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:

Human keratinocytes have two interconvertible modes of proliferation

Abstract

Single stem cells, including those in human epidermis, have a remarkable ability to reconstitute tissues in vitro, but the cellular mechanisms that enable this are ill-defined. Here we used live imaging to track the outcome of thousands of divisions in clonal cultures of primary human epidermal keratinocytes. Two modes of proliferation were seen. In ‘balanced’ mode, similar proportions of proliferating and differentiating cells were generated, achieving the ‘population asymmetry’ that sustains epidermal homeostasis in vivo. In ‘expanding’ mode, an excess of cycling cells was produced, generating large expanding colonies. Cells in expanding mode switched their behaviour to balanced mode once local confluence was attained. However, when a confluent area was wounded in a scratch assay, cells near the scratch switched back to expanding mode until the defect was closed. We conclude that the ability of a single epidermal stem cell to reconstitute an epithelium is explained by two interconvertible modes of proliferation regulated by confluence.

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: Live imaging of cultured keratinocytes.
Figure 2: Lineage trees of NFSKs cultured at clonal density.
Figure 3: Division outcomes of sister cells.
Figure 4: Transcriptional analysis of colonies.
Figure 5: Expanding colonies switch towards balance.
Figure 6: Cell density and ROCK2 kinase activity regulate the switch from expanding to balanced mode of proliferation.
Figure 7: Effects of loss of confluence on mode of proliferation in large colonies.
Figure 8: Two interconvertible modes of proliferation underpin epithelial reconstitution in vitro.

Similar content being viewed by others

References

  1. Rheinwald, J. G. & Green, H. Serial cultivation of strains of human epidermal keratinocytes: the formation of keratinizing colonies from single cells. Cell 6, 331–343 (1975).

    Article  CAS  PubMed  Google Scholar 

  2. Watt, F. M. & Green, H. Stratification and terminal differentiation of cultured epidermal cells. Nature 295, 434–436 (1982).

    Article  CAS  PubMed  Google Scholar 

  3. Sato, T. & Clevers, H. Growing self-organizing mini-guts from a single intestinal stem cell: mechanism and applications. Science 340, 1190–1194 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Huch, M. et al. Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. EMBO J. 32, 2708–2721 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Huch, M. et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature 494, 247–250 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Jones, P. H. & Watt, F. M. Separation of human epidermal stem cells from transit amplifying cells on the basis of differences in integrin function and expression. Cell 73, 713–724 (1993).

    Article  CAS  PubMed  Google Scholar 

  7. Barrandon, Y. & Green, H. Three clonal types of keratinocyte with different capacities for multiplication. Proc. Natl Acad. Sci. USA 84, 2302–2306 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Janich, P. et al. Human epidermal stem cell function is regulated by circadian oscillations. Cell Stem Cell 13, 745–753 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Luis, N. M. et al. Regulation of human epidermal stem cell proliferation and senescence requires polycomb-dependent and -independent functions of Cbx4. Cell Stem Cell 9, 233–246 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. Mulder, K. W. et al. Diverse epigenetic strategies interact to control epidermal differentiation. Nat. Cell Biol. 14, 753–763 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Connelly, J. T. et al. Actin and serum response factor transduce physical cues from the microenvironment to regulate epidermal stem cell fate decisions. Nat. Cell Biol. 12, 711–718 (2010).

    CAS  PubMed  Google Scholar 

  12. Tan, D. W. et al. Single-cell gene expression profiling reveals functional heterogeneity of undifferentiated human epidermal cells. Development 140, 1433–1444 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Li, A., Simmons, P. J. & Kaur, P. Identification and isolation of candidate human keratinocyte stem cells based on cell surface phenotype. Proc. Natl Acad. Sci. USA 95, 3902–3907 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lowell, S., Jones, P., Le Roux, I., Dunne, J. & Watt, F. M. Stimulation of human epidermal differentiation by δ-notch signalling at the boundaries of stem-cell clusters. Curr. Biol. 10, 491–500 (2000).

    Article  CAS  PubMed  Google Scholar 

  15. Rochat, A., Kobayashi, K. & Barrandon, Y. Location of stem cells of human hair follicles by clonal analysis. Cell 76, 1063–1073 (1994).

    Article  CAS  PubMed  Google Scholar 

  16. Gallico, G. G. III, O’Connor, N. E., Compton, C. C., Kehinde, O. & Green, H. Permanent coverage of large burn wounds with autologous cultured human epithelium. New Engl. J. Med. 311, 448–451 (1984).

    Article  PubMed  Google Scholar 

  17. Compton, C. C. et al. Skin regenerated from cultured epithelial autografts on full-thickness burn wounds from 6 days to 5 years after grafting. A light, electron microscopic and immunohistochemical study. Lab Invest. 60, 600–612 (1989).

    CAS  PubMed  Google Scholar 

  18. Jones, P. H., Harper, S. & Watt, F. M. Stem cell patterning and fate in human epidermis. Cell 80, 83–93 (1995).

    Article  CAS  PubMed  Google Scholar 

  19. Fortunel, N. O. et al. Exploration of the functional hierarchy of the basal layer of human epidermis at the single-cell level using parallel clonal microcultures of keratinocytes. Exp. Dermatol. 19, 387–392 (2010).

    Article  PubMed  Google Scholar 

  20. Clayton, E. et al. A single type of progenitor cell maintains normal epidermis. Nature 446, 185–189 (2007).

    Article  CAS  PubMed  Google Scholar 

  21. Doupe, D. P., Klein, A. M., Simons, B. D. & Jones, P. H. The ordered architecture of murine ear epidermis is maintained by progenitor cells with random fate. Dev. Cell 18, 317–323 (2010).

    Article  CAS  PubMed  Google Scholar 

  22. Mascre, G. et al. Distinct contribution of stem and progenitor cells to epidermal maintenance. Nature 489, 257–262 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Lim, X. et al. Interfollicular epidermal stem cells self-renew via autocrine Wnt signaling. Science 342, 1226–1230 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Alcolea, M. P. et al. Differentiation imbalance in single oesophageal progenitor cells causes clonal immortalization and field change. Nat. Cell Biol. 16, 615–622 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Coutu, D. L. & Schroeder, T. Probing cellular processes by long-term live imaging—historic problems and current solutions. J. Cell Sci. 126, 3805–3815 (2013).

    CAS  PubMed  Google Scholar 

  26. Thon, J. N., Devine, M. T., Jurak Begonja, A., Tibbitts, J. & Italiano, J. E. Jr High-content live-cell imaging assay used to establish mechanism of trastuzumab emtansine (T-DM1)-mediated inhibition of platelet production. Blood 120, 1975–1984 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Watt, F. M. Epidermal stem cells as targets for gene transfer. Hum. Gene Ther. 11, 2261–2266 (2000).

    Article  CAS  PubMed  Google Scholar 

  28. He, J. et al. How variable clones build an invariant retina. Neuron 75, 786–798 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Simpson, P. Lateral inhibition and the development of the sensory bristles of the adult peripheral nervous system of Drosophila. Development 109, 509–519 (1990).

    Article  CAS  PubMed  Google Scholar 

  30. Rangarajan, A. et al. Notch signaling is a direct determinant of keratinocyte growth arrest and entry into differentiation. EMBO J. 20, 3427–3436 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Doupe, D. P. et al. A single progenitor population switches behavior to maintain and repair esophageal epithelium. Science 337, 1091–1093 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Alcolea, M. P. & Jones, P. H. Tracking cells in their native habitat: lineage tracing in epithelial neoplasia. Nat. Rev. Cancer 13, 161–171 (2013).

    Article  CAS  PubMed  Google Scholar 

  33. Cicalese, A. et al. The tumor suppressor p53 regulates polarity of self-renewing divisions in mammary stem cells. Cell 138, 1083–1095 (2009).

    Article  CAS  PubMed  Google Scholar 

  34. van den Bogaerdt, A. J. et al. Upside-down transfer of porcine keratinocytes from a porous, synthetic dressing to experimental full-thickness wounds. Wound Repair Regen. 12, 225–234 (2004).

    Article  PubMed  Google Scholar 

  35. O’Shaughnessy, R. F., Seery, J. P., Celis, J. E., Frischauf, A. & Watt, F. M. PA-FABP, a novel marker of human epidermal transit amplifying cells revealed by 2D protein gel electrophoresis and cDNA array hybridisation. FEBS Lett. 486, 149–154 (2000).

    Article  PubMed  Google Scholar 

  36. Eckert, R. L. et al. S100 proteins in the epidermis. J. Invest. Dermatol. 123, 23–33 (2003).

    Article  Google Scholar 

  37. Signer, R. A., Magee, J. A., Salic, A. & Morrison, S. J. Haematopoietic stem cells require a highly regulated protein synthesis rate. Nature 509, 49–54 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Liu, J., Xu, Y., Stoleru, D. & Salic, A. Imaging protein synthesis in cells and tissues with an alkyne analog of puromycin. Proc. Natl Acad. Sci. USA 109, 413–418 (2012).

    Article  CAS  PubMed  Google Scholar 

  39. Shyh-Chang, N. et al. Lin28 enhances tissue repair by reprogramming cellular metabolism. Cell 155, 778–792 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zhang, Q., Shalaby, N. A. & Buszczak, M. Changes in rRNA transcription influence proliferation and cell fate within a stem cell lineage. Science 343, 298–301 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Buszczak, M., Signer, R. A. J. & Morrison, S. J. Cellular differences in protein synthesis regulate tissue homeostasis. Cell 159, 242–251 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Taguchi, S. et al. Overexpression of the transcription factor Yin-Yang-1 suppresses differentiation of HaCaT cells in three-dimensional cell culture. J. Invest. Dermatol. 131, 37–45 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Shaw, T. & Martin, P. Epigenetic reprogramming during wound healing: loss of polycomb-mediated silencing may enable upregulation of repair genes. EMBO Rep. 10, 881–886 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Perekatt, A. O. et al. YY1 is indispensable for Lgr5 + intestinal stem cell renewal. Proc. Natl Acad. Sci. USA 111, 7695–7700 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Xu, X. et al. Yin-yang 1 negatively regulates the differentiation-specific transcription of mouse loricrin gene in undifferentiated keratinocytes. J. Invest. Dermatol. 123, 1120–1126 (2004).

    Article  CAS  PubMed  Google Scholar 

  46. Somervaille, T. C. et al. Hierarchical maintenance of MLL myeloid leukemia stem cells employs a transcriptional program shared with embryonic rather than adult stem cells. Cell Stem Cell 4, 129–140 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Wongtawan, T., Taylor, J. E., Lawson, K. A., Wilmut, I. & Pennings, S. Histone H4K20me3 and HP1α are late heterochromatin markers in development, but present in undifferentiated embryonic stem cells. J. Cell Sci. 124, 1878–1890 (2011).

    Article  CAS  PubMed  Google Scholar 

  48. Barrandon, Y. & Green, H. Cell migration is essential for sustained growth of keratinocyte colonies: the roles of transforming growth factor-α and epidermal growth factor. Cell 50, 1131–1137 (1987).

    Article  CAS  PubMed  Google Scholar 

  49. Rheinwald, J. G. & Green, H. Epidermal growth factor and the multiplication of cultured human epidermal keratinocytes. Nature 265, 421–424 (1977).

    Article  CAS  PubMed  Google Scholar 

  50. Bellas, E. & Chen, C. S. Forms, forces, and stem cell fate. Curr. Opin. Cell Biol. 31, 92–97 (2014).

    Article  CAS  PubMed  Google Scholar 

  51. Barker, N. et al. Lgr5(+ ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell 6, 25–36 (2010).

    Article  CAS  PubMed  Google Scholar 

  52. Sato, T. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009).

    Article  CAS  PubMed  Google Scholar 

  53. Chapman, S., McDermott, D. H., Shen, K., Jang, M. K. & McBride, A. A. The effect of Rho kinase inhibition on long-term keratinocyte proliferation is rapid and conditional. Stem Cell Res. Ther. 5, 60 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Chapman, S., Liu, X., Meyers, C., Schlegel, R. & McBride, A. A. Human keratinocytes are efficiently immortalized by a Rho kinase inhibitor. J. Clin. Invest. 120, 2619–2626 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Luxenburg, C. et al. Wdr1-mediated cell shape dynamics and cortical tension are essential for epidermal planar cell polarity. Nat. Cell Biol. 17, 592–604 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Lee, B. et al. Transcriptional mechanisms link epithelial plasticity to adhesion and differentiation of epidermal progenitor cells. Dev. Cell 29, 47–58 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Amelio, I. et al. miR-24 triggers epidermal differentiation by controlling actin adhesion and cell migration. J. Cell Biol. 199, 347–363 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Nanba, D. et al. Cell motion predicts human epidermal stemness. J. Cell Biol. 209, 305–315 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Lecuit, T. & Yap, A. S. E-cadherin junctions as active mechanical integrators in tissue dynamics. Nat. Cell Biol. 17, 533–539 (2015).

    Article  CAS  PubMed  Google Scholar 

  60. Zhu, A. J., Haase, I. & Watt, F. M. Signaling via β1 integrins and mitogen-activated protein kinase determines human epidermal stem cell fate in vitro. Proc. Natl Acad. Sci. USA 96, 6728–6733 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Nanba, D. et al. Actin filament dynamics impacts keratinocyte stem cell maintenance. EMBO Mol. Med. 5, 640–653 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. McMullan, R. et al. Keratinocyte differentiation is regulated by the Rho and ROCK signaling pathway. Curr. Biol. 13, 2185–2189 (2003).

    Article  CAS  PubMed  Google Scholar 

  63. Iglesias-Bartolome, R. et al. mTOR inhibition prevents epithelial stem cell senescence and protects from radiation-induced mucositis. Cell Stem Cell 11, 401–414 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Wabik, A. & Jones, P. H. Switching roles: the functional plasticity of adult tissue stem cells. EMBO J. 34, 1164–1179 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Centanin, L. et al. Exclusive multipotency and preferential asymmetric divisions in post-embryonic neural stem cells of the fish retina. Development 141, 3472–3482 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Rompolas, P., Mesa, K. R. & Greco, V. Spatial organization within a niche as a determinant of stem-cell fate. Nature 502, 513–518 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Buczacki, S. J. et al. Intestinal label-retaining cells are secretory precursors expressing Lgr5. Nature 495, 65–69 (2013).

    Article  CAS  PubMed  Google Scholar 

  68. Aspinall-O’Dea, M. et al. Antibody-based detection of protein phosphorylation status to track the efficacy of novel therapies using nanogram protein quantities from stem cells and cell lines. Nat. Protoc. 10, 149–168 (2015).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  70. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The initial association of holoclone and paraclone type behaviour in clonal cultures of NFSKs with stem and balanced progenitor dynamics was recognized by B.D.S. working in collaboration with P.H.J., V.N.-N., D. Doupé and A. Klein, based on the quantitative analysis of published and unpublished colony size distributions6. We thank G. Akdeniz and D. Doupé for experimental work that led up to the project that was analysed by A. Klein and G. Zhang, P. Lombard at the Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute for Bioinformatics analysis and E. Choolun for technical assistance. We acknowledge the support of the Wellcome Trust, Cambridge Cancer Centre, Medical Research Council, the NC3Rs (National Centre for the Replacement, Refinement and Reduction of Animals in Research) and Cancer Research UK (Programme grant C609/A17257).

Author information

Authors and Affiliations

Authors

Contributions

A.R., K.M., J.F., V.N.-N. and P.H.J. designed and performed experiments and analysed and interpreted data. B.D.S. proposed using growth rate from 144 to 168 h to classify division mode in dye-labelled cells. A.R., K.M., J.F. and P.H.J. wrote the manuscript.

Corresponding author

Correspondence to Philip H. Jones.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Lineage trees of primary adult human keratinocytes in vitro.

Keratinocytes cultured directly from adult epidermis in standard media. Scale indicates time since plating in hours. Magenta indicates cells that did not divide within 48 h, green cells observed to divide, grey cells those which could not be tracked for 48 h. a, balanced, b expanding trees. Horizontal brackets in b, marked by , indicate representative cells tracked within a single expanding colony.

Supplementary Figure 2 Live imaging of primary adult human keratinocytes and validation of two-mode model.

ac, Live imaging of keratinocytes cultured directly from adult epidermis. a: Cell cycle time distribution b, c: Division outcomes (% with 95% confidence interval) in balanced, b (547 divisions in 50 colonies), and expanding, c lineages (609 divisions, 6 colonies). See Supplementary Fig. 1a, b for lineage trees. d, e Predictions from a large simulation of keratinocyte growth. Simulated and observed colony size distributions. d: Box plots showing day 7 colony size distributions from a computer simulation assuming two modes of growth with division outcomes shown in Fig. 1 (Simulated, n = 30000 colonies) compared with observed sizes from cultured NFSK (Observed, n = 1,631 colonies pooled from 3 independent experiments). Box boundaries indicate the 25th and 75th percentiles. Line across box is the median. Whiskers indicate 1st and 99th percentiles. There is no statistically significant difference between the distributions (Kolmogorov–Smirnov test P = 0.15). e: Proportions of simulated colonies containing one or more cycling Roshan Keratinocyte Proliferation 2 cells after 7 days, red line indicates balanced colonies, red shading balanced colonies with one or more proliferating cells and green shading expanding colonies. Arrow indicates overlap of colonies containing 50–150 cells, which may be from cells proliferating in either mode.

Supplementary Figure 3 Tracking colony expansion with PKH26 labelled cells.

a: Protocol. NFSK were labelled with PKH26 and plated at clonal density. Every 24 h the coordinates of each colony and the number of cells per colony were determined by fluorescence microscopy. At 168 h dishes were fixed and stained. b,c Images of typical colonies, showing phase contrast images overlaid with PKH26 fluorescence (orange) for time points up to 168 h with the number of cells/colony and the fixed 168 h colonies stained for Dapi, blue, pan cytokeratin (green) and EdU (red). Scale bars: 100 μm. Images are representative examples of 333 colonies in 3 independent experiments.

Supplementary Figure 4 Analysis of PKH26 tracking data.

a: Live imaging data (Fig. 2b) shows that in larger balanced mode colonies the accumulation of differentiating cells (red) resulted in a proportionate increase in cell number between 144 and 168 h 150 cells are expanding. d: Cells per clone in PKH26 labelled colonies. Each line represents a single colony growth with, inset showing enlarged view of colonies between 50 and 150 cells. Full data set is given in Supplementary Table 3. e: Mean cells per colony versus time in balanced mode colonies (n = 304 colonies). The increase is linear (r2 = 0.98), dotted lines indicate 95% confidence intervals. f: Mean cells per colony versus time in expanding mode colonies (n = 29 colonies) showing an exponential increase, dotted lines indicate 95% confidence intervals.

Supplementary Figure 5 Effect of increased cell density on NFSK proliferation.

a: Immunoblot analysis of EGF signalling in NFSK. Lysates were collected at the times indicated after plating keratinocytes in media containing 0, 10 or 20 ng ml−1 added EGF. Arrowheads indicate EGFR degradation product. Position of size markers (kDa) is as indicated. Blots shown are representative of 3 independent experiments. b: Cell cycle time distribution of NFSK cultured in media without supplemented EGF (EGF0). c, d Lineage trees NFSK cultured in EGF0. Scale indicates time since plating in hours. Magenta indicates cells that did not divide within 48 h, green cells observed to divide, grey cells those that could not be tracked for 48 h. c, balanced, d expanding Roshan Keratinocyte Proliferation 4 trees. Horizontal brackets marked by in b indicate representative cells tracked within a single expanding colony. In d large green circles, arrowed, indicate cells lying in outer third (by area) of expanding colony, other green cells lie within inner two thirds of colonies at 96 h.

Supplementary Figure 6 Effect of Y27632 on NFSK proliferation.

a,b: Lineage trees of NFSK cultured in standard media in the presence of 10 μM Y27632. Scale indicates time since plating in hours. Magenta indicates cells that did not divide within 48 h, green cells observed to divide, grey cells those that could not be tracked for 48 h. a, balanced, b expanding trees. Horizontal brackets in b, marked by , indicate representative cells tracked within a single expanding colony.

Supplementary Figure 7 Scans of Western blots.

Scans of four blots (1 to 4) presented in cropped form in Supplementary Fig. 5a.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2727 kb)

Supplementary Table 1

Supplementary Information (XLSX 1407 kb)

Supplementary Table 2

Supplementary Information (XLSX 35 kb)

Supplementary Table 3

Supplementary Information (XLSX 53 kb)

Supplementary Table 4

Supplementary Information (XLSX 32 kb)

Supplementary Table 5

Supplementary Information (XLSX 47 kb)

Supplementary Table 6

Supplementary Information (XLSX 38 kb)

Supplementary Table 7

Supplementary Information (XLSX 8 kb)

Supplementary Table 8

Supplementary Information (XLSX 46 kb)

Supplementary Table 9

Supplementary Information (XLSX 43 kb)

Supplementary Table 10

Supplementary Information (XLSX 48 kb)

Supplementary Table 11

Supplementary Information (XLSX 47 kb)

Supplementary Table 12

Supplementary Information (XLSX 9 kb)

Time lapse imaging of typical NFSK expanding type colony.

Clock indicates time since plating, yellow dashed line NFSK cells, other cells are mouse 3T3 J2 fibroblast feeder cells, scale bar 100 μm. (MP4 10471 kb)

Time lapse imaging of typical NFSK balanced type colony.

Clock indicates time since plating, yellow dashed line NFSK cells, other cells are mouse 3T3 J2 fibroblast feeder cells, scale bar 100 μm. (MP4 10627 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Roshan, A., Murai, K., Fowler, J. et al. Human keratinocytes have two interconvertible modes of proliferation. Nat Cell Biol 18, 145–156 (2016). https://doi.org/10.1038/ncb3282

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3282

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