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.

  • Resource
  • Published:

Human melanocyte development and melanoma dedifferentiation at single-cell resolution

Abstract

In humans, epidermal melanocytes are responsible for skin pigmentation, defence against ultraviolet radiation and the deadliest common skin cancer, melanoma. Although there is substantial overlap in melanocyte development pathways between different model organisms, species-dependent differences are frequent and the conservation of these processes in human skin remains unresolved. Here, we used a single-cell enrichment and RNA-sequencing pipeline to study human epidermal melanocytes directly from the skin, capturing transcriptomes across different anatomical sites, developmental age, sexes and multiple skin tones. We uncovered subpopulations of melanocytes that exhibit anatomical site-specific enrichment that occurs during gestation and persists through adulthood. The transcriptional signature of the volar-enriched subpopulation is retained in acral melanomas. Furthermore, we identified human melanocyte differentiation transcriptional programs that are distinct from gene signatures generated from model systems. Finally, we used these programs to define patterns of dedifferentiation that are predictive of melanoma prognosis and response to immune checkpoint inhibitor therapy.

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

Fig. 1: Melanocyte transcriptomic profiles differ based on development and anatomical location.
Fig. 2: Characterization of divergent pigment developmental trajectories in volar and non-volar melanocytes.
Fig. 3: Anatomical site-specific melanocyte subpopulation enrichment arises during development and persists in adulthood.
Fig. 4: Defining transcriptomic programs specific to human melanocyte development.
Fig. 5: Evaluation of the expression of melanocyte developmental programs from mammalian models in human non-volar cutaneous melanocyte developmental groups.
Fig. 6: Identification of distinct patterns of developmental programs reacquired in metastasized melanomas.
Fig. 7: Reacquisition of specific developmental programs in heterogeneous melanoma is prognostic.

Similar content being viewed by others

Data availability

All healthy human skin scRNA-seq data generated for this study has been deposited in the Gene Expression Omnibus (GEO) database repository and are available under accession number GSE151091. Human melanoma datasets were obtained from publicly accessible repositories: GSE65904, GSE72056, GSE115978, dbGAP phs001036.v1.p1, TCGA Research Network (https://www.cancer.gov/tcga), and the Single Cell portal (https://portals.broadinstitute.org/single_cell/study/melanoma-immunotherapy-resistance). All other data supporting the findings of this study are available from the corresponding authors on reasonable request; lead contact: R.L.J.-T. Source data are provided with this paper.

Code availability

Jupyter notebooks with detailed analysis scripts are available at GitHub (https://github.com/danledinh/human_melanocytes)83.

References

  1. Yamaguchi, Y. et al. Mesenchymal-epithelial interactions in the skin: Increased expression of dickkopf1 by palmoplantar fibroblasts inhibits melanocyte growth and differentiation. J. Cell Biol. 165, 275–285 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Adameyko, I. et al. Schwann cell precursors from nerve innervation are a cellular origin of melanocytes in skin. Cell 139, 366–379 (2009).

    Article  CAS  PubMed  Google Scholar 

  3. Mort, R. L., Jackson, I. J. & Elizabeth Patton, E. The melanocyte lineage in development and disease. Development 142, 620–632 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hayward, N. K. et al. Whole-genome landscapes of major melanoma subtypes. Nature 545, 175–180 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Rabbie, R., Ferguson, P., Molina-Aguilar, C., Adams, D. J. & Robles-Espinoza, C. D. Melanoma subtypes: genomic profiles, prognostic molecular markers and therapeutic possibilities. J. Pathol. 247, 539–551 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Malta, T. M. et al. Machine learning identifies stemness features associated with oncogenic dedifferentiation. Cell 173, 338–354 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Gupta, P. B. et al. The melanocyte differentiation program predisposes to metastasis after neoplastic transformation. Nat. Genet. 37, 1047–1054 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Vorstandlechner, V. et al. Deciphering the functional heterogeneity of skin fibroblasts using single-cell RNA sequencing. FASEB J. 34, 3677–3692 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Solé-Boldo, L. et al. Single-cell transcriptomes of the human skin reveal age-related loss of fibroblast priming. Commun. Biol. 3, 188 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Cheng, J. B. et al. Transcriptional programming of normal and inflamed human epidermis at single-cell resolution. Cell Rep. 25, 871–883 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Takahashi, R. et al. Defining transcriptional signatures of human hair follicle cell states. J. Invest. Dermatol. 140, 764–773 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Popescu, D. M. et al. Decoding human fetal liver haematopoiesis. Nature 574, 365–371 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gao, S. et al. Tracing the temporal-spatial transcriptome landscapes of the human fetal digestive tract using single-cell RNA-sequencing. Nat. Cell Biol. 20, 721–734 (2018).

    Article  CAS  PubMed  Google Scholar 

  14. Cao, J. et al. A human cell atlas of fetal gene expression. Science 370, eaba7721 (2020).

  15. Sridhar, A. et al. Single-cell transcriptomic comparison of human fetal retina, hPSC-derived retinal organoids, and long-term retinal cultures. Cell Rep. 30, 1644–1659 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Belote, R. L. & Simon, S. M. Ca2+ transients in melanocyte dendrites and dendritic spine-like structures evoked by cell-to-cell signaling. J. Cell Biol. 219, e201902014 (2020).

    Article  PubMed  CAS  Google Scholar 

  17. Norris, A., Todd, C., Graham, A., Quinn, A. G. & Thody, A. J. The expression of the c-kit receptor by epidermal melanocytes may be reduced in vitiligo. Br. J. Dermatol. 134, 299–306 (1996).

    Article  CAS  PubMed  Google Scholar 

  18. Randall, V. A., Jenner, T. J., Hibberts, N. A., De Oliveira, I. O. & Vafaee, T. Stem cell factor/c-Kit signalling in normal and androgenetic alopecia hair follicles. J. Endocrinol. 197, 11–23 (2008).

    Article  CAS  PubMed  Google Scholar 

  19. Picelli, S. et al. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat. Methods 10, 1096–1100 (2013).

    Article  CAS  PubMed  Google Scholar 

  20. Hsiao, C. J. et al. Characterizing and inferring quantitative cell cycle phase in single-cell RNA-seq data analysis. Genome Res. 30, 611–621 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lu, R. et al. Transcription factor TCF4 maintains the properties of human corneal epithelial stem cells. Stem Cells 30, 753–761 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Li, Z., Li, Y. & Jiao, J. Neural progenitor cells mediated by H2A.Z.2 regulate microglial development via Cxcl14 in the embryonic brain. Proc. Natl Acad. Sci. USA 116, 24122–24132 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Denecker, G. et al. Identification of a ZEB2-MITF-ZEB1 transcriptional network that controls melanogenesis and melanoma progression. Cell Death Differ. 21, 1250–1261 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Nishikawa, S.-I. & Osawa, M. Generating quiescent stem cells. Pigment Cell Res. 20, 263–270 (2007).

    Article  PubMed  Google Scholar 

  25. Joshi, S. S. et al. CD34 defines melanocyte stem cell subpopulations with distinct regenerative properties. PLOS Genet. 15, e1008034 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Choi, H. R., Park, S. H., Choi, J. W., Kim, D. S. & Park, K. C. A simple assay method for melanosome transfer. Ann. Dermatol. 24, 90–93 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Nakamura, M. et al. Site-specific migration of human fetal melanocytes in volar skin. J. Dermatol. Sci. 78, 143–148 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. Cramer, S. F. & Fesyuk, A. On the development of neurocutaneous units—Implications for the histogenesis of congenital, acquired, and dysplastic nevi. Am. J. Dermatopathol. 34, 60–81 (2012).

    Article  PubMed  Google Scholar 

  29. Baxter, L. L., Watkins-Chow, D. E., Pavan, W. J. & Loftus, S. K. A curated gene list for expanding the horizons of pigmentation biology. Pigment Cell Melanoma Res. 32, 348–358 (2019).

    Article  PubMed  Google Scholar 

  30. Adhikari, K. et al. A GWAS in Latin Americans highlights the convergent evolution of lighter skin pigmentation in Eurasia. Nat. Commun. 10, 358 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Crawford, N. G. et al. Loci associated with skin pigmentation identified in African populations. Science 358, eaan8433 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Han, J. et al. A genome-wide association study identifies novel alleles associated with hair color and skin pigmentation. PLoS Genet. 4, e1000074 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Sturm, R. A. A golden age of human pigmentation genetics. Trends Genet. 22, 464–468 (2006).

    Article  CAS  PubMed  Google Scholar 

  34. Antunes, L. C. M. et al. Tropomyosin-related kinase receptor and neurotrophin expression in cutaneous melanoma is associated with a poor prognosis and decreased survival. Oncology 97, 26–37 (2019).

    Article  CAS  PubMed  Google Scholar 

  35. DiVito, K. A., Simbulan-Rosenthal, C. M., Chen, Y. S., Trabosh, V. A. & Rosenthal, D. S. Id2, Id3 and Id4 overcome a Smad7-mediated block in tumorigenesis, generating TGF-β-independent melanoma. Carcinogenesis 35, 951–958 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Yamaguchi, Y. et al. Epithelial-mesenchymal interactions in wounds: treatment of palmoplantar wounds by nonpalmoplantar pure epidermal sheet grafts. Arch. Dermatol. 137, 621–628 (2001).

    CAS  PubMed  Google Scholar 

  37. Bolognia, J., Schaffer, J. & Cerroni, L. Dermatology 4th edn (Elsevier, 2018).

  38. Bradford, P. T., Goldstein, A. M., McMaster, M. L. & Tucker, M. A. Acral lentiginous melanoma: Incidence and survival patterns in the United States, 1986-2005. Arch. Dermatol. 145, 427–434 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Mahendraraj, K. et al. Malignant melanoma in African-Americans. Medicine 96, e6258 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Ashburner, M. et al. Gene ontology: tool for the unification of biology. Nat. Genet. 25, 25–29 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Carbon, S. et al. The Gene Ontology resource: enriching a GOld mine. Nucleic Acids Res. 49, D325–D334 (2021).

    Article  CAS  Google Scholar 

  42. Hou, L., Arnheiter, H. & Pavan, W. J. Interspecies difference in the regulation of melanocyte development by SOX10 and MITF. Proc. Natl Acad. Sci. USA 103, 9081–9085 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Marie, K. L. et al. Melanoblast transcriptome analysis reveals pathways promoting melanoma metastasis. Nat. Commun. 11, 333 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Rezza, A. et al. Signaling networks among stem cell precursors, transit-amplifying progenitors, and their niche in developing hair follicles. Cell Rep. 14, 3001–3018 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Sennett, R. et al. An integrated transcriptome atlas of embryonic hair follicle progenitors, their niche, and the developing skin. Dev. Cell 34, 577–591 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Mica, Y., Lee, G., Chambers, S. M., Tomishima, M. J. & Studer, L. Modeling neural crest induction, melanocyte specification, and disease-related pigmentation defects in hESCs and patient-specific iPSCs. Cell Rep. 3, 1140–1152 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Osawa, M. et al. Molecular characterization of melanocyte stem cells in their niche. Development 132, 5589–5599 (2005).

    Article  CAS  PubMed  Google Scholar 

  48. Lu, Z. et al. Hair follicle stem cells regulate retinoid metabolism to maintain the self-renewal niche for melanocyte stem cells. eLife 9, e52712 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Saxena, N., Mok, K. W. & Rendl, M. An updated classification of hair follicle morphogenesis. Exp. Dermatol. 28, 332–344 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Gleason, B. C., Crum, C. P. & Murphy, G. F. Expression patterns of MITF during human cutaneous embryogenesis: evidence for bulge epithelial expression and persistence of dermal melanoblasts. J. Cutan. Pathol. 35, 615–622 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Holbrook, K. A., Underwood, R. A., Vogel, A. M., Gown, A. M. & Kimball, H. The appearance, density and distribution of melanocytes in human embryonic and fetal skin revealed by the anti-melanoma monoclonal antibody, HMB-45. Anat. Embryol. 180, 443–455 (1989).

    Article  CAS  Google Scholar 

  52. Hoek, K. S. et al. In vivo switching of human melanoma cells between proliferative and invasive states. Cancer Res. 68, 650–656 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. Tsoi, J. et al. Multi-stage differentiation defines melanoma subtypes with differential vulnerability to drug-induced iron-dependent oxidative stress. Cancer Cell 33, 890–904 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Richard, G. et al. ZEB 1-mediated melanoma cell plasticity enhances resistance to MAPK inhibitors. EMBO Mol. Med. 8, 1143–1161 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Landsberg, J. et al. Melanomas resist T-cell therapy through inflammation-induced reversible dedifferentiation. Nature 490, 412–416 (2012).

    Article  CAS  PubMed  Google Scholar 

  56. Grzywa, T. M., Paskal, W. & Włodarski, P. K. Intratumor and intertumor heterogeneity in melanoma. Transl. Oncol. 10, 956–975 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Jerby-Arnon, L. et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 175, 984–997 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Akbani, R. et al. Genomic classification of cutaneous melanoma. Cell 161, 1681–1696 (2015).

    Article  CAS  Google Scholar 

  60. Cirenajwis, H. et al. Molecular stratification of metastatic melanoma using gene expression profiling—prediction of survival outcome and benefit from molecular targeted therapy. Oncotarget 6, 12297–12309 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Widmer, D. S. et al. Systematic classification of melanoma cells by phenotype-specific gene expression mapping. Pigment Cell Melanoma Res. 25, 343–353 (2012).

    Article  CAS  PubMed  Google Scholar 

  62. Raskin, L. et al. Transcriptome profiling identifies HMGA2 as a biomarker of melanoma progression and prognosis. J. Invest. Dermatol. 133, 2585–2592 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Webster, M. R. et al. Wnt5A promotes an adaptive, senescent-like stress response, while continuing to drive invasion in melanoma cells. Pigment Cell Melanoma Res. 28, 184–195 (2015).

    Article  CAS  PubMed  Google Scholar 

  64. Rambow, F. et al. Toward minimal residual disease-directed therapy in melanoma. Cell 174, 843–855 (2018).

    Article  CAS  PubMed  Google Scholar 

  65. Guo, B. et al. Humanin peptide suppresses apoptosis by interfering with Bax activation. Nature 423, 456–461 (2003).

    Article  CAS  PubMed  Google Scholar 

  66. Newman, A. M. et al. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods 12, 453–457 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Wishart, D. S. et al. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res. 46, D1074–D1082 (2018).

    Article  CAS  PubMed  Google Scholar 

  68. Okamoto, N. et al. A melanocyte-melanoma precursor niche in sweat glands of volar skin. Pigment Cell Melanoma Res. 27, 1039–1050 (2014).

    Article  CAS  PubMed  Google Scholar 

  69. Nitzan, E., Pfaltzgraff, E. R., Labosky, P. A. & Kalcheim, C. Neural crest and Schwann cell progenitor-derived melanocytes are two spatially segregated populations similarly regulated by Foxd3. Proc. Natl Acad. Sci. USA 110, 12709–12714 (2013).

  70. Goydos, J. S. & Shoen, S. L. in Cancer Treatment and Research Vol. 167 321–329 (Kluwer Academic Publishers, 2016).

  71. Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, 2020. CA Cancer J. Clinicians 70, 7–30 (2020).

    Article  Google Scholar 

  72. Sennepin, A. et al. The human penis is a genuine immunological effector site. Front. Immunol. 8, 1732 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Lezcano, C., Jungbluth, A. A., Nehal, K. S., Hollmann, T. J. & Busam, K. J. PRAME expression in melanocytic tumors. Am. J. Surgical Pathol. 42, 1456–1465 (2018).

    Article  Google Scholar 

  74. Drey, E. A., Kang, M. S., McFarland, W. & Darney, P. D. Improving the accuracy of fetal foot length to confirm gestational duration. Obstet. Gynecol. 105, 773–778 (2005).

    Article  PubMed  Google Scholar 

  75. Darmanis, S. et al. A survey of human brain transcriptome diversity at the single cell level. Proc. Natl Acad. Sci. USA 112, 7285–7290 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Liang, W. S. et al. Integrated genomic analyses reveal frequent TERT aberrations in acral melanoma. Genome Res. 27, 524–532 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  78. Anders, S., Pyl, P. T. & Huber, W. HTSeq-A Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  79. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  80. Hsiao, C. J. et al. Characterizing and inferring quantitative cell cycle phase in single-cell RNA-seq data analysis. Genome Res. 30, 611–621 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

  82. 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 

  83. Jupyter note books: danledinh/human_melanocytes. Zenodo https://doi.org/10.5281/zenodo.5076159 (2021).

  84. Liu, J. et al. An integrated TCGA pan-cancer clinical data resource to drive high-quality survival outcome analytics. Cell 173, 400–416 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the staff at the University of California, San Francisco Program for Breakthrough Biomedical Research Sandler Fellows Program for funding (to R.L.J.-T.); the staff at the University of California, San Francisco Biospecimen Resource Program for support with tissue acquisition; N. Neff and M. Tan for assistance with library quality control and sequencing; the staff at the Huntsman Cancer Institute Bioinformatic Analysis Shared Resource and University of Utah Center for High Performance Computing for supporting analyses of acral tumour samples at the Huntsman Cancer Institute; and the staff at Life Science Editors for critical editing of the manuscript. Funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: R.L.B., D.L., A.D.T, S.D. and R.L.J.-T. Methodology: R.L.B. and A.M. Validation: R.L.B. and D.L. Formal analysis: R.L.B. and D.L. Investigation: R.L.B., D.L. and A.M. Resources: U.E.L., A.S., B.K.L., V.P.-P., L.B. and A.D.T. Data curation: D.L., A.M. and B.K.L. Writing—original draft: R.L.B. and R.L.J.-T. Writing—review and editing: D.L., A.M., U.E.L. and S.D. Visualization: R.L.B. and D.L. Supervision: S.D. and R.L.J.-T. Funding acquisition: S.D. and R.L.J.-T.

Corresponding authors

Correspondence to Spyros Darmanis or Robert L. Judson-Torres.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Cell Biology thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Single cell RNA sequencing quality control, cell-type specific markers, and donor age.

a) FACS gate protocol for representative sort. Melanocytes (blue circles in live, scatter, and singlets) were isolated as KIT+ cells from the CD11c- gate. b) Fraction of cells from each indexed FACS gate assignment. c) Number of reads and d) number of genes per cell for all 14,370 sequenced cells. Dashed line: quality control threshold, cells with < 50,000 reads and < 500 genes were excluded from further analysis. e) Genes expressed in more than 3 cells (dashed line) were included for subsequent analysis. f) Cell-type specific gene expression overlay on UMAPs. Genes indicated in upper left corner of each plot. g-i) UMAPs with donor age overlay for g) adult, h) neonatal, and i) fetal cells. j) Heatmap of expression values (row z-score) for all cell types in our dataset of differentially expressed genes (DEG)s from melanocyte clusters identified in previous fresh from human skin sequencing studies10,11.

Source data

Extended Data Fig. 2 Characterization of cell cycle state.

a) UMAPs of cycling cell program score used to determine which cells were designated as b) cycling (blue, in G2 & M phase) vs non-cycling (red). c) Fraction of cycling and non-cycling cells for each cell type identified in Fig. 1b.

Source data

Extended Data Fig. 3 FACS BSC is a correlate of relative melanocyte pigmentation.

a) Representative FACS plots of BSC and FSC for melanocytes from three non-volar cutaneous skin donors of varying skin pigmentation levels: light (L), light-medium (LM), and medium (M). b) Increase in BSC corresponds to increase in pigmentation. Mean raw BSC value with corresponding histogram for melanocytes isolated from non-volar cutaneous skin donors.

Source data

Extended Data Fig. 4 NTRK2 and HPGD expression (summarized in Fig. 3d) by anatomic site and donor age.

Expression level of v-mel gene, NTRK2, and c-mel gene, HPGD, in volar melanocytes compared to non-volar cutaneous melanocytes at each age (n=22 donors). Interquartile range with median, standard deviation, and outliers (grey circles).

Source data

Extended Data Fig. 5 Pseudotime and pairwise differential expression analysis of developmental ages and groups.

Pseudotime and pairwise differential expression analysis of developmental ages and groups. a) Melanocytes cluster by developmental age in diffusion component space DC1 and DC2. b) Pseudotime overlay onto DC space. c) Progression from fetal to adult through an intermediate neonatal transcriptional state. Diffusion pseudotime from b) is plotted for each cell, binned by donor age. d) Volcano plot highlighting the top ten DEGs between MSC (yellow) and FET (teal) non-volar cutaneous melanocyte populations. e) Volcano plot showing the top ten DEGs between FET (teal) and ADT (magenta) non-volar cutaneous melanocytes. (d-e) DEGs determined by Two-sided Wilcoxon Rank Sum Test and adjusted p-value computed using Benjamini-Hochberg multiple testing procedure. f) Heatmap visualization of the relative expression (column z score) of DEGs from (d) and (e) for all four non-volar cutaneous developmental groups. Both MSC and FET were enriched for known developmental genes (SOX11, LYPD1) and genes involved in extracellular matrix establishment/remodeling (COL1A2, PXDN). The ADT group expressed genes involved in innate immunity, inflammation and regulating apoptosis/cell stress in other cell types and tissues (HLAs, APOD, CLU, LGALS3). The NEO group exhibited high expression of a subset of genes from both the FET and ADT stages, consistent with neonatal melanocytes being an intermediate developmental state. See Supplementary Table 4 for the full list.

Source data

Extended Data Fig. 6 Identification of enriched biological processes in MSC, FET, NEO and ADT melanocytes.

Significantly enriched biological processes between temporally adjacent developmental groups a) FET vs MSC; b) NEO vs FET; and c) ADT vs NEO. Each dot represents an individual GO-bp term, plotted according to their associated NES. Dot color correspond to the FDR q-value for each GO-bp term and size corresponds to number of enriched genes from each GO-bp term.

Source data

Extended Data Fig. 7 Biological processes associated with DevMel transcriptional programs.

PercayAI, an augmented artificial intelligence software platform, identified biological concepts (processes/pathways) associated with the positively correlated genes in each DevMel transcriptional program. Two dimensional representation of biological themes (circles) comprised of genes related by associated biological concepts arrange in three dimensional space based on relatedness of each themes for a) prg[MSC]; b) prg[FET]; c) prg[NEO]; and d) prg[ADT]. Highly related themes are connected by grey lines.

Extended Data Fig. 8 Characterization of melanoma cells and tumors classified by in situ human melanocyte developmental programs.

a-b) Density plots showing the expression of a) the Widmer et al. from ref. 61 invasive and proliferative programs and b) the Tirosh et al. from ref. 58 AXL and MITF programs for individual cells in MALADT, MALNEO, MALFET and MALMSC groups. c) Pairwise Fisher (one-sided) exact test showing negative log10 adjusted (Bonferroni multiple testing) p-values for the gene set enrichment analysis conducted using TCGA et al., 2015, Cirenajwis et al., 2015 and Tsoi et al., 2018 gene signatures. Significant enrichment determined as adjusted p-value < 0.05. d) Heatmap showing the relative expression levels (row z score) of WNT5A high, TP53 high slow cycling cell associated genes in each normal melanocyte and MAL developmental group. e) Heatmap showing the relative expression levels (row z score) of the four minimal residual disease states identified by Rambow et al., 2018 in each normal melanocyte and MAL developmental group. f) Pairwise Fisher (one-sided) exact test showing negative log10 adjusted (Bonferroni multiple testing) p-values for clinicopathological feature and transcriptional categorization within each SKCM group (SKCMADT, SKCMNEO, SKCMFET, SKCMMSC). There is little to no difference in the enrichment of pigment level, mutation category, or tissue origin between SKCM groups in Fig. 7. g) Heatmap showing the relative expression levels (row z-score) of immune infiltration program, immune evasion program and FDA-approved therapeutic targets in SKCM groups. h) The MALNEO signature is enriched for genes down regulated in tumors that respond to Nivolumab treatment (green text). Pairwise Fisher (one-sided) exact test showing negative log10 adjusted (Bonferroni multiple testing) p-values for the gene set enrichment analysis conducted using previously identified prognostic signatures (Supplementary Table 8).

Source data

Supplementary information

Reporting Summary

Supplementary Table 1–8

Supplementary Tables 1–8 in a single Excel file.

Peer Review Information

Source data

Source Data Fig. 1

Numerical and statistical source data.

Source Data Fig. 2

Numerical and statistical source data.

Source Data Fig. 3

Numerical and statistical source data.

Source Data Fig. 4

Numerical and statistical source data.

Source Data Fig. 5

Numerical and statistical source data.

Source Data Fig. 6

Numerical and statistical source data.

Source Data Fig. 7

Numerical and statistical source data.

Source Data Extended Data Fig. 1

Numerical and statistical source data.

Source Data Extended Data Fig. 2

Numerical and statistical source data.

Source Data Extended Data Fig. 3

Numerical and statistical source data.

Source Data Extended Data Fig. 4

Numerical and statistical source data.

Source Data Extended Data Fig. 5

Numerical and statistical source data.

Source Data Extended Data Fig. 6

Numerical and statistical source data.

Source Data Extended Data Fig. 8

Numerical and statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Belote, R.L., Le, D., Maynard, A. et al. Human melanocyte development and melanoma dedifferentiation at single-cell resolution. Nat Cell Biol 23, 1035–1047 (2021). https://doi.org/10.1038/s41556-021-00740-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-021-00740-8

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer