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:

Single-cell transcriptomic analyses provide insights into the developmental origins of neuroblastoma

Abstract

Neuroblastoma is a pediatric tumor of the developing sympathetic nervous system. However, the cellular origin of neuroblastoma has yet to be defined. Here we studied the single-cell transcriptomes of neuroblastomas and normal human developing adrenal glands at various stages of embryonic and fetal development. We defined normal differentiation trajectories from Schwann cell precursors over intermediate states to neuroblasts or chromaffin cells and showed that neuroblastomas transcriptionally resemble normal fetal adrenal neuroblasts. Importantly, neuroblastomas with varying clinical phenotypes matched different temporal states along normal neuroblast differentiation trajectories, with the degree of differentiation corresponding to clinical prognosis. Our work highlights the roles of oncogenic MYCN and loss of TFAP2B in blocking differentiation and may provide the basis for designing therapeutic interventions to overcome differentiation blocks.

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

Fig. 1: Cell types in the developing human adrenal gland.
Fig. 2: Differentiation dynamics during adrenal medullary development.
Fig. 3: Projection of neuroblastomas onto developmental trajectories.
Fig. 4: Regulators in neuroblastoma differentiation.
Fig. 5: Neuroblastoma cell type composition deduced from bulk transcriptomes.
Fig. 6: Evaluation of neuroblastoma differentiation therapy.

Similar content being viewed by others

Data availability

Raw adrenal gland and neuroblastoma sequencing data have been deposited in the European Genome-phenome Archive under study accession no. EGAS00001004388. Processed data can be downloaded and explored interactively in a Shiny App: https://adrenal.kitz-heidelberg.de/developmental_programs_NB_viz/. Raw and processed sequencing data for the neuroblastoma cell lines have been deposited in the Gene Expression Omnibus (GEO) under accession no. GSE163431. Bulk RNA-seq data of neuroblastomas from the SEQC cohort were obtained from the GEO (accession no. GSE49711). Bulk RNA-seq data of neuroblastomas from the TARGET cohort were obtained from https://portal.gdc.cancer.gov/projects/TARGET-NBL. The single-cell RNA-seq data of the neuroblastoma cell line SK-N-SH were obtained from the GEO under accession no. GSE158130. Expression data of IMR-32 cells on inducible overexpression of TFAP2B were obtained from the GEO under accession no. GSE74350. Whole-genome sequencing data for tumors studied by scRNA-seq were obtained from the European Genome-phenome Archive (accession nos. EGAS00001004349 and EGAS00001001308). Matching tumor IDs are provided in Supplementary Table 8. Source data are provided with this paper.

References

  1. Maris, J. M., Hogarty, M. D., Bagatell, R. & Cohn, S. L. Neuroblastoma. Lancet 369, 2106–2120 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Peifer, M. et al. Telomerase activation by genomic rearrangements in high-risk neuroblastoma. Nature 526, 700–704 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Brodeur, G. M., Seeger, R. C., Schwab, M., Varmus, H. E. & Bishop, J. M. Amplification of N-myc in untreated human neuroblastomas correlates with advanced disease stage. Science 224, 1121–1124 (1984).

    Article  CAS  PubMed  Google Scholar 

  4. Ackermann, S. et al. A mechanistic classification of clinical phenotypes in neuroblastoma. Science 362, 1165–1170 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Mossé, Y. P. et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature 455, 930–935 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Janoueix-Lerosey, I. et al. Somatic and germline activating mutations of the ALK kinase receptor in neuroblastoma. Nature 455, 967–970 (2008).

    Article  CAS  PubMed  Google Scholar 

  7. van Groningen, T. et al. Neuroblastoma is composed of two super-enhancer-associated differentiation states. Nat. Genet. 49, 1261–1266 (2017).

    Article  CAS  PubMed  Google Scholar 

  8. Boeva, V. et al. Heterogeneity of neuroblastoma cell identity defined by transcriptional circuitries. Nat. Genet. 49, 1408–1413 (2017).

    Article  CAS  PubMed  Google Scholar 

  9. Anderson, D. J., Carnahan, J. F., Michelsohn, A. & Patterson, P. H. Antibody markers identify a common progenitor to sympathetic neurons and chromaffin cells in vivo and reveal the timing of commitment to neuronal differentiation in the sympathoadrenal lineage. J. Neurosci. 11, 3507–3519 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. De Preter, K. et al. Human fetal neuroblast and neuroblastoma transcriptome analysis confirms neuroblast origin and highlights neuroblastoma candidate genes. Genome Biol. 7, R84 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Furlan, A. et al. Multipotent peripheral glial cells generate neuroendocrine cells of the adrenal medulla. Science 357, eaal3753 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Cooper, M. J., Hutchins, G. M. & Israel, M. A. Histogenesis of the human adrenal medulla. An evaluation of the ontogeny of chromaffin and nonchromaffin lineages. Am. J. Pathol. 137, 605–615 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Molenaar, W. M., Lee, V. M. & Trojanowski, J. Q. Early fetal acquisition of the chromaffin and neuronal immunophenotype by human adrenal medullary cells. An immunohistological study using monoclonal antibodies to chromogranin A, synaptophysin, tyrosine hydroxylase, and neuronal cytoskeletal proteins. Exp. Neurol. 108, 1–9 (1990).

    Article  CAS  PubMed  Google Scholar 

  14. Magro, G. & Grasso, S. Immunohistochemical identification and comparison of glial cell lineage in foetal, neonatal, adult and neoplastic human adrenal medulla. Histochem. J. 29, 293–299 (1997).

    Article  CAS  PubMed  Google Scholar 

  15. Katsetos, C. D. et al. Class III β-tubulin isotype (β III) in the adrenal medulla: I. Localization in the developing human adrenal medulla. Anat. Rec. 250, 335–343 (1998).

    Article  CAS  PubMed  Google Scholar 

  16. Lake, B. B. et al. Integrative single-cell analysis of transcriptional and epigenetic states in the human adult brain. Nat. Biotechnol. 36, 70–80 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. La Manno, G. et al. Molecular diversity of midbrain development in mouse, human, and stem cells. Cell 167, 566–580.e19 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Zhong, S. et al. Decoding the development of the human hippocampus. Nature 577, 531–536 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. Gartlgruber, M. et al. Super enhancers define regulatory subtypes and cell identity in neuroblastoma. Nat. Cancer 2, 114–128 (2021).

    Article  PubMed  Google Scholar 

  20. Zimmerman, M. W. et al. MYC drives a subset of high-risk pediatric neuroblastomas and is activated through mechanisms including enhancer hijacking and focal enhancer amplification. Cancer Discov. 8, 320–335 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Ikram, F. et al. Transcription factor activating protein 2 beta (TFAP2B) mediates noradrenergic neuronal differentiation in neuroblastoma. Mol. Oncol. 10, 344–359 (2016).

    Article  CAS  PubMed  Google Scholar 

  22. Zhang, W. et al. Comparison of RNA-seq and microarray-based models for clinical endpoint prediction. Genome Biol. 16, 133 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ambros, I. M. et al. Role of ploidy, chromosome 1p, and Schwann cells in the maturation of neuroblastoma. N. Engl. J. Med. 334, 1505–1511 (1996).

    Article  CAS  PubMed  Google Scholar 

  24. Wei, J. S. et al. Clinically relevant cytotoxic immune cell signatures and clonal expansion of T-cell receptors in high-risk MYCN-not-amplified human neuroblastoma. Clin. Cancer Res. 24, 5673–5684 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Sidell, N. Retinoic acid-induced growth inhibition and morphologic differentiation of human neuroblastoma cells in vitro. J. Natl Cancer Inst. 68, 589–596 (1982).

    CAS  PubMed  Google Scholar 

  26. Matthay, K. K. et al. Long-term results for children with high-risk neuroblastoma treated on a randomized trial of myeloablative therapy followed by 13-cis-retinoic acid: a children’s oncology group study. J. Clin. Oncol. 27, 1007–1013 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Huber, K. The sympathoadrenal cell lineage: specification, diversification, and new perspectives. Dev. Biol. 298, 335–343 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Dong, R. et al. Single-cell characterization of malignant phenotypes and developmental trajectories of adrenal neuroblastoma. Cancer Cell 38, 716–733.e6 (2020).

    Article  CAS  PubMed  Google Scholar 

  29. Huber, K. Segregation of neuronal and neuroendocrine differentiation in the sympathoadrenal lineage. Cell Tissue Res. 359, 333–341 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Lumb, R. & Schwarz, Q. Sympathoadrenal neural crest cells: the known, unknown and forgotten? Dev. Growth Differ. 57, 146–157 (2015).

    Article  PubMed  Google Scholar 

  31. Janoueix-Lerosey, I., Lopez-Delisle, L., Delattre, O. & Rohrer, H. The ALK receptor in sympathetic neuron development and neuroblastoma. Cell Tissue Res. 372, 325–337 (2018).

    Article  CAS  PubMed  Google Scholar 

  32. Unsicker, K., Huber, K., Schober, A. & Kalcheim, C. Resolved and open issues in chromaffin cell development. Mech. Dev. 130, 324–329 (2013).

    Article  CAS  PubMed  Google Scholar 

  33. Ernst, K. J. Establishment of a simplified preparation method for single-nucleus RNA-sequencing and its application to long-term frozen tumor tissues. Preprint at bioRxiv https://doi.org/10.1101/2020.10.23.351809 (2020).

  34. Jaitin, D. A. et al. Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types. Science 343, 776–779 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Tirosh, I. et al. Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma. Nature 539, 309–313 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Angerer, P. et al. destiny: diffusion maps for large-scale single-cell data in R. Bioinformatics 32, 1241–1243 (2016).

    Article  CAS  PubMed  Google Scholar 

  38. Street, K. et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genomics 19, 477 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Qiu, X. et al. Reversed graph embedding resolves complex single-cell trajectories. Nat. Methods 14, 979–982 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bergen, V., Lange, M., Peidli, S., Wolf, F. A. & Theis, F. J. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat. Biotechnol. 38, 1408–1414 (2020).

    Article  PubMed  Google Scholar 

  44. Teschendorff, A. E. & Enver, T. Single-cell entropy for accurate estimation of differentiation potency from a cell’s transcriptome. Nat. Commun. 8, 15599 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Gulati, G. S. et al. Single-cell transcriptional diversity is a hallmark of developmental potential. Science 367, 405–411 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Patel, A. P. et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 344, 1396–1401 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Aran, D. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 20, 163–172 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Baron, M. et al. A single-cell transcriptomic map of the human and mouse pancreas reveals inter- and intra-cell population structure. Cell Syst. 3, 346–360.e4 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. 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 

  51. Muth, D. et al. Transcriptional repression of SKP2 is impaired in MYCN-amplified neuroblastoma. Cancer Res. 70, 3791–3802 (2010).

    Article  CAS  PubMed  Google Scholar 

  52. Henrich, K.-O. et al. Integrative genome-scale analysis identifies epigenetic mechanisms of transcriptional deregulation in unfavorable neuroblastomas. Cancer Res. 76, 5523–5537 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  54. Tarasov, A., Vilella, A. J., Cuppen, E., Nijman, I. J. & Prins, P. Sambamba: fast processing of NGS alignment formats. Bioinformatics 31, 2032–2034 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. DeLuca, D. S. et al. RNA-SeQC: RNA-seq metrics for quality control and process optimization. Bioinformatics 28, 1530–1532 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  57. McCarthy, D. J., Chen, Y. & Smyth, G. K. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 40, 4288–4297 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  59. Robinson, M. D. & Oshlack, A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 11, R25 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Pihlajoki, M., Dörner, J., Cochran, R. S., Heikinheimo, M. & Wilson, D. B. Adrenocortical zonation, renewal, and remodeling. Front. Endocrinol. (Lausanne) 6, 27 (2015).

    Article  Google Scholar 

  61. Goncharov, N. V., Nadeev, A. D., Jenkins, R. O. & Avdonin, P. V. Markers and biomarkers of endothelium: when something is rotten in the state. Oxid. Med. Cell. Longev. 2017, 9759735 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Xie, T. et al. Single-cell deconvolution of fibroblast heterogeneity in mouse pulmonary fibrosis. Cell Rep. 22, 3625–3640 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Foote, A. G., Wang, Z., Kendziorski, C. & Thibeault, S. L. Tissue specific human fibroblast differential expression based on RNAsequencing analysis. BMC Genomics 20, 308 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Donovan, J. A. & Koretzky, G. A. CD45 and the immune response. J. Am. Soc. Nephrol. 4, 976–985 (1993).

    Article  CAS  PubMed  Google Scholar 

  65. Irving, B. A., Chan, A. C. & Weiss, A. Functional characterization of a signal transducing motif present in the T cell antigen receptor zeta chain. J. Exp. Med. 177, 1093–1103 (1993).

    Article  CAS  PubMed  Google Scholar 

  66. Kadowaki, T. et al. Reconsideration of macrophage and dendritic cell classification. Anticancer Res. 32, 2257–2261 (2012).

    CAS  PubMed  Google Scholar 

  67. Affer, M. et al. Gene expression differences between enriched normal and chronic myelogenous leukemia quiescent stem/progenitor cells and correlations with biological abnormalities. J. Oncol. 2011, 798592 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Castiglioni, I. et al. The Trithorax protein Ash1L promotes myoblast fusion by activating Cdon expression. Nat. Commun. 9, 5026 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Chal, J. & Pourquié, O. Making muscle: skeletal myogenesis in vivo and in vitro. Development 144, 2104–2122 (2017).

    Article  CAS  PubMed  Google Scholar 

  70. Robin, Y.-M. et al. Transgelin is a novel marker of smooth muscle differentiation that improves diagnostic accuracy of leiomyosarcomas: a comparative immunohistochemical reappraisal of myogenic markers in 900 soft tissue tumors. Mod. Pathol. 26, 502–510 (2013).

    Article  CAS  PubMed  Google Scholar 

  71. Hinz, B. et al. The myofibroblast: one function, multiple origins. Am. J. Pathol. 170, 1807–1816 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Hsia, L.-T. et al. Myofibroblasts are distinguished from activated skin fibroblasts by the expression of AOC3 and other associated markers. Proc. Natl Acad. Sci. USA 113, E2162–E2171 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Peters, D. T. et al. Asialoglycoprotein receptor 1 is a specific cell-surface marker for isolating hepatocytes derived from human pluripotent stem cells. Development 143, 1475–1481 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Aizarani, N. et al. A human liver cell atlas reveals heterogeneity and epithelial progenitors. Nature 572, 199–204 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Kim, H.-S. et al. Schwann cell precursors from human pluripotent stem cells as a potential therapeutic target for myelin repair. Stem Cell Rep. 8, 1714–1726 (2017).

    Article  CAS  Google Scholar 

  76. Liu, Z. et al. Specific marker expression and cell state of Schwann cells during culture in vitro. PLoS ONE 10, e0123278 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  77. Chan, W. H., Anderson, C. R. & Gonsalvez, D. G. From proliferation to target innervation: signaling molecules that direct sympathetic nervous system development. Cell Tissue Res. 372, 171–193 (2018).

    Article  CAS  PubMed  Google Scholar 

  78. Rohrer, H. Transcriptional control of differentiation and neurogenesis in autonomic ganglia. Eur. J. Neurosci. 34, 1563–1573 (2011).

    Article  PubMed  Google Scholar 

  79. Chan, W. H. et al. RNA-seq of isolated chromaffin cells highlights the role of sex-linked and imprinted genes in adrenal medulla development. Sci. Rep. 9, 3929 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the patients and their parents for making available the tumor specimens analyzed in this study. We also thank the German Neuroblastoma Biobank for providing these samples. The human embryonic and fetal materials were provided by the joint MRC/Wellcome Trust (grant no. MR/R006237/1) Human Developmental Biology Resource (www.hdbr.org). The institutional review board approved the collection and use of all specimens in this study. We thank J.-P. Mallm and the Single-cell Open Lab, Genomics and Proteomics Core Facility and Light Microscopy Facility at the German Cancer Research Center. The results published in this article are in part based on data generated by the Therapeutically Applicable Research to Generate Effective Treatments (https://ocg.cancer.gov/programs/target) initiative (phs000218). The data used for this analysis are available at https://portal.gdc.cancer.gov/projects. This work was supported by the Fördergesellschaft Kinderkrebs-Neuroblastom-Forschung e.V. (F.W. and S.J.), German-Israeli Helmholtz Research School in Cancer Biology (S.J.), German Ministry of Science and Education (e:Med initiative, grant no. 01ZX1307 to F.W.; grant no. 031L0238 to F.W. and T.H.), EU as part of the EraCoSysMed initiative (Optimize-NB and Infer-NB to F.W. and T. H.), German Cancer Research Center intramural program for interaction projects (Single-cell Open Lab) and DKFZ-Heidelberg Center for Personalized Oncology (HIPO) and National Center for Tumor Disease (NCT 3.0—ENHANCE to F.W.). The laboratory of T.G.P.G. is supported by the Barbara & Wilfried Mohr Foundation.

Author information

Authors and Affiliations

Authors

Contributions

S.J. and F.W. designed and coordinated the experiments and data analyses. S.J. performed most of the experiments and data analyses. A.K.S. analyzed the transcription factor activity in the snRNA-seq data. V.K. analyzed the copy number variation in the snRNA-seq data. U.H.T. contributed to the preprocessing of the snRNA-seq and bulk RNA-seq data. E.M.W. performed the RNAscope and western blot experiments. A.G., A.Q., E.C., C.H., A.T. and T.H. contributed to the single-cell data analysis. M.G. performed the ATRA treatment. T.G.P.G. coordinated the immunohistochemical staining. S.J. and F.W. wrote the manuscript with input from K.-O.H. and T.H. F.W. supervised the project.

Corresponding author

Correspondence to Frank Westermann.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Genetics thanks Guoji Guo and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Adrenal gland histology and cell types.

a, b, H&E and immunohistochemical staining of adrenal gland serial sections for CHGA, ALK, ISL1 and KI67. Left panels show overviews over entire glands. Boxes mark regions of insets on the right. Scale bars: left: 1000/500 µm, inset: 200 µm. Sections are derived from 17 pcw (AG07) (a) and 14 pcw (AG08) (b) adrenal glands. Shown are representative results for two out of four samples analyzed in independent experiments.

Extended Data Fig. 2 Adrenal gland cluster, age and sample contribution.

a, Uniform Manifold Approximation and Projection (UMAP) embedding of single cells derived from developing human adrenal glands colored by 45 unique clusters of transcriptionally similar cells. Inset shows the cells colored by cell types as in Fig. 1b. b, c, UMAP embedding from (a) colored by time point of sample collection (b) and individual samples (c). d, Heat map depicting relative expression of marker genes for adrenal gland cell populations as in Fig. 1b. Normalized expression is shown as z-score. e, f, UMAP plot of adrenal medullary cells colored by 16 clusters of transcriptionally similar cells (d) and individual samples (e). Inset shows the cells colored by cell types as in Fig. 1c. g, UMAP visualizations showing adrenal medullary cells from each individual time point integrated using harmony. h, Barplot with fractions of cells in each cell cycle phase in adrenal medullary cell types. i, RNAscope fluorescent RNA in situ hybridization for bridge marker ASCL1, neuroblast marker ISL1 and chromaffin marker TH in human adrenal glands at 8 pcw and 14 pcw. Left panels show composite overviews over entire glands, boxes mark the regions of insets shown on the right. Scale bars: left: 800 µm, inset: 20 µm. Shown are representative results for two out of four samples analyzed in independent experiments.

Extended Data Fig. 3 Adrenal medulla potency and pseudotime.

a, Diffusion map of adrenal medullary cells as shown in Fig. 2a, colored by 19 clusters of transcriptionally similar cells. Start of the pseudotime trajectory is marked with an arrow (cluster 16). b, c, UMAP embedding of adrenal medullary cells without cycling populations colored by 19 clusters of transcriptionally similar cells (b) or cell types (c). Start of the pseudotime trajectory is marked with an arrow (cluster 16). d, f, UMAP plot showing adrenal medullary cells without cycling populations colored by differentiation potential as determined by CytoTRACE (d) and differentiation potency as determined by LandSCENT (f). e, Boxplot with differentiation potencies for adrenal medullary cell populations as determined by LandSCENT. Here, the center line represents the median, upper and lower boundaries the 75th and 25th percentile and whiskers the 1.5x interquartile range. Number of cells: late SCPs: n = 632, SCPs: n = 817, cycling SCPs: n = 288, Bridge: n = 612, con. Prog: n = 1499, Chrom: n = 1346, late Chrom: n = 963, cycling Nbs: n = 1064, Nbs: n = 1388, late Nbs: n = 2130. g, Diffusion map of adrenal medullary cells colored by SCP-to-late SCP pseudotime trajectory as determined by slingshot. h, UMAP visualization illustrating adrenal medullary cells without cycling populations colored by pseudotime as determined by monocle3. Start of the pseudotime trajectory is marked with an arrow. i, Gene expression of selected genes over pseudotime in chromaffin cell and neuroblast trajectories.

Extended Data Fig. 4 Neuroblastoma cluster, sample and cell type contribution.

a, UMAP visualization illustrating 64.769 single neuroblastoma cells integrated using harmony and colored by cell types. b, Heat map showing expression of canonical cell type markers in cells derived from neuroblastoma tumors. Color indicates relative expression as z-score. c, UMAP embedding showing single neuroblastoma cells integrated using harmony and colored by inferred copy number status. d, Barplot with annotated cell type fractions for each neuroblastoma tumor. e, Boxplots showing expression of published adrenergic and mesenchymal neuroblastoma signatures in single neuroblastoma cells grouped by subgroup. Here, the center line represents the median, upper and lower boundaries the 75th and 25th percentile and whiskers the 1.5x interquartile range. Number of cells: LR: n = 21,086, ALT/TERT: n = 12,719, MES: n = 8980, MYCN: n = 16,527. f, g, UMAP plot showing neuroblastoma cells integrated using harmony and colored by clusters of transcriptionally similar cells (f) and sample (g).

Extended Data Fig. 5 Neuroblastoma copy number variations.

a–n Inferred copy number profiles of neuroblastoma cells using InferCNV with normal neuroblasts as reference. Below: Copy number profiles of the same tumors derived from whole genome sequencing data.

Extended Data Fig. 6 Similarity of single neuroblastoma cells to normal developing adrenal medullary cell populations.

a–l, Heat maps with similarity scores of single neuroblastoma cells and fetal adrenal medullary cell populations for neuroblastoma tumors not shown in Fig. 3. m, Bar plots illustrating proportion of neuroblastoma cells assigned to adrenal medullary cells or cells from published fetal and adult brain data sets by neuroblastoma subgroup. Cells are assigned to the normal population with highest similarity score.

Extended Data Fig. 7 Similarity of single cells from neuroblastoma cell lines to normal developing adrenal medullary cell populations.

a–c, UMAP visualization of single SK-N-SH cells colored by adrenergic, mesenchymal and intermediate population (a), published mesenchymal neuroblastoma signature expression (b) or adrenergic neuroblastoma signature expression (c). d, Heat map with similarity scores of single SK-N-SH cells to fetal adrenal medullary cell populations. Top bar indicates SK-N-SH subpopulation identity. e, Bar plots illustrating proportion of SK-N-SH cells assigned to each normal adrenal medullary cell population. Cells are assigned to the normal population with highest similarity score. f-h, UMAP embedding of single SK-N-AS cells colored by clusters of transcriptionally similary cells (f), published mesenchymal neuroblastoma signature expression (g) or adrenergic neuroblastoma signature expression (h). i, Heat map with similarity scores of single SK-N-AS cells to fetal adrenal medullary cell populations. j, Bar plots illustrating proportion of SK-N-AS cells assigned to each normal adrenal medullary cell population. Cells are assigned to the normal population with highest similarity score.

Extended Data Fig. 8 Projection of single neuroblastoma cells onto adrenal medullary cell populations.

a–l, Diffusion maps of adrenal medullary cells as shown in Fig. 2a with single neuroblastoma cells projected onto the embedding for tumors not shown in Fig. 3.

Extended Data Fig. 9 Regulation of neuroblastoma by markers of normal adrenal medullary development.

a, Dot plot showing expression of genes with strong enhancers described to engage in enhancer hijacking events in neuroblastoma, in adrenal medullary cell populations. b, Violin plots with normalized expression of adrenal medullary developmental genes in neuroblastoma subgroups. c, Heat map illustrating activity of adrenal medullary transcription factors in neuroblastoma shown as fraction of cells in which the regulon was found active by subgroup. d, UMAP embedding showing single MYCN-amplified IMR5/75 neuroblastoma cells with tetracyclin-inducible shRNAs targeting MYCN and generating MYCN high (-Tet) and MYCN low (+Tet) cells. Cells are colored by levels of regulatable MYCN. e, UMAP visualization of MYCN low and MYCN high cells colored by cell cycle phase. f, Barplot with fractions of cells in each cell cycle phase in MYCN high and MYCN low cells. g, MYCN protein levels in MYCN high and MYCN low IMR5/75 neuroblastoma cells. Detection by western blot using vinculin as loading control. Analysis was performed once on cells from the same experiment as used for single-cell sequencing. h, Violin plots show expression of signatures for Bridge (p < 2.22 × 10−16), con. Progenitor cells (p < 2.22 × 10−16), Chromaffin cells (p = 0.034), late Chromaffin cells (p < 2.22 × 10−16), SCPs (p < 2.22 × 10−16) and late SCPs (p < 2.22 × 10−16) in MYCN-amplified (MYCN high) neuroblastoma cells and MYCN-amplified cells upon MYCN knockdown (MYCN low). Statistical significance was determined by two-sided Wilcoxon rank-sum test.

Extended Data Fig. 10 Association of neuroblastoma cell type composition with clinical parameters.

a, Hierarchical clustering of patient samples based on abundance of fetal adrenal medullary cell populations without cycling populations determined by deconvolution with BSEQ-sc (SEQC cohort). b, Composition of neuroblastoma tumors by subgroup based on deconvolution of bulk RNA-seq data with fetal adrenal cell populations without cycling populations using BSEQ-sc (SEQC cohort). c, Hierarchical clustering of patient samples based on abundance of fetal adrenal medullary cell populations determined by BSEQ-sc (TARGET cohort). d-i, Kaplan-Meier analysis of event-free (d, f, h) or overall (e, g, i) survival in neuroblastoma patients according to the relative abundance of late neuroblasts (d–g), neuroblasts (h) or cycling neuroblasts (i) determined by deconvolution of tumors with fetal adrenal cell populations. Panels f and g show deconvolution results for the TARGET cohort, panels e and i show deconvolution results for the SEQC cohort and panels d and h show deconvolution results for the SEQC cohort when excluding cycling cells from the analysis. P-values were calculated using the two-sided log-rank test.

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1–11.

Source data

Source Data Fig. 1

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jansky, S., Sharma, A.K., Körber, V. et al. Single-cell transcriptomic analyses provide insights into the developmental origins of neuroblastoma. Nat Genet 53, 683–693 (2021). https://doi.org/10.1038/s41588-021-00806-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-021-00806-1

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