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 analysis and functional characterization uncover the stem cell hierarchies and developmental origins of rhabdomyosarcoma

Abstract

Rhabdomyosarcoma (RMS) is a common childhood cancer that shares features with developing skeletal muscle. Yet, the conservation of cellular hierarchy with human muscle development and the identification of molecularly defined tumor-propagating cells has not been reported. Using single-cell RNA-sequencing, DNA-barcode cell fate mapping and functional stem cell assays, we uncovered shared tumor cell hierarchies in RMS and human muscle development. We also identified common developmental stages at which tumor cells become arrested. Fusion-negative RMS cells resemble early myogenic cells found in embryonic and fetal development, while fusion-positive RMS cells express a highly specific gene program found in muscle cells transiting from embryonic to fetal development at 7–7.75 weeks of age. Fusion-positive RMS cells also have neural pathway-enriched states, suggesting less-rigid adherence to muscle-lineage hierarchies. Finally, we identified a molecularly defined tumor-propagating subpopulation in fusion-negative RMS that shares remarkable similarity to bi-potent, muscle mesenchyme progenitors that can make both muscle and osteogenic cells.

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: scRNA-seq reveals distinct cell states and intertumoral heterogeneity in human RMS.
Fig. 2: Single RMS cells can remake all tumor cell heterogeneity within the cancer.
Fig. 3: LARRY barcoding of human FN-RMS RD cells reveals that the mesenchymal-enriched cell subfraction is capable of driving tumor growth following culture in low-serum, stress conditions.
Fig. 4: Tumor-propagating potential is found within the mesenchymal-enriched cell fraction of FN-RMS.
Fig. 5: Limiting dilution cell transplantation confirms that the FN-RMS mesenchymal-enriched subfraction has tumor-propagating potential in vivo.
Fig. 6: RMS subtypes share common gene expression patterns and are arrested at distinct stages of fetal and embryonic muscle development.
Fig. 7: Mesenchymal-enriched FN-RMS TPCs share transcriptional and functional similarities with the bi-potent SkM. Mesen.

Similar content being viewed by others

Data availability

scRNA-seq and snRNA-seq data are available at the Gene Expression Omnibus (GEO) under accession GSE195709. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

Code availability

All of the analysis scripts have been deposited at GitHub and can be accessed using the links: https://github.com/qinqian/sc_normal_muscle, and https://github.com/qinqian/rms_analysis.

References

  1. Reya, T., Morrison, S. J., Clarke, M. F. & Weissman, I. L. Stem cells, cancer, and cancer stem cells. Nature 414, 105–111 (2001).

    Article  CAS  PubMed  Google Scholar 

  2. Miyamoto, T., Weissman, I. L. & Akashi, K. AML1/ETO-expressing nonleukemic stem cells in acute myelogenous leukemia with 8;21 chromosomal translocation. Proc. Natl Acad. Sci. USA 97, 7521–7526 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ginestier, C. et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 1, 555–567 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Merlos-Suarez, A. et al. The intestinal stem cell signature identifies colorectal cancer stem cells and predicts disease relapse. Cell Stem Cell 8, 511–524 (2011).

    Article  CAS  PubMed  Google Scholar 

  5. Quintana, E. et al. Efficient tumour formation by single human melanoma cells. Nature 456, 593–598 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kenney, A. M., Cole, M. D. & Rowitch, D. H. Nmyc upregulation by sonic hedgehog signaling promotes proliferation in developing cerebellar granule neuron precursors. Development 130, 15–28 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Gilbertson, R. J. & Ellison, D. W. The origins of medulloblastoma subtypes. Annu. Rev. Pathol. 3, 341–365 (2008).

    Article  CAS  PubMed  Google Scholar 

  8. Wong, D. J. et al. Module map of stem cell genes guides creation of epithelial cancer stem cells. Cell Stem Cell 2, 333–344 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Parham, D. M. & Barr, F. G. Classification of rhabdomyosarcoma and its molecular basis. Adv. Anat. Pathol. 20, 387–397 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Horn, R. C. Jr. & Enterline, H. T. Rhabdomyosarcoma: a clinicopathological study and classification of 39 cases. Cancer 11, 181–199 (1958).

    Article  PubMed  Google Scholar 

  11. Yohe, M. E. et al. Insights into pediatric rhabdomyosarcoma research: challenges and goals. Pediatr. Blood Cancer 66, e27869 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Langenau, D. M. et al. Effects of RAS on the genesis of embryonal rhabdomyosarcoma. Genes Dev. 21, 1382–1395 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Chen, X. et al. Targeting oxidative stress in embryonal rhabdomyosarcoma. Cancer Cell 24, 710–724 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Shern, J. F. et al. Comprehensive genomic analysis of rhabdomyosarcoma reveals a landscape of alterations affecting a common genetic axis in fusion-positive and fusion-negative tumors. Cancer Discov. 4, 216–231 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Linardic, C. M., Downie, D. L., Qualman, S., Bentley, R. C. & Counter, C. M. Genetic modeling of human rhabdomyosarcoma. Cancer Res. 65, 4490–4495 (2005).

    Article  CAS  PubMed  Google Scholar 

  16. Hibbitts, E. et al. Refinement of risk stratification for childhood rhabdomyosarcoma using FOXO1 fusion status in addition to established clinical outcome predictors: a report from the Children’s Oncology Group. Cancer Med. 8, 6437–6448 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Agaram, N. P. et al. MYOD1-mutant spindle cell and sclerosing rhabdomyosarcoma: an aggressive subtype irrespective of age. A reappraisal for molecular classification and risk stratification. Mod. Pathol. 32, 27–36 (2019).

    Article  CAS  PubMed  Google Scholar 

  18. Sorensen, P. H. et al. PAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicators in alveolar rhabdomyosarcoma: a report from the children’s oncology group. J. Clin. Oncol. 20, 2672–2679 (2002).

    Article  CAS  PubMed  Google Scholar 

  19. Heske, C. M. et al. Survival outcomes of patients with localized FOXO1 fusion-positive rhabdomyosarcoma treated on recent clinical trials: a report from the Soft Tissue Sarcoma Committee of the Children’s Oncology Group. Cancer 127, 946–956 (2021).

    Article  CAS  PubMed  Google Scholar 

  20. Shern, J. F. et al. Genomic classification and clinical outcome in rhabdomyosarcoma: a report from an international consortium. J. Clin. Oncol. 39, 2859–2871 (2021).

    Article  CAS  PubMed  Google Scholar 

  21. Sebire, N. J. & Malone, M. Myogenin and MyoD1 expression in paediatric rhabdomyosarcomas. J. Clin. Pathol. 56, 412–416 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tenente, I. M. et al. Myogenic regulatory transcription factors regulate growth in rhabdomyosarcoma. eLife https://doi.org/10.7554/eLife.19214 (2017).

  23. Rubin, B. P. et al. Evidence for an unanticipated relationship between undifferentiated pleomorphic sarcoma and embryonal rhabdomyosarcoma. Cancer Cell 19, 177–191 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ignatius, M. S. et al. In vivo imaging of tumor-propagating cells, regional tumor heterogeneity, and dynamic cell movements in embryonal rhabdomyosarcoma. Cancer Cell 21, 680–693 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hettmer, S. et al. Sarcomas induced in discrete subsets of prospectively isolated skeletal muscle cells. Proc. Natl Acad. Sci. USA 108, 20002–20007 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Preussner, J. et al. Oncogenic amplification of zygotic Dux factors in regenerating p53-deficient muscle stem cells defines a molecular cancer subtype. Cell Stem Cell 23, 794–805.e4 (2018).

    Article  CAS  PubMed  Google Scholar 

  27. Drummond, C. J. et al. Hedgehog pathway drives fusion-negative rhabdomyosarcoma initiated from non-myogenic endothelial progenitors. Cancer Cell 33, 108–124.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Xi, H. et al. A human skeletal muscle atlas identifies the trajectories of stem and progenitor cells across development and from human pluripotent stem cells. Cell Stem Cell 27, 181–185 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhang, A. W. et al. Probabilistic cell-type assignment of single-cell RNA-seq for tumor microenvironment profiling. Nat. Methods 16, 1007–1015 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Becht, E. et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat. Biotechnol. https://doi.org/10.1038/nbt.4314 (2018).

  31. 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 

  32. Weinreb, C., Rodriguez-Fraticelli, A., Camargo, F. D. & Klein, A. M. Lineage tracing on transcriptional landscapes links state to fate during differentiation. Science https://doi.org/10.1126/science.aaw3381 (2020).

  33. Chen, E. Y. et al. Glycogen synthase kinase 3 inhibitors induce the canonical WNT/β-catenin pathway to suppress growth and self-renewal in embryonal rhabdomyosarcoma. Proc. Natl Acad. Sci. USA 111, 5349–5354 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Davicioni, E. et al. Molecular classification of rhabdomyosarcoma—genotypic and phenotypic determinants of diagnosis: a report from the Children’s Oncology Group. Am. J. Pathol. 174, 550–564 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Gryder, B. E. et al. PAX3-FOXO1 establishes myogenic super enhancers and confers BET bromodomain vulnerability. Cancer Discov. 7, 884–899 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Qin, Q. et al. Lisa: inferring transcriptional regulators through integrative modeling of public chromatin accessibility and ChIP-seq data. Genome Biol. 21, 32 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Walter, D. et al. CD133 positive embryonal rhabdomyosarcoma stem-like cell population is enriched in rhabdospheres. PLoS ONE 6, e19506 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Patel, A. G. et al. The myogenesis program drives clonal selection and drug resistance in rhabdomyosarcoma. Dev. Cell. 57, 1226–1240 (2022).

  39. Yan, C. et al. Single-cell imaging of T cell immunotherapy responses in vivo. J. Exp. Med. https://doi.org/10.1084/jem.20210314 (2021).

  40. Fletcher, C. D. M. et al. WHO Classification of Tumours of Soft Tissue and Bone (IARC Press, 2013).

  41. Gonzalez Curto, G. et al. The PAX-FOXO1s trigger fast trans-differentiation of chick embryonic neural cells into alveolar rhabdomyosarcoma with tissue invasive properties limited by S phase entry inhibition. PLoS Genet. 16, e1009164 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Khalatbari, M. R., Jalaeikhoo, H., Hamidi, M. & Moharamzad, Y. Primary spinal epidural rhabdomyosarcoma: a case report and review of the literature. Childs Nerv. Syst. 28, 1977–1980 (2012).

    Article  PubMed  Google Scholar 

  43. Chikhalkar, S. et al. Alveolar rhabdomyosarcoma arising in a giant congenital melanocytic nevus in an adult—case report with review of literature. Int. J. Dermatol. 52, 1372–1375 (2013).

    Article  PubMed  Google Scholar 

  44. Fu, N. Y., Nolan, E., Lindeman, G. J. & Visvader, J. E. Stem cells and the differentiation hierarchy in mammary gland development. Physiol. Rev. 100, 489–523 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. Stewart, E. et al. Orthotopic patient-derived xenografts of paediatric solid tumours. Nature 549, 96–100 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hayes, M. N. et al. Vangl2/RhoA signaling pathway regulates stem cell self-renewal programs and growth in rhabdomyosarcoma. Cell Stem Cell 22, 414–427.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Tomayko, M. M. & Reynolds, C. P. Determination of subcutaneous tumor size in athymic (nude) mice. Cancer Chemother. Pharmacol. 24, 148–154 (1989).

    Article  CAS  PubMed  Google Scholar 

  48. Skoda, J. et al. Serial xenotransplantation in NSG mice promotes a hybrid epithelial/mesenchymal gene expression signature and stemness in rhabdomyosarcoma cells. Cancers (Basel) https://doi.org/10.3390/cancers12010196 (2020).

  49. Mehra, S. et al. Detection of FOXO1 (FKHR) gene break-apart by fluorescence in situ hybridization in formalin-fixed, paraffin-embedded alveolar rhabdomyosarcomas and its clinicopathologic correlation. Diagn. Mol. Pathol. 17, 14–20 (2008).

    Article  CAS  PubMed  Google Scholar 

  50. Slyper, M. et al. A single-cell and single-nucleus RNA-seq toolbox for fresh and frozen human tumors. Nat. Med. 26, 792–802 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, 281–291.e289 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Yu, G., Wang, L. G., Han, Y. & He, Q. Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Liberzon, A. et al. Molecular Signatures Database (MSigDB) 3.0. Bioinformatics 27, 1739–1740 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).

    CAS  PubMed  Google Scholar 

  57. Wang, M., Zhao, Y. & Zhang, B. Efficient test and visualization of multi-set intersections. Sci. Rep. 5, 16923 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Robertson, D., Savage, K., Reis-Filho, J. S. & Isacke, C. M. Multiple immunofluorescence labelling of formalin-fixed paraffin-embedded (FFPE) tissue. BMC Cell Biol. 9, 13 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Slemmons, K. K. et al. A method to culture human alveolar rhabdomyosarcoma cell lines as rhabdospheres demonstrates an enrichment in stemness and Notch signaling. Biol. Open https://doi.org/10.1242/bio.050211 (2021).

  60. Hu, Y. & Smyth, G. K. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J. Immunol. Methods 347, 70–78 (2009).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by NIH grants no. R01CA154923 (D.M.L.), no. R01CA215118 (D.M.L.), no. R01CA211734 (D.M.L.), no. U54CA231630 (D.M.L.), no. R00HG008399 (L.P.), no. R35HG010717 (L.P.) and no. R01AR064327 (A.D.P.). Additional funding included the Liddy Shriver Sarcoma Initiative (D.M.L.), the MGH Research Scholars Program (D.M.L.), the Infinite Love for Kids Fighting Cancer Grant (D.M.L., F.D.C), the Rally Foundation (D.M.L.), The Truth 365 (D.M.L.), the Summer’s Way/Friends of TJ Young Investigator Award (Y.W.), the Tosteson & Fund for Medical Discovery Fellowship from MGH (C.Y.), the Alex’s Lemonade Stand Foundation Young Investigator Award (C.Y.), CIRM Quest DISC2-10696 (A.D.P.), UCLA BSCRC (A.D.P.), the Ayoub Centennial Chair (A.D.P.), the Paulie Strong Foundation (F.D.C.), The Grayson Fund (F.D.C.), Willens Family Fund (F.D.C.) and the Pediatric Cancer Foundation (F.D.C.). We thank the MGH Department of Pathology Flow and Image Cytometry Research Core which has been supported by NIH grants no. 1S10OD012027-01A1, no. 1S10OD016372-01, no. 1S10RR020936-01 and no. 1S10RR023440-01A1. We thank M. Dyer and E. Stewart from the Childhood Solid Tumor Network (CSTN) at St. Jude for a subset of PDX models used in this work. We thank A. Friedmann and D. Ebb from MGH Pediatric Hematology/Oncology department. We also thank D.C. O’Neill and L. Millet for helpful and stimulating discussions.

Author information

Authors and Affiliations

Authors

Contributions

Y.W., D.M.L. and M.N.H. designed the project. Q.Q., S.P.G. and L.P. performed computational modeling and data analysis. Y.W., C.Y., D.D., A.H.J., A.A., K.M.M. and T.C.E. performed sequencing, mouse work and sample processing. H.X. and A.D.P. shared single-cell sequencing data of normal muscle development before publication. M.L.O. and A.J.I. validated fusion status by FOXO1-FISH break-apart assay. D.S., G.P.N., L.H.W. and F.D.C. contributed PDX models and patient samples. M.L.S. aided in high-throughput sequencing and computational approaches for single-cell sequencing data.

Corresponding authors

Correspondence to Luca Pinello or David M. Langenau.

Ethics declarations

Competing interests

A.J.I. receives royalties from ArcherDx and consults for Paige.AI, Repare Therapeutics, Oncoclinicas Brasil and Kinnate Biopharma. M.L.S. is an equity holder, scientific co-founder and advisory board member of Immunitas Therapeutics. L.P. has financial interests in Edilytics and SeQure Dx, Inc. All potential competing interests are reviewed and managed by Massachusetts General Hospital and Mass General Brigham HealthCare in accordance with their conflict-of-interest policies. D.M.L. receives sponsored research funds from NextCure for an unrelated project.

Peer review

Peer review information

Nature Cancer thanks Junyue Cao, Max Wicha and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Frozen RMS patient samples have similar cell states as those in PDX models.

a,b, UMAP showing all cells sequenced from representative FN-RMS 20696. Non-tumor cells were assigned using Cellassign and clusterprofiler enricher analysis (a, n = 1,726 cells) and tumor cells analyzed for expression of diagnostic markers for rhabdomyosarcoma (b, n = 1,676 cells). In total, 3,402 cells were analyzed in this experiment. c, UMAP visualization of tumor cells from primary FN-RMS 20696 (n = 1,676 cells). d, Heatmap showing single cells (x axis) and genes enriched for specific transcription modules (y axis, FN-RMS 20696). Cells are arranged by UMAP clusters, combined based on expression similarity, and then assigned a specific cell state as noted. Cell numbers same as panel c. e, UMAP renderings for primary RMS samples juxtaposed with graphical analysis showing detected genes/tumor cells when analyzed across different cell states (n = 1,676 cells for sample 20696; n = 9,675 cells for 29806; n = 11,902 cells for 21202; and n = 6,188 cells for 20082).

Extended Data Fig. 2 Cell states are identified by scRNA sequencing in all RMS PDXs and similar numbers of detected genes per cell are found across cell states.

a, UMAP renderings of all PDXs, with exception of MAST111, MAST139, MAST85-r2 and MSK72117, which were shown in Figs. 1a and 2d. Number of tumor cells analyzed are denoted within the figure panels. b, Representative examples of FN-RMS (left) and FP-RMS (right). UMAP showing genes detected per cell (left). Violin plots showing genes detected within each cell for a given RMS subpopulation (right). c All PDX models assessed by violin plots denoting the number of detected genes per cell across RMS subpopulations.

Extended Data Fig. 3 A subset of fusion-positive RMS contain unique and tumor-specific cell clusters that express neural genes.

a, Top enriched molecular signatures from MSigDB are shown for each unique cell cluster identified from individual FP-RMS PDX models. False Discovery Rate (FDR) Q values noted. Tumor and cluster number are noted (that is, MSK74711-8). b, Venn diagram showing little overlap in gene expression across unique transcription clusters identified from different tumors. c, Upset plot quantifying the gene set enrichment of unique clusters with the GO_NEUROGENESIS gene set (P values defined by Fisher’s exact test).

Extended Data Fig. 4 RMS cells ubiquitously express a subset of muscle lineage and cancer-specific genes.

a,b, UMAP visualizations showing cell states (left panels) and compared with gene expression for MYOD1, DESMIN and MYC. Representative examples shown for fusion-negative (MAST39, n = 10,577 cells) and fusion-positive RMS (MAST95, n = 4,771 cells). c,d, Histological analysis of PDXs grown in NSG mice. Representative sections of three tumors shown with similar results. Hematoxylin and Eosin (left) and immunohistochemistry for MYOD1 and DESMIN (right). Fusion-negative (FN, c) and fusion-positive RMS (FP, d). Scale bar = 50 μm. n = 3 independently engrafted tumors were stained and imaged.

Extended Data Fig. 5 Immunofluorescence antibody staining reveals intermingling of cell states in PDX tumors.

a, Immunofluorescence staining within the central tumor mass. Dashed lines indicate clustered cell populations. Arrows denote rare cells detected by IF staining. Scale bar = 50 μm. b,c Heterogeneity identified by single-cell RNA sequencing (b) and compared with immunofluorescence staining of the central tumor mass (IF-C, c) or at the invasive edge (IF-E, c). Color coding denotes that immunofluorescence was detected in tumor cells within the sections analyzed. ND, not detected; NA, not applicable. Evenly distributed through tumor (ED) or clustered (C) based on immunofluorescence staining. d Quantitation of cell state percentages assessed by scRNA-sequencing or immunofluorescence (center of mass, C compared with tumor edge, E). Error bar equals s.e.m. (n = 4 image fields analyzed per condition, range 207–643 cells/field).

Extended Data Fig. 6 Cell state heterogeneity in primary patient samples, PDX models, single cell engrafted tumors and RD cells grown in mouse xenografts.

ac, 3D renderings of gene expression for muscle (x axis), proliferation (z axis), mesenchymal-like (y axis) gene modules identified in RMS samples (a, FN-PDXs; b, FP-PDXs; c, primary patient samples). Individual cells are noted by dots and color coded based on cell assignments shown in Fig. 1d. Not detected (ND) denotes lack of a given cell state both in the initial UMAP cell cluster annotations and in 3D gene expression space. d, Combined UMAP visualization for all parental PDXs profiled (n = 10, and includes both replicates of MAST85) and single cell derived PDXs (n = 3). See Supplementary Table 1 for detailed information and cell numbers. e,f, Single cell RNA sequencing of RD xenograft (n = 2,619 tumor cells profiled). Heatmap showing single cells (x axis) and genes enriched for specific transcription modules (y axis, e). Cells are arranged by UMAP clusters, combined based on expression similarity, and then assigned a specific cell state as noted. f UMAP rendering of xenografted RD cells following single cell sequencing (left) and quantification of cell state composition of all 2,619 RD cells profiled (right). Similar cell states are observed in RD cells raised in 2D cell culture (see Fig. 3).

Source data

Extended Data Fig. 7 Tumor-propagating potential is enriched in the mesenchymal-enriched tumor cell fraction in FN-RMS.

a,e,i,m, Flow cytometry analysis of FN-MSK74711 cells harvested directly from PDX tumors grown in NSG mouse (a) or cell line models (e,i,m) prior to (left) and after FACS enrichment (right two panels). b,f,j,n, Quantitative PCR confirming cell state enrichment following FACS (n = 2 independently engrafted tumors (MSK74711) and analyzed in triplicate from independent sorts, 6 datum points shown). Three replicates were analyzed from independent sorting of RD, 381 T and SMS-CTR cells. Two-way ANOVA followed by two-sided Student’s t-test comparison (*P < 0.05, **P < 0.01; ***P < 0.001 and ****P < 0.0001). c,g,k,o, Representative images of sphere size following FACS enrichment and plating for two weeks (scale bar = 20 μm). Shown is a representative micrograph of tumorspheres (left) with similar results seen across replicate wells. Combined quantification from two independent experiments (c,g,k) and three independent experiments in panel o. Three wells/experiment were completed and the combined analysis shown (o, +/− SEM noted). d,h,l,p, Quantification of tumor sphere numbers from representative limiting dilution experiments (datum points represent number of spheres found within individual wells from a single experiment). Experiments replicated twice (d,h,l) or three times (p) with similar results. Mean + /− SEM noted for SMS-CTR, two-way ANOVA followed by two-sided Student’s t-test (*P < 0.05, **P < 0.01, ***P < 0.001). Mesenchymal-enriched (Mesen, Mes or Me, muscle (Musc, Mu), interferon (INF), proliferative (Prolif).

Source data

Extended Data Fig. 8 Limiting dilution cell transplantation confirms that mesenchymal-enriched cells from FN-RMS PDX 74711 are enriched for tumor propagating potential in vivo.

a Representative images of NSG mice engrafted with CHODL + /CD90 + mesenchymal-enriched or CHODL-/CD90- MSK74711 PDX RMS cells (all three mice from 10,000 cells/mouse group are shown). Mice were imaged at days post-transplantation as noted. Dashed lines delineate tumor. b, Latency of tumor regrowth following engraftment into NSG mice. TPC frequency + /− 95% confidence interval noted per condition in parenthesis. Quantification by ELDA *P < 0.05, **P < 0.01. n = 3 animals were transplanted for each limiting dilution across each of the four sorted cell populations analyzed. c, Flow analysis of tumors generated from sorted cell populations, mean + /− SEM noted, n = 3 independent tumors, *P < 0.05, **P < 0.01, ***P < 0.001 by two-sided Student’s t-test comparing the Mesen+ vs. Mesen− populations. d, Immunostaining of Ki67 proliferation and MF20 differentiation muscle markers in animals engrafted with FACs sorted cells. n = 3 independent tumors. For each tumor, four random fields were selected for quantification. mean + /− SEM., *P < 0.05, **P < 0.01, ***P < 0.001, by two-sided Student’s t-test.

Source data

Extended Data Fig. 9 Subtype-specific RMS core signatures are expressed at specific muscle development stages.

a,b Dot plot renderings showing the expression of ten representative genes that comprise the fusion-negative or fusion-positive core signature across all PDXs and their identified cell states (a) and across normal muscle cells stratified by age (b). c UMAP rendering of scRNA sequencing data from embryonic (n = 5 samples), fetal (n = 4 samples), and adult skeletal muscle (n = 4 samples) showing expression of representative subtype-specific core signature genes. Week or year of life is noted (Wk and Yr, respectively). n = 3,251 cells analyzed across all stages of muscle development.

Extended Data Fig. 10 Osteogenic markers are expressed in the mesenchymal-enriched FN-RMS tumor propagating cells.

a, TSNE renderings denoting cell state (left) and compared with OGN and MGP expression in representative FN-RMS PDXs, MAST111 (n = 2,199 cells) and MAST39 (n = 10,577 cells). b, FACS sorting of RD and 381 T FN-RMS cells followed by Q-PCR validates the enrichment of osteogenic markers OGN and MGP within the mesenchymal-enriched subpopulation. FACs plots are representative across three independent experiments completed on different days (left). Q-PCR samples are the same as those rendered in Fig. 4 and Extended Data Fig. 7, Mean+/−SEM (n = 3 samples isolated from independent sorts completed on different days). ***P < 0.001, ****P < 0.0001 by ANOVA followed by two-sided Student’s t-test. c, Representative flow cytometric analysis confirming cell surface expression of OGN and MGP in mesenchymal-enriched subfractions of RD and 381 T RMS cells (completed in three independent biological replicates).

Source data

Supplementary information

Supplementary Information

Representative example of sorting strategy used for FACS experiments.

Reporting Summary

Supplementary Table 1

Supplementary tables.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wei, Y., Qin, Q., Yan, C. et al. Single-cell analysis and functional characterization uncover the stem cell hierarchies and developmental origins of rhabdomyosarcoma. Nat Cancer 3, 961–975 (2022). https://doi.org/10.1038/s43018-022-00414-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-022-00414-w

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