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:

Unified rhombic lip origins of group 3 and group 4 medulloblastoma

Abstract

Medulloblastoma, a malignant childhood cerebellar tumour, segregates molecularly into biologically distinct subgroups, suggesting that a personalized approach to therapy would be beneficial1. Mouse modelling and cross-species genomics have provided increasing evidence of discrete, subgroup-specific developmental origins2. However, the anatomical and cellular complexity of developing human tissues3—particularly within the rhombic lip germinal zone, which produces all glutamatergic neuronal lineages before internalization into the cerebellar nodulus—makes it difficult to validate previous inferences that were derived from studies in mice. Here we use multi-omics to resolve the origins of medulloblastoma subgroups in the developing human cerebellum. Molecular signatures encoded within a human rhombic-lip-derived lineage trajectory aligned with photoreceptor and unipolar brush cell expression profiles that are maintained in group 3 and group 4 medulloblastoma, suggesting a convergent basis. A systematic diagnostic-imaging review of a prospective institutional cohort localized the putative anatomical origins of group 3 and group 4 tumours to the nodulus. Our results connect the molecular and phenotypic features of clinically challenging medulloblastoma subgroups to their unified beginnings in the rhombic lip in the early stages of human development.

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: Molecular signatures align group 3/4 MB with the human fetal RL.
Fig. 2: Conservation of human fetal RLSVZ gene signatures in group 3/4 MB.
Fig. 3: Genetic targeting of RLSVZ markers in group 3/4 MB.
Fig. 4: Absence of the human RLSVZ photoreceptor signature in the mouse cerebellum.
Fig. 5: MRI mapping of group 3/4 MB origins in the cerebellum.
Fig. 6: Proposed model of group 3/4 MB origins.

Similar content being viewed by others

Data availability

Previously unpublished scRNA-seq, bulk RNA-seq and array-based DNA methylation datasets described in this study have been deposited in the GEO with the accession code GSE207266. The harmonized mouse single-cell cerebellar atlas expression matrix is accessible in the GEO under the same accession code (GSE207266).

References

  1. Northcott, P. A. et al. Medulloblastoma. Nat. Rev. Dis. Primers 5, 11 (2019).

    Article  PubMed  Google Scholar 

  2. Hovestadt, V. et al. Medulloblastomics revisited: biological and clinical insights from thousands of patients. Nat. Rev. Cancer 20, 42–56 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Haldipur, P., Millen, K. J. & Aldinger, K. A. Human cerebellar development and transcriptomics: implications for neurodevelopmental disorders. Annu. Rev. Neurosci. 45, 515–531 (2022).

    Article  PubMed  Google Scholar 

  4. Jones, D. T. W. et al. Molecular characteristics and therapeutic vulnerabilities across paediatric solid tumours. Nat. Rev. Cancer 19, 420–438 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. Hovestadt, V. et al. Resolving medulloblastoma cellular architecture by single-cell genomics. Nature 572, 74–79 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. Vladoiu, M. C. et al. Childhood cerebellar tumours mirror conserved fetal transcriptional programs. Nature 572, 67–73 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Haldipur, P. et al. Spatiotemporal expansion of primary progenitor zones in the developing human cerebellum. Science 366, 454–460 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  8. Aldinger, K. A. et al. Spatial and cell type transcriptional landscape of human cerebellar development. Nat. Neurosci. 24, 1163–1175 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Jessa, S. et al. Stalled developmental programs at the root of pediatric brain tumors. Nat. Genet. 51, 1702–1713 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gibson, P. et al. Subtypes of medulloblastoma have distinct developmental origins. Nature 468, 1095–1099 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Capper, D. et al. DNA methylation-based classification of central nervous system tumours. Nature 555, 469–474 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Consalez, G. G., Goldowitz, D., Casoni, F. & Hawkes, R. Origins, development, and compartmentation of the granule cells of the cerebellum. Front. Neural Circuits 14, 611841 (2020).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Englund, C. et al. Unipolar brush cells of the cerebellum are produced in the rhombic lip and migrate through developing white matter. J. Neurosci. 26, 9184–9195 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hagan, N. & Zervas, M. Wnt1 expression temporally allocates upper rhombic lip progenitors and defines their terminal cell fate in the cerebellum. Mol. Cell. Neurosci. 49, 217–229 (2012).

    Article  CAS  PubMed  Google Scholar 

  16. McDonough, A. et al. Unipolar (dendritic) brush cells are morphologically complex and require Tbr2 for differentiation and migration. Front. Neurosci. 14, 598548 (2020).

    Article  PubMed  Google Scholar 

  17. Furukawa, T., Morrow, E. M. & Cepko, C. L. Crx, a novel otx-like homeobox gene, shows photoreceptor-specific expression and regulates photoreceptor differentiation. Cell 91, 531–541 (1997).

    Article  CAS  PubMed  Google Scholar 

  18. Kozareva, V. et al. A transcriptomic atlas of mouse cerebellar cortex comprehensively defines cell types. Nature 598, 214–219 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Nayler, S., Agarwal, D., Curion, F., Bowden, R. & Becker, E. B. E. High-resolution transcriptional landscape of xeno-free human induced pluripotent stem cell-derived cerebellar organoids. Sci. Rep. 11, 12959 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Cho, Y. J. et al. Integrative genomic analysis of medulloblastoma identifies a molecular subgroup that drives poor clinical outcome. J. Clin. Oncol. 29, 1424–1430 (2011).

    Article  PubMed  Google Scholar 

  21. Garancher, A. et al. NRL and CRX define photoreceptor identity and reveal subgroup-specific dependencies in medulloblastoma. Cancer Cell 33, 435–449 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kool, M. et al. Integrated genomics identifies five medulloblastoma subtypes with distinct genetic profiles, pathway signatures and clinicopathological features. PLoS One 3, e3088 (2008).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  23. Northcott, P. A. et al. Medulloblastoma comprises four distinct molecular variants. J. Clin. Oncol. 29, 1408–1414 (2011).

    Article  PubMed  Google Scholar 

  24. Northcott, P. A. et al. The whole-genome landscape of medulloblastoma subtypes. Nature 547, 311–317 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Sharma, T. et al. Second-generation molecular subgrouping of medulloblastoma: an international meta-analysis of Group 3 and Group 4 subtypes. Acta Neuropathol. 138, 309–326 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Lin, C. Y. et al. Active medulloblastoma enhancers reveal subgroup-specific cellular origins. Nature 530, 57–62 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bunt, J. et al. OTX2 directly activates cell cycle genes and inhibits differentiation in medulloblastoma cells. Int. J. Cancer 131, E21–E32 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Sweet-Cordero, E. A. & Biegel, J. A. The genomic landscape of pediatric cancers: implications for diagnosis and treatment. Science 363, 1170–1175 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Grobner, S. N. et al. The landscape of genomic alterations across childhood cancers. Nature 555, 321–327 (2018).

    Article  ADS  PubMed  Google Scholar 

  30. Northcott, P. A. et al. Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma. Nature 511, 428–434 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Goodrich, L. V., Milenkovic, L., Higgins, K. M. & Scott, M. P. Altered neural cell fates and medulloblastoma in mouse patched mutants. Science 277, 1109–1113 (1997).

    Article  CAS  PubMed  Google Scholar 

  32. Kawauchi, D. et al. A mouse model of the most aggressive subgroup of human medulloblastoma. Cancer Cell 21, 168–180 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Pei, Y. et al. An animal model of MYC-driven medulloblastoma. Cancer Cell 21, 155–167 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Swartling, F. J. et al. Distinct neural stem cell populations give rise to disparate brain tumors in response to N-MYC. Cancer Cell 21, 601–613 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Perreault, S. et al. MRI surrogates for molecular subgroups of medulloblastoma. Am. J. Neuroradiol. 35, 1263–1269 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Wefers, A. K. et al. Subgroup-specific localization of human medulloblastoma based on pre-operative MRI. Acta Neuropathol. 127, 931–933 (2014).

    Article  PubMed  Google Scholar 

  37. Gajjar, A. et al. Outcomes by clinical and molecular features in children with medulloblastoma treated with risk-adapted therapy: results of an international phase III trial (SJMB03). J. Clin. Oncol. 39, 822–835 (2021).

    Article  CAS  PubMed  Google Scholar 

  38. Robinson, G. W. et al. Risk-adapted therapy for young children with medulloblastoma (SJYC07): therapeutic and molecular outcomes from a multicentre, phase 2 trial. Lancet Oncol. 19, 768–784 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Patay, Z. et al. MR imaging characteristics of Wingless-type-subgroup pediatric medulloblastoma. Am. J. Neuroradiol. 36, 2386–2393 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Aldape, K. et al. Challenges to curing primary brain tumours. Nat. Rev. Clin. Oncol. 16, 509–520 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Gajjar, A. J. & Robinson, G. W. Medulloblastoma—translating discoveries from the bench to the bedside. Nat. Rev. Clin. Oncol. 11, 714–722 (2014).

    Article  CAS  PubMed  Google Scholar 

  42. Cavalli, F. M. G. et al. Intertumoral heterogeneity within medulloblastoma subgroups. Cancer Cell 31, 737–754 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Poli, V. et al. MYC-driven epigenetic reprogramming favors the onset of tumorigenesis by inducing a stem cell-like state. Nat. Commun. 9, 1024 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  44. Friedmann-Morvinski, D. et al. Dedifferentiation of neurons and astrocytes by oncogenes can induce gliomas in mice. Science 338, 1080–1084 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  45. Morfouace, M. et al. Pemetrexed and gemcitabine as combination therapy for the treatment of Group 3 medulloblastoma. Cancer Cell 25, 516–529 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Pei, Y. et al. HDAC and PI3K antagonists cooperate to inhibit growth of MYC-driven medulloblastoma. Cancer Cell 29, 311–323 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Tao, R. et al. MYC drives group 3 medulloblastoma through transformation of Sox2+ astrocyte progenitor cells. Cancer Res. 79, 1967–1980 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Kawauchi, D. et al. Novel MYC-driven medulloblastoma models from multiple embryonic cerebellar cells. Oncogene 36, 5231–5242 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Behesti, H., Kocabas, A., Buchholz, D. E., Carroll, T. S. & Hatten, M. E. Altered temporal sequence of transcriptional regulators in the generation of human cerebellar granule cells. eLife 10, e67074 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Polanski, K. et al. BBKNN: fast batch alignment of single cell transcriptomes. Bioinformatics 36, 964–965 (2020).

    CAS  PubMed  Google Scholar 

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

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  52. 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  CAS  PubMed  Google Scholar 

  53. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. 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 

  55. Wang, X., Park, J., Susztak, K., Zhang, N. R. & Li, M. Bulk tissue cell type deconvolution with multi-subject single-cell expression reference. Nat. Commun. 10, 380 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  56. 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  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  57. Riemondy, K. A. et al. Neoplastic and immune single cell transcriptomics define subgroup-specific intra-tumoral heterogeneity of childhood medulloblastoma. Neuro-oncology 24, 273–286 (2021).

    Article  PubMed Central  Google Scholar 

  58. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Hanzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21.29.21–21.29.29 (2015).

    Article  Google Scholar 

  64. Corces, M. R. et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat. Methods 14, 959–962 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Li, H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. Preprint at https://arxiv.org/abs/1303.3997 (2013).

  66. Ramírez, F., Dündar, F., Diehl, S., Grüning, B. A. & Manke, T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–W191 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Vo, B. T. et al. Mouse medulloblastoma driven by CRISPR activation of cellular Myc. Sci Rep. 8, 8733 (2018).

    Article  ADS  MathSciNet  PubMed  PubMed Central  Google Scholar 

  69. Lee, C. et al. Lsd1 as a therapeutic target in Gfi1-activated medulloblastoma. Nat. Commun. 10, 332 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was principally supported by the American Lebanese Syrian Associated Charities and St Jude (P.A.N.), The Sontag Foundation (Distinguish Scientist Award; P.A.N.), St. Baldrick’s Foundation (Robert J. Arceci Innovation Award; P.A.N.) and the National Cancer Institute (P.A.N.; P01CA096832-16A1). P.A.N. is a Pew-Stewart Scholar for Cancer Research (Margaret and Alexander Stewart Trust). L.B. was supported by a Future Leaders Award from The Brain Tumour Charity (GN-000518). V.V.C. acknowledges funding from R01NS093009. From St Jude, we thank the Flow Cytometry Core Laboratory (Department of Developmental Neurobiology), the Flow Cytometry and Cell Sorting Shared Resource Facility, the Hartwell Center (supported in part by NCI grant P30 CA021765) and the Center for In Vivo Imaging and Therapeutics (supported in part by NCI grants R50CA211481 and P30 CA021765 (Cancer Center)). We thank G. Campbell for assistance with immunostaining. Images were acquired at the Cell & Tissue Imaging Center, which is supported by St Jude and NCI P30 CA021765. We also thank A. Vasilyeva for coordination of clinical study information, M. Batts for animal care support and B. Stelter for assistance with artwork. Healthy human samples were provided by the Birth Defects Research Laboratory at the University of Washington (supported by NICHD R24 HD000836 to I.A.G.) and the Joint Medical Research Council/Wellcome (MR/R006237/1) Human Developmental Biology Resource. Human tissue used in this study was covered by a material transfer agreement between SCRI and HDBR.

Author information

Authors and Affiliations

Authors

Contributions

K.S.S., L.B., B.L.G., K.J.M. and P.A.N. designed the study. K.S.S., B.L.G., Q.G. and Y.L. performed computational analyses. L.B. performed mouse lineage-tracing experiments, scRNA-seq and ATAC-seq. R.T. generated the developmental mouse scRNA-seq dataset and performed OTX2 functional studies in MB cells. I.Y.I. performed microdissection of the mouse RL. P.H., J.M., A.H.S. and J.G. performed mouse and human ISH experiments and immunofluorescence imaging of human fetal cerebellum. M.D. performed LMD and DNA extraction from human fetal cerebellum. H.S. and B.A.O. performed immunofluorescence and immunohistochemistry in MB and human adult cerebellum. Z.P. reviewed MRI evaluations, A.O.-T. and Z.P. analysed tumour measurements and locations and Z.P., M.S., S.Z. and A.E. generated the heat maps of MB location. K.A.A., V.V.C., I.A.G., L.M.O., J.H., K.M., A.G., G.W.R., V.H. and Z.P. supported the project. K.S.S., L.B., B.L.G., V.H., K.J.M. and P.A.N. wrote the manuscript with contributions from all authors. K.J.M. and P.A.N supervised and funded the study.

Corresponding author

Correspondence to Paul A. Northcott.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Mary Elizabeth Hatten and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Transcriptional atlas of the human fetal cerebellum.

(a) Schematic summary of the molecular datasets used to interconnect mouse and human cerebellar profiles with MB. Single-cell and bulk sample counts for each data source are indicated. (b, c) UMAP plots showing published cell-type annotation (b) and predicted cell-cycle phase (c) of the human fetal cerebellar atlas8. (d) Correlation heat map of human cerebellar cell types. (e) Violin plots showing expression of cerebellar lineage markers in the human fetal cerebellar atlas. (f) Inferred cerebellar cell-type proportions in deconvoluted bulk MB transcriptomes. (g) Proportion of cerebellar lineage-associated genes upregulated in MB subgroups. (h) MB subgroup-specific gene set enrichment in cerebellar cell types.

Extended Data Fig. 2 Reciprocal classification of human fetal cerebellar and MB datasets using developmental and tumorigenic signatures.

(a) Predicted pseudotime of MB single cells according to underlying cellular states. (b) RL–GlutaCN/UBC cell state metagenes projected onto group 3/4-MB single cells. MB single cells are positioned according to the previously reported cellular hierarchy5. (c) Predicted pseudotime of bulk MB expression profiles according to underlying developmental cell states. Molecular subgroup, subtype, and driver gene alteration status is annotated for each individual tumour. Inferred cellular proportions of GluCN/UBC cell states are annotated for each tumour profile. (d) UMAP plot of MB Smart-seq2 training dataset, annotated with published subgroup assignments5. (e) UMAP plot of MB 10x scRNA-seq test dataset, annotated with published subgroup assignments57. (f) UMAP plot of MB 10x scRNA-seq test dataset, labelled with predicted subgroup assignments based on the neural-net classifier. (g) Confusion matrix summarizing performance of the neural-net classifier. (h) (Left panel) Pseudotime diffusion map of glutamatergic lineages extracted from the human cerebellar atlas. (Right panel) Classification of glutamatergic single cells according to MB subgroup based on the neural-net classifier. (i) Heat map showing unsupervised clustering of human RL (n = 7), EGL (n = 7), and MB (n=559) DNA methylation profiles. (j) DNA methylation-based classification of microdissected human fetal EGL (n = 7) and RL (n = 7) samples. Reference entities represent an aggregated set of CNS tumour and non-neoplastic methylation classes. Error bars represent standard error of the mean.

Extended Data Fig. 3 Enrichment of photoreceptor and UBC gene signatures in the RLSVZ.

(a) Heat maps showing expression of RLSVZ-enriched photoreceptor (upper panel) and UBC (lower panel) marker genes in human cerebellar subcompartments. (b) UMAP plots showing mean expression of the photoreceptor (upper panel) and UBC (lower panel) gene sets across 77 human cell types13. (c) Heat maps showing mean expression of RLSVZ-enriched UBC (left panel) and photoreceptor (right panel) marker genes across 77 human cell types. (d, e) Quantification of RLSVZ-enriched photoreceptor (d) and UBC (e) gene sets in human cerebellar glutamatergic lineages.

Extended Data Fig. 4 Status of RLSVZ photoreceptor and RLSVZ UBC gene signatures in adult human cerebellum and cerebellar organoids.

(a) IHC of CRX (photoreceptor marker) and EOMES (UBC marker) in the adult human cerebellum. The precise localization of EOMES positivity was unconfirmed. Scale bars, 40 μm. (b) UMAP summarizing snRNA-seq data derived from >150,000 nuclei isolated from the adult human cerebellum according to published cell types18. (c) Quantification of the RLSVZ, photoreceptor, and UBC gene expression signatures in the adult human cerebellar atlas by cell type. Proportion of excitatory neurons positive for each expression signature is indicated. P-values were calculated as described in Fig. 4d. (d) Expression of select marker genes in the adult human cerebellar atlas. (e) UMAP summary of cell types annotated in published human cerebellar organoids19. (f) Violin plots quantifying enrichment of the RLSVZ, photoreceptor, and UBC gene sets in cell types annotated in human cerebellar organoids. P-values were calculated as described in Fig. 4d. (g) Photoreceptor and UBC marker gene expression in cell types annotated in published human cerebellar organoids.

Extended Data Fig. 5 Expression of RLSVZ marker genes across cerebellar subcompartments and MB subgroups.

(a, b) Heat maps of RLSVZ-photoreceptor (a) and RLSVZ-UBC (b) marker gene expression in group 3/4-MB. (c, d) Box plots of select RLSVZ-photoreceptor (c) and RLSVZ-UBC (d) marker genes summarizing expression across human cerebellar subcompartments and MB subgroups. All box plots were created as described in Fig. 2c.

Extended Data Fig. 6 Correlation of OTX2 expression and transcription factor activity with the RLSVZ-photoreceptor signature.

(a, b) Correlation of OTX2 and RLSVZ-photoreceptor gene set expression across human cerebellar subcompartments (a) and group 3/4-MB (b). P-values were calculated using a two-sided Pearson’s correlation test. (c) iRegulon analysis of the RLSVZ-photoreceptor gene set. (d) Enrichment of the RLSVZ-derived photoreceptor gene set in the RL–GlutaCN/UBC lineage trajectory. (e) OTX2 TF activity in the RL–GlutaCN/UBC lineage trajectory. (f) Correlation of OTX2 TF activity and expression of the RLSVZ-photoreceptor gene set in single cells derived from the RL–GlutaCN/UBC lineage. P-values were calculated using a two-sided Pearson’s correlation test.

Extended Data Fig. 7 OTX2 is required for maintenance of the photoreceptor signature in group 3 MB.

(a) Experimental approach for inactivating OTX2 in group 3-MB cells. (b) Western blot showing efficient reduction of OTX2 protein expression at day 7 in OTX2-edited D283 MB cells. (c) Volcano plot highlighting differentially expressed genes in OTX2-edited versus non-targeted D283 cells. P-values were calculated using a two-sided Wald test and were adjusted for multiple comparisons using FDR correction. (d) GSEA results showing the top upregulated and downregulated gene sets in OTX2-edited D283 cells compared to non-targeted controls. (e) Significant upregulation of the RLSVZ-derived UBC gene set in OTX2-edited D283 cells. (f) Significant downregulation of the RLSVZ-photoreceptor gene set in OTX2-edited D283 cells. P-values for all gene set enrichment analyses were calculated using the Monte Carlo method. (g) Pairwise analysis of recurrent genetic alterations in group 3/4-MB. P-values were calculated using a linear regression model and adjusted for multiple comparisons using the FDR correction. (h) Smoothed expression of DDX31, BARHL1, and OTX2 in the human fetal GlutaCN/UBC lineage sorted by pseudotime.

Extended Data Fig. 8 Single-cell profiles of the developing mouse cerebellum and lineage-enriched subpopulations.

(a, b) UMAP plots of the harmonized mouse cerebellar atlas summarized by data origin (a) and developmental stage (b). (c) UMAP plot of the harmonized mouse cerebellar atlas indicating consensus cell-type annotations. (d) Heat map showing relative enrichment of mouse cerebellar lineage gene sets in human fetal cerebellar cell types. (e,f) UMAP plots showing enrichment of the human RLVZ (e) and RLSVZ (f) gene signatures in the mouse cerebellar atlas. (g) Violin plots quantifying enrichment of RLVZ (upper panel) and RLSVZ (lower panel) gene sets in mouse cerebellar cell types. Cell types with enrichment in >25% of cells are indicated; adjusted p < 0.05. (h) Enrichment of the mouse RL gene signature in the human cerebellar atlas. (i) Violin plots quantifying enrichment of the mouse RL gene signature in human cerebellar cell types. (j) Violin plots quantifying enrichment of the human RLSVZ-UBC gene signature across mouse (upper panel) and human (lower panel) cell types. P-values for all violin plots were calculated as described in Fig. 4d. Additional details regarding the harmonization of the mouse cerebellar atlas are summarized in the Supplementary Information.

Extended Data Fig. 9 Quantification of photoreceptor and UBC gene sets in mouse MB models.

(a) Enrichment strategy for isolating the mouse RL–UBC lineage. (b) UMAP plots of the enriched mouse RL–UBC trajectory, showing experimental source and select marker gene expression. (c) (Left panel) Latent time analysis of RL–UBC lineage-enriched single cells. (Right panels) Enrichment quantification of the RLSVZ-photoreceptor and -UBC gene sets in the mouse RL–UBC lineage trajectory. (d) ISH of photoreceptor marker genes in mouse cerebellum. Scale bars, 200μm. (e, f) Box plots summarizing enrichment of the RLSVZ-photoreceptor (e) and UBC (f) gene sets across published mouse MB transcriptional datasets (n = 14). Human MB and normal P7 cerebellum datasets were included as a reference. All box plots were created as described in Fig. 2c.

Extended Data Fig. 10 Anatomical mapping of MB diagnosis in the cerebellum.

(a–d) Select pre- and post-surgical MRIs of ‘small’ MBs according to subgroup. Group 3 (a) and group 4 (b) tumours indicate a common anatomical point of origin in the nodulus. Additional details for each ‘small’ MB are summarized in the Supplementary Information.

Supplementary information

Supplementary Information

This file contains Supplementary Figs. 1–4 and Supplementary Notes.

Reporting Summary

Peer Review File

Supplementary Table 1

Summary of human and mouse molecular datasets.

Supplementary Table 2

Human cerebellar atlas annotations by developmental stage.

Supplementary Table 3

Enrichment of MB subgroup-specific gene sets in human cerebellar lineages.

Supplementary Table 4

DNA methylation classification of human cerebellar subcompartments.

Supplementary Table 5

Human cerebellar compartment-specific signatures and associated gene sets.

Supplementary Table 6

Enrichment of Descartes CNS cell-type gene sets in human cerebellar subcompartments.

Supplementary Table 7

Mouse cerebellar atlas annotations by developmental stage.

Supplementary Table 8

Summary of MB tumour size and location for institutional MRI cohort.

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Smith, K.S., Bihannic, L., Gudenas, B.L. et al. Unified rhombic lip origins of group 3 and group 4 medulloblastoma. Nature 609, 1012–1020 (2022). https://doi.org/10.1038/s41586-022-05208-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-05208-9

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: 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