Main

Malignant transformation of the human rhombic lip results in medulloblastoma, with group 3 (G3), group 4 (G4) and sonic hedgehog (SHH) tumors arising from the upper rhombic lip, and wingless/integrated (WNT) medulloblastoma arising from the lower rhombic lip1,2,3,4,5,6,7,8,9,10,11,12,13. There are a number of well-known driver genes for medulloblastoma, particularly SHH pathway genes in SHH medulloblastoma. However, G4 medulloblastoma is less well understood, with mutations of histone modifier genes, members of the CBFA complex and amplifications of MYCN and OTX2 (refs. 3,14). A tail of less well understood but recurrent somatically altered genes has been observed across medulloblastoma subgroups14.

The zinc finger protein in the cerebellum (ZIC) family of transcription factors (TFs) has crucial roles in the development of the central nervous system (CNS), including hindbrain development15,16,17. There are five human ZIC family genes (ZIC1ZIC5), all of which contain conserved tandem C2H2 zinc finger motif repeats that can interact with DNA or other proteins15,16,17,18. While ZICs exhibit some overlapping expression patterns throughout the CNS, different mutations are associated with distinct congenital disorders15,16,19. Somatic mutations of ZIC1 have been identified in distinct medulloblastoma subgroups, and although ZIC1 is a pan-medulloblastoma master TF associated with an active super-enhancer (SE)20, the specific role of ZIC TFs in the etiology of medulloblastoma is obscure.

ZIC1 and ZIC4 have multiple critical roles in cerebellar development15,16,21. Heterozygous deletion of the ZIC1/ZIC4 locus in humans22 is a rare cause of Dandy–Walker malformation (DWM), which includes cerebellar hypoplasia16. Gain-of-function (GOF) mutations at the carboxy terminus of ZIC1 have been identified in children with craniosynostosis and learning disabilities23. We now demonstrate that ZIC1 mutations in medulloblastoma are context dependent, with loss-of-function (LOF) mutations and epigenetic alterations in G4 medulloblastoma, contrasted with GOF mutations in SHH medulloblastoma. Concordantly, expression of ZIC1 represses malignant phenotypes in G3/G4 medulloblastoma while enhancing malignant phenotypes in SHH medulloblastoma in model systems. ZIC1 is therefore a stark example of how the same gene can have distinct driver mechanisms in highly similar cancers depending on their specific lineage of origin.

Results

The subgroup-specific H3K27ac/H3K27me3 landscape of medulloblastoma

Due to the high prevalence and recurrence of somatic mutations in genes associated with chromatin modulation in medulloblastoma (~30% of medulloblastomas)14, we hypothesized that some medulloblastomas might acquire somatic histone modification alterations (chromatin variants24,25) for driver genes. To test this hypothesis, we profiled H3K27ac and H3K27me3 landscapes across the four medulloblastoma subgroups (including 123 matching samples for H3K27ac and 63 matching samples for H3K27me3) and integrated the data with matching RNA sequencing (RNA-seq), as well as an independent cohort of tumors characterized by H3K27ac HiChIP (Fig. 1a, Extended Data Fig. 1a and Supplementary Tables 1 and 2). Hierarchical clustering using either H3K27ac or H3K27me3 chromatin immunoprecipitation followed by sequencing (ChIP–seq) data recapitulated the four subgroups (Fig. 1b). We categorized subgroup-specific H3K27 modification as either subgroup-enriched peaks (signal enrichment) or subgroup-recurrent peaks (peak called recurrently for one subgroup; Fig. 1c–e). A subset of the identified peaks was shared by either SHH/WNT (enriched in SHH versus G3 or G4, but not WNT) or G3/G4 (enriched in G3 versus SHH or WNT, but not G4; Fig. 1d, e) and were documented as such.

Fig. 1: Characterization of subgroup-specific chromatin landscape of medulloblastoma.
figure 1

a, Summary of the newly generated and public datasets. Number within the bracket indicates the number of tumors with previously published data. b, Hierarchical clustering plots generated using the top 10,000 variable H3K27ac and H3K27me3 ChIP–seq peaks. c, Schematic representation summarizing different types of ChIP–seq peaks used in downstream analysis. Subgroup-specific peaks were defined by identifying peaks that (1) exhibit subgroup enrichment in ChIP–seq read counts or (2) are recurrently present only for specific subgroups even if the average ChIP–seq read count is not strongly subgroup enriched on average. d, Number of subgroup-specific peaks for each subgroup in the H3K27ac cohort. After batch correction, peaks annotated as subgroup enriched for ChIP–seq reads or subgroup recurrent were characterized separately. e, Number of subgroup-specific H3K27me3 peaks using the same annotations/criteria as d. f, Number of peaks and proportion of genome covered by H3K27ac and H3K27me3 peaks across the medulloblastoma subgroups. P values were calculated by the tailed Mann–Whitney U test. Biological sample size for H3K27ac—G3/G4/SHH/WNT = 27/47/39/10 and H3K27me3—G3/G4/SHH/WNT = 14/24/22/3. Center of box, median. Bounds of box, 25% and 75% percentile. Whiskers show minimum and maximum values within the 1.5× interquartile range. g, Schematic representation summarizing how high-confidence enhancer–promoter interactions were identified from HiChIP and ChIP–seq data. Adjusted P values were calculated using Pearson correlation between target gene transcript and enhancer H3K27ac read levels, which was corrected for multiple testing. h, Summary of distance distribution for high-confidence enhancer–promoter interactions. Proportion of SCLs (g; Methods) over a total number of loops is depicted as overlapping Venn diagrams. Double asterisk (**) indicates a significant correlation (P.adj < 0.1).

The average number of peaks and the proportion of genome coverage for H3K27ac did not significantly differ between subgroups (Fig. 1f). However, H3K27me3 deposition was markedly increased in G3 medulloblastoma (Fig. 1f). Additionally, G3/G4 medulloblastoma-enriched H3K27me3 peaks exhibited a strong preference for gene promoters as compared to WNT/SHH (Extended Data Fig. 1b). Core regulatory circuit analysis of H3K27ac ChIP–seq data identified known and new medulloblastoma subgroup-specific master TFs, including the pan-subgroup master TFs ZIC1 and ZIC4 as we reported previously (Extended Data Fig. 1c–e)20. Additionally, H3K27ac HiChIP was used to define the enhancer–promoter interactome across medulloblastoma subgroups (Fig. 1g). Integration of H3K27ac HiChIP, H3K27ac ChIP–seq and RNA-seq allowed the identification of loops connecting enhancers and promoters of protein-coding genes. Among the enhancer–promoter interacting loops, those with enhancer H3K27ac read counts exhibiting significant positive correlations with the expression of target genes were also identified (adjusted P < 0.1) and defined as significantly correlated loops (SCL; Fig. 1g,h). Many SCL-associated enhancers target more than one gene (Extended Data Fig. 1f), and notably, enhancers frequently target genes that are not the most proximal gene (Extended Data Fig. 1g).

We conclude that post-translational modification of H3K27 in medulloblastoma varies by subgroup.

Recurrent single-nucleotide variations (SNVs) and hemizygous H3K27me3 affect ZIC1 in G4 medulloblastoma

We hypothesized that a subset of medulloblastoma LOF driver genes somatically altered by SNVs, small insertions/deletions (InDels) or copy number aberrations (CNAs) might also be targeted through somatic H3K27me3-mediated repression to achieve the common endpoint of tumor suppressor gene LOF. We determined the intersection between genes affected by genetic mutations and those overlapping either ‘enriched’ or ‘recurrent’ subgroup-specific H3K27me3 peaks (Fig. 2a and Extended Data Fig. 2a)14. While no overlapping genes were identified for WNT or G3, BCOR for SHH, and both ZIC1 and FLG in G4 are affected by both mutation and H3K27me3-modified chromatin. H3K27me3 peaks on the BCOR promoter (chromosome Xp11.4) were found predominantly in female SHH tumors, suggesting a link to X chromosome inactivation (Extended Data Fig. 2b,c). Broadening the analysis to genes encompassed by focal deletions identified from our published Affymetrix SNP6 array data26,27 identified genes targeted by both deletions and H3K27me3, including the MIR4786 locus in G3 and G4 medulloblastoma (Extended Data Fig. 2d,e and Supplementary Tables 313).

Fig. 2: ZIC1 is recurrently mutated and repressed by H3K27me3 in G4 medulloblastoma.
figure 2

a, Overlap between genes regulated by subgroup-specific H3K27me3 peaks in G3, G4 medulloblastoma and genes recurrently mutated in each subgroup. b, Ranking of SEs across medulloblastoma subgroups, showcasing the number of total SEs identified (in gray) as well as the proportion of subgroup-enriched SEs in pie charts. c, ZIC1 and ZIC4 expression patterns across medulloblastoma subgroups. Biological sample size—G3/G4/SHH/WNT = 72/122/93/24. P values from two-tailed Mann–Whitney U test. Center of box, median. Bounds of box, 25% and 75% percentile. Whiskers show minimum and maximum values within 1.5× interquartile range. d, Sequencing depth normalized bigwig tracks showcasing recurrent (n ≥ 3 per subgroup) ZIC1 and ZIC4 chromatin states across four subgroups. e, Summary of chromatin states observed at the ZIC1 promoter across all samples in the ChIP–seq libraries with both H3K27ac and H3K27me3 modifications. f, Expression levels of ZIC1 and ZIC4 in G3/G4 medulloblastoma samples that harbor both H3K27ac and H3K27me3 (AM) or just H3K27ac (A) peaks on the ZIC1 promoter. Biological sample size for G4—AM/A = 6/18 (24 total) and G3—AM/A = 3/11 (14 total). P values from two-tailed Mann–Whitney U test. Same whisker box plot parameters as c. g, Density plot summarizing H3K27ac versus H3K27me3 signal at H3K27ac and H3K27me3 peaks. Correlations between H3K27ac and H3K27me3 were calculated by Pearson correlation on merged peak coordinates. h, Method for inferring heterozygous SNPs using H3K27ac and H3K27me3; two mutually exclusive histone modification marks. i, Distribution of inferred heterozygous SNPs across H3K27ac and H3K27me3 libraries of four G4 samples and one SHH sample with H3K27ac and H3K27me3 peaks on the ZIC1 promoter.

The ZIC1 and ZIC4 genomic loci are separated by an interposed, shared, bidirectional promoter (Extended Data Fig. 2g). They are coregulated by a SE that is highly active across all four subgroups (Fig. 2b and Extended Data Fig. 2f,g). Both genes are highly expressed across all medulloblastoma subgroups as previously described20, particularly in the G4 (Fig. 2c and Extended Data Fig. 2h). We now describe a subset of G3 and G4 tumors that exhibit atypical hemizygous H3K27me3 deposition across the ZIC1/ZIC4 SE locus while showing a robust H3K27ac mark in trans on the other allele (Fig. 2d,e). This pattern was associated with reduced ZIC1/ZIC4 transcript levels (Fig. 2f) and was not recurrently observed in either SHH or WNT medulloblastoma (Fig. 2e). These two functionally opposing marks are usually mutually exclusive at the vast majority of loci, with the ‘H3K27ac–H3K27me3 hemizygous state’ being exceedingly rare (Fig. 2g). We hypothesized therefore that somatic repression of ZIC1 through acquisition of the ‘H3K27ac–H3K27me3 hemizygous state’ is a chromatin-based driver event in G4 medulloblastoma.

To determine if the H3K27ac and H3K27me3 are indeed found in trans on separate alleles within the same cells, allelic frequencies for dbSNP151 annotated heterozygous single-nucleotide polymorphisms (SNPs) were examined in our H3K27ac and H3K27me3 libraries for samples harboring the H3K27ac–H3K27me3 hemizygous state at the ZIC1/ZIC4 locus (Fig. 2h). While the G3 samples lacked heterozygous SNPs, all SNPs within the examined G4 samples exhibited a strong bias for distinct alleles in the H3K27ac versus H3K27me3 libraries (Fig. 2i), suggesting that the two chromatin marks occur in trans within single cells. Inferred SNPs were verified with matching whole-genome sequencing (WGS) data when possible (Extended Data Fig. 2i). While a plurality of G4 medulloblastomas alter activity of ZIC1 through genetic mutation, an additional nonoverlapping cohort (Supplementary Table 1) of G4 tumors reduce ZIC1/ZIC4 expression through uni-allelic chromatin variant repression mediated by H3K27me3 deposition, suggesting a convergence of mechanisms underlying ZIC1 alteration and that ZIC1 might be a LOF driver gene in G4 medulloblastoma.

Mono-allelic SEs regulate ZIC1/ZIC4 expression in G3/G4 medulloblastoma

Our observation that the ZIC1/ZIC4 locus undergoes recurrent repression in G4 medulloblastoma through hemizygous deposition of H3K27me3 on its SE prompted us to look for additional mono-allelic SEs in a cohort of 51 medulloblastoma tumors with matching H3K27ac ChIP–seq and WGS data (Fig. 3a). Mono-allelic SEs were rare in SHH medulloblastoma, although a number of further examples were identified for G3 and G4 medulloblastoma, including the known example of PRDM6 enhancer hijacking in G4 (Fig. 3a)14. Of the 19 G4 medulloblastoma samples harboring heterozygous SNPs at the ZIC1/ZIC4 SE locus (to allow assessment of heterozygosity), 9/19 tumors (47% of cases) exhibited a mono-allelic SE in keeping with the H3K27ac–H3K27me3 hemizygous state. A similar, albeit less frequent pattern, was observed in G3 medulloblastoma, but only very rarely in SHH medulloblastoma. Notably, samples with mono-allelic ZIC1/ZIC4 SE exhibit expression of ZIC1/ZIC4 mRNA predominantly from the H3K27ac allele (Extended Data Fig. 3a), in keeping with a bona fide repression effect of H3K27me3 deposition. Aside from the SE directly overlapping the ZIC1/ZIC4 locus, several other genomically proximate SEs that target ZIC1/ZIC4 were also identified to be recurrently mono-allelic (Extended Data Fig. 3b, c).

Fig. 3: ZIC1/ZIC4 exhibit mono-allelic expression patterns in G3 and G4.
figure 3

a, SEs that are recurrently (n ≥ 3 for G4 and n ≥ 2 for others) mono-allelic across different medulloblastoma subgroups. SEs that harbor SNPs (phased and pooled for each allele) that are heterozygous in WGS but homozygous (normalized allelic frequency ≥ 0.9) in H3K27ac ChIP–seq reads (same SNPs) from the same sample were defined as mono-allelic. Dot plots above each SE show differences in pooled allelic frequencies for heterozygous SNPs (allele A–B) in (1) H3K27ac reads from the SE (left) and (2) RNA-seq reads from the SE target gene (right). Matching samples are connected by lines between SE and RNA. b, Allelic frequency summary for heterozygous germline SNPs for ZIC1 and ZIC4 transcripts in RNA-seq within the validation cohort (251 samples with both WGS and RNA-seq data). Adjusted P values from two-tailed pairwise Fisher’s exact test. c, Whisker box plots summarizing ZIC1 and ZIC4 expression cross the medulloblastoma subgroups, but G3 and G4 are divided according to mono-allelic (mono) versus bi-allelic (bi) expression of ZIC1 or ZIC4. Biological sample size: G3_bi/G3_mono = 19/8, G4_bi/G4_mono = 24/44 and SHH/WNT = 93/24. P values from two-tailed Mann–Whitney U test. Center of box, median. Bounds of box, 25% and 75% percentile. Whiskers show minimum and maximum values within the 1.5× interquartile range. d, Mutational landscape of ZIC1 in G4 and SHH. e, Allelic frequency distribution for ZIC1 mutations in G4 (n = 3) and SHH (n = 2) samples from the assembled validation cohort. f, ZIC1 VAF obtained from published medulloblastoma RNA-seq data. P value from two-tailed Mann–Whitney U test.

We determined the mono-allelic expression pattern of ZIC1/ZIC4 in a validation cohort of 251 medulloblastomas with matching RNA-seq and WGS data, assembled by combining publicly available and newly generated datasets3,4,14,27,28. We found frequent mono-allelic expression in G3 and G4, but neither SHH nor WNT medulloblastomas (Fig. 3b). Indeed, 55% of G4 tumors (36/65) and 24% of G3 tumors (7/29) exhibit mono-allelic expression of ZIC1, and 48.5% (33/68) of G4 tumors and 18.9% (7/37) of G3 tumors have mono-allelic expression of ZIC4 (Fig. 3b and Extended Data Fig. 3d). In both G3 and G4, mono-allelic expression is associated with reduced expression of ZIC1/ZIC4, consistent with chromatin-based suppression (Fig. 3c). The importance of diminished, mono-allelic expression of the ZIC1/ZIC4 locus in medulloblastomas arising from the rhombic lip is underscored by humans who have hypoplastic cerebella (DWM) secondary to germline hemizygous deletions of ZIC1/ZIC4 (ref. 16). We conclude that haploinsufficiency of ZIC1 due to either germline or somatic events, with consequent diminished transcription, has critical effects on the biology of the rhombic lip, either in toto (DWM) or possibly in distinct somatic subclones (medulloblastoma).

ZIC1 is a presumed medulloblastoma driver gene that recurrently harbors SNVs in G4 and SHH medulloblastoma14. We now demonstrate that ZIC1 mutations in G4 medulloblastoma are found in the DNA-binding zinc finger ___domain, whereas SHH medulloblastoma SNVs are found in the 3′ end of the gene, encoding a carboxy-terminal intrinsically disordered region (IDR) of currently unknown function (Fig. 3d)14. Intriguingly, SHH medulloblastoma ZIC1 somatic mutations are found in the same 3′ region of the ZIC1 gene as previously reported germline GOF ZIC1 mutations in humans with craniosynostosis23. Within our 251 medulloblastoma validation cohort, three G4 tumors and two SHH tumors with ZIC1 mutations were identified. In all three G4 tumors, the variant allele frequency (VAF) of mutants comprised nearly 100% of all ZIC1 reads from RNA-seq, whereas they were below 50% in the matching WGS libraries (Fig. 3e). Conversely, SHH medulloblastoma mutants exhibited VAF near 50% in both WGS and RNA-seq reads. Examination of ZIC1 VAF from our published medulloblastoma RNA-seq cohort3,27 produced similar results (Fig. 3f). These data are consistent with a model in which G4 medulloblastomas acquire LOF genetic and chromatin variants, while SHH medulloblastomas acquire GOF variants.

Mono-allelic ZIC1 expression occurs in a subset of G4 medulloblastoma

PRDM6 overexpression secondary to a tandem duplication of the SNCAIP locus is a suspected G4 medulloblastoma driver gene14, and in our dataset it is found only in G4 tumors with mono-allelic expression of ZIC1 or ZIC4 (Fig. 4a and Extended Data Fig. 4a). G4 ZIC1/ZIC4 mono-allelic samples were significantly enriched (P = 0.0196) for mutations in chromatin modifiers including KDM6A, KMT2C and KMT2D (Fig. 4b). In G3, KMT2D mutation was significantly enriched (P = 0.0215) in ZIC1/ZIC4 mono-allelic samples (Fig. 4c,d). Conversely, KBTBD4 InDel mutations were enriched (P = 0.0041) in G3/4 ZIC1/ZIC4 bi-allelic samples (Fig. 4b,c). SHH tumors with ZIC1 mutations always co-occurred with mutations of the U1 splicing factor (Extended Data Fig. 4b), consistent with our previous publication in which ZIC1 mutations were found in SHHα and SHHδ tumors where U1 mutations occur27. Notably, we observe cases of G4 medulloblastoma with mono-allelic ZIC1/ZIC4 expression but without H3K27me3 deposition, suggesting that additional cryptogenic genetic/epigenetic routes to allelic silencing of ZIC1/ZIC4 exist (Fig. 4e–h). G3/G4 medulloblastoma tumors exhibit a spectrum of ZIC1 expression levels as well as differentiation signatures (Supplementary Table 14), with G4 medulloblastoma exhibiting higher levels of both (Extended Data Fig. 4c,d), potentially rehighlighting the known role of ZIC1 in cerebellar development29.

Fig. 4: ZIC1/ZIC4 mono-allelic and bi-allelic G3/G4 medulloblastomas enrich for distinct mutations.
figure 4

a, Whisker box plot of normalized PRDM6 transcript counts in bi-allelic versus mono-allelic ZIC1/ZIC4 G4 samples. PRDM6 transcription occurs exclusively in the context of single allele inactivation of ZIC1/ZIC4. b, Oncoplot showcasing mutation status of previously published recurrently mutated genes in mono-allelic and bi-allelic G4 samples. Each column represents different samples. Each row represents different genes that are recurrently mutated in medulloblastoma. Distinct types of mutations for a gene in each patient are depicted with different size/colored bars. c, Oncoplot showcasing mutation status of previously published recurrently mutated genes in mono-allelic and bi-allelic G3 samples. d, Sample distribution summary and two-tailed Fisher’s exact test outputs for the significance of enrichment for chromatin modifier mutations in ZIC1/ZIC4 mono-allelic G4 and G3, as well as KBTBD4 mutation in ZIC1/ZIC4 bi-allelic G4. e, Summary of different proportions of G4 medulloblastoma samples exhibiting transcriptional repression within the chromatin (H3K27me3) data or RNA (mono-allelic expression) data. f, Sequencing depth normalized bigwig tracks for H3K27ac and H3K27me3 in one G4 sample with bi-allelic ZIC1/ZIC4 SE and two G4 samples with mono-allelic ZIC1/ZIC4 locus SE. Not all G4 samples with mono-allelic ZIC1/ZIC4 SE harbor H3K27me3 peak on the locus. g,h, Allelic frequencies for heterozygous SNPs in WGS, H3K27ac and H3K27me3 ChIP–seq data in the two mono-allelic G4 samples: MDT-AP-1168, where H3K27me3 is observed, and MDT-AP-2673, where H3K27me3 is absent on the ZIC1/ZIC4 locus.

One possible explanation for the H3K27ac–H3K27me3 hemizygous state is that it occurs naturally during the differentiation of the rhombic lip subventricular zone (RL-SVZ), where G4 medulloblastoma is thought to arise2,3. However, hierarchical clustering of G3 and G4 medulloblastoma by both overall transcriptome or neuronal gene expression does not segregate tumors by ZIC1/ZIC4 expression status, suggesting that the observed repression of the ZIC1/ZIC4 locus from chromatin variants is not purely secondary to a transient developmental state in the RL-SVZ (Extended Data Fig. 4e,f). mono-allelic ZIC1/ZIC4 expression may also arise from local or distal mutations/structural variations affecting ZIC1/ZIC4 transcription. However, mutational mining of the region surrounding the ZIC1/ZIC4 locus for the presence of noncoding mutations that could account for the observed epigenetic repression failed to yield any likely candidates (Extended Data Fig. 4g,h). Taken together, we hypothesize that the acquisition of somatic mutations and/or aberrant activity of histone-modifying complexes may result in unusual regulation of the ZIC1/ZIC4 locus, although this concept remains largely speculative.

Opposing ZIC1/ZIC4 CNAs in G3/G4 versus SHH medulloblastoma

Previous studies have reported recurrent copy loss of chromosome 3q (chr3q), which contains the ZIC1/ZIC4 locus, in G4 medulloblastoma26,30. Examining CNAs at the ZIC1/ZIC4 locus using published SNP6 array data26 validates this finding and further showcases an intriguing pattern—the ZIC1/ZIC4 locus was recurrently deleted in G3/G4; however, the same locus exhibits recurrent genomic gains in SHH (Fig. 5a), as determined by GISTIC31, and pairwise comparison of CNAs across subgroups (Fig. 5b,c). Frequencies of chr3q deletions and focal deletions harboring the ZIC1/ZIC4 locus within G4 medulloblastoma were examined at the subtype level as we annotated previously30. These deletions exhibited subtype specificity, being notably depleted in G4β (Fig. 5d), whereas chromatin-based repression of the locus is very frequent in G4β (Fig. 5e). Tumors that target ZIC1 through either a genetic or a chromatin route show loss of heterozygosity at the level of mRNA (Fig. 5f,g). SHH samples affected by copy number gains exhibited concomitant increased expression of both ZIC1 and ZIC4 (Fig. 5h). SNP6 and expression array data26,30 demonstrate that G4γ samples with focal and broad deletions of the ZIC1/ZIC4 locus exhibit diminished expression of ZIC1 and ZIC4 transcripts as compared to balanced controls (Fig. 5i). Because the ZIC1/ZIC4 locus can be targeted by both genetic- and chromatin-based mechanisms, we examined the overall proportion of samples within the validation cohort medulloblastomas (251 tumors with RNA-seq and WGS) affected by either chromatin or genetic variants. We identified the copy number status for the ZIC1/ZIC4 locus within these samples using control-FREEC on the WGS data32. Annotating samples by ZIC1/ZIC4 allelic expression status, copy gain within SHH, copy loss within G3/G4 medulloblastoma and ZIC1 SNV status revealed that close to 20% of SHH samples harbor genetic variants promoting ZIC1/ZIC4 expression (Fig. 5j). Conversely, approximately 33% of G3 and 60% of G4 samples harbored genetic/epigenetic variants associated with repression of ZIC1/ZIC4 expression (Fig. 5j). These results are consistent with a model in which ZIC1, and possibly ZIC4, are LOF drivers in G4 medulloblastoma and GOF drivers in SHH medulloblastoma.

Fig. 5: ZIC1/ZIC4 locus exhibits distinct genomic rearrangements in G3/G4 and SHH medulloblastoma.
figure 5

a, CNA track for medulloblastoma samples exhibiting ZIC1/ZIC4 locus copy gain/loss. b, GISTIC output for SHH medulloblastoma, highlighting 2p24.3 (MYCN), 2q14.2 (GLI2) and 3q23 (ZIC1/ZIC4) gain. FDR, false discovery rate. c, CNA summary for the ZIC1/ZIC4 locus per medulloblastoma subgroups. Adjusted P values from two-tailed pairwise Fisher’s exact test. d, Chr3q and ZIC1/ZIC4 focal copy deletion frequency across three subtypes of G4 medulloblastoma. P values from two-tailed Fisher’s exact test and Hochberg correction. e, Breakdown of chromatin repression of a single allele of ZIC1/ZIC4 locus across three subtypes of G4 medulloblastoma. P values were calculated by two-tailed Fisher’s exact test followed by Hochberg multiple correction. f,g. Allelic frequencies for heterozygous germline SNPs across normal tumor DNA and tumor RNA from a representative G4 sample with (f) chr3q deletion and (g) epigenetic suppression of the ZIC1/ZIC4 locus. h, Whisker box plots for ZIC1 and ZIC4 expression in SHH medulloblastoma tumors with chr3 copy gain versus neutral. Expression values from RNA-seq data with matching SNP6 array data. P values were calculated from the two-tailed Mann–Whitney U test. Center of box, median. Bounds of box, 25% and 75% percentile. Whiskers show minimum and maximum values within the 1.5× interquartile range. i, Whisker box plots for ZIC1 and ZIC4 expression in G4γ medulloblastoma with chr3q copy loss versus copy neutral. Expression values from expression array data with matching SNP6 array data. Same statistical test and whisker box plot parameters as h. j, Breakdown of ZIC1/ZIC4 allelic expression pattern, ZIC1/ZIC4 CNA and ZIC1 SNVs in medulloblastoma samples with both RNA-seq and WGS data available, as well as harboring heterozygous germline SNPs in ZIC1/ZIC4 exons.

ZIC1/ZIC4 represses G3 medulloblastoma model growth in vitro and in vivo

Due to the lack of accurate, robust G4 medulloblastoma cell lines, we examined the functional importance of ZIC1/ZIC4 by overexpressing blue fluorescence protein (BFP) empty vector, ZIC1, ZIC4 or ZIC1 and ZIC4 together in D425 and D283 G3 medulloblastoma cell lines. Because G3 and G4 medulloblastomas are (1) molecularly similar and (2) exhibit highly similar genetic and epigenetic dysregulation of the ZIC1/ZIC4 locus, G3 medulloblastoma cell lines were considered relevant for these experiments. Overexpression of ZIC1 led to a significant reduction in the proliferative potential of D425 with evidence for some additive activity with ZIC4 (Fig. 6a). Similar results were observed for D283 in a cell proliferation assay (Fig. 6b,c). Overexpression of ZIC1/ZIC4 in G3 medulloblastoma lines followed by transcriptional profiling revealed increased expression of genes involved in neuronal differentiation, consistent with a model in which LOF of ZIC1/ZIC4 might hinder differentiation (Fig. 6d). Cerebellar xenografting of NOD SCID γ (NSG) mice with D425 cells overexpressing ZIC1/ZIC4 or BFP empty vector demonstrated a significant difference in both bioluminescence imaging (BLI) signal and survival (Fig. 6e–g). The patient-derived G3 xenograft, MB051, harbors single allele chromatin-based suppression of the ZIC1/ZIC4 locus (Fig. 6h,i and Supplementary Table 15). Restoring ZIC1/ZIC4 expression in MB051 significantly reduces BLI signal, as well as prolonging survival in vivo (Fig. 6j–m) in a setting with pre-existing ZIC1/ZIC4 chromatin repression. Upon endpoint, ZIC1 expression was minimal with the ZIC1/ZIC4 overexpression construct (but higher than an empty vector), suggesting a possible negative selection for cells highly expressing ZIC1 over time in vivo (Extended Data Fig. 5a). MB051 also exhibited upregulation of neuronal differentiation-associated genes with ZIC1/ZIC4 overexpression in vivo (Extended Data Fig. 5b–f), although morphological changes were not evident (Extended Data Fig. 6). Taken together, our results show tumor suppressive roles of genes in the ZIC1/ZIC4 locus, especially ZIC1.

Fig. 6: ZIC1/ZIC4 reduces G3 medulloblastoma cell proliferation both in vitro and in vivo.
figure 6

a, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) cell proliferation assay results (mean ± s.d.) for D425. Three biological replicates. P values from two-tailed Welch t-test. b, Cell proliferation assay results for D283. P values from two-tailed Welch t-test. Data points show mean ± s.d. Five biological replicates. Center of box, median. Bounds of box, 25% and 75% percentile. Whiskers show minimum and maximum values within the 1.5× interquartile range. c, Western blot validation of ZIC1/ZIC4 overexpression in D283 and D425. d, Pathway analysis for ZIC1/ZIC4 versus EV (BFP) overexpressing D425 (RNA-seq, biological n = 3). e,f. Representative images (e) and whisker box plots (f) summarizing BLI signals in BFP versus ZIC1/ZIC4 overexpressing D425-injected mice. P values were calculated by two-tailed Welch t-test. Same whisker box plot parameters as b. g, Survival curves for BFP versus ZIC1/ZIC4-transduced D425-injected mice. P values from two-tailed log-rank test. h, Normalized bigwig tracks showcasing chromatin state of ZIC1/ZIC4 locus in patient-derived G3 xenograft line MB051. i, Allelic frequency of heterozygous SNP rs6766244 on coding exon of ZIC4 from MB051 RNA-seq and H3K27me3 ChIP–seq counts, and Sanger sequencing result from the tumor DNA for the same SNP. j, ZIC1/ZIC4-normalized counts from RNA-seq in MB051 (biological n = 3 for EV and ZIC1/ZIC4 constructs). Mean ± s.d. k,l. Representative images (k) and whisker box plots (l) summarizing BLI signals in BFP versus ZIC1/ZIC4 overexpressing MB051-injected mice. P values were calculated by two-tailed Welch t-test. Same whisker box plot parameters as b. m, Survival curves for BFP versus ZIC1/ZIC4-transduced MB051-injected mice. P values from two-tailed log-rank test. H3, histone 3; EV, empty vector.

Source data

SHH and G4 medulloblastoma ZIC1 mutants exert opposite phenotypes

As the CNAs in SHH (gain) and G4 (deletion) are diametrically opposed, we hypothesized that the SHH medulloblastoma SNVs would have divergent biological activity compared to G4 medulloblastoma SNVs, consistent with GOF and LOF phenotypes, respectively. To test this hypothesis, we generated ZIC1 expression constructs with mutations from G4 medulloblastoma (G4 medulloblastoma ZIC1 mutants) in the zinc finger regions or with mutations from SHH medulloblastoma (SHH medulloblastoma ZIC1 mutants) in the carboxy terminus IDR (Fig. 7a). Consistent with our hypothesis, cell proliferation assays in D425 and cell competition assays in D283 demonstrated a reduced antiproliferative effect for the G4 medulloblastoma ZIC1 mutants compared to the wild-type (WT) ZIC1, whereas SHH medulloblastoma ZIC1 mutants exhibited even more profound growth repression (Fig. 7b–d). We noted marked overexpression after Western blotting for SHH medulloblastoma ZIC1 mutant proteins as compared to WT controls or G4 medulloblastoma ZIC1 mutant proteins (Fig. 7e,f). Cycloheximide pulse-chase assays demonstrated that SHH medulloblastoma ZIC1 mutant proteins exhibit significantly higher protein stability, as compared to WT ZIC1, or G4 medulloblastoma ZIC1 mutant proteins, suggesting that the carboxy terminus IDR exerts control over the stability of the ZIC1 protein (Fig. 7g,h). Overexpression of G4 medulloblastoma ZIC1 mutant constructs in G3 medulloblastoma cell lines leads to tenfold fewer upregulated genes, as compared to WT ZIC1, whereas overexpression of the SHH medulloblastoma ZIC1 mutant constructs resulted in more differentially expressed genes as compared to WT controls (Fig. 7i,j and Extended Data Fig. 7a–c). WT ZIC1 overexpression led to activation of pathways involved in development and organogenesis, which was dampened with the G4 medulloblastoma ZIC1 mutants but further augmented with the SHH medulloblastoma ZIC1 mutants (Extended Data Fig. 7d–f). ChIP–seq against Flag-ZIC1 demonstrates reduced DNA-binding affinity of G4 medulloblastoma ZIC1 mutant proteins, offering a mechanistic insight underlying the reduction of ZIC1 target gene induction (Fig. 7k and Extended Data Fig. 7g). As the G4 medulloblastoma ZIC1 point mutations occur in the DNA-binding ___domain, we conclude therefore that loss of DNA binding is at least partially responsible for the phenotype of G4 medulloblastoma ZIC1 mutants.

Fig. 7: ZIC1 mutations from G4 and SHH medulloblastoma are functionally distinct.
figure 7

a, AlphaFold2 predicted structure of ZIC1. Mutant constructs generated and used in the study are summarized in the structure. b, Proliferation assay for D425 G3 cell line transduced with ZIC1 mutant constructs and mCherry EV. Three technical replicates for each construct. Mean ± s.d. P values from two-tailed Welch t-test. c, Schematic representation for the cell competition assay using D283. d, Cell competition assay results using D283 transduced with ZIC1 mutant constructs and mCherry EV. Three technical replicates for each construct. Mean ± s.d. P values from two-tailed Welch t-test. e, Representative western blot visualization of exogenous ZIC1 expression in D283 transduced with FLAG-ZIC1 constructs. f, Whisker box plots showing exogenous ZIC1 expression in D283 transduced with FLAG-ZIC1 constructs. Signals were normalized by transduction efficiency and GAPDH levels. Center of box—median. Bounds of box—25% and 75% percentile. Whiskers show minimum and maximum values within the 1.5× interquartile range. P values from two-tailed Welch t-test. g, Representative cycloheximide chase results for WT and mutant ZIC1 constructs in D283. h, Comparison of ZIC1 protein level across varying exposure times to cycloheximide for WT (n = 2), G4 medulloblastoma mutant (n = 4) and SHH medulloblastoma ZIC1 mutant (n = 4) constructs. n, biological replicates. Mean ± s.d. P values from two-tailed Welch t-test. i, Number of DEG (DESeq2 output) for ZIC1 constructs when compared against EV or WT ZIC1. Q value cutoff of 0.05. j, Volcano plot summarizing differentially expressed genes between WT ZIC1 and EV. k, Distribution of normalized reads from FLAG ChIP–seq peaks from FLAG-tagged WT versus G4 medulloblastoma mutant ZIC1-transduced D283. DEG, differentially expressed genes.

Source data

ZIC1 is a GOF driver in SHH medulloblastoma

Contrary to ZIC1 suppressing the growth of G3 medulloblastoma, we hypothesized that ZIC1 would promote the growth of SHH medulloblastoma. Indeed, overexpression of ZIC1 constructs in mouse granule neuron progenitor (GNPs) cells (the cell of origin for SHH medulloblastoma)10,12 results in increased cellular proliferation, which was more pronounced with the SHH medulloblastoma ZIC1 mutants as compared to WT ZIC1 or G4 medulloblastoma ZIC1 mutants (Fig. 8a,b). Cycloheximide chase in GNPs transduced with ZIC1 mutant constructs revealed that SHH medulloblastoma ZIC1 mutants also increase protein stability in GNPs, demonstrating the conservation of mutant mechanism across different cell types (Fig. 8c,d). ZIC1 ChIP–seq in GNPs transduced with ZIC1 mutant constructs also demonstrated reduced DNA-binding affinity for G4 medulloblastoma ZIC1 mutants similar to results observed in D283 (Extended Data Fig. 8a,b). Transduction of GNPs with ZIC1 constructs promoted higher expression of cell cycle pathway genes as well as Gli2, the main effector of SHH signaling (Fig. 8e–g and Extended Data Fig. 8c,d)14,26. Gli2 is a known oncogene for SHH medulloblastoma, which exhibits a highly SHH medulloblastoma-enriched expression pattern as well as ZIC1-binding motif enrichment in its promoter (Extended Data Fig. 8e and Supplementary Table 16). Re-analysis of published datasets33 demonstrates that Zic1 binds the Gli2 promoter in the mouse cerebellum and that loss of Zic1 is associated with diminished expression of Gli2 (Extended Data Fig. 8f–h). These data are consistent with a model in which ZIC1 expression represses cell growth in maturing unipolar brush cell (UBC) progenitors of the RL-SVZ (origin of G4 medulloblastoma)2,3, whereas it promotes growth of GNPs (origin of SHH medulloblastoma) in the developing cerebellar external granule layer (EGL). In the mouse, after the generation of eomesodermin (EOMES)+ excitatory deep cerebellar nuclear neuron committed cells at E10.5–E12.5 (refs. 34,35), the RL-SVZ arises as a bipotent progenitor zone capable of producing both GNPs and UBCs from E13.5 (refs. 35,36). Publicly available data on developing human cerebellum3,37, as well as newly generated RNA-scope results, demonstrated that both ZIC1 and ZIC4 are highly expressed in UBC progenitors of the RL-SVZ (Extended Data Fig. 9a–g). The genetic and chromatin variants of ZIC1 and ZIC4 in G4 and SHH medulloblastoma suggest a model in which the activity of ZIC TFs has context-dependent roles in UBC and granule neuron lineage cells, which cumulatively constitute the majority of the neurons in a human brain (Fig. 8h,i).

Fig. 8: ZIC1 is a GOF driver oncogene in SHH medulloblastoma.
figure 8

a, Schematic representation summarizing GNP 5-ethynyl-2'-deoxyuridine (EdU) proliferation assay. FACS, fluorescence activate cell sorting. b, Summary of EdU proliferation assay for GNP transduced with ZIC1 mutant constructs and mCherry EV. GNPs enriched from multiple mouse cerebellums were used to generate biological triplicates for each construct. Mean ± s.d. as error bars. P values were calculated by two-way ANOVA. c, Representative results from two independent replicates from running cycloheximide (CHX) chase on GNP transduced with WT ZIC1 construct, two G4 medulloblastoma ZIC1 mutant constructs and two SHH medulloblastoma ZIC1 mutant constructs. d, Comparison of ZIC1 protein level from GNP across varying exposure times to cycloheximide for WT (n = 2), G4 medulloblastoma mutant (n = 4) and SHH medulloblastoma ZIC1 mutant (n = 4) constructs. n, biological replicates. Mean ± s.d. P values were calculated by two-tailed Welch t-test. e, RNA-seq-derived volcano plot summarizing DEG (DESeq2 output) between ZIC1 (mch+ ZIC1+) and EV (mch+) transduced granule cells. Two biological replicates were generated for bulk granule cells and sorted GNPs (biological n = 4). Q value cutoff of 0.05. f, Normalized RNA-seq counts for Gli2 transcript in EV (mch+) and ZIC1 (mch+ ZIC1+) transduced GNPs. Adjusted P value from differential expression was calculated from DESeq2 differential expression analysis. g, Top ten pathways upregulated with ZIC1 overexpression in bulk granule cells and GNPs. h,i, Summary of normal rhombic lip development (h) as well as epigenetic and genetic events (i) that lead to ZIC1 LOF in G3 and G4 medulloblastoma and ZIC1 GOF in SHH medulloblastoma. ANOVA, analysis of variance; NSC, neural stem cell.

Source data

Discussion

G3 and G4 medulloblastoma are molecularly distinct medulloblastoma subgroups that are highly related to each other and share many oncogenic drivers38. We report similar ZIC1 LOF phenotypes manifesting in G3 and G4 (epigenetic suppression, copy deletion and LOF mutation), albeit at different proportions, suggesting that the ZIC1/ZIC4 locus has similar roles within each subgroup and possibly within their cells of origin. On the other hand, while SHH medulloblastoma shares a direct developmental relationship with G4 medulloblastoma, ZIC1/ZIC4 events confer a GOF phenotype. These findings suggest that ZIC1/ZIC4 has opposing roles in G3/G4 medulloblastoma versus SHH medulloblastoma, raising the possibility that these genes may also have distinct roles in the cells of origin for these similar but distinct tumor types.

Our genetic and experimental data provide robust support for a model in which LOF mutations/chromatin variants in the ZIC1/ZIC4 locus promote G4 medulloblastoma, while GOF mutations promote SHH medulloblastoma within the different lineages of the rhombic lip. ZIC1 events in the current cohort are found in 20% of SHH medulloblastoma and 60% of G4 medulloblastoma, making ZIC1 one of the most frequently affected driver genes in medulloblastoma biology. While ZIC4 is coregulated with ZIC1 through recurrent epigenetic suppression and copy number changes, the functional role of ZIC4 in G3 medulloblastoma cell lines is minimal compared to that of ZIC1. Furthermore, somatic point mutations have only been identified for ZIC1 and not for ZIC4. As such, we predict that ZIC1 has a more dominant role in medulloblastoma tumorigenesis, with ZIC4 potentially providing some additive effects.

Our discovery of a H3K27me3/H3K27ac heterozygous chromatin state in G4 medulloblastomas at the ZIC1/ZIC4 locus demonstrates a convincing complementation group in which some tumors achieve repression of ZIC1 through deletion or somatic mutations of genomic DNA, while other tumors reach the same phenotype through chromatin variants that impose epigenetic repression. This may be through somatic acquisition of chromatin variants, akin to de novo allele-specific ‘epimutations’ that have been described to be associated with oncogenesis39,40. Indeed, this robust complementation group provides strong evidence for the biological importance of somatic chromatin variants in the pathogenesis of cancer. We suggest that the observed chromatin events drive the clonal selection of tumor cells and are not merely passenger events.

We were unable to use current technologies to identify local or distal cryptic noncoding mutations driving the H3K27me3/H3K27ac heterozygous chromatin state, although we acknowledge that these may occur and be currently cryptogenic. It is also possible that there exists a minor unidentified population in the rhombic lip that is temporally or anatomically restricted and passes through a state with the H3K27me3/H3K27ac heterozygous chromatin state, and that these particular cells are at increased risk for transforming to G4 medulloblastoma. An additional possible mechanism is somatic ‘epimutation’, in which aberrant H3K27me3 marks repress ZIC1 expression, and this heritable chromatin state results in clonal expansion and eventually G4 medulloblastoma. The consistent co-occurrence of somatic mutations of histone lysine modifier genes in G4 medulloblastomas that also harbor somatic chromatin variants of ZIC1 is consistent with a model in which aberrant control of the epigenome leads to ‘epigenetic instability’, with clones that by error contain ZIC1 silencing chromatin events undergoing clonal selection. Similarly, it has been previously shown that succinate dehydrogenase deficiency can induce aberrant epigenetic remodeling mono-allelically41. Which of the three outlined mechanisms, or mechanisms not currently suspected, is responsible for the H3K27me3/H3K27ac heterozygous chromatin state is, however, not currently known, nor readily determined using current technologies, although we favor the somatic chromatin variant model.

G4 medulloblastoma comprises cells similar to the UBC progenitors within RL-SVZ, while SHH medulloblastoma cells resemble GNPs of the EGL. These highly related cell types likely arise from the same bipotential progenitors. The clear difference between the LOF phenotypes (G4) versus GOF phenotypes (SHH) suggests a model in which ZIC1 and/or ZIC4 have context-dependent roles in UBC progenitors and GNP during rhombic lip development. In GNPs, ZIC1/ZIC4 may work in conjunction with other SHH pathway genes, such as GLI2, to promote cell proliferation and granule-cell-like transcriptome. Tight regulation of ZIC1/ZIC4 activity is likely critical to prevent overexpansion of GNPs during EGL formation. Conversely, UBC progenitors likely require higher levels of ZIC1/ZIC4 activity for normal differentiation, as shown by the UBC lineage-enriched ZIC1/ZIC4 expression pattern. Perturbation of ZIC1/ZIC4 activities in these different contexts likely contributes to improper rhombic lip development and favors oncogenic transformation, where LOF genetic/chromatin variants promote the transformation of the UBC progenitors and GOF variants promote the transformation of the GNPs.

We maintain that LOF/GOF mutations of ZIC1 are true driver events, as overexpression of ZIC1 represses malignant phenotypes in G3 medulloblastoma models while promoting malignancy in SHH medulloblastoma precursor cells, both in vitro and in vivo. Indeed, our data support a model in which ZIC1 is the paramount example of a context-specific cancer driver gene, as it appears to show diametrically opposing biological activity in these two different cell types that arise from the exact same progenitors and which occur on either side of a very specific cell fate decision during rhombic lip development.

Methods

Research ethics board (REB)

This study obtained full ethics approval from the Hospital for Sick Children (REB 0020020238 and REB 1000055059) as well as McGill University Health Centre (REB MCH003-26). All materials were collected after receiving written informed consent from patients, including consent to publish the generated data. All primary sample collection and experimental procedures (in vitro and in vivo) were done in accordance with guidelines from the REB of Hospital for Sick Children (REB 0020020238 and REB 1000055059), McGill University Health Centre (REB MCH003-26) and the Centre for Phenogenomics (AUP 22-0151H).

Experimental model and subject details

Primary tumor collection

Primary tumors used in the study were obtained from the Medulloblastoma Advanced Genomics International Consortium and International Cancer Genome Consortium. All materials were collected after receiving written informed consents, including consent to publish the generated data, as per guidelines from REB from the following institutes: Agostino Gemelli University Hospital, Children’s Hospital of Minnesota, Cooperative Human Tissue Network, David Geffen School of Medicine at University of California Los Angeles, Duke University, Emory University, Erasmus University Medical Centre, German Cancer Research Centre (DKFZ), Hospital Cantonal De Geneve, Hospital Infantil de Mexico Federico Gomez, Hospital Sant Joan de Deu, Ludwig Maximilans University, Masaryk University, McGill University, McMaster University, Memorial Sloan Kettering Cancer Centre, Miami Children’s Hospital, Portugese Cancer Institute, Queensland Children’s Tumor Bank, Seattle Children’s Hospital Fred Hunchinson Cancer Research Centre, Seoul National University Children’s Hospital, Stanford University School of Medicine, the Chinese University of Hong Kong, Tohoku University, University of California San Francisco, University Health Network, Universitats Kinderklinik, Universite de Lyon, University of Arkansas, University of Calgary, University of Debrecen Medical and Health Science Centre, University of Pittsburgh, University of Ulsan Asan Medical Centre, University of Warsaw Children’s Memorial Health Institute, Vanderbilt Medical Centre and Wolfson Children’s Hospital. Statistical methods were not used to predetermine the sample size. Age, sex, subgroup and subtype information for used tumors are available in Supplementary Table 1. Primary tumor tissues were snap-frozen in liquid nitrogen and stored at −80 °C until use.

Mouse housing and husbandry

All mouse breeding and procedures were performed as approved by the Toronto Centre for Phenogenomics.

Method details

G3 medulloblastoma cell lines and xenograft line

D425 and D283 cell lines were derived at Duke University (Supplementary Table 2) and verified with short tandem repeats before being used for experiments. MB051 patient-derived xenograft line was generated at the Hospital for Sick Children and passaged only by serial intracranial injection in NSG mice without expansion in vitro.

Source of NOD-SCID-IL2Rγ null mice

NOD-SCID-IL2Rγ null (NSG) mice were obtained from the Toronto Centre for Phenogenomics in-house breeding colony.

Intracranial injection of G3 medulloblastoma tumor cells

Intracranial injection was performed on NSG mice (age range of 6–10 weeks, ~50% males and females for all conditions) using D425 and MB051 xenograft lines as previously described42 using slightly modified stereotactic coordinates—2 mm posterior to λ, 1 mm lateral and 2 mm deep. In total, 2,000 Green fluorescent protein luciferase-tagged D425 cells transduced with BFP empty vector or ZIC1/ZIC4 vector were injected per mouse. In total, 4,000 GFP luciferase-tagged MB051 cells transduced with BFP empty vector or ZIC1/ZIC4 vector were injected per mouse. Humane endpoint was called independently by staff at the Toronto Centre for Phenogenomics based on physiological conditions exhibited by the injected mice. These staff were blinded from construct information. Mice that did not exhibit any BLI signal above the background (2.5 × 104 p s−1 cm2 sr−1) by the third week after injection were excluded from the cohort.

Bioluminescence measurement

Bioluminescence was measured in NSG mice injected with GFP Luciferase-tagged tumor cells as previously described42. For D425, measurements were taken on week 1 (6–7 days after injection), week 2 (13–14 days after injection) and week 3 (20 days after injection). For MB051, measurements were taken on week 1 (7 days after injection) and week 2 (14 days after injection).

RNA-scope on developing human cerebellum slides

Manufacturer-recommended protocols were used for RNA-scope in situ hybridization (ISH) assays as previously described37 using RNA-scope 2.5 High Definition-RED Assay (ACDBio, 322350). Briefly, RNA-scope was performed on mid-sagittal sections of the developing vermis, fixed in 10% formalin for 4 weeks. Manufacturer-recommended protocols (ACDBio/Bio-Techne) were used to assay the following probes: Hs-ZIC4 (525661) and Hs-ZIC1 (542991). All sections were counterstained with hematoxylin or methyl green. Stained slides were imaged using the Nanozoomer Digital Pathology slide scanner (Hamamatsu).

ZIC1 mutant construct generation

WT ZIC1 was cloned into pCDH-mCherry or pCDH-GFP empty lentiviral vector using the In-Fusion Snap Assembly Starter Bundle (Takara). Mutagenesis, or N-terminal FLAG tagging of ZIC1, was also done using the In-Fusion kit.

Isolation of cerebellar granule cells or GNPs

Cerebellar cells were isolated from the cerebellum as described previously43. Briefly, cerebellum from postnatal day 5 (P5) mice was digested with high glucose Dulbecco’s Phosphate Buffered Saline (DPBS) (Thermo Fisher Scientific) containing 10 U ml−1 papain (Worthington), 200 μg ml−1 l-cysteine and 250 U ml−1 DNase (Sigma) for 30 min. Tissue was triturated to obtain a single-cell suspension and then centrifuged through a 35% and 65% Percoll gradient (Sigma). Cells in the layer between 35% and 65% Percoll were washed once with DPBS containing 0.02% BSA and resuspended in GNP culture medium (neurobasal supplemented with B27 (50×), sodium pyruvate (100×), penicillin–streptomycin (100×) and glutamax (100×)). Granule cells or GNPs were enriched by depleting the adherent cells through two incubations in poly-D-lysine(PDL)-coated plates for 20 min each time. Enriched granule cells and GNPs were cultured with GNP culture medium supplemented with 3 μg ml−1 SHH (Peprotech) in PDL-coated plates. For the isolation of pure GNPs, cerebellar cells were isolated from Atoh1-GFP mice at P5 as described above. After washing once with DPBS containing 0.02% BSA, cells were suspended with DPBS containing 5% FBS (Thermo Fisher Scientific). GNPs with strong GFP expression (~40%) were sorted and cultured with the GNP culture medium as described above.

5-ethynyl-2′-deoxyuridine (EdU) assay in GNPs

GNPs isolated from P5 Atoh1-GFP mice, as described above, were infected with control (pCDH-mCherry) or ZIC1 viruses (pCDH-mCherry_ZIC1 WT/mutants) in triplicates. Cells were cultured in a GNP culture medium with SHH in PDL-coated 48-well plates. At each time point, cells were treated with 10 μM 5-ethynyl-2′-deoxyuridine (EdU) for 6 h and then dissociated for EdU staining (Click-iT Plus EdU Pacific Blue Flow Cytometry Assay Kit) and flow cytometry analysis. For data analysis, cells were first gated for mCherry+ cells. The percentage of proliferating cells (EdU+) was then calculated for each sample.

Quantification and statistical analysis

ChIP–seq data processing

Raw ChIP–seq reads were aligned to hg19 genome assembly using bowtie2 (v2.2.1)44. PCR duplicates were removed using Picard MarkDuplicates. Reads with mapping quality lower than 20 were removed. Reads from nonchromosomal contigs, mitochondria or ENCODE blacklist regions were also filtered out before peak calling. H3K27ac peaks were identified using MACS2 (v2.1.1.20160309) with the following code: MACS2 callpeak -t IP_bam_file -f BAMPE -g hs --nomodel -B -q 1e-2 (ref. 45). H3K27me3 peaks were identified using the following parameters: MACS2 callpeak -t 27me3_IP_bam_file -c input_bam_file -f BAMPE -g hs --nomodel --broad -B -q 1e-5–broad-cutoff 1e-4. Peaks that could not be identified in at least two primary medulloblastomas were excluded from any further analysis. Library sizes for samples in H3K27ac and H3K27me3 samples were calculated using SAMtools46 and average fragment sizes of three different batches of H3K27ac and H3K27me3 were evaluated by deeptools47 (v3.1.3). H3K27ac and H3K27me3 peaks in each sample were annotated according to their closest genes and then categorized into different classes based on their distributions over different types of features, for example, promoter, exon, intron and distal intergenic. The distance between peaks and their assigned genes was calculated by using the center of the peak and the transcription start site as coordinates.

For ChIP–seq data from D283 cells transduced with FLAG-tagged ZIC1 constructs, peaks were called using Q value threshold of 1 × 10−5. For ChIP–seq data from GNP cells transduced with FLAG-tagged ZIC1 constructs, peaks were called using a Q value threshold of 0.05.

SNP inference from ChIP–seq libraries

For samples harboring both H3K27ac and H3K27me3 peaks on the ZIC1/ZIC4 locus, ‘H3K27ac–H3K27me3 hemizygous region’ was defined for each sample with bedtools (v2.27) intersect on the called peaks48. From the bivalent region containing the ZIC1/ZIC4 locus, allelic frequencies were calculated for each dbSNP151 annotated heterozygous SNP positions from H3K27ac and H3K27me3 library reads using bedtools multicov. Heterozygous SNPs were identified by first calculating allelic frequency r = absolute value of (reference (REF) alternate (ALT) allelic frequency). Afterward, SNPs with r ≥ 0.6 in both H3K27Ac and H3K27me3, but biased for different alleles in each, were used to infer heterozygous SNPs (ex, H3K27ac enriched for REF allele and H3K27me3 enriched for ALT allele). Alternatively, SNPs with r < 0.6 in either H3K27ac or H3K27me3 libraries were also used to identify SNPs. Only SNPs that are supported by at least ten reads from each library were used.

SEs analysis and subgroup consensus peak sets

SEs were defined using the Rank Ordering of Super Enhancers (v0.1) algorithm using H3K27ac peaks as input49. For all samples, the stitching distance was fixed at 12.5 kb to facilitate comparisons between samples. All other parameters used the default setting. Once SEs were generated for each sample, SEs were merged from samples within the same subgroup using GenomicRanges Bioconductor package50. Only SEs that were present at least two times per subgroup were considered for merging.

RNA-seq data processing

Custom hs37d5 genome assembly generated in previous study27 was used to align raw RNA-seq reads using STAR aligner (2.7.4) with the following parameters: --outFilterMultimapNmax 20 --alignSJoverhangMin 8 --alignMatesGapMax 200000 --alignIntronMax 200000 --alignSJDBoverhangMin 10 --alignSJstitchMismatchNmax 5 -1 5 5 --outSAMmultNmax 20 --twopassMode Basic51. Gene expression level was quantified using HTSeq (0.6.0) based on Gencode v19 annotations with the argument ‘-stranded reverse -m union’52. Differential gene expression analysis between subgroups was performed using the R Bioconductor package DESeq2 (v1.26.0)53. An adjusted P value of 0.05 was used for differentially expressed gene identifications.

H3K27ac HiChIP data process and loop call

Raw HiChIP reads were aligned using bowtie2 (2.3.4) and HiC-pro (2.9.0) using the default parameters in HiC-pro54. Output directory was used as input for hichipper (v0.7.3) to call significant loops using the following parameters: min-dist 5000, max-dist 20000000, read-length 150, ‘macs2-string -q 0.01 --extsize 315 –nomodel’55. Intrachromosomal loops with Q value less than 0.01 and read counts greater than 5 were used for downstream enhancer gene interactome analysis.

WGS data processing and germline variants calling

WGS data were aligned to the ‘hs37d5’ reference genome from 1000 Genomes Project Phase II as previously described28, using Burrows–Wheeler aligner–MEM (v0.7.8) with the ‘-T 0’ parameter56. For germline variant call, variants identified in both normal and tumor DNA from Platypus (v0.8.1) run with default parameters were used (https://github.com/andyrimmer/Platypus). To have the final heterozygous SNP list for each sample in WGS data, we only selected those passed Platypus quality control (minBaseQual and minMapQual: 20; alleleBias and strandBias: 0.001 and badReadsWindow: 11). Second, we retained SNPs with allele depth in tumor samples ≥10, allele depth in paired blood samples ≥7, allele ratio in blood between (0.3, 0.7) and allele ratio in tumor between (0.2, 0.8). Third, only bi-allelic sites and InDels shorter than three nucleotides were used. The final heterozygous SNP candidates were retained in the following allele imbalance analysis. We used EAGLE2 for haplotype phase estimation on bcftools (v1.9)57 normalized variants, using a phased reference panel in 1000 Genomes Project58.

Affymetrix SNP6 array data processing

SNP6 Affymetrix array data were mapped to hg19 and processed using Affymetrix Power Tools (v1.18.2) as previously described27.

Identification of focal recurrent CNAs from SNP6 array

To identify recurrent focal copy gains and losses for each subgroup, SNP6 array-derived segmentation files were used as input for GISTIC2 (v2.0.23) from gene pattern with the following options: refgene file = Human_Hg19.mat, maxspace = 10,000, gene gistic = yes, confidence = 0.90, Q value threshold = 0.25, run broad analysis = no, max sample segs = 10,000, arm peel = yes, gene collapse method = extreme, amplification threshold = 0.5, deletion threshold = −0.5, focal length cutoff = 0.5, armlevelpeel = on, confidence level = 0.95, Q value = 0.25, run broad analysis = no, max sample segs = 10,000 (ref. 31). Other parameters were left as default.

Single-cell RNA-seq (scRNA-seq) data analysis

Publicly available scRNA-seq data were analyzed as previously described with minor modifications3,59. Specifically, RL-SVZ cells from the glutamatergic lineage cells were further divided into three smaller cell clusters using the following criteria: RL-SVZ (KI67 high, EOMES+)—RL-SVZ residing UBC progenitor cells; RL-SVZ (KI67 high, ATOH1+)—RL-SVZ cells more committed to GCP lineage; RL-SVZ (KI67 low, EOMES+)—RL-SVZ residing UBC progenitor cells likely mixed with some early UBC.

Pathway enrichment analysis

Enriched pathways for differentially expressed genes were identified by using g-profiler at default parameters, using Q value threshold of 0.05 (ref. 60). Gene Ontology-biological term outputs were used for the final list of pathways. Top ten enriched/depleted pathways were identified for ZIC1 mutant construct experiments using G3 medulloblastoma cell lines or GNP cells in vitro and G3 medulloblastoma xenograft experiments in vivo.

Calling CNA events from WGS data

Copy number information was derived from WGS data using Control-FREEC (v10.3)32 as previously described with the following parameters: breakPointType = 4, ploidy = ‘2,3,4’, step = 10,000, window = 50,000 (ref. 28).

Before focal CNA call from WGS data for known medulloblastoma driver genes, ploidy for all WGS samples was predicted with Control-FREEC. For samples with inferred ploidy greater than 3.5, pileup ratio was used from ploidy = 4 output. All other samples used pileup ratio from ploidy = 2 output. Median ratio values for each segmented genomic locus were used to generate a segmented (.seg) format for each sample. Merged seg file for each subgroup was used as input for GISTIC2 (v2.0.23) from gene pattern with the following options: refgene file = human_Hg19.mat, maxspace = 10,000, gene gistic = yes, confidence = 0.90, Q value threshold = 0.25, run broad analysis = no, max sample segs = 10,000, arm peel = yes, gene collapse method = extreme, amplification threshold = 0.25, deletion threshold = −0.25, focal length cutoff = 0.5, armlevelpeel = on, confidence level = 0.95, Q value = 0.25, run broad analysis = no, max sample segs = 10,000 (ref. 31). Other parameters were left as default. Output from focal_data_by_genes was used for genes previously identified to undergo recurrent CNA gain in G3/G4—MYC, MYCN, OTX2 and CDK6, which have been previously reported14,26.

For CNA identification from WGS data for the ZIC1/ZIC4 locus, both broad chromosomal events and focal CNA were identified using the seg files generated above. An amplification threshold of 0.25 and a copy loss threshold of −0.25 were used to estimate the proportion of samples with copy number changes in SHH or G3/G4 samples, respectively.

Oncoplot generation

Highly expressed genes were identified by performing k-means clustering on size factor normalized RNA-seq counts with k = 2 for the following genes: GFI1, GFI1B and PRDM6. Group with higher expression of genes were categorized as highly expressing. Somatic SNVs, InDels, CNA amplifications and high expression samples for each gene were annotated for all samples using complexheatmap (v2.2.0) R package61.

Statistics and reproducibility

No statistical method was used to predetermine the sample size. Randomizing and blinding were not used for the experiments. For experiments involving the injection of mice with medulloblastoma cell lines or patient-derived xenograft lines, independent staff at the Toronto Centre for Phenogenomics were blinded from the experimental arm conditions before calling the endpoints. For mouse BLI experiments, mice that failed to reach the minimal detectable signal of 2.5 × 104 p s−1 cm2 sr−1 by the third week postinjection were removed from the cohort (failure to engraft).

Reporting summary

Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.