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:

Comprehensive multi-omic profiling of somatic mutations in malformations of cortical development

Abstract

Malformations of cortical development (MCD) are neurological conditions involving focal disruptions of cortical architecture and cellular organization that arise during embryogenesis, largely from somatic mosaic mutations, and cause intractable epilepsy. Identifying the genetic causes of MCD has been a challenge, as mutations remain at low allelic fractions in brain tissue resected to treat condition-related epilepsy. Here we report a genetic landscape from 283 brain resections, identifying 69 mutated genes through intensive profiling of somatic mutations, combining whole-exome and targeted-amplicon sequencing with functional validation including in utero electroporation of mice and single-nucleus RNA sequencing. Genotype–phenotype correlation analysis elucidated specific MCD gene sets associated with distinct pathophysiological and clinical phenotypes. The unique single-cell level spatiotemporal expression patterns of mutated genes in control and patient brains indicate critical roles in excitatory neurogenic pools during brain development and in promoting neuronal hyperexcitability after birth.

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: Comprehensive genetic profiling and validation of somatic variants in 283 MCD patients.
Fig. 2: Genes mutated in MCD highlight four major gene networks.
Fig. 3: Selected new MCD somatic variants show functional defects in embryonic mouse brain and patient samples.
Fig. 4: Clinical phenotypic outcomes correlate with genotype-based classifications in MCD.
Fig. 5: Single-nucleus transcriptomes reveal MCD gene enrichment in radial glia and excitatory neurons in the developing human cortex.
Fig. 6: Single-nucleus transcriptomes showed MCD gene expression enriched in MCD-specific cell types.

Similar content being viewed by others

Data availability

WES and AmpliSeq data are deployed on the NIMH Data Archive under study number 1484 ‘Comprehensive multi-omic profiling of somatic mutations in malformations of cortical development’ and SRA under accession number PRJNA821916: ‘Comprehensive multi-omic profiling of somatic mutations in malformations of cortical development’. The BSMN neurotypical brain data are available at NIMH Data Archive (NDA study 644, 792 and 919, https://nda.nih.gov/study.html?tab=result&id=644, https://nda.nih.gov/study.html?tab=result&id=792 and https://nda.nih.gov/study.html?tab=result&id=919) and SRA PRJNA736951. The raw and processed snRNA-seq dataset was deposited in the Gene Expression Omnibus (GEO) under accession number GSE218022. gnomAD frequencies were extracted from https://gnomad.broadinstitute.org/; the implication in cancers for each sSNV was evaluated in COSMIC, https://cancer.sanger.ac.uk/cosmic/; STRING analysis was performed using STRING, https://string-db.org/; single-cell RNA sequencing data from the developing cortex (Nowakowski et al.48), https://cells.ucsc.edu/?ds=cortex-dev; GRCh37 human reference genome under accession number SRA PRJNA31257 was used for the alignment of all types of sequencing data. Source data are provided with this paper.

Code availability

Code to generate the figures and analyze the data are publicly available on GitHub71 (https://github.com/shishenyxx/MCD_mosaic).

References

  1. Leventer, R. J., Guerrini, R. & Dobyns, W. B. Malformations of cortical development and epilepsy. Dialogues Clin. Neurosci. 10, 47–62 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Barkovich, A. J., Dobyns, W. B. & Guerrini, R. Malformations of cortical development and epilepsy. Cold Spring Harb. Perspect. Med. 5, a022392 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Blumcke, I. et al. The clinicopathologic spectrum of focal cortical dysplasias: a consensus classification proposed by an ad hoc Task Force of the ILAE Diagnostic Methods Commission. Epilepsia 52, 158–174 (2011).

    Article  PubMed  Google Scholar 

  4. Choi, S. A. & Kim, K. J. The surgical and cognitive outcomes of focal cortical dysplasia. J. Korean Neurosurg. Soc. 62, 321–327 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Krsek, P. et al. Different features of histopathological subtypes of pediatric focal cortical dysplasia. Ann. Neurol. 63, 758–769 (2008).

    Article  PubMed  Google Scholar 

  6. Bast, T., Ramantani, G., Seitz, A. & Rating, D. Focal cortical dysplasia: prevalence, clinical presentation and epilepsy in children and adults. Acta Neurol. Scand. 113, 72–81 (2006).

    Article  CAS  PubMed  Google Scholar 

  7. Poduri, A. et al. Somatic activation of AKT3 causes hemispheric developmental brain malformations. Neuron 74, 41–48 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lee, J. H. et al. De novo somatic mutations in components of the PI3K-AKT3-mTOR pathway cause hemimegalencephaly. Nat. Genet. 44, 941–945 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Baldassari, S. et al. Dissecting the genetic basis of focal cortical dysplasia: a large cohort study. Acta Neuropathol. 138, 885–900 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lai, D. et al. Somatic variants in diverse genes leads to a spectrum of focal cortical malformations. Brain 145, 2704–2720 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Bedrosian, T. A. et al. Detection of brain somatic variation in epilepsy-associated developmental lesions. Epilepsia 63, 1981–1997 (2022).

    Article  CAS  PubMed  Google Scholar 

  12. Sim, N. S. et al. Precise detection of low-level somatic mutation in resected epilepsy brain tissue. Acta Neuropathol. 138, 901–912 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Dou, Y. et al. Accurate detection of mosaic variants in sequencing data without matched controls. Nat. Biotechnol. 38, 314–319 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Yang, X. et al. Control-independent mosaic single nucleotide variant detection with DeepMosaic. Nat. Biotechnol. https://doi.org/10.1038/s41587-022-01559-w (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Huang, A. Y. et al. MosaicHunter: accurate detection of postzygotic single-nucleotide mosaicism through next-generation sequencing of unpaired, trio, and paired samples. Nucleic Acids Res. 45, e76 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wang, Y. et al. Comprehensive identification of somatic nucleotide variants in human brain tissue. Genome Biol. 22, 92 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Breuss, M. W. et al. Autism risk in offspring can be assessed through quantification of male sperm mosaicism. Nat. Med. 26, 143–150 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Yang, X. et al. Developmental and temporal characteristics of clonal sperm mosaicism. Cell 184, 4772–4783.e15 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Garcia, C. A. B. et al. mTOR pathway somatic variants and the molecular pathogenesis of hemimegalencephaly. Epilepsia Open 5, 97–106 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Pelorosso, C. et al. Somatic double-hit in MTOR and RPS6 in hemimegalencephaly with intractable epilepsy. Hum. Mol. Genet. 28, 3755–3765 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Breuss, M. W. et al. Somatic mosaicism in the mature brain reveals clonal cellular distributions during cortical development. Nature 604, 689–696 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lee, W. S. et al. Gradient of brain mosaic RHEB variants causes a continuum of cortical dysplasia. Ann. Clin. Transl. Neurol. 8, 485–490 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Bae, T. et al. Analysis of somatic mutations in 131 human brains reveals aging-associated hypermutability. Science 377, 511–517 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Bozic, I. et al. Accumulation of driver and passenger mutations during tumor progression. Proc. Natl Acad. Sci. USA 107, 18545–18550 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Alexandrov, L. B. et al. The repertoire of mutational signatures in human cancer. Nature 578, 94–101 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kim, M. & Costello, J. DNA methylation: an epigenetic mark of cellular memory. Exp. Mol. Med 49, e322 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Szklarczyk, D. et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47, D607–D613 (2019).

    Article  CAS  PubMed  Google Scholar 

  28. Tarabeux, J. et al. Rare mutations in N-methyl-D-aspartate glutamate receptors in autism spectrum disorders and schizophrenia. Transl. Psychiatry 1, e55 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Bezprozvanny, I. Calcium signaling and neurodegenerative diseases. Trends Mol. Med. 15, 89–100 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. van den Berg, D. L. C. et al. Nipbl interacts with Zfp609 and the integrator complex to regulate cortical neuron migration. Neuron 93, 348–361 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Yang, Y. J. et al. Microcephaly gene links trithorax and REST/NRSF to control neural stem cell proliferation and differentiation. Cell 151, 1097–1112 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Massimino, L. et al. TBR2 antagonizes retinoic acid dependent neuronal differentiation by repressing Zfp423 during corticogenesis. Dev. Biol. 434, 231–248 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Su, M. Y. et al. Hybrid structure of the RagA/C-Ragulator mTORC1 activation complex. Mol. Cell 68, 835–846.e3 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chen, J. et al. KLHL22 activates amino-acid-dependent mTORC1 signalling to promote tumorigenesis and ageing. Nature 557, 585–589 (2018).

    Article  CAS  PubMed  Google Scholar 

  35. Behar, T. N. et al. Glutamate acting at NMDA receptors stimulates embryonic cortical neuronal migration. J. Neurosci. 19, 4449–4461 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Paoletti, P., Bellone, C. & Zhou, Q. NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat. Rev. Neurosci. 14, 383–400 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Strehlow, V. et al. GRIN2A-related disorders: genotype and functional consequence predict phenotype. Brain 142, 80–92 (2019).

    Article  PubMed  Google Scholar 

  38. Prickett, T. D. & Samuels, Y. Molecular pathways: dysregulated glutamatergic signaling pathways in cancer. Clin. Cancer Res. 18, 4240–4246 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Tate, J. G. et al. COSMIC: the Catalogue Of Somatic Mutations In Cancer. Nucleic Acids Res. 47, D941–D947 (2019).

    Article  CAS  PubMed  Google Scholar 

  40. Ruvinsky, I. & Meyuhas, O. Ribosomal protein S6 phosphorylation: from protein synthesis to cell size. Trends Biochem. Sci. 31, 342–348 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Sumimoto, H., Imabayashi, F., Iwata, T. & Kawakami, Y. The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J. Exp. Med. 203, 1651–1656 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ornitz, D. M. & Itoh, N. The fibroblast growth factor signaling pathway. Wiley Interdiscip. Rev. Dev. Biol. 4, 215–266 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Chen, X. et al. TNF-alpha-induced NOD2 and RIP2 contribute to the up-regulation of cytokines induced by MDP in monocytic THP-1 cells. J. Cell. Biochem. 119, 5072–5081 (2018).

    Article  CAS  PubMed  Google Scholar 

  44. Yates, T. M. et al. SLC35A2-related congenital disorder of glycosylation: defining the phenotype. Eur. J. Paediatr. Neurol. 22, 1095–1102 (2018).

    Article  PubMed  Google Scholar 

  45. Paganini, C. et al. Calcium activated nucleotidase 1 (CANT1) is critical for glycosaminoglycan biosynthesis in cartilage and endochondral ossification. Matrix Biol. 81, 70–90 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lee, N. et al. Neuronal migration disorders: positron emission tomography correlations. Ann. Neurol. 35, 290–297 (1994).

    Article  CAS  PubMed  Google Scholar 

  47. Kim, Y. H. et al. Neuroimaging in identifying focal cortical dysplasia and prognostic factors in pediatric and adolescent epilepsy surgery. Epilepsia 52, 722–727 (2011).

    Article  PubMed  Google Scholar 

  48. Nowakowski, T. J. et al. Spatiotemporal gene expression trajectories reveal developmental hierarchies of the human cortex. Science 358, 1318–1323 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kim, J. K. et al. Brain somatic mutations in MTOR reveal translational dysregulations underlying intractable focal epilepsy. J. Clin. Invest. 129, 4207–4223 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Li, R. J. et al. Regulation of mTORC1 by lysosomal calcium and calmodulin. eLife 5, e19360 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Hoeffer, C. A. & Klann, E. mTOR signaling: at the crossroads of plasticity, memory and disease. Trends Neurosci. 33, 67–75 (2010).

    Article  CAS  PubMed  Google Scholar 

  52. Laplante, M. & Sabatini, D. M. Regulation of mTORC1 and its impact on gene expression at a glance. J. Cell Sci. 126, 1713–1719 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Coe, B. P. et al. Neurodevelopmental disease genes implicated by de novo mutation and copy number variation morbidity. Nat. Genet. 51, 106–116 (2019).

    Article  CAS  PubMed  Google Scholar 

  54. Ridley, A. J. et al. Cell migration: integrating signals from front to back. Science 302, 1704–1709 (2003).

    Article  CAS  PubMed  Google Scholar 

  55. Brini, M., Cali, T., Ottolini, D. & Carafoli, E. Neuronal calcium signaling: function and dysfunction. Cell. Mol. Life Sci. 71, 2787–2814 (2014).

    Article  CAS  PubMed  Google Scholar 

  56. Lamparello, P. et al. Developmental lineage of cell types in cortical dysplasia with balloon cells. Brain 130, 2267–2276 (2007).

    Article  PubMed  Google Scholar 

  57. Englund, C., Folkerth, R. D., Born, D., Lacy, J. M. & Hevner, R. F. Aberrant neuronal-glial differentiation in Taylor-type focal cortical dysplasia (type IIA/B). Acta Neuropathol. 109, 519–533 (2005).

    Article  CAS  PubMed  Google Scholar 

  58. Kim, S. et al. Strelka2: fast and accurate calling of germline and somatic variants. Nat. Methods 15, 591–594 (2018).

    Article  CAS  PubMed  Google Scholar 

  59. Benjamin, D. et al. Calling Somatic SNVs and Indels with Mutect2. Preprint at https://doi.org/10.1101/861054 (2019).

  60. Yang, X. et al. Genomic mosaicism in paternal sperm and multiple parental tissues in a Dravet syndrome cohort. Sci. Rep. 7, 15677 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  61. O’Roak, B. J. et al. Multiplex targeted sequencing identifies recurrently mutated genes in autism spectrum disorders. Science 338, 1619–1622 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Krumm, N. et al. Excess of rare, inherited truncating mutations in autism. Nat. Genet. 47, 582–588 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Piovesan, A. et al. Human protein-coding genes and gene feature statistics in 2019. BMC Res. Notes 12, 315 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Untergasser, A. et al. Primer3Plus, an enhanced web interface to Primer3. Nucleic Acids Res. 35, W71–W74 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Untergasser, A. et al. Primer3–new capabilities and interfaces. Nucleic Acids Res. 40, e115 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Lee, J. et al. Mutalisk: a web-based somatic MUTation AnaLyIS toolKit for genomic, transcriptional and epigenomic signatures. Nucleic Acids Res. 46, W102–W108 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Bindea, G. et al. ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics 25, 1091–1093 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Koizumi, H., Tanaka, T. & Gleeson, J. G. Doublecortin-like kinase functions with doublecortin to mediate fiber tract decussation and neuronal migration. Neuron 49, 55–66 (2006).

    Article  CAS  PubMed  Google Scholar 

  69. Langfelder, P. & Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9, 559 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Wang, F. et al. RNAscope: a novel in situ RNA analysis platform for formalin-fixed, paraffin-embedded tissues. J. Mol. Diagn. 14, 22–29 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Yang, X. & Chung, C. shishenyxx/MCD_mosaic: MCD mosaic codes v1.0.0. Zenodo https://doi.org/10.5281/ZENODO.7196403 (2022).

Download references

Acknowledgements

AmpliSeq, TASeq and snRNA-seq were supported by NIH P30CA023100 and S10OD026929 at the UCSD IGM Genomics Center. Rady Children’s Institute for Genomic Medicine, Broad Institute (U54HG003067, UM1HG008900), the Yale Center for Mendelian Disorders (U54HG006504) and the New York Genome Center provided WES. The UCSD Microscopy Core (NINDS P30NS047101) provided imaging support. The UCSD Tissue Technology Shared Resources Team (National Cancer Institute Cancer Center Support Grant, P30CA23100) supported paraffin sectioning and H&E staining. This study was supported by the 2021 NARSAD Young Investigator Grant from the Brain & Behavior Research Foundation (30598 to C.C.), NIH (NIMH U01MH108898 and R01MH124890 to J.G.G. and G.W.M. and NIA R21AG070462, NINDS R01NS083823 to J.G.G.), the Regione Toscana under the Call for Health 2018 (DECODE-EE to R.G.) and Fondazione Cassa di Risparmio di Firenze (to R.G.). Figures 1b and 3a were created with BioRender.com. The funders had no role in the study design, data collection, analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

C.C., X.Y., Sa.B., St.B. and J.G.G. designed the study. C.C., S.M. and S.K. conducted functional validation. C.B., V.S., A.S.N., E.R., C.C. and G.H. coordinated the clinical database. X.Y., C.C., M.W.B., L.L.B., R.D.G., J.G., M.X., A.P.L.M. and K.N.J. organized, handled and sequenced human samples. X.Y., C.C., T.B., Y.W., A.A., X.X., Z.L. and B.C. performed bioinformatics and data analysis. C.C. and K.I.V. performed the RNAscope experiment. C.D., H.W.P., C.A.B.G., S.H.K., H.K., H.U., M.P., A.S., C.A.H., D.D.L., C.A.G., M.D.S., S.S., M.N., D.D.G., K.I., Y.T., H.C., J.T., V.C., R.G., O.D., W.A.S., H.R.M. and G.W.M. provided resected brain tissues and clinical data from FCD patients. C.C., X.Y. and J.G.G. wrote the manuscript. All authors read and commented on the manuscript before submission.

Corresponding author

Correspondence to Joseph G. Gleeson.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Jean-Baptiste Rivière 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

Extended Data Fig. 1 Workflow of genetic discovery and bioinformatic pipeline to detect sSNVs in the MCD cohort.

(a) Workflow chart describing the flow of cases for each phase of genetic discovery. QNS: quantity not sufficient. 2 cases labeled by a star are sequenced in phase 1 but not phase 2. (b) The pipeline for paired samples. Notably, the dashed square indicates that the sharing variants between MuTect2 paired mode and Strelka2 were used for the downstream analysis. BSMN common pipeline and DeepMosaic were used only for WES datasets. The DeepMosaic input variants were generated by MuTect2 single mode. (c) The pipeline for unpaired samples. The pipeline is similar except that MuTect2 single mode without Strelka2 is used. PM: paired mode, SM: single mode.

Extended Data Fig. 2 The locations of the selected MCD variants.

(a) The location of two recurrent SNV calls is at the same position between the coiled-coil domain (CC) and the first SAM domain (S) of Liprin-α4. (b) Two different variants in SERCA1. p.R524C mutation is at the nucleotide ATP-binding (N) domain, whereas the p.A846T variant is in the 7th transmembrane (M7) domain. A: Actuator domain, P: Phosphorylation domain, M: Transmembrane domain. (c) Left: The location of p.H226R variant in RAGA protein. GTPase: GTPase domain, CRD: C-terminal roadblock domain. Right: UCSC genome browser screenshot describing that p.H226 is a conserved site across all vertebrates. (d) The location of the p.R38Q variant in the N-terminal region before the BTB (Broad-Complex, Tramtrack, and Bric-à-brac) domain of KLHL22. (e) A variant in the S1 domain of NR2C. S1 and S2 together make the ligand-binding domain (LBD), the target of glutamate. ATD: Amino-terminal domain. (f) RHOA p.P75S variant in the interdomain space between the second GTP/GDP binding domain and Rho insert domain.

Extended Data Fig. 3 Mutational signature analysis shows cell-division-related clock-like signatures in the MCD cohort.

SBS5 (39.1%) and SBS1 (33%) revealed by Mutalisk are clock-like mutational signatures. SBS1 especially correlates with stem cell division and mitosis.

Extended Data Fig. 4 Four major gene networks were reconstructed from the WES dataset.

(a) STRING DB pathway analysis of the 59 MCD discovered genes and six novel genes (a total of 65 genes) from recent publications identifies mTOR/MAP kinase pathway (pink, Cluster 1), Calcium dynamics (green, Cluster 2), Synapse (purple, Cluster 3), Gene expression (blue, Cluster 4). Edge thickness: confidence score calculated by STRING. Size and color of a node: square root transformed (sqrt-t) number of patients carrying a given mutation and average VAF across all patients, respectively. Non-clustered orphan genes are listed on the right. (b) Gene Ontology (GO) analysis results confirmed the functions of compositions in each network. Top GO terms or KEGG pathways. Strength calculation and cluster generation were performed by STRING.

Extended Data Fig. 5 ClueGO analysis using the MCD genes result identifies the biological processes and molecular pathways.

The main cluster is related to TOR signaling, regulation of cell-matrix adhesion, regulation of focal adhesion assembly, and artery morphogenesis. Notably, there are also isolated clusters that were not covered in previous studies, for example, cardiac muscle cell contraction, calcium ion import, and protein localization to the synapse. Corrected p-value with Bonferroni step down was reflected in node size (two-sided hypergeometric test, Large: p < 0.0005, medium: p < 0.005, small: p < 0.05). All p-values are in Supplementary Table 9.

Extended Data Fig. 6 Additional functional analyses for new RRAGA and RHOA mutations.

(a) Over-expression of RHOA WT and P75S mutant form in cortical neurogenic pool induce both significant defects in migration. Notably, some portion of WT form-expressing cells migrate to the superior cortical area (white arrow), whereas mutant form-expressing cells did not show any migrating cells at all. The dashed square area is magnified to the right side images. Scale bar: 100 µm and 20 µm for left and right images, respectively. Right, Quantification of the migration level. EV data was exported from Fig. 3b. Two-way ANOVA and Sidak multiple comparisons with p-values of interaction between genotype and bin factor. Ten bins from the surface of the cortex (top) to SVZ (bottom). n = 3, 3, 2 biologically independent mice for EV, RHOA WT and RHOA P75S, respectively. Mean ± SEM. (b) Immunofluorescence in postnatal day 21 mouse cortices for RRAGA wild-type (WT) or mutant isoform. Yellow dashed lines: examples of cell body size quantification. Dashed lines and dotted lines in the violin plots indicate median and quartiles, respectively. Two-tailed Mann-Whitney U–test. Scale bar: 20 μm. n = 61 cells (3 mice), 61 (2) for RRAGA WT and RRAGA H226R, respectively * or # indicates a p-value in comparison between WT and mutant group, or EV and mutant group, respectively. ####P < 0.0001; ##P < 0.01; #P < 0.05; **P < 0.01; ns, non-significant.

Source data

Extended Data Fig. 7 Cell-type identification by DEGs and WGCNA in the MCD snRNA-seq dataset.

(a) DEG analysis using FindAllMarker function in Seurat v4 package. The top 10 genes for each cluster were presented. Some notable marker genes are presented on the left side. Color: scaled gene expression level. (b) Description of WGCNA. The most variable 3000 genes were used for generating six co-expression module eigengenes (ME1 to ME11). The members of each ME are described in Supplementary Table 5,b. (c) Enrichment of module eigengenes in cell type clusters. Atypical clusters showing similar patterns with a normal cell cluster were classified as the same lineage. We identified 5 different lineages (Ast, OD, ExN, InN, OPC) coded as different colors. Notably, Ast-L1/2/3 and OPC-L1/2 show excessively increased expression of ME6 or ME7, Ast or OPC signature ME, respectively. OPC-L2 shows upregulation of ME4, related to the cell cycle, implying that HME has many over-proliferating OPC-L cells. Excitatory neuronal lineage typically expresses ME5 and ME10, but ExN-L1/2 also shows increased expression of ME9, a signature of inhibitory neurons, compared to ExN1/2/3. OD-L cells are classified as OD lineage because they express excessive ME11, a signature to OD. U cluster, dominant in TSC, does not show a clear signature. The size and color of the dot plot are the Pearson correlation coefficient and corresponding non-adjusted asymptotic p-value derived from a two-sided Student’s t-test, respectively.

Extended Data Fig. 8 The validation of the snRNA-seq result from HME-6593 shows that MCD dominant clusters are highly correlated with dysplastic cells in MCD.

(a–b) H&E (left) and RNAscope (right) for genes expressed highly in MCD brain (FGFR2, FGFR1, EGFR, top) or (FGFR3, PDGFRA bottom). Dashed lines: blood vessels. White/black arrows: dysplastic cells. One representative section is shown for each probe combination.

Extended Data Fig. 9 Expression patterns of individual MCD genes in the MCD snRNA-seq dataset.

The gene members of each eigen module shown in Fig. 6d were colored according to the name of a given eigengene.

Extended Data Fig. 10 Functional implication of MCD genes in MCD snRNA-seq dataset.

(a) The 75 MCD genes overlap with DEGs of MCDs in contrast to controls. p-values derived from permutation tests (10,000 times) show a chance to show this overlap in a random sampling of DEGs from 19909 protein-coding genes used in these DEGs. Red or blue coloring of gene names indicates upregulated or downregulated DEGs in MCDs compared to CTRLs, respectively. HME and TSC show significant overlap with the MCD genes whereas an FCD case with low VAF did not, which is probably because of low VAF. One-sided permutation test. (b) Cell-lineage specific DEGs compared to the according to normal cell lineage of CTRL represent alterations of mTOR downstream pathways, calcium dynamics, and synaptic functions across. Red dots in UMAPs indicate the cells that participated in the comparison. Top 10 enriched GO or KEGG terms representing lineage-specific DEGs.

Supplementary information

Supplementary Information

Supplementary Tables 1–9.

Reporting Summary

Peer Review File

Supplementary Tables

Supplementary Table 1. Cohort list and corresponding sequencing methods. The 293 cases are listed in each row and corresponding sequencing methods used for a given sample are described. Supplementary Table 2. AmpliSeq primer pool designs. a, Ampliseq primer pool design used in phase 1. b, Highlighting table for the known genes where somatic or germline variants are previously detected in FCD/HMEs and the candidate ones known as PI3K–AKT3–mTOR interactors. c, Ampliseq primer pool design used in phase 3. Supplementary Table 3. Summary of SNV calls across three phases of genetic discovery. a, 1181 raw sSNV calls derived from the combination of variant callers described in Extended Data Fig. 1b. b, Validated 108 brain sSNVs from a. c, Annotation table of the genes listed in b based on GO terms. Supplementary Table 4. Summary of phenotype and genotype information for the ‘genetically solved’ cases. Supplementary Table 5. Summary table for snRNA-seq DEG and WGCNA analysis. a, Cell markers are generated by the FindAllMarkers function in the Seurat v,4 package. b, 11 MEs and their gene members are generated by WGCNA. c, DEG analysis results include all cell types or in a cell-type-restricted manner. d, All enriched terms from DEGs. Supplementary Table 6. Summary table for false discovery estimation. Supplementary Table 7. Estimation of probability of observing recurrency in mutated genes in MCD. Supplementary Table 8. Primer sequences used in TASeq for orthogonal validation and quantification of mosaic mutations detected in AmpliSeq and WES sequencing. Supplementary Table 9. Detailed statistical information on Fig. 1d, Fig. 3 and Extended Data Figs. 5 and 6.

Source data

Source Data Fig. 3

Actual per-cell immunofluorescence values in Fig. 3c violin plots.

Source Data Extended Data Fig. 6

Actual per-cell immunofluorescence values in Extended Data Fig. 6b violin plots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) 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

Chung, C., Yang, X., Bae, T. et al. Comprehensive multi-omic profiling of somatic mutations in malformations of cortical development. Nat Genet 55, 209–220 (2023). https://doi.org/10.1038/s41588-022-01276-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-022-01276-9

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing