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.

Genetic identification of brain cell types underlying schizophrenia

Abstract

With few exceptions, the marked advances in knowledge about the genetic basis of schizophrenia have not converged on findings that can be confidently used for precise experimental modeling. By applying knowledge of the cellular taxonomy of the brain from single-cell RNA sequencing, we evaluated whether the genomic loci implicated in schizophrenia map onto specific brain cell types. We found that the common-variant genomic results consistently mapped to pyramidal cells, medium spiny neurons (MSNs) and certain interneurons, but far less consistently to embryonic, progenitor or glial cells. These enrichments were due to sets of genes that were specifically expressed in each of these cell types. We also found that many of the diverse gene sets previously associated with schizophrenia (genes involved in synaptic function, those encoding mRNAs that interact with FMRP, antipsychotic targets, etc.) generally implicated the same brain cell types. Our results suggest a parsimonious explanation: the common-variant genetic results for schizophrenia point at a limited set of neurons, and the gene sets point to the same cells. The genetic risk associated with MSNs did not overlap with that of glutamatergic pyramidal cells and interneurons, suggesting that different cell types have biologically distinct roles in schizophrenia.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Specificity metric calculated from single-cell transcriptome sequencing data can be used to test for increased burden of schizophrenia-SNP heritability in brain cell types.
Fig. 2: Evaluation of enrichment of common-variant CLOZUK schizophrenia GWAS results in the KI brain scRNA-seq dataset from mouse.
Fig. 3: Comparison of scRNA-seq and snRNA-seq, and evaluation of enrichment of common-variant CLOZUK schizophrenia genome-wide association results in brain snRNA-seq datasets from adult humans.
Fig. 4: Cell-type enrichment of gene sets associated with schizophrenia, neurological disorders and the evolutionary divergence between human and mouse.
Fig. 5: CA1 pyramidal neurons, medium spiny neurons and cortical interneurons are independently associated with schizophrenia, and distinct molecular pathways contribute to each cell type.

References

  1. Sullivan, P. F., Daly, M. J. & O’Donovan, M. Genetic architectures of psychiatric disorders: the emerging picture and its implications. Nat. Rev. Genet. 13, 537–551 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Purcell, S. M. et al. A polygenic burden of rare disruptive mutations in schizophrenia. Nature 506, 185–190 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Schizophrenia Working Group of the Psychiatric Genomics Consortium. Biological insights from 108 schizophrenia-associated genetic loci. Nature 511, 421–427 (2014).

    Article  PubMed Central  CAS  Google Scholar 

  4. Fromer, M. et al. De novo mutations in schizophrenia implicate synaptic networks. Nature 506, 179–184 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Genovese, G. et al. Increased burden of ultra-rare protein-altering variants among 4,877 individuals with schizophrenia. Nat. Neurosci. 19, 1433–1441 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Singh, T. et al. Rare loss-of-function variants in SETD1A are associated with schizophrenia and developmental disorders. Nat. Neurosci. 19, 571–577 (2016).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  7. Sekar, A. et al. Schizophrenia risk from complex variation of complement component 4. Nature 530, 177–183 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Marshall, C. R. et al. Contribution of copy-number variants to schizophrenia from a genome-wide study of 41,321 subjects. Nat. Genet. 49, 27–35 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Finucane, H. K. et al. Partitioning heritability by functional category using GWAS summary statistics. Nat. Genet. 47, 1228–1235 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Lek, M. et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 536, 285–291 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Lips, E. S. et al. Functional gene group analysis identifies synaptic gene groups as risk factor for schizophrenia. Mol. Psychiatry 17, 996–1006 (2012).

    Article  PubMed  CAS  Google Scholar 

  12. Darnell, J. C. et al. FMRP stalls ribosomal translocation on mRNAs linked to synaptic function and autism. Cell 146, 247–261 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Goudriaan, A. et al. Specific glial functions contribute to schizophrenia susceptibility. Schizophr. Bull. 40, 925–935 (2014).

    Article  PubMed  Google Scholar 

  14. Fromer, M. et al. Gene expression elucidates functional impact of polygenic risk for schizophrenia. Nat. Neurosci. 19, 1442–1453 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Pers, T. H. et al. Comprehensive analysis of schizophrenia-associated loci highlights ion channel pathways and biologically plausible candidate causal genes. Hum. Mol. Genet. 25, 1247–1254 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Zeisel, A. et al. Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq. Science 347, 1138–1142 (2015).

    Article  PubMed  CAS  Google Scholar 

  17. Romanov, R. A. et al. Molecular interrogation of hypothalamic organization reveals distinct dopamine neuronal subtypes. Nat. Neurosci. 20, 176–188 (2017).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. de Leeuw, C. A., Mooij, J. M., Heskes, T. & Posthuma, D. MAGMA: generalized gene-set analysis of GWAS data. PLoS Comput. Biol. 11, e1004219 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Pardiñas, A. F. et al. Common schizophrenia alleles are enriched in mutation-intolerant genes and in regions under strong background selection. Nat. Genet. 50, 381–389 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. GTEx Consortium. The Genotype–Tissue Expression (GTEx) pilot analysis: multi-tissue gene regulation in humans. Science 348, 648–660 (2015).

    Article  PubMed Central  CAS  Google Scholar 

  22. Finucane, H. K. et al. Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types. Nat. Genet. 50, 621–629 (2018).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  23. Gokce, O. et al. Cellular taxonomy of the mouse striatum as revealed by single-cell RNA-seq. Cell Rep. 16, 1126–1137 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Habib, N. et al. Div-Seq: single-nucleus RNA-seq reveals dynamics of rare adult newborn neurons. Science 353, 925–928 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Tasic, B. et al. Adult mouse cortical cell taxonomy revealed by single-cell transcriptomics. Nat. Neurosci. 19, 335–346 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Habib, N. et al. Massively parallel single-nucleus RNA-seq with DroNc-seq. Nat. Methods 14, 955–958 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Abdelmoez, M.N. et al. Correlation of gene expressions between nucleus and cytoplasm reflects single-cell physiology. Preprint at bioRxiv https://www.biorxiv.org/content/early/2017/10/20/206672 (2017).

  28. Cajigas, I. J. et al. The local transcriptome in the synaptic neuropil revealed by deep sequencing and high-resolution imaging. Neuron 74, 453–466 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Darmanis, S. et al. A survey of human brain transcriptome diversity at the single-cell level. Proc. Natl Acad. Sci. USA 112, 7285–7290 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Lake, B. B. et al. Neuronal subtypes and diversity revealed by single-nucleus RNA sequencing of the human brain. Science 352, 1586–1590 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Skene, N. G. & Grant, S. G. Identification of vulnerable cell types in major brain disorders using single-cell transcriptomes and expression-weighted cell-type enrichment. Front. Neurosci. 10, 16 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Gaspar, H. A. & Breen, G. Drug enrichment and discovery from schizophrenia genome-wide association results: an analysis and visualisation approach. Sci. Rep. 7, 12460 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Anttila, V. et al. Analysis of shared heritability in common disorders of the brain. Science (in the press).

  34. Lambert, J. C. et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat. Genet. 45, 1452–1458 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Bertram, L., McQueen, M. B., Mullin, K., Blacker, D. & Tanzi, R. E. Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nat. Genet. 39, 17–23 (2007).

    Article  PubMed  CAS  Google Scholar 

  36. Patsopoulos, N. et al. The Multiple Sclerosis Genomic Map: role of peripheral immune cells and resident microglia in susceptibility. Preprint at bioRxiv https://www.biorxiv.org/content/early/2017/07/13/143933 (2017).

  37. Yang, H., Robinson, P. N. & Wang, K. Phenolyzer: phenotype-based prioritization of candidate genes for human diseases. Nat. Methods 12, 841–843 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Burow, D. A. et al. Dynamic regulation of mRNA decay during neural development. Neural Dev. 10, 11 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Akbarian, S. et al. The PsychENCODE project. Nat. Neurosci. 18, 1707–1712 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. 1000 Genomes Project Consortium. A global reference for human genetic variation. Nature 526, 68–74 (2015).

    Article  CAS  Google Scholar 

  41. de Leeuw, C. A., Neale, B. M., Heskes, T. & Posthuma, D. The statistical properties of gene-set analysis. Nat. Rev. Genet. 17, 353–364 (2016).

    Article  PubMed  CAS  Google Scholar 

  42. Brown, M. B. A method for combining non-independent, one-sided tests of significance. Biometrics 31, 987–992 (1975).

    Article  Google Scholar 

  43. Lun, A. T., McCarthy, D. J. & Marioni, J. C. A step-by-step workflow for low-level analysis of single-cell RNA-seq data with Bioconductor. F1000Res 5, 2122 (2016).

    PubMed  PubMed Central  Google Scholar 

  44. Vu, T. N. et al. Beta-Poisson model for single-cell RNA-seq data analyses. Bioinformatics 32, 2128–2135 (2016).

    Article  PubMed  CAS  Google Scholar 

  45. Okbay, A. et al. Genome-wide association study identifies 74 loci associated with educational attainment. Nature 533, 539–542 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Sniekers, S. et al. Genome-wide association meta-analysis of 78,308 individuals identifies new loci and genes influencing human intelligence. Nat. Genet. 49, 1107–1112 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Nalls, M. A. et al. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease. Nat. Genet. 46, 989–993 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Wood, A. R. et al. Defining the role of common variation in the genomic and biological architecture of adult human height. Nat. Genet. 46, 1173–1186 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Paternoster, R., Brame, R., Mazerolle, P. & Piquero, A. Using the correct statistical test for the equality of regression coefficients. Criminology 36, 859–866 (1998).

    Article  Google Scholar 

  50. Wagnon, J. L. et al. CELF4 regulates translation and local abundance of a vast set of mRNAs, including genes associated with regulation of synaptic function. PLoS Genet. 8, e1003067 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Collins, M. O. et al. Molecular characterization and comparison of the components and multiprotein complexes in the postsynaptic proteome. J. Neurochem. 97, 16–23 (2006). (Suppl. 1).

    Article  PubMed  CAS  Google Scholar 

  52. Bayés, A. et al. Characterization of the proteome, diseases and evolution of the human postsynaptic density. Nat. Neurosci. 14, 19–21 (2011).

    Article  PubMed  CAS  Google Scholar 

  53. Fernández, E. et al. Targeted tandem affinity purification of PSD95 recovers core postsynaptic complexes and schizophrenia susceptibility proteins. Mol. Syst. Biol. 5, 269 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Weyn-Vanhentenryck, S. M. et al. HITS–CLIP and integrative modeling define the Rbfox splicing-regulatory network linked to brain development and autism. Cell Rep. 6, 1139–1152 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Thul, P. J. et al. A subcellular map of the human proteome. Science 356, eaal3321 (2017).

    Article  PubMed  CAS  Google Scholar 

  56. Brozzi, A., Urbanelli, L., Germain, P. L., Magini, A. & Emiliani, C. hLGDB: a database of human lysosomal genes and their regulation. Database 2013, bat024 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Calvo, S. E., Clauser, K. R. & Mootha, V. K. MitoCarta2.0: an updated inventory of mammalian mitochondrial proteins. Nucleic Acids Res. 44, D1251–D1257 (2016). (D1).

    Article  PubMed  CAS  Google Scholar 

  58. Shigeoka, T. et al. Dynamic axonal translation in developing and mature visual circuits. Cell 166, 181–192 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Boyken, J. et al. Molecular profiling of synaptic vesicle docking sites reveals novel proteins but few differences between glutamatergic and GABAergic synapses. Neuron 78, 285–297 (2013).

    Article  PubMed  CAS  Google Scholar 

  60. Takamori, S. et al. Molecular anatomy of a trafficking organelle. Cell 127, 831–846 (2006).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

J.H.-L. was funded by the Swedish Research Council (Vetenskapsrådet, award 2014-3863), StratNeuro, the Wellcome Trust (108726/Z/15/Z) and the Swedish Brain Foundation (Hjärnfonden). P.F.S. gratefully acknowledges support from the Swedish Research Council (Vetenskapsrådet, award D0886501). N.G.S. was supported by the Wellcome Trust (108726/Z/15/Z). J.B. was supported by the Swiss National Science Foundation. The PGC has received major funding from the US National Institute of Mental Health (U01 MH109528 and U01 MH109532). H.A.G. was supported by PGC grant 1U01MH109514-01. Primary schizophrenia GWAS data were generated with support from the Medical Research Council (MRC) Centre (MR/L010305/1), program grant G0800509 and project grant MR/L011794/1, and funding from the European Union’s Seventh Framework Programme for research, technological development and demonstration under grant agreement no. 279227 (CRESTAR Consortium).

Author information

Authors and Affiliations

Authors

Consortia

Contributions

N.G.S., J.B., P.F.S. and J.H.-L. designed the study and wrote and reviewed the manuscript; N.G.S. performed the LDSC analyses; J.B. performed the MAGMA analyses; T.E.B., R.D.H., J.A.M. and E.S.L. generated the human mid-temporal cortex data; A.B.M.-M., J.R., S.L. and J.H.-L. generated the KI single-cell data; the Major Depressive Disorder Working Group of the PGC performed the MDD GWAS; J.T.R.W., J.J.C., P.G.-R., M.C.O., M.J.O. and A.F.P. performed the schizophrenia CLOZUK GWAS; G.B. and H.A.G. analyzed the antipsychotic drug targets; and all authors read and approved the manuscript.

Corresponding authors

Correspondence to Patrick F. Sullivan or Jens Hjerling-Leffler.

Ethics declarations

Competing interests

P.F.S. is on the advisory committee at Lundbeck, is a Scientific Advisory Board member at Pfizer and has received speaker reimbursement and grant funding from Roche. J.H.-L. is a Scientific Advisor at Cartana and has received grant funding from Roche.

Additional information

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

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–21, Supplementary Note and Supplementary Tables 1–3

Reporting Summary

Supplementary Table 4

Specificity values for Karolinska scRNA-seq superset

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Skene, N.G., Bryois, J., Bakken, T.E. et al. Genetic identification of brain cell types underlying schizophrenia. Nat Genet 50, 825–833 (2018). https://doi.org/10.1038/s41588-018-0129-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-018-0129-5

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