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.

  • Resource
  • Published:

Coexpression network architecture reveals the brain-wide and multiregional basis of disease susceptibility

Abstract

Gene networks have yielded numerous neurobiological insights, yet an integrated view across brain regions is lacking. We leverage RNA sequencing in 864 samples representing 12 brain regions to robustly identify 12 brain-wide, 50 cross-regional and 114 region-specific coexpression modules. Nearly 40% of genes fall into brain-wide modules, while 25% comprise region-specific modules reflecting regional biology, such as oxytocin signaling in the hypothalamus, or addiction pathways in the nucleus accumbens. Schizophrenia and autism genetic risk are enriched in brain-wide and multiregional modules, indicative of broad impact; these modules implicate neuronal proliferation and activity-dependent processes, including endocytosis and splicing, in disease pathophysiology. We find that cell-type-specific long noncoding RNA and gene isoforms contribute substantially to regional synaptic diversity and that constrained, mutation-intolerant genes are primarily enriched in neurons. We leverage these data using an omnigenic-inspired network framework to characterize how coexpression and gene regulatory networks reflect neuropsychiatric disease risk, supporting polygenic models.

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: Human whole-brain coexpression atlas.
Fig. 2: Cell-type heterogeneity relates to coexpression modules, mutation intolerance and evolution.
Fig. 3: Creating a catalog of cell-type-specific isoforms.
Fig. 4: Region-specific gene upregulation reflects region-specific cell types and ribosomal turnover.
Fig. 5: Gene-level module enrichment for de novo protein-truncating variants, GWAS summary statistics and differential expression.
Fig. 6: Ontologies, PPI networks and expression profiles of ASD-associated modules.
Fig. 7: Characterizing core–periphery structure of high-impact neuropsychiatric disease genes across multiple networks.

Similar content being viewed by others

Data availability

Processed data are available at http://geschwindlab.org/gclabapps/hubgene/home/.

Code availability

Supporting code for network construction and network genetic analysis is available at https://github.com/dhglab/multiregional-networks/.

References

  1. Smoller, J. W. et al. Psychiatric genetics and the structure of psychopathology. Mol. Psychiatry 24, 409–420 (2018).

  2. Félix, M.-A. & Barkoulas, M. Pervasive robustness in biological systems. Nat. Rev. Genet. 16, 483–496 (2015).

    Article  PubMed  CAS  Google Scholar 

  3. Geschwind, D. H. & Flint, J. Genetics and genomics of psychiatric disease. Science 349, 1489–1494 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gandal, M. J. et al. Shared molecular neuropathology across major psychiatric disorders parallels polygenic overlap. Science 359, 693–697 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Horn, H. et al. NetSig: network-based discovery from cancer genomes. Nat. Methods 15, 61–66 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Mostafavi, S. et al. Parsing the interferon transcriptional network and its disease associations. Cell 164, 564–578 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Oldham, M. C. et al. Functional organization of the transcriptome in human brain. Nat. Neurosci. 11, 1271–1282 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. GTEx Consortium. Genetic effects on gene expression across human tissues. Nature 550, 204–213 (2017).

  9. Boyle, E. A., Li, Y. I. & Pritchard, J. K. An expanded view of complex traits: from polygenic to omnigenic. Cell 169, 1177–1186 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kelley, K. W., Nakao-Inoue, H., Molofsky, A. V. & Oldham, M. C. Variation among intact tissue samples reveals the core transcriptional features of human CNS cell classes. Nat. Neurosci. 21, 1171–1184 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. McKenzie, A. T. et al. Brain cell-type-specific gene expression and coexpression network architectures. Sci. Rep. 8, 8868 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Wang, H.-Y. et al. Rate of evolution in brain-expressed genes in humans and other primates. PLoS Biol. 5, e13 (2006).

    Article  PubMed Central  Google Scholar 

  13. Shohat, S., Ben-David, E. & Shifman, S. Varying intolerance of gene pathways to mutational classes explain genetic convergence across neuropsychiatric disorders. Cell Rep. 18, 2217–2227 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Parikshak, N. N. et al. Genome-wide changes in lncRNA, splicing and regional gene expression patterns in autism. Nature 540, 423–427 (2016).

  16. Gandal, M. J. et al. Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia and bipolar disorder. Science 362, eaat8127 (2018).

  17. Bennett, V. & Lorenzo, D. N. An adaptable spectrin/ankyrin-based mechanism for long-range organization of plasma membranes in vertebrate tissues. Curr. Top. Membr. 77, 143–184 (2016).

  18. Dörrbaum, A. R., Kochen, L., Langer, J. D. & Schuman, E. M. Local and global influences on protein turnover in neurons and glia. eLife 7, e34202 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

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

  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. Battista, D., Ferrari, C. C., Gage, F. H. & Pitossi, F. J. Neurogenic niche modulation by activated microglia: transforming growth factor-β increases neurogenesis in the adult dentate gyrus. Eur. J. Neurosci. 23, 83–93 (2006).

    Article  PubMed  Google Scholar 

  22. Phoenix, T. N. & Temple, S. Spred1, a negative regulator of Ras-MAPK-ERK, is enriched in CNS germinal zones, dampens NSC proliferation, and maintains ventricular zone structure. Genes Dev. 24, 45–56 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Parikshak, N. et al. Integrative functional genomic analyses implicate specific molecular pathways and circuits in autism. Cell 155, 1008–1021 (2013).

  24. Selimbeyoglu, A. et al. Modulation of prefrontal cortex excitation/inhibition balance rescues social behavior inCNTNAP2-deficient mice. Sci. Transl. Med. 9, eaah6733 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Nelson, S. B. & Valakh, V. Excitatory/inhibitory balance and circuit homeostasis in autism spectrum disorders. Neuron 87, 684–698 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang, D. et al. Comprehensive functional genomic resource and integrative model for the human brain. Science 362, eaat8464 (2018).

  27. Boulting, G. L. et al. Activity-dependent regulome of human GABAergic neurons reveals new patterns of gene regulation and neurological disease heritability. Nat. Neurosci. 24, 437–448 (2021).

  28. Schanzenbächer, C. T., Langer, J. D. & Schuman, E. M. Time- and polarity-dependent proteomic changes associated with homeostatic scaling at central synapses. eLife 7, e33322 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Marbach, D. et al. Tissue-specific regulatory circuits reveal variable modular perturbations across complex diseases. Nat. Methods 13, 366–370 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Wray, N. R., Wijmenga, C., Sullivan, P. F., Yang, J. & Visscher, P. M. Common disease is more complex than implied by the core gene omnigenic model. Cell 173, 1573–1580 (2018).

    Article  CAS  PubMed  Google Scholar 

  31. Miller, J. A., Oldham, M. C. & Geschwind, D. H. A systems-level analysis of transcriptional changes in Alzheimer’s disease and normal aging. J. Neurosci. 28, 1410–1420 (2008).

  32. Voineagu, I., et al. Transcriptomic analysis of autistic brain reveals convergent molecular pathology. Nature 474, 380–384 (2011).

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

  34. Wang, Q. et al. The landscape of multiscale transcriptomic networks and key regulators in Parkinson’s disease. Nat Commun. 10, 5234 (2019).

  35. Miller, J. A., Horvath, S. & Geschwind, D. H. Divergence of human and mouse brain transcriptome highlights Alzheimer disease pathways. Proc. Natl Acad. Sci. USA 107, 12698–12703 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Howrigan, D. P. et al. Exome sequencing in schizophrenia-affected parent–offspring trios reveals risk conferred by protein-coding de novo mutations. Nat. Neurosci. 23, 185–193 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. 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  CAS  PubMed  PubMed Central  Google Scholar 

  38. Sanders, S. J. et al. Insights into autism spectrum disorder genomic architecture and biology from 71 risk loci. Neuron 87, 1215–1233 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Skene, N. G. et al. Genetic identification of brain cell types underlying schizophrenia. Nat. Genet. 50, 825–833 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ruzzo, E. K. et al. Inherited and de novo genetic risk for autism impacts shared networks. Cell 178, 850–866 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Wang, Q. et al. A Bayesian framework that integrates multi-omics data and gene networks predicts risk genes from schizophrenia GWAS data. Nat. Neurosci. 22, 691–699 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Walker, R. L. et al. Genetic control of expression and splicing in developing human brain informs disease mechanisms. Cell 179, 750–771 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Satterstrom, F. K. et al. Large-scale exome sequencing study implicates both developmental and functional changes in the neurobiology of autism. Cell 180, 568–584 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Polioudakis, D. et al. A single-cell transcriptomic atlas of human neocortical development during mid-gestation. Neuron 103, 785–801 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Nicholson-Fish, J. C., Kokotos, A. C., Gillingwater, T. H., Smillie, K. J. & Cousin, M. A. VAMP4 is an essential cargo molecule for activity-dependent bulk endocytosis. Neuron 88, 973–984 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kokotos, A. C., Peltier, J., Davenport, E. C., Trost, M. & Cousin, M. A. Activity-dependent bulk endocytosis proteome reveals a key presynaptic role for the monomeric GTPase Rab11. Proc. Natl Acad. Sci. USA 115, E10177–E10186 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Doll, C. A. & Broadie, K. Impaired activity-dependent neural circuit assembly and refinement in autism spectrum disorder genetic models. Front. Cell. Neurosci. 8, 30 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Dobin, A. et al. STAR: ultrafast and universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

  49. Nieuwenhuis, T. O. et al. Consistent RNA-sequencing contamination in GTEx and other datasets. Nat. Comm. 11, 1933 (2020).

  50. Pedregosa, F. et al. Scikit-learn: machine learning in Python. JMLR 12, 2825–2830 (2011).

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

  52. Langfelder, P., Luo, R., Oldham, M. & Horvath, S. Is my network module preserved and reproducible? PLoS Comput. Biol. 7, e1001057 (2011).

  53. Crow, M., Paul, A., Ballouz, S., Huang, Z. J. & Gillis, J. Characterizing the replicability of cell types defined by single-cell RNA-sequencing data using MetaNeighbor. Nat. Commun. 9, 884 (2018).

  54. Nehme, R. et al. Combining NGN2 programming with developmental patterning generates human excitatory neurons with NMDAR-mediated synaptic transmission. Cell Rep. 23, 2509–2523 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Tychele N. T. et al. denovo-db: a compendium of human de novo variants Nucleic Acids Res. 45, D804–D811 (2016).

  56. Ruzzo, E. K. et al. Inherited and de novo genetic risk for autism impacts shared networks. Cell 178, 850–866 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  Google Scholar 

  58. Won, H., Huang, J., Opland, C. K., Hartl, C. L. & Geschwind, D. H. Human evolved regulatory elements modulate genes involved in cortical expansion and neurodevelopmental disease susceptibility. Nat. Commun. 10, 2396 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Schork, A. J. et al. A genome-wide association study of shared risk across psychiatric disorders implicates gene regulation during fetal neurodevelopment. Nat. Neurosci. 22, 353–361 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Cross-Disorder Group of the Psychiatric Genomics Consortium. Identification of risk loci with shared effects on five major psychiatric disorders: a genome-wide analysis. Lancet https://doi.org/10.1016/S0140-6736(12)62129-1 (2013).

  61. 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  CAS  PubMed  PubMed Central  Google Scholar 

  62. Andlauer, T. F. et al. Novel multiple sclerosis susceptibility loci implicated in epigenetic regulation. Sci. Adv. 2, e1501678 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Nguyen, H. T. et al. Integrated Bayesian analysis of rare exonic variants to identify risk genes for schizophrenia and neurodevelopmental disorders. Genome Med. https://doi.org/10.1186/s13073-017-0497-y (2017).

  65. Du, Y. et al. Nonrandom occurrence of multiple de novo coding variants in a proband indicates the existence of an oligogenic model in autism. Genet. Med. 22, 170–180 (2019).

  66. Ionita-Laza, I., Lange, C. & Laird, M. N. Estimating the number of unseen variants in the human genome. Proc. Natl Acad. Sci. USA 106, 5008–5013 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Lambert, S. A. et al. The human transcription factors. Cell 172, 650–665 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

D.H.G., C.L.H. and G.R. were supported by the National Institute of Mental Health (NIMH; R01MH110927 and U01MH115746) and the Simons Foundation Autism Research Initiative. G.R. was supported by NIMH award 1F32MH114620. K.L. and G.P. are supported by institutional funding from the Stanley Center for Psychiatric Research at the Broad Institute, the NIMH (R01 MH109903 and U01 MH121499), the Simons Foundation Autism Research Initiative (awards 515064 and 735604), the Lundbeck Foundation (R223-2016-721 and R350-2020-963). A.B., A.S. and P.P. were supported by NIMH award R01MH110927. We thank members of the laboratories of D.H.G., K.L. and A.B. for stimulating discussions. We are also grateful for data made publicly available by the GTEx consortium and S. Deverasetty for creating the HUBgene web browser.

Author information

Authors and Affiliations

Authors

Contributions

D.H.G., C.L.H., A.B. and K.L. conceived the study design. A.B., A.S. and P.P. reprocessed RNA-seq data. C.L.H., S.M., P.P., W.G.P., G.R. and A.S. built networks and performed analyses. G.P. differentiated cell lines and performed the ANK2 western blot. C.L.H. and D.H.G. prepared the manuscript and figures. A.B. and K.L. contributed to editing.

Corresponding author

Correspondence to Daniel H. Geschwind.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Neuroscience thanks the anonymous reviewers for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Brain network QC and algorithm comparisons (related to Fig. 1).

a, Standard boxplot (box: quartiles, whiskers: 1.5xIQR) of ePC and HCP loadings onto canonical cell type genes, showing significant heterogeneity of loadings across cell types, N = 114 (Neuron), 79 (Astrocyte), 242 (Microglia), 103 (Oligodendrocyte), 176 (Endothelial). b, Standard boxplot (box: quartiles, whiskers: 1.5xIQR) of ePC loadings after covariate correction using HCP and LM base correction, showing that cell type heterogeneity of the 1st component of expression is lost after HCP correction. Gene set sizes as in a; significance (two-sided T-test) ***: < 0.001. c, Network-based GO prediction accuracy for each brain region. The same gene holdouts are used in 10-fold cross validation, generating 10 values for the AUC difference of each GO category, which are used to generate a Z-score for the expected AUC difference. d, Relative improvement to the integrated correlation coefficient for BRNHYP genes, for linear model and HCP based corrections. e, Pairwise co-clustering statistics for the 4 algorithms compared in Fig. 1. X-axis denotes which modules are taken as the reference set. f-h, Pairwise module overlaps between 3 of the 4 algorithms compared in Fig. 1 (GLASSO yielded too many modules to visualize here). i, t-SNE embedding of gene features from whole-brain tensor decomposition, colored by DBSCAN clusters. j, As (i), but colored and annotated with whole-brain modules. k,l, Overlap between whole-brain consensus and tensor-decomposition+DBSCAN modules. Color scheme as in (f-h). m, Standard boxplot (box: quartiles, whiskers: 1.5xIQR, N = 10 bootstrap re-samplings) of within-module recall values for hub-gene co-clustering, demonstrating that at 100 samples, the recall is above 50% for most modules.

Extended Data Fig. 2 Exploration of cell-type modules (related to Fig. 2).

a, Cell-type marker enrichment for brain-wide modules, extended with markers of microglial activation and deactivation, and markers of reactive gliosis and A1/A2 reactive astrocytes. b, Plots of the marginal rate (solid: mean, shade: 95% CI of GAM) of LoF-intolerant (pLI>0.9) genes, as a function of BW-M1 (most enriched) and BW-M2 (most depleted) kME. c, Gene ontology enrichment for BW-M5. d, Marginal LoF-intolerance rates (solid: mean, shade: 95% CI of GAM), by gene kME, for neuronal subtype modules. e,f, Standard boxplots (box: quartiles, whiskers: 1.5xIQR) of module mean topological overlap, and gene expression, for 5 whole-brain modules in ASD cases and matched controls (Parikshak 2016). The case/control difference in lncRNA is closely matched by the same difference in randomly-selected, matched coding genes. g, LoF-intolerance enrichment for neuronal subtype modules, using pLI and o/e bins as response variables, and a linear model correcting for gene GC and length (logit link, p-values: coefficient T-test). All modules except BROD-M8 show strong enrichment, and BROD-M8 shows enrichment when using soft-membership instead of hard membership.

Extended Data Fig. 3 Glial cell-type isoforms (related to Fig. 3).

a, Replicate of main Fig. 3(b) in astrocytes, showing a strong positive relationship between astrocyte module membership, and relative expression in astrocyte cells. b-e, Relationship between module kME and cell type relative expression for transcripts across 4 neuron/astrocyte isoform switch genes, demonstrating concordance between high kME, and high relative expression. f, Unsigned Fisher’s exact test of the contingency of ‘assigned to module’ and ‘top-ranked cell type marker’ for varying kME thresholds for (left) oligodendrocytes and (right) astrocytes; for marker rankings based on both absolute and relative expression within the cell-sorted data. Thresholds in the range 0.45-0.55 appear to balance significance and odds ratio across absolute and relative rankings.

Extended Data Fig. 4 Validation of neuron-astrocyte isoform switching (related to Fig. 3).

(left) Unmodified Western Blot corresponding to Fig. 3 (right) Same blot, annotated with source of input material and band identities.

Extended Data Fig. 5 Overlap with published modules containing disease genes.

a–d, Overlaps between published modules and the consensus whole-brain co-expression modules identified in this paper, demonstrating that the majority of modules show a high overlap, particularly to the neuronal module BW-M4. P-values: signed Fisher’s exact test. These modules were been selected because of published enrichment for neuropsychiatric disease risk genes. (see Methods).

Extended Data Fig. 6 Module BW-M4 functional annotation (related to Fig. 5).

a, Signed gene ontology enrichments (logistic regression controlling for gene length and GC, p-value from coefficient T-test) for MAGMA-significant ASD genes in module set BW-M4 across all regions in which a BW-M4 module is present. Bar color reflects the enrichment odds ratio (black: 5, light blue: 25); outline color reflects the GWAS source (pink: Grove et al., 2017; green: iPsych (Robinson et al., Nat. Genet. 2016), blue: PGC-2017). b, Meta-GSEA scores for significant MAGMA genes in BW-M4 across all tissues, implicating synaptic transmission and calcium transport as sources of neuronal dysfunction in SCZ. Red line denotes FDR significance level.

Extended Data Fig. 7 Regional AUPR curves for CEREB-M1 and CTX-M1.

Nearest-neighbor precision-recall curves for CEREB-M1 labels across all region-level co-expression networks; showing significantly higher AUPR for cerebellar regions, but substantial AUPR for all remaining regions. Right. Nearest-neighbor precision-recall curves for CTX-M3.

Extended Data Fig. 8 Exploration of an omnigenic-like model across different networks (related to Fig. 7).

Plot of Phi statistics for InWeb brain PPI network (‘PPI’) and four regulatorycircuits.org (‘RC’) networks: Hippocampus (‘Hippo’), amygdala (‘Amy’), NEU+ neurons, astrocytes, and neuroprogenitor cells (‘NPC’). Vertical breaks represent the study used to calculate phi, while the colors represent those studies used to define proposed core genes, or network central genes.

Supplementary information

Supplementary Information

Supplementary Note.

Reporting Summary

Supplementary Table 1

Per-region and multiregional module definitions and gene-module kME values.

Supplementary Table 2

Pathway and cell marker enrichments for regional and multiregional modules.

Supplementary Table 3

Correlations between module eigengenes and nonnegative matrix factorization loadings onto PsychEncode factors.

Supplementary Table 4

Table of gene kME values with pLI and overexpression values from ExAC.

Supplementary Table 5

Table of lncRNAs, their inferred module from xgboost-based projection, expression in cell types from single-cell data, and differential expression statistics in neuropsychiatric disease.

Supplementary Table 6

Table of isoform quantification correlation to total-expression modules across all regions. This is a very large file and may be difficult to download or open; please contact the authors for alternative formats.

Supplementary Table 7

RCT results for regional and multiregional contrasts.

Supplementary Table 8

Table of significant overlapping odds ratios and P values between whole-brain modules and previously published disease modules.

Supplementary Table 9

Phi statistics and P values for blood, brain and developing brain networks.

Supplementary Table 10

Preservation statistics for GLASSO, vMF and ARACNe–produced modules in orthogonal microarray datasets.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hartl, C.L., Ramaswami, G., Pembroke, W.G. et al. Coexpression network architecture reveals the brain-wide and multiregional basis of disease susceptibility. Nat Neurosci 24, 1313–1323 (2021). https://doi.org/10.1038/s41593-021-00887-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-021-00887-5

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