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.

A myelin-related transcriptomic profile is shared by Pitt–Hopkins syndrome models and human autism spectrum disorder

Abstract

Autism spectrum disorder (ASD) is genetically heterogeneous with convergent symptomatology, suggesting common dysregulated pathways. In this study, we analyzed brain transcriptional changes in five mouse models of Pitt–Hopkins syndrome (PTHS), a syndromic form of ASD caused by mutations in the TCF4 gene, but not the TCF7L2 gene. Analyses of differentially expressed genes (DEGs) highlighted oligodendrocyte (OL) dysregulation, which we confirmed in two additional mouse models of syndromic ASD (Ptenm3m4/m3m4 and Mecp2tm1.1Bird). The PTHS mouse models showed cell-autonomous reductions in OL numbers and myelination, functionally confirming OL transcriptional signatures. We also integrated PTHS mouse model DEGs with human idiopathic ASD postmortem brain RNA-sequencing data and found significant enrichment of overlapping DEGs and common myelination-associated pathways. Notably, DEGs from syndromic ASD mouse models and reduced deconvoluted OL numbers distinguished human idiopathic ASD cases from controls across three postmortem brain data sets. These results implicate disruptions in OL biology as a cellular mechanism in ASD pathology.

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: RNA-seq of mice with multiple Tcf4 mutations reveals age-specific differential gene expression.
Fig. 2: OL-specific deficits in PTHS model mice.
Fig. 3: Shared myelination gene regulation between mouse models of syndromic ASD.
Fig. 4: Validation of myelination defects due to Tcf4 mutation.
Fig. 5: The proportion of myelinated axons in the CC is reduced in Tcf4-mutant mice.
Fig. 6: In vitro biological validation of myelination defects due to Tcf4 mutation.
Fig. 7: TCF4 regulation of OLs is cell autonomous.
Fig. 8: Human–mouse convergence of gene expression in idiopathic and syndromic ASD.

Data availability

RNA-seq data from the cortex and whole brain of Tcf4-mutant mice are available via Globus: http://research.libd.org/globus/NatNeuro_TCF4_Data/ and at BioProject under accession number PRJNA601252.

Code availability

R code used to analyze data in this study and analyzed data are available at https://github.com/LieberInstitute/PTHS_mouse.

References

  1. Iossifov, I. et al. The contribution of de novo coding mutations to autism spectrum disorder. Nature 515, 216–221 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. 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  CAS  Google Scholar 

  3. Gaugler, T. et al. Most genetic risk for autism resides with common variation. Nat. Genet. 46, 881–885 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Silverman, J. L., Yang, M., Lord, C. & Crawley, J. N. Behavioural phenotyping assays for mouse models of autism. Nat. Rev. Neurosci. 11, 490–502 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sweatt, J. D. Pitt–Hopkins syndrome: intellectual disability due to loss of TCF4-regulated gene transcription. Exp. Mol. Med. 45, e21 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Rannals, M. D. et al. Psychiatric risk gene transcription factor 4 regulates intrinsic excitability of prefrontal neurons via repression of scn10a and KCNQ1. Neuron 90, 43–55 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. D’Rozario, M. et al. Type I bHLH proteins daughterless and Tcf4 restrict neurite branching and synapse formation by repressing neurexin in postmitotic neurons. Cell Rep. 15, 386–397 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Page, S. C. et al. The schizophrenia- and autism-associated gene, transcription factor 4 regulates the columnar distribution of layer 2/3 prefrontal pyramidal neurons in an activity-dependent manner. Mol. Psychiatry 23, 304–315 (2018).

    Article  CAS  PubMed  Google Scholar 

  9. Sepp, M. et al. The intellectual disability and schizophrenia associated transcription factor TCF4 is regulated by neuronal activity and protein kinase A. J. Neurosci. 37, 10516–10527 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kennedy, A. J. et al. Tcf4 regulates synaptic plasticity, DNA methylation, and memory function. Cell Rep. 16, 2666–2685 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Thaxton, C. et al. Common pathophysiology in multiple mouse models of Pitt–Hopkins syndrome. J. Neurosci. 38, 918–936 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Rubenstein, J. L. R. & Merzenich, M. M. Model of autism: increased ratio of excitation/inhibition in key neural systems. Genes Brain Behav. 2, 255–267 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Courchesne, E. & Pierce, K. Why the frontal cortex in autism might be talking only to itself: local over-connectivity but long-distance disconnection. Curr. Opin. Neurobiol. 15, 225–230 (2005).

    Article  CAS  PubMed  Google Scholar 

  14. Ismail, M. M. T. et al. Studying autism spectrum disorder with structural and diffusion magnetic resonance imaging: a survey. Front. Hum. Neurosci. 10, 211 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Pagnozzi, A. M., Conti, E., Calderoni, S., Fripp, J. & Rose, S. E. A systematic review of structural MRI biomarkers in autism spectrum disorder: a machine learning perspective. Int. J. Dev. Neurosci. 71, 68–82 (2018).

    Article  PubMed  Google Scholar 

  16. Zikopoulos, B. & Barbas, H. Changes in prefrontal axons may disrupt the network in autism. J. Neurosci. 30, 14595–14609 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Rannals, M. D. et al. Neurodevelopmental models of transcription factor 4 deficiency converge on a common ion channel as a potential therapeutic target for Pitt–Hopkins syndrome. Rare Dis. 4, e1220468 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Willsey, A. J. et al. Coexpression networks implicate human midfetal deep cortical projection neurons in the pathogenesis of autism. Cell 155, 997–1007 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Tilot, A. K. et al. Neural transcriptome of constitutional Pten dysfunction in mice and its relevance to human idiopathic autism spectrum disorder. Mol. Psychiatry 21, 118–125 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Gabel, H. W. et al. Disruption of DNA-methylation-dependent long gene repression in Rett syndrome. Nature 522, 89–93 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Fraser, M. M., Bayazitov, I. T., Zakharenko, S. S. & Baker, S. J. Phosphatase and tensin homolog, deleted on chromosome 10 deficiency in brain causes defects in synaptic structure, transmission and plasticity, and myelination abnormalities. Neuroscience 151, 476–488 (2008).

    Article  CAS  PubMed  Google Scholar 

  22. Sharma, K., Singh, J., Pillai, P. P. & Frost, E. E. Involvement of MeCP2 in regulation of myelin-related gene expression in cultured rat oligodendrocytes. J. Mol. Neurosci. 57, 176–184 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Olmos-Serrano, J. L. et al. Down syndrome developmental brain transcriptome reveals defective oligodendrocyte differentiation and myelination. Neuron 89, 1208–1222 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Marques, S. et al. Oligodendrocyte heterogeneity in the mouse juvenile and adult central nervous system. Science 352, 1326–1329 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Chen, Y. et al. Isolation and culture of rat and mouse oligodendrocyte precursor cells. Nat. Protoc. 2, 1044–1051 (2007).

    Article  CAS  PubMed  Google Scholar 

  26. 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 

  27. Satterstrom, F. K. et al. Large-scale exome sequencing study implicates both developmental and functional changes in the neurobiology of autism. Preprint at bioRxiv https://doi.org/10.1101/484113 (2018).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wright, C. et al. Altered expression of histamine signaling genes in autism spectrum disorder. Transl. Psychiatry 7, e1126 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Jaffe, A. E. et al. Developmental and genetic regulation of the human cortex transcriptome illuminate schizophrenia pathogenesis. Nat. Neurosci. 21, 1117–1125 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Petrelli, F., Pucci, L. & Bezzi, P. Astrocytes and microglia and their potential link with autism spectrum disorders. Front. Cell Neurosci. 10, 21 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Suzuki, K. et al. Microglial activation in young adults with autism spectrum disorder. JAMA Psychiatry 70, 49–58 (2013).

    Article  PubMed  Google Scholar 

  35. Velmeshev, D. et al. Single-cell genomics identifies cell type-specific molecular changes in autism. Science 364, 685–689 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ercan, E. et al. Neuronal CTGF/CCN2 negatively regulates myelination in a mouse model of tuberous sclerosis complex. J. Exp. Med. 214, 681–697 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Pacey, L. K. K. et al. Delayed myelination in a mouse model of fragile X syndrome. Hum. Mol. Genet. 22, 3920–3930 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Scott, R. et al. Loss of Cntnap2 causes axonal excitability deficits, developmental delay in cortical myelination, and abnormal stereotyped motor behavior. Cereb. Cortex 29, 586–597 (2019).

    Article  PubMed  Google Scholar 

  39. Jung, H. et al. Sexually dimorphic behavior, neuronal activity, and gene expression in Chd8-mutant mice. Nat. Neurosci. 21, 1218–1228 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. Bagot, R. C. et al. Circuit-wide transcriptional profiling reveals brain region-specific gene networks regulating depression susceptibility. Neuron 90, 969–983 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Jin, C. et al. Integrative brain transcriptome analysis reveals region-specific and broad molecular changes in Shank3-overexpressing mice. Front. Mol. Neurosci. 11, 250 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Zhao, C. et al. Dual requirement of CHD8 for chromatin landscape establishment and histone methyltransferase recruitment to promote CNS myelination and repair. Dev. Cell 45, 753–768 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Deoni, S. C. L. et al. White-matter relaxation time and myelin water fraction differences in young adults with autism. Psychol. Med. 45, 795–805 (2015).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Modi, M. E. & Sahin, M. Translational use of event-related potentials to assess circuit integrity in ASD. Nat. Rev. Neurol. 13, 160–170 (2017).

    Article  CAS  PubMed  Google Scholar 

  46. Carmody, D. P. & Lewis, M. Regional white matter development in children with autism spectrum disorders. Dev. Psychobiol. 52, 755–763 (2010).

    Article  PubMed  Google Scholar 

  47. Broek, J. A., Guest, P. C., Rahmoune, H. & Bahn, S. Proteomic analysis of post mortem brain tissue from autism patients: evidence for opposite changes in prefrontal cortex and cerebellum in synaptic connectivity-related proteins. Mol. Autism 5, 41 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Butler, M. G. et al. Subset of individuals with autism spectrum disorders and extreme macrocephaly associated with germline PTEN tumour suppressor gene mutations. J. Med. Genet. 42, 318–321 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Renieri, A. et al. Diagnostic criteria for the Zappella variant of Rett syndrome (the preserved speech variant). Brain Dev. 31, 208–216 (2009).

    Article  CAS  PubMed  Google Scholar 

  50. Monje, M. Myelin plasticity and nervous system function. Annu. Rev. Neurosci. 41, 61–76 (2018).

    Article  CAS  PubMed  Google Scholar 

  51. Zhuang, Y., Cheng, P. & Weintraub, H. B-lymphocyte development is regulated by the combined dosage of three basic helix-loop-helix genes, E2A, E2-2, and HEB. Mol. Cell. Biol. 16, 2898–2905 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Bergqvist, I. et al. The basic helix-loop-helix transcription factor E2-2 is involved in T lymphocyte development. Eur. J. Immunol. 30, 2857–2863 (2000).

    Article  CAS  PubMed  Google Scholar 

  53. Zawadzka, M. et al. CNS-resident glial progenitor/stem cells produce Schwann cells as well as oligodendrocytes during repair of CNS demyelination. Cell Stem Cell 6, 578–590 (2010).

    Article  CAS  PubMed  Google Scholar 

  54. Goebbels, S. et al. Elevated phosphatidylinositol 3,4,5-trisphosphate in glia triggers cell-autonomous membrane wrapping and myelination. J. Neurosci. 30, 8953–8964 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Maher, B. J. & LoTurco, J. J. Disrupted-in-schizophrenia (DISC1) functions presynaptically at glutamatergic synapses. PLoS One 7, e34053 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Leek, J. T. svaseq: removing batch effects and other unwanted noise from sequencing data. Nucleic Acids Res. 42, e161 (2014).

    Article  PubMed Central  CAS  Google Scholar 

  60. Yu, G., Wang, L.-G., Han, Y. & He, Q.-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Xu, X., Wells, A. B., O’Brien, D. R., Nehorai, A. & Dougherty, J. D. Cell type-specific expression analysis to identify putative cellular mechanisms for neurogenetic disorders. J. Neurosci. 34, 1420–1431 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Zhang, Y. et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Newman, A. M. et al. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods 12, 453–457 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Abrahams, B. S. et al. SFARI Gene 2.0: a community-driven knowledgebase for the autism spectrum disorders (ASDs). Mol. Autism 4, 36 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Banerjee-Basu, S. & Packer, A. SFARI Gene: an evolving database for the autism research community. Dis. Model. Mech. 3, 133–135 (2010).

    Article  PubMed  Google Scholar 

  66. Durinck, S., Spellman, P. T., Birney, E. & Huber, W. Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt. Nat. Protoc. 4, 1184–1191 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Jaffe, A. E. et al. qSVA framework for RNA quality correction in differential expression analysis. Proc. Natl Acad. Sci. USA 114, 7130–7135 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Jaffe, A. E. et al. Practical impacts of genomic data ‘cleaning’ on biological discovery using surrogate variable analysis. BMC Bioinformatics 16, 372 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  69. 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  CAS  PubMed  PubMed Central  Google Scholar 

  70. Houseman, E. A. et al. DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinformatics 13, 86 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful for the vision and generosity of the Lieber and Maltz families, who made this work possible. We thank D.R. Weinberger for his helpful comments and feedback. We thank the Johns Hopkins School of Medicine Microscope Core Facility and specifically L. Roker for generating TEM images of CC used in this study. This work was supported by the Lieber Institute for Brain Development, the Pitt–Hopkins Research Foundation Awards (to B.J.M., B.D.P., C.T., D.S. and A.J.K.), National Institute of Mental Health (NIMH) grant R56MH104593 (to B.J.M.), NIMH grant R01MH110487 (to B.J.M.), a Johns Hopkins PURA grant (to B.N.P.), UPenn Orphan Disease Center Million Dollar Bike Ride grant MDBR-15-108-PH (to B.D.P. and C.T.), NARSAD Young Investigator grant 20653 from the Brain Behavior Research Foundation (to C.T.), National Institute of Neurological Disorders and Stroke grant P30NS045892 (to J.M.S.), National Institute of Child Health and Human Development grant P30HD03110 (to J.M.S.), NIMH grant R01MH104158 (to D.S. and A.J.K.), National Institute of General Medical Sciences training grant T32GM008208 (to B.N.P.) and NIMH training grant T32MH015330 (to B.A.D.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

B.N.P. and A.E.J. performed RNA-seq analysis. H.K., S.C.P. and M.N.C. collected samples and performed qRT–PCR and western blot experiments. J.F.B., B.A.D., S.R.S., H.L.S. and B.M. performed western blot, ICC, IHC and EM experiments. Z.Y. and H.Y.C. performed electrophysiology experiments. D.G. performed animal husbandry and genotyping. C.L.T., J.M.S., A.J.K., J.D.S. and B.D.P. contributed RNA-seq datasets and mouse models. J.H.S. performed RNA sequencing. E.E.B. contributed to RNA-seq data processing. B.N.P., J.F.B, A.E.J. and B.J.M. contributed to experimental design, data analysis and writing. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Andrew E. Jaffe or Brady J. Maher.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 Heterozygous truncation of Tcf4 decreases levels of Tcf4 mRNA and protein.

Comparison of lifespan expression patterns of TCF4 in heterozygous (Tcf4tr/+) mice and wild-type (Tcf4+/+) littermates in qRT-PCR and RNA-sequencing analyses. mRNA and protein were extracted from frontal cortex of mice across developmental ages. (A) qRT-PCR analysis of full-length Tcf4 transcripts from mouse frontal cortex. Tcf4tr/+ mice show overall reduced expression compared to Tcf4+/+ mice (n = 66 mice, ANOVA p = 0.02) with the greatest decrease in Tcf4 expression around postnatal days 1–4 (P1–4, n = 18 mice, Posthoc p<0.05). (B) RNA-seq analysis also shows Tcf4 expression decreased in the Tcf4tr/+ mouse in the exon after the truncation (n = 35 mice). Tcf4tr/+ mice had significant decrease of Tcf4 exons (differentially expressed exon by genotype FDR = 3.35 × 10−35). The boxplot shows the quartile breaks of residualized variance stablilized count of a Tcf4 exon after the truncation (see methods on residualization for visual interpretation). (C) Western blot of endogenous mouse TCF4 at three ages (E12, P1, and P42). A single full-length (TCF4 fl; 80kDa) protein is observed in lysates from Tcf4+/+mouse brain and Tcf4tr/+ mouse brain expresses a truncated (TCF4 tr) and full-length TCF4 protein. These representative gel images are compiled across several different gel images and stitched together. (D) Full-length TCF4 protein is decreased in the Tcf4tr/+ mouse brain (n = 3 mice per genotype per timepoint, pAnova = 0.0009) with the largest effect occuring at P1 in the TCF4tr/+ mice (n = 3 mice per condition, two-sided unpaired t-test, p<0.01). Center values indicate mean and errors bars are the S.E.M., *<0.05, **<0.01, ***< 0.001.

Extended Data Fig. 2 Replicated differential expression across PTHS models.

a, Table of DEGs (NP1 = 28, NAdult = 69, using the two-sided differential expression cutoff of FDR<0.05) and percent of differential expression replication across different forms of Tcf4 mutation P1 and adult mice. Most DEGs and replication occur in adult mice. The replication rate was defined as the proportion/percentage of DE genes that were p < 0.01 in at least one other mouse model of the same age group divided by those DEGs in the reference mouse model. b, Differential expression log2 fold-change heatmap comparing replicated DEGs across various models of Tcf4 mutations in P1 (replication defined the same gene having differential expression with two-sided p<0.05).

Extended Data Fig. 3 Cell type-specific expression analysis in PTHS mice.

Bulls-eye plots from CSEA analysis of DEGs in (a) P1 and (b) adult Tcf4tr/+ mice. The bulls-eye plot size is scaled to the number of genes specific to a cell type at increasing levels of specificity as published by Xu et al., 201461. The FDR-adjusted hypergeometric test p-value is plot for each level of specificity, with unenriched groups colored gray. Cell type bulls-eye plots are arranged by hierarchical distance of their specific gene expression levels. (a) P1 DEGs (N = 36 DEG at Padj < 0.05) enriched for D1+, D2+, and cholinergic neurons (Padj<0.05). (b) Adult DEGs (N = 1832 DEG at Padj < 0.05) strongly enrich for OLs among other neuronal cell types.

Extended Data Fig. 4 Analysis of TEM images.

a, Plot of gRatio and corresponding radius for all axons assessed. Axon radius is significantly correlated with gRatio (p = 2.86e-34), and this correlation is different by genotype (p = 0.03). (b–f) No significant differences were observed between genotypes for gRatio (p = 0.796), axon area (p = 0.844), myelin area (p = 0.852), myelin + axon radius (p = 0.615), or axon radius (p = 0.873).

Extended Data Fig. 5 Conduction velocity does not differ between TCF4 genotypes.

The peak time of N1 and N2 waveform (y-axis) is the amount of time between stimulation artifact and the amplitude peak of the compound action potential. The peak time is plotted against distance (x-axis) which is the distance between the stimulating electrode and recording electrode. The slope of the line generated from both N1(a) and N2 (b) does not differ between genotypes (N1 slope p = 0.96, N2 slope p = 0.36, N = 30 slices from 4 Tcf4+/+ and 5 Tcf4tr/+ mice) Center values indicate mean and errors bars are the S.E.M.

Extended Data Fig. 6 Tcf4 is abundantly expressed at all stages of oligodendrocyte development.

a, Example images of fluorescent in situ hybridization showing Tcf4 transcript co-localizes with both Pdgfrα and Mbp. b, Summary plots of single-cell RNA-seq data across oligodendrocyte development showing expression levels for Pdgfrα, Tcf4, Olig2, and Mbp. This data was adapted from Marques et al.24.

Extended Data Fig. 7 Primary OL cultures are devoid of neurons and astrocytes.

a, Primary neuronal culture stained with CNP and GFAP as a positive control for antibody staining. b1, Primary OL culture stained with CNP and GFAP. b2, Cell counts showing primary OL cultures have very few neurons (Tuj1+) or astrocytes (GFAP+). Numbers indicate number of cells counted for that condition. c, Primary neuronal cultures stained with OLIG2, NeuN, and GFAP as a positive control for antibody staining. d, Primary OL culture stained with OLIG2, NeuN, and GFAP. d1, Cell counts showing primary OL cultures have very few neurons (NeuN+) or astrocytes (GFAP+).

Extended Data Fig. 8 OPCs derived from Tcf4tr/+ mice show inefficient maturation into oligodendrocytes.

a, Representative images of OPCs (PDGFRα) and mature OLs (MBP, CNP) derived from Tcf4+/+ and Tcf4tr/+ mice. To control for cell numbers all cell counts are normalized by the pan-OL marker Olig2 that labels both OPC and mature OLs. Tcf4tr/+ produce significantly more OPCs (n = 23 mice, two-tailed unpaired t-test, p<0.0001) and fewer MBP positive OLs (Tcf4+/+ 0.19 ± 0.02 vs. Tcf4tr/+ 0.05 ± 0.01, n = 23 mice, two-tailed unpaired t-test, p<0.0001). b, Representative images of OPCs (PDGFRα) and mature OLs (CNP). Tcf4tr/+ produce significantly more OPCs (two-tailed unpaired t-test, n = 17 mice, two-tailed unpaired t-test, p<0.0001) and fewer CNP positive OLs (Tcf4+/+ 0.56 ± 0.04 vs. Tcf4tr/+ 0.22 ± 0.02, n = 17 mice, two-tailed unpaired t-test, p<0.0001). All scale bars equal 100µm. For all bar graphs, center values represent the mean and error bars are S.E.M.,***p<0.001, ****p<0.0001.

Extended Data Fig. 9 Concordant gene regulation between PTHS mice and human ASD.

Comparison of differential expression in adult PTHS mice with human ASD and 15q duplication (15q Dup) in postmortem frontal, temporal, and cerebellum. (NTemp = 68, NFrontal = 73, NVermis = 63, Human two-sided differential expression p<0.05, mouse DEGs, FDR<0.01). a, Log2 fold-change comparison of adult PTHS mouse DEGs replicated in human ASD and 15q Dup in each tissue region (p<0.05). Gene regulation in PTHS mice cluster closest with ASD differential expression in cortex. b, More than 50% of replicated PTHS DEGs had concordant fold-change directionality. Null permutation for empirical p-value significance of human-mouse gene fold-change concordance from 1000 permutations are reported (Two-sided Fisher’s exact test, *, padj<0.05; **, padj<0.01; *** padj<0.001). c, Replicated DEGs in ASD and 15q Dup are significantly enriched in all tissues, mostly in the cortex (FDR-adjusted Fisher Exact test for overlap of Tcf4 mouse DEG with ASD DEG, padj<0.05). d, Venn diagram showing overlap of PTHS mouse DEGs with human ASD or 15q Dup in cortical tissues. e, Gene ontology analysis shows tissue-specific biological processes and cellular components between overlap of PTHS mouse and human ASD or 15q Dup (NASD = 10896 and N15qDup = 13149 DEGs at p < 0.01,s q-adjusted two-sided hypergeometric test). The gene sets are largely brain region specific and concordant between human ASD and 15q Dup. The color of the dot plots shows the q-adjusted hypergeometric test p-value for gene set enrichment of the DEG of each diagnosis group.

Extended Data Fig. 10 Mouse concordant ASD genes (CAGs) are not convergent with Schizophrenia or Down Syndrome.

a, The eigengene of the CAGs found across the three models of syndromic ASD explains 65.8% of the gene expression variance and is not associated with Schizophrenia diagnosis (linear regression two-sided p-value = 0.538). b, The eigengene of the CAGs found across the three models of syndromic ASD explains 53.2% of the gene expression variance and is not associated with Down Syndrome diagnosis (linear regression two-sided p-value = 0.34). c, Estimated cellular composition differences between individuals with schizophrenia and controls using reference-based deconvolution. There were significant increases of astrocytes (p = 0.0002) and endothelial cells (p = 0.0118) and decreases in microglia (p = 0.0076) in individuals with schizophrenia compared to controls using linear regression analysis.

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1–8.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Phan, B.N., Bohlen, J.F., Davis, B.A. et al. A myelin-related transcriptomic profile is shared by Pitt–Hopkins syndrome models and human autism spectrum disorder. Nat Neurosci 23, 375–385 (2020). https://doi.org/10.1038/s41593-019-0578-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-019-0578-x

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