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:

Widespread RNA editing dysregulation in brains from autistic individuals

Abstract

Transcriptomic analyses of postmortem brains have begun to elucidate molecular abnormalities in autism spectrum disorder (ASD). However, a crucial pathway involved in synaptic development, RNA editing, has not yet been studied on a genome-wide scale. Here we profiled global patterns of adenosine-to-inosine (A-to-I) editing in a large cohort of postmortem brains of people with ASD. We observed a global bias for hypoediting in ASD brains, which was shared across brain regions and involved many synaptic genes. We show that the Fragile X proteins FMRP and FXR1P interact with RNA-editing enzymes (ADAR proteins) and modulate A-to-I editing. Furthermore, we observed convergent patterns of RNA-editing alterations in ASD and Fragile X syndrome, establishing this as a molecular link between these related diseases. Our findings, which are corroborated across multiple data sets, including dup15q (genomic duplication of 15q11.2-13.1) cases associated with intellectual disability, highlight RNA-editing dysregulation in ASD and reveal new mechanisms underlying this disorder.

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: Transcriptome-wide differential editing in the frontal cortex of subjects with ASD.
Fig. 2: Global analysis reveals potential regulators of differential editing in the frontal cortex of ASD.
Fig. 3: FMRP and FXR1P regulate RNA editing.
Fig. 4: Transcriptome-wide differential editing in the frontal cortex of Fragile X subjects and controls.
Fig. 5: RNA editing dysregulation in different brain regions.
Fig. 6: Hypoediting in three brain regions of dup15q subjects.

Similar content being viewed by others

Data availability

eCLIP-seq data on FMRP and FXR1P from postmortem human brain have been deposited in GEO with accession code GSE107895. RNA-seq data of Fragile X subjects, carriers and controls have been deposited in GEO with accession codes GSE107867 (NeuroBiobank data set) and GSE117776 (UC Davis FXTAS data set). Fastq files of RNA-seq from the idiopathic ASD, dup15q and control brains were obtained from our previous study9 and are available in the PsychENCODE website (https://www.synapse.org//#!Synapse:syn4921369/wiki/235539). Fastq files of RNA-seq data from the replicate ASD and control cohort are available in GEO (accession GSE51264 / GSE59288).

References

  1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders 4th edn, text rev. (APA Publishing, Washington, DC, 2000).

    Google Scholar 

  2. Rojas, D. C. The role of glutamate and its receptors in autism and the use of glutamate receptor antagonists in treatment. J. Neural Transm. (Vienna) 121, 891–905 (2014).

    Article  CAS  Google Scholar 

  3. Guo, Y. P. & Commons, K. G. Serotonin neuron abnormalities in the BTBR mouse model of autism. Autism Res. 10, 66–77 (2017).

    Article  PubMed  Google Scholar 

  4. Ha, S., Sohn, I. J., Kim, N., Sim, H. J. & Cheon, K. A. Characteristics of brains in autism spectrum disorder: structure, function and connectivity across the lifespan. Exp. Neurobiol. 24, 273–284 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  5. 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 

  6. Marchetto, M. C. et al. Altered proliferation and networks in neural cells derived from idiopathic autistic individuals. Mol. Psychiatry 22, 820–835 (2017).

    Article  CAS  PubMed  Google Scholar 

  7. de la Torre-Ubieta, L., Won, H., Stein, J. L. & Geschwind, D. H. Advancing the understanding of autism disease mechanisms through genetics. Nat. Med. 22, 345–361 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Gupta, S. et al. Transcriptome analysis reveals dysregulation of innate immune response genes and neuronal activity-dependent genes in autism. Nat. Commun. 5, 5748 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. 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 

  10. Irimia, M. et al. A highly conserved program of neuronal microexons is misregulated in autistic brains. Cell 159, 1511–1523 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wu, Y. E., Parikshak, N. N., Belgard, T. G. & Geschwind, D. H. Genome-wide, integrative analysis implicates microRNA dysregulation in autism spectrum disorder. Nat. Neurosci. 19, 1463–1476 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Nishikura, K. A-to-I editing of coding and non-coding RNAs by ADARs. Nat. Rev. Mol. Cell Biol. 17, 83–96 (2016).

    Article  CAS  PubMed  Google Scholar 

  13. Behm, M. & Öhman, M. RNA editing: a contributor to neuronal dynamics in the mammalian brain. Trends Genet. 32, 165–175 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. Slotkin, W. & Nishikura, K. Adenosine-to-inosine RNA editing and human disease. Genome Med. 5, 105 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Khermesh, K. et al. Reduced levels of protein recoding by A-to-I RNA editing in Alzheimer’s disease. RNA 22, 290–302 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Eran, A. et al. Comparative RNA editing in autistic and neurotypical cerebella. Mol. Psychiatry 18, 1041–1048 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Iossifov, I. et al. De novo gene disruptions in children on the autistic spectrum. Neuron 74, 285–299 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. 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 

  19. Bahn, J. H. et al. Accurate identification of A-to-I RNA editing in human by transcriptome sequencing. Genome Res. 22, 142–150 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Porath, H. T., Carmi, S. & Levanon, E. Y. A genome-wide map of hyper-edited RNA reveals numerous new sites. Nat. Commun. 5, 4726 (2014).

    Article  CAS  PubMed  Google Scholar 

  21. Picardi, E., D’Erchia, A. M., Lo Giudice, C. & Pesole, G. REDIportal: a comprehensive database of A-to-I RNA editing events in humans. Nucleic Acids Res. 45,(D1), D750–D757 (2017).

    Article  Google Scholar 

  22. Levanon, E. Y. et al. Systematic identification of abundant A-to-I editing sites in the human transcriptome. Nat. Biotechnol. 22, 1001–1005 (2004).

    Article  CAS  PubMed  Google Scholar 

  23. Oakes, E., Anderson, A., Cohen-Gadol, A. & Hundley, H. A. Adenosine deaminase that acts on RNA 3 (ADAR3) binding to glutamate receptor subunit B pre-mRNA inhibits RNA editing in glioblastoma. J. Biol. Chem. 292, 4326–4335 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Rakic, P. Evolution of the neocortex: a perspective from developmental biology. Nat. Rev. Neurosci. 10, 724–735 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. 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 

  26. Hwang, T. et al. Dynamic regulation of RNA editing in human brain development and disease. Nat. Neurosci. 19, 1093–1099 (2016).

    Article  CAS  PubMed  Google Scholar 

  27. Liu, X. et al. Disruption of an evolutionarily novel synaptic expression pattern in autism. PLoS Biol. 14, e1002558 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  29. Davis, J. K. & Broadie, K. Multifarious functions of the Fragile X mental retardation protein. Trends Genet. 33, 703–714 (2017).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhang, Y. et al. The Fragile X mental retardation syndrome protein interacts with novel homologs FXR1 and FXR2. EMBO J. 14, 5358–5366 (1995).

  32. Van Nostrand, E. L. et al. Robust transcriptome-wide discovery of RNA-binding protein binding sites with enhanced CLIP (eCLIP). Nat. Methods 13, 508–514 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Ascano, M. et al. FMRP targets distinct mRNA sequence elements to regulate protein expression. Nature 492, 382–386 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Vasudevan, S. & Steitz, J. A. AU-rich-element-mediated upregulation of translation by FXR1 and Argonaute 2. Cell 128, 1105–1118 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Van Nostrand, E. L. et al. A large-scale binding and functional map of human RNA binding proteins. Preprint at biorXiv https://www.biorxiv.org/content/early/2017/08/23/179648 (2017).

  36. Hagerman, R., Hoem, G. & Hagerman, P. Fragile X and autism: intertwined at the molecular level leading to targeted treatments. Mol. Autism 1, 12 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Abbeduto, L., McDuffie, A. & Thurman, A. J. The fragile X syndrome-autism comorbidity: what do we really know? Front. Genet. 5, 355 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Pinto, Y., Cohen, H. Y. & Levanon, E. Y. Mammalian conserved ADAR targets comprise only a small fragment of the human editosome. Genome. Biol. 15, R5 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Irimia, M. et al. Evolutionarily conserved A-to-I editing increases protein stability of the alternative splicing factor Nova1. RNA Biol. 9, 12–21 (2012).

    Article  CAS  PubMed  Google Scholar 

  40. DiStefano, C. et al. Identification of a distinct developmental and behavioral profile in children with Dup15q syndrome. J. Neurodev. Disord. 8, 19 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Battaglia, A. et al. The inv dup(15) syndrome: a clinically recognizable syndrome with altered behavior, mental retardation, and epilepsy. Neurology 48, 1081–1086 (1997).

    Article  CAS  PubMed  Google Scholar 

  42. Frith, C. & Dolan, R. The role of the prefrontal cortex in higher cognitive functions. Brain. Res. Cogn. Brain Res. 5, 175–181 (1996).

  43. Jansen, A. et al. Gene-set analysis shows association between FMRP targets and autism spectrum disorder. Eur. J. Hum. Genet. 25, 863–868 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Pinto, D. et al. Convergence of genes and cellular pathways dysregulated in autism spectrum disorders. Am. J. Hum. Genet. 94, 677–694 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Fatemi, S. H. & Folsom, T. D. Dysregulation of fragile X mental retardation protein and metabotropic glutamate receptor 5 in superior frontal cortex of individuals with autism: a postmortem brain study. Mol. Autism 2, 6 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Patzlaff, N. E., Nemec, K. M., Malone, S. G., Li, Y. & Zhao, X. Fragile X related protein 1 (FXR1P) regulates proliferation of adult neural stem cells. Hum. Mol. Genet. 26, 1340–1352 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Charman, T. et al. IQ in children with autism spectrum disorders: data from the Special Needs and Autism Project (SNAP). Psychol. Med. 41, 619–627 (2011).

    Article  CAS  PubMed  Google Scholar 

  48. Finucane, B. M. et al. 15q duplication syndrome and related disorders. in GeneReviews (eds Adam, M. P. et al.) (University of Washington, Seattle, 2016).

  49. Ahn, J. & Xiao, X. RASER: reads aligner for SNPs and editing sites of RNA. Bioinformatics 31, 3906–3913 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lee, J. H., Ang, J. K. & Xiao, X. Analysis and design of RNA sequencing experiments for identifying RNA editing and other single-nucleotide variants. RNA 19, 725–732 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Feldmeyer, D. et al. Neurological dysfunctions in mice expressing different levels of the Q/R site-unedited AMPAR subunit GluR-B. Nat. Neurosci. 2, 57–64 (1999).

    Article  CAS  PubMed  Google Scholar 

  52. Wang, K., Li, M. & Hakonarson, H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Gu, Z., Gu, L., Eils, R., Schlesner, M. & Brors, B. circlize Implements and enhances circular visualization in R. Bioinformatics 30, 2811–2812 (2014).

    Article  CAS  PubMed  Google Scholar 

  54. Josse, J. & Husson, F. missMDA: a package for handling missing values in multivariate data analysis. J. Stat. Softw. 70, 31 (2016).

    Article  Google Scholar 

  55. Langfelder, P., Zhang, B. & Horvath, S. Defining clusters from a hierarchical cluster tree: the Dynamic Tree Cut package for R. Bioinformatics 24, 719–720 (2008).

    Article  CAS  PubMed  Google Scholar 

  56. Langfelder, P. & Horvath, S. Eigengene networks for studying the relationships between co-expression modules. BMC Syst. Biol. 1, 54 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Wheeler, E. C., Van Nostrand, E. L. & Yeo, G. W. Advances and challenges in the detection of transcriptome-wide protein-RNA interactions. Wiley Interdiscip. Rev. RNA https://doi.org/10.1002/wrna.1436 (2018).

  58. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Bailey, T. L., Johnson, J., Grant, C. E. & Noble, W. S. The MEME Suite. Nucleic Acids Res. 43(W1), W39–W49 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Aken, B. L. et al. The Ensembl gene annotation system. Database (Oxford) https://doi.org/10.1093/database/baw093 (2016).

  61. Nojima, T., Gomes, T., Carmo-Fonseca, M. & Proudfoot, N. J. Mammalian NET-seq analysis defines nascent RNA profiles and associated RNA processing genome-wide. Nat. Protoc. 11, 413–428 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Sundararaman, B. et al. Resources for the comprehensive discovery of functional RNA elements. Mol. Cell 61, 903–913 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Bahn, J. H. et al. Genomic analysis of ADAR1 binding and its involvement in multiple RNA processing pathways. Nat. Commun. 6, 6355 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Postmortem brain samples used in this study were obtained from the University of Maryland Brain and Tissue Bank, which is a component of the US National Institutes of Health (NIH) NeuroBioBank. We are grateful to the subjects and families who participate in the tissue donation programs. This work was funded by grants from the NIH to X.X. (HG009417 and HG006264), G.W.Y. (HG004659, HG009417, HG007005 and MH107367), S.T. (T32HG002536), E.L.V.N. (HG009530), V.M.C. (MH094681) and R.J.H. (HD 036071). S.T is supported by the UCLA Eureka Scholarship. E.L.V.N. is a Merck Fellow of the Damon Runyon Cancer Research Foundation (DRG-2172-13). G.A.P. is supported by the National Science Foundation Graduate Research Fellowship. G.R. is supported by NIH fellowship 1F32MH114620.

Author information

Authors and Affiliations

Authors

Contributions

S.S.T. carried out data analyses with input from G.R. H.I.J., J.H.B. and A.A. performed molecular biology experiments. E.L.V.N., T.B.N., G.A.P. and G.W.Y carried out eCLIP experiments and data processing. Y.H.E.H. contributed to data visualization. C.L. carried out ASD RNA-seq data generation. V.M.C. and R.J.H. provided postmortem Fragile X and control samples. S.S.T, D.H.G. and X.X. designed the study, interpreted the results and wrote the manuscript.

Corresponding authors

Correspondence to Daniel H. Geschwind or Xinshu Xiao.

Ethics declarations

Competing interests

G.W.Y. is a cofounder of Locana and Eclipse Bioinnovations and member of the scientific advisory boards of Locana, Eclipse Bioinnovations and Aquinnah Pharmaceuticals. E.V.N. is a cofounder and member of the scientific advisory board of Eclipse BioInnovations. The terms of these arrangements have been reviewed and approved by the University of California San Diego in accordance with its conflict of interest policies.

Additional information

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

Supplementary information

Supplementary Figures 1–31

Supplementary Figures 1–31

Reporting Summary

Supplementary Table 1

Meta data of brain samples used in this study.

Supplementary Table 2

Differential RNA editing sites identified from ASD-control samples in three brain regions.

Supplementary Table 3

List of primer sequences used in this study.

Supplementary Table 4

Differential editing sites in frontal cortex that correlate with expression of harboring gene.

Supplementary Table 5

Module memberships of editing sites from WGCNA.

Supplementary Table 6

List of FMPR and FXR1P eCLIP peaks.

Supplementary Table 7

Differential RNA editing sites identified in Fragile X samples.

Supplementary Table 8

Genes with brain-region-specific differential editing.

Supplementary Software

Supplementary Software.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tran, S.S., Jun, HI., Bahn, J.H. et al. Widespread RNA editing dysregulation in brains from autistic individuals. Nat Neurosci 22, 25–36 (2019). https://doi.org/10.1038/s41593-018-0287-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-018-0287-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