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:

AUTS2 isoforms control neuronal differentiation

Abstract

Mutations in AUTS2 are associated with autism, intellectual disability, and microcephaly. AUTS2 is expressed in the brain and interacts with polycomb proteins, yet it is still unclear how mutations in AUTS2 lead to neurodevelopmental phenotypes. Here we report that when neuronal differentiation is initiated, there is a shift in expression from a long isoform to a short AUTS2 isoform. Yeast two-hybrid screen identified the splicing factor SF3B1 as an interactor of both isoforms, whereas the polycomb group proteins, PCGF3 and PCGF5, were found to interact exclusively with the long AUTS2 isoform. Reporter assays showed that the first exons of the long AUTS2 isoform function as a transcription repressor, but the part that consist of the short isoform acts as a transcriptional activator, both influenced by the cellular context. The expression levels of PCGF3 influenced the ability of the long AUTS2 isoform to activate or repress transcription. Mouse embryonic stem cells (mESCs) with heterozygote mutations in Auts2 had an increase in cell death during in vitro corticogenesis, which was significantly rescued by overexpressing the human AUTS2 transcripts. mESCs with a truncated AUTS2 protein (missing exons 12–20) showed premature neuronal differentiation, whereas cells overexpressing AUTS2, especially the long transcript, showed increase in expression of pluripotency markers and delayed differentiation. Taken together, our data suggest that the precise expression of AUTS2 isoforms is essential for regulating transcription and the timing of neuronal differentiation.

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
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Sanders SJ, Murtha MT, Gupta AR, Murdoch JD, Raubeson MJ, Willsey AJ, et al. De novo mutations revealed by whole-exome sequencing are strongly associated with autism. Nature. 2012;485:237–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Hamdan FF, Srour M, Capo-Chichi J-M, Daoud H, Nassif C, Patry L, et al. De novo mutations in moderate or severe intellectual disability. PLoS Genet. 2014;10:e1004772.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Ben-David E, Shifman S. Combined analysis of exome sequencing points toward a major role for transcription regulation during brain development in autism. Mol Psychiatry. 2013;18:1054–6.

    Article  CAS  PubMed  Google Scholar 

  4. Pinto D, Delaby E, Merico D, Barbosa M, Merikangas A, Klei L, et al. Convergence of genes and cellular pathways dysregulated in autism spectrum disorders. Am J Hum Genet. 2014;94:677–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ronan JL, Wu W, Crabtree GR. From neural development to cognition: unexpected roles for chromatin. Nat Rev Genet. 2013;14:347–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Suliman R, Ben-David E, Shifman S. Chromatin regulators, phenotypic robustness, and autism risk. Front Genet. 2014;5:81.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. De Rubeis S, He X, Goldberg AP, Poultney CS, Samocha K, Ercument Cicek A, et al. Synaptic, transcriptional and chromatin genes disrupted in autism. Nature. 2014;515:209–15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Ernst C. Proliferation and differentiation deficits are a major convergence point for neurodevelopmental disorders. Trends Neurosci. 2016;39:290–9.

    Article  CAS  PubMed  Google Scholar 

  9. Casanova EL, Casanova MF. Genetics studies indicate that neural induction and early neuronal maturation are disturbed in autism. Front Cell Neurosci. 2014;8:397.

    PubMed  PubMed Central  Google Scholar 

  10. Kaushik G, Zarbalis KS. Prenatal neurogenesis in autism spectrum disorders. Front Chem. 2016;4:12. https://doi.org/10.3389/fchem.2016.00012.

  11. Packer A. Neocortical neurogenesis and the etiology of autism spectrum disorder. Neurosci Biobehav Rev. 2016;64:185–95.

    Article  PubMed  Google Scholar 

  12. Fromer M, Pocklington AJ, Kavanagh DH, Williams HJ, Dwyer S, Gormley P, et al. De novo mutations in schizophrenia implicate synaptic networks. Nature. 2014;506:179–84.

    Article  CAS  PubMed  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. 2017;18:2217–27.

    Article  CAS  PubMed  Google Scholar 

  14. Zhu X, Need AC, Petrovski S, Goldstein DB. One gene, many neuropsychiatric disorders: lessons from Mendelian diseases. Nat Neurosci. 2014;17:773–81.

    Article  CAS  PubMed  Google Scholar 

  15. Sultana R, Yu C-E, Yu J, Munson J, Chen D, Hua W, et al. Identification of a novel gene on chromosome 7q11. 2 interrupted by a translocation breakpoint in a pair of autistic twins. Genomics. 2002;80:129–34.

    Article  CAS  PubMed  Google Scholar 

  16. Kalscheuer VM, FitzPatrick D, Tommerup N, Bugge M, Niebuhr E, Neumann LM, et al. Mutations in autism susceptibility candidate 2 (AUTS2) in patients with mental retardation. Hum Genet. 2007;121:501–9.

    Article  PubMed  Google Scholar 

  17. Huang X-L, Zou YS, Maher TA, Newton S, Milunsky JM. A de novo balanced translocation breakpoint truncating the autism susceptibility candidate 2 (AUTS2) gene in a patient with autism. Am J Med Genet Part A. 2010;152:2112–4.

    Article  CAS  Google Scholar 

  18. Ben-David E, Granot-Hershkovitz E, Monderer-Rothkoff G, Lerer E, Levi S, Yaari M, et al. Identification of a functional rare variant in autism using genome-wide screen for monoallelic expression. Hum Mol Genet. 2011;20:3632–41.

    Article  CAS  PubMed  Google Scholar 

  19. Elia J, Gai X, Xie HM, Perin JC, Geiger E, Glessner JT, et al. Rare structural variants found in attention-deficit hyperactivity disorder are preferentially associated with neurodevelopmental genes. Mol Psychiatry. 2009;15:637–46.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Mefford HC, Muhle H, Ostertag P, von Spiczak S, Buysse K, Baker C, et al. Genome-wide copy number variation in epilepsy: novel susceptibility loci in idiopathic generalized and focal epilepsies. PLoS Genet. 2010;6:e1000962.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Amarillo IE, Li WL, Li X, Vilain E, Kantarci S. De novo single exon deletion of AUTS2 in a patient with speech and language disorder: a review of disrupted AUTS2 and further evidence for its role in neurodevelopmental disorders. Am J Med Genet A. 2014;164A:958–65.

    Article  PubMed  CAS  Google Scholar 

  22. Beunders G, Voorhoeve E, Golzio C, Pardo LM, Rosenfeld Ja, Talkowski ME, et al. Exonic deletions in AUTS2 cause a syndromic form of intellectual disability and suggest a critical role for the C terminus. Am J Hum Genet. 2013;92:210–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Bedogni F, Hodge RD, Nelson BR, Frederick EA, Shiba N, Daza RA, et al. Autism susceptibility candidate 2(Auts2) encodes a nuclear protein expressed in developing brain regions implicated in autism neuropathology. Gene Expr Patterns. 2010;10:9–15.

    Article  CAS  PubMed  Google Scholar 

  24. Oksenberg N, Stevison L, Wall JD, Ahituv N. Function and regulation of AUTS2, a gene implicated in autism and human evolution. PLoS Genet. 2013;9:e1003221.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hori K, Nagai T, Shan W, Sakamoto A, Taya S, Hashimoto R, et al. Cytoskeletal regulation by AUTS2 in neuronal migration and neuritogenesis. Cell Rep. 2014;9:2166–79.

    Article  CAS  PubMed  Google Scholar 

  26. Gao Z, Lee P, Stafford JM, von Schimmelmann M, Schaefer A, Reinberg D. An AUTS2–Polycomb complex activates gene expression in the CNS. Nature. 2014;516:349–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gao Z, Zhang J, Bonasio R, Strino F, Sawai A, Parisi F, et al. PCGF homologs, CBX proteins, and RYBP define functionally distinct PRC1 family complexes. Mol Cell. 2012;45:344–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Oksenberg N, Haliburton GDE, Eckalbar WL, Oren I, Nishizaki S, Murphy K, et al. Genome-wide distribution of Auts2 binding localizes with active neurodevelopmental genes. Transl Psychiatry. 2014;4:e431.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Gaspard N, Bouschet T, Herpoel A, Naeije G, van den Ameele J, Vanderhaeghen P. Generation of cortical neurons from mouse embryonic stem cells. Nat Protoc. 2009;4:1454–63.

    Article  CAS  PubMed  Google Scholar 

  30. Gaspard N, Bouschet T, Hourez R, Dimidschstein J, Naeije G, van den Ameele J, et al. An intrinsic mechanism of corticogenesis from embryonic stem cells. Nature. 2008;455:351–7.

    Article  CAS  PubMed  Google Scholar 

  31. Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F. Genome engineering using the CRISPR-Cas9 system. Nat Protoc. 2013;8:2281–308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ernst J, Kellis M. ChromHMM: automating chromatin-state discovery and characterization. Nat Methods. 2012;9:215–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ge H, Liu Z, Church GM, Vidal M. Correlation between transcriptome and interactome mapping data from Saccharomyces cerevisiae. Nat Genet. 2001;29:482–6.

    Article  CAS  PubMed  Google Scholar 

  34. Jansen R, Greenbaum D, Gerstein M. Relating whole-genome expression data with protein-protein interactions. Genome Res. 2002;12:37–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Deane CM, Salwiński Ł, Xenarios I, Eisenberg D. Protein interactions: two methods for assessment of the reliability of high throughput observations. Mol Cell Proteomics. 2002;1:349–56.

    Article  CAS  PubMed  Google Scholar 

  36. Jansen R, Yu H, Greenbaum D, Kluger Y, Krogan NJ, Chung S, et al. A Bayesian networks approach for predicting protein-protein interactions from genomic data. Science. 2003;302:449–53.

    Article  CAS  PubMed  Google Scholar 

  37. Chen J, Xu H, Aronow BJ, Jegga AG. Improved human disease candidate gene prioritization using mouse phenotype. BMC Bioinformatics. 2007;8:392.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Isono K, Mizutani-Koseki Y, Komori T, Schmidt-Zachmann MS, Koseki H. Mammalian polycomb-mediated repression of Hox genes requires the essential spliceosomal protein Sf3b1. Genes Dev. 2005;19:536–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kfir N, Lev-Maor G, Glaich O, Alajem A, Datta A, Sze SK, et al. SF3B1 association with chromatin determines splicing outcomes. Cell Rep. 2015;11:618–29.

    Article  CAS  PubMed  Google Scholar 

  40. Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell. 2006;125:315–26.

    Article  CAS  PubMed  Google Scholar 

  41. Harikumar A, Meshorer E Chromatin remodeling and bivalent histone modifications in embryonic stem cells. EMBO Rep. 2015;16:1609–19.

  42. Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, Giannoukos G, et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature. 2007;448:553–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Endoh M, Endo TA, Endoh T, Isono K, Sharif J, Ohara O, et al. Histone H2A mono-ubiquitination is a crucial step to mediate PRC1-dependent repression of developmental genes to maintain ES cell identity. PLoS Genet. 2012;8:e1002774.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ku M, Koche RP, Rheinbay E, Mendenhall EM, Endoh M, Mikkelsen TS, et al. Genomewide analysis of PRC1 and PRC2 occupancy identifies two classes of bivalent domains. PLoS Genet. 2008;4:e1000242.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. McEvilly RJ, de Diaz MO, Schonemann MD, Hooshmand F, Rosenfeld MG. Transcriptional regulation of cortical neuron migration by POU domain factors. Science. 2002;295:1528–32.

    Article  CAS  PubMed  Google Scholar 

  46. Brennaman LH, Zhang X, Guan H, Triplett JW, Brown A, Demyanenko GP, et al. Polysialylated NCAM and ephrinA/EphA regulate synaptic development of GABAergic interneurons in prefrontal cortex. Cereb Cortex. 2013;23:162–77.

    Article  PubMed  Google Scholar 

  47. Nishikimi M, Oishi K, Tabata H, Torii K, Nakajima K. Segregation and pathfinding of callosal axons through EphA3 signaling. J Neurosci. 2011;31:16251–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Sado T, Nakajima N, Tada M, Takagi N. A novel mesoderm-specific cDNA isolated from a mouse embryonal carcinoma cell line (embryonal carcinoma cell/cDNA/in situ hybridization/mesoderm/mouse embryo). Dev Growth Differ. 1993;35:551–60.

    Article  CAS  Google Scholar 

  49. Martin BL, Kimelman D. Brachyury establishes the embryonic mesodermal progenitor niche. Genes Dev. 2010;24:2778–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Bedogni F, Hodge RD, Elsen GE, Nelson BR, Daza RAM, Beyer RP, et al. Tbr1 regulates regional and laminar identity of postmitotic neurons in developing neocortex. Proc Natl Acad Sci USA. 2010;107:13129–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Edupuganti RR, Harikumar A, Aaronson Y, Biran A, Sailaja BS, Nissim-Rafinia M, et al. Alternative SET/TAFI promoters regulate embryonic stem cell differentiation. Stem Cell Reports. 2017;9:1291–303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Oksenberg N, Ahituv N. The role of AUTS2 in neurodevelopment and human evolution. Trends Genet. 2013;29:600–8.

    Article  CAS  PubMed  Google Scholar 

  53. Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, Südhof TC, Wernig M. Direct conversion of fibroblasts to functional neurons by defined factors. Nature. 2010;463:1035–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This research was supported by German-Israeli Foundation for Scientific Research and Development (GIF) (grant number 2224/2009 to SS), by the Israel Science Foundation (grant numbers 688/12 to SS, 575/17 to SS and 1140/17 to EM), and supported in part by ‘Investissements dʼAvenir’ program (Labex Biopsy, ANR-11-IDEX-0004-02 to MG) and Fondation Lejeune (to MG).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sagiv Shifman.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Monderer-Rothkoff, G., Tal, N., Risman, M. et al. AUTS2 isoforms control neuronal differentiation. Mol Psychiatry 26, 666–681 (2021). https://doi.org/10.1038/s41380-019-0409-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-019-0409-1

This article is cited by

Search

Quick links