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:

The exon junction complex component Magoh controls brain size by regulating neural stem cell division

This article has been updated

Abstract

Brain structure and size require precise division of neural stem cells (NSCs), which self-renew and generate intermediate neural progenitors (INPs) and neurons. The factors that regulate NSCs remain poorly understood, and mechanistic explanations of how aberrant NSC division causes the reduced brain size seen in microcephaly are lacking. Here we show that Magoh, a component of the exon junction complex (EJC) that binds RNA, controls mouse cerebral cortical size by regulating NSC division. Magoh haploinsufficiency causes microcephaly because of INP depletion and neuronal apoptosis. Defective mitosis underlies these phenotypes, as depletion of EJC components disrupts mitotic spindle orientation and integrity, chromosome number and genomic stability. In utero rescue experiments showed that a key function of Magoh is to control levels of the microcephaly-associated protein Lis1 during neurogenesis. Our results uncover requirements for the EJC in brain development, NSC maintenance and mitosis, thereby implicating this complex in the pathogenesis of microcephaly.

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

Figure 1: Mutation of Magoh causes microcephaly and reduced body size.
Figure 2: E18.5 MagohMos2/+ brains contain disorganized layers and fewer neurons.
Figure 3: Magoh is required for proper numbers of INPs but not NSCs.
Figure 4: Magoh is required to prevent premature neuronal differentiation and apoptosis.
Figure 5: Magoh and core EJC components regulate the mitotic spindle, ploidy, mitosis and genomic stability.
Figure 6: Magoh acts upstream of the microcephaly-associated protein Lis1 to regulate neurogenesis.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

Change history

  • 19 April 2010

    In the version of this article initially published online, the last sentence of the sixth paragraph in the Discussion section read “Future studies will reveal, for example, whether any of the genes altered in our microarray or proteomics experiments are also essential targets of MAGOH.” MAGOH has been corrected to Magoh, denoting the mouse gene. Also, the second sentence of the last paragraph in the Discussion section initially read “Of note, Magoh is found within a 55-gene deletion on chromosome 1p32.3 that is associated with mental retardation and abnormalities in brain size31.” Magoh has been corrected to MAGOH, denoting the human gene. These errors have been corrected in the print, HTML and PDF versions of the article.

References

  1. Molyneaux, B.J., Arlotta, P., Menezes, J.R. & Macklis, J.D. Neuronal subtype specification in the cerebral cortex. Nat. Rev. Neurosci. 8, 427–437 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Haubensak, W., Attardo, A., Denk, W. & Huttner, W.B. Neurons arise in the basal neuroepithelium of the early mammalian telencephalon: a major site of neurogenesis. Proc. Natl. Acad. Sci. USA 101, 3196–201 (2004).

    Article  CAS  PubMed  Google Scholar 

  3. Pontious, A., Kowalczyk, T., Englund, C. & Hevner, R.F. Role of intermediate progenitor cells in cerebral cortex development. Dev Neurosci 30, 24–32 (2008).

    Article  CAS  Google Scholar 

  4. Chenn, A. & McConnell, S.K. Cleavage orientation and the asymmetric inheritance of Notch1 immunoreactivity in mammalian neurogenesis. Cell 82, 631–641 (1995).

    Article  CAS  PubMed  Google Scholar 

  5. Sanada, K. & Tsai, L.H. G protein βγ subunits and AGS3 control spindle orientation and asymmetric cell fate of cerebral cortical progenitors. Cell 122, 119–131 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Zhong, W., Feder, J.N., Jiang, M.M., Jan, L.Y. & Jan, Y.N. Asymmetric localization of a mammalian numb homolog during mouse cortical neurogenesis. Neuron 17, 43–53 (1996).

    Article  CAS  Google Scholar 

  7. Bond, J. & Woods, C.G. Cytoskeletal genes regulating brain size. Curr. Opin. Cell Biol. 18, 95–101 (2006).

    Article  CAS  Google Scholar 

  8. Bond, J. et al. ASPM is a major determinant of cerebral cortical size. Nat. Genet. 32, 316–320 (2002).

    Article  CAS  Google Scholar 

  9. Bond, J. et al. A centrosomal mechanism involving CDK5RAP2 and CENPJ controls brain size. Nat. Genet. 37, 353–355 (2005).

    Article  CAS  Google Scholar 

  10. Jackson, A.P. et al. Identification of microcephalin, a protein implicated in determining the size of the human brain. Am. J. Hum. Genet. 71, 136–142 (2002).

    Article  CAS  PubMed  Google Scholar 

  11. do Carmo Avides, M. & Glover, D.M. Abnormal spindle protein, Asp, and the integrity of mitotic centrosomal microtubule organizing centers. Science 283, 1733–1735 (1999).

    Article  CAS  Google Scholar 

  12. Bi, W. et al. Increased LIS1 expression affects human and mouse brain development. Nat. Genet. 41, 168–177 (2009).

    Article  CAS  PubMed  Google Scholar 

  13. Faulkner, N.E. et al. A role for the lissencephaly gene LIS1 in mitosis and cytoplasmic dynein function. Nat. Cell Biol. 2, 784–791 (2000).

    Article  CAS  Google Scholar 

  14. Reiner, O. et al. Isolation of a Miller-Dieker lissencephaly gene containing G protein β-subunit-like repeats. Nature 364, 717–721 (1993).

    Article  CAS  Google Scholar 

  15. Griffith, E. et al. Mutations in pericentrin cause Seckel syndrome with defective ATR-dependent DNA damage signaling. Nat. Genet. 40, 232–236 (2008).

    Article  CAS  Google Scholar 

  16. O'Driscoll, M., Ruiz-Perez, V.L., Woods, C.G., Jeggo, P.A. & Goodship, J.A. A splicing mutation affecting expression of ataxia-telangiectasia and Rad3-related protein (ATR) results in Seckel syndrome. Nat. Genet. 33, 497–501 (2003).

    Article  CAS  Google Scholar 

  17. Rauch, A. et al. Mutations in the pericentrin (PCNT) gene cause primordial dwarfism. Science 319, 816–819 (2008).

    Article  CAS  Google Scholar 

  18. Matera, I. et al. A sensitized mutagenesis screen identifies Gli3 as a modifier of Sox10 neurocristopathy. Hum. Mol. Genet. 17, 2118–2131 (2008).

    Article  CAS  PubMed  Google Scholar 

  19. Diem, M.D., Chan, C.C., Younis, I. & Dreyfuss, G. PYM binds the cytoplasmic exon-junction complex and ribosomes to enhance translation of spliced mRNAs. Nat. Struct. Mol. Biol. 14, 1173–1179 (2007).

    Article  CAS  Google Scholar 

  20. Kataoka, N., Diem, M.D., Kim, V.N., Yong, J. & Dreyfuss, G. Magoh, a human homolog of Drosophila mago nashi protein, is a component of the splicing-dependent exon-exon junction complex. EMBO J. 20, 6424–6433 (2001).

    Article  CAS  PubMed  Google Scholar 

  21. Le Hir, H., Gatfield, D., Braun, I.C., Forler, D. & Izaurralde, E. The protein Mago provides a link between splicing and mRNA localization. EMBO Rep. 2, 1119–1124 (2001).

    Article  CAS  PubMed  Google Scholar 

  22. Nott, A., Le Hir, H. & Moore, M.J. Splicing enhances translation in mammalian cells: an additional function of the exon junction complex. Genes Dev. 18, 210–222 (2004).

    Article  CAS  PubMed  Google Scholar 

  23. Palacios, I.M., Gatfield, D., St. Johnston, D. & Izaurralde, E. An eIF4AIII-containing complex required for mRNA localization and nonsense-mediated mRNA decay. Nature 427, 753–757 (2004).

    Article  CAS  Google Scholar 

  24. Pawlisz, A.S. et al. Lis1-Nde1-dependent neuronal fate control determines cerebral cortical size and lamination. Hum. Mol. Genet. 17, 2441–2455 (2008).

    Article  CAS  PubMed  Google Scholar 

  25. Gambello, M.J. et al. Multiple dose-dependent effects of Lis1 on cerebral cortical development. J. Neurosci. 23, 1719–1729 (2003).

    Article  CAS  Google Scholar 

  26. Yingling, J. et al. Neuroepithelial stem cell proliferation requires Lis1 for precise spindle orientation and symmetric division. Cell 132, 474–486 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Micklem, D.R. et al. The mago nashi gene is required for the polarisation of the oocyte and the formation of perpendicular axes in Drosophila. Curr. Biol. 7, 468–478 (1997).

    Article  CAS  Google Scholar 

  28. Azzalin, C.M. & Lingner, J. The human RNA surveillance factor UPF1 is required for S phase progression and genome stability. Curr. Biol. 16, 433–439 (2006).

    Article  CAS  Google Scholar 

  29. van der Weele, C.M., Tsai, C.W. & Wolniak, S.M. Mago nashi is essential for spermatogenesis in Marsilea. Mol. Biol. Cell 18, 3711–3722 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Yamada, M. et al. Inhibition of calpain increases LIS1 expression and partially rescues in vivo phenotypes in a mouse model of lissencephaly. Nat. Med. 15, 1202–1207 (2009).

    Article  CAS  PubMed  Google Scholar 

  31. Mulatinho, M., Llerena, J., Leren, T.P., Rao, P.N. & Quintero-Rivera, F. Deletion (1)(p32.2-p32.3) detected by array-CGH in a patient with developmental delay/mental retardation, dysmorphic features and low cholesterol: A new microdeletion syndrome? Am. J. Med. Genet. A. 146A, 2284–2290 (2008).

    Article  CAS  Google Scholar 

  32. Brunetti-Pierri, N. et al. Recurrent reciprocal 1q21.1 deletions and duplications associated with microcephaly or macrocephaly and developmental and behavioral abnormalities. Nat. Genet. 40, 1466–1471 (2008).

    Article  CAS  PubMed  Google Scholar 

  33. Mefford, H.C. et al. Recurrent rearrangements of chromosome 1q21.1 and variable pediatric phenotypes. N. Engl. J. Med. 359, 1685–1699 (2008).

    Article  CAS  PubMed  Google Scholar 

  34. Lundsteen, C. & Lind, A.M. A test of a climate room for preparation of chromosome slides. Clin. Genet. 28, 260–262 (1985).

    Article  CAS  Google Scholar 

  35. Lichter, P., Cremer, T., Borden, J., Manuelidis, L. & Ward, D.C. Delineation of individual human chromosomes in metaphase and interphase cells by in situ suppression hybridization using recombinant DNA libraries. Hum. Genet. 80, 224–234 (1988).

    Article  CAS  Google Scholar 

  36. Pinkel, D., Straume, T. & Gray, J.W. Cytogenetic analysis using quantitative, high-sensitivity, fluorescence hybridization. Proc. Natl. Acad. Sci. USA 83, 2934–2938 (1986).

    Article  CAS  Google Scholar 

  37. Liyanage, M. et al. Multicolour spectral karyotyping of mouse chromosomes. Nat. Genet. 14, 312–315 (1996).

    Article  CAS  Google Scholar 

  38. Schrock, E. et al. Multicolor spectral karyotyping of human chromosomes. Science 273, 494–497 (1996).

    Article  CAS  Google Scholar 

  39. Gaiano, N., Nye, J.S. & Fishell, G. Radial glial identity is promoted by Notch1 signaling in the murine forebrain. Neuron 26, 395–404 (2000).

    Article  CAS  PubMed  Google Scholar 

  40. Kittler, R. et al. An endoribonuclease-prepared siRNA screen in human cells identifies genes essential for cell division. Nature 432, 1036–1040 (2004).

    Article  CAS  Google Scholar 

  41. Visel, A., Thaller, C. & Eichele, G. GenePaint.org: an atlas of gene expression patterns in the mouse embryo. Nucleic Acids Res. 32, D552–D556 (2004).

    Article  CAS  PubMed  Google Scholar 

  42. Saito, T. In vivo electroporation in the embryonic mouse central nervous system. Nat. Protocols 1, 1552–1558 (2006).

    Article  CAS  Google Scholar 

  43. Buac, K. et al. NRG1 / ERBB3 signaling in melanocyte development and melanoma: inhibition of differentiation and promotion of proliferation. Pigm. Cell Melanoma Res. 22, 773–784 (2009).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

For advice, we thank Pavan laboratory members, including L. Baxter for reading the manuscript. For technical assistance, we thank A. Incao (mouse husbandry); G. Elliot and A. Chen (mouse transgenics); A. Dutra, E. Pak and S. Witchovitch (metaphase, SKY, microscopy assistance); S. Anderson and M. Kirby (FACS analysis); B. Bhorate (microarray statistics); M. Bryant (pathology analysis); Harvard Partners Center for Genetics and Genomics (candidate gene sequencing); and J. Fekecs and D. Leja (assistance with figures). This research was funded in part by an National Institute of General Medical Sciences PRAT fellowship and K99/R00 Pathway to Independence Award (to D.L.S.), by the Intramural Research program of NIH/NHGRI (to W.J.P., K.M.) and by the NIH/NHGRI (to N.G.). C.A.W. is an Investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

D.L.S. designed the study, performed all experiments except where noted otherwise and wrote the paper. D.E.W.-C. performed all mRNA analyses and assisted with quantitative analyses. K.C.S. and T.J.P. performed all in utero electroporations and dissections of electroporated brains. D.M.L. performed staining of E18.5 markers. A.J.B. assisted with HeLa cell analyses and quantification of NSCs and INPs. H.L. performed RPE cell analyses. D.L.S., W.J.P., C.A.W., N.G. and K.M. were involved in research design, and all authors were involved in data analysis. D.L.S. and W.J.P. prepared the manuscript. All authors have agreed to the content in the manuscript, including the data as presented.

Corresponding author

Correspondence to William J Pavan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9 and Supplementary Tables 1–3 (PDF 12447 kb)

Supplementary Data

Serum chemistries, hematology, and organ weights of control and mutant animals (XLS 83 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Silver, D., Watkins-Chow, D., Schreck, K. et al. The exon junction complex component Magoh controls brain size by regulating neural stem cell division. Nat Neurosci 13, 551–558 (2010). https://doi.org/10.1038/nn.2527

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.2527

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