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.

Aspm knockout ferret reveals an evolutionary mechanism governing cerebral cortical size

Abstract

The human cerebral cortex is distinguished by its large size and abundant gyrification, or folding. However, the evolutionary mechanisms that drive cortical size and structure are unknown. Although genes that are essential for cortical developmental expansion have been identified from the genetics of human primary microcephaly (a disorder associated with reduced brain size and intellectual disability)1, studies of these genes in mice, which have a smooth cortex that is one thousand times smaller than the cortex of humans, have provided limited insight. Mutations in abnormal spindle-like microcephaly-associated (ASPM), the most common recessive microcephaly gene, reduce cortical volume by at least 50% in humans2,3,4, but have little effect on the brains of mice5,6,7,8,9; this probably reflects evolutionarily divergent functions of ASPM10,11. Here we used genome editing to create a germline knockout of Aspm in the ferret (Mustela putorius furo), a species with a larger, gyrified cortex and greater neural progenitor cell diversity12,13,14 than mice, and closer protein sequence homology to the human ASPM protein. Aspm knockout ferrets exhibit severe microcephaly (25–40% decreases in brain weight), reflecting reduced cortical surface area without significant change in cortical thickness, as has been found in human patients3,4, suggesting that loss of ‘cortical units’ has occurred. The cortex of fetal Aspm knockout ferrets displays a very large premature displacement of ventricular radial glial cells to the outer subventricular zone, where many resemble outer radial glia, a subtype of neural progenitor cells that are essentially absent in mice and have been implicated in cerebral cortical expansion in primates12,13,14,15,16. These data suggest an evolutionary mechanism by which ASPM regulates cortical expansion by controlling the affinity of ventricular radial glial cells for the ventricular surface, thus modulating the ratio of ventricular radial glial cells, the most undifferentiated cell type, to outer radial glia, a more differentiated progenitor.

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: Aspm knockout ferrets robustly model human microcephaly.
Fig. 2: Aspm knockout ferrets show displaced NPCs.
Fig. 3: Loss of Aspm changes cell type proportions but not transcriptional programs.
Fig. 4: ASPM controls localization of apical polarity complex proteins to the centrosome.

References

  1. Faheem, M. et al. Molecular genetics of human primary microcephaly: an overview. BMC Med. Genomics 8, S4 (2015).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  3. Passemard, S. et al. Abnormal spindle-like microcephaly-associated (ASPM) mutations strongly disrupt neocortical structure but spare the hippocampus and long-term memory. Cortex 74, 158–176 (2016).

    Article  Google Scholar 

  4. Desir, J., Cassart, M., David, P., Van Bogaert, P. & Abramowicz, M. Primary microcephaly with ASPM mutation shows simplified cortical gyration with antero-posterior gradient pre- and post-natally. Am. J. Med. Genet. A 146A, 1439–1443 (2008).

    Article  CAS  Google Scholar 

  5. Pulvers, J. N. et al. Mutations in mouse Aspm (abnormal spindle-like microcephaly associated) cause not only microcephaly but also major defects in the germline. Proc. Natl Acad. Sci. USA 107, 16595–16600 (2010).

    Article  ADS  CAS  Google Scholar 

  6. Fujimori, A. et al. Disruption of Aspm causes microcephaly with abnormal neuronal differentiation. Brain Dev. 36, 661–669 (2014).

    Article  Google Scholar 

  7. Capecchi, M. R. & Pozner, A. ASPM regulates symmetric stem cell division by tuning cyclin E ubiquitination. Nat. Commun. 6, 8763 (2015).

    Article  ADS  CAS  Google Scholar 

  8. Williams, S. E. et al. Aspm sustains postnatal cerebellar neurogenesis and medulloblastoma growth in mice. Development 142, 3921–3932 (2015).

    Article  CAS  Google Scholar 

  9. Jayaraman, D. et al. Microcephaly proteins Wdr62 and Aspm define a mother centriole complex regulating centriole biogenesis, apical complex, and cell fate. Neuron 92, 813–828 (2016).

    Article  CAS  Google Scholar 

  10. Montgomery, S. H. & Mundy, N. I. Microcephaly genes evolved adaptively throughout the evolution of eutherian mammals. BMC Evol. Biol. 14, 120 (2014).

    Article  Google Scholar 

  11. Bae, B. I., Jayaraman, D. & Walsh, C. A. Genetic changes shaping the human brain. Dev. Cell 32, 423–434 (2015).

    Article  CAS  Google Scholar 

  12. Fietz, S. A. et al. OSVZ progenitors of human and ferret neocortex are epithelial-like and expand by integrin signaling. Nat. Neurosci. 13, 690–699 (2010).

    Article  CAS  Google Scholar 

  13. Reillo, I., de Juan Romero, C., García-Cabezas, M. A. & Borrell, V. A role for intermediate radial glia in the tangential expansion of the mammalian cerebral cortex. Cereb. Cortex 21, 1674–1694 (2011).

    Article  Google Scholar 

  14. Johnson, M. B. et al. Single-cell analysis reveals transcriptional heterogeneity of neural progenitors in human cortex. Nat. Neurosci. 18, 637–646 (2015).

    Article  CAS  Google Scholar 

  15. Smart, I. H., Dehay, C., Giroud, P., Berland, M. & Kennedy, H. Unique morphological features of the proliferative zones and postmitotic compartments of the neural epithelium giving rise to striate and extrastriate cortex in the monkey. Cereb. Cortex 12, 37–53 (2002).

    Article  Google Scholar 

  16. Hansen, D. V., Lui, J. H., Parker, P. R. & Kriegstein, A. R. Neurogenic radial glia in the outer subventricular zone of human neocortex. Nature 464, 554–561 (2010).

    Article  ADS  CAS  Google Scholar 

  17. Bond, J. et al. Protein-truncating mutations in ASPM cause variable reduction in brain size. Am. J. Hum. Genet. 73, 1170–1177 (2003).

    Article  CAS  Google Scholar 

  18. Hutchinson, E. B. et al. Population based MRI and DTI templates of the adult ferret brain and tools for voxelwise analysis. Neuroimage 152, 575–589 (2017).

    Article  CAS  Google Scholar 

  19. Martínez-Cerdeño, V. et al. Comparative analysis of the subventricular zone in rat, ferret and macaque: evidence for an outer subventricular zone in rodents. PLoS ONE 7, e30178 (2012).

    Article  ADS  Google Scholar 

  20. Martínez-Martínez, M. A. et al. A restricted period for formation of outer subventricular zone defined by Cdh1 and Trnp1 levels. Nat. Commun. 7, 11812 (2016).

    Article  ADS  Google Scholar 

  21. Pollen, A. A. et al. Molecular identity of human outer radial glia during cortical development. Cell 163, 55–67 (2015).

    Article  CAS  Google Scholar 

  22. Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015).

    Article  CAS  Google Scholar 

  23. Kodani, A. et al. Centriolar satellites assemble centrosomal microcephaly proteins to recruit CDK2 and promote centriole duplication. eLife 4, e07519 (2015).

    Article  Google Scholar 

  24. Paridaen, J. T., Wilsch-Bräuninger, M. & Huttner, W. B. Asymmetric inheritance of centrosome-associated primary cilium membrane directs ciliogenesis after cell division. Cell 155, 333–344 (2013).

    Article  CAS  Google Scholar 

  25. Wang, X. et al. Asymmetric centrosome inheritance maintains neural progenitors in the neocortex. Nature 461, 947–955 (2009).

    Article  ADS  CAS  Google Scholar 

  26. Singh, S. & Solecki, D. J. Polarity transitions during neurogenesis and germinal zone exit in the developing central nervous system. Front. Cell. Neurosci. 9, 62 (2015).

    Article  CAS  Google Scholar 

  27. Dauber, A. et al. Novel microcephalic primordial dwarfism disorder associated with variants in the centrosomal protein ninein. J. Clin. Endocrinol. Metab. 97, E2140–E2151 (2012).

    Article  CAS  Google Scholar 

  28. Zhang, X. et al. Cell-type-specific alternative splicing governs cell fate in the developing cerebral cortex. Cell 166, 1147–1162 (2016).

    Article  CAS  Google Scholar 

  29. Lehtinen, M. K. et al. The cerebrospinal fluid provides a proliferative niche for neural progenitor cells. Neuron 69, 893–905 (2011).

    Article  CAS  Google Scholar 

  30. Mashiko, D. et al. Generation of mutant mice by pronuclear injection of circular plasmid expressing Cas9 and single guided RNA. Sci. Rep. 3, 3355 (2013).

    Article  Google Scholar 

  31. Li, Z., Sun, X., Chen, J., Leno, G. H. & Engelhardt, J. F. Factors affecting the efficiency of embryo transfer in the domestic ferret (Mustela putorius furo). Theriogenology 66, 183–190 (2006).

    Article  Google Scholar 

  32. Chahboune, H. et al. Neurodevelopment of C57B/L6 mouse brain assessed by in vivo diffusion tensor imaging. NMR Biomed. 20, 375–382 (2007).

    Article  Google Scholar 

  33. Petersen, K. F. et al. The role of skeletal muscle insulin resistance in the pathogenesis of the metabolic syndrome. Proc. Natl Acad. Sci. USA 104, 12587–12594 (2007).

    Article  ADS  CAS  Google Scholar 

  34. Rueckert, D. et al. Nonrigid registration using free-form deformations: application to breast MR images. IEEE Trans. Med. Imaging 18, 712–721 (1999).

    Article  CAS  Google Scholar 

  35. Papademetris, X., Jackowski, A. P., Schultz, R. T., Staib, L. H. & Duncan, J. S. Integrated intensity and point-feature nonrigid registration. In International Conference on Medical Image Computing and Computer-Assisted Intervention Vol. 3216 (eds Barillot, C., et al.) 763–770 (Springer, Berlin, 2001).

  36. Sawada, K. & Watanabe, M. Development of cerebral sulci and gyri in ferrets (Mustela putorius). Congenit. Anom. (Kyoto) 52, 168–175 (2012).

    Article  Google Scholar 

  37. Joshi, A. et al. Unified framework for development, deployment and robust testing of neuroimaging algorithms. Neuroinformatics 9, 69–84 (2011).

    Article  Google Scholar 

  38. Rahman, A. S., Parvinjah, S., Hanna, M. A., Helguera, P. R. & Busciglio, J. Cryopreservation of cortical tissue blocks for the generation of highly enriched neuronal cultures. J. Vis. Exp 45, e2384 (2010).

    Google Scholar 

  39. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc 7, 562–578 (2012).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  41. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  Google Scholar 

  42. Villani, A. C. et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science 356, eaah4573 (2017).

    Article  Google Scholar 

  43. Higgins, J. et al. Human ASPM participates in spindle organisation, spindle orientation and cytokinesis. BMC Cell Biol 11, 85 (2010).

    Article  Google Scholar 

Download references

Acknowledgements

We thank the late R. W. Guillery, who first introduced ferrets as a model for developmental neuroscience; J. K. Joung for advice on genome editing; J. Bond for the ASPM antibody; L. Vasung, P. Herman, J. Neil and C. D. Kroenke for advice on ferret brain MRI; A. Lee, the G. M. Church laboratory (S. Biwas), the S. McCarroll laboratory (S. Burger), the P. Kharchenko laboratory (J. Fan), and the R. Satija laboratory (A. Butler) for advice on scRNA-seq; S. Wasiuk, E. Feiner, A. S. Kamumbu and M. Lee for technical assistance; Marshall BioResources for animal husbandry; and E. Pollack and the veterinary staff at Boston Children’s Hospital and Yale School of Medicine for surgical support. Animal silhouettes in Fig. 1 were designed by Freepik from https://www.flaticon.com/. This work was supported by P30NS052519 (F.H. and Yale’s QNMR Core Center), 2R01MH067528 (F.H.), 1R24MH114805 (X.P.), R21HD083956 (K.I.), R01EB017337 (P.E.G.), R24HL123482 (J.F.E.), 5R01NS032457 (C.A.W.), 5R21NS091865 (B.-I.B.) and the Allen Discovery Center program through The Paul G. Allen Frontiers Group. C.A.W. is an Investigator of the Howard Hughes Medical Institute.

Reviewer information

Nature thanks S. Juliano, F. Tissir and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

B.-I.B., C.A.W. and J.F.E. conceived the project. B.-I.B. generated genome-editing reagents. X.S., Z.Y. and B.L. injected ferret zygotes under J.F.E.’s supervision. M.B.J., K.M.G., P.P.W. and D.M.G. performed immunohistochemistry experiments. R.S.S., M.C., K.I., J.B.M., P.E.G., D.C., X.P., L.H.S. and F.H. performed MRI analyses. M.B.J., R.B.-M., Y.M.W. and H.K. performed scRNA-seq. M.B.J. and R.B.-M. performed single-molecule fluorescence in situ hybridization. A.K. and S.C.R. characterized molecular defects with K.P. and B.-I.B. With input from all authors, M.B.J., B.-I.B. and C.A.W interpreted the data and wrote the paper.

Corresponding authors

Correspondence to Christopher A. Walsh or Byoung-Il Bae.

Ethics declarations

Competing interests

X.P. is a consultant for Electrical Geodesics Inc. The other 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 Figures and Tables

Extended Data Fig. 1 Cytoarchitecture and neuronal subtype lamination in the cortex of mature Aspm knockout ferrets.

a, Nissl stains of coronal sections from the brains of P41 littermates, as shown in Fig. 1l, with additional Aspm+/− and Aspm−/− littermates shown. b, c, Brain sections of P41 littermates immunostained for cortical layer-specific projection neurons including SATB2 (layer II–IV), CTIP2 (layer V), and FoxP2 (layer VI). The experiments were repeated independently three times with similar results. Scale bars, 2 mm (a, top), 200 μm (a, bottom) and 100 μm (b, c).

Extended Data Fig. 2 SOX2 and Ki-67 immunostaining in additional E35 and P0 littermates dorsal cortex.

Additional results to Fig. 2e, f. Each set of panels is from the brain of a different littermate, showing the high penetrance of the neural progenitor cell basal displacement phenotype. The experiments were repeated independently three times with similar results. Scale bar, 200 μm.

Extended Data Fig. 3 Displaced progenitors in Aspm knockout ferrets have basal fibres.

Additional results for Fig. 2h, i. Immunostaining of SOX2, Ki-67 and VIM shows that displaced neural progenitors have basal radial fibres. The experiments were repeated independently three times with similar results. Scale bars, 100 μm.

Extended Data Fig. 4 Aspm knockout mice do not demonstrate displaced progenitors in the intermediate zone.

a, b, Unlike Aspm−/− ferrets, Aspm−/− mice do not have displaced NPCs in the intermediate zone. However, they show a variable increase in the number of intermediate progenitors (PAX6Ki-67+ cells in a and TBR2+ cells in b), which is enhanced by heterozygous, compound mutation in Wdr62, a microcephaly gene causing more severe microcephaly9. The experiments were repeated independently three times with similar results. Scale bars, 100 μm.

Extended Data Fig. 5 Modest increase in apoptosis throughout the germinal zones of the Aspm knockout telencephalon.

Apoptotic cells (yellow) are indicated by enzymatic fluorescence detection of double-stranded DNA damage with DAPI nuclear counterstaining (blue). The experiments were repeated independently three times with similar results. a, Whole section. b, c, Cortical wall columns. Scale bars, 500 μm (a) and 100 μm (b, c).

Extended Data Fig. 6 Additional immunohistochemical analyses of displaced progenitors in the Aspm knockout cortex.

a, E35 knockout cortex stained for VRG and ORG markers SOX2 and HOPX reveals extensive co-labelling in both the ventricular zone (VZ) and SVZ, including in displaced OSVZ progenitors. b, In the E35 knockout OSVZ, clusters of supernumerary displaced neural progenitor cells include numerous TBR2+ intermediate progenitors and are surrounded by DCX+ newborn neurons, indicating preserved neurogenesis within the precocious OSVZ niche of the Aspm knockout cortex. The experiments were repeated independently three times with similar results. Scale bars, 50 μm.

Extended Data Fig. 7 scRNA-seq batch, sample and cluster analyses.

at-SNE plot from Fig. 3a with cells coloured by biological replicate (that is, animal). Most clusters include cells from all samples, except for a cluster expressing blood genes and a cluster expressing choroid plexus epithelial cells that are mostly from animal WT5E. These two cell clusters were not included in downstream analyses. HET, heterozygote; KO, knockout; WT, wild type. Numbers and letters indicate litter and animal identification number, respectively. b, t-SNE plot from Fig. 3a with cells coloured by the batch they were processed in. Clusters are composed of cells from all batches. c, Per-cell gene count and UMI count per sample. Each violin plot is one biological replicate and each dot is one cell. Sample WT5D was not included in the analysis due to the lower gene and UMI count compared to other samples as well as the inconsistent clustering compared to other wild-type samples (data not shown). d, Per-cell gene count and UMI count for identified clusters. Each violin plot is one cell cluster and each dot is one cell. The three clusters in grey (EN4, BL, CPE) were not included in downstream analyses. See Methods for details. This scRNA-seq experiment was performed once with n = 22,211 cells (8,037 cells from two Aspm+/+ and one Aspm+/− ferrets and 14,174 cells from four Aspm−/− ferrets). RG1, cycling radial glial progenitors; RG2, interphase radial glial progenitors; IP, intermediate progenitors; EN1, upper-layer excitatory neurons; EN2, deep-layer excitatory neurons; EN3, Cajal–Retzius cells; IN1, immature inhibitory neurons; IN2, SST+ inhibitory neurons; IN3, ventral/inhibitory progenitors; ENDO1, endothelial cells 1; ENDO2, endothelial cells 2; OPC, oligodendrocyte precursors; MG, microglia; EN4, mixed excitatory neuron identity; BL, blood cells; CPE, choroid plexus epithelial cells.

Extended Data Fig. 8 Loss of Aspm disrupts centriole duplication in FEFs.

Mitotic Aspm knockout FEFs, identified by staining for pH3 and co-stained for the centriolar marker centrin, display a significant loss of centrioles. The percentage of cells with an abnormal number (less than 4) of centrioles is increased eightfold in Aspm−/− FEFs compared to Aspm+/+ FEFs (n = 100 cells per genotype for three independent experiments; P = 0.003). The experiments were repeated independently three times with similar results. Statistical analysis was performed using a two-tailed t-test; data are mean ± s.e.m.

Extended Data Table 1 Region-specific changes in volume and surface area by loss of Aspm in ferrets
Extended Data Table 2 Cluster identifiers of E35 ferret cerebral cortical cells analysed by scRNA-seq

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Johnson, M.B., Sun, X., Kodani, A. et al. Aspm knockout ferret reveals an evolutionary mechanism governing cerebral cortical size. Nature 556, 370–375 (2018). https://doi.org/10.1038/s41586-018-0035-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-018-0035-0

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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