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:

Gene co-regulation by Fezf2 selects neurotransmitter identity and connectivity of corticospinal neurons

Abstract

The neocortex contains an unparalleled diversity of neuronal subtypes, each defined by distinct traits that are developmentally acquired under the control of subtype-specific and pan-neuronal genes. The regulatory logic that orchestrates the expression of these unique combinations of genes is unknown for any class of cortical neuron. Here, we report that Fezf2 is a selector gene able to regulate the expression of gene sets that collectively define mouse corticospinal motor neurons (CSMN). We find that Fezf2 directly induces the glutamatergic identity of CSMN via activation of Vglut1 (Slc17a7) and inhibits a GABAergic fate by repressing transcription of Gad1. In addition, we identify the axon guidance receptor EphB1 as a target of Fezf2 necessary to execute the ipsilateral extension of the corticospinal tract. Our data indicate that co-regulated expression of neuron subtype–specific and pan-neuronal gene batteries by a single transcription factor is one component of the regulatory logic responsible for the establishment of CSMN identity.

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: Fezf2 overexpression in cortical progenitors induces genes that label corticospinal motor neurons.
Figure 2: Fezf2-induced genes identify native CSMN and label subsets of the broad CTIP2-positive population in layer V.
Figure 3: Genome-wide binding analysis for Fezf2 shows preferential association with proximal promoter regions of CSMN genes.
Figure 4: Fezf2 promotes glutamatergic and inhibits GABAergic neurotransmitter pathways.
Figure 5: Fezf2 controls Ephb1 selective expression in CSMN by direct association with the Ephb1 promoter.
Figure 6: Cortical neurons aberrantly project through the anterior commissure in the absence of EphB1.
Figure 7: Ephb1−/− mice recapitulate the axon guidance phenotype observed in Fezf2 null mutants.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Gene Expression Omnibus

NCBI Reference Sequence

References

  1. Finlay, B.L. & Darlington, R.B. Linked regularities in the development and evolution of mammalian brains. Science 268, 1578–1584 (1995).

    CAS  PubMed  Google Scholar 

  2. Migliore, M. & Shepherd, G.M. Opinion: an integrated approach to classifying neuronal phenotypes. Nat. Rev. Neurosci. 6, 810–818 (2005).

    CAS  PubMed  Google Scholar 

  3. Arlotta, P. et al. Neuronal subtype-specific genes that control corticospinal motor neuron development in vivo. Neuron 45, 207–221 (2005).

    CAS  PubMed  Google Scholar 

  4. 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).

    CAS  PubMed  Google Scholar 

  5. Molyneaux, B.J., Arlotta, P., Hirata, T., Hibi, M. & Macklis, J.D. Fezl is required for the birth and specification of corticospinal motor neurons. Neuron 47, 817–831 (2005).

    CAS  PubMed  Google Scholar 

  6. Chen, J.G., Rasin, M.R., Kwan, K.Y. & Sestan, N. Zfp312 is required for subcortical axonal projections and dendritic morphology of deep-layer pyramidal neurons of the cerebral cortex. Proc. Natl. Acad. Sci. USA 102, 17792–17797 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Chen, B., Schaevitz, L.R. & McConnell, S.K. Fezl regulates the differentiation and axon targeting of layer 5 subcortical projection neurons in cerebral cortex. Proc. Natl. Acad. Sci. USA 102, 17184–17189 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Rouaux, C. & Arlotta, P. Fezf2 directs the differentiation of corticofugal neurons from striatal progenitors in vivo. Nat. Neurosci. 13, 1345–1347 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Rouaux, C. & Arlotta, P. Direct lineage reprogramming of post-mitotic callosal neurons into corticofugal neurons in vivo. Nat. Cell Biol. 15, 214–221 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. De la Rossa, A. et al. In vivo reprogramming of circuit connectivity in postmitotic neocortical neurons. Nat. Neurosci. 16, 193–200 (2013).

    CAS  PubMed  Google Scholar 

  11. Lodato, S. et al. Excitatory projection neuron subtypes control the distribution of local inhibitory interneurons in the cerebral cortex. Neuron 69, 763–779 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Ayoub, A.E. et al. Transcriptional programs in transient embryonic zones of the cerebral cortex defined by high-resolution mRNA sequencing. Proc. Natl. Acad. Sci. USA 108, 14950–14955 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Kowalczyk, T. et al. Intermediate neuronal progenitors (basal progenitors) produce pyramidal-projection neurons for all layers of cerebral cortex. Cereb. Cortex 19, 2439–2450 (2009).

    PubMed  PubMed Central  Google Scholar 

  14. Sessa, A., Mao, C.-A., Hadjantonakis, A.-K., Klein, W.H. & Broccoli, V. Tbr2 directs conversion of radial glia into basal precursors and guides neuronal amplification by indirect neurogenesis in the developing neocortex. Neuron 60, 56–69 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Franco, S.J. et al. Fate-restricted neural progenitors in the mammalian cerebral cortex. Science 337, 746–749 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Dominguez, M.H., Ayoub, A.E. & Rakic, P. POU-III transcription factors (Brn1, Brn2, and Oct6) influence neurogenesis, molecular identity, and migratory destination of upper-layer cells of the cerebral cortex. Cereb. Cortex 23, 2632–2643 (2013).

    PubMed  Google Scholar 

  17. Molyneaux, B.J. et al. Novel subtype-specific genes identify distinct subpopulations of callosal projection neurons. J. Neurosci. 29, 12343–12354 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Park, P.J. ChIP-seq: advantages and challenges of a maturing technology. Nat. Rev. Genet. 10, 669–680 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Guo, Y., Mahony, S. & Gifford, D.K. High resolution genome wide binding event finding and motif discovery reveals transcription factor spatial binding constraints. PLoS Comput. Biol. 8, e1002638 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Chen, L., Zheng, J., Yang, N., Li, H. & Guo, S. Genomic selection identifies vertebrate transcription factor Fezf2 binding sites and target genes. J. Biol. Chem. 286, 18641–18649 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Xie, Z., Hu, S., Blackshaw, S., Zhu, H. & Qian, J. hPDI: a database of experimental human protein-DNA interactions. Bioinformatics 26, 287–289 (2010).

    CAS  PubMed  Google Scholar 

  22. Fremeau, R.T. et al. The expression of vesicular glutamate transporters defines two classes of excitatory synapse. Neuron 31, 247–260 (2001).

    CAS  PubMed  Google Scholar 

  23. Eiraku, M. et al. Self-organized formation of polarized cortical tissues from ESCs and its active manipulation by extrinsic signals. Cell Stem Cell 3, 519–532 (2008).

    CAS  PubMed  Google Scholar 

  24. Canty, A.J. & Murphy, M. Molecular mechanisms of axon guidance in the developing corticospinal tract. Prog. Neurobiol. 85, 214–235 (2008).

    CAS  PubMed  Google Scholar 

  25. Luria, V., Krawchuk, D., Jessell, T.M., Laufer, E. & Kania, A. Specification of motor axon trajectory by ephrin-B:EphB signaling: symmetrical control of axonal patterning in the developing limb. Neuron 60, 1039–1053 (2008).

    CAS  PubMed  Google Scholar 

  26. Lee, R., Petros, T.J. & Mason, C.A. Zic2 regulates retinal ganglion cell axon avoidance of ephrinB2 through inducing expression of the guidance receptor EphB1. J. Neurosci. 28, 5910–5919 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Petros, T.J., Shrestha, B.R. & Mason, C. Specificity and sufficiency of EphB1 in driving the ipsilateral retinal projection. J. Neurosci. 29, 3463–3474 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Englund, C. et al. Pax6, Tbr2, and Tbr1 are expressed sequentially by radial glia, intermediate progenitor cells, and postmitotic neurons in developing neocortex. J. Neurosci. 25, 247–251 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Cahoy, J.D. et al. A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J. Neurosci. 28, 264–278 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Alcamo, E.A. et al. Satb2 regulates callosal projection neuron identity in the developing cerebral cortex. Neuron 57, 364–377 (2008).

    CAS  PubMed  Google Scholar 

  31. Leone, D.P., Srinivasan, K., Chen, B., Alcamo, E. & McConnell, S.K. The determination of projection neuron identity in the developing cerebral cortex. Curr. Opin. Neurobiol. 18, 28–35 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Jouandet, M.L.M., Lachat, J.J.J. & Garey, L.J.L. Distribution of the neurons of origin of the great cerebral commissures in the cat. Anat. Embryol. (Berl.) 171, 105–120 (1985).

    CAS  Google Scholar 

  33. Tuch, D.S., Reese, T.G., Wiegell, M.R. & Wedeen, V.J. Diffusion MRI of complex neural architecture. Neuron 40, 885–895 (2003).

    CAS  PubMed  Google Scholar 

  34. Takahashi, E. et al. Developing neocortex organization and connectivity in cats revealed by direct correlation of diffusion tractography and histology. Cereb. Cortex 21, 200–211 (2011).

    PubMed  Google Scholar 

  35. Kwan, K.Y. et al. SOX5 postmitotically regulates migration, postmigratory differentiation, and projections of subplate and deep-layer neocortical neurons. Proc. Natl. Acad. Sci. USA 105, 16021–16026 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Lai, T. et al. SOX5 controls the sequential generation of distinct corticofugal neuron subtypes. Neuron 57, 232–247 (2008).

    CAS  PubMed  Google Scholar 

  37. Han, W. et al. TBR1 directly represses Fezf2 to control the laminar origin and development of the corticospinal tract. Proc. Natl. Acad. Sci. USA 108, 3041–3046 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. McKenna, W.L. et al. Tbr1 and Fezf2 regulate alternate corticofugal neuronal identities during neocortical development. J. Neurosci. 31, 549–564 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Chen, B. et al. The Fezf2-Ctip2 genetic pathway regulates the fate choice of subcortical projection neurons in the developing cerebral cortex. Proc. Natl. Acad. Sci. USA 105, 11382–11387 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Srinivasan, K. et al. A network of genetic repression and derepression specifies projection fates in the developing neocortex. Proc. Natl. Acad. Sci. USA 109, 19071–19078 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Hirata, T. et al. Zinc finger gene fez-like functions in the formation of subplate neurons and thalamocortical axons. Dev. Dyn. 230, 546–556 (2004).

    CAS  PubMed  Google Scholar 

  42. Telese, F., Gamliel, A., Skowronska-Krawczyk, D., Garcia-Bassets, I. & Rosenfeld, M.G. 'Seq-ing' insights into the epigenetics of neuronal gene regulation. Neuron 77, 606–623 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Mann, R.S. & Carroll, S.B. Molecular mechanisms of selector gene function and evolution. Curr. Opin. Genet. Dev. 12, 592–600 (2002).

    CAS  PubMed  Google Scholar 

  44. Cochella, L. & Hobert, O. Diverse functions of microRNAs in nervous system development. Curr. Top. Dev. Biol. 99, 115–143 (2012).

    CAS  PubMed  Google Scholar 

  45. Braun, T. & Gautel, M. Transcriptional mechanisms regulating skeletal muscle differentiation, growth and homeostasis. Nat. Rev. Mol. Cell Biol. 12, 349–361 (2011).

    CAS  PubMed  Google Scholar 

  46. García-Bellido, A.A. Genetic control of wing disc development in Drosophila. Ciba Found. Symp. 0, 161–182 (1975).

    PubMed  Google Scholar 

  47. Williams, S.E. et al. Ephrin-B2 and EphB1 mediate retinal axon divergence at the optic chiasm. Neuron 39, 919–935 (2003).

    CAS  PubMed  Google Scholar 

  48. García-Frigola, C. & Herrera, E. Zic2 regulates the expression of Sert to modulate eye-specific refinement at the visual targets. EMBO J. 29, 3170–3183 (2010).

    PubMed  PubMed Central  Google Scholar 

  49. Robichaux, M.A. et al. EphB receptor forward signaling regulates area-specific reciprocal thalamic and cortical axon pathfinding. Proc. Natl. Acad. Sci. USA. 111, 2188–2193 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Flames, N. & Hobert, O. Gene regulatory logic of dopamine neuron differentiation. Nature 458, 885–889 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Saito, T. & Nakatsuji, N. Efficient gene transfer into the embryonic mouse brain using in vivo electroporation. Dev. Biol. 240, 237–246 (2001).

    CAS  PubMed  Google Scholar 

  52. Catapano, L.A., Arnold, M.W., Perez, F.A. & Macklis, J.D. Specific neurotrophic factors support the survival of cortical projection neurons at distinct stages of development. J. Neurosci. 21, 8863–8872 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Barrett, M.T. et al. High-quality RNA and DNA from flow cytometrically sorted human epithelial cells and tissues. Biotechniques 32, 888–896 (2002).

    CAS  PubMed  Google Scholar 

  54. Arlotta, P. et al. Neuronal subtype-specific genes that control corticospinal motor neuron development in vivo. Neuron 45, 207–221 (2005).

    CAS  PubMed  Google Scholar 

  55. Eberwine, J. et al. Analysis of gene expression in single live neurons. Proc. Natl. Acad. Sci. USA 89, 3010–3014 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Weng, L. et al. Rosetta error model for gene expression analysis. Bioinformatics 22, 1111–1121 (2006).

    CAS  PubMed  Google Scholar 

  57. Wu, F.X., Zhang, W.J. & Kusalik, A.J. Modeling gene expression from microarray expression data with state-space equations. Pac. Symp. Biocomput. 2004, 581–592 (2004).

    Google Scholar 

  58. Tusher, V.G., Tibshirani, R. & Chu, G. Significance analysis of microarrays applied to the ionizing radiation response. Proc. Natl. Acad. Sci. USA 98, 5116–5121 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Arlotta, P., Molyneaux, B.J., Jabaudon, D., Yoshida, Y. & Macklis, J.D. Ctip2 controls the differentiation of medium spiny neurons and the establishment of the cellular architecture of the striatum. J. Neurosci. 28, 622–632 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Rosen, G.D. et al. Bilateral subcortical heterotopia with partial callosal agenesis in a mouse mutant. Cereb. Cortex 23, 859–872 (2013).

    CAS  PubMed  Google Scholar 

  62. Takahashi, E. et al. Development of cerebral fiber pathways in cats revealed by diffusion spectrum imaging. Neuroimage 49, 1231–1240 (2010).

    PubMed  Google Scholar 

  63. Lodato, S. et al. Excitatory projection neuron subtypes control the distribution of local inhibitory interneurons in the cerebral cortex. Neuron 69, 763–779 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Tiveron, M.C., Hirsch, M.R. & Brunet, J.F. The expression pattern of the transcription factor Phox2 delineates synaptic pathways of the autonomic nervous system. J. Neurosci. 16, 7649–7660 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Heinrich, C. et al. Generation of subtype-specific neurons from postnatal astroglia of the mouse cerebral cortex. Nat. Protoc. 6, 214–228 (2011).

    CAS  PubMed  Google Scholar 

  66. Landt, S.G. et al. ChIP-seq guidelines and practices of the ENCODE and modENCODE consortia. Genome Res. 22, 1813–1831 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Nelson, J.D., Denisenko, O., Sova, P. & Bomsztyk, K. Fast chromatin immunoprecipitation assay. Nucleic Acids Res. 34, e2 (2006).

    PubMed  PubMed Central  Google Scholar 

  68. Trapnell, C., Pachter, L. & Salzberg, S.L. TopHat: discovering splice junctions with RNA-seq. Bioinformatics 25, 1105–1111 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    PubMed  PubMed Central  Google Scholar 

  70. Trapnell, C. et al. Differential analysis of gene regulation at transcript resolution with RNA-seq. Nat. Biotechnol. 31, 46–53 (2013).

    CAS  PubMed  Google Scholar 

  71. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Huang, D.W. et al. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    CAS  Google Scholar 

  73. Mazzoni, E.O. et al. Embryonic stem cell–based mapping of developmental transcriptional programs. Nat. Methods 8, 1056–1058 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Iacovino, M. et al. Inducible cassette exchange: a rapid and efficient system enabling conditional gene expression in embryonic stem and primary cells. Stem Cells 29, 1580–1588 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Eiraku, M. et al. Self-organized formation of polarized cortical tissues from ESCs and its active manipulation by extrinsic signals. Cell Stem Cell 3, 519–532 (2008).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank A. McMahon and J. Macklis for insightful advice in the early stages of the work; S. McConnell (Stanford University) for her generous sharing of the Fezf2PLAP line; J. Macklis (Harvard University), R. Hevner (Seattle Children's Research Institute), A. Catic (Massachusetts General Hospital) and S. Sykes (Massachusetts General Hospital) for sharing of antibodies and expression vectors; C. O'Donnell and D. Cacchiarelli for discussions on ChIP-seq analysis.; Z. Trayes-Gibson, E. Stronge and A. Iannone for outstanding technical support and for reading the manuscript; and A. Goodwin for careful editing of the manuscript and comments. This work was supported by grants from the US National Institutes of Health (NS062849, MH101268, NS078164 to P.A.), (HD078561 and HD069001 to E.T.), the New York Stem Cell Foundation (Robertson Investigator award to P.A.) and the Harvard Stem Cell Institute to P.A.

Author information

Authors and Affiliations

Authors

Contributions

S.L. and P.A. conceived the work, designed the experiments, analyzed the data and wrote the manuscript. S.L. performed the majority of the experiments. B.J.M. contributed to experimental design and performed microarray analysis. E.Z. performed the ChIP-seq experiments. L.A.G. analyzed the RNA-seq data. H.-H.C. performed the in vitro differentiation experiment and assisted in manuscript preparation. W.Y. performed the electrophoretic mobility shift assay experiment. A.M. assisted with FACS purification and the microarray experiments. E.T. performed the HARDI analysis. S.M. analyzed the ChIP-seq data. D.K.G. and J.L.R. supervised the bioinformatics analyses. P.A. supervised all aspects of the project.

Corresponding author

Correspondence to Paola Arlotta.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 FACS-purification of cortical progenitors electroporated in vivo with Fezf2GFP and CtrlGFP expression constructs at E14.5.

(a) Schematic of the experimental approach. (b) Coronal sections of CtrlGFP and Fezf2GFP-electroporated brains, 48 hours after electroporation, showing GFP-positive progenitors within the proliferative zones. CTIP2 immunohistochemistry (red) marks corticofugal neurons in the developing cortical plate. (c) Dissociated cortical cells before and after FACS purification; only a very small percentage of dissociated cells are electroporated (GFP-positive). FACS purification of GFP-positive cells results in virtually pure populations of labeled cells. CP, cortical plate. Scale bars, 100μm (b).

(Related to Figure 1)

Supplementary Figure 2 Expression profiles of Fezf2-induced genes at multiple embryonic and postnatal stages.

Expression profiles of 12 genes up-regulated by Fezf2 (shown in Figure 1) were examined by in situ hybridization in wild type cortices of E15.5, E18.5, P3, P7 and P14 wild type mice. CP, cortical plate. Scale bars, 50 μm.

(Related to Figure 1).

Supplementary Figure 3 Expression of Fezf2 target genes in Layer Vb subcerebral projection neurons is dependent on Fezf2 expression.

(a) In situ hybridizations for selected Fezf2-induced genes (blue signal) followed by immunocytochemistry for CTIP2 (brown signal) were performed on coronal sections of wild type mouse brains. (b) In situ hybridizations of selected Fezf2-induced genes on coronal sections of Fezf−/− brains (right) and wild type littermate controls (left). CP, cortical plate. Scale bars, 50 μm (a) left panels and (b), and 20 μm (a) right panels.

(Related to Figure 2).

Supplementary Figure 4 Fezf2-induced genes are expressed in nascent CSMN.

(a) Left, ternary plot of gene expression probabilities in VZ, SVZ/IZ and CP for Fezf2-induced targets shows enrichment in the cortical plate. Right, cluster representation of 186 genes induced by Fezf2 and also significantly enriched in the E14.5 CP. Y axis, FPKM (Fragments Per Kilobase RNA per Million mapped reads). Complete gene list is available in Supplementary Table 2. (b) Upper most left panel) In situ hybridization for Fezf2 on an E14.5 embryo shows the position of the cortical plate (CP). In situ hybridizations (www.genepaint.org) for prototypical genes induced by Fezf2 and enriched in the E14.5 CP (insets, enlarged from boxed areas). (c-d) In situ hybridizations for Hivep2 and Lmo3 at E14.5 (left panels) and at P4 (middle panels, http://developingmouse.brain-map.org) show specific expression in the developing CP and in layer V CSMN, confirming that Fezf2 induces expression of CSMN genes from early stages of development. Right panels, expression levels of Hivep2 (c) and Lmo3 (d) in CtrlGFP (blue line) and Fezf2GFP samples (red line) at 24 hours and 48 hours. Error bars indicate standard errors of the mean (SEM). CP, cortical plate; SVZ, subventricular zone; VZ, ventricular zone; IZ intermediate zone. Images shown in b; c and d left 2 panels are from the Genepaint database and c and d middle panels are from the Allen Brain Atlas database.

(Related to Figure 2).

Supplementary Figure 5 Fezf2-repressed genes are expressed in E14.5 VZ and SVZ/IZ cortical progenitors and in their progeny.

(a) Left, ternary plot of expression profiles collapsed to probability distributions in VZ, SVZ/IZ and CP for Fezf2-repressed genes shows enrichment in the VZ and the SVZ/IZ. Right, cluster representation of 73 genes repressed by Fezf2 and also significantly enriched in the E14.5 VZ-SVZ/IZ. Y axis, FPKM (Fragments Per Kilobase RNA per Million mapped reads). Complete gene list is available in Supplementary Table 2. (b) In situ hybridizations (www.genepaint.org) for prototypical genes repressed by Fezf2 and preferentially expressed in the E14.5 VZ-SVZ/IZ (insets, enlarged from boxed areas). (c,d) In situ hybridizations for Cux1 and Cux2 at E14.5 (left panels) showing specific expression in cortical progenitors. Middle panel, in situ hybridizations for Cux1 and Cux2 P4 showing specific expression in mature upper layer CPN (http://developingmouse.brain-map.org). Far right, expression levels of Cux1 and Cux2 in CtrlGFP (blue line) and Fezf2GFP samples (red line) are shown at 24 hours and 48 hours. Error bars indicate standard errors of the mean (SEM). CP, cortical plate; SVZ, subventricular zone; VZ, ventricular zone; IZ, intermediate zone. Images shown in b; c and d left 2 panels are from the Genepaint database and c and d middle panels are from the Allen Brain Atlas database.

(Related to Figure 2).

Supplementary Figure 6 3xFlag-Fezf2 recapitulates Fezf2 overexpression phenotype in vivo.

N-terminally FLAG-tagged Fezf2, employed for ChIP-seq and RNA-seq experiments, retains the ability to instruct the molecular identity and connectivity of deep layer projection neurons when ectopically expressed in progenitors of the upper layers at E14.5 (a) Experimental design of the ChIP-seq and RNA-seq approach. (b) Schematic of the experimental approach. (c) 3xFLAG-Fezf2 overexpression results in ectopic GFP+ cell aggregates below the corpus callosum, phenocopying untagged Fezf2 overexpression. (d) Immunohistochemistry for GFP, CTIP2 and TBR1 in electroporated brains harvested at P7 shows that 3xFLAG-Fezf2 is sufficient to induce a switch of fate to CTIP2+ and TBR1+ corticofugal projection neurons. (e) Immunohistochemistry for GFP shows axonal projections through the internal capsule (IC) (left panel), toward the thalamus (Th) (middle panel) and through the cerebral peduncle (CP) (right panel) upon 3xFLAG-Fezf2 overexpression. Ctx, cortex; Str, striatum; cc, corpus callosum. Scale bars, 100 μm (b, d), and 20 μm (c).

(Related to Figure 3).

Supplementary Figure 7 Overexpression of Fezf2 in neurosphere-derived neurons in vitro directs transcriptional changes toward the nascent CSMN state.

(a) Left, ternary plot of expression profiles collapsed to probability distributions in VZ, SVZ/IZ and CP for Fezf2-induced genes (identified from RNA-seq analysis) shows enrichment in the CP. Genes are classified as CP-specific (red), VZ-specific (blue), SVZ-specific (green), or non-specific (grey) using a specificity score threshold of 0.65 (detailed in Methods). Right, top 10 Gene Ontology categories from GO analysis of 224 genes up-regulated by Fezf2 in vitro. (b) Left, ternary plot of expression profiles collapsed to probability distributions in VZ, SVZ/IZ and CP for Fezf2-repressed genes (identified from RNA-seq analysis) shows enrichment in the VZ- SVZ/IZ. Right, top 10 Gene Ontology categories from GO analysis of 155 genes down-regulated by Fezf2 in vitro and specifically expressed in E14.5 VZ or SVZ/IZ. CP, cortical plate; VZ, ventricular zone; SVZ, subventricular zone; IZ, intermediate zone.

(Related to Figure 3).

Supplementary Figure 8 Fezf2 directly controls expression of its transcriptional targets via association with their promoter regions.

(a) Bar plots showing the percentage of FEZF2-bound genes among all genes (grey), Fezf2-regulated genes (green), Fezf2-induced genes (red), and Fezf2-repressed genes (blue) in in vitro cortical progenitors. (b) Table of gene counts, percentages of total, and p-values (hypergeometric test and bootstrap method) showing significant enrichment for FEZF2-bound genes in both the Fezf2-induced and Fezf2-repressed significant gene sets from in vitro cortical progenitors. (c) Bar plots showing the percentage of FEZF2-bound genes among all genes (grey), Fezf2-regulated genes (green), Fezf2-induced genes (red), and Fezf2-repressed genes (blue) in in vivo purified cortical progenitors. (d) Table of gene counts, percentages of total, and p-values (χ2-test) demonstrating significant enrichment for FEZF2-bound genes in both the Fezf2-induced and Fezf2-repressed significant gene sets from in vivo purified E14.5 cortical progenitors collected at 24h and 48h after Fezf2 over-expression. (e) Examples of 3xFLAG-Fezf2 peaks at the proximal promoters of CSMN-specific, Fezf2-induced genes (red peaks) and CPN-specific, Fezf2-repressed genes (blue peaks). Source data is shown in Supplementary Table 6.

(Related to Figure 3).

Supplementary Figure 9 Fezf2 directly associates with a CG-rich consensus sequence.

(a) The CG-rich FEZF2 consensus motif identified by GEM differs from two previously predicted FEZF2 motifs (SELEX and hPDI). (b) The motif-independent approach GPS found the GEM-defined motif more represented at the FEZF2-bound sites compared to the two previously defined motifs. FEZF2-bound Transcriptional Start Sites (TSSs) were also found preferentially associated with the GEM-defined motif compared to unbound TSSs. (c) Coomassie blue staining of purified GST-tFEZF2 protein (see Methods). The expected molecular weight of the GST-fusion truncated protein is 43.5 kDa, (arrow). (d) Electrophoretic mobility shift assay of probes for regions within the promoters of Ascl1 and EphB1 in the presence of GST-tFEZF2. Probes containing FEZF2 consensus motifs (EphB1-positive and Ascl1-positive) showed specific interaction with GST-tFEZF2 compared to probes without the consensus motifs in neighboring region of the same promoters (Ephb1-negative and Ascl1-negative). Arrow, probes bound to GST-tFEZF2; Asterisk, unbound probes.

(Related to Figure 3).

Supplementary Figure 10 Generation of inducible Fezf2 mouse ES cell lines.

(a) Strategy used for derivation of TET-ON Fezf2 mouse ES cell lines using the inducible cassette exchange method (see Methods). (b) Uncropped original immunoblots of Figure 4c.

Supplementary Figure 11 Axon guidance signaling molecules are highly represented in the GO analysis of Fezf2-induced genes and include the tyrosine kinase receptor Ephb1.

(a) GO analysis using Fezf2-induced genes at 24 hours and 48 hours identifies “axon guidance signaling” among the most significant categories (ranking as the most significant category at 48 hours). (b, far left panel) Schematic representation of the EphB1 gene targeting strategy (Deltagen Inc.). Arrows show positions and orientations of the primers employed for genotyping PCR. (b, middle panel) Genotyping PCR using primers flanking the targeted region identifies a 350 bp product corresponding to the wild type allele and a 300 bp product corresponding to the EphB1−/− allele. (b, most right panel) Western blot of brain lysate from P1 wild type, EphB1+/– pups, and EphB1−/− P1 pups using anti-EPHB1 antibody shows absence of EPHB1 protein (expected MW=100 kDa) in the EphB1−/− sample and a reduction in the EphB1+/– sample compared to wild type. Anti-β-tubulin is used as loading control. (c) Uncropped original immunoblots shown in panel (b, most right panel).

(Related to Figure 5).

Supplementary Figure 12 Expression patterns of EphB ligands in the mouse brain.

In situ hybridization for ephrin B1, ephrin B2 and ephrin B3 on horizontal wild type brain sections at E18.5 shows expression of ephrin B3 at the ventral forebrain midline. AC, anterior commissure. Scale bars, 100 μm.

(Related to Figure 7).

Supplementary Figure 13 Class-specific identity and distribution of cortical projection neurons is unaltered in Ephb1−/− mice.

(a) Schematic representation of the axon pathfinding phenotypes observed in both EphB1−/− and Fezf2−/− mice. (b) Schematic drawing of a P7 brain showing the position of area imaged. (c-d) Immunocytochemistry for CTIP2, TBR1, CUX1 and SATB2 show no difference in the layer distribution of these markers in EphB1−/− (d) compared to wild type mice (c). Scale bars, 100 μm.

(Related to Figure 7).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–13 (PDF 2814 kb)

Supplementary Methods Checklist (PDF 395 kb)

Supplementary Table 1

Genes induced and repressed upon Fezf2 overexpression in E14.5 cortical progenitors in vivo as shown in Figure 1. Gene lists include Fezf2- induced genes (fold change >1.5; p value <0.001) at 24 hours (263 genes), at 48 hours (441 genes), and all unique genes induced at both time points (589 genes); Fezf2-repressed genes (fold change >1.5; p value <0.001) at 24 hours (90 genes), at 48 hours (89 genes), and all unique genes repressed at both time points (153 genes). Relevant and complete Gene Ontology terms for each gene are tabulated. Results from in situ hybridization across multiple developmental stages are summarized from data shown in Fig. 1 and Supplementary Fig. 2. (XLSX 134 kb)

Supplementary Table 2

Fezf2-induced and Fezf2-repressed genes that are expressed in the E14.5 developing cortex in Supplementary Figures 4 and 5. Lists include genes induced by Fezf2 and enriched in E14.5 CP, and genes repressed by Fezf2 and enriched in E14.5 VZ and SVZ progenitors. Each transcript is assigned to a cluster, which includes genes with analogous expression profiles in VZ, SVZ, and CP at E14.5. (XLSX 16 kb)

Supplementary Table 3

Early postnatal (P3/P6) CSMN-specific genes induced by Fezf2 overexpression in vivo including those shown in Figure 2. 30 genes preferentially expressed in purified CSMN3 P3 & P6, p<0.0001, CSMN/CPN >1.5) that are also induced by Fezf2 overexpression in vivo (p<0.001, Fezf2GFP/CtrlGFP >1.5). (XLSX 9 kb)

Supplementary Table 4

Genes bound by Fezf2 at their TSS and signature genes used for GSEA analysis as shown in Figure 3. High-confidence genes bound by 3xFLAG-Fezf2 in two independent ChIP-seq replicates (3xFLAG-Fezf2 bound genes) are listed with Ensembl IDs. A separate list of CSMN genes3 and one of axon guidance signaling molecules induced by Fezf2 in vivo are shown with related FEZF2 promoter-binding information. CSMN signature genes are defined as genes with 3 fold or higher expression levels in CSMN than CPN at both P3 and P6 (103 genes, CPN/CSMN ≤ 1/3). CPN signature genes are defined as genes with 3 fold or higher expression levels in CPN than CSMN at both P3 and P6 (144 genes, CPN/CSMN ≥ 3). Signature genes that are also bound by 3xFLAG-Fezf2 are shown in red and in a separate column (47 CSMN signature genes and 77 CPN signature genes). (XLSX 381 kb)

Supplementary Table 5

Fezf2-regulated genes identified from RNA-seq analysis of cortical progenitors (cultured as neurospheres) in vitro that are also differentially expressed between E14.5 CP and SVZ/VZ as shown in Supplementary Figure 7. Lists include genes that are up-regulated and down-regulated by Fezf2 in vitro, genes that are both up-regulated by Fezf2 in vitro and enriched in E14.5 CP, and genes that are both down-regulated by Fezf2 and enriched in VZ- SVZ/IZ. (XLSX 95 kb)

Supplementary Table 8

Combined analysis of in vivo Fezf2-overexpression microarray and in vitro Fezf2 ChIP-seq as shown in Supplementary Figure 8. χ2 test results of the enrichment for FEZF2-bound genes in both the Fezf2-induced and Fezf2-repressed significant gene lists. (XLSX 14 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lodato, S., Molyneaux, B., Zuccaro, E. et al. Gene co-regulation by Fezf2 selects neurotransmitter identity and connectivity of corticospinal neurons. Nat Neurosci 17, 1046–1054 (2014). https://doi.org/10.1038/nn.3757

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3757

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