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:

Hominini-specific regulation of CBLN2 increases prefrontal spinogenesis

Abstract

The similarities and differences between nervous systems of various species result from developmental constraints and specific adaptations1,2,3,4. Comparative analyses of the prefrontal cortex (PFC), a cerebral cortex region involved in higher-order cognition and complex social behaviours, have identified true and potential human-specific structural and molecular specializations4,5,6,7,8, such as an exaggerated PFC-enriched anterior–posterior dendritic spine density gradient5. These changes are probably mediated by divergence in spatiotemporal gene regulation9,10,11,12,13,14,15,16,17, which is particularly prominent in the midfetal human cortex15,18,19,20. Here we analysed human and macaque transcriptomic data15,20 and identified a transient PFC-enriched and laminar-specific upregulation of cerebellin 2 (CBLN2), a neurexin (NRXN) and glutamate receptor-δ GRID/GluD-associated synaptic organizer21,22,23,24,25,26,27, during midfetal development that coincided with the initiation of synaptogenesis. Moreover, we found that species differences in level of expression and laminar distribution of CBLN2 are, at least in part, due to Hominini-specific deletions containing SOX5-binding sites within a retinoic acid-responsive CBLN2 enhancer. In situ genetic humanization of the mouse Cbln2 enhancer drives increased and ectopic laminar Cbln2 expression and promotes PFC dendritic spine formation. These findings suggest a genetic and molecular basis for the anterior-posterior cortical gradient and disproportionate increase in the Hominini PFC of dendritic spines and a developmental mechanism that may link dysfunction of the NRXN–GRID–CBLN2 complex to the pathogenesis of neuropsychiatric disorders.

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

Fig. 1: Phylogenetic, transcriptomic and regulatory characterization of developmental PFC upregulation of CBLN2.
Fig. 2: SOX5 represses CBLN2 through Hominini-specific regulatory deletions.
Fig. 3: In situ genetic humanization of the mouse Cbln2 enhancer drives upregulation and ectopic laminar Cbln2 expression in the neonatal frontal cortex.
Fig. 4: Increased density of dendritic spines in the mPFC of hCbln2 E2 mice.

Similar content being viewed by others

Code availability

All software and code used in this study are publicly available.

References

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

    Article  ADS  CAS  PubMed  Google Scholar 

  2. Barton, R. A. & Harvey, P. H. Mosaic evolution of brain structure in mammals. Nature 405, 1055–1058 (2000).

    Article  ADS  CAS  PubMed  Google Scholar 

  3. Krubitzer, L. & Kaas, J. The evolution of the neocortex in mammals: how is phenotypic diversity generated? Curr. Opin. Neurobiol. 15, 444–453 (2005).

    Article  CAS  PubMed  Google Scholar 

  4. Passingham, R. E. & Wise, S. P. The Neurobiology of the Prefrontal Cortex: Anatomy, Evolution, and the Origin of Insight (Oxford Univ. Press, 2015).

  5. Elston, G. N. et al. Specializations of the granular prefrontal cortex of primates: implications for cognitive processing. Anat. Rec. A Discov. Mol. Cell. Evol. Biol. 288, 26–35 (2006).

    Article  PubMed  Google Scholar 

  6. Semendeferi, K. et al. Spatial organization of neurons in the frontal pole sets humans apart from great apes. Cereb. Cortex 21, 1485–1497 (2011).

    Article  PubMed  Google Scholar 

  7. Kwan, K. Y. et al. Species-dependent posttranscriptional regulation of NOS1 by FMRP in the developing cerebral cortex. Cell 149, 899–911 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Gabi, M. et al. No relative expansion of the number of prefrontal neurons in primate and human evolution. Proc. Natl Acad. Sci. USA 113, 9617–9622 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Caceres, M. et al. Elevated gene expression levels distinguish human from non-human primate brains. Proc. Natl Acad. Sci. USA 100, 13030–13035 (2003).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Khaitovich, P. et al. Regional patterns of gene expression in human and chimpanzee brains. Genome Res. 14, 1462–1473 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Uddin, M. et al. Sister grouping of chimpanzees and humans as revealed by genome-wide phylogenetic analysis of brain gene expression profiles. Proc. Natl Acad. Sci. USA 101, 2957–2962 (2004).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Konopka, G. et al. Human-specific transcriptional networks in the brain. Neuron 75, 601–617 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Bauernfeind, A. L. et al. Evolutionary divergence of gene and protein expression in the brains of humans and chimpanzees. Genome Biol. Evol. 7, 2276–2288 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Sousa, A. M. M. et al. Molecular and cellular reorganization of neural circuits in the human lineage. Science 358, 1027–1032 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Zhu, Y. et al. Spatiotemporal transcriptomic divergence across human and macaque brain development. Science 362, eaat8077 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Pollen, A. A. et al. Establishing cerebral organoids as models of human-specific brain evolution. Cell 176, 743–756 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kanton, S. et al. Organoid single-cell genomic atlas uncovers human-specific features of brain development. Nature 574, 418–422 (2019).

    Article  ADS  CAS  PubMed  Google Scholar 

  18. Johnson, M. B. et al. Functional and evolutionary insights into human brain development through global transcriptome analysis. Neuron 62, 494–509 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Pletikos, M. et al. Temporal specification and bilaterality of human neocortical topographic gene expression. Neuron 81, 321–332 (2014).

    Article  CAS  PubMed  Google Scholar 

  20. Li, M. et al. Integrative functional genomic analysis of human brain development and neuropsychiatric risks. Science 362, eaat7615 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. Urade, Y. et al. Precerebellin is a cerebellum-specific protein with similarity to the globular domain of complement C1q B chain. Proc. Natl Acad. Sci. USA 88, 1069–1073 (1991).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hirai, H. et al. Cbln1 is essential for synaptic integrity and plasticity in the cerebellum. Nat. Neurosci. 8, 1534–1541 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Uemura, T. et al. Trans-synaptic interaction of GluRδ2 and neurexin through Cbln1 mediates synapse formation in the cerebellum. Cell 141, 1068–1079 (2010).

    Article  CAS  PubMed  Google Scholar 

  24. Matsuda, K. et al. Cbln1 is a ligand for an orphan glutamate receptor δ2, a bidirectional synapse organizer. Science 328, 363–368 (2010).

    Article  ADS  CAS  PubMed  Google Scholar 

  25. Yasumura, M. et al. Glutamate receptor delta1 induces preferentially inhibitory presynaptic differentiation of cortical neurons by interacting with neurexins through cerebellin precursor protein subtypes. J. Neurochem. 121, 705–716 (2012).

    Article  CAS  PubMed  Google Scholar 

  26. Wei, P. et al. The Cbln family of proteins interact with multiple signaling pathways. J. Neurochem. 121, 717–729 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Seigneur, E. & Sudhof, T. C. Genetic ablation of all cerebellins reveals synapse organizer functions in multiple regions throughout the brain. J. Neurosci. 38, 4774–4790 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Elston, G. N. Pyramidal cells of the frontal lobe: all the more spinous to think with. J. Neurosci. 20, RC95 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Jacobs, B. et al. Regional dendritic and spine variation in human cerebral cortex: a quantitative golgi study. Cereb. Cortex 11, 558–571 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Bianchi, S. et al. Synaptogenesis and development of pyramidal neuron dendritic morphology in the chimpanzee neocortex resembles humans. Proc. Natl Acad. Sci. USA 110, 10395–10401 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Molliver, M. E. et al. The development of synapses in cerebral cortex of the human fetus. Brain Res. 50, 403–407 (1973).

    Article  CAS  PubMed  Google Scholar 

  32. Voigt, T. et al. Synaptophysin immunohistochemistry reveals inside-out pattern of early synaptogenesis in ferret cerebral cortex. J. Comp. Neurol. 330, 48–64 (1993).

    Article  CAS  PubMed  Google Scholar 

  33. Rakic, P. et al. Concurrent overproduction of synapses in diverse regions of the primate cerebral cortex. Science 232, 232–235 (1986).

    Article  ADS  CAS  PubMed  Google Scholar 

  34. Shibata, M. et al. Regulation of prefrontal patterning and connectivity by retinoic acid. Nature https://doi.org/10.1038/s41586-021-03953-x (2021).

  35. Kang, H. J. et al. Spatiotemporal transcriptome of the human brain. Nature 478, 483–489 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  36. Lambert, N. et al. Genes expressed in specific areas of the human fetal cerebral cortex display distinct patterns of evolution. PLoS ONE 6, e17753 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  37. Miller, J. A. et al. Transcriptional landscape of the prenatal human brain. Nature 508, 199–206 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. ENCODE Project Consortium et al. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  ADS  CAS  Google Scholar 

  39. Chiang, M. Y. et al. An essential role for retinoid receptors RARβ and RXRγ in long-term potentiation and depression. Neuron 21, 1353–1361 (1998).

    Article  CAS  PubMed  Google Scholar 

  40. Krezel, W. et al. Impaired locomotion and dopamine signaling in retinoid receptor mutant mice. Science 279, 863–867 (1998).

    Article  ADS  CAS  PubMed  Google Scholar 

  41. 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).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  42. Shim, S. et al. Cis-regulatory control of corticospinal system development and evolution. Nature 486, 74–79 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  43. Clarke, R. A. & Eapen, V. Balance within the neurexin trans-synaptic connexus stabilizes behavioral control. Front. Hum. Neurosci. 8, 52 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. State, M. W. & Sestan, N. The emerging biology of autism spectrum disorders. Science 337, 1301–1303 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  45. Sudhof, T. C. Synaptic neurexin complexes: a molecular code for the logic of neural circuits. Cell 171, 745–769 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Willsey, A. J. et al. Coexpression networks implicate human midfetal deep cortical projection neurons in the pathogenesis of autism. Cell 155, 997–1007 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Gulsuner, S. et al. Spatial and temporal mapping of de novo mutations in schizophrenia to a fetal prefrontal cortical network. Cell 154, 518–529 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Lewis, D. A. & Mirnics, K. Transcriptome alterations in schizophrenia: disturbing the functional architecture of the dorsolateral prefrontal cortex. Prog. Brain Res. 158, 141–152 (2006).

    Article  CAS  PubMed  Google Scholar 

  49. Dy, P., Han, Y. & Lefebvre, V. Generation of mice harboring a Sox5 conditional null allele. Genesis 46, 294–299 (2008).

    Article  CAS  PubMed  Google Scholar 

  50. Mathelier, A. et al. JASPAR 2016: a major expansion and update of the open-access database of transcription factor binding profiles. Nucleic Acids Res. 44, D110–D115 (2016).

    Article  CAS  PubMed  Google Scholar 

  51. Khan, A. et al. JASPAR 2018: update of the open-access database of transcription factor binding profiles and its web framework. Nucleic Acids Res. 46, D260–D266 (2018).

    Article  CAS  PubMed  Google Scholar 

  52. Shim, S. et al. Regulation of EphA8 gene expression by TALE homeobox transcription factors during development of the mesencephalon. Mol. Cell. Biol. 27, 1614–1630 (2007).

    Article  CAS  PubMed  Google Scholar 

  53. Wang, H. et al. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell 153, 910–918 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Liu, P. et al. A highly efficient recombineering-based method for generating conditional knockout mutations. Genome Res. 13, 476–484 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  56. Wilkinson, D. G. & Nieto, M. A. Detection of messenger RNA by in situ hybridization to tissue sections and whole mounts. Methods Enzymol. 225, 361–373 (1993).

    Article  CAS  PubMed  Google Scholar 

  57. Hunt, C. A. et al. PSD-95 is associated with the postsynaptic density and not with the presynaptic membrane at forebrain synapses. J. Neurosci. 16, 1380–1388 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Essrich, C. et al. Postsynaptic clustering of major GABAA receptor subtypes requires the γ2 subunit and gephyrin. Nat. Neurosci. 1, 563–571 (1998).

    Article  CAS  PubMed  Google Scholar 

  59. Ippolito, D. M. & Eroglu, C. Quantifying synapses: an immunocytochemistry-based assay to quantify synapse number. J. Vis. Exp. 45, 2270 (2010).

    Google Scholar 

  60. Fiala, J. C. Reconstruct: a free editor for serial section microscopy. J. Microsc. 218, 52–61 (2005).

    Article  MathSciNet  CAS  PubMed  Google Scholar 

  61. Risher, W. C. et al. Rapid Golgi analysis method for efficient and unbiased classification of dendritic spines. PLoS ONE 9, e107591 (2014).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  62. Kaur, N. et al. Neural stem cells direct axon guidance via their radial fiber scaffold. Neuron 107, 1197–1211 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Meijering, E. et al. Design and validation of a tool for neurite tracing and analysis in fluorescence microscopy images. Cytometry A 58, 167–176 (2004).

    Article  CAS  PubMed  Google Scholar 

  64. Hinrichs, A. S. et al. The UCSC Genome Browser database: update 2006. Nucleic Acids Res. 34, D590–D598 (2006).

    Article  CAS  PubMed  Google Scholar 

  65. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24-26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Kent, W. J. et al. The human genome browser at UCSC. Genome Res. 12, 996–1006 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Rosenbloom, K. R. et al. ENCODE data in the UCSC Genome Browser: year 5 update. Nucleic Acids Res. 41, D56–D63 (2013).

    Article  CAS  PubMed  Google Scholar 

  68. Shibata, M. et al. MicroRNA-9 regulates neurogenesis in mouse telencephalon by targeting multiple transcription factors. J. Neurosci. 31, 3407–3422 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Morozov, Y. M., Ayoub, A. E. & Rakic, P. Translocation of synaptically connected interneurons across the dentate gyrus of the early postnatal rat hippocampus. J. Neurosci. 26, 5017–5027 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Morozov, Y. M., Mackie, K. & Rakic, P. Cannabinoid type 1 receptor is undetectable in rodent and primate cerebral neural stem cells but participates in radial neuronal migration. Int. J. Mol. Sci. 21, 1–19 (2020).

    Article  CAS  Google Scholar 

  71. Thompson, C. L. et al. A high-resolution spatiotemporal atlas of gene expression of the developing mouse brain. Neuron 83, 309–323 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank S. Bai, T. Nottoli and X. Xing for technical help with generating transgenic mice and genetically humanized mice; A. Sousa for providing tissue; B. Lorente-Galdos and G. Santpere for helping with data analysis; A. Duque for using equipment from MacBrainResource (MH113257); V. Lefebvre for providing floxed Sox5 mice; and the members of the Sestan laboratory for comments. This work was supported by the National Institutes of Health (HG010898, MH106874, MH106934, MH110926 and MH116488) and the Simons Foundation Autism Research Institute (SFARI) (572080; to N.S.). Additional support was provided by the National Institutes of Health T32 fellowship (MH18268 to K.P.), the National Science Foundation Graduate Research Fellowship Program (to S.K.M.), the Kavli Foundation and the James S. McDonnell Foundation (to N.S.).

Author information

Authors and Affiliations

Authors

Contributions

M.S., K.P. and N.S. designed the research. M.S. and K.P. performed the overall experiments and analysed the data. S.K.M. analysed the RNA sequencing, ChIP sequencing and genomic sequence data. M.S. generated the transgenic and knock-in mice. N.K. performed retrograde neuronal tracing with the AAV and analysed the data. Y.M.M. performed and analysed the electron microscopy experiments. X.C. and S.G.W. performed and analysed the preliminary electrophysiological analysis. N.S. conceived the study. M.S., K.P. and N.S. wrote the manuscript. All authors discussed the results and implications and commented on the manuscript at all stages.

Corresponding author

Correspondence to Nenad Sestan.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Alex Pollen and the other, anonymous, reviewer(s) for the peer review of this work.

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 Spatiotemporal expression of CBLN2 in the human and macaque neocortex.

ac, Spatiotemporal expression of CBLN2 expression in the human and macaque cerebral cortex during development base on BrainSpan (brainspan.org) and PsychENCODE (development.psychencode.org, evolution.psychencode.org) human and macaque RNA-seq data15,20. The RNA-seq data consisted of tissue-level samples comprising the pial surface, marginal zone, cortical plate (layers 2-6) and adjacent subplate zone, of eleven prospective neocortical areas. Red and blue lines indicate human and macaque PFC CBLN2 expression, respectively, and dotted lines represent the non-PFC CBLN2 expression. Vertical grey box demarcates mid-fetal developmental periods. Predicted ages, timeline of human and macaque development and the associated periods are shown below. Visual representation of CBLN2 in human cortex with heatmaps of regional expression in human and macaque below during period 4-6 (b) and period 12-13 (c). Darker reds represent high expression levels. d, Anteroposterior visual representation of human CBLN2 expression at PCW 21 from the BrainSpan human prenatal laser microdissection microarray data (brainspan.org)37.

Extended Data Fig. 2 Expression profile of CBLN1,3,4 genes related to dendrite and synapse development and CBLN2 binding partners in macaque and human.

a, Developmental trajectory of CBLN2 expression compared to the expression of key genes related to dendrite development (i.e., MAP1A and CAMK2A) and synapse development genes (i.e., SYP, SYPL1, SYPL2 and SYN1). The lists of genes related to synapse and dendrite development were previously complied and analyzed for their expression trajectories by Kang et al.35. b, Developmental trajectory of CBLN2 expression compared to the expression of genes encoding CBLN2 binding partners (i.e., GRID1, GRID2, NRXN1, NRXN2 and NRXN3). Left and right panels show gene expression in human and macaque, respectively. Gene expression in PFC and non-PFC are indicated by solid and dashed line, respectively. Vertical grey box highlights the mid-fetal developmental periods. Predicted ages, timeline of human and macaque development and the associated periods are shown below. Gene expression values are represented as log2(RPKM+1). For all of these plots, the shading around the lines represents the 95% confidence interval. Abbreviations are as described in Fig. 1 legend.

Extended Data Fig. 3 Mouse Cbln2 expression at multiple stages and Cbln2 E2 transgenic lines.

a, b, Visualization of Cbln2 expression in wholemount (a) and sagittal sections (b) at PCD 11.5, 13.5, 15.5 18.5 and PD 4, 14 from Allen Brain Atlas developing mouse brain atlas (developingmouse.brain-map.org)71. Arrowheads highlight early rostral expression, UL, upper layer; L5, layer 5; SP, subplate. c, β-Galactosidase activity in two additional mouse brains from independent transgenic mouse carrying Cbln2 E2 conjugated with lacZ reporter at PCD 17. See Fig. 1c for the third replicate. FP, frontal pole; OB, olfactory bulb. Scale bars, 1 mm.

Extended Data Fig. 4 Comparative analysis of CBLN2 E2 deletions across mammals.

a, Schematic representation of CBLN2 E2 from mouse and primate species including apes (human, common chimpanzee, bonobo, gorilla, orangutan and gibbon), old world monkey (Rhesus macaque), new world monkey (marmoset) and prosimians (tarsier and lemur). HSD1, HSD2 and I3 are shaded. Putative RAR-RXR tandem binding sites indicated as red lines and putative SOX5-binding sites as blue lines. b, Sequence alignments of HSD1, HSD2 and I3 from macaque, mouse, and rat. Putative SOX5 binding sites are shown in red boxes. c, Sequence alignments of HSD1 and HSD2 from primates shown in a.

Extended Data Fig. 5 Conservation of CBLN2 E2 and deleted regions across species.

Phylogenetic tree of selected chordates including placental and non-placental mammals with information about presence of the CBLN2/Cbln2 gene and CBLN2 E2/Cbln2 E2. The last three columns describe the presence of HSD1, HSD2 and I3. The mouse E2 sequence was searched in each most updated animal genome browser at UCSC genome browser (as of December 22, 2019). E2 conservation criteria are: 1) identity over 80%; and 2) alignment length over 600 bp compared with mouse E2 (1005 bp).

Extended Data Fig. 6 SOX5 directly suppresses RXRG-RARB responsive human, chimpanzee, gorilla and mouse Cbln2 E2 enhancer.

a, Luciferase assay in Neuro-2a cell line from luciferase reporters conjugated to chimpanzee and gorilla wild CBLN2 E2. Two-tailed Student’s t-test; *P = 0.01; ***P = 9e-5 (Chimpanzee), 1e-5 (Gorilla); NS, not significant. Error bars; S.E.M.; N = 3 per condition. b, Overexpression of human SOX5 exerts a similar effect to mouse Sox5 on human and mouse Cbln2 E2 reporters. Two-tailed Student’s t-test; *P = 5e-3; ***P = 4e-5; ****P = 2e-6; 2e-6, 1e-6 (Human SOX5 + mouse Cbln2 E2); NS, not significant; Error bars, S.E.M.; N = 3 per condition. c, Cbln2 expression is upregulated in Sox5 conditional knockout brain at PD 0. Additional two replicates (Repl #2 and 3) not shown in Fig. 2d are shown. Scale bar, 200 μm (left); 100 μm (right). d, Constructs used for luciferase assay and generation of transgenic animals shown in e, f. e, Transgenic mouse brain at PCD 17 carrying Cbln2 E2 (N = 3) or Cbln2 E2 Fr1-lacZ reporters (N = 3). Cbln2 expression in the PFC is indicated by arrowheads. Endogenous Cbln2 expression is also shown for comparison (N = 4). Scale bar, 200 μm. f, Luciferase assay for Cbln2 E2 and Cbln2 E2-Fr1. Two-tailed Student’s t-test; **P = 1e-4 (Cbln2 E2), 2e-4 (Cbln2 E2-Fr1); ***P = 8e-5; NS, not significant; Error bars: S.E.M.; N = 3 per condition.

Extended Data Fig. 7 Humanized Cbln2 E2 knock-in mouse shows increased Cbln2 expression in neonatal neocortex.

a, Positions of single-guide RNAs (sgRNA1 and 2) to introduce double-strand breaks in the genomic DNA and targeting vector to replace WT mouse Cbln2 E2 region with that of human CBLN2 E2 (humanized hCbln2 E2) are shown. b, Genotyping strategy for F1 mice. Germline transmission in the F1 generation was confirmed by nested PCR using the primer set of mP3/mP4, followed by hP1/P2 as indicated in a. Two founders #13 and #25 were obtained. c, Mice in the following generation were genotyped by PCR with hP1/P2 and mP1/mP2. An example of genotyping for F2 of line #13 mice is shown (c). d, Comparison of Cbln2 expression between WT Cbln2 E2 (Mm;Mm) and hCbln2 E2 (Hs;Hs) neocortex at PD 0 using quantitative reverse transcription-PCR. RNA was extracted from the neocortex following the removal of hippocampus, olfactory bulb and subpallial regions. Two-tailed Student’s t-test; *P = 0.007; Error bars: S.E.M.; N = 5 per genotype. Genotyping were repeated at least two times in b and c.

Extended Data Fig. 8 Additional replicates.

Additional replicates from Fig. 3. a, b, The neocortex of the humanized hCbln2 E2 knock-in prenatal and neonatal mice exhibits upregulated Cbln2 in both upper and deeper layers compared to mice carrying WT Cbln2 E2. Additional two replicates (Repl #2 and 3) not shown in Fig. 3a are shown for PND 16 and PND18. Scale bars, 200 μm (left); 100 μm (right). All analysis and three replicates for PD1. Neocortex was divided into six equal bins spanning from pia to ventricular zone, and Cbln2 signal intensity was quantified for each bin and compared between WT and hCbln2 E2 (Hs;Hs). Two-tailed Student’s t-test; *P = 0.02 (PCD18), 0.02, 0.04 (PD1), **P = 1e-3 (PCD18), 1e-3, 3e-3, 3e-3 (PD1), ***P = 1e-4, 2e-4 (PCD18); N = 3 per genotype. Scale bars, 100 μm. c, Expression of the upper layer marker, Cux2, and SOX5 in adjacent tissue sections were detected by in situ hybridization and immunostaining, respectively. Scale bars, 100 μm. N = 3.

Extended Data Fig. 9 Expression of Sox5 in the developing mouse and human neocortex.

a, Double immunofluorescent staining for SOX5 and BCL11B in PD 0 and PD 7 mouse neocortex. N = 3 per condition. Scale bar, 100 μm. b, SOX5 expression in the PFC and non-PFC areas of the cerebral cortex of human and macaque across development. Red and blue lines indicate human and macaque, respectively. Vertical grey box demarcates mid-fetal developmental periods. For all of those plots, the shading around the lines represents the 95% confidence interval. Predicted ages, timeline of human and macaque development and the associated periods are shown below.

Extended Data Fig. 10 Quantification of excitatory and inhibitory postsynaptic puncta.

ad, PD 0 WT and hCbln2 E2 mPFC L2-3 and L5 (BCL11B-immunopositive) immunostained for PSD-95/DLG4 or GPHN. Density of PSD-95/DLG4+ excitatory and GPHN+ inhibitory postsynaptic puncta in PD 0 and PD 60 mPFC, primary somatosensory area (SSp), and primary visual area (VISp) of WT (blue), hCbln2 E2 (orange), and Sox5 cKO mice (green). Two-tailed Student’s test; *P = 0.018; ***P = 4e-4, 1e-4 (b, WT vs. humanized hCbln2 E2 L2-3, and L5, respectively), 1e-4, 7e-5, 4e-4, 2e-4 (d, WT vs. humanized hCbln2 E2 L2-3 and L5, respectively; WT vs. Sox5 cKO L2-3 and L5, respectively); ****P < 1e-5. Error bars: S.E.M.; N = 5 per PD 0 genotype; N = 3 per PD 60 genotype. Scale bar, 25 μm.

Extended Data Fig. 11 Analysis of dendritic spines and dendritic complexity.

ac, Quantification of Sholl analysis and representative images (inset) of Golgi stained dendrites and dendritic spines per 10 μm in layer 2-3 and 5 of mPFC (a), SSp (b) and VISp (c) in WT Cbln2 E2 and humanized hCbln2 E2. Two-way ANOVA with Sidak’s multiple comparison method was applied; *P = 0.04; **P = 1e-3, ****P < 0001; Error bars: S.E.M.; N = 16 (WT Cbln2 E2), and 16 (humanized hCbln2 E2) of PD 60 brains (N = 3). Scale bars: 5 mm. d, e, Quantification and representative images of juxtaposed synaptophysin (SYP) and DLG4/PSD-95 immunolabelled puncta in upper (UL) and deep layer (DL) mPFC of WT Cbln2 E2 and humanized hCbln2 E2. Two-tailed Student’s t-test was applied * P = 0.03; Error bars: S.E.M. (N = 3); Scale bar: 10 mm. f, Electron microscopy (EM) image shows dendrite emitting spine with postsynaptic density (PSD) and spine apparatus (SA) (left) in PD 60 humanized hCbln2 E2 mouse. 3D reconstruction (right) of the dendrite with numerous spines showing spine heads and synaptic contacts (red). Double arrow points to the spine shown in EM image. Thin spines without head (TS) and not innervated thin extensions (arrow) are also detected. Abbreviation: M, mitochondria. Sixteen-representable dendrite fragments were traced in the serial images (see Methods). g, Quantification of percentage of innervated spines in mPFC of WT Cbln2 E2 and humanized hCbln2 E2. Two-tailed Student’s t-test was applied. NS, Not significant. Error bars: S.E.M. (N = 3).

Supplementary information

Supplementary Fig. 1

Full image of ChIP-PCR assays from Figure 2c in PD 0 mouse neocortex using anti-RNA polymerase II (anti-POL2), IgG, and anti-SOX5 and Cbln2 E1 and E2 primers with relevant marker sizes defined.

Reporting Summary

Supplementary Table 1

Comparative analysis of the transcription factor binding sites in CBLN2 E2.

Supplementary Table 2

List and sequence of relevant primers.

Supplementary Table 3

Statistics for all comparisons in Figure 1a.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shibata, M., Pattabiraman, K., Muchnik, S.K. et al. Hominini-specific regulation of CBLN2 increases prefrontal spinogenesis. Nature 598, 489–494 (2021). https://doi.org/10.1038/s41586-021-03952-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-03952-y

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