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:

Patterned cPCDH expression regulates the fine organization of the neocortex

Abstract

The neocortex consists of a vast number of diverse neurons that form distinct layers and intricate circuits at the single-cell resolution to support complex brain functions1. Diverse cell-surface molecules are thought to be key for defining neuronal identity, and they mediate interneuronal interactions for structural and functional organization2,3,4,5,6. However, the precise mechanisms that control the fine neuronal organization of the neocortex remain largely unclear. Here, by integrating in-depth single-cell RNA-sequencing analysis, progenitor lineage labelling and mosaic functional analysis, we report that the diverse yet patterned expression of clustered protocadherins (cPCDHs)—the largest subgroup of the cadherin superfamily of cell-adhesion molecules7—regulates the precise spatial arrangement and synaptic connectivity of excitatory neurons in the mouse neocortex. The expression of cPcdh genes in individual neocortical excitatory neurons is diverse yet exhibits distinct composition patterns linked to their developmental origin and spatial positioning. A reduction in functional cPCDH expression causes a lateral clustering of clonally related excitatory neurons originating from the same neural progenitor and a significant increase in synaptic connectivity. By contrast, overexpression of a single cPCDH isoform leads to a lateral dispersion of clonally related excitatory neurons and a considerable decrease in synaptic connectivity. These results suggest that patterned cPCDH expression biases fine spatial and functional organization of individual neocortical excitatory neurons in the mammalian brain.

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: Patterned expression of cPcdh isoforms linked to lineage relationship and developmental origin.
Fig. 2: Coupling between cPcdh expression and spatial configuration of the clone.
Fig. 3: PCDHγ removal causes a lateral clustering of clonally related excitatory neurons.
Fig. 4: Pcdhgc3 overexpression leads to a lateral dispersion of clonally related excitatory neurons.
Fig. 5: PCDHγ removal enhances the preferential synaptic connectivity between clonally related excitatory neurons.
Fig. 6: Pcdhgc3 overexpression disrupts preferential synaptic connectivity between clonally related excitatory neurons.

Similar content being viewed by others

Data availability

The scRNA-seq matrix of cPcdh isoform expression is provided in Supplementary Table 4Source data are provided with this paper.

Code availability

No code or model was generated in this study.

References

  1. Luo, L. Architectures of neuronal circuits. Science 373, eabg7285 (2021).

    Article  CAS  Google Scholar 

  2. Sanes, J. R. & Zipursky, S. L. Synaptic specificity, recognition molecules, and assembly of neural circuits. Cell 181, 536–556 (2020).

    Article  CAS  Google Scholar 

  3. Mountoufaris, G., Canzio, D., Nwakeze, C. L., Chen, W. V. & Maniatis, T. Writing, reading, and translating the clustered protocadherin cell surface recognition code for neural circuit assembly. Annu. Rev. Cell Dev. Biol. 34, 471–493 (2018).

    Article  CAS  Google Scholar 

  4. Yogev, S. & Shen, K. Cellular and molecular mechanisms of synaptic specificity. Annu. Rev. Cell Dev. Biol. 30, 417–437 (2014).

    Article  CAS  Google Scholar 

  5. Sudhof, T. C. The cell biology of synapse formation. J. Cell Biol. 220, e202103052 (2021).

    Article  CAS  Google Scholar 

  6. Yagi, T. Molecular codes for neuronal individuality and cell assembly in the brain. Front. Mol. Neurosci. 5, 45 (2012).

    Article  CAS  Google Scholar 

  7. Wu, Q. & Maniatis, T. A striking organization of a large family of human neural cadherin-like cell adhesion genes. Cell 97, 779–790 (1999).

    Article  CAS  Google Scholar 

  8. Kriegstein, A. & Alvarez-Buylla, A. The glial nature of embryonic and adult neural stem cells. Annu. Rev. Neurosci. 32, 149–184 (2009).

    Article  CAS  Google Scholar 

  9. Malatesta, P., Hartfuss, E. & Gotz, M. Isolation of radial glial cells by fluorescent-activated cell sorting reveals a neuronal lineage. Development 127, 5253–5263 (2000).

    Article  CAS  Google Scholar 

  10. Noctor, S. C., Flint, A. C., Weissman, T. A., Dammerman, R. S. & Kriegstein, A. R. Neurons derived from radial glial cells establish radial units in neocortex. Nature 409, 714–720 (2001).

    Article  ADS  CAS  Google Scholar 

  11. Noctor, S. C., Martinez-Cerdeno, V., Ivic, L. & Kriegstein, A. R. Cortical neurons arise in symmetric and asymmetric division zones and migrate through specific phases. Nat. Neurosci. 7, 136–144 (2004).

    Article  CAS  Google Scholar 

  12. Gao, P. et al. Deterministic progenitor behavior and unitary production of neurons in the neocortex. Cell 159, 775–788 (2014).

    Article  CAS  Google Scholar 

  13. Luskin, M. B., Pearlman, A. L. & Sanes, J. R. Cell lineage in the cerebral cortex of the mouse studied in vivo and in vitro with a recombinant retrovirus. Neuron 1, 635–647 (1988).

    Article  CAS  Google Scholar 

  14. Rakic, P. Specification of cerebral cortical areas. Science 241, 170–176 (1988).

    Article  ADS  CAS  Google Scholar 

  15. Shen, Z. et al. Distinct progenitor behavior underlying neocortical gliogenesis related to tumorigenesis. Cell Rep. 34, 108853 (2021).

    Article  CAS  Google Scholar 

  16. Walsh, C. & Cepko, C. L. Clonally related cortical cells show several migration patterns. Science 241, 1342–1345 (1988).

    Article  ADS  CAS  Google Scholar 

  17. Yu, Y. C., Bultje, R. S., Wang, X. Q. & Shi, S. H. Specific synapses develop preferentially among sister excitatory neurons in the neocortex. Nature 458, 501–504 (2009).

    Article  ADS  CAS  Google Scholar 

  18. Yu, Y. C. et al. Preferential electrical coupling regulates neocortical lineage-dependent microcircuit assembly. Nature 486, 113–117 (2012).

    Article  ADS  CAS  Google Scholar 

  19. Li, Y. et al. Clonally related visual cortical neurons show similar stimulus feature selectivity. Nature 486, 118–121 (2012).

    Article  ADS  CAS  Google Scholar 

  20. Ohtsuki, G. et al. Similarity of visual selectivity among clonally related neurons in visual cortex. Neuron 75, 65–72 (2012).

    Article  CAS  Google Scholar 

  21. He, S. J., Li, Z. Z., Ge, S. Y., Yu, Y. C. & Shi, S. H. Inside-out radial migration facilitates lineage-dependent neocortical microcircuit assembly. Neuron 86, 1159–1166 (2015).

    Article  CAS  Google Scholar 

  22. Wu, Q. et al. Comparative DNA sequence analysis of mouse and human protocadherin gene clusters. Genome Res. 11, 389–404 (2001).

    Article  CAS  Google Scholar 

  23. Canzio, D. et al. Antisense lncRNA transcription mediates DNA demethylation to drive stochastic protocadherin alpha promoter choice. Cell 177, 639–653 (2019).

    Article  CAS  Google Scholar 

  24. Tasic, B. et al. Promoter choice determines splice site selection in protocadherin α and γ pre-mRNA splicing. Mol. Cell 10, 21–33 (2002).

    Article  CAS  Google Scholar 

  25. Jia, Z. et al. Tandem CTCF sites function as insulators to balance spatial chromatin contacts and topological enhancer-promoter selection. Genome Biol. 21, 75 (2020).

    Article  CAS  Google Scholar 

  26. Esumi, S. et al. Monoallelic yet combinatorial expression of variable exons of the protocadherin-alpha gene cluster in single neurons. Nat. Genet. 37, 171–176 (2005).

    Article  CAS  Google Scholar 

  27. Mountoufaris, G. et al. Multicluster Pcdh diversity is required for mouse olfactory neural circuit assembly. Science 356, 411–413 (2017).

    Article  ADS  CAS  Google Scholar 

  28. Toyoda, S. et al. Developmental epigenetic modification regulates stochastic expression of clustered protocadherin genes, generating single neuron diversity. Neuron 82, 94–108 (2014).

    Article  CAS  Google Scholar 

  29. Thu, C. A. et al. Single-cell identity generated by combinatorial homophilic interactions between α, β, and γ protocadherins. Cell 158, 1045–1059 (2014).

    Article  CAS  Google Scholar 

  30. Rubinstein, R. et al. Molecular logic of neuronal self-recognition through protocadherin domain interactions. Cell 163, 629–642 (2015).

    Article  CAS  Google Scholar 

  31. Brasch, J. et al. Visualization of clustered protocadherin neuronal self-recognition complexes. Nature 569, 280–283 (2019).

    Article  ADS  CAS  Google Scholar 

  32. Schreiner, D. & Weiner, J. A. Combinatorial homophilic interaction between γ-protocadherin multimers greatly expands the molecular diversity of cell adhesion. Proc. Natl Acad. Sci. USA 107, 14893–14898 (2010).

    Article  ADS  CAS  Google Scholar 

  33. Kostadinov, D. & Sanes, J. R. Protocadherin-dependent dendritic self-avoidance regulates neural connectivity and circuit function. eLife 4, e08964 (2015).

    Article  Google Scholar 

  34. Lefebvre, J. L., Kostadinov, D., Chen, W. S. V., Maniatis, T. & Sanes, J. R. Protocadherins mediate dendritic self-avoidance in the mammalian nervous system. Nature 488, 517–521 (2012).

    Article  ADS  CAS  Google Scholar 

  35. Ing-Esteves, S. et al. Combinatorial effects of alpha- and gamma-protocadherins on neuronal survival and dendritic self-avoidance. J. Neurosci. 38, 2713–2729 (2018).

    Article  CAS  Google Scholar 

  36. Chen, W. S. V. et al. Pcdhαc2 is required for axonal tiling and assembly of serotonergic circuitries in mice. Science 356, 406–410 (2017).

    Article  ADS  CAS  Google Scholar 

  37. Zong, H., Espinosa, S., Su, H. H., Muzumdar, M. D. & Luo, L. Q. Mosaic analysis with double markers in mice. Cell 121, 479–492 (2005).

    Article  CAS  Google Scholar 

  38. Bonaguidi, M. A. et al. In vivo clonal analysis reveals self-renewing and multipotent adult neural stem cell characteristics. Cell 145, 1142–1155 (2011).

    Article  CAS  Google Scholar 

  39. Lv, X. et al. TBR2 coordinates neurogenesis expansion and precise microcircuit organization via protocadherin 19 in the mammalian cortex. Nat. Commun. 10, 3946 (2019).

    Article  ADS  Google Scholar 

  40. Kessaris, N. et al. Competing waves of oligodendrocytes in the forebrain and postnatal elimination of an embryonic lineage. Nat. Neurosci. 9, 173–179 (2006).

    Article  CAS  Google Scholar 

  41. Kaneko, R. et al. Allelic gene regulation of Pcdh-α and Pcdh-γ clusters involving both monoallelic and biallelic expression in single Purkinje cells. J. Biol. Chem. 281, 30551–30560 (2006).

    Article  CAS  Google Scholar 

  42. Yao, Z. Z. et al. A transcriptomic and epigenomic cell atlas of the mouse primary motor cortex. Nature 598, 103–110 (2021).

    Article  ADS  CAS  Google Scholar 

  43. Prasad, T., Wang, X. Z., Gray, P. A. & Weiner, J. A. A differential developmental pattern of spinal interneuron apoptosis during synaptogenesis: insights from genetic analyses of the protocadherin-gamma gene cluster. Development 135, 4153–4164 (2008).

    Article  CAS  Google Scholar 

  44. Lefebvre, J. L., Zhang, Y. F., Meister, M., Wang, X. Z. & Sanes, J. R. γ-Protocadherins regulate neuronal survival but are dispensable for circuit formation in retina. Development 135, 4141–4151 (2008).

    Article  CAS  Google Scholar 

  45. Cadwell, C. R. et al. Cell type composition and circuit organization of clonally related excitatory neurons in the juvenile mouse neocortex. eLife 9, e52951 (2020).

    Article  CAS  Google Scholar 

  46. Schmucker, D. et al. Drosophila Dscam is an axon guidance receptor exhibiting extraordinary molecular diversity. Cell 101, 671–684 (2000).

    Article  CAS  Google Scholar 

  47. Hattori, D. et al. Dscam diversity is essential for neuronal wiring and self-recognition. Nature 449, 223–227 (2007).

    Article  ADS  CAS  Google Scholar 

  48. Hattori, D., Millard, S. S., Wojtowicz, W. M. & Zipursky, S. L. Dscam-mediated cell recognition regulates neural circuit formation. Annu. Rev. Cell Dev. Biol. 24, 597–620 (2008).

    Article  CAS  Google Scholar 

  49. Liu, C. et al. Dscam1 establishes the columnar units through lineage-dependent repulsion between sister neurons in the fly brain. Nat. Commun. 11, 4067 (2020).

    Article  ADS  CAS  Google Scholar 

  50. Garrett, A. M., Schreiner, D., Lobas, M. A. & Weiner, J. A. γ-Protocadherins control cortical dendrite arborization by regulating the activity of a FAK/PKC/MARCKS signaling pathway. Neuron 74, 269–276 (2012).

    Article  CAS  Google Scholar 

  51. Hippenmeyer, S. et al. Genetic mosaic dissection of Lis1 and Ndel1 in neuronal migration. Neuron 68, 695–709 (2010).

    Article  CAS  Google Scholar 

  52. Wang, X., Qiu, R., Tsark, W. & Lu, Q. Rapid promoter analysis in developing mouse brain and genetic labeling of young neurons by doublecortin-DsRed-express. J. Neurosci. Res. 85, 3567–3573 (2007).

    Article  CAS  Google Scholar 

  53. Dursun, I. et al. Effects of early postnatal exposure to ethanol on retinal ganglion cell morphology and numbers of neurons in the dorsolateral geniculate in mice. Alcohol Clin. Exp. Res. 35, 2063–2074 (2011).

    Article  CAS  Google Scholar 

  54. Tasic, B. et al. Shared and distinct transcriptomic cell types across neocortical areas. Nature 563, 72–78 (2018).

    Article  ADS  CAS  Google Scholar 

  55. Cadwell, C. R. et al. Multimodal profiling of single-cell morphology, electrophysiology, and gene expression using Patch-seq. Nat. Protoc. 12, 2531–2553 (2017).

    Article  CAS  Google Scholar 

  56. Gu, Z. G., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).

    Article  CAS  Google Scholar 

  57. Yao, Z. Z. et al. A taxonomy of transcriptomic cell types across the isocortex and hippocampal formation. Cell 184, 3222–3241 (2021).

    Article  CAS  Google Scholar 

  58. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Shi laboratory and H. Jiang for discussions and input; and J. L. Lefebvre and J. A. Weiner for the Pcdhgfcon3 and Pcdhgc3 mouse lines. This work was supported by grants from the Ministry of Science and Technology of China (2021ZD0202300), the National Natural Science Foundation of China (32021002 to S.-H.S. and 31630039 to Q.W.), Beijing Outstanding Young Scientist Program (BJJWZYJH01201910003012), Beijing Municipal Science & Technology Commission (Z20111000530000 and Z211100003321001), the Chinese Institute for Brain Research (Beijing) and the Simons Foundation (SFARI GC232866) (to S.-H.S.) and the Postdoctoral Program of the Tsinghua Centre for Life Sciences (to S.L.).

Author information

Authors and Affiliations

Authors

Contributions

X.L. and S.-H.S. conceived the project. X.L. performed the majority of the experiments and data analysis. S.L., B.L. and S.H. performed electrophysiological recordings and analysis. X.G. and S.L. helped with the scRNA-seq experiments. J.L., X.-Y.Y., X.G., S.Z. and J. Yang performed the scRNA-seq data analysis. Y.L. helped with the immunostaining. X.Z. and J. Yan helped with the mouse genotyping. A.L.J. advised on the project. H.S. helped with isoform detection and sequencing design. X.L., Q.W. and S.-H.S. wrote the paper with input from all of the other authors.

Corresponding author

Correspondence to Song-Hai Shi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.

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 MADM-labelling of individual excitatory neuron clones in the neocortex and quality check of cell aspiration and extraction.

a, Schematic diagram of MADM labelling strategy. b, c, Representative 3D reconstruction images of symmetric (b) or asymmetric (c) excitatory neuron clones induced at E12 and analysed at P21. Different coloured dots represent the cell bodies of labelled neurons. The x-/y-/z-axes indicate the spatial orientation of the clone with the x-axis parallel to the brain pial surface and the y-axis perpendicular to the pial surface. Coloured lines indicate the layer boundaries. Scale bars, 100 μm. d, e, Representative fluorescence images of individual clones in live brain slices subjected to glass pipette aspiration and extraction shown in Fig. 1band c. Scale bars, 50 μm. fh, Quantifications of amplified cDNAs from negative controls (f, g) or individual extracted neurons (h) in the same experiment setting. i, Quantification of the cDNA library of individual extracted neurons. Data are representative of at least three independent experiments.

Extended Data Fig. 2 Quality check of the scRNA-seq analysis.

ac, Quality control of individual extracted neurons after sequencing. Neurons with a low total detected gene number (n = 14), or a high mitochondrial gene (n = 2), or ERCC (n = 7) percentage (orange) were excluded from further analysis. Each symbol represents a neuron. d, e, Quantification of the number of the total detected genes (d) and cPcdh isoforms (e) in our clone dataset (n = 188). Each symbol represents a neuron. f, Histogram of the number of the detected cPcdh isoforms per neuron in our clone dataset (n = 188). g, h, Quantification of the number of the total detected gene (g) and cPcdh isoforms (h) in the previously published randomly collected neocortical excitatory neuron scRNA-seq dataset from the Allen Institute42 (n = 4,152). Each symbol represents a neuron. i, Histogram of the number of the detected cPcdh isoforms in the previously published randomly collected neocortical excitatory neuron scRNA-seq dataset from the Allen Institute42 (n = 4,152). j, Quantification of the number of the total detected cPcdh isoforms in clonally related excitatory neurons from different neocortical regions (motor cortex/MO, n = 32; somatosensory cortex/SS, n = 116; visual cortex/VIS, n = 40). k, Pearson correlation analysis between the average expression level of each cPcdh isoform in clonally related excitatory neurons of SS (n = 116) and VIS (n = 40). Each dot represents a cPcdh isoform. The grey bar indicates the 95% confidence interval. A similar display is used in subsequent panels (l–n). l, Pearson correlation analysis between the average expression level of each cPcdh isoform in clonally related excitatory neurons of SS (n = 116) and MO (n = 32). m, Pearson correlation analysis between the average expression level of each cPcdh isoform in clonally related excitatory neurons of MO (n = 32) and VIS (n = 40). n, Pearson correlation analysis between the average expression level of each cPcdh isoform in randomly collected excitatory neurons of MO (n = 3,893) and VIS (n = 7,347) in the previously published scRNA-seq datasets from the Allen Institute54. o, Frequency distribution of the mean similarities of cPcdh isoform expression pattern in the non-clonal dataset (n = 1,000 trails, grey bars), and the mean similarity of the clonal dataset (red line) from 32 clones. The non-clonal dataset was generated by a random permutation of the clonal dataset and statistics were performed using the permutation test (see Methods). p, Quantification of the pairwise similarity of cPcdh isoform expression for clonally related neocortical excitatory neurons in the same or different layers per clone (same layers, n = 31; different layers, n = 20). q, Quantification of the pairwise similarity of cPcdh isoform expression for neurons in the same or different layers of the randomly collected 150 neocortical excitatory neurons from the previously published Allen Institute dataset42 (same layers, n = 600; different layers, n = 400). r, Quantification of the pairwise similarity of cPcdh isoform expression for randomly collected excitatory neurons across different layers from the previously published Allen Institute dataset42 (L2/3-L2/3, n = 200; L2/3-L5, n = 200; L2/3-L6, n = 200). The n numbers indicate neurons (a–n), clones (p), and neuron pairs (q, r). Data are representative of at least three independent experiments. One-way ANOVA test without adjusted P value (j); Two-tailed Pearson correlation analysis (kn); One-tailed permutation test (o); Two-tailed unpaired Student’s t-test (pr). Box plots as in Fig. 1.

Source data

Extended Data Fig. 3 Combinatorial expression of cPcdh isoforms in individual neocortical excitatory neurons in the clone dataset.

Heatmap of cPcdh transcripts in 188 individual neocortical excitatory neurons from 32 clones. The existence of individual cPcdh isoforms is indicated by the red or green coloured boxes, the colour reflects fluorescence labelling, and the expression level is indicated by the colour gradient. Note that the majority of neurons express multiple cPcdh isoforms from three Pcdh clusters (a, b, and g) and the C-type Pcdh isoforms (ac2 and gc5).

Extended Data Fig. 4 Combinatorial expression of cPcdh isoforms in individual excitatory neurons of the motor cortex in the previously published Allen Institute dataset.

Heatmap of cPcdh transcripts in 200 individual excitatory neurons in the motor cortex randomly selected from the published Allen Institute dataset42. The existence of individual cPcdh isoforms is indicated by the blue coloured boxes and the expression level is indicated by the colour gradient. Note that the majority of neurons express multiple cPcdh isoforms from three Pcdh clusters (a, b, and g) and the C-type Pcdh isoforms (ac2 and gc5), similar to the neurons in the clone dataset.

Extended Data Fig. 5 Confirmation of layer identities for individual neocortical excitatory neurons in the clone dataset.

a, Uniform manifold approximation and projection (UMAP) plot for alignment of individual neurons in our clone dataset (L2/3, n = 95; L4, n = 26; L5, n = 41; L6, n = 26) to the previously published neocortical excitatory neuron scRNA-seq dataset from the Allen Institute57 (n = 9,100). Large dots with black outlines represent individual clonally related excitatory neuron in our dataset and small dots with no outline represent individual excitatory neurons in the reference Allen Institute dataset. Different colours reflect different subtypes of neocortical excitatory neurons. The n numbers indicate neurons. b, Sankey plot for the result of transferring subtype labels from the reference dataset (predicted) to our clone dataset (raw) based on scRNA-seq. Different colours reflect different subtypes of neocortical excitatory neurons. The mapped cell numbers are shown in the graph inset. c, No significant difference in the percentage of cells mapped to the corresponding reference neuron subtypes/layers between our dataset and a previously published dataset using a similar method45. The mapped cell numbers are shown in the bar graph. Two-tailed paired Student’s t-test was used for statistical analysis.

Extended Data Fig. 6 PCDHγ removal causes a lateral clustering of sister and cousin excitatory neurons in the neocortex.

a, b, Representative 3D reconstruction images of P21 WT (left) and Pcdhg cKO (right) symmetric (a) and asymmetric (b) excitatory neuron clones labelled by MADM. Coloured lines indicate the layer boundaries and coloured dots represent the cell bodies of labelled neurons. The x-/y-/z-axes indicate the spatial orientation of the clone with the x-axis parallel to the brain pial surface and the y-axis perpendicular to the pial surface. Scale bars: 100 μm. cf, Quantification of the pairwise (c, d) and maximal (e, f) lateral and radial distances between sister neurons in individual WT and Pcdhg cKO clones (WT, n = 90 clones; Pcdhg cKO, n = 90 clones). gj, Quantification of the pairwise (g, h) and maximal (i, j) lateral and radial distances between cousin neurons in individual WT and Pcdhg cKO clones (WT, n = 68 clones; Pcdhg cKO, n = 74 clones). Data are representative of four independent experiments. Two-tailed unpaired Student’s t-test was used for statistical analysis. Box plots as in Fig. 1.

Source data

Extended Data Fig. 7 PCDHγ removal does not affect the overall layer formation, the excitatory neuron density, or the dendritic morphology.

a, Quantification of the number of neurons in individual WT and Pcdhg cKO clones (WT, n = 158 clones; Pcdhg cKO, n = 164 clones). b, Representative confocal images of P35 WT (left) and Pcdhg cKO (Emx1-Cre;Pcdhgfcon3/fcon3) (right) neocortices stained for CUX1 (red), a superficial layer excitatory neuron marker, and CTIP2 (green), a deep layer excitatory neuron marker, and counter-stained with DAPI (blue). Note no change in either layer formation or neuronal density in the Pcdhg cKO neocortex compared with the WT control. Scale bars: 100 μm (left) and 20 μm (right). c, d, Quantification of the numbers of CUX1+ (c) and CTIP2+ (d) cells in the 10,000 μm2 rectangle area (WT, n = 4 brains; Pcdhg cKO, n = 3 brains). e, Representative reconstructed dendritic morphologies of WT and Pcdhg cKO excitatory neurons in different layers. Scale bar, 50 μm. fi, Quantification of the neurite length (f), branch number (g), planar angle (h), or local angle (i) of WT and Pcdhg cKO neurons in different layers (L2/3: WT, n = 71; Pcdhg cKO, n = 55; L4: WT, n = 16; Pcdhg cKO, n = 24; L5: WT, n = 33; Pcdhg cKO, n = 46; L6: WT, n = 23; Pcdhg cKO, n = 40). The n numbers indicate neurons. j, Confocal images of representative E16 Doublecortin (Dcx) promoter driven eGFP expressing (green) cortices electroporated at E14 and stained for DCX (red) and DAPI (blue). The arrow points to the example cell. Note that eGFP-expressing cells are mostly located in the intermediate zone and positive for DCX. Scale bars: 100 µm (left) and 5 µm (right). k, Schematic diagram of in utero intraventricular injection of low-titre retroviruses containing Dcx promoter driven eGFP (green, Ctrl) and Cre/tdTomato (red, Pcdhg cKO) into the Pcdhgfcon3 mice at E12. l, Representative 3D reconstruction images of the control and Pcdhg cKO clones. Note that the Pcdhg cKO clone is more laterally clustered than the control clone. Scale bar, 100 μm. m, Quantification of the number of neurons in individual control and Pcdhg cKO clones (Ctrl, n = 67 clones from ~305 brain slices/5 brains; Pcdhg cKO, n = 78 clones from ~315 brain slices/5 brains). nq, Quantification of the pairwise (n, o) and maximal (p, q) lateral and radial distances between neurons in individual control and Pcdhg cKO clones (Ctrl, n = 67 clones; Pcdhg cKO, n = 78 clones). Data are representative of four (a), three (bd, jq), or ten (ei) independent experiments. Two-tailed unpaired Student’s t-test was used for statistical test. Data are presented as mean ± SEM (c, d). Box plots as in Fig. 1.

Source data

Extended Data Fig. 8 PCDHγ removal causes a lateral clustering of excitatory neuron clones during embryonic and neonatal neocortical development.

a, Representative 3D reconstruction images of E15 WT (left) and Pcdhg cKO (right) clones labelled by MADM at E12. Coloured lines indicate the layer boundaries and coloured dots represent the cell bodies of labelled cells. The x-/y-/z-axes indicate the spatial orientation of the clone with the x-axis parallel to the brain pial surface and the y-axis perpendicular to the pial surface. Similar symbols and displays are used in subsequent panels. Scale bar, 100 μm. be, Quantification of the pairwise (b, c) and maximal (d, e) lateral and radial distances between cells in individual E15 WT and Pcdhg cKO clones (WT, n = 54 clones from ~160 brain slices/4 brains; Pcdhg cKO, n = 62 clones from ~150 brain slices/4 brains). f, Representative 3D reconstruction images of E18 WT (left) and Pcdhg cKO (right) clones labelled by MADM at E12. Scale bar, 100 μm. gj, Quantification of the pairwise (g, h) and maximal (i, j) lateral and radial distances between cells in individual E18 WT and Pcdhg cKO clones (WT, n = 60 clones from ~140 brain slices/4 brains; Pcdhg cKO, n = 76 clones from ~135 brain slices/4 brains). k, l, Representative 3D reconstruction images of P7 WT (left) and Pcdhg cKO (right) symmetric (k) and asymmetric (l) clones labelled by MADM at E12. Coloured lines indicate the layer boundaries and coloured dots represent the cell bodies of labelled neurons. Note that the Pcdhg cKO clones are more laterally clustered than the WT clones. Scale bars, 100 μm. mp, Quantification of the pairwise (m, n) and maximal (o, p) lateral and radial distances between neurons in individual P7 WT and Pcdhg cKO clones (WT, n = 66 clones from ~204 brain slices/4 brains; Pcdhg cKO, n = 71 clones from ~215 brain slices/4 brains). Data are representative of three independent experiments. Two-tailed unpaired Student’s t-test was used for statistical test. Box plots as in Fig. 1.

Source data

Extended Data Fig. 9 PCDHγC3 overexpression leads to a lateral dispersion of sister and cousin excitatory neurons.

a, b, Representative 3D reconstruction images of P21 WT (left) and Pcdhgc3 OE (right) symmetric (a) and asymmetric (b) excitatory neuron clones labelled by MADM. Coloured lines indicate the layer boundaries and coloured dots represent the cell bodies of labelled neurons. The x-/y-/z-axes indicate the spatial orientation of the clone with the x-axis parallel to the brain pial surface and the y-axis perpendicular to the pial surface. Scale bars: 100 μm. cf, Quantification of the pairwise (c, d) and maximal (e, f) lateral and radial distances between sister neurons in individual WT and Pcdhgc3 overexpressing clones (WT, n = 99 clones; Pcdhgc3 OE, n = 68 clones). gj, Quantification of the pairwise (g, h) and maximal (i, j) lateral and radial distances between cousin neurons in individual WT and Pcdhgc3 OE clones (WT, n = 79 clones; Pcdhgc3 OE, n = 86 clones). Data are representative of four independent experiments. Two-tailed unpaired Student’s t-test was used for statistical test. Box plots as in Fig. 1.

Source data

Extended Data Fig. 10 PCDHγC3 overexpression does not affect the number of clonally related excitatory neurons or the dendritic morphology.

a, Quantification of the number of neurons in individual WT and Pcdhgc3 OE clones in the neocortex (WT, n = 178 clones; Pcdhgc3 OE, n = 154 clones). b, Representative reconstructed dendritic morphologies of WT and Pcdhgc3 OE excitatory neurons in different layers. Scale bar, 50 μm. cf, Quantification of the neurite length (c), branch number (d), planar angle (e), or local angle (f) of WT and Pcdhgc3 OE neurons in different layers (L2/3: WT, n = 58; Pcdhgc3 OE, n = 55; L4: WT, n = 17; Pcdhgc3 OE, n = 29; L5: WT, n = 34; Pcdhgc3 OE, n = 30; L6: WT, n = 23; Pcdhgc3 OE, n = 31). The n numbers indicate neurons. g, Schematic diagram of in utero intraventricular injection of low-titre retroviruses with Doublecortin (Dcx) promoter driven eGFP (green, Ctrl) and Cre/tdTomato (red, Pcdhgc3 OE) into the Pcdhgc3/c3 mice at E12. h, Representative 3D reconstruction images of the control and Pcdhgc3 OE clones. Note that the Pcdhgc3 OE clones are more laterally dispersed than the control clones. Scale bar, 100 μm. i, Quantification of the number of neurons in individual control and Pcdhgc3 OE clones (Ctrl, n = 62 clones from ~255 brain slices/4 brains; Pcdhgc3 OE, n = 66 clones from ~260 brain slices/4 brains). jm, Quantification of the pairwise (j, k) and maximal (l, m) lateral and radial distances between neurons in individual control and Pcdhgc3 OE clones (Ctrl, n = 62 clones; Pcdhgc3 OE, n = 66 clones). Data are representative of four (a), ten (bf), or three (gm) independent experiments. Two-tailed unpaired Student’s t-test was used for statistical test. Box plots as in Fig. 1.

Source data

Extended Data Fig. 11 PCDHγC3 overexpression leads to a lateral dispersion during early neocortical development.

a, Representative 3D reconstruction images of E15 WT (left) and Pcdhgc3 OE (right) clones labelled by MADM at E12. Coloured lines indicate the layer boundaries and coloured dots represent the cell bodies of labelled cells. The x-/y-/z-axes indicate the spatial orientation of the clone with the x-axis parallel to the brain pial surface and the y-axis perpendicular to the pial surface. Similar symbols and displays are used in subsequent panels. Scale bar, 100 μm. be, Quantification of the pairwise (b, c) and maximal (d, e) lateral and radial distances between cells in individual E15 WT and Pcdhgc3 OE clones (WT, n = 60 clones from ~180 brain slices/5 brains; Pcdhgc3 OE, n = 67 clones from ~160 brain slices/4 brains). f, Representative 3D reconstruction images of E18 WT (left) and Pcdhgc3 OE (right) clones labelled by MADM at E12. Scale bar, 100 μm. gj, Quantification of the pairwise (g, h) and maximal (i, j) lateral and radial distances between cells in individual E18 WT and Pcdhgc3 OE clones (WT, n = 53 clones from ~106 brain slices/3 brains; Pcdhgc3 OE, n = 39 clones from ~100 brain slices/3 brains). k, l, Representative 3D reconstruction images of P7 WT (left) and Pcdhgc3 OE (right) symmetric (k) and asymmetric (l) clones labelled by MADM at E12. Coloured lines indicate the layer boundaries and coloured dots represent the cell bodies of labelled neurons. Note that the Pcdhgc3 OE clones are more laterally dispersed than the WT clones. Scale bars, 100 μm. mp, Quantification of the pairwise (m, n) and maximal (o, p) lateral and radial distances between neurons in individual P7 WT and Pcdhgc3 OE clones (WT, n = 62 clones from ~160 brain slices/3 brains; Pcdhgc3 OE, n = 61 clones from ~156 brain slices/3 brains). Data are representative of three independent experiments. Two-tailed unpaired Student’s t-test was used for statistical test. Box plots as in Fig. 1.

Source data

Extended Data Fig. 12 PCDHγ removal does not affect the basic membrane properties of neocortical excitatory neurons.

a, Quantification of the inter-soma distance between clonally related WT and Pcdhg cKO excitatory neurons and nearby non-clonally related excitatory neurons (clonally related: WT, n = 415; Pcdhg cKO, n = 330; non-clonally related: WT, n = 157; Pcdhg cKO, n = 141). b, Summary of the rate of chemical synaptic connections between the clonally related WT and Pcdhg cKO excitatory neurons and nearby non-clonally related excitatory neurons at P14–20 (WT clone & non-clone, n = 337 from ~53 brain slices/15 brains; Pcdhg cKO clone & non-clone, n = 318 from ~52 brain slices/17 brains) and P21–35 (WT clone & non-clone, n = 235 from ~46 brain slices/16 brains; Pcdhg cKO clone & non-clone, n = 225 from ~40 brain slices/12 brains). The specific numbers of recorded pairs are shown in the bar graphs. c, Representative sample traces of the responses of excitatory neurons to somatic current injections in the WT (left) and Pcdhg cKO (right) neocortices. Scale bars: 50 mV and 200 msec. df, Summary of the resting membrane potential (RMP) (d, WT, n = 45; Pcdhg cKO, n = 48), input resistance (e, WT, n = 44; Pcdhg cKO, n = 48), and maximal firing rate (f, WT, n = 44; Pcdhg cKO, n = 47) of WT and Pcdhg cKO excitatory neurons. g, Quantification of the inter-soma distance between WT and Pcdhg cKO sister (left) or cousin (right) excitatory neurons (sister: WT, n = 216; Pcdhg cKO, n = 177; cousin: WT, n = 199; Pcdhg cKO, n = 153). h, i, Summary of the rate of synaptic connections between WT and Pcdhg cKO sister or cousin neuronal pairs with regard to the angular orientation of their cell bodies relative to the pia. Each symbol represents a neuronal pair. The numbers of recorded pairs and the rates of synaptic connections are shown in the graphs. The n numbers indicate neuron pairs (a, b, gi) or neurons (df). Data are representative of at least 20–30 independent experiments. Two-tailed unpaired Student’s t-test (a, dg); Two-tailed χ2 test (b, h, i). Box plots as in Fig. 1.

Source data

Extended Data Fig. 13 PCDHγC3 overexpression does not affect the basic membrane properties of neocortical excitatory neurons.

a, Quantification of the inter-soma distance between clonally related WT and Pcdhgc3 OE excitatory neurons and nearby non-clonally related excitatory neurons (clonally related: WT, n = 318; Pcdhgc3 OE, n = 324; non-clonally related: WT, n = 128; Pcdhgc3 OE, n = 123). b, Summary of the frequency of chemical synaptic connections between the clonally related WT and Pcdhgc3 OE excitatory neurons and nearby non-clonal excitatory neurons in P14–20 (WT clone & non-clone, n = 242 from ~44 brain slices/13 brains; Pcdhgc3 OE clone & non-clone, n = 273 from ~53 brain slices/18 brains) and P21–35 (WT clone & non-clone, n = 204 from ~31 brain slices/11 brains; Pcdhgc3 OE clone & non-clone, n = 204 from ~32 brain slices/11 brains). The specific numbers of recorded pairs are shown in the bar graphs. c, Representative sample traces of the responses of excitatory neurons to somatic current injections in the WT (left) and Pcdhgc3 OE (right) neocortices. Scale bars: 50 mV and 200 msec. df, Summary of the resting membrane potential (RMP) (d, WT, n = 45; Pcdhgc3 OE, n = 54), input resistance (e, WT, n = 44; Pcdhgc3 OE, n = 62), and maximal firing rate (f, WT, n = 42; Pcdhgc3 OE, n = 62) of WT and Pcdhgc3 OE excitatory neurons. g, Quantification of the inter-soma distance between WT and Pcdhgc3 OE sister (left) or cousin (right) excitatory neurons (sister: WT, n = 167; Pcdhgc3 OE, n = 215; cousin: WT, n = 151; Pcdhgc3 OE, n = 109). h, i, Summary of the rate of synaptic connections between WT and Pcdhgc3 OE sister or cousin neuronal pairs with regard to the angular orientation of their cell bodies relative to the pia. Each symbol represents a neuronal pair. The numbers of recorded pairs and the rates of synaptic connections are shown in the graphs. The n numbers indicate neuron pairs (a, b, g-i) or neurons (df). Data are representative of at least 20–30 independent experiments. Two-tailed unpaired Student’s t-test (a, dg); Two-tailed χ2 test (b, h, i). Box plots as in Fig. 1.

Source data

Supplementary information

Reporting Summary

Peer Review File

Supplementary Table 1

A summary of statistical tests (P values) for Extended Data Figs. 12h,i and 13h,i.

Supplementary Table 2

Mouse information used in individual experiments.

Supplementary Table 3

A summary of the numbers of brains, brain slices/sections, clones or neurons in individual experiments.

Supplementary Table 4

The scRNA-seq matrix of cPcdh isoform expression in individual clonally related neocortical excitatory neurons.

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lv, X., Li, S., Li, J. et al. Patterned cPCDH expression regulates the fine organization of the neocortex. Nature 612, 503–511 (2022). https://doi.org/10.1038/s41586-022-05495-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-05495-2

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