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.

  • Resource
  • Published:

Interneuron origin and molecular diversity in the human fetal brain

Abstract

Precise generation of excitatory neurons and inhibitory interneurons is crucial for proper formation and function of neural circuits in the mammalian brain. Because of the size and complexity of the human brain, it is a challenge to reveal the rich diversity of interneurons. To decipher origin and diversity of interneurons in the human fetal subpallium, here we show molecular features of diverse subtypes of interneuron progenitors and precursors by conducting single-cell RNA sequencing and in situ sequencing. Interneuron precursors in the medial and lateral ganglionic eminence simultaneously procure temporal and spatial identity through expressing a combination of specific sets of RNA transcripts. Acquisition of various interneuron subtypes in adult human brains occurs even at fetal stages. Our study uncovers complex molecular signatures of interneuron progenitors and precursors in the human fetal subpallium and highlights the logic and programs in the origin and lineage specification of various interneurons.

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: Molecular diversity of the human fetal subpallium.
Fig. 2: Characterization of heterogeneity of interneuron progenitors in the human fetal subpallium.
Fig. 3: Expression patterns of subpallial genes in the GE of the human fetal brain as detected by ISS.
Fig. 4: Specification of MGE-derived interneuron precursors.
Fig. 5: Two lineages of LGE-derived interneuron precursors.
Fig. 6: Heterogeneity of CGE-derived interneuron precursors.
Fig. 7: Early acquisition of adult interneuron diversity in the human fetal subpallium.

Similar content being viewed by others

Data availability

The raw single-cell sequencing data are available in the Gene Expression Omnibus at https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE165388, with the accession code GSE165388. All images of ISS are available upon request at https://github.com/yuanbell/Single-cell-sequencing-of-subpallium.

Code availability

All codes generated during this study are available upon request at https://github.com/yuanbell/Single-cell-sequencing-of-subpallium.

References

  1. Tremblay, R., Lee, S. & Rudy, B. GABAergic interneurons in the neocortex: from cellular properties to circuits. Neuron 91, 260–292 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Lim, L., Mi, D., Llorca, A. & Marin, O. Development and functional diversification of cortical interneurons. Neuron 100, 294–313 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Marín, O. Interneuron dysfunction in psychiatric disorders. Nat. Rev. Neurosci. 13, 107–120 (2012).

    Article  PubMed  Google Scholar 

  4. Pauly, M. C., Dobrossy, M. D., Nikkhah, G., Winkler, C. & Piroth, T. Organization of the human fetal subpallium. Front. Neuroanat. 7, 54 (2013).

    PubMed  Google Scholar 

  5. Silberberg, S. N. et al. Subpallial enhancer transgenic lines: a data and tool resource to study transcriptional regulation of GABAergic cell fate. Neuron 92, 59–74 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Nery, S., Fishell, G. & Corbin, J. G. The caudal ganglionic eminence is a source of distinct cortical and subcortical cell populations. Nat. Neurosci. 5, 1279–1287 (2002).

    Article  CAS  PubMed  Google Scholar 

  7. Hu, J. S. et al. Coup-TF1 and Coup-TF2 control subtype and laminar identity of MGE-derived neocortical interneurons. Development 144, 2837–2851 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Bandler, R. C., Mayer, C. & Fishell, G. Cortical interneuron specification: the juncture of genes, time and geometry. Curr. Opin. Neurobiol. 42, 17–24 (2017).

    Article  CAS  PubMed  Google Scholar 

  9. Xu, Q., Tam, M. & Anderson, S. A. Fate mapping Nkx2.1-lineage cells in the mouse telencephalon. J. Comp. Neurol. 506, 16–29 (2008).

    Article  CAS  PubMed  Google Scholar 

  10. Su-Feher, L. et al. Single cell enhancer activity maps neuronal lineages in embryonic mouse basal ganglia. Preprint at https://www.biorxiv.org/content/10.1101/2021.01.11.426285v1.full (2021).

  11. Flames, N. et al. Delineation of multiple subpallial progenitor domains by the combinatorial expression of transcriptional codes. J. Neurosci. 27, 9682–9695 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Hu, J. S., Vogt, D., Sandberg, M. & Rubenstein, J. L. Cortical interneuron development: a tale of time and space. Development 144, 3867–3878 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Tosches, M. A. et al. Evolution of pallium, hippocampus, and cortical cell types revealed by single-cell transcriptomics in reptiles. Science 360, 881–888 (2018).

    Article  CAS  PubMed  Google Scholar 

  14. Krienen, F. M. et al. Innovations present in the primate interneuron repertoire. Nature 586, 262–269 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ma, T. et al. Subcortical origins of human and monkey neocortical interneurons. Nat. Neurosci. 16, 1588–1597 (2013).

    Article  CAS  PubMed  Google Scholar 

  16. Hansen, D. V. et al. Non-epithelial stem cells and cortical interneuron production in the human ganglionic eminences. Nat. Neurosci. 16, 1576–1587 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Boldog, E. et al. Transcriptomic and morphophysiological evidence for a specialized human cortical GABAergic cell type. Nat. Neurosci. 21, 1185–1195 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Close, J. L. et al. Single-cell profiling of an in vitro model of human interneuron development reveals temporal dynamics of cell type production and maturation. Neuron 93, 1035–1048 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sun, T. & Hevner, R. F. Growth and folding of the mammalian cerebral cortex: from molecules to malformations. Nat. Rev. Neurosci. 15, 217–232 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Fan, X. et al. Spatial transcriptomic survey of human embryonic cerebral cortex by single-cell RNA-seq analysis. Cell Res. 28, 730–745 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhong, S. et al. A single-cell RNA-seq survey of the developmental landscape of the human prefrontal cortex. Nature 555, 524–528 (2018).

    Article  CAS  PubMed  Google Scholar 

  22. Zeng, Z., Miao, N. & Sun, T. Revealing cellular and molecular complexity of the central nervous system using single cell sequencing. Stem Cell Res. Ther. 9, 234 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. O’Rahilly, R. & Müller, F. The Embryonic Human Brain: An Atlas of Developmental Stages, Third Edition, 1–358 (Wiley, 2005).

  24. Clancy, B., Darlington, R. B. & Finlay, B. L. Translating developmental time across mammalian species. Neuroscience 105, 7–17 (2001).

    Article  CAS  PubMed  Google Scholar 

  25. Otis, E. M. & Brent, R. Equivalent ages in mouse and human embryos. Anat. Rec. 120, 33–63 (1954).

    Article  CAS  PubMed  Google Scholar 

  26. Nowakowski, T. J. et al. Spatiotemporal gene expression trajectories reveal developmental hierarchies of the human cortex. Science 358, 1318 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Mayer, C. et al. Developmental diversification of cortical inhibitory interneurons. Nature 555, 457–462 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Mi, D. et al. Early emergence of cortical interneuron diversity in the mouse embryo. Science 360, 81–85 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Harkin, L. F. et al. Distinct expression patterns for type II topoisomerases IIA and IIB in the early foetal human telencephalon. J. Anat. 228, 452–463 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Bansod, S., Kageyama, R. & Ohtsuka, T. Hes5 regulates the transition timing of neurogenesis and gliogenesis in mammalian neocortical development. Development 144, 3156–3167 (2017).

    Article  CAS  PubMed  Google Scholar 

  31. Marques, S. et al. Oligodendrocyte heterogeneity in the mouse juvenile and adult central nervous system. Science 352, 1326–1329 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhao, Q. et al. Single-cell transcriptome analyses reveal endothelial cell heterogeneity in tumors and changes following antiangiogenic treatment. Cancer Res. 78, 2370–2382 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Petros, T. J., Bultje, R. S., Ross, M. E., Fishell, G. & Anderson, S. A. Apical versus basal neurogenesis directs cortical interneuron subclass fate. Cell Rep. 13, 1090–1095 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lindtner, S. et al. Genomic resolution of DLX-orchestrated transcriptional circuits driving development of forebrain GABAergic neurons. Cell Rep. 28, 2048–2063 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ke, R. et al. In situ sequencing for RNA analysis in preserved tissue and cells. Nat. Methods 10, 857–860 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Wang, X. et al. Three-dimensional intact-tissue sequencing of single-cell transcriptional states. Science 361, eaat5691 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Alzu’bi, A. et al. The transcription factors COUP-TFI and COUP-TFII have distinct roles in arealisation and GABAergic interneuron specification in the early human fetal telencephalon. Cereb. Cortex 27, 4971–4987 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Butt, S. J. et al. The temporal and spatial origins of cortical interneurons predict their physiological subtype. Neuron 48, 591–604 (2005).

    Article  CAS  PubMed  Google Scholar 

  39. Kanatani, S., Yozu, M., Tabata, H. & Nakajima, K. COUP-TFII is preferentially expressed in the caudal ganglionic eminence and is involved in the caudal migratory stream. J. Neurosci. 28, 13582–13591 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Rudy, B., Fishell, G., Lee, S. & Hjerling-Leffler, J. Three groups of interneurons account for nearly 100% of neocortical GABAergic neurons. Dev. Neurobiol. 71, 45–61 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Kepecs, A. & Fishell, G. Interneuron cell types are fit to function. Nature 505, 318–326 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Turrero Garcia, M. & Harwell, C. C. Radial glia in the ventral telencephalon. FEBS Lett. 591, 3942–3959 (2017).

    Article  CAS  PubMed  Google Scholar 

  43. Silva, T. P. et al. Transcriptome profiling of human pluripotent stem cell-derived cerebellar organoids reveals faster commitment under dynamic conditions. Biotechnol. Bioeng. 118, 2781–2803 (2021).

    Article  CAS  PubMed  Google Scholar 

  44. Enterría-Morales, D. et al. Molecular targets for endogenous glial cell line-derived neurotrophic factor modulation in striatal parvalbumin interneurons. Brain Commun. 2, fcaa105 (2020).

  45. Flandin, P., Kimura, S. & Rubenstein, J. L. The progenitor zone of the ventral medial ganglionic eminence requires Nkx2-1 to generate most of the globus pallidus but few neocortical interneurons. J. Neurosci. 30, 2812–2823 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Tiveron, M. C. et al. Zic-proteins are repressors of dopaminergic forebrain fate in mice and C. elegans. J. Neurosci. 37, 10611–10623 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sousa, V. H., Miyoshi, G., Hjerling-Leffler, J., Karayannis, T. & Fishell, G. Characterization of Nkx6-2-derived neocortical interneuron lineages. Cereb. Cortex 19 i1–i10 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Lu, K. M., Evans, S. M., Hirano, S. & Liu, F. C. Dual role for Islet-1 in promoting striatonigral and repressing striatopallidal genetic programs to specify striatonigral cell identity. Proc. Natl Acad. Sci. USA 111, E168–E177 (2014).

    CAS  PubMed  Google Scholar 

  49. Li, J. et al. Transcription factors Sp8 and Sp9 coordinately regulate olfactory bulb interneuron development. Cereb. Cortex 28, 3278–3294 (2018).

    Article  PubMed  Google Scholar 

  50. O’Leary, D. D. & Nakagawa, Y. Patterning centers, regulatory genes and extrinsic mechanisms controlling arealization of the neocortex. Curr. Opin. Neurobiol. 12, 14–25 (2002).

    Article  PubMed  Google Scholar 

  51. Xu, Q. et al. Sonic hedgehog signaling confers ventral telencephalic progenitors with distinct cortical interneuron fates. Neuron 65, 328–340 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ma, T. et al. A subpopulation of dorsal lateral/caudal ganglionic eminence-derived neocortical interneurons expresses the transcription factor Sp8. Cereb. Cortex 22, 2120–2130 (2012).

    Article  PubMed  Google Scholar 

  53. Torigoe, M., Yamauchi, K., Kimura, T., Uemura, Y. & Murakami, F. Evidence that the laminar fate of LGE/CGE-derived neocortical interneurons is dependent on their progenitor domains. J. Neurosci. 36, 2044–2056 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Rubin, A. N. et al. The germinal zones of the basal ganglia but not the septum generate GABAergic interneurons for the cortex. J. Neurosci. 30, 12050–12062 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Hern, W. M. Correlation of fetal age and measurements between 10 and 26 weeks of gestation. Obstet. Gynecol. 63, 26–32 (1984).

    CAS  PubMed  Google Scholar 

  56. Gayoso, A., Shor, J., Carr, A. J., Sharma, R. & Pe’er, D. JonathanShor/DoubletDetection: HOTFIX: correct setup.py installation. Zenodo (2019). https://doi.org/10.5281/zenodo.3376859

  57. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Pedregosa, F. et al. Scikit-learn: machine learning in Python. J. Mach. Learn. Res. 12, 2825–2830 (2011).

    Google Scholar 

  60. Wolf, F. A. et al. PAGA: graph abstraction reconciles clustering with trajectory inference through a topology preserving map of single cells. Genome Biol. 20, 59 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Angerer, P. et al. destiny: diffusion maps for large-scale single-cell data in R. Bioinformatics 32, 1241–1243 (2016).

    Article  CAS  PubMed  Google Scholar 

  64. Street, K. et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genomics 19, 477 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Stevant, I. et al. Dissecting cell lineage specification and sex fate determination in gonadal somatic cells using single-cell transcriptomics. Cell Rep. 26, 3272–3283 (2019).

    Article  CAS  PubMed  Google Scholar 

  66. Yu, G., Wang, L. G., Han, Y. & He, Q. Y. clusterProfiler: an R package for comparing biological themes among gene clusters. Omics 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Korotkevich, G., Sukhov, V. & Sergushichev, A. Fast gene set enrichment analysis. Preprint at https://www.biorxiv.org/content/10.1101/060012v3 (2021).

Download references

Acknowledgements

This study was supported by the Scientific Research Funds of Huaqiao University (Z16Y0017 to T.S.), the Fundamental Research Funds for the Central Universities (ZQN-715 to Y.S.), the Innovation Awards of Quanzhou Talents (2018C057R to Y.C.), the Natural Science Foundation of Fujian Province of China (2018J01585 to S.H.), the National Key Research and Development Program of China (2017YFA0106800 to R.K.) and the National Natural Science Foundation of China (31771141 to T.S., 11802096 to Y.S. and 31770927 to R.K.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Y.Y. and T.S. conceived and designed the study. Y.Y. performed the single-cell sequencing experiments. Y.Y., Z.Z., R.C. and S.H. analyzed sequencing data. Y.Y., D.X. and R.K. performed the ISS experiment. Y.S., W.J.C., W.H.C. and W.L. prepared brain tissues. Y.Y. and T.S. wrote the paper. T.S. supervised the entire study.

Corresponding author

Correspondence to Tao Sun.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Neuroscience thanks the anonymous reviewers for their contribution to 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

Extended Data Fig. 1 Single-cell transcriptomic maps of the human fetal subpallium.

a, Cell clustering of human fetal subpallial samples at gestational weeks (GW) 9 to 12 integrated using harmony and depicted using t-SNE. b, The distribution of each subclusters in the subpallium at four developing stages. Colors indicate cell clusters as shown in a. c, Clustering of all cells from four developing stages after batch correction visualized using t-SNE. d, Heatmap illustrating differentially expressed genes (DEGs) in five major clusters and 17 subclusters. e, Assessment of 71 genes from 2,922 DEGs using Random Forest Classifier from scikit-learn of the remaining 16 subclusters after removing excitatory lineages. ‘True label’ indicates the manual annotation based on the 71 genes.

Extended Data Fig. 2 Expression patterns of canonical genes from each cell types.

a-f, Representative genes expressed in cell clusters from human fetal subpallium visualized using t-SNE. Each dot represents one cell. Major clusters include excitatory lineages (a), neural progenitor cells (NPCs, b) and interneuron precursors (INPs, c) derived from the medial ganglionic eminence (MGE_INPs, d), caudal ganglionic eminence (CGE_INPs, e) and lateral ganglionic eminence (LGE_INPs, f).

Extended Data Fig. 3 Heterogeneity of interneuron progenitors in the human fetal subpallium.

a-c, Expression of DLX2 and GAD2 at gestational weeks (GW) 9 (a), GW11 (b), GW12 (c), visualized by the t-SNE plot. d, Violin plots of expression patterns of progenitor markers expressed at the ventricular zone (VZ) and subventricular zone (SVZ) in the subpallium at GW12. e, Violin plots of expression patterns of ganglionic eminence regional markers in the progenitors from GW9 to GW12.

Extended Data Fig. 4 Expression patterns of subpallial genes in the ganglionic eminence of the human fetal brain as detected by in situ sequencing (ISS).

a and b, Expression patterns of 6 subpallial genes in the medial ganglionic eminence (MGE) (a), lateral ganglionic eminence (LGE) (a) and caudal ganglionic eminence (CGE) (b) at gestational week 12 (GW12). c-e, Boxed areas in a and b are shown in high power views. Scale bars: 1 mm in a and b; 100 µm in c-e.

Extended Data Fig. 5 Expression patterns of genes in the subpallium of the human fetal brain as detected by in situ sequencing (ISS).

a-d, Expression patterns of genes expressed in the ventricular zone (VZ) and subventricular zone (SVZ) at gestational week 12 (GW12) are shown in green and purple pseudo-colors, respectively, in the medial ganglionic eminence (MGE), lateral ganglionic eminence (LGE) and caudal ganglionic eminence (CGE). Scale bars: 1 mm.

Extended Data Fig. 6 Expression patterns of genes expressed in the ventricular zone (VZ) in the subpallium of the human fetal brain as detected by in situ sequencing (ISS).

a, Merged expression of CLU, LIX1, PTN and SPARC in the medial ganglionic eminence (MGE), lateral ganglionic eminence (LGE) and caudal ganglionic eminence (CGE) at gestational week 12 (GW12). b-e, Expression of CLU (b), LIX1 (c), PTN (d) and SPARC (e) in the MGE, LGE and CGE. Scale bars: 100 µm.

Extended Data Fig. 7 Specification of medial ganglionic eminence (MGE)-derived interneuron precursors.

a, Clustering of MGE-derived interneuron precursors from GW9 to GW12 visualized by t-SNE embedding. Cluster 5, 1, 2, 4 and 7 were similar to ANGPT2+/CRABP1+, ZEB2+/MAF+, POU3F2+/CNTNAP2+, NR2F1+/MEIS2+ and LHX8+/NKX2-1+, respectively, in the Fig. 4a, cluster 3, 6, 8 were raised from the GW12. b, Heatmap of differentially expressed genes from subclusters in a. c, Diffusion map of the most variable genes and reconstruction of the cell lineages. Dots represent cells, and black lines represent predicted cell lineage-1 and -2. As examples, the cluster 6 was predicted to the lineage 1 and the cluster 3 to the lineage 2. d. Expression patterns of genes highly expressed in the dorsal (MEIS2 and NR2F1) and ventral (LHX8 and ZIC4) MGE in human fetal brains at gestational week 12 (GW12) as detected using in situ sequencing (ISS). e, Box plots representing relative numbers of positive dots of MGE marker genes in boxed areas in the dorsal and ventral MGE in d. Box: 25–75th percentiles, whiskers: 10–90th percentiles, horizontal line in box: median. Scale bars: 1 mm.

Extended Data Fig. 8 Expression patterns of medial ganglionic eminence (MGE) specific genes of the human fetal brain as detected by in situ sequencing (ISS).

a, Coronal sections labeled with DAPI to illustrate cell nucleoli of a human fetal brain at gestational week 12 (GW12). Boxed areas highlight the MGE. The dorsal (D), ventral (V), medial (M) and lateral (L) orientations of the sections are labeled. b, High expression of CNTNAP2 and MAF at the root of lineage-1 and -2 based on the diffusion map in lineage analyses. c-e, Expression patterns of 10 MGE-precursor marker genes in coronal sections of human fetal brains. The dorsal and ventral MGEs are labeled as dMGE (c) and vMGE (d). Scale bars: 1 mm.

Extended Data Fig. 9 Lateral ganglionic eminence (LGE)-derived interneuron precursors.

a and b, Expression patterns of 8 LGE specific genes in coronal sections of the human fetal brain at gestational week 12 (GW12) as detected by in situ sequencing (ISS). The ventral (green box) and dorsal (blue box) LGE are shown as vLGE and dLGE. The boxed areas also are shown in high power views. Scale bars: 1 mm and 100 µm.

Extended Data Fig. 10 Caudal ganglionic eminence (CGE)-derived interneuron precursors.

a, The transcription network regulated by CGE-specific transcription factors in interneuron precursors. b, Gene ontology (GO) enrichment analysis of target genes for CGE-specific transcription factors. c, A coronal section labeled with DAPI to illustrate cell nucleoli of one human fetal brain at gestational week 12 (GW12). d, Expression patterns of CALB2, NPAS3, ST18 and SP9 in the CGE in coronal sections of the human fetal brain at GW12, as detected by in situ sequencing (ISS). Scale bar: 1 mm.

Supplementary information

Supplementary Information

Supplementary Figs. 1–5

Reporting Summary

Supplementary Tables

Supplementary Tables 1–18

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yu, Y., Zeng, Z., Xie, D. et al. Interneuron origin and molecular diversity in the human fetal brain. Nat Neurosci 24, 1745–1756 (2021). https://doi.org/10.1038/s41593-021-00940-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-021-00940-3

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