Abstract
Immature dentate granule cells (imGCs) arising from adult hippocampal neurogenesis contribute to plasticity, learning and memory, but their evolutionary changes across species and specialized features in humans remain poorly understood. Here we performed machine-learning-augmented analysis of published single-nucleus RNA-sequencing datasets and identified macaque imGCs with transcriptome-wide immature neuronal characteristics. Our cross-species comparisons among humans, monkeys, pigs and mice showed few shared (such as DPYSL5), but mostly species-specific gene expression in imGCs that converged onto common biological processes regulating neuronal development. We further identified human-specific transcriptomic features of imGCs and demonstrated the functional roles of human imGC-enriched expression of a family of proton-transporting vacuolar-type ATPase subtypes in the development of imGCs derived from human pluripotent stem cells. Our study reveals divergent gene expression patterns but convergent biological processes in the molecular characteristics of imGCs across species, highlighting the importance of conducting independent molecular and functional analyses for adult neurogenesis in different species.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout





Similar content being viewed by others
Data availability
All scRNA-seq or single-nucleus RNA-seq data were previously published (summarized in Supplementary Table 1). Additional information required for reanalyzing the data presented in this study can be obtained from the corresponding authors upon request. Source data are provided with this paper.
Code availability
Scripts for bioinformatic analyses used in this study are available at https://github.com/zhoujoeyyi/imGC_species/.
References
Gage, F. H. Adult neurogenesis in mammals. Science 364, 827–828 (2019).
Ming, G. L. & Song, H. Adult neurogenesis in the mammalian brain: significant answers and significant questions. Neuron 70, 687–702 (2011).
Denoth-Lippuner, A. & Jessberger, S. Formation and integration of new neurons in the adult hippocampus. Nat. Rev. Neurosci. 22, 223–236 (2021).
Salta, E. et al. Adult hippocampal neurogenesis in Alzheimer’s disease: a roadmap to clinical relevance. Cell Stem Cell 30, 120–136 (2023).
Terreros-Roncal, J. et al. Impact of neurodegenerative diseases on human adult hippocampal neurogenesis. Science 374, 1106–1113 (2021).
Miller, S. M. & Sahay, A. Functions of adult-born neurons in hippocampal memory interference and indexing. Nat. Neurosci. 22, 1565–1575 (2019).
Ge, S. et al. GABA regulates synaptic integration of newly generated neurons in the adult brain. Nature 439, 589–593 (2006).
Ge, S., Yang, C. H., Hsu, K. S., Ming, G. L. & Song, H. A critical period for enhanced synaptic plasticity in newly generated neurons of the adult brain. Neuron 54, 559–566 (2007).
Christian, K. M., Song, H. & Ming, G. L. Functions and dysfunctions of adult hippocampal neurogenesis. Annu. Rev. Neurosci. 37, 243–262 (2014).
Li, Y. D., Luo, Y. J. & Song, J. Optimizing memory performance and emotional states: multi-level enhancement of adult hippocampal neurogenesis. Curr. Opin. Neurobiol. 79, 102693 (2023).
Hochgerner, H., Zeisel, A., Lonnerberg, P. & Linnarsson, S. Conserved properties of dentate gyrus neurogenesis across postnatal development revealed by single-cell RNA sequencing. Nat. Neurosci. 21, 290–299 (2018).
Shin, J. et al. Single-cell RNA-Seq with waterfall reveals molecular cascades underlying adult neurogenesis. Cell Stem Cell 17, 360–372 (2015).
Lu, Z. et al. Tracking cell-type-specific temporal dynamics in human and mouse brains. Cell 186, 4345–4364 (2023).
Gao, Y. et al. Integrative single-cell transcriptomics reveals molecular networks defining neuronal maturation during postnatal neurogenesis. Cereb. Cortex 27, 2064–2077 (2017).
Artegiani, B. et al. A single-cell RNA sequencing study reveals cellular and molecular dynamics of the hippocampal neurogenic niche. Cell Rep. 21, 3271–3284 (2017).
Ratz, M. et al. Clonal relations in the mouse brain revealed by single-cell and spatial transcriptomics. Nat. Neurosci. 25, 285–294 (2022).
Habib, N. et al. Div-Seq: single-nucleus RNA-Seq reveals dynamics of rare adult newborn neurons. Science 353, 925–928 (2016).
Rasetto, N. B. et al. Transcriptional dynamics orchestrating the development and integration of neurons born in the adult hippocampus. Sci. Adv. 10, eadp6039 (2024).
Zhang, H. et al. Single-nucleus transcriptomic landscape of primate hippocampal aging. Protein Cell 12, 695–716 (2021).
Hao, Z. Z. et al. Single-cell transcriptomics of adult macaque hippocampus reveals neural precursor cell populations. Nat. Neurosci. 25, 805–817 (2022).
Wang, W. et al. Transcriptome dynamics of hippocampal neurogenesis in macaques across the lifespan and aged humans. Cell Res. 32, 729–743 (2022).
Franjic, D. et al. Transcriptomic taxonomy and neurogenic trajectories of adult human, macaque, and pig hippocampal and entorhinal cells. Neuron 110, 452–469 (2022).
Habib, N. et al. Massively parallel single-nucleus RNA-seq with DroNc-seq. Nat. Methods 14, 955–958 (2017).
Tosoni, G. et al. Mapping human adult hippocampal neurogenesis with single-cell transcriptomics: reconciling controversy or fueling the debate? Neuron https://doi.org/10.1016/j.neuron.2023.03.010 (2023).
Zhou, Y. et al. Molecular landscapes of human hippocampal immature neurons across lifespan. Nature 607, 527–533 (2022).
Ayhan, F. et al. Resolving cellular and molecular diversity along the hippocampal anterior-to-posterior axis in humans. Neuron 109, 2091–2105 (2021).
Zhong, S. et al. Decoding the development of the human hippocampus. Nature 577, 531–536 (2020).
Chen, X. et al. A brain cell atlas integrating single-cell transcriptomes across human brain regions. Nat. Med. https://doi.org/10.1038/s41591-024-03150-z (2024).
Ramnauth, A. D. et al. Spatiotemporal analysis of gene expression in the human dentate gyrus reveals age-associated changes in cellular maturation and neuroinflammation. Cell Rep. 44, 115300 (2025).
Zhou, Y., Su, Y., Ming, G. L. & Song, H. Special properties of adult neurogenesis in the human hippocampus: Implications for its clinical applications. Clin. Transl. Med. 13, e1196 (2023).
Kempermann, G. et al. Human adult neurogenesis: evidence and remaining questions. Cell Stem Cell 23, 25–30 (2018).
Zhou, Y., Song, H. & Ming, G. L. Genetics of human brain development. Nat. Rev. Genet. 25, 26–45 (2024).
Wallace, J. L. & Pollen, A. A. Human neuronal maturation comes of age: cellular mechanisms and species differences. Nat. Rev. Neurosci. 25, 7–29 (2024).
Vanderhaeghen, P. & Polleux, F. Developmental mechanisms underlying the evolution of human cortical circuits. Nat. Rev. Neurosci. 24, 213–232 (2023).
Knoth, R. et al. Murine features of neurogenesis in the human hippocampus across the lifespan from 0 to 100 years. PLoS ONE 5, e8809 (2010).
Sorrells, S. F. et al. Human hippocampal neurogenesis drops sharply in children to undetectable levels in adults. Nature 555, 377–381 (2018).
Kornack, D. R. & Rakic, P. Continuation of neurogenesis in the hippocampus of the adult macaque monkey. Proc. Natl Acad. Sci. USA 96, 5768–5773 (1999).
Ammothumkandy, A. et al. Altered adult neurogenesis and gliogenesis in patients with mesial temporal lobe epilepsy. Nat. Neurosci. 25, 493–503 (2022).
Moreno-Jimenez, E. P. et al. Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer’s disease. Nat. Med. 25, 554–560 (2019).
Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).
Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).
Han, L. et al. Cell transcriptomic atlas of the non-human primate Macaca fascicularis. Nature 604, 723–731 (2022).
Kohler, S. J., Williams, N. I., Stanton, G. B., Cameron, J. L. & Greenough, W. T. Maturation time of new granule cells in the dentate gyrus of adult macaque monkeys exceeds six months. Proc. Natl Acad. Sci. USA 108, 10326–10331 (2011).
Ngwenya, L. B., Heyworth, N. C., Shwe, Y., Moore, T. L. & Rosene, D. L. Age-related changes in dentate gyrus cell numbers, neurogenesis, and associations with cognitive impairments in the rhesus monkey. Front. Syst. Neurosci. 9, 102 (2015).
Hodge, R. D. et al. Conserved cell types with divergent features in human versus mouse cortex. Nature 573, 61–68 (2019).
Lein, E. S. et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature 445, 168–176 (2007).
Hart, J. C., Ellis, N. A., Eisen, M. B. & Miller, C. T. Convergent evolution of gene expression in two high-toothed stickleback populations. PLoS Genet. 14, e1007443 (2018).
Foster, C. S. P. et al. Different genes are recruited during convergent evolution of pregnancy and the placenta. Mol. Biol. Evol. 39, msac077 (2022).
Bontonou, G. et al. Evolution of chemosensory tissues and cells across ecologically diverse drosophilids. Nat. Commun. 15, 1047 (2024).
Foote, A. D. et al. Convergent evolution of the genomes of marine mammals. Nat. Genet. 47, 272–275 (2015).
She, R. et al. Comparative landscape of genetic dependencies in human and chimpanzee stem cells. Cell 186, 2977–2994 (2023).
Whitmarsh-Everiss, T. & Laraia, L. Small molecule probes for targeting autophagy. Nat. Chem. Biol. 17, 653–664 (2021).
Yoshimori, T., Yamamoto, A., Moriyama, Y., Futai, M. & Tashiro, Y. Bafilomycin A1, a specific inhibitor of vacuolar-type H+-ATPase, inhibits acidification and protein degradation in lysosomes of cultured cells. J. Biol. Chem. 266, 17707–17712 (1991).
Huss, M. et al. Concanamycin A, the specific inhibitor of V-ATPases, binds to the Vo subunit c. J. Biol. Chem. 277, 40544–40548 (2002).
Van Galen, P. et al. Single-cell RNA-Seq reveals AML hierarchies relevant to disease progression and immunity. Cell 176, 1265–1281 (2019).
Bielecki, P. et al. Skin-resident innate lymphoid cells converge on a pathogenic effector state. Nature 592, 128–132 (2021).
Peng, Y. R. et al. Molecular classification and comparative taxonomics of foveal and peripheral cells in primate retina. Cell 176, 1222–1237 (2019).
Ichihara, A. & Yatabe, M. S. The (pro)renin receptor in health and disease. Nat. Rev. Nephrol. 15, 693–712 (2019).
Bracke, A. & von Bohlen Und Halbach, O. Roles and functions of Atp6ap2 in the brain. Neural Regen. Res 13, 2038–2043 (2018).
Schafer, S. T. et al. The Wnt adaptor protein ATP6AP2 regulates multiple stages of adult hippocampal neurogenesis. J. Neurosci. 35, 4983–4998 (2015).
Hirose, T. et al. ATP6AP2 variant impairs CNS development and neuronal survival to cause fulminant neurodegeneration. J. Clin. Invest. 129, 2145–2162 (2019).
Leeman, D. S. et al. Lysosome activation clears aggregates and enhances quiescent neural stem cell activation during aging. Science 359, 1277–1283 (2018).
Lange, C. et al. The H+ vacuolar ATPase maintains neural stem cells in the developing mouse cortex. Stem Cells Dev. 20, 843–850 (2011).
Qu, Q. et al. Lithocholic acid binds TULP3 to activate sirtuins and AMPK to slow down ageing. Nature https://doi.org/10.1038/s41586-024-08348-2 (2024).
Song, Q., Meng, B., Xu, H. & Mao, Z. The emerging roles of vacuolar-type ATPase-dependent Lysosomal acidification in neurodegenerative diseases. Transl. Neurodegener. 9, 17 (2020).
Aoto, K. et al. ATP6V0A1 encoding the a1-subunit of the V0 domain of vacuolar H+-ATPases is essential for brain development in humans and mice. Nat. Commun. 12, 2107 (2021).
Guerrini, R. et al. Phenotypic and genetic spectrum of ATP6V1A encephalopathy: a disorder of lysosomal homeostasis. Brain 145, 2687–2703 (2022).
Wang, M. et al. Transformative network modeling of multi-omics data reveals detailed circuits, key regulators, and potential therapeutics for Alzheimer’s disease. Neuron 109, 257–272 (2021).
Sorrells, S. F. et al. Immature excitatory neurons develop during adolescence in the human amygdala. Nat. Commun. 10, 2748 (2019).
Nascimento, M. A. et al. Protracted neuronal recruitment in the temporal lobe of young children. Nature https://doi.org/10.1038/s41586-023-06981-x (2023).
Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).
La Manno, G. et al. Molecular diversity of midbrain development in mouse, human, and stem cells. Cell 167, 566–580 (2016).
Pedregosa, F. et al. Scikit-learn: machine learning in Python. J. Mach. Learn. Res. 12, 2825–2830 (2011).
Marques, S. et al. Oligodendrocyte heterogeneity in the mouse juvenile and adult central nervous system. Science 352, 1326–1329 (2016).
Bindea, G. et al. ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics 25, 1091–1093 (2009).
Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).
Yu, W., Clyne, M., Khoury, M. J. & Gwinn, M. Phenopedia and Genopedia: disease-centered and gene-centered views of the evolving knowledge of human genetic associations. Bioinformatics 26, 145–146 (2010).
Sun, Y. et al. Brain-wide neuronal circuit connectome of human glioblastoma. Nature https://doi.org/10.1038/s41586-025-08634-7 (2025).
Wen, Z. et al. Synaptic dysregulation in a human iPS cell model of mental disorders. Nature 515, 414–418 (2014).
Kim, N. S. et al. Pharmacological rescue in patient iPSC and mouse models with a rare DISC1 mutation. Nat. Commun. 12, 1398 (2021).
Chiang, C. H. et al. Integration-free induced pluripotent stem cells derived from schizophrenia patients with a DISC1 mutation. Mol. Psychiatry 16, 358–360 (2011).
Huang, W. K. et al. Generation of hypothalamic arcuate organoids from human induced pluripotent stem cells. Cell Stem Cell 28, 1657–1670 (2021).
Jgamadze, D. et al. Structural and functional integration of human forebrain organoids with the injured adult rat visual system. Cell Stem Cell 30, 137–152 (2023).
Gracia-Diaz, C. et al. Gain and loss of function variants in EZH1 disrupt neurogenesis and cause dominant and recessive neurodevelopmental disorders. Nat. Commun. 14, 4109 (2023).
Zhou, Y. et al. Autocrine Mfge8 signaling prevents developmental exhaustion of the adult neural stem cell pool. Cell Stem Cell 23, 444–452 (2018).
Schwarzkopf, M. et al. Hybridization chain reaction enables a unified approach to multiplexed, quantitative, high-resolution immunohistochemistry and in situ hybridization. Development 148, dev.199847 (2021).
Sun, G. J. et al. Latent tri-lineage potential of adult hippocampal neural stem cells revealed by Nf1 inactivation. Nat. Neurosci. 18, 1722–1724 (2015).
Sun, G. J. et al. Tangential migration of neuronal precursors of glutamatergic neurons in the adult mammalian brain. Proc. Natl Acad. Sci. USA 112, 9484–9489 (2015).
Chan, S. et al. A method for manual and automated multiplex RNAscope in situ hybridization and immunocytochemistry on cytospin samples. PLoS ONE 13, e0207619 (2018).
Acknowledgements
We thank members of the Song, Ming and Zhou laboratories, and S. Zhu (CAS Inst. of Neurosci./CEBSIT) for discussion; K. M. Christian (U Penn) for comments; Z. Xu (CAS Inst. of Genet. & Dev. Biol.), X. Teng (ACD, Bio-Techne), L. Gu (from S.X. lab), C. Chen (from S.X. lab) and L. Xie (from Y. Chen lab at CAS Inst. of Neurosci./CEBSIT) for help on in situ hybridization experiments; B. Temsamrit (U Penn), E. LaNoce (U Penn), A. Garcia-Epelboim (U Penn), G. Alepa (U Penn), A. Angelucci (U Penn) and Y. Lu (CAS Inst. of Neurosci./CEBSIT) for laboratory support; and W. Luo, Q. Wu and Y. Chen from U Penn for providing reagents. Some schematic illustrations were created using images modified from BioRender.com. This work was supported by grants from the National Institutes of Health (R35NS137480 to G.-l.M., R35NS116843 and RF1AG079557 to H.S. and R01NS127913 to Y.S.), the Lieber Institute for Brain Development (to J.E.K., T.M.H. and D.R.W.), Dr. Miriam and Sheldon G. Adelson Medical Research Foundation (to G.-l.M.), the National Key Research and Development Program of China (2024YFA1803400 to Y. Zhou), Shanghai Science and Technology Development Funds (24QA2710400 to Y. Zhou), Shanghai Pujiang Program (23PJ1414500 to Y. Zhou), Science and Technology Commission of Shanghai Municipality (22JC1403100 to S.X.) and the National Science Foundation of China (32321003 and 32371072 to S.X.).
Author information
Authors and Affiliations
Contributions
Y. Zhou led the project and contributed to all aspects. Y.S., T.G., X.M. and L.W. contributed to computational data analyses. Q.Y. and Y.H. developed the in vitro human hippocampal neuron culture system and Q.Y. contributed to in vitro culture experiments. J.L., Y. Zhong, M.J., X.L., L.Y., C. Li, S.X. and J.H. contributed to histology analysis. B.C.K., A.N.V., I.H., S.K.K., J.E.K., T.M.H., D.W.N. and D.R.W. provided human hippocampal specimens. C. Liu provided marmoset specimens. Y. Zhong, N.Y., Z.L., Z.S. and C. Li provided macaque specimens. Y. Zhou, Y.S., G.-l.M. and H.S. conceived the project and wrote the manuscript with inputs from all authors.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Neuroscience thanks the anonymous reviewers for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data
Extended Data Fig. 1 Identification and molecular characteristics of immature neurons in a published macaque hippocampal single-cell RNA sequencing dataset using traditional unsupervised clustering.
a, Gene module analysis of the previously annotated immature dentate granule cells (imGCs) and mature dentate granule cells (mGCs) in an adult macaque hippocampus single-nucleus RNA sequencing dataset20 using a well-recognized published list of mouse immature progeny (neuroblast and imGC)-enriched and mGC-enriched genes11. Box plots as in Fig. 3a. b, Top Gene Ontology (GO) term groups for genes enriched in imGCs (using published annotations with traditional unsupervised clustering20). Schematics created using BioRender.com. Reg., regulation.
Extended Data Fig. 2 Performance of machine learning model for macaque imGCs and feature extraction and comparison of gene weights defining imGCs in different species.
a, Measuring performance of our machine learning model for macaque datasets. Line plot showing the accuracy score of the machine learning classifier varying with decreasing regularization strength as estimated by cross-validation. The red line shows a 95% confidence interval on the estimation of the accuracy score. #Sum abs (coeffs): sum of the absolute value of regression coefficients. b, Heatmap showing expression of top gene weights in top-scoring cells of each prototype determined by our machine learning model for macaque datasets. The genes listed are the top 25 weights defining macaque imGCs. c, Venn diagram of the positive gene weights defining imGCs in humans, macaques, and mice that were generated by separate machine learning models (weights for human and mouse imGCs were generated in ref. 25). Schematics in c created using BioRender.com. Astro: astrocyte; OPC: oligodendrocyte progenitor cell; mOli: mature oligodendrocyte.
Extended Data Fig. 3 Specificity of our machine learning approach for identification of immature neurons in the macaque brain.
a, The fractions of cells with high similarity scores (pimGC ≥ 0.85) among excitatory dentate granule cell (GC), non-GC excitatory neuron, GABAergic interneuron, and non-neuronal cell clusters in various single-cell or single-nucleus RNA sequencing (scRNA-seq) datasets of the macaque hippocampus. Each dot represents data from one specimen from each study (noted by first author’s last name). Note that all imGCs identified reside in the GC clusters. b, No immature neurons were identified using our machine learning model in a scRNA-seq dataset of two 6-year macaque neocortex (one male and one female)42.
Extended Data Fig. 4 Shared molecular signatures of immature neurons in the hippocampus of different species.
a, GO network of biological processes associated with imGC-enriched genes in different species in comparison to mGCs, colored by FDR-adjusted P value. Only significantly enriched nodes are displayed (one-sided hypergeometric test, FDR-adjusted P < 0.05). The node size represents the term enrichment significance. Examples of the most substantial terms per group are shown. b, Violin plots showing normalized expression of 8 imGC-enriched genes shared across four species in imGCs and GCs (one-way Wilcoxon rank-sum test). A total of nine shared genes were identified and eight are shown here (see Fig. 3d for the plot for the ninth gene, DPYSL5). Schematics created using BioRender.com.
Extended Data Fig. 5 Enrichment of immature neuronal gene features in imGCs of different species.
a, Expression patterns of nine shared imGC-enriched genes across species in the dentate gyrus (DG) of adult mice, eight of which show enrichment in the neurogenic subgranular zone (except for PROX1). Images are from the Allen Brain in situ hybridization database46 (https://mouse.brain-map.org/). P: postnatal day. b, A second set of sample confocal immunostaining images of DPYSL5 enrichment in imGCs in the hippocampi of infant and adult humans, postnatal macaques and marmosets, and adult mice. Scale bars: 10 µm. Asterisks indicate DPYSL5+ cells among STMN1+PROX1+ imGCs (Fig. 3e). c, Venn diagram depicting the overlap of GC-enriched genes across different species when compared to other cell types within the same datasets. d, A schematic illustration of our working model. In contrast to the traditional concept that a single genetic variant can drive cross-species cellular innovations in immature neuron regulation, our study revealed substantial interspecies variance in highly expressed genes enriched in imGCs, which converged onto conserved biological processes, suggesting imGCs in different species may recruit and use species-unique molecular features to drive similar biological processes regulating neuronal development. Schematics in c and d created using BioRender.com.
Extended Data Fig. 6 A schematic of imGC-enriched feature analysis in different species.
To explore species-specific imGC molecular features in a nonbiased manner, gene expression patterns of each individual gene associated with ‘ion transport’ and ‘synaptic transmission’, the two major GO terms showing the most species-specific enrichment, were plotted (Extended Data Fig. 7). Schematics created using BioRender.com.
Extended Data Fig. 7 Unbiased examination of divergent imGC molecular features across four species.
Red-blue heatmaps depict expression patterns of each individual gene associated with ‘ion transport’ and ‘synaptic transmission’, the two major GO terms showing the most species-specific enrichment. Exemplary genes in these two categories, such as those encoding various ion channels, glutamate receptors, GABA receptors, ATPases and transmembrane proteins, among others are highlighted with square boxes. The color bar represents z scores of gene expression, scaled to range from −2 to 2. Schematics created using BioRender.com.
Extended Data Fig. 8 Role of lysosomal vacuolar-type H+-transporting ATPase in the in vitro human hippocampal immature neuron culture.
a, A schematic illustration of the experimental design. Human induced pluripotent stem cell (iPSC) lines (C65 and WTC11) were differentiated into DCX+PROX1+ hippocampal imGCs before treatment with bafilomycin A1 (BafA1) and concanamycin A (ConA), two specific blockers against lysosomal vacuolar-type H+-transporting ATPases to measure neurite growth and neuronal activities. b–e, Characterization of hippocampal imGC culture derived from two independent iPSC lines. Sample confocal images (b) and quantification of DCX and PROX1 enrichment in the hippocampal neuron in vitro culture (c,d) and its cell death level (e) with different treatments. Scale bar: 10 µm. Asterisks indicate cleaved Caspase 3 (cCasp3)+DCX+PROX1+ imGCs (b). Box colors match treatment conditions. Dots represent data from individual images; the centerline represents the mean, box edges show s.e.m. and whiskers extend to the maximum and minimum values (n = 3 cultures per condition) (c–e). None of the quantifications were statistically significant using ANOVA posthoc test; exact P values in Supplementary Table 7 (c–e).
Supplementary information
Supplementary Table 1
Summary of published scRNA-seq or single-nucleus RNA-seq datasets used in the current study.
Supplementary Table 2
Molecular weights defining immature dentate granule cells in macaques.
Supplementary Table 3
GO terms related to the positive gene weights defining macaque immature dentate granule cells.
Supplementary Table 4
Lists of GO terms associated with genes enriched in immature dentate granule cells across four species.
Supplementary Table 5
Lists of GO terms associated with genes uniquely enriched in immature dentate granule cells in four species.
Supplementary Table 6
Summary of human specimens used for histology validation.
Supplementary Table 7
Exact P values for Fig. 5 and Extended Data Fig. 8.
Source data
Source Data Fig. 1
Statistical source data for Fig. 1b.
Source Data Fig. 2
Statistical source data for Fig. 2b.
Source Data Fig. 3
Statistical source data for Fig. 3b,f.
Source Data Fig. 4
Statistical source data for Fig. 4a.
Source Data Fig. 5
Statistical source data for Fig. 5c,e,g.
Source Data Extended Data Fig. 1
Statistical source data for ED Fig. 1.
Source Data Extended Data Fig. 3
Statistical source data for ED Fig. 3a,b.
Source Data Extended Data Fig. 8
Statistical source data for ED Fig. 8c–e.
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.
About this article
Cite this article
Zhou, Y., Su, Y., Yang, Q. et al. Cross-species analysis of adult hippocampal neurogenesis reveals human-specific gene expression but convergent biological processes. Nat Neurosci 28, 1820–1829 (2025). https://doi.org/10.1038/s41593-025-02027-9
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41593-025-02027-9