Abstract
Understanding the molecular logic of cortical cell-type diversity can illuminate cortical circuit function and evolution. Here, we performed single-nucleus transcriptome and chromatin accessibility analyses to compare neurons across three- to six-layered cortical areas of adult mice and across tetrapod species. We found that, in contrast to the six-layered neocortex, glutamatergic neurons of the three-layered mouse olfactory (piriform) cortex displayed continuous rather than discrete variation in transcriptomic profiles. Subsets of piriform and neocortical glutamatergic cells with conserved transcriptomic profiles were distinguished by distinct, area-specific epigenetic states. Furthermore, we identified a prominent population of immature neurons in piriform cortex and observed that, in contrast to the neocortex, piriform cortex exhibited divergence between glutamatergic cells in laboratory versus wild-derived mice. Finally, we showed that piriform neurons displayed greater transcriptomic similarity to cortical neurons of turtles, lizards and salamanders than to those of the neocortex. In summary, despite over 200 million years of coevolution alongside the neocortex, olfactory cortex neurons retain molecular signatures of ancestral cortical identity.
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
27,99 € / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
209,00 € per year
only 17,42 € 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
Raw and processed sn-multiome-seq and RNA-seq data have been deposited and available in Gene Expression Omnibus under accession no. GSE239477. Seurat and Scanpy integrated objects are available upon request. All other data are included in the main paper or the supplementary materials. Source data are provided with this paper.
Code availability
The R and Python analysis scripts used for this paper are available at GitLab at https://gitlab.com/fleischmann-lab/papers/zeppilli-et-al-2023.
References
Bear, D. M., Lassance, J.-M., Hoekstra, H. E. & Datta, S. R. The evolving neural and genetic architecture of vertebrate olfaction. Curr. Biol. 26, R1039–R1049 (2016).
Cisek, P. Evolution of behavioural control from chordates to primates. Phil. Trans. R. Soc. B 377, 20200522 (2022).
Roberts, R. J. V., Pop, S. & Prieto-Godino, L. L. Evolution of central neural circuits: state of the art and perspectives. Nat. Rev. Neurosci. 23, 725–743 (2022).
MacIver, M. A. & Finlay, B. L. The neuroecology of the water-to-land transition and the evolution of the vertebrate brain. Phil. Trans. R. Soc. B 377, 20200523 (2022).
Niimura, Y. Evolutionary dynamics of olfactory receptor genes in chordates: interaction between environments and genomic contents. Hum. Genom. 4, 107 (2009).
Weiss, L., Manzini, I. & Hassenklöver, T. Olfaction across the water–air interface in anuran amphibians. Cell Tissue Res. 383, 301–325 (2021).
Kaas, J. H. The evolution of brains from early mammals to humans. WIREs Cognitive Sci. 4, 33–45 (2013).
Rowe, T. B., Macrini, T. E. & Luo, Z.-X. Fossil evidence on origin of the mammalian brain. Science 332, 955–957 (2011).
Rowe, T. B. & Shepherd, G. M. Role of ortho-retronasal olfaction in mammalian cortical evolution. J. Comp. Neurol. 524, 471–495 (2016).
Kaas, J. H. Evolution of the neocortex. Curr. Biol. 16, R910–R914 (2006).
Carroll, S. B. Endless forms: the evolution of gene regulation and morphological diversity. Cell 101, 577–580 (2000).
Striedter, G. F. The telencephalon of tetrapods in evolution. Brain Behav. Evol. 49, 179–194 (2008).
García-Cabezas, M, Á., Zikopoulos, B. & Barbas, H. The Structural Model: a theory linking connections, plasticity, pathology, development and evolution of the cerebral cortex. Brain Struct. Funct. 224, 985–1008 (2019).
Kappers, C. A. The Phylogenesis of the Palaeo-Cortex and Archi-Cortes Compared with the Evolution of the Visual Neo-Cortex. (J. Truscott and Son, 1909).
Laurent, G. et al. in Micro-, Meso- and Macro-Dynamics of the Brain (eds Buzsáki, G. & Christen, Y.) Ch. 2 (Springer, 2016).
Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).
Yao, Z. et al. A taxonomy of transcriptomic cell types across the isocortex and hippocampal formation. Cell 184, 3222–3241.e26 (2021).
Bravo González-Blas, C. et al. SCENIC+: single-cell multiomic inference of enhancers and gene regulatory networks. Nat. Methods https://doi.org/10.1038/s41592-023-01938-4 (2023).
Diodato, A. et al. Molecular signatures of neural connectivity in the olfactory cortex. Nat. Commun. 7, 12238 (2016).
Delás, M. J. & Briscoe, J. Repressive interactions in gene regulatory networks: when you have no other choice. Curr. Top. Dev. Biol. 139, 239–266 (2020).
Greig, L. C., Woodworth, M. B., Galazo, M. J., Padmanabhan, H. & Macklis, J. D. Molecular logic of neocortical projection neuron specification, development and diversity. Nat. Rev. Neurosci. 14, 755–769 (2013).
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).
Mu, L. et al. SoxC transcription factors are required for neuronal differentiation in adult hippocampal neurogenesis. J. Neurosci. 32, 3067–3080 (2012).
Rotheneichner, P. et al. Cellular Plasticity in the Adult Murine Piriform Cortex: Continuous Maturation of Dormant Precursors Into Excitatory Neurons. Cerebral Cortex 28, 2610–2621 (2018).
Zhang, M. et al. Molecularly defined and spatially resolved cell atlas of the whole mouse brain. Nature 624, 343–354 (2023).
Qu, R. et al. Gene trajectory inference for single-cell data by optimal transport metrics. Nat. Biotechnol. https://doi.org/10.1038/s41587-024-02186-3 (2024).
Habib, N. et al. Div-seq: single nucleus RNA-seq reveals dynamics of rare adult newborn neurons. Science 353, 925–928 (2016).
Duan, Y. et al. Semaphorin 5A inhibits synaptogenesis in early postnatal- and adult-born hippocampal dentate granule cells. eLife 3, e04390 (2014).
Buckwalter, M. S. et al. Chronically increased transforming growth factor-β1 strongly inhibits hippocampal neurogenesis in aged mice. Am. J. Pathol. 169, 154–164 (2006).
Kastriti, M. E. et al. Ablation of CNTN2+ pyramidal neurons during development results in defects in neocortical size and axonal tract formation. Front. Cell. Neurosci. 13, 454 (2019).
Rasetto, N. B. et al. Transcriptional dynamics orchestrating the development and integration of neurons born in the adult hippocampus. Sci. Adv. 10, eadp6039 (2024).
Kerloch, T. et al. The atypical Rho GTPase Rnd2 is critical for dentate granule neuron development and anxiety-like behavior during adult but not neonatal neurogenesis. Mol. Psychiatry 26, 7280–7295 (2021).
Chatzi, C., Zhang, Y., Shen, R., Westbrook, G. L. & Goodman, R. H. Transcriptional profiling of newly generated dentate granule cells using TU tagging reveals pattern shifts in gene expression during circuit integration. eNeuro 3, ENEURO.0024-16.2016 (2016).
Gómez-Climent, M. Á. et al. A population of prenatally generated cells in the rat paleocortex maintains an immature neuronal phenotype into adulthood. Cereb. Cortex 18, 2229–2240 (2008).
La Rosa, C. et al. Phylogenetic variation in cortical layer II immature neuron reservoir of mammals. eLife 9, e55456 (2020).
Miller, R. A. et al. Mouse (Mus musculus) stocks derived from tropical islands: new models for genetic analysis of life-history traits. J. Zool. 250, 95–104 (2000).
Zilkha, N. et al. Sex-dependent control of pheromones on social organization within groups of wild house mice. Curr. Biol. 33, 1407–1420.e4 (2023).
Tran, Q. H. et al. Unbalanced CO-optimal transport. Proc. AAAI Conf. Artificial Intel. 37, 10006–10016 (2023).
Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).
Lopez, R., Regier, J., Cole, M. B., Jordan, M. I. & Yosef, N. Deep generative modeling for single-cell transcriptomics. Nat. Methods 15, 1053–1058 (2018).
Siletti, K. et al. Transcriptomic diversity of cell types across the adult human brain. Science 382, eadd7046 (2023).
Woych, J. et al. Cell-type profiling in salamanders identifies innovations in vertebrate forebrain evolution. Science 377, eabp9186 (2022).
Hain, D. et al. Molecular diversity and evolution of neuron types in the amniote brain. Science 377, eabp8202 (2022).
Norimoto, H. et al. A claustrum in reptiles and its role in slow-wave sleep. Nature 578, 413–418 (2020).
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).
Ulinski, P. S. & Rainey, W. T. Intrinsic organization of snake lateral cortex. J. Morphol. 165, 85–116 (1980).
Lust, K. et al. Single-cell analyses of axolotl telencephalon organization, neurogenesis, and regeneration. Science 377, eabp9262 (2022).
Bonfanti, L., La Rosa, C., Ghibaudi, M. & Sherwood, C. C. Adult neurogenesis and ‘immature’ neurons in mammals: an evolutionary trade-off in plasticity? Brain Struct. Funct. https://doi.org/10.1007/s00429-023-02717-9 (2023).
Gerhart, J. & Kirschner, M. The theory of facilitated variation. Proc. Natl Acad. Sci. USA 104, 8582–8589 (2007).
Wittkopp, P. J. & Kalay, G. Cis-regulatory elements: molecular mechanisms and evolutionary processes underlying divergence. Nat. Rev. Genet. 13, 59–69 (2012).
Dugas-Ford, J., Rowell, J. J. & Ragsdale, C. W. Cell-type homologies and the origins of the neocortex. Proc. Natl Acad. Sci. USA 109, 16974–16979 (2012).
Tosches, M. A. & Laurent, G. Evolution of neuronal identity in the cerebral cortex. Curr. Opin. Neurobiol. 56, 199–208 (2019).
Kaslin, J., Ganz, J. & Brand, M. Proliferation, neurogenesis and regeneration in the non-mammalian vertebrate brain. Philos. Trans. R Soc. Lond. B Biol. Sci. 363, 101–122 (2008).
Tosches, M. A. From cell types to an integrated understanding of brain evolution: the case of the cerebral cortex. Annu. Rev. Cell Dev. Biol. 37, 495–517 (2021).
Luzzati, F. A hypothesis for the evolution of the upper layers of the neocortex through co-option of the olfactory cortex developmental program. Front. Neurosci. 9, 162 (2015).
Striedter, G. F. & Northcutt, R. G. The independent evolution of dorsal pallia in multiple vertebrate lineages. Brain Behav. Evol. 96, 200–211 (2021).
Zeppilli, S. et al. Molecular characterization of projection neuron subtypes in the mouse olfactory bulb. eLife 10, e65445 (2021).
Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).
Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, 281–291.e9 (2019).
Lun, A. T. L., Bach, K. & Marioni, J. C. Pooling across cells to normalize single-cell RNA sequencing data with many zero counts. Genome Biol. 17, 75 (2016).
Traag, V. A., Waltman, L. & van Eck, N. J. From Louvain to Leiden: guaranteeing well-connected communities. Sci. Rep. 9, 5233 (2019).
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).
Zhang, Y. et al. Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).
Yao, Z. et al. A high-resolution transcriptomic and spatial atlas of cell types in the whole mouse brain. Nature 624, 317–332 (2023).
Wang, Q. et al. The Allen Mouse Brain Common Coordinate Framework: a 3D reference atlas. Cell 181, 936–953.e20 (2020).
Pedregosa, F. et al. Scikit-learn: machine learning in Python. J. Mach. Learn. Res. 12, 2825–2830 (2011).
Cao, K., Hong, Y. & Wan, L. Manifold alignment for heterogeneous single-cell multi-omics data integration using Pamona. Bioinformatics 38, 211–219 (2021).
Demetci, P., Tran, Q. H., Redko, I. & Singh, R. Jointly aligning cells and genomic features of single-cell multi-omics data with co-optimal transport. Preprint at bioRxiv https://doi.org/10.1101/2022.11.09.515883 (2022).
Acknowledgements
We thank Z. Herbert and M. Berkeley from the Molecular Biology Core Facilities at the Dana-Farber Cancer Institute for sequencing services. We thank N. Eckart from 10x Genomics for excellent technical support. We thank K. Babcock, F. Mosti, D. Silver, R. Van Drunen, J. Ritt, S. Firestein, H. Berry, K. Franks, S. Pantalacci, G. Beslon, H. Wichterle, B. Mensh and T. Pievani for critical comments on the manuscript. We thank A. Pierre for software support and C. Bravo González-Blas for analysis support with SCENIC+. We thank N. Nisim for technical assistance with the wild-derived mice and the Brown and Weizmann animal facilities for animal care. Work in the A.F., R.S. and A.C. laboratories was supported by National Institutes of Health (NIH) grant NIDCD R01DC020478. Work in the A.F. laboratory was supported by grants from the NIH, NIDCD R01DC017437 to A.F., the Robert J. and Nancy D. Carney Institute for Brain Science to A.F. and the Carney Graduate Award in Brain Science to S.Z. Carney Institute computational resources used in this work were supported by the NIH Office of the Director grant S10OD025181. Work in the A.C. laboratory was supported by project AI4scMed, France 2030 ANR-22-PESN-0002. 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
Contributions
S.Z., A.C. and A.F. conceptualized the study with input from all authors. S.Z. conducted all experiments and supervised all computational analyses. S.Z., R.A. and A.C. performed the transcriptomic analysis. A.C. performed the epigenomic analysis. A.O.G. performed the cross-species transcriptomic analysis. P.D. performed the OT integration between datasets. T.M.P. performed the gene trajectory analysis. D.H.B. performed the immature neuron-related analysis. T.K. and N.Z. assisted with the wild-derived mice. S.Z., T.K., S.R.D., R.S., M.A.T., A.C. and A.F. provided supervision and scientific guidance. S.Z., A.C. and A.F. wrote the manuscript with input from all authors.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Neuroscience thanks Paul Greer, Fenna Krienen, and the other, anonymous, reviewer(s) 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 Relative abundance, main cell types, and histological assessment across biological replicates and cortical areas of lab mice.
(a) Relative abundance of main cell types across biological replicates (donors) integrated from single-nucleus multiome sequencing (sn-multiome seq) experiments. From left to right: anterior piriform cortex (aPir) replicates, indicated by A; posterior piriform cortex (pPir) replicates, indicated by P; agranular insular cortex (AI) replicates, indicated by T; primary somatosensory cortex (SSp) replicates, indicated by N. Numbers correspond to the ID of mice. VLMC: vascular leptomeningeal cells; Micro: microglia; OPC_diff: differentiating oligodendrocytes; OPC: oligodendrocyte precursors; Oligo: oligodendrocytes; Astro: astrocytes; IN_MGE and IN_CGE: inhibitory neurons from medial and caudal ganglionic eminence, respectively; SL: semilunar cells; Pyr: pyramidal cells. (b) Post-hoc histological assessment of aPir and AI dissections from anterior coronal sections of adult lab mice ordered by ID mouse number. Asterisks indicate the microdissected area. Neurotrace counterstain in gray. (c) Same as in (b) but for pPir and SSp. (d) From left to right: UMAPs of aPir, pPir, AI, and SSp datasets color-coded by main cell types. (e) Gene expression levels of representative markers for each cell type across the four cortical areas, from left to right: aPir, pPir, AI, and SSp.
Extended Data Fig. 2 Quality control of single-nucleus multiome (RNA and ATAC) sequencing data of lab mice.
(a) Transcriptome quality control. Left: number of genes per nucleus quantified for each biological replicate. A indicates aPir replicates, P indicates pPir, T indicates AI, N indicates SSp. Numbers correspond to the ID of mice. Right: number of genes per nucleus identified for each main cell type. (b) Same as in (a) but for the fraction of mitochondrial content per nucleus. (c) Epigenome quality control for aPir biological replicates. From left to right: barcode rank plot, fragment size distribution, Transcription Start Site (TSS) enrichment and Fraction of Reads In Peaks (FRIP). (d) Same as in (c) but for pPir biological replicates. (e) Same as in (c) but for AI biological replicates. (f) Same as in (c) but for SSp biological replicates.
Extended Data Fig. 3 Piriform cortex-specific markers and histological validation.
(a) Left: gene expression levels of generic or subtype-specific markers for semilunar cells in the combined aPir and pPir datasets. Right: in situ hybridization images from the Allen Brain Atlas for some of the markers shown in the dotplot on the left, with the exception of the marker SATB1, which was validated using immunohistochemistry in Vglut1-CRE/INTACT-GFP transgenic mice. (b) Same as (a), but for generic or subtype-specific markers for pyramidal cells in the combined aPir and pPir datasets. (c) High magnifications in Pir layer 3 of immunohistochemical experiments using Vglut1-CRE/INTACT-GFP transgenic mice showing combinatorial expression of the specific marker for Pyr11, EBF2 (in magenta), with specific or generic markers for glutamatergic neurons (in cyan): EBF1 (Pyr10-11-specific); TBR1 and CUX1 (pan-excitatory/pyramidal neuron marker); MEIS1 (Vglut2-specific). Scale bar, 100 μm. (d) Same as (a), but for generic or subtype-specific markers for Vglut2 cells in the combined aPir and pPir datasets. (e) High magnifications in Pir layer 3 of immunohistochemical experiments using Vglut1-CRE/INTACT-GFP transgenic mice showing combinatorial expression of the specific marker for Vglut2 cells, PAPPA2 (in magenta), with generic markers for semilunar cells (RELN), INs (GABA), immature neurons (SOX11), and pyramidal cells (CUX1), or with markers highly specific to Vglut2 cells (MEIS1), or expressed in Pir layer 3 (BARHL1) (in cyan), to understand the identity of the uncharacterized Vglut2-expressing neuronal population. Scale bar, 100 μm. (f) Same as (a), but for generic or subtype-specific markers for immature neurons in the combined aPir and pPir datasets. (g) Same as (a), but for generic or subtype-specific markers for INs in the combined aPir and pPir datasets.
Extended Data Fig. 4 Integration of neurons from this study and a mouse single-cell reference atlas.
(a) Integration of neurons from this study and a mouse single-cell reference atlas17. UMAP of integrated neurons (n = 30,553). (b) UMAPs as shown in (a) with gene expression of generic markers for glutamatergic neurons (Slc17a7 and Slc17a6), inhibitory neurons (INs) (Gad2), and other established markers for neocortical projection neurons: Rorb (layer 4), Cux1 (layer 2/3), Ctip2 (layer 5), Fezf2 (layer 5), Foxp2 (layer 6), Nfia (layer 6). (c) Quantification of co-clustering between piriform, hippocampal formation and transition areas neurons from integration shown in (a). Neurons are grouped into main types per cortical area. Rectangles indicate co-clustering of neurons in the Seurat integrated clusters. Color represents the percentage of cells in an integrated cluster. L: layer; IT: intratelencephalic; NP: near projecting; CT: corticothalamic; Sub: subiculum; ProS: prosubiculum; PPP: para/post/pre subiculum; RHP: retrohippocampal region; DG: dentate gyrus; CA 1/2/3: hippocampal fields; IG: induseum griseum; FC: fasciola cinereal; AI: agranular insular; ENT: entorhinal (medial and lateral); TPE: Temporal association areas, Perirhinal area, Ectorhinal area. (d) Quantification of co-clustering between piriform glutamatergic neuron subtypes and hippocampal formation glutamatergic neurons from the integration shown in (a). Rectangles indicate co-clustering of neurons in the Seurat integrated clusters. Color represents the percentage of cells in an integrated cluster. Dentate gyrus (DG) glutamatergic neurons aligned with 13%, 31% and 7% of piriform subtypes Pyr7-8-9, respectively. CA 1 glutamatergic neurons aligned with 4% of piriform subtype Pyr7. DG: dentate gyrus; CA 1/2/3: hippocampal fields. (e) UMAPs of gene expression of transcription factors (TFs) highly enriched in piriform cortex compared to other cortical areas. (f) UMAPs of integrated neurons from this study and the mouse single-cell reference atlas (bottom) (Yao 2021), and visualization of only SSp neurons from the two studies (top: this study, bottom: single-cell reference atlas). (g) Quantification of co-clustering between SSp neurons of this study and of the single-cell reference atlas. Only SSp datasets were included in the integration to transfer projection neuron profile nomenclature to this study. SSp_yao indicates neurons from (Yao 2021), the rest corresponds to SSp clusters of this study. Dots indicate co-clustering of neurons in the Seurat integrated clusters. Size of the dots represents the percentage of cells in the integrated cluster. (h) Gene expression levels of Car3 in the SSp dataset. Car3 is expressed in SSp neurons co-clustering with piriform neurons in the immature neuron supertype. These Car3+ SSp neurons co-cluster with L4/5/6 intratelencephalic Car3+ neurons of the single-cell reference atlas. Given also the lack of DCX expression in SSp, we do not consider SSp neurons falling in the immature type as immature neurons. (i) Gene expression levels of the TF Rorb, which is highly enriched in the SL1 supertype (left), and of established TFs present in the SSp-specific supertypes (right). Pyr 14-15-16 of layers 4 and 5 are characterized by Rorb and Fezf2, Pyr17 is characterized by Foxp2, Bcl11b (Ctip2) and Nfia, corresponding to CT neurons of layers 6.
Extended Data Fig. 5 Epigenetic divergence of transcriptome-based supertypes across mouse cortical areas.
(a) UMAP of multiome ATAC data colored by aPir, pPir, AI, and SSp datasets. Neurons are integrated using Harmony. (b) UMAP as in (a) colored by the corresponding transcriptome-based supertype. For interneurons areas mix, while for glutamatergic neurons piriform separates from SSp, while AI overlaps with both (see (a)). (c) UMAP as in (a) colored by epigenome-based leiden clusters. (d) Mapping of transcriptome-based clusters (RNA, supertypes) to epigenome-based (ATAC, leiden) clusters. For glutamatergic neurons, multiple area-specific epigenome-based clusters correspond to a single supertype. Adjusted Rand Indices (ARIs) quantify the cluster overlap: for all neurons= 0.43; for INs= 0.88; for glutamatergic neurons= 0.37. (e) High-quality e-regulons are selected for downstream analysis based on the correlation between AUC (Area Under the Curve) scores for target genes and target CREs (Cis Regulatory Elements). A correlation cut-off of 0.4 and a minimum number of target genes of 10 are used. (f) Upset plot of the intersection of target genes for the e-regulon Rorb(+), which is shared across aPir, pPir, AI and SSp. Vertical bars show the number of target genes in the corresponding intersection of the matrix below. Horizontal bars show the total number of target genes for each cortical area. Of note, main text states 9% overlap between aPir and SSp target genes. That overlap is computed with a Jaccard similarity index and equivalent to taking in the upset plot the (relative) size of a combination of intersections. (g) Upset plot of the intersection of target CREs for the e-regulon Rorb(+). See (f) for details. (h) From left to right, average log-normalized expression of target genes (TGs) of Rorb identified in aPir, pPir, AI, and SSp. TGs of a given area are shown across areas (even if they are regulated by other TFs). (i) Difference of gene expression between target genes identified in aPir and SSp for the e-regulon Rorb(+) remains within a 2-fold change. The average log- normalization expression (y-axis) is only used to spread data points. (j) From left to right, average log-normalized expression of a random set of genes (n = 500) for aPir, pPir, AI, and SSp, similar to (h). Expression patterns are qualitatively equivalent as in (h). (k) Difference of gene expression between set of random genes in aPir and SSp datasets remains within a 2-fold change and indicates that Rorb TGs in (i) behave as a set of random genes.
Extended Data Fig. 6 Cell type discreteness and TF coexpression and repression analyses.
(a) Kernel-density estimate plots showing cluster distance per cortical area computed on integrated clusters composed of glutamatergic neurons from all areas using highly variable transcription factors. Integration was performed between our data (aPir, pPir, AI, SSp) and cortical areas from Yao et al.17: SSp (as internal control to compare with our SSp dataset), primary motor cortex (MOp), visual cortex (VIS), and dentate gyrus (DG). (b) Top 500 positively (left) and negatively (right) correlated TF pairs projected from one cortical area (source, rows) to another (target, columns). Distances are normalized to be 1.0 when source is target. Lower distance indicates lower correlation match. TF-TF correlations were computed from log-normalized expression matrices for each area (see Supplementary Fig. 3c). (c) Repressive (top row) and activating (middle row) TF-TF interactions identified per cortical area e-GRN (aPir, pPir, SSp). AI was excluded for lack of laminar information. Repression and activation were determined using SCENIC + , which considered (anti)correlations for each e-regulon as computed between the TF, its target genes and its target CREs. Results in main text are based on an (anti)correlation value of 0.4 (red line) and minimum number of target genes 10. Only TFs expressed in at least 20% of neurons in a given cortical layer are considered. Gray area indicates bounds of pattern stability. (d) Activating and repressing ATAC regions recovered from leave-10%-out datasets. Each dot represents the ratio of regions in the leave-10%-out dataset over the original e-GRN, for both activating and repressing regions. Dots are colored by cortical area. To indicate the trend per cortical area, linear regressions (solid lines) with +/- bootstrapped 95% confidence intervals (shaded area) are shown. As a reference, the dashed black line is the equal ratio of recovery for activating and repressing regions. (e) Gene expression levels of predicted piriform repressors shown in Fig. 3i in aPir (top) and pPir (bottom). Neurons are labeled by cortical layer (SL and Pyr cells), neuron type (INs and Vglut2 cells), or “Unknown” if layer information was unavailable.
Extended Data Fig. 7 Adult piriform cortex immature neurons and the transcriptomic divergence of pyramidal cells between lab and wild mice and between human individuals.
(a) Left: distribution of piriform cortex mature and immature neurons in reanalyzed MERFISH data (Zhang et al.25). Right: cumulative distribution along the anterior–posterior axis of piriform cortex of all pyramidal cells, immature neurons, and pyramidal cells from Yao 2023 aligning with subtype Pyr7 (Yao et al.64; Zhang et al.25). (b) Classification accuracy of support vector machine (SVM) trained on combined RNA and ATAC data of the pPir dataset. Each neuron type is distinguished. (c) Same as (b) but applied to subtypes. Each neuron subtype is distinguished. (d) Conservation scores for aPir, pPir, and SSp. Scores are median probabilities of transcriptomic similarity between lab and wild datasets, quantified by considering all mature neurons, or neurons of a given cell type (SL, Pyr, Vglut2, and IN). A conservation score of 0.5 indicates perfect mixing between lab and wild datasets. 95% confidence intervals (CIs) for each neuron type are reported in brackets. (e) Statistical significance of pairwise differences in conservation score between cortices reported per neuron type (see also (d)). P-values are calculated using two-sided Mann–Whitney U-Tests and Bonferroni-corrected. P-values in red are smaller than α0.01(Bonf.)=8.34e-4, p values in yellow are (only) smaller than α0.05(Bonf.)=4.167e-3. (f) Similarity (as cosine distance) between lab and wild neuron types upon integrating lab and wild pPir datasets using scVI. Pyramidal cells were on average less similar to each other than the other neuron types. (g) Percentage of lab pPir neighbors in scVI integration when computing nearest neighbor graphs from 5 to 100. Data shown as mean +/- bootstrapped 95% CIs, for 7861 aligned (other Pyr cells) and 184 misaligned cells (wild-specific Pyr cells). Alignment of cells between lab and wild datasets is taken from the OT integration shown in (c). (h) Classification accuracy of SVM trained on combined lab and wild pPir data. Neuron types are distinguished with 98.8% accuracy. (i) Immunohistochemistry using lab (left) and wild-derived mice (right) of markers for piriform neuronal populations (in cyan), namely RELN for SL, CUX1 for Pyr, GABA for INs, co-stained with DCX (in magenta), a canonical marker for immature neurons. Inset white boxes with higher magnification show coexpression of CUX1 with DCX in both lab and wild-derived mice. Scale bar, 100 μm. (j) Conservation scores per neuron type for adult human piriform (PIR) and primary somatosensory cortex (S1C) computed using published adult human whole-brain sn-RNA-seq data (Siletti et al.41). Scores are computed per pair of three human donors. Abbreviated donor IDs: h18 for H.18.30.001, h19.1 for h19.30.001, and h19.2 for H19.30.002. Within violin plots, black circles mark medians and black bars indicate 95% CIs. Bonferroni-corrected significance thresholds were α0.01(Bonf.)=1.59e-4 and α0.05(Bonf.)=7.94e-4. IT: intratelencephalic; NP: near projecting (see Supplementary Table 1 for sample size, see (l) for statistical significance). (k) Conservation scores as in (d), but for human piriform (left) and primary somatosensory cortex (right). Scores are median probabilities of transcriptomic similarity per pair of donors. (l) Statistical significance of pairwise differences in conservation score as shown in (e), but between donors. P-values are calculated using two-sided Mann–Whitney U-Tests and reported per neuron type.
Extended Data Fig. 8 Transcriptomically-defined cell types across cortical areas of wild-derived mice.
(a) Relative abundance of main cell types across biological replicates (donors) integrated from single-nucleus RNA (sn-RNA-seq) and multiome (sn-multiome seq) sequencing experiments. Replicates IDs 1 and 2 derive from sn-multiome seq experiments, replicates IDs 3, 4, 5 and 6 derive from sn-RNA-seq experiments. From left to right: aPir replicates, indicated by A; pPir replicates, indicated by P; SSp replicates, indicated by N. Numbers correspond to the ID of the donor mouse. W: wild. (b) Post-hoc histological assessment of aPir, pPir, and SSp dissections from anterior and posterior coronal sections of adult wild-derived mice ordered by ID mouse number. Asterisks indicate the microdissected area. Neurotrace counterstain in gray. (c) Left: number of genes per nucleus quantified for biological replicates. A indicates aPir replicates, P indicates pPir, N indicates SSp. Numbers correspond to the ID of mice. Right: number of genes per nucleus identified for main cell types. (d) Same as in (c) but for the fraction of mitochondrial content per nucleus. (e) From left to right: UMAPs of aPir, pPir, and SSp datasets color-coded by main cell types. (f) Gene expression levels of representative markers for each cell type across the three cortical areas, from left to right: aPir, pPir, and SSp. (g) Optimal transport (OT) alignment of main cell types between lab and wild datasets for each cortical area, from left to right: aPir, pPir, and SSp. Color and size of dots indicate the probability of alignment.
Extended Data Fig. 9 Quantification of co-clustering between cortical glutamatergic neurons of mice, reptiles, and salamander grouped by areas and LISI scores.
(a) Broad quantification of co-clustering in the integrated clusters between glutamatergic neurons from mouse and non-mammalian cortical areas, highlighting greater transcriptomic similarity of piriform glutamatergic neurons to those of non-mammals than to those of the neocortex. Pir: piriform; NCx: neocortex; DCtx, LCtx: dorsal, lateral cortex; aDVR: anterior dorsal ventricular ridge; dDP, LP, dVP: deep dorsal, lateral, deep ventral pallium. Rectangles indicate co-clustering of neurons (rows) in the integrated clusters (columns). Color of the rectangle represents the percentage of neurons in the integrated cluster. (b) Distribution of LISI scores across neuronal clusters of the datasets Tosches et al.45 (turtle and lizard), Hain et al.43 (lizard), and Woych et al.42 (salamander). A mean value close to 4 indicates a cell type that is well-mixed with neurons from other species, while a value close to 0 indicates a cell type mixed only with neurons from the same species. Box-and-whisker plots show min to max, center (median), 25th and 75th percentile box bounds, and whiskers extending to 1.5 * interquartile range (see Supplementary Table 1 for sample size).
Extended Data Fig. 10 Cross-species gene module score and TF coexpression analyses.
(a) Expression enrichment of gene modules Pyr-like (top, blue) and SL-like (bottom, orange) in this study (left), in the salamander dataset (right), and in Yao., 2021 (middle). NTS: neurotensin neurons. A positive score indicates that the set of genes in the module are expressed in a particular cluster more highly compared to the average expression across all clusters of the dataset (see Supplementary Table 1 for sample size). (b) UMAPs of SL-like (top) and Pyr-like (bottom) gene module scores across datasets shown in (a), from left to right: this study, Yao et al.17, and Woych et a.l42. Color bar indicates the enrichment score of a module.
Supplementary information
Supplementary Information
Supplementary text: abbreviations, Additional Methods, Supplementary Figs. 1–6 and Supplementary Tables 1–3.
Source data
Source Data
Statistical source data for Figs. 4k and 5e and Extended Data Figs. 7f, 9b and 10a.
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
Zeppilli, S., Gurrola, A.O., Demetci, P. et al. Single-cell genomics of the mouse olfactory cortex reveals contrasts with neocortex and ancestral signatures of cell type evolution. Nat Neurosci 28, 937–948 (2025). https://doi.org/10.1038/s41593-025-01924-3
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41593-025-01924-3