Abstract Background Ephrin-B2 (EB2) signaling plays a crucial role in regulating memory and synaptic plasticity. Comprehensive identification of cell-type-specific transcriptomic changes in EB2 knockout mice is expected to shed light on potential mechanisms associated with EB2 signaling in cognitive functions. Results Our study captures changes in cell populations in response to EB2 manipulation and reveals previously uncharacterized cell types (CPA6 + inhibitory neurons) in the mPFC. We validated the differential transcriptomic activity of Pbx1 and Meis1 in CPA6 + neurons using fluorescence in situ hybridization (ISH) in EB2-vGATCre mice. The aberrant presence of CPA6 + neurons in the mPFC may correlate with cognitive impairments induced by EB2 deletion in vGAT + neurons. Analyzing differentially expressed genes (DEGs) in individual cell clusters, we identified alterations related to synapse organization and development, cognition, amyloid-beta formation, and locomotor behavior. Additionally, our DEGs overlapped with human genome-wide association study (GWAS) candidate genes related to cognition and anxiety, underscoring the relevance of our mouse model to human disease. Conclusions We present a comprehensive atlas of cell-type-specific gene expression changes in this synaptic deficiency model and identify novel cell-type-specific targets implicated in cognitive deficits. Our investigation provides a detailed map of the cell types, genes, and pathways altered in this inhibitory synaptic deficiency model. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-025-02333-5. Keywords: Ephrin-B2, Cognition, Single-nucleus RNA sequencing, GABAergic neurons Background GABAergic inhibitory neurons are essential components of neural circuits, working together with excitatory neurons and glia to maintain the balance necessary for normal brain development and function. They constitute approximately 20–30% of the cortical neurons. Dysfunction in their synapse can disrupt the delicate balance between excitation and inhibition, potentially leading to neurological disorders or cognitive impairments [[42]1]. Molecular mechanisms likely underlie the development of different inhibitory synapses [[43]2]. These synaptic molecules play a crucial role in shaping cellular and synaptic programs in inhibitory subpopulations, both during developmental phase and adulthood, influencing behavioral processes like learning and memory [[44]3]. Genetic manipulation of these synaptic molecules within inhibitory neurons could impact a heterogeneous population of neuronal cells, and subsequently coordinate an array of fundamental behaviors. However, our understanding of how genetic alterations in synaptic molecules affect the diverse population of inhibitory neuronal population and their interaction with other brain cell types, as well as the broader functional connectivity between different brain regions, remains limited. Ephrin-Eph ligand-receptor synaptic signaling is a crucial molecular guidance system in the central nervous system with versatile roles in synaptic function and dysfunction [[45]4]. Specifically, Ephrin-B2 (EB2), which is a transmembrane molecule, can transduce a reverse signal into EB2-expressing cells [[46]5]. In excitatory neurons, EB2 regulates synaptic strength by modulating AMPA receptor trafficking [[47]6, [48]7]. While EB2’s importance in excitatory neurons was well documented, its function in inhibitory neurons has received less attention until recent studies, which detected EB2 expression in nearly all inhibitory neurons and suggested its role in transducing reverse signals to guide their migration into neocortex during embryonic development [[49]8]. Loss of Ephrin-B in the major class of GABAergic interneurons (GAD65/67 + interneurons) affected distinct interneuron subpopulations in the cortex [[50]9]. Despite these advancements, our knowledge of EB2 reverse signaling pathways in GABAergic neurons which are critical for shaping higher brain connectivity and other cellular functions remains rudimentary. Further investigation is also required to determine how EB2 loss affects distinct cell subtypes in the brain which is not confined to inhibitory neurons. We hypothesized that EB2 ablation in GABAergic neurons influences distinct subtypes of cell population at transcriptomic level. To investigate our hypothesis, we generated mice with Ephrin-B2 specifically knocked out in vGAT + neurons (vGAT-Cre; Ephrin-B2^loxP, EB2-vGATCre mice). Selective restoration of EB2 expression in GABAergic neurons of the PFC reversed learning and memory deficits in EB2-vGATCre mice. Additionally, these mice exhibited impaired resting-state network connectivity between the PFC and the reticular nucleus of the thalamus (RNT) region, along with impaired synaptic spatial plasticity in the prefrontal cortex (PFC), underscoring the impact of EB2 loss in inhibitory neurons. To comprehensively assess changes in cell-type diversity following EB2 ablation in vGAT + neurons, we used a single-nucleus RNA sequencing to identify novel populations and uncover transcriptomic diversity within known populations. Our snRNA-seq data enabled us to identify a novel population of transcriptionally and topographically distinct neurons following EB2 deletion in vGAT + neurons (CPA6 + neurons). Furthermore, we validated the differential transcriptomic expression of Pbx1 and Meis1 using fluorescence in situ hybridization (ISH), confirming their expression changes within EB2-vGATCre mice. Taken together, these findings enhance our understanding of the EB2-mediated molecular basis of cell-type heterogeneity within the PFC, laying the groundwork for targeted investigations into circuit-specific manifestations of neurocognitive disorders. Results Cognitive deficits in GABAergic neuron-specific EB2 KO mice are rescued by targeted EB2 over-expression in the PFC GABAergic neuron-specific EB2 KO mice were used for this study [[51]10]. The specific knockout of EB2 in GABAergic neurons is illustrated in Additional file [52]1: Fig. S1A and B. Cells expressing vGAT-Cre showed tdTomato fluorescence, marking EB2 KO. In the PFC, tdTomato-positive neurons colocalized with GABA and GAD65/67, confirming vGAT-Cre specificity in GABAergic neurons (Fig. [53]1A and B), with a minor activity of vGATCre in PV neurons (Additional file [54]1: Fig. S1C), but not in vGluT2 and CaMK2α neurons (Additional file [55]1: Fig. S1D, E). Fig. 1. [56]Fig. 1 [57]Open in a new tab Specific ablation of EB2 in vGAT + neurons induced cognitive deficits. A and B In vGATCre mice, the neurons labeled with Ai9 Rosa26 reporter tdTomato were expressing GABA and GAD65/67. DAPI indicated the nuclei location. Scale bar, 20 μm. C NORT discrimination index in EB2-vGATCre mice as compared with EB2-flox mice (left) adolescent, **P = 0.0097, Mann–Whitney U test. (right) Adult, **P = 0.0012, Mann–Whitney U test. D Left, schematic representation of the tamoxifen treatment protocol. Behavioral tests were carried out during postnatal week 4 and week 10. Bar plots showing the NORT discrimination index in Cre + T mice compared with W/OCre + T and Cre + V mice. Middle, P = 0.0051, Kruskal–Wallis ANOVA test, W/OCre + T vs. Cre + V, P > 0.9999, W/OCre + T vs. Cre + T, *P = 0.0153, Cre + V vs. Cre + T, *P = 0.0227. Right, P = 0.0298, ordinary one-way ANOVA test, W/OCre + T vs. Cre + V, P = 0.9987, W/OCre + T vs. Cre + T, *P = 0.0416, Cre + V vs. Cre + T, P = 0.0658. E NORT discrimination index in Ephrin-B2.^lacZ/+mice as compared with WT mice (*P = 0.0317, Mann–Whitney U test). F Specific restoration of Ephrin-B2 by the precise bilateral injections of pAAV-EF1a-DIOEfnb22A-EGFP (OE virus) rescues NOR behavior deficit. P = 0.0038, ordinary one-way ANOVA test, EB2-vGATCre + Control vector vs. EB2-vGATCre + OE vector, **P = 0.0041, EB2-vGATCre + Control vector vs. Off target + OE vector, P = 0.7480, EB2-vGATCre + OE vector vs. Off target + OE vector, *P = 0.0274. G Mice injected with control vector and pAOV-CAMKIIα-GFP-2A-Cre showed no difference in NORT discrimination index (P = 0.9623, Mann–Whitney U test) To investigate the behavioral consequence of EB2 deletion in vGAT + neurons, we conducted a series of tests on EB2-vGATCre mice and their littermates (EB2-flox mice). In the novel object recognition (NOR) test, EB2-vGATCre mice exhibited pronounced cognitive impairments. Unlike EB2-flox mice, neither adolescent nor adult EB2-vGATCre mice preferred the novel object compared over the familiar one, as indicated by the reduced exploration. The impairment was quantified using as a preference index, with EB2-vGATCre mice showing significantly lower novelty preference indices for duration and frequency compared to EB2-flox mice (Fig. [58]1C). Similar cognitive deficits were observed in EB2-flox Cre mice and EB2^lacZ/+ mice (Fig. [59]1D, E). Additionally, mice with EB2 ablation in vGAT + neurons showed mild deficits in anxiety-like and depression-like behaviors (Additional file [60]1: Fig. S2A, B), but no significant abnormalities in prepulse inhibition (PPI), or compulsive behavior paradigms (Additional file [61]1: Fig. S2C, D). Numerous psychiatric and neurological conditions are associated with cognitive deficits linked to PFC functions [[62]11]. Inspired by these studies, we investigated the involvement of the PFC in the abnormal cognitive behaviors observed in EB2 mutants. We bilaterally microinjected a Cre-dependent AAV vector (pAAV-EF1a-DIO-Efnb2-2A-EGFP) into the mPFC of mice to specifically overexpress EB2 in the vGAT Cre-expressing neurons of EB2-vGATCre mice. Control animals received viruses expressing only EGFP (pAAV-EF1α-DIO-EGFP). Three weeks after virus injection, we validated the overexpression in the injected animals and performed NOR behavioral assays. EB2-vGATCre mice with EB2 overexpression in the PFC showed significant restoration of cognitive impairments (Fig. [63]1F). To further confirm whether EB2 contribute to cognitive impairment specifically though its absence in inhibitory neurons, we use a viral assay to knock out EB2 in excitatory neurons in the PFC, followed by cognitive behavioral studies. The results showed that knockout of EB2 in CamKIIα + excitatory neurons did not impair cognitive function (Fig. [64]1G). Together, these findings demonstrate that deletion of EB2 in vGAT + neurons impaired recognition memory. Impaired paired-pulse ratio (PPR) and altered PFC-thalamic connectivity from EB2 deletion in PFC inhibitory neurons EB2 deletion from vGAT + neurons disturbed local excitatory and inhibitory synaptic formation and transmission [[65]8, [66]10]. To evaluate functional changes in the PFC neural network, we performed microelectrode array recordings (Fig. [67]2A) in mice with specific EB2 knockout in inhibitory neurons. As shown in Fig. [68]2B and C, EB2-vGATCre mice exhibited significantly reduced activity across the entire population of cells compared to EB2-flox mice, with a progressive decrease from proximal to distal regions relative to the stimulation electrode point. In Fig. [69]2D (upper panels), the slope of field excitatory postsynaptic potentials (fEPSPs) showed a notable alteration in the EB2-vGATCre group of mice. PPR values were calculated at graded stimulus intensities of 10–70 μA. A reduction in paired-pulse ratio was observed in EB2-vGATCre mice (Fig. [70]2D, lower panels), indicating impaired short-term synaptic plasticity due to altered presynaptic vesicle release probability. Fig. 2. [71]Fig. 2 [72]Open in a new tab Impaired synaptic plasticity in mice with EB2 ablation in vGAT + neurons. A Schematic diagram showing the location of a MED64 probe placed on the coronal PFC slice (left) and the arrangement of the 8 × 8 recording array. Light microscopy photograph showing the location of the MED64 probe relative to the PFC cortex. The red dot indicates the stimulation site. B At various stimulation intensities (20, 40, 70 μA), the response intensity around the stimulation point (black) is shown (each square is 100 μm per side). The bar on the right indicates the color changes in response intensity. C Sample traces of evoked potentials in both groups of mice. D Significant decrease in fEPSP at positions 100 μm above, below, and to the left of the stimulation point (the top row). Upper 100 μm: two-way ANOVA, interaction, F(8,72) = 1.416, P = 0.2045, row factor, F(8,72) = 8.936, P < 0.0001, column factor, F(1,72) = 49.26, P < 0.0001, Sidak’s multiple comparisons test, 50 μA, 60 μA, 70 μA, **P < 0.01; left 100 μm: two-way ANOVA, interaction, F(8,72) = 1.584, P = 0.1447, row factor, F(8,72) = 12.26, P < 0.0001, column factor, F(1,72) = 78.53, P < 0.0001, Sidak’s multiple comparisons test, 40 μA, 50 μA, 60 μA, **P < 0.01, 70 μA, ***P < 0.001; lower 100 μm: two-way ANOVA, interaction, F(8,72) = 2.891, P = 0.0075, row factor, F(8,72) = 8.489, P < 0.0001, column factor, F(1,72) = 59.16, P < 0.0001, Sidak’s multiple comparisons test, 40 μA, *P < 0.05, 50 μA, ****P < 0.0001, 70 μA, ***P < 0.001. At positions 100 μm above, below, and to the left of the stimulation point, there is a significant decrease in the paired-pulse ratio of fEPSP (the bottom row). Upper 100 μm: two-way ANOVA, interaction, F(8,72) = 0.6037, P = 0.7689, row factor, F(8,72) = 0.7938, P = 0.6098, column factor, F(1,72) = 61.17, P < 0.0001, Sidak’s multiple comparisons test, 25 μA *P = 0.0388, 30 μA *P = 0.0243, 40 μA, *P = 0.0175, 60 μA, *P = 0.0421, 70 μA, *P = 0.0139; left 100 μm: two-way ANOVA, interaction, F(8,72) = 0.4780, P = 0.8678, row factor, F(8,72) = 0.3884, P = 0.9234, column factor, F(1,72) = 41.39, P < 0.0001, Sidak’s multiple comparisons test, 25 μA, *P = 0.0495, 40 μA, *P = 0.0211; lower 100 μm: two-way ANOVA, interaction, F(8,72) = 0.9763, P = 0.4618, row factor, F(8,72) = 1.159, P = 0.3359, column factor, F(1,72) = 14.1, P = 0.0004, Sidak’s multiple comparisons test To investigate the impact of EB2 deletion on PFC neural circuitry of the at a systemic level, we used functional ultrasound imaging (fUSi), which accurately identifies the PFC as a major connectivity hub. Most ROI pairs were shown in Fig. [73]3A, and the heatmap presents level variations across ROI pairs for both groups of mice (Fig. [74]3B). Connectivity between the infralimbic PFC and the RNT was significantly impaired in the resting state in EB2-vGATCre mice (Fig. [75]3C). These findings suggest that EB2 deletion in inhibitory neurons profoundly disrupts local PFC function and connectivity with related brain regions, such as the RNT, potentially contributing the cognitive changes observed in EB2-vGATCre mice. Fig. 3. [76]Fig. 3 [77]Open in a new tab Functional connectivity changes between EB2-vGATCre and EB2-flox group of mice. A Upper panel: head fixation and skull thinning. Lower panel: 3D registration enabled linear probe positioning (gray bar) through the PFC. Right panel: anatomical delineations derived from the Allen Common Coordinate Framework overlaid on a representative fUS Doppler image show coverage of ROIs. B Heatmap of Pearson correlation coefficient between selected ROIs in both groups of mice. C Differences in functional connectivity z-values between EB2-vGATCre and EB2-flox group of mice for RNT-IA (Mann–Whitney U test, **P = 0.0020). N = 5 per group Cellular and transcriptional alterations in the PFC of EB2-vGATCre and EB2-flox mice via snRNA-seq Neural activity induces widespread transcriptional changes in neurons, resulting in structural and/or functional changes [[78]12]. To investigate cell-type-specific changes linked to cognitive abnormalities and impaired neural connectivity in the PFC of EB2-vGATCre mice, we performed snRNA-seq. A total of 11,308 single nuclei were sequenced. After quality filtering (Methods, Additional file [79]1: Figs. S3A, S4A–C), 11,090 high-quality nuclei were obtained, including 5336 cells from EB2-flox mouse and 5754 from EB2-vGATCre mouse. These nuclei were grouped into 17 cell clusters (Additional file [80]1: Fig. S4D–H), which were further assigned to 9 major cell types based on cell-type-specific markers: excitatory neurons (Slc17a7^+), inhibitory neurons (Gad2^+), astrocytes (Gja1^+), microglia (C1qa^+), oligodendrocytes (Aspa^+), oligodendrocyte precursors (Pdgfra^+), smooth muscle cell (Uaca^+), endothelial (Flt1^+) and ependymal (Dynlrb2^+) cells (Fig. [81]4A, C, Additional file [82]1: Fig. S3B). Fig. 4. [83]Fig. 4 [84]Open in a new tab Widespread transcriptional changes in PFC cell types between EB2-flox and EB2-vGATCre mouse. A t-SNE plot showing the broad clustering of PFC cells (5336 from EB2-flox mouse PFC and 5754 from EB2-vGAT mouse PFC). B t-SNE plot showing the broad clustering of PFC cell (5336 from EB2-flox mouse PFC and 5754 from EB2-vGAT mouse PFC). t-SNE plot showing the distribution of the merged PFC cells from EB2-flox mouse (red) and EB2-vGATCre mouse (green). C t-SNE plot showing the expression of well-established marker genes in each broad cell cluster. D Bar plot showing the proportion of PFC cell types in EB2-flox and EB2-vGAT mice. E Strip chart shows the logFC of all detected genes (dots) across all 9 cell types. Genes in colored dots are significantly (Padj < 0.05 and |logFC|> 0.25) upregulated or downregulated. Genes in gray are not significantly changed. F Representative volcano plots showing altered gene expression that are significantly upregulated (red dots) or downregulated (blue dots) in the interneuron. G Overlap of differentially expressed genes between neurons and glia. H Expression changes of selected genes involved in synapse, learning and memory, and locomotory behavior. I Enriched pathways related to metabolism, cellular stress, synapse, and channel activity per cell type. The ancestor terms selected from the Gene Ontology and numbers of their associated child terms are listed on the left. On the right, the red and green colors represent numbers and percentages of upregulated and downregulated enriched pathways Comparison of cell composition in the PFC of EB2-vGATCre mouse with EB2-flox mouse revealed significant changes in neuronal populations (Fig. [85]4B, Additional file [86]2: Table S1). Consistent with the reported excitatory/inhibitory (E/I) ratio [[87]13], excitatory neurons constituted the largest cell class (54.31%) in the PFC of EB2-flox mouse, while inhibitory neurons accounted for 19.45%. Interestingly, EB2 deletion in vGAT^+ neurons disrupted the delicate E/I balance, resulting in comparable proportions of excitatory (37.81%) and inhibitory (34.93%) neurons, which might underlie cognitive deficits observed in these mice (Fig. [88]4D). The disruption of E/I homeostasis in the GABAergic/glutamatergic system is implicated in the pathophysiology of mental disorders [[89]14]. Comparison of gene expression in the PFC of EB2-vGATCre mouse and EB2-flox mouse revealed substantial changes across major cell types. Using an adjusted P value cutoff of < 0.05 and |logFC|> 0.25, we identified significantly alterations in the expression of 540 genes in at least one cell type (Additional file [90]2: Table S2). Excitatory and inhibitory neurons showed the most pronounced transcriptional changes (292 and 221 DEGs, respectively) (Fig. [91]4E, F and Additional file [92]1: Fig. S5A–H). Comparing DEGs across major cell clusters revealed that only four genes (Hbb-bs, Cd81, Apoe, Gm28928) out of 540 DEGs were shared by all neurons and glia, while 413 DEGs were cell-type-specific, indicating minimal overlap in expression profiles between cell types (Fig. [93]4G and Additional file [94]2: Table S3). Cell-type-specific transcriptional changes associated with distinct functional phenotypes were identified in this study. Among the 540 DEGs, many genes involved in cognition, locomotion, and synapse development showed altered expression across various cell types (Fig. [95]4H). For example, Apoe, which was upregulated in all neurons and glia clusters, is a well-known gene related to cognition and locomotory behavior. Differentially expressed genes such as Syp, Atp1a3, Ndrg4, and Pbx3, involved in synapse development, cognition, and locomotion, were upregulated in neuronal populations. In addition, Neurexin1 (Nrxn1) and Lsamp identified in glial cells were associated with locomotor functions and synaptic activity. A subset of genes, including Reln, Apoe, and Gpr88 in inhibitory neurons was implicated in both cognition and anxiety, both of which were observed in EB2-vGATCre mouse. To explore the biological impact of transcriptional changes across cell types, we performed gene set enrichment analysis and identified significant effects on relevant pathways. Using an adjusted P value cutoff of < 0.05, each cluster exhibited significantly affected pathways related to synaptic development, neurotransmitter, metabolism, etc. (Additional file [96]2: Table S4). Minimal overlap between cell clusters, except for Olig and OPC (Additional file [97]1: Fig. S6). Based on the hierarchical GO tree structure and previous literature [[98]15], we selected a set of GO ancestor terms associated with cellular stress, metabolism, synapse, and cation homeostasis, reflecting the E/I balance, and a hallmark of behavioral deficit in EB2-vGATCre mice (i.e., cognitive anomaly). We then filtered pathways linked to these terms (Fig. [99]4I). Pathways related to cellular stress, ATP metabolic process, cellular response to DNA damage, and proteolysis (e.g., protein catabolic process and protein ubiquitination) were upregulated across all cell clusters, indicating that EB2 deletion increased metabolism and cellular stress. Conversely, pathways involved in synapse signaling (e.g., synaptic signaling, chemical synaptic transmission, neurotransmitter transport) and synapse development (e.g., axon and dendrite development, cell junction organization, and cytoskeleton organization) were significantly downregulated, especially in glia and stromal cells. Notably, G protein-coupled receptor (GPCR) signaling and cation hemostasis pathways were primarily upregulated in neurons but downregulated in glia and stromal cells. Additionally, pathways related to amyloid processes (e.g., amyloid-beta/amyloid precursor protein formation, metabolism, and clearance) were broadly upregulated. Amyloid deposition in the brain, a hallmark of Alzheimer’s disease, is strongly linked to cognitive deficits. Collectively, these pathways highlight the roles of metabolism, cellular stress, synaptic signaling, and development in EB2 deletion-induced dysfunction. Molecularly distinct neuronal subtypes in the PFC following EB2 deletion Many studies have reported distinct functional subpopulations within the same neuron types, classified by morphology, anatomical location, electrical properties, histological features, and transcriptomic profiles [[100]13]. Consistently, we found that inhibitory and excitatory neurons in the PFC comprise distinct cell subtypes with EB2 deletion in vGAT + neurons causing significant changes in cellular composition and transcriptomic profiles. We observed a substantial increase in inhibitory neurons in the PFC of EB2-vGATCre mouse (Additional file [101]1: Fig. S7). Re-clustering these inhibitory neurons revealed 12 transcriptionally distinct subtypes (Fig. [102]5A), each identified by unique or combinational marker expression. Some subtypes corresponded to well-known inhibitory neuron populations, such as Pvalb^+ (Int_2), Sst^+ (Int_5), and Vip^+ (Int_7) (Fig. [103]5B and Additional file [104]1: Fig. S8A and B), whose expression in the PFC was validated using in situ hybridization (Allen Brain Atlas; Additional file [105]1: Fig. S8C). Additionally, we identified novel inhibitory subpopulations, i.e., Int_8, 10 (Igfbpl1^+, with or without Atad2^+), Int_0, 1, 3 (Rgs9^+, with or without Mhrt^+/Sh3rf2^+), Int_6 (Cpa6^+), Int_4, 9 (Ndst4^+, with or without Gfra1^+), and Int_11 (Atad2^+), which did not belong to the aforementioned populations. Fig. 5. [106]Fig. 5 [107]Open in a new tab Inhibitory neuronal subclusters. A t-SNE plot showing that inhibitory neurons of PFC can be classified into 12 subtypes based on their transcriptome (left), and the distribution of the merged cells from EB2-flox (1038 cells, red) and EB2-vGAT (2010 cells, green) PFC cells (right). B t-SNE plots highlight marker genes for inhibitory neuronal subclusters. C Enrichment map network of GO pathways impacted in Int_6 based on marker genes. Each node represents a GO term and each edge represents the overlap between two GO terms. D Heatmap of the mean value of AUCell scores of expression regulation by transcription factors, as estimated using SCENIC, per inhibitory neuronal subcluster. E t-SNE plots of inhibitory neurons, color-coded for expression of Pbx1 and Meis1 (up), for AUC of the estimated regulon activity of Pbx1 and Meis1 (bottom), corresponding to the degree of expression regulation of their target genes. F Strip chart shows logFC of all detected genes (dots) across all 12 subclusters. Genes in colored dots are significantly (Padj < 0.05 and |logFC|> 0.25) upregulated or downregulated. Genes in gray are not significantly changed. G Heatmap showing the functional GO pathways impacted in many of subclusters based on gene expression changes between EB2-flox and EB2-vGAT mice Surprisingly, we discovered that Int_6 was a cell population detected exclusively in EB2-vGATCre (Fig. [108]5A, Additional file [109]1: Fig. S8D). RNAscope validation confirmed the emergence of int_6 following EB2 deletion (Fig. [110]6A and B). Functional enrichment analysis of its marker genes revealed that Int_6 is associated with functional categories including membrane signaling, cation hemostasis, synaptic structure and development, synaptic signaling/neurotransmission, and metabolism (Fig. [111]5C). These pathways collectively support functions critical for cognition (e.g., learning or memory) and locomotory behavior. Fig. 6. [112]Fig. 6 [113]Open in a new tab Validation of gene expression changes in PFC of EB2-vGATCre group of mice. A Multi-channel FISH detecting subtypes within a novel inhibitory neuronal subpopulation in PFC (CPA6 +, arrow, and CPA6 −, arrowhead within vGAT neurons: enlarged view of single cells in box area shown in side panel). B Violin plots with boxplots overlaid (right) showing the quantification of the RNAscope data (data presents differential expression of CPA6 in vGAT + neurons (n = 82 cells from 4 brains of EB2-flox mice, n = 442 cells from 4 brains of EB2-vGATCre mice, ****P < 0.0001, Mann–Whitney U test)). C and D Ablation of EB2 induced the upregulation of the Pbx1 and Meis1 genes in vGAT + neurons (Pbx1 + cells; indicated by arrows), scale bar, 50 μm and 10 μm. E Enlarged view of single cells in box area shown, scale bar, 10 μm. F Violin plots with boxplots overlaid (right) showing the quantification of the RNAscope data (data presents median expression of Pbx1 in vGAT + neurons (n = 493 cells from 4 brains of EB2-flox mice, n = 540 cells from 4 brains of EB2-vGATCre mice, *P = 0.0102, Mann–Whitney U test) and G Meis1 + vGAT + neurons (n = 493 cells from 4 brains of EB2-flox mice, n = 564 cells from 4 brains of EB2-vGATCre mice; ****P < 0.0001, Mann–Whitney U test)) To identify transcription factors underlying differences in expression between Int_6 and other subtypes, we applied single-cell regulatory network inference and clustering (SCENIC). This analysis identified co-expressed transcription factors and their putative target genes, highlighting Pbx1, Meis1, Sp4, Sall1, and Nfatc1, with Pbx1 and Meis1 being prominent candidates (Fig. [114]5D and Additional file [115]1: Fig. S9). The expression of Pbx1 and Meis1 increased following EB2 deletion (Fig. [116]6C–G), and their target genes were highly upregulated in Int_6 (Fig. [117]5E). Pbx1 and Meis1 frequently form heterodimeric complexes that regulate the transcription of targeting genes. These finding suggest that Pbx1 and Meis1 likely function cooperatively to orchestrate precise gene expression patterns in Int_6, contributing to the abnormal behavior observed in the EB2-vGATCre mice. To investigate the contributions of other inhibitory neuron subtypes to abnormal behaviors, we compared gene expression between EB2-vGATCre and EB2-flox Mice and performed an enrichment analysis on these DEGs. Using the aforementioned cutoffs, we identified differentially expressed genes, with Int_0, Int_1, Int_2, Int_5, and Int_7 exhibiting the most significant transcriptional changes (Fig. [118]5F and Additional file [119]2: Table S5). Enrichment analysis revealed that these transcriptional changes impacted various functional pathways, including synapse-related processes (structure, neurotransmitter, long-term synaptic potential, synaptic plasticity, etc.), Rac GTPase, ERBB signaling, and amyloid protein across different inhibitory subtypes (Fig. [120]5G). These findings shed light on potential pathways through which EB2 deletion may contribute to behavioral abnormalities. Excitatory neurons were significantly affected by EB2 deletion. Consistent with a previous study [[121]16], they accounted for the largest cell population in the PFC, but their proportion decreased following EB2 deletion. Sub-clustering revealed 14 distinct subtypes of excitatory neurons (Additional file [122]1: Fig. S10A and B). Layer identity and specific functions of these subtypes were assessed using t-SNE overlaid with unique layer marker gene expression (Additional file [123]1: Fig. S10C and D). Comparison of gene expression between EB2-vGATCre and EB2-flox mice identified 1 (Exc-8) to 138 (Exc-0) DEGs across subtypes, with Exc_0, Exc_2, Exc_4, Exc_5, Exc_6, and Exc_11—distributed in different layers, showing the most transcriptional changes (Additional file [124]1: Fig. S10E and Additional file [125]2: Table S6). These findings suggest that EB2 deletion likely affects distinct cortical layers involved in neuronal projection, connections, and related functions, contributing to abnormal animal behaviors. Enrichment analysis of DEGs further indicated that pathways related to synapse function, nervous system development, amyloid protein, and so on might may play roles in the pathogenic mechanisms of EB2 deletion (Additional file [126]1: Fig. S10F). EB2 knockout in vGAT + neurons affects genes linked to neuropsychiatric diseases Many neuropsychiatric disorders are characterized by cognitive impairment, emotional symptoms, and locomotor abnormalities. The PFC plays an important role in cognition and emotion, and the pathophysiology of disorders like schizophrenia, Alzheimer’s disease, anxiety, and bipolar disorder. Genome-wide association studies (GWAS) have identified numerous candidate genes linked to these disorders, which show cell-type-specific expression in the PFC [[127]13]. Our study demonstrated that EB2 deletion resulted in novel inhibitory neuron subtypes, E/I imbalance, and cell-specific transcriptional changes in the PFC. Thus, we conclude that EB2 deletion may be closely associated with neuropsychiatric disorders. To gain preliminary insights into the potential role of EB2 in neuropsychiatric disorders, we compared the expression of GWAS candidate genes of 12 disorders/phenotypes between EB2-vGATCre mice and EB2-flox mice. These disorders/phenotypes include cognition, schizophrenia, bipolar disorder, depression, suicide, obsessive–compulsive disorder, autism, attention-deficit hyperactivity disorder, dementia, anxiety, and Parkinson’s disease. Many GWAS genes associated with these disorders showed significant expression changes after EB2 deletion, with inhibitory and excitatory neurons accounting for the largest portion of genes related to cognition and anxiety (Fig. [128]7A and B and Additional file [129]2: Table S7). Some candidate genes exhibited selective differential expression in distinct cell clusters, while others showed broad changes across neurons and glia (Fig. [130]7C: shows some examples). For example, the well-known cognition-related gene Apoe was upregulated to varying degrees among neurons and glia, while Nrg1 was specifically downregulated in excitatory neurons (Fig. [131]7C top). Similarly, the anxiety-related gene Elavl2 was downregulated only in inhibitory neurons, whereas Ptprd was downregulated in excitatory neurons and most glias (Fig. [132]7C down). Given the significant number of differentially expressed GWAS genes in inhibitory and excitatory neurons, we analyzed transcriptional changes within their subpopulations. Int_0 showed the highest number of differential candidate genes across most disorders/phenotypes in inhibitory neurons, while Exc_0, Exc_2, and Exc_6 identified the most genes in excitatory neurons. These transcriptional changes also exhibited cell-specific patterns (Additional file [133]2: Table S7, Additional file [134]1: Fig. S11). For instance, Erbb4, a gene critical for learning and memory, was upregulated in Int_8 and Exc_6, while Clu, an anxiety-related gene, was upregulated in Int_5, Int_7, and Exc_2. Finally, phenotype analysis of Int-6 marker genes and DEGs in neuron and glia revealed significant enrichment in pathways related to cognition and synapse development (Fig. [135]7D). Fig. 7. [136]Fig. 7 [137]Open in a new tab EB2 deletion links to phenotypes of mice and neuropsychiatric diseases. A Heatmap showing the number of differentially expressed GWAS candidate genes relevant to 12 disorders/phenotypes in the PFC cell types between EB2-flox and EB2-vGAT mice. B t-SNE plot indicating the number of differentially expressed cognition and anxiety GWAS candidates per cluster (shown in distinct colors). C Heatmap showing the representative differential expressed GWAS candidates relative to cognition (up) and anxiety (down) in PFC cell types. Cognition and anxiety are shown as examples. D The phenotypes are found to be associated to markers of Int_6 and DEGs of neurons and glia. The left panel shows the downregulated DEGs, and the right panel shows the upregulated DEGs. The Fisher’s exact test and Benjamini–Hochberg FDR correction were used for P values The cell-type-specific transcriptional changes in GWAS candidate genes suggested that EB2 deletion may lead to abnormal behaviors by altering the expression of risk genes, offering a cellular perspective on the pathological mechanisms. Discussion In this study, we manipulated endogenous EB2 expression in transgenic mice and found that EB2 loss disrupts several behavioral domains, including recognition memory and increased anxiety, highlighting its indispensable role in the brain. Restoration of EB2 after early brain development ameliorated cognition-related impairments. EB2 deletion in vGAT + neurons also impaired PPR and disrupted PFC-thalamic functional connectivity. To investigate these alterations at the cellular level, we conducted a single-cell investigation in EB2 knockout mice and their EB2-flox control littermates. Our study revealed cell population changes in response to EB2 deficiency in vGAT + neurons and identified previously uncharacterized cell types. Differentially expressed gene analysis showed that EB2 deficiency primarily affects genes involved in synapse organization and development, cognition, amyloid-beta formation, and locomotor behavior, overlapping with human GWAS candidate genes linked to cognition and anxiety. This underscores the relevance of our mice model to human disease. Overall, our investigation provides a comprehensive map of cell types, genes, and pathways altered in this synaptic deficiency model. Previous studies have highlighted the role of Ephrin Bs in regulating neuronal migration [[138]17]. Here, we demonstrate for the first time that the relative numbers of neuronal subtypes in the PFC change when EB2, the most abundantly expressed Ephrin B molecule, is specifically knocked out in inhibitory neurons. During early brain development, synapses form to guide the migration of newborn neurons to the neocortex [[139]18]. Disruption of EB2-mediated signaling significantly impairs neuronal migration, affecting brain maturation and population dynamics. We confirmed an increase in inhibitory neurons in EB2-vGATCre mice, leading to an imbalance in E/I signaling (Additional file [140]1: Fig. S10). PPR analysis revealed abnormalities in the PFC, further supporting disrupted E/I balance. In PPR, a larger second fEPSP indicates paired-pulse facilitation [[141]19], a fundamental form of short-term potentiation (STP) that we also observed. STP underpins higher cognitive functions, including associative learning, working memory, and short-term memory [[142]20]. In addition to abnormal electrophysiological signals in the local PFC region caused by EB2 knockout in vGAT + neurons, ultrasound functional imaging revealed disrupted functional connectivity between the PFC and the RNT, a region known to be functionally linked to the PFC. The midline/paramidline thalamus, which receives projections from the PFC, plays a crucial role in working memory, with PFC interneurons modulating its function [[143]21]. Thus, EB2 knockout in vGAT neurons may impair PFC and RNT connectivity, potentially contributing to the cognitive deficits observed in EB2 knockout mice. The synaptic balance between excitation and inhibition is a fundamental principle of cortical circuits, and disruptions in this balance are closely linked to cognitive impairments [[144]22]. Our results demonstrate that deleting EB2 in inhibitory neurons during early development, but not in adulthood, induces cognitive impairments. Evidence suggests that activating the EB2 signaling pathway can restore cognitive function [[145]23, [146]24]. Consistent with this, we found that targeted viral overexpression of EB2 in inhibitory neurons reversed cognitive impairments caused by its loss, while EB2 knockout in excitatory neurons does not cause deficits. Since early EB2 deficiency in inhibitory neurons disrupts synaptic balance and leads to cognitive and behavioral deficits, we used single-nucleus sequencing to identify affected cell subpopulations and functional pathways associated with differentially expressed genes. These findings provide insights into the cellular mechanisms linking EB2 deficiency, E/I imbalance, and altered cognitive behaviors. Our single-nucleus sequencing revealed that a novel inhibitory neuron subpopulation (Int_6) exclusively present in the EB2-vGATCre mice, representing a significant finding warranting further investigation. The emergence of this transcriptionally distinct subtype suggests that EB2 deletion in vGAT + neurons profoundly impact the cellular composition and transcriptional landscape of the PFC. Int_6 neurons are marked by the expression of carboxypeptidase A6 (CPA6), a member of the M14 metallocarboxypeptidase family, which is highly expressed in the adult mouse olfactory bulb but broadly expressed in the brain and other tissues during embryonic development [[147]25]. In EB2-vGATCre mice, CPA6 is widely observed in Int_6 neurons within the PFC, suggesting potential migration issues for CPA6-expressing neurons. Cognitive processing relies on the refinement of limbic circuitry during the first 2 weeks of life, with the olfactory bulb influencing limbic activity and communication during development [[148]26, [149]27]. The reduction of CPA6 in the olfactory bulb (Additional file [150]1: Fig. S12) and the presence of CPA + neurons in the adult PFC may contribute to the cognitive impairments caused by EB2 deletion in vGAT + neurons. Functional enrichment analysis indicated that Int_6 is involved in pathways related to membrane signaling, cation homeostasis, synaptic structure/development, and neurotransmission, suggesting a role in synaptic plasticity, neuronal migration, and E/I balance-processes critical for cognition and behavior. Furthermore, transcription factors Pbx1 and Meis1 emerged as key regulators of the Int_6 population. Evidence linked Pbx1 and Meis1 to mood instability and psychotic disorder [[151]28–[152]30], and Meis1 knockout mice exhibited impaired cognitive abilities [[153]31]. These findings collectively suggested that the synaptic protein EB2 modulates transcriptional networks governing Int-6’s unique gene expression patterns that are associated with cognitive impairment. Apart from the newly identified Int_6 population, DEG analysis revealed widespread transcriptional adaptations across subpopulations, primarily involving cognition, locomotion, and synaptic function. In excitatory neuronal subpopulations, we identified from layer 5/6 excitatory neurons in the PFC with significantly differentially expressed genes between EB2-vGATCre and EB2-flox mice. Dysfunctional projections from the hippocampal CA1 area to the PFC, which are associated with poor cognitive performance in mental disorders, preferentially target layer 5/6 pyramidal neurons and interneurons [[154]32–[155]34]. Thus, the DEGs in these excitatory subpopulations may underlie the cognitive impairments observed in EB2-vGATCre mice. While some pathways are cell-type-specific, common enriched pathways include synaptic signaling, neurotransmission, and cell junction assembly. These transcriptional alterations across diverse brain cell types, combined with disruptions in the E/I neuronal ratio, likely underlie the synaptic and circuit-level dysfunctions contributing to cognitive and behavioral deficits. Some DEGs were consistently changed across cell types, while others were neuron-specific. Notably, transcriptional adaptations in neurons affected common signaling pathways involved in neuronal development, neurotransmitter transport, and neuropsychological behaviors. These findings provide a foundation for investigating the molecular mechanisms underlying behavioral deficits, including cognitive impairments, and suggest possible changes in both neurons and supportive cells. Notably, several GWAS candidate genes linked to neuropsychiatric disorders, including cognition, schizophrenia, and anxiety, exhibited cell-type-specific expression changes upon EB2 deletion. This underscores the relevance of the EB2-vGATCre model to human disorders and suggests potential cellular mechanisms through which EB2 deletion contributes to abnormal behaviors by dysregulating risk gene expression across neuronal populations. Conclusions Overall, our study unveiled novel cell types, detailed transcriptional remodeling, and links to neuropsychiatric risk genes, paving the way for targeted investigations into circuit-specific mechanisms underlying EB2-mediated behavioral phenotypes. Further exploration of distinct neuronal subtypes like Int_6, their connectivity patterns, and the interplay between excitatory and inhibitory populations may provide crucial insights into the etiology and pathophysiology of cognitive and neuropsychiatric disorders. Methods Animals and behavioral assessment Mice were maintained in a mixed CD1/B6 genetic background. All experiments involving mice were conducted in accordance with National Institute of Health Guide for the Care and Use of Laboratory Animals and approved by Animal Care and Use Committee in Shanghai Jiao Tong University. Mice used in this analysis were previously described [[156]10]. They carried loxP-flanked alleles in Ephrin-B2 (EB2^loxP), and the Ai9 Rosa26-STOP-tdTomato Cre reporter strain was used to monitor Cre activity. Specifically, EB2-flox mice were crossed with Vgat-Cre mice [B6.FVB-Tg(Slc32a1-cre)2.1Hzo/FrkJ, provided by Prof. Huang Zhili, Fudan University]. Genotyping with WT-EB2, Mutant-EB2, and vGAT-Cre primers yielded products of 500 bp, 350 bp, and 400 bp, respectively. Genotypes were revealed only after analysis. Additionally, mice with loxP-flanked EB2 alleles were crossed with tamoxifen-inducible CreER^T2 mice [homozygous CreER^T2 (B6.129-Gt(ROSA)26Sor tm1(CreER^T2)tyj/J)], allowing EB2 deletion 5–7 days post-tamoxifen administration (Sigma T5648-5G). Ephrin-B2^lacZ/+ mice were also used to validate behavioral phenotypes. Mice were housed in standard conditions with a standard 12-h dark/light cycle (lights were on at AM 7:00 and off at PM 7:00), a room temperature of 24 ± 1 °C, and ad libitum access to food and water. Novel object recognition A plastic arena (44 cm × 44 cm × 44 cm) and objects with distinct shapes was used for examining novel object recognition. Mice were habituated to the arena for 5 min per day over 3 days without the test objects. Each mouse was initially placed into the box with two identical (familiar) objects for 10 min, and then returned to their home cage. One hour later, the mouse was reintroduced to the arena, now containing one familiar object and one novel object with a different shape, for 5 min. Object-related behaviors including the number of object contacts and the duration of contacts were recorded and analyzed by EthoVision XT 8.5. Novel object recognition task (NORT), [DI = (Novel Object Exploration Time/Total Exploration Time) − (Familiar Object Exploration Time/Total Exploration Time) × 100]. Elevated plus maze The elevated plus maze (EPM) apparatus was made of dark gray plastic and comprised two open arms (30*7*0.25 cm) opposed to two enclosed arms (30*7*15 cm) elevated 60 cm from the floor. Animals were placed in the central area facing an enclosed arm (test duration, 5 min). Digitized video recordings with EthoVision software (Noldus Information Technology) were used for behavioral analysis. The percentages of time spent in open arms and open-arm entries were evaluated. Tailing suspension test The mouse is suspended 50 cm above the ground, ensuring it cannot touch any surface. The test lasts 6 min, during which movements are recorded, and immobility is measured. Immobility is defined as the absence of any limb or body movement except those caused by respiration. The first 2 min serve as an acclimation period, with data collected during the last 4 min. The percentage of immobility time is calculated. Prepulse inhibition test The prepulse inhibition (PPI) test measures sensorimotor gating in mice by assessing their ability to inhibit a startle response when a weaker prepulse precedes a stronger startle-inducing stimulus. Mice are habituated to a sound-attenuated chamber equipped with a startle platform to detect movements. After establishing a baseline startle response to 120 dB noise bursts, prepulses (e.g., 75 dB, 80 dB, 85 dB) are presented 100 ms before the startle stimuli. Trials include startle stimuli alone, prepulse followed by startle, and control (background noise). Startle response amplitudes are recorded and PPI is calculated using the formula: PPI (%) = (1 − (startle response with prepulse/startle response alone)) × 100. Compulsive checking The protocol for assessing compulsive checking behavior in mice typically involves the following steps: Mice are habituated to an open field arena with several objects for exploration. During testing, specific objects or locations are designated as “checkpoints.” The mouse is placed in the arena and allowed to explore freely for a set period, usually 30–60 min. The number of returns to and interactions with the checkpoints is recorded. High frequency and repetitive returns to these checkpoints are indicative of compulsive checking behavior. This behavior can be further analyzed to evaluate the effects of pharmacological interventions or genetic modifications on compulsive behavior in mice. Immunohistochemistry Anesthetized mice were transcardially perfused with PBS followed by 4% paraformaldehyde (PFA). Brains were fixed overnight with 4% PFA and dehydrated in 30% sucrose at 4 °C. Brains were sectioned coronally in 30 µm slices. Slices between bregma 1.98 to 2.58 mm were collected. Sections were washed with PBS, blocked with 10% BSA in 0.1% Triton-X-100 (CAS No. 9002–93-1) for 2 h at room temperature (RT), and incubated with primary antibodies of anti-rabbit GABA, anti-mouse GAD65/67, anti-goat Ephrin-B2, anti-rabbit parvalbumin (1:2000), anti-vGLUT2, and anti-goat CamkII at 4 °C overnight. Secondary antibodies were used as follows: Alexa 647 donkey anti-mouse IgG (1:1000, Jackson lab, Shanghai, China, RRID: AB_2340863) and Alexa 488 donkey anti-rabbit (1:1000, Jackson lab, RRID: AB_2313584) for 2 h at room temperature. Cells were counted in every sixth section. Images were captured using Thunder image systems (Leica). RNA scope In situ RNA analysis was conducted using the RNAscope® Multiplex Fluorescent Reagent Kit v2 Assay (Advanced Cell Diagnostics, Hayward, CA, USA, Cat. No. 323100), following the manufacturer’s guidelines. Briefly, paraffin-embedded mouse brains were sectioned into 5 µm slices and mounted on Superfrost® plus slides (Thermo Fisher Scientific, Waltham, MA, USA, Cat. No. 4951PLUS4). After pretreatment with hydrogen peroxide, target retrieval, and protease, the slices were hybridized with probes for Pbx1 (Cat. No. 550901-C2), Meis1 (Cat. No. 445231), vGAT (Cat. No. 400951), and CAP6 (Cat. No. 311491) using a HybEz hybridization system. Following amplification steps, the sections were stained with DAPI and mounted using Prolong Gold® Antifade Mountant (Thermo Fisher Scientific, Cat. No. [157]P10144). Imaging was performed with FV1200 confocal microscopes (Olympus, Tokyo, Japan). Recording multi-channel field potentials in medial frontal cortical slices The method for recording multi-channel field potentials from medial frontal cortical slices follows established protocols [[158]35]. After incubation, a slice is placed on the MED64 probe ensuring the insular cortex is fully covered by the recording chamber on an inverted microscope (CKX41; Olympus). Placement follows an anatomical atlas for precise localization. To stabilize the slice, a fine mesh anchor (Warner Instruments, Harvard Apparatus) is used during recording. The slice is continuously perfused with oxygenated extracellular solution (95% O2/5% CO2) containing ACSF: 125 mM NaCl, 2 mM CaCl2, 1.25 mM NaH2PO4, 3 mM KCl, 26 mM NaHCO3, 1 mM MgCl2, and 10 mM D‐glucose, pH 7.3, at a flow rate of 2 ml/min, maintained at 30 ± 1 °C using a peristaltic pump (Minipuls 3; Gilson). After a 20-min recovery period, a single microelectrode from the 64 available on the probe is selected for stimulation, identified via a charge-coupled device camera (DP70; Olympus) linked to the inverted microscope. Monopolar, biphasic constant-current pulses (0.2 ms duration) are generated by MOBIUS software (Panasonic Alpha-Med Sciences) and applied at 0.008 Hz to the deep layer of the insular slice. Field excitatory postsynaptic potentials (fEPSPs) evoked at other sites are amplified using a 64-channel amplifier, displayed on a monitor, and stored for analysis. Stimulation intensities are sequentially adjusted from 15 to 70 µA in increments of 5 µA. fUS headframe attachment surgery To conduct fUS recordings on awake mice, we developed a lightweight head fixation device compatible with fUS. The design incorporates a standard mounting headband with mouse support brackets. Headframe placement procedure: Mice were anesthetized with isoflurane gas (1–1.5%). During surgery, mice breathed a mixture of air and supplemental oxygen, with body temperature maintained at 36.5 °C using a feedback-controlled heating pad and monitored via a rectal probe. The scalp was carefully incised, using a specialized cranial drill and grinder. Care was taken to avoid any abrasion to the skull and the head fixation device was securely attached to the skull and surrounding muscles (Super-Bond C&B, LOCTTLF 454). Afterward, a protective layer was applied to safeguard the skull. Post-surgery, mice were given a 3-day recovery period with careful monitoring and pain management as needed. Starting on day 3, they were habituated to head fixation, with session duration gradually increased from 5 min on the first day to 60 min by day 6 [[159]24]. This acclimation step was critical to reduce stress and ensure consistent fUS measurements. fUS imaging fUS hardware and software (Iconeus, Paris, France) were used for imaging sessions. A linear ultrasonic probe (15 MHz central frequency) with 128 piezo-electric was connected to an ultrafast ultrasound scanner (Iconeus One—128 channels). The animal was positioned in a stereotaxic frame, and we pre-trained the mice to adapt to a conscious state of restraint beforehand, in preparation for acquiring resting-state fUS imaging. After animal fixation, the scalp was covered with an isotonic coupling gel and the ultrasonic probe was lowered to 1 mm from the scalp for complete immersion into the gel. The probe’s location was determined using Iconeus Studio software to target an oblique plane, which encompassed the neuroanatomical regions of interest (ROIs) associated with the PFC. Twenty minutes of Power Doppler images were captured for each session. These normalized values were used to generate bar graphs and conduct statistical analyses between the two groups. Brain tissue dissociation and nucleus isolation Two mice from EB2-flox and EB2-vGATCre group respectively were anesthetized with isoflurane and rapidly decapitated. Brains were extracted and transferred into ice-cold Hibernate A/B27 medium. The PFC was removed from slices and isolated intact nucleus suspension. The nucleus was isolated and purified as previously described with some modifications [[160]36]. Briefly, the frozen tissue was homogenized in NLB buffer containing 25 mM sucrose, 10 mM Tris–HCl, 3 mM MgAc2, 0.1% Triton X-100 (Sigma-Aldrich), 0.1 mM EDTA, and 0.2 U/μL RNase Inhibitor (Takara). Various concentrations of sucrose were used to purify the nuclei. Nuclei counts were determined by PI staining. Finally, the concentration was adjusted to 1000 nuclei/μL for snRNA-Seq. Single-nucleus RNA sequencing The snRNA-Seq libraries were generated using the 10X Genomics Chromium Controller Instrument and Chromium Single Cell 3′ V3 Reagent Kits (10X Genomics, Pleasanton, CA). Briefly, nuclei were concentrated to 1000 nuclei/μL and approximately 10,000 nuclei were loaded into each channel to generate single-nuclei gel bead-in-emulsions (GEMs), which results into expected mRNA barcoding of ~ 6000 single nuclei for each sample. After the reverse transcription (RT) step, GEMs were broken and barcoded cDNA was purified and amplified. The amplified barcoded cDNA was fragmented, A-tailed, ligated with adaptors, and index PCR amplified. The final libraries were quantified using the Qubit High Sensitivity DNA assay (Thermo Fisher Scientific) and the size distribution of the libraries were determined using a High Sensitivity DNA chip on a Bioanalyzer 2200 (Agilent). All libraries were sequenced by Novaseq 6000 (Illumina, San Diego, CA) on a 150-bp paired-end run. Single-nucleus gene expression quantification and determination of cell-type identity Raw data processing were performed by NovelBio Bio-Pharm Technology Co., Ltd. with NovelBrain Cloud Analysis Platform. First, we applied fastp with default parameter to filter adaptor sequence and remove the low-quality reads to achieve the clean data [[161]37]. The feature-barcode matrices were then obtained by aligning reads to the mouse genome (GRCm38 ensembl: version 92) using CellRanger v3.1.0. We performed downsampling analysis among samples based on the mapped barcoded reads per nucleus of each sample and obtained the aggregated matrix. Nuclei with fewer than 200 or more than 10,000 expressed genes, or with more than 20% mitochondria UMI rate, were removed. Mitochondria genes were also removed in the expression table. For the remaining 11,090 nuclei, the Seurat R package (v.2.3.4) [[162]38] was used for cell normalization and regression based on UMI counts and mitochondrial rate, generating scaled data. Highly variable genes were identified by FindVariableGenes function with default parameters. The top 2000 highly variable genes were used to calculate principal components (PCs) and the top 10 PCs were selected for t-SNE construction. Utilizing graph-based cluster method, unsupervised cell cluster was generated based on the top 10 PCs. Marker genes were calculated with FindAllMarkers function using the Wilcoxon rank-sum test and the following criteria: 1. lnFC > 0.25; 2. P value < 0.05; 3. min.pct > 0.1. Clusters were annotated based on well-known marker genes of major brain cell types as reported in previous studies [[163]39]. To identify subclusters within neurons, inhibitory and excitatory neurons were reclustered separately using similar methods and parameters. Differential gene expression analysis Differential gene expression analysis between EB2-flox and EB2-vGATCre mouse was performed using the FindMarker function in Seurat. The analysis generated logFC (natural log of the FCs), P values, pct (percentage of cells where the gene is detected in the case or control group), and adjusted P values (based on Bonferroni correction using all genes in the dataset). Genes with |logFC|> 0.25 and adjusted P value < 0.05 were considered significantly changed. DEGs were shown in strip charts and volcano plots, which were generated using ggplot2 R package [[164]40]. Overlapping DEGs across cell clusters was displayed in Venn diagrams created with jveen [[165]41]. Pathway analysis Pathway enrichment analysis was performed to elucidate the biological implications of differentially expressed genes. The clusterProfiler package [[166]42] in R was used to identify the Gene Ontology (GO) terms in the biological process, considering only pathways with an adjusted P value < 0.05 as significant. The GO.db package ([167]https://web.mit.edu/~r/current/arch/i386_linux26/lib/R/library/GO .db/html/00Index.html) was used to explore the ancestor-child relationship between representative pathways (ancestor pathways), associated with cognition and anxiety [[168]15], and significant pathways (child pathways). SCENIC analysis To assess transcription factor regulation strength, we used the single-cell regulatory network inference and clustering (pySCENIC, v0.9.5) workflow with the 20-thousand motifs database for RcisTarget and GRNboost [[169]43]. Association of GWAS genes to cell clusters To investigate the relationship between EB2 and neuropsychiatric diseases, we downloaded GWAS candidate genes for 12 selected diseases from the NHGRI-EBI GWAS catalog (v. 1.0.2) [[170]44] and compared them with differentially expressed genes in each cluster. Phenotype analyses were performed on Int-6 marker genes and DEGs of neurons and glia using model organism Phenotype Enrichment Analysis (modPhEA, [171]https://evol.nhri.org.tw/phenome2/) [[172]45]. Statistical analysis Statistical analysis was performed using a two-sample Student’s t-test or one-way/two-way ANOVA, depending on the experimental design. For multiple group comparisons, Bonferroni’s post hoc test was used. Results are expressed as the mean ± standard error of the mean (SEM). Prior to analysis, data were log-transformed and tested for normality using the Shapiro–Wilk test. Nonparametric tests were applied for data that did not follow to a Gaussian distribution. Statistical significance was defined as P < 0.05 (two-sided). Analyses were conducted using GraphPad Prism 8.0.2., and the figures were prepared with CorelDraw 2019. Supplementary Information [173]12915_2025_2333_MOESM1_ESM.zip^ (21.7MB, zip) Additional file 1: Figures S1–S12. Fig. S1 Immunostaining of EphrinB2 and GAD65/67 in PFC slices from EB2-flox and EB2-vGATCre mice. Fig. S2 Behavioral assessments showing mild anxiety- and depression-like phenotypes in mutant mice, with no schizophrenia- or OCD-like deficits. Fig. S3 Overview of the experimental workflow and transcriptome-based classification of PFC cells. Fig. S4 Quality control and initial data processing of single-cell RNA-seq from mouse PFC. Fig. S5 Cell-type-specific transcriptional changes in PFC between EB2-flox and EB2-vGATCre mice. Fig. S6 Heatmap of enriched GO pathways across PFC cell clusters. Fig. S7 Increased vGAT mRNA expression in PFC of EB2-vGATCre mice visualized by RNAscope. Fig. S8 Classification of inhibitory neuronal subtypes in PFC based on transcriptomic profiles and in situ validation. Fig. S9 SCENIC-based TF motif activity (AUC scores) in PFC inhibitory neurons. Fig. S10 Identification and characterization of excitatory neuronal subclusters in PFC, including layer-specific markers and pathway analysis. Fig. S11 Number of differentially expressed GWAS candidate genes in each PFC neuronal subcluster. Fig. S12 RNAscope showing reduced CPA6 mRNA in olfactory bulb of EB2-vGATCre mice. [174]12915_2025_2333_MOESM2_ESM.zip^ (20.3MB, zip) Additional file 2: Tables S1–S7. Table S1 Number of cells detected by cell clusters. Table S2 Differential gene expression results for major cell types between EB2-vGATCre and EB2-flox. Table S3 Matrix of DEGs across neurons and glias. Table S4 GO pathways across major cell types. Table S5 Differential gene expression data for inhibitory neuronal subclusters between EB2-vGATCre and EB2-flox. Table S6 Differential gene expression data for excitatory neuronal subclusters between EB2-vGATCre and EB2-flox. Table S7 Differential expression of GWAS candidate genes. Acknowledgements