Abstract Sleep loss is a key trigger for a manic episode of bipolar disorder (BD), but the underlying microglial and molecular mechanisms remain unclear. Sleep loss induces microglial and inflammatory responses. Microglia, resident macrophages in the central nervous system, regulate synaptic pruning by engulfing dendritic spines. Here, we introduce a modified paradoxical sleep deprivation (SD) paradigm as a BD mouse model. After intermittent 16-h daily SD for 4 days, the mice showed mania-like behavior, reduced cytokine/chemokine production, mitochondrial damage, microglial loss, decreased synaptic engulfment by microglia, and synaptic gain. Single-nucleus RNA sequencing (snRNA-seq) revealed cell-type-specific inflammation- and synapse-related gene expression profiles in the prefrontal cortex (PFC) and hippocampus of SD-treated male mice. Interestingly, much more differentially expressed genes were observed in SD-treated female versus male mouse brain, especially in the PFC. Pharmacological depletion of microglia by colony stimulating factor-1 receptor (CSF1R) inhibitor PLX3397 blocked SD-induced inflammation-related and senescence-associated abnormalities in a sex-specific manner. Microglial elimination reversed SD-induced synapse gain and mania-like behavior in males but not in females. However, microglial inhibition by minocycline had no effect on SD-induced behaviors in a sex-independent manner. These findings demonstrate that microglia-mediated neuroinflammation and synaptic pruning contribute to SD-induced mania-like behavior in a mouse model of BD in a sex-specific manner. graphic file with name 41398_2025_3525_Figa_HTML.jpg Subject terms: Molecular neuroscience, Bipolar disorder Introduction People with insomnia or sleep loss often experience mental issues with the increasing pressure of study, work, and life [[40]1, [41]2]. Sleep loss can trigger a mood switch from depression to mania in bipolar disorder (BD) [[42]3]. BD significantly affects patients’ quality of work, study, and life and increases individual’s social burdens [[43]4]. Patients with BD often experience sleep disturbances. During manic episodes, most BD patients (69–99%) experience a decrease in sleep demand [[44]3]. Transcriptome sequencing data from peripheral blood samples from BD patients indicate that sleep deprivation (SD) causes a dysregulated inflammatory response [[45]5] and an increase in the levels of inflammatory factors [[46]6]. In addition, abnormal expression of inflammation-related genes has been found in microglia-like cells differentiated from monocytes derived from peripheral blood of BD donors [[47]7]. A common BD animal model of rapid eye movement (REM) SD is established by a modified multi-platform water method, which can induce manic-like behavior in rodents [[48]8, [49]9]. Four days of continuous and prolonged SD exposure triggers cytokine-storm-like systemic inflammation using a “curling prevention by water” paradigm [[50]10]. Acute 48-h SD leads to an increase in the density of microglia (the resident innate immune cells of the central nervous system (CNS)) in brain regions in young mice [[51]11]. Microglia regulate synaptic density by engulfing dendritic spines [[52]12]. Abnormal synaptic transmission and synaptic pruning are observed in sleep-deprived mice [[53]13, [54]14]. Sleep fragmentation causes an increase in colony stimulating factor-1 (CSF1) levels [[55]15]. CSF1 receptor (CSF1R) in the brain is mainly expressed in microglia and regulates the survival and distribution of microglia [[56]16]. CSF1R inhibitor reduces microglia and complement C1qb, avoiding neuronal damage and synaptic pruning abnormalities [[57]17]. However, the detailed mechanisms underlying microglia-mediated inflammation and synaptic pruning in BD have not been elucidated. Although some studies have suggested sex-dependent effects of SD on emotional behaviors [[58]3, [59]18], sex-dependent gene expression levels in the brain and treatment response in BD remain unresolved. Here, we used a modified SD paradigm as a BD model to investigate mania-like behavior in mice. As a consequence of intermittent 16-h daily SD for 4 days, mice exhibited inflammation- and synapse-related gene expression profiles using transcriptome sequencing. Chronic SD exposure reduced cytokine/chemokine levels in the plasma with a multicytokine assay. Additionally, single-nucleus RNA sequencing (snRNA-seq) revealed cell-type-specific changes. Microglial loss and synapse gain were found after SD using immunohistochemistry, Golgi staining, and transmission electron microscopy. Finally, pharmacological depletion of microglia by CSF1R inhibitor inhibited SD-induced mania-like behavior in a sex-specific manner and reversed SD-induced synapse gain in male mice. Methods and materials Animals Adult male and female C57BL/6 mice (2 months old) were obtained from GemPharmatech (Chengdu, China). CX3CR1^CreERT2 (JAX#020940) and Ai9 (JAX#007909) mice were obtained from the Jackson Laboratory. CX3CR1^CreERT2:: Ai9 mice were used for this experiment. CX3CR1^CreERT2:: Ai9 mice were intraperitoneally injected with tamoxifen (100 mg/kg) once daily for 4 days at five to six weeks old to induce microglia to constantly express tdTomato. The animals were housed in animal facilities with food and water available ad libitum and were maintained on a normal 12-h light/dark cycle (lights on at 7:00 AM, lights off at 7:00 PM). Ethics approval and consent to participate All procedures were performed in accordance with the Institutional Animal Care and Use Committee of Sichuan University (Approval Number: 20230504005). All experimental procedures were in compliance with the ARRIVE guidelines. The guidelines used in the present study were made to minimize animal suffering as well as the number of animals used. Drug administration To deplete microglia in the brain, mice were injected with the CSF1R inhibitor PLX3397 (pexidartinib, PLX, MedChemExpress, Cat# HY-16749, USA) dissolved in 5% dimethyl sulfoxide (DMSO), 10% sulfobutylether-β-cyclodextrin (SBE-β-CD) based on previous studies [[60]19]. PLX was administered (i.p. injection) at a dose of 50 mg/kg twice a day for 8 days (4-day treatment with PLX, 3-day withdrawal, and 4-day treatment with PLX). The control mice received vehicle solution (5% DMSO/10% SBE-β-CD/85% saline, vol/vol/vol). Minocycline (MCL, MedChemExpress, Cat# HY-17412) was administered orally in drinking water at 0.5 mg/mL (delivering daily doses of approximately 100 mg/kg) for 14 days based on previous studies [[61]20]. Mice were intraperitoneally treated with valproic acid (VPA) at a dose of 200 mg/kg twice a day for 7 days. Administration of PLX, MCL, or VPA was stopped 12 h before behavioral testing. To efficiently induce the CreER-dependent recombination, tamoxifen (100 mg/kg, MedChemExpress, Cat# HY-13757A) dissolved in corn oil was intraperitoneally administered for 4 consecutive days as previously described [[62]21]. Sleep deprivation procedure All SD-treated mice were first habituated to environmental conditions and submitted to rapid eye movement (REM) sleep deprivation stress in a Plexiglas box for 16 h daily (starting time: 6:00 p.m., ending time: 10:00 a.m. the next day) for 4 days. The SD period of 6 p.m. to 10 a.m. was adapted based on previous studies [[63]22–[64]24]. Paradoxical SD stress was conducted using a modified multiple-platform method, according to the protocol described [[65]25]. Briefly, the Plexiglas box contained seven visible escape platforms (3 cm in diameter, 1 cm above the water surface) surrounded by water. Mice were allowed to freely move on the platform with food and water available ad libitum. The control mice were kept in their home cages in the same experimental room. Behavioral analysis All behavioral measurements were conducted by trained experimenters in a blinded manner. All mice were habituated to the test room for at least 30 min prior to behavioral testing. Behavioral testing was conducted under dim light between 9:00 a.m. and 05:00 p.m. according to the protocol described [[66]20, [67]26]. Mice were tested successively in the open-field test (OFT) and elevated plus-maze test (EPM). The recorded videos were analyzed using EthoVision (12.0) tracking software (Noldus, Netherlands). One day after behavioral testing (from 01:00 p.m. to 05:00 p.m. of the next day), the mice were randomly selected and sacrificed. In tail suspension test (TST), the mice were suspended by their tails using an elastic band based on a previously described procedure [[68]27]. A video recording device captured the behavior of each mouse throughout the 6-min testing period. Immobility time, defined as the time with no body movement, was quantified during the last 4 min of the 6-min test session. In the novel object recognition test (NOR), the test was conducted in an open field arena (50 × 50 × 50 cm) based on a previously described procedure [[69]28]. During the familiarization phase, mice were allowed to explore two identical objects positioned 20 cm apart for 5 min. Five hours later, one of the objects was replaced with a novel object during the test phase, and mice were allowed to explore both objects for 5 min. The time spent exploring each object was recorded, and an object discrimination index (ODI) was calculated as the time spent exploring the novel object divided by the total exploration time. In the sociability test (ST), the test involved two phases based on a previously described procedure [[70]29, [71]30]. First, mice were habituated to the open field (50 × 50 × 50 cm) and empty enclosures for 5 min. Next, during the sociability phase, a novel male conspecific (social object) was placed in one enclosure, while a novel object (non-social object) was placed in the other. The experimental mouse was then allowed to explore the open field for 5 min, and the time spent in each quadrant (social vs. non-social) was recorded. The social discrimination index (SDI) was calculated as (time spent with social object) / (total exploration time). Tissue preparation, immunohistochemistry, and imaging Twenty-four hours after behavioral testing, mice under anesthesia were transcardially perfused with PBS, followed by 4% paraformaldehyde (PFA), and the brains were removed, post-fixed in 4% PFA for 24 h. Tissues were dehydrated with 30% sucrose and sectioned at 40 μm on a Leica microtome (Leica CM3050S, Germany). Immunostaining with a rabbit anti-ionized calcium-binding adaptor molecule 1 primary antibody (Iba1; #019–19741, Japan, RRID: AB_839504) and the specificity of this antibody has been previously validated in rodents [[72]31, [73]32]. A series of prefrontal and hippocampal sections was collected from experimental mice (at 240 µm intervals). These brain sections were processed for immunohistochemical detection of Iba1 (microglial marker), as described previously [[74]11]. Briefly, the free-floating sections were treated with 0.3% hydrogen peroxide (H[2]O[2]) for 30 min to quench endogenous peroxidase activity, incubated with 5% normal goat serum (Solarbio, China) for 30 min to block nonspecific binding, and immunoreacted with a rabbit anti-Iba1 antibody (Wako, #019-19741, at 1:1000 dilution, RRID: AB_839504) at 4 °C overnight. The free-floating sections were performed with biotinylated goat anti-rabbit immunoglobulin G (IgG) (at 1:200 dilution, Cat# BA-1400, RRID: AB_2336187, Vector Laboratories, USA) for 1 h, avidin-biotin peroxidase complex (at 1:200 dilution, Cat# PK-6100, RRID: AB_2336819, Vector Laboratories, USA) for 1 h, and 0.05% diaminobenzidine (Sigma-Aldrich) containing 0.03% H[2]O[2] and 0.25% nickel ammonium sulfate (Sigma-Aldrich) for 10 min. All sections were mounted on gelatin-coated glass, dehydrated in graded ethanol, treated with TO-type biological transparentizing agent, coverslipped using neutral quick drying glue, and scanned using a digital whole-slide scanner (NanoZoomer-XR, Japan). All digital images of brain sections were adjusted for cropping, brightness/contrast, and image size using Adobe Photoshop 2020 (Adobe Systems, USA). The image files were processed in the .tif format. The quantification of microglial density and morphology was performed using NIH Image J software as described in our previous study [[75]11]. For immunofluorescence analysis, three sections per sample from the hippocampus of CX3CR1^CreERT2:: Ai9 mice were incubated overnight at 4 °C with the appropriate antibodies (rabbit anti-PSD95 antibody, 1:500, ab18258, RRID: AB_444362, Abcam; rat anti-CD68 antibody, 1:500, MCA1957, RRID: AB_322219, Bio-Rad). Binding sites were visualized using Alexa Fluor-conjugated secondary antibodies at 50 μg/mL for 2 h at room temperature (Alexa Fluor® 488-AffiniPure donkey anti-rabbit IgG, 711-545-152, RRID: AB_2313584; Alexa Fluor® 488 AffiniPure goat anti-rat IgG, 112-545-003, RRID: AB_2338351; Jackson ImmunoResearch Inc). Nuclei were counterstained with Hoechst 33,342 (1:2000; Cat# 4082, RRID: AB_10626776, Cell Signaling Technology) during incubation with the secondary antibody. The slides were coverslipped with Citiflour® mounting medium. The fluorescent signals were observed using an Olympus VS200 slide scanner and a Spinning-Disc confocal microscopy (Nikon/EclipsTi2-E with Andor Dragonfly 200, Nikon) with a 60 × oil-immersion objective. The imaging parameters (laser power, gain, offset, resolution, and steps in the z direction) were consistent across all slides. Three-dimensional (3D) reconstruction All images were analyzed using IMARIS 10 software (Bitplane). Three images randomly picked from each mouse were reconstructed. PSD95 + puncta were reconstructed using the “Spots” function in IMARIS. CX3CR1 + microglia (tdTomato) were accurately reconstructed using the “Surface” function in IMARIS. The “Split into Surface Objects” function in IMARIS was used to assess the number of PSD95 + puncta entirely within the microglial surface. Each mouse had at least eight cells (n = 5–6 mice per group). The mean result was used for morphological analysis. Transcriptome sequencing and data analysis The brains used for RNA sequencing (RNA-seq) were removed from euthanized mice within 1–2 h after sleep deprivation, and the prefrontal cortex was excised. Samples of the prefrontal cortex from eight control and SD-treated mice were subjected to RNA extraction, RNA quantification, cDNA library preparation, and library quality assessment according to the protocols provided by Novogene (Novogene Co., Ltd., Beijing, China). Libraries were prepared for Illumina sequencing using the Illumina Nova-seq platform at Novogene. All the downstream analyses were based on the high-quality clean Unique Molecular Identifiers reads. Furthermore, we performed differential expression analysis of two groups using the DESeq R package and adjusted P-values using Benjamini and Hochberg’s approach for controlling the false discovery rate. We performed Gene Ontology (GO) enrichment analysis of differentially expressed genes (DEGs) by the GOseq R package with an adjusted P-value < 0.05 and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis using the DEGs to identify functional pathways ([76]http://www.genome.jp/kegg/). Nuclei isolation and snRNA-seq on the 10x genomics platform The brains used for snRNA-seq were removed from euthanized mice within 1–2 h after SD, and the prefrontal cortex (PFC) and hippocampus (HIP) were excised and pooled into a single sample. Samples from the six control and SD groups were lysed in lysis buffer and homogenized with a glass Dounce homogenizer. Nuclei extraction and library preparation were processed according to the protocols provided by Novogene (Novogene Co., Ltd., Beijing, China). Single-nucleus cells were manually counted on Luna FL using an AO/PI cell viability kit (Logos Biosystems) after each centrifugation and resuspension. Single-nucleus cell suspensions were loaded into chromium microfluidic chips with a 10 × Chromium Controller (10X Genomics). RNA from the barcoded cells was subsequently reverse-transcribed and sequencing libraries were constructed with reagents from a Chromium Single Cell 3’v2 reagent kit (10X Genomics) according to the manufacturer’s instructions. Single-nucleus RNA sequencing was performed with an Illumina platform (Illumina NovaSeq 6000) according to the manufacturer’s instructions (Illumina). snRNA-seq data processing and filtering Raw read sequences produced by the Illumina pipeline in fastq format were preprocessed through Trimmomatic software as follows: (1) low-quality reads were removed; (2) trailing low-quality or N bases were removed; (3) adapters were removed; (4) reads below 26 bases long were dropped; and (5) reads that could not form pairs were discarded. Downstream analyses were performed on the filtered data reanalyzed with Cell Ranger (version 7.0) ([77]https://github.com/10XGenomics/cellranger) and R package Seurat (version 4.3.0) ([78]https://satijalab.org/seurat/articles/pbmc3k_tutorial.html). Briefly, resulting fastq files were aligned to the mouse genome (mm10 1.2.0) using Bowtie (version 1.1.2). Unique molecule identifiers were counted for individual genes and cells (filtered by CellRanger) to construct digital expression matrices. The Seurat package filtered the data to screen for genes expressed in more than three cells, as well as for cells expressing more than 200 genes. Dimensionality reduction, clustering, and UMAP Integration of multiple sample datasets was performed using the canonical correlation analysis (CCA) algorithm within Seurat, followed by data scaling via the ScaleData function. Principal component analysis (PCA) was conducted on the top 2000 highly variable genes to reduce linear dimensionality, with the first 20 principal components used in subsequent analysis. Cell clusters were identified using the FindClusters function in Seurat with a resolution = 0.5, and visualization was achieved through uniform manifold approximation and projection (UMAP). Cluster-enriched genes were identified using the FindAllMarkers function, with the settings “min.pct = 0.25”. Functional enrichment analysis, single-cell pseudotime analysis, and cell‒cell interaction analysis Gene functional enrichment analysis was conducted using the clusterProfiler package, with the following parameters: ont = BP, pAdjustMethod = BH, pvalueCutoff = 0.01, and qvalueCutoff = 0.05. Pseudotime analysis was performed using the R package Monocle 2, with the starting point determined by CytoTRACE. High dispersion genes across cells were utilized to select and arrange cells of interest, followed by division into two components using the discriminative dimensionality reduction via learning a tree (DDRTree) approach. Branch-dependent genes were identified using branched expression analysis modeling (BEAM). Cell‒cell interactions were assessed using CellChat (version 2.1.0) to infer and visualize intercellular communication based on known ligand‒receptor pairs. All categories of ligand-receptor interactions in the database were utilized, and differences in communication probabilities were visualized using the netVisual_bubble function in the CellChat R package. Quantitative real-time polymerase chain reaction (qRT‒PCR) At 1–2 h after SD, the prefrontal cortex and hippocampus were quickly removed and excised from euthanized mice. The specific sequences of primers used for qRT‒PCR are listed in Extended Data Table [79]1 (36 inflammation-related, 12 senescence-associated, and 11 synapse-related candidate genes) and were designed according to reference sequences in PrimerBank or the NCBI database with Primer-BLAST. The primer sequences were synthesized by Sangon Biotech (Shanghai, China). Total RNA was isolated from the PFC and HIP using TRIzol reagent and reverse-transcribed into cDNA using a HiScript III All-In-One RT SuperMix Perfect for qPCR Kit (Cat# R333, Vazyme, China). qRT‒PCR was performed using the QuantStudio™ 3 Applied Biosystem Real-Time PCR System and PowerUp SYBR Green Master Mix (Cat. No. A25742; Applied Biosystems, USA). The relative mRNA expression levels of selected genes were calculated with the 2^−ΔΔCt method [[80]33]. The relative mRNA expression levels were normalized to the expression level of GAPDH. Each sample was tested in duplicate. The experimental group values were normalized to those of the control group. Multicytokine assay At 24 h after SD, the plasma from each treatment and control group was collected for a multicytokine assay. Cytokine and chemokine levels were evaluated with a Luminex X-200 system (R&D Systems, USA) using a panel of 31 mouse cytokines (LX-MultiDTM-31, R&D Systems, USA) in accordance with the manufacturer’s instructions. The selected cytokines included CXCL13, CCL27, CXCL5, CCL11, CCL24, CX3CL1, GM-CSF, CCL1, IFN-γ, IL-1β, IL-2, IL-4, IL-6, IL-10, IL-16, CXCL10, CXCL11, CXCL1, CCL2, CCL7, CCL12, CCL22, CCL3, CCL4, CCL20, CCL19, CCL5, CXCL16, CXCL12, CCL17, and TNF-α. Data acquisition and analysis were conducted using Luminex xPONENT software (Luminex Corporation). The experimental group values were normalized to those of the control group. Electron microscopy Synaptic density and mitochondrial damage were determined by electron microscopy as described previously [[81]34]. At 1–2 h after SD, the mice were deeply anesthetized and transcardially perfused with 4% PFA. Hippocampal slices were post-fixed in cold 1% OsO4 for 2 h. Hippocampal samples were prepared for electron microscopy using standard procedures. Ultrathin sections (70 nm, Ultra 45°, Daitome) were stained with uranyl acetate/lead acetate. Synaptic density and mitochondrial damage were viewed by transmission electron microscopy (TEM, HT7700, accelerating voltage 120 kV). Seven mice were examined in the two groups. Synapses were identified by the presence of synaptic vesicles and postsynaptic densities. Each mitochondrion was classified as either intact (the mitochondrial membranes and internal cristae structures were clear and complete) or damaged (loss of the outer membrane or loss of the inner cristae structure). Golgi staining At 1–2 h after SD, the mice were deeply anesthetized and transcardially perfused with 4% PFA. Golgi staining was performed using the FD Rapid GolgiStain™ Kit according to the manufacturer’s recommended protocol (PK401, FD NeuroTechnologies). Briefly, hippocampal samples were immersed in impregnation solution made by mixing equal volumes of solutions A and B for 3 weeks and then transferred to solution C for 5 days at room temperature in the dark. The hippocampus was sliced using a Leica microtome (Leica CM3050S, Germany) at a thickness of 100 μm. Each section was rinsed and then placed in staining solution (solution D/solution E/double-distilled water = 1:1:2) for 10 min. The sections were dehydrated and cover-slipped. Dendritic spines were examined using a Nikon ECLIPSE Ni-E microscope (60 × oil objective). The qualitative and morphometric properties of the neurons were investigated as described previously [[82]35]. Statistical analysis Randomization was used to assign animals to experimental groups. Furthermore, samples for histopathological and bioinformatics analyses were randomly selected from each group. For the histology experiments, the damaged brain sections were excluded from the quantitative analysis. Drowning mice in the SD model were excluded. All histopathological analyses were conducted by trained personnel in a blinded manner. Statistical analysis was analyzed using SPSS 25 software (SPSS, Inc., Chicago, IL, USA). All results are expressed as the mean ± standard error of the mean (SEM). For normally distributed data, differences between group means were tested using Student’s t-test when the experiment contained two groups or one-way analysis of variance (ANOVA) followed by Tukey’s post hoc test and two-way ANOVA followed by Bonferroni post hoc correction when the experiment contained more than two groups. To ensure 80% power to detect a 25% difference between groups (Cohen’s d, 5% Type I error rate), a sample size of at least eight per group was selected. Significance was set at P < 0.05. The data were plotted using GraphPad Prism (GraphPad Software, San Diego, CA, USA). Results Intermittent SD induces mania-like behavior In this study, we provided a new paradigm of intermittent 16-h daily SD (from 6 P.M. to 10 A.M. the next day) for 4 days, to mimic sleep loss as the result of multiple sleep pressures (Fig. [83]1a, b). Intermittent SD in male mice induced significant weight loss, an increase in the total distance traveled, and an increase in the frequency of entries into the central area in the OFT (Fig. [84]1c–f). In the EPM test, SD also significantly increased the total distance moved and the frequency of entries into the open arms (Fig. [85]1g–i). VPA, a common mood stabilizer for the treatment of bipolar disorder [[86]36], reversed these mania-like behavioral alterations in both male and female mice after 7 days of VPA pretreatment (Fig. [87]1j–l and Extended Data Fig. [88]1). Furthermore, we incorporated the TST and the ST to further validate the efficacy of the sleep deprivation-induced BD model (Extended Data Fig. [89]2). The TST results showed a significant reduction in immobility time in the SD group (Extended Data Fig. [90]2a), indicating a marked antidepressant effect (mania-like behavior). Furthermore, the NOR test revealed that both the control and SD groups exhibited a significant increase in exploration time for the novel object (Extended Data Fig. [91]2b). Importantly, there was no significant difference in the ODI between the two groups (Extended Data Fig. [92]2c), suggesting that SD did not impair novel object recognition ability in mice. In the sociability test, the results demonstrated that SD mice displayed a significantly increased exploration time towards the social mice compared to the novel object (Extended Data Fig. [93]2d). Moreover, the SDI was significantly higher in the SD group, indicating enhanced social interaction (Extended Data Fig. [94]2e). Fig. 1. Paradoxical sleep deprivation (SD) induces mania-like behavior and transcriptomic changes, and treatment with valproate (VPA) reverses these behavioral alterations in male mice. [95]Fig. 1 [96]Open in a new tab a, j Schematic of the experimental procedures. b Schematic of the SD apparatus. c Body weights of control (Con) and SD mice. d Representative traces of movement in the open field test (OFT) of control- and SD-treated mice. e, f Quantification of total distance moved and frequency of entry into the central area of the open field after intermittent 16-h SD for 4 days. g Representative traces of movement from the elevated plus-maze test (EPM). h, i Quantification of the total distance moved and duration in the open arms. k, l Quantification of the total distance moved during the 5-min OFT and total cumulative duration of immobility (velocity < 1.75 cm/s). m Volcano plot of differentially expressed genes (DEGs) in the prefrontal cortex between control and SD mice (n = 4, each group). Red dots represent genes with upregulated expression, green dots represent genes with downregulated expression, and blue dots represent non-DEGs. The labels are given as gene symbols for the top gene by unadjusted P value rank. n The top significantly enriched Gene Ontology (GO) categories of biological process, cellular component and molecular function between the two groups. The X-axis indicates the value of -log10 (P-value). The Y-axis indicates GO terms. o The scatter plots show the top 30 enriched KEGG pathways between the two groups. The Y-axis indicates KEGG pathways. The X-axis indicates the gene ratio. p Validation of DEGs expression by quantitative real-time polymerase chain reaction (n = 5–6, each group). *P < 0.05, **P < 0.01, ***P < 0.001 (n = 9–12 each group for behavioral test). Statistical analysis was performed using unpaired t test a–i, p or two-way ANOVA followed by Bonferroni post hoc correction k, l. Transcriptome sequencing reveals inflammation- and synapse-related gene expression profiles in SD-induced BD model We first tested for differential gene expression in the PFC between control and SD-treated male mice using RNA-seq (Fig. [97]1 and Extended Data Fig. [98]3). DEGs (1031 upregulated genes and 782 downregulated genes) were identified by RNA-Seq and are shown in a volcano plot (Fig. [99]1m). Gene Ontology enrichment analysis revealed that both inflammation- and synapse-related DEGs were enriched including the MHC class I peptide loading complex, the MHC protein complex, T-cell receptor binding, CD8 receptor binding, negative regulation of immune system process, synaptic membrane, postsynaptic density membrane, dendritic spine, and dendrite development (Fig. [100]1n). In addition, KEGG analysis from the RNA-seq data further revealed enrichment of DEGs associated with immune responses (e.g., IL-17 signaling pathway, TNF signaling pathway, T-cell leukemia virus 1 infection, Epstein‒Barr virus infection, and autoimmune thyroid disease) and synaptic transmission (e.g., axon guidance and adherens junction) (Fig. [101]1o). To validate the RNA-seq data, we confirmed the expression of several randomly chosen DEGs by qRT‒PCR (Fig. [102]1p). SD reduces the cytokine/chemokine levels in the plasma, expression of microglial marker genes, and microglia density in the brain The SD-treated mice showed significantly lower levels of cytokine and chemokine levels in the plasma compared with the control group by a Luminex-based multicytokine assay (Fig. [103]2a). To examine the changes in microglia (CNS-resident immune cells) after chronic SD exposure, we performed qRT‒PCR to detect CD11b, CSF1R, and TREM2 (microglial marker genes) mRNA levels and immunohistochemistry to analyze microglial morphology in the brain of male mice (Fig. [104]2b–k). We found that the gene expression levels of these microglial genes were significantly lower in the PFC and HIP SD-treated mice than in the untreated controls (Fig. [105]2b, c). Furthermore, intermittent SD stress significantly reduced the density of Iba1-ir cells and the microglial soma area in the PFC and HIP (Fig. [106]2da–f, i, j) and increased microglial total process length and the number of branching points only in the PFC (Fig. [107]2g, h, k). Fig. 2. Effects of SD on microglial marker gene expression and morphology in the brain. [108]Fig. 2 [109]Open in a new tab a The levels of 31 cytokines and chemokines in the plasma of the two groups with a Luminex liquid chip (red, upregulated; blue, downregulated; n = 5, each group). b, c The expression of CD11b (microglial marker), CSF1R (microglial marker), and Ki67 in the PFC and hippocampus (HIP). d Representative high-magnification photomicrographs showing Iba1 immunoreactivity in the PFC of control and SD-treated mice. e–k Changes in microglial density and morphology in the PFC and HIP of the two groups. *P < 0.05, **P < 0.01, ***P < 0.001. Scale bars = 50 μm in d. Statistical analysis was performed using unpaired t test or Mann-Whitney test. Single-nucleus RNA sequencing reveals cell-type-specific changes in SD-induced BD model To further identify the cell populations potentially involved in SD-induced mania-like behavior, we performed snRNA-seq of approximately 84,124 nuclei isolated from the prefrontal and hippocampal tissues after 16-h daily SD for 4 days (Fig. [110]3a). After raw data quality control, alignment and UMI filtering, 75,201 nuclei were retained for further analysis (Extended Data Table [111]2 and Extended Data Fig. [112]4). As a result, we identified 29 distinct clusters through an unsupervised graph-based clustering algorithm (Fig. [113]3b). We next confirmed the cell types identified above and annotated the main cell types by examining cell marker gene expression reported in previous studies [[114]37, [115]38]: microglia (n = 4463; PTPRC, APBB1IP, DOCK8, MERTK, CSF1R), excitatory neurons (n = 41,826; marked by CUX2, TLE4, RORB, SLC17A7), interneurons (n = 11,604; SST, GAD1, GAD2, SV2C), astrocytes (n = 5358; SLC1A2, SLC1A3, GFAP, GLI3, AQP4), oligodendrocytes (n = 9027; MBP, PLP1, MOBP), and oligodendrocyte progenitor cells (n = 1982; VCAN, LHFPL3, PDGFRA, PCDH15) (Fig. [116]3c, d). Moreover, SD stress did not result in an imbalance of glial/neuronal ratio or distribution of cell type-specific clusters between the two groups. Fig. 3. Effects of SD on the single-cell transcriptome in the prefrontal and hippocampal tissues of male mice by single-nucleus RNA sequencing. [117]Fig. 3 [118]Open in a new tab a Schematic of the experimental design and analysis. b, c tSNE visualization showing clustering of 75,201 nuclei by unsupervised cluster (b) and annotated cell types (c). d Bubble plot showing the expression levels, distribution of representative cell-type markers and numbers annotated by cell types. e Numbers of up- and downregulated DEGs in each cell type. f DEGs involved in pathways in each cell type. g Representative tSNE depicting clustering of microglial nuclei from the brains of control (n = 3) versus SD (n = 3). Ast astrocyte, Exn excitatory neuron, Int interneuron, Mic microglia, Oli oligodendrocyte, OPC oligodendrocyte precursors. Next, we compared cell type-specific gene expression differences between control and SD-treated mice, and identified 4181 DEGs across six cell types, including 3172 upregulated and 1009 downregulated genes (Fig. [119]3e). More upregulated DEGs than downregulated DEGs were found in excitatory neurons, interneurons, oligodendrocytes, and astrocytes but not in microglia or oligodendrocyte progenitor cells (Fig. [120]3e). To gain insight into the biological pathways associated with these DEGs, we performed KEGG analysis on DEGs of different cell types. The significantly enriched KEGG pathways showed enrichment of DEGs in microglia participating in ubiquitin-mediated proteolysis, the B-cell receptor signaling pathway, the mTOR signaling pathway, autophagy-animal, the T-cell receptor signaling pathway, the chemokine signaling pathway, the PI3K-Akt signaling pathway, and the TNF signaling pathway (Fig. [121]3f). Additionally, the DEGs in excitatory neurons and interneurons were mostly implicated in axon guidance, dopaminergic synapse, the calcium signaling pathway, glutamatergic synapse, circadian entrainment, morphine addiction, cholinergic synapse, and GABAergic synapse (Fig. [122]3f). We performed unsupervised cluster to confirm the identity of microglia in control versus SD-treated mice (Fig. [123]3g). Damaged mitochondria, synapse gain, and decreased synaptic engulfment by microglia during SD To evaluate the consequences of SD on synaptic structures, we analyzed prefrontal and hippocampal sections from adult male mice using Golgi staining and transmission electron microscopy. TEM imaging of the HIP revealed that SD caused severe damage of mitochondria (Fig. [124]4a). Meanwhile, the results of electron microscopic analysis showed SD-induced increase in synaptic density and decrease in the lengths of active zones of synapses (Fig. [125]4b–d). Consistent with these findings, Golgi staining revealed a significant increase in the total spine density in the HIP of SD-treated male mice (Fig. [126]4e–g). Additionally, our data showed a relatively low percentage of engulfed PSD95 inside microglia after SD exposure (Fig. [127]4h, i). Fig. 4. Effects of sleep deprivation (SD) on microglia, mitochondria and synapses in the brains of male mice. [128]Fig. 4 [129]Open in a new tab a The percentage of damaged mitochondria in the HIP of control and SD-treated male mice by electron microscopy. The green shadow indicates mitochondria. b Images of synapses in the HIP by electron microscopy. The yellow shadow indicates the presynaptic terminal. The red shadow indicates the postsynaptic spine. c, d The number of synapses and the lengths of the active zones of synapses (n = 3–4, each group). e, f Representative Golgi-Cox staining images of dendritic spines in the HIP. g Histograms showing spine densities of dendritic spines. h Representative images and 3D reconstruction rendering of microglia (red, tdTomato) containing PSD95 + puncta (green) in the HIP from CX3CR1^CreERT2:: Ai9 mice treated with SD. The white arrowheads show engulfed PSD95 + puncta inside microglia. i Quantification of the percentage of engulfed PSD95 puncta inside microglia (n = 5–6 per group). The data are shown as the mean ± SEM. The data represent three independent experiments. *P < 0.05, **P < 0.01. Scale bars = 200 μm in e; 10 μm in f; 5 μm in h. Statistical analysis was performed using unpaired t test. Microglial depletion prevents SD-induced mania-like behavior in a sex-specific manner Interestingly, microglial inhibition by MCL did not prevent SD-induced mania-like behavior in either male or female mice (Extended Data Fig. [130]5). To determine whether microglial depletion in the brain influenced SD-induced behavior, we investigated changes in the SD-induced BD model after PLX pretreatment at a dose of 50 mg/kg (efficiency of microglia depletion > 80% [[131]20]) (Fig. [132]5a). In the OFT and EPM tests, pharmacological depletion of microglia completely reversed SD-induced mania-like behavior in male mice (Fig. [133]5b, c, e–g). In contrast, microglial elimination had no significant effect on SD-induced mania in female mice (Fig. [134]5d, e, h, i). Fig. 5. Effects of the colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX3397 (pexidartinib, PLX) on SD-induced mania-like behaviors and inflammation-related gene expression changes in a sex-specific manner in mice. [135]Fig. 5 [136]Open in a new tab a Schematic of the experimental procedures. b, d Representative heatmaps showing the cumulative duration spent by each mouse throughout the compartment during the 5-min open field test (OFT) in male (left) and female (right) mice. c, e Quantification of the frequency of entry into the central area of the open field in PLX- and/or SD-treated mice. f, h Representative heatmaps showing the cumulative duration spent by each mouse throughout the maze in the 5-min elevated plus-maze test (EPM). g, i Quantification of the frequency of mouse entry into the open arms. j Senescence-associated and inflammation-related gene expression changes in the PFC (left) and HIP (right) in PLX- and/or SD-treated male and female mice (red, upregulated; blue, downregulated). * indicates comparison with Veh + Con. # indicates comparison with Veh + SD (n = 6, each group). k Venn diagram representing the overlap of DEGs among the four groups in the PFC and HIP of male and female mice (blue, Veh + Con vs Veh + SD; yellow, Veh + SD vs PLX + SD; red, Veh + Con vs PLX + Con). N.S., no significant difference; *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, ^#P < 0.05, ^##P < 0.01, ^###P < 0.001, ^####P < 0.0001. Statistical analysis was performed using two-way ANOVA followed by Bonferroni post hoc correction. Effects of microglial depletion on sex-specific changes in inflammation-related, senescence-associated, and synapse-related gene expression in SD-induced BD model To further examine the effects of microglial depletion on SD-induced inflammation-related and senescence-associated gene expression abnormalities, we performed qRT‒PCR to test these genes in the PFC and HIP. After 16-h daily SD exposure for 4 days, we identified 16 inflammation-related and senescence-associated DEGs (4 upregulated and 12 downregulated genes) in the PFC in males and 25 DEGs (5 upregulated and 20 downregulated genes) in females between control and SD-treated mice (Fig. [137]5j, k). In the HIP, we identified 18 DEGs (6 upregulated and 12 downregulated genes) in males and 21 DEGs (3 upregulated and 19 downregulated genes) in females between control and SD-treated mice (Fig. [138]5j, k). Among them, 6 DEGs in the male PFC (6/16, 37.5%) and 10 DEGs in the male HIP (10/18, 55.6%) were significantly regulated by PLX treatment, while 13 DEGs in the female PFC (13/25, 52%) and 6 DEGs in the female HIP (6/21, 28.6%) were significantly affected by PLX treatment (Fig. [139]5k). Together, these results indicate a sex-specific functional role of microglia in the HIP. Next, we sought to determine whether there were sex differences in synapse-related gene expression in the HIP in SD- and/or PLX-treated mice. We identified 2 synapse-related DEGs (1 upregulated and 1 downregulated gene) in the HIP in males and 2 DEGs (0 upregulated and 2 downregulated genes) in females between control and SD-treated mice (Fig. [140]6a, b). However, synapsin III (Syn3), synaptotagmin-1 (Syt1) and synaptotagmin-7 (Syt7) were upregulated after treatment with PLX in SD-treated males but not in SD-treated females (Fig. [141]6a). Fig. 6. Effects of PLX3397 (PLX) on SD-induced microglial and synaptic changes in the PFC and HIP in male mice. [142]Fig. 6 [143]Open in a new tab a Synapse-related gene expression changes in the HIP in PLX- and/or SD-treated male and female mice (red, upregulated; blue, downregulated). * indicates comparison with Veh + Con. # indicates comparison with Veh + SD (n = 6, each group). b Venn diagram representing the overlap of DEGs among the four groups in the HIP of male and female mice (blue, Veh + Con vs Veh + SD; yellow, Veh + SD vs PLX + SD; red, Veh + Con vs PLX + Con). c, f Representative images of Iba1 immunoreactivity in the PFC and HIP. d–h Changes in microglial density and soma area in the PFC d, e and HIP g, h after treatment of male mice with PLX and/or SD. i Representative high-magnification Golgi-Cox staining images of dendritic spines in the HIP. j, k Histograms showing the total dendritic spine density in males and females. *P < 0.05, **P < 0.01. Scale bars = 50 μm in c, f; 10 μm in i. Statistical analysis was performed using two-way ANOVA followed by Bonferroni post hoc correction. PLX3397 pretreatment eliminates microglia and reverses SD-induced synaptic gain in males but not in females Subsequently, we conducted immunohistochemistry and Golgi staining to observe the effect of CSF1R inhibitor PLX on microglia and neurons. We found a significant reduction in the density of microglia and soma area in the PFC and HIP (Fig. [144]6c–h). Short-term pretreatment with PLX significantly blocked the microglia response to SD stimulation (Fig. [145]6d, g). Importantly, the elimination of microglia significantly decreased the total spine density in the HIP in SD-treated male mice but not in SD-treated female mice (Fig. [146]6i–k). Discussion Here, we introduce the modified SD paradigm as a BD model in mice to mimic sleep loss during the whole daytime and partial nighttime in the presence of REM sleep pressure. We then used this model to elucidate the underlying mechanism by which intermittent SD in male mice triggers mania-like behavior and a cytokine-deficiency state. Microglia-mediated immune and synaptic dysfunctions were detected not only in the bulk transcriptome, but also in the single-cell transcriptome. Furthermore, our results confirmed SD-induced mania-like behaviors, inflammation-related, senescence-associated, and synapse-related gene expression changes in a sex-specific manner. Microglial loss and synapse gain were observed in SD-treated mice. Finally, we demonstrated that a CSF1R inhibitor can eliminate microglia and prevent SD-induced mania-like behavior and synapse gain in male mice. The validity of our chronic SD model was demonstrated using a range of behavioral paradigms. The OFT and EPM are commonly used to assess anxiety-like behavior [[147]39, [148]40], but are also widely employed for behavioral assessment of mania-like behavior (hyperactivity and decreased anxiety-like behaviors) in BD models [[149]41–[150]43]. This hyperactivity may be related to impulsivity in clinical BD patients. The TST and the ST further validate the efficacy of the sleep deprivation-induced BD model. Results showed a significant reduction in immobility time in the SD-treated group in the TST, indicating a marked antidepressant effect, which may relate to impulsivity and irritability in clinical BD patients. In the sociability test, SD mice displayed a significantly increased exploration time towards the social mice compared to the novel object. Moreover, the SDI was significantly higher in the SD group, indicating enhanced social interaction. This may mimic the talkativeness observed in patients with bipolar disorder (BD) [[151]44]. The chronic SD model elicits behavioral changes that are sustained for several hours (Extended Data Fig. [152]6), while the associated microglial reduction is maintained for several days (Extended Data Fig. [153]7). Additionally, our findings demonstrate that acute and chronic SD exert differential effects on hippocampal inflammation-related gene expression (Extended Data Fig. [154]8). Previous studies have used acute or severe SD models to investigate the effects of sleep loss on brain inflammation [[155]10, [156]45]. Here, single-nucleus and bulk RNA-seq analyses emphasized the enrichment of DEGs in microglia participating in inflammatory signaling pathways, including the TNF signaling pathway, IL-17 signaling pathway, B-cell receptor signaling pathway, and autophagy, in the chronic BD model. Liu et al. reported that DEGs enriched in the TNF signaling pathway were significantly upregulated during acute paradoxical SD [[157]45]. In addition, acute SD in mice induces activation of the IL-17 signaling pathway [[158]46], impaired B-cell responses [[159]47], and excessive autophagy [[160]48]. Four days of prolonged and continuous SD exposure leads to a cytokine-storm-like phenotype in mice [[161]10]. In contrast, we detected decreased expression levels of inflammatory genes in the brain and decreased levels of inflammatory cytokines in the plasma after 4 days of intermittent 16-h daily SD exposure. These findings expand the understanding of the role of microglia-mediated neuroinflammation in BD. Importantly, Zandi et al. reported that downregulated neuroimmune and synaptic pathways are associated with transcriptional changes in the amygdala and anterior cingulate from post-mortem brains of individuals with BD [[162]49], which is consistent to our findings in the hippocampus of SD-induced mice. Here, we detected the downregulation of microglia-specific genes in the PFC and HIP in both SD-treated male and female mice. Furthermore, snRNA-seq and bulk RNA-seq analyses showed that DEGs related to the synaptic membrane and function were enriched in SD-treated male mice. Abnormal synaptic transmission and synaptic pruning have been found in sleep-deprived mice [[163]13, [164]14, [165]50]. The synaptic homeostasis hypothesis proposes an unsustainable increase in synaptic strength during wakefulness and increased synaptic pruning during sleep [[166]51, [167]52]. A more recent study showed synapse gains during sleep deprivation and synapse loss during sleep in zebrafish [[168]53]. Our data confirmed the decreased engulfment of synapses in microglia and synapse gains during chronic SD. SD-induced behavioral and synaptic changes were prevented by the in vivo depletion of microglia with a CSF1R inhibitor (PLX) but not by the inhibition of microglia with MCL. Notably, the upregulation of Syn3, Syt1, and Syt7 induced by PLX may protect against behavioral and synaptic changes in SD-treated male mice. Strong evidence has indicated that Syt7 deficiency in the HIP induces mania-like behavior in mice by attenuating N-methyl-D-aspartate receptor activity [[169]54]. These findings strengthen the role of neuroinflammation and microglia-mediated synaptic pruning in BD. Microglia contribute to the inflammatory response and synaptic dysfunction induced by SD stimulation. Pretreatment with PLX may block the microglia-mediated detrimental processes induced by SD stimulation. Previous studies have shown that both Clock knockdown mice and Rev-erbα knockout mice exhibit marked mania-like behavior (hyperactivity and decreased anxiety-like behaviors) [[170]55, [171]56]. Depleting microglia using the CSF1R inhibitor ameliorated neuroinflammatory changes and mania-like behavior in Rev-erbα knockout mice [[172]55]. These literature reports are similar to our findings in SD-induced mice. Our findings show decrease in the number of microglia following SD contributes to loss of synaptic pruning. Sleep-deprived male mice impaired microglia-mediated surveillance, phagocytic microglial contraction and synaptic pruning deficits [[173]50, [174]57, [175]58]. Depletion of microglia with PLX pretreatment, rather than inhibition (MCL), blocked these SD-induced abnormalities. These data implicate microglial dyshomeostasis as a driver of SD-induced pathogenesis and show that pretreatment with the microglial depletion strategy can prevent this phenotype. Clinically, sex differences are present in BD symptoms and treatment response [[176]59–[177]61]. This study further examined sex-specific changes in SD-induced behaviors, gene expression levels, and treatment responses. We confirmed that VPA reversed SD-induced manic-like behavior in a sex-independent manner (Extended Data Fig. [178]1). Although long-term pretreatment with MCL has no effect on SD-induced behaviors with no sex difference, long-term pretreatment with PLX relieves SD-induced mania-like behaviors and synaptic gain in males but not females. Interestingly, more DEGs were detected in the PFC and HIP in SD-treated females than in SD-treated males, consistent with a higher burden of transcriptomic dysfunction observed in human brain specimens from females than in those from males with BD [[179]61], whereas PLX pretreatment had a much greater impact on DEGs in the HIP in SD-treated males than in SD-treated females. Microglial depletion by pretreatment with PLX prevents microglia-mediated inflammation-related and senescence-associated genes in the hippocampus in male mice. Sleep-deprived male mice develop microglia-mediated synaptic pruning dysfunction [[180]50], which may be mediated by the C1qb signaling pathway [[181]17, [182]62, [183]63] but not by the SIRPα-CD47 signal axis [[184]64]. Together, sex differences in BD may result from sex-specific differences in microglia-mediated neuroinflammation and complement-mediated microglial synaptic elimination. It remains to be confirmed whether the C1qb signaling pathway plays a causal role in BD in both males and females. In summary, our findings provide evidence that microglia-mediated inflammation, senescence, and synaptic pruning contribute to paradoxical SD-induced mania-like behavior in a mouse model of BD in a sex-specific manner. However, it remains to be understood at the single-cell level how sex-specific inflammation-related and senescence-associated DEGs affect microglia-mediated synaptic pruning in the pathophysiology of BD. Further elaborating the causal link between neuroimmune, neuroplasticity, and behavioral states will provide mechanistic insights into the pathophysiology and neurodevelopment of bipolar disorder and pave the way for curative therapies for BD patients. Supplementary information [185]Supplementary materials^ (76.9MB, pdf) Acknowledgements