Abstract The microbiota-gut-brain axis plays a pivotal role in neuropsychiatric disorders, particularly in depression. Escitalopram (ESC) is a first-line antidepressant, however, its regulatory mechanisms on the microbiota-gut-brain axis in the treatment of depression remain unclear. The antidepressant effects of ESC were evaluated using the forced swim test in Wistar-Kyoto (WKY) rats, while damage in the gut and brain regions was assessed through H&E staining and immunohistochemistry. The therapeutic mechanisms in WKY rats with depression-like behavior were investigated through 16S rRNA sequencing of the gut microbiota, serum untargeted metabolomics, and hippocampal proteomics. Results indicated that ESC intervention improved depressive-like behaviors, as evidenced by increased swimming times in WKY rats, and also restored intestinal permeability and brain tissue integrity. Significant changes in the gut microbiota composition, particularly an increase in Bacteroides barnesiae, as well as increases in serum sphingolipid metabolites (Sphinganine 1-phosphate, Sphingosine, and Sphingosine-1-phosphate) and hippocampal proteins (Sptlc1, Enpp5, Enpp2), were strongly correlated. These robust correlations suggest that ESC may exert its antidepressant effects by modulating sphingolipid metabolism through the influence of gut microbiota. Accordingly, this research elucidates novel mechanisms underlying the antidepressant efficacy of ESC and highlights the pivotal importance of the microbiota-gut-brain axis in mediating these effects. Subject terms: Depression, Hippocampus Introduction Depression is a common mental disorder, characterized by persistent low mood, loss of interest, and impaired cognitive function, affecting an estimated 300 million people of all ages globally [[38]1]. The impact of depression extends beyond psychological health, increasing the risk of chronic conditions such as cardiovascular diseases and diabetes, as well as being closely associated with a significantly higher suicide rate compared to the general population [[39]2]. The hippocampus plays a crucial role in the pathophysiology of depression, with alterations in its structure and function potentially being closely linked to the symptoms of the disorder [[40]3]. Studies have shown that the number of hippocampal neurons and glial cells may be reduced in individuals with depression, which is negatively correlated with the severity of the condition [[41]4]. Furthermore, chronic stress and depression are associated with synaptic inhibition and neuronal atrophy in the hippocampus [[42]5]. In addition to these neural changes, emerging evidence suggests that the microbiota-gut-brain (MGB) axis, which involves complex interactions between the gastrointestinal system and the central nervous system (CNS), may influence brain function through neural, endocrine, immune, metabolic, and other pathways [[43]6]. Over the past decade, mounting evidence has suggested a potential link between alterations in the fecal microbiome composition and the development of depression [[44]7, [45]8]. Depression phenotypes are accompanied by changes in the gut microbiota composition, which in turn influences depression-like behaviors and increases the risk of depression [[46]6]. Currently, pharmacological interventions remain the cornerstone of depression treatment. The interaction between medications and the microbiome can alter bacterial metabolism as well as drug activity and efficacy [[47]9]. Antidepressants possess antimicrobial and anti-inflammatory properties, which are associated with the ecology of the gut microbiota [[48]10, [49]11]. The antidepressant effects of fluoxetine and ketamine are correlated with the gut microbiome. For example, Actinobacteria and genera producing short-chain fatty acids (SCFAs), such as Butyricimonas and Turicibacter, might be related to the antidepressant effects of ketamine [[50]12]. Mice treated with fluoxetine exhibit changes in gut microbiome diversity and composition, such as decreased abundance of Ruminococcus and Adlercreutzia [[51]13]. Escitalopram (ESC), a selective serotonin reuptake inhibitor, is considered a first-line treatment option for patients with depression [[52]14]. In the depression CUMS model, significant differences in gut microbial communities and metabolites are observed between responders and non-responders to escitalopram treatment [[53]15]. Nowadays, targeting the gut microbiome and integrating MGB axis mechanisms in disease treatment has become a focal point of research. Therefore, studying the regulatory effects of escitalopram on the gut microbiome will aid in advancing our understanding of its antidepressant mechanisms. The Wistar-Kyoto rat (WKY rat) is a widely used spontaneous model of depression in research, exhibiting a variety of behavioral and physiological traits associated with depression, such as reduced social interaction, decreased exploratory behavior, and heightened sensitivity to stressful environments [[54]16]. These characteristics make the WKY rat an important model for understanding the pathophysiological mechanisms of depression and its treatment. The Forced Swim Test (FST) is one of the classic experimental methods for assessing antidepressant behavior in animals [[55]17]. In this test, animals are placed in a pool of water from which they cannot escape or touch the bottom, typically exhibiting a state referred to as “behavioral despair.” The effectiveness of antidepressants is generally assessed in the FST by reducing the time spent in passive floating and increasing the time struggling and swimming, thus reflecting the potential antidepressant activity of the compounds. This method is not only a standard tool for evaluating antidepressants but also key for understanding behavioral changes associated with depression [[56]18, [57]19]. However, despite the WKY rat and FST providing significant tools for depression research, the mechanisms by which the gut microbiota influences the antidepressant effects of escitalopram, a commonly used selective serotonin reuptake inhibitor, especially within the WKY rat model, remain unclear. In this study, we utilized the WKY rat model of depression to investigate changes in the gut microbiome, serum metabolomic profiles, and hippocampal proteomic patterns following administration of ESC. Furthermore, this research analyzed the interactions between microbes, metabolites, and proteins, exploring how these differential elements synergistically function within the MGB axis to mediate the antidepressant effects of ESC. This study aims to deepen our understanding of the gut-brain interactions in the treatment of depression, hoping to provide a theoretical basis and experimental support for novel antidepressant treatment strategies based on microbiome modulation. Materials and methods Animal and experimental design This study utilized 8 male Wistar rats and 16 WKY rats, a depression model, weighing 150 ± 20 g and aged 6–8 weeks, obtained from Vital River Laboratories (Beijing, China). No formal sample size estimation was performed. The chosen sample size was based on prior research experience and the feasibility of the experimental design. Animals were included in the study if they met the following criteria: healthy, male rats of the specified strains, with no signs of illness or physical abnormalities. Exclusion criteria included any animals that exhibited abnormal behavior, signs of injury, or illness during the acclimation period or prior to the start of the experiment. The animals were housed under conditions of 21–25 °C temperature, 35 ± 5% humidity, and a 12 h light/dark cycle. The study was approved by the First Affiliated Hospital of Henan University of Science and Technology Animal Ethics Committee (2023-D-H0006), and conducted in accordance with the Regulations for the Management of Laboratory Animals by the Ministry of Science and Technology of the People’s Republic of China. The compound escitalopram oxalate was acquired from MCE (HY-14258A, MCE, China). Escitalopram oxalate was dissolved in 0.85% saline to achieve a dose of 10 mg/kg. Age and weight-matched WKY rats were randomly divided into two groups using computer-based randomization software, each consisting of 8 animals: one group received the prepared ESC solution (ESC group), and the other received the vehicle solution (WKY group). Additionally, 8 Wistar rats served as the control group (CON group), receiving the vehicle solution without any treatment. Rats in the ESC group were administered 10 mg/kg/day of ESC solution, while those in the WKY and control groups received an equivalent volume of vehicle solution, all via intraperitoneal injection for 21 consecutive days. Post-administration, daily assessments and records of rat body weight were conducted. Furthermore, any physiological changes, including the occurrence of diarrhea and any lethal effects, were carefully monitored. Blinding was not implemented during the treatment phase, but all outcome assessments were conducted without prior knowledge of group assignment to minimize bias. The experimental procedures are shown in Fig. [58]1A. Fig. 1. ESC improves behavioral changes and MGB axis damage in rats with depression-like behavior. [59]Fig. 1 [60]Open in a new tab A Schematic flow of the experiment. B The immobility (%) in the forced swim test between the three groups at DAY0 and DAY22 was analyzed using a two-way repeated-measures ANOVA, with post-hoc comparisons performed using Bonferroni correction. C Growth rates of body weight in different rat groups were analyzed using a two-way repeated-measures ANOVA, with post-hoc comparisons performed using Bonferroni correction. The significance between CON and WKY is indicated by #, and between CON and ESC by *. D HE staining of the cerebral cortex and hippocampus. Scale bar, 50 μm. E Immunohistochemical staining and H-score assessment of colon sections for claudin-1, occludin, and ZO-1. Scale bar, 50 μm. Data are expressed as mean ± SEM. ^*P < 0.05, ^**P < 0.01, ^***P < 0.001, ns, no significance. Forced swim test A transparent cylindrical glass tank (20 × 60 cm) is filled with water to approximately two-thirds of its height, with the water temperature maintained at 24 ± 1 °C [[61]20, [62]21]. A backlight is placed behind the swim tank to prevent light reflection from affecting experimental outcomes. Rats are placed in the swim tank for a 5 min trial, with their activity continuously recorded by a camera. It should be noted that no pretest was performed before the initial 5 min trial. A pretest is typically used to acclimate animals to the testing environment, enhancing the consistency and reliability of behavioral data [[63]22]. This absence should be considered when interpreting and extrapolating the results of this study. The primary evaluation metric is the immobility time, defined as minimal movement just sufficient to keep the rat afloat, with occasional gentle movements to prevent drowning. FST is conducted both before (Day0) and 21 days after drug administration (Day22) to assess changes in depressive-like behaviors in rats. Hematoxylin-eosin staining The brain tissues were fixed in 4% paraformaldehyde solution for 48 h, followed by embedding in paraffin. Paraffin-embedded tissue sections were prepared, stained with hematoxylin and eosin (HE). Samples were immersed in alcohol, stained with hematoxylin for 5 min, immersed in alcohol for 1 min, stained with eosin for 15 s and re-immersed in alcohol and xylene. Then subsequently examined under a light microscope to observe the morphology and structure of the brain tissues and capture images. Immunohistochemistry The colon tissues were fixed and embedded in paraffin, and immunohistochemistry was employed to analyze the expression and distribution of colonic tight junction proteins. Paraffin-embedded tissue sections were deparaffinized through a series of rehydration steps and subjected to antigen retrieval. To block endogenous peroxidases, each slide was treated with 3% hydrogen peroxide for 25 min and subsequently blocked with 3% normal goat serum. Sections were incubated overnight at 4 °C with specific primary antibodies against claudin-1, occludin, and zonula occludens-1 (ZO-1). Following PBST washing, slides were incubated with secondary antibodies at room temperature for 1 h. Visualization was achieved using 3,3′-diaminobenzidine (DAB) chromogen, followed by counterstaining with hematoxylin. Immunohistochemistry scoring (H-score) was conducted to evaluate staining intensity: H-score = (percentage of weak intensity ×1) + (percentage of moderate intensity ×2) + (percentage of strong intensity ×3). The percentage of stained area and intensity of stained cells were graded as 0, negative; 1+, weak; 2+, moderate; 3+, strong [[64]23]. 16S rRNA gene sequencing analysis Fresh colonic content was collected into sterile tubes, rapidly frozen in liquid nitrogen, and stored at −80 °C pending further processing. Total genomic DNA extraction of microbial communities was performed, with PCR amplification and library construction targeting the V3-V4 variable regions of the 16S rRNA gene. Sequencing was carried out on Illumina’s PE250 platform to generate 250 bp paired-end raw reads, and raw paired-end sequences were quality-controlled using fastp (version 0.19.6) and assembled using FLASH (version 1.2.11). Post-quality control, optimized sequences were denoised using the DADA2 plugin within the Qiime2 pipeline with default parameters to obtain amplicon sequence variants (ASVs). ASVs underwent sequence rarefaction [[65]24] for bioinformatics analysis, where α-diversity indices (Chao, Ace, Simpson, and Shannon) were compared using Kruskal-Wallis tests. Beta diversity analysis was visualized using principal coordinates analysis (PCoA) to assess differences in microbial community structure between groups. Furthermore, we employed the Wilcoxon rank-sum test to analyze the gut microbiota health index (GMHI) and the microbiota dysbiosis index (MDI). The GMHI, a composite index used to assess the overall health status of the gut microbiota, is generally considered a marker of gut health and ecological dysbiosis [[66]25]. The MDI serves as an index to determine the extent of ecological imbalance, with higher values indicating greater degrees of microbial dysregulation [[67]26]. Differential abundance of taxa between groups was determined using bar plot analysis and heatmap visualization. Taxonomic differences at the genus level between groups were identified using linear discriminant analysis (LDA) effect size (LEfSe) (LDA score >2, P < 0.05). Wilcoxon rank-sum tests and Kruskal-Wallis tests were used to respectively determine statistical differences in gut microbiota composition between two and three groups. Microbial functions were predicted using PICRUSt2 [[68]27] in conjunction with KEGG pathways. Metabolome Metabolites were extracted from serum samples. Briefly, each sample underwent metabolite precipitation with 400 μL of extraction solvent (methanol:water = 4:1). Solid samples were ground using a cryogenic tissue grinder for 6 min (−10 °C), followed by a 30 min freeze (5 °C) and a 30 min incubation at −20 °C. The supernatant was collected after centrifugation for 15 min (4 °C, 13,000 g) and subjected to LC-MS/MS analysis for metabolite identification. Chromatographic separation was performed using an ACQUITY HSS T3 column (100 mm * 2.1 mm, 1.8 μm, Waters Corporation, USA). Metabolites eluted from the column were detected using high-resolution tandem mass spectrometry on a UHPLC-Q-Exactive HF-X system (Thermo, USA) operating in both positive and negative ion modes. To assess the stability of LC-MS/MS throughout the collection process, a quality control sample (pool of all samples) was collected every 5–15 samples. Raw LC-MS/MS data were processed using Progenesis QI software (Waters Corporation, USA) to generate sample information, metabolite names, and mass spectrometry response intensity data. Metabolite annotation was conducted using the KEGG database (Kyoto Encyclopedia of Genes and Genomes, [69]https://www.kegg.jp/) and HMDB database ([70]http://www.hmdb.ca/). PCoA and Orthogonal Partial Least Squares Discriminant Analysis were performed. Significant differential metabolites were selected based on Variable Importance in Projection (VIP) values from the Orthogonal Partial Least Squares Discriminant Analysis model and student’s t-test (VIP > 1 and P < 0.05). Additionally, pathway enrichment analysis of differential metabolites was conducted using the KEGG database. Proteome Rat bilateral hippocampal tissues were extracted, rapidly frozen in liquid nitrogen, and stored at −80 °C. For protein extraction, hippocampal tissues were homogenized in protein lysis buffer, incubated on ice for 30 min with vortex mixing (5–10 s every 5 min), followed by centrifugation at 12,000 g for 30 min at 4 °C to collect the supernatant. Protein concentration in the supernatant was determined using the BCA assay. Enzymatic digestion and peptide quantification of proteins were performed, and liquid chromatography separation was carried out on an Ionopticks UPLC C18 Column (1.6 µm, 250 mm × 75 μm, Ionopticks, USA). The resulting fractions were analyzed using the timsTOF Pro2 mass spectrometer (Bruker, Germany) for LC-MS/MS analysis of protein samples. Group differences in protein expression were calculated using the t.test function in R, with significance set at P < 0.05 and fold change criteria of >1.2 or <0.83 for differential expression proteins. Gene Ontology (GO) database ([71]http://geneontology.org/) was used for annotation analysis of the differentially expressed protein (DEPs) in terms of biological processes, cellular components, and molecular functions. Differential pathway analysis of DEPs was conducted using the KEGG pathway database. The rich factor was calculated as the ratio of the number of differential proteins in a specific pathway to the total number of proteins in that pathway. Statistical analysis Statistical analyses were performed using GraphPad Prism version 9.0 (GraphPad Software Inc, La Jolla, CA, USA) and SPSS version 23.0 (IBM Inc, Armonk, NY, USA). Data are presented as mean ± SEM. For independent sample experiments, we used the Shapiro-Wilk test for normality and Levene’s test for homogeneity of variances. For normally distributed and equal variance data, differences between two groups were assessed with a two-tailed Student’s t-test, and differences among three groups were assessed with one-way ANOVA followed by Bonferroni post-hoc tests. For non-normally distributed data, we used the Wilcoxon rank-sum test for two-group comparisons and the Kruskal-Wallis H test with Dunn’s post-hoc for three-group comparisons. For analysis of body weight gain and immobility time in the forced swim test, we applied two-way repeated measures ANOVA to examine group effects (CON, WKY, ESC), time effects, and group-time interactions, using Bonferroni post-hoc for multiple comparisons. A P-value of <0.05 was considered statistically significant. Notably, for comparisons between WKY and ESC groups in microbial and protein data, we did not apply additional P-value corrections, intending to identify differences of potential biological significance (Supplementary Table [72]1). Spearman correlation analysis was employed to explore co-occurrence relationships among gut microbiota, metabolites, and proteins, and visualized using Cytoscape (version 3.7.2), displaying only edges with correlations >0.5 and P < 0.05. Results ESC alleviates depressive-like behaviors and gut-brain damage in WKY rats To assess the antidepressant effects of ESC, we utilized the FST to evaluate depressive-like behaviors in rats. In the FST, using a two-way repeated measures ANOVA, we found a significant interaction between group and time (F(2,14) = 11.310, P = 0.001). At baseline, there was a significant difference in immobility time among the three groups (F(2, 14) = 16.227, P < 0.001). After the intervention, the difference in immobility time between the CON and the ESC group was not statistically significant (P = 0.272). Only the ESC group showed a significant reduction in immobility time before and after treatment (F(1, 7) = 36.902, P = 0.001). No significant changes were observed in the CON and WKY groups before and after treatment. The above results indicate that ESC treatment can alleviate depression-like behaviors, particularly behaviors associated with despair (Fig. [73]1B). We also employed a two-way repeated measures ANOVA to assess changes in body weight. The results indicated that the interaction between treatment and time was not statistically significant (F(4, 28) = 0.893, P = 0.481). However, significant differences in body weight between treatment groups were observed on day 7 (F(2, 14) = 27.449, P < 0.001), with significant differences between the CON group and the WKY group (P = 0.012), and between the CON group and the ESC group (P < 0.001), while the difference between the WKY and ESC groups was not significant (P = 0.099). Similar significant differences between groups were observed on days 14 and 21. In terms of the effect of time, all groups exhibited significant changes in weight at each of the three time points (F values of 320.284, 139.878, and 265.814, all P < 0.001), and there were significant differences in the rate of weight gain over time. Body weight measurements revealed that both the WKY and ESC groups exhibited slower weight gain compared to the control group (Fig. [74]1C). To further evaluate the impact of ESC on the MGB axis, we conducted histopathological examinations of brain and colon tissues in rats. HE staining of brain tissues showed that the control group had abundant, regularly shaped, and densely packed cortical and hippocampal neurons with no apparent abnormalities. In the WKY group, more neurons exhibited shrunken and deeply stained appearances in the cortex, with a few similarly affected neurons in the hippocampus. In the ESC group, fewer cortical neurons were shrunken and deeply stained, and hippocampal structure appeared normal (Fig. [75]1D). ESC also had a mitigating effect on intestinal barrier integrity; claudin-1 and ZO-1 protein expressions were significantly reduced in the WKY rats compared to the control group (P < 0.05), but markedly improved after ESC treatment (Fig. [76]1E). Intestinal homeostasis in WKY rats was affected by ESC We characterized changes in the gut microbiota induced by ESC treatment through 16S rRNA sequencing of rat colonic contents. The control group exhibited 1928 ASVs, whereas the WKY group showed a reduction in ASVs (1412). Additionally, the ESC group displayed increased ASV counts (1709), though still lower than the CON group (Fig. [77]2A). Alpha diversity indices Ace, Chao, Shannon, and Simpson were used to evaluate microbial diversity. Results indicated that the WKY group had lower Ace and Chao indices compared to CON, and higher Simpson index, suggesting reduced richness and diversity of microbial communities in rats with depression-like behavior. ESC intervention appeared to influence this trend, albeit not statistically significant (Fig. [78]S1A). PCoA analysis assessed differences in intestinal microbiota composition among groups, our results indicate significant differences in microbial β-diversity among the three groups (Fig. [79]2B). Specifically, the PCoA analysis based on the weighted UniFrac distance revealed significant differences along the PC2 axis (explaining 18.62% of the variation) between the CON and WKY groups, as well as between the ESC and WKY groups (Supplementary Table [80]1). However, no significant differences between the ESC and WKY groups were observed along the PC1 axis (explaining 35.28% of the variation). Moreover, the PCoA analysis based on the Bray-Curtis distance showed that the CON group could be significantly separated from both the ESC and WKY groups, while no significant differences were observed between the ESC and WKY groups (Fig. [81]S1B). Thus, our findings suggest that ESC may partially alter the intestinal microbial diversity in the WKY rats. Subsequently, we analyzed GMHI and MDI. Both WKY and ESC groups showed significant decreases in GMHI compared to CON (P < 0.05, Fig. [82]2C, Fig. [83]S1C). Moreover, MDI was higher in both WKY and ESC groups than in CON (Fig. [84]S1D, P < 0.05), with ESC showing a slight decrease compared to WKY (Fig. [85]2C). We further evaluated microbial composition at different taxonomic levels. Based on the analysis of compositional ratios, at the phylum level, Firmicutes predominated in all groups, followed by Bacteroidota. Firmicutes were most abundant in CON (92.99%), followed by WKY (88.54%) and ESC (85.83%). Bacteroidota was highest in ESC (11.41%), followed by WKY (9.22%) and CON (3.83%) (Fig. [86]2D). At the genus level, Turicibacter and Blautia are found in higher proportions in the WKY and ESC groups compared to the CON group. Additionally, the proportion of Lactobacillus is highest in the CON group, followed by the ESC group, and lowest in the WKY group (Fig. [87]2D). Using LEfSe analysis, we identified consistent microbial changes between CON vs. WKY and CON vs. ESC, with 40 and 41 differential microbes identified, respectively (Fig. [88]S2A). Analysis of these differential microbes revealed 31 common differential species. A heatmap depicted the expression abundance and LDA values of these 31 differential microbes across the three groups (Fig. [89]S2B). Representatively, Bacillus and Lachnospiraceae_NK4A136 were predominant in CON (LDA > 4), while Turicibacter and Blautia were predominant in WKY and ESC groups (LDA > 4, Fig. [90]S2C). Furthermore, we conducted LEfSe analysis between WKY vs. ESC to identify important ESC-influenced microbes (Fig. [91]2E). Results revealed differential abundance at various taxonomic levels, including 1 at the phylum level (Patescibacteria), 1 at the class level (Saccharimonadia), 4 at the order level (Staphylococcales, Saccharimonadales, Corynebacteriales and Bifidobacteriales), 6 at the family level (Staphylococcaceae, Saccharimonadaceae, Rikenellaceae, Marinifilaceae, Corynebacteriaceae and Bifidobacteriaceae), 11 at the genus level (unclassified_f__Anaerovoracaceae, UCG-005, Staphylococcus, Odoribacter, norank_f__Christensenellaceae, Intestinimonas, Corynebacterium, CHKCI002, Candidatus_Saccharimonas, Bifidobacterium and Alistipes), and 17 at the species level (Only Bacteroides barnesiae has complete annotation information). Notably, Bacteroides barnesiae at the species level showed significantly higher expression in ESC than in WKY (Fig. [92]2F). The PICRUSt2-based functional predictions provide insights into the microbial community’s functional capabilities by assessing the composition and abundance of functional attributes. The KEGG pathways predicted by PICRUSt2 indicated significant differences between the WKY and ESC groups in the level 2 pathways, such as biosynthesis of other secondary metabolites and glycan biosynthesis and metabolism. Specifically, at the level 3 pathway within lipid metabolism, fatty acid degradation and glycerolipid metabolism were significantly enriched in the WKY group, whereas sphingolipid metabolism was significantly enriched in the ESC group (Fig. [93]2G, H). Fig. 2. Characteristics of microbial communities in colon contents. [94]Fig. 2 [95]Open in a new tab A Veen diagram of ASVs in three groups. B PCoA of the three groups. C Analysis of differences in MDI (left) and GMHI (right) between WKY and ESC groups. D Bacterial taxonomic analysis at the phylum level across the three groups (left) and bacterial taxonomic analysis at the genus level across the three groups (right). E Differential microbiota between the WKY and ESC groups based on LEfSe analysis (LDA > 2.0, P < 0.05). LDA effect size showing the most differentially significant abundant microbiota between WKY and ESC groups. F Bar graph showing the abundance of differential microbiota at the genus and species levels. G Microbiota function predicted based on the KEGG database (at level II) by using PICRUSt2. H Microbiota function predicted based on the KEGG database (at level III) by using PICRUSt2. ^*P < 0.05, ^**P < 0.01, ^***P < 0.001. ESC affects the metabolic profile in rats with depression-like behavior To explore the potential association between ESC and its effects on serum metabolites relevant to communication within the MGB axis, we analyzed changes in serum metabolites. Initially, PCoA was used, revealing significant separation of metabolic profiles between the CON and WKY/ESC groups, while no significant separation was observed between the ESC and WKY groups (Fig. [96]3A). Subsequently, differential metabolites were screened based on a standard of P < 0.05 and VIP values > 1. Compared with the CON group, 104 metabolites were significantly upregulated and 89 metabolites were significantly downregulated in the WKY group (Fig. [97]3B). Similarly, 125 metabolites were significantly upregulated and 64 metabolites were significantly downregulated in the ESC group compared with the CON group (Fig. [98]3C). Additionally, there were 131 differential metabolites between the ESC and WKY group (Fig. [99]3D). Furthermore, compared to the CON group, there are 58 commonly upregulated and 105 commonly downregulated differential metabolites in both the WKY and ESC groups, exhibiting the same trend of changes in these groups (Fig. [100]3E). KEGG pathway annotations of the metabolites revealed that the majority belonged to Phospholipids (Fig. [101]3F). Further analysis of 131 metabolites affected by ESC intervention was conducted, there were 83 significantly upregulated metabolites and 48 significantly downregulated metabolites in the ESC group compared with the WKY group (Fig. [102]3G). The top 5 differential metabolites based on fold change in the ESC group were Allylestrenol, Mipafox, P-Tolyl Sulfate, 2-Hydroxypyridine and P-Cresol glucuronide. The top 5 differential metabolites based on fold change in the WKY group were CDP-DG(18:2(10E,12Z)+=O(9)/20:3(8Z,11Z,14Z)), CDP-DG(18:1(9Z)/PGJ2), PS(15:0/20:3(8Z,11Z,14Z)), PGP(20:4(6Z,8E,10E,14Z)-2OH(5S,12 R)/i-19:0) and Leukotriene F4. Metabolite enrichment analysis was performed on differential metabolites between the WKY and ESC groups. Results showed that the top two pathways by P value were Glycerophospholipid metabolism and Sphingolipid metabolism (Fig. [103]3H). Within the Glycerophospholipid metabolism pathway, 15 metabolites were enriched; compared to the WKY group, glycerylphosphorylcholine, LysoPC (18:0/0:0), PC (18:0/22:6 (4Z,7Z,10Z,13Z,16Z,19Z)), and PS (20:4 (8Z,11Z,14Z,17Z)/22:2 (13Z,16Z)) were expressed at higher levels in the ESC group, while the remaining 11 differential metabolites were downregulated. In the Sphingolipid metabolism pathway, three metabolites, Sphinganine 1-phosphate, Sphingosine, and Sphingosine-1-phosphate, were highly expressed in the ESC group (Fig. [104]3I). These data indicate that ESC intervention promotes changes in serum metabolites in rats with depression-like behavior. Fig. 3. Serum Metabolomic Features. [105]Fig. 3 [106]Open in a new tab A Principal Coordinate Analysis (PCoA) plots for CON, WKY, and ESC groups. B Volcano plot for CON vs. WKY (red: increased in WKY group; blue: increased in CON group). C Volcano plot for CON vs. ESC (red: increased in ESC group; blue: increased in CON group). D Volcano plot for WKY vs. ESC (red: increased in ESC group; blue: increased in WKY group). E Venn diagram showing metabolites with the same trends between CON vs. WKY and CON vs. ESC. F KEGG pathway annotations for 142 common differential metabolites. G Bar chart displaying the Log2FC of differential metabolites (red: increased in ESC group; blue: increased in WKY group); and heatmap of differential metabolites between WKY and ESC groups. High expression is marked in red, and low expression is marked in blue. The “value” represents the relative expression value (Z-score) obtained after normalization. H KEGG topology analysis of differential metabolites between WKY and ESC groups. The impact value represents the importance of metabolites in the metabolic pathway’s topology, while the P-value reflects the statistical significance of the metabolic pathway. The horizontal coordinate and size of the bubble represent the impact factor of the pathway in the topology analysis, with larger bubbles indicating a larger impact factor. The vertical coordinate and color of the bubble represent the P-value of the enrichment analysis, with darker colors indicating lower P-values and greater enrichment significance. I Heatmap of differential metabolites in Glycerophospholipid metabolism and Sphingolipid metabolism, with high expression shown in red and low expression shown in blue. The “value” represents the relative expression value (Z-score) obtained after normalization. Higher positive values indicate higher expression, while larger negative values indicate lower expression. ESC influences rat proteome in hippocampus To investigate changes in the hippocampal proteome of WKY rats following ESC intervention, we conducted a proteomic analysis. Principal component analysis showed no significant separation of protein profiles among the three groups (Fig. [107]4A). However, differential proteins between groups were identified based on a fold change criterion of >1.2 or <0.83 and a P-value < 0.05 for further analysis. The results indicated that there were 127 upregulated DEPs and 231 downregulated DEPs in the WKY group (Fig. [108]4B), and 94 upregulated DEPs and 244 downregulated DEPs in the ESC group compared with the CON group (Fig. [109]4C). Meanwhile, 103 DEPs were upregulated and 160 DEPs were downregulated in the ESC group compared with the WKY group (Fig. [110]4D). Differential proteins that were upregulated or downregulated in both WKY and ESC groups, as compared to the control group, were considered potentially due to genetic factors, as they were unaffected by the treatment (Fig. [111]4E). GO annotation analysis of these proteins identified 44, 78, and 100 entries in molecular function (MF), cellular component (CC), and biological process (BP), respectively. Fig. [112]4F depicts the top five enriched terms. MF analysis showed that most identified proteins are involved in binding to small molecules, proteins, and compounds. CC category analysis indicated that most proteins are predicted to be localized in organelles. Many proteins in the BP category are related to metabolic processes. KEGG pathway enrichment analysis identified 26 significantly enriched signaling pathways (P value < 0.05), with the alpha-Linolenic acid metabolism pathway showing the highest Rich factor (Rich factor = 0.375, P value = 0.0017), enriched with one upregulated protein Acc1a and two downregulated proteins Fads1 and Cyb5a (Fig. [113]4G). Further analysis was conducted on differential proteins between the ESC and WKY groups. GO annotation results revealed that most identified proteins in the MF analysis are involved in binding to proteins and organic cyclic compounds, while CC analysis showed most proteins are predicted to be located in organelles and cell membranes. BP analysis indicated that most proteins are related to metabolic processes (Fig. [114]4H). Additionally, KEGG enrichment analysis of differential proteins, in conjunction with the above metabolomics results, focused on proteins enriched in the Glycerophospholipid metabolism and Sphingolipid metabolism pathways. Two proteins in the Glycerophospholipid metabolism pathway, Lysophosphatidylcholine acyltransferase 1 (Lpcat1) and Pnpla7, and four proteins in the Sphingolipid metabolism pathway, Bin2a, Ectonucleotide pyrophosphatase/phosphodiesterase 5 (Enpp5), Serine palmitoyltransferase 1 (Sptlc1), and Ectonucleotide pyrophosphatase/phosphodiesterase 2 (Enpp2), were analyzed. Compared to the WKY group, Lpcat1, Enpp5, Sptlc1, and Enpp2 were significantly upregulated (P value < 0.05) in the ESC group, while Pnpla7 and Bin2a were significantly downregulated (P value < 0.05, Fig. [115]4I, J). Fig. 4. Proteomic Characteristics of the Hippocampus. [116]Fig. 4 [117]Open in a new tab A Principal component analysis of hippocampal proteomics for CON, WKY, and ESC groups. B Volcano plot for CON vs. WKY (red: increased in WKY group; blue: increased in CON group). C Volcano plot for CON vs. ESC (red: increased in ESC group; blue: increased in CON group). D Volcano plot for WKY vs. ESC (red: increased in ESC group; blue: increased in WKY group). E Venn diagram showing proteins with the same expression trends between CON vs. WKY and CON vs. ESC. F GO annotation analysis for 163 differentially expressed proteins. G KEGG enrichment analysis for 163 differentially expressed proteins. The bubble color indicates the p-value and the size indicates the number of proteins. The horizontal axis indicates the Rich factor, which means the ration of the number of DEPs that enriched in one pathway and the number of all annotated proteins under this pathway term. H GO annotation analysis of differential proteins between WKY and ESC. I Bar graph of protein expression enriched in the Glycerophospholipid metabolism pathway. J Bar graph of protein expression enriched in the Sphingolipid metabolism pathway. ^*P < 0.05, ^**P < 0.01. Co-occurrence analysis of intestinal bacteria, serum metabolites and hippocampal proteins To investigate the complex interactions among the gut microbiome, metabolites, and proteins, spearman correlation analysis was performed. The analysis assessed the correlations among differential microbes, metabolites, proteins and the immobility time of FST between the WKY and ESC groups, including 9 differential microbes, 18 differential metabolites, and 6 differential proteins. Correlation pairs with a coefficient greater than 0.5 or less than −0.5 and a P value below 0.05 were selected for presentation, revealing a network comprising eight interconnected clusters, including Bacteroidota, Actinobacteriota, Patescibacteria, Firmicutes, as well as metabolites and proteins enriched in the Glycerophospholipid metabolism and Sphingolipid metabolism pathways (Fig. [118]5A). Several key factors emerged within these clusters. For instance, Bacteroides barnesiae had the most connections (degree = 15), followed by Enpp2, Pnpla7, and Bin2a (degree = 10), Lpcat1 and PE-NMe2(15:0/20:2(11Z,14Z)) (degree = 8), Staphylococcus (degree = 7), and Sptlc1, Sphinganine 1-phosphate and PS(15:0/20:3(8Z,11Z,14Z)) (degree = 6) were involved (Supplementary Table [119]2). Bacteroides barnesiae showed a positive correlation with metabolites enriched in the Sphingolipid metabolism pathway (sphingosine-1-phosphate, sphingosine, and sphinganine 1-phosphate). Moreover, it negatively correlated with the protein Bin2a (r < −0.9, P < 0.05) in the Sphingolipid metabolism pathway, while positively correlating with Sptlc1 and Enpp2 (r > 0.8, P < 0.05). Enpp2 was positively correlated with sphingosine-1-phosphate, sphingosine, and sphinganine 1-phosphate (r > 0.8, P < 0.05), Enpp5 was positively correlated with sphingosine-1-phosphate, sphinganine 1-phosphate, LysoPC(18:0/0:0), PC(18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z)), PE-NMe2(15:0/18:1(11Z)) (r > 0.8, P < 0.05), whereas Bin2a showed negative correlations with these metabolites (r < −0.9, P < 0.05). Sptlc1 was positively correlated with sphingosine (r > 0.8, P < 0.05). Additionally, the immobility time of FST was positively correlated with 1-Palmitoylphosphatidylcholine, PC(16:0/18:3(9Z,12Z,15Z)), PE(15:0/22:1(13Z)), PE(22:6(4Z,7Z,10Z,13Z,16Z,19Z)/22:1(13Z)), PE-NMe2(15:0/20:2(11Z,14Z)), PE-NMe2(18:0/18:2(9Z,12Z)), PE-NMe2(18:1(9Z)/16:0), PE-NMe2(18:2(9Z,12Z)/16:0) and PS(15:0/20:3(8Z,11Z,14Z)), while it was negatively correlated with Candidatus_Saccharimonas, Bacteroides_barnesiae, UCG-005, Sphinganine 1-phosphate, Sphingosine-1-phosphate, Glycerylphosphorylcholine and LysoPC(18:0/0:0), PC(18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z)) (Supplementary Table [120]3). KEGG pathway diagrams indicated that Glycerophospholipid metabolism and Sphingolipid metabolism pathways were interconnected through the same metabolite (Phosphoethanolamine). Notably, Sptlc1 could positively promote the feedback generation of sphinganine 1-phosphate and sphingosine, and Enpp2/5 were involved in the catalysis of sphingosine, with sphingosine-1-phosphate being a downstream metabolite of sphingosine (Fig. [121]5B). These findings demonstrate that the gut microbe Bacteroides barnesiae, along with the phospholipid metabolism pathway and associated proteins Sptlc1 and Enpp2/5, may play a crucial role in the antidepressant efficacy of ESC. Fig. 5. Correlation analysis of differential microbiota, metabolites, proteins and FST. [122]Fig. 5 [123]Open in a new tab A Co-occurrence network analysis. Purple nodes represent microbiota, yellow nodes represent metabolites, blue nodes represent FST and green nodes represent proteins; the size of the nodes indicates the degree, or the number of connections. Lines between nodes represent Spearman negative (gray) or positive (red) correlations. B Map of key components in glycerophospholipid and sphingolipid metabolism pathways. Red indicates high expression in the ESC group, while blue indicates low expression in the ESC group. Discussion The interaction between the gut microbiome and the brain has increasingly become a focus of neuroscience research. It is well understood that the stability of the gut microbiota is important for maintaining host health, and specific changes in the gut microbiota, such as variations in alpha and beta diversity and alterations in the abundance of bacterial families like Christensenellaceae, Lachnospiraceae, and Ruminococcaceae, are associated with depression [[124]28]. Under stress and depression, the gut microbiota becomes disordered, adversely affecting the CNS and exacerbating the disease [[125]29]. Studies have confirmed that transplanting the fecal microbiota of patients with severe depression into germ-free mice induces depressive symptoms in the mice, whereas restoring the gut microbiota can alleviate depression [[126]30, [127]31]. In our study, we analyzed the changes in the gut microbiota response to ESC treatment in WKY rats and the associations between alterations in the gut microbiota and changes in the host metabolic profile and hippocampal proteome, to explore the potential mechanisms by which ESC exerts therapeutic effects on depressive-like phenotypes through the MGB axis. In this study, we employed a WKY rat depression model. The FST, a behavioral test widely used in preclinical studies, assesses depressive-like behaviors in animal models [[128]32]. ESC has been shown to ameliorate depressive-like behaviors in our study, as evidenced by the reduced immobility time in WKY rats during FST. Although the FST is a reliable measure of depressive-like behavior, the lack of general locomotor activity assessment may limit the interpretation of the results. Therefore, when testing antidepressants in WKY rats, incorporating behavioral paradigms not reliant on locomotor activity in addition to the FST may provide a more comprehensive evaluation of the depressive phenotype in WKY rats. Pathological findings suggest that ESC intervention may alleviate brain damage and reduce intestinal permeability in WKY rats, potentially decreasing the translocation of microbial metabolites or microbiota and the invasion of harmful substances into the CNS. The expression of tight junction proteins is a critical marker for assessing intestinal health and disease recovery. Occludin and claudins, the major components of intestinal tight junctions, regulate the barrier properties of the paracellular space by forming tight junction strands. ZO-1 plays a key role in assembling intestinal tight junctions by linking transmembrane proteins to the intracellular actin cytoskeleton [[129]33]. Our study found that ESC intervention alleviated the underexpression of these three tight junction proteins, indicating that ESC can influence intestinal barrier function. ESC is a first-line antidepressant and anxiolytic medication [[130]34]. Most research on ESC’s antidepressant effects has focused on the serotonin transporter, which is responsible for the uptake of serotonin into serotonergic neurons; ESC inhibits this uptake, subsequently increasing serotonin levels at the postsynaptic membrane in the serotonergic synapse [[131]35, [132]36]. Interestingly, the therapeutic effects of ESC cannot be solely attributed to the inhibition of the serotonin transporter and involve multiple mechanisms of action. Studies have reported that alterations in the gut microbiome, characterized by changes in microbial diversity and imbalances in key microbial taxa, are observed in patients with depression [[133]30]. This suggests that investigating how antidepressants interact with these microbiome changes could be a promising direction for future research. To date, few studies have focused on the relationship between ESC’s therapeutic effects and the gut microbiome, and currently, there is no research on changes in the gut microbiota following ESC treatment in WKY rats. Therefore, our study aims to provide evidence for the antidepressant mechanisms of ESC through the perspective of the MGB axis. Through analysis of 16S rRNA sequencing data, we observed that the gut microbiota diversity in WKY rats was lower compared to the CON group, but there was an increase in both richness and diversity of the gut microbiota in the depressive group following ESC treatment. These results indicate a potential link between ESC treatment and changes in gut microbiota composition in WKY rats. The gut microbiota primarily consisted of the phyla Firmicutes and Bacteroidota, which have been reported to be the most affected in depression [[134]37, [135]38]. Additionally, our study found that the abundance of Bacteroidota was higher in the ESC group compared to the other two groups, while Firmicutes was less abundant. Antidepressant medications can influence the composition of the microbiome; higher abundances of Bacteroidetes and Proteobacteria and lower abundances of Firmicutes, Actinobacteria, and Fusobacteria have been observed during treatment with selective serotonin reuptake inhibitors or serotonin-norepinephrine reuptake inhibitors [[136]11]. The analysis of gut microbiota differences showed that 16 genera were significantly enriched in the WKY and ESC groups compared to the control group, and ESC intervention did not reverse the abundance of these bacteria, which may reflect potential genetic factors. Due to these genetic background differences, WKY rats inherently exhibit physiological and behavioral characteristics that differ fundamentally from those of the CON group, potentially directly affecting their microbiota structure. Future studies should consider using control and experimental groups with the same genetic backgrounds combined with depression modeling to validate the consistency and reproducibility of results. Our results identified 11 distinct microbes at the genus level and 17 at the species level between the WKY and ESC groups, of which 8 genera and 1 species had clear annotation information. Firmicutes included Intestinimonas, Staphylococcus, and UCG-005, while Bacteroidota included Alistipes, Odoribacter, and Bacteroides barnesiae. It was found that Intestinimonas, Alistipes, and Odoribacter, which produce SCFAs, correlate negatively with the severity of depression and increase in abundance following antidepressant treatment [[137]11, [138]39]. Consistently, in our study, the abundance of these three bacteria increased following ESC intervention. Furthermore, in the ESC group, the abundance of Staphylococcus and UCG-005 decreased. Staphylococcus can produce toxins and staphylococcal enterotoxins, which stimulate the vagus nerve to send signals to the brain, potentially inducing disease behavior [[139]40]. In this study, UCG-005 belongs to the Oscillospiraceae family, which has been reported to be a key pathogenic gut bacterium associated with the development of colitis [[140]41]. It is noteworthy that Bacteroides_barnesiae is the only bacterium at the species level that has been clearly annotated. It belongs to the Bacteroides, which is one of the most abundant Gram-negative genera in the human gut. Bacteroides acts as a commensal bacterium that can establish a stable relationship with the host and play a potentially probiotic role. It is considered to be a major synthesiser of vitamin K and also maintains host intestinal homeostasis by regulating the levels of SCFAs [[141]42]. Interestingly, we observed an increase in the abundance of Bacteroides barnesiae in the ESC group compared to the WKY group. Moreover, correlational analysis revealed that the abundance of Bacteroides barnesiae is negatively associated with immobility time in the FST. These results suggest a potential association that might contribute to understanding the complex interactions within the microbiota-gut-brain axis in depressive-like behaviors. Our study utilized a 3-week escitalopram regimen, based on prior rodent and human studies [[142]43–[143]48], which typically describe escitalopram treatment as chronic, ranging from 3 to 12 weeks [[144]13, [145]15, [146]49–[147]52]. This duration aligns with research indicating that chronic antidepressants like escitalopram and vortioxetine can reshape the gut microbiome in depression, impacting diversity and altering key microbial phyla [[148]15, [149]50, [150]52, [151]53]. While acute administration of escitalopram is less studied, evidence from other antidepressants suggests it also affects the gut microbiota. For instance, (S)-norketamine decreased harmful bacteria like Escherichia-Shigella and increased beneficial ones like Harryflintia 24 h after injection [[152]54]. Therefore, our findings highlight the influence of chronic antidepressant treatment on the gut microbiome, suggesting the need to also explore the impacts of acute dosing in future studies. The gut microbiome interacts with the CNS through microbial-derived metabolites and neurotransmitters such as gamma-aminobutyric acid and serotonin [[153]55]. In general, alterations in the composition of the gut microbiota may result in alterations in microbial metabolites, which may subsequently influence the entire metabolome of the host. In our research, we analyzed and compared the serum metabolomic profiles across groups to explore the mechanisms of communication along the MGB axis. Initial annotation analysis of differential metabolites between WKY and ESC group revealed that lipid metabolism abnormalities are one of the main manifestations. The CNS comprises three major classes of lipids: phospholipids, sphingolipids, and cholesterol. Lipids account for over 50% of the dry weight of the brain and their composition can influence perception and emotional behavior, potentially leading to conditions such as depression and anxiety [[154]56, [155]57]. Specifically, phospholipids and sphingolipids are closely linked to the development of neuropsychiatric disorders such as depression and anxiety [[156]58]. Plasma metabolites involved in lipid metabolism are key intermediates in the MGB axis [[157]59]. Studies have shown that traditional herbal medicines like kudzu can reverse depressive-like behaviors by regulating disturbances in phospholipid metabolism [[158]60]. Through the enrichment analysis of serum differential metabolites with or without ESC intervention, we found significant changes in glycerophospholipid and sphingolipid metabolism in the ESC group. The focus was particularly on the sphingolipid metabolism pathway, with sphingosine identified as a key differential metabolite. Research has linked elevated levels of sphingosine with reductions in depressive-like behavior and improvements in cognitive function [[159]61]. Sphingosine, a crucial intermediate in the sphingolipid metabolic pathway, can be phosphorylated to form sphingosine-1-phosphate, which is then cleaved into phosphoethanolamine [[160]62]. Many studies report that sphingosine-1-phosphate is involved in various neurological and psychiatric disorders, including Alzheimer’s disease [[161]63], depression, and anxiety [[162]64]. Sphingosine-1-phosphate has been identified as a pharmacological target in mental illness research, exemplified by FTY720 (fingolimod), an S1P receptor agonist that modulates neuroinflammation and is approved for treating multiple sclerosis [[163]65]. In our study, WKY rats exhibited extremely low levels of sphingosine, indicating an abnormality in sphingolipid metabolism. In the ESC group, we observed an increase in sphingosine levels, which may relate to the observed changes in depressive-like behaviors. Additionally, glycerophospholipid metabolism was another significant pathway enriched in differential metabolites with or without ESC intervention. Glycerophospholipids, critical components of neuronal membranes and brain phospholipids, play an essential role in regulating synaptic function and can be subdivided into phosphatidylserine (PS), phosphatidylethanolamine (PE), and phosphatidylcholine (PC). Our research observed changes in levels of PS, PE, and PC in the serum associated with ESC treatment, which may reflect alterations in glycerophospholipid metabolic pathways in the periphery. Although this study primarily focuses on the mechanisms by which ESC modulates sphingolipid metabolism through reshaping the gut microbiome, it is noteworthy that ESC might also affect sphingolipid metabolism through other pathways. Firstly, sphingolipid metabolism is closely related to immune responses and inflammatory states [[164]66], which are often exhibited in patients with depression [[165]67]. The anti-inflammatory properties of ESC may play a role by modulating the metabolism of sphingolipid signaling molecules associated with neuroinflammation [[166]68, [167]69]. Secondly, the role of antidepressants in reducing oxidative stress should not be overlooked [[168]70], as it may help maintain the lipid balance of neural cells, particularly the stability of membrane sphingolipids, which are essential for neuronal function and survival [[169]71]. Lastly, our study’s data also show that ESC has impact on the glycerophospholipid metabolism pathway (Fig. [170]3I), which may further affect the synthesis and degradation of sphingolipids, impacting neural cell function [[171]72]. The presence of these potential mechanisms suggests a multifaceted regulatory role of ESC on sphingolipid metabolism, providing new directions for future research. The hippocampus is a critical brain region for the generation and regulation of emotions and cognition, and depression is commonly characterized by memory loss and cognitive impairments, indicating hippocampal neuronal dysfunction as a pathophysiological feature of this disorder [[172]73, [173]74]. In our study, pathological analysis revealed hippocampal neuronal atrophy [[174]75]. Given that proteins are the primary executors of physiological functions in organisms, we conducted a proteomic analysis of the hippocampus to enhance our understanding of the antidepressant mechanisms of ESC. Based on our microbiota and metabolomics findings, we are particularly interested in proteins related to sphingolipid metabolism. The analysis revealed significant differences in sphingolipid metabolism-related proteins between the WKY and ESC groups. Specifically, Sptlc1, Enpp5, and Enpp2 were significantly enriched in the ESC group, while Bin2a was significantly enriched in the WKY group. Notably, ENPP5 (Ectonucleotide Pyrophosphatase/ Phosphodiesterase 5) showed a significant negative correlation with immobility time in the FST. ENPP5 is an enzyme that plays a crucial role in extracellular signal transduction. Studies have confirmed that ENPP expression in immature oligodendrocytes can enhance myelin regulatory factors via the mTOR signaling pathway, providing oxidative damage protection to myelin membranes and thereby reducing the risk of psychiatric disorders such as depression [[175]76]. Additionally, members of the ENPP family influence neuroprotection and damage responses by regulating autophagy and generating lysophosphatidic acid (LPA), which affects neuronal signaling in neurodegenerative diseases [[176]77]. This suggests that ENPP proteins play a critical role in maintaining neural homeostasis and mental health, highlighting the potential of this protein family as emerging targets for psychiatric disorders like depression. Finally, to further elucidate the potential role of the microbiota-gut-brain (MGB) axis in the antidepressant mechanism of escitalopram (ESC), we performed multi-omics correlation analysis. In our study, co-occurrence network analysis revealed a significant increase in the gut microbiota Bacteroides barnesiae in ESC group, which was strongly positively correlated with key sphingolipid metabolism-related metabolites (such as Sphingosine and Sphingosine-1-phosphate) in the serum, as well as with Sptlc1, a protein associated with sphingolipid metabolism in the hippocampus. Increased activity of Sptlc1 promotes the production of sphingosine and Sphinganine 1-phosphate, which essential for neuroprotection [[177]78]. Correlation analysis revealed that UCG-005 was positively correlated with FST, while Candidatus_Saccharimonas and Bacteroides barnesiae were negatively correlated. Eighteen differential metabolites were identified, with positive correlations found with 1-Palmitoylphosphatidylcholine, PC(16:0/18:3(9Z,12Z,15Z)), PE(15:0/22:1(13Z)), PE(22:6(4Z,7Z,10Z,13Z,16Z,19Z)/22:1(13Z)), PE-NMe2(15:0/20:2(11Z,14Z)), PE-NMe2(18:0/18:2(9Z,12Z)), PE-NMe2(18:1(9Z)/16:0), PE-NMe2(18:2(9Z,12Z)/16:0), and PS(15:0/20:3(8Z,11Z,14Z)); and negative correlations with Sphinganine 1-phosphate, Sphingosine-1-phosphate, Glycerylphosphorylcholine, LysoPC(18:0/0:0), and PC(18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z)). Six differential proteins were identified, with Enpp5 showing a negative correlation with FST. The correlation results support the potential association of these biomarkers with the improvement of depressive symptoms. Previous studies have shown that Bacteroides can modify sphingolipids through its metabolic activities, influencing the biosynthesis and degradation pathways of sphingolipids [[178]79, [179]80]. Sphingolipid metabolites and related proteins play a crucial role in regulating neuronal function and maintaining neural health [[180]81]. These findings suggest that Bacteroides barnesiae may indirectly promote the antidepressant effects of ESC by modulating sphingolipid metabolism. Specifically, by increasing the levels of sphingosine and sphingosine-1-phosphate, Bacteroides barnesiae may enhance the function of Sptlc1 and Enpp5, key proteins in the sphingolipid metabolic pathway that are closely associated with neuroprotection and anti-inflammatory effects. In this way, Bacteroides barnesiae not only impacts the gut environment as a gut microbiota member but may also play an important role in the antidepressant effects of ESC by modulating key neuro-metabolic pathways. This study has several limitations: (1) The analysis of gut microbiota was conducted using 16S sequencing. Although this method is effective for taxonomic identification, it offers lower resolution and less comprehensive functional insights compared to metagenomic sequencing. Future studies employing metagenomics and other sequencing techniques could effectively address these limitations; (2) The conclusions of this study are largely based on correlational analyses, necessitating cautious interpretation. Future interventions using fecal microbiota transplantation (FMT) or targeted microbial interventions may serve as effective approaches to explore causal relationships; (3) We did not apply traditional multiple comparisons adjustments to the microbial and protein comparisons between WKY and ESC groups, aiming to uncover biologically significant differences. However, this may increase the risk of false positives. Future studies will involve more rigorous validation to confirm these findings; (4) In this study, the choice of intraperitoneal administration of escitalopram differs from the commonly used oral route in clinical settings. This route of administration may not fully replicate the effects of oral administration on the microbiota, and thus might limit the direct extrapolation of our results to clinical application. Future studies specifically testing oral administration of escitalopram are needed to more accurately assess its effects on the gut-brain axis; (5) This study included only male subjects, and did not investigate potential sex-related differences that might affect the results. Future research should include female subjects to ensure broader applicability and relevance of the findings. Conclusion In summary, our study explored new mechanisms underlying the antidepressant efficacy of ESC. We utilized WKY rats as a model for depression and assessed the antidepressant effects of ESC using the forced swim test. Histological examination using Hematoxylin and Eosin (HE) staining and immunohistochemistry (IHC) assessed the impact of ESC on intestinal permeability and brain damage. Additionally, multi-omics analyses were conducted to elucidate the pivotal role of gut microbiota in this context. We observed that ESC significantly increased the abundance of certain bacteria, including Bacteroides barnesiae. Furthermore, in the ESC group compared to the WKY group, there were notable alterations in the serum metabolome and hippocampal proteome, which influenced key metabolites and proteins involved in sphingolipid metabolism, such as sphinganine 1-phosphate, sphingosine, sphingosine-1-phosphate, Sptlc1, and Enpp2. The strong correlations among these key substances are consistent with the hypothesis that ESC may influence the microbiota-gut-brain axis, potentially associated with depressive-like behaviors via the Bacteroides barnesiae/sphingolipid metabolism pathway. The discovery of this gut-brain interaction provides potential targets for antidepressant therapy, potentially optimizing treatment outcomes by regulating specific gut microbiota and sphingolipid metabolic pathways. Future research should further explore the specific roles of these differential microbiota, metabolites and proteins in the treatment of depression, providing a scientific basis for developing new therapeutic strategies. Supplementary information [181]Supplementary information^ (14.1KB, docx) [182]Figure S1^ (7.3MB, tif) [183]Figure S2^ (11.6MB, tif) [184]Supplementary tables^ (21.9KB, docx) Acknowledgements