Abstract Depression, a major global health issue, is closely associated with imbalances in gut microbiota and altered intestinal functions. This study investigates the antidepressant potential of a composite of Geniposide (GP) and Nanocrystalline Cellulose (NCC), focusing on its effects on the gut-brain axis. Utilizing network pharmacology, GP was identified as a key compound targeting the BCL2 gene in depression management. Experimental approaches, including a chronic unpredictable mild stress (CUMS) model in mice, cellular assays, and fecal microbiota transplantation (FMT), were used to evaluate the composite’s effectiveness. Results indicate that GP activates the adenosine monophosphate-activated protein kinase (AMPK) pathway by upregulating BCL2, enhancing intestinal barrier integrity, and balancing gut flora. These mechanisms contribute to its positive effects on hippocampal function and depressive-like behaviors in mice, suggesting that the GP-NCC composite could be a promising avenue for developing depression therapies that target gut health. graphic file with name 42003_2025_7934_Figa_HTML.jpg Subject terms: Biotechnology, Diseases __________________________________________________________________ NCC-GP complex enhances gut-brain axis modulation to treat depression. It strengthens the intestinal barrier, balances gut microbiota, and activates the BCL2-AMPK pathway, offering a novel therapeutic approach. Introduction Depression is a prevalent mental disorder that significantly impacts the quality of life and social functioning of individuals^[34]1–[35]3. With the rise in societal stress and lifestyle changes, the incidence of depression is increasing and has emerged as a crucial issue in global health^[36]4. Research indicates that the development of depression is closely associated with various factors, including genetics, environment, and lifestyle, necessitating comprehensive regulation and treatment^[37]5–[38]7. Geniposide (GP), a vital active ingredient of Gardenia jasminoides J. Ellis traditional medicine, exhibits diverse biological activities and has been found to have a positive impact on mood regulation, cognitive enhancement, and anti-inflammatory effects^[39]8,[40]9. Simultaneously, nanocrystalline cellulose (NCC), known for its excellent biocompatibility and drug delivery performance, is increasingly being utilized in the pharmaceutical field for drug applications^[41]10. Leveraging the advantages of GP and NCC, exploring a novel therapeutic strategy for depression by combining the two holds significant research significance. Recent studies have revealed a close association between intestinal flora, intestinal barrier function, and the onset and progression of Depression^[42]11–[43]13. The gut, being one of the body’s largest immune organs, plays a vital role in maintaining the balance of intestinal flora, intestinal barrier function, and neurotransmitters^[44]14–[45]16. Imbalanced intestinal flora and compromised intestinal barrier function may lead to inflammatory responses and neurological abnormalities, subsequently affecting mood and behavior^[46]17–[47]19. The rise of network pharmacology has brought new perspectives and methodologies to traditional medicine research^[48]20,[49]21. In this study, through network pharmacology screening, GP was identified as the primary active component for treating Depression in Gardenia jasminoides J. Ellis. Furthermore, the regulatory role of GP’s target, BCL2, is crucial in depression treatment^[50]22. BCL2’s modulation plays a significant role in processes such as cell apoptosis and inflammatory responses, thus warranting further investigation into its role in Depression^[51]23,[52]24. A comprehensive research approach was undertaken. Initially, utilizing network pharmacology, the main active component and key targets of GP in treating depression were identified, followed by the construction of a mouse chronic unpredictable mild stress (CUMS) model. Sequencing data and proteomic data of mouse intestinal tissue pre-and post-GP treatment were obtained using high-throughput sequencing and proteomics techniques, enabling the identification of key factors related to CUMS^[53]25. The aim of this study is to elucidate the molecular mechanisms of the GP and NCC composite in alleviating depression-like behaviors by improving intestinal barrier function and restoring microbial balance to exert a positive effect on depression. Through experimentation, it was demonstrated that NCC-GP can balance intestinal flora, repair the intestinal barrier, and affect the brain’s neurological system via the gut-brain axis, thereby holding potential value and scientific significance for treating depression. This research provides valuable insights into new approaches and strategies in depression therapy, offering a scientific basis for clinical applications. Results Analysis of the potential mechanisms of GP in the treatment of depression based on network pharmacology Depression, also known as depressive disorder, is characterized by persistent and prominent mood disturbances and cognitive impairments. Patients often exhibit suicidal tendencies^[54]26–[55]28. According to the World Health Organization, the global prevalence of depression is expected to reach 15%-20%^[56]29,[57]30, with high rates of disability and mortality. It is one of the leading causes of Disability-Adjusted Life Years (YLD) loss^[58]31. Gardenia jasminoides J. Ellis, a shrub in the Rubiaceae family, has been traditionally used in Chinese medicine for jaundice hepatitis, sprains, hypertension, diabetes, and other conditions^[59]32–[60]35. Recent studies have shown that Gardenia jasminoides J. Ellis and its bioactive components exhibit significant anti-tumor activity, can lower blood sugar and lipids, protect the liver and gallbladder, gastric mucosa, and improve depression symptoms^[61]36–[62]38. In order to investigate the potential application of Gardenia jasminoides J. Ellis and its bioactive components in depression, we conducted an in-depth analysis of its bioactive components and targets based on network pharmacology. The workflow of this analysis is depicted in Fig. [63]1A. Using OB > 10% and DL > 0.4 as selection criteria, we identified 19 effective bioactive components from Gardenia jasminoides J. Ellis in the traditional Chinese medicine systems pharmacology database and analysis platform (TCMSP) database (Table [64]S1). By mapping these effective bioactive components to their corresponding 867 target proteins in the Uniprot database, we obtained a total of 251 target genes. Subsequently, using the GeneCards database with “depression” as the key term, we screened depression-related genes with a Relevance score > 5 threshold. The intersection of depression-related genes and the target genes of effective bioactive components was considered the potential therapeutic target of the bioactive components (Fig. [65]1B). Fig. 1. Potential target screening and functional enrichment analysis of Gardenia jasminoides J. Ellis components for treating depression. [66]Fig. 1 [67]Open in a new tab A Illustrative Overview of the Bioinformatics Analysis Process; B Venn Diagram Showing the Intersection Genes between Depression-Related Genes and Gardenia jasminoides J. Ellis Active Compound Target Genes; C Network Illustrating the Drug-Active Component-Target-Depression Relationship of Gardenia jasminoides J. Ellis, where green and red colors denote Gardenia jasminoides J. Ellis and Depression, respectively, yellow signifies 52 active compounds with target genes, and blue represents 47 intersecting target genes; D Hierarchical Tree Map displaying the enrichment of target genes in different GO categories (Biological Process, Cellular Component, Molecular Function); E Network Diagram of KEGG Pathway Enrichment Analysis; F Protein Interaction Node Statistics Chart of the 47 intersecting target genes; G PPI Network Graph based on the Degree value of the 47 intersecting target genes, wherein larger nodes with redder color indicate higher importance in the network, and thicker, redder lines denote stronger interaction relationships. Subsequently, we selected 47 intersecting target genes as potential targets and integrated them with the 52 active compounds of Gardenia jasminoides J. Ellis into Cytoscape, resulting in the construction of a network model representing the Chinese herbal medicine-active compounds-target genes-Depression (Fig. [68]1C). Through this network, crucial active compounds and their target genes can be identified. To elucidate the roles of these potential therapeutic targets in the treatment of depression, functional enrichment analysis was conducted on these genes. The Gene Ontology (GO) analysis revealed that the BP of the potential targets was significantly enriched in cellular responses to oxidative stress, regulation of cell development, and positive regulation of monooxygenase activity, among others. The CCs were mainly enriched in transport vesicles, endoplasmic reticulum lumens, and phagocytic cups. Likewise, the MFs were primarily enriched in cytokine activity, cytokine receptor binding, and signaling receptor activator activity (Fig. [69]1D, Fig. [70]S1). Furthermore, based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, these genes were found to mainly participate in pathways such as the AMPK signaling pathway, Neurotrophin signaling pathway, and Dopaminergic synapse, which are potentially related to the occurrence and development of Depression (Fig. [71]1E). To facilitate the subsequent selection of key therapeutic targets, the aforementioned potential targets were subjected to protein-protein interaction (PPI) analysis using the String database. A PPI network graph was then redrawn based on the Degree value, revealing clear interaction relationships among the targets (Fig. [72]1F). A statistical graph based on the number of adjacent nodes showed that 11 targets had a Number of adjacent nodes > 30, namely IL6, AKT1, IL1B, INS, TNF, BCL2, FOS, BDNF, IL10, CXCL8, and IFNG. These genes were proposed as potential candidates for further exploration in the treatment of depression (Fig. [73]1G). Moreover, further analysis demonstrated that the 11 therapeutic targets corresponded to 13 active compounds (Table [74]S2). Among these 11 targets identified, only BCL2 was found to be a target of GP, prompting us to choose BCL2 for further investigation, with different pathways enriched by these compounds’ target genes. For example, the target genes of caffeic acid, chexanal, farnesol, and methyl palmitate were mainly associated with cell apoptosis and inflammation. On the other hand, the target genes of kaempferol, lauric acid, methyl palmitate, oleic acid, paeonol, quercetin, rutin, and ursolic acid were primarily involved in intracellular receptor signaling, receptor activity regulation, and hormone-mediated signal transduction pathways. The target genes of β-sitosterol and GP were associated with multiple pathways like signal transduction, substance metabolism, cell cycle, and regulation of cell structure. Given that GP is involved in various neuro-signaling pathways, as well as depression-related apoptosis and inflammation pathways within the PPI network, we speculate that GP may serve as the main active component for treating depression. Research has shown that GP significantly improves some depressive-like behaviors in mice and rats, suppresses neuroinflammation, and enhances cognitive dysfunction and depressive/anxiety symptoms. Previous studies by the research group have also corroborated these findings^[75]39,[76]40. Thus, it is speculated that GP may serve as the main effective component for treating depression, with BCL2. BCL2 was prioritized due to its robust regulatory effects on apoptosis and intestinal barrier function, both of which are critical in the pathophysiology of depression. The crucial role of the gut-brain axis in depression treatment: evidence of the therapeutic effect of GP The interaction between the digestive system and brain function has emerged as a vital area of focus in psychiatric research in recent years. These multifaceted interactions occur within the microbiota-gut-brain axis. The microbial community throughout the entire gastrointestinal tract, known as intestinal flora, closely correlates with anxiety and depressive disorders^[77]41,[78]42. Emerging scientific evidence indicates that the intestinal flora plays a significant role in regulating the central nervous system (CNS)^[79]43. During negative emotional states such as stress, anxiety, or depression, there is an increase in pro-inflammatory markers expression and alterations in the intestinal flora and gut permeability, leading to dysregulation of the gut-brain axis, which subsequently causes dysfunction in the CNS^[80]44. To validate the therapeutic effect of GP on Depression, we initially established a CUMS animal model, with the CUMS procedures conducted prior to behavioral tests such as the FST and SPT. Subsequently, we assessed the success of modeling through the tail suspension test (TST), sucrose preference test (SPT), forced swim test (FST), Morris water maze (MWM), and histopathological staining of the hippocampal tissues in mice (Fig. [81]2A). The results consistently demonstrated that, compared to the control group, the CUMS-induced mice exhibited significantly reduced sucrose preference and memory but increased tail suspension and forced swim times, indicative of pronounced depressive-like behavior. The hippocampal staining results showed significant changes in the hippocampal structure of the depression mice induced by CUMS, with a notable increase in apoptotic neurons and a significantly lower proportion of neuron cells compared to the control group (Fig. [82]S2A-G). Fig. 2. Validation of the therapeutic effect of GP on Depression. [83]Fig. 2 [84]Open in a new tab A Overview of Mouse Depressive-Like Behavior Detection (Created by BioRender); B–E Evaluation of depressive-like behavior in mice by TST, SPT, FST, and MWM experiments; F H&E staining results of the hippocampal tissues of mice in various groups, Scale bar=250 / 25 μm for the leftmost column, and Scale bar=25 μm for the remaining two columns; G Assessment of neuron cell counts in the hippocampal tissues of mice across different groups by Nissl staining, Scale bar=250 / 25 μm; H TUNEL staining to detect neuron cell apoptosis in the hippocampal tissues of mice in different groups, Scale bar=250/ 25 μm; One-way ANOVA was used for comparisons among different groups; I Immunohistochemical analysis of GFAP and BDNF expression in hippocampal tissues, Scale bar=50μm; J Weight changes in mice from each group; K ELISA analysis of inflammatory factor content in colonic tissues of mice in each group; L Immunohistochemical evaluation of E-cadherin content in colonic tissues of mice in various groups, Scale bar=100 μm; M H&E staining results of colonic tissues of mice in different groups, Scale bar=100 μm; N–Q Assessment of depressive-like behavior in mice by TST, SPT, FST, and MWM experiments. For the comparison of two sets of data, the independent samples t-test was utilized, whereas for the comparison of data among multiple groups, ANOVA was employed. To compare data across different time points within each group, two-way ANOVA was conducted. The line between groups represents the specific p-value, with p < 0.05 indicating a statistically significant difference between groups, with 6 mice in each group. In the colonic epithelial tissue, E-cadherin plays a critical role in maintaining cellular barrier function and structural integrity^[85]45. Subsequent examinations of the mice revealed changes in body weight and colonic tissue induced by CUMS; the results showed that CUMS-induced mice experienced a significant decrease in body weight compared to the control group. In the colonic tissue, levels of TNF-α, IL-1β, and IL-6 were markedly elevated by enzyme-linked immunosorbent assay (ELISA) (Fig. [86]S2I). The H&E staining results showed that the colonic tissue of CUMS mice exhibited inflammatory cell infiltration in the lamina propria, goblet cell loss, mucosal hyperplasia, crypt abscesses, and crypt ulcers. Immunohistochemistry results revealed a substantial decrease in E-cadherin content in the colonic epithelial tissue of mice (Fig. [87]S2H–K). Furthermore, follow-up FMT experiments confirmed that alleviation of depressive behavior in CUMS-induced mice occurred when they were orally administered fecal pellets from the control group, underscoring the pivotal role of intestinal flora in the onset and progression of Depression (Fig. [88]S2L–O). Subsequently, we treated the successfully CUMS modeled mice with 100 mg/kg of GP. The selective serotonin reuptake inhibitor (SSRI) fluoxetine was used as a positive control for comparison and evaluation. Behavioral tests revealed a significant therapeutic effect of GP on the mice compared to the DMSO group. The therapeutic effects of the SSRI fluoxetine were comparable to those of GP, with no significant differences (Fig. [89]2B–E). H&E staining results demonstrated that fluoxetine, as a positive control, exhibited significant therapeutic effects compared to the negative control. Additionally, treatment with GP significantly alleviated hippocampal lesions in mice. Nissl staining and TUNEL staining results showed a significant increase in the proportion of neuronal cells in the hippocampal tissue of mice post-GP or fluoxetine treatment, accompanied by a marked reduction in apoptotic cell numbers (Fig. [90]2F–H). In the CUMS model, GFAP and BDNF immunoexpression were significantly reduced. GP or fluoxetine treatment markedly increased the expression of these key neuroproteins (Fig. [91]2I). Furthermore, GP or fluoxetine treatment resulted in a significant increase in mouse body weight and a significant alleviation of inflammation in colonic tissue, with a notable elevation in E-cadherin content (Fig. [92]2J–L). Moreover, FMT experiments showed that administering fecal pellets from GP-treated mice to the DMSO-treated mice relieved depressive behavior in both groups (Fig. [93]2N–Q). Overall, these findings collectively suggest that GP can treat depression by repairing the intestinal barrier, balancing intestinal flora, and further impacting the hippocampal tissue in the brain. We speculate that GP alleviates depressive symptoms by modulating GFAP and BDNF expression in the hippocampus. Mechanistic analysis of the therapeutic effect of GP on depression: BCL2 as a key downstream target Studies have shown that BCL2 may play a potential role in improving intestinal barrier dysfunction^[94]46. Recent research indicates a close relationship between the gut-brain axis and neurological disorders such as depression^[95]47,[96]48. To analyze GP’s target in improving intestinal dysfunction associated with depression, tandem mass tag (TMT)-labeled quantitative proteomics was used to analyze colonic tissues from six mice treated with DMSO or 100 mg/kg GP (GP100). A total of 10,971 proteins were identified, yielding a set of 108 differentially expressed proteins (DEPs), including 38 upregulated and 70 downregulated proteins (Fig. [97]3A–C). High-throughput sequencing of colonic tissues from the DMSO and GP100 groups identified 306 differentially expressed genes (Fig. [98]3D, E). LASSO regression and multivariable Cox analysis identified gene set B (Fig. [99]3F, G), while a random forest algorithm was used to evaluate gene importance, yielding gene set C (Fig. [100]3H). Depression-related genes were extracted using SVM-RFE to obtain gene set D (Fig. [101]3I). The intersection of these sets revealed a key factor: BCL2 (Fig. [102]3J). Proteomics and transcriptomics confirmed significant upregulation of BCL2 expression after GP treatment, validated by reverse transcription quantitative polymerase chain reaction (RT-qPCR). When drug molecules interact with targets, they require active conformations with geometric and energy matching. Therefore, we employed molecular docking techniques to predict the binding of GP to the active pocket of BCL2. The detailed analysis process is illustrated in Fig. [103]3K. Initially, the 2D and 3D molecular structures of GP were obtained from the PubChem database (Fig. [104]S3). Subsequently, molecular docking analysis of BCL2 and GP was conducted using software such as AutoDockTools 1.5.6 and Vina 1.5.6. The 3D model displayed the binding mode of the target protein receptor with the compound GP and its interactions with surrounding amino acid residues (Fig. [105]3L). Molecular docking demonstrated the binding affinity between GP and BCL2, with BCL2 upregulation confirmed by transcriptomics, proteomics, and RT-qPCR. Results from proteomics and transcriptomics studies demonstrated a significant upregulation of BCL2 expression levels following GP treatment, which was further validated using RT-qPCR in the colon tissues of mice treated with GP (Fig. [106]3M). Fig. 3. Machine learning algorithm and proteomics combined identification of key targets for GP treatment in depression. [107]Fig. 3 [108]Open in a new tab A Proportion of differentially expressed proteins (DEPs) between colonic tissues of three DMSO group mice and three GP100 group mice. B Comparison of the number of upregulated and downregulated DEPs, with red representing upregulated proteins and blue representing downregulated proteins, N = 6. C Heatmap showing the differential expression of the top 20 DEPs in the proteomics data, with DMSO (N = 3) and GP100 (N = 3) representing colonic tissue samples from three mice in each group. D Volcano plot of differentially expressed mRNA between colonic tissues of three DMSO group mice and three GP100 group mice in high-throughput sequencing data. E Heatmap showing the differential expression of selected DEGs in the high-throughput sequencing data, with DMSO (N = 3) and GP100 (N = 3) representing colonic tissue samples from three mice in each group. F–G Lasso coefficient screening plot. H Random forest algorithm result plot. I SVM-RFE algorithm result plot. J Venn diagram showing the overlap of Depression-related mRNA and DEPs identified by three machine learning algorithms: lasso regression, random forest, and SVM-RFE. K Schematic workflow of bioinformatics analysis in Fig. 3 (Created by BioRender). L Molecular docking result between GP molecule and target protein BCL2. M RT-qPCR analysis of BCL2 expression changes in colonic tissues of mice after GP treatment, analyzed by independent t-test (N = 6 mice per group). In the volcano plot, blue dots represent significantly downregulated mRNA in the GP100 group, red dots represent significantly upregulated mRNA in the GP100 group, and gray dots represent mRNA with no significant difference. The line between groups represents the specific p-value, with p < 0.05 indicating a statistically significant difference between groups. In conclusion, the aforementioned results indicate that BCL2 is a downstream target of GP in the treatment of depression. GP promotes intestinal barrier integrity by regulating BCL2 expression in colonic epithelial cells To validate whether BCL2 is a target of GP, we established an in vitro cell model of colonic epithelial cells induced by LPS in mice (Fig. [109]4A). ELISA and in vitro barrier integrity experiments showed a significant upregulation of inflammatory factors and a decrease in intestinal barrier integrity in colonic epithelial cells, confirming the successful model construction (Fig. [110]4B, C). RT-qPCR results revealed a significant downregulation of BCL2 expression in colonic epithelial cells after LPS induction, which was significantly upregulated upon treatment with GP (Fig. [111]S4A, B). Subsequently, we used lentivirus to knock down or overexpress BCL2, verifying lentiviral transfection efficiency using RT-qPCR and Western blot techniques. The results demonstrated a significant increase in BCL2 expression after overexpressing lentivirus, with the most significant knockdown efficiency observed in sh-BCL2-1, which was consequently chosen for further experiments (Fig. [112]S4C–F). Fig. 4. GP-mediated BCL2 expression alleviates depression. [113]Fig. 4 [114]Open in a new tab A Schematic diagram of in vitro cell model; B Determination of inflammatory factor levels in cells of each group using ELISA; C Results of in vitro intestinal barrier integrity in different cell groups. The left graph shows TEER values representing cell barrier integrity, and the right graph shows FITC-Dex values representing cell barrier permeability; D Schematic diagram of cell experiment process (Created by BioRender); E Measurement of cell viability in each group using MTT assay; F Evaluation of cell proliferation capacity in each group using EdU assay (Scale bar=50 μm); G Re-assessment of inflammatory factor levels in cells of each group using ELISA; H Detection of E-cadherin expression in cells of each group using Western blot; I Same as (C), depicting the results of in vitro intestinal barrier integrity in different cell groups; J Assessment of cell viability in each group using MTT assay; K Evaluation of cell proliferation capacity in each group using EdU assay (Scale bar=50 μm); L Reinforced assessment of inflammatory factor levels in cells of each group using ELISA; M Re-examination of E-cadherin expression in cells of each group using Western blot; N Reiteration of in vitro intestinal barrier integrity results in different cell groups, with TEER values indicating cell barrier integrity on the left and FITC-Dex values representing cell barrier permeability on the right. Comparisons between two groups were analyzed using t-tests, while one-way ANOVA was used for comparisons among different groups. The line between groups represents the specific p-value, with p < 0.05 indicating a statistically significant difference between groups. Each cell experiment was replicated three times. Next, we overexpressed BCL2 with DMSO treatment and knocked down BCL2 with GP treatment. Cell viability was measured using the MTT assay, cell proliferation capacity was assessed with EdU incorporation, changes in inflammatory factor levels were evaluated by ELISA, E-cadherin expression was analyzed by Western blot, and the integrity of the intestinal barrier was assessed through in vitro barrier integrity experiments (permeability assessment) (Fig. [115]4D). The results showed that in the DMSO group, overexpression of BCL2 led to increased cell viability, enhanced cell proliferation, reduced inflammatory factor levels, significant upregulation of E-cadherin expression, and improved integrity of the intestinal barrier. The results showed that there was no significant difference in the therapeutic effects between the GP-treated group and the BCL2 overexpression group. When GP was combined with the BCL2 overexpression group, GP significantly enhanced the therapeutic effects compared to the BCL2 overexpression group alone, further supporting the role of BCL2 in the mechanism of GP action (Fig. [116]4E–I). Conversely, in the GP treatment group, cell viability decreased, proliferation capacity diminished, inflammatory factor levels increased, E-cadherin expression significantly decreased, and the in vitro intestinal barrier integrity declined after knocking down BCL2 (Fig. [117]4J–N). These results convincingly demonstrate that GP can enhance the integrity of the intestinal barrier by upregulating BCL2 expression in colonic epithelial cells. GP alleviates depression by activating the BCL2-mediated AMPK pathway Numerous studies have shown that activation of the AMPK signaling pathway is beneficial in promoting the balance of intestinal flora^[118]49,[119]50. Dorsomorphin, a selective ATP-competitive AMPK inhibitor, has been proven to suppress the phosphorylation levels of AMPK^[120]51,[121]52. The results in Fig. [122]1E suggest a potential association between GP and the treatment of depression through the AMPK pathway, while predictive results from the GeneMANIA database indicate interactions between BCL2 and AMPK (including PRKAA1 and PRKAA2) (Fig. [123]5A). Therefore, we hypothesize that GP might activate the AMPK pathway by mediating the expression of BCL2, promoting the balance of intestinal flora and thus alleviating depression. Fig. 5. GP mediates BCL2 expression to activate the AMPK pathway, alleviating depression. [124]Fig. 5 [125]Open in a new tab A Predicted results from the GeneMANIA database; B Evaluation of AMPK phosphorylation levels in cells of each group using Western blot; C Detection of BCL2 expression levels and AMPK phosphorylation levels in cells of each group through Western blot; D Assessment of cell viability in each group with MTT assay; E Examination of cell proliferation capacity in each group using EdU experiment (Scale bar=50 μm); F Measurement of inflammatory factor levels in cells of each group with ELISA; G Assessment of E-cadherin expression in cells of each group, analyzed by Western blot; H Measurement of in vitro intestinal barrier integrity in cells of each group, where TEER values in the left graph represent cell barrier integrity, and FITC-Dex values in the right graph represent cell barrier permeability; I Presentation of H&E staining results of mouse hippocampal tissues in each group, Scale bar=250 / 25 μm; J Examination of the number of neuronal cells in mouse hippocampal tissues of each group through Nissl staining, Scale bar=250 / 25 μm; K Detection of neuronal cell apoptosis status in mouse hippocampal tissues of each group using TUNEL staining, Scale bar=250 / 25 μm; L Recording of the body weight changes in mice of each group; M Demonstration of H&E staining results of mouse colon tissues in each group, with Scale bar=100 μm; N Determination of in vivo barrier integrity experiment results; (O-R) Evaluation of depressive-like behaviors in mice of each group using TST, SPT, FST, and MWM experiments. One-way ANOVA was used for comparisons among different groups. The line between groups represents the specific p-value, with p < 0.05 indicating a statistically significant difference between groups. Cell experiments were repeated three times, with 6 mice in each group. To validate our hypothesis, we initially examined the phosphorylation levels of AMPK upon knocking down or overexpressing BCL2, revealing a decrease in AMPK phosphorylation levels with BCL2 knockdown and an increase with BCL2 overexpression (Fig. [126]5B). Subsequently, while overexpressing BCL2, we inhibited the phosphorylation levels of AMPK using Dorsomorphin (Dor). The results showed that compared to the oe-NC + DMSO group, the cells in the oe-BCL2 + DMSO group exhibited significantly enhanced phosphorylation levels of AMPK, increased expression of BCL2, enhanced cell viability and proliferation capabilities, reduced cellular inflammation, upregulated E-cadherin expression, and improved intestinal barrier integrity. Conversely, compared to the oe-BCL2 + DMSO group, the cells in the oe-BCL2 + Dor group showed a significant decrease in AMPK phosphorylation levels, no significant change in BCL2 expression, weakened cell viability and proliferation capabilities, exacerbated cellular inflammation, downregulated E-cadherin expression, and decreased intestinal barrier integrity (Fig. [127]5C–H). Subsequently, we verified this mechanism in vivo. Western blot analysis revealed a significant increase in AMPK phosphorylation levels and BCL2 expression in mouse colon tissues of the oe-BCL2 + DMSO group compared to the oe-NC + DMSO group (Fig. [128]S5A). Further examinations on the impact on mouse depression revealed that compared to the oe-NC + DMSO group, the inflammation infiltration in mouse colon tissues of the oe-BCL2 + DMSO group was alleviated, intestinal barrier integrity was strengthened, hippocampal tissue lesions were mitigated, the proportion of neuronal cells significantly increased, depression-like behaviors in mice were alleviated, accompanied by a slight increase in body weight. In contrast, the oe-BCL2 + Dor group showed a worsening of colorectal tissue inflammation, a significant decrease in intestinal barrier integrity, exacerbation of hippocampal tissue lesions, reduction in neuronal cells, worsening of depression in mice, and decrease in body weight (Figs. [129]5I–N and Fig. [130]S5B–F). Furthermore, results from FMT experiments confirmed that administration of fecal pellets from oe-BCL2 + DMSO group mice to the oe-NC + DMSO/oe-BCL2 + Dor group mice alleviated depression behaviors, highlighting the essential role of intestinal flora in the development of Depression (Fig. [131]5O–R). In conclusion, the results demonstrate that GP can alleviate depression by upregulating the expression of BCL2 in colonic epithelial tissues to activate the AMPK pathway. GP complexed with NCC: enhancing oral drug release and intestinal absorption Table [132]S1 results revealed a mere 14.64% oral bioavailability of GP. Previous studies have highlighted that NCC, originating from plant fibers, boasts excellent biocompatibility and high biosafety, enabling the promotion of intestinal barrier repair and the balance of intestinal flora^[133]53. Additionally, NCC presents a substantial surface area and negative charge, facilitating the binding of a plethora of drugs, thus unleashing the potential for high drug loading and optimal dosage control^[134]54. Building upon these investigations, we successfully synthesized the GP complex with NCC (Fig. [135]6A). Fig. 6. Synthesis and characterization of NCC-GP. [136]Fig. 6 [137]Open in a new tab A Schematic representation of the preparation and physical characterization process of the NCC-GP composite (Created by BioRender); B Scanning electron microscope images of the NCC-GP composite (Scale bar=500 nm); C Determination of the binding efficiency of NCC with GP using UV-Vis spectrophotometry; D Assessment of the Zeta potential of NCC-GP solution by dynamic light scattering (DLS); E Evaluation of the drug release rate of NCC-GP composite using UV-Vis spectrophotometry; F Investigation of intracellular uptake of fluorescently labeled NCC-GP after cellular internalization (Scale bar=50 μm). Cell experiments were repeated thrice. In the preparation process, we initially examined the morphology of NCC using scanning electron microscopy, revealing individual NCC fibers to be approximately 10 nm wide and 500 nm long (Fig. [138]6B). Following the preparation of NCC-GP, the drug binding rate was calculated using a UV-visible spectrophotometer. The results indicated a significant increase in the quantity of GP bound to NCC with an escalation in the drug mass added to the NCC suspension. When PBS was utilized as the dispersing medium, over 100 μg of GP was shown to bind to NCC, with a binding efficiency exceeding 50% (Fig. [139]6C). Additionally, no significant alteration in zeta potential was observed after GP was bound to NCC (Fig. [140]6D). Subsequently, we assessed the drug release efficacy of NCC-GP. The findings demonstrated a rapid release of GP from NCC, with approximately 60% of the bound GP being released within 4 hours. By the end of day 1, drug release had stabilized, surpassing an 80% release rate of GP (Fig. [141]6E). Furthermore, visualization of the uptake of fluorescently labeled NCC by murine colonic epithelial cells indicated substantial uptake of both NCC and NCC-GP, with no apparent cellular breakdown within 24 hours, suggesting the absence of significant cytotoxicity with NCC and NCC-GP (Fig. [142]6F). In conclusion, we have successfully developed a GP complexed with NCC, exhibiting a rapid and cost-effective drug release rate, along with efficient uptake by colonic epithelial cells. NCC-GP enhanced GP’s restorative effect on the intestinal barrier In this study, the toxicity and therapeutic effects of NCC and NCC-GP on colonic epithelial cells were further investigated through in vitro cell experiments. Initially, Western blot analysis was employed to assess the impact of NCC-GP on the expression of BCL2 and the phosphorylation levels of AMPK. The results demonstrated that compared to the NCC/DMSO group, NCC-GP exhibited similar effects to GP, significantly upregulating the expression of BCL2 and the phosphorylation levels of AMPK (Fig. [143]7A). Subsequently, cells from the Normal group were treated with NCC or NCC-GP, and the toxicity and therapeutic effects of NCC-GP on the cells were evaluated using the MTT assay, EdU labeling, and TUNEL staining, with DMSO serving as the control group. The findings revealed no significant changes in the viability, proliferation capacity, or apoptosis status of cells in the NCC + Nor/NCC-GP + Nor groups compared to the DMSO + Nor group, indicating that NCC and NCC-GP did not exhibit significant toxicity to the cells (Fig. [144]7B–D). Moreover, compared to the DMSO group, the viability and proliferation capacity of cells in the NCC group, GP group, and NCC-GP group gradually increased while the apoptotic ability decreased. Among these groups, cells in the NCC-GP group demonstrated the highest viability and proliferation capacity and the lowest apoptosis rate (Fig. [145]7E–G). Fig. 7. Effects of NCC-GP on colonic epithelial cells. [146]Fig. 7 [147]Open in a new tab A Evaluation of BCL2 expression levels and AMPK phosphorylation levels in cells of each group using Western blot; B Assessment of cell viability in each group with MTT assay; C Measurement of cell proliferation capacity in each group through EdU experiment (Scale bar=50 μm); D Evaluation of cell apoptosis status in each group using TUNEL staining (Scale bar=50 μm); E Reassessing cell viability in each group with MTT assay; F Analysis of cell proliferation capacity in each group through EdU experiment (Scale bar=50 μm); G Determination of cell apoptosis status in each group using TUNEL staining (Scale bar=50 μm); H Measurement of inflammatory factor levels in cells of each group with ELISA; I Detection of E-cadherin expression in cells of each group through Western blot; J Assessment of in vitro intestinal barrier integrity in cells of each group, where TEER values in the left graph represent cell barrier integrity, and FITC-Dex values in the right graph represent cell barrier permeability. Comparisons between two groups were analyzed using t-tests, while one-way ANOVA was used for comparisons among different groups. The line between groups represents the specific p-value, with p < 0.05 indicating a statistically significant difference between groups. Each cell experiment was repeated three times. Subsequent analysis involved measuring the levels of inflammatory factors within the cells using ELISA. The results illustrated a gradual reduction in the inflammatory factor levels in cells from the NCC group, GP group, and NCC-GP group compared to the DMSO group, with the NCC-GP group showing the most significant alleviating effect on cellular inflammation (Fig. [148]7H). Further Western blot and immunohistochemistry experiments yielded consistent outcomes, indicating that the expression of E-cadherin in cells from the NCC group, GP group, and NCC-GP group significantly increased compared to the DMSO group. This led to an enhancement in the integrity of the in vitro intestinal barrier, with NCC-GP demonstrating the most significant effect (Fig. [149]7I, J). In summary, the results suggest that NCC-GP effectively enhances GP’s reparative action on the intestinal barrier. Intestinal barrier repair and depression treatment: mechanisms and efficacy of NCC-GP In the final analysis, it was verified in vivo that NCC-GP mediates the alleviation of depression through enhancement of the intestinal barrier in the gut-brain axis. Western blot analysis revealed significantly increased levels of BCL2 expression and AMPK phosphorylation in the colon tissues of mice treated with NCC-GP, consistent with the results of in vitro cell experiments (Fig. [150]8A). Subsequently, behavioral tests were conducted to assess the degree of depression in mice, showing a noticeable reduction in depressive behaviors in the NCC group, GP100 group, and NCC-GP group compared to the DMSO group, with the efficacy gradually increasing and being most pronounced in the NCC-GP group (Fig. [151]8B–E). H&E staining results indicated alleviation of lesions in the hippocampal tissues of mice in the NCC group, GP100 group, and NCC-GP group, with the therapeutic effect progressively increasing. Nissl and TUNEL staining further demonstrated a gradual increase in the proportion of neuronal cells and a reduction in apoptosis, with the most significant effect observed in the NCC-GP group (Fig. [152]8F–H). Additionally, compared to the DMSO group, mice in the NCC group, GP100 group, and NCC-GP group exhibited gradual weight gain, reduced intestinal inflammation, increased E-cadherin levels, and improved integrity of the intestinal barrier (Fig. [153]8I–L). Fig. 8. In vivo validation of NCC-GP alleviating depression via the gut-brain axis. [154]Fig. 8 [155]Open in a new tab A Expression levels of BCL2 and AMPK in each group of cells were detected using Western blot; B–E Depressive-like behaviors of mice in each group were assessed through TST, SPT, FST, and MWM experiments; F display of H&E staining results of hippocampal tissues in each group of mice, Scale bar = 250 μm / 25 μm; G Neuronal cell count in the hippocampal tissues of each group of mice was evaluated through Nissl staining, Scale bar = 250 / 25 μm; H Apoptosis of neuronal cells in the hippocampal tissues of each group of mice was examined using TUNEL staining, Scale bar = 250 / 25 μm; I Monitoring of weight changes in each group of mice with analyzed using two-way ANOVA; J Presentation of H&E staining results of colonic tissues in each group of mice, Scale bar = 100 μm; K Content of E-cadherin in colonic tissues of each group of mice was detected through Western blot; L Display of in vivo barrier integrity experiment results; M–P Assessment of depressive-like behaviors in each group of mice through TST, SPT, FST, and MWM experiments; Q H&E staining results of hippocampal tissues in each group of mice, Scale bar = 250/25 μm; R Neuronal cell count in the hippocampal tissues of each group of mice was evaluated via Nissl staining, Scale bar = 250 / 25 μm; S Evaluation of neuronal cell apoptosis in the hippocampal tissues of each group of mice using TUNEL staining, Scale bar = 250 / 25 μm. Comparisons between two groups were analyzed using t-tests, while one-way ANOVA was used for comparisons among different groups. The line between groups represents the specific p-value, with p < 0.05 indicating a statistically significant difference between groups, with 6 mice per group. Furthermore, results from FMT experiments indicated that administering fecal pellets from NCC mice or NCC-GP mice via gavage to DMSO group mice led to relief in depressive behaviors, with the NCC-GP group showing the best therapeutic efficacy (Fig. [156]8M–P). H&E staining illustrated a notable alleviation of lesions in the hippocampal tissues of mice in the DMSO + FMT-NCC group and DMSO + FMT-NP group, while Nissl and TUNEL staining revealed a significant increase in the proportion of neuronal cells and a marked decrease in apoptosis in these groups (Fig. [157]8Q–S). Finally, an assessment of the in vivo toxicity of NCC-GP in mice was conducted, with major organs subjected to H&E staining post-treatment, revealing no significant pathological changes (Fig. [158]S6), indicating the excellent biocompatibility of NCC-GP. These findings collectively demonstrate that NCC-GP can ameliorate depression by alleviating intestinal inflammation, repairing the intestinal barrier, balancing the intestinal flora, and further influencing the hippocampal tissues in the brain through the gut-brain axis. Discussion In previous studies, GP, as the main component of the traditional Chinese medicine Gardenia jasminoides J. Ellis, has been a subject of considerable attention due to its confirmed antidepressant effect^[159]55,[160]56. However, to the best of our knowledge, this study investigates a previously unreported mechanism of the GP-NCC combination in regulating intestinal barrier function and balancing intestinal flora. By employing various research tools, including network pharmacology screening, proteomic techniques, and machine learning algorithms, this study delves deeper into unveiling the molecular mechanism of GP in combination with NCC on depression-like behaviors, offering a fresh perspective for depression treatment research. Through the construction of a CUMS model and in vitro experimental validation, we discovered that GP improves intestinal barrier function and alleviates depression-like behaviors in mice by upregulating the expression of BCL2 to activate the AMPK pathway. This finding aligns with previous research on the regulatory role of GP in cellular survival environments. However, our study further elucidates its key factors correlated with CUMS, providing a new angle for mechanistic exploration^[161]57,[162]58. In vitro cell experiments revealed that GP modulates the vitality, proliferative capacity, and apoptosis of colonic epithelial cells by regulating BCL2 expression, thereby enhancing the integrity of the intestinal barrier. This discovery supports previous research findings, validating the significant role of GP in regulating cellular functions and inflammatory responses. Concurrently, the compound of GP and NCC, NCC-GP, demonstrates remarkable effects in intestinal barrier repair, offering new avenues and possibilities for depression treatment^[163]22,[164]59,[165]60. By preparing the composite of NCC and GP, NCC-GP, we confirmed its reparative effect on the intestinal barrier. This innovative preparation method opens new pathways for the application of traditional Chinese medicine ingredients in depression therapy while also unveiling fresh possibilities for the potential use of nanotechnology in treating neurological and psychiatric disorders^[166]61. Animal experiments further substantiated the effectiveness of NCC-GP in alleviating depression-like behaviors, reinforcing the compound’s therapeutic effect on depression through the restoration of the intestinal barrier and microbiota balance^[167]62. Compared to previous research outcomes, our study offers a more comprehensive and in-depth experimental validation, providing more reliable evidence for the application of this compound in depression treatment. Based on the aforementioned results, we can preliminarily conclude the following: GP, by upregulating the expression of BCL2 in colonic epithelial tissues, activates the AMPK pathway to alleviate depression. The complex formed by NCC binding with GP can further impact the brain’s hippocampal tissue through the gut-brain axis by alleviating intestinal inflammation, repairing the intestinal barrier, and balancing the intestinal flora, ultimately treating Depression. This study introduces a novel therapeutic strategy to address the low oral bioavailability issue of GP, which conventional treatments struggle to overcome. Our research lays a theoretical foundation for understanding the development of depression and discovering new therapeutic targets. Nevertheless, the study has certain limitations. While positive results were observed in in vitro cell experiments and animal models, the successful translation of these findings into clinical applications requires further validation. Additionally, the long-term safety and potential side effects remain unclear, prompting our future endeavors to delve deeper into these areas. The study’s scientific and clinical significance lies in the in-depth exploration of the active component GP from the traditional Chinese medicine Gardenia jasminoides J. Ellis and its binding with NCC to treat depression, elucidating the molecular mechanisms involved. Through network pharmacology screening and experimental validation, GP was identified as the primary active component for treating depression, revealing its mechanism of action in treating depression by modulating key pathways such as BCL2 and AMPK, thus providing new pharmacological evidence for traditional Chinese medicine in depression treatment. Furthermore, utilizing high-throughput sequencing and proteomic technologies, the study unveiled the impact of GP on balancing intestinal flora and improving intestinal barrier function in mice exhibiting depressive behaviors, offering crucial insights into the relationship between depression and the gut-brain axis. In terms of clinical applications, our study offers a theoretical basis for developing new drugs to treat depression. Moreover, the cost-effective preparation method of the complex formed by NCC and GP exhibits a favorable drug release profile, making it a promising candidate for a novel depression treatment. Additionally, the new strategy for treating depression through balancing intestinal flora, repairing the intestinal barrier, and regulating brain neural tissues via the gut-brain axis presents fresh perspectives for the treatment of mental illnesses. Nonetheless, the study has some limitations. Firstly, the research was conducted on mouse models, and further confirmation of its efficacy and safety in humans is needed. The study results indicated that the absolute bioavailability of GP followed the order: i.m. (72.69%) > i.n. (49.54%) > i.g. (9.74%). It was also found that GP can permeate the skin both in vitro and in vivo, allowing for rapid distribution into subcutaneous tissues and the bloodstream in mice after administration with ShanYi aerosol^[168]63,[169]64. We acknowledge that pharmacokinetic and pharmacodynamic data on NCC are currently limited, and future studies will explore the pharmacokinetics and pharmacology of GP treatment in greater depth. We also plan to include germ-free mice or antibiotic-treated groups in subsequent research to further confirm whether GP exerts its antidepressant effects through modulation of the gut microbiota. By introducing such controls, we aim to delve deeper into the mechanism of GP in the CUMS mouse model. Secondly, despite unveiling the molecular mechanisms of GP in treating depression through various technological approaches, further investigation into its effects and applicability across different types of depression patients is required to better guide clinical applications. Based on previous CUMS research selections^[170]65,[171]66, male mice were chosen in this study for several scientific considerations: The endocrine cycles of female mice significantly impact the reproducibility of behavioral experiments, especially in depression models, where fluctuations in female hormones can interfere with experimental outcomes, increasing the variability and complexity of data interpretation. In contrast, male mice have more stable endocrine profiles, facilitating clearer and more consistent results. Standardized research protocols often prioritize male mice to minimize sex-related variables when investigating new biological mechanisms, as this is a common strategy in experimental model design. This choice not only helps to clarify the relationship between experimental interventions and improvements in depressive symptoms but also provides a comparative basis for future studies in female models. Building on this study, we will explore whether the mechanisms of GP-mediated BCL2 upregulation and AMPK pathway activation in alleviating depression are equally effective in female mice and will address potential sex differences. Looking ahead, future exploration can focus on uncovering the potential mechanisms and clinical applications of the complex formed by GP and NCC in depression treatment. By integrating the concept of personalized medicine and conducting clinical trials tailored to different types of depression, patients can lead to individualized precision therapies. Preclinical studies have shown that subdiaphragmatic vagotomy (SDV) can block depressive-like behaviors in mice following fecal microbiota transplantation (FMT) from mice exhibiting depressive-like behaviors^[172]67. We will further investigate whether SDV affects the antidepressant effects of the GP-NCC complex in the CUMS model, which promises to be an intriguing area of exploration. Furthermore, investigating the combined use of GP with other drugs or treatment modalities may unveil additional treatment strategies, providing more choices and hope for patients with depression. Materials and methods Gardenia bioactive components Chemical components of Gardenia jasminoides J. Ellis were retrieved using the TCMSP. Active components with oral bioavailability (OB) > 10% and drug-likeness (DL) > 0.4 were selected as effective, along with their respective targets. The identified targets were converted into corresponding genes using the Uniprot database ([173]http://www.uniprot.org/)^[174]68. Analysis of active ingredient targets Genes related to depression were searched in the GeneCards database ([175]https://www.genecards.org/) using “depression” as the keyword, with a relevance score > 5 as the threshold for selecting depression-related genes. An intersection analysis was conducted using the XenoTox academic online analysis platform between genes related to depression and the target genes of the aforementioned active components to create a Venn diagram, obtaining the intersection genes as potential therapeutic targets. The selected potential therapeutic targets and active components were imported into Cytoscape software (version 3.10.0) to construct a network named “Chinese Medicine - Active Components - Targets - Depression.” Subsequently, core components and targets were identified based on this network, and further analysis of the underlying mechanisms was conducted^[176]69. PPI The potential therapeutic targets identified through the aforementioned screening were mapped onto the String database platform ([177]https://string-db.org/), with Homo sapiens selected as the research species for PPI analysis. A medium confidence level interaction score (0.400) was set, unconnected nodes were hidden, and other parameters were kept at default settings to obtain a PPI network of potential therapeutic targets for depression treatment, with the protein interaction data downloaded. The connectivity node count statistics graph for each protein was generated using R software. Subsequently, the PPI network graph was redrawn using Cytoscape software based on the Degree values and the proteins’ importance in the network as determined in the PPI analysis^[178]70. Functional enrichment analysis GO and KEGG functional enrichment analyses were conducted on the potential therapeutic targets of active compounds to differentiate the enriched pathways of different target genes of active compounds. The “ClusterProfiler” package in R software was utilized for GO and KEGG enrichment analyses. The GO enrichment analysis included three categories: biological process (BP), cellular component (CC), and molecular function (MF), with a significance threshold of P < 0.05 for selection and visualization of the enriched pathways and genes. KEGG enrichment analysis was performed with a significant enrichment selection criteria of P < 0.05, followed by visualization of the enriched pathways and genes. Based on the differential enrichment pathways of the targets, key active compounds and their targets were selected in conjunction with relevant literature to speculate on the potential mechanisms of action^[179]71. Molecular simulation docking GP’s 2D and 3D chemical structures were obtained from the PubChem database ([180]https://pubchem.ncbi.nlm.nih.gov/) with Compound CID: 187808 and saved in “SDF” format. The crystal structure of the candidate target protein BCL2 (PDB ID: 1G5M) for GP was downloaded from the Protein Data Bank (PDB) database ([181]https://www.rcsb.org). The compound GP’s structure was converted to a three-dimensional structure using Chem3DUltra 14.0 software and subjected to energy minimization using the MM2 algorithm. Subsequently, the target protein receptor underwent dehydration and organic compound removal using PyMOL software. AutoDockTools 1.5.6 was employed for hydrogenation and charge computation of the target protein receptor molecules. The compound and target protein receptor were converted into “pdbqt” files, and appropriate box center and grid parameters were set. Finally, Vina 1.5.6 was utilized to assess molecular docking and calculate the binding free energy. By integrating PPI network analysis, pharmacophore modeling, and molecular docking analysis, drug-target interactions were identified^[182]72. Cell culture Mouse colonic epithelial cells (CP-M159, Procell, China) were cultured in a specialized medium (CM-M159, Procell) containing 10% FBS (12484028, Gibco, USA) and 1% penicillin/streptomycin (15140148, Gibco, USA). To establish an in vitro model of colonic epithelial cells, 100 μg/mL of LPS (L5293, Sigma-Aldrich, USA) was added to the culture medium and the cells were treated for 24 hours. For studying the action of AMPK, 10 μM Dorsomorphin (AMPK inhibitor; HY-13418A, MedChemExpress, USA) was added to the cell culture medium, followed by subsequent experiments after 18 hours^[183]51. The cell groups were designated as follows: Control/Nor group (normal mouse colonic epithelial cells), LPS group (mouse colonic epithelial cells treated with LPS), DMSO group (mouse colonic epithelial cells cultured with 10 μM DMSO in the medium), Dor group (mouse colonic epithelial cells cultured with 10 μM Dorsomorphin in the medium). 293 T cell line was obtained from ATCC (CRL-3216) and cultured in DMEM medium (11965092, Gibco, USA) containing 10% FBS, 10 μg/mL streptomycin, and 100 U/mL penicillin. All cells were cultured at 37 °C with 5% CO[2] in a humidified CO[2] incubator. Passaging was performed when the cells reached 80%-90% confluence^[184]73. Construction of virus vector and cell transfection Mouse cDNA sequences were analyzed for potential short hairpin RNA (shRNA) target sequences based on GeneBank. Three sequences targeting BCL2 were initially designed, with one sequence lacking interfering sequences serving as the negative control (sh-NC). The primer sequences are listed in Table [185]S3, and the oligonucleotides were synthesized by GenePharma® (Shanghai, China). A lentiviral packaging system was constructed using pLKO.1 (lentivirus gene silencing vector). The packaged virus and target vector were co-transfected into 293 T cells using Lipofectamine 2000 (11668500, Invitrogen, USA) with a cell confluence of 80-90%. After 48 hours of cell culture, the supernatant was collected, and virus particles were present in the filtered supernatant after centrifugation. The virus was harvested during the logarithmic growth phase for virus titer detection. Lentivirus for BCL2 overexpression was constructed and packaged by Genechem (Shanghai, China), with the overexpression vector being LV-PDGFRA. The packaged virus and target vector were co-transfected into 293 T cells using Lipofectamine 2000 (11668500, Invitrogen, USA) with a cell confluence of 80-90%. After 48 hours of cell culture, the supernatant was collected, and virus particles were present in the filtered supernatant after centrifugation. The virus was harvested during the logarithmic growth phase for virus titer detection. Additionally, for the convenience of subsequent bioluminescence imaging experiments, the silence or overexpression vectors carried the GFP/mCherry genes. When the cells reached the logarithmic growth phase, they were digested with trypsin, suspended, and seeded at a concentration of 5 × 10^4 cells/mL in 6-well plates, with 2 mL per well. Prior to establishing the in vitro cell model, lentivirus for BCL2 overexpression or knockdown (MOI = 10, virus titer of 1 × 10^8 TU/mL) was added separately to the cell culture medium for 48 hours, followed by screening stable cell lines with 2 µg/mL puromycin (UC0E03, Sigma-Aldrich, USA) for 2 weeks^[186]74–[187]77. The cell transfection groups were as follows: sh-NC group (mouse colonic epithelial cells without BCL2 knockdown), sh-BCL2 group (mouse colonic epithelial cells with BCL2 knockdown), oe-NC group (mouse colonic epithelial cells without BCL2 overexpression), and oe-BCL2 group (mouse colonic epithelial cells with BCL2 overexpression). Construction and grouping of the CUMS model in mice Male C57BL/6 J mice weighing 20-25 grams and aged 8-10 weeks were purchased from Hunan Siley Jingda Experimental Animal Co., Ltd., in Hunan, China. The mice were housed individually in cages in an SPF-grade animal facility maintained at a humidity of 60%-65% and a temperature of 25 ± 2  °C, with a 12-hour light-dark cycle and ad libitum access to food and water. After one week of acclimation, the mice were observed for their health status before the start of the experiment. The experimental procedures and animal usage were approved by the Animal Ethics Committee^[188]78. The mice were divided into a control group and a CUMS group. In the CUMS group, mice were subjected to the following mild stressors (Fig. [189]S7) individually: (i) 24-hour fasting; (ii) 24-hour water deprivation; (iii) 1-hour empty bottle exposure; (iv) 7-hour cage tilt (45 °); (v) overnight illumination; (vi) 24-hour living in dirty cages (bedding with 100 g sawdust and 200 mL water); (vii) 30 minutes of forced swimming at 8 °C; (viii) 2 hours of restraint; (ix) 24-hour exposure to a novel object (such as a piece of plastic). These stress stimuli were randomly applied over a span of 6 weeks^[190]79,[191]80.After successful modeling with CUMS, behavioral tests such as the FST and SPT were conducted. The grouping information of the mice in the modeling experiment is as follows (6 mice per group): Control group (non-modeled control mice); CUMS group (mice modeled with CUMS); DMSO group (mice modeled and treated with DMSO); GP100 group (mice modeled and treated with 100 mg/kg GP); SSRI group (mice modeled treated with fluoxetine at a dosage of 20 mg/kg);CUMS + FMT group (CUMS modeled mice subjected to FMT with 200 µL of fecal pellets from control mice); DMSO + FMT-GP group (DMSO-treated mice subjected to FMT with 200 µL of fecal pellets from GP100 group); oe-NC + DMSO group (mice injected with lentivirus constructed with oe-NC vector and intraperitoneally injected with 10 mg/kg of DMSO); oe-BCL2 + DMSO group (mice injected with lentivirus constructed with oe-BCL2 vector and intraperitoneally injected with 10 mg/kg of DMSO); oe-BCL2 + Dor group (mice injected with lentivirus constructed with oe-BCL2 vector and intraperitoneally injected with 10 mg/kg of Dorsomorphin); oe-NC + DMSO + FMT-BCL2 group (mice from oe-NC + DMSO group subjected to FMT with 200 µL of fecal pellets from oe-BCL2 + DMSO group); oe-BCL2 + Dor + FMT-BCL2 group (mice from oe-BCL2 + Dor group subjected to FMT with 200 µL of fecal pellets from oe-BCL2 + DMSO group); NCC group (mice treated with 100 mg/kg NCC by gastric lavage); NCC-GP group (mice treated with 100 mg/kg NCC-GP by gastric lavage); DMSO + FMT-NCC group (DMSO-treated mice subjected to FMT with 200 µL of fecal pellets from NCC group); DMSO + FMT-NP group (DMSO-treated mice subjected to FMT with 200 µL of fecal pellets from NCC-GP group). GP (HY-N0009, MedChemExpress, USA) was dissolved in DMSO (D2650, Sigma-Aldrich, USA) to prepare a 10 μM stock solution. The dosage of GP was based on previous studies demonstrating its antidepressant effects in mouse models^[192]81. The fluoxetine dosage followed established research, with fluoxetine hydrochloride obtained from Dar Al Dawa Pharmaceuticals (Jordan). Fluoxetine hydrochloride was dissolved in 0.03% sodium carboxymethylcellulose and administered daily by gastric gavage at a dose of 20 mg/kg^[193]82. Sixty minutes before CUMS induction, mice were orally gavaged with a dose of 10 mL/kg once daily, Lentiviruses overexpressing BCL2 were constructed and packaged by Genechem (Shanghai, China), with LV-PDGFRA as the viral gene overexpression vector. Packaging plasmids (1 µg) and target plasmid (1 µg) were co-transfected into 293 T cells in a 6-well plate (80-90% cell confluence) using Lipofectamine 2000 (11668500, Invitrogen, USA) at a ratio of 1:2. After 48 hours of culture, the supernatant containing viral particles was collected and centrifuged. The viral titer was subsequently determined. Mice in the lentivirus-transfected groups were injected with 1 × 10^8 TU/mL lentivirus into the colon tissue to establish overexpression or knockdown models, with two injections per week. Behavioral tests were conducted six weeks after Chronic Unpredictable Mild Stress (CUMS) induction to confirm the successful establishment of the CUMS mouse model. Mice were deeply anesthetized with an appropriate concentration of isoflurane gas administered through a breathing mask. Blood samples were collected via retro-orbital puncture following cervical dislocation, and serum was separated by centrifugation at 4 °C, 2500 rpm for 10 minutes, then stored at -80 °C. Subsequently, the brain and hippocampal tissues were collected, colonic tissues were dissected, frozen in liquid nitrogen, and stored at -80 °C^[194]52,[195]79. Tail suspension test (TST) Each mouse had its tail taped to a position 60 centimeters above the ground and was filmed for 6 minutes. To prevent climbing behavior during the test, a transparent cylindrical obstacle with a diameter of 1.5 cm and a length of 4 cm (XinRuan Technology) was placed on the tail of each mouse. An observer blinded to the mouse groups recorded the total immobility time of each mouse during the entire 6 minutes (passively suspended without any movement)^[196]80,[197]83. Sucrose preference test (SPT) Initially, animals were trained to drink a 1% sucrose solution from two different bottles (conducted 48 hours before the formal experiment). After 24 hours, the animals were given free access to a 1% sucrose solution and water from the two different bottles. To prevent side preference, the positions of the two bottles were switched. The consumption of sucrose from the two bottles was measured in the following 24 hours, and the sucrose preference (%) was calculated using the formula: Sucrose Consumption / (Sucrose Consumption + Water Consumption) × 100%^[198]80,[199]84. FST Mice were placed in a cylindrical acrylic tank filled with water at a temperature of 23 ± 1 °C and a depth of 13 cm (diameter 10 cm, height 20 cm). The mice were forced to swim for 6 minutes, and the immobility time during the last 5 minutes was recorded using the Visutrack motion tracking system from Shanghai Xinhua Software Information Technology Co., Ltd.^[200]80. MWM The MWM utilized in this study consists of a circular water tank with a diameter of 120 cm, filled with water maintained at a temperature of 22 ± 2 °C. The platform within the maze is utilized as a spatial reference point and is surrounded by walls bearing distinct shapes. A camera mounted above the maze records the swimming paths of the mice as they navigate through the water maze. During the acquisition phase trials, the platform is submerged 1-2 cm below the water’s surface, with mice placed in one of the cardinal directions (north, south, east, or west) facing the walls of the tank. Mice are given 60 seconds to locate the platform, and if unsuccessful, they are guided to it and allowed to remain there for 10 seconds. Four trials are conducted each day, separated by at least one hour, with the escape latency for spatial memory acquisition recorded in each trial. On the 7th day, the platform is removed for a probe test (Fig. [201]S8), where the time spent in each quadrant, the number of crossings in the target (platform) area, average speed, and total distance covered are measured^[202]80. Histopathological staining techniques Hematoxylin and eosin (H&E) staining: Mouse hippocampal or colon tissue samples were fixed, sectioned, dewaxed in xylene, rehydrated in 100%, 95%, and 70% ethanol, and then embedded or rinsed in water. The prepared sections were stained with hematoxylin staining solution (H8070, Solarbio, Beijing) for 5-10 minutes at room temperature, followed by rinsing with distilled water, dehydration in 95% ethanol, and immersion in eosin staining solution (G1100, Solarbio, Beijing) for 5-10 minutes. The sections were dehydrated, cleared, and coverslipped^[203]85. Nissl staining: Intact hippocampal tissue sections were air-dried at room temperature, fixed for 30 seconds, placed in cresyl violet staining solution (C0117, Beyotime Biotechnology Co., Ltd, Shanghai) for 1 hour in the dark, sequentially differentiated in 70%, 80%, and 95% ethanol (10 seconds each), followed by staining differentiation solution (ethanol: chloroform: diethyl ether = 1:1:1) until clear differentiation was observed. The sections were immersed twice in ethanol (5 minutes each), permeated twice in dichloromethane (4 minutes each), and mounted in Canada balsam. Neuronal counts were conducted using a fluorescence microscope (BX63, Olympus). TUNEL staining: Frozen tissue sections were immersed in DMSO and incubated in 200 mL citrate buffer (0.1 mL, pH 6.0), heated in a microwave at 90-95 °C for 1 minute at 680 W (80% power), cooled by adding 80 mL double-distilled water (20-25 °C). Subsequently, the sections were incubated in 20% normal bovine serum at room temperature for 30 minutes, followed by a 90-minute incubation with 50 μL TUNEL reaction mixture (negative control without TUNEL reaction mixture). The sections were then incubated at room temperature for 10 minutes with 3% H[2]O[2], followed by a 90-minute incubation at 37 °C, incubated for 30 minutes at 37 °C with peroxidase (POD) (50 μL per section), visualized using diaminobenzidine (DAB)/H[2]O[2], counterstained with hematoxylin, dehydrated, cleared, and mounted in neutral balsam. Cell observations were made using a fluorescence microscope. The number of TUNEL-positive cells with dark brown nuclear staining was counted, and the apoptotic cell count was determined^[204]86. High-throughput sequencing and analysis Three samples of colonic epithelial tissue were randomly selected from mice in the DMSO treatment group and three from the 100 mg/kg GP treatment group. Total RNA was extracted using the Total RNA Isolation Kit (12183555, Invitrogen, USA). The total RNA OD value was measured quantitatively using a UV spectrophotometer (BioSpectrometer basic, Eppendorf, USA), and RNA integrity was assessed via agarose gel electrophoresis. High-quality total RNA was reverse-transcribed to cDNA to construct an RNA library, and sequencing was performed on Illumina’s NextSeq 500. The raw image data obtained from sequencing were converted to raw reads through base calling. For quality control, sequencing adapters were removed and low-quality sequences were filtered out using cutadapt, leaving “clean reads.” These reads were aligned to the mouse reference genome, followed by gene expression quantification using the R package to obtain a gene expression matrix^[205]87. The “limma” package in R was used to identify differentially expressed genes in the high-throughput sequencing data, using |log2FC | > 2 & P.value < 0.05 as the selection criteria. Volcano and heatmaps were created using the ggplot2 and pheatmap packages, respectively^[206]88–[207]90. Protein sample preparation and analysis in proteomics Colonic epithelial tissue samples from three mice in the DMSO-treated group and three from the 100 mg/kg GP-treated group were ground in a mortar with liquid nitrogen and transferred to a 5 cm³ centrifuge tube. Subsequently, ultrasonication was performed using an ultrasonic cell disruptor (SCIENTZ-IID, Scientz, Ningbo) under ice-cold conditions. The processing medium consisted of phenol extraction buffer containing 10 mM DTT (R0861, Solarbio, Beijing), a 1% proteinase inhibitor cocktail (P6731, Solarbio, Beijing), and 2 mM EDTA (E1170, Solarbio, Beijing). This step was repeated 8 times. Afterward, an equal volume of Tris-saturated phenol (HC1380, BIOFOUNT, Beijing) at pH=8.0 was added, followed by vortex mixing for 4 minutes. The mixture was then centrifuged at 5000 g for 10 minutes at 4 °C, and the upper layer of phenol was transferred to a new centrifuge tube. Ammonium sulfate (0.1 M, 101217, Sigma-Aldrich, USA) saturated methanol (106035, Sigma-Aldrich, USA) was added to the phenol solution in a volumetric ratio of 1:5 and left overnight to precipitate the proteins. After centrifugation at 4 °C for 10 minutes, the supernatant was removed. The remaining precipitate was washed once with ice-cold methanol and three times with ice-cold acetone. The washed proteins were then re-dissolved in 8 M urea (U8020, Solarbio, Beijing), and the protein concentration was determined using a BCA assay kit (P0012, Beyotime Biotechnology Co., Ltd, Shanghai)^[208]91. The procedures were carried out according to the manufacturer’s instructions. Protein digestion and mass spectrometry analysis workflow Each sample received 50 µg for enzymatic digestion. Initially, the protein solution was mixed with DTT to a concentration of 5 mM and left at 56 °C for 30 minutes. Subsequently, acrylamide was added to reach a concentration of 11 mM and left at room temperature for 15 minutes. Finally, the urea concentration in the samples was diluted below 2 M, followed by the addition of trypsin (25200056, Thermo Fisher, USA) at a ratio of 1:50 (w/w) for overnight digestion at 37 °C. Subsequently, trypsin was added again at a ratio of 1:100 (trypsin: protein) for another 4 hours of digestion. After digestion, peptide desalting was performed using the HyperSep™ C18 purification column (60108-302, Thermo Fisher, USA), followed by vacuum drying. The peptides were reconstituted in 0.5 M TEAB (90114, Thermo Fisher, USA) and processed according to the manufacturer’s instructions of the TMT reagent kit (90064CH, Thermo Fisher, USA). In brief, a unit of TMT reagent was thawed and dissolved in acetonitrile (113212, Sigma-Aldrich, USA). The peptide mixture was then incubated at room temperature for 2 hours, desalted and dried using a vacuum centrifuge. Equal amounts of labeled peptides from each group were mixed and subjected to hierarchical separation using the Pierce™ High-pH Reversed-Phase Peptide Isolation Kit (84868, Thermo Fisher, USA). Subsequently, the samples were collected and combined into 15 fractions, and the dried peptides in each fraction were dissolved in 0.1% formic acid (159002, Sigma-Aldrich, USA). Each sample was prepared by taking 2 µg of peptide and separated using the nano-UPLC liquid chromatography system Easy nLC 1200 (Thermo Fisher, USA). The samples were first pre-treated using a Trap C18 column (100 µm × 20 mm, 5 µm), followed by gradient separation using a C18 analytical column (75 µm × 150 mm, 3 µm) at a flow rate of 300 nL/min. The mobile phase A consisted of a 0.1% formic acid aqueous solution, while mobile phase B consisted of a 0.1% formic acid aqueous solution with 95% acetonitrile. The gradient elution process included: 0-2 minutes, 2%-8% B; 2-71 minutes, 8%-28% B; 71-79 minutes, 28%-40% B; 79-81 minutes, 40%-100% B; 81-90 minutes, 100% B. The liquid chromatography-separated peptides were analyzed by a Q-Exactive HF-X mass spectrometer (Thermo Fisher, USA). The analysis duration was 60 minutes, with an electrospray voltage set at 2.1 kV, operating in positive ion mode, with a precursor ion scan range of 350-1200 m/z and a primary mass resolution of 60,000 at m/z 200. The AGC target was set at 3e6, and the maximum injection time for the primary scan was 30 ms. The secondary mass resolution was 15,000 @ m/z 200 with an AGC target of 1e6 and a maximum injection time of 25 ms. The MS2 activation type was set as HCD, with an isolation window of 20 Th and a normalized collision energy of 32^[209]91,[210]92. Database retrieval and data processing The LC-MS/MS data obtained were processed using MaxQuant software (v1.5.2.8), which involved peptide identification and protein quantification. The UniProt 14.1 database (2009) ([211]https://www.uniprot.org/) was utilized for tandem mass spectrometry retrieval in conjunction with a decoy database. Trypsin/P was designated as the cleaving enzyme, allowing for up to 2 missed cleavages. The initial search was set at 20 ppm, while the main search was set at 5 ppm with a fragment ion mass tolerance of 0.02 Da. Criteria for peptide false discovery rate (FDR) ≤ 0.01, protein FDR ≤ 0.01, and peptide score distribution were established for database qualification. Differential expression proteins (DEPs) between colonic tissue samples were screened using the “limma” package in R software (with thresholds set at |log2FC | > 2 and P.value < 0.05)^[212]91,[213]93. PPI network analysis DEPs identified in colonic tissue samples were utilized to construct PPI networks by importing them into the STRING database ([214]https://cn.string-db.org/) and further analyzed using Cytoscape v3.10.0 software to generate the protein interaction network diagram^[215]94. MTT assay for assessing colonic epithelial cell viability The MTT assay involved seeding the cells onto a 96-well cell culture plate at a density of 3-5 × 10^4 cells/mL and then culturing them for 48 hours. Subsequently, MTT solution (10 mg/mL, ST316, Beyotime Biotechnology Co., Ltd, Shanghai, China) was added to the cell suspension and incubated for 4 hours, followed by the addition of dimethyl sulfoxide (DMSO) with shaking for 10 minutes. Absorbance at 490 nm was measured using a spectrophotometer (Laspec, China). Cell viability was calculated as (ΔA_sample - ΔA_blank)/(ΔA_control - ΔA_blank), where ΔA_sample represents the absorbance difference of the sample, ΔA_blank is the absorbance difference of the blank, and ΔA_control is the absorbance difference of the control group^[216]95. EdU experiment Colonic epithelial cells were seeded in a 24-well plate and incubated in a culture medium containing EdU ([217]C10337, Invitrogen, USA) at a concentration of 10 µmol/L for 2 hours in a CO[2] incubator. Subsequently, the culture medium was removed, and the cells were fixed with a DMSO solution containing 4% paraformaldehyde at room temperature for 15 minutes. The cells were then washed twice with DMSO containing 3% BSA, permeabilized at room temperature with DMSO containing 0.5% Triton X-100 for 20 minutes, followed by another two washes with DMSO containing 3% BSA. 100 µL of staining solution was added to each well and incubated at room temperature in the dark for 30 minutes. DAPI nuclear staining was performed for 5 minutes, and the slides were then observed using a fluorescence microscope (BX63, Olympus, Japan) to randomly examine 6-10 fields of view and record the number of positive cells in each field. The EdU labeling rate (%) was calculated as the number of positive cells divided by the sum of positive and negative cells multiplied by 100%^[218]96. Each experiment was repeated three times. TUNEL staining Colonic epithelial cells from each group were fixed with 4% paraformaldehyde at room temperature for 15 minutes, followed by permeabilization with 0.25% Triton X-100 for 20 minutes. Samples were then blocked with 5% bovine serum albumin (BSA, 36101ES25, Yeasen Biotechnology (Shanghai) Co., Ltd., China) and stained with TUNEL (C1086, Beyotime Biotechnology Co., Ltd, Shanghai, China) reagent. Subsequently, the sections were counterstained in the dark with a DAPI staining solution (C1002, Beyotime Biotechnology Co., Ltd, Shanghai, China). Apoptotic cells were visualized under a confocal microscope (LSM 880, Carl Zeiss AG, Germany). TUNEL-positive cells (green fluorescence) indicated apoptotic cells, while DAPI labeled the cell nuclei, with blue fluorescence representing the total cell count. Five different fields were selected from each group to calculate the percentage of apoptotic cells, which is calculated as the number of apoptotic cells divided by the total cell count multiplied by 100%^[219]97. Substance permeability assessment (SPE) To establish a single-cell barrier model, 0.3 × 10^5 murine colonic epithelial cells were suspended on polycarbonate membranes of transwell chambers (3413, Corning, USA) and cultured at 37 °C with 5% CO[2] for 21 days. The transepithelial electrical resistance (TEER) of the monolayer was measured using the Millicell-ERS resistance system (Millipore, Billerica, MA, USA), and after 21 days of cultivation, the average TEER value exceeded 400 Ω·cm^2 (Fig. [220]S9). Induction of barrier dysfunction in the single-cell barrier was achieved by treating with 100 μg/mL lipopolysaccharide (LPS) for 48 hours, followed by replacing the culture medium in the upper and lower chambers with a serum-free medium. Fluorescein isothiocyanate (FITC)-labeled dextran (70 kDa, 2 mg/mL; FD70, Sigma-Aldrich, USA) was added to the upper chamber, and the concentration of albumin-FITC in the lower chamber was measured at 495 nm wavelength using an ELISA^[221]98. Preparation of GP and NCC composite (NCC-GP) GP was dissolved in 10 mM phosphate buffer (AM9624, Invitrogen, USA) at pH 7.4. A 1.5 mL GP solution was added to a 2 mL microcentrifuge tube containing a 4 mg/mL suspension of NCC (C909405, MACKLIN, China), and the mixture was stirred at 37 °C with a stirring speed of 8 revolutions per minute for 30 minutes. The suspension, containing phosphate buffer, formed a flocculated NCC/GP suspension, which was centrifuged at 18000 g for 10 minutes to precipitate the GP and NCC composite. The concentration of unbound GP ([drug[unbound]]) in the supernatant was determined using a Varian 50 Bio UV Vis spectrophotometer (Varian, Inc., Canada) at wavelengths of 482 nm and 364 nm. The concentration of GP bound to NCC ([drug[bound]]) was calculated using the formula: [MATH: [drugbound]=[drugadded][drugunbound] :MATH] ^[222]54. Characterization of NCC physicochemical properties The zeta potential of NCC in phosphate buffer was measured in an Eppendorf UVette centrifuge tube using Zeta Plus (Brookhaven Instruments Corp., USA) at a scattering angle of 90 °. Three measurements were conducted. The structure and morphology of NCC were examined using scanning electron microscopy (JEOL 6700, JEOL, Japan). The method for scanning electron microscopy examination involved dropping the NCC suspension onto a clean silicon wafer to form a small droplet (approximately 5 μL), followed by overnight drying for preparation^[223]99. Drug release study For the release study, the NCC samples loaded with the drug were resuspended in PBS and incubated at 37 °C with a speed of 8 rpm. At specified time points, the suspension was centrifuged at 18,000 g for 10 minutes to remove the supernatant, and drug quantification was performed by measuring GP using UV-Vis spectroscopy or high-performance liquid chromatography (HPLC). The amount of unbound drug in the supernatant was determined by HPLC using a Waters HPLC system equipped with Millennium software and UV-visible detection (Waters Ltd, Canada). A Novapak C18 column with an injection volume of 20 μL, a flow rate of 1 mL/min, and a mobile phase consisting of acetonitrile and formic acid water (containing 0.05% formic acid) was used for separation. Detection was performed at 273 nm. At each sampling point, fresh PBS was added to the tube, and the NCC/GP nanocomplex was resuspended^[224]54,[225]61. Study on cell binding and uptake The test cells were seeded at a concentration of 10,000 cells/well in a 96-well cell culture plate and allowed to equilibrate overnight. A suspension of 2 mg NCC-GP in a 5 mm NaCl solution was prepared and incubated at 37  °C for 30 minutes. The suspension was centrifuged at 18000 g for 30 seconds to remove the supernatant. The pellet was resuspended in a 5 mm NaCl solution containing 100 μg of fluorescein (HY-D0251, MedChemExpress, USA) per milliliter and then further incubated at 37  °C for 30 minutes. After another centrifugation at 18000 g for 30 seconds and removal of the supernatant, the pellet was resuspended in DMSO and diluted to obtain a 10 mg/mL sample. Subsequently, 200 μL of NCC-GP suspension was added to each well of the cell culture plate, and the cells were allowed to incubate for 24 hours. The culture medium was removed, and the cells were washed three times with PBS, followed by lysis with 2% Triton X-100. The fluorescence intensity of the bound NCC-GP/fluorescein on the cells was measured using a fluorescence spectrophotometer with excitation and emission wavelengths of 480 and 520 nm, respectively. These fluorescence intensity values were converted to the mass of bound NCC-GP/fluorescein on the cells using a standard curve previously constructed. The BCA total protein analysis kit (P0012, Beyotime Biotechnology Co., Ltd., Shanghai, China) was used to ensure an equal number of cells in each well. For the observation of cell uptake of NCC, the test cells were cultured following the binding study protocol. Disinfected glass coverslips with a diameter of 1 cm were placed at the bottom of a 6-well tissue culture plate, and 50,000 cells were added to each well and incubated overnight under culture conditions. The cells were then incubated at 37  °C with a suspension of 1.25 mg/mL NCC/cetyltrimethylammonium bromide (CTAB)/fluorescein for varying durations. The coverslips were removed, washed three times in PBS, treated with DAPI dye for 30 minutes, washed again, and then treated with a 3.7% formaldehyde solution in deionized water for 30 minutes. The coverslips with the cells facing down were mounted on glass microscope slides and sealed with transparent nail polish. Visualization of the fluorescein and DAPI-stained cell nuclei was performed using an FV-1000 laser confocal scanning microscope (Olympus, Japan)^[226]54. The subsequent cell experimental groups were as follows: (1) DMSO + Nor group: untreated normal colonic epithelial cells cultured with 200 μL of DMSO solution; (2) NCC + Nor group: untreated normal colonic epithelial cells cultured with 200 μL of 10 mg/mL NCC solution; (3) NCC-GP + Nor group: untreated normal colonic epithelial cells cultured with 200 μL of 10 mg/mL NCC-GP solution; (4) DMSO group: LPS-stimulated colonic epithelial cells cultured with 200 μL of DMSO solution; (5) GP group: LPS-stimulated colonic epithelial cells cultured with 200 μL of 10 mg/mL GP solution; (6) NCC group: LPS-stimulated colonic epithelial cells cultured with 200 μL of 10 mg/mL NCC solution; (7) NCC-GP group: LPS-stimulated colonic epithelial cells cultured with 200 μL of 10 mg/mL NCC-GP solution. ELISA Colonic epithelial cells and murine colonic tissue levels of inflammatory cytokines were determined using ELISA kits purchased from Invitrogen (USA), as indicated in Table [227]S4^[228]100,[229]101. RT-qPCR Total RNA was extracted from tissues or cells using Trizol reagent (15596026, Invitrogen, USA). Subsequently, the concentration and purity of the total RNA were assessed at 260/280 nm using NanoDrop LITE (ND-LITE-PR, Thermo Scientific™, Germany). Next, cDNA was synthesized from the extracted total RNA using the PrimeScript RT reagent Kit with gDNA Eraser (RR047Q, TaKaRa, Japan). The expression levels of various genes were then determined by RT-qPCR using SYBR Green PCR Master Mix (4364344, Applied Biosystems, USA) and the ABI PRISM 7500 sequence detection system (Applied Biosystems). The primers for the genes were synthesized by TaKaRa (Table [230]S5), with GAPDH serving as the reference gene. Finally, the relative expression levels of the genes were analyzed using the 2^-ΔΔCt method, where △△Ct = (average Ct value of the target gene in the experimental group - average Ct value of the reference gene in the experimental group) - (average Ct value of the target gene in the control group - average Ct value of the reference gene in the control group)^[231]102–[232]104. All RT-qPCR analyses were performed in triplicate. Western blot To begin, tissues or cells were collected and lysed using an enhanced RIPA lysis buffer containing protease inhibitors (P0013B, Beyotime Biotechnology Co., Ltd, Shanghai, China). The protein concentration was then determined using a BCA protein quantification kit (P0012, Beyotime Biotechnology Co., Ltd, Shanghai, China). Subsequently, proteins were separated by 10% SDS-PAGE and transferred onto a PVDF membrane (FFP39, Beyotime Biotechnology Co., Ltd). The membrane was blocked with 5% BSA (ST023, Beyotime Biotechnology Co., Ltd) at room temperature for 2 hours to prevent nonspecific binding, followed by the addition of diluted primary antibodies and incubation at room temperature for 1 hour. The primary antibodies used were rabbit anti-human (details in Table [233]S6). After washing the membrane, an HRP-conjugated goat anti-rabbit secondary antibody (ab6721, 1:2000, Abcam, UK) was added and incubated at room temperature for 1 hour. Pierce™ ECL Western Blotting Substrate (32209, Thermo Scientific™, Germany) A and B solutions were mixed in equal amounts in a dark room, added to the membrane, and exposed in a gel imaging system. Images were captured using a Bio-Rad imaging system (BIO-RAD, USA) and the bands in the Western blot images were quantified for grayscale using Image J analysis software, with GAPDH as an internal control^[234]103. Each experiment was repeated 3 times. Assessment of intestinal barrier integrity The test is based on examining the integrity of the intestinal barrier using 4 kD fluorescein isothiocyanate-dextran (FD4, Sigma-Aldrich). After 4 hours of fasting, mice were orally administered FITC-dextran (0.6 g per kg body weight). One hour later, 200 μL of blood was collected into Microvette tubes (Sarstedt, France, Marnay). The tubes were then centrifuged at 10,000 g for 5 minutes at room temperature to extract serum. The collected serum was diluted with an equal volume of DMSO and analyzed for FITC concentration using an excitation wavelength of 485 nm and an emission wavelength of 535 nm. All analyses were conducted under blinded experimental conditions^[235]45. FMT Recipient mice were provided with a synthetic antibiotic combination in their drinking water for six consecutive days, including vancomycin (0.5 g/L; V2002), ampicillin (1 g/L; 171254), streptomycin (5 g/L; 85886), neomycin (1 g/L; N6386), and metronidazole (0.5 g/L; M1547), all sourced from Sigma-Aldrich. After 24 hours, the animals were orally administered the microbial inoculum twice, with a three-day interval between administrations. The inoculated animals were housed in separate sterile isolators. Fresh fecal pellets were collected directly from the donor mouse’s rectum, homogenized in 1 mL of sterile phosphate-buffered saline (DMSO) containing 100 mg (approximately 5-6 fecal pellets). The homogenate was then filtered through a 20 μm nylon filter to remove large particles and fibrous materials. The solution was collected and administered to recipient mice orally in a volume of 200 µL to minimize microbial content changes within 15 minutes. Prior to any analysis, the recipient animals were housed in isolators for eight weeks. The choice of an eight-week delay between FMT and analysis was made because the C57BL/6 J mouse strain is considered to have relatively strong stress resistance and requires at least eight weeks to exhibit depression-like behavior (Fig. [236]S10)^[237]79. Immunohistochemistry (IHC) staining For a list of antibodies, please refer to Table [238]S7. Mouse tissues were fixed with 4% paraformaldehyde overnight and then processed for paraffin embedding with a slice thickness of 4 μm, followed by deparaffinization in xylene. The tissue sections were rehydrated in a series of graded ethanol washes (absolute ethanol, 95% ethanol, and 75% ethanol for 3 minutes each). Antigen retrieval was achieved by boiling the sections in 0.01 M citrate buffer for 15-20 minutes, followed by quenching endogenous peroxidase activity at room temperature for 30 minutes with 3% H[2]O[2]. The sections were then blocked with goat serum blocking solution at room temperature for 20 minutes, excess liquid was removed, and primary antibodies were applied and left to incubate at room temperature for 1 hour. Subsequently, the sections were washed with DMSO and incubated with a secondary IgG goat anti-mouse antibody at 37 °C for 20 minutes, followed by washing with DMSO and incubation with SP (streptavidin-peroxidase) at 37 °C for 30 minutes. After washing with DMSO, the sections were developed using DAB (P0202, Beyotime Biotechnology) for 5-10 minutes, followed by a 10-minute water rinse to stop the reaction. Counterstaining was performed with hematoxylin (C0107, Beyotime Biotechnology) for 2 minutes, differentiation with acid alcohol, a 10-minute water rinse, dehydration in graded alcohols (xylene clearing), and mounting with 2-3 drops of neutral resin. Observations and counting were done under a bright-field microscope: 5 high-power fields per section were randomly selected, with 100 cells counted per field, to determine the Ki67-positive cell percentage^[239]105. Statistics and reproducibility Our study utilized the R statistical programming language, version 4.2.1, with code compilation facilitated through the integrated development environment RStudio (version 2022.12.0-353). For file processing, Perl programming language version 5.30.0 was employed. Additionally, data analysis involved the utilization of GraphPad Prism software (version 8.0). Descriptive statistics were presented using mean ± standard deviation for continuous variables. Group comparisons were conducted using an independent samples t-test^[240]106. Analysis of variance (ANOVA) was employed for comparing data among different groups, while two-way ANOVA was utilized for assessing data variations across different time points. Post-hoc testing was performed using Bonferroni adjustment. The threshold for statistical significance was set at P < 0.05^[241]107. For animal experiments, each group consisted of at least six mice (n = 6 per group), determined based on power analysis to ensure sufficient statistical significance. Behavioral tests were conducted in biological triplicates, considering each mouse as an independent biological replicate. In vitro experiments, including cellular assays, Western blot, RT-qPCR, and ELISA, were performed in technical triplicates and repeated in at least three independent biological replicates (n ≥ 3). Details of sample sizes and statistical tests applied for specific experiments are provided in the figure legends and Methods section. To ensure reproducibility, all experiments were performed independently at least three times, and results were verified across different experimental batches. Blinding was applied where applicable, particularly in behavioral assessments and histological analyses, to minimize bias. Data consistency was evaluated across independent experiments, ensuring robust and reproducible findings. Reporting summary Further information on research design is available in the [242]Nature Portfolio Reporting Summary linked to this article. Supplementary information [243]Supplementary Information^ (3.4MB, pdf) [244]Reporting summary^ (1.6MB, pdf) Acknowledgements