Animals
Male WT C57BL/6 J mice, TrdcCreERT2 mice, Il7rflox/flox mice, Cd8aflox/flox mice, and Cx3cr1Cre mice (all on a C57BL/6 background), weighing between 23.0 and 25.0 grams and aged between 8 and 10 weeks, were obtained from Vital River Laboratory Animal Technology Co Ltd., Beijing, China. TrdcCreERT2 mice were crossed with Il7rflox/flox mice and Cd8aflox/flox mice to generate TrdcCreERT2Il7rflox/flox and TrdcCreERT2Cd8aflox/flox offspring. To induce Cre activity in γδ T cells, tamoxifen (150 mg kg− 1) was administered intraperitoneally to TrdcCreERT2Il7rflox/flox mice and TrdcCreERT2Cd8aflox/flox mice for five consecutive days. The CLP model was performed one week after tamoxifen induction. Kaede-transgenic (Kaede-Tg) mice (B6. Cg-Gt (ROSA)26Sor < tm1.1(CAG-kikGR) Kgwa>) were generously provided by M. Tomura of Kyoto University. The mice were housed in a controlled, specific pathogen-free environment. They were exposed to a 12:12 light/dark cycle, maintained at a regulated temperature and humidity, and provided with unrestricted access to food and water. Ethical approval for all experiments was obtained from the Experimental Animals Committee of Tongji Medical College (permission number: 4028), in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals. The study adhered to ARRIVE guidelines (Animals in Research: Reporting In Vivo Experiments).
Animal model
Mice were anesthetized with sodium pentobarbital (0.3% solution) at a dose of 40 mg kg–1 body weight. A midline laparotomy was performed to conduct a central lymphadenectomy, following which the abdominal region was shaved and sterilized.
A 1 cm incision was made along the midline to expose the cecum, which was ligated 1 cm from the distal end using a 4 − 0 silk suture. A 20-gauge needle was then used to puncture the cecum, allowing a small amount of cecal content to extrude from both openings. After the procedure, the ligated cecum was returned to the abdominal cavity, and the incision was closed in multiple layers. Either 50 mg kg–1 of 4-OI or 10 ml kg–1 of 0.9% saline was administered intraperitoneally immediately after CLP or sham-operation. Inhibitor H151 was administered at a concentration of 750 nM via intraperitoneal injection, three times per week for three weeks prior to the CLP procedure. Survival rates were monitored and assessed 7 days post-CLP.
To increase IL-17A expression in the brain, mice received adenoviruses (Adv) via lateral ventricle injection three weeks before sham surgery or CLP. Each Adv injection contained 1 × 1012 plaque-forming units (PFU) of IL-17A-expressing recombinant AAV (IL-17A+-Adv). Control mice received an equivalent dose of Adv expressing GFP. In a separate experimental group, IL-17A neutralizing antibody was administered intraperitoneally at 100 µg per day for 5 days, with four doses given prior to the CLP challenge and one dose immediately after. Control mice received equivalent doses of normal hamster serum IgG.
To specifically interfere with gene expression in microglia, we injected Cx3cr1Cre mice with DIO-AAV vectors. The AAVs were obtained from Brainvta (Wuhan, China) and included the following constructs: rAAV-SFFV-DIO-cGAS-2a-EGFP-WPREs, rAAV-SFFV-DIO-RNF5-His-2a-EGFP-WPREs, rAAV-CWV-DIO-(EGFP-U6)-shRNA1(Acod1), and rAAV-SFFV-DIO-STING1 (K150R)-2a-EGFP-W.
Kaede photoconversion
To track immune cells in the small intestines in vivo, we performed photoconversion following the method described in a previous study8. Briefly, following CLP or sham surgery, the small intestine of Kaede-transgenic mice was exposed to a 405-nm laser for 10 min, while the surrounding tissue was shielded from light with aluminum foil.
Brain stereotaxic injection
A previous study35 detailed the stereotaxic injection of viruses and neutralizing antibodies into the hippocampus. Briefly, mice were anesthetized with 1–2% isoflurane. Openings were made at specific coordinates targeting the unilateral hippocampal CA1 region (x: ± 2.15 mm; y: − 2.5 mm; z: − 2.25 mm). A volume of 400 nanoliters (nL) was injected into each hippocampus at a rate of 26.67 nL min–1. Following the injection, the needle was left in place for 5 min to ensure proper diffusion before removal. The CLP model was performed 21 days post-viral injection and 1 day after C1q neutralizing antibody administration.
Morris water maze
Experimental data were collected in a 120 cm circular pool filled with opaque water maintained at 20–22 ℃. Mice were trained four times daily for four days to locate a hidden platform submerged 1 cm below the water surface. The average latency to find the platform was calculated from four trials. Each mouse was allowed 60 seconds per trial to search for the platform. Mice remained on the platform for 15 seconds if they found it, or were placed on it for 10 seconds if they failed to locate it. A probe trial without the platform was conducted 24 h after the final hidden platform test. Mice were tested for 60 seconds to locate the platform. The number of times the mice crossed the platform target and the total time spent in the target quadrant were recorded. Mice were monitored via video cameras throughout training and probe trials. Data were analyzed using TopScan Lite (Clever Sys. Inc.).
Y-maze test
Three identical arms (30 cm long, 5 cm wide, 20 cm high) were positioned at 120° angles in the Y-maze (YM). In the initial 10-minute training session, mice explored two arms while the third arm was blocked. One hour later, mice were given free access to all arms during the retention test. Mice were recorded exploring the novel arm for 5 min. Each trial was separated by cleaning the Y-maze arms with 75% ethanol. Arm entries and time spent in the novel arm were recorded, and short-term memory was calculated as the ratio of novel arm time to total exploration time. Data were examined using TopScan Lite (Clever Sys. Inc.).
Open filed test
The testing area was an acrylic box measuring 40 cm long, 40 cm wide, and 30 cm high. The center of the base contained a 20 x 20 cm square core region. Each mouse was gently placed in the center of a dimly lit open-field arena and allowed to explore for 5 min. A mobile camera automatically recorded and tracked their movement. The time spent in the central region was used to assess exploratory behavior. To eliminate odors, the chamber floor was wiped with 75% ethanol after each session. Data were examined using TopScan Lite (Clever Sys. Inc.).
Novel objection recognition
Two similar objects were placed at the edges of a 40 × 40 × 30 cm box. Mice were then gently placed in the box and allowed to explore the objects freely for 5 min, with the time spent on each object recorded. Exploration was defined as sniffing or touching an object within 0–2 cm with the nose. One hour later, one object was replaced with a novel one, and mice were given another 5 min to explore both objects, with the time spent on each recorded. The novel object recognition rate was calculated as (time spent on the novel object / total time spent on both objects) × 100%.
Tissue preparation
Cold PBS was perfused transcardially into sedated mice. Brains and small intestines were then collected. For immunofluorescent labeling, tissues were fixed overnight at 4 ℃ in 4% paraformaldehyde and cryoprotected in 30% sucrose for at least 2 days. Brains were sectioned at 30 µm thickness using a Leica CM1950 cryostat. Small intestines were paraffin-embedded and sectioned at 4 µm thickness. Blood was collected from sedated mice via cardiac puncture into anticoagulant tubes for biochemical analysis. Plasma was obtained and stored at − 80 ℃. Small intestines were stored at − 80 ℃ after a gentle flush with cold PBS. After dissociation, the hippocampus was snap-frozen in liquid nitrogen and stored at − 80 ℃ for protein extraction.
Immunofluorescence
Brain slices were washed in PBS and blocked for 2 h at room temperature with 5% BSA (Biofroxx, Germany) and 0.3% Triton X-100 in PBS. The sections were incubated overnight at 4 ℃ with mouse anti-Iba1 (1:100, Abcam) and rabbit anti-PSD95 (1:250, CST) primary antibodies. After PBS washing, the sections were incubated with Alexa Fluor 488-conjugated goat anti-rabbit (1:1000, Abcam) and Alexa Fluor 549-conjugated goat anti-mouse (1:1000, Abcam) for 1 h at room temperature the following day. The slices were mounted with SouthernBiotech DAPI Fluoromount-G after a final wash.
After deparaffinization in xylene, intestinal sections were rehydrated through a graded ethanol series to water. Sections were heated in citrate buffer (pH 6.0) for 20 min for antigen retrieval. After cooling to room temperature, the sections were blocked for 1 h with 5% BSA and 0.3% Triton X-100 in PBS. Sections were incubated overnight at 4 ℃ with rabbit anti-ZO-1, rabbit anti-MUC2, and rabbit anti-Occludin primary antibodies. The following day, sections were washed in PBS and incubated with Alexa Fluor 449-conjugated goat anti-rabbit (1:1000, Abcam) for 1 h at room temperature. The slices were mounted with SouthernBiotech DAPI Fluoromount-G after a final wash.
Microscopy and analysis
Immunofluorescent-stained brain slices were imaged using a Dragonfly spinning disk confocal microscope (Andor Technology) equipped with 405, 488, and 561 nm laser lines. Z-stack images were captured using a 60x oil immersion objective. Image processing and analysis were performed using Imaris 10.2 (Bitplane). Z-stack images were generated using Imaris 10.2 for 3D reconstruction and quantification of Iba1 and PSD95 expression. Immunofluorescent signal intensity in ROIs was compared across experimental groups.
Immunofluorescent-stained small intestine sections were scanned at 20x magnification using an Olympus VS200 slide scanner. High-resolution images were analyzed using Image J (1.8.0, NIH, USA). The fluorescence intensity of ZO-1, MUC2, and Occludin was quantified in randomly selected fields of view.
Primary microglia culture
Based on a recent study36, we made slight modifications to the primary microglia culture procedure. Cortical brain tissues from 1-day-old C57BL/6J mice were dissected in cold HBSS (Servicebio, China, cat. no. G4203). After removing the meninges, the brain tissues were minced and rinsed three times with HBSS. The tissues were then digested in 0.25% trypsin-EDTA (ThermoFisher, cat. no. 25200056) for 20 min at 37 ℃ and triturated into a single-cell suspension. The primary cells were plated on poly-L-lysine-coated plates in Neurobasal medium supplemented with 10% FBS (Gibco, USA. cat. no. 10099-141), 1% GlutaMAX (Gibco, USA. cat. no. 35050061), and 1% penicillin-streptomycin (P.S.) (Gibco, USA. cat. no. 15140122). After 10 days of culture, the cell culture flasks were shaken at 250 rpm at 37 ℃ for 2 h. The collected culture media were centrifuged at 1000 rpm for 10 min, and the cells were resuspended in DMEM (Gibco, USA. cat. no. 11995073) with 10% FBS for inoculation
Primary γδ T culture
Primary γδ T cells were isolated from mouse spleens. Briefly, T25 cell culture flasks were coated with 5 µg mL–1 TCR γδantibody one day prior. After anesthesia, mouse spleens were extracted under sterile conditions, homogenized with 2 mL of precooled mouse lymphatic separation solution, filtered through a 70-mesh filter, gently layered over 5 mL of precooled DMEM medium, and centrifuged at 2000g for 20 min. The intermediate layer was carefully aspirated, washed with PBS, and centrifuged at 420 × g for 6 min at 4 ℃. Cells were resuspended in inoculation medium (DMEM + 10% FBS + 1% P.S + 0.1 mM β-mercaptoethanol + 5 µM Zoledronic acid monohydrate + 1000 IU IL-2 + 20 ng mL–1 IL-7) at 1 × 10⁵ cells mL–1 and seeded into six-well plates for growth. The culture medium was replaced every 3 days with DMEM containing 10% FBS, 1% P.S, 0.1 mM β-mercaptoethanol, 1000 IU IL-2, and 20 ng mL–1 IL-7. On day 12, primary cells were transfected and stimulated.
Primary γδ T cells-microglia co-culture
Primary γδ T cells were isolated from mouse spleens, and microglia were obtained from the brains of postnatal day 1 (P1) mice. For co-culture experiments, microglia were seeded in the bottom chamber of a transwell device with 0.4 µm pore-size polycarbonate membrane inserts, while γδ T cells were seeded in the top chamber. To investigate the impact of IL-17A, γδ T cells were transfected with siIL-17A or siCtrl using Lipofectamine RNAiMAX. Additionally, γδ T cells were treated with recombinant mouse IL-17A (100 ng mL–1) and LPS (1000 ng mL–1) for 6 h, followed by a medium change. The transwell inserts containing γδ T cells and microglia were co-cultured for 24 h post-treatment. After co-culture, microglia were harvested for analysis.
Cell Culture and treatment
BV2 murine microglial cells were obtained from Punosai Life Science and Technology Co., Ltd. and cultured in RPMI-1640 medium (Gibco, USA. cat. no. 11875119) supplemented with 10% FBS and 1% P.S. at 37 ℃ in 5% CO2. To model SAE, BV2 cells were treated with 1000 µg mL–1 LPS for 6 h. After incubation, Cells were collected and processed for further assays. To block the cGAS-STING pathway, BV2 cells were treated with H151 (0.75 µM) for 2 h prior to LPS stimulation. To generate BV2ρ0 cells, which are devoid of mitochondria, BV2 cells were treated with ethidium bromide (50 ng mL–1) for 4 weeks37.
Cell Transfection
For gene silencing, primary γδ T cells were transfected with IL-17A siRNA, and BV2 microglial cells transfected with RNF5 siRNA. Additionally, BV2 cells were transfected with STING protein plasmids containing site-directed mutations for ubiquitination site analysis. All siRNAs and plasmids were obtained from Obio Technology Co., Ltd., Shanghai. Following the manufacturer’s instructions. Lipofectamine RNAiMAX was used for siRNA transfections, and Lipofectamine 3000 was used for plasmid transfections. Transfection efficiency was confirmed by Western blot analysis.
Western blot
Total proteins were extracted from cells and tissues, and protein concentrations were measured using the BCA assay. Equal amounts of protein were separated by SDS-PAGE and transferred to PVDF membranes (Millipore). Membranes were incubated overnight at 4 ℃ with primary antibodies against ZO-1, MUC-2, Occludin, cGAS, STING, C1q, PSD95, Synaptophysin (SYN), ACOD1, RNF5, Ubiquitin (UB), β-actin,GAPDH and a custom-made antibody for the K150 ubiquitination site on STING (Abclonal). After incubation, membranes were treated with HRP-conjugated anti-mouse or anti-rabbit IgG. Protein bands were visualized using ECL Western blot Detection Reagents (Beyotime) and captured with a UVP gel documentation system (UVP, LLC, Phoenix). Band intensity was quantified using ImageJ (1.8.0, NIH, USA).
Synaptosomal proteins were extracted from mouse hippocampi using the Syn-PER Synaptic Protein Extraction Reagent (Thermofisher) following the manufacturer’s instructions. Western blot analysis was then performed on the isolated synaptosomal proteins.
FCM analysis
As previously dicribed8, cell suspensions from the meninges and small intestine were prepared. After counting 1 × 106 cells, they were resuspended in 100 µL of 1% BSA in PBS, blocked with 5 ng µL–1 anti-CD16/CD32 antibody for 5 min at 4 ℃, and then stained with the desired antibodies. Antibodies used for extracellular staining included fixable viability dye, CD45, CD3, TCR γδ, IL7R, CD8, F4/80, CD206, CD86, and CD11B. For intracellular staining, cells were first labeled with surface markers, then frozen and permeabilized before being labeled with IL-17A antibody (4 ng µL–1). Samples were analyzed using a Beckman CytoFLEX or BD LSRFortessa X-20 cytometer (BD Biosciences, San Jose, CA). Data were analyzed using FlowJo 10.0 (FlowJo, Oregon, USA).
JC-1 Assay
Mitochondrial membrane potential in BV2 microglial cells was measured using the JC-1 detection kit (Elabscience, China) according to the manufacturer’s instructions. BV2 cells were seeded in 6-well plates following experimental criteria. After treatment, cells were stained with JC-1 at 37 ℃ for 20 min. The cells were rinsed with buffer solution and analyzed using a fluorescence microscope or flow cytometer after incubation. The mitochondrial membrane potential was assessed by the red-to-green fluorescence ratio.
Ubiquitination level detection
MG132 (100 µM) was added to cells 6 h before collection to inhibit proteasome activity and accumulate ubiquitinated proteins in cell culture studies. Hippocampus tissues were lysed in IP lysis buffer containing protease and phosphatase inhibitors to extract proteins. The lysates were incubated overnight at 4 ℃ with an anti-STING antibody and protein A/G magnetic beads for co-immunoprecipitation. After washing, the co-immunoprecipitated proteins were eluted from the beads. Eluted proteins were analyzed by Western blot using an anti-UB antibody (1:1000, Proteintech) to measure ubiquitination.
Detection of ubiquitination modification sites
Immunoprecipitation (IP) was performed to pull down STING from BV2 microglial cells for the identification of ubiquitination modification sites. Ubiquitination sites were identified by mass spectrometry following STING isolation. The identified modification sites were used to create mutant plasmids for each ubiquitination site using site-directed mutagenesis. These mutant plasmids were then transfected into BV2 cells to specifically alter STING ubiquitination sites. Ubiquitination levels of WT and mutant STING proteins were compared in subsequent assays to identify key ubiquitination sites.
Microglia depletion
PLX3397 (Selleck, China) was used to deplete brain microglia. As previously reported38, PLX3397 was incorporated into AIN-76A chow at a concentration of 300 mg kg–1. The PLX3397-supplemented chow was prepared by Jiangsu Xietong, Inc., Nanjing. Mice were fed AIN-76A chow or PLX3397-supplemented chow (300 mg kg–1 in AIN-76A) for 21 days. Microglia depletion was evaluated by quantifying microglia numbers and assessing Iba1 expression after the 21-day feeding period.
Golgi staining
After anesthesia, mouse brains were immediately placed in Golgi stain fixing solution and immersed in dye solution for 14 days at room temperature in the dark. The treatment solution was changed after 1 h and then stored at 4 ℃ in the dark for 3 days. Brain samples were sectioned into 60 µm coronal slices using a vibrating microtome. Dendritic spine morphology was examined under a microscope.
Reactive Oxygen Species (ROS) Detection by DHE Staining
Oxidative stress in the hippocampal CA1 region was measured using dihydroethidium (DHE) staining. Brain slices from treated animals were processed as previously described. Sections were treated with Beyotime DHE staining reagent according to the manufacturer’s instructions. F Slices were incubated with DHE (10 µM) for 30 min in a light-protected, humidified chamber at 37 ℃. Superoxide anions oxidize DHE to ethidium, which intercalates with DNA and fluoresces red, indicating tissue ROS levels.
After incubation, sections were washed in PBS and mounted with anti-fade media. Fluorescent signals were captured using an Olympus fluorescence microscope (Olympus, Japan) with DHE filters. Red fluorescence intensity in the hippocampal CA1 region was quantified using ImageJ (NIH, USA) to assess oxidative stress. Oxidative stress levels between experimental groups were assessed by statistical analysis of mean fluorescence intensity.
TEM
Mice were perfused transcardially with 2.5% glutaraldehyde. 1-mm coronal brain slices were collected immediately after perfusion. The CA1 region of the hippocampus was meticulously microdissected and post-fixed in 2.5% glutaraldehyde in 0.1 M sodium cacodylate buffer at 4 ℃ for 24 h. After treatment, BV2 and primary microglia were centrifuged and fixed in 2.5% glutaraldehyde in 0.1 M sodium cacodylate buffer at 4 ℃ for 24 h.
Both tissue samples and cells were processed similarly after fixation. After three washes with 0.1 M sodium cacodylate buffer, the samples were post-fixed in 1% OsO₄ in the same buffer at 4 ℃ for 1.5 h. After dehydration in graded ethanol (50%, 70%, 90%, 100%), the samples were embedded in epoxy resin. Thin sections (60–70 nm) were cut using a Leica EM UC7 ultramicrotome and placed on copper grids. For contrast enhancement, sections were stained with 2% uranyl acetate for 15 min and lead citrate for 10 min. Electron micrographs were captured using an 80 kV Hitachi HT7800 transmission electron microscope (HITACHI, Japan). Ultrastructural characteristics of hippocampal CA1 synapses, BV2 cells, and primary microglia were identified and analyzed.
MDA and SOD activity determination
MDA and SOD activity were measured using kits (Beyotime, China) according to the manufacturer’s instructions. The hippocampus was excised, weighed, and homogenized in MDA and SOD assay solutions. Plasma and homogenate supernatants were added to the reaction system. MDA activity was measured at 532 nm by absorbance and reported as µmol mg–1 or µM. SOD activity in the samples was measured by absorbance at 450 nm and expressed as U mg–1 or U mL–1 of total protein.
Determination of cytokine levels
Cytokine levels in mouse plasma and hippocampus were measured using the ABplex Mouse Cytokine 8-Plex Assay Kit (Abclonal). The analyzed cytokines included IL-2, IL-4, IL-12p70, IL-1β, IL-6, IFN-γ, TNF-α, and IL-17A. The samples were clarified by centrifugation and analyzed by FCM.
Scratch Assay
BV2 microglial cell migration was assessed using a scratch assay. BV2 cells were seeded into 12-well plates and cultured until 90% confluence. A linear scratch was made in the cell monolayer using a sterile 200 µL pipette tip. After scratching, wells were gently rinsed with PBS to remove floating cells and debris before fresh media was added. Cell migration into the scratch region was tracked for 24 h using the Opera Phenix Plus high-content imaging system (Revvity). Images were frequently captured to monitor cell migration. The migration rate was calculated as the proportion of the scratch area covered by migrating cells relative to the control group. ImageJ (NIH, USA) was used for image analysis to compare cell migration rates between experimental conditions.
Phagocytosis Assay
The phagocytic capability of primary microglial cells was assessed using the Cell Meter™ Fluorimetric Phagocytosis Assay Kit (Red Fluorescence, Cat#21225). I Primary microglial cells were allowed to adhere overnight in a 96-well confocal cell culture plate. After experimental treatments, cells were incubated with Protonex™ 600-labeled beads according to the manufacturer’s instructions. After incubation, non-internalized beads were washed away, and fluorescence from ingested beads was detected using a Zeiss LSM 880 confocal laser scanning microscope. Phagocytic activity was quantified as bead engulfment (%), calculated as the ratio of red fluorescent area (engulfed beads) to total cell area. ImageJ software (NIH, USA) was used to calculate this ratio and statistically compare microglial phagocytic capability between experimental groups.
PET-CT Imaging for Brain Metabolism
Brain metabolic activity was assessed via PET-CT imaging on mice 24 h after CLP. To minimize glucose fluctuations that could affect 18F-fluorodeoxyglucose (18F-FDG) uptake, mice were fasted for 4–6 h with access to water before imaging. After fasting, mice were intravenously administered 18F-FDG at a dose of 15.6 ± 1.7 MBq in 0.5 mL saline. Following injection, mice were housed in a warm, calm environment for 30–45 min to enhance tracer uptake and minimize stress and muscular activity that could affect glucose distribution.
Small-animal PET-CT scanners with high resolution and sensitivity were utilized under isoflurane anesthesia. Optimized scanning covered the entire brain, with a focus on the hippocampi. CT scans provided anatomical reference, followed by PET scans to assess glucose metabolism.
The SUVmean, which represents the average 18F-FDG uptake adjusted for body weight and injected dose, was used to measure brain metabolic activity, particularly in the hippocampal region. SUVmean data were statistically compared between experimental groups to evaluate post-CLP brain metabolism.
Laser Speckle Contrast Imaging (LSCI) for Cerebral Blood Flow
Cerebral blood flow dynamics in mice were assessed by LSCI 24 h post-CLP. After anesthesia, the scalp was gently removed to expose the skull. LSCI uses coherent laser light to illuminate cortical tissue. Red blood cells in cerebral vessels alter the speckle pattern captured by a high-resolution camera. These oscillations are used to map blood flow across the cortical surface in real time. Data were collected using a laser-synchronized high-sensitivity CCD camera. Relative blood flow changes were computed based on speckle contrast. To evaluate the impact of CLP on cerebral perfusion, blood flow was quantified and statistically compared between experimental groups.
TMT-based quantitative proteomics analysis
Sample preparation
Proteomic analysis was conducted on CLP and sham-operated mice (n = 3 per group). After anesthesia, intestinal tissues were promptly removed and snap-frozen in liquid nitrogen. Weighed protein samples were added to SDS L3-EDTA lysis buffer. The samples were homogenized (25,000 g, 4 ℃, 5 min). The homogenate was then treated with 10 mM DTT and 45 mM IAM and incubated in the dark for 45 min. Proteins were precipitated with cold acetone, followed by centrifugation (25,000g, 4 ℃, 15 min). After air-drying the pellet, SDS L3-free lysis solution was added to fully solubilize the proteins. Following another centrifugation (25,000 g, 4 ℃, 15 min), protein concentration was measured using the Bradford method. Proteins were desalted after trypsin digestion. The freeze-dried peptides were separated using a Shimadzu LC-20AB liquid chromatography system.
High-performance liquid chromatography and mass spectrometry
Peptides were separated using gradient elution on a self-packed C18 column on an Easy nLC 1200 system. The isolated peptides were analyzed using Data Dependent Acquisition (DDA) tandem mass spectrometry. TMT-proteomic analysis was performed by BGI-Shenzhen, China.
Bioinformatics analysis
The UniProtKB database (Release 2016_10) was used to obtain FASTA protein sequences of differentially expressed proteins for Gene Ontology (GO) mapping and annotation. KEGG Orthology (KO) identities and pathways were determined by aligning the FASTA sequences of significantly altered proteins with the Kyoto Encyclopedia of Genes and Genomes (KEGG) database (http://geneontology.org/). Protein expression data from relevant KEGG pathways were visualized using a hierarchical clustering heat map.
Statistical analysis
Data reported as mean ± standard deviation (Mean ± SD) were analyzed using GraphPad Prism 10.0 software. Data normality was assessed using the D’Agostino & Pearson test or Shapiro-Wilk test. One-way analysis of variance (ANOVA) and two way ANOVA with Tukey’s or Šídák’s multiple comparisons test was used for multiple-group comparisons. Unpaired Student’s t-test or Mann-Whitney test was used for group comparisons, and Log-rank (Mantel–Cox) test was performed for survival rate analysis. P values < 0.05 were considered statistically significant. All data analyses and statistical figures were generated using GraphPad Prism 10.0.