Abstract Developmental neurotoxicity (DNT) induced by sevoflurane exposure poses significant risks to pediatric anesthesia, yet effective protective strategies remain limited. Here, we developed self-assembling Angiopep-2/SIRT1 nanoparticles (Ang/SIRT1-NPs) with favorable biocompatibility and brain-targeting properties. Through in vitro and in vivo studies, we demonstrate that Ang/SIRT1-NPs effectively alleviate sevoflurane-induced neuronal apoptosis, neuroinflammation, and dendritic spine loss. Multi-omics analyses identified SIRT1-mediated suppression of necroptosis and oxidative stress pathways as key mechanisms underlying neuroprotection. Behavioral assays further confirmed improved cognitive and motor function in nanoparticle-treated mice. Our findings highlight the potential of Ang/SIRT1-NPs as a promising neuroprotective strategy for preventing anesthesia-related DNT and support their translational application in pediatric neuroprotection. Graphical abstract [30]graphic file with name 12951_2025_3639_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-025-03639-w. Keywords: Sevoflurane, Developmental neurotoxicity, Angiopep-2/SIRT1 nanoparticles, Necroptosis, Multi-omics Introduction Sevoflurane is a volatile anesthetic commonly used in clinical anesthesia due to its ability to induce a rapid and controllable anesthetic state [[31]1, [32]2]. However, increasing research indicates that sevoflurane may have adverse effects on the developing brain, particularly in infants and children [[33]3, [34]4]. The issue of sevoflurane-induced developmental neurotoxicity (DNT) has garnered significant attention because it can lead to long-term cognitive impairment and behavioral abnormalities [[35]5–[36]7]. The developing brain is extremely vulnerable, and external factors such as the use of anesthetics can profoundly affect neuronal growth and differentiation [[37]8]. Therefore, studying the mechanisms of sevoflurane-induced DNT is crucial for protecting children’s brain health [[38]5, [39]9, [40]10]. Current research on sevoflurane-induced DNT primarily focuses on its induction of neuronal apoptosis and inflammatory responses [[41]3, [42]11]. Necroptosis, a form of programmed cell death, plays a crucial role in sevoflurane-induced neurotoxicity [[43]12–[44]14]. Multi-omics approaches, such as high-throughput sequencing, proteomics, and metabolomics, have been widely employed to elucidate the molecular mechanisms underlying sevoflurane-induced DNT [[45]15, [46]16]. However, despite numerous studies revealing the direct impact of sevoflurane on neural cells, its precise molecular mechanisms remain incompletely understood [[47]5, [48]9, [49]17]. Particularly in clinical settings, effective interventions are still lacking [[50]5, [51]18, [52]19]. Therefore, exploring new therapeutic strategies and molecular targets is of paramount importance for mitigating sevoflurane-induced DNT. Sirtuin 1 (SIRT1) is a crucial deacetylase that plays a key role in neuroprotection and anti-apoptosis [[53]20]. Studies have shown that SIRT1 regulates cell survival and apoptosis by deacetylating specific target proteins [[54]21, [55]22]. SIRT1 is particularly recognized for its protective effects in inflammatory responses and necroptosis [[56]23–[57]25]. Its neuroprotective role has been widely acknowledged in various neurotoxic and neurodegenerative disease studies [[58]26, [59]27]. For instance, research on Alzheimer’s and Parkinson’s diseases indicates that SIRT1 can reduce neuronal damage and improve cognitive function [[60]28–[61]30]. Therefore, SIRT1 presents a promising therapeutic target with broad application potential [[62]28, [63]31, [64]32]. In this context, self-assembled Angiopep-2/SIRT1 nanoparticles have been introduced as a novel drug delivery system in the study of sevoflurane-induced DNT. Angiopep-2 is a peptide capable of crossing the blood-brain barrier (BBB), and when combined with SIRT1, it forms nanoparticles with excellent stability and biocompatibility [[65]33–[66]35]. The application of nanoparticle technology in drug delivery, particularly in crossing the BBB, shows immense potential [[67]36–[68]38]. Through targeted delivery using nanoparticles, drugs can more efficiently reach damaged brain regions and exert their therapeutic effects [[69]39]. Previous studies have demonstrated the significant efficacy of nanoparticles in treating neurological diseases such as stroke and brain tumors [[70]40–[71]42]. Therefore, Angiopep-2/SIRT1 nanoparticles exhibit innovative and important application value in the research of sevoflurane-induced DNT. This study aims to investigate the molecular mechanisms by which self-assembled Angiopep-2/SIRT1 nanoparticles regulate necroptosis in sevoflurane-induced DNT. Through both in vivo and in vitro experiments, we evaluate the stability, biocompatibility, and intervention effects of Angiopep-2/SIRT1 nanoparticles on sevoflurane-induced DNT. Multi-omics analysis is employed to uncover the key genes, proteins, and related metabolic pathways involved in the nanoparticles’ amelioration of DNT. The scientific significance of this study lies in elucidating the molecular mechanisms of sevoflurane-induced DNT, proposing a novel therapeutic strategy, and providing new molecular targets for clinical application. This research has the potential for significant clinical value, offering new therapeutic approaches to enhance the safety of pediatric anesthesia. Materials and methods Preparation of Angiopep-2/SIRT1 nanoparticles Angiopep-2/SIRT1 nanoparticles were prepared using the anti-solvent precipitation method. To prepare the SIRT1 solution, 10 mg of SIRT1 (ab287914, Abcam) was added to 10 ml of distilled water, and the mixture was stirred at room temperature for 1 h. The pH was adjusted to 7.0 using 0.1 M HCl or NaOH. The protein solution was then heated to 90 °C or higher for 30 min, resulting in a final concentration of 1 mg/ml SIRT1. Separately, 1 mg of Angiopep-2 and 1 mg of SMCC (22360, Thermo, USA) were each dissolved in 1 ml of water, creating Angiopep-2 (1 mg/ml) and SMCC (1 mg/ml) solutions. Add 1 mL of water to a 2 mL centrifuge tube, followed by the addition of 100 µL of Angiopep-2 solution. Stir vigorously. Next, add 100 µL of SMCC solution to the mixture and stir for 3 h at 1500 rpm in an ice bath to prepare the Angiopep-2-SMCC solution. Withdraw 100 µL of the Angiopep-2-SMCC solution and slowly add it to the SIRT1 solution. Stir vigorously for 2 h in an ice bath to obtain the Angiopep-2/SIRT1. Transmission electron microscopy (TEM) and scanning electron microscopy (SEM) were used to analyze the Angiopep-2/SIRT1 nanoparticles. The morphology of the Angiopep-2/SIRT1 nanoparticles was examined using a TEM (TECNAI G2 F20, FEI, USA) at an acceleration voltage of 30 kV. The nanoparticles were dried using a freeze dryer. For SEM analysis, the samples were sputter-coated for 45 s at 20 mA and observed with an SEM (Quanta 250, FEI, Hillsboro, OR, USA) to assess cross-sectional morphology and serum stability. For flow cytometry analysis of SIRT1, SH-SY5Y cells were treated with 0.25% trypsin (25200072, Gibco, USA) and counted. Approximately 1 × 10^6 cells were resuspended in 200 µL of fluorescence-activated cell sorting (FACS) buffer (660585, BD Biosciences, USA). Fluorescent antibodies (2 µL each) were added, and the cells were incubated on ice for 30 min. Subsequently, the SH-SY5Y cells were washed with FACS buffer, fixed with 10% formalin ([72]R04587, Merck, USA), and analyzed for antigen positivity using a BD FACSCalibur flow cytometer (BD Biosciences, USA). The antibody used was a fluorescently labeled SIRT1 antibody (PA5-85921, 1:200, Invitrogen, USA). Stability assessment of Angiopep-2/SIRT1 using dynamic light scattering (DLS) DLS was employed to measure the size, polydispersity index (PDI), and surface zeta potential of the Angiopep-2/SIRT1 nanoparticles. The measurements were conducted using a nanoparticle size and zeta potential analyzer (Nano-ZS90, Malvern Instruments, UK) equipped with a 633 nm He-Ne laser. All experiments were performed at 25 °C and repeated three times. To investigate the physiological stability of the nanoparticles, they were resuspended in water, saline, phosphate-buffered saline (PBS), and RPMI 1640 and incubated for 24 h. Additionally, Angiopep-2/SIRT1 was resuspended in solutions with different pH values (4.5, 5.5, 6.5, 7.5, and 8.5) and incubated for 24 h. After incubation, DLS was used to measure the size, distribution, and scattering intensity of the nanoparticles. SEM (S-4800, Hitachi, purchased from Shanghai Fulai Optical Technology Co., Ltd.) was employed to observe the morphology of the nanoparticles. In vitro biocompatibility assessment Live/Dead Cell Staining: SH-SY5Y cells at a density of 1 × 10^6 were seeded into 12-well culture plates and co-cultured with PBS, 0.5 µM SIRT1, and 0.5 µM Angiopep-2/SIRT1 samples for 1, 2, and 3 days. A live/dead cell staining solution (ZY140632, Zeye Biotechnology, China) was prepared in a ratio of 1 mL PBS: 3 µL calcein-AM: 5 µL PI, then added to each group and incubated at 30 °C for 37 min. The live (green) and dead (red) cells in each field of view were then counted using a fluorescence microscope (IMT-2, Olympus, Japan). CCK-8 Assay: SH-SY5Y cells at a density of 1 × 10^6 were co-cultured with CON, SIRT1, and Angiopep-2/SIRT1 samples for 1, 2, and 3 days. After the incubation period, 100 µL of CCK-8 solution (C0037, Beyotime) was added to each well and incubated for 2 h. Subsequently, 100 µL of the supernatant was transferred to a 96-well plate, and the absorbance was measured at 450 nm using a microplate reader (BioTek Synergy Neo2 Hybrid, Agilent, USA). Hemolysis assay To conduct the hemolysis assay of Angiopep-2/SIRT1 nanoparticles, prepare solutions with varying concentrations of Angiopep-2/SIRT1 nanoparticles, ranging from 100 µg/mL to 3.13 µg/mL. Use PBS as the negative control and deionized water as the positive control. Fresh human whole blood containing an anticoagulant (EDTA) (Human Whole Blood, IPHASE, Suzhou, China) is then mixed with the different concentrations of nanoparticle solutions and control liquids in specific proportions. The mixtures are incubated at 37 °C for 3 h. After incubation, the samples are centrifuged at 2000 g for 10 min to separate the undissolved cellular components (CC) from the plasma. The supernatant is collected, and the absorbance is measured at 540 nm using a spectrophotometer to quantify the hemoglobin content dissolved in the blood. Evaluation of Angiopep-2/SIRT1 crossing the BBB Cellular Assay: HBMEC cells were cultured in a Transwell system. Successful formation of the BBB was confirmed when the transendothelial electrical resistance reached 150 ohms×cm². Subsequently, 0.5 µM SIRT1 and 0.5 µM Angiopep-2/SIRT1 were added to the upper chamber of the Transwell. After 4 h, the fluorescence intensity of FITC (green) in HBMEC cells (upper chamber) and SH-SY5Y cells (lower chamber) incubated with Angiopep-2/SIRT1/FITC was detected using a confocal laser scanning microscope (CLSM) (FV3000, Olympus, Japan). The HBMEC cell membranes were labeled with Dil fluorescent dye (red), and the SH-SY5Y cells were stained with DAPI (blue). After a 4-hour incubation, the fluorescence intensity in the upper and lower chambers of each group was measured using a microplate reader. When red and green fluorescence were simultaneously present in the images, the red fluorescence was pseudocolored to purple. The penetration ratio of SIRT1 and Angiopep-2/SIRT1 across the BBB was determined by calculating the ratio of the fluorescence intensity in the lower chamber to the total fluorescence intensity in both chambers using the following formula: Penetration Rate (%) = Fluorescence Intensity in the Upper Chamber/(Fluorescence Intensity in the Upper Chamber + Fluorescence Intensity in the Lower Chamber) × 100%. Animal Experiment: Angiopep-2/SIRT1 nanoparticles were labeled with Cy5.5 NHS and injected into mice via the tail vein at a dose of 10 mg/kg in a 100 µL volume. The distribution of the labeled Angiopep-2/SIRT1 was observed at 6, 12, 24, and 48 h post-injection using the IVIS Lumina Series III small animal imaging system (PerkinElmer, USA). This experimental protocol and animal study were approved by the hospital’s Institutional Animal Care and Use Committee (IACUC). All animal experiments in this study were conducted in strict accordance with institutional ethical guidelines and approved by the Institutional Animal Care and Use Committee (IACUC). Mice were housed in a standard SPF-grade animal facility under controlled environmental conditions (temperature: 22–25 °C; humidity: 60–65%; 12-hour light/dark cycle) to ensure their basic physiological needs. All procedures were performed by professionally trained personnel using appropriate anesthesia and analgesia to minimize pain and stress. Animal health was regularly monitored, and humane endpoints were applied when necessary. Throughout the study, the principles of Replacement, Reduction, and Refinement (3Rs) were followed to ensure ethical use of animals while enhancing the scientific validity and reproducibility of the experimental data. Construction and grouping of sevoflurane-induced DNT mouse model SPF-grade C57BL/6J mice (8–12 weeks old, weighing 20–30 g) were obtained from Beijing Vital River Laboratory Animal Technology Co., Ltd., Beijing, China. All mice were housed in an SPF-grade animal laboratory with controlled conditions: a 12-hour light/dark cycle, 60–65% humidity, and a temperature range of 22 to 25 °C. The mice had free access to food and water. After a one-week acclimation period, during which their health status was monitored, the experiment commenced. To facilitate mating, 1 to 3 female mice (8–12 weeks old) were housed with one proven fertile male mouse. Vaginal plugs were checked daily between 08:00 and 10:00 as evidence of successful mating. The presence of a vaginal plug marked the first day of pregnancy. The pregnant female mice were then separated from the male and housed individually under standardized conditions until delivery. Silencing SIRT1: Lentiviral packaging kits (V48820, Invitrogen, USA) were used to transfect HEK-293T cells (CRL-3216, ATCC) with control plasmid sh-NC, sh-SIRT1-1, and sh-SIRT1-2, constructed using the pLKO.1 vector from Shanghai HanHeng Biotechnology Co., Ltd. After 48 h of transfection, the supernatant was collected and concentrated using a lentivirus concentrator (631231, Takara). The concentrated lentivirus was stored at −80 °C. The shRNA sequences are listed in Table S1. Establishment of sevoflurane-induced DNT mouse model: Newborn mice with SIRT1 gene silencing were subjected to lentivirus intervention via tail vein injection (100 µL, 1 × 10^11 PFU/mL) on days 1, 3, and 5. After the injections, on postnatal days 6, 8, and 10 (PNDs), mice in the sevoflurane group were placed in a sealed chamber. Using the Datex-Ohmeda anesthesia system (Madison, WI, USA), the mice were anesthetized with a mixture of 3% sevoflurane and 60% oxygen at a flow rate of 2 L/min for induction [[73]5], followed by maintenance at 1 L/min for 2 h. The control group was exposed to 60% oxygen without sevoflurane for 2 h. The concentrations of sevoflurane and oxygen were continuously monitored (Vamos, Dräger Medical, Germany), and the mice were kept on a heating pad at 37 ± 1 °C throughout the procedure. After sevoflurane exposure, the mice were immediately returned to their home cages and given standard care. Angiopep-2/SIRT1 nanoparticles were administered at a dose of 10 mg/kg via tail vein injection (100 µL each time) on PND 27 and PND 29. Detailed group information can be found in Table S2 [[74]43]. Hematoxylin and eosin (H&E) staining and TEM Mouse brain tissue fixed in paraformaldehyde was dehydrated, embedded in paraffin, and sectioned into 5 μm thick slices. Continuous sections were taken from the internal and external compartments at 200 μm intervals. Selected sections were deparaffinized in xylene and rehydrated through a series of ethanol gradients. For H&E staining, samples were stained in Harris alum hematoxylin solution (ST2067, Beyotime) for 5 min. They were then washed with 0.5% hydrochloric acid ethanol (C0165S, Beyotime) for 10 s, followed by staining with eosin solution (C0109, Beyotime) for 40 s. Finally, the sections were dehydrated, cleared, and mounted using neutral resin for observation under a light microscope. For TEM analysis, following H&E staining, the striatum was cut into 1 mm³ cubes and fixed in 2.5% glutaraldehyde (G5882, Sigma, USA). After primary fixation, the samples were post-fixed in 1% osmium tetroxide solution (1.60333, Sigma, USA) to enhance electron density and contrast. The samples were then dehydrated through a graded ethanol series, infiltrated, and embedded in acrylic resin. Ultrathin sections of 70 nm thickness were cut using an ultramicrotome and collected on grids for TEM analysis. Nissl staining Nissl staining was performed according to standard protocols. After deep anesthesia, mice were perfused transcardially, and their brains were extracted, fixed in formaldehyde, dehydrated, and embedded in paraffin. The brain tissues were then sectioned into 6 μm slices for staining. The sections were deparaffinized and stained with Nissl staining solution (C0117, Beyotime, China) at 60 °C for 45 min. Following staining, the sections were soaked in absolute ethanol for 2 min and then in xylene for 10 min before being coverslipped. Images were captured using the ScanScope scanning system (Olympus, Japan) and analyzed with Image-Pro Plus 6.0 software (Media Cybernetics, Silver Spring, USA). Golgi staining Golgi staining was performed using the FD Rapid Golgi Stain Kit (PK401, FD NeuroTechnologies, United States) according to the manufacturer’s instructions. Brain samples were collected and immersed in a mixture of Solutions A and B (1:1) at room temperature in the dark for 14 days. Subsequently, the samples were transferred to Solution C and kept in the dark for 5 days. The samples were then embedded in an optimal cutting temperature compound and frozen at −80 °C for 24 h. Using a cryostat (Leica, Wetzlar, Germany), the brains were sectioned coronally at 100 μm thickness. The staining procedure was carried out as per the manufacturer’s instructions. The slides were observed using an optical microscope equipped with a 63× oil immersion objective (Olympus, FV1200). Pyramidal neurons in the CA1 region that were well-impregnated and distinctly separated from other neurons were analyzed. From each pyramidal neuron, five basal dendritic segments, each 30 μm or longer, were randomly selected. Dendritic spine density was analyzed using ImageJ software. Short-term neurological assessment This study employed random group assignment, with mice randomly allocated to each experimental group to ensure baseline comparability between groups. In addition, all behavioral tests were conducted by well-trained observers who were blinded to the group assignments to prevent subjective bias. On days 31, 35, and 37, after the construction of the mouse model, 6 mice from each group were randomly selected for behavioral tests to evaluate short-term neurological function. The tests included negative geotaxis and front-limb suspension. Each mouse was subjected to three trials for each test, and the average of the three scores was recorded. For the negative geotaxis test, Each mouse was placed on an inclined board (30 cm long, 45° inclination) with its head facing downward. After holding the mouse in place for 5 s, it was released. The time taken for the mouse to turn its head upwards within 30 s was recorded. For the front-limb suspension Test, Each mouse’s forepaws were placed on a suspended wire, and the time it took for the mouse to fall from the wire within a 60-second period was recorded. Long-term neurological assessment After completing the treatments, 6 mice from each group were randomly selected to undergo two behavioral tests to evaluate long-term neurological function. Novel object recognition test (NORT): Before the test, mice were placed in an open field (60 × 40 × 40 cm) for 10 min per day for three consecutive days to acclimate them to the testing environment. At the beginning of the test, mice were placed in the open field for 10 min. After this acclimation period, two identical balls of the same color were introduced into the open field. The duration of interaction with the balls, including nose or mouth contact, was recorded for 20 min. Between trials, the apparatus and objects were cleaned with 50% ethanol to eliminate any olfactory cues. One hour after the initial test, a second test was conducted by replacing one of the familiar objects with a novel object of a different shape and color. The mice were then observed for another 20 min. The time spent investigating the novel object versus the familiar object was recorded. The discrimination ratio was calculated as follows: Discrimination Ratio = Time Spent on Novel Object/(Time Spent on Novel Object + Time Spent on Familiar Object). Morris water maze test (MWMT): The MWMT was conducted in a black cylindrical tank (120 cm in diameter, 60 cm deep). An animal tracking system was used to monitor the movements of the mice during the test. The water maze was divided into four equal quadrants, with a platform submerged just below the water surface in the center of the third quadrant. For five consecutive days prior to the test, all mice underwent training to locate the platform. During the training, mice from different quadrants were placed in the maze, and the time taken to find the platform was recorded using tracking software. On the sixth day, the platform was moved to a new location, and the mice were placed in the maze to swim freely for 60 s. The time taken for each mouse to find the new platform location was recorded. The SMART video tracking system consists of both hardware and analysis software components. It measures indices of memory behavior and automatically tracks and monitors the mice. Using color image processing algorithms, the system tracks the movement trajectories of the mice in real-time. It can analyze the paths and time spent in each of the four quadrants, the efficiency over six cycles, orientation angle, average swimming speed, time spent crossing the virtual platform, and time spent on the virtual platform. High-throughput RNA sequencing Brain tissues were collected from the DNT group (n = 3) and the Angiopep-2/SIRT1 group (n = 3) for RNA sequencing. The sequencing libraries were generated and sequenced by CapitalBio Technology (Beijing, China). A total of 5 µg of RNA was used for each sample. Briefly, ribosomal RNA (rRNA) was removed from the total RNA using the Ribo-Zero Magnetic Kit (MRZG12324, Epicentre, USA). Sequencing libraries were constructed using the NEBNext Ultra RNA Library Prep Kit for Illumina (E7760S, NEB, USA). RNA was then fragmented into approximately 300 bp (bp) fragments in NEBNext First Strand Synthesis Reaction Buffer (5x). First-strand cDNA was synthesized using reverse transcriptase and random primers. The second strand was synthesized in the presence of dUTP Mix (10x) to create strand-specific libraries. The cDNA fragments were end-repaired, a polyA tail was added, and sequencing adapters were ligated. After ligation of Illumina sequencing adapters, the second strand of cDNA was digested with USER Enzyme (M5508, NEB, USA) to construct a strand-specific library. The library DNA was amplified, purified, and enriched via PCR. The libraries were then validated using the Agilent 2100 Bioanalyzer and quantified with the KAPA Library Quantification Kit (kk3605, Merck, USA). Finally, paired-end sequencing was performed on the Illumina NextSeqCN500 sequencing platform. RNA sequencing data analysis The quality of the raw paired-end sequencing reads was assessed using FastQC software v0.11.8. Preprocessing of the raw data was conducted using Cutadapt software v1.18 to remove Illumina sequencing adapters and poly(A) tail sequences. Reads with more than 5% N content were removed using a Perl script. The FASTX Toolkit v0.0.13 was then used to extract reads with at least 70% of bases having a quality score above 20. Paired-end sequences were repaired using BBMap software. Finally, the filtered high-quality reads were aligned to the mouse reference genome using Hisat2 software v0.7.12. Differential Analysis: The raw counts matrix was normalized using the logCPM (Counts Per Million) method provided by the edgeR package. Multiple comparisons were adjusted using the Benjamini-Hochberg method (i.e., FDR correction). Genes with adjusted p-values (adjP) < 0.05 and |log2FC| >2 were considered differentially expressed. A heatmap of intersecting gene expression was generated using the heatmap package in R. Lasso regression modeling was conducted with the “glmnet” package in R. By setting the penalty coefficient λ, Lasso regression enables compression estimation and variable selection of metabolites, thereby identifying genes significantly associated with osteoarthritis (OA). In the Lasso regression model, the optimal value of λ was determined through cross-validation using the “cv.glmnet” function in R, ensuring the model’s best predictive ability. After analysis, a coefficient path plot was generated to illustrate the changes in gene variable coefficients across different λ values using the “plot.glmnet” function, and significant differential genes were extracted as key features. First, a Support Vector Machine (SVM) model was implemented using the “e1071” package in R. In the SVM model, the Radial Basis Function (RBF) was used as the kernel function. Model parameters, such as the cost parameter C and the kernel parameter γ, were optimized using the grid search method via the “tune.svm” function. Next, the Recursive Feature Elimination (RFE) strategy was applied to optimize the feature subset by recursively removing the least contributing features. In each iteration, the importance of each feature was assessed using the weight coefficients of the SVM model, and the least important features were removed until a predefined number of features was reached. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were conducted using the “clusterProfiler”, “org.Hs.eg.db”, “enrichplot”, “DOSE”, and “ggplot2” packages in R. Quantitative proteomics detection Brain tissues from DNT group (n = 3) and Angiopep-2/SIRT1 group (n = 3) mice were collected. Tumor tissues were extracted using RIPA buffer containing protease inhibitors (87786, Thermo, USA). During extraction, samples were sonicated for 30 s every 5 minutes, repeating this process three times to ensure complete cell lysis and protein release. The concentration of the extracted protein samples was determined using a BCA Protein Assay Kit (23225, Thermo Fisher, USA), ensuring each sample’s concentration fell within the acceptable range based on the standard curve. After adjusting the pH to 8.0, trypsin was added at a ratio of 1:50 (enzyme to protein) and incubated at 37 °C for 16 h for protein digestion. After enzymatic digestion, the samples were cleaned using ZipTip C18, loaded onto a high-performance liquid chromatography (HPLC) system, and connected to a mass spectrometer for mass spectrometry (MS)/MS analysis. Data processing, including protein identification and quantification, was performed using MaxQuant software. Desalted peptides were labeled with iTRAQ reagent containing 0.1% formic acid (FA) for MS analysis. Each sample was analyzed three times using the QSTAR Elite Hybrid MS (Applied Biosystems/MDS-SCIEX) and an online HPLC system (Shimadzu, Japan). For each analysis, 30 µl of peptide solution was injected and separated on a homemade nanopore C18 column with a micro-nano spray (75 μm ID × 15 cm, 5 μm particles) (New Objectives, Wubrun, MA). A 90-minute HPLC gradient was established using mobile phase A (0.1% FA/2% ACN) and mobile phase B (0.1% FA/100% ACN) with an effective flow rate of 0.2 µl/min. A constant flow rate of 30 µl/min was achieved using a splitter. The mass spectrometer was set to operate in positive ion mode for data acquisition. The mass range was set from 300 to 2000 m/z, and precursor ions with charge states of + 2 to + 4 were selected for fragmentation. For each MS/MS spectrum, the three most abundant peptide ions with a count threshold exceeding 5 were selected. The selected precursor ions were dynamically excluded for 30 s, with a mass tolerance of 30 mDa. Intelligent information-dependent acquisition was activated using automated collision energy and automatic MS/MS accumulation. The fragment intensity multiplier was set to 20, with a maximum accumulation time of 2 s. Three LC-MS/MS injections (technical replicates = 3) were performed to ensure better coverage of the target proteome and to achieve better statistical consistency. Proteomics data validation was conducted on the same samples using Western Blot (WB) analysis. The MS parameters were: scan range (m/z) = 350–1500, resolution = 120,000, AGC target = 4e^5, and maximum injection time = 50 ms. The HCD-MS/MS parameters were: resolution = 30,000, AGC target = 1e^5, and collision energy = 33. For data-independent acquisition (DIA), each window overlapped by 1 m/z, with a total of 47 windows. The iRT kit (Ki3002, Biognosys AG, Switzerland) was added to calibrate the retention time of the extracted peptide peaks. The DIA dataset was generated using Spectronaut V13 (Biognosys, Switzerland), including data normalization and relative protein quantification. Welch’s ANOVA test was used for statistical analysis, and multiple comparisons were corrected using the Benjamini-Hochberg method to control the false discovery rate. Differentially expressed proteins were filtered based on an adjusted p-value (adjP) < 0.05 and |log2FC| >1. Metabolomics analysis Brain tissues from the DNT group (n = 6) and the Angiopep-2/SIRT1 group (n = 6) were collected. Each sample was transferred to a 1.5 mL polypropylene tube containing 300 µL of tissue, mixed with 900 µL of 80% methanol and 0.1% FA, and vortexed for 2 min. The mixture was then centrifuged at 12,000 g for 10 min. The supernatant was transferred to autosampler vials. Plasma metabolomics analysis was performed using an LC20 ultra-high-performance liquid chromatography (UHPLC) system (Shimadzu, Japan) coupled with a Triple TOF-6600 mass spectrometer (AB Sciex). Chromatographic separation was achieved using a Waters ACQUITY UPLC HSS T3 C18 column (100 × 2.1 mm, 1.8 μm). The column temperature was maintained at 40 °C, and the flow rate was set at 0.4 mL/min. The mobile phase consisted of acetonitrile with 0.1% FA in water. The gradient elution program for mobile phase B was as follows: 5% from 0.0 to 11.0 min, 90% from 11.0 to 12.0 min, and 5% from 12.1 to 14.0 min. The eluent was directly introduced into the mass spectrometer without splitting. The mass spectrometry conditions were as follows: ionization voltage, 5500 V; capillary temperature, 550 °C; nebulizer gas flow rate, 50 psi; and auxiliary heating gas flow rate, 60 psi. Orthogonal partial least squares-discriminant analysis (OPLS-DA) and 100-permutation tests were performed on the preprocessed data to prevent overfitting. In the OPLS-DA model, variables with a variable importance in projection (VIP) value > 1 were considered significant. Subsequently, univariate statistical analysis was conducted using Welch’s t-test, which is appropriate for samples with unequal variances. Multiple comparisons were corrected using the Benjamini-Hochberg (BH) method to control the false discovery rate (FDR). Metabolites were considered differentially expressed (DMs) if they met the criteria of adjusted p-value (adjP) < 0.05 and |log₂FC| >1. Metabolites that satisfied both VIP > 1 and the statistical significance criteria were defined as final differential metabolites. MetaboAnalyst (Version 5.0) was used to identify the associated metabolic pathways. Cell transfection and grouping Human neuroblastoma SH-SY5Y cells were obtained from the American Type Culture Collection (SCSP-5014, Cell Bank of the Chinese Academy of Sciences) and routinely cultured in Dulbecco’s Modified Eagle Medium/Ham’s F-12 (31765035, Gibco, USA) supplemented with 10% fetal bovine serum (FBS) (12483020, Gibco, USA) at 37 °C in a humidified atmosphere with 5% CO[2]. Appropriate cell densities were used for various assays according to our experimental requirements. Silencing SIRT1: Lentivirus packaging was performed using a packaging kit (V48820, Invitrogen, USA). Control plasmid sh-NC, sh-SIRT1-1, and sh-SIRT1-2 (constructed in pLKO.1 vector) with shRNA sequences listed in Table S3 were transfected into HEK-293T cells (CRL-3216, ATCC). After 48 h of transfection, the supernatant was collected and concentrated using a lentivirus concentration solution (631231, Takara). The concentrated lentivirus was stored at −80 °C. For lentivirus-mediated cell transfection, 5 × 10⁵ SH-SY5Y cells were seeded into 6-well plates. When cell confluence reached 70–90%, the medium containing lentivirus (MOI = 10, working titer approximately 5 × 10⁶ TU/mL) and 5 µg/mL polybrene (TR-1003, Sigma-Aldrich, UK) were added. After 4 h, the medium was diluted with an equal volume of fresh medium to reduce polybrene concentration. After 24 h, the medium was replaced with a fresh culture medium. After 48 h, transfection efficiency was observed using a luciferase reporter gene. Stable cell lines were selected using an appropriate concentration of puromycin (E607054, Sangon Biotech, Shanghai, China). Once the cells no longer died in the puromycin-containing medium, they were collected, and knockdown efficiency was confirmed by RT-qPCR. Establishment of the sevoflurane-induced DNT cell model: Cells in the exponential growth phase, between passages 35 and 60, were exposed to different concentrations of sevoflurane for 24 h. Freshly prepared sevoflurane stock solution (4 g/L in distilled water) was used to create exposure concentrations of 20, 40, and 60 mg/L (equivalent to 0.476, 0.952, and 1.429 mM) [[75]44]. Based on the results of the CCK-8 assay, 60 mg/L was selected for modeling. On days 1, 9, and 11, general cytotoxicity, neuronal precursor cell migration, and neurite outgrowth were measured in 96-well plates. Grouping details are provided in Table S4. General cytotoxicity: Cells were thawed and grown to 90% confluence in T175 flasks (Greiner, Hamburg, Germany). After trypsinization, the cells were resuspended in DMEM/F12 medium without RA and seeded at 10,000 cells per well in 200 µL medium in uncoated 96-well plates (Corning Costar, Kaiserslautern, Germany). After 1 h of adhesion, cells were exposed to Angiopep-2/SIRT1. Following a 24-hour incubation, the cells underwent the Resazurin reduction assay (V23110, Invitrogen, USA) and were then fixed in a 4% paraformaldehyde phosphate-buffered solution (PFA) for 15 min. Cell analysis and imaging were performed using a Zeiss Axiovert 200 inverted microscope equipped with a Colibri LED light source, Zeiss Axiocam50 monochrome digital camera, and Zeiss ZENlite 2.6 blue edition software. Cell migration assay: Cells were seeded at a density of 4 × 10⁶ to 5 × 10⁶ cells per dish in 96 mm bacteriological-grade culture dishes (Greiner, Hamburg, Germany). Within 24 h, cells formed free-floating spheroid aggregates with diameters ranging from 300 to 800 μm. On day 1, 10 mL of medium was added to each dish. Over the following days, 10 µM retinoic acid was added, and the medium was changed every 2–3 days. Cell suspensions were transferred to centrifuge tubes and centrifuged at 200 × g for 7 min. After 9 days of culture, aggregates were gently resuspended using a Pasteur pipette and seeded at 60,000 cells per well in black 96-well plates with flat, clear bottoms (Nunc). The plates were coated with poly-D-lysine (A3890401, Invitrogen, USA) and laminin ([76]A29249, Invitrogen, USA) (each at 10 µg/mL) and equipped with silicone stoppers from the Oris Cell Migration Assay (AMS Biotechnology, Abingdon, UK). Allow the cells to adhere overnight. On the next day, remove the stoppers, leaving a 2 mm diameter circular hole in the monolayer of cells. Incubate the cells with seven different concentrations of copper sulfide nanoparticles in DMEM/F12/RA medium for 44 h, with six technical replicates for each concentration. Perform the Resazurin reduction assay for 2 h. Wash the cells once with PBS, fix them with PFA for 15 min, wash twice with PBS-T, stain with DAPI (0.5 µg/mL) for 5 min, and wash twice with PBS. Attach a black plastic mask with 96 holes to the bottom of the plate, leaving a 2 mm diameter central circular area for each well. This setup allows observation of cells that migrated into the free area left by the silicone stopper during seeding (Fig. [77]1B). Quantify cell migration by taking two photographs per well and measuring the distance from the migration front to the black edge of the mask using ImageJ. To compensate for occasional asymmetry due to slight inaccuracies in the placement of silicone stoppers, average measurements from four quadrants per well. Use 100 nM cytochalasin D (PHZ1063, Invitrogen, USA) in six wells per plate as a reference for no migration, which completely inhibits cell movement over 44 h without affecting cell viability. Subtract the average zero-migration distance from the measured migration distances before evaluation. As an endpoint-specific control, add the Rho kinase inhibitor Y-27,632 (HY-10071, MedChemExpress, USA) at 50 µM to cells in six wells, which increases precursor cell migration rates to over 125% without affecting overall cell survival. Discard cell cultures that do not meet these standards. Fig. 1. [78]Fig. 1 [79]Open in a new tab Preparation and Characterization of Angiopep-2/SIRT1 Nanoparticles. A Schematic diagram of the preparation process of Angiopep-2/SIRT1 nanoparticles; (B) Transmission electron microscopy (TEM) image of Angiopep-2/SIRT1 nanoparticles, scale bar: 50 nm; (C) Particle size distribution of Angiopep-2/SIRT1 nanoparticles; (D) Zeta potential of Angiopep-2/SIRT1 nanoparticles; (E) Scanning electron microscopy (SEM) image of Angiopep-2/SIRT1 nanoparticles, scale bar:200 nm; (F) Serum stability of Angiopep-2/SIRT1 nanoparticles; (G) Flow cytometry analysis of protein expression in SH-SY5Y cells.All experiments were performed in triplicate, and data are presented as mean ± standard deviation Ca²^+ concentration detection To detect intracellular free Ca²⁺ levels, use Fluo-3 AM (S1056, Beyotime, China). Briefly incubate the cells with Fluo-3 AM at 37 °C for 1 h. Then, wash the cells three times with PBS. Measure the absorbance of all wells using a microplate reader (Tecan Infinite F50, Switzerland) with excitation at 488 nm and emission at 530 nm. Convert the fluorescence values to Ca²⁺ concentrations using a standard curve. Propidium iodide (PI) and Acridine orange (AO) staining Cell experiments: Seed the cells in 6-well plates prior to the designated treatments. Incubate the cells with 2 µg/mL PI (#P4170, Sigma Aldrich) for 10 min or with 200 µM AO (#A6014, Sigma Aldrich) for 15 min. Wash all PI-stained wells three times with PBS, and wash all AO-stained wells three times with PBS. After fixation with 4% PFA for 20 min, directly capture images using an Olympus IX73 microscope. Stain primary neurons with anti-TUJ1 antibody (PA5-80198, 5 µg/mL, Invitrogen). Animal experiment: For each group, 5 mice were intraperitoneally injected with PI 3 h before euthanasia. Thirty minutes after the last administration, the mice were rapidly perfused through the heart with physiological saline followed by 4% paraformaldehyde. The brains were extracted, fixed, dehydrated in a gradient series, embedded in an OCT compound, and sectioned (protected from light). The sections were incubated overnight with an anti-NEUN antibody (PA5-78499, 1:100, Invitrogen). The next day, the sections were washed three times with PBS, each wash lasting 5 min. The sections were then incubated in the dark at room temperature for 2 h with goat anti-rabbit IgG (H + L) secondary antibody (1:500 dilution). After incubation with the secondary antibody, the sections were washed again three times with PBS, each wash lasting 5 min. The specimens were then stained with DAPI and imaged using a fluorescence microscope. The images were analyzed using Image LabTM software. TUNEL staining The TUNEL Kit (C1086, Beyotime, China) was used to detect apoptosis in mouse brain tissue. Sections were treated with 3% H₂O₂ and then incubated with 50 µL of TUNEL reaction mixture for 60 min at 37 °C, avoiding light exposure. The cell nuclei were stained with DAPI for 30 min, followed by three PBS washes. Images of the tumor tissues were captured at 400× magnification using a fluorescence microscope equipped with a camera (BX53, Olympus, Japan). The TUNEL fluorescence area and DAPI fluorescence area were calculated using ImageJ software. Each group consisted of 5 mice, and for each mouse, 5 sections were analyzed. Randomly selected fields of view (6–10 per section) were observed, and the number of positive cells was recorded. The total number of cells and the number of apoptotic cells were counted to calculate the apoptosis rate. Enzyme-linked immunosorbent assay (ELISA) Collected mouse serum and used a total SOD activity assay kit (S0101S, Beyotime, China) to measure SOD levels, and a lipid peroxidation (MDA) assay kit (S0131S, Beyotime, China) to measure MDA levels. First, the antigen was diluted with coating buffer to the appropriate concentration. The wells of the microplate were blocked with 5% calf serum (F8318, MSK, Wuhan, China) at 37 °C for 40 min. Next, the diluted samples were added to the wells, followed by the addition of the enzyme-labeled antibody, and then the substrate solution. Finally, 50 µL of stop solution was added to each well to terminate the reaction, and the results were measured within 20 min. The absorbance was read at 450 nm using a microplate reader (Bio-Rad, USA), the standard curve was plotted, and the data were analyzed. Immunofluorescence staining for protein expression detection SH-SY5Y cells or mouse brain tissues were fixed with 4% paraformaldehyde at room temperature for 15 min, followed by two washes with PBS and permeabilization with 0.5% Triton X-100 (P0096, Beyotime Biotechnology, China) for 10 min. The cells and tissues were then incubated overnight at 4 °C with the following primary antibodies: rabbit anti-β-III-tubulin (PA5-80198, 5 µg/mL), rabbit anti-PSD95 (MA1-046, 1:500), p-RIPK1 (PA5-104645, 1:100), p-RIPK3-Human (93654, 1:800), p-RIPK3-Mouse (91702, 1:100), p-MLKL (#AF3903, 1:100), SIRT1 (MA5-27217, 1:100), Synaptophysin (MA5-14532, 1:250), MAP2 (PA1-10005, 1:5000), GFP (A-11122, 1:200), and nNOS (PA1-033, 1:200). After washing three times with PBS, the samples were stained with FITC-labeled phalloidin (F432, Invitrogen) or rhodamine-labeled phalloidin (R415, Invitrogen) to label the cytoskeleton. Subsequently, they were incubated with goat anti-rabbit Alexa Fluor 647 (ab150083, 1:200) or goat anti-rabbit Alexa Fluor 488 (ab150077, 1:200) secondary antibodies for 1 h. Afterward, the samples were washed three times with PBS and stained with DAPI (D3571, 10 µg/mL) at room temperature for 10 min. The sections were stored at 4 °C and then observed under a fluorescence microscope (IMT-2, Olympus, Japan). The fluorescence area of the relevant proteins relative to the nucleus fluorescence area was calculated using ImageJ software. For cell experiments, five sections per sample were randomly observed in 6–10 fields of view, with each experiment repeated three times per group. Except for the p-RIPK3-Human and p-RIPK3-Mouse antibodies, which were purchased from Cell Signaling, and Alexa Fluor 647 and Alexa Fluor 488 from Abcam, the p-MLKL antibody from Affinity, all other antibodies were purchased from Invitrogen. WB Cells and brain tissues were lysed in RIPA lysis buffer (P0013B, Beyotime Biotechnology, China). Protein concentrations were quantified using a BCA assay kit ([80]A53226, Thermo Fisher Scientific, USA). After separation by polyacrylamide gel electrophoresis, proteins were transferred onto PVDF membranes (PVH85R, Millipore, Germany) using the wet transfer method. The membranes were blocked at room temperature for 1 h with 10% BSA (37520, Thermo, USA) and then incubated overnight at 4 °C with the following primary antibodies: p-RIPK1 (PA5-104645, 1:1000), RIPK1 (PA5-20811, 1 µg/mL), p-RIPK3-Human (93654, 1:8000), p-RIPK3-Mouse (91702, 1:1000), RIPK3 (PA5-141114, 1:1000), p-MLKL (PA5-105678, 1:1000), MLKL (PA5-115578, 1:1000), SIRT1 (MA5-27217, 1:1000), and GAPDH (MA5-15738, 1:1000). After washing, the membranes were incubated with HRP-conjugated secondary IgG antibody (AN-170-250UG, 1:5000) for 2 h. The membranes were then washed three times with TBST for 5 min each. Visualization, imaging, and analysis of all protein bands were performed using the Syngene G: BOX F3 imaging system (Antpedia, China) and ImageJ software. The relative protein levels were expressed as the grayscale value of the target protein bands relative to the grayscale value of the GAPDH protein bands. Each cell experiment was repeated three times, and the mouse experiments included six mice per group. All antibodies mentioned were purchased from Invitrogen. RT-qPCR Total RNA was extracted from cells and tumor tissues using TRIzol (15596026, ThermoFisher, USA). The concentration and purity of the extracted RNA were measured using a Nanodrop 2000 spectrophotometer (ThermoFisher, USA). RNA was reverse-transcribed into cDNA according to the PrimeScript RT reagent Kit instructions (RR047A, Takara, Japan). The synthesized cDNA was subjected to RT-PCR using the Fast SYBR Green PCR kit (11736059, ThermoFisher, USA), with each reaction set up in triplicate. GAPDH was used as the internal control, and relative expression levels were calculated using the 2^−ΔΔCt method. The experiment was repeated three times. The primer sequences used for RT-qPCR are listed in Table S5. Statistical analysis Statistical analyses in this study were performed using SPSS software (version 21.0, IBM, USA). Data are expressed as mean ± standard deviation. Normality and homogeneity of variances were first tested. For data that were normally distributed and had homogeneous variances, an unpaired t-test was used for comparisons between two groups, while one-way ANOVA or repeated measures ANOVA was used for comparisons among multiple groups. A p-value of less than 0.05 was considered statistically significant. Results Preparation and characterization of Angiopep-2/SIRT1 nanoparticles Sevoflurane is a widely used inhalation anesthetic in pediatric anesthesia due to its rapid onset and favorable tolerance. It is often employed in short-duration surgeries for children [[81]5]. However, an increasing number of studies indicate that sevoflurane may have long-term adverse effects on brain development in infants, leading to DNT [[82]45]. This neurotoxicity is primarily manifested as cognitive dysfunction and behavioral abnormalities. Epidemiological data show that infants frequently or prolongedly exposed to sevoflurane have a significantly increased risk of these issues [[83]46]. SIRT1, through its antioxidant and anti-apoptotic functions, can significantly mitigate neurocyte damage induced by sevoflurane, offering a novel therapeutic strategy for DNT [[84]47]. However, effectively delivering SIRT1 to specific neural cells remains a challenge. Here, the self-assembling peptide nanoparticle technology provides a promising solution. By encapsulating SIRT1 activators or SIRT1 itself, these nanoparticles can more effectively penetrate the BBB and directly target brain cells. Angiopep-2, a peptide that can specifically recognize and penetrate the BBB, is used to modify SIRT1 nanoparticles, further enhancing the neuro-specificity and therapeutic efficiency of the treatment [[85]48]. In this context, self-assembling peptide nanoparticles serve as a unique drug delivery system platform, enhancing the stability and targeting of therapeutics [[86]49]. Specifically, Angiopep-2/SIRT1 nanoparticles combine the brain-targeting capability of Angiopep-2 with the antioxidant and anti-apoptotic properties of SIRT1. This study demonstrates that these nanoparticles can effectively cross the BBB and directly target the damaged nervous system. By activating SIRT1, they effectively inhibit sevoflurane-induced necroptosis and other cellular damage pathways, significantly improving the symptoms of DNT. The Angiopep-2/SIRT1 nanoparticles were prepared according to the protocol outlined in Fig. [87]1A. TEM results show that the Angiopep-2/SIRT1 nanoparticles have a diameter of 144.86 ± 2.17 nm and a zeta potential of −19.06 ± 9.08 mV (Fig. [88]1B-D). SEM analysis reveals that the Angiopep-2/SIRT1 nanoparticles are spherical with a uniform size distribution (Fig. [89]1E). In the serum stability test, both SIRT1 and Angiopep-2/SIRT1 nanoparticles remained stable for 7 days in PBS containing 10% FBS (Fig. [90]1F). Flow cytometry analysis indicated that SH-SY5Y cells had internalized the Angiopep-2/SIRT1 nanoparticles, as evidenced by the presence of SIRT1 protein within the cells (Fig. [91]1G). Following the successful preparation of Angiopep-2/SIRT1 nanoparticles, we further assessed their stability under various physiological conditions (water, PBS, saline, and RPMI 1640) using DLS. The results indicated slight variations in the size of the Angiopep-2/SIRT1 nanoparticles, but overall, they exhibited good stability in all tested environments (Fig. S1A). Additionally, in different pH buffer solutions, the Angiopep-2/SIRT1 nanoparticles demonstrated a decrease in particle size with increasing pH, particularly showing brain pH responsiveness at pH 7.4 (Fig. S1B). We evaluated the impact of Angiopep-2/SIRT1 nanoparticles on SH-SY5Y cell viability and proliferation using LIVE/DEAD staining and the CCK-8 assay. The CCK-8 assay results showed an increase in cell number over time with no significant differences between groups (Fig. S1C). LIVE/DEAD staining indicated that nearly all cells remained viable over a 3-day culture period, with no differences observed between groups (Fig. S1D). Furthermore, the hemolysis assay results revealed negligible hemolysis (< 5%) at all concentrations of Angiopep-2/SIRT1 nanoparticles (Fig. S1E). These findings demonstrate that Angiopep-2/SIRT1 nanoparticles possess good biocompatibility and biosafety. SIRT1 and Angiopep-2/SIRT1 were respectively added to the upper chamber of a Transwell system, and their uptake by cells in both the upper and lower chambers was observed using CLSM. After a 4-hour incubation, the fluorescence intensity of FITC was measured using an ELISA reader. In SH-SY5Y cells treated with Angiopep-2/SIRT1, the average FITC fluorescence intensity was significantly higher compared to cells treated with SIRT1 alone (Fig. S2A). To evaluate the targeting efficacy, Angiopep-2/SIRT1 nanoparticles conjugated with Cyanine 5.5 (Cy5.5) were intravenously injected into mice. The Angiopep-2/SIRT1 nanoparticles group exhibited stronger Cy5.5 fluorescence in the brain at 6, 12, 24, and 48 h post-injection, indicating enhanced targeting, permeability, and retention effects of Angiopep-2/SIRT1 (Fig. S2B). This study demonstrates that Angiopep-2/SIRT1 nanoparticles, due to their excellent stability and biocompatibility, successfully achieved efficient delivery of SIRT1 protein in both in vitro and in vivo models. Notably, in the brain environment, these nanoparticles exhibited significant targeting and enhanced cellular uptake, providing a potential strategy for the future treatment of related neuropathological conditions. Multi-omics analysis reveals potential pathways through which Angiopep-2/SIRT1 nanoparticles alleviate sevoflurane-induced DNT In this study, we explored the molecular mechanisms underlying sevoflurane-induced DNT, focusing on the regulatory role of Angiopep-2/SIRT1 nanoparticles. Through a systematic evaluation of cellular morphology and functional damage in the DNT model, we analyzed how these nanoparticles affect necroptosis-related pathways, providing potential therapeutic targets [[92]13, [93]50]. First, we established a DNT model by inducing sevoflurane exposure in neonatal mice. H&E staining and TEM results showed that, compared to the control group, the DNT group exhibited nuclear condensation, vacuolization, and an increase in striatal neurons (Fig. S3A). Nissl staining results indicated a significant reduction in Nissl-stained cells in the DNT group, suggesting neuronal loss in the striatum compared to the control group (Fig. S3B). Golgi staining was used to examine dendritic spine density, revealing a significant decrease in the number of spines on pyramidal neurons in the DNT group compared to the control group (Fig. S3C). Immunofluorescence results showed that β-III-tubulin and PSD95 were significantly reduced in the DNT group compared to the control group (Fig. S3D-E). After establishing the DNT model, we conducted negative geotaxis and front-limb suspension tests on days 31, 35, and 37. The results of the negative geotaxis test (Fig. A) showed that the time required for the DNT group mice to turn their heads was significantly longer compared to the control group (Fig. S4B). In the front-limb suspension test (Fig. S4C), the DNT group mice spent significantly less time hanging on the wire compared to the control group (Fig. S4D). To further assess the long-term neurofunctional damage induced by sevoflurane in DNT mice, we performed NORT and MWMT. The NORT results (Fig. S4E) indicated a significantly lower discrimination index in the DNT group compared to the control group, suggesting a marked decline in their ability to differentiate between new and familiar objects, which implies impairment in cognitive functions, particularly memory and learning abilities (Fig. S4F). The MWMT results showed that the DNT group mice spent significantly more time finding the platform and less time in the target quadrant compared to the control group, indicating more severe cognitive impairment in the DNT group (Fig. S4G). However, there was no difference in swimming speed between the two groups (Fig. S4H), suggesting that the longer escape latency was primarily due to impaired spatial memory rather than slower swimming speed (Fig. S4I-J). After 5 days of training, the platform was removed from the water maze, and the mice’s movement trajectories were recorded for 1 min. The target quadrant, where the platform was previously located, was considered the escape zone. The control group spent approximately 50% of their time in the target quadrant, while the DNT group spent similar amounts of time in all four quadrants, indicating significant declines in cognitive function and memory in the DNT group (Fig. S4K-L). These results confirm the successful establishment of the sevoflurane-induced DNT model in neonatal mice, demonstrating both short-term and long-term neurofunctional deficits. The processing and differential analysis of high-throughput sequencing data revealed that, using a threshold of |log2FC|>2 and adjP < 0.05, compared to the DNT group, the Angiopep-2/SIRT1 group had 441 significantly upregulated and 593 significantly downregulated genes in the brain tissue (Fig. [94]2A-B). Subsequently, two machine learning algorithms were used to screen the differential genes. A Lasso regression model and the SVM-RFE algorithm were employed to identify feature genes. The Lasso regression algorithm identified nine key genes: Ptx3, Gadd45a, Ripk1, Lims2, Txnip, Polr3gl, Nedd9, Rpa2, and Cers1 (Fig. [95]2C). The SVM-RFE algorithm identified ten key genes: Igf1, Aven, Ift70a1, Dtwd2, Ripk1, Psmd6, Tg, Gm32856, Nfyc, and Mfsd12 (Fig. [96]2D). By intersecting the genes identified by both algorithms, the common feature gene Ripk1 (RIPK1) was obtained, as illustrated in the Venn diagram (Fig. [97]2E). Following the identification of differential genes, enrichment analysis was conducted. GO analysis results indicated significant enrichment of differential genes in various categories. In the biological process (BP) category, the genes were primarily enriched in programmed necrotic cell death, necroptotic process, regulation of programmed necrotic cell death, and necrotic cell death. In the CC category, enrichment was observed in receptor complex, membrane microdomain, membrane raft, and collagen-containing extracellular matrix. In the molecular function (MF) category, the genes were mainly enriched in tumor necrosis factor receptor superfamily binding, ATP hydrolysis activity, endodeoxyribonuclease activity producing 5’-phosphomonoesters, and lyase activity (Fig. [98]2F). KEGG pathway analysis showed that the differential genes were predominantly enriched in the TNF signaling pathway, NF-kappa B signaling pathway, apoptosis, and IL-17 signaling pathway (Fig. [99]2G). Using machine learning algorithms, RIPK1 was identified as a key gene involved in necroptosis. The results of GO and KEGG, pathway enrichment analyses, indicate that the differential genes are closely related to necroptosis. Fig. 2. [100]Fig. 2 [101]Open in a new tab Identification of Potential Pathways for Angiopep-2/SIRT1 Nanoparticles in Alleviating Sevoflurane-Induced DNT Using Multi-Omics Data. A Volcano plot of differentially expressed genes between DNT group (n = 3) and Angiopep-2/SIRT1 group (n = 3) based on high-throughput sequencing data. Green dots indicate significantly upregulated genes, blue dots indicate significantly downregulated genes. Screening criteria: |log₂FC| >2 and adjP < 0.05; (B) Heatmap of differentially expressed genes between DNT and Angiopep-2/SIRT1 groups; green indicates high expression and blue indicates low expression; (C) LASSO algorithm identified 9 feature genes; (D) SVM-RFE algorithm identified 10 feature genes; (E) Venn diagram showing the overlap of the two machine learning results, yielding 1 shared gene; (F) Bubble plot of GO enrichment analysis of differentially expressed genes; (G) Bubble plot of KEGG pathway enrichment analysis of differentially expressed genes; (H) Venn diagram of proteomics data showing 1 overlapping protein identified by both machine learning algorithms; (I) KEGG pathway analysis of 21 differential metabolites (VIP > 1, adjP < 0.05, |log₂FC| >1) To further investigate the pathways through which Angiopep-2/SIRT1 nanoparticles mitigate sevoflurane-induced DNT, we collected brain tissues from three mice in both the DNT and Angiopep-2/SIRT1 groups. Principal component analysis (PCA) results and the loading plot showed a clear separation between the DNT group and the Angiopep-2/SIRT1 group (Fig. S5A-C). The OPLS-DA results also demonstrated a clear separation between the DNT and Angiopep-2/SIRT1 groups in the OPLS-DA score plot (Fig. S5D-F). Differential analysis identified, using a threshold of |log2FC|>1 and adjP < 0.05, a total of 278 differential proteins, with 24 upregulated and 20 downregulated proteins (Fig. S6A-B). Subsequently, two machine learning algorithms were used to screen the differential proteins. The Lasso regression model identified two key proteins: Ripk1 and Aarsd1 (Fig. S6C). The SVM-RFE algorithm also identified two key proteins: Mospd1 and Ripk1 (Fig. S6D). The intersection of the proteins identified by the two algorithms resulted in the key protein, RIPK1, as illustrated in the Venn diagram (Fig. [102]2H). Next, we performed an enrichment analysis on the differential proteins. According to GO analysis, the differential proteins in the BP category were mainly enriched in the regulation of necrotic cell death, regulation of the necroptotic process, regulation of programmed necrotic cell death, and necrotic cell death. In the CC category, they were primarily enriched in NuRD complex, CHD-type complex, histone deacetylase complex, SWI/SNF superfamily-type complex, and U5 snRNP. In the MF category, they were mainly enriched in aminoacyl-tRNA editing activity, death receptor binding, transcription corepressor activity, and aminoacyl-tRNA ligase activity (Fig. S6E). KEGG pathway analysis showed that the differential proteins were predominantly enriched in necroptosis, NOD-like receptor signaling pathway, TNF signaling pathway, and cytosolic DNA-sensing pathway (Fig. S6F). These experimental results indicate that the differential proteins are mainly enriched in the necroptosis pathway, consistent with the high-throughput sequencing results. We collected brain tissues from six mice in both the DNT group and the Angiopep-2/SIRT1 group. PCA results and the loading plot showed a clear separation between the DNT and Angiopep-2/SIRT1 groups (Fig. S7A-C). The OPLS-DA results also demonstrated a clear separation between the DNT and Angiopep-2/SIRT1 groups, with a permutation analysis showing R²Y = 0.944 > 0.8, indicating that the model is stable (Fig. S7D-F). Using thresholds of VIP > 2, |log₂FC| >1, and adjusted p-value (adjP) < 0.05, a total of 21 DMs were identified. Subsequent KEGG pathway analysis revealed that these metabolites were primarily enriched in the glyoxylate and dicarboxylate metabolism, alanine, aspartate and glutamate metabolism, nitrogen metabolism, and pyruvate metabolism pathways (Fig. [103]2I, Table S6). Glyoxylate and dicarboxylate metabolism involve key energy production pathways, such as the tricarboxylic acid cycle, and their dysfunction may lead to mitochondrial damage and necroptosis [[104]51]. Alanine, aspartate, and glutamate metabolism regulate intracellular amino acid and nitrogen balance, affecting cell survival signals. Dysregulation in these pathways may trigger cell death mechanisms [[105]52]. Additionally, pyruvate, as a key junction between glycolysis and the tricarboxylic acid cycle, is directly linked to necroptosis caused by energy depletion when its metabolism is disrupted [[106]53]. The disruption of these metabolic pathways is closely related to necroptosis in various pathological conditions, suggesting that metabolic regulation could provide new strategies for preventing and treating diseases associated with necroptosis. The results indicate that Angiopep-2/SIRT1 nanoparticles significantly ameliorate sevoflurane-induced DNT. The underlying mechanisms primarily involve the regulation of multiple key metabolic pathways related to necroptosis. These findings not only deepen our understanding of the pathological mechanisms of DNT but also provide a crucial molecular basis for developing new therapeutic strategies. With further research, we hope to more comprehensively uncover the potential of these nanoparticles in regulating cell death, offering effective treatment options for DNT. Angiopep-2/SIRT1 nanoparticles activate SIRT1 to inhibit necroptosis and alleviate sevoflurane-induced DNT in SH-SY5Y cells In this study, we utilized an SH-SY5Y cell model to explore the neurodevelopmental toxicity effects of sevoflurane and the regulatory role of Angiopep-2/SIRT1 nanoparticles. Neuronal migration is a critical process in brain development, and its disruption is often associated with DNT. Such toxicity can alter migration pathways, affecting the formation and function of neural networks and potentially leading to cognitive and behavioral abnormalities [[107]54, [108]55]. Therefore, proper neuronal migration is essential for maintaining the health and function of the nervous system. Through detailed cellular and molecular biological analyses, we observed the effects of sevoflurane on apoptosis and migration, as well as its specific impact on neuronal morphology and function. These analyses helped to elucidate the potential mechanisms by which Angiopep-2/SIRT1 nanoparticles regulate intracellular signaling pathways. Our findings demonstrate that Angiopep-2/SIRT1 nanoparticles can activate SIRT1, inhibit necroptosis, and significantly reduce sevoflurane-induced DNT in SH-SY5Y cells. The technical route for the sevoflurane-induced DNT cell model is depicted in Fig. S8A. SH-SY5Y cells were seeded at a density of 10,000 cells per well in a 96-well plate using a normal culture medium. Within 24 h, the cells proliferated to reach 90% confluency. Treatment with 60 mg/L sevoflurane caused the culture medium to turn green, and this concentration was subsequently used for model induction (Fig. S8B). The cell migration assay results showed that cell migration significantly increased in the DNT group compared to the PBS group (Fig. S8C). Immunofluorescence results indicated a significant downregulation of β-III-tubulin expression in the DNT group compared to the PBS group (Fig. S8D). These findings confirm the successful establishment of the sevoflurane-induced DNT cell model. Initially, two shRNA constructs targeting SIRT1 (sh-SIRT1) were transfected into SH-SY5Y cells. RT-qPCR results indicated that sh-SIRT1-1 exhibited the highest silencing efficiency, and thus, sh-SIRT1-1 (sh-SIRT1) was used for subsequent experiments (Fig. S8E). WB results showed that, compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group had significantly lower relative expression levels of p-RIPK1/RIPK1, p-RIPK3/RIPK3, and p-MLKL/MLKL, and significantly higher expression of SIRT1 in SH-SY5Y cells. In contrast, the sh-SIRT1 group exhibited significantly higher relative expression levels of p-RIPK1/RIPK1, p-RIPK3/RIPK3, and p-MLKL/MLKL, and significantly lower expression of SIRT1. Moreover, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group showed significantly higher relative expression levels of p-RIPK1/RIPK1, p-RIPK3/RIPK3, and p-MLKL/MLKL, and significantly lower expression of SIRT1 in SH-SY5Y cells (Fig. [109]3A). Immunofluorescence results demonstrated that, compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group showed significantly lower fluorescence intensities of p-RIPK1, p-RIPK3, and p-MLKL and a significantly higher fluorescence intensity of SIRT1 in SH-SY5Y cells. Conversely, the sh-SIRT1 group exhibited significantly higher fluorescence intensities of p-RIPK1, p-RIPK3, and p-MLKL and significantly lower fluorescence intensity of SIRT1. Furthermore, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group displayed significantly higher fluorescence intensities of p-RIPK1, p-RIPK3, and p-MLKL, and significantly lower fluorescence intensity of SIRT1 in SH-SY5Y cells (Fig. [110]3B-E). Fig. 3. [111]Fig. 3 [112]Open in a new tab Effects of Angiopep-2/SIRT1 Nanoparticles on Necroptosis-Related Proteins. A WB analysis of the expression levels of p-RIPK1/RIPK1, p-RIPK3/RIPK3, p-MLKL/MLKL, and SIRT1 in SH-SY5Y cells; (B) Immunofluorescence staining showing the expression of SIRT1 in SH-SY5Y cells, scale bar: 25 μm; (C) Immunofluorescence staining showing the expression of p-RIPK1 in SH-SY5Y cells, with GFP-labeled neurons used for protein quantification, scale bar: 25 μm; (D) Immunofluorescence staining showing the expression of p-RIPK3 in SH-SY5Y cells, with GFP-labeled neurons used for protein quantification, scale bar: 25 μm; (E) Immunofluorescence staining showing the expression of p-MLKL in SH-SY5Y cells, with GFP-labeled neurons used for protein quantification, scale bar: 25 μm. Each experiment was repeated three times, and values are presented as mean ± standard deviation. *** indicates p < 0.001 CCK-8 assays were used to assess cell viability. The results showed that, compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group had increased cell viability in SH-SY5Y cells, while the sh-SIRT1 group had decreased cell viability. Additionally, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group exhibited reduced cell viability in SH-SY5Y cells (Fig. [113]4A). PI staining results indicated that, compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group showed significantly reduced PI fluorescence in SH-SY5Y cells, whereas the sh-SIRT1 group exhibited significantly increased PI fluorescence. Furthermore, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group displayed significantly increased PI fluorescence in SH-SY5Y cells (Fig. [114]4B). AO staining results showed that, compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group exhibited increased cytoplasmic (red fluorescence) and nuclear (green fluorescence) integrity in SH-SY5Y cells, whereas the sh-SIRT1 group showed decreased cytoplasmic and nuclear integrity. Furthermore, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group exhibited decreased cytoplasmic and nuclear integrity in SH-SY5Y cells (Fig. [115]4C). RT-qPCR results indicated that, compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group had significantly downregulated mRNA levels of Interleukin-6 (IL-6), Tumor Necrosis Factor-alpha (TNF-α), and Transforming Growth Factor-beta (TGF-β) in SH-SY5Y cells, while the sh-SIRT1 group showed significantly upregulated levels of these cytokines. Additionally, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group had significantly upregulated mRNA levels of IL-6, TNF-α, and TGF-β in SH-SY5Y cells (Fig. [116]4D). Fig. 4. [117]Fig. 4 [118]Open in a new tab Effects of Angiopep-2/SIRT1 Nanoparticles on Necroptosis. A CCK-8 assay to detect the cell viability of SH-SY5Y cells; (B) PI staining to detect apoptosis in SH-SY5Y cells, scale bar: 25 μm; (C) AO staining to examine DNA, RNA, and neurons in SH-SY5Y cells, scale bar: 25 μm; (D) RT-qPCR analysis of IL-6, TNF-α, and TGF-β mRNA levels in SH-SY5Y cells. Each experiment was repeated three times, and values are presented as mean ± standard deviation. *** indicates p < 0.001 Fluo-3 AM was used to measure intracellular Ca²⁺ levels. The results showed that, compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group had significantly increased Ca²⁺ levels in SH-SY5Y cells, whereas the sh-SIRT1 group exhibited significantly reduced Ca²⁺ levels. Additionally, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group showed significantly reduced Ca²⁺ levels in SH-SY5Y cells (Fig. [119]5A). Immunofluorescence analysis was used to detect the expression of PSD95 and β-III-tubulin in SH-SY5Y cells. The results indicated that, compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group had significantly upregulated expression of PSD95 and β-III-tubulin, while the sh-SIRT1 group showed significantly downregulated expression of these proteins. Furthermore, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group exhibited significantly downregulated expression of PSD95 and β-III-tubulin in SH-SY5Y cells (Fig. [120]5B-C). The cell migration assay results showed that compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group exhibited a significant reduction in cell migration, while the sh-SIRT1 group demonstrated a significant increase in cell migration. Additionally, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group showed a significant increase in cell migration (Fig. [121]5D). Immunofluorescence staining revealed that compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group had a significant increase in the total length of neurites and the average number of dendritic spines in SH-SY5Y cells. In contrast, the sh-SIRT1 group exhibited a significant decrease in both neurite length and dendritic spine density. Moreover, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group also showed a significant reduction in neurite length and dendritic spine number (Fig. [122]5E). Fig. 5. [123]Fig. 5 [124]Open in a new tab Angiopep-2/SIRT1 Nanoparticles Inhibit Necroptosis to Alleviate Sevoflurane-Induced DNT in SH-SY5Y Cells. A Fluo-3 AM assay to detect Ca^2+ levels in SH-SY5Y cells; (B) Immunofluorescence staining to detect the expression of PSD95 in SH-SY5Y cells, scale bar: 25 μm; (C) Immunofluorescence staining to detect the expression of β-III-tubulin in SH-SY5Y cells, scale bar: 25 μm; (D) Cell migration assay to assess migration in SH-SY5Y cells, scale bar: 200 μm; (E) Immunofluorescence staining of green (Synaptophysin), red (MAP-2), and blue (DAPI) in SH-SY5Y cells, scale bar: 25 μm. Each experiment was repeated three times, and values are presented as mean ± standard deviation. *** indicates p < 0.001 The above results indicate that Angiopep-2/SIRT1 nanoparticles effectively inhibit the necroptosis signaling pathway induced by sevoflurane, including the reduction of p-RIPK1, p-RIPK3, and p-MLKL expression, thereby promoting neuronal cell survival and structural integrity. Furthermore, these nanoparticles significantly improve the physiological state of cells by enhancing cell migration ability and reducing changes in intracellular calcium ion concentration. This provides strong experimental evidence for the development of new therapeutic strategies. These findings offer important molecular targets and potential drug delivery systems for future DNT treatment research. Angiopep-2/SIRT1 nanoparticles activate SIRT1 to inhibit necroptosis and alleviate sevoflurane-induced DNT in mice In this study, we rigorously evaluated the effects of Angiopep-2/SIRT1 nanoparticles on sevoflurane-induced DNT through comprehensive cellular and animal experiments. Utilizing a mouse model, we conducted a series of biochemical and histological analyses to explore how these nanoparticles modulate the SIRT1-related necroptosis pathway and to elucidate their potential neuroprotective mechanisms. We paid particular attention to the nanoparticles’ effects on nuclear condensation, vacuolization, dendritic spine density, and neurite outgrowth, as well as their impact on performance in behavioral tests. Using the model of sevoflurane-induced DNT in mice established according to Fig. [125]6A results from H&E staining and TEM revealed the following: Compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group exhibited significantly reduced nuclear condensation and vacuolization, as well as an increased number of striatal neurons. Conversely, the sh-SIRT1 group showed significantly increased nuclear condensation and vacuolization, along with a decreased number of striatal neurons. Additionally, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group showed significantly increased nuclear condensation and vacuolization, and a reduced number of striatal neurons (Fig. [126]6B). Nissl staining results indicated that, compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group had a significant increase in Nissl-stained cells, whereas the sh-SIRT1 group had a significant decrease. Furthermore, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group exhibited a significant decrease in Nissl-stained cells (Fig. [127]6C). Immunofluorescence results showed that β-III-tubulin and PSD95 were significantly increased in the brain tissues of the sh-NC + Angiopep-2/SIRT1 group compared to the sh-NC group, whereas they were significantly decreased in the sh-SIRT1 group. Additionally, β-III-tubulin and PSD95 levels were significantly reduced in the Angiopep-2/SIRT1 + sh-SIRT1 group compared to the sh-NC + Angiopep-2/SIRT1 group (Fig. [128]6D-E). Golgi staining examined the density of dendritic spines, and the results indicated that, compared to the sh-NC group, the number of pyramidal neuron spines was significantly increased in the sh-NC + Angiopep-2/SIRT1 group, while significantly decreased in the sh-SIRT1 group. Furthermore, the number of pyramidal neuron spines was significantly reduced in the Angiopep-2/SIRT1 + sh-SIRT1 group compared to the sh-NC + Angiopep-2/SIRT1 group (Fig. [129]6F). Fig. 6. [130]Fig. 6 [131]Open in a new tab Effects of Angiopep-2/SIRT1 Nanoparticles on Sevoflurane-Induced DNT in Mice. A Technical roadmap for establishing a sevoflurane-induced DNT model in mice; (B) H&E staining and TEM analysis of cellular morphology and structure in mouse brains, scale bars: 100 μm (top row), 2 μm (second row), 500 nm (bottom row), with blue arrows indicating mitochondria and green arrows indicating autophagic vacuoles; (C) Nissl staining to detect striatal neurons in mouse brains, black arrows indicating neurons, scale bar: 100 μm; (D) Immunofluorescence staining to detect the expression of β-III-tubulin in mouse brains, scale bar: 50 μm; (E) Immunofluorescence staining to detect the expression of PSD95 in mouse brains, scale bar: 50 μm; (F) Golgi staining to detect dendritic spines in the hippocampus of mice, scale bar: 10 μm. Each group consisted of 6 mice, and values are presented as mean ± standard deviation. *** indicates p < 0.001 On days 31, 35, and 37, after establishing the DNT model, we conducted negative geotaxis and front-limb suspension tests. The results of the negative geotaxis test showed that, compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group required significantly less time to turn their heads, while the sh-SIRT1 group required significantly more time. Furthermore, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group took significantly more time to turn their heads (Fig. [132]7A). In the front-limb suspension test, the sh-NC + Angiopep-2/SIRT1 group spent significantly more time hanging on the wire compared to the sh-NC group, whereas the sh-SIRT1 group spent significantly less time. Additionally, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group showed a significant decrease in the hanging time (Fig. [133]7B). To further assess the long-term neurological damage caused by sevoflurane in DNT mice, we conducted NORT and MWMT. The NORT results indicated that the discrimination index was significantly higher in the sh-NC + Angiopep-2/SIRT1 group compared to the sh-NC group, while it was significantly lower in the sh-SIRT1 group. Additionally, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group showed a significantly lower discrimination index (Fig. [134]7C). MWMT results showed that the sh-NC + Angiopep-2/SIRT1 group spent significantly less time finding the platform compared to the sh-NC group, whereas the sh-SIRT1 group spent significantly more time. Furthermore, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group took significantly more time to find the platform. However, there was no significant difference in swimming speed between the two groups (Fig. [135]7D-G). In the MWMT, the sh-NC + Angiopep-2/SIRT1 group spent significantly more time in the escape zone compared to the sh-NC group, while the sh-SIRT1 group spent significantly less time. Compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group also showed a significantly reduced duration in the escape zone (Fig. [136]7H). Fig. 7. [137]Fig. 7 [138]Open in a new tab Effects of Angiopep-2/SIRT1 Nanoparticles on Neurological Reflexes in Sevoflurane-Induced DNT in Mice. A Negative geotaxis test results on days 31, 35, and 37 post-construction of the sevoflurane-induced DNT model in different groups of mice; (B) Front-limb suspension test results on days 31, 35, and 37 post-construction of the sevoflurane-induced DNT model in different groups of mice; (C) NORT test results post-construction of the sevoflurane-induced DNT model in different groups of mice, n = 6; (D-G) MWMT results post-construction of the sevoflurane-induced DNT model in different groups of mice, including swimming speed, escape latency, swimming distance, and time spent in the target quadrant; (H) Representative Morris water maze swimming trajectory images post-construction of the sevoflurane-induced DNT model in different groups of mice. Each group consisted of 6 mice, and values are presented as mean ± standard deviation. *** indicates p < 0.001 First, the WB results showed that, compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group exhibited significantly downregulated relative expression levels of p-RIPK1/RIPK1, p-RIPK3/RIPK3, and p-MLKL/MLKL in the brain tissue, while the expression of SIRT1 was significantly upregulated. Conversely, in the sh-SIRT1 group, the relative expression levels of p-RIPK1/RIPK1, p-RIPK3/RIPK3, and p-MLKL/MLKL were significantly upregulated, and SIRT1 expression was significantly downregulated. Additionally, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group showed significantly upregulated relative expression levels of p-RIPK1/RIPK1, p-RIPK3/RIPK3, and p-MLKL/MLKL, with a significant downregulation of SIRT1 expression (Fig. S9A). Immunofluorescence results showed that, compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group exhibited significantly reduced fluorescence intensity of p-RIPK1, p-RIPK3, and p-MLKL in the brain tissue, while the fluorescence intensity of SIRT1 was significantly increased. In contrast, the sh-SIRT1 group showed significantly increased fluorescence intensity of p-RIPK1, p-RIPK3, and p-MLKL, with a significant decrease in the fluorescence intensity of SIRT1. Additionally, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group exhibited significantly increased fluorescence intensity of p-RIPK1, p-RIPK3, and p-MLKL, and a significantly decreased fluorescence intensity of SIRT1 (Fig. S9B-E). TUNEL results showed that, compared to the sh-NC group, neuronal apoptosis in the brain tissue was significantly reduced in the sh-NC + Angiopep-2/SIRT1 group, while it was significantly increased in the sh-SIRT1 group. Furthermore, compared to the sh-NC + Angiopep-2/SIRT1 group, neuronal apoptosis was significantly increased in the Angiopep-2/SIRT1 + sh-SIRT1 group (Fig. [139]8A). ELISA results indicated that, compared to the sh-NC group, the sh-NC + Angiopep-2/SIRT1 group exhibited a significant increase in the content of SOD and a significant decrease in MDA in brain tissue. Conversely, the sh-SIRT1 group showed a significant decrease in SOD content and a significant increase in MDA content. Additionally, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group exhibited a significant decrease in SOD content and a significant increase in MDA content (Fig. [140]8B-C). PI staining results showed that compared to the sh-NC group, PI fluorescence in the brain tissue was significantly reduced in the sh-NC + Angiopep-2/SIRT1 group, while it was significantly increased in the sh-SIRT1 group. Furthermore, compared to the sh-NC + Angiopep-2/SIRT1 group, PI fluorescence was significantly increased in the Angiopep-2/SIRT1 + sh-SIRT1 group (Fig. [141]8D). RT-qPCR results indicated that compared to the sh-NC group, the mRNA levels of IL-6, TNF-α, and TGF-β in brain tissue were significantly downregulated in the sh-NC + Angiopep-2/SIRT1 group, while they were significantly upregulated in the sh-SIRT1 group. Additionally, compared to the sh-NC + Angiopep-2/SIRT1 group, the Angiopep-2/SIRT1 + sh-SIRT1 group exhibited significantly upregulated mRNA levels of IL-6, TNF-α, and TGF-β (Fig. [142]8E). Fig. 8. [143]Fig. 8 [144]Open in a new tab Effects of Angiopep-2/SIRT1 Nanoparticles on Necroptosis. A TUNEL staining to detect apoptosis in mouse brain tissue, scale bar: 50 μm; (B) ELISA to measure SOD content in mouse brain tissue; (C) ELISA to measure MDA content in mouse brain tissue; (D) PI staining to detect apoptosis in mouse brain tissue, scale bar: 50 μm; (E) RT-qPCR to measure mRNA levels of IL-6, TNF-α, and TGF-β in mouse brain tissue. Each group consisted of 6 mice, and values are presented as mean ± standard deviation. *** indicates p < 0.001 These experimental results demonstrate that Angiopep-2/SIRT1 nanoparticles effectively mitigate sevoflurane-induced cellular and tissue damage by reducing the expression of necroptosis-related proteins and enhancing the cellular antioxidant defense system. These nanoparticles not only improved cell survival and structural integrity at the cellular level but also enhanced the cognitive and motor functions of the treated mice in vivo. This finding highlights the potential therapeutic value of Angiopep-2/SIRT1 nanoparticles in the prevention and treatment of DNT. Discussion This study utilizes multi-omics approaches to elucidate the mechanisms by which self-assembled Angiopep-2/SIRT1 nanoparticles alleviate sevoflurane-induced DNT, showcasing significant innovation compared to previous research. Earlier studies have primarily focused on sevoflurane-induced neuronal apoptosis and inflammatory responses, lacking in-depth exploration of the specific molecular mechanisms involved [[145]56, [146]57]. Through systematic multi-omics analysis, our research reveals key changes in genes and proteins, offering new insights into the understanding of sevoflurane-induced DNT. This approach not only provides comprehensive molecular-level information but also uncovers complex biological networks and signaling pathways, laying a solid foundation for future research. SIRT1, an important deacetylase, has been confirmed to play a neuroprotective role in multiple studies [[147]58]. Our study found that SIRT1 significantly alleviates sevoflurane-induced neurotoxicity by inhibiting necroptosis, which is consistent with previous research findings. However, past studies have primarily focused on the regulation of apoptosis by SIRT1, whereas our study is the first to reveal its crucial role in necroptosis [[148]59–[149]61]. Compared to other studies on neurotoxicity and neurodegenerative diseases, our research further expands the protective scope of SIRT1, highlighting its broad application potential under different pathological conditions. The development of self-assembled Angiopep-2/SIRT1 nanoparticles represents another major innovation in this study. Compared to other nano-drug delivery systems, Angiopep-2/SIRT1 nanoparticles offer superior stability and biocompatibility, efficiently crossing the BBB to achieve targeted delivery [[150]62]. Previous research has shown that many nanoparticles suffer from poor stability and targeting ability in vivo, limiting their clinical applications. Our study employed various characterization methods and biocompatibility tests to confirm the superior performance of Angiopep-2/SIRT1 nanoparticles, demonstrating their significant potential in the treatment of neurological diseases. The application of multi-omics approaches in this study further enhances the reliability and comprehensiveness of the research findings. Through high-throughput sequencing, proteomics, and metabolomics analyses, we not only identified key genes and proteins involved in sevoflurane-induced DNT but also analyzed related metabolic pathways. Compared to single-omics methods, multi-omics approaches provide more comprehensive biological information, revealing complex biological networks and signaling pathways [[151]63–[152]65]. The results of our multi-omics analysis offer new insights and directions for understanding the molecular mechanisms underlying sevoflurane-induced DNT. The importance of in vivo and in vitro experiments in validating the intervention effects of nanoparticles cannot be overstated. In this study, we systematically evaluated the protective effects of Angiopep-2/SIRT1 nanoparticles using both in vivo mouse models and in vitro cell experiments. The in vivo experiments demonstrated that the nanoparticles significantly alleviated sevoflurane-induced neurological deficits and neuronal apoptosis in mice. The in vitro experiments further confirmed the protective effects of the nanoparticles at the cellular level. The consistency between in vivo and in vitro results strengthens the credibility and reproducibility of our findings. Necroptosis plays a crucial role in sevoflurane-induced DNT [[153]66–[154]68]. Through various experimental techniques, this study detailed the regulatory mechanisms of self-assembled Angiopep-2/SIRT1 nanoparticles on necroptosis (Fig. [155]9). Unlike previous studies [[156]12], this research systematically explored the role of necroptosis in sevoflurane-induced DNT, filling a significant gap in the field. By inhibiting the expression of p-RIPK1, p-RIPK3, and p-MLKL, SIRT1 effectively reduced necroptosis, thereby protecting neuronal cells. Fig. 9. [157]Fig. 9 [158]Open in a new tab Molecular Mechanisms of Self-Assembled Angiopep-2/SIRT1 Nanoparticles in Alleviating Sevoflurane-Induced DNT by Regulating Necroptosis Through SIRT1 Activation Inflammatory responses are a major pathological process in sevoflurane-induced DNT [[159]69–[160]71]. This study found that Angiopep-2/SIRT1 nanoparticles significantly attenuate inflammatory responses by regulating the expression of IL-6, TNF-α, and TGF-β. Compared to other anti-inflammatory strategies, our approach not only effectively reduces inflammation but also reveals its precise molecular mechanisms through multi-omics analysis. The dual role of nanoparticles in modulating inflammatory responses underscores their broad application potential in neuroprotection. In conclusion, this study successfully developed self-assembled Angiopep-2/SIRT1 nanoparticles and explored their effects on inhibiting sevoflurane-induced DNT. Compared to existing research, the innovation of this study lies in using Angiopep-2 as a carrier, which effectively enhances the intracellular delivery efficiency of SIRT1. Although previous studies have demonstrated that SIRT1 can protect neurons through antioxidant and anti-inflammatory actions, its in vivo application has been limited by the lack of an effective delivery system. By utilizing Angiopep-2/SIRT1 nanoparticles, this study not only improved the stability and bioavailability of SIRT1 in neural cells but also significantly enhanced its efficacy against DNT. This study, through multi-omics approaches and in vivo and in vitro experiments, revealed the mechanisms by which self-assembled Angiopep-2/SIRT1 nanoparticles alleviate sevoflurane-induced DNT, providing new molecular targets and therapeutic strategies. By activating SIRT1 and inhibiting necroptosis and inflammatory responses, the nanoparticles significantly improved neural function impairment. These findings not only deepen our understanding of the mechanisms underlying sevoflurane-induced neurotoxicity but also offer a potential clinical intervention method that could enhance the safety of pediatric anesthesia and reduce the risk of long-term neurological damage [[161]5, [162]43]. In this study, Angiopep-2/SIRT1 self-assembling nanoparticles were successfully prepared using the anti-solvent precipitation method. The resulting nanoparticles demonstrated good particle size uniformity and biocompatibility, making them suitable for laboratory-scale preparation and application. However, the industrial-scale production of nanoparticles still faces numerous challenges, including control of batch-to-batch consistency, stability of physicochemical properties during scale-up, and optimization of production costs. Future studies should focus on the standardization and optimization of process parameters to ensure the stability and functionality of the nanoparticles during large-scale manufacturing, thereby promoting their clinical translation potential [[163]72, [164]73]. Although this study verified the biocompatibility of Angiopep-2/SIRT1 nanoparticles through in vitro cell viability assays, hemolysis tests, and short-term animal experiments, the long-term in vivo metabolism, accumulation, and potential toxicity of the nanoparticles have not yet been systematically evaluated. Nanomaterials may undergo slow degradation in vivo or accumulate in specific organs (such as the liver, kidneys, spleen, and brain), potentially triggering chronic inflammation or immune responses [[165]74]. Therefore, future work should include long-term toxicological studies, covering the nanoparticles’ in vivo biodistribution, metabolic pathways, immune activation, and potential tissue toxicity, with particular emphasis on safety monitoring within brain tissue to comprehensively assess clinical application risks. Angiopep-2, as a brain-penetrating peptide, primarily mediates brain-targeted delivery of nanoparticles through low-density lipoprotein receptor-related protein 1 (LRP1). Given that LRP1 is expressed in various tissues, there is a risk of off-target distribution of nanoparticles in non-target organs. Such off-target effects may lead to unintended biological responses, thereby affecting therapeutic safety and efficacy. In future studies, we will use high-sensitivity imaging techniques and multi-organ distribution analyses to systematically evaluate the targeting specificity of the nanoparticles. Moreover, strategies such as adjusting Angiopep-2 density and surface modifications (e.g., PEGylation) will be explored to reduce off-target effects and enhance therapeutic precision [[166]75]. Additionally, since nanoparticles may activate the immune system and trigger immune responses such as complement activation, comprehensive immune compatibility testing will be essential for safety assessment. This study demonstrated that Angiopep-2/SIRT1 nanoparticles alleviate sevoflurane-induced DNT by activating SIRT1 and inhibiting necroptosis, exhibiting favorable brain-targeting ability and biocompatibility. In recent years, lipid nanoparticles (LNPs) delivering mRNA encoding the necroptosis effector MLKL have been shown to induce tumor cell necroptosis and activate immune responses, exhibiting strong antitumor potential [[167]76]. Furthermore, CRISPR-Cas9 systems are being developed for gene regulation in central nervous system (CNS) diseases, enabling precise modulation of pathological gene expression, reduction of off-target effects, and improved therapeutic specificity [[168]77]. In comparison, the Angiopep-2/SIRT1 nanoparticles in this study primarily activate endogenous protective factor SIRT1, inhibit necroptosis signaling pathways, and reduce inflammatory factor expression. This approach focuses on regulating intracellular signaling and metabolic pathways, offering advantages in terms of biocompatibility and safety. Unlike RNA therapy and gene editing, this strategy avoids the direct introduction of exogenous genes, potentially reducing immunogenicity and off-target risks; however, it may be relatively limited in its capacity for precise gene-level modulation [[169]78]. In the future, integrating this nanoparticle platform with RNA or CRISPR technologies may yield synergistic effects, enabling more efficient regulation of programmed necrosis and neuroprotection. Despite the significant progress made in elucidating the mechanisms of sevoflurane-induced DNT, this study has certain limitations. First, the relatively small sample size may affect the generalizability of the findings. Second, the experimental conditions and selected models may not fully replicate clinical scenarios, thus limiting the extrapolation of the results. Moreover, the long-term safety and efficacy of the nanoparticles remain to be further validated. Future research should focus on increasing the sample size and optimizing the experimental design to validate the broad applicability and consistent efficacy of self-assembled Angiopep-2/SIRT1 nanoparticles. Additionally, the potential application of these nanoparticles in various neurotoxic and neurodegenerative diseases should be further explored, and their long-term safety and efficacy in clinical settings should be assessed. Through continued research and clinical trials, we aim to develop more effective and safer therapeutic strategies for sevoflurane-induced DNT, thereby enhancing anesthesia safety and protecting children’s brain health. Supplementary Information [170]Supplementary material 1^ (588.3KB, jpg) [171]Supplementary material 2^ (402.6KB, jpg) [172]Supplementary material 3^ (479.7KB, jpg) [173]Supplementary material 4^ (442.4KB, jpg) [174]Supplementary material 5^ (480.3KB, jpg) [175]Supplementary material 6^ (598KB, jpg) [176]Supplementary material 7^ (1.1MB, jpg) [177]Supplementary material 8^ (1.2MB, jpg) [178]Supplementary material 9^ (2.2MB, jpg) [179]Supplementary material 10^ (24.4KB, docx) Abbreviations AO Acridine Orange BBB Blood-Brain Barrier BP Biological Process CC Cellular Components CLSM Confocal Laser Scanning Microscope DIA Data-Independent Acquisition DLS Dynamic Light Scattering DMs Differential Metabolites DNT Developmental Neurotoxicity FACS Fluorescence-Activated Cell Sorting FA Formic Acid FBS Fetal Bovine Serum GO Gene Ontology H&E Hematoxylin and Eosin HPLC High-Performance Liquid Chromatography IL-6 Interleukin-6 KEGG Kyoto Encyclopedia of Genes and Genomes MDA Malondialdehyde MF Molecular Function MWMT Morris Water Maze Tests NORT Novel Object Recognition Test OPLS-DA Orthogonal Partial Least Squares-Discriminant Analysis PCA Principal Component Analysis PBS Phosphate-Buffered Saline PDI Polydispersity Index PI Propidium Iodide rRNA Ribosomal RNA RBF Radial Basis Function RFE Recursive Feature Elimination SEM Scanning Electron Microscopy SIRT1 Sirtuin 1 SOD Superoxide Dismutase SVM Support Vector Machine TEM Transmission Electron Microscopy TGF-β Transforming Growth Factor-beta TNF-α Tumor Necrosis Factor-alpha WB Western Blot Author contributions Y.C. and X.Z. contributed equally to this work. Y.C. and S.Z. designed the research. X.Z. and G.Q. performed the experiments and data analysis. Y.C. conducted the behavioral assays and contributed to data interpretation. S.Z. and G.Q. supervised the project and provided critical revisions to the manuscript. All authors reviewed and approved the final version of the manuscript. Funding Not applicable. Data availability All data can be provided as needed. Declarations Ethics approval and consent to participate All animal experiments were approved by the Animal Ethics Committee of the First Hospital of China Medical University. Consent for publication All authors participating in this research gave their full consent to publish the findings. Competing interests The authors declare no competing interests. Footnotes Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Yi Chang and Xue Zhang are regarded as co-first authors. Contributor Information Shuo Zhang, Email: zhangshuo1005@sina.com. Ge Qu, Email: 546653916@qq.com. References