Abstract Objective The exacerbation of extreme high-temperature events due to global climate change poses a significant challenge to public health, particularly impacting the central nervous system through heat stroke. This study aims to develop Poly(amidoamine) (PAMAM) nanoparticles loaded with curcumin (PAMAM@Cur) to enhance its therapeutic efficacy in hypothalamic neural damage in a heat stroke model and explore its potential mechanisms. Methods Curcumin (Cur) was encapsulated into PAMAM nanoparticles through a hydrophobic interaction method, and various techniques were employed to characterize their physicochemical properties. A heat stroke mouse model was established to monitor body temperature and serum biochemical parameters, conduct behavioral assessments, histological examinations, and biochemical analyses. Transcriptomic and proteomic analyses were performed to investigate the therapeutic mechanisms of PAMAM@Cur, validated in an N2a cell model. Results PAMAM@Cur demonstrated good stability, photostability, cell compatibility, significant blood–brain barrier (BBB) penetration capability, and effective accumulation in the brain. PAMAM@Cur markedly improved behavioral performance and neural cell structural integrity in heat stroke mice, alleviated inflammatory responses, with superior therapeutic effects compared to Cur or PAMAM alone. Multi-omics analysis revealed that PAMAM@Cur regulated antioxidant defense genes and iron death-related genes, particularly upregulating the PCBP2 protein, stabilizing SLC7A11 and GPX4 mRNA, and reducing iron-dependent cell death. Conclusion By enhancing the drug delivery properties of Cur and modulating molecular pathways relevant to disease treatment, PAMAM@Cur significantly enhances the therapeutic effects against hypothalamic neural damage induced by heat stroke, showcasing the potential of nanotechnology in improving traditional drug efficacy and providing new strategies for future clinical applications. Significance This study highlights the outlook of nanotechnology in treating neurological disorders caused by heat stroke, offering a novel therapeutic approach with potential clinical applications. Graphical abstract [28]graphic file with name 12951_2024_2771_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-024-02771-3. Keywords: Heat Stroke Treatment, PAMAM Nanoparticles, Curcumin, Hypothalamus Nerve Injury, Antioxidant Defense Introduction Global climate change has had a serious impact on the frequent occurrence of extreme high-temperature events, exacerbating heat stress reactions, including heat stroke (HS), which has become an urgent issue in the field of global public health [[29]1, [30]2]. Heat shock (HS) is a condition triggered by high temperatures and a humid environment, characterized mainly by symptoms such as elevated body temperature and fluid loss, which can lead to organ dysfunction and death in severe cases [[31]3, [32]4]. With the increasing hot weather conditions due to climate change, people are more susceptible to heat stress, resulting in a rising incidence of HS [[33]5–[34]8]. Therefore, studying the mechanisms of HS and finding effective treatment methods are crucial to safeguarding people's health [[35]9, [36]10]. The pathophysiology of HS involves the disruption of the body's heat balance regulation system, leading to elevated body temperature and further disarray in heat stress reactions, triggering systemic physiological and biochemical responses [[37]11, [38]12]. During an HS episode, the body faces severe heat stress, resulting in damage to vital organs such as the central nervous system, cardiovascular system, and kidneys [[39]13–[40]15]. The impact of the central nervous system on HS is crucial; heat injuries cause brain inflammation responses, neuronal damage, and disrupted nerve conduction, potentially leading to cerebral edema and neuronal apoptosis in severe cases [[41]16–[42]18]. Thus, protecting the central nervous system and reducing inflammation responses are particularly important in the treatment of HS [[43]19–[44]21]. With the advancement of nanotechnology, the application of nano-carriers in drug delivery is receiving increasing attention. As a novel nano-carrier, PAMAM@Cur encapsulating Cur shows promise in improving drug stability, bioavailability, and targeting ability, potentially enhancing the therapeutic efficacy in the treatment of HS. PAMAM nanoparticles exhibit excellent biocompatibility and cellular penetration ability, capable of crossing the BBB to reach the central nervous system, offering a new avenue for treating neuro-related diseases [[45]22–[46]24]. Cur, a natural active ingredient renowned for its anti-inflammatory, antioxidant, and anti-apoptotic properties, when combined with PAMAM nano-carriers, can further enhance its pharmacological effects and mitigate adverse reactions, thus opening up new possibilities for treating HS [[47]25]. To investigate the therapeutic effects and mechanism of action of PAMAM@Cur on HS, we designed a series of systematic experimental protocols. Initially, employing a hydrophobic interaction method, we successfully encapsulated Cur into PAMAM nanoparticles and ensured the stability and biocompatibility of the nanoparticles through detailed physicochemical characterization [[48]26]. Subsequently, we established a mouse model of HS by exposing them to 42 °C for 1 h followed by recovery at 24 °C, monitoring body temperature changes, assessing serum biochemical indicators, and evaluating behavioral performance [[49]27–[50]29]. Furthermore, through histological examinations and molecular mechanism studies, we investigated the therapeutic effects of PAMAM@Cur on hypothalamus nerve injury. Additionally, through in vitro and in vivo experiments, we validated the drug delivery properties and cell compatibility of PAMAM@Cur. These comprehensive experimental designs will help elucidate the mechanisms and potential of PAMAM@Cur in the treatment of HS, providing a scientific basis for future clinical applications. This study aims to explore the therapeutic effects and underlying mechanisms of PAMAM@Cur on hypothalamus nerve injury induced by HS. Our research findings indicate that PAMAM@Cur effectively improves behavioral performance, and neural cell structural integrity, and reduces inflammation response in HS mice, demonstrating superior efficacy. By modulating the expression of antioxidant defense genes and ferroptosis-related genes, PAMAM@Cur exhibits significant therapeutic effects. This study is of vital significance in enhancing our understanding of the pathophysiological mechanisms of HS, developing novel therapeutic drugs, and advancing clinical treatment. It provides strong support for future strategies in treating HS. Materials and methods Ethical statement All mouse experiments conducted in this study have adhered to international and national ethical guidelines and standards regarding the use of experimental animals, including but not limited to the "Three Rs principle" (Replacement, Reduction, Refinement). The experimental procedures in this study were approved by the Animal Ethics Committee (Ethical approval number: DWLL20200111). We ensured that all experimental animals were treated with respect and in a humane manner throughout the research process. The housing and handling of all mice were carried out under conditions aimed at minimizing their pain and distress. Following the conclusion of the experiments, all mice were humanely euthanized. Establishment of an HS mouse model The Animal Ethics Committee (DWLL20230101). Male C57BL/6 mice aged 9 to 10 weeks (strain code: 219, Beijing Vital River Laboratory Animal Technology Co., Ltd.) were selected for the experiments. One week before the start of the experiment, the mice were acclimatized to a room temperature of 22 ± 2 °C, relative humidity of 50 ± 8%, with a 12-h light–dark cycle, and provided ad libitum access to food and water. Subsequently, the mice were exposed to a whole-body heat (WBH) environment at 42 °C and 50–55% relative humidity for 1 h to simulate HS conditions. Following the WBH exposure, the mice were returned to a room temperature of 24 °C for recovery. The criteria for successful modeling included signs such as deep breathing, cyanosis of the mouth, nose, and limbs, abdomen closely attached to the treadmill surface, and a rectal temperature reaching 42.0 °C as the HS standard. Additionally, proper feeding and hydration were provided to the mice after 1 h of WBH exposure. To evaluate the HS mouse model, physiological and biochemical tests were conducted on the mice's blood. After the experiment, whole blood was collected from the mice and serum was separated. The blood samples were left at room temperature for 2 h, then centrifuged at 3000 rpm for 15 min at 2–8 °C to obtain the upper layer of serum for biochemical testing. Biochemical parameters analyzed included alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), creatinine (CREA), urea (UREA), creatine kinase (CK), among others. Finally, the physiological and biochemical status of the HS mice as well as multi-organ function were evaluated [[51]30]. Open field test (OFT) The experimental setup for the OFT comprises a square testing arena divided into four sections, each measuring 50 cm × 50 cm × 40 cm. Boundary lines are marked on the floor of the area. Mice (Control group: 10 mice, HS group: 8 mice) are placed into the arena and given a 30-s habituation period. Each mouse undergoes a 10-min test in the arena, which is recorded by a video camera positioned 120 cm away from the testing area. Subsequently, the time spent by the mouse in the central area (defined as the time when all four limbs are simultaneously within the central square) and the distance traveled within this zone are analyzed. The testing environment is maintained under normal lighting conditions (800 lx) [[52]31]. Novel object recognition (NOR) Test Before the training session, mice are placed in the apparatus for 3 min to acclimate to the environment. Following acclimation, the mice (Control group: 10 mice, HS group: 8 mice) undergo a training session during which two identical objects are introduced into the apparatus for 5 min. The test session is conducted 2 h post-training, during which one of the familiar objects is replaced with a new object of a different shape but similar properties. Mice are allowed to explore these two objects for 5 min. The time spent by the mice exploring each object is recorded using the Any-maze video tracking system, and the recognition index and frequency of interactions with the new object are calculated. The recognition index is defined as the ratio of time spent by the mice on the new object to the total test duration [[53]32]. Immunofluorescent staining of C-Fos and NeuN After the completion of behavioral experiments, the mice in each group were euthanized, and their brain tissues were immediately fixed in a 4% paraformaldehyde solution (P0099, Beyotime, Shanghai, China). The fixed tissues were dehydrated, embedded, and cut into 5 μm thick consecutive sections. The sections were subjected to antigen retrieval and washed three times with PBS. Subsequently, the sections were treated with a 0.3% hydrogen peroxide solution (National Medicinal Code 10,011,208, National Medicinal Group Chemical Reagents Co., Ltd.) for 30 min to block endogenous peroxidase activity. Following this, the sections were incubated in a PBS solution containing 5% goat serum (C0265, Beyotime, Shanghai, China) to block nonspecific binding sites for 1 h. The sections were then separately incubated overnight at 4 °C with primary antibodies against C-Fos (ab208942, 1:200, Abcam, UK) and NeuN (ab177487, 1:300, Abcam, UK). Subsequently, the sections were incubated for 1 h at room temperature in the dark with a secondary antibody, goat anti-rabbit IgG H&L (Alexa Fluor® 488) (ab150077, 1:200, Abcam, UK). In the final step, the sections were counterstained with DAPI (D9542, Sigma-Aldrich), mounted with an antifade reagent, and observed under a confocal microscope (Leica, STELLARIS 5, Germany) [[54]30]. Preparation of PAMAM@Cur In the first step, the fourth generation (G4) PAMAM dendrimer polymer (412,449, Sigma-Aldrich) was fluorescently labeled with Cy5.5 (170C0, Lumiprobe). The labeled dendrimer polymer was dialyzed overnight in a 0.9% sodium chloride solution and further dialyzed in water. The Cy5.5-labeled dendrimer polymer was freeze-dried for 48 h and stored at -20 °C until further use. Subsequently, 1 mL of Cur (CAS No: 458–37-7, Sigma-Aldrich) dissolved in methanol (1 mg/mL) was added dropwise to the PAMAM solution in 1 mL of methanol (1 mg/mL) and 2 mL of PBS. The mixture solution was vigorously stirred for 2 days to encapsulate Cur within the PAMAM cavities. Methanol was removed using a rotary evaporator, leaving behind a PBS suspension. The PAMAM@Cur mixture was centrifuged (5000 rpm, 15 min at 4 °C) to remove the precipitate of non-complexed free Cur insoluble in PBS. The precipitate was collected and dissolved in 1 mL of methanol, and the amount of unloaded drug was determined by UV–visible spectroscopy, with the supernatant stored light-protected at 4 °C [[55]33]. Enzyme-linked immunosorbent assay (ELISA) The levels of TNF-α, IL-1β, and IL-6 in the hypothalamic lysates of the mice groups were determined using the Mouse TNF-α ELISA Kit (MTA00B, R&D Systems, USA), Mouse IL-6 ELISA Kit (M6000B, R&D Systems, USA), and Mouse IL-1β ELISA Kit (MLB00C, R&D Systems, USA). The experimental procedures were conducted according to the instructions provided in the respective kits [[56]30]. Physical and chemical characterization of nanoparticles The Malvern Zetasizer Nano-ZS (Malvern Instruments, Worcestershire, England) was utilized to measure the hydrodynamic size and Zeta potential of PAMAM-Cur formulation and PAMAM at a 20:1 N/P ratio before and after Cur loading using dynamic light scattering (DLS). The results were evaluated using the standard deviation (SD) ± method. To ascertain the encapsulation of Cur in the system (PAMAM@Cur), ultraviolet–visible absorption spectroscopy measurements were performed on free Cur, PAMAM, and PAMAM@Cur [[57]33]. Cur Release from PAMAM@Cur To determine the release of Cur from PAMAM@Cur complexes, a phosphate buffer solution (PBS, pH 7.4) was used as the medium, and the experiment was conducted using a dialysis bag method. Initially, 1 mg/mL of Cur-containing PAMAM@Cur was dissolved in 5 mL of PBS, followed by placing this solution into a dialysis bag with a molecular weight cutoff of 10 kDa. Subsequently, the dialysis bag was oscillated at 200 rpm in a water bath shaker at 37 °C with 50 mL of external PBS as the medium, allowing free Cur molecules to diffuse into the external medium. At specific time points of incubation (for 3 days), 500 μL of samples were withdrawn for analysis, and an equal volume of fresh PBS was immediately replenished to maintain system constancy. The released amount of Cur was quantified using high-performance liquid chromatography (HPLC) analysis. The procedure involved extracting Cur from the samples with chloroform (in a 1:1 volume ratio), followed by its dissolution in 50 μL of methanol. This solution was then injected into a reverse-phase HPLC system equipped with a Cecil 1100 series UV detector for analysis, with the mobile phase comprising a mixture of 75% acetonitrile and 25% water with 5% acetic acid, a flow rate of 1 mL/min, and a detection wavelength of 430 nm. The amount of released Cur was calculated by comparing it against the experimentally determined standard curve (linear range of 0.1–50 μg/mL) to ascertain the effective payload release [[58]33]. Photostability profiles of Cur and PAMAM@Cur Free Cur or PAMAM@Cur (at a concentration of 1 mg/mL) was dissolved in 4 mL of PBS and exposed to direct light at various time intervals from 1 to 72 h. This procedure aimed to compare the photostability of Cur enclosed in PAMAM dendrimer with free Cur by measuring the degradation by-products and reduction in absorption peaks of Cur and PAMAM-Cur. Sample measurements were performed using HPLC to calculate the remaining amount in each sample within 72 h [[59]33]. Cell viability staining Live and dead cells were identified using Calcein AM (C3099, Thermo Fisher, USA) and Propidium Iodide (PI) (DN1005-010, inibio, USA). Cells were incubated with Calcein AM (1 μM) at 37 ℃ for 30 min, followed by three washes with PBS. Subsequently, PI (1 μM) was added and incubated for 10 min, followed by three washes with PBS. Images were captured using a confocal microscope (880, Carl Zeiss AG, Germany), with each image representing a different field of view. The experiments were conducted with three independent samples. Cell viability was calculated using ImageJ software (V1.8.0) [[60]34]. CCK-8 Cells were treated according to the instructions of the CCK-8 kit (C0041, Beyotime, Shanghai, China). Cell viability was assessed using the CCK-8 assay at 48 h post-culturing. For each measurement, 10 μL of CCK-8 detection solution was added, and the cells were then incubated in a CO[2] incubator for 4 h. Subsequently, the absorbance at 450 nm was measured using a microplate reader to determine cell viability [[61]35]. Animal grouping The animal experiment was divided into the following groups: HS group (HS model group), HS + PAMAM group (HS model with intracarotid injection of PAMAM nanoparticles), HS + Cur group (HS model with intracarotid injection of 20 mg/kg Cur), and HS + PAMAM@Cur group (HS model with intracarotid injection of PAMAM@Cur nanoparticles containing 20 mg/kg Cur). In Vitro BBB permeability An in vitro BBB model was established using mouse brain endothelial bEnd.3 cells (CL-0598, Wuhan Punuosi Life Science and Technology Co., Ltd.). The bEnd.3 cells were cultured on transwell membranes with a pore size of 0.4 μm, positioned above Na2 cells. When the cells formed a confluent monolayer, this model was considered a reasonable approximation of the BBB in vitro. An endothelial electrical resistance of at least 200 Ω•cm^2 indicated the stability of the cell layer for subsequent experiments. Samples from the lower chamber were collected at regular intervals, and their fluorescence intensity was measured using a fluorescence spectrophotometer to evaluate the efficiency of PAMAM@Cur crossing the BBB. The bottom chamber was observed using a confocal microscope (Leica, STELLARIS 5, Germany), and ImageJ software was used to analyze the obtained images [[62]36]. In Vivo BBB permeability PAMAM@Cur and Cur were intracarotidly injected into male C57BL/6 mice (9–10 weeks old) in groups of six. Brain fluorescence images were obtained at 2, 4, 8, 12, and 24 h post-injection using an in vivo imaging system (IVIS) (Perkin Elmer). Additionally, brain, heart, liver, spleen, lung, and kidney samples were collected for ex vivo imaging 48 h post-administration. The recorded images were analyzed using Living Image 4.3.1 software [[63]36]. Biochemical parameter analysis The metabolic parameters in the serum of each group of mice were analyzed using the ALT activity assay kit (E1010, Sigma-Aldrich, USA), AST activity assay kit (E1020, Sigma-Aldrich, USA), blood urea nitrogen (BUN) assay kit (60–1100, BioAssay Systems, USA), and creatinine assay kit (CR200, Randox Laboratories Ltd, UK). Specific procedures were performed according to the instructions provided with the respective assay kits. Hematoxylin and Eosin (H&E) staining Hypothalamus tissues of the mice in each group were collected, fixed in 10% neutral formalin, embedded in paraffin for sectioning, and deparaffinized in xylene. The sections were then stained with hematoxylin (C0107, Beyotime, Shanghai, China), rinsed with distilled water, immersed in 95% ethanol, stained with eosin (C0109, Beyotime, Shanghai, China), dehydrated with a gradient of ethanol, cleared in xylene, air-dried, fixed with neutral resin, and observed under an optical microscope [[64]37]. Detection of neuronal apoptosis Hypothalamus slices were first incubated with 10% normal donkey serum and then labeled with a series of antibodies: rabbit anti-mouse neuronal nuclear protein NeuN (ab104224, 1:200, Abcam, UK) and rabbit anti-cleaved caspase-3 (PA5-114,687, 1:100, Invitrogen, USA). Subsequently, the slices were incubated with fluorescently labeled secondary antibodies, including goat anti-mouse IgG H&L (Alexa Fluor® 568) (ab175473, 1:200, Abcam, UK) and goat anti-rabbit IgG H&L (Alexa Fluor® 488) (ab150077, Abcam, UK). Next, nuclear counterstaining was performed with DAPI (D9542, Sigma-Aldrich) to enhance nuclear visualization. In addition, selected sections were stained using the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay kit (#630,108, Clontech, Palo Alto, CA, USA). Images were observed with a confocal microscope (Leica, STELLARIS 5, Germany), and neuronal apoptosis was quantitatively analyzed using ImageJ software [[65]38]. qRT-PCR RNA was extracted from tissues and cells using Trizol (Cat# 16,096,020, Thermo Fisher Scientific, USA). The concentration and purity of RNA were determined using the NanoDrop One/OneC spectrophotometer (Thermo Fisher Scientific) with an A260/A280 ratio of 2.0 and a concentration greater than 5 μg/μL. The cDNA first-strand synthesis kit (Cat# D7168L, Beyotime, Shanghai) was used for RNA reverse transcription. RT-qPCR experiments were performed using the RT-qPCR kit (Cat# Q511-02, Vazyme Biotech, Nanjing) following the manufacturer's instructions. For each reaction, 2 μL of cDNA template, 0.2 μL of each forward and reverse primer, and 10 μL of RT-qPCR Mix were mixed and brought to a final volume of 20 μL with RNase-free water. PCR amplification was carried out using the Bio-Rad CFX96 real-time PCR system with the following cycling conditions: initial denaturation at 95 ℃ for 30 s, followed by denaturation at 95 ℃ for 10 s, annealing at 60 ℃ for 30 s, and extension at 72 ℃ for 30 s, repeated for 40 cycles, with a melt curve analysis from 65 ℃ to 95 ℃. The primer sequences were designed and provided by Shanghai Sangon Biotech and are listed in Table S1. The relative expression of the target gene in the experimental group compared to the control group was calculated using the 2^−ΔΔCt method with GAPDH as the reference gene [[66]39]. The formula used was ΔΔCt = ΔCt(exp)—ΔCt(control), where ΔCt = Ct(target gene)—Ct(reference gene), and the experiment was repeated three times. Western blot Cell extracts for total protein were prepared using RIPA lysis buffer (Cat# P0013B, Beyotime, Shanghai) containing 1% PMSF. Nuclear proteins were extracted using the NE-PER™ Nuclear and Cytoplasmic Extraction Kit (Cat# 78,833, ThermoFisher Scientific). The total protein concentration of each sample was determined using the BCA assay kit (Cat# P0011, Beyotime, Shanghai). SDS-PAGE gels ranging from 8 to 12% were prepared based on the target protein band size, and equal amounts of protein samples were loaded into each lane for electrophoresis using a micropipette. Proteins separated on the gel were transferred to a PVDF membrane (Cat# 1,620,177, BIO-RAD, USA) and then blocked with 5% non-fat milk at room temperature for 1 h. The membrane was probed with primary antibodies: Anti-BNDF (ab108319, 1:1000), Anti-HSP70 (ab2787, 1:1000), Anti-HIF-1α (ab308433, 1:1000), Anti-IL-1β (ab283818, 1:1000), Anti-TNF-α (ab183218, 1:1000), Anti-PCBP2 (ab200835, 1:1000), Anti-SLC7A11 (xCT) (ab307601, 1:1000), Anti-GPX4 (ab125066, 1:1000), Anti-CytoC (ab133504, 1:1000), Anti-Bax (ab32503, 1:1000), Anti-Bcl-2 (ab182858, 1:1000), and anti-GAPDH (ab8245, 1:2000), and then incubated overnight at 4 ℃. The membrane was washed three times for 5 min each with 1 × TBST at room temperature, followed by incubation with HRP-conjugated secondary antibodies: goat anti-rabbit IgG (ab6721, 1:2000) or goat anti-mouse IgG (ab6728, 1:2000) for 1 h at room temperature, all antibodies were purchased from Abcam, UK. After three washes with 1 × TBST buffer at room temperature, the membrane was incubated with ECL reagent (Cat# 1,705,062, Bio-Rad, USA), and the bands were visualized using the Image Quant LAS 4000C gel imaging system (GE, USA). The protein of interest was normalized to GAPDH, and the relative protein expression levels were analyzed using ImageJ software (V1.8.0.112) by comparing the grayscale values of the target band to the reference band, to detect the protein expression levels [[67]40]. Each experiment was performed in triplicate. High-throughput transcriptome sequencing RNA was extracted from the hypothalamus tissues of different groups of mice (HS, n = 3; HS + PAMAM@Cur, n = 3) using Trizol reagent (Thermo Fisher Scientific, catalog no. 16096020, USA). The RNA concentration, purity, and integrity were assessed using the Qubit®2.0 Fluorometer® (Thermo Fisher Scientific, catalog no. [68]Q33238, USA) with the Qubit® RNA Analysis Kit (Shanghai Bogoo Biotechnology Co., Ltd., catalog no. HKR2106-01, Shanghai, China), Nanophotometer (IMPLEN, USA) and Bioanalyzer 2100 system with the RNA Nano 6000 Assay Kit (Agilent, catalog no. 5067–1511, USA), ensuring a RNA concentration greater than 100 ng/μL and a 260/280 ratio between 1.8 and 2.1. A total of 3 μg of RNA from each sample was used as input material for RNA library preparation. Following the manufacturer's instructions, the NEBNext® UltraTM RNA Library Prep Kit (New England Biolabs, catalog no. E7435L, Beijing, China) suitable for the Illumina® platform was used to generate cDNA libraries, which were then evaluated for quality on the Agilent Bioanalyzer 2100 system. The indexed samples were clustered using the TruSeq PE Cluster Kit v3 cBot HS (Illumina, catalog no. PE-401–3001, USA) on the cBot cluster generation system. After cluster generation, library preparation was sequenced on the Illumina HiSeq 550 platform, producing 125 bp/150 bp paired-end reads [[69]41, [70]42]. The quality of the paired-end reads from the raw sequencing data was assessed using FastQC software v0.11.8. Preprocessing of the raw data was performed using Cutadapt software version 1.18 to remove Illumina sequencing adapters and poly(A) tail sequences. Reads with N content exceeding 5% were filtered out using a perl script. Reads with 70% base quality over 20 were extracted using FASTX Toolkit software version 0.0.13. The BBMap software was used for repairing paired-end sequences. Finally, the filtered high-quality read fragments were mapped to the mouse genome using HISAT2 software (version 0.7.12) [[71]43, [72]44]. Proteomic analysis In proteomic analysis, the precipitate was first sonicated on ice in a lysis buffer containing 4% SDS, 0.1 M dithiothreitol (DTT), and 20 mM Tris–HCl. Subsequently, the supernatant was collected by centrifuging the sample at 14,000 g for 10 min at 4 °C. Ice-cold acetone (5 volumes) was added to each sample and allowed to precipitate overnight at -20 °C. The resulting protein precipitate was resuspended and quantified for concentration using the BCA Protein Assay Kit (cat. no. 23225, Thermo Fisher Scientific, USA) following the manufacturer's instructions. Next, FASP technology was employed to perform trypsin digestion of 100 μg of each sample on a 30 kD filter (Millipore, Billerica, USA) according to established protocols. Following peptide collection, drying was carried out using a freeze dryer (Labconco, Kansas City, Missouri, USA). Eight-plex iTRAQ reagents (Applied Biosystem Inc., CA, USA) were used to label 20 μg of each sample from a total of 4 samples per group, after which the labeled samples were combined. The combined samples were then separated using the Pierce High pH Reversed-Phase Peptide Fractionation Kit (Thermo Fisher Scientific, USA). The eluate from each fraction was vacuum-concentrated, reconstituted in 8 μL of 0.1% formic acid, and subsequently analyzed by nanoLC-orbitrap MS, following established protocols. Three replicates were conducted for each sample. The collected MS/MS spectra were converted to MGF files and searched against the SwissProt mouse database using Mascot Daemon. A filtering criterion was set at a 1% false discovery rate to minimize the possibility of erroneous peptide identification. Finally, significantly differentially expressed proteins were determined with a standard of P < 0.05 and |log2FC|> 1.00 [[73]30]. Analysis of differentially expressed genes (DEGs) DEGs were identified using the "limma" package in R with a threshold of |log2FC|> 2 and a P-value < 0.05. The differential gene expression volcano plots were generated using the "ggplot2" package in R [[74]45, [75]46]. GO and KEGG enrichment analysis The DEGs were subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis using the "clusterProfiler," "org.Hs.eg.db," "enrichplot," and "ggplot2" packages in R. Enrichment plots for the biological process (BP), cellular component (CC), and molecular function (MF) categories in GO were generated alongside a bubble plot for KEGG enrichment analysis results [[76]47]. Protein–protein interaction (PPI) analysis Proteins encoded by the relevant DEGs were analyzed for PPI using the String website, filtering out non-interacting proteins to construct the PPI network [[77]48]. The PPI network was imported into Cytoscape software for further analysis using the MCODE plugin. By leveraging the network's topological characteristics, the MCODE plugin identifies highly connected subgraphs within the network, which often represent protein complexes or closely interacting protein groups involved in specific biological processes [[78]49]. Determination of Fe^2+ content The level of Fe^2+ was determined using the iron ion detection assay kit (ab83366, Abcam) following the manufacturer's instructions. Briefly, cellular or tissue samples were weighed, homogenized in the iron ion detection assay kit buffer on ice, and rinsed with cold PBS. Subsequently, the homogenate was centrifuged at 16,000 g for 10 min at 4 °C; 5 μL of the assay kit buffer was added to each supernatant. The mixture was then incubated at 37 °C for 30 min, and the absorbance was measured at 593 nm using a microplate reader to determine the optical density (OD) [[79]50]. GSH content analysis The glutathione (GSH) content in cells/tissues was measured using a GSH fluorescence assay kit (EIAGSHF, Thermo Scientific, USA). Samples or standards were mixed with the assay reagent and incubated at room temperature for 15 min. The fluorescence product was then read at 510 nm in a fluorescence microplate reader with an excitation wavelength of 390 nm. Initially, free GSH was measured after 15 min, followed by the addition of the reaction mixture to convert all oxidized glutathione (GSSG) to free GSH, which reacted with excess assay reagent to generate a signal related to the total GSH content. The total concentration of generated GSH in the samples was calculated based on the signal produced. [[80]51]. ELISA detection of MDA and SOD levels The levels of Malondialdehyde (MDA) and Superoxide Dismutase (SOD) were assessed using the MDA Colorimetric Assay Kit (EEA015, Invitrogen, USA) and the SOD Colorimetric Activity Assay Kit (EIASODC, Invitrogen, USA) following the specific protocols outlined in the respective assay kit manuals [[81]52]. Measurement of mitochondrial membrane potential The changes in mitochondrial membrane potential were evaluated using the mitochondrial staining assay kit with the cationic fluorescent dye JC-1 ([82]M34152, Sigma-Aldrich, St. Louis, MO, USA). Briefly, cells were seeded in a 96-well black plate at a density of 5 × 10^3 cells per well. After 48 h of treatment, the cells were co-incubated with the JC-1 dye for 20 min. To image JC-1 monomers, a live-cell imaging system (BD Pathway™ Bioimager System, BD Biosciences) was utilized with an excitation wavelength of 490 nm and emission wavelength of 530 nm; for J-aggregates, the excitation wavelength was set at 525 nm and emission wavelength at 590 nm [[83]53]. Detection of intracellular reactive oxygen species (ROS) Hypothalamic tissue samples from various groups of mice were treated with 20 μM 2,7-dichlorodihydrofluorescein diacetate (DCFH-DA, C2938, Invitrogen, USA) and then incubated for 1 h. Following the completion of the incubation, the tissue samples were washed three times with PBS to remove any unabsorbed DCFH-DA dye. Subsequently, the samples were exposed to 850 nm light for 20 min to activate the fluorescence of DCF. Prior to imaging, the samples were stained with 1.0 μM Hoechst 33,342 (62,249, Thermo Fisher Scientific) for 10 min to label the cell nuclei. Fluorescence signals generated by 2,7-dichlorofluorescein (DCF) were monitored using a 488 nm excitation laser, while Hoechst 33,342 was excited at 405 nm. All imaging procedures were performed using a confocal microscope (Leica, STELLARIS 5, Germany) [[84]54]. Establishment of HS cell model In vitro experiments were conducted using N2a cells (CL-0168, Wuhan Punose Life Technology Co., Ltd), cultured in RPMI-1640 medium (11,875,093) supplemented with 1% glutamine (25,030,081), 10% fetal bovine serum (10099141C), and 1% penicillin/streptomycin (10,378,016), and incubated at 37 °C in a humidified atmosphere containing 5% CO[2]. The aforementioned reagents were procured from ThermoFisher [[85]55]. For HS treatment, the cells were subjected to a 2-h incubation at 43 °C followed by a 6-h recovery period at 37 °C [[86]56]. Cell infection and grouping N2a cells were manipulated through lentivirus infection for either overexpression or silencing, with lentivirus packaging services provided by Sangon Biotech (Shanghai), China. The pHAGE-puro series plasmid, along with the helper plasmids pSPAX2 and pMD2.G, and the pSuper-retro-puro series plasmid with the helper plasmids gag/pol and VSVG were co-transfected into 293 T cells (CL-0005, Wuhan Punose Life Technology Co., Ltd). After 48 h of cell culture, the supernatant was collected, filtered through a 0.45 μm filter, centrifuged, and the viral supernatant was collected. Subsequently, the supernatant was concentrated by centrifugation after 72 h, the two batches of virus were mixed, and the titer was determined. When the cells were in the logarithmic growth phase, they were trypsinized and seeded into 6-well plates at 1 × 10^5 cells per well. Following 24 h of routine culture, when the cell confluency reached approximately 75%, the culture medium containing an appropriate amount of lentivirus (MOI = 10, working titer around 5 × 10^6 TU/mL) and 5 μg/mL polybrene (Merck, TR-1003, USA) was added to infect the cells. After 4 h of infection, an equal amount of medium was added to dilute the polybrene, and 24 h post-infection, the medium was replaced with a fresh culture medium. To establish stable cell lines, 4 μg/mL puromycin (Sangon Biotech, A100339, Shanghai, China) was used for resistance selection to obtain stable cell lines. The sequences for silencing lentivirus are listed in Table S2. Cell grouping: Control group (control group), HS group (HS model group), HS + PAMAM@Cur (cells treated with PAMAM@Cur before HS induction), HS + PAMAM@Cur + sh-PCBP2 (cells transfected with sh-PCBP2 lentivirus and treated with PAMAM@Cur before HS induction), HS + PAMAM@Cur + sh-PCBP2 + oe-SLC7A11 (cells transfected with sh-PCBP2 + oe-SLC7A11 lentivirus and treated with PAMAM@Cur before HS induction), HS + PAMAM@Cur + sh-PCBP2 + oe-GPX4 (cells transfected with sh-PCBP2 + oe-GPX4 lentivirus and treated with PAMAM@Cur before HS induction). RIP-qPCR Initially, rinse the cells once with cold PBS, then add 2 mL of cold PBS to a 10 cm culture dish for cell collection. Extract the cell nuclei from these cells and proceed with sonication. Bind 2 μg of PCBP2 antibody (ab184962, Abcam) or corresponding rabbit IgG control (ab172730, Abcam) to Protein A/G magnetic beads (88,802, Thermo Fisher Scientific), and incubate the mixture at 4 °C for 4 h. Subsequently, wash the beads three times and then incubate them with pre-cleared nuclear extract in RIP buffer (150 mM KCl, 25 mM Tris (pH 7.4), 5 mM EDTA, 0.5 mM DTT, 0.5% NP-40, 1 × protease inhibitor, 10 U/mL RNase inhibitor) at 4 °C overnight. Following incubation, wash the beads three times with RIP buffer, resuspend the beads in 80 μL of PBS, and perform DNA digestion at 37 °C for 30 min, followed by treatment with 2 μL of proteinase K (25,530,031, 20 mg/mL, Thermo Fisher) at 55 °C for 30 min. Finally, extract RNA from the Input and IP samples using the TRIzol method and analyze the levels of SLC7A11 and GPX4 by quantitative PCR [[87]57]. Experiment on RNA half-life Cells were treated with streptomycin D (129,935, Millipore) at a concentration of 5 mg/ml. After incubation for 0, 0.5, 1.5, and 2 h, cells were collected, and RNA was extracted for RT-qPCR analysis. The rate of RNA degradation was estimated using the formula Nt/N0 = e^−kt, where t represents the time of transcriptional inhibition, and Nt and N0 denote the RNA expression levels at time t and time 0, respectively. The RNA half-life (t[1/2]) was calculated from the degradation rate as follows: t[1/2] = ln[2]/k [[88]58]. Statistical analysis Our study utilized R language version 4.2.1, with R language compilation performed through the integrated development environment RStudio, version 2022.12.0–353. Data was processed using GraphPad Prism 8.0, and continuous data were presented as mean ± standard deviation (Mean ± SD). Unpaired t-tests were used for comparisons between two groups, while one-way analysis of variance was applied for comparisons among multiple groups. The homogeneity of variance was tested using the Levene test. In cases of homogeneity, Dunnett’s T3 and LSD-t tests were used for pairwise comparisons. When variance was not homogeneous, Dunnett’s T3 test was employed. A p-value < 0.05 indicated statistical significance for differences between the two groups [[89]59]. Results Successful establishment of a HS mouse model As extreme high-temperature events become more frequent and intense due to global climate change, HS, a severe heat stress response triggered by high-temperature environments, is on the rise annually, becoming a significant public health concern. HS not only results in the failure of the body's temperature regulation mechanism but also triggers systemic inflammatory responses and multi-organ dysfunction, with notable damage to the central nervous system, particularly the hypothalamus. The hypothalamus plays a crucial role in maintaining temperature stability, water-electrolyte balance, and various endocrine functions [[90]60, [91]61]. Therefore, this study initially established an HS mouse model to investigate the effects of high-temperature exposure (Fig. [92]1A). In the model group, mice were exposed to a high-temperature climate chamber at 42 °C for 1 h and then allowed to recover at room temperature of 24 °C. The rectal temperature of HS model group mice gradually increased, with 8 mice reaching 42 °C within 100 min (Fig. [93]1B). These 8 mice were selected as the HS group for subsequent experiments. Subsequently, the change in body weight before and after modeling was measured for each mouse. It was observed that the body weight of the HS group mice decreased, likely due to dehydration from sweating (Fig. [94]1C). Blood samples were collected from mice in each group, and serum was isolated for biochemical analysis, which revealed that compared to the control group, markers such as ALT, AST, ALP, CREA, UREA, and CK showed significant increases (Fig 1D-I), confirming the successful establishment of the HS mouse model. Fig. 1. [95]Fig. 1 [96]Open in a new tab Construction of an HS mouse model. Note: A Schematic diagram of the construction process of the HS mouse model (mice exposed to high-temperature conditions in a climatic chamber, allowed to recover at room temperature, and blood collected for biochemical analysis); B Monitoring of rectal temperature in mice from each group; C Comparative graph of body weight changes before and after modeling in mice from each group; D-I Evaluation of changes in serum levels of ALT, AST, ALP, CREA, UREA, and CK in mice from each group. * indicates P < 0.05 compared to the Control group, ** indicates P < 0.01, *** indicates P < 0.001, ns indicates no significant change. The Control group comprised 10 mice, while the HS group had 8 mice HS induces hypothalamic nerve injury in mice Following the successful establishment of an HS mouse model, we conducted behavioral tests to observe the effects of HS on the brain (Fig. [97]2A). In the OFT, mice in the HS group exhibited a significant reduction in the time spent in the central zone compared to the control group, indicating a potential decrease in cognitive abilities and induction of anxiety-like behavior (Fig. [98]2B-C). In the NOR test, mice in the HS group showed a notable decrease in the recognition index and the number of entries to the new object compared to the control group (Fig. [99]2D-E). These results suggest that HS leads to increased stress and anxiety-like behavior in mice, accompanied by brain damage. Fig. 2. [100]Fig. 2 [101]Open in a new tab Impact of HS on hypothalamic nerve injury in mice. Note: A Evaluation process of the HS mouse model, including behavioral tests, histological analysis, and biochemical assays; B Tracing path diagram of OFT; C Comparison of total distance traveled and time spent in the central zone in the OFT; D Tracing path diagram of NOR test; E Comparison of exploratory behavior towards a novel object in the NOR test; F Immunofluorescence staining showing c-Fos protein expression in the hypothalamus (scale bar = 50 μm); G Immunofluorescence staining showing NeuN-positive cells in the hypothalamus (scale bar = 25 μm); H-J ELISA detection of TNF-α, IL-1β, and IL-6 in hypothalamic lysates of mice from each group; K Western blot analysis of HSP70, Bax, cyto C, and Bcl2 protein expression in the hypothalamus of mice from each group. * indicates P < 0.05 compared to the Control group, ** indicates P < 0.01, *** indicates P < 0.001. The Control group consisted of 10 mice, while the HS group had 8 mice Based on the aforementioned observations and experimental findings, further research focused on elucidating how HS specifically affects the structure and function of the hypothalamus. Through immunofluorescence analysis, we found an increase in c-Fos expression in the hypothalamus of HS mice, indicating stress response and activation of hypothalamic neurons in response to high-temperature exposure (Fig. [102]2F). Furthermore, the decrease in NeuN-positive cells suggests damage to hypothalamic neurons due to HS (Fig. [103]2G). Examination of inflammatory markers TNF-α, IL-1β, and IL-6 levels in hypothalamic lysates revealed a 1.4-fold increase in TNF-α, a 1.2-fold increase in IL-1β, and a 1.3-fold increase in IL-6 levels post HS, further confirming the inflammatory response triggered by HS (Fig. [104]2H-J). To ascertain the heat stress-related biological responses in the hypothalamus, Western blot analysis was performed to assess the expression changes of heat shock protein (HSP70), apoptosis-related proteins Bax and Bcl-2, and mitochondrial functional protein cytochrome C. The results indicated an increase in the expression of HSP70, Bax, and cytochrome C, along with a decrease in Bcl-2 expression in the hypothalamus post-HS, highlighting the stress response, apoptosis, and mitochondrial dysfunction caused by HS (Fig. [105]2K). In conclusion, this study successfully established an HS mouse model and uncovered that HS leads to hypothalamic neuronal damage, heightened inflammatory response, and apoptotic reactions. Preparation and characterization of PAMAM nanoparticles loaded with Cur After identifying the specific impacts of HS-induced hypothalamus damage, this study proceeded to evaluate potential intervention strategies. Considering the known anti-inflammatory and antioxidant benefits of Cur, as well as its demonstrated protective effects in other neural injury models (such as Alzheimer's disease, Parkinson's, ischemic brain injury, spinal cord injury) [[106]62–[107]65], supporting its potential use as a therapeutic strategy for neurological disorders, including hypothalamus damage caused by HS. However, due to the limited solubility and bioavailability of Cur, this study employed PAMAM nanoparticles as carriers, aiming to enhance its therapeutic efficiency by improving its physicochemical properties. The excellent biocompatibility and surface modifiability of PAMAM nanoparticles are expected to enhance the stability and efficacy of Cur, facilitating its targeted delivery to the hypothalamus to more effectively alleviate neural damage induced by HS. Initially, Cur was encapsulated into the cavities of PAMAM (4th generation) dendrimer via hydrophobic interactions in a 1:1 mass ratio, forming PAMAM@Cur (Fig. [108]3A). UV–visible absorption spectroscopy revealed a significant absorption peak at 420 nm and a shoulder peak at about 370 nm, indicating enhanced solubility and stability of Cur within the dendritic structure, resulting in a redshift in absorbance (Fig. [109]3B). DLS results showed average hydrodynamic diameters of 24.5 nm for PAMAM and 90.2 nm for PAMAM@Cur (Fig. [110]3C), with zeta potentials of + 6.6 mV and + 2.1 mV, respectively (Fig. [111]3D). TEM observation depicted spherical Cur-DA nanoparticles with an average diameter of 83.5 ± 18.6 nm, consistent with the DLS results (Fig. [112]3E). In PBS, an in vitro release curve of Cur from PAMAM@Cur at physiological pH 7.4 over 24 h showed approximately 48% release within 24 h (Fig. [113]3F). Further evaluation of the photostability of PAMAM@Cur revealed that the absorption of free Cur dissolved in PBS decreased gradually from 100 to 15% after 72 h, while PAMAM@Cur only decreased by 20%, indicating higher photostability (Fig. [114]3G). Fig. 3. [115]Fig. 3 [116]Open in a new tab Preparation and characterization of PAMAM nanoparticles loaded with Cur. Note: A Schematic illustration of Cur encapsulated into the cavities of PAMAM (4th generation) dendrimers through hydrophobic interactions to form PAMAM@Cur complexes; B UV–Vis absorption spectra showing characteristic absorption peaks of encapsulated Cur and potential changes in solubility and stability; C DLS data indicating the average hydrodynamic diameters of PAMAM and PAMAM@Cur nanoparticles; D Zeta potential measurement of PAMAM and PAMAM@Cur nanoparticles; E TEM images displaying the spherical morphology and average diameter of nanoparticles (scale bar = 100 nm); F In vitro release curve of Cur from PAMAM@Cur in PBS over 24 h; G Comparative evaluation of photo-stability of free Cur and PAMAM@Cur in PBS over 72 h; H CCK-8 assay showing the cytotoxicity of different concentrations of Cur and PAMAM@Cur after 24 h of treatment on Na2 cells; I Live/dead cell staining images after treatment with 50 mg/mL Cur and PAMAM@Cur for 24 h, demonstrating cell viability (scale bar = 25 μm). Cell experiments were repeated three times. * indicates P < 0.05 compared to the PAMAM group, ** indicates P < 0.01; # indicates P < 0.05 compared to the Cur group, ### indicates P < 0.001 Furthermore, we evaluated the cytotoxicity of PAMAM@Cur in Na2 cells using the CCK-8 assay and live/dead cell staining. The results demonstrated that after 24 h of cultivation with varying concentrations of Cur and PAMAM@Cur, cell viability in the Cur group decreased as the concentration increased to 20 mg/mL, reaching around 80% at 100 mg/mL. In contrast, in the PAMAM@Cur group, possibly due to its good sustained-release properties, cell viability remained above 90% throughout (Fig. [117]3H). Treatment with a concentration of 50 mg/mL for 24 h and observation using live/dead cell staining showed that the cell survival rate was approximately 90% in the Cur group and around 96% in the PAMAM@Cur group (F[118]ig. [119]3I), indicating excellent cell compatibility of PAMAM@Cur. In summary, by utilizing PAMAM nanoparticles, we improved the physicochemical properties of Cur. The in vitro experimental results demonstrated that PAMAM@Cur exhibits good cell compatibility and enhanced photostability. This supports the potential of PAMAM@Cur in the treatment of hypothalamus damage induced by HS. The ability of PAMAM@Cur to penetrate the BBB both in Vitro and in Vivo To investigate the role of PAMAM@Cur in hypothalamic damage induced by HS, we initially validated the penetration ability of PAMAM@Cur through the BBB both in vitro and in vivo (Fig. [120]4A). In the in vitro experiments, a monolayer model of brain microvascular endothelial cells (bEnd.3 cells) mimicking the BBB was established to assess the penetration ability of PAMAM@Cur. Cy5.5 fluorescent-labeled PAMAM@Cur was introduced into the upper chamber of the model, samples from the lower chamber were collected at timed intervals, and the fluorescence intensity was measured using a fluorescence spectrophotometer to evaluate the efficiency of PAMAM@Cur crossing the BBB. The results indicated that compared to the standalone Cur group, the fluorescence intensity in the lower chamber of the in vitro BBB model significantly increased with PAMAM@Cur, demonstrating successful penetration of the simulated BBB (Fig. [121]4B). Immunofluorescence staining of Na2 cells also confirmed the successful penetration of PAMAM@Cur through the simulated BBB (Fig. [122]4C). Fig. 4. [123]Fig. 4 [124]Open in a new tab Evaluation of the ability of PAMAM@Cur to penetrate the BBB in Vitro and in Vivo. Note: A Schematic representation of the in vitro and in vivo experimental procedures for investigating the penetration of PAMAM@Cur across the BBB; B Statistical analysis of the fluorescence intensity of Cy5.5-labeled PAMAM@Cur in an in vitro BBB model; C Assessment of the effectiveness of PAMAM@Cur crossing the simulated BBB using immunofluorescence staining technique (scale bar = 25 μm); D-E Time-dependent fluorescence images of Cy5.5-labeled PAMAM@Cur distribution in the mouse brain recorded by IVIS imaging. Each group consisted of 6 mice. * indicates P < 0.05 compared to the Cur group, ** indicates P < 0.01, *** indicates P < 0.001 In the in vivo experiments, C57BL/6 mice were intravenously injected with Cy5.5-labeled PAMAM@Cur via the carotid artery, and the fluorescence signal in the brain was observed and recorded at different time points using an IVIS to evaluate the BBB penetration ability and distribution of PAMAM@Cur in the brain. The results showed a significant enhancement of the fluorescence signal in the brains of mice injected with PAMAM@Cur compared to the Cur group, particularly within 2 to 24 h post-injection, with the fluorescence signal concentrating in specific brain regions and gradually diminishing thereafter (Fig. [125]4D-E). Additionally, H&E staining of mouse heart, liver, spleen, lungs, and kidneys revealed no notable morphological changes in the treated tissues (Figure S1A), indicating that PAMAM@Cur nanoparticles did not induce significant tissue damage. Furthermore, the biochemical parameters such as ALT, AST, BUN, and CR in the serum of treated mice remained within normal levels, showing no significant differences compared to the normal control group (Figure S1B-E), demonstrating excellent biocompatibility and safety. These series of experiments demonstrate that PAMAM@Cur effectively crosses a simulated BBB in vitro and accumulates in the brain in vivo, providing experimental evidence for the potential of PAMAM@Cur as a drug delivery system for treating brain disorders. PAMAM@Cur improves hypothalamic damage caused by HS Following the confirmation of PAMAM@Cur's effective penetration of the BBB, our study aimed to assess the therapeutic effects of PAMAM@Cur on HS-induced hypothalamic damage. Experimental animals were divided into four groups: HS group, HS + PAMAM group, HS + Cur group, and HS + PAMAM@Cur group. Results from the NOR test revealed that compared to the HS and HS + PAMAM groups, mice in the HS + PAMAM@Cur group exhibited significantly enhanced ability to recognize a new object, suggesting a marked improvement in cognitive function. In contrast, although mice in the HS + Cur group showed behavioral improvements compared to the HS model group, indicating some degree of improvement, the limited ability of Cur to penetrate the BBB resulted in an inferior outcome compared to the HS + PAMAM@Cur group (Fig. [126]5A-C). In the OFT, mice in the HS + PAMAM@Cur group displayed a significant increase in exploration activity in the central area, reflecting a marked reduction in anxiety behavior. While the HS + Cur group also showed an increase in central area activity, the extent of improvement was not as significant as that in the HS + PAMAM@Cur group (Fig. [127]5D-F). Fig. 5. [128]Fig. 5 [129]Open in a new tab The therapeutic effect of PAMAM@Cur on hypothalamic damage induced by HS. Note: A Tracing path diagram of NOR test; B-C Comparison of exploratory behavior towards a novel object in the NOR test; D Tracing path diagram of OFT; E–F Comparison of total distance traveled and time spent in the central zone in the OFT; G Histopathological changes observed in hypothalamic tissue sections stained with H&E (scale bar = 50 μm); H Immunofluorescence double staining of neuronal-specific nuclear antigen (NeuN) and apoptosis-executing protein (cleaved-caspase-3) in hypothalamic tissue sections (scale bar = 25 μm); I Immunofluorescence double staining of NeuN and TUNEL in hypothalamic tissue sections (scale bar = 100, 50, 25 μm); J-K Western blot analysis of the protein expression of BDNF, HSP70, HIF-1α, IL-1β, and TNF-α in the hypothalamus of mice from each group. * indicates P < 0.05 compared to the HS group, ** indicates P < 0.01; # indicates P < 0.05 compared to the HS + Cur group, ## indicates P < 0.01. Each group comprised 6 mice Observation of hypothalamic tissue slices stained with H&E revealed that treatment with PAMAM@Cur significantly improved the structural integrity of nerve cells, and reduced inflammation and cell death, surpassing the outcomes of the HS group and HS + PAMAM group. Although the HS + Cur group showed neuroprotective effects and reduced inflammation, limitations in drug delivery efficiency resulted in inferior outcomes compared to the HS + PAMAM@Cur group (Fig. [130]5G). Further support for these findings was provided by immunofluorescence labeling, showing a significant decrease in c-Fos expression and a significant increase in NeuN-positive neurons in the HS + PAMAM@Cur group compared to the HS group, indicating a reduction in neural stress response and an increase in neuronal survival rate (Figure S2A-B). To investigate the role of HS + PAMAM@Cur in HS-induced hypothalamic neuronal apoptosis, we conducted immunofluorescent double staining on hypothalamic slices. The results revealed that compared to the HS group and HS + PAMAM group, the HS + Cur and HS + PAMAM@Cur groups exhibited significantly reduced NeuN + cleaved-caspase3 + and NeuN + TUNEL + positive cells, indicating a significant reduction in HS-induced hypothalamic neuronal apoptosis with both PAMAM@Cur and Cur treatments, with the efficacy of PAMAM@Cur still superior to Cur (Fig. 5H-I). Additionally, Western blot analysis unveiled a significant increase in BDNF, HSP70, and HIF-1α expression, along with a notable decrease in IL-1β and TNF-α expression in the HS + PAMAM@Cur group, reflecting significant neuroprotection and anti-inflammatory effects (Fig. [131]5J-K). In conclusion, PAMAM@Cur, by enhancing the BBB penetration capability and bioavailability of Cur, significantly ameliorated hypothalamic nerve injury in the HS model. This improvement encompasses enhanced cognitive function, reduced anxiety behavior, protection of neural cell structure, alleviation of neural stress response, and mitigation of inflammatory reactions. In contrast, while standalone Cur treatment exhibited some therapeutic effects, its limited BBB penetration capability resulted in outcomes inferior to the significant effects of PAMAM@Cur. Multi-omics analysis reveals the therapeutic mechanism of PAMAM@Cur in treating HS by regulating antioxidant defense genes and ferroptosis-related genes To delve deeper into the molecular mechanisms of PAMAM@Cur in treating hypothalamic nerve injury caused by HS, we systematically analyzed the transcriptome and proteome of hypothalamic tissues from mice in the HS group and the HS + PAMAM@Cur group. Transcriptomic data revealed that after PAMAM@Cur treatment, the expression levels of 47 genes were upregulated, while 72 genes were downregulated (Fig. [132]6A). Heatmap analysis further demonstrated a consistent regulatory pattern of these genes' expression in the HS + PAMAM@Cur group, highlighting the specific gene expression changes induced by the treatment (Fig. [133]6B). KEGG pathway enrichment analysis indicated that these DEGs underwent significant changes in biological pathways closely related to neuronal damage and repair, such as ferroptosis, oxidative stress response, and central nervous system myelination (Fig. [134]6C). Furthermore, GO analysis emphasized the roles of these genes in biological processes such as oxidative stress response, and neuronal development, as well as in molecular functions related to structural composition and receptor activity (Fig. [135]6D). By conducting protein interaction analysis using the STRING database and the MCODE plugin in Cytoscape software on 119 DEGs, we found that Cluster 1 mainly comprised genes associated with neuronal signal transduction and protein stability (Fig. [136]6E), while Cluster 2 focused on genes closely linked to ferroptosis, further confirming the crucial role of ferroptosis in hypothalamic nerve injury induced by HS (Fig. [137]6F). Fig. 6. [138]Fig. 6 [139]Open in a new tab Analysis of transcriptome sequencing results. Note: A Volcano plot of gene expression in transcriptome sequencing, where the x-axis (logFC) represents the logarithm of fold change in gene expression, and the y-axis (-log10(p-value)) represents the statistical significance. The colors of data points indicate the direction of gene expression changes: red for upregulated genes, green for downregulated genes, and black for genes with insignificant changes; B Heatmap illustrating differential gene expression, where each row represents a gene and each column represents a sample. The color intensity reflects the level of gene expression: blue for decreased expression and yellow for increased expression; C Display of KEGG enrichment analysis results for DEGs, depicting gene-pathway associations through lines. Line colors represent various diseases or biological processes, while the thickness of lines and the size of circles next to genes indicate the extent of gene expression changes; D Results of GO enrichment analysis for DEGs, showcasing the biological functions (BP: Biological Process), cellular components (CC: Cellular Component), and molecular functions (MF: Molecular Function) affected by gene expression changes. Each point represents an enriched GO term, where point size denotes the number of genes (Count), color depth (q-value) signifies statistical significance, and the x-axis (GeneRatio) indicates the proportion of significant genes in a specific GO term; E–F Analysis of DEGs in the PPI network from the STRING database using the MCODE plugin in Cytoscape software. The clusters in these graphs represent highly interactive protein groups, where proteins in each cluster may collectively participate in the same biological process. HS n = 3, HS + PAMAM@Cur n = 3 Principal component analysis of the proteomic data revealed a significant overall difference in protein profiles between the HS group and the HS + PAMAM@Cur group (Fig. [140]7A), with consistent expression patterns of proteins of different molecular weights across the two groups (Fig. [141]7B). Differential protein analysis identified 18 upregulated and 11 downregulated proteins (Table S3). This was further corroborated through heatmap analysis, demonstrating that PAMAM@Cur significantly influenced the expression levels of key proteins, including GPX4 and SLC7A11, which play crucial roles in antioxidant defense, iron metabolism, and ferroptosis processes (Fig. [142]7C). The enrichment results of KEGG pathways for differential proteins showed significant enrichment of multiple biological pathways related to ferroptosis, particularly the ferroptosis pathway itself, further confirming our findings from transcriptomic analysis and protein expression heatmaps (Fig. [143]7D). GO enrichment analysis revealed significant enrichment of biological processes closely related to oxidative stress defense, electron transport chain, and GSH metabolism (Fig. [144]7E). The significance of these biological processes emphasizes the potential role of PAMAM@Cur in promoting redox balance and antioxidant stress response. Fig. 7. [145]Fig. 7 [146]Open in a new tab Proteomic analysis results. Note: A Principal component analysis illustrating the changes in protein profiles between the HS group and the HS + PAMAM@Cur group; B Box plot presenting reliable protein expression in the HS and HS + PAMAM@Cur groups; C Heatmap displaying differential protein expression, where each row represents a protein and each column represents a sample. Color variations indicate relative changes in protein expression levels, with red denoting increased expression and blue denoting decreased expression; D Enrichment analysis of KEGG pathways for differential proteins, where each point in the graph represents a specific KEGG pathway. The color of points reflects the significance of enrichment (p-value), with smaller p-values represented by a redder color, indicating greater statistical significance. The size of points indicates the number of differentially enriched proteins in that pathway. A higher enrichment score on the x-axis suggests more significant enrichment; E GO enrichment analysis results for differential proteins, encompassing biological processes (in green), cellular components (in orange), and molecular functions (in blue). The bar lengths in the graph indicate the significance of enrichment for each GO term, with higher -log10 p-values signifying greater statistical significance; F-G Analysis results of interactions between differential proteins using the MCODE plugin in Cytoscape software based on the STRING protein interaction data. The clusters in these graphs represent highly interacting protein groups, where proteins within each cluster may jointly participate in the same biological processes. HS n = 3, HS + PAMAM@Cur n = 3 Furthermore, analysis using the MCODE module in Cytoscape unveiled that Cluster 1 was associated with signal transduction and protein stability (Fig. [147]7F), while Cluster 2 pointed to key genes involved in metabolic processes (Fig. [148]7G), aligning with the previous analytical results. Notably, the identification of Pcbp2, Slc7a11, and GPX4 not only confirmed findings from transcriptomic and proteomic data (Figure S3A-C) but also underscored their potential importance in regulating oxidative stress response and ferroptosis. Overall, integrating the multi-omics data, our study indicates that PAMAM@Cur may reduce ferroptosis and promote the repair of hypothalamic nerve injury induced by HS by modulating genes associated with antioxidant defense and neuroprotection. PAMAM@Cur ameliorates hypothalamic nerve injury induced by HS through mitigation of ferroptosis Through multi-omics analysis, we have identified that PAMAM@Cur may improve hypothalamic nerve injury induced by HS by modulating ferroptosis. To further confirm the role of iron accumulation and oxidative stress in hypothalamic damage caused by HS, Prussian Blue staining and assessment of oxidative stress markers (SOD activity, MDA, and GSH levels) were conducted. The results revealed that compared to the Control group, the HS group exhibited elevated levels of Fe^2+ accumulation and MDA, along with decreased SOD and GSH levels. In contrast to both the HS and HS + PAMAM groups, the HS + PAMAM@Cur group demonstrated significant reductions in Fe^2+ accumulation and MDA levels, along with increased SOD activity and GSH content, indicating the effective inhibition of ferroptosis and oxidative stress by PAMAM@Cur (Fig. [149]8A-E). Additionally, Western blot analysis of the expression levels of PCBP2, SLC7A11, and GPX4 in the hypothalamus of mice across different groups revealed a significant increase in the protein expression levels of these markers in the HS + PAMAM@Cur group compared to the HS and HS + PAMAM groups, while the HS + Cur group showed only modest changes (Fig. [150]8F). Fig. 8. [151]Fig. 8 [152]Open in a new tab The impact of PAMAM@Cur on ferroptosis regulation in hypothalamus nerve injury induced by HS. Note: A Prussian blue staining observing the iron ion deposition in the hypothalamus of mice (scale bar = 50 μm); B Detection of Fe^2+ content in the hypothalamus tissue of mice using an assay kit; C-E Measurement of SOD activity, MDA levels, and GSH content in the hypothalamus tissue using assay kits; F Expression levels of PCBP2, SLC7A11, and GPX4 proteins in the hypothalamus of mice in different experimental groups assessed by Western blot; G-H Evaluation of mitochondrial membrane potential changes using JC-1 fluorescent staining (scale bar = 25 μm); -JI Detection of intracellular ROS levels using DCFDA fluorescent probe (scale bar = 25 μm).* indicates P < 0.05 compared to the HS group, ** indicates P < 0.01; # indicates P < 0.05 compared to the HS + Cur group, ## indicates P < 0.01. Six mice per group Mitochondrial dysfunction, a critical link in the ferroptosis pathway, is one of the direct causes of cell death induced by iron overload [[153]66]. Therefore, measurements of mitochondrial membrane potential and ROS levels were performed to assess mitochondrial health and function directly. Evaluation of mitochondrial membrane potential using JC-1 dye and detection of intracellular ROS levels with the DCFDA fluorescent probe revealed that compared to the Control group, mice in the HS group exhibited decreased mitochondrial membrane potential and significantly increased ROS production, indicating compromised mitochondrial function and exacerbated oxidative stress under HS conditions. However, in the HS + PAMAM@Cur group, the decrease in mitochondrial membrane potential was less pronounced, and ROS production was significantly reduced. These changes, in comparison to the HS group and HS + PAMAM group, suggest that PAMAM@Cur exerts a protective effect on mitochondrial function, effectively alleviating HS-induced mitochondrial damage and oxidative stress (Fig. [154]8G-J). Through a comprehensive analysis combining iron levels, oxidative stress markers, and mitochondrial function, we further confirmed that PAMAM@Cur mitigates hypothalamic nerve injury induced by HS by reducing ferroptosis. PAMAM@Cur upregulates PCBP2 to stabilize SLC7A11 and GPX4 and reduce ferroptosis In a previous proteomic analysis, we observed a significant upregulation of PCBP2 expression following treatment with PAMAM@Cur, a finding that captured our attention. PCBP2, an RNA-binding protein, has the potential to regulate the expression of specific proteins through interactions with mRNA [[155]67]. Based on this hypothesis, we focused on the potential targets of PCBP2, namely SLC7A11 and GPX4, both closely associated with the process of ferroptosis. To investigate the relationship between PCBP2 and these proteins, we conducted RIP experiments, confirming a direct interaction between PCBP2 and the mRNA of SLC7A11 and GPX4 (Fig. [156]9A). Furthermore, data from Starbase-CLIP supported this finding, demonstrating that PCBP2 can bind to both mRNAs (Figure S4A-B). Following PCBP2 knockdown and overexpression in the Na2 cell model, the most efficient sh-PCBP2-1 sequence was selected for further studies (Figure S4C-D). Subsequent to the PCBP2 knockdown, mRNA levels of SLC7A11 and GPX4 significantly decreased (Fig. [157]9B). Treatment with actinomycin D revealed that in cells where PCBP2 was silenced, the half-life of SLC7A11 and GPX4 mRNA significantly shortened, indicating an impact on mRNA stability (Fig. [158]9C-D); conversely, in cells with PCBP2 overexpression, the half-life of these mRNAs extended significantly, suggesting that overexpression of PCBP2 enhanced the stability of SLC7A11 and GPX4 mRNA (Figure S4E-F). These experiments indicate that PCBP2 may enhance the stability of SLC7A11 and GPX4 mRNA by directly binding to them, thereby enhancing cellular regulation of iron ions and reducing ferroptotic cell death. Fig. 9. [159]Fig. 9 [160]Open in a new tab The influence of PAMAM@Cur on regulating PCBP2 stability to stabilize SLC7A11 and GPX4 in HS-induced ferroptosis. Note: A RIP experiment detecting the interaction between PCBP2 and SLC7A11 and GPX4 mRNA; B Detection of SLC7A11 and GPX4 mRNA levels after PCBP2 knockout; C-D Assessment of the effect of PCBP2 knockdown on the mRNA half-life of SLC7A11 and GPX4 following actinomycin D treatment; E Examination of the protein expression levels of PCBP2, SLC7A11, and GPX4 in cells from different experimental groups; F-I Utilization of assay kits to evaluate intracellular Fe^2+ accumulation, as well as changes in SOD, MDA, and GSH levels; J Evaluation of cell viability through CCK-8 experiments. * indicates P < 0.05 compared to the Control group, ** indicates P < 0.01; # indicates P < 0.05 compared to the HS group, ## indicates P < 0.01; & indicates P < 0.05 compared to the HS + PAMAM@Cur group, && indicates P < 0.01; @ indicates P < 0.05 compared to the HS + PAMAM@Cur + sh-PCBP2 group, @@ indicates P < 0.01. Cell experiments were repeated three times To determine whether PAMAM@Cur can stabilize SLC7A11 expression through PCBP2 and consequently reduce ferroptosis, we overexpressed SLC7A11 and GPX4 in PCBP2-knockdown N2a cells to test if the effects of ferroptosis could be reversed, as demonstrated in Figure S4G-H. Initially, the expression of relevant proteins in each group of cells was assessed. The results showed that compared to the Control group, the expression of PCBP2, SLC7A11, and GPX4 decreased in the HS group; in contrast, in the HS + PAMAM@Cur group, the expression of these proteins was upregulated compared to the HS group. When PCBP2 was silenced, the expression of PCBP2, SLC7A11, and GPX4 decreased again. Worth noting, through overexpression of SLC7A11 or GPX4 (HS + PAMAM@Cur + sh-PCBP2 + oe-SLC7A11 and HS + PAMAM@Cur + sh-PCBP2 + oe-GPX4 groups), the levels of these proteins could be partially restored (Fig. [161]9E). Further assessment of intracellular Fe^2+ accumulation, SOD, MDA, and GSH levels using an iron ion detection kit was conducted to evaluate the status of ferroptosis and oxidative stress. The results revealed that in comparison to the Control group, the HS group exhibited significant increases in Fe^2+ accumulation and MDA levels, accompanied by notable decreases in SOD and GSH levels, indicating exacerbated oxidative stress and ferroptosis. Relative to the HS group, the HS + PAMAM@Cur group displayed considerable improvement in the levels of these oxidative stress markers, reflecting the protective effect of the treatment. However, in the PCBP2 knockdown HS + PAMAM@Cur group, significant increases in Fe^2+ accumulation and MDA levels were observed, while SOD and GSH levels decreased significantly, suggesting that inhibition of PCBP2 may attenuate the protective effect of PAMAM@Cur. Notably, in cells overexpressing SLC7A11 and GPX4, there was a reduction in Fe^2+ accumulation, decreased MDA levels, and increased SOD and GSH levels, indicating that overexpression of SLC7A11 and GPX4 could reverse the adverse effects caused by PCBP2 silencing (Fig. 9F-I). These findings suggest that PAMAM@Cur may enhance cellular resistance to ferroptosis by upregulating PCBP2, thereby stabilizing the mRNA of SLC7A11 and GPX4. Subsequently, cell viability was assessed using the CCK-8 assay. The results revealed that compared to the Control group, the HS group exhibited a significant decrease in cell viability. Conversely, the HS + PAMAM@Cur group demonstrated increased cell viability compared to the HS group, while cell viability decreased in the HS + PAMAM@Cur + sh-PCBP2 group. Cell viability was restored or enhanced in the HS + PAMAM@Cur + sh-PCBP2 + oe-SLC7A11 and HS + PAMAM@Cur + sh-PCBP2 + oe-GPX4 groups, indicating a positive impact of SLC7A11 and GPX4 overexpression on reducing ferroptosis and improving cell survival (Fig. [162]9J). In conclusion, our study illustrates that PAMAM@Cur can mitigate HS-induced ferroptosis by upregulating the PCBP2 protein, thereby stabilizing and enhancing the levels of SLC7A11 mRNA effectively. Discussion Previous research has brought significant attention to the application of nanotechnology in drug delivery systems [[163]68–[164]70]. However, studies on nano drug delivery systems for the treatment of HS, a severe heat stress reaction, remain limited. This study introduces a novel PAMAM nanoparticles loaded with Cur (PAMAM@Cur) and successfully demonstrates its significant neuroprotective effects in an HS model. Compared to traditional therapeutics, PAMAM@Cur nanoparticles exhibit improved bioavailability and stability of Cur, showcasing the potential to enhance therapeutic outcomes in HS treatment. The ability of PAMAM@Cur to effectively cross the BBB and target the hypothalamus makes it a promising alternative to existing treatments that often face limitations in drug delivery and efficacy. Traditional Cur, a natural compound with various biological activities such as anti-inflammatory and antioxidant properties [[165]71], faces limitations in drug delivery and low bioavailability [[166]72, [167]73]. Using PAMAM nanoparticles as carriers, this study encapsulates Cur within the nanoparticles, boosting its stability and bioavailability, thereby enhancing its efficacy in HS treatment. Compared to studies solely focusing on Cur, PAMAM@Cur exhibits a more significant therapeutic effect, showcasing the advantages of nanotechnology in drug delivery [[168]33]. Furthermore, the use of green nanomaterials in the formulation of PAMAM@Cur underscores the sustainability and biocompatibility of the approach, addressing environmental concerns associated with synthetic nanomaterials. Green nanomaterials, derived from natural sources, minimize toxicity and adverse effects, making PAMAM@Cur a safer and more eco-friendly option for clinical applications. This study significantly improved the bioavailability and stability of Cur by encapsulating it in PAMAM nanoparticles. This innovation not only demonstrated excellent efficacy in treating hypothalamic neural damage induced by heat shock but also showcased the broad prospects of nanotechnology in drug delivery. The PAMAM@Cur nanoparticles, with their remarkable Blood–brain barrier penetration capability and excellent cell compatibility, offer a novel and effective approach for neuroprotection and anti-inflammatory therapy. Furthermore, the development of this nanodelivery system provides insights for the delivery of other drugs that have difficulty crossing the Blood–brain barrier, thereby expanding the application of nanotechnology in the treatment of neurodegenerative diseases, brain injuries, and other neurological disorders. By utilizing green nanomaterials, this study contributes to the development of sustainable therapeutic strategies, aligning with global efforts to reduce the environmental footprint of medical interventions. Research on HS predominantly focuses on aspects like temperature regulation, inflammatory responses, and neural damage [[169]3, [170]74, [171]75]. This study, through behavioral assessments, histological examinations, and biochemical analyses, reveals the therapeutic effects of PAMAM@Cur on hypothalamus nerve injury in an HS mouse model. The results demonstrate improvements in behavior and reduction of inflammation, offering a comprehensive therapeutic approach for HS treatment. In comparison to previous studies concentrating on singular aspects, this research comprehensively explores the treatment mechanisms of HS, highlighting the potential of PAMAM@Cur as a robust alternative to traditional therapeutics. Moreover, in cellular biology experiments, this study further validates the cell compatibility and penetration capability of PAMAM@Cur in a BBB model. These experimental findings provide deeper support for the effectiveness of PAMAM@Cur in treating HS. Compared to the interaction mechanisms between traditional drugs and cells, nano-drug delivery systems exhibit superior cellular penetration and bioavailability, laying a solid foundation for their clinical applications. Multi-omics analysis plays a crucial role in elucidating disease treatment mechanisms. This study delves deep into the potential mechanisms of PAMAM@Cur in treating hypothalamus injury induced by HS through transcriptomic and proteomic analyses. The results demonstrate that PAMAM@Cur exerts its effects by regulating antioxidant defense genes and ferroptosis-related genes, playing a significant role in alleviating neural damage caused by HS. In comparison to past studies focusing solely on surface phenomena, this research delves more profoundly into the molecular-level regulatory effects of the drug [[172]76–[173]78]. In summary, this study successfully developed PAMAM@Cur nanoparticles and validated their efficacy and mechanism in treating hypothalamus nerve injury induced by HS through a series of in vivo and in vitro experiments (Fig. [174]10). By loading Cur into PAMAM nanoparticles, the bioavailability and stability of Cur were enhanced, allowing it to effectively penetrate the BBB and directly act on the damaged hypothalamus region. The research findings indicate that PAMAM@Cur not only alleviates neural cell damage and reduces inflammation caused by HS but also, by regulating antioxidant defense genes and ferroptosis-related genes, particularly through the upregulation of PCBP2 protein, stabilizing SLC7A11 and GPX4 mRNA, enhancing the cell's regulation of iron ions, and reducing iron-dependent cell death. These discoveries offer new strategies for the treatment of HS and related neural injuries, showcasing the immense potential of nanotechnology in enhancing the efficacy of traditional drugs. Fig. 10. [175]Fig. 10 [176]Open in a new tab Mechanistic investigation of PAMAM nanoparticles loaded with Cur regulating PCBP2-mediated cell ferroptosis in the treatment of hypothalamus nerve injury induced by HS The scientific and clinical significance of this study lies in proposing an innovative treatment strategy for HS, a global public health concern, by utilizing PAMAM nanoparticles loaded with Cur to treat hypothalamus nerve injury induced by HS. Detailed investigations into nanoparticle preparation and drug delivery characteristics confirm the significant efficacy of PAMAM@Cur in the HS model, not only improving behavioral performance and neural cell structure but also reducing inflammation, paving the way for novel directions in HS treatment. The results of this study hold important scientific significance and hold promising potential for clinical applications by providing insights into the mechanisms of neural damage induced by HS and exploring new drug treatment approaches. Nevertheless, this study has certain limitations. For instance, potential biases and constraints in experimental design need further validation in terms of the reproducibility and reliability of the experimental results. Additionally, more in-depth research and exploration are required to investigate the stability, biocompatibility, and feasibility of the clinical application of PAMAM@Cur in long-term treatment. Looking ahead, building upon the outcomes of this study, future research could explore the potential application of PAMAM@Cur in a broader range of disease models, expanding its possibilities in treating other neurological disorders. By integrating high-throughput technologies such as transcriptomics and proteomics, and delving deeper into the therapeutic mechanisms, it is promising to provide new insights and strategies for the development of nanomedicine in the field of neural injury treatment, advancing the application and development of nanotechnology in clinical medicine. Therefore, despite the challenges and outstanding issues, this study brings hope and enlightenment for future research and clinical therapy in related fields. Supplementary Information [177]Additional file1 (JPG 2678 KB)^ (2.6MB, jpg) [178]Additional file2 (JPG 2189 KB)^ (2.1MB, jpg) [179]Additional file3 (JPG 631 KB)^ (630.9KB, jpg) [180]Additional file4 (JPG 1306 KB)^ (1.3MB, jpg) [181]Additional file5 (DOCX 18 KB)^ (17.8KB, docx) Acknowledgements