2.1 Chemicals and materials
All chemicals were obtained from commercial suppliers and used as received without further purification. Zinc nitrate hexahydrate (Zn(NO3)2·6H2O), selenocystine, methanol, 30% H2O2, and tert-Butyl hydroperoxide (TBHP) were purchased from Macklin (Shanghai, China). 2-methylimidazole (2-MIM), polyvinylpyrrolidone (PVP, Mw = 40,000), dopamine hydrochloride, Sodium selenite pentahydrate (Na2SeO3·5H2O), NaOH were purchased from Aladdin (Shanghai, China). 3,3’,5,5’-tetramethyl benzidine (TMB), 2,2′-Azinobis-(3-ethylbenzthiazoline-6-sulphonate) (ABTS), Cell Cycle and Apoptosis Analysis Kit, Annexin V-FITC Apoptosis Detection Kit, 2′,7′-Dichlorofluorescein diacetate (DCFH-DA), Calcein AM, BCA Protein Assay Kit and crystal violet dye were purchased from Beyotime Biotechnology (Shanghai, China). Cell staining buffer, Human VEGF-a, TNF-α, IL-6 and IL-4 ELISA Kit were purchased from Elabscience (Wuhan, China). Chlorpromazine (CPZ), Methyl-β-cyclodextrin (MβCD), chloroquine (CHL), LY294002, MitoSOX Red, Cell Counting Kit-8 (CCK-8), radioimmunoprecipitation assay (RIPA) lysis and cocktail are purchased from MedChemExpress (USA). PC12 cell line (Undifferentiated) and Human Umbilical Vein Endothelial Cells (HUVECs) were supplied by Procell Life Science&Technology (wuhan, China). Sodium dodecyl sulfate-polyacrylamide gels (SDS-PAGE) are purchased from Epizyme Biotech (Shanghai, China). Cell culture plate, Matrigel and transwell chambers (8.0 µm pore size, 24-well) were purchased from Corning (USA). Hematoxylin and Eosin (H&E), Nissl Stain Kit, and TUNEL Apoptosis Assay Kit were purchased from Solarbio Science & Technology (Beijing, China). N- (6-Methoxy-8-quinolyl)-p-toluenesulfonamide) (TSQ) was purchased from enzo life science (USA). Roswell Park Memorial Institute 1640 medium (RPMI-1640), dulbecco’s modified eagle medium (DMEM), fetal bovine serum (FBS), Horse Serum (HS), Phosphate-buffered saline (PBS), and polyvinylidene fluoride (PVDF) membrane were purchased from Thermo Fisher Scientific (USA). The GelMA hydrogel is purchased from QiYue Biology (Xian, Shanxi, China). Endothelial cell medium (ECM) was purchased from Sigma-Aldrich. PE-Cy7 Rat Anti-Mouse CD86, Alexa Fluor 647 Rat Anti-Mouse CD206, Fixation/Permeablization Kit were purchased from BD Pharmingen (USA).
2.2 Synthesis of nanomaterials
2.2.1 Synthesis of ZIF-8: 5 mL of 62.3 mM 2-MIM (25.6 mg) methanol solution AND 5 mL of 16.9 mM Zn(NO3)2·6H2O (25.2 mg) methanol solution were mixed and stirred for 30 min at 37 ℃. The products were centrifuged at 12,000 rpm for 5 min and washed with methanol three times.
2.2.2 Synthesis of SeNPs: Small, medium and large size nano-selenium was prepared by mixing 1 ml 25 mM sodium selenite with 4 ml 25 mM glutathione (containing 200, 20 and 2 mg BSA), respectively. The pH of the solution was adjusted to 7.2 with 1.0 M sodium hydroxide to form red element Se and glutathione oxide (GSSG). The red solution was dialyzed on double distilled water for 96 h, and the water was changed every 24 h to separate GSSG and Nano-Se. The final solution containing nano-selenium and bovine serum albumin was freeze-dried and stored at room temperature. Transmission electron microscopy (TEM) revealed that the size of red element Se is 5–15 nm (small size, designed as SSSe), 20–60 nm(medium size, designed as SSe) and 80–200 nm(large size, designed as Se).
2.2.3 Synthesis of Se@ZIF-8: 10 mg SeNPs and 500 mg PVP were added to 10 mL methanol and stirred for 2 h at room temperature. The PVP-SSe was collected by centrifugation at 100,000 g for 60 min. The obtained PVP-SSSe was added to 5 mL of 62.3 mM 2-MIM (25.6 mg) methanol solution and stirred for 15 min at 37 ℃. Subsequently, 5 mL of 16.9 mM Zn(NO3)2·6H2O (25.2 mg) methanol solution was added to this mixed solution and stirred for another 30 min at 37 ℃. The products were centrifuged at 12,000 rpm for 5 min and washed with methanol three times.
2.2.4 Synthesis of Fer-1@SSe@ZIF-8 (FSZ): 40mg SSe@ZIF-8 and 26.2mg Fer-1 were added to 1 mL DMSO and the mixture stirred continuously at room temperature and dark for 24 h. The products were centrifuged at 12,000 rpm for 5 min and washed with DMSO three times. The resultants were dried at ambient temperature in a vacuum oven overnight.
2.3 Nanomaterials characterization
TEM and energy spectra analysis was observed with JEM-2100 and JEM-F200 TEM (JEOL, Japan). X-ray diffraction (XRD) data was obtained with a XPert Pro diffractometer (Panalytical, Holland).
2.4 Cell Culture
PC12 cells were incubated in RPMI-1640 medium with 15% HS and 5% FBS at 37°C in a 5% CO2 environment. The culture medium was changed every 3 days. HUVECs were incubated with ECM at 37°C in a 5% CO2 environment. The culture medium was changed every 1 days. RAW264.7 cells were incubated with DMEM containing 10% FBS at 37°C in a 5% CO2 environment. The culture medium was changed every 1 days. MSCs were extracted and identified as before[23]. MSCs were incubated with α-MEM containing 10% FBS at 37°C in a 5% CO2 environment. The culture medium was changed every 3 days.
2.5 Intracellular uptake of nanoparticles
To visualize SSe@ZIF-8, coumarin 6 (C6) is mixed with 2-MIM solution along with PVP-Se before adding Zn(NO3)2·6H2O. After similar steps, C6-labeled Se@ZIF-8 (C6-SSe@ZIF-8) is obtained. PC12 cells (5×104 cells per well) were seeded in a 12-well plate and attached overnight. The cells were stained with Hoechst 33342 (blue) for nucleus and LysoTracker (red) for lysosome, and C6-SSe@ZIF-8 (green) was added. At 0, 1, 2, and 4 h, the medium was removed and cleaned 3 times with pre-cooled PBS. real-time living cell imaging were captured by an inverted fluorescence microscope (IX71, OLYMPUS). To further explore the intracellular uptake of SSe@ZIF-8, endocytotic inhibitor CPZ (10 µg/mL), MβCD(5 mM), CHL(20 µM), and LY294002 (100 µM) was used to pretreat cells for 1 h.
2.6 TEM analysis for cellular localization of nanoparticles
PC12 cells (5×104 cells per well) were seeded in a 12-well plate and incubated with 20 µg/mL SSe@ZIF-8 for after adhesion. After 6 h, the cells were collected and washed. After fixation and gradient dehydration, the samples were coated with epoxy resin and kept overnight in an oven at 60 ° C. Ultrathin sections were prepared and stained with uranyl acetate and lead citrate for 15 min, and observed under TEM.
2.7 PH-release drug release
Fer-1 was labeled with Cy5.5 and Cy5.5-Fer-1@SSe@ZIF-8 was prepared for detection of drug release properties. SSe@ZIF-8 (20µg/mL) was dispersed in 10mL solution of different pH (pH = 5.5, 6.5, and 7.4, respectively) at 37℃. At each time point, 1mL of solution was taken and centrifuged at 12,000 rpm for 5 min, and the absorbance of supernatant was measured at 450 nm by a microplate reader (PerkinElmer, EnSight).
2.8 TMB and ABTS free radical scavenging assay
To assess antioxidant activity of nanoparticles, the TMB and ABTS scavenging assays were used. According to the instruments, TMB (80 mM) was added into 1 mL of solution with 100 µM H2O2. The absorbance changes of TMB at 654 nm were monitored. The ABTS•+ was mixed with different concentrations of nanoparticles. The absorbance of ABTS at 734 nm was examined using a a microplate reader (PerkinElmer, EnSight),
2.9 Mitochondrial ROS detection
Mito-Tracker Red probes are used to detect mitochondrial superoxide production. PC12 cells (5×104 cells per well) were seeded in a 35 mm confocal dish and attached. After 24 h stimulation with 200 µM H2O2, the cells were incubated with Mito-Tracker Red for 10 min at 37℃ and stained with Hoechst 33342. Intracellular fluorescence was observed by a confocal laser scanning microscopy (CLSM, FV1200, OLYMPUS).
2.10 Mitochondrial membrane potential detection
PC12 cells were treated with TBHP and FSZ NPs for 24 h. After the culture-medium was removed, the cells were gently washed with PBS, and 1 ml JC-1 staining solution (Beyotime, Shanghai, China) was added to each well and incubated ina cell incubator at 37°C for 15 minutes. Wash gently with washing buffer 2 times and add fresh medium. Fluorescence microscopy (Leica DM3000 LED) measures fluorescence intensity.
2.11 Cell viability assay
The cytotoxicity of SSe, ZIF-8 and SSe@ZIF-8 was measured using CCK-8. PC12 cells (5×103 cells per well) were seeded in a 96-well plate and adhered overnight. Different concentrations of SSe, ZIF-8 and SSe@ZIF-8 were added to the medium and the cells were cultured for 24 h. After incubation with freshly prepared CCK-8 solution at 37 ℃ for 2 h, the absorbance was measured at 450 nm by a microplate reader (PerkinElmer, EnSight).
2.12 Cell cycle analysis
The effect and recovery ability of SSe@ZIF-8 on PC12 cell cycle were tested by flow cytometry. With or without TBHP (100 µM) treatment, cells were incubated with different concentrations of SSe@ZIF-8 for 24 h. The cells were fixed overnight with 75% ethanol solution at 4℃, incubated with propidiumiodide (PI) and ribonuclease (RNase) at 37℃ for 30 min, and then analyzed by flow cytometry (CytoFlex, Beckman Coulter).
2.13 Intracellular ROS detection
Intracellular ROS levels were evaluated using DCFH-DA probes. MSCs (5×104 cells per well) were seeded in a 12-well plate and attached. SSe (20 µg/mL), ZIF-8 (20 µg/mL), Fer-1 (0.1 µM), SSe@ZIF-8 (40 µg/mL) and FSZ (40 µg/mL) were added according to the groups, and the cells were incubated with TBHP (100 µM) for 24 h. Then cells were incubated DCFH-DA (10 µM) at 37 ℃ for 30 min and observed by a CLSM (FV1200, OLYMPUS). The ratio of average fluorescence intensity of ROS-stained cells was measured in each field by ImageJ.
2.14 Cell apoptosis analysis
Apoptosis was evaluated by Annexin V/PI staining of flow cytometry. Under the stimulation of TBHP (100 µM), PC-12 cells were incubated with SSe (20 µg/mL), ZIF-8 (20 µg/mL), Fer-1 (0.1 µM), SSe@ZIF-8 (40 µg/mL), and FSZ (40 µg/mL) according to the groups. After 24 h, cells were stained with Annexin V FITC and (PI), and analyzed using a flow cytometer (CytoFlex, Beckman Coulter). In post-analysis, cell apoptosis was determined by the following rules (active cells: annexin V‐ and PI‐; early apoptotic cells: annexin V + and PI‐; late apoptotic cells: annexin V + and PI+; dead cells: annexin V‐ and PI+).
2.15 Measurement of neurite growth
The direct effects of nanomaterials on synaptic growth in PC12 cells and the correlation with dose or time were explored. PC12 cells were treated with different concentrations (0, 5, 10, 20, 40, and 80 µg/mL) of SSe@ZIF-8 and cultured in ordinary medium. After 6 days, the cells were observed under a optical microscope. Next, PC12 cells with the same treatment were cultured in DMEM with 10 ng/mL NGF. At 2, 4, and 6 d, the PC12 cells were stained with Calcein AM (1 µM) for 30 min at 37℃ in the dark and then replaced with fresh medium for another 30 min. The morphology of PC12 cells was observed and the number and length of neurites were analyzed under a fluorescence microscope (BX53, OLYMPUS). In growth ratio analysis, differentiated PC12 cells are divided into L0-L6 according to the ratio of their longest neurite length to the diameter of the cell body. For example, L0 is defined as cells whose longest neurite length is shorter than the diameter of the cell body; L1 is defined as cells whose longest neurite length is between the original and twice the diameter of the cell body.
2.16 Transcriptome analysis
The detailed steps were shown in our previous study. In short, 1 x 107 MSCs were incubated in B-27 Plus Neuronal Culture System (20 ng/ml EGF and 20 ng/ml bFGF), treated with 40 µg/mL FSZ for 6 days. Total RNA was extracted and qualified. Strand RNA sequencing libraries were prepared. PCR products corresponding to 200–500 bps were enriched, quantified and sequenced. RNA-Seq data analysis Raw sequencing data were first filtered by Trimmomatic (version 0.36). Limma packets were used to identify genes that were differentially expressed between groups. A p-value cutoff of 0.05 and a fold-change cutoff of 1.5 were used to judge the statistical significance of differenced gene expressions (DEGs). Gene ontology (GO) analysis and Kyoto encyclopedia of genes and genomes (KEGG) enrichment analysis for DEGs were both implemented by DAVID (https://david.ncifcrf.gov) with a p-value cutoff of 0.05 to judge statistically significant enrichment.
2.17 Western blot analysis
After transfection with WNT4-siRNA or control siRNA (the sequences was shown in Table S1), PC12 cells were treated with a series of concentrations of FSZ and differentiated for 6 days. Raw264.7 cells are stimulated by LPS (1 mg/mL) and incubated with Fer-1 (0.1 µM), SSe@ZIF-8 (40 µg/mL), and FSZ (40 µg/mL) for 24 h. Proteins were extracted with RIPA lysis containing cocktail and their concentrations were detected by BCA. These protein samples were separated by SDS-PAGE electrophoresis and transferred to PVDF membranes. After blocking, the membranes were incubated overnight at 4℃ with primary antibody including anti-Nestin (Santa Cruz, sc-23927 trail size, 1:200), anti-TUJ1 (abcam, ab105389, 1:5000), anti-VEGF-a (abcam, ab46154, 1:5000), anti-GAPDH (Servicebio, GB15002, 1:10,000), anti-iNOS (CST, 10320, 1:2000), anti-Arg1 (abcam, ab239731, 1:5000), anti-TNFα (CST, 11948, 1:10,000), anti-IL10 (Cell Signaling Technology, 12163, 1:5000), Axin2 (Proteintech, 20540-1-AP, 1:2000), Active β-catenin (Cell Signaling Technology, 8814S, 1:2000), β-catenin (Servicebio, GB11015, 1:1000), GSK3β (abcam, ab32391, 1:2500), p-GSK3β (Abcam, ab75814, 1:2500), anti-JNK1 (Cell Signaling Technology, 9252P, 1:500), anti-p-JNK1 (Cell Signaling Technology, 9255S, 1:500), anti-p38 (Cell Signaling Technology, 8690P, 1:500), and anti-p-p38 (Cell Signaling Technology, 4511S, 1:500), primary antibody. The next day, the bands were incubated with the corresponding secondary antibody and scanned with gel imaging system (ChemiDoc, BIO-RAD).
2.18 Enzyme-linked immunosorbent assay (ELISA)
PC12 cells were incubated with a series of concentrations of FSZ NPs, respectively. The supernatants were collected and subjected to VEGF-a/IL-4/IL-6/TNF-α ELISA assay according to the specification.
2.19 Transwell migration assay
PC12 cells were cultured with ECM medium with different concentrations of SSe@ZIF-8 for 24 h. HUVECs (1×104 cells per well) were resuspended in 100 µL ECM from PC12 cells (with different concentrations of SSe@ZIF-8) and then seed in the upper chambers of the 24-well transwell nest. Meanwhile, medium containing FBS was added into the lower chamber. After incubation at 37°C for 24 h, the cells on the surface of the membrane were washed with PBS, fixed with 4% paraformaldehyde for 15 min and stained with 0.1% crystal violet for 30 min at room temperature. Cells on the upper surface of the transwell membrane were scraped out. After that, cells were photographed and five fields were randomly selected to count the number of migrated cells on the back surface.
2.20 Cell homing detection
PC12 cells (2×104 cells per well) were seeded in the upper chambers of the transwell nest, while different concentration of FSZ-GelMA hydorgel were fixed in bottom of the upper chambers with visible light source. The cell viability test was performed by CCK-8 assay.
2.21 Tube formation assay
The matrigel-coated plate was obtained by adding 60 µL matrigel (1:1) to each hole of the 96-well plate and leaving it at room temperature for 15 minutes and 37 ℃ for 60 minutes. HUVECs (1×104 cells per well) were seeded in each matrigel-coated well and incubated with ECM containing different concentrations (0, 5, 10, 20, 40, and 80 µg/mL) of SSe@ZIF-8 for 6 h. The tubular structures were observed under inverted fluorescence microscope (IX71, OLYMPUS). The tube length and the number of nodes and meshes were determined by the ImageJ software (Media Cybernetics).
2.22 Flow cytometry
Except the control group, RAW264.7 cells were stimulated with LPS (1 mg/mL) and the cells in experimental group were treated with Fer-1 (0.1 µM), SSe@ZIF-8 (40 µg/mL), and Fer-1@SSe@ZZIF-8 (40 µg/mL), respectively. After incubation for 24 h, single-cell suspension was blocked with Fc Block and stained with CD86 at 4 ℃ for 30 min. Then samples were stained with CD206 at 4 ℃ for 30 min after fixation and permeabilization. A flow cytometer (CytoFlex, Beckman Coulter) is used to analyze the ratio of M1/M2 cell polarization.
2.23 Animals and ethics statement
36 female Sprague-Dawley rats (4 weeks old) were purchased from the Experimental Animal Center of Three Gorges University (Yichang, Hubei, China) and adaptively fed at room temperature and adequate diet for a week. All surgical procedures were approved by the Experimental Animal Ethics Committee of Renmin Hospital of Wuhan University (20220704B).
2.24 Surgical procedure and treatment
To establish the SCI model, rats were anesthetized by intraperitoneal injection of 3% phenobarbital (0.1 mL/100 g) and the skin was disinfected. Then a longitudinal incision is made, and the T10 laminectomy was conducted to expose the spinal cord. Except the sham operation group, the spinal cord was compressed by a vascular clamp with a closing force of 50 g for 15 s, and the tail of rats swung from side to side and then suddenly drooped, indicating the model is successful. The muscle, fascia and skin were sutured layer by layer. After the establishment of the model, the sensory and motor functions of the hind limbs of rats were lost with a Basso, Beattie, and Bresnahan (BBB) score of < 2. For 3 days after surgery, penicillin was injected intramuscularly once a day. The bladder was manually emptied twice daily until the urinary function was restored.
All rats were randomly divided into 6 groups (n = 6): (1) sham operation group, (2)SCI model group, (3) GelMA (the pure GelMA hydrogel), (4) Fer-1 (GelMA with 1 µM Fer-1), (5) SSe@ZIF-8 (GelMA with 40 µg/mL SSe@ZIF-8), (6) Fer-1@ZIF-8(GelMA with 40 µg/mL FSZ). Rats in the sham operation group only received laminectomy without spinal cord compression. At 24 h post injury, 10 µL of corresponding GelMA hydrogel was injected into the spinal cord injury site by stereotaxic instrument and microinjection pump, and solidified with a light curing lamp for 10 s. The SCI model group was treated with an equal volume of saline as the control.
2.25 Behavioral tests
The spinal cord function in rats was evaluated by video recordings, Basso, Beattie & Bresnahan (BBB) Locomotor Rating Scale, oblique plate test, and footprint analysis on 1, 7, 14, 21 and 28 d post-SCI. The rats were asked to walk along the wall, and videos were taken. height from the ground, less foot error, spasm duration and forward distance were analyzed. BBB scores were performed by three trained observers who were blinded to the experimental design. For footprint analysis, the rats were asked to run in a narrow dark track (10 cm × 100 cm) lined with paper. The red ink on the sole of the foot could record the position of footprints when rats touched. For the slope test, the rats were placed on an angle-adjustable inclined plate and the angle between the inclined plate and the desktop was slowly increased. When the rat could only stay in the original position for 5 s, the angle was recorded.
2.26 Blood routine and biochemical tests
On Day 28 after SCI, Anticoagulant blood for rat tail was taken for routine blood test provided by Servicebio company.
2.27 H&E staining
On Day 28 after SCI, the rats were given saline and 4% PFA for cardiac perfusion. The spinal cord from T9-T11 was carefully separated and fixed with 4% paraformaldehyde. The tissues were embedded in paraffin after dehydration and made into paraffin sections (5 µM thickness). The sections were baked at 60°C for 1 h and then dewaxed with xylene and gradient ethanol. The sections were stained with H & E Stain Kit and photographed under an optical microscope.
The heart, liver, spleen, lung and kidney of the rats were also extracted and stained.
2.28 Nissl staining
Slices were incubated with Nissl staining solution at 37 ℃ for 3 min, and then washed with distilled water and 95% ethanol. After dehydration, transparency and sealing, images were obtained under an optical microscope.
2.29 Immunofluorescence staining
The tissue sections were incubated with 1% BSA for 30 min after antigen repair. Subsequently, the samples were incubated with anti-iNOS (CST, 10320), anti-Arg1 (abcam, ab239731), anti-Tuj1 (CST, 5568), anti-GFAP (CAT, 3670), anti-Nrf2 (CST, 12721), and anti-c-cap3 (santa cruz, sc-56053) primary antibody, and then incubated with corresponding FITC-labeled goat anti-mouse and Cy3-labeled goat anti-rabbit secondary antibody for 1 h at RT in the dark. Finally, they were stained with DAPI, followed by sealing with anti-fluorescence quenching sealing tablets. The fluorescence distribution of these cells was observed by a fluorescence microscope (BX53, OLYMPUS) and analyzed by the ImageJ software.
Raw264.7 cell samples were obtained by the same treatment as those for FCM. Cells were fixed with 4% paraformaldehyde and permeabilized with 0.2% Triton X-100. The next protocol was similar to the above.
2.30 Immunohistochemical staining
The sections were blocked with 1% bovine serum albumin (BSA) at RT for 60 min, and incubated with anti-CD31 (abcam, ab28364) primary antibody overnight at 4 ℃. Afterwards, the samples were incubated with the secondary antibody at RT for 1 h and stained with DAPI. Images were collected with a fluorescence microscope (BX53, OLYMPUS) and analyzed using ImageJ.
2.31 TUNEL staining
The sections were incubated with the TUNEL reaction mixture for 60 min at 37°C in the dark, followed by stained with DAPI for 5 min at RT. Images were obtained with a fluorescence microscope (BX53, OLYMPUS) and analyzed using ImageJ. The apoptotic cell proportion was calculated as the number of TUNEL-positive cells / total number of cells.
2.32 TSQ staining
TSQ is a fluorophore that binds intracellular Zn ion[24]. The sections were immersed with 4.5 µM TSQ in 140 mM sodium barbital and 140 mM sodium acetate buffer (pH 10) for 90 s. Images were obtained with a fluorescence microscope (BX53, OLYMPUS) and analyzed using ImageJ.
2.33 Statistical analysis
All experiments in this paper were repeated three times. Data were analyzed using GraphPad Prism (Version 9.0) were expressed as mean ± standard deviation (Mean ± SD). Statistical analysis of experimental data was performed using one-way ANOVA followed with Bonferroni's post-hoc test or paired t-test (*P < 0.05, **, ##P < 0.01, and ***P < 0.001).