Ethics Statement
This study utilized male C57BL/6J mice procured from Changsheng Biotechnology Technology Co., Ltd. (Liaoning, China). Standard environmental conditions were provided for the mice to minimize environmental variability. All animal protocols were implemented in strict accordance with ARRIVE guidelines (https://arriveguidelines.org) and approved by the Ethics Committee of Hubei University of Chinese Medicine (Approval No. HUCMS22702282).
Establishment of the transient middle cerebral artery occlusion (tMCAO) model
A total of 90 male C57BL/6J mice (aged 6–8 weeks and weighing 20–22 g) were used in short-term tMCAO experiments. Six groups were formed at random: control group, sham group, vehicle group, 30 mg/kg Nef group, 60 mg/kg Nef group, and 60 mg/kg NBP group.
Dl-3n-butylphthalide (NBP), a synthetic medication derived from Apium graveolens Linn seeds (celery), has been approved by the Food and Drug Administration (FDA) for phase II clinical trials in patients with acute ischemic stroke since 201610. NBP has been found to possess diverse pharmacological actions, such as promoting cerebral microcirculation and improving mitochondrial function 11–13. Due to its protective effect on BBB during IS 14, it was adopted as a positive drug. Each group consisted of 12 animals that underwent a successful operation for subsequent experiments. A total of 1% of mice failed to achieve successful reperfusion following the tMCAO procedure, and approximately 20% died within one day.
The tMCAO procedure was carried out in accordance with the methods described previously. 15. To mitigate the influence of hormonal disturbances in female mice post-tMCAO surgery, only male mice were studied. Briefly, mice were anesthetized by intraperitoneal injection of pentobarbital sodium at a dose of 70mg/kg, and a monofilament nylon suture with a round tip was inserted into the internal carotid artery via the right common carotid artery, avoid pterygopalatine artery and finally accessed the middle cerebral artery. It was placed for 45 min until the blood flow was restored by withdrawing the filament for reperfusion. The wounds were carefully sutured to prevent infection. The sham group underwent all surgical procedures, excluding the insertion of nylon sutures. Additionally, all mice were allowed free access to food and water.
Drug administration
Neferine (Nef, CAS number 2292-16-2, purity over 98%) was obtained from Biopurify (Chengdu, China). The vehicle solution used for dissolving Nef consisted of 10% DMSO and 90% corn oil. Dl-3n-butylphthalide (NBP, CAS number 6066-49-5, purity over 95%) was obtained from Macklin (Shanghai, China). NBP was diluted with corn oil. At 2 and 12 hours after surgery, separate groups of mice were treated with a volume of 0.2 mL via intragastric gavage of NEF (30, 60 mg/kg), NBP (60 mg/kg) or vehicle.
Measurement of assessment
At the end of 24 h reperfusion, neurological status of mice was assessed using the Zea Longa neurological scoring system, as mentioned earlier 16. For this study, mice with scores ranging from 1 to 3 were included as the tMCAO model mice.
Quantification of the infarct volume, cerebral edema volume, and cerebral water content
At 24 h post-reperfusion, mice were euthanized by cervical dislocation. All brains were collected and divided into five sections, then immersed immediately in a 0.5% solution of 2,3,5-triphenyltetrazolium chloride (TTC, G1017, Servisebio, Wuhan, China) at 37 ℃ for approximately 30 min. A red stain appeared on normal brain tissues, while infarcted parts displayed in white. To correct the brain swelling volume due to cerebral edema, the area of the ipsilateral uninfarcted brain slice was subtracted from the contralateral hemisphere brain slice area to determine as infarct volume. Once the brain tissues were removed, any surplus water in the vicinity of the tissue was soaked up, and the weight was documented as wet weight. Afterward, a 72-hour desiccation process in an oven set at 80°C was employed to ensure complete removal of moisture in brain tissues. The weight obtained following the drying phase was denoted as the dry weight. Subtract dry weight from wet weight to determine brain water content.
BBB permeability measurement
Mice were injected with 2% Evans Blue (E104208, Aladin, Shanghai, China) staining solution (4ml/kg) through the tail vein 1 hour before the end of reperfusion in accordance with the earlier descriptions to evaluate BBB permeability17,18, and blue coloration of the conjunctiva, ears, and limbs was observed. After complete anesthesia of mice, open the chest to completely expose the heart, excise the right atrial appendage, and saline solution was infused into the mice via the left ventricle, take the brain from the head after the fluid flowing out from the right atrium becomes clear. Cut out the brain tissue from the right ischemic area, add an appropriate amount of formamide in a ratio of 100: 1 for the brain tissue (mg): formamide (ml), and prepare the brain homogenate by thorough grinding using a homogenizer. The homogenate was incubated at 45°C under light-protected conditions for 48 h. Afterward, the homogenate was centrifuged (1000 × g, 15 min) to obtain the supernatant. OD values at 632 nm were determined for each group utilizing a microplate reader (Tecan SunriseTM, Austria), and brain EB concentration of each group were measured by utilizing a standard curve as a reference.
Histopathological examination
The whole brain tissue was fixed for 24 hours in 4% paraformaldehyde at room temperature before undergoing paraffin embedding and subsequent sectioning into 5µm thick coronal slices, including the cortical infarct region. Utilizing the Hematoxylin/Eosin (H&E) staining to provide a clear visualization of the tissue morphology and cellular architecture of the cortical region in the brain, and examine the stained sections of each group using an optical microscope (Olympus, Tokyo, Japan).
Immunohistochemistry
Saline perfusion was carried out in the mice to eliminate blood from the vasculature, and then fix brain tissue with 4% paraformaldehyde. Following fixation, the tissues were dehydrated and sliced into 20 µm frozen sections. Sections were then blocked with 3% BSA (Vector Laboratories, Burlingame, CA, USA) for 30 min, and incubated with the following primary antibodies overnight at 4°C: NLRP3, PGC-1α, Occludin, and ZO-1. After that, the sections were incubated with appropriate HPR-labeled secondary antibodies for 50 min at room temperature followed by with a 3 min staining of Hematoxylin dye solution (Servicebio, Wuhan, China) at room temperature. Fluorescence images of the specimen were acquired using the Olympus fluorescence microscope FV3000 and Image J software was employed for the measurement of fluorescence intensity. The working dilution of specific antibodies can be found in Supplementary materials.
Cell cultures
Mouse brain endothelial cells (bEnd.3, No. CL-0598, Procell, Wuhan, China) were cultured in the complete endothelial cell medium (CM-0598, Procell, Wuhan, China) in a humidified incubator at 37°C with 5% CO2 and 95% air.
Oxygen-glucose deprivation/reoxygenation (OGD/R) establishment and drug treatment
Cells were subjected to OGD/R to simulate I/R injury. Upon reaching 80–90% confluence, bEnd.3 cells were placed in glucose-free DMEM (Procell, Wuhan, China) that had been degassed by ultrasonication. Then, cells were treated with Nef at concentrations of 0.1µM, 0.5µM or 1µM, and transferred to a chamber consisting of 5% CO2 and 95% N2 for 9 hours. Following the hypoxia exposure, replace the media with fresh growth medium and incubated another 12 h under normoxic conditions for reperfusion.
Cell viability assays
A colorimetric CCK-8(GK10001, GLPBIO, CA, USA) assay was utilized for cell viability measurement. After OGD/R treatment, cell viability data exserts a relative percentage change compared to the untreated control group. Furthermore, a commercial assay kit (LDH, C0016, Beyotime, Shanghai, China) was employed to measure LDH release, a marker of cellular toxicity. Briefly, 50µl of culture medium from each group was transferred to another 96-well plate, followed by LDH measurement using the manufacturer-provided test reagent. Additionally, the visualization of live and dead cells was conducted employing a Live/Dead assay kit (BB-4126, BestBio, Shanghai, China). Fluorescence images of the specimen were acquired using the Olympus fluorescence microscope FV3000 and Image J software was employed for the measurement of fluorescence intensity.
Tube Formation Assay
The analysis of tube formation was conducted using Matrigel basement membrane matrix with growth factor-reduced (082701, ABWbio, Shanghai, China). Briefly, a volume of 60 µL of Matrigel matrix was added to a 24-well plate. The pretreated bEnd.3 cells were then seeded and incubated for 6 hours. Microscope photos were then obtained from three randomly chosen optical fields at a magnification of ×100. Analysis of the tube branch length was conducted by an uninformed experimenter, utilizing the "Angiogenesis Analyzer" plugin in Image J software. Each experiment was conducted a minimum of three times.
Endothelial Barrier Permeability Measurement
To assess endothelial barrier permeability, transmembrane electrical resistance (TEER) values of endothelial cell monolayer were measured using transmembrane resistance measuring instruments (EVOM2, WPI, Florida, USA) at time points of OGD, reoxygenation initiation, and 12 hours of reperfusion. TEER values were calculated as followed:
TEER (Ω × cm2) = (total resistance − blank resistance) (Ω) × insert area (cm2)
The flux of Evans blue-labeled albumin (EBA: 1% BSA + 167.5 µg/ml Evans blue; 67 kDa) across the cell monolayer was measured according to previous report19. Following 12h reoxygenation, 50 µL of EBA was added to the transwell inserts. Over the next six hours, media from the lower chambers from each group were collected hourly to measure optical density at 630 nm.
Oxidative Stress Assay
Levels of superoxide dismutase (SOD, S0101S, Beyotime, Shanghai, China) and malondialdehyde (MDA, S0131S, Beyotime, Shanghai, China) were measured to assess intracellular oxidative stress using commercially available kits. The experiments were conducted in triplicate using 6-well plates for each condition.
Western blot analysis
Cellular protein extraction was accomplished using Cell lysis buffer for Western and IP (P0013, Beyotime, Shanghai, China) in accordance with the manufacturer’s instructions. SDS-PAGE was employed to separate the protein samples (30 µg per group), followed by transferred onto a PVDF membrane (0.45 µm pore size, Millipore, MA, USA). The membrane blocked for 30 min and incubated overnight at 4°C with primary antibodys against ZO-1 (1: 5000; 21773-1-AP, Proteintech Group, Wuhan, China), Occludin (1: 5000; 66378-1-Ig, Proteintech Group, Wuhan, China), PGC-1α (1: 5000; 66369-1-Ig, Proteintech Group, Wuhan, China), NLRP3 (1: 1000; #15101, CST, MA, USA), AIM2 (1: 1000; #63660, CST, MA, USA), ASC/TMS1 (1: 1000; #67824, CST, MA, USA), IL-1β (1: 1000; #31202, CST, MA, USA), Cleaved-Caspase-1 (1: 1000; #89332, CST, MA, USA), Caspase-1 (1: 1000; #24232, CST, MA, USA), GSDMD (1: 2000, bs-14287R, Bioss, Beijing, China), IL-18(1: 2000, bs-42148R, Bioss, Beijing, China) β-actin (1: 5000; bs-0061R, Bioss, Beijing, China). After that, the membrane were interacted with the respective secondary antibody for 1.5 h. β-actin was used as a loading reference. The protein bands were detected using ECL method (MA0186-1, Meilunbio, Dalian, China) and developed with gel densitometric scanning.
Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR)
TRIzol reagent (BS258A, Biosharp, Anhui, China) was employed to obtain total RNA from bEnd.3 cells. Subsequently, DNA contamination was eliminated from the total RNA, and cDNA was synthesized using the ABScript II cDNA First Strand Synthesis Kit (RK20400, ABclone, Wuhan, China). In the end, cDNA was mixed with specific primers and qRT-PCR was performed using TOYOBO SYBR Green Realtime PCR Master Mix (QPK-201, TOYOBO, Japan) to validate the expression of specific mRNAs. The specific primers listed below were utilized for PCR amplification of the cDNA fragment. NLRP3 forward: 5′-GCATTGCTTCGTAGATAGAGG-3′, reverse: 5′-GA GAAGGACCCACAGTGTAA-3′; PGC-1α forward:5′-AGTTTTTGGTGAAATTGAGGAAT-3′, reverse: 5′- TCATACTTGCTCTTGGTGGAAGC';Occludin forward:5′- AGGACGGACCCTGACCACTA-3′, reverse: 5′- CCTGCAGACCTGCATCAAAA′༛ZO-1 forward: 5′- CACAAGGAGCCATTCCTGAAG-3′, reverse: 5′-ATCACTAGGGGGCTCAGCAG′; β-actin forward: 5′-CGTGCGTGACATCAAAGAGA-3′, reverse: 5′-CCCAAGAAGGAAGGCTGGA-3′. The 2 − ΔΔCt method was used to calculate the relative gene expression levels, and β-actin expression served as endogenous internal control.
Mitochondrial ROS (mtROS) measurement
The mtROS level was assessed with MitoSOX Red Mitochondrial Superoxide Indicator (C1071S, Beyotime biotechnology, Shanghai, China). Following OGD/R injury, the cells underwent centrifugation at 1000g for 5 minutes and were then washed with PBS. Mito-Tracker Red CMXRos and Hoechst 33342 were then added and incubated in dark for 30 min. Fluorescence images of the specimen were acquired using the Olympus fluorescence microscope FV3000 and Image J software was employed for the measurement of fluorescence intensity.
Mitochondrial membrane potential (MMP) measurement
Mitochondrial Membrane Potential Assay Kit (E-CK-A301, Elabscience, Wuhan, China) was employed to assess the MMP in bEnd.3 cell. Briefly, JC-1 dye solution was added to cells and incubated for 20 min after OGD/R injury, followed by 5 min incubation with Hoechst 33342. Olympus Fluorescence FV3000 microscope was employed to observe the fluorescence and Image J software was employed for the measurement of fluorescence intensity.
Molecular Docking
To initiate the molecular docking process, the PGC-1α protein structure (PDBID: 3B1M) was imported from the RSCB PDB database in PDB format, while the 3D structure of Neferine was obtained from PubChem. Using PyMOL software, water molecules were removed, and the ligand was isolated. After employing AutoDock Tools 1.5.6 to perform tasks such as hydrogen addition, charge calculation, and atomic type assignment. Subsequently, AutoDock Vina 1.1.2 was utilized for molecular docking simulations to investigate the binding characteristics between the Nef ligand and PGC-1α protein. The binding energy, which serves as an evaluation index for molecular docking, was calculated. The optimal conformations of Nef and PGC-1α were determined by analyzing the clusters within the docking results, which were selected based on their respective binding energies. Typically, a lower docking energy indicates a stronger interaction force between the components, and a threshold of -7 kcal/mol is often used. The epitope with the lowest affinity scores predicted by AutoDock Vina was subjected to LigPlot + and PyMol for further visualization of the interactions.
Short Interfering (Si) RNA and Plasmid Transfection
Cells cultivated in a 6-well plate with OPTI medium (31985062, Invitrogen, CA, USA) were transfected with either PGC-1α siRNA (3′- CCG CAA UUC UCC CUU GUA UTT-5′) or negative control siRNA (3′-ACG UGA CAC GUU CGG AGA A-5′) using Lipofectamine 3000 transfection reagent (L3000001, Invitrogen, CA, USA). The siRNAs were obtained from Sangon (Shanghai, China). Following 12 h incubation, all medium was substituted with growth medium, and cells were cultured for an additional 24 hours. Cells were then subjected to OGD/R injury and harvested for Western blot analysis.
Statistical analysis
GraphPad Prism 7 Software (La Jolla, CA, USA) was used for statistical analysis, and all results were expressed as means ± SD. One-way ANOVA with Bonferroni’s test was utilized to determine the differences between groups for data with a single dosage or time point. For comparisons between two groups, including OGD and drug treatment, PGC-1α siRNA and drug treatment, the t-test was used. Statistical significance was defined as a p-value of 0.05 or less.