
Key Words: blood-brain barrier, Dl-3-n-butylphthalide, hypoxia inducible factor 1α, mitochondria, neuroprotection, oxidative stress, reactive oxygen species, stroke, transcription factor, ubiquitination
Abstract
Dl-3-n-butylphthalide is used to treat mild and moderate acute ischemic stroke. However, the precise underlying mechanism requires further investigation. In this study, we investigated the molecular mechanism of Dl-3-n-butylphthalide action by various means. We used hydrogen peroxide to induce injury to PC12 cells and RAW264.7 cells to mimic neuronal oxidative stress injury in stroke in vitro and examined the effects of Dl-3-n-butylphthalide. We found that Dl-3-n-butylphthalide pretreatment markedly inhibited the reduction in viability and reactive oxygen species production in PC12 cells caused by hydrogen peroxide and inhibited cell apoptosis. Furthermore, Dl-3-n-butylphthalide pretreatment inhibited the expression of the pro-apoptotic genes Bax and Bnip3. Dl-3-n-butylphthalide also promoted ubiquitination and degradation of hypoxia inducible factor 1α, the key transcription factor that regulates Bax and Bnip3 genes. These findings suggest that Dl-3-n-butylphthalide exhibits a neuroprotective effect on stroke by promoting hypoxia inducible factor-1α ubiquitination and degradation and inhibiting cell apoptosis.
Introduction
Stroke is the second leading cause of death and disability worldwide, with ischemic cerebrovascular disease accounting for 80% of all strokes (Koh and Park, 2017). Thrombolytic therapy and arterial intervention are the most effective treatment methods for stroke (Brott et al., 1994). These methods are aimed at restoring the blood supply to the ischemic area caused by the stroke as soon as possible (Prabhakaran et al., 2015). However, calcium overload (Zhou et al., 2021) and oxidative stress (Deng et al., 2021; Jiang et al., 2022; Ma et al., 2022; Zhu et al., 2022) after stroke can lead to a cascade of pathologies, subsequently resulting in neuronal necrosis and blood-brain barrier (BBB) damage (Jiang et al., 2018). Reducing oxidative stress injury is a key part of the treatment of ischemic stroke and a priority in research.
Dl-3-n-butylphthalide (NBP) is a small molecule drug used to treat mild to moderate acute ischemic stroke (Huang et al., 2018; Wang et al., 2018; Chen et al., 2019; Fan et al., 2021). NBP has been reported to inhibit apoptosis after ischemic injury (Liu et al., 2021). Another study found that NBP treatment promotes neurological functional recovery in a cerebral ischemia mouse model (Wang et al., 2019). The protective effects of NBP on vascular endothelial cells, BBB and neurons suggest the potential of NBP in rescuing brain function after ischemic injury (Li et al., 2019a). However, its precise role and molecular mechanism have not been completely elucidated.
In this study, we investigated the neuroprotective effects of NBP in response to oxidative stress injury by simulating oxidative stress injury in neuronal cells with H2O2 treatment. We further explored the potential molecular mechanism of NBP.
Methods
Cell culture
PC12 (Cat# TCR9) and RAW 264.7 (Cat# TCM13) cells, which are common in vitro models for neurological diseases and macrophage research, were purchased from the Chinese Type Culture Collection, Chinese Academy of Sciences (Shanghai, Chinese). Cells were cultured in Dulbecco’s modified Eagle’s medium (Invitrogen, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (Biological Industries, Jerusalem, Israel) in a humidified atmosphere at 37°C containing 5% CO2. To establish an oxidative stress injury model, PC12 cells (1 × 104 cells/well) were inoculated in 96 well plates and treated with a final concentration of 200 µM H2O2 (Sigma, St. Louis, MO, USA) for 4 hours. The cells were pretreated with 20 and 40 µM NBP (Shiyao Pharmaceutical Co., Ltd., Shijiazhuang, Chinese) for 24 hours. Untreated cells served as controls.
Cell viability analysis
Cells were incubated with Cell Counting Kit-8 solution (CCK8, 10 µL; MCE, Shanghai, China) for 60 minutes. Absorbance was measured at 450 nm using a spectrophotometric microplate reader (Gene Company Ltd., Shenyang, China).
5-Ethynyl-2’-deoxyuridine incorporation assay
PC12 cells (1 × 104 cells/well) were inoculated in appropriate numbers into 96-well plates and treated as indicated. Viability was evaluated using the BeyoClick™ EdU Cell Proliferation Kit (APExBIO, Houston, TX, USA). EdU pre-warmed at 37°C was added to 96-well plates and cells were then examined following the kit instructions. Cells were observed and photographed using a fluorescent inverted microscope (Leica, Wetzlar, Germany).
Caspase-3 activity assay
Caspase-3 activity was measured using GreenNuc Caspase-3 substrate (Beyotime, Shanghai, China) following the manufacturer’s protocol. In brief, PC12 cells were plated into 96 wells (5 × 104 cells/mL) and treated as indicated. After washing cells twice with phosphate buffered saline (PBS), GreenNuc Caspase-3 substrate was added to each well and cells were incubated for 30 minutes. Hoechst 33342 solution (Beyotime) was then added to each well and plates were incubated for 10 minutes at 18–26°C and protected from light. Cells were analyzed under a fluorescence microscope (Leica).
Mitochondrial membrane potential assay
PC12 cells were treated with 5 μM tetramethylrhodamine ethyl ester (TMRE, MCE) at 37°C for 15 minutes; the supernatant was removed and cells were washed twice with pre-warmed cell culture medium to remove excess dye. After Hoechst 33342 staining, photographs were taken under a fluorescence microscope.
Intracellular reactive oxygen species assay
Intracellular reactive oxygen species (ROS) production was detected using 2,7-dichlorodihydrofluorescein diacetate (DCFH-DA; MCE). Briefly, after NBP treatment, PC12 cells were washed with prewarmed PBS and stained with 10 µM DCFH-DA for 20 minutes at 37°C. Cells were washed three times with pre-warmed cell culture medium to remove excess dye and subjected to H2O2 treatment for 4 hours. Hoechst 33342 was used to stain cells for 10 minutes, and images were acquired with a fluorescence microscope.
Transcriptome sequencing and differentially expressed gene analysis
Total RNA was extracted from PC12 cells, and whole transcriptome sequencing was sequenced on an Illumina Novaseq platform and bioinformatics data were analyzed by Origin-Gene Company (Shanghai, China). The differentially expressed genes (DEGs) results were analyzed in the RNA sequencing results after transcriptome sequencing. Regulatory interactions were examine between transcription factors and target genes using the TRRUST database (https://www.grnpedia.org/trrust/) (Han et al., 2018). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of the DEGs were conducted by clusterProfiler package (version 3.14.3; https://bioconductor.org/packages/release/bioc/html/clusterProfiler.html).
Real-time polymerase chain reaction
Trizol reagent (Thermo Fisher, Waltham, MA, USA) was used to isolate RNA from PC12 and RAW264.7 cells. cDNA was synthesized from 3 µg of RNA template with the RevertAid Master Mix Reagent (Thermo Fisher) following the manufacturer’s instructions. Real-time polymerase chain reaction (qPCR) was performed using Platinum SYBR Green qPCR SuperMix (Thermo Fisher). Gene expression was calculated using the ΔΔCt method (Livak and Schmittgen, 2001); glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA was used as the internal control. Primer sequences were as follows: Bax: forward: 5′-AGA CAG GGG CCT TTT TGC TAC-3′, reverse: 5′-AAT TCG CCG GAG ACA CTC G-3′; Bnip3: forward: 5′-CTG GGT AGA ACT GCA CTT CAG-3′, reverse: 5′-GGA GCT ACT TCG TCC AGA TTC AT-3′; Mmp-2: forward: 5′-CAA GTT CCC CGG CGA TGT C-3′, reverse: 5′-TTC TGG TCA AGG TCA CCT GTC-3′; Mmp-9: forward: 5′-CTG GAC AGC CAG ACA CTA AAG-3′, reverse: 5′-CTC GCG GCA AGT CTT CAG AG-3′; and GAPDH: forward: 5′-AGG TCG GTG TGA ACG GAT TTG-3′, reverse: 5′-GGG GTC GTT GAT GGC AAC A-3′.
Immunoprecipitation
PC12 cells were incubated with 10 µM MG132 (Beyotime) for 9 hours. Immunoprecipitation was performed using an anti-hypoxia-inducible factor 1-alpha antibody (HIF-1α; rabbit, 1:100, Proteintech, Chicago, IL, USA, Cat# 20960-1-AP, RRID: AB_10732601). Immunoprecipitated proteins were subjected to western blot as described below.
Western blot assay
The PC12 cells were lysed using mixed cell lysates containing protease inhibitors at 4°C for 30 minutes, and 12,000 × g for 15 minutes, and the supernatant was collected. Protein samples were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred to polyvinylidene fluoride membranes. After blocking, the membranes were probed with anti-HIF-1α and anti-GAPDH (1:1000, Proteintech, Cat# 10494-1-AP) overnight at 4°C. For immunoblotting of immunoprecipitation complexes, membranes were incubated with the ubiquitin primary antibody (mouse, 1:1000, Proteintech, Cat# 10201-2-AP, RRID: AB_671515) overnight at 4°C. Membranes were then incubated with horseradish peroxidase-conjugated secondary antibody (mouse, 1:1000, Abcam, Cambridge, MA, USA, Cat# ab131366, RRID: AB_2892718) for 1 hour at room temperature. Protein bands were visualized by electrochemiluminescence (Beyotime) using Quantity One (Bio-Rad).
Chromatin immunoprecipitation
PC12 cells were incubated in 1% formaldehyde for 15 minutes and cells were lysed using mixed cell lysates at 4°C for 30 minutes. DNA was digested with micrococcal nuclease (NEB, Ipswich, MA, USA). After the addition of chromatin immunoprecipitation dilution buffer (Abcam, Cambridge, MA, USA), part of the sample (50 µL) was saved as input. Samples (500 µL) were then incubated with HIF-1α antibody overnight at 4°C. Protein A/G magnetic beads (MCE) were added to the antibody/chromatin complex and the mixture was incubated at room temperature for 2 hours. The complexes were resuspended in wash buffer, and the protein/DNA cross-links were cleaved to obtain free DNA. The purified DNA was resuspended in tris-ethylenediaminetetraacetic acid buffer (Sango Biotech, Shanghai, China) for polymerase chain reaction.
Matrix degradation assay
Glass coverslips were coated with 0.2 mg/mL of fluoresceine isothiocyanate-gelatin (Invitrogen, Carlsbad, CA, USA) and cross-linked with 0.5% glutaraldehyde. Coverslips were incubated in 10% fetal calf serum-containing medium overnight at 37°C. RAW 264.7 cells were cultured on the coated coverslips for 8 hours. Cells were stained with phalloidin (Beyotime), and nuclei were stained with 4′,6-diamidino-2-phenylindole (Beyotime). Cells were examined under a confocal microscope (Leica TCS SP8).
Statistical analysis
Statistical analyses were performed using GraphPad Prism 6.01 statistical software (GraphPad Software, San Diego, CA, USA, www.graphpad.com). Comparisons between groups were made using one-way analysis of variance followed by Bonferroni correction. P < 0.05 was considered statistically significant.
Results
NBP improves cell viability and inhibits apoptosis in H2O2-treated cells
PC12 cells are commonly used as an in vitro model to study central nervous system neurons (Wu et al., 2020). We first examined the effects of NBP on the viability of PC12 cells treated with H2O2, as a model of oxidative stress, using CCK8. While H2O2 reduced the viability of cells compared with controls, NBP pretreatment significantly prevented the decrease in viability of H2O2-treated cells (P = 0.0344; Figure 1A). EdU assay showed that NBP pretreatment reversed the inhibition of cell proliferation after H2O2 treatment (Figure 1B). Caspase-3 plays a key role in regulating apoptosis (Porter and Jänicke, 1999). Caspase-3 activity assays showed that pretreatment with NBP significantly reduced the induction of caspase-3 activity caused by H2O2 treatment (Figure 1C).
Figure 1.

NBP improves cell viability and inhibits apoptosis in H2O2-treated cells.
(A) PC12 cells were treated with 200 µM H2O2 for 4 hours. The cells were pretreated with 20 and 40 µM NBP for 24 hours. CCK8 assay was used to explore the effect of NBP on cell viability. (B) PC12 cells were treated with 200 µM H2O2 for 4 hours. The cells were pretreated with 40 µM NBP for 24 hours. EdU was used to examine the effects of NBP on cell proliferation (original magnification, 100×). (C) Caspase-3 activity assay in PC12 cells (original magnification, 100×). Data are presented as mean ± SD. The experiment was repeated three times. *P < 0.05, **P < 0.01, ****P < 0.0001 (one-way analysis of variance followed by Bonferroni correction). CCK8: Cell Counting Kit-8; CTL: control; EdU: 5-ethynyl-2′-deoxyuridine; NBP: Dl-3-n-butylphthalide.
NBP mitigates H2O2-induced mitochondrial dysfunction and ROS contents in PC12 cells
To assess whether NBP protected PC12 cells against H2O2 treatment by mitigating mitochondrial dysfunction associated with apoptosis, we measured the mitochondrial membrane potential by TRME staining assay. TRME intensity decreased following H2O2 exposure; however, NBP cotreatment prevented H2O2-induced changes in the mitochondrial membrane potential (Figure 2A). These results demonstrated that NBP mitigated H2O2 exposure-induced mitochondrial dysfunction.
Figure 2.

NBP protects mitochondrial function and inhibits ROS production in PC12 cells exposed to H2O2.
(A) PC12 cells were treated with 200 µM H2O2 for 4 hours. The cells were pretreated with 40 µM NBP for 24 hours. Mitochondrial membrane potential was determined by evaluation of TMRE staining under a fluorescence microscope (100× magnification). (B) Detection of ROS by DCFH-DA staining in PC12 cells (100× magnification). ROS levels were increased after H2O2 exposure compared with controls, and NBP pretreatment inhibited the increase in ROS levels. The experiment was repeated three times. CTL: Control; DCFH-DA: 2,7-dichlorodi -hydrofluorescein diacet; NBP: Dl-3-n-butylphthalide; ROS: reactive oxygen species; TRME: N,N,N’,N’-tetra-methylethylenediamine.
We next investigated the effects of NBP on intracellular ROS levels in PC12 cells after H2O2 treatment using DCFH-DA. The results showed that ROS levels increased after H2O2 exposure compared with controls, and NBP pretreatment inhibited the increase in ROS levels (Figure 2B).
Transcriptome sequencing to obtain DEGs and enrichment analysis
We next performed transcriptome sequencing analysis of H2O2-treated cells, NBP pretreated and H2O2-treated cells and control cells and identified DEGs. The overall distribution of the DEGs was represented by volcano plots (Figure 3A and B). The results identified 1338 DEGs in the H2O2 group compared with the control group (722 up-regulated and 616 down-regulated DEGs) (Figure 3A) and 1233 DEGs were found in the NBP + H2O2 group compared with the H2O2 group (643 up-regulated and 590 down-regulated DEGs) (Figure 3B). We then obtained 474 overlapping DEGs between the two sets of DEGs (Figure 3C).
Figure 3.

Identification of DEGs from transcriptome sequencing analysis.
(A) PC12 cells were treated with 200 µM H2O2 for 4 hours. The cells were pretreated with 40 µM NBP for 24 hours. Volcano plot visualizing DEGs between H2O2-treated and control PC12 cells. (B) Volcano plot of the DEGs between NBP-pretreated H2O2-treated and H2O2-treated cells. (C) Venn diagram showing the overlapping DEGs. CTL: Control; DEG: differentially expressed gene; NBP: Dl-3-n-butylphthalide.
Enrichment analysis was performed to analyze the functions of the overlapping DEGs. KEGG pathway analysis revealed that the DEGs were mainly enriched in phosphatidylinositol 3 kinase (PI3K)/protein kinase B (AKT), apoptosis and cellular senescence signaling pathways (Figure 4A). GO functional analysis revealed that DEGs were mainly enriched in biological processes of mRNA processing (Figure 4B). GO functional analysis revealed that the DEGs were enriched in nuclear speck and chromatin binding in the top 10 enrichment classes (Figure 4C and D).
Figure 4.

GO and KEGG pathway enrichment analyses of the overlapping DEGs.
(A) Bubble plot of KEGG pathway enrichment analysis of the DEGs. (B) Bubble plot of biological process enrichment analysis of the DEGs. (C) Bubble plot of cellular component enrichment analysis of the DEGs. (D) Bubble plot of molecular function enrichment analysis of the DEGs. DEG: Differentially expressed gene; GO: Gene Ontology; KEGG: Kyoto Encyclopedia of Genes and Genomes; NBP: Dl-3-n-butylphthalide.
NBP promotes HIF-1α degradation and may affect target gene transcription to inhibit apoptosis
Mitochondria play an important role in regulating caspase activity to promote apoptosis. Our results showed that NBP affects the mitochondrial membrane potential and ROS production in PC12 cells, which in turn inhibits apoptosis. Enrichment analysis of the DEGs obtained by transcriptome sequencing revealed that the overlapping DEGs were enriched in the endogenous apoptotic pathway (Additional Table 1), suggesting that NBP may regulate apoptosis by affecting the expression of these genes. The results of the enrichment analysis showed that Bcl2l1, Bax and Bnip3 genes were enriched in the apoptotic signaling pathway in PC12 cells. Therefore, we examined the expression levels of these genes in PC12 cells by real-time PCR. The results showed that H2O2 upregulated the expression of Bax and Bnip3 genes compared with controls, while NBP reversed this effect (Figure 5A).
Additional Table 1.
List of 31 differentially expressed genes involved in apoptosis
| ID | Description | Gene ratio | P-value | FDR | Gene ID | Size |
|---|---|---|---|---|---|---|
| GO:200 1233 |
Regulation of apoptotic signaling pathway | 23/40 6 |
4.07E-0 5 |
0.005 724 |
Ybx3/Selenos/Rffl/Bcl2l1/Pten/Eno1/Rpl26/Vdac2/Bax/Gsn/Becn1/Arhgef2/Eif2ak3/M ff/Slc35f6/Bnip3/Rps7/Tfdp1/Gnai2/Ywhaq/Ywhah/Ctnnb1/Raf1 | 23 |
| GO:009 7193 | Intrinsic apoptotic signalingpathway | 15/40 6 |
0.00171 2 |
0.047 472 |
Ybx3/Selenos/Bcl2l1/Eno1/Rpl26/Vdac2/Bax/Becn1/Arhgef2/Eif2ak3/Aifm1/Bnip3/Br sk2/Rps7/Stk25 | 15 |
FDR: False discovery rate.
Figure 5.
NBP promotes HIF-1α ubiquitination in H2O2-treated cells.
(A) PC12 cells were treated with 200 µM H2O2 for 4 hours. The cells were pretreated with 40 µM NBP for 24 hours. The mRNA expression of Bnip3 and Bax were detected by real-time polymerase chain reaction. GAPDH mRNA was used as the internal control. Data normalized by control group are presented as mean ± SD. The experiment was repeated three times. **P < 0.01, ***P < 0.001 (one-way analysis of variance followed by Bonferroni correction). (B) Analysis of HIF-1α ubiquitination (Ub) by immunoblot analysis. (C) Chromatin immunoprecipitation-polymerase chain reaction analysis of HIF-1α binding to Bnip3 and Bax promoters using antibodies against HIF-1α in PC12 cells. Bax: BCL2-associated X; Bnip3: BCL2 interacting protein 3; CTL: control; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; HIF-1α: hypoxia-inducible factor 1-alpha; IP: immunoprecipitation; NBP: Dl-3-n-butylphthalide.
The TRRUST database can be used to predict the regulatory interactions between transcription factors and target genes (Han et al., 2018). We used TRRUST to predict transcription factors of the DEGs involved in regulating the apoptotic process (Additional Table 2), and the results showed that HIF-1α was the key transcription factor regulating the DEGs.
AdditionalTable 2.
Predicting transcription factors of differentially expressed genes
| Key transcription factor | Numbers of genes | P-value | Q-value | List of overlapped genes |
|---|---|---|---|---|
| HIF1A JUN | 3 2 | 3.59E-05 0.00608 | 0.00018 0.0122 | Bax, Bnip3, Eno1 Bcl2l1, Becn1 |
| TP53 | 2 | 0.00732 | 0.0122 | Bax, Bcl2l1 |
| RELA | 2 | 0.0232 | 0.0235 | Bcl2l1, Bax |
| NFKB1 | 2 | 0.0235 | 0.0235 | Bax, Bcl2l1 |
We next examined the effect of NBP on HIF-1α protein expression after H2O2 treatment in PC12 cells by western blot analysis. The results showed that HIF-1α protein increased after H2O2 treatment compared with controls and it was down-regulated in the NBP pretreatment group compared with the H2O2 treatment group (Figure 5B). However, the HIF-1α gene was not identified as a DEG in transcriptome sequencing analysis. The alteration of HIF-1α protein levels but not mRNA levels in response to NBP may involve modulation of steady state protein levels through ubiquitination-mediated modification. Thus, we next analyzed the ubiquitination level of HIF-1α. While HIF-1α was non-ubiquitinated under H2O2 conditions, NBP promoted HIF-1α ubiquitination (Figure 5B).
To confirm that Bnip3 and Bax genes are transcriptionally regulated by HIF-1α, we performed chromatin immunoprecipitation assays. The results showed that HIF-1α may bound to Bnip3 and Bax promoters (Figure 5C).
Together these results suggest that NBP treatment results in the ubiquitination and subsequent degradation of HIF-1α under H2O2 conditions, leading to inhibition of the transcription of HIF-1α downstream genes.
NBP inhibits H2O2-induced activation of macrophage-derived matrix metalloproteinases
BBB damage is a critical pathological process that contributes to poor prognosis in cerebral ischemia (Li et al., 2019b), inhibition of matrix metalloproteinases protects the BBB (Li et al., 2019b; Candelario-Jalil et al., 2022). Given the reported protective effects of NBP on the BBB (Li et al., 2019a; Wu et al., 2020), we determined the effect of NBP on extracellular matrix (ECM) degradation in H2O2-treated RAW 264.7 macrophages, which have been reported as a potential source of MMPs that can degrade the BBB (Jiang et al., 2018). Matrix degradation assays showed that H2O2 treatment caused ECM degradation around the periphery of macrophages, and this was attenuated by NBP (Figure 6A). We next sought to investigate the mechanisms by which NBP treatment blocked H2O2-induced BBB degradation. Real-time PCR of RAW 264.7 macrophages demonstrated that H2O2 increased Mmp-2 and Mmp-9 mRNA expressions compared with controls, while NBP blocked the increase of Mmp-2 and Mmp-9 mRNA expression (Figure 6B). Previous studies showed that HIF-1α transcriptionally upregulates the expression of MMPs, and in the absence of HIF-1α, the level of MMPs decreased (Du et al., 2008; Palazon et al., 2014). Our findings suggest that NBP promotion of the degradation of HIF-1α may be responsible for the decrease in Mmp-9 levels in RAW 264.7 macrophages.
Figure 6.

NBP inhibits H2O2-induced ECM degradation.
(A) RAW 264.7 cells were treated with 200 µM H2O2 and 40 µM NBP for 8 hours. NBP inhibited H2O2-induced ECM degradation in RAW 264.7 cells. Cells were plated on coverslips coated with FITC-gelatin (green) for 8 hours. F-actin was stained with phalloidin (red), and nuclei were stained with DAPI (blue) (100× magnifications). Black areas indicate gelatin degradation. (B) Mmp-2 and Mmp-9 mRNA levels were detected by real-time polymerase chain reaction. Data normalized by control group are presented as mean ± SD. The experiment was repeated three times. **P < 0.01, vs. control group (one-way analysis of variance followed by Bonferroni correction). CTL: Control; DAPI: 4′,6-diamidino-2-phenylindole; ECM: extracellular matrix; Mmp: matrix metalloproteinase; NBP: Dl-3-n-butylphthalide.
Discussion
Stroke is a common and frequent occurrence in the elderly, often leaves survivors with chronic disability. Stroke is accompanied by cerebral ischemia-reperfusion injury and the death of neurons and other functional cells, leading to motor and cognitive impairment. After acute ischemic stroke, large amounts of ROS are produced, leading to further tissue damage. These ROS damage cellular macromolecules, leading to apoptosis and necrosis. Dysfunction of the BBB, characterized by degradation and increased permeability of the BBB, is an important pathophysiological feature of acute ischemic stroke. NBP has been shown to improve microcirculation and cerebral blood flow; it exhibits anti-inflammatory properties, inhibits apoptosis and protects the vascular endothelium to achieve brain protection (Huang et al., 2018; Wang et al., 2018; Chen et al., 2019; Fan et al., 2021). However, the exact molecular mechanisms of how NBP exerts these effects are unclear.
Most of neuroprotective drugs exert their protective effects by increasing the antioxidant capacity of cells (Tao et al., 2020; Griñán-Ferré et al., 2021). Several studies have shown that NBP exerts a significant neuroprotective effect in cerebral ischemia, which may be from its ability to inhibit oxidative stress (Chen et al., 2018; Que et al., 2021) and reduce brain edema (Mamtilahun et al., 2020; Zeng et al., 2020), neuronal death and inflammation (Liu et al., 2021; Que et al., 2021). Our study showed that NBP exerts a protective effect by affecting the mitochondrial apoptotic pathway, inhibiting Bax and Bnip3 expression. NBP reversed the reduction of mitochondrial membrane potential and activation of caspase 3 caused by H2O2. Chromatin immunoprecipitation experiments confirmed that the HIF-1α transcription factor binds the Bax and Bnip3 promoters, and NBP promoted the ubiquitinated-mediated degradation of HIF-1α, thereby inhibiting Bax and Bnip3 expression. HIF-1α protein level is modified by binding to Hippel-Lindau protein (VHL) to form a complex that is subsequently degraded by ubiquitination. We speculate that the effect on the degradation of ubiquitination may be due to the involvement of NBP in the interaction between ubiquitin and HIF-1α, and NBP may contribute to the formation of a more stable binding of the HIF-1α and VHL complex during ubiquitination, thus enhancing the degradation of HIF-1α.
In addition to exhibiting anti-apoptotic and ROS inhibitory effects, NBP treatment also inhibited the degradation of the basement membrane by macrophages, exerting exerting neuroprotective. The underlying mechanism needs further study. Given that NBP affects the ubiquitinated degradation of HIF-1α, which promotes the expression of matrix metalloproteinases, this may be related to the degradation of the basement membrane by macrophages affected by NBP. Further studies are required to clarify the mechanism. In addition, in vivo experiments may further compensate for the limitations of the results of in vitro experiments in this study.
In conclusion, our study provides evidence that NBP induces ubiquitination and degradation of the HIF-1α transcription factor and influences mitochondrial membrane potential and caspase-3 activation, resulting in anti-apoptotic effects and inhibition of ROS-mediated damage. However, how NBP as a small molecule is involved in influencing HIF-1α ubiquitination requires further study. Further studies with more cell types as well as animal experiments may provide further insights. Our study paves the way for further studies on the neuroprotective effects and mechanisms of NBP.
Additional files:
Additional Table 1: List of 31 differentially expressed genes involved in apoptosis.
Additional Table 2: Predicting transcription factors of differentially expressed genes.
Footnotes
Conflicts of interest: The authors declare no potential conflicts of interest.
Data availability statement: All data relevant to the study are included in the article or uploaded as Additional files.
C-Editor: Zhao M; S-Editors: Yu J, Li CH; L-Editors: Yu J, Song LP; T-Editor: Jia Y
References
- 1.Brott T, Broderick J, Kothari R. Thrombolytic therapy for stroke. Curr Opin Neurol. 1994;7:25–35. doi: 10.1097/00019052-199402000-00006. [DOI] [PubMed] [Google Scholar]
- 2.Candelario-Jalil E, Dijkhuizen RM, Magnus T. Neuroinflammation, stroke, blood-brain barrier dysfunction, and imaging modalities. Stroke. 2022;53:1473–1486. doi: 10.1161/STROKEAHA.122.036946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Chen N, Zhou Z, Li J, Li B, Feng J, He D, Luo Y, Zheng X, Luo J, Zhang J. 3-n-butylphthalide exerts neuroprotective effects by enhancing anti-oxidation and attenuating mitochondrial dysfunction in an in vitro model of ischemic stroke. Drug Des Devel Ther. 2018;12:4261–4271. doi: 10.2147/DDDT.S189472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Chen XQ, Qiu K, Liu H, He Q, Bai JH, Lu W. Application and prospects of butylphthalide for the treatment of neurologic diseases. Chin Med J (Engl) 2019;132:1467–1477. doi: 10.1097/CM9.0000000000000289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Deng H, Zhang Y, Li GG, Yu HH, Bai S, Guo GY, Guo WL, Ma Y, Wang JH, Liu N, Pan C, Tang ZP. P2X7 receptor activation aggravates NADPH oxidase 2-induced oxidative stress after intracerebral hemorrhage. Neural Regen Res. 2021;16:1582–1591. doi: 10.4103/1673-5374.303036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Du R, Lu KV, Petritsch C, Liu P, Ganss R, Passegué E, Song H, Vandenberg S, Johnson RS, Werb Z, Bergers G. HIF1alpha induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell. 2008;13:206–220. doi: 10.1016/j.ccr.2008.01.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Fan X, Shen W, Wang L, Zhang Y. Efficacy and safety of DL-3-n-butylphthalide in the treatment of poststroke cognitive impairment:a systematic review and meta-analysis. Front Pharmacol. 2021;12:810297. doi: 10.3389/fphar.2021.810297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Griñán-Ferré C, Bellver-Sanchis A, Izquierdo V, Corpas R, Roig-Soriano J, Chillón M, Andres-Lacueva C, Somogyvári M, Sőti C, Sanfeliu C, Pallàs M. The pleiotropic neuroprotective effects of resveratrol in cognitive decline and Alzheimer's disease pathology:From antioxidant to epigenetic therapy. Ageing Res Rev. 2021;67:101271. doi: 10.1016/j.arr.2021.101271. [DOI] [PubMed] [Google Scholar]
- 9.Han H, Cho JW, Lee S, Yun A, Kim H, Bae D, Yang S, Kim CY, Lee M, Kim E, Lee S, Kang B, Jeong D, Kim Y, Jeon HN, Jung H, Nam S, Chung M, Kim JH, Lee I. TRRUST v2:an expanded reference database of human and mouse transcriptional regulatory interactions. Nucleic Acids Res. 2018;46:D380–D386. doi: 10.1093/nar/gkx1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Huang L, Wang S, Ma F, Zhang Y, Peng Y, Xing C, Feng Y, Wang X, Peng Y. From stroke to neurodegenerative diseases:The multi-target neuroprotective effects of 3-n-butylphthalide and its derivatives. Pharmacol Res. 2018;135:201–211. doi: 10.1016/j.phrs.2018.08.007. [DOI] [PubMed] [Google Scholar]
- 11.Jiang C, Wang ZN, Kang YC, Chen Y, Lu WX, Ren HJ, Hou BR. Ki20227 aggravates apoptosis, inflammatory response, and oxidative stress after focal cerebral ischemia injury. Neural Regen Res. 2022;17:137–143. doi: 10.4103/1673-5374.314318. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 12.Jiang X, Andjelkovic AV, Zhu L, Yang T, Bennett MVL, Chen J, Keep RF, Shi Y. Blood-brain barrier dysfunction and recovery after ischemic stroke. Prog Neurobiol 163- 2018;164:144–171. doi: 10.1016/j.pneurobio.2017.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Koh SH, Park HH. Neurogenesis in stroke recovery. Transl Stroke Res. 2017;8:3–13. doi: 10.1007/s12975-016-0460-z. [DOI] [PubMed] [Google Scholar]
- 14.Li J, Liu Y, Zhang X, Chen R, Zhang L, Xue J, Gao X. Dl-3-N-butylphthalide alleviates the blood-brain barrier permeability of focal cerebral ischemia reperfusion in mice. Neuroscience. 2019a;413:99–107. doi: 10.1016/j.neuroscience.2019.06.020. [DOI] [PubMed] [Google Scholar]
- 15.Li Y, Zhong W, Jiang Z, Tang X. New progress in the approaches for blood-brain barrier protection in acute ischemic stroke. Brain Res Bull. 2019b;144:46–57. doi: 10.1016/j.brainresbull.2018.11.006. [DOI] [PubMed] [Google Scholar]
- 16.Liu X, Liu R, Fu D, Wu H, Zhao X, Sun Y, Wang M, Pu X. Dl-3-n-butylphthalide inhibits neuroinflammation by stimulating foxp3 and Ki-67 in an ischemic stroke model. Aging (Albany NY) 2021;13:3763–3778. doi: 10.18632/aging.202338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25:402–408. doi: 10.1006/meth.2001.1262. [DOI] [PubMed] [Google Scholar]
- 18.Ma YH, Deng WJ, Luo ZY, Jing J, Pan PW, Yao YB, Fang YB, Teng JF. Inhibition of microRNA-29b suppresses oxidative stress and reduces apoptosis in ischemic stroke. Neural Regen Res. 2022;17:433–439. doi: 10.4103/1673-5374.314319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Mamtilahun M, Wei Z, Qin C, Wang Y, Tang Y, Shen FX, Tian HL, Zhang Z, Yang GY. DL-3n-butylphthalide improves blood-brain barrier integrity in rat after middle cerebral artery occlusion. Front Cell Neurosci. 2020;14:610714. doi: 10.3389/fncel.2020.610714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Palazon A, Goldrath AW, Nizet V, Johnson RS. HIF transcription factors, inflammation, and immunity. Immunity. 2014;41:518–528. doi: 10.1016/j.immuni.2014.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Porter AG, Jänicke RU. Emerging roles of caspase-3 in apoptosis. Cell Death Differ. 1999;6:99–104. doi: 10.1038/sj.cdd.4400476. [DOI] [PubMed] [Google Scholar]
- 22.Prabhakaran S, Ruff I, Bernstein RA. Acute stroke intervention:a systematic review. JAMA. 2015;313:1451–1462. doi: 10.1001/jama.2015.3058. [DOI] [PubMed] [Google Scholar]
- 23.Que R, Zheng J, Chang Z, Zhang W, Li H, Xie Z, Huang Z, Wang HT, Xu J, Jin D, Yang W, Tan EK, Wang Q. Dl-3-n-butylphthalide rescues dopaminergic neurons in Parkinson's disease models by inhibiting the NLRP3 inflammasome and ameliorating mitochondrial impairment. Front Immunol. 2021;12:794770. doi: 10.3389/fimmu.2021.794770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Shi K, Tian DC, Li ZG, Ducruet AF, Lawton MT, Shi FD. Global brain inflammation in stroke. Lancet Neurol. 2019;18:1058–1066. doi: 10.1016/S1474-4422(19)30078-X. [DOI] [PubMed] [Google Scholar]
- 25.Tao T, Liu M, Chen M, Luo Y, Wang C, Xu T, Jiang Y, Guo Y, Zhang JH. Natural medicine in neuroprotection for ischemic stroke:Challenges and prospective. Pharmacol Ther. 2020;216:107695. doi: 10.1016/j.pharmthera.2020.107695. [DOI] [PubMed] [Google Scholar]
- 26.Wang S, Ma F, Huang L, Zhang Y, Peng Y, Xing C, Feng Y, Wang X, Peng Y. Dl-3-n-butylphthalide (NBP):a promising therapeutic agent for ischemic stroke. CNS Neurol Disord Drug Targets. 2018;17:338–347. doi: 10.2174/1871527317666180612125843. [DOI] [PubMed] [Google Scholar]
- 27.Wang Y, Shen Y, Liu Z, Gu J, Xu C, Qian S, Zhang X, Zhou B, Jin Y, Sun Y. Dl-NBP (Dl-3-N-butylphthalide) treatment promotes neurological functional recovery accompanied by the upregulation of white matter integrity and HIF-1α/VEGF/Notch/Dll4 expression. Front Pharmacol. 2019;10:1595. doi: 10.3389/fphar.2019.01595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Wu F, Xu K, Xu K, Teng C, Zhang M, Xia L, Zhang K, Liu L, Chen Z, Xiao J, Wu Y, Zhang H, Chen D. Dl-3n-butylphthalide improves traumatic brain injury recovery via inhibiting autophagy-induced blood-brain barrier disruption and cell apoptosis. J Cell Mol Med. 2020;24:1220–1232. doi: 10.1111/jcmm.14691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Zeng Z, Gong X, Hu Z. L-3-n-butylphthalide attenuates inflammation response and brain edema in rat intracerebral hemorrhage model. Aging (Albany NY) 2020;12:11768–11780. doi: 10.18632/aging.103342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Zhou X, Chen H, Wang L, Lenahan C, Lian L, Ou Y, He Y. Mitochondrial dynamics:a potential therapeutic target for ischemic stroke. Front Aging Neurosci. 2021;13:721428. doi: 10.3389/fnagi.2021.721428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Zhu G, Wang X, Chen L, Lenahan C, Fu Z, Fang Y, Yu W. Crosstalk between the oxidative stress and glia cells after stroke:from mechanism to therapies. Front Immunol. 2022;13:852416. doi: 10.3389/fimmu.2022.852416. [DOI] [PMC free article] [PubMed] [Google Scholar]

