Calycosin‑7‑O‑β‑D‑glucoside downregulates mitophagy by mitigating mitochondrial fission to protect HT22 cells from oxygen‑glucose deprivation/reperfusion‑induced injury
- Authors:
- Published online on: January 13, 2025 https://doi.org/10.3892/mmr.2025.13436
- Article Number: 71
-
Copyright: © Yan et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Stroke is the leading cause of mortality and disability worldwide (1). According to the World Health Organization, stroke causes ~5.9 million deaths annually, with ischemic stroke accounting for ~87% of these cases (2). Mechanical thrombectomy and early pharmacological thrombolysis using tissue plasminogen activator are clinically effective interventions for restoring cerebral blood flow (reperfusion) in ischemic stroke (3). However, reperfusion can trigger cerebral ischemia-reperfusion injury (CIRI), a form of secondary damage that critically affects clinical outcomes (4–6). CIRI is a complex pathological process characterized by excitotoxicity, oxidative stress, inflammation and mitochondrial dysfunction, eventually triggering neuronal apoptosis (7). As such, there is an urgent need for effective therapeutic strategies for CIRI.
Mitochondria maintain cellular homeostasis through the induction of dynamic processes, including fission, fusion, autophagy and regeneration (8). Disturbances in these processes, particularly mitochondrial fission, facilitate neuronal apoptosis (9). Excessive mitochondrial fission is a prominent early upstream event that drives neuronal apoptosis during CIRI (10). Dynamin-related protein 1 (Drp1), a critical regulator of mitochondrial fission, is primarily activated by the phosphorylation of serine 616, which promotes its translocation from the cytoplasm to the mitochondrial membrane, triggering mitochondrial fission (11). Inhibition of Drp1 phosphorylation by pharmacological inhibition or Drp1 knockdown has been shown to alleviate CIRI by reducing infarct size and neuronal apoptosis (12,13).
Drp1-mediated excessive fission not only causes mitochondrial fragmentation, reactive oxygen species (ROS) production, reduced mitochondrial membrane potential (MMP) and increased cytochrome c release, but also leads to the overactivation of mitophagy (14–17). Although previous studies have shown that mitophagy exerts neuroprotective effects and improves the prognosis of CIRI (18,19), several studies have demonstrated that type II programmed cell death, induced by overactivated mitophagy, aggravates brain damage (20–22). Peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) is a multifunctional co-transcription factor involved in mitochondrial biosynthesis and function (23), which negatively regulates Drp1 expression by directly binding to the Drp1 promoter, thereby preventing excessive mitochondrial fission (24). As a direct upstream regulator of PGC-1α, sirtuin 1 (SIRT1) serves a crucial role in maintaining mitochondrial function by regulating its expression (25). Previous studies have demonstrated that activation of the SIRT1/PGC-1α pathway can inhibit excessive mitochondrial fission and attenuate the development of diabetes-induced cardiac dysfunction (24,26). Moreover, the inhibition of Drp1-mediated excessive mitochondrial fission has been shown to mitigate mitophagy, thereby offering protection against CIRI (27).
Astragali Radix (Huangqi) is a widely used traditional Chinese medicine, which exhibits potent pharmacological effects on cardiovascular diseases and immune regulation (28). Calycosin-7-O-β-D-glucoside (CG), a major bioactive ingredient of Astragali Radix, has further been reported to exert neuroprotective effects through anti-inflammatory, antioxidant and anti-apoptotic mechanisms (29–31). Although our previous study showed that CG can alleviate oxygen-glucose deprivation/reperfusion (OGD/R)-induced HT22 cell damage (32), whether this action is related to the prevention of excessive mitochondrial fission and mitophagy overactivation remains unclear. Therefore, the present study investigated the protective effects of CG against OGD/R-induced injury by focusing on the inhibition of Drp1-mediated mitochondrial fission. Additionally, the study investigated whether CG could inhibit the overactivation of mitophagy.
Materials and methods
Materials
The following reagents were acquired for use in the present study: CG (Fig. 1A; purity >98%; Shanghai Aladdin Biochemical Technology Co., Ltd.); edaravone (EDA; purity >99%; MilliporeSigma); Cell Counting Kit-8 (CCK-8; Abbkine Scientific Co., Ltd.); lactate dehydrogenase (LDH) kit (Nanjing Jiancheng Bioengineering Institute); MitoTracker™ Orange CMTMRos kit (Thermo Fisher Scientific, Inc.); MitoSOX™ kit (Thermo Fisher Scientific, Inc.); JC-1, MitoTracker Green kits and Hoechst 33342 (Beyotime Institute of Biotechnology); BCA kit (Beijing Solarbio Science & Technology Co., Ltd.); Dulbecco's modified Eagle's medium (DMEM), fetal bovine serum (FBS) and penicillin/streptomycin (HyClone; Cytiva); and glucose-free DMEM (Gibco; Thermo Fisher Scientific, Inc.). The following antibodies were also purchased: SQSTM1/p62 (cat. no. P0067; MilliporeSigma); LC3B (cat. no. A19665), Bcl-2 (cat. no. A19693), Drp1 (cat. no. A21968) and HRP-conjugated secondary antibody (cat. no. AS014) (all from ABclonal Biotech Co., Ltd.); translocase of outer mitochondrial membrane 20 (TOM20; cat. no. 42406S), phosphorylated p-Drp1 (Ser616) (cat. no. 4494S) and caspase-3 (cat. no. 14220S) (all from Cell Signaling Technology, Inc.); PGC-1α (cat. no. ab313559), Bax (cat. no. ab32503) and SIRT1 (cat. no. ab12193) (all from Abcam); β-actin (cat. no. K101527P; Beijing Solarbio Science & Technology Co., Ltd.).
OGD/R model and drug treatment
HT22 mouse hippocampal neurons were purchased from Pricella (cat. no. CL-0697), and were cultured in DMEM supplemented with 10% FBS and antibiotics (penicillin, 100 IU/ml; streptomycin, 100 µg/ml) at 37°C in an incubator under 5% CO2. To construct the OGD/R model, cells were cultured in glucose-free DMEM and then transferred to a hypoxia incubator chamber (MIC-101; Embrient, Inc.) containing 5% CO2 and 95% N2 at 37°C. After 10 h, the cells were transferred to an incubator with 5% CO2 and 95% air, and the culture medium was replaced with standard DMEM for 6 h at 37°C (reperfusion). CG (20, 40 and 80 µM) and EDA (50, 100 and 200 µM) were added for 6 h during the reperfusion phase. The control group was maintained in standard culture medium without OGD. The concentration of CG was selected based on a previous study (31).
Cell viability assay
HT22 cells were cultured in 96-well plates at a density of 5×103 cells/well. After OGD/R modelling and drug treatment, images were captured under an optical microscope (Nikon Corporation), after which the medium was discarded, 10 µl CCK-8 solution was added, and the cells were cultured for 30 min in a regular incubator. The absorbance was measured at 450 nm using a microplate reader (BioTek; Agilent Technologies, Inc.).
Measurement of LDH activity
The LDH activity in the supernatant was measured using an LDH assay kit, according to the manufacturer's instructions. After OGD/R modelling and drug treatment, the cell medium was collected and mixed with substrate solution at 37°C for 15 min. The mixture was subsequently incubated with 2,4-dinitrophenylhydrazine for 15 min at 37°C and the absorbance was measured at 450 nm using a microplate reader (BioTek; Agilent Technologies, Inc.).
Cell apoptosis assay
Cell apoptosis was determined using an Annexin V-FITC/PI kit (Beijing Solarbio Science & Technology Co., Ltd.), according to the manufacturer's protocol. After treatment, the cells were harvested by centrifugation at 100 × g for 5 min at 37°C, followed by trypsin digestion. The cells were then resuspended in binding buffer mixed with 5 µl Annexin V-FITC and were incubated in the dark for 5 min at 37°C. Subsequently, 1 µl PI was added and incubated for another 5 min at 37°C. The apoptosis rate in each group was analyzed using flow cytometry (CytoFLEX V2-B2-R0; Beckman Coulter, Inc.).
Mitochondrial ROS (mtROS) detection
According to the manufacturer's instructions, following treatment, the supernatants were discarded, and the cells were incubated with MitoSOX™ (3 µM) for 30 min at 37°C, followed by Hoechst staining for 15 min at 37°C to label the nuclei. The cells were then washed three times with PBS and observed under an inverted fluorescence microscope (TS-2; Nikon Corporation).
MMP detection
JC-1 is a widely used probe for monitoring MMP. In healthy cells with normal MMP levels, JC-1 forms J-aggregates that emit intense red fluorescence. By contrast, in apoptotic or unhealthy cells with a low MMP, JC-1 remains in its monomeric form and emits green fluorescence. Thus, mitochondrial depolarization can be measured as a decrease in the red/green fluorescence intensity ratio. According to the manufacturer's protocol, following treatment, the supernatants were discarded, and cells were incubated with 1 ml JC-1 solution at 37°C for 50 min. The JC-1 solution was then aspirated and the cells were washed twice with dye buffer. The fluorescence intensity of the JC-1 aggregate/monomer was subsequently observed under an inverted fluorescence microscope (TS-2; Nikon Corporation).
Mitochondrial morphology observation
MitoTracker Green, a live cell mitochondria-specific fluorescent dye, was applied to distinguish changes in mitochondrial morphology. Briefly, HT22 cells were incubated with 100 nM MitoTracker Green for 15 min at 37°C. The staining solution was subsequently removed and the cells were washed twice with HBSS (Beyotime Institute of Biotechnology). Images were then captured using a laser confocal microscope (Leica Microsystems GmbH) and the length of the mitochondria was measured using Image-Pro Plus 6.0 (Media Cybernetics, Inc.).
Immunofluorescence
For immunofluorescence detection, HT22 cells were first stained with MitoTracker™ Orange CMTMRos (200 nM) at 37°C for 10 min, and then fixed with 4% paraformaldehyde for 15 min at 37°C. After three washes with PBS, the cells were permeabilized with 0.3% Triton X-100 in PBS for 15 min at 37°C. The cells were then blocked with 5% BSA (Beijing Solarbio Science & Technology Co., Ltd.) for 30 min at 37°C, and incubated overnight at 4°C with primary antibodies against LC3 (1:500). The next day, after washing three times with PBS, the samples were incubated with a Alexa Fluor™ 488-conjugated goat anti-rabbit IgG secondary antibody (1:500; cat. no. A11008; Thermo Fisher Scientific, Inc.) for 1 h at room temperature in the dark, followed by DAPI staining for 30 min at 37°C to label the nuclei. Finally, the fluorescence intensity was analyzed using an inverted fluorescence microscope (TS-2; Nikon Corporation).
Molecular docking
The 3D structure of SIRT1 [Protein Data Bank (PDB) ID: 4KXQ] was downloaded from the PDB (https://www.rcsb.org/), and protein structure was optimized to ensure it can be used for molecular docking using the Protein Preparation Wizard module in Schrödinger 12.9 (Schrödinger, Inc.). The 3D structure of CG was obtained from the PubChem database (compound CID: 5318267; http://pubchem.ncbi.nlm.nih.gov/compound/5318267). Molecular docking between CG and SIRT1 was performed using the Glide module (Schrödinger, Inc.), and the molecular mechanics generalized Born surface area (MM/GBSA) was calculated. The binding sites were visualized using PyMOL 3.0 (Schrödinger, Inc.).
Western blotting
Total proteins were extracted from the cells using RIPA lysis buffer (cat. no. CW2333S; CWBio), whereas protein concentration was detected using a BCA kit. Proteins (10 µg) were separated by SDS-PAGE on 10 or 12% gels (the percentage gel used depended on the molecular weight of the targeted protein), were transferred to polyvinylidene difluoride membranes and were blocked for 2 h with 5% skim milk in PBS containing 0.1% Tween-20 (PBST) at 37°C. The membranes were subsequently incubated with primary antibodies against the following proteins at 4°C overnight: Bax (1:2,000), Bcl-2 (1:500), caspase-3 (1:4,000), TOM20 (1:4,000), p62 (1:4,000), LC3 (1:1,500), Drp1 (1:1,000), p-Drp1 (1:1,000), PGC-1α (1:1,000), SIRT1 (1:1,000) and β-actin (1:5,000). After washing three times with PBST, the membranes were incubated with an HRP-conjugated secondary antibody for 50 min at room temperature. The membranes were then scanned using an imaging system (Bio-Rad Laboratories, Inc.), and Image-Pro Plus 6.0 (Media Cybernetics, Inc.) was used to analyze the optical density of the bands. The relative protein expression levels were normalized to β-actin.
Statistical analysis
The data are presented as the mean ± SEM and were analyzed using SPSS 27.0 (IBM Corporation). Figures were drawn using GraphPad Prism 8.0 (Dotmatics). One-way ANOVA followed by Tukey's post hoc test was used for data analysis. P<0.05 was considered to indicate a statistically significant difference.
Results
CG ameliorates damage to HT22 cells induced by OGD/R challenge
To investigate the protective effects of CG against OGD/R-induced HT22 cell injury, the viability of HT22 cells was measured using the CCK-8 assay. The results showed that cell viability was significantly decreased in the OGD/R group compared with that in the control group, but it was significantly increased by CG (40 µM) and EDA (100 and 200 µM) treatment (Fig. 1B); therefore, 40 µM CG and 100 µM EDA were selected for subsequent experiments. The morphological characteristics of the OGD/R group included irregular shrinkage, unclear boundaries and floating (Fig. 1C). Additionally, the release of LDH was markedly increased in the OGD/R group compared with that in the control group (Fig. 1D). However, these trends were markedly reversed by CG and EDA.
CG alleviates the OGD/R-induced apoptosis of HT22 cells
Neuronal apoptosis is the final event of CIRI (33). To assess the effects of CG on the apoptosis of HT22 cells, flow cytometry and western blot analysis were performed. The results revealed a significant increase in the percentage of apoptotic cells and caspase-3 expression, accompanied by a marked decrease in the Bcl-2/Bax ratio in the OGD/R group compared with those in the control group (Fig. 2). However, these changes were reversed by the CG and EDA treatment. These findings confirmed the anti-apoptotic effect of CG and indicated that CG ameliorated OGD/R by reducing neuronal apoptosis.
CG improves the OGD/R-induced mitochondrial dysfunction of HT22 cells
Mitochondrial dysfunction is an important characteristic of CIRI (22,34). To investigate whether CG could restore the mitochondrial dysfunction caused by OGD/R, mtROS levels were analyzed using MitoSOX Red. As shown in Fig. 3A and B, mtROS levels were significantly increased in the OGD/R group compared with those in the control group, but were notably reduced following CG and EDA treatment. By contrast, MMP, a hallmark of mitochondrial integrity, was significantly elevated by CG and EDA treatment compared with that in the OGD/R group (Fig. 3C and D). Collectively, these results indicated that CG may protect cells from OGD/R-induced mitochondrial dysfunction.
CG prevents OGD/R-induced excessive mitochondrial fission in HT22 cells
Mitochondria are highly dynamic organelles that undergo fission and fusion, both of which are processes closely related to mitochondrial function (8). The MitoTracker Green probe was used to examine the effects of CG on mitochondrial morphology in OGD/R-treated HT22 cells. As shown in Fig. 4A and B, the average length of mitochondria in OGD/R-treated cells was significantly shorter than that in control cells, indicating a marked decrease in the linear shape and diffusely interspersed fragmented or dotted mitochondria. As hypothesized, CG and EDA mitigated mitochondrial fragmentation in OGD/R-treated HT22 cells. Mitochondrial fission is mediated by p-Drp1 (Ser616); to confirm whether CG regulated mitochondrial fission, the expression levels of this marker of mitochondrial fission were examined. As shown in Fig. 4C and D, an increased p-Drp1/Drp1 ratio was observed in the OGD/R group compared with that in the control group, which was effectively interrupted by CG and EDA treatment. These results indicated the protective effect of CG against OGD/R-induced mitochondrial fission.
CG downregulates OGD/R-induced mitophagy overactivation
To investigate whether CG can affect mitophagy, the expression levels of the mitophagy-related proteins LC3, p62 and TOM20 were investigated (Fig. 5A). The results showed an increased LC3II/LC3I ratio in the OGD/R group compared with that in the control group, and, as expected, this increase was markedly inhibited by CG treatment (Fig. 5D). By contrast, a significant increase in the expression levels of p62 and TOM20 were observed following CG and EDA administration, indicating that CG may reduce the overactivation of mitophagy induced by OGD/R (Fig. 5B and C). To further observe overactivated mitophagy, mitochondria and autophagosomes were colocalized using immunofluorescence staining. As presented in Fig. 5E and F, the ratio of colocalization of mitochondria stained with LC3 was greatly increased in the OGD/R group compared with that in the control group, and was markedly abrogated by treatment with CG and EDA. Taken together, these results indicated that CG downregulated OGD/R-induced mitophagy overactivation.
CG upregulates the expression of SIRT1 and PGC-1α after OGD/R
The SIRT1/PGC-1α signaling pathway serves an essential role in mitochondrial protection (24). Thus, it was hypothesized that the SIRT1/PGC-1α pathway may be involved in the protective effect of CG against neuronal apoptosis by inhibiting mitophagy overactivation. To assess whether SIRT1 is a target of CG, the affinity between CG and SIRT1 was analyzed using molecular docking. As presented in Fig. 6A, CG can bind well with amino acids in SIRT1, including Asn465, Glu467, Gly263, Arg274 and Phe273, and the docking score of CG and SIRT1 was −8.02 kcal/mol. Furthermore, the effects of CG on the protein expression levels of SIRT1 and PGC-1α were examined, with results showing that the levels of SIRT1 and PGC-1α were markedly reduced in the OGD/R group compared with those in the control group, while CG and EDA treatment upregulated the expression of SIRT1 and PGC-1α (Fig. 6B-D). These results indicated that CG may attenuate mitochondrial dysfunction through the expression of SIRT1 and PGC-1α.
Discussion
High rates of disability and recurrence are characteristic of ischemic stroke, a common cerebral disease that represents a serious global public health concern (35). According to our previous study, CG may mitigate OGD/R-induced injury by improving mitochondrial function (32). Notably, abundant evidence has demonstrated that mitophagy overactivation, triggered by excessive mitochondrial fission, is central to neuronal apoptosis following cerebral ischemia/reperfusion (36,37). However, whether the protective effects of CG against OGD/R-induced injury are related to the modulation of mitochondrial fission and mitophagy has not yet been investigated, to the best of our knowledge.
The results of the present study indicated that CG notably enhanced cell viability, increased the Bcl-2/Bax ratio, and decreased caspase-3 expression and LDH release following OGD/R, thus validating the protective effects of CG against OGD/R-induced neuronal apoptosis. It has previously been well established that mitochondrial dysfunction damages neurons and is a major factor in the etiology of CIRI (22). One of the main causes of CIRI is the overproduction of mtROS (6). This process contributes to and permeates all aspects of CIRI, including oxidative stress, mitochondrial swelling, membrane instability and reduced MMP (38). Furthermore, cerebral ischemia-reperfusion causes mitochondrial morphological fragmentation, which ultimately triggers neurological injury (39,40). Improving mitochondrial morphology has been shown to effectively reduce the damage caused by CIRI (39,41). EDA was first approved for the treatment of ischemic stroke in Japan in 2001 (42). Notably, ~50% of EDA is present in the body as the EDA anion, which transfers electrons to remove different types of radicals, such as H2O2 and O-2. The produced EDA radical at that time then combines with the oxygen molecules in the reaction system to form EDA peroxyradical, and ultimately 2-oxo-(phenylhydrazono)-butanoic acid, a reaction product unrelated to radicals (43–45). Thus, in order to assess the effectiveness and mechanism of CG, EDA was selected as a positive control medication. The present study verified that CG protected the mitochondria of OGD/R-induced HT22 cells, as demonstrated by decreased mtROS, increased MMP and improved mitochondrial morphology. The present data further suggested that CG may reduce OGD/R-induced damage by improving mitochondrial dysfunction.
Under normal physiological conditions, Drp1 is typically found in the cytoplasm and is an essential regulator of mitochondrial fission. Upon stimulation, Drp1 is recruited to the mitochondrial surface to induce fission (11). Previous studies have shown that when Drp1 is translocated to the mitochondria, it releases Beclin1 and interacts with LC3 to activate autophagy (16,46). Excessive mitochondrial fission during cerebral ischemia-reperfusion triggers mitophagy, a crucial process in maintaining mitochondrial quality control (47,48). Although mitophagy is essential for cellular homeostasis, it is a double-edged sword and its overactivation can lead to apoptosis (49). Prior research has shown that preventing mitophagy overactivation caused by excessive mitochondrial fission can alleviate CIRI (39,40,50). In the present study, excessive mitochondrial fission and mitophagy overactivation were detected following OGD/R, as evidenced by the upregulation of p-Drp1/Drp1 and LC3II/LC3I, and decreased p62 and TOM20 expression. As expected, treatment with CG reversed the changes induced by OGD/R.
SIRT1 is the most researched member of the SIRT family (51), with numerous studies showing that SIRT1 serves a role in autophagy and apoptosis following cerebral ischemia (52–54). PGC-1α, a transcription factor downstream of SIRT1, is crucial for controlling mitochondria, and may be used as a therapeutic target for a variety of neurodegenerative illnesses (23). To explore whether the downregulation of p-Drp1 by CG is related to the expression of SIRT1 and PGC-1α, the binding ability of CG to SIRT1 was simulated through molecular docking, and the protein expression levels of SIRT1 and PGC-1α were detected. The results of these analyses demonstrated that CG has the ability to directly bind to SIRT1, and regulate the expression of SIRT1 and PGC-1α. These results indicated that CG may reduce Drp1 phosphorylation via regulation of the expression of SIRT1 and PGC-1α, thereby preventing the overactivation of mitophagy induced by excessive mitochondrial fission.
In conclusion, the present study provided compelling evidence that CG alleviated OGD/R-induced HT22 cell apoptosis by preventing the overactivation of mitophagy induced by excessive mitochondrial fission by regulating the expression of SIRT1 and PGC-1α (Fig. 7). These findings provide a theoretical basis for the development of clinical strategies for the use of CG in the management of CIRI. However, the current study only focused on the protective effects of CG in vitro; the precise mechanism by which CG protects against CIRI through the regulation of mitochondrial fission and mitophagy, and how excessive mitochondrial fission leads to mitophagy overactivation, still requires further elucidation in vivo. Furthermore, establishing the pharmacokinetics and ideal dosage of CG in the treatment of ischemic stroke is essential for its possible integration into clinical practice.
Acknowledgements
The authors would like to thank Dr Yong Yuan (Academy of Chinese Medicine of Henan University of Chinese Medicine) for their technological help with laser confocal microscopy.
Funding
This study was supported by the National Natural Science Foundation of China (grant nos. 82304771 and 82274496), the Joint Fund of Science and Technology Research and Development Project of Henan Province (grant no. 232301420018) and the Science and Technology Research Project of Henan Province (grant nos. 232102311193 and 232102310419).
Availability of data and materials
The data generated in the present study may be requested from the corresponding author.
Authors' contributions
XY designed the experiments. SQ and RG performed the experiments. XY and SQ wrote the original draft. ZL, MB and BW analyzed the data. PS analyzed the data and confirmed the version to be published. EX and YL designed the experiments, drafted the manuscript and confirm the authenticity of all the raw data. All authors contributed to the critical revision of the manuscript. All authors have read and approved the final manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Glossary
Abbreviations
Abbreviations:
CIRI |
cerebral ischemia-reperfusion injury |
OGD/R |
oxygen-glucose deprivation/reperfusion |
CG |
calycosin-7-O-β-D-glucoside |
EDA |
edaravone |
mtROS |
mitochondrial reactive oxygen species |
MMP |
mitochondrial membrane potential |
Drp1 |
dynamin-related protein 1 |
PGC-1α |
peroxisome proliferator-activated receptor γ coactivator-1α |
SIRT1 |
sirtuin 1 |
References
Saini V, Guada L and Yavagal DR: Global epidemiology of stroke and access to acute ischemic stroke interventions. Neurology. 97 (20 Suppl 2):S6–S16. 2021. View Article : Google Scholar : PubMed/NCBI | |
Iadecola C, Buckwalter MS and Anrather J: Immune responses to stroke: Mechanisms, modulation, and therapeutic potential. J Clin Invest. 130:2777–2788. 2020. View Article : Google Scholar : PubMed/NCBI | |
Jolugbo P and Ariëns RAS: Thrombus composition and efficacy of thrombolysis and thrombectomy in acute ischemic stroke. Stroke. 52:1131–1142. 2021. View Article : Google Scholar : PubMed/NCBI | |
Ma R, Xie Q, Li Y, Chen Z, Ren M, Chen H, Li H, Li J and Wang J: Animal models of cerebral ischemia: A review. Biomed Pharmacother. 131:1106862020. View Article : Google Scholar : PubMed/NCBI | |
Paul S and Candelario-Jalil E: Emerging neuroprotective strategies for the treatment of ischemic stroke: An overview of clinical and preclinical studies. Exp Neurol. 335:1135182021. View Article : Google Scholar : PubMed/NCBI | |
Zeng X, Zhang YD, Ma RY, Chen YJ, Xiang XM, Hou DY, Li XH, Huang H, Li T and Duan CY: Activated Drp1 regulates p62-mediated autophagic flux and aggravates inflammation in cerebral ischemia-reperfusion via the ROS-RIP1/RIP3-exosome axis. Mil Med Res. 9:252022.PubMed/NCBI | |
Zhang M, Liu Q, Meng H, Duan H, Liu X, Wu J, Gao F, Wang S, Tan R and Yuan J: Ischemia-reperfusion injury: Molecular mechanisms and therapeutic targets. Signal Transduct Target Ther. 9:122024. View Article : Google Scholar : PubMed/NCBI | |
Song J, Herrnann JM and Becker T: Quality control of the mitochondrial proteome. Nat Rev Mol Cell Biol. 22:54–70. 2020. View Article : Google Scholar : PubMed/NCBI | |
Hou Y, Fan F, Xie N, Zhang Y, Wang X and Meng X: Rhodiola crenulata alleviates hypobaric hypoxia-induced brain injury by maintaining BBB integrity and balancing energy metabolism dysfunction. Phytomedicine. 128:1555292024. View Article : Google Scholar : PubMed/NCBI | |
Anaell AR, Maizy R, Przyklenk K and Sanderson TH: Mitochondrial quality control and disease: Insights into ischemia-reperfusion injury. Mol Neurobiol. 55:2547–2564. 2018. View Article : Google Scholar : PubMed/NCBI | |
Duan CY, Wang L, Zhang J, Xiang X, Wu Y, Zhang Z, Li Q, Tian K, Xue M, Liu L and Li T: Mdivi-1 attenuates oxidative stress and exerts vascular protection in ischemic/hypoxic injury by a mechanism independent of Drp1 GTPase activity. Redox Biol. 37:1017062020. View Article : Google Scholar : PubMed/NCBI | |
Xu Y, Wang Y, Wang G, Ye X, Zhang J, Cao G, Zhao Y, Gao Z, Zhang Y, Yu B and Kou J: YiQiFuMai powder injection protects against ischemic stroke via inhibiting neuronal apoptosis and PKCδ/Drp1-mediated excessive mitochondrial fission. Oxid Med Cell Longev. 2017:18320932017. View Article : Google Scholar : PubMed/NCBI | |
Wu B, Luo H, Zhou X, Cheng CY, Lin L, Liu BL, Liu K, Li P and Yang H: Succinate-induced neuronal mitochondrial fission and hexokinase II malfunction in ischemic stroke: Therapeutical effects of kaempferol. Biochim Biophys Acta Mol Basis Dis. 1863:2307–2318. 2017. View Article : Google Scholar : PubMed/NCBI | |
Mao K and Klionsky DJ: Mitochondrial fission facilitates mitophagy in Saccharomyces cerevisiae. Autophagy. 9:1900–1901. 2013. View Article : Google Scholar : PubMed/NCBI | |
Vásquez-Trincado C, García-Carvajal I, Pennanen C, Parra V, Hill JA, Rothermel BA and Lavandero S: Mitochondrial dynamics, mitophagy and cardiovascular disease. J Physiol. 594:509–525. 2016. View Article : Google Scholar : PubMed/NCBI | |
Shirihai OS, Song M and Dorn GW II: How mitochondrial dynamism orchestrates mitophagy. Circ Res. 116:1835–1849. 2015. View Article : Google Scholar : PubMed/NCBI | |
Ikeda Y, Shirakabe A, Maejima Y, Zhai P, Sciarretta S, Toli J, Nomura M, Mihara K, Egashira K, Ohishi M, et al: Endogenous Drp1 mediates mitochondrial autophagy and protects the heart against energy stress. Circ Res. 116:264–278. 2015. View Article : Google Scholar : PubMed/NCBI | |
Cai Y, Yang E, Yao X, Zhang X, Wang Q, Wang Y, Liu J, Fan W, Yi K, Kang C and Wu J: FUNDC1-dependent mitophagy induced by tPA protects neurons against cerebral ischemia-reperfusion injury. Redox Biol. 38:1017922021. View Article : Google Scholar : PubMed/NCBI | |
Wu Q, Liu J, Mao Z, Tian L, Wang N, Wang G, Wang Y and Seto S: Ligustilide attenuates ischemic stroke injury by promoting Drp1-mediated mitochondrial fission via activation of AMPK. Phytomedicine. 95:1538842022. View Article : Google Scholar : PubMed/NCBI | |
Li TH, Sun HW, Song LJ, Yang B, Zhang P, Yan DM, Liu XZ and Luo YR: Long non-coding RNA MEG3 regulates autophagy after cerebral ischemia/reperfusion injury. Neural Regen Res. 17:824–831. 2022. View Article : Google Scholar : PubMed/NCBI | |
Ling J, Cai H, Lin M, Qi S, Du J and Chen L: RTN1-C mediates cerebral ischemia/reperfusion injury via modulating autophagy. Acta Biochim Biophys Sin (Shanghai). 53:170–178. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yuan ZL, Mo YZ, Li DL, Xie L and Chen MH: Inhibition of ERK downregulates autophagy via mitigating mitochondrial fragmentation to protect SH-SY5Y cells from OGD/R injury. Cell Commun Signal. 21:2042023. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Xu E, Musich PR and Lin F: Mitochondrial dysfunction in neurodegenerative diseases and the potential countermeasure. CNS Neurosci Ther. 25:816–824. 2019. View Article : Google Scholar : PubMed/NCBI | |
Ding M, Feng N, Tang D, Feng J, Li Z, Jia M, Liu Z, Gu X, Wang Y, Fu F and Pei J: Melatonin prevents Drp1-mediated mitochondrial fission in diabetic hearts through SIRT1-PGC1α pathway. J Pineal Res. 65:1–16. 2018. View Article : Google Scholar | |
Lei MY, Cong L, Liu ZQ, Liu ZF, Ma Z, Liu K, Li J, Deng Y, Liu W and Xu B: Resveratrol reduces DRP1-mediated mitochondrial dysfunction via the SIRT1-PGC1α signaling pathway in manganese-induced nerve damage in mice. Environ Toxicol. 37:282–298. 2022. View Article : Google Scholar : PubMed/NCBI | |
Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM and Puigserver P: Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1. Nature. 434:113–118. 2005. View Article : Google Scholar : PubMed/NCBI | |
Tang J, Hu Z, Tan J, Yang S and Zeng L: Parkin protects against oxygen-glucose deprivation/reperfusion insult by promoting Drp1 degradation. Oxid Med Cell Longev. 2016:84743032016. View Article : Google Scholar : PubMed/NCBI | |
Li M, Han B, Zhao H, Xu C, Xu D, Sieniawska E, Lin X and Kai G: Biological active ingredients of Astragali Radix and its mechanisms in treating cardiovascular and cerebrovascular diseases. Phytomedicine. 98:1539182022. View Article : Google Scholar : PubMed/NCBI | |
Xu W, Zhou F, Zhu Q, Bai M, Luo T, Zhou L and Deng R: Calycosin-7-O-β-D-glucoside attenuates palmitate-induced lipid accumulation in hepatocytes through AMPK activation. Eur J Pharmacol. 925:1749882022. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Che G, Di Z, Sun W, Tian J and Ren M: Calycosin-7-O-β-D-glucoside attenuates myocardial ischemia-reperfusion injury by activating JAK2/STAT3 signaling pathway via the regulation of IL-10 secretion in mice. Mol Cell Biochem. 463:175–187. 2019. View Article : Google Scholar : PubMed/NCBI | |
Park KR, Park JE, Kim B, Kwon IK, Hong JT and Yun HM: Calycosin-7-O-β-glucoside isolated from astragalus membranaceus promotes osteogenesis and mineralization in human mesenchymal stem cells. Int J Mol Sci. 22:113622021. View Article : Google Scholar : PubMed/NCBI | |
Yan X, Yu A, Zheng H, Wang S, He Y and Wang L: Calycosin-7-O-β-D-glucoside attenuates OGD/R-induced damage by preventing oxidative stress and neuronal apoptosis via the SIRT1/FOXO1/PGC-1α pathway in HT22 cells. Neural Plast. 2019:87980692019. View Article : Google Scholar : PubMed/NCBI | |
Xu S, Huang P, Yang J, Du H, Wan H and He Y: Calycosin alleviates cerebral ischemia/reperfusion injury by repressing autophagy via STAT3/FOXO3a signaling pathway. Phytomedicine. 115:1548452023. View Article : Google Scholar : PubMed/NCBI | |
Mao Z, Tia L, Liu J, Wu Q, Wang N, Wang G, Wang Y and Seto S: Ligustilide ameliorates hippocampal neuronal injury after cerebral ischemia reperfusion through activating PINK1/Parkin-dependent mitophagy. Phytomedicine. 101:1541112022. View Article : Google Scholar : PubMed/NCBI | |
Feske SK: Ischemic stroke. Am J Med. 134:1457–1464. 2021. View Article : Google Scholar : PubMed/NCBI | |
Quintana DD, Garcia JA, Sarkar SN, Jun S, Engler-Chiurazzi EB, Russell AE, Cavendish JZ and Simpkins JW: Hypoxia-reoxygenation of primary astrocytes results in a redistribution of mitochondrial size and mitophagy. Mitochondrion. 47:244–255. 2019. View Article : Google Scholar : PubMed/NCBI | |
Sanderson TH, Raghunayakula S and Kumar R: Neuronal hypoxia disrupts mitochondrial fusion. Neuroscience. 301:71–78. 2015. View Article : Google Scholar : PubMed/NCBI | |
Xu L, Gao Y, Hu M, Dong Y, Xu J, Zhang J and Lv P: Edaravone dexborneol protects cerebral ischemia reperfusion injury through activating Nrf2/HO-1 signaling pathway in mice. Fundam Clin Pharmacol. 36:790–800. 2022. View Article : Google Scholar : PubMed/NCBI | |
Grohm J, Kin SW, Mamrak U, Tobaben S, Cassidy-Stone A, Nunnari J, Plesnila N and Culmsee C: Inhibition of Drp1 provides neuroprotection in vitro and in vivo. Cell Death Differ. 19:1446–1458. 2012. View Article : Google Scholar : PubMed/NCBI | |
Zhao YX, Cui M, Chen SF, Dong Q and Liu XY: Amelioration of ischemic mitochondrial injury and Bax-dependent outer membrane permeabilization by Mdivi-1. CNS Neurosci Ther. 20:528–538. 2014. View Article : Google Scholar : PubMed/NCBI | |
Guo X, Sesaki H and Qi X: Drp1 stabilizes p53 on the mitochondria to trigger necrosis under oxidative stress conditions in vitro and in vivo. Biochem J. 461:137–146. 2014. View Article : Google Scholar : PubMed/NCBI | |
Higashi Y, Jitsuiki D, Chayama K and Yoshizumi M: Edaravone (3-methyl-1-phenyl-2-pyrazolin-5-one), a novel free radical scavenger, for treatment of cardiovascular diseases. Recent Pat Cardiovasc Drug Discov. 1:85–93. 2006. View Article : Google Scholar : PubMed/NCBI | |
Yang DX, Li Y, Yu D, Guan B, Ming Q, Li Y and Chen LQ: Human urinary kallidinogenase combined with edaravone in treating acute ischemic stroke patients: A meta-analysis. Brain Behav. 11:e24312021. View Article : Google Scholar : PubMed/NCBI | |
Watanabe T, Tanaka M, Watanabe K, Takamatsu Y and Tobe A: Research and development of the free radical scavenger edaravone as a neuroprotectant. Yakugaku Zasshi. 124:99–111. 2004.(In Japanese). View Article : Google Scholar : PubMed/NCBI | |
Matsumoto S, Murozono M, Kanazawa M, Nara T, Ozawa T and Watanabe Y: Edaravone and cyclosporine A as neuroprotective agents for acute ischemic stroke. Acute Med Surg. 5:213–221. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wu H, Li G, Chen W, Luo W, Yang Z, You Z and Zou Y: Drp1 knockdown represses apoptosis of rat retinal endothelial cells by inhibiting mitophagy. Acta Histochem. 124:1518372022. View Article : Google Scholar : PubMed/NCBI | |
Feng J, Chen X, Guan B, Li C, Qiu J and Shen J: Inhibition of peroxynitrite-induced mitophagy activation attenuates cerebral ischemia-reperfusion injury. Mol Neurobiol. 55:6369–6386. 2018. View Article : Google Scholar : PubMed/NCBI | |
Feng J, Chen X, Lu S, Li W, Yang D, Su W, Wang X and Shen J: Naringin attenuates cerebral ischemia-reperfusion injury through inhibiting peroxynitrite-mediated mitophagy activation. Mol Neurobiol. 55:9029–9042. 2018. View Article : Google Scholar : PubMed/NCBI | |
Li L, Yang S, Lu X, Zhang Y and Li T: Research progress on the mechanism of mitochondrial autophagy in cerebral stroke. Front Aging Neurosci. 13:6986012021. View Article : Google Scholar : PubMed/NCBI | |
Yuan Y, Zhang X, Zheng Y and Chen Z: Regulation of mitophagy in ischemic brain injury. Neurosci Bull. 31:395–406. 2015. View Article : Google Scholar : PubMed/NCBI | |
Wu J, Zhang D, Chen L, Li J, Wang J, Ning C, Yu N, Zhao F, Chen D, Chen X, et al: Discovery and mechanism study of SIRT1 activators that promote the deacetylation of fluorophore-labeled substrate. J Med Chem. 56:761–780. 2013. View Article : Google Scholar : PubMed/NCBI | |
Tang H, Wen J, Qin T, Chen Y, Huang J and Yang Q, Jiang P, Wang L, Zhao Y and Yang Q: New insights into Sirt1: Potential therapeutic targets for the treatment of cerebral ischemic stroke. Front Cell Neurosci. 17:12287612023. View Article : Google Scholar : PubMed/NCBI | |
Huang S, Hong Z, Zhang L, Guo J, Li Y and Li K: CERKL alleviates ischemia reperfusion-induced nervous system injury through modulating the SIRT1/PINK1/Parkin pathway and mitophagy induction. Biol Chem. 403:691–701. 2022. View Article : Google Scholar : PubMed/NCBI | |
Li M, Li SC, Dou BK, Zou YX, Han HZ, Liu DX, Ke ZJ and Wang ZF: Cycloastragenol upregulates SIRT1 expression, attenuates apoptosis and suppresses neuroinflammation after brain ischemia. Acta Pharmacol Sin. 41:1025–1032. 2020. View Article : Google Scholar : PubMed/NCBI |