1.Causal effects of chronic kidney disease on Alzheimer's disease and its prevention based on "kidney-brain interaction" theory.
Sen-Lin CHEN ; Zhi-Chen WANG ; Geng-Zhao CHEN ; Hang-Bin ZHENG ; Sai-E HUANG
China Journal of Chinese Materia Medica 2025;50(12):3431-3440
Based on the traditional Chinese medicine(TCM) theory of "kidney-brain interaction", a two-sample Mendelian randomization(MR) analysis was conducted to investigate the causal effects of chronic kidney disease(CKD) on Alzheimer's disease(AD) and analyze the potential mechanisms of kidney-tonifying and essence-replenishing TCM to improve AD. From the perspective that CKD is closely related to the core pathogenesis of AD, namely "kidney deficiency, essence loss, and marrow reduction", genome-wide association study(GWAS) data was used, with the inverse variance weighting(IVW) method as the main approach to reveal the causal association between CKD and AD. Sensitivity analysis was conducted to evaluate the robustness of the results. To further investigate the causal effects of CKD on AD, two different AD datasets were used as outcomes, and the urinary albumin-to-creatinine ratio(UACR) data was used as the exposure for a supplementary analysis. On this basis, the modern scientific mechanism of the kidney-tonifying and essence-replenishing method for improving AD was further explored. The IVW analysis show that CKD(ieu-b-2: OR=1.084, 95%CI[1.011, 1.163], P=0.024; ieu-b-5067: OR=1.001, 95%CI[1.000, 1.001], P=0.002) and UACR(ieu-b-2: OR=1.247, 95%CI[1.021, 1.522], P=0.031; ieu-b-5067: OR=1.001, 95%CI[1.000, 1.003], P=0.015) both have significant causal effects on AD in different datasets, with CKD increasing the risk of AD. The sensitivity analysis further confirmed the reliability of the results. Genetic studies have shown that CKD has a significant causal effect on AD, suggesting that controlling CKD is an important intervention measure for preventing and treating AD. Therefore, further research on CKD's role in AD is crucial in clinical practice. The research enriches the theoretical implication of "kidney-brain interaction", deepens the understanding of AD' etiology, and provides further insights and directions for the prevention and treatment of AD with TCM, specifically from a kidney-based perspective.
Humans
;
Alzheimer Disease/genetics*
;
Renal Insufficiency, Chronic/genetics*
;
Kidney/metabolism*
;
Brain/physiopathology*
;
Genome-Wide Association Study
;
Medicine, Chinese Traditional
;
Mendelian Randomization Analysis
2.Mechanism of immediate administration of Angong Niuhuang Pills in intervention of traumatic brain injury based on metabolomics and transcriptomics.
Xiao-Tong ZHU ; Liang-Liang TIAN ; Jing-Jing ZHANG ; Hong-Jun YANG
China Journal of Chinese Materia Medica 2025;50(10):2750-2760
This study integrates metabolomics and transcriptomics to explore the immediate effects of Angong Niuhuang Pills(ANP) in intervening traumatic brain injury(TBI) in rats. A TBI model was successfully established in rats using the optimized Feeney free-fall impact technique. Rats were randomly divided into sham operation(sham) group, model(Mod) group, positive drug(piracetam) group, ANP low-dose(ANP-L) group, and ANP high-dose(ANP-H) group according to a random number table. Nissl staining and immunofluorescence were used to count the number of Nissl bodies and detect B-cell lymphoma-2(Bcl-2) gene, caspase-3, and tumor protein 53(TP53) expression in brain tissue, and enzyme-linked immunosorbent assay(ELISA) was used to measure prostaglandin-endoperoxide synthase 2(PTGS2) level in rat brain tissue. Metabolomics and transcriptomics analyses were conducted for brain tissue from sham, Mod, and ANP-H groups. Gene Ontology(GO) and Kyoto Encyclopedia of Genes and Genomes(KEGG) enrichment analyses were carried out to indicate the mechanisms of ANP in the intervention of TBI. Integrative metabolomics and transcriptomics analysis revealed the metabolic pathways involved in ANP's intervention in TBI. The results showed that ANP significantly increased the number of Nissl bodies in TBI rat brain tissue, upregulated Bcl-2 expression, and downregulated the levels of caspase-3, TP53, and PTGS2. Compared to the Mod group, the ANP-H group significantly upregulated 12 differential metabolites(DMs) and downregulated 25 DMs. Five key metabolic pathways were identified, including glycerophospholipid metabolism, pyrimidine metabolism, glycine, threonine, and serine metabolism, arginine and proline metabolism, and D-amino acid metabolism. Transcriptomics identified 730 upregulated and 612 downregulated differentially expressed genes(DEGs). Enrichment analysis highlighted that biological functions related to inflammatory responses and apoptotic processes, and key signaling pathways, including phosphoinositide 3-kinase(PI3K)/protein kinase B(Akt) and mitogen-activated protein kinase(MAPK) were significantly enriched. The data of transcriptomics and metabolomics pinpointed three key metabolic pathways, i.e., glycerophospholipid metabolism, pyrimidine metabolism, and glycine, threonine, and serine metabolism.
Animals
;
Drugs, Chinese Herbal/administration & dosage*
;
Rats
;
Brain Injuries, Traumatic/metabolism*
;
Male
;
Metabolomics
;
Rats, Sprague-Dawley
;
Transcriptome/drug effects*
;
Cyclooxygenase 2/genetics*
;
Brain/metabolism*
;
Caspase 3/genetics*
;
Humans
;
Tumor Suppressor Protein p53/genetics*
3.Rhodiolae Crenulatae Radix et Rhizoma protects brain microvascular endothelial cells from ischemia and hypoxia injury by regulating PI3K/AKT/GSK3β pathway.
Li TANG ; Qiu-Yue YANG ; Hong-Fa CHENG ; Ya-Hui XIE ; Qiu-Xia ZHANG
China Journal of Chinese Materia Medica 2025;50(11):3127-3136
This study elucidates the mechanism of Rhodiolae Crenulatae Radix et Rhizoma(RCRR) in protecting brain microvascular endothelial cells from oxygen-glucose deprivation(OGD) injury and reveals the modern pharmacological mechanism of RCRR's traditional use in nourishing Qi and promoting blood circulation to protect endothelial cells. The scratch assay was employed to assess the migratory capacity of endothelial cells. Immunofluorescence and Western blot techniques were employed to assess the protein expression of tight junction proteins zonula occludens-1(ZO-1), occludin, claudin-5, and proteins of the phosphoinositide 3-kinase(PI3K)/protein kinase B(AKT)/glycogen synthase kinase-3beta(GSK3β) pathway. The results demonstrated that 63 bioactive components and 125 potential core targets of RCRR were identified from the ETCM, TCMBank, and SwissTargetPrediction databases, as well as from the literature. A total of 1 708 brain microvascular endothelial cell-related targets were identified from the GeneCards and OMIM databases, and 52 targets were obtained by intersecting drug components with cell targets. The protein-protein interaction(PPI) network analysis revealed that AKT1, epidermal growth factor receptor(EGFR), matrix metalloproteinase 9(MMP9), estrogen receptor 1(ESR1), proto-oncogene tyrosine-protein kinase(SRC), peroxisome proliferator-activated receptor gamma(PPARG), GSK3β, and matrix metalloproteinase 2(MMP2) were considered hub genes. The KEGG enrichment analysis identified the PI3K/AKT pathway as the primary signaling pathway. Cell experiments demonstrated that RCRR-containing serum could enhance the migratory capacity of brain microvascular endothelial cells and the expression of tight junction proteins following OGD injury, which may be associated with the downregulation of the PI3K/AKT/GSK3β pathway. This study elucidates the pharmacological mechanism of RCRR in protecting brain microvascular endothelial cells through network pharmacology, characterized by multiple components and targets. These findings were validated through in vitro experiments and provide important ideas and references for further research into the molecular mechanisms of RCRR in protecting brain microvascular endothelial cells.
Endothelial Cells/cytology*
;
Glycogen Synthase Kinase 3 beta/genetics*
;
Proto-Oncogene Proteins c-akt/genetics*
;
Drugs, Chinese Herbal/pharmacology*
;
Phosphatidylinositol 3-Kinases/genetics*
;
Signal Transduction/drug effects*
;
Brain/metabolism*
;
Humans
;
Animals
;
Rhizome/chemistry*
;
Microvessels/metabolism*
;
Brain Ischemia/drug therapy*
4.Mechanism of inhibiting miR-34a-5p expression and promoting bone growth in mouse brain tissue by Semen Ziziphi Spinosae extract.
Yuan-Yuan PEI ; Yan XIE ; Na YIN ; Wen-Long MA ; Wei-Peng XING ; Gui-Zhi WANG ; Qing-Feng WANG
China Journal of Orthopaedics and Traumatology 2025;38(10):1061-1070
OBJECTIVE:
To explore the mechanism by which the extract of Semen Ziziphi Spinosae extract promotes bone growth in mice by modulation of the expression of miR-34a-5p in brain tissue.
METHODS:
Mice were assigned to four experimental groups:a normal control group, a drug administration group (receiving 0.320 mg·g-1 body weight of Semen Ziziphi Spinosae extract via intragastric administration), a positive control group (receiving 0.013 mg·g-1 body weight of jujube seed saponin via intragastric administration), and a combination group administration with Semen Ziziphi Spinosae extract plus a 5-hydroxytryptamine 2A receptor (5-HT2AR) agonist (intragastric administration of Semen Ziziphi Spinosae extract combined with intracerebroventricular injection of 8 μg P-MPPF per mice for the final three days of the experiment). Following a 20-day administration period, the effects of the interventions on bone growth, serum growth hormone (GH) levels, and 5-HT2AR expression in brain tissue were evaluated. MicroRNAs (miRNAs) that were differentially expressed in the brain tissues of mice exhibiting bone growth induced by Semen Ziziphi Spinosae extract, as compared to those in normal mice, were identified using a gene chip approach. The interaction between miR-34a-5p and 5-HT2AR was subsequently validated through quantitative reverse transcription polymerase chainreaction (RT-qPCR) and dual-luciferase reporter gene assays. Subsequently, by utilizing the miR-34a-5p inhibitor group and mimics group, along with the normal control group, the drug administration group, the positive control group, and the drug administration combined with miR-34a-5p inhibitor group, the variations in 5-HT2AR expression in mouse brain tissue across all groups were examined, and the binding activity of 5-hydroxytryptamine (5-HT) to the 5-hydroxytryptamine 1A receptor (5-HT1AR) in mice was assessed.
RESULTS:
The body lengths of the normal control group and the drug administration group were(8.9±0.3) and(10.4±0.4) cm;femur lengths were (8.5±0.3) and (9.1±0.5) mm;tibia lengths were (10.7±0.3) and (11.2±0.4) mm, respectively. The contents of GH levels were (58.6±8.2) and (72.9±6.1) ng·ml-1;and the contents of 5-HT2AR were (32.0±5.0) and (21.9± 5.5) ng·ml-1, respectively. Compared with the normal control group, the drug administration group promoted the growth of body length, femur, and tibia in mice, and increased GH secretion, showing statistically significant differences (P<0.05). Additionally, it significantly reduced the content of 5-HT2AR in brain tissue, with statistical significance (P<0.01). The gene chip analysis identified a total of 16 differentially expressed miRNAs, of which 13 were up-regulated and 3 were down-regulated. Bioinformatics analysis predicted that the up-regulated miR-34a-5p could regulate the expression of 5-HT2AR, a prediction that was confirmed through a dual-luciferase reporter gene assay, demonstrating a direct regulatory interaction between the two. Furthermore, in vivo experiments in mice revealed that overexpression and silencing of miR-34a-5p resulted in corresponding changes in the expression levels of 5-HT2AR in brain tissues/cells, as well as in the binding activity between 5-HT and 5-HT1AR.
CONCLUSION
The Semen Ziziphi Spinosae extract promotes animal bone growth by enhancing miR-34a-5p expression in brain tissue, downregulating the expression level of 5-HT2AR, improving the binding activity between 5-HT and 5-HT1AR, and extending slow-wave sleep duration, thereby stimulating GH secretion.
Animals
;
MicroRNAs/metabolism*
;
Mice
;
Male
;
Brain/metabolism*
;
Ziziphus/chemistry*
;
Bone Development/drug effects*
;
Drugs, Chinese Herbal/pharmacology*
;
Plant Extracts/pharmacology*
5.Homer 1a overexpression alleviates nerve injury in mice with traumatic brain injury by regulating autophagy mediated by PI3K/AKT/mTOR pathway.
Yuan WANG ; Mengyang WANG ; Xiumin ZHANG ; Ming LUO
Chinese Journal of Cellular and Molecular Immunology 2025;41(1):31-37
Objective To investigate the effects and molecular mechanism of Homer protein homolog 1a (Homer 1a) overexpression on nerve injury in mice with traumatic brain injury (TBI). Methods Sixty male C57BL/6 mice were randomly divided into five groups: sham group, TBI group, empty lentivirus (Lv-NC) group, Homer 1a overexpression lentivirus (Lv-Homer 1a) group and Lv-Homer 1a + 740 Y-P group, with 12 mice in each group. The lentivirus was orthotopic injected into the cerebral cortex of mice 5 d before modeling, while 740 Y-P was injected intraperitoneally 1 d before modeling. The TBI model was established using the free-fall impact method, and the modified neurological severity scores (mNSS) of the mice was assessed 72 h post-surgery. The water content of brain tissue was quantified, and the histopathological damage and neuronal loss in brain tissue were assessed using HE staining and Nissl staining respectively. The formation of autophagosomes in brain tissue was observed by transmission electron microscopy. The protein expression levels of Homer 1a, microtubule-associated protein 1 light chain 3B (LC3B), Beclin 1, phosphatidylinositol 3-kinase (PI3K), phosphorylation PI3K(p-PI3K), protein kinase B (AKT), p-AKT, mammalian target of rapamycin (mTOR), and p-mTOR in brain tissue were detected by Western blot analysis. Results Compared to the sham group, the mice in the TBI group exhibited a significant increase in mNSS and cerebral water content. Moreover, severe brain tissue pathological damage was observed, accompanied by a substantial loss of neurons and an increase in autophagosome formation. The protein expressions of Homer 1a and Beclin 1, as well as the protein ratio of LC3B-II/LC3B-I, in brain tissues were significantly elevated, while the protein ratios of p-PI3K/PI3K, p-AKT/AKT, and p-mTOR/mTOR were significantly reduced. Compared to the TBI group, the Lv-Homer 1a group exhibited reduced mNSS and brain water content. Additionally, there was an improvement in pathological brain tissue damage and neuron loss. Furthermore, there was an increase in autophagosome formation and expression of autophagy-related proteins, while the protein ratios of p-PI3K/PI3K, p-AKT/AKT, and p-mTOR/mTOR were decreased. Compared to the Lv-Homer 1a group, the nerve injury in the Lv-Homer 1a+740 Y-P group was exacerbated, accompanied by a reduction in autophagosome formation and expression of autophagy-related proteins, while the PI3K/AKT/mTOR signaling pathway was activated. Conclusion Overexpression of Homer 1a effectively mitigates neurological damage in TBI mice, potentially through modulation of autophagy mediated by the PI3K/AKT/mTOR signaling pathway.
Animals
;
TOR Serine-Threonine Kinases/genetics*
;
Autophagy
;
Brain Injuries, Traumatic/pathology*
;
Male
;
Proto-Oncogene Proteins c-akt/genetics*
;
Phosphatidylinositol 3-Kinases/genetics*
;
Homer Scaffolding Proteins/metabolism*
;
Mice, Inbred C57BL
;
Signal Transduction
;
Mice
6.Correlation analysis of low expression of LY86-AS1 and KHDRBS2 with immune cell invasion and prognosis in glioblastoma.
Shasha WANG ; Wenhao ZHAO ; Xining HE ; Yangyang ZHANG ; Wenli CHANG
Chinese Journal of Cellular and Molecular Immunology 2025;41(3):245-253
Objective To investigate the expression and correlation of LY86-AS1 and KHDRBS2 in glioblastoma (GBM), and their impacts on the prognosis of patients and immune cell infiltration. Methods Based on the GSE50161 dataset from the Gene Expression Omnibus (GEO) database, LY86-AS1 and KHDRBS2, which are closely related to the development of GBM, were identified by WGCNA and differential expression analysis. The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA) databases were used to analyze the relationship between the expression of LY86-AS1 and KHDRBS2 and the prognosis of GBM patients. Multiple datasets were employed to analyze the correlation between the expression levels of LY86-AS1 and KHDRBS2 and its relationship with immune cell infiltration. Real-time quantitative PCR was used to verify the expression of LY86-AS1 and KHDRBS2 in GBM and normal brain tissues. The Human Protein Atlas (HPA) database was accessed to obtain the protein expression of KHDRBS2, and immunohistochemical staining was conducted to verify the protein expression of KHDRBS2. Results LY86-AS1 and KHDRBS2 were lowly expressed in GBM tissues and were closely related to the development of GBM, showing a significant positive correlation. Patients with low expression levels of LY86-AS1 and KHDRBS2 had a lower overall survival rate than those with high expression levels. LY86-AS1 was positively correlated with naive B cells, plasma cells, activated NK cells, M1 macrophages, activated mast cells and monocytes. KHDRBS2 was positively correlated with naive B cells, plasma cells, helper T cells, activated NK cells and monocytes. Conclusion The low expression levels of LY86-AS1 and KHDRBS2 in GBM, which is associated with poor prognosis, affect the tumor immune microenvironment and may serve as potential new biomarkers for the diagnosis of GBM and the prognosis assessment of patients.
Humans
;
Glioblastoma/metabolism*
;
Prognosis
;
Brain Neoplasms/pathology*
;
Gene Expression Regulation, Neoplastic
;
RNA-Binding Proteins/metabolism*
7.Sialyltransferase ST3GAL1 promotes malignant progression in glioma.
Zihao ZHAO ; Wenjing ZHENG ; Lingling ZHANG ; Wenjie SONG ; Tao WANG
Chinese Journal of Cellular and Molecular Immunology 2025;41(4):308-317
Objective To investigate the clinical relevance and diagnostic or prognostic value of ST3β-galactoside α-2, 3-sialyltransferase 1 (ST3GAL1) in glioma and to confirm its role in promoting malignant phenotypes. Methods Using data from The Cancer Genome Atlas (TCGA) database, we analyzed the correlation between ST3GAL1 expression levels in glioma and clinical parameters to evaluate its diagnostic and prognostic value. The impact of ST3GAL1 on malignant phenotypes of glioma cells-including proliferation, cell cycle progression, apoptosis, and invasion was further validated through ST3GAL1 knockdown experiments. Results The expression level of ST3GAL1 was significantly higher in glioma tissues compared to healthy brain tissues and showed a strong correlation with clinical characteristics of glioma patients. Survival analysis and receiver operating characteristic (ROC) curve demonstrated that ST3GAL1 could serve as a potential diagnostic and prognostic biomarker for glioma. Knockdown of ST3GAL1 suppressed proliferation, invasion, and migration capabilities of glioma cell lines, and induced G1-phase cell cycle arrest. Conclusion ST3GAL1 promotes malignant phenotypes in glioma and plays a critical role in its malignant progression, suggesting its potential as a biomarker for glioma diagnosis and prognosis.
Humans
;
Sialyltransferases/metabolism*
;
Glioma/diagnosis*
;
Cell Proliferation/genetics*
;
Cell Line, Tumor
;
Brain Neoplasms/enzymology*
;
beta-Galactoside alpha-2,3-Sialyltransferase
;
Disease Progression
;
Prognosis
;
Cell Movement/genetics*
;
Apoptosis/genetics*
;
Male
;
Female
;
Gene Expression Regulation, Neoplastic
;
Biomarkers, Tumor/metabolism*
;
Middle Aged
8.Effects of Zhuang medicine Shuanglu Tongnao Formula on neuroinflammation in ischemic stroke model rats via the P2X7R/NLRP3 pathway.
Liangji GUO ; Ligui GAN ; Zujie QIN ; Hongli TENG ; Chenglong WANG ; Jiangcun WEI ; Xiaoping MEI
Chinese Journal of Cellular and Molecular Immunology 2025;41(11):985-991
Objective To explore the effects of Shuanglu Tongnao Formula on neuroinflammation in ischemic stroke (IS) rats via the P2X purinoceptor 7 receptor (P2X7R)/NLR family pyrin domain-containing 3 (NLRP3) pathway. Methods The rats were divided into five groups: the IS group, control group, Shuanglu Tongnao Formula group, P2X7R inhibitor brilliant blue G (BBG) group, and Shuanglu Tongnao Formula combined with P2X7R activator adenosine triphosphate (ATP) group, with 18 rats in each group. Except for the control group, rats in all other groups were used to construct an IS model using the suture method. After successful modeling, the drug was given once a day for 2 weeks. Neurological function scores and cerebral infarction volume ratios were measured in rats. Pathological examination of the ischemic penumbra brain tissue was performed. Immunofluorescence staining was used to quantify the proportions of microglia co-expressing both inducible nitric oxide synthase (iNOS) and ionized calcium-binding adapter molecule 1 (Iba1), as well as arginase 1 (Arg1) and Iba1, in the ischemic penumbra brain tissue. ELISA was used to detect tumor necrosis factor-alpha (TNF-α), transforming growth factor-beta (TGF-β), interleukin 6 (IL-6) and IL-10 in the ischemic penumbra brain tissue. Western blotting was used to measure P2X7R, NLRP3, and IL-1β proteins in the ischemic penumbra brain tissue. Results Compared with the control group, the IS group showed disordered neuronal arrangement, nuclear condensation, and obvious infiltration of inflammatory cells in the ischemic penumbra; significantly elevated neurological function scores, cerebral infarction volume ratios, proportions of microglia co-expressing iNOS and Iba1, and levels of TNF-α, IL-6, and P2X7R, NLRP3, IL-1β proteins; along with reduced proportions of microglia co-expressing Arg1 and Iba1 and levels of TGF-β and IL-10. Compared with the IS group, the Zhuang medicine Shuanglu Tongnao Formula and BBG groups demonstrated alleviated brain tissue damage; reduced neurological function scores, cerebral infarction volume ratios, proportions of microglia co-expressing iNOS and Iba1, and levels of TNF-α, IL-6, and P2X7R, NLRP3, IL-1β proteins; along with increased proportions of microglia co-expressing Arg1 and Iba1 and levels of TGF-β and IL-10. ATP reversed the effects of Zhuang medicine Shuanglu Tongnao Formula on microglial polarization and neuroinflammation in IS rats. Conclusion Zhuang medicine Shuanglu Tongnao Formula may promote the transformation of microglia from M1 type to M2 type by inhibiting the P2X7R/NLRP3 pathway, thereby improving neuroinflammation in IS rats.
Animals
;
NLR Family, Pyrin Domain-Containing 3 Protein/metabolism*
;
Receptors, Purinergic P2X7/metabolism*
;
Male
;
Drugs, Chinese Herbal/pharmacology*
;
Rats
;
Ischemic Stroke/pathology*
;
Rats, Sprague-Dawley
;
Disease Models, Animal
;
Signal Transduction/drug effects*
;
Neuroinflammatory Diseases/metabolism*
;
Tumor Necrosis Factor-alpha/metabolism*
;
Nitric Oxide Synthase Type II/metabolism*
;
Interleukin-10/metabolism*
;
Brain Ischemia/drug therapy*
;
Microglia/metabolism*
9.In vitro cultured calculus bovis alleviates cerebral ischemia/reperfusion injury through regulating microglial polarization and inhibiting NLRP3.
Tanlu CHU ; Wei ZHANG ; Jingwen CHEN ; Zeyue PAN ; Lingfeng WANG ; Xiaoming ZHONG ; Fengmei QIU ; Zhen HUANG
Journal of Zhejiang University. Medical sciences 2025;54(3):360-371
OBJECTIVES:
To investigate the effect of in vitro cultured calculus bovis (ICCB) on cerebral ischemia/reperfusion injury (CIRI) and its mechanism.
METHODS:
A CIRI rat model and a cell model were induced by middle cerebral artery occlusion (MCAO) in Sprague Dawley rats and oxygen glucose deprivation/reperfusion (OGD/R) in BV2 cells, respectively. The CIRI rat model was evaluated using the modified neurological severity score (mNSS), brain water content, and cerebral infarction volume after 1.5 h of ischemia followed by 72 h of reperfusion. Histopathological changes in the cortex and hippocampal CA1 region were observed with hematoxylin and eosin staining. Microglial polarization and NOD-like receptor thermal protein domain associated protein (NLRP) 3 inflammasome expression in the cortex were examined by immunofluorescence. BV2 cell viability was measured via MTT assay after treatment with ICCB and Nigericin. The expressions of NLRP3, ASC, caspase-1 proteins and inflammatory cytokines were detected with Western blotting in OGD/R treated BV2 cells (0.5 h OGD+24 h reperfusion) and in cells pretreated with Nigericin for 24 h.
RESULTS:
ICCB treatment significantly improved neurological function, reduced cerebral infarct volume and brain water content, and mitigated pathological damage in the cortical and hippocampal CA1 regions of rats subjected to CIRI (all P<0.05). ICCB promoted the transition of cortical microglia from M1 to M2 phenotypes and suppressed NLRP3 activation in microglial cells (all P<0.01). ICCB significantly down-regulated the expression of NLRP3, ASC, and caspase-1 proteins, and reduced the secretion of IL-18 and IL-1β in BV2 cells of OGD/R model (all P<0.01). In addition, Nigericin significantly reversed the salvage effect of ICCB on model cells (both P<0.01) and the modulation of inflammatory cytokines (P<0.05).
CONCLUSIONS
ICCB exerts a protective effect against CIRI by mitigating neuroinflammation, through the reduction of M1 microglial polarization, promotion of M2 conversion, and suppression of the NLRP3/ASC/caspase-1 signaling pathway.
Animals
;
Rats, Sprague-Dawley
;
Reperfusion Injury/prevention & control*
;
Microglia/metabolism*
;
Rats
;
NLR Family, Pyrin Domain-Containing 3 Protein
;
Brain Ischemia/metabolism*
;
Male
10.Study on the targets and mechanisms of 7-hydroxyethyl chrysin in prevention and treatment of high-altitude cerebral edema using proteomics technology.
Dongmei ZHANG ; Xiaolin LI ; Chenyu YANG ; Linlin JING ; Lei HE ; Huiping MA
Journal of Zhejiang University. Medical sciences 2025;54(4):549-558
OBJECTIVES:
To investigate the targets and mechanisms of 7-hydroxyethyl chrysin (7-HEC) in prevention and treatment of high-altitude cerebral edema (HACE) in rats.
METHODS:
Fifty-four male Wistar rats were randomly divided into normal control group, HACE model group, and 7-HEC-treated group (18 rats in each group). Except for the normal control group, rats in the two other groups were exposed to a hypobaric hypoxic chamber simulating a 7000 m altitude for 72 h to establish the HACE model. The 7-HEC-treated group was intraperitoneally injected with 7-HEC (150 mg·kg-¹·d-¹) for 3 consecutive days before modeling, while the model group received equivalent isotonic sodium chloride solution. Tandem Mass Tag (TMT) proteomics technology was used to detect differentially expressed proteins (DEPs) with screening criteria set at a fold change >1.2 and P<0.05. Western blotting was used to verify the expression levels of target proteins. Gene Ontology (GO) enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, and protein-protein interaction (PPI) network analysis were performed.
RESULTS:
Compared with the normal control group, 256 DEPs were identified in the HACE model group. Compared with the HACE model group, 87 DEPs were identified in the 7-HEC-treated group. Among them, 19 DEPs that were dysregulated in the HACE model group were restored after 7-HEC intervention, of which seven (HSPA4, Arhgap20, SERT, HACL1, CCDC43, POLR3A, and PCBD1) were confirmed by Western blotting. GO enrichment analysis of the DEPs between the HACE model and 7-HEC-treated groups revealed their involvement in 13 biological processes, five cellular components, and two molecular functions. KEGG pathway analysis indicated associations with the mRNA surveillance pathway, Th17 cell differentiation, serotonergic synapse, RNA polymerase, protein processing in the endoplasmic reticulum, peroxisome, neuroactive ligand-receptor interaction, folate biosynthesis. PPI network analysis demonstrated that HSPA4, POLR3A, and HACL1, which were validated by Western blotting, interacted with multiple signaling pathways and ranked among the top 20 hub proteins by degree value, suggesting their potential role as core regulatory factors. Arhgap20, SERT and PCBD1 also exhibited interactions with several proteins, suggesting their potential as key regulatory proteins, whereas no interactions for CCDC43 were identified.
CONCLUSIONS
This study applied TMT proteomics to identify seven potential therapeutic targets of 7-HEC for the prevention and treatment of HACE. These targets may be involved in the pathogenesis of HACE through multiple pathways, including maintaining cellular homeostasis, ameliorating oxidative stress, regulating energy metabolism, and reducing vascular permeability.
Animals
;
Male
;
Proteomics/methods*
;
Rats, Wistar
;
Flavonoids/therapeutic use*
;
Rats
;
Brain Edema/etiology*
;
Altitude Sickness/metabolism*
;
Protein Interaction Maps

Result Analysis
Print
Save
E-mail