1.Mechanism of acupuncture on cerebral ischemia-reperfusion injury via p53/SLC7A11/GPX4 signaling pathway in rat models.
Qi WANG ; Ziwen HOU ; Yaoyao LIU ; Dan WEI ; Qingjie KONG ; Xia CHEN
Chinese Acupuncture & Moxibustion 2025;45(8):1099-1110
OBJECTIVE:
To explore the neuroprotective effect and underlying mechanism of Xingnao Kaiqiao acupuncture (acupuncture for regaining consciousness and opening orifices) in the rat models of cerebral ischemia-reperfusion injury (CIRI) based on the p53 protein (p53)/solute carrier family 7 member 11 (SLC7A11)/glutathione peroxidase 4 (GPX4) signaling pathway.
METHODS:
Of 102 male Wistar rats, 20 rats were randomly collected as a sham-operation group. Using a modified external carotid artery filament insertion method, CIRI models were prepared by occluding the middle cerebral artery in the rest rats. After modeling and excluding 1 non-successfully modeled rat and 1 dead one, the other modeled rats were randomized into a model group, an agonist group, an acupuncture group, and an acupuncture + agonist group, 20 rats in each one. Xingnao Kaiqiao acupuncture therapy was delivered in the rats of the acupuncture group and the acupuncture + agonist group. The acupoints included "Shuigou" (GV26), bilateral "Neiguan" (PC6), and "Sanyinjiao" (SP6) on the affected side. Electroacupuncture was attached to "Neiguan" (PC6) and "Sanyinjiao" (SP6) on the affected side, with dense-disperse wave, a frequency of 2 Hz/15 Hz and intensity of 1 mA. The intervention was delivered twice daily, 20 min each time and for 7 consecutive days. In the agonist group and acupuncture+agonist group, p53 agonist, COTI-2 was intraperitoneally injected (15 mg/kg), once daily for 7 consecutive days. Neurological deficit was evaluated using Zausinger's six-point scale. Cerebral infarction volume was quantified by triphenyl tetrazolium chloride (TTC) staining. Histopathological changes were observed using hematoxylin-eosin (HE) staining. Iron deposition was assessed by Prussian blue staining. Mitochondrial ultrastructure in the ischemic cortex was examined under transmission electron microscopy (TEM). Serum iron (Fe2+) was measured with chromometry. Malondialdehyde (MDA) and glutathione (GSH) levels in the ischemic hippocampus were determined using thiobarbituric acid and microplate assays, respectively. The mean fluorescence intensity of reactive oxygen species (ROS) in the ischemic cortex was analyzed by flow cytometry. The mRNA and protein expression of GPX4, SLC7A11, and p53 in the ischemic hippocampus were evaluated using quantitative real-time PCR (qRT-PCR) and Western blotting, respectively.
RESULTS:
Compared with the sham-operated group, the model group exhibited the decrease in neurological deficit score (P<0.01), and the increase in cerebral infarction volume percentage (P<0.01). The changes of brain tissue were presented in extensive cellular necrosis, pyknotic and deeply-stained nuclei, and vacuolar degeneration. The iron deposition was elevated in cortex and hippocampus (P<0.01), mitochondrial membrane density increased, the cristae was broken or reduced, and the outer membrane ruptured. The levels of Fe2+ and MDA, as well as the mean flourscence intensity of ROS were elevated (P<0.01) and the level of GSH was reduced (P<0.01). The mRNA and protein expression of GPX4 and SLC7A11 was reduced (P<0.01), while that of p53 rose (P<0.01). When compared with the model group, in the agonist group, the neurological deficit score was reduced (P<0.05), the percentage of infarction volume was higher (P<0.01), the histopathological damage was further exacerbated, and the percentage of iron deposition increased in the cortex and hippocampus (P<0.01). The mitochondrial quantity decreased, the membrane density increased, the mitochondrial cristae were broken or reduced, and the outer membrane was ruptured. The levels of Fe2+ and MDA, as well as the mean flourscence intensity of ROS were higher (P<0.01, P<0.05) and the level of GSH was reduced (P<0.05). The mRNA and protein expression of GPX4 and SLC7A11 decreased (P<0.01, P<0.05), while that of p53 was elevated (P<0.01). Besides, in comparison with the model group, the neurological deficit score was higher in the acupuncture group and the acupuncture + agonist group (P<0.01, P<0.05), the percentage of cerebral infarction volume was lower in the acupuncture group (P<0.01), the pathological damage of brain tissue was alleviated in the acupuncture group and the acupuncture + agonist group, and the percentage of iron depositiondecreased in the cortex and hippocampus (P<0.01). The mitochondrial structure was relatively clear, the mitochondrial cristae were fractured or reduced mildly in the acupuncture group and the acupuncture + agonist group. The levels of Fe2+ and MDA, as well as the mean flourscence intensity of ROS were lower (P<0.01) and the level of GSH was higher (P<0.01) in the acupuncture group. The mean fluorescence intensity of ROS were dropped (P<0.01) in the acupuncture + agonist group. The mRNA expression of GPX4 and SLC7A11 was elevated (P<0.01) and that of p53 was reduced (P<0.01, P<0.05) in either the acupuncture group or the acupuncture + agonist group; the protein expression of GPX4 and SLC7A11 rose (P<0.05, P<0.01) and that of p53 was dropped (P<0.01) in the acupuncture group; and the protein expression of p53 was also lower in the acupuncture + agonist group (P<0.05). When compared with the agonist group, in the acupuncture + agonist group, neurological deficit score increased (P<0.01), the percentage of cerebral infarction volume decreased (P<0.01), the pathological brain tissue damage was reduced, the percentage of iron deposition in cortex and hippocampus decreased (P<0.01), the mitochondrial structure was relatively clear and the cristae broken or reduced slightly; the levels of Fe2+ and MDA, as well as the mean fluorescence intensity of ROS were dropped (P<0.01), while the level of GSH increased (P<0.05); the mRNA and protein expression of GPX4 and SLC7411 was elevated (P<0.01, P<0.05), and that of p53 reduced (P<0.01). In comparison with the acupuncture + agonist group, in the acupuncture group, the neurological deficit score increased (P<0.05), the percentage of cerebral infarction volume decreased (P<0.05), the pathological brain tissue damage was alleviated, the percentage of iron deposition in cortex and hippocampus decreased (P<0.01), the mitochondrial structure was normal in tendency; the levels of Fe2+ and MDA, as well as the mean fluorescence intensity of ROS were reduced (P<0.05), while the level of GSH rose (P<0.01); the mRNA and protein expression of GPX4 and SLC7411 was elevated (P<0.01, P<0.05), and that of p53 reduced (P<0.01, P<0.05).
CONCLUSION
Xingnao Kaiqiao acupuncture can alleviate neurological damage in CIRI rats, which is obtained probably by inhibiting ferroptosis through p53/SLC7A11/GPX4 pathway.
Animals
;
Reperfusion Injury/metabolism*
;
Male
;
Acupuncture Therapy
;
Rats
;
Tumor Suppressor Protein p53/genetics*
;
Brain Ischemia/metabolism*
;
Rats, Wistar
;
Signal Transduction
;
Humans
;
Phospholipid Hydroperoxide Glutathione Peroxidase/genetics*
;
Disease Models, Animal
;
Acupuncture Points
;
Amino Acid Transport System y+/genetics*
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.Effects of ROCK-siRNA transfection on Ang II-induced endothelial cell senescence and endothelial microparticles.
Kai WANG ; Yan WANG ; Tianqi CHEN ; Fang PENG ; Hui ZHOU ; Qin SHI
Chinese Journal of Cellular and Molecular Immunology 2025;41(9):778-783
Objective To investigate the effects of ROCK-siRNA transfection on endothelial cell senescence and endothelial microparticles (EMPs) induced by angiotensin II (Ang II). Methods Human umbilical vein endothelial cells (HUVECs) were treated with Ang II (1.0 μmo/L) to induce cellular senescence models, followed by transfection with ROCK-siRNA. The cells were divided into four groups: control group, model group, negative transfection control group (Ang II combined with NC-siRNA), and ROCK-siRNA transfection group (Ang II combined with ROCK-siRNA). Cellular senescence was assessed by SA-β-Gal staining. EMP levels in cell supernatants and intracellular reactive oxygen species (ROS) levels were assessed using flow cytometry. The expression levels of silenced information regulator 1(SIRT1) and p53 protein in each group were analyzed by Western blotting. Results Following ROCK-siRNA transfection, the number of senescent cells induced by Ang II was significantly reduced, accompanied by decreased CD31+ EMP levels and suppressed intracellular ROS levels. Meanwhile, the expression levels of SIRT1 were up-regulated, while the expression levels of p53 were down-regulated. Conclusion Silencing ROCK expression suppresses EMP release, reduces ROS generation, regulates the expression of SIRT1 and p53, and ultimately attenuates Ang II-induced endothelial cell senescence.
Humans
;
Angiotensin II/pharmacology*
;
Cellular Senescence/genetics*
;
Human Umbilical Vein Endothelial Cells/cytology*
;
RNA, Small Interfering/metabolism*
;
Reactive Oxygen Species/metabolism*
;
Sirtuin 1/genetics*
;
Transfection
;
Tumor Suppressor Protein p53/genetics*
;
Cell-Derived Microparticles/drug effects*
;
rho-Associated Kinases/metabolism*
;
Endothelial Cells/metabolism*
;
Cells, Cultured
4.Hydroxysafflor Yellow A Ameliorates the Replicative Senescence of Human Umbilical Cord Mesenchymal Stem Cells by Suppressing Oxidative Stress.
Si-Yun WANG ; Qi ZHU ; Chun-Xia TAN ; Fang LU ; Tao LU
Journal of Experimental Hematology 2025;33(5):1507-1515
OBJECTIVE:
To investigate the effects and mechanisms of hydroxysafflor yellow A (HSYA) on replicative senescence in human umbilical cord mesenchymal stem cells (hUC-MSCs).
METHODS:
hUC-MSCs were cultured to construct a replicative senescence model through continuous amplification in vitro. Cells at passage 2 served as the control group, while cells at passage 10 were designated as the senescence group. The senescent cells were cultured in a culture medium containing HSYA. Cell viability was detected by the CCK-8 assay, and cell confluence was analyzed using the Incucyte S3 live-cell analysis system. The optimal concentration and time point were determined and utilized for subsequent experiments. Senescent cells were pretreated with 0.01 mg/ml HSYA, and the proportion of senescence-associated β-galactosidase (SA-β-gal) positive cells was detected to assess the senescence state. The relative telomere length was detected by qPCR. Reactive oxygen species (ROS) levels were measured using the fluorescent probe DCFH-DA. Mitochondrial membrane potential was assessed by JC-1 staining. The expression of p53, p16, p21, OCT4, and SOX2 genes was detected by qPCR. The expression of p16, p53, OCT4, and SOX2 proteins was analyzed by Western blot.
RESULTS:
HSYA significantly decreased the SA-β-gal positive staining rate, inhibited telomere attrition, reduced the ROS accumulation, increased mitochondrial membrane potential in senescent cells. Additionally, HSYA downregulated the expression of p53 and p16, and upregulated the expression of OCT4. HSYA decreased p16 protein level and increased OCT4 and SOX2 protein levels.
CONCLUSION
HSYA may ameliorate replicative senescence in hUC-MSCs by modulating the p53 and p16 signaling pathways and suppressing oxidative stress.
Humans
;
Mesenchymal Stem Cells/drug effects*
;
Cellular Senescence/drug effects*
;
Chalcone/pharmacology*
;
Oxidative Stress/drug effects*
;
Quinones/pharmacology*
;
Umbilical Cord/cytology*
;
Reactive Oxygen Species/metabolism*
;
Cells, Cultured
;
Cyclin-Dependent Kinase Inhibitor p16/metabolism*
;
Tumor Suppressor Protein p53/metabolism*
;
Membrane Potential, Mitochondrial
;
Cell Proliferation
5.Mechanism of Hedyotis diffusa-Scutellaria barbata D. Don for treatment of primary liver cancer: analysis with network pharmacology, molecular docking and in vitro validation.
Meng XU ; Lina CHEN ; Jinyu WU ; Lili LIU ; Mei SHI ; Hao ZHOU ; Guoliang ZHANG
Journal of Southern Medical University 2025;45(1):80-89
OBJECTIVES:
To investigate the active ingredients in Hedyotis diffusa-Scutellaria barbata D. Don and the main biological processes and signaling pathways mediating their inhibitory effect on primary hepatocellular carcinoma (HCC).
METHODS:
The core intersecting genes of HCC and the two drugs were screened from TCMSP, Uniport, Genecards, and String databases using Cytoscape software, and GO and KEGG enrichment analyses of the intersecting genes were conducted. Molecular docking between the active ingredients of the drugs and the core genes was carried out using Pubcham, RCSB and Autoduckto to identify the active ingredients with the highest binding energy, whose inhibitory effect on HepG2 cells was verifies using CCK-8 assay, flow cytometry and Western blotting.
RESULTS:
TP53 and ESR1 were identified as the core genes of HCC and the two drugs. GO and KEGG analyses showed that the two genes were mainly involved in regulation of apoptotic signaling pathway, cell population proliferation, methane raft, and protein kinase activity, and participated in the signaling pathways of apoptosis, proteoglycans in cancer, PI3K Akt signaling pathway, and hepatitis B. Molecular docking studies showed that the active ingredients of the drugs could be docked with TP53 and ESR1 genes under natural conditions, and ursolic acid had the highest binding energy to ESR1 (-4.98 kcal/mol). The results of CCK-8 assay, flow cytometry and Western blotting all demonstrated significant inhibitory effect of ursolic acid on HepG2 cells.
CONCLUSIONS
The inhibitory effect of Hedyotis diffusa-scutellariae barbatae on HCC is mediated by multiple active ingredients in the two drugs.
Humans
;
Molecular Docking Simulation
;
Liver Neoplasms/drug therapy*
;
Hep G2 Cells
;
Network Pharmacology
;
Carcinoma, Hepatocellular/drug therapy*
;
Hedyotis/chemistry*
;
Signal Transduction/drug effects*
;
Cell Proliferation/drug effects*
;
Tumor Suppressor Protein p53/metabolism*
;
Apoptosis/drug effects*
;
Estrogen Receptor alpha/metabolism*
;
Drugs, Chinese Herbal/pharmacology*
6.GPSM2 is highly expressed in gastric cancer to affect patient prognosis by promoting tumor cell proliferation.
Xue SONG ; Yue CHEN ; Min ZHANG ; Nuo ZHANG ; Lugen ZUO ; Jing LI ; Zhijun GENG ; Xiaofeng ZHANG ; Yueyue WANG ; Lian WANG ; Jianguo HU
Journal of Southern Medical University 2025;45(2):229-238
OBJECTIVES:
To explore the association between GPSM2 expression level and gastric cancer progression and analyze the functional pathways and action mechanism of GPSM2.
METHODS:
We analyzed GPSM2 expression levels in gastric cancer tumors based on data from the GEPIA database and the clinical data of 109 patients. Public databases enrichment analysis were used to assess the impact of GPSM2 expression level on survival outcomes and the functional pathways and action mechanism of GPSM2. We further observed the effects of GPSM2 knockdown and overexpression on proliferation, migration and apoptosis of MGC803 cells using CCK-8 assay, colony formation assay, flow cytometry and immunoblotting and on the growth of MGC803 cell xenografts in nude mice.
RESULTS:
Bioinformatic analysis and immunohistochemical staining of the clinical specimens both revealed high GPSM2 expressions in gastric cancer (P<0.01). A high GPSM2 expression was significantly correlated with T3-4 stages, N2-3 stages, a carcinoembryonic antigen (CEA) level ≥5 μg/L, and a carbohydrate antigen (CA) 19-9 level ≥37 kU/L (P<0.05). Cox regression analysis identified high GPSM2 expression as an independent risk factor affecting 5-year survival of the patients (P<0.05). Gene ontology (GO) analysis suggested that GPSM2 was involved in cell cycle regulation. In MGC803 cells, GPSM2 overexpression significantly promoted cell proliferation and G1/S transition and xenograft growth in nude mice. KEGG pathway enrichment analysis indicated that GPSM2 executed its biological functions by regulating the p53 signaling pathway, which was confirmed by the results of immunoblotting experiments showing suppression of p53 signaling pathway activity in GPSM2-over expressing MGC803 cells.
CONCLUSIONS
GPSM2 is highly expressed in gastric cancer to affect patient prognosis by promoting tumor cell proliferation and G1/S transition possibly via inhibiting the p53 pathway.
Stomach Neoplasms/metabolism*
;
Humans
;
Cell Proliferation
;
Prognosis
;
Animals
;
Mice, Nude
;
Cell Line, Tumor
;
Mice
;
Apoptosis
;
Tumor Suppressor Protein p53/metabolism*
;
Cell Movement
7.Quercetin inhibits proliferation and migration of clear cell renal cell carcinoma cells by regulating TP53 gene.
Junjie GAO ; Kai YE ; Jing WU
Journal of Southern Medical University 2025;45(2):313-321
OBJECTIVES:
To identify potential molecular targets of quercetin in the treatment of clear cell renal carcinoma (ccRCC).
METHODS:
The therapeutic targets of quercetin were screened from multiple databases by network pharmacology analysis, and the targets significantly correlated with ccRCC were screened from 4907 plasma proteins using a Mendelian randomization method. The drug-disease network model was constructed to screen the potential key targets. The functions of these targets were evaluated via bioinformatics analysis, and the screened targets were verified in cultured ccRCC cells.
RESULTS:
Network pharmacology analysis combined with Mendelian randomization identified TP53 (OR=3.325, 95% CI: 1.805-6.124, P=0.0001), ARF4 (OR=0.173, 95% CI: 0.065-0.456, P=0.0003), and DPP4 (OR=0.463, 95% CI: 0.302-0.711, P=0.0004) as the core targets in quercetin treatment of ccRCC. Bioinformatics analysis showed that TP53 was highly expressed in ccRCC, and patients with high TP53 expressions had worse survival outcomes. Molecular docking studies showed that the binding energy between quercetin and TP53 was -5.83 kcal/mol. In cultured 786-O cells, CCK-8 assay and wound healing assay showed that treatment with quercetin significantly inhibited cell proliferation and migration. Quercetin treatment also strongly suppressed the expression of TP53 at both the mRNA and protein levels in 786-O cells as shown by RT-qPCR and Western blotting.
CONCLUSIONS
TP53 may be the key target of quercetin in the treatment of ccRCC, which sheds light on potential molecular mechanism that mediate the therapeutic effect of quercetin.
Humans
;
Quercetin/pharmacology*
;
Carcinoma, Renal Cell/genetics*
;
Cell Proliferation/drug effects*
;
Kidney Neoplasms/genetics*
;
Cell Movement/drug effects*
;
Tumor Suppressor Protein p53/metabolism*
;
Cell Line, Tumor
;
Computational Biology
8.High expression of CDKN3 promotes migration and invasion of gastric cancer cells by regulating the p53/NF-κB signaling pathway and inhibiting cell apoptosis.
Yi ZHANG ; Yu SHEN ; Zhiqiang WAN ; Song TAO ; Yakui LIU ; Shuanhu WANG
Journal of Southern Medical University 2025;45(4):853-861
OBJECTIVES:
To investigate the expression of CDKN3 in gastric cancer and its impact on prognosis of gastric cancer patients.
METHODS:
We analyzed CDKN3 expression in clinical specimens from 114 gastric cancer patients and assessed its association with 5-year postoperative survival of the patients. GO and KEGG enrichment analyses were used to predict the biological function and possible mechanism of CDKN3. The effects of lentivirus-mediated CDKN3 knockdown on biological behaviors of gastric cancer cells were evaluated using Transwell assay, CCK-8 assay, TUNEL staining, flow cytometry, and Western blotting.
RESULTS:
CDKN3 expression was significantly higher in gastric cancer tissues than in the adjacent tissues with significant correlations with CEA level, CA19-9 level, and T and N staging (P<0.05). High CDKN3 expression was an independent risk factor affecting 5-year postoperative survival of the patients and predictive for long-term prognosis (P<0.01). Enrichment analyses suggested a probable association of CDKN3 with apoptosis. In MGC-803 cells, CDKN3 knockdown significantly lowered migration and invasion capacities of the cells, while CDKN3 overexpression produced the opposite effects. TUNEL staining revealed a significantly lower level of cell apoptosis in gastric cancer tissues than in adjacent tissues (P<0.01). CDKN3 knockdown obviously inhibited proliferation and increased apoptosis of MGC-803 cells. CDKN3 overexpression down-regulated the expressions of p53, p21 and Bax and up-regulated the expressions of p-p65 and Bcl-2.
CONCLUSIONS
CDKN3 is highly expressed in gastric cancer tissues and affects patient prognosis. CDKN3 overexpression promotes proliferation, invasion and migration and suppressed apoptosis of gastric cancer cells possibly through the p53/NF-κB signaling pathway.
Humans
;
Stomach Neoplasms/pathology*
;
Apoptosis
;
Signal Transduction
;
Tumor Suppressor Protein p53/metabolism*
;
Cell Movement
;
Cell Line, Tumor
;
NF-kappa B/metabolism*
;
Prognosis
;
Cyclin-Dependent Kinase Inhibitor Proteins/metabolism*
;
Cell Proliferation
;
Neoplasm Invasiveness
;
Male
;
Female
;
Dual-Specificity Phosphatases
9.Shuangshu Decoction inhibits growth of gastric cancer cell xenografts by promoting cell ferroptosis via the P53/SLC7A11/GPX4 axis.
Xinyuan CHEN ; Chengting WU ; Ruidi LI ; Xueqin PAN ; Yaodan ZHANG ; Junyu TAO ; Caizhi LIN
Journal of Southern Medical University 2025;45(7):1363-1371
OBJECTIVES:
To explore the mechanism of Shuangshu Decoction (SSD) for inhibiting growth of gastric cancer xenografts in nude mice.
METHODS:
Network pharmacology analysis was conducted to identify the common targets of SSD and gastric cancer cell ferroptosis, and bioinformatics analysis and molecular docking were used to validate the core targets. In the cell experiment, AGS cells were treated with SSD-medicated serum, Fer-1 (a ferroptosis inhibitor), or both, and the changes in cell viability, ferroptosis markers (ROS, Fe2+ and GSH), expressions of P53, SLC7A11 and GPX4, and mitochondrial morphology were examined. In a nude mouse model bearing gastric cancer xenografts, the effects of gavage with SSD, intraperitoneal injection of Fer-1, or their combination on tumor volume/weight, histopathology, and expressions of P53, SLC7A11 and GPX4 levels were evaluated.
RESULTS:
The active components in SSD (quercetin and wogonin) showed strong binding affinities to P53. In AGS cells, SSD treatment dose-dependently inhibited cell proliferation, increased ROS and Fe2+ levels, upregulated P53 expression, and downregulated the expressions of SLC7A11 and GPX4, but these effects were effectively attenuated by Fer-1 treatment. SSD also induced mitochondrial shrinkage and increased the membrane density, which were alleviated by Fer-1. In the tumor-bearing mouse models, gavage with SSD significantly reduced tumor size and weight, caused tumor cell necrosis, upregulated P53 and downregulated SLC7A11 and GPX4 expression in the tumor tissue, and these effects were obviously mitigated by Fer-1 treatment.
CONCLUSIONS
SSD inhibits gastric cancer growth in nude mice by inducing cell ferroptosis via the P53/SLC7A11/GPX4 axis.
Ferroptosis/drug effects*
;
Animals
;
Stomach Neoplasms/metabolism*
;
Tumor Suppressor Protein p53/metabolism*
;
Mice, Nude
;
Phospholipid Hydroperoxide Glutathione Peroxidase
;
Drugs, Chinese Herbal/pharmacology*
;
Humans
;
Amino Acid Transport System y+/metabolism*
;
Mice
;
Cell Line, Tumor
;
Cell Proliferation/drug effects*
;
Xenograft Model Antitumor Assays
10.Dysregulation of Iron Homeostasis Mediated by FTH Increases Ferroptosis Sensitivity in TP53-Mutant Glioblastoma.
Xuejie HUAN ; Jiangang LI ; Zhaobin CHU ; Hongliang ZHANG ; Lei CHENG ; Peng LUN ; Xixun DU ; Xi CHEN ; Qian JIAO ; Hong JIANG
Neuroscience Bulletin 2025;41(4):569-582
Iron metabolism is a critical factor in tumorigenesis and development. Although TP53 mutations are prevalent in glioblastoma (GBM), the mechanisms by which TP53 regulates iron metabolism remain elusive. We reveal an imbalance iron homeostasis in GBM via TCGA database analysis. TP53 mutations disrupted iron homeostasis in GBM, characterized by elevated total iron levels and reduced ferritin (FTH). The gain-of-function effect triggered by TP53 mutations upregulates itchy E3 ubiquitin-protein ligase (ITCH) protein expression in astrocytes, leading to FTH degradation and an increase in free iron levels. TP53-mut astrocytes were more tolerant to the high iron environment induced by exogenous ferric ammonium citrate (FAC), but the increase in intracellular free iron made them more sensitive to Erastin-induced ferroptosis. Interestingly, we found that Erastin combined with FAC treatment significantly increased ferroptosis. These findings provide new insights for drug development and therapeutic modalities for GBM patients with TP53 mutations from iron metabolism perspectives.
Ferroptosis/drug effects*
;
Humans
;
Iron/metabolism*
;
Glioblastoma/metabolism*
;
Tumor Suppressor Protein p53/metabolism*
;
Homeostasis/physiology*
;
Ferritins/metabolism*
;
Brain Neoplasms/genetics*
;
Mutation
;
Astrocytes/drug effects*
;
Cell Line, Tumor
;
Piperazines/pharmacology*
;
Quaternary Ammonium Compounds/pharmacology*
;
Ferric Compounds

Result Analysis
Print
Save
E-mail