1.Mechanism of salidroside in inhibiting expression of adhesion molecules in oxLDL-induced endothelial cells by regulating ferroptosis mediated by SIRT1/Nrf2.
Meng ZHANG ; Min XIAO ; Jing-Jing LI ; Jiang-Feng LI ; Guang-Hui FAN
China Journal of Chinese Materia Medica 2025;50(10):2787-2797
This article investigated the effect and mechanism of salidroside(SAL) on the expression of adhesion molecules in oxidized low-density lipoprotein(oxLDL)-induced mouse aortic endothelial cell(MAEC). The oxLDL-induced endothelial cell injury model was constructed, and the safe concentration and action time of SAL were screened. The cells were divided into control group, oxLDL group, low and high concentration groups of SAL, and ferrostatin-1(Fer-1) group. The cell viability was detected by CCK-8 assay; lactate dehydrogenase(LDH) leakage was measured by colorimetry; the expression of intercellular adhesion molecule 1(ICAM-1) and recombinant vascular cell adhesion molecule 1(VCAM-1) were detected by immunofluorescence; Fe~(2+),glutathione(GSH),malondialdehyde(MDA),and 4-hydroxynonenal(4-HNE) levels were detected by kit method; reactive oxygen species(ROS) was detected by DCFH-DA probe; the levels of glutathione peroxidase 4(GPX4),silent mating type information regulation 2 homolog 1(SIRT1), and nuclear factor erythroid 2-related factor 2(Nrf2) were determined by using Western blot. The inhibitors of Nrf2 and SIRT1 were used, and endothelial cell were divided into control group, oxLDL group, SAL group, ML385 group(Nrf2 inhibitor), and EX527 group(SIRT1 inhibitor). The ultrastructure of mitochondria was observed by electron microscope; mitochondrial membrane potential(MMP) was detected by flowcytometry; the expressions of SIRT1,Nrf2,solute carrier family 7 member 11(SLC7A11),GPX4,ferroportin 1(FPN1),ferritin heavy chain 1(FTH1),ICAM-1, and VCAM-1 were detected by Western blot. The results showed that similar to Fer-1,low and high concentrations of SAL could improve cell viability, inhibit LDH release and the expression of ICAM-1 and VCAM-1 in oxLDL-induced endothelial cells(P<0.05 or P<0.01). It was related to increase in GSH level, decrease in Fe~(2+),ROS,MDA, and 4-HNE level, and up-regulation of SIRT1,Nrf2, and GPX4 expression to inhibit ferroptosis(P<0.05 or P<0.01). The intervention effect of high concentration SAL was the most significant. ML385 and EX527 could partially offset the protection of SAL on mitochondrial structure and MMP and reverse the ability of SAL to up-regulate the expression of SIRT1,Nrf2,SLC7A11,GPX4,FPN1, and FTH1 and down-regulate the expression of ICAM-1 and VCAM-1(P<0.05 or P<0.01).To sum up, SAL could reduce the expression of ICAM-1 and VCAM-1 in oxLDL-induced endothelial cell, which may relate to activation of SLC7A11/GPX4 antioxidant signaling pathway mediated by SITR1/Nrf2, up-regulation of FPN1 and FTH1 expression, and inhibition of ferroptosis.
Sirtuin 1/genetics*
;
Animals
;
Ferroptosis/drug effects*
;
Lipoproteins, LDL/metabolism*
;
NF-E2-Related Factor 2/genetics*
;
Mice
;
Endothelial Cells/cytology*
;
Glucosides/pharmacology*
;
Phenols/pharmacology*
;
Cell Adhesion Molecules/genetics*
;
Reactive Oxygen Species/metabolism*
;
Intercellular Adhesion Molecule-1/genetics*
;
Vascular Cell Adhesion Molecule-1/genetics*
;
Cell Survival/drug effects*
2.Research progress in pharmacological activities and pharmacokinetics of geniposidic acid.
Zi-Wei LI ; Sheng-Lan QI ; Qing-Guang ZHANG ; Ling CHEN ; Jing HU ; Guang-Bo GE ; Feng HUANG
China Journal of Chinese Materia Medica 2025;50(13):3679-3691
Geniposidic acid(GA), a natural iridoid, exists in the roots, stems, leaves, flowers, bark, fruits, and seeds of medicinal plants of Rubiaceae, Eucommiaceae, and Plantaginaceae. Modern pharmacological studies have revealed that GA has multiple pharmacological activities, including organ-protective, anti-inflammatory, antioxidative, anti-osteoporosis, anti-neurodegenerative, and anti-cardiovascular effects. GA can enhance cell/organism defenses by upregulating key anti-inflammatory and antioxidant cytokines, while downregulating key node proteins in pro-inflammatory signaling pathways such as AhR and TLR4/MyD88, thereby exerting pharmacological effects such as organ protection. Pharmacokinetic investigations have suggested that after oral administration, GA can be distributed in multiple organs(kidney, liver, heart, spleen, lung, etc.). In addition, the pharmacokinetic behavior of GA could be significantly altered under disease conditions, as demonstrated by a marked increase in systematic exposure. This article comprehensively summarizes the reported pharmacological activities and mechanisms and systematically analyzes the pharmacokinetic characteristics and key parameters of GA, with the aim of providing a theoretical basis and scientific reference for the precise clinical application of GA-related Chinese patent medicines, as well as for the investigation and development of innovative drugs based on GA.
Humans
;
Drugs, Chinese Herbal/chemistry*
;
Animals
;
Iridoid Glucosides/chemistry*
;
Plants, Medicinal/chemistry*
;
Anti-Inflammatory Agents/pharmacology*
3.Regulation of JAK2/STAT3 signaling pathway by polydatin in the treatment of hormone-induced femoral head necrosis in rats.
Xiang-Jun YANG ; Cong-Yue WANG ; Xi-Lin XU ; Hai HU ; Yi-Wei SHEN ; Xiao-Feng ZHANG
China Journal of Orthopaedics and Traumatology 2025;38(2):195-203
OBJECTIVE:
To explore the therapeutic effect of polygonum cuspidatum glycoside on steroid-induced osteonecrosis of the femoral head(SONFH) in rats and its potential mechanism of protecting bone tissue by regulating the Janus kinase 2/signal transducer and activator of transcription 3 signaling pathway(JAK2/STAT3).
METHODS:
Fifty male SD rats were randomly divided into control group, model group, low-dose polygonum cuspidatum glycoside group (polygonum cuspidatum glycoside-L), high-dose polygonum cuspidatum glycoside group (polygonum cuspidatum glycoside-H), and polygonum cuspidatum glycoside-H+Colivelin (JAK2/STAT3 pathway activator) group. SONFH model was induced by lipopolysaccharide and dexamethasone. The treatment groups were given polygonum cuspidatum glycoside orally(polygonum cuspidatum glycoside-L 10 mg·kg-1, polygonum cuspidatum glycoside-H 20 mg·kg-1, and the polygonum cuspidatum glycoside-H+Colivelin group was injected with Colivelin (1 mg·kg-1) intraperitoneally once a day, while the control and model groups were given an equal volume of saline for 6 weeks. The observed indicators included serum calcium(Ca), serum phosphorus (P), alkaline phosphatase, and transforming growth factor β1(TGF-β1) levels, micro-CT scanning, hematoxylin-eosin staining, and Western blot detection of JAK2/STAT3 signaling pathway and osteogenic differentiation marker genes, including Runt-related transcription factor 2 (Runx2), bone morphogenetic protein 2 (BMP2), and osteopontin (OPN) protein expression.
RESULTS:
Compared with the model group, the trabecular bone area percentage in the polygonum cuspidatum glycoside-L and polygonum cuspidatum glycoside-H groups was significantly increased, and the empty lacunar rate was significantly decreased (P<0.05). Micro-CT analysis showed that the bone volume fraction, trabecular number, and thickness increased, and the trabecular separation decreased in the polygonum cuspidatum glycoside-treated groups(P<0.05). Serum biochemical tests found that the serum Ca and P concentrations in the polygonum cuspidatum glycoside-L and polygonum cuspidatum glycoside-H groups were restored, the alkaline phosphatase levels decreased, and the transforming growth factor β1 levels increased (P<0.05). Western blot analysis showed that polygonum cuspidatum glycoside significantly inhibited the activation of the JAK2/STAT3 signaling pathway in the model group and promoted the expression of osteogenic differentiation marker genes such as Runx2, BMP2, and OPN (P<0.05). Compared with the polygonum cuspidatum glycoside-H group, the improvements in the polygonum cuspidatum glycoside-H+Colivelin group were somewhat weakened, indicating the importance of the JAK2/STAT3 signaling pathway in the action of polygonum cuspidatum glycoside.
CONCLUSION
polygonum cuspidatum glycoside promotes osteogenic differentiation, improves bone microstructure, and has significant therapeutic effects on rat SONFH by regulating the JAK2/STAT3 signaling pathway.
Animals
;
Male
;
Janus Kinase 2/physiology*
;
Rats, Sprague-Dawley
;
Rats
;
Signal Transduction/drug effects*
;
Glucosides/pharmacology*
;
STAT3 Transcription Factor/genetics*
;
Femur Head Necrosis/chemically induced*
;
Stilbenes/pharmacology*
4.Research on the mechanism of gentiopicroside preventing macrophage-mediated liver fibrosis by regulating the MIF-SPP1 signaling pathway in hepatic stellate cells.
Jixu WANG ; Yingbin ZHU ; Maoli CHEN ; Yongfeng HAN
Chinese Journal of Cellular and Molecular Immunology 2025;41(7):593-602
Objective To explore the mechanism by which gentiopicroside (GPS) prevents macrophage-mediated hepatic fibrosis by regulating the macrophage migration inhibitory factor (MIF)-secreted phosphoprotein 1 (SPP1) signaling pathway in hepatic stellate cells. Methods LX-2 cells were divided into control group, transforming growth factor β(TGF-β) group, and TGF-β combined with GPS (25, 50, 100, 150 μmol/mL) groups. Cell proliferation was detected by EDU assay, cell invasion was assessed by TranswellTM assay, and the protein expressions of α-smooth muscle actin (α-SMA) and type I collagen (COL1A1) were measured by Western blot. M1-type macrophage-conditioned medium (M1-CM) was used to treat LX-2 cells in the TGF-β group and TGF-β combined with GPS group. The concentrations of inducible nitric oxide synthase (iNOS) and arginase 1 (Arg1) in the cell supernatant, as well as cell proliferation, invasion ability, and the expressions of α-SMA and COL1A1 were detected. Bioinformatics analysis was performed to identify the target intersections of GPS, hepatic fibrosis, and macrophage-related genes. Drug affinity responsive target stability (DARTS) experiments and Western blot were used to verify the regulatory effect of GPS on MIF. Furthermore, LX-2 cells were divided into control group, TGF-β group, TGF-β combined with M2-CM group, TGF-β and oe-NC combined with M2-CM group, and TGF-β and oe-MIF combined with M2-CM group to analyze the concentrations of iNOS and Arg1 in the cell supernatant, as well as changes in cell proliferation, invasion, and the expressions of α-SMA and COL1A1. LX-2 cells were also divided into control group, TGF-β group, TGF-β combined with oe-NC group, TGF-β combined with oe-MIF group, and TGF-β and oe-MIF combined with GPS group to determine the protein expressions of MIF and SPP1 by Western blot. A rat model of hepatic fibrosis was constructed to explore the potential therapeutic effects of GPS on hepatic fibrosis in vivo. Results Compared with the control group, the proliferation and invasion abilities of LX-2 cells in the TGF-β group were increased, and the protein expressions of α-SMA and COL1A1 were enhanced. GPS intervention inhibited the proliferation and invasion of LX-2 cells under TGF-β conditions and reduced the expressions of α-SMA and COL1A1. Compared with the control group, the concentration of iNOS in the cell supernatant of the TGF-β group was upregulated, while the concentration of Arg1 was decreased. M1-CM treatment further increased the concentration of iNOS, decreased the concentration of Arg1, and promoted cell proliferation and invasion, as well as upregulated the expressions of α-SMA and COL1A1 on the basis of TGF-β intervention. However, GPS could reverse the effects of M1-CM intervention. Bioinformatics analysis revealed that MIF was one of the target intersections of GPS, hepatic fibrosis, and macrophage-related genes, and GPS could target and inhibit its expression. Compared with the TGF-β group, after M2-CM intervention, the concentration of iNOS in the cell supernatant decreased, the concentration of Arg1 increased, the proliferation and invasion abilities of LX-2 cells were reduced, and the expressions of α-SMA and COL1A1 were weakened. However, overexpression of MIF reversed the effects of M2-CM intervention. Western blot results showed that compared with the control group, the protein expressions of MIF and SPP1 were enhanced in the TGF-β group. Overexpression of MIF further enhanced the expressions of MIF and SPP1, while GPS intervention inhibited the expressions of MIF and SPP1. In the animal experiment, GPS intervention treatment alleviated liver injury in rats with hepatic fibrosis and inhibited the expressions of MIF and SPP1, as well as α-SMA and COL1A1 in liver tissue. Conclusion GPS may prevent macrophage-mediated hepatic fibrosis by inhibiting the MIF-SPP1 signaling pathway in hepatic stellate cells.
Hepatic Stellate Cells/metabolism*
;
Signal Transduction/drug effects*
;
Macrophage Migration-Inhibitory Factors/genetics*
;
Liver Cirrhosis/prevention & control*
;
Macrophages/drug effects*
;
Iridoid Glucosides/pharmacology*
;
Humans
;
Cell Proliferation/drug effects*
;
Animals
;
Cell Line
;
Collagen Type I/metabolism*
;
Collagen Type I, alpha 1 Chain
;
Intramolecular Oxidoreductases/genetics*
;
Rats
;
Transforming Growth Factor beta/pharmacology*
;
Actins/metabolism*
5.The effect of gentiopicroside on osteogenic differentiation of human bone marrow mesenchymal stem cells by regulating the SDF-1/CXCR4 signaling pathway.
Ruifang WANG ; Yingchun YANG ; Haibing QIAO ; Ying YANG
Chinese Journal of Cellular and Molecular Immunology 2025;41(9):784-789
Objective To investigate the effect of gentiopicroside on osteogenic differentiation of human bone marrow mesenchymal stem cells (BMSCs), and to determine whether its mechanism involves the stromal cell-derived factor 1(SDF-1)/C-X-C chemokine receptor 4 (CXCR4) pathway. Methods BMSCs were divided into six groups: normal culture control group, osteogenic induction model group, low-dose gentiopicroside (L-gentiopicroside, 10 μmol/L) group, medium-dose gentiopicroside (M-gentiopicroside, 20 μmol/L) group, high-dose gentiopicroside (H-gentiopicroside, 40 μmol/L) group, and H-gentiopicroside+SDF-1/CXCR4 pathway inhibitor (AMD3100) group (H-gentiopicroside+AMD3100, 40 μmol/L gentiopicroside+10 μg/mL AMD3100). Cell viability, apoptosis, ALP activity, mineralized nodule formation, and protein levels of the SDF-1/CXCR4 pathway were assessed using the CCK-8 assay, flow cytometry, ALP staining, Alizarin Red S staining, and Western blotting, respectively. Results No mineralized nodules were observed in either the control and model group, although the color of the model group deepened. Compared with the control group, the model group showed significantly increased A value, ALP activity, expression levels of Runt related transcription factor 2 (RUNX2), osteopontin (OPN), SDF-1, CXCR4 proteins, along with a lower apoptosis rate. Compared with the model group, the L-gentiopicroside, M-gentiopicroside and H-gentiopicroside groups showed dose-dependently (L
6.Catalpol reduces liver toxicity of triptolide in mice by inhibiting hepatocyte ferroptosis through the SLC7A11/GPX4 pathway: testing the Fuzheng Zhidu theory for detoxification.
Linluo ZHANG ; Changqing LI ; Lingling HUANG ; Xueping ZHOU ; Yuanyuan LOU
Journal of Southern Medical University 2025;45(4):810-818
OBJECTIVES:
To investigate the protective effect of catalpol against triptolide-induced liver injury and explore its mechanism to test the Fuzheng Zhidu theory for detoxification.
METHODS:
C57BL/6J mice were randomized into blank control group, catalpol group, triptolide group and triptolide+catalpol group. After 13 days of treatment with the agents by gavage, the mice were examined for liver tissue pathology, liver function, hepatocyte subcellular structure, lipid peroxidation, ferrous ion deposition and expressions of ferroptosis-related proteins in the liver. In a liver cell line HL7702, the effect of catalpol or the ferroptosis inhibitor Fer-1 on triptolide-induced cytotoxicity was tested by examining cell functions, Fe2+ concentration, lipid peroxidation, ROS level and the ferroptosis-related proteins.
RESULTS:
In C57BL/6J mice, catalpol significantly alleviated triptolide-induced hepatic injury, lowered the levels of ALT, AST and LDH, and reversed the elevation of Fe2+ concentration and MDA level and the reduction of GPX level. In HL7702 cells, inhibition of ferroptosis by Fer-1 significantly reversed triptolide-induced elevation of ALT, AST and LDH levels. Western blotting and qRT-PCR demonstrated that catalpol reversed abnormalities in expressions of SLC7A11, FTH1 and GPX4 at both the mRNA and protein levels in triptolide-treated HL7702 cells.
CONCLUSIONS
The combined use of catalpol can reduce the hepatotoxicity of triptolide in mice by inhibiting excessive hepatocyte ferroptosis through the SLC7A11/GPX4 pathway.
Animals
;
Phenanthrenes/toxicity*
;
Ferroptosis/drug effects*
;
Diterpenes/toxicity*
;
Epoxy Compounds/toxicity*
;
Mice, Inbred C57BL
;
Hepatocytes/metabolism*
;
Mice
;
Phospholipid Hydroperoxide Glutathione Peroxidase
;
Iridoid Glucosides/pharmacology*
;
Liver/metabolism*
;
Chemical and Drug Induced Liver Injury/prevention & control*
;
Male
;
Amino Acid Transport System y+/metabolism*
7.Salidroside inhibits proliferation of gastric cancer cells by regulating the miR-1343-3p-OGDHL/PDHB glucose metabolic axis.
Xinrui HOU ; Zhendong ZHANG ; Mingyuan CAO ; Yuxin DU ; Xiaoping WANG
Journal of Southern Medical University 2025;45(6):1226-1239
OBJECTIVES:
To investigate the mechanism through which salidroside inhibits proliferation of gastric cancer (GC) cells focusing on glucose metabolic reprogramming pathways.
METHODS:
High-throughput sequencing combined with bioinformatics analysis was employed to identify the potential targets of salidroside in human GC MGC-803 cells. Liposome-mediated transfection experiments were carried out to validate the functional and mechanistic roles of these targets. CCK-8 and colony formation assays were used to assess the effects of salidroside on GC cell viability and clonogenic ability. qRT-PCR, Western blotting, and biochemical assay kits were used to analyze the regulatory effects of salidroside on the miR-1343-3p-OGDHL/PDHB enzyme complex-pyruvate metabolic pathway in GC cells.
RESULTS:
Bioinformatics analysis suggested that the tumor-suppressive factor miR-1343-3p negatively regulated the key glycolytic enzyme gene oxoglutarate dehydrogenase-like (OGDHL) in GC cells, and OGDHL and pyruvate dehydrogenase E1 subunit beta (PDHB) were both significantly upregulated in GC tissues, which was close by correlated with reduced survival rates of GC patients. In MGC-803 cells, salidroside treatment significantly enhanced the expression level of miR-1343-3p and downregulated OGDHL expression, resulting in disruption of the stability of PDHB, reduced pyruvate oxidative decarboxylation, and consequently decreased production of acetyl-CoA and ATP.
CONCLUSIONS
Salidroside inhibits GC cell proliferation possibly by regulating the miR-1343-3p-OGDHL/PDHB enzyme complex-pyruvate metabolic pathway, which provides new insights into its anti-tumor mechanisms and suggests new strategies for targeted therapy for GC.
Humans
;
Stomach Neoplasms/pathology*
;
MicroRNAs/genetics*
;
Cell Proliferation/drug effects*
;
Glucosides/pharmacology*
;
Phenols/pharmacology*
;
Cell Line, Tumor
;
Glucose/metabolism*
;
Pyruvate Dehydrogenase (Lipoamide)/metabolism*
8.Gastrodin inhibits ferroptosis to alleviate hypoxic-ischemic brain damage in neonatal mice by activating GPX4/SLC7A11/FTH1 signaling.
Tao GUO ; Bolin CHEN ; Jinsha SHI ; Xianfeng KUANG ; Tengyue YU ; Song WEI ; Xiong LIU ; Rong XIAO ; Juanjuan LI
Journal of Southern Medical University 2025;45(10):2071-2081
OBJECTIVES:
To evaluate the therapeutic effect of gastrodin against hypoxic-ischemic brain damage (HIBD) in neonatal mice and explore the role of GPX4/SLC7A11/FTH1 signaling in mediating its effect.
METHODS:
Twenty-four 9- to 11-day-old C57BL/6J mice were randomized equally into 4 groups for sham operation, HIBD modeling by right common carotid artery ligation and subsequent exposure to hypoxia for 1 h, or gastrodin treatment at 100 or 200 mg/kg before and at 1 and 2 days after modeling. The mice then underwent neurological assessment (Zea-Longa scores), and the cerebral cortical penumbra tissue were collected for HE and Nissl staining, detection of ferroptosis biomarkers and protein expressions of GPX4, SLC7A11, and FTH1 with Western blotting and immunofluorescence co-localization, and observation of mitochondrial ultrastructure with electron microscopy. In cultured HT22 neuronal cells with oxygen-glucose deprivation (OGD) for 2 h, the effects of pretreatments with 0.5 mmol/L gastrodin, 10 μmol/L RSL3 (a GPX4 inhibitor), alone or in combination, were analyzed on expressions of ferroptosis-related proteins, cellular Fe²⁺, ROS, lipid peroxidation, MDA, and GSH levels, mitochondrial membrane potential (JC-1), and cell viability.
RESULTS:
Gastrodin treatment at the two doses both significantly ameliorated HIBD and neurological deficits of the mice, reduced mitochondrial damage and Fe²⁺, MDA and ROS levels, increased GSH level, and upregulated GPX4, SLC7A11, and FTH1 protein expressions. In HT22 cells, gastrodin pretreatment obviously attenuated OGD-induced ferroptosis and improved cell viability and mitochondrial function. Co-treatment with RSL3 potently abrogated the inhibitory effects of gastrodin on Fe²⁺, ROS, BODIPY-C11, and MDA levels and attenuated its protective effects on GSH level, cell viability, and mitochondrial membrane potential.
CONCLUSIONS
Gastrodin provides neuroprotective effects in neonatal mice with HIBD by suppressing neuronal ferroptosis via upregulating the GPX4/SLC7A11/FTH1 signaling pathway.
Animals
;
Ferroptosis/drug effects*
;
Hypoxia-Ischemia, Brain/drug therapy*
;
Mice
;
Mice, Inbred C57BL
;
Signal Transduction/drug effects*
;
Phospholipid Hydroperoxide Glutathione Peroxidase
;
Glucosides/pharmacology*
;
Animals, Newborn
;
Benzyl Alcohols/pharmacology*
;
Amino Acid Transport System y+/metabolism*
9.Salidroside alleviates high glucose and ethanol-induced pyroptosis in insulinoma cells.
Xiao-Han LI ; Xu GUO ; Shi-Qi CHEN ; Yan-Guo GAO ; Jun ZHOU ; Yong-Hong ZHANG ; Qi-Bin WANG ; Li CHEN ; Tao ZHENG
China Journal of Chinese Materia Medica 2024;49(22):6181-6189
This study established a pyroptosis injury model by stimulating insulinoma cells(INS-1) of rats with high glucose(HG) and observed the impact of additional ethanol(ET) exposure on cell pyroptosis, as well as the intervention effect of salidroside(SAL). INS-1 cells were cultured and divided into a normal control group(NG), an HG group, an HG + ET(100 mmol·L~(-1)) group, and an HG + ET + SAL(1-100 μmol·L~(-1)) group. After 72 hours of treatment, cell viability was assessed using the cell counting kit-8(CCK-8) assay. The number of pyroptotic bodies was observed under a microscope. Western blot was used to detect changes in the intracellular Nod-like receptor protein 3(NLRP3)/gasdermin D(GSDMD) signaling pathway and adenosine monophosphate-activated protein kinase(AMPK) activity. A fluorescence probe was used to detect changes in intracellular reactive oxygen species(ROS) levels. Time-resolved fluorescence resonance energy transfer(TR-FRET) technology was employed to observe the effect of SAL on recombinant AMPK protein kinase activity in vitro. The results showed that compared to the NG group, HG exposure induced an increase in the number of pyroptotic bodies, elevated ROS levels, and activation of the NLRP3/GSDMD signaling pathway in INS-1 cells. Compared to the HG group, HG + ET exposure further exacerbated these changes. Compared to the HG + ET group, SAL dose-dependently increased cell viability, reduced the formation of pyroptotic bodies in INS-1 cells, and inhibited excessive ROS production, overactivation of the NLRP3/GSDMD signaling pathway, and the decrease in AMPK activity. TR-FRET experiments indicated that SAL could directly activate AMPK. When INS-1 cells were pretreated with an AMPK inhibitor, the effects of SAL on increasing cell viability, alleviating the formation of pyroptotic bodies, and inhibiting excessive ROS production were abolished. These results suggest that SAL can alleviate HG combined with ET-induced exacerbation of INS-1 pyroptosis by activating AMPK.
Pyroptosis/drug effects*
;
Animals
;
Rats
;
Glucose/metabolism*
;
Insulinoma/metabolism*
;
Ethanol/pharmacology*
;
Reactive Oxygen Species/metabolism*
;
Glucosides/pharmacology*
;
Phenols/pharmacology*
;
Cell Line, Tumor
;
Signal Transduction/drug effects*
;
NLR Family, Pyrin Domain-Containing 3 Protein/genetics*
;
Cell Survival/drug effects*
;
AMP-Activated Protein Kinases/metabolism*
;
Phosphate-Binding Proteins/genetics*
10.Mechanism of aucubin in regulating ribosome biogenesis and inhibiting injury of nucleus pulposus cells and extracellular matrix degradation.
Ling-Hui LI ; Shang-Quan WANG ; Kai SUN ; Xun-Lu YIN ; Li-Guo ZHU ; Xu WEI
China Journal of Chinese Materia Medica 2024;49(21):5713-5720
This study aimed to investigate the effect of aucubin(AU) on injury of nucleus pulposus cells and extracellular matrix(ECM) degradation and its mechanism. The nucleus pulposus cell injury model was established by interleukin-1β(IL-1β) and treated with AU or phosphatidylinositol 3-kinase(PI3K) inhibitor LY294002. CCK-8 experiment was conducted to test cell proliferation. EdU staining method was employed to detect cell injury. Flow cytometry was used to detect cell apoptosis. Western blot was used to detect protein levels of cleaved-caspase-3, B-cell lymphoma(Bcl-2), Bcl-2 associated X protein(Bax), type Ⅱ collagen(collagen Ⅱ), aggregation proteoglycans(aggrecan), PI3K, and mammalian target of rapamycin(mTOR). qPCR was used to detect the rRNA level of 5S, 18S, and 28S. Ethynyluridine was used to label nascent RNA. The results showed that IL-1β could significantly cause injury of nucleus pulposus cells and increase the apoptosis rate of nucleus pulposus cells and the expression of apoptosis protein cleaved-caspase-3 and Bax. At the same time, IL-1β down-regulated the expression of anti-apoptotic protein Bcl-2 and collagen Ⅱ and aggrecan, the main components of ECM. On this basis, AU intervention could improve the injury of nucleus pulposus cells, reduce the apoptosis of nucleus pulposus cells and the expression of cleaved-caspase-3 and Bax, and increase the expression of Bcl-2, collagen Ⅱ, and aggrecan. Compared with IL-1β, AU could up-regulate the phosphorylation level of PI3K and mTOR, and LY294002 could reverse the injury of nucleus pulposus cells and improve ECM degradation induced by AU. In addition, AU also could save lowered rRNA levels of 5S, 18S, and 28S induced by IL-1β and improve RNA synthesis. PI3K inhibitor LY294002 intervention could reduce the promoting effect of AU on ribosome biogenesis. The above results suggest that AU can improve the injury of nucleus pulposus cells and ECM degradation, and its mechanism of action is related to its activation of the PI3K/mTOR pathway to promote ribosome biogenesis.
Nucleus Pulposus/cytology*
;
Extracellular Matrix/drug effects*
;
Animals
;
Iridoid Glucosides/pharmacology*
;
Apoptosis/drug effects*
;
Interleukin-1beta/metabolism*
;
Phosphatidylinositol 3-Kinases/genetics*
;
Rats
;
Cell Proliferation/drug effects*
;
TOR Serine-Threonine Kinases/genetics*
;
Rats, Sprague-Dawley
;
Humans
;
Signal Transduction/drug effects*
;
Caspase 3/genetics*
;
Proto-Oncogene Proteins c-bcl-2/genetics*

Result Analysis
Print
Save
E-mail