1.Clematichinenoside AR protects bone marrow mesenchymal stem cells from hypoxia-induced apoptosis by maintaining mitochondrial homeostasis.
Zi-Tong ZHAO ; Peng-Cheng TU ; Xiao-Xian SUN ; Ya-Lan PAN ; Yang GUO ; Li-Ning WANG ; Yong MA
China Journal of Chinese Materia Medica 2025;50(5):1331-1339
This study aims to elucidate the role and mechanism of clematichinenoside AR(CAR) in protecting bone marrow mesenchymal stem cells(BMSCs) from hypoxia-induced apoptosis. BMSCs were isolated by the bone fragment method and identified by flow cytometry. Cells were cultured under normal conditions(37℃, 5% CO_2) and hypoxic conditions(37℃, 90% N_2, 5% CO_2) and treated with CAR. The BMSCs were classified into eight groups: control(normal conditions), CAR(normal conditions + CAR), hypoxia 24 h, hypoxia 24 h + CAR, hypoxia 48 h, hypoxia 48 h + CAR, hypoxia 72 h, and hypoxia 72 h + CAR. The cell counting kit-8(CCK-8) assay and terminal-deoxynucleoitidyl transferase mediated nick end labeling(TUNEL) were employed to measure cell proliferation and apoptosis, respectively. The number of mitochondria and mitochondrial membrane potential were measured by MitoTracker®Red CM-H2XRo staining and JC-1 staining, respectively. The level of reactive oxygen species(ROS) was measured with the DCFH-DA fluorescence probe. The protein levels of B-cell lymphoma-2 associated X protein(BAX), caspase-3, and optic atrophy 1(OPA1) were determined by Western blot. The results demonstrated that CAR significantly increased cell proliferation. Compared with the control group, the hypoxia groups showed increased apoptosis rates, reduced mitochondria, elevated ROS levels, decreased mitochondrial membrane potential, upregulated expression of BAX and caspase-3, and downregulated expression of OPA1. In comparison to the corresponding hypoxia groups, CAR intervention significantly decreased the apoptosis rate, increased mitochondria, reduced ROS levels, elevated mitochondrial membrane potential, downregulated the expression of BAX and caspase-3, and upregulated the expression of OPA1. Therefore, it can be concluded that CAR may exert an anti-apoptotic effect on BMSCs under hypoxic conditions by regulating OPA1 to maintain mitochondrial homeostasis.
Mesenchymal Stem Cells/metabolism*
;
Apoptosis/drug effects*
;
Mitochondria/metabolism*
;
Animals
;
Rats
;
Cell Hypoxia/drug effects*
;
Homeostasis/drug effects*
;
Reactive Oxygen Species/metabolism*
;
Rats, Sprague-Dawley
;
Membrane Potential, Mitochondrial/drug effects*
;
Saponins/pharmacology*
;
Caspase 3/genetics*
;
Male
;
bcl-2-Associated X Protein/genetics*
;
Bone Marrow Cells/metabolism*
;
Cell Proliferation/drug effects*
;
Protective Agents/pharmacology*
;
Cells, Cultured
2.Alleviation of hypoxia/reoxygenation injury in HL-1 cells by ginsenoside Rg_1 via regulating mitochondrial fusion based on Notch1 signaling pathway.
Hui-Yu ZHANG ; Xiao-Shan CUI ; Yuan-Yuan CHEN ; Gao-Jie XIN ; Ce CAO ; Zi-Xin LIU ; Shu-Juan XU ; Jia-Ming GAO ; Hao GUO ; Jian-Hua FU
China Journal of Chinese Materia Medica 2025;50(10):2711-2718
This paper explored the specific mechanism of ginsenoside Rg_1 in regulating mitochondrial fusion through the neurogenic gene Notch homologous protein 1(Notch1) pathway to alleviate hypoxia/reoxygenation(H/R) injury in HL-1 cells. The relative viability of HL-1 cells after six hours of hypoxia and two hours of reoxygenation was detected by cell counting kit-8(CCK-8). The lactate dehydrogenase(LDH) activity in the cell supernatant was detected by the lactate substrate method. The content of adenosine triphosphate(ATP) was detected by the luciferin method. Fluorescence probes were used to detect intracellular reactive oxygen species(Cyto-ROS) levels and mitochondrial membrane potential(ΔΨ_m). Mito-Tracker and Actin were co-imaged to detect the number of mitochondria in cells. Fluorescence quantitative polymerase chain reaction and Western blot were used to detect the mRNA and protein expression levels of Notch1, mitochondrial fusion protein 2(Mfn2), and mitochondrial fusion protein 1(Mfn1). The results showed that compared with that of the control group, the cell activity of the model group decreased, and the LDH released into the cell culture supernatant increased. The level of Cyto-ROS increased, and the content of ATP decreased. Compared with that of the model group, the cell activity of the ginsenoside Rg_1 group increased, and the LDH released into the cell culture supernatant decreased. The level of Cyto-ROS decreased, and the ATP content increased. Ginsenoside Rg_1 elevated ΔΨ_m and increased mitochondrial quantity in HL-1 cells with H/R injury and had good protection for mitochondria. After H/R injury, the mRNA and protein expression levels of Notch1 and Mfn1 decreased, while the mRNA and protein expression levels of Mfn2 increased. Ginsenoside Rg_1 increased the mRNA and protein levels of Notch1 and Mfn1, and decreased the mRNA and protein levels of Mfn2. Silencing Notch1 inhibited the action of ginsenoside Rg_1, decreased the mRNA and protein levels of Notch1 and Mfn1, and increased the mRNA and protein levels of Mfn2. In summary, ginsenoside Rg_1 regulated mitochondrial fusion through the Notch1 pathway to alleviate H/R injury in HL-1 cells.
Ginsenosides/pharmacology*
;
Receptor, Notch1/genetics*
;
Signal Transduction/drug effects*
;
Mice
;
Animals
;
Mitochondrial Dynamics/drug effects*
;
Mitochondria/metabolism*
;
Cell Line
;
Reactive Oxygen Species/metabolism*
;
Oxygen/metabolism*
;
Cell Hypoxia/drug effects*
;
Cell Survival/drug effects*
;
Membrane Potential, Mitochondrial/drug effects*
;
Humans
3.Protective mechanism of Chaihu Shugan San against CORT-induced damage in PC12 cells based on mitochondrial dynamics.
Ling-Yuan ZHANG ; Qi-Qi ZHENG ; Jia-Li SHI ; Pei-Fang WANG ; Jia-Li LU ; Jian-Ying SHEN
China Journal of Chinese Materia Medica 2025;50(16):4546-4554
In this report, the protective effect and molecular mechanism of Chaihu Shugan San-containing serum on corticosterone(CORT)-induced mitochondrial damage in pheochromocytoma(PC12) cells was studied based on CORT-induced rat PC12 cell model. The cultured cells were divided into five groups: blank control group, CORT group(400 μmol·L~(-1) CORT), Chaihu Shugan San-containing serum group(400 μmol·L~(-1) CORT + 10% Chaihu Shugan San-containing serum), control serum group(400 μmol·L~(-1) CORT + 10% control serum), and fluoxetine group(400 μmol·L~(-1) CORT + 10% fluoxetine-containing serum). The study was carried out by cell activity detection, mitochondrial morphology observation, membrane potential measurement, energy metabolism analysis, and mitochondrial dynamics-related protein detection. The results showed that CORT treatment significantly reduced the survival rate of PC12 cells, altered mitochondrial morphology, and decreased mitochondrial membrane potential and adenosine triphosphate(ATP) synthetic rate. Both Chaihu Shugan San-and fluoxetine-containing serum significantly increased the survival rate of CORT-treated PC12 cells and the ATP synthetic rate in the mitochondria. Unlike fluoxetine, Chaihu Shugan San-containing serum significantly inhibited the decrease in mitochondrial membrane potential caused by CORT and increased the oxygen consumption rate(OCR) values of both mitochondrial maximum respiration and reserve respiration capacity. Western blot analysis showed that CORT induced upregulated protein expressions of dynamin-related protein 1(Drp1) and peroxisome proliferator-activated receptor gamma co-activator 1α(PGC-1α) in PC12 cells and specific protein expression of optic atrophy protein 1(OPA1), yet it repressed the protein expressions of silent information regulator 1(SIRT1) and mitochondrial fusion protein 1(Mfn1) in PC12 cells. Both Chaihu Shugan San-and fluoxetine-containing serum significantly inhibited the protein expression of Drp1. However, only Chaihu Shugan San-containing serum could significantly inhibit the CORT-induced upregulation protein of PGC-1α. RESULTS:: herein suggest that Chaihu Shugan San-containing serum can alleviate CORT-induced damage in PC12 cells, which may be related to the mitochondrial fragmentation/lipid peroxidation protection by Drp1 inhibition, as well as mitochondrial dynamics and energy metabolism mediated by PGC-1α/SIRT1 signaling pathway.
Animals
;
PC12 Cells
;
Rats
;
Mitochondrial Dynamics/drug effects*
;
Mitochondria/metabolism*
;
Corticosterone/adverse effects*
;
Membrane Potential, Mitochondrial/drug effects*
;
Drugs, Chinese Herbal/pharmacology*
;
Protective Agents/pharmacology*
;
Cell Survival/drug effects*
4.Impact of tyrosine phosphorylation site mutation in FUNDC1 protein on mitophagy in H9c2 cardiomyocytes.
Zhaoyang ZHANG ; Yanli YU ; Jieyun WU ; Wei TIAN ; Jingman XU
Chinese Journal of Cellular and Molecular Immunology 2025;41(7):629-636
Objective To investigate the effect of FUNDC1 tyrosine phosphorylation site mutations on mitophagy in H9c2 myocardial cells by constructing tyrosine site mutant plasmids (Y11 and Y18) of the FUN14 domain-containing protein 1 (FUNDC1). Methods The mutant plasmids constructed by whole-gene synthesis were transfected into rat myocardial H9c2 cells and divided into five groups: empty plasmid group, FUNDC1 overexpression group, Y11 mutant group, Y18 mutant group, and Y11 combined with Y18 mutant group. The viability of H9c2 cells was assessed using the CCK-8 assay. Additionally, tetramethylrhodamine ethyl ester (TMRE) staining was utilized to detect mitochondrial membrane potential. The protein expression levels of FUNDC1, translocase of the outer mitochondrial membrane 20 (TOM20), and cytochrome c oxidase IV (COX IV) were detected by Western blot analysis. Confocal microscopy was used to evaluate transfection efficiency as well as the co-localization of mitochondria and lysosomes. Results The FUNDC1 overexpression, Y11, Y18, and Y11 combined with Y18 mutant plasmids were successfully constructed. After plasmid transfection, widespread GFP fluorescence expression was observed under confocal microscopy. Compared with the empty plasmid group, FUNDC1 protein expression levels were significantly increased in the FUNDC1 overexpression group, Y11 mutation group, Y18 mutation group, and Y11 combined with Y18 mutation group, while cell viability and mitochondrial membrane potential showed no significant changes. Compared to the empty plasmid group, cells transfected with Y18 and Y11 combined with Y18 mutant plasmids showed increased TOM20 and COX IV expression levels and decreased mitochondrial-lysosomal co-localization. Conclusion Transfection with FUNDC1 Y18 or Y11 combined with Y18 mutant plasmids inhibited mitophagy in H9c2 myocardial cells.
Animals
;
Rats
;
Mitophagy/genetics*
;
Myocytes, Cardiac/cytology*
;
Mitochondrial Proteins/metabolism*
;
Mutation
;
Phosphorylation
;
Tyrosine/genetics*
;
Cell Line
;
Membrane Proteins/metabolism*
;
Membrane Potential, Mitochondrial
5.Study on the effect of ATPIF1 on the anti-tumor activity of CAR-NK92 cells by regulating glycolytic capacity.
Biao LIU ; Xue GONG ; Biliang HU ; Chunlei GUO ; Genshen ZHONG
Chinese Journal of Cellular and Molecular Immunology 2025;41(10):865-874
Objective To investigate the effect of ATP synthase inhibitory factor 1 (ATPIF1) on the antitumor activity of chimeric antigen receptor (CAR)-NK92 cells. Methods HER2-targeted CAR-NK92 cells with ATPIF1 overexpression or knockdown were constructed. CAR-positive expression rate was detected by flow cytometry. Cell proliferation capacity was measured using CCK-8 assay. Glycolytic capacity was analyzed by Seahorse metabolic analyzer. Mitochondrial membrane potential levels were detected using JC-1 probe. Target cell lysis rate was evaluated by firefly luciferase reporter assay. Expression levels of CD107a, natural-killer group 2 member D (NKG2D), granzyme B (GzmB), perforin, and interleukin 2 (IL-2) were detected via flow cytometry. Quantitative real-time PCR was used to measure the expression of interferon-induced protein with tetratricopeptide repeats 1 (IFIT1), tumor necrosis factor α (TNF-α), ATPIF1, and hexokinase 1 (HK1). The impact of glycolytic inhibition by 2-Deoxy-D-glucose (2-DG) on CAR-NK92 antitumor capacity was examined. Results Successfully generated HER2-targeting control CAR-NK92 cells, as well as ATPIF1-overexpressing and ATPIF1 knockdown CAR-NK92 cells. The ATPIF1-overexpressing CAR-NK92 cells showed significantly enhanced target cell lysis rate, elevated expression levels of NKG2D and CD107a, increased secretion capacities of Granzyme B (GzmB) and IL-2, and upregulated mRNA expression levels of IFIT1 and TNF-α, while ATPIF1-knockdown cells exhibited opposite effects. ATPIF1 overexpression induced metabolic reprogramming in CAR-NK92 cells, manifested by significantly decreased mitochondrial membrane potential (δpsim), markedly upregulated HK1 mRNA expression, and enhanced basal glycolysis and glycolytic capacity. After glycolysis inhibition with 2-DG (5 μmol/L), both ATPIF1-overexpressing and knockdown CAR-NK92 cells showed no significant differences in NKG2D and CD107a expression levels compared to control cells. Conclusion ATPIF1 regulates the antitumor activity of CAR-NK92 cells through modulating glycolytic metabolism. Overexpression of ATPIF1 can enhance the antitumor efficacy of CAR-NK92 cells.
Humans
;
Glycolysis
;
Killer Cells, Natural/metabolism*
;
Receptors, Chimeric Antigen/immunology*
;
Granzymes/genetics*
;
Hexokinase/metabolism*
;
Cell Line, Tumor
;
Interleukin-2/genetics*
;
Cell Proliferation
;
NK Cell Lectin-Like Receptor Subfamily K/genetics*
;
Membrane Potential, Mitochondrial
6.In vitro effects of antidepressants on human sperm function.
Rita António SANTOS ; Ana Paula SOUSA ; Teresa ALMEIDA-SANTOS ; João RAMALHO-SANTOS ; Renata Santos TAVARES
Asian Journal of Andrology 2025;27(1):30-36
Depression currently affects about 280 million people worldwide and its prevalence has been increasing dramatically, especially among the young and people of reproductive age, which consequently leads to an increase in antidepressant consumption. Antidepressants are associated with sexual dysfunction in both men and women; however, their role in male fertility has been scarcely studied. Fluoxetine and sertraline, two serotonin reuptake inhibitors (SSRIs), are among the most prescribed antidepressants worldwide. To determine their possible effects, human sperm cells were exposed to either sertraline or fluoxetine at concentrations previously found in blood and seminal fluid of patients undergoing treatment. Spermatozoa were incubated for up to 24 h at 37°C and 5% CO 2 , and important functional parameters such as sperm motility, viability, mitochondrial membrane potential, cellular reactive oxygen species (ROS) production, chromatin/DNA integrity, acrosome status, and tyrosine phosphorylation were assessed. At low levels, fluoxetine consistently decreased progressive motility throughout time while promoting fluctuations in ROS levels and sperm capacitation. Nevertheless, it did not affect viability, mitochondrial membrane potential, acrosome reaction nor chromatin/DNA integrity. Sertraline, on the other hand, had little to nonsignificant impact at low doses, but affected almost all tested parameters at supratherapeutic concentrations. Altogether, our results suggest that both antidepressants may impair sperm function, possibly through different mechanisms of action, but fluoxetine is the only exhibiting mild negative effects at doses found in vivo .
Humans
;
Male
;
Spermatozoa/drug effects*
;
Fluoxetine/pharmacology*
;
Sperm Motility/drug effects*
;
Sertraline/pharmacology*
;
Reactive Oxygen Species/metabolism*
;
Antidepressive Agents/pharmacology*
;
Membrane Potential, Mitochondrial/drug effects*
;
Sperm Capacitation/drug effects*
;
Selective Serotonin Reuptake Inhibitors/pharmacology*
;
Cell Survival/drug effects*
;
Acrosome Reaction/drug effects*
7.Triclocarban impacts human sperm motility by inhibiting glycolysis and oxidative phosphorylation.
Long-Long FU ; Wei-Zhou WANG ; Yan FENG ; Fu CHEN ; Bin LIU ; Liang HUANG ; Lin-Yuan ZHANG ; Lei CHEN
Asian Journal of Andrology 2025;27(6):707-713
Triclocarban (TCC) is a broad-spectrum antimicrobial widely used in various personal care products, textiles, and children's toys. TCC has potential reproductive and developmental toxicity in animals. However, little is known regarding the effect of TCC on human sperm function. In this study, an in vitro assay was used to investigate the effects of TCC on normal human spermatozoa and the possible underlying mechanisms involved. Semen from healthy male donors was collected and cultured in complete Biggers, Whitten and Whittingham (BWW) and low-sugar BWW media, followed by treatment with TCC at concentrations of 0, 0.1 µmol l -1 , 1 µmol l -1 , 10 µmol l -1 , and 100 µmol l -1 for 4 h. TCC was found to reduce the sperm total motility and progressive motility. Moreover, the sperm kinematic parameters, straight-line velocity (VSL), average path velocity (VAP), and curvilinear velocity (VCL) were affected in a dose-dependent manner. After treatment with TCC at the lowest effective concentration of 10 µmol l -1 , TCC caused a significant decrease in mitochondrial adenosine triphosphate (ATP) production and mitochondrial membrane potential (MMP) and a significant increase in reactive oxygen species (ROS), similar to the observations with the positive control carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (FCCP), suggesting that TCC may decrease sperm motility by affecting the oxidative phosphorylation (OXPHOS) pathway. In a sugar-free and low-sugar BWW culture environment, TCC enhanced the damaging effect on sperm motility and ATP, MMP, and lactate decreased significantly, suggesting that TCC may also affect the glycolytic pathway that supplies energy to spermatozoa. This study demonstrates a possible mechanism of TCC toxicity in spermatozoa involving both the OXPHOS and glycolysis pathways.
Male
;
Sperm Motility/drug effects*
;
Humans
;
Carbanilides/pharmacology*
;
Oxidative Phosphorylation/drug effects*
;
Glycolysis/drug effects*
;
Membrane Potential, Mitochondrial/drug effects*
;
Adenosine Triphosphate/metabolism*
;
Spermatozoa/metabolism*
;
Reactive Oxygen Species/metabolism*
;
Mitochondria/metabolism*
8.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
9.Roles of reactive oxygen species and lactate dehydrogenase isoenzyme X in changes of sperm mitochondrial membrane in patients with varicocele-induced infertility.
Xiao-Xia ZHANG ; Ru-Yao LI ; Liang-Liang YU ; Jun ZHOU
National Journal of Andrology 2025;31(1):19-24
OBJECTIVE:
To explore the role of reactive oxygen species (ROS) and lactate dehydrogenase isoenzyme X (LDH-X) in the changes of sperm mitochondrial membrane potential (MMP) in infertility patients with varicocele (VC).
METHODS:
This study included 38 infertility patients with VC (VCinf), 35 non-VC infertile males (NVCinf), and 30 normal fertile men as controls. We obtained the routine semen parameters using the sperm quality analysis system, examined the contents of LDH-X in the seminal plasma and sperm with the automatic biochemical analyzer, measured the level of malondialdehyde (MDA) in seminal plasma by thiobarbituric acid (TBA) colorimetry, and determined the expressions of mitochondrial membrane potential (MMP) and LDH-X mRNA in the sperm using JC-1 fluorescence probe and RT-PCR.
RESULTS:
No statistically significant differences were observed among the three groups of subjects in age, semen pH value, semen volume and sperm concentration (P > 0.05). Compared with the normal fertile controls, the patients in the VCinf and NVCinf groups showed significantly decreased sperm motility ([52.36 ± 12.48]% vs [34.74 ± 15.23]% vs [25.76 ± 13.73]%, P< 0.05), percentage of progressively motile sperm (PMS) ([42.54 ± 13.58]% vs [29.10 ± 14.17]% vs [20.95 ± 12.33]%, P< 0.05), sperm LDH-X ([16.46 ± 5.47] vs [13.63 ± 4.50] vs [10.18 ± 3.00] mU/106, P< 0.05), sperm MMP ([48.04 ± 11.62]% vs [40.86 ± 12.69]% vs [34.41 ± 13.93]%, P< 0.05) and expression of sperm LDH-X mRNA (P< 0.05). but increased seminal plasma LDH-X ([935.36 ± 229.48] vs [1241.05 ± 337.07] vs [1425.08 ± 469.35] U/L, P< 0.05), seminal plasma/whole sperm LDH-X ([1.06 ± 0.35] vs [1.40 ± 0.34] vs [1.63 ± 0.66], P< 0.05), and content of seminal plasma MDA ([1.10 ± 0.19] vs [1.59 ± 0.27] vs [2.00 ± 0.22] nmol/ml, P< 0.05).
CONCLUSION
Excessive ROS in the reproductive system of VCinf patients reduces the content of MMP and causes the overflow of LDH-X out of sperm cells. Therefore the decrease of sperm LDH-X may be accompanied by that of MMP.
Humans
;
Male
;
Infertility, Male/etiology*
;
Varicocele/metabolism*
;
Adult
;
Reactive Oxygen Species/metabolism*
;
Spermatozoa/metabolism*
;
L-Lactate Dehydrogenase/metabolism*
;
Membrane Potential, Mitochondrial
;
Isoenzymes/metabolism*
;
Case-Control Studies
;
Young Adult
;
Mitochondrial Membranes/metabolism*
10.Chrysophanol Induces Cell Death and Inhibits Invasiveness through Alteration of Calcium Levels in HepG2 Human Liver Cancer Cells.
Shu-Chao CHEN ; Qiao-Wen CHEN ; Chih-Yuan KO
Chinese journal of integrative medicine 2025;31(5):434-440
OBJECTIVE:
To investigate the effect of chrysophanol, a phytochemical derived from Radix et Rhizoma Rhei on HepG2 liver cancer cells.
METHODS:
HepG2 cell line was treated with different concentrations chrysophanol (0-100 μmol/L) for 24 h. The cell counting kit 8 assay was employed to assess cell viability. Intracellular calcium levels were examined using Fluo-4 AM and Mag-fluo-4 AM staining, followed by flow cytometry analysis. Mitochondrial membrane potential was measured with JC-1 assay kit. Additionally, the expressions of key proteins such as p-JNK, Bax, cytochrome c (Cyt C), cleaved caspase-3 (cCaspase-3), and caspase-8 were analyzed by Western blot. The inhibitory effects of chrysophanol on the invasion of cells were determined using a Transwell assay. Analysis of invasiveness was conducted by wound healing assay.
RESULTS:
Chrysophanol significantly reduced the proliferation of HepG2 liver cancer cells by affecting intracellular calcium distribution, diminishing mitochondrial membrane potential, and enhancing the expressions of p-JNK, Bax, Cyt C, cCaspase-3, and caspase-8 in the groups treated with 75 or 100 μmol/L chrysophanol compared to the control group (P<0.05). Additionally, 75 and 100 μmol/L chrysophanol exhibited inhibitory effects on cell migration and wound healing.
CONCLUSION
Chrysophanol demonstrates potential against HepG2 liver cancer cells, suggesting its potential use as a therapeutic agent for liver cancer treatment.
Humans
;
Calcium/metabolism*
;
Hep G2 Cells
;
Liver Neoplasms/metabolism*
;
Neoplasm Invasiveness
;
Membrane Potential, Mitochondrial/drug effects*
;
Anthraquinones/pharmacology*
;
Cell Proliferation/drug effects*
;
Cell Death/drug effects*
;
Apoptosis/drug effects*
;
Cell Movement/drug effects*
;
Cell Survival/drug effects*

Result Analysis
Print
Save
E-mail