1.Emd-D inhibited ovarian cancer progression via PFKFB4-dependent glycolysis and apoptosis.
Xin ZHAO ; Chao CHEN ; Xuefei FENG ; Haoqi LEI ; Lingling QI ; Hongxia ZHANG ; Haiying XU ; Jufeng WAN ; Yan ZHANG ; Baofeng YANG
Chinese Journal of Natural Medicines (English Ed.) 2025;23(4):431-442
Ovarian cancer poses a significant threat to women's health, necessitating effective therapeutic strategies. Emd-D, an emodin derivative, demonstrates enhanced pharmaceutical properties and bioavailability. In this study, Cell Counting Kit 8 (CCK8) assays and Ki-67 staining revealed dose-dependent inhibition of cell proliferation by Emd-D. Migration and invasion experiments confirmed its inhibitory effects on OVHM cells, while flow cytometry analysis demonstrated Emd-D-induced apoptosis. Mechanistic investigations elucidated that Emd-D functions as an inhibitor by directly binding to the glycolysis-related enzyme PFKFB4. This was corroborated by alterations in intracellular lactate and pyruvate levels, as well as glucose transporter 1 (GLUT1) and hexokinase 2 (HK2) expression. PFKFB4 overexpression experiments further supported the dependence of Emd-D on PFKFB4-mediated glycolysis and SRC3/mTORC1 pathway-associated apoptosis. In vivo experiments exhibited reduced xenograft tumor sizes upon Emd-D treatment, accompanied by suppressed glycolysis and increased expression of Bax/Bcl-2 apoptotic proteins within the tumors. In conclusion, our findings demonstrate Emd-D's potential as an anti-ovarian cancer agent through inhibition of the PFKFB4-dependent glycolysis pathway and induction of apoptosis. These results provide a foundation for further exploration of Emd-D as a promising drug candidate for ovarian cancer treatment.
Female
;
Humans
;
Ovarian Neoplasms/physiopathology*
;
Phosphofructokinase-2/genetics*
;
Apoptosis/drug effects*
;
Glycolysis/drug effects*
;
Animals
;
Cell Line, Tumor
;
Mice
;
Cell Proliferation/drug effects*
;
Emodin/administration & dosage*
;
Mice, Nude
;
Mice, Inbred BALB C
;
Hexokinase/metabolism*
;
Xenograft Model Antitumor Assays
2.Astragalus polysaccharides induces ferroptosis in ovarian adenocarcinoma cells through Nrf2/SLC7A11/GPX4 signaling pathway.
Yong-Gen ZHANG ; Xiao-Fei YAN ; Feng LIU ; Wen-Zhe HAO ; Yue CAI ; Ying LIU ; Lan-Lin LIU ; Xue-Jun LI
China Journal of Chinese Materia Medica 2024;49(23):6459-6467
This study primarily investigated the mechanism of Astragalus polysaccharides(APS), a Chinese medicinal material, in regulating the Nrf2/SLC7A11/GPX4 signaling pathway to induce ferroptosis in ovarian cancer cells(Caov-3 and SKOV3 cells). Caov-3 and SKOV3 cells were divided into control(Vehicle) group, APS group, glutathione peroxidase 4 inhibitor(RSL3) group, and APS+RSL3 group. After 48 h of intervention, the activity and morphology of the cells in each group were observed. The cell counting kit-8(CCK-8) method was used to determine the half-maximal inhibitory concentration(IC_(50)), while colony formation and EdU assays were conducted to assess cell proliferation. Biochemical reagents were used to detect lipid reactive oxygen species(L-ROS), malondialdehyde(MDA), divalent iron ions(Fe~(2+)), and glutathione(GSH) in Caov-3 cells. Transmission electron microscopy was employed to observe the morphological changes of mitochondria in Caov-3 cells. Bioinformatics analysis were used to screen potential target genes of APS in ovarian cancer cells. Western blot and RT-PCR were applied to measure the protein and mRNA expression of Nrf2, SLC7A11, and GPX4. The results revealed that APS effectively inhibited the activity and proliferation of ovarian cancer cells, significantly increased the expression levels of L-ROS, MDA, and Fe~(2+)(P<0.001), and significantly reduced the expression level of GSH(P<0.001). Under electron microscopy, the mitochondria of Caov-3 cells appeared significantly smaller, with a marked increase in the density of the bilayer membrane, disappearance of mitochondrial cristae, and rupture of the outer mitochondrial membrane. These effects were more pronounced when APS was combined with RSL3. Bioinformatics screening identified Nrf2, SLC7A11, and GPX4 as potential target genes for APS in ovarian cancer cells. APS was shown to reduce the protein and mRNA expression of Nrf2, SLC7A11, and GPX4(P<0.01), with the APS+RSL3 showing even more significant effects(P<0.001). In conclusion, APS can induce ferroptosis in ovarian cancer cells, and its mechanism may be related to the regulation of the Nrf2/SLC7A11/GPX4 signaling pathway, providing an experimental basis for the use of APS injections in the treatment of ovarian cancer.
Humans
;
Ferroptosis/drug effects*
;
Female
;
NF-E2-Related Factor 2/genetics*
;
Ovarian Neoplasms/physiopathology*
;
Phospholipid Hydroperoxide Glutathione Peroxidase/genetics*
;
Signal Transduction/drug effects*
;
Cell Line, Tumor
;
Amino Acid Transport System y+/genetics*
;
Polysaccharides/pharmacology*
;
Astragalus Plant/chemistry*
;
Drugs, Chinese Herbal/pharmacology*
;
Adenocarcinoma/physiopathology*
;
Cell Proliferation/drug effects*
;
Glutathione Peroxidase/genetics*
;
Reactive Oxygen Species/metabolism*
3.Research advances on the role of exosomes in chemotherapy resistance of ovarian cancer.
Journal of Zhejiang University. Medical sciences 2019;48(1):116-120
Chemotherapy resistance is one of the biggest challenges in treatment of ovarian cancer. Mounting evidence shows that the exosomes shedding from tumor cells are considered to be involved in chemotherapy resistance of ovarian cancer by enhanced exosomal export of drugs, transferring RNAs or proteins and interfering with the bioactivity of therapeutic anti-tumor antibodies. In this review, we display the correlation between exosomes and chemotherapy resistance of ovarian cancer, the mechanism of exosomes involved in chemotherapy resistance of ovarian cancer, and discuss the potential clinical values of exosomes in chemotherapy resistance of ovarian cancer.
Antineoplastic Agents
;
therapeutic use
;
Carcinoma, Ovarian Epithelial
;
drug therapy
;
physiopathology
;
Drug Resistance, Neoplasm
;
Exosomes
;
metabolism
;
Female
;
Humans
;
Ovarian Neoplasms
;
drug therapy
;
physiopathology
4.Effects of methylseleninic acid on cisplatin-resistant ovarian cancer cells (SKOV3/DDP) and the mechanisms.
Ying TAN ; Qing FENG ; Xin SUN ; Min XUE ; Ning JIANG ; Xinliang DENG
Journal of Central South University(Medical Sciences) 2016;41(12):1305-1311
To investigate the reversal effect of methylseleninic acid on cisplatin (DDP)-resistant ovarian cancer cells and the underlying mechanisms.
Methods: SKOV3/DDP cells were incubated with cisplatin at different concentrations for 48 h, then the proliferation rate of SKOV3/DDP cells was detected by MTT assays, and the expression of β-catenin in SKOV3/DDP cells was examined by Western blot. The inhibitory effect of methyl-seleninic acid (MSA) combined with DDP at different concentrations on SKOV3/DDP cells was assayed by MTT method. Western blot was used to detect the expression of β-catenin protein in the cells.
Results: The inhibitory rate for proliferation in DDP-treated SKOV3/DDP cells with different concentrations is lower than that in the SKOV3 cells (P<0.05); β-catenin expression in SKOV3/DDP cells was significantly higher than that in the SKOV3 cells (P<0.05). The inhibitory rate for proliferation in SKOV3/DDP cells with different concentrations of MSA was increased with the increase in concentration (P<0.05). The inhibitory rate for proliferation in SKOV3/DDP cells with 2 or 6 μmol/L MSA plus cisplatin was lower than that in cisplatin alone group (P<0.05). β-catenin expression in SKOV3 /DDP cells with 2 or 6 μmol/L MSA plus cisplatin was higher than that in the cisplatin alone group (P<0.05).
Conclusion: MSA can reverse cisplatin resistance on SKOV3 / DDP cells, which may be related to the decrease in β-catenin expression.
Antineoplastic Agents
;
pharmacology
;
Carcinoma
;
physiopathology
;
Cell Line, Tumor
;
physiology
;
Cell Proliferation
;
drug effects
;
Cisplatin
;
pharmacology
;
Drug Resistance, Neoplasm
;
drug effects
;
genetics
;
Female
;
Humans
;
Organoselenium Compounds
;
pharmacology
;
Ovarian Neoplasms
;
physiopathology
;
beta Catenin
;
drug effects
;
metabolism
5.Effect of antibacterial peptide hCAP18/LL-37 on ovarian cancer microenvironment and the regulatory mechanism of its expression.
Qian LU ; Wenqiang QUAN ; Junlu WU ; Xian ZHANG ; Wei MA ; Li PANG ; Dong LI ; Email: 186LD@163.COM.
Chinese Journal of Oncology 2015;37(10):725-730
OBJECTIVETo investigate the effect of antibacterial peptide hCAP18/LL-37 on ovarian cancer microenvironment and the regulatory mechanism of its expression.
METHODSWe assessed the effect of macrophage-promoted ovarian cancer cells invasion using BioCoat Matrigel invasion chamber. The expressions of hCAP18/LL-37 and versican V1 were determined by real-time PCR and Western blot analysis. SKOV3 cells were transfected with shRNA plasmid to abrogate the expression of versican V1, and then the expression of hCAP18/LL-37 in macrophages and the invasiveness of SKOV3 cells were assayed.
RESULTSThe Matrigel invasion assay showed that after co-culture with macrophages for 4 days, the number of penetrated SKOV3 cells was 112.8±17.1/per high power field, significantly higher than that in the SKOV3 cells cultured alone (8.2±1.9/per high power field) (P<0.05). Addition of hCAP/LL-37 neutralizing antibody into the co-cultured macrophage-SKOV3 cells markedly inhibited the macrophage-promoted SKOV3 cells invasion. The penetrated SKOV3 cells was 22.2±5.6/per high power field, significantly lower than the 100.6±25.2/per high power field in the control macrophage- SKOV3 co-cultured cells (P<0.05). The expressions of hCAP18/LL-37 mRNA and protein in macrophages were remarkably enhanced upon co-culture with SKOV3 cells, but not changed in SKOV3 cells cultured alone. The expression and secretion of versican V1 in the ovarian cancer cells were also significantly increased after co-cultured with macrophages. Knockdown of versican V1 in SKOV3 cells by small interfering RNA significantly reduced the expression of hCAP18/LL-37 mRNA and protein in the macrophages, as well as decreased the invasiveness of SKOV3 cells (P<0.05).
CONCLUSIONSIn the cancer microenvironment, the macrophage-secreted hCAP18/LL-37 promote the invasiveness of ovarian cancer cells, and the hCAP18/LL-37 expression is regulated by versican V1 protein released by ovarian cancer cells.
Antimicrobial Cationic Peptides ; metabolism ; pharmacology ; Cell Line, Tumor ; Coculture Techniques ; Collagen ; Drug Combinations ; Female ; Humans ; Laminin ; Macrophages ; metabolism ; Neoplasm Invasiveness ; Neoplasm Proteins ; metabolism ; Ovarian Neoplasms ; metabolism ; pathology ; physiopathology ; Plasmids ; Proteoglycans ; RNA, Messenger ; metabolism ; RNA, Small Interfering ; Real-Time Polymerase Chain Reaction ; Transfection ; Tumor Microenvironment ; drug effects ; Versicans ; metabolism
6.Suppression of EphB4 improves the inhibitory effect of mTOR shRNA on the biological behaviors of ovarian cancer cells by down-regulating Akt phosphorylation.
Xiangyi MA ; Danfeng LUO ; Kezhen LI ; Ronghua LIU ; Yan LIU ; Tao ZHU ; Dongrui DENG ; Jianfeng ZHOU ; Li MENG ; Shixuan WANG ; Ding MA
Journal of Huazhong University of Science and Technology (Medical Sciences) 2012;32(3):358-363
The aim of the present study was to examine the effects of suppression of EphB4 and/or mTOR on the biological behaviors of ovarian cancer cells, and the potential regulatory pathways. Antisense EphB4 vectors and shRNA vectors targeting mammalian target of rapamycin (mTOR) were constructed and transfected into A2780 and SKOV3 cells (two ovarian cancer cell lines). The effects of the antisense EphB4 vectors and the shRNA vectors on the proliferation, apoptosis and invasion of ovarian cancer cells were measured, and the expression of EphB4, mTOR and Akt detected. The results showed that transfection with mTOR shRNA could inhibit growth, induce apoptosis, and reduce invasive ability of ovarian cancer cells, which was accompanied by downregulation of EphB4, mTOR and Akt. The inhibitory effects on cell growth caused by mTOR shRNA alone were weaker than those by antisense pEGFP-C1-EphB4. In the antisense pEGFP-C1-EphB4-transfected cells, it was found that EphB4 knockdown could decrease the mTOR expression and slightly reduce the Akt phosphorylation. Significant suppressive effects on cell growth were observed in cells co-transfected with antisense pEGFP-C1-EphB4 and mTOR shRNA. In co-transfection group, the expression levels of EphB4, mTOR and Akt were distinctly lower than those in other groups. It was concluded that suppression of EphB4 may inhibit the growth of ovarian cancer cells by downregulation of the PI3K/Akt/mTOR pathway, and reverse Akt phosphorylation induced by mTOR shRNA. Inhibition of EphB4 and mTOR combined may cooperatively suppress the biological behaviors of ovarian cancer cells.
Apoptosis
;
genetics
;
Cell Line, Tumor
;
Cell Proliferation
;
Down-Regulation
;
genetics
;
Female
;
Humans
;
Ovarian Neoplasms
;
pathology
;
physiopathology
;
Proto-Oncogene Proteins c-akt
;
genetics
;
metabolism
;
RNA, Small Interfering
;
genetics
;
Receptor, EphB4
;
genetics
;
metabolism
;
Suppression, Genetic
;
genetics
7.Surgical high ligation of the ovarian vein and preservation of ovarian function for twisted ovarian tumors.
Ju LI ; Yue-Xin YU ; Chun-Yan SUN ; Dan XUE ; Dong-Ying QU
Chinese Medical Journal 2012;125(20):3744-3746
BACKGROUNDAppendectomy is the traditional surgical procedure for correcting torsion of the adnexa. Although it prevents pulmonary embolism, ovarian necrosis, and secondary infection, it can have critical adverse effects on the ovarian function.
METHODSWe performed surgery for adnexal torsion in 12 patients, using high ligation of the ovarian vein, followed by removal of the ovarian tumor.
RESULTSBlood flow in the residual ovary gradually returned to normal within 1 - 3 months, and a dominant follicle could be seen in the residual ovary within 2 - 6 months post-surgery in all the 12 cases. Menstruation recovered in these three cases within 2 - 3 months. Postoperative intrauterine pregnancies occurred in two cases, with a corpus luteum graviditatis in the residual ovary in one case, while the other patient underwent labor after 13 months and a normal ovary on the affected side was seen at cesarean section.
CONCLUSIONSThis new surgical technique involving high ligation of the ovarian vein for adnexal torsion allowed successful preservation of the residual ovary and ovarian blood distribution, and can thus be used for the treatment of primary diseases of the ovary. The surgical procedure is simple, safe, and effective, and warrants extensive application in clinical practice.
Adolescent ; Adult ; Female ; Humans ; Ligation ; Ovarian Neoplasms ; physiopathology ; surgery ; Ovary ; blood supply ; physiopathology ; Torsion Abnormality ; surgery ; Ultrasonography, Doppler, Color
8.Effects of survivin antisense oligonucleotides on survivin and Smac gene expression in ovarian cancer cells.
Yan SUN ; Yong LI ; Li-Qiao FAN ; Xiao-Hua WU ; Jiang-Xin CHENG ; Qu ZHAO
Journal of Southern Medical University 2007;27(10):1571-1574
OBJECTIVETo observe the effect of survivin antisense oligonucleotides (ASODN) on survivin and Smac gene expression in ovarian cancer SKOV3 cells and explore the role of survivin and Smac genes in ASODN-induced ovarian cancer cell cycle alteration and apoptosis and its molecular mechanisms.
METHODSSurvivin ASODN was introduced via Lipofectamine(TM)2000 into SKOV3 cells, whose growth activity was detected subsequently with MTT assay. The apoptosis index, cell cycle and changes in survivn and Smac protein expression were determined using flow cytometry. The changes in survivn and Smac mRNA expression were detected by RT-PCR.
RESULTSIn comparison with the control cells, cells transfected with difference concentrations of survivin ASODN exhibited significantly inhibited growth, and 48 h after the transfection, the IC(50) was about 600 nmol/L. After a 48-hour transfection of the cells with 600 nmol/L ASODN, the apoptosis index significantly decreased (t=6.3671, P<0.05), and the cell percentage in (1)/G(0) phase increased (t=10.3832, P<0.01), resulting also in significantly down-regulated survivin mRNA and protein expressions (t=3.5031, P<0.05; t=7.8146, P<0.01) and up-regulated Smac mRNA and protein expressions (t= 2.8011, P<0.05; t= 11.3917, P<0.01).
CONCLUSIONASODN against survivin can induce apoptosis of ovarian cancer cell line SKOV3 and results cell growth arrest in G(1)/G(0) phase by up-regulating Smac and down-regulating survivin expression. Survivin and Smac are closely correlated in their action on SKOV3 cell cycle and apoptosis, which is one of the important mechanisms of ovarian cancer development.
Apoptosis ; Cell Line, Tumor ; Female ; Gene Expression Regulation, Neoplastic ; Humans ; Inhibitor of Apoptosis Proteins ; Intracellular Signaling Peptides and Proteins ; genetics ; metabolism ; Microtubule-Associated Proteins ; genetics ; metabolism ; Mitochondrial Proteins ; genetics ; metabolism ; Oligonucleotides, Antisense ; genetics ; metabolism ; Ovarian Neoplasms ; genetics ; metabolism ; physiopathology
9.Effect of chemotherapy to ovary function in ovarian malignancy patients undergoing conservative surgery.
Zheng-yi SUN ; Keng SHEN ; Jing-he LANG ; Hui-fang HUANG
Acta Academiae Medicinae Sinicae 2003;25(4):431-433
OBJECTIVETo determine the effect of chemotherapy on ovarian endocrine function and menstruation.
METHODSMenstruation and serum luteinizing hormone (LH), follicle-stimulating hormone (FSH), and estradiol (E2) levels were observed when the patients with ovarian cancer undergoing one side ovarectomy were followed by chemotherapy.
RESULTSAbnormal menstruation occurred in 86.7% cases, including 80% amenorrhea and 6.7% oligomenstruate. Menstruation resumed around 2 months after chemotherapy. Serum LH, FSH levels rose and E2 level declined during amenorrhea. No significant change was observed in progesterone or testosterone levels.
CONCLUSIONSOvary function impairment may occur in ovarian cancer patient treated by one side ovarectomy followed by chemotherapy. Serum LH, FSH, and E2 levels change and abnormal menstruation are two common manifestations. However, chemotherapy-related amenorrhea is reversible.
Adolescent ; Adult ; Amenorrhea ; etiology ; Child ; Estradiol ; blood ; Female ; Follicle Stimulating Hormone ; blood ; Humans ; Luteinizing Hormone ; blood ; Menstruation ; Ovarian Neoplasms ; drug therapy ; surgery ; Ovariectomy ; Ovary ; physiopathology

Result Analysis
Print
Save
E-mail