Open Access

Oleanolic acid inhibits osteosarcoma cell proliferation and invasion by suppressing the SOX9/Wnt1 signaling pathway

  • Authors:
    • Xianming Chen
    • Yulin Zhang
    • Sen Zhang
    • Aiming Wang
    • Quanyin Du
    • Ziming Wang
  • View Affiliations

  • Published online on: February 28, 2021     https://doi.org/10.3892/etm.2021.9883
  • Article Number: 443
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Osteosarcoma is the most common primary bone malignancy in children and adolescents. Inhibition of SOX9/Wnt1‑mediated signaling might suppress osteosarcoma metastasis, and oleanolic acid (OA) might decrease the activity of the SOX9/Wnt1 signaling pathway. The aim of the present study was to determine the role of OA in osteosarcoma cell proliferation and invasion. Osteosarcoma cell lines (KHOS and U2OS) and an osteoblastic cell line (hFOB1.19) were used for cell viability, proliferation and invasion analysis. The data suggested that OA significantly inhibited cell viability on days 3, 4 and 5 compared with the control (Ctrl) group in both U2OS and KHOS cells. Cell proliferation in the OA‑treated group was significantly decreased compared with the Ctrl group in the osteosarcoma cell lines. Analysis of the cell cycle indicated that OA significantly reduced the percentage of U2OS and KHOS cells in the S phase compared with the Ctrl group. The wound healing assay results indicated that the OA group displayed significantly decreased cell re‑colonization of the wound at 48 h compared with the Ctrl group. The Transwell chamber assay results also indicated that cell invasion was significantly inhibited by OA compared with the Ctrl group. Furthermore, OA significantly increased osteosarcoma cell apoptosis compared with the Ctrl group. Similarly, the protein expression levels of SOX9 and Wnt1 were significantly decreased in OA‑treated U2OS and KHOS cells compared with Ctrl cells. OA‑mediated downregulation of Wnt1 expression was reversed following SOX9 small interfering RNA transfection. Collectively, the results indicated that OA inhibited SOX9/Wnt1‑associated osteosarcoma cell proliferation, migration and invasion.

Introduction

Osteosarcoma is the most common primary bone malignancy in children and adolescents worldwide (1). The majority of patients with osteosarcoma display invasion and metastasis, leading to a reduction in the efficiency of anticancer agents and the failure of therapy (2). Therefore, identification of potential therapeutic targets to suppress osteosarcoma cell proliferation and invasion is required.

Various mechanisms underlying osteosarcoma cell proliferation and invasion have been previously described (1,2). The Wnt signaling pathway, which regulates cell proliferation, differentiation and migration, serves important functions in osteosarcoma cell proliferation and invasion (3,4). Previous studies have reported that Wnt1 expression is increased in osteosarcoma tissues (3,4). Activation of Wnt signaling activates genes associated with cell proliferation, including c-Myc, matrix metallopeptidases and cyclin D1 (3-5), leading to osteosarcoma cell proliferation and invasion. Moreover, it has been reported that SOX9 regulates hyperexpression of Wnt1 in osteosarcoma tissues and cells (3). Therefore, it was hypothesized that inhibition of SOX9/Wnt1-mediated signaling might suppress osteosarcoma metastasis.

Oleanolic acid (OA), a naturally occurring triterpenoid, displays potential antitumor activity in several tumor cells, and has also has been reported to inhibit osteosarcoma cell proliferation and induce cell apoptosis (6-8). However, the mechanisms underlying OA during osteosarcoma are not completely understood. Previous studies have demonstrated that OA inhibits the Wnt signaling pathway in hepatoma cells (9) and derivatives of OA decrease the expression levels of SOX2(10). The present study investigated the hypothesis that OA functions as an anticancer agent to protect against osteosarcoma.

Materials and methods

Osteosarcoma cell lines culture

Osteosarcoma cell lines (KHOS and U2OS) and an osteoblastic cell line (hFOB1.19) were purchased from American Type Culture Collection. Osteosarcoma cells were cultured in DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.), and osteoblastic cells were cultured in RPMI-1640 (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS at 37˚C with 5% CO2. Cells were incubated with 50 µM OA (cat. no. O5504; Sigma-Aldrich; Merck KGaA) for 24 h at 37˚C.

Cell viability analysis

The Cell Counting Kit-8 (CCK-8) assay (cat. no. C0038; Beyotime Institute of Biotechnology) was performed to evaluate cell viability according to the manufacturer's protocol. Briefly, cells (1x104 cells/well) were seeded into a 96-well plate and incubated with 10 µl CCK-8 reagent for 2 h at 37˚C. The absorbance of each well was measured at a wavelength of 450 nm using a microplate spectrophotometer.

Colony formation assay

Cell proliferation was assessed by conducting a colony formation assay. Cells were seeded (1x103 cells/well) into a 6-well plate and cultured at 37˚C with 5% CO2. Following culture for 21 days, cell colonies, which were defined as 0.5-1 mm in diameter cell assemblies, were fixed with 4% paraformaldehyde for 30 min at room temperature, stained using crystal violet for 10 min at room temperature and observed using a light microscope (magnification, x100).

Ki67 staining

Cell proliferation was also assessed by performing Ki67 staining. Briefly, cells were grown on coverslips and fixed with 4% paraformaldehyde for 30 min at room temperature. Subsequently, cells were incubated with an Alexa Fluor 647-conjugated anti-Ki67 antibody (1:100; cat. no. ab196907; Abcam) for 30 min at 37˚C and the nuclei were stained with DAPI for 1 min at room temperature. Ki67-positive cells were counted in ten randomly selected fields of view using a fluorescence microscope (magnification, x100) and NIS-Elements Viewer software (version 4.2.0; Nikon Corporation).

Flow cytometry

The cell cycle distribution was analyzed by performing by flow cytometry. Cells were harvested and washed with PBS. Subsequently, cells were fixed with ice-cold 70% ethanol for 4 h at 4˚C. After permeabilization by 0.1% Triton X-100 for 30 min at room temperature, the cells were stained with PI containing RNase A (cat. no. C1052; Beyotime Institute of Biotechnology) at 37˚C for 1 h. The cell cycle distribution was analyzed via flow cytometry using a BD flow cytometry system (FACSVerse; BD Biosciences) and FlowJo software (version 10.0; BD Biosciences).

Wound healing assay

The wound healing assay was performed to assess cell migration. Cells were cultured in a 6-well plate. At 90% confluence, the cell layer was scratched using a 200-µl sterile pipette tip. Subsequently, cells were cultured in DMEM or RPMI-1640 without FBS at 37˚C with 5% CO2. At 0 and 48 h, the wounds were observed using a phase contrast light microscope (magnification, x200). Taking the distance between the wound edges at 0 h as a baseline, the image of the same location after 48 h was captured. The percentage of the wound healing area was analyzed using ImageJ software (version 1.46r; National Institutes of Health).

Transwell assay

To assess cell invasion, cells (5x104) were seeded into the upper chambers of Transwell plates with 50 mg/l Matrigel (pore size, 8 µm; cat. no. CLS3374; Corning Inc.) and serum-free medium after Matrigel precoating at 37˚C for 1 h. DMEM or RPMI-1640 medium supplemented with 10% FBS was filled into the lower chambers. Following incubation for 8 h, cells on the upper surface of the membrane were removed and invading cells were fixed with 4% paraformaldehyde for 15 min at room temperature and stained with crystal violet for 30 min at room temperature. Invading cells were visualized using an inverted light microscope (magnification, x100).

TUNEL assay

To detect osteosarcoma cell apoptosis, a TUNEL assay (cat. no. C1088; Beyotime Institute of Biotechnology) was performed. Briefly, 104 cells/well were cultured on coverslips in DMEM or RPMI-1640 supplemented with 10% FBS at 37˚C with 5% CO2. Subsequently, cells were fixed with 4% paraformaldehyde for 30 min at room temperature. After rinsing with PBS, cells were stained using the TUNEL assay kit at 37˚C for 60 min and the nuclei were stained with DAPI (0.3 mM, Vector Laboratories, Inc.) at room temperature for 1 min. Following mounting with an anti-fluorescence quenching mounting medium (cat. no. P0126; Beyotime Institute of Biotechnology), the number of apoptotic nuclei per section was calculated using the following formula: The number of TUNEL-positive cell nuclei/the total number of DAPI-positive nuclei. The number of apoptotic nuclei were counted in ten randomly selected fields of view using a fluorescence microscope (magnification, x100).

Western blotting

After washing twice with ice-cold PBS, total protein was extracted from osteosarcoma cells using ice-cold lysis buffer (cat. no. P0013; Beyotime Institute of Biotechnology) and quantified using the bicinchoninic acid assay (cat. no. P0012; Beyotime Institute of Biotechnology). Protein homogenates (30 µg) were separated via 8-10% SDS-PAGE and transferred onto polyvinylidene fluoride membranes. The membranes were washed with TBS and blocked with 5% milk powder in TBS for 1 h at room temperature. Subsequently, the membranes were incubated at 4˚C overnight with the following primary antibodies: Rabbit anti-caspase-3 (1:1,000; cat. no. 9662; Cell Signaling Technology, Inc.), rabbit anti-cleaved- caspase-3 (1:1,000; cat. no. 9664; Cell Signaling Technology, Inc.), rabbit anti-Bcl2 (1:1,000; cat. no. 3498; Cell Signaling Technology, Inc.), rabbit anti-SOX9 (1:1,000; cat. no. 82630; Cell Signaling Technology, Inc.), rabbit anti-β-catenin (1:1,000; cat. no. 9582; Cell Signaling Technology, Inc.), rabbit anti-Wnt1 (1:1,000; cat. no. 2915; Cell Signaling Technology, Inc.) and anti-GAPDH (1:1,000; cat. no. sc-47724; Santa Cruz Biotechnology, Inc.). Following primary incubation, the membranes were washed with TBS and incubated with an HRP-conjugated goat anti-rabbit-IgG secondary antibody (1:4,000; cat. no. BA1039; Boster Biological Technology) for 1 h. Protein bands were visualized using enhanced chemiluminescence (EMD Millipore). Densitometry levels were analyzed using Quantity One software (version 4.6.6; Bio-Rad Laboratories, Inc.) with GAPDH as the loading control.

Small interfering (si)RNA) transfection

Cells in 6-well plates (2x105 cells/well) were transfected with 10 nM SOX9-specific siRNA (5'-GCGACGUCAUCUCCAACAU-3') or scramble siRNA (5'-UAGAGCUAGAGCAAGGGUA-3') (both GE Healthcare Dharmacon, Inc.) using 6 µl Oligofectamine in Opti-MEM medium (Invitrogen; Thermo Fisher Scientific, Inc.). At 24 h post-transfection, cells were cultured in DMEM supplemented with 10% FBS for 24 h at 37˚C with 5% CO2. Subsequently, transfection efficiency was determined via RT-qPCR and western blotting (Fig. S1).

Statistical analysis

Statistical analyses were performed using SPSS software (version 22.0; IBM Corp.). Comparisons among multiple groups were analyzed using one-way ANOVA followed by Holm-Sidak's post hoc test. Comparisons between two groups were analyzed using unpaired Student's t-test. Data are presented as the mean ± SEM. P<0.05 was considered to indicate a statistically significant difference.

Results

OA inhibits osteosarcoma cell proliferation and invasion

To investigate the protective effect of OA against osteosarcoma, the primary low-metastatic U2OS (11) and high-metastatic KHOS (12) cell lines were treated with OA. The CCK-8 assay was performed to assess cell viability, and the results indicated that OA significantly inhibited cell viability compared with the control (Ctrl) group on days 3, 4 and 5 in both U2OS and KHOS cells (Fig. 1A). Similar results were obtained in the colony formation assay (Fig. 1B). The Ki67 staining results indicated that cell proliferation in the OA-treated group was significantly lower compared with the Ctrl group in both osteosarcoma cell lines (Fig. 1C). Cell cycle analysis indicated that OA significantly reduced the percentage of U2OS and KHOS cells in the S phase compared with the Ctrl group (Fig. 1D).

Further studies investigated the effects of OA on osteosarcoma cell migration and invasion. The wound healing assay results indicated that the OA group displayed significantly decreased cell re-colonization of the wound after 48 h compared with the Ctrl group (Fig. 2A). Moreover, the Transwell assay results indicated that OA significantly inhibited cell invasion compared with the Ctrl group (Fig. 2B). An additional osteoblastic cell line, hFOB1.19, was used to assess the effect of OA on non-tumor cells. Compared with the Ctrl group, OA did not significantly alter hFOB1.19 cell viability, colony formation, proliferation, migration or invasion, as determined by performing CCK-8, colony formation, Ki67 staining, wound healing and Transwell assays, respectively (Fig. S2).

OA enhances osteosarcoma cell apoptosis

Previous studies have reported that OA increased cell apoptosis in other tumors (6,13); therefore, the effects of OA on osteosarcoma cell apoptosis were assessed. The TUNEL staining results suggested that OA significantly increased osteosarcoma cell apoptosis compared with the Ctrl group (Fig. 3A). The western blotting results also suggested OA-mediated induction of cell apoptosis. Caspase-3, a key enzyme involved in apoptosis (14), was significantly activated in OA-treated osteosarcoma cells compared with control osteosarcoma cells. In addition, OA treatment markedly reduced Bcl-2 expression compared with the Ctrl group (Fig. 3B and C).

OA inactivates the SOX9/Wnt1 signaling pathway in osteosarcoma cells

Subsequently, whether OA downregulated the SOX9/Wnt1 signaling pathway was investigated. The protein expression levels of SOX9, β-catenin and Wnt1 were significantly decreased in OA-treated U2OS and KHOS cells compared with Ctrl cells (Fig. 4A and B). Moreover, OA-mediated downregulation of Wnt1 expression was significantly reversed by transfection with SOX9 siRNA (Fig. 4C).

Discussion

The results of the present study were consistent with the hypothesis that OA, the 3β-hydroxy-olean-12-en-28-oic acid that is widely distributed in the plant kingdom as free acid or as aglycone of triterpenoid saponins (13), reduced osteosarcoma cell proliferation and invasion, and promoted osteosarcoma cell apoptosis compared with the Ctrl group. Although OA displays protective effects against inflammation and oxidative stress-induced cell apoptosis (15), OA also displays a wide range of anticancer pharmacological activities (16,17). Similar to the results of the present study, previous studies also demonstrated that OA inhibits cell proliferation and invasion, and induces cell apoptosis in other tumor cells, such as thyroid, prostate and breast cancer cells, as well as hepatocellular carcinoma and glioblastoma cells (13,18-20).

OA serves an antioncogenic role during malignant tumor development by inhibiting cancer cell proliferation, migration and invasion, and triggering cell death via apoptosis, autophagy or mitophagy (6,17,21). Multiple signaling pathways are associated with OA-mediated anticancer activities. Previous studies have indicated that OA induces cancer cell apoptosis by inhibiting the Akt-mTOR signaling cascade (22-24), as well as ERK, STAT3 and NF-κB signaling pathways (21).

Previous studies have also reported that OA induces osteosarcoma cell apoptosis and inhibits osteosarcoma cell proliferation; however, the underlying mechanism is not completely understood. Xu et al (6) reported that OA inhibits osteosarcoma cell proliferation and viability and interrupts the balance between proapoptotic and antiapoptotic factors by inhibiting the Notch signaling pathway. Moreover, the derivative of OA, N-formyl morpholine substituent of CDDO (CDDO-NFM), leads to degradation of c-Myc and decreases glucose uptake, lactate generation and adenosine triphosphate production to block glycolysis (7). In the present study, compared with the Ctrl group, OA inhibited osteosarcoma cell proliferation and invasion, and enhanced cell apoptosis by inactivating the SOX9/Wnt1 signaling pathway.

SOX9 is a member of the SOX family of transcription factors, which is closely associated with the development of a variety of malignant tumors (25). SOX9 enhances tumorigenesis by reactivating Wnt signaling (26,27). Moreover, the SOX9/Wnt1 signaling pathway also serves a key role in osteosarcoma (3). Suh et al (28) indicated that SOX9 regulates hyperexpression of Wnt1 in human osteosarcoma tissues and cells, and siRNA-mediated SOX9 knockdown inhibits human osteosarcoma cell proliferation by downregulating Wnt1 expression. Furthermore, the synthetic oleanane triterpenoids, CDDO-Imidazolide (CDDO-Im) and CDDO-Ethyl amide (CDDO-EA), upregulate SOX9 expression in normal cartilage and induce chondrogenic differentiation. OA displays a contrary effect in normal and tumor cells (3,28-30); however, the results of the present study indicated that the expression levels of SOX9 and Wnt1 were significantly decreased in OA-treated osteosarcoma cells compared with Ctrl cells, which suggested that OA inhibited SOX9/Wnt1-associated osteosarcoma cell proliferation, migration and invasion.

The mechanisms underlying OA-mediated regulation of SOX9 expression have not been previously reported. Previous studies have indicated that SOX9 activity is regulated via multiple layers, including posttranslational modifications, such as Small Ubiquitin-like Modifier (SUMO)ylation. SUMOylation represses the gene transcriptional activity of SOX9, leading to decreased SOX9 expression (31-33). Meanwhile, Momordin Ιc, an analog of OA, inhibits the activity of SUMO-specific protease 1, a member of the de-SUMOylation protease family (34), which might elevate the level of SOX9 SUMOylation and decease SOX9 expression. Future studies are required to identify the mechanisms underlying OA-mediated regulation of SOX9 expression. The current study has several limitations. Firstly, in vivo studies should be performed to identify the role of OA in tumor growth. Secondly, further research is required on the mechanisms that underlie the role of OA in the inhibition of SOX9 expression and the inactivation of the SOX9/Wnt1 signaling pathway.

In conclusion, the present study indicated that OA-mediated inactivation of the SOX9/Wnt1 signaling pathway may serve as a potential therapeutic strategy for osteosarcoma. The results of the present study indicated that OA significantly inhibited osteosarcoma cell proliferation, migration and invasion compared with the Ctrl group; therefore, OA may display potent antitumorigenic activities in osteosarcoma. For centuries, several medicinal plant extracts containing OA have been used in Asian countries for medical purposes (29) and have become registered drugs for the treatment of liver diseases in China (30). Chemically modified OA has been evaluated in several clinical trials for the treatment of various types of cancer such as breast, colorectal and lung cancer among others (8); however, further clinical trials are required to confirm the potential therapeutic application of OA.

Supplementary Material

Inhibitory effect of siSOX9. The transfection efficiency of siRNA was determined by performing (A) western blotting and (B) reverse transcription-quantitative PCR (n=4). *P<0.05 vs. NC. si, small interfering RNA; NC, negative control.
OA treatment does not inhibit non-tumor osteoblastic cell viability, proliferation, migration and invasion. (A) OA did not alter osteosarcoma cell viability, as determined by performing the Cell Counting Kit-8 assay (n=8). (B) OA did not alter osteosar-coma cell colony formation. Cell colonies were stained using crystal violet and observed under a microscope (magnification, x40; n=5). (C) Ki67 staining of osteosarcoma cells cultured with or without OA treatment (scale bar, 40 μm; n=5). Cell migration and invasion were assessed by performing (D) wound healing and (E) Transwell assays, respectively (magnification, x100; n=5). OA, oleanolic acid; Ctrl, control.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

XC and ZW contributed to the conception or design of the work and drafting the article or revising it for important intellectual content. YZ, SZ, AW and QD performed the experiments and contributed to the acquisition of data, analysis and interpretation of data and drafting and revising the article. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sadykova LR, Ntekim AI, Muyangwa-Semenova M, Rutland CS, Jeyapalan JN, Blatt N and Rizvanov AA: Epidemiology and risk factors of osteosarcoma. Cancer Invest. 38:259–269. 2020.PubMed/NCBI View Article : Google Scholar

2 

Morrow JJ, Bayles I, Funnell APW, Miller TE, Saiakhova A, Lizardo MM, Bartels CF, Kapteijn MY, Hung S, Mendoza A, et al: Positively selected enhancer elements endow osteosarcoma cells with metastatic competence. Nat Med. 24:176–185. 2018.PubMed/NCBI View Article : Google Scholar

3 

Liu H, Chen Y, Zhou F, Jie L, Pu L, Ju J, Li F, Dai Z, Wang X and Zhou S: Sox9 regulates hyperexpression of Wnt1 and Fzd1 in human osteosarcoma tissues and cells. Int J Clin Exp Pathol. 7:4795–4805. 2014.PubMed/NCBI

4 

Wang S, Zhang D, Han S, Gao P, Liu C, Li J and Pan X: Fibulin-3 promotes osteosarcoma invasion and metastasis by inducing epithelial to mesenchymal transition and activating the Wnt/β-catenin signaling pathway. Sci Rep. 7(6215)2017.PubMed/NCBI View Article : Google Scholar

5 

Zou J, Zhang W and Li XL: Effects of SOST gene silencing on proliferation, apoptosis, invasion, and migration of human osteosarcoma cells through the wnt/β-catenin signaling pathway. Calcif Tissue Int. 100:551–564. 2017.PubMed/NCBI View Article : Google Scholar

6 

Xu Y, Shu B, Tian Y, Wang G, Wang Y, Wang J and Dong Y: Oleanolic acid induces osteosarcoma cell apoptosis by inhibition of Notch signaling. Mol Carcinog. 57:896–902. 2018.PubMed/NCBI View Article : Google Scholar

7 

Gao F, Zuo Q, Jiang T, Song H and Zhou J: A newly synthesized oleanolic acid derivative inhibits the growth of osteosarcoma cells in vitro and in vivo by decreasing c-MYC-dependent glycolysis. J Cell Biochem. 120:9264–9276. 2019.PubMed/NCBI View Article : Google Scholar

8 

Shanmugam MK, Dai X, Kumar AP, Tan BK, Sethi G and Bishayee A: Oleanolic acid and its synthetic derivatives for the prevention and therapy of cancer: Preclinical and clinical evidence. Cancer Lett. 346:206–216. 2014.PubMed/NCBI View Article : Google Scholar

9 

Fan X, Wang P, Sun Y, Jiang J, Du H, Wang Z, Duan Z, Lei H and Li H: Oleanolic acid derivatives inhibit the Wnt/β-catenin signaling pathway by promoting the phosphorylation of beta-catenin in human SMMC-7721 cells. Pharmazie. 71:398–401. 2016.PubMed/NCBI View Article : Google Scholar

10 

Wang YY, Yang YX, Zhao R, Pan ST, Zhe H, He ZX, Duan W, Zhang X, Yang T, Qiu JX and Zhou SF: Bardoxolone methyl induces apoptosis and autophagy and inhibits epithelial-to-mesenchymal transition and stemness in esophageal squamous cancer cells. Drug Des Devel Ther. 9:993–1026. 2015.PubMed/NCBI View Article : Google Scholar

11 

Landers JE, Cassel SL and George DL: Translational enhancement of mdm2 oncogene expression in human tumor cells containing a stabilized wild-type p53 protein. Cancer Res. 57:3562–3568. 1997.PubMed/NCBI

12 

Rhim JS, Cho HY, Vernon ML, Arnstein P, Huebner RJ and Gilden RV: Characterization of non-producer human cells induced by Kirsten sarcoma virus. Int J Cancer. 16:840–849. 1975.PubMed/NCBI View Article : Google Scholar

13 

Duan L, Yang Z, Jiang X, Zhang J and Guo X: Oleanolic acid inhibits cell proliferation migration and invasion and induces SW579 thyroid cancer cell line apoptosis by targeting forkhead transcription factor A. Anticancer Drugs. 30:812–820. 2019.PubMed/NCBI View Article : Google Scholar

14 

Hotchkiss RS, Strasser A, McDunn JE and Swanson PE: Cell death. N Engl J Med. 361:1570–1583. 2009.PubMed/NCBI View Article : Google Scholar

15 

Zhang SL, Yang ZN, He C, Liao HB, Wang HS, Chen ZF and Liang D: Oleanane-type triterpenoid saponins from lysimachia fortunei maxim. Phytochemistry. 147:140–146. 2018.PubMed/NCBI View Article : Google Scholar

16 

Peng XP, Li XH, Li Y, Huang XT and Luo ZQ: The protective effect of oleanolic acid on NMDA-induced MLE-12 cells apoptosis and lung injury in mice by activating SIRT1 and reducing NF-kB acetylation. Int Immunopharmacol. 70:520–529. 2019.PubMed/NCBI View Article : Google Scholar

17 

Žiberna L, Šamec D, Mocan A, Nabavi SF, Bishayee A, Farooqi AA, Sureda A and Nabavi SM: Oleanolic acid alters multiple cell signaling pathways: Implication in cancer prevention and therapy. Int J Mol Sci. 18(643)2017.PubMed/NCBI View Article : Google Scholar

18 

Zhu YY, Huang HY and Wu YL: Anticancer and apoptotic activities of oleanolic acid are mediated through cell cycle arrest and disruption of mitochondrial membrane potential in HepG2 human hepatocellular carcinoma cells. Mol Med Rep. 12:5012–5018. 2015.PubMed/NCBI View Article : Google Scholar

19 

Liese J, Hinrichs TM, Lange M and Fulda S: Cotreatment with sorafenib and oleanolic acid induces reactive oxygen species-dependent and mitochondrial-mediated apoptotic cell death in hepatocellular carcinoma cells. Anticancer Drugs. 30:209–217. 2019.PubMed/NCBI View Article : Google Scholar

20 

Kim GJ, Jo HJ, Lee KJ, Choi JW and An JH: Oleanolic acid induces p53-dependent apoptosis via the ERK/JNK/AKT pathway in cancer cell lines in prostatic cancer xenografts in mice. Oncotarget. 9:26370–26386. 2018.PubMed/NCBI View Article : Google Scholar

21 

Zhu B, Ren C, Du K, Zhu H, Ai Y, Kang F, Luo Y, Liu W, Wang L, Xu Y, et al: Olean-28,13b-olide 2 plays a role in cisplatin-mediated apoptosis and reverses cisplatin resistance in human lung cancer through multiple signaling pathways. Biochem Pharmacol. 170(113642)2019.PubMed/NCBI View Article : Google Scholar

22 

Khan MW, Zhao P, Khan A, Raza F, Raza SM, Sarfraz M, Chen Y, Li M, Yang T, Ma X and Xiang G: Synergism of cisplatin-oleanolic acid co-loaded calcium carbonate nanoparticles on hepatocellular carcinoma cells for enhanced apoptosis and reduced hepatotoxicity. Int J Nanomedicine. 14:3753–3771. 2019.PubMed/NCBI View Article : Google Scholar

23 

Shi Y, Song Q, Hu D, Zhuang X, Yu S and Teng D: Oleanolic acid induced autophagic cell death in hepatocellular carcinoma cells via PI3K/Akt/mTOR and ROS-dependent pathway. Korean J Physiol Pharmacol. 20:237–243. 2016.PubMed/NCBI View Article : Google Scholar

24 

Nie H, Wang Y, Qin Y and Gong XG: Oleanolic acid induces autophagic death in human gastric cancer cells in vitro and in vivo. Cell Biol Int. 40:770–778. 2016.PubMed/NCBI View Article : Google Scholar

25 

Khurana N and Sikka SC: Interplay between SOX9, Wnt/β-catenin and androgen receptor signaling in castration-resistant prostate cancer. Int J Mol Sci. 20(2066)2019.PubMed/NCBI View Article : Google Scholar

26 

Yu Y, Yin W, Yu ZH, Zhou YJ, Chi JR, Ge J and Cao XC: miR-190 enhances endocrine therapy sensitivity by regulating SOX9 expression in breast cancer. J Exp Clin Cancer Res. 38(22)2019.PubMed/NCBI View Article : Google Scholar

27 

Domenici G, Aurrekoetxea-Rodriguez I, Simoes BM, Rábano M, Lee SY, Millán JS, Comaills V, Oliemuller E, López-Ruiz JA, Zabalza I, et al: A Sox2-Sox9 signalling axis maintains human breast luminal progenitor and breast cancer stem cells. Oncogene. 38:3151–3169. 2019.PubMed/NCBI View Article : Google Scholar

28 

Suh N, Paul S, Lee HJ, Yoon T, Shah N, Son AI, Reddi AH, Medici D and Sporn MB: Synthetic triterpenoids, CDDO-Imidazolide and CDDO-Ethyl amide, induce chondrogenesis. Osteoarthritis Cartilage. 20:446–450. 2012.PubMed/NCBI View Article : Google Scholar

29 

Borella R, Forti L, Gibellini L, De Gaetano A, De Biasi S, Nasi M, Cossarizza A and Pinti M: Synthesis and anticancer activity of CDDO and CDDO-Me, two derivatives of natural triterpenoids. Molecules. 24(4097)2019.PubMed/NCBI View Article : Google Scholar

30 

Lin C, Wen X and Sun H: Oleanolic acid derivatives for pharmaceutical use: A patent review. Expert Opin Ther Pat. 26:643–655. 2016.PubMed/NCBI View Article : Google Scholar

31 

Oh HJ, Kido T and Lau YF: PIAS1 interacts with and represses SOX9 transactivation activity. Mol Reprod Dev. 74:1446–1455. 2007.PubMed/NCBI View Article : Google Scholar

32 

Hattori T, Eberspaecher H, Lu J, Zhang R, Nishida T, Kahyo T, Yasuda H and de Crombrugghe B: Interactions between PIAS proteins and SOX9 result in an increase in the cellular concentrations of SOX9. J Biol Chem. 281:14417–14428. 2006.PubMed/NCBI View Article : Google Scholar

33 

Saotome H, Ito A, Kubo A and Inui M: Generation of a quantitative luciferase reporter for Sox9 SUMOylation. Int J Mol Sci. 21(1274)2020.PubMed/NCBI View Article : Google Scholar

34 

Wu J, Lei H, Zhang J, Chen X, Tang C, Wang W, Xu H, Xiao W, Gu W and Wu Y: Momordin Ic, a new natural SENP1 inhibitor, inhibits prostate cancer cell proliferation. Oncotarget. 7:58995–59005. 2016.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

May-2021
Volume 21 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen X, Zhang Y, Zhang S, Wang A, Du Q and Wang Z: Oleanolic acid inhibits osteosarcoma cell proliferation and invasion by suppressing the SOX9/Wnt1 signaling pathway. Exp Ther Med 21: 443, 2021
APA
Chen, X., Zhang, Y., Zhang, S., Wang, A., Du, Q., & Wang, Z. (2021). Oleanolic acid inhibits osteosarcoma cell proliferation and invasion by suppressing the SOX9/Wnt1 signaling pathway. Experimental and Therapeutic Medicine, 21, 443. https://doi.org/10.3892/etm.2021.9883
MLA
Chen, X., Zhang, Y., Zhang, S., Wang, A., Du, Q., Wang, Z."Oleanolic acid inhibits osteosarcoma cell proliferation and invasion by suppressing the SOX9/Wnt1 signaling pathway". Experimental and Therapeutic Medicine 21.5 (2021): 443.
Chicago
Chen, X., Zhang, Y., Zhang, S., Wang, A., Du, Q., Wang, Z."Oleanolic acid inhibits osteosarcoma cell proliferation and invasion by suppressing the SOX9/Wnt1 signaling pathway". Experimental and Therapeutic Medicine 21, no. 5 (2021): 443. https://doi.org/10.3892/etm.2021.9883