Open Access

Oxyresveratrol induces apoptosis and inhibits cell viability via inhibition of the STAT3 signaling pathway in Saos‑2 cells

  • Authors:
    • Tao Lv
    • Zhen Jian
    • Dejian Li
    • Rongguang Ao
    • Xu Zhang
    • Baoqing Yu
  • View Affiliations

  • Published online on: October 14, 2020     https://doi.org/10.3892/mmr.2020.11591
  • Pages: 5191-5198
  • Copyright: © Lv 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

Oxyresveratrol (ORES) is a natural phenolic compound with multiple biological functions including antioxidation, anti‑inflammation and neuroprotection; however, the inhibitory effect of ORES on osteosarcoma remains largely unknown. The present study aimed to determine the effects of ORES on osteosarcoma cell Saos‑2. Cell Counting Kit‑8 assay was performed to detect Soas‑2 cell viability. Annexin‑FITC/PI staining and JC‑1 staining were used to measure cell apoptosis and the change of mitochondrial membrane potential. In addition, western blotting was conducted to determine the expression levels of apoptotic proteins and the phosphorylation of STAT3. It was found that ORES inhibited cell viability and induced apoptosis of osteosarcoma Saos‑2 cells in a concentration‑dependent manner. In addition, ORES increased the expression levels of apoptotic proteases caspase‑9 and caspase‑3 and reduced mitochondrial membrane potential. In response to ORES treatment, the expression levels of pro‑apoptotic proteins, Bad and Bax, were enhanced, whereas those of anti‑apoptotic proteins, Bcl‑2 and Bcl‑xL, were reduced. In addition, the phosphorylation of STAT3 was attenuated in Saos‑2 cells after treatment with ORES. Inhibition of cell viability and apoptosis induction by ORES were rescued by enhancement of STAT3 activation upon treatment with IL‑6. Collectively, the present study indicated that ORES induced apoptosis and inhibited cell viability, which may be associated with the inhibition of STAT3 activation; thus, ORES represents a promising agent for treating osteosarcoma.

Introduction

Osteosarcoma is the most common primary malignant bone tumor of mesenchymal origin that occurs primarily in children and adolescents (1). It is characterized by the production of calcified osteoid extracellular matrix and a high frequency of inducing lung metastases, which directly results in a high mortality rate (2). The 5-year survival rate of patients with local osteosarcoma ranges between 65 and 75%, whereas that of patients with metastatic osteosarcoma is <30% (3). Surgical resection, chemotherapy and radical resection are established strategies for treating patients with osteosarcoma (4,5). Immunotherapy or a combination of radiotherapy and immunotherapy using nanomaterial delivery systems have also been a research hotspot in the field (68). However, the 5-year survival rate of patients with osteosarcoma demonstrates little improvement after treatment with chemotherapy drugs and traditional amputation (4). Therefore, it is important to identify new effective agents for treating patients with osteosarcoma.

Certain malignant tumors present an anti-apoptotic phenotype, which allows cancer cells to survive in local tissues (9). Most of the current anticancer medicines target apoptotic signaling pathways (10). Activation of caspases can be initiated via both the extrinsic and the mitochondria-mediated apoptotic signaling pathways (1012). The extrinsic pathway is triggered by activation of caspase-8 at the plasma membrane upon ligation of the death receptor, and subsequent cleavage of downstream effector caspases, such as caspase-3 (10). The mitochondria-mediated signaling pathway relies on interactions among Bcl-2 family proteins at the mitochondrial outer membrane and the release of cytochrome c (11). Among Bcl-2 family proteins, the activators directly bind both anti-apoptotic proteins and pro-apoptotic effector proteins, while sensitizers such as Bad bind only anti-apoptotic proteins (11,12). By competing for the BH3 binding site, sensitizers displace the binding of activators to anti-apoptotic proteins, including Bcl-2 and Bcl-xL (11). By interacting with the activators, pro-apoptotic effector proteins such as Bax and Bak create openings in the outer mitochondrial membrane and release cytochrome c, which promotes caspase-9 activation, followed by a cascade of apoptotic proteases, including caspase-3, resulting in cell death (12). The increased permeability of the mitochondrial outer membrane also results in mitochondrial depolarization with reduced mitochondrial membrane potential (MMP) and the release of apoptotic factors, triggering the downstream cell death processes (1315). Venetoclax, a direct small molecular inhibitor of Bcl-2, was initially approved by the Food and Drug Administration (FDA) in April 2016 and exhibited impressive outcomes particularly in chronic lymphocytic leukemia, small lymphocytic lymphoma and acute myeloid leukemia (16).

STAT3-mediated signaling pathways have an oncogenic potential in cancer cell proliferation and metastasis (17). The expression levels of STAT3 have been reported to be significantly higher in osteosarcoma tissue compared with those in normal bone or cartilage tissue, and high levels of STAT3 are associated with poor tumor differentiation and metastasis (18). Five-year overall survival and relapse-free survival rates in patients with high STAT3 expression levels are lower compared with in those with low STAT3 expression levels (18). Inhibition of STAT3 has also been reported to suppress osteosarcoma cell growth and induce apoptosis (19). Therefore, STAT3 may have an important therapeutic and prognostic value in osteosarcoma. In addition, STAT3 is directly involved in transcriptional regulation of osteopontin (OPN) (20), and increased OPN expression levels serve a critical role in osteosarcoma, which is a potential biomarker and promising drug target for osteosarcoma (21).

As a natural ingredient, which is primarily found in Morus alba L., oxyresveratrol (ORES) has extensive biological effects. Over the previous two decades, ORES has been reported as a powerful tyrosinase activity inhibitor (22,23), and also as having antioxidative (24,25), anti-inflammatory (26,27), anticancer (2830) and anti-lipogenesis properties (31). ORES has also been observed to exert strong neuroprotective effects, as it reduces neuronal oxidative damage (32,33). Notably, ORES and its derivatives have been reported to serve an efficient role against various types of cancer, such as head and neck carcinoma (28), neuroblastoma (29), prostate (30), kidney (34) and lung cancer (35). However, it remains unknown whether ORES has an effect on the inhibition of osteosarcoma cells and the mechanism by which ORES inhibits tumor cell viability.

In the present study, the inhibitory effect of ORES on Saos-2 osteosarcoma cells was determined, which indicates the ORES is a promising agent for treating osteosarcoma.

Materials and methods

Compound and reagents

ORES (2,3′,4,5′-Tetrahydroxy-trans-stilbene, C14H12O4; molecular weight: 244.24; purity ≥97.0%; cat. no. 29700-22-9) was purchased from Sigma-Aldrich (Merck KGaA). DMSO was used as control. DMEM, penicillin and streptomycin solution (100 IU/ml; 100 µg/ml), PBS, 0.25% trypsin-EDTA and enhanced chemiluminescent (ECL) substrate were all provided by Thermo Fisher Scientific, Inc. Fetal bovine serum (FBS) was obtained from Hangzhou Sijiqing Biological Engineering Materials Co., Ltd. Cell Counting Kit (CCK)-8 assay kit, MMP assay kit with JC-1, bicinchoninic acid (BCA) protein assay kit and RIPA lysis buffer (cat. no. P0013B) were acquired from Beyotime Institute of Biotechnology. Annexin V-FITC/propidium iodide (PI) apoptosis detection kit was purchased from MultiSciences (Lianke) Biotech Co., Ltd. Tris, non-fat milk and Tween-20 were purchased from Sangon Biotech Co., Ltd. IL-6 was purchased from PeproTech, Inc. Primary antibodies against cleaved caspase-9 (cat. no. 20750), cleaved caspase-3 (cat. no. 9664), GAPDH (cat. no. 5174), Bcl-2 (cat. no. 4223), Bcl-xL (cat. no. 2764), Bad (cat. no. 9239), Bax (cat. no. 5023), phophorylated-STAT3 (P-STAT3; cat. no. 9145) and total-STAT3 (T-STAT3; cat. no. 12640) were obtained from Cell Signaling Technologies, Inc. An antibody against OPN (cat. no. 7C5H12) was obtained from Thermo Fisher Scientific, Inc. Horseradish peroxidase (HRP)-conjugated anti-rabbit antibody (cat. no. 111-035-003) and HRP-conjugated anti-mouse antibody (cat. no. 115-035-003) were supplied by Jackson ImmunoResearch Laboratories, Inc. Methanol and ethanol were obtained from Sinopharm Chemical Reagent Co., Ltd.

Cell culture and cell viability assay

Saos-2 cells were obtained from the American Type Culture Collection. Cells were grown in DMEM containing 10% FBS and 1% penicillin and streptomycin solution with 5% CO2 at 37°C. Cell passage was performed with 0.25% trypsin-EDTA.

Cell viability was detected using the CCK-8 assay kit according to the manufacturer's instructions. Briefly, cells were seeded in 12-well plates at a density of 4×105 cells/well. After attachment, the cells were incubated with ORES at 0, 5, 15 and 45 µM at 37°C for 48 h. The CCK-8 solution (10 µl) was added to each well and after 1.5 h incubation, the viability of Saos-2 cells was detected using a microplate reader at 450 nm. In certain experiments, the Saos-2 cells were incubated with 30 µM ORES or 30 µM ORES and 20 ng/ml IL-6 at 37°C for 24 h.

Apoptosis assay

Early and late apoptosis was detected using the Annexin V-FITC/PI apoptosis detection kit according to the manufacturer's instructions. Cells were seeded in 12-well plates at a density of 4×105 cells/well. After attachment, the cells were incubated with ORES at 0, 5, 15 and 45 µM at 37°C for 24 h. The cells were then washed twice with ice-cold PBS. After digestion with 0.25% trypsin-EDTA, cells were collected and washed with PBS. Cells were resuspended in 400 µl Annexin-binding buffer, and then incubated with 5 µl Annexin V-FITC and 10 µl PI at room temperature for 5 min. Apoptosis of Saos-2 cells was detected with a LSRFortessa TM X-20 flow cytometer (BD Biosciences). Given that ORES significantly inhibited Saos-2 cell viability at the concentration between 15 and 45 µM, the Saos-2 cells was incubated with 30 µM ORES or 30 µM ORES and 20 ng/ml IL-6 at 37°C for 24 h. FlowJo 7.6 software (Tree Star, Inc.) was used for data processing.

MMP measurement

Changes in MMP were detected using the MMP assay kit with JC-1 staining, according to the manufacturer's instructions. Cells were seeded in 12-well plates at a density of 4×105 cells/well. After attachment, cells were incubated with ORES at 0, 5, 15 and 45 µM at 37°C for 24 h. The cells were then harvested and washed with PBS. After incubation with JC-1 staining solution for 20 min at 37°C in the dark, cells were washed twice with JC-1 buffer. Finally, cells were resuspended in PBS and detected using a LSRFortessa TM X-20 flow cytometer (BD Biosciences). FlowJo 7.6 software (FlowJo LLC) was used for data processing. The polymer/monomer (red/green) fluorescence intensity ratio was used to quantify the MMP.

Western blotting

Saos-2 cells were seeded in 6-well plates at 1×106 cells/well overnight at 37°C, and were then incubated with ORES at 0, 5, 15 and 45 µM at 37°C for 24 h. In some experiments, the Saos-2 cells were incubated with 30 µM ORES or 30 µM ORES and 20 ng/ml IL-6 at 37°C for 24 h. After washing with cold phosphate-buffered saline (PBS) three times, cells were lysed in RIPA lysis buffer and the protein concentration was detected using a BCA protein assay kit, according to the manufacturer's instructions. Each sample was adjusted to the same concentration with lysis buffer and 20 µg protein was loaded per lane. Proteins were then separated by SDS-PAGE on 10% gels and were transferred to 0.45 µm nitrocellulose filter membranes. The membranes were washed and blocked in blocking buffer (5% non-fat milk) for 1 h at room temperature. Subsequently, the membranes were incubated with primary antibodies against cleaved caspase-9, cleaved caspase-3, GAPDH, Bcl-2, Bcl-xL, Bad, Bax, P-STAT3, T-STAT3 and OPN (diluted at 1:1,000) with gentle agitation overnight at 4°C. After three washes, the membrane was incubated with HRP-conjugated anti-rabbit secondary antibody (diluted at 1:3,000) with gentle agitation for 2 h at room temperature. Finally, the ECL reagent was used to detect the target proteins. Semi-quantification of the bands was performed using ImageJ software (1.48v; National Institutes of Health).

Statistical analysis

The experiments were repeated at least three times independently, and data were presented as the mean ± SEM. One-way ANOVA followed by Tukey's post hoc test was applied to compare the differences between groups, using GraphPad Prism 5 (GraphPad Software, Inc.). *P<0.05 was considered to indicate a statistically significant difference.

Results

ORES inhibits the viability of Saos-2 cells

Although ORES has been shown to inhibit prostate and colon cancer cell viability (30), it remains unknown whether ORES can inhibit osteosarcoma cell viability. As a natural phenolic compound, ORES is a resveratrol analogue that contains an additional hydroxyl group in the aromatic ring (22). The chemical structure of ORES is shown in Fig. 1A. To determine the effect of ORES on the proliferation of osteosarcoma cells, the cell viability following ORES treatment in the Saos-2 cell line was detected. Cells were incubated with ORES at 0, 5, 15 and 45 µM for 48 h, and cell viability was detected using the CCK-8 assay. The results demonstrated that ORES significantly reduced cell viability in a concentration-dependent manner (Fig. 1B). These results indicated that ORES inhibits osteosarcoma cell viability.

ORES induces apoptosis of Saos-2 cells

Although ORES has been reported to induce apoptosis of neuroblastoma cells (29), the effect on osteosarcoma cells remains unclear. Following incubation with ORES at 0, 5, 15 and 45 µM for 24 h, apoptotic cells were detected using Annexin-FITC/PI staining. The results demonstrated that treatment with ORES significantly increased the percentage of apoptotic cells in a concentration-dependent manner compared with that of the untreated group (Fig. 1C and D).

A decrease in MMP is a marker of early apoptosis. In the mitochondria of normal cells, JC-1 is present as a polymer with bright red fluorescence. Once the mitochondria-mediated apoptotic pathway is initiated, JC-1 cannot aggregate and the JC-1 monomer generates green fluorescence, according to the manufacturer's instructions. The relative ratio of polymer/monomer (red/green) fluorescence was used to measure the mitochondrial depolarization. In response to ORES treatment at 0, 5, 15 and 45 µM for 24 h, cells were clearly divided into two groups, with polymer at the top and monomer at the bottom (Fig. 2A). The ratio of JC-1 polymer/monomer significantly decreased in a concentration-dependent manner in response to ORES compared with that of the untreated group (Fig. 2B).

In the mitochondria-mediated pathway, cytochrome c is released from mitochondria and induces the cleavage of caspase-3 and caspase-9, which ultimately triggers apoptosis via proteolysis of several cytoskeletal proteins, as well as the cleavage of DNA via caspase-activated DNase (12). The results of the present study demonstrated that compared with those of the untreated group, the expression levels of cleaved caspase-9 and cleaved caspase-3 were significantly increased by ORES treatment in a concentration-dependent manner (Fig. 3A-C). These results suggested that ORES induces osteosarcoma apoptosis.

ORES modulates apoptotic signaling pathways in Saos-2 cells

Given that ORES was demonstrated to promote apoptosis, the mechanism by which ORES regulates the apoptotic signaling pathways was further investigated. Western blotting was used to detect the expression levels of Bcl-2 family proteins, including Bcl-2, Bcl-xL, Bad and Bax (Fig. 4A). Mitochondria-mediated apoptosis is regulated by Bcl-2 family proteins (10). Pro-apoptotic sensitizer protein Bad can bind anti-apoptotic proteins Bcl-2 and Bcl-xL, which cause pro-apoptotic activator proteins to dissociate from these anti-apoptotic proteins and then bind to pro-apoptotic effector proteins, such as Bax, to initiate apoptosis (11). Compared with those of the untreated group, following treatment with ORES the expression levels of anti-apoptotic proteins Bcl-2 (Fig. 4B) and Bcl-xL (Fig. 4C) were significantly decreased, whereas the expression levels of pro-apoptotic proteins Bad (Fig. 4D) and Bax (Fig. 4E) were significantly increased. These results suggested that ORES may regulate the mitochondria-mediated apoptotic signaling pathway in Saos-2 cells.

ORES induces apoptosis and inhibits cell viability via STAT3 phosphorylation in Saos-2 cells

Given that STAT3 serves an important role in the proliferation and apoptosis of osteosarcoma cells (17), STAT3 activation in Saos-2 cells upon treatment with ORES was further examined. Compared with those of the untreated group, after treatment with ORES at 5, 15 and 45 µM for 24 h, the expression levels of P-STAT3 were significantly reduced in a concentration-dependent manner (Fig. 5A and B), whereas no significant differences were observed in the expression levels of T-STAT3 (Fig. 5A and C). OPN is regulated by STAT3 signaling, which is involved in osteosarcoma pathogenesis (21). Compared with those of the untreated group, treatment with ORES significantly decreased the expression levels of OPN (Fig. 5A and D). To determine whether ORES inhibits Saos-2 cell viability via a reduction in STAT3 activation, IL-6 was used to activate STAT3 signaling (36) in Saos-2 cells and reverse the effects of ORES. Upon treatment with 30 µM ORES for 24 h, the phosphorylation of STAT3 was induced by co-stimulation with 20 ng/ml IL-6 (Fig. 6A and B). After incubation with 20 ng/ml IL-6, the effects of ORES on the percentage of apoptotic cells (Fig. 6C and D) and cell viability (Fig. 6E) were reversed. These results indicated that ORES induces apoptosis and inhibits cell viability via partially reducing STAT3 signaling in Saos-2 cells.

Discussion

The results of the present study demonstrated that ORES effectively inhibited cell viability and induced apoptosis of Saos-2 cells. Upon treatment with ORES, the expression levels of cleaved caspase-9 and cleaved caspase-3 were significantly increased. Additionally, the expression levels of anti-apoptotic proteins Bcl-2 and Bcl-xL were decreased, whereas the expression levels of the pro-apoptotic proteins Bad and Bax were upregulated. These results indicated that ORES has specific antitumor activity against Saos-2 cells.

ORES is a natural compound derived from Morus alba L., and its roles have been revealed in various disease models. In macrophages, ORES has been proven to suppress LPS-stimulated inflammatory responses by blocking the MAPK signaling pathway (26). In addition to its anti-inflammatory effects, ORES significantly inhibited the proliferation of hepatocellular carcinoma (HCC) cells (37), and head and neck squamous cell carcinoma cells (28). In both cases, the expression levels of vascular endothelial growth factor (VEGF) were inhibited by ORES treatment, which indicates that ORES may reduce micro-blood vessel density and micro-lymphatic vessel density in tumors. As VEGF expression is regulated directly by STAT3 in diverse human cancer cell lines (38), in addition to inhibition of cell viability, ORES may target STAT3-mediated VEGF expression in osteosarcoma. The inhibitory concentrations of ORES in various tumor cells were different. The IC50 value of ORES in T24 bladder cancer cells was 47.46 µM (39). In HCC cells, the IC50 values were nearly 80 µM (37). As for neuroblastoma cells, the IC50 values reached 140 µM, with little inhibitory effect against non-cancer cells, such as Rat-2, HEK293, NIH3T3 and human peripheral blood mononuclear cells (29). Therefore, Saos-2 cells were relatively sensitive to ORES with an IC50 of <15 µM. In view of the broad biological activities of ORES, especially its significant antitumor effects, further research should focus on both cancer cells and tumor-surrounding cells, including endothelial cells and immune cells.

It has been reported that multiple signaling pathways influence the process of osteosarcoma cell growth and anti-apoptosis, which can be interfered with using small molecule compounds (4042). For instance, caffeine has been shown to induce apoptosis of osteosarcoma cells by inhibiting the AKT/mTOR/ribosomal protein S6 kinase, NF-κB and MAPK pathways (40). Glycogen synthase kinase 3 inhibitors have also been demonstrated to inhibit proliferation in osteosarcoma cell lines U2OS and MG-63 in vitro (41). Messerschmitt et al (42) reported that specific tyrosine kinase inhibitors, such as inhibitors of EGF-R, insulin-like growth factor 1 and Met, decreased motility and colony formation in human osteosarcoma cell lines. Signaling pathways, such as the hedgehog (43), Wnt (44) and PI3K/AKT signaling pathways (45), are all involved in osteosarcoma proliferation. Notably, recently discovered types of cell death, such as receptor-interacting protein (RIP)1- and RIP3-dependent necroptosis, as well as Ca2+/calmodulin-dependent protein kinase type II a activity-related autophagic degradation, have been considered as efficient anti-osteosarcoma strategies in recent research (46,47). Resveratrol, which is structurally similar to ORES, has been shown to induce the apoptosis of osteosarcoma cells via modulating the microRNA139-5p/NOTCH1 signaling pathway (48). Additionally, resveratrol can inhibit osteosarcoma MG-63 and MNNG/HOS cell proliferation and tumorigenesis via blocking janus kinase (JAK)2/STAT3 signaling (49). Consistent with this, in the present study, as a potent tyrosinase inhibitor, ORES inhibited the growth of osteosarcoma cells and promoted apoptosis by affecting the STAT3 signaling pathway, which indicated that STAT3 may be another drug target for ORES.

STAT3 has already been confirmed to serve a crucial role in selectively inducing and maintaining a carcinogenic inflammatory microenvironment in response to cytokine stimulation (17). The IL-6-JAK-STAT3 signaling axis contributes to the pathogenesis of myeloma cells by preventing apoptosis (50). By blocking the IL-6-OPN-STAT3 signaling pathway, the viability and tumorigenesis of osteosarcoma cells are inhibited (21). This is consistent with the findings of the present study, which demonstrated that compared with in the control group, the phosphorylation of STAT3 was attenuated by ORES treatment in Saos-2 cells. In addition, when P-STAT3 was partially activated by IL-6, the inhibition of cell viability and apoptosis promotion induced by ORES were also rescued. As a major cytokine in the tumor microenvironment, IL-6 protects cancer cells against apoptosis via multiple signaling pathways (51). The rescue of STAT3 phosphorylation may explain in part the reversal effect of ORES. Given that completely blocking STAT3 was demonstrated to be a risk factor causing other diseases, such as hyper-immunoglobulin E syndrome (52), inhibition of STAT3 should be under strict control. Local suppression of STAT3 for a limited period of time may convert inflammation in the tumor microenvironment from promoting tumor growth to inhibition without serious side effects (17). FDA-approved tyrosine kinase inhibitors, such as sorafenib (53) and sunitinib (54), are already in clinical use and can indirectly inhibit STAT3 signaling, resulting in tumor cell cycle arrest and the induction of apoptosis. The development of new STAT3 inhibitors may inhibit the viability and survival of multiple tumor cells.

In conclusion, the present study demonstrated the interaction between ORES and osteosarcoma. ORES induced apoptosis and inhibited cell viability by reducing STAT3 signaling in Saos-2 cells. More comprehensive studies of ORES may establish it as a promising agent for the future treatment of osteosarcoma.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81971753), the Program for Medical Key Department of Shanghai (grant no. ZK2019C01), the Program for the Outstanding Clinical Discipline Project of Shanghai Pudong (grant no. PWYgy2018-09), the Key Disciplines Group Construction Project of Pudong Health Bureau of Shanghai (grant no. PWZxq2017-11), the Program for Outstanding Leader of Shanghai (grant no. 046), and the Talents Training Program of Pudong Hospital affiliated to Fudan University (grant no. PJ201903).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

BY designed the project and drafted the manuscript. TL and ZJ performed the experiments and drafted the manuscript. DL, RA and XZ participated in data analysis and revised the manuscript. 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 

Mirabello L, Troisi RJ and Savage SA: International osteosarcoma incidence patterns in children and adolescents, middle ages and elderly persons. Int J Cancer. 125:229–234. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Heymann MF, Lezot F and Heyman D: The contribution of immune infiltrates and the local microenvironment in the pathogenesis of osteosarcoma. Cell Immunol. 343:1037112019. View Article : Google Scholar : PubMed/NCBI

3 

Gorlick R, Janeway K, Lessnick S, Randall RL and Marina N; COG Bone Tumor Committee, : Children's oncology group's 2013 blueprint for research: Bone tumors. Pediatr Blood Cancer. 60:1009–1015. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Mirabello L, Troisi RJ and Savage SA: Osteosarcoma incidence and survival rates from 1973 to 2004: Data from the surveillance, epidemiology, and end results program. Cancer. 115:1531–1543. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Lu KH, Lin RC, Yang JS, Yang WE, Reiter RJ and Yang SF: Molecular and cellular mechanisms of melatonin in osteosarcoma. Cells. 8:16182019. View Article : Google Scholar

6 

Zhang Y, Cai L, Li D, Lao YH, Liu D, Li M, Ding J and Chen X: Tumor microenvironment-responsive hyaluronate-calcium carbonate hybrid nanoparticle enables effective chemotherapy for primary and advanced osteosarcomas. Nano Res. 11:4806–4822. 2018. View Article : Google Scholar

7 

Feng X, Xu W, Li Z, Song W, Ding J and Chen X: Immunomodulatory nanosystems. Adv Sci (Weinh). 6:19001012019. View Article : Google Scholar : PubMed/NCBI

8 

Wang J, Li Z, Wang Z, Yu Y, Li D, Li B and Ding J: Nanomaterials for combinational radio-immuno oncotherapy. Adv Funct Mater. Oct 7–2020.(Epub ahead of print).

9 

Lowe SW and Lin AW: Apoptosis in cancer. Carcinogenesis. 21:485–495. 2000. View Article : Google Scholar : PubMed/NCBI

10 

Fulda S and Debatin KM: Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene. 25:4798–4811. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Bhola PD and Letai A: Mitochondria-judges and executioners of cell death sentences. Mol Cell. 61:695–704. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Adams JM and Cory S: The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene. 26:1324–1337. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Chen G, Wang F, Trachootham D and Huang P: Preferential killing of cancer cells with mitochondrial dysfunction by natural compounds. Mitochondrion. 10:614–625. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Hu Y, Yu K, Wang G, Zhang D, Shi C, Ding Y, Hong D, Zhang D, He H, Sun L, et al: Lanatoside C inhibits cell proliferation and induces apoptosis through attenuating Wnt/β-catenin/c-Myc signaling pathway in human gastric cancer cell. Biochem Pharmacol. 150:280–292. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Wang Z, Yu K, Hu Y, Su F, Gao Z, Hu T, Yang Y, Cao X and Qian F: Schisantherin A induces cell apoptosis through ROS/JNK signaling pathway in human gastric cancer cells. Biochem Pharmacol. 173:1136732020. View Article : Google Scholar : PubMed/NCBI

16 

Ashkenazi A, Fairbrother WJ, Leverson JD and Souers AJ: From basic apoptosis discoveries to advanced selective BCL-2 family inhibitors. Nat Rev Drug Discov. 16:273–284. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Yu H, Pardoll D and Jove R: STATs in cancer inflammation and immunity: A leading role for STAT3. Nat Rev Cancer. 9:798–809. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Wang YC, Zheng LH, Ma BA, Zhou Y, Zhang MH, Zhang DZ and Fan QY: Clinical value of signal transducers and activators of transcription 3 (STAT3) gene expression in human osteosarcoma. Acta Histochem. 113:402–408. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Wang X, Goldstein D, Crowe PJ and Yang JL: Impact of STAT3 inhibition on survival of osteosarcoma cell lines. Anticancer Res. 34:6537–6545. 2014.PubMed/NCBI

20 

Goel S, Sahu S, Minz RW, Singh S, Suri D, Oh YM, Rawat A, Sehgal S and Saikia B: STAT3-mediated transcriptional regulation of osteopontin in STAT3 loss-of-function related hyper IgE syndrome. Front Immunol. 9:10802018. View Article : Google Scholar : PubMed/NCBI

21 

Zhang C, Ma K and Li WY: Cinobufagin suppresses the characteristics of osteosarcoma cancer cells by inhibiting the IL-6-OPN-STAT3 pathway. Drug Des Devel Ther. 13:4075–4090. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Kim YM, Yun J, Lee CK, Lee H, Min KR and Kim Y: Oxyresveratrol and hydroxystilbene compounds. Inhibitory effect on tyrosinase and mechanism of action. J Biol Chem. 277:16340–16344. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Ortiz-Ruiz CV, Ballesta de Los Santos M, Berna J, Fenoll J, Garcia-Ruiz PA, Tudela J and Garcia-Canovas F: Kinetic characterization of oxyresveratrol as a tyrosinase substrate. IUBMB Life. 67:828–836. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Heo JI, Kim JH, Lee JM, Kho YJ, Lim SS, Park JB, Kim J, Kim SC and Lee JY: FOXO3a Activation by oxyresveratrol of Morus bombycis koidzumi extract mediates antioxidant activity. Anim Cells Syst. 20:39–47. 2016. View Article : Google Scholar

25 

Jia YN, Lu HP, Peng YL, Zhang BS, Gong XB, Su J, Zhou Y, Pan MH and Xu L: Oxyresveratrol prevents lipopolysaccharide/D-galactosamine-induced acute liver injury in mice. Int Immunopharmacol. 56:105–112. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Lee HS, Kim DH, Hong JE, Lee JY and Kim EJ: Oxyresveratrol suppresses lipopolysaccharide-induced inflammatory responses in murine macrophages. Hum Exp Toxicol. 34:808–818. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Wei J, Chen JR, Pais EMA, Wang TY, Miao L, Li L, Li LY, Qiu F, Hu LM, Gao XM and Fan GW: Oxyresveratrol is a phytoestrogen exerting anti-inflammatory effects through NF-κB and estrogen receptor signaling. Inflammation. 40:1285–1296. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Sintuyanon N, Phoolcharoen W, Pavasant P and Sooampon S: Resveratrol demonstrated higher antiproliferative and antiangiogenic efficacy compared with oxyresveratrol on head and neck squamous cell carcinoma cell lines. Natural Prod Commun. 12:1781–1784. 2017.

29 

Rahman MA, Bishayee K, Sadra A and Huh SO: Oxyresveratrol activates parallel apoptotic and autophagic cell death pathways in neuroblastoma cells. Biochim Biophys Acta Gen Subj. 1861:23–36. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Matencio A, Dhakar NK, Bessone F, Musso G, Cavalli R, Dianzani C, García-Carmona F, López-Nicolás JM and Trotta F: Study of oxyresveratrol complexes with insoluble cyclodextrin based nanosponges: Developing a novel way to obtain their complexation constants and application in an anticancer study. Carbohydr Polym. 231:1157632020. View Article : Google Scholar : PubMed/NCBI

31 

Lee JH, Baek SY, Jang EJ, Ku SK, Kim KM, Ki SH, Kim CE, Park KI, Kim SC and Kim YW: Oxyresveratrol ameliorates nonalcoholic fatty liver disease by regulating hepatic lipogenesis and fatty acid oxidation through liver kinase B1 and AMP-activated protein kinase. Chem Biol Interact. 289:68–74. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Tadtong S, Chatsumpun N, Sritularak B, Jongbunprasert V, Ploypradith P and Likhitwitayawuid K: Effects of oxyresveratrol and its derivatives on cultured P19-derived neurons. Trop J Pharm Res. 15:2619–2628. 2016. View Article : Google Scholar

33 

Lee HJ, Feng JH, Sim SM, Lim SS, Lee JY and Suh HW: Effects of resveratrol and oxyresveratrol on hippocampal cell death induced by kainic acid. Anim Cells Syst (Seoul). 23:246–252. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Duan C, Han J, Zhang C, Wu K and Lin Y: Inhibition of kidney cancer cell growth by Mulberroside-A is mediated via mitochondrial mediated apoptosis, inhibition of cell migration and invasion and targeting EGFR signalling pathway. J BUON. 24:296–300. 2019.PubMed/NCBI

35 

Li ZR, Ma T, Guo YJ, Hu B, Niu SH, Suo FZ, Du LN, You YH, Kang WT, Liu S, et al: Sanggenon O induced apoptosis of A549 cells is counterbalanced by protective autophagy. Bioorg Chem. 87:688–698. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Che Q, Xiao X, Liu M, Lu Y, Doug X and Liu S: IL-6 promotes endometrial cancer cells invasion and migration through signal transducers and activators of transcription 3 signaling pathway. Pathol Res Pract. 215:1523922019. View Article : Google Scholar : PubMed/NCBI

37 

Liu Y, Ren W, Bai Y, Wan L, Sun X, Liu Y, Xiong W, Zhang YY and Zhou L: Oxyresveratrol prevents murine H22 hepatocellular carcinoma growth and lymph node metastasis via inhibiting tumor angiogenesis and lymphangiogenesis. J Nat Med. 72:481–492. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Niu G, Wright KL, Huang M, Song L, Haura E, Turkson J, Zhang S, Wang T, Sinibaldi D, Coppola D, et al: Constitutive Stat3 activity up-regulates VEGF expression and tumor angiogenesis. Oncogene. 21:2000–2008. 2002. View Article : Google Scholar : PubMed/NCBI

39 

Yang Y, Zhang G, Li C, Wang S, Zhu M, Wang J, Yue H, Ma X, Zhen Y and Shu X: Metabolic profile and structure-activity relationship of resveratrol and its analogs in human bladder cancer cells. Cancer Manag Res. 11:4631–4642. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Miwa S, Sugimoto N, Yamamoto N, Shirai T, Nishida H, Hayashi K, Kimura H, Takeuchi A, Igarashi K, Yachie A and Tsuchiya H: Caffeine induces apoptosis of osteosarcoma cells by inhibiting AKT/mTOR/S6K, NF-κB and MAPK pathways. Anticancer Res. 32:3643–3649. 2012.PubMed/NCBI

41 

Lu K, Wang X, Chen Y, Liang D, Luo H, Long L, Hu Z and Bao J: Identification of two potential glycogen synthase kinase 3β inhibitors for the treatment of osteosarcoma. Acta Biochim Biophys Sin (Shanghai). 50:456–464. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Messerschmitt PJ, Rettew AN, Brookover RE, Garcia RM, Getty PJ and Greenfield EM: Specific tyrosine kinase inhibitors regulate human osteosarcoma cells in vitro. Clin Orthop Relat Res. 466:2168–2175. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Kumar RM and Fuchs B: Hedgehog signaling inhibitors as anti-cancer agents in osteosarcoma. Cancers (Basel). 7:784–794. 2015. View Article : Google Scholar : PubMed/NCBI

44 

McQueen P, Ghaffar S, Guo Y, Rubin EM, Zi XL and Hoangn BH: The Wnt signaling pathway: Implications for therapy in osteosarcoma. Expert Rev Anticancer Ther. 11:1223–1232. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Qian H, Huang T, Chen Y, Li X, Gong W, Jiang G, Zhang W, Cheng S, Li X and Li P: X-linked inhibitor of apoptosis protein inhibitor Embelin induces apoptosis via PI3K/Akt pathway and inhibits invasion in osteosarcoma cells. J Cancer Res Ther. 14 (Supplement):S648–S655. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Li S, Zhang T, Xu W, Ding J, Yin F, Xu J, Sun W, Wang H, Sun M, Cai Z and Hua Y: Sarcoma-targeting peptide-decorated polypeptide nanogel intracellularly delivers shikonin for upregulated osteosarcoma necroptosis and diminished pulmonary metastasis. Theranostics. 8:1361–1375. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Xu J, Wang H, Hu Y, Zhang YS, Wen L, Yin F, Wang Z, Zhang Y, Li S, Miao Y, et al: Inhibition of CaMKIIα activity enhances antitumor effect of fullerene C60 nanocrystals by suppression of autophagic degradation. Adv Sci (Weinh). 6:18012332019. View Article : Google Scholar : PubMed/NCBI

48 

Xiao X, Zhang Y, Pan W and Chen F: MiR-139-mediated NOTCH1 regulation is crucial for the inhibition of osteosarcoma progression caused by resveratrol. Life Sci. 242:1172152020. View Article : Google Scholar : PubMed/NCBI

49 

Peng L and Jiang D: Resveratrol eliminates cancer stem cells of osteosarcoma by STAT3 pathway inhibition. PLoS One. 13:e02059182018. View Article : Google Scholar : PubMed/NCBI

50 

Catlett-Falcone R, Landowski TH, Oshiro MM, Turkson J, Levitzki A, Savino R, Ciliberto G, Moscinski L, Fernández-Luna JL, Nuñez G, et al: Constitutive activation of Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells. Immunity. 10:105–115. 1999. View Article : Google Scholar : PubMed/NCBI

51 

Kumari N, Dwarakanath BS, Das A and Bhatt AN: Role of interleukin-6 in cancer progression and therapeutic resistance. Tumour Biol. 37:11553–11572. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Minegishi Y, Saito M, Tsuchiya S, Tsuge I, Takada H, Hara T, Kawamura N, Ariga T, Pasic S, Stojkovic O, et al: Dominant-negative mutations in the DNA-binding domain of STAT3 cause hyper-IgE syndrome. Nature. 448:1058–1062. 2007. View Article : Google Scholar : PubMed/NCBI

53 

Yang F, Van Meter TE, Buettner R, Hedvat M, Liang W, Kowolik CM, Mepani N, Mirosevich J, Nam S, Chen MY, et al: Sorafenib inhibits signal transducer and activator of transcription 3 signaling associated with growth arrest and apoptosis of medulloblastomas. Mol Cancer Ther. 7:3519–3526. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Xin H, Zhang C, Herrmann A, Du Y, Figlin R and Yu H: Sunitinib inhibition of Stat3 induces renal cell carcinoma tumor cell apoptosis and reduces immunosuppressive cells. Cancer Res. 69:2506–2513. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2020
Volume 22 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lv T, Jian Z, Li D, Ao R, Zhang X and Yu B: Oxyresveratrol induces apoptosis and inhibits cell viability via inhibition of the STAT3 signaling pathway in Saos‑2 cells. Mol Med Rep 22: 5191-5198, 2020
APA
Lv, T., Jian, Z., Li, D., Ao, R., Zhang, X., & Yu, B. (2020). Oxyresveratrol induces apoptosis and inhibits cell viability via inhibition of the STAT3 signaling pathway in Saos‑2 cells. Molecular Medicine Reports, 22, 5191-5198. https://doi.org/10.3892/mmr.2020.11591
MLA
Lv, T., Jian, Z., Li, D., Ao, R., Zhang, X., Yu, B."Oxyresveratrol induces apoptosis and inhibits cell viability via inhibition of the STAT3 signaling pathway in Saos‑2 cells". Molecular Medicine Reports 22.6 (2020): 5191-5198.
Chicago
Lv, T., Jian, Z., Li, D., Ao, R., Zhang, X., Yu, B."Oxyresveratrol induces apoptosis and inhibits cell viability via inhibition of the STAT3 signaling pathway in Saos‑2 cells". Molecular Medicine Reports 22, no. 6 (2020): 5191-5198. https://doi.org/10.3892/mmr.2020.11591