Open Access

Total flavonoids suppress lung cancer growth via the COX‑2‑mediated Wnt/β‑catenin signaling pathway

Retraction in: /10.3892/ol.2024.14212

  • Authors:
    • Lei Han
    • Shu Fang
    • Guangtao Li
    • Minghuan Wang
    • Renzhi Yu
  • View Affiliations

  • Published online on: January 9, 2020     https://doi.org/10.3892/ol.2020.11271
  • Pages: 1824-1830
  • Copyright: © Han 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

The aim of the present study was to explore the anti‑cancer effects of total flavonoids (TF) on lung cancer and to investigate the underlying mechanism. The inhibitory effect of TF on the proliferation of A549 cells in vitro was measured using an MTT assay. The apoptotic rate of TF‑treated A549 cells was analyzed using flow cytometry and terminal deoxynucleotidyl transferase‑mediated biotinylated UTP nick end labeling. Migration and invasion assays were performed to investigate the anti‑migration effect of TF on A549 cells. Reverse‑transcription quantitative PCR was used to analyze BCL2‑like 2, BCL2, Bax, Bad, cyclooxygenase 2 (COX‑2), Wnt and β‑catenin mRNA expression levels in A549 cells. The in vivo anti‑cancer effect of TF was investigated in a subcutaneous xenograft model of lung cancer in BALB/c nude mice. The results obtained in the present study revealed that TF exerted a significant inhibitory effect on the proliferation of A549 cells in a dose‑dependent manner (P<0.01). TF induced apoptosis of A549 cells, which exhibited increased and decreased expression of pro‑ and anti‑ apoptotic genes, respectively. Furthermore, TF had a significant inhibitory effect on the migration and invasion of A549 cells (P<0.01). The mRNA expression levels of COX‑2, Wnt and β‑catenin were significantly downregulated in TF‑treated A549 cells compared with controls. Additionally, treatment with TF inhibited tumor growth in mice, with a tumor inhibition rate of 64.07% compared with the controls. TF exhibited significant tumor inhibitory effects in vivo by promoting the apoptosis of tumor cells. In conclusion, the results suggested that TF may regulate lung cancer growth via the COX‑2‑Wnt/β‑catenin signaling pathway. TF may serve as a novel anti‑cancer agent for the treatment of lung cancer.

Introduction

Non-small cell lung cancer (NSCLC) is one of the most common human cancers and is characterized by rapid growth, high metastatic potential and reoccurrence (1). Approximately 75% of patients are diagnosed with advanced stage NSCLC, which is associated with a poor 5-year survival rate of 15% (2). NSCLC includes adenocarcinoma, large cell carcinoma and squamous cell carcinoma, the latter of which is the most common type of lung cancer and accounts for approximately 80% of all lung cancer cases (35). A systematic review and meta-analysis have indicated the efficacy of targeted agents in the treatment of elderly patients with advanced NSCLC (6). Cancer cell migration and invasion are key events in the development, metastasis and reoccurrence of NSCLC, and previous studies have demonstrated the importance of inhibiting growth and metastasis when selecting clinical treatment regimens for NSCLC (79).

A previous study has shown that total flavonoids (TF) reduce the risk of developing chronic diseases, such as cancer and cardiovascular diseases (10). Currently, few studies have investigated the biological activity of TF, particularly in human cancer cells (1113). Wang et al (14) revealed that TF extracted from Cotinus coggygria exhibit potential therapeutic effect by reducing the proliferation and inducing apoptosis by regulating the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/ERK signaling pathway in glioblastoma cancer cells (14). The antitumor activity of TF isolated from Daphne genkwa has been analyzed in colorectal cancer, and revealed that the action of TF is likely associated with the regulation of immune function and decreased production of inflammatory cytokines (15). However, the therapeutic effects of TF in NSCLC are not well understood, particularly in terms of their in vivo anti-cancer efficacy (16).

The present study investigated whether TF exert anti-cancer effects in NSCLC cells by promoting apoptosis and inhibiting growth and migration. The results indicated that TF treatment significantly promoted apoptosis and inhibited the growth of A549 cells via the cyclooxygenase 2 (COX-2)/Wnt/β-catenin signaling pathway, which suggested that TF may serve as a novel therapeutic agent in NSCLC.

Materials and methods

Cell culture

A549 cells were purchased from the American Type Culture Collection and were cultured in Dulbecco's Modified Eagle Medium (DMEM; Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% heat-inactivated fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin/streptomycin (Sigma-Aldrich, Merck KGaA). Cells were maintained at 37°C and 5% CO2.

Reverse-transcription quantitative PCR (RT-qPCR)

Total RNA was extracted from A549 cells using the RNeasy Mini kit (Qiagen, Inc.) according to the manufacturer's protocol. The mRNA expression levels of BCL2-like 2 (BCL2L2), BCL2 apoptosis regulator (BCL2), BCL2 associated agonist of cell death (BAD) and BCL2 associated X apoptosis regulator (BAX), COX-2, Wnt and β-catenin in A549 cells were measured by RT-qPCR with β-actin as an endogenous control as previously described (17). The forward and reverse primers used for qPCR were synthesized by Invitrogen, Thermo Fisher Scientific, Inc., and are presented in Table I. qPCR was performed using SYBR-Green Master Mix (Takara Bio, Inc.) according to the manufacturer's instructions and an ABI 7500 Fast Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The following thermocycling conditions were used: 95°C for 90 sec, followed by 45 cycles of 95°C for 30 sec, 57.5°C for 20 sec and 72°C for 30 sec. mRNA expression levels were calculated using the 2−ΔΔCq method (18) and normalized to β-actin levels.

Table I.

Primer sequences used for quantitative PCR.

Table I.

Primer sequences used for quantitative PCR.

Gene nameSequence (5′→3′)
BCL2 associated X apoptosis regulatorF: CTTCTCACTGTCGACTACCGC
R: GCGTCTCCTGTGCATTCG
BCL2 associated agonist of cell deathF: GCAAGGACAAGATTCGATACT
R: GCCAGACTACATGGAAATCTA
BCL2 apoptosis regulatorF: CATGCTGGGGCCGTACAG
R: TTGTCCGACCTTTGGCAACT
BCL2-like 2F: ATTTGCGTGTGGAGTATTTGG
R: GCTGTTCCGTCCCAGTAGATTA
Cyclooxygenase 2F: CCGGGTACAATCGCACTTAT
R: GGCGCTCAGCCATACAG
WntF: CTCGTCGTACTCCTGCTTGGT
R: ACTGGTGAGACCTGCGTGTA
β-cateninF: CATCTACACAGTTTGATGCTGCT
R: GCAGTTTTGTCAGTTCAGGGA
β-actinF: CATGTACGTTGCTATCCAGGC
R: CTCCTTAATGTCACGCACGAT
Overexpression of COX-2

The human COX-2 gene was cloned into a pcDNA3.1 plasmid (Invitrogen; Thermo Fisher Scientific, Inc.) to produce the pcDNA3.1-COX-2 vector. A549 cells (1×105 cells/well) were cultured in six-well plates until 90% confluence was reached and subsequently transfected with the pcDNA3.1-COX-2 vector (100 nM) or empty pcDNA3.1 (100 nM) plasmid using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol.

MTT cytotoxicity assay

A549 cells (1×103 cells/well) were incubated in 96-well plates with 2.5, 5.0 and 7.5 mg/ml TF (purity ≥95%, Sigma-Aldrich; Merck KGaA) for 24, 48 and 72 h at 37°C. TF were originally extracted from Daphne genkwa and dissolved in 40% ethanol. For the control group, cells were incubated with PBS instead of TF. A total of 20 µl MTT (5 mg/ml) solution in PBS was added to the wells at each time point, and the cells were incubated for an additional 4 h. Subsequently, 100 µl dimethyl sulfoxide were added to the wells to dissolve the formazan crystals and the optical density was measured at wavelength of 490 nm using a plate reader. Each experiment was performed in triplicate.

Cells invasion and migration assays

A549 cells were incubated with 5 mg/ml TF for 24 h at 37°C based on the maximum inhibitory effect on cell growth. A total of 1×105 A549 cells in 500 µl serum-free DMEM were pipetted into the upper chamber of Transwell inserts (8-µm pore size; Corning Life Sciences) and incubated for 48 h at 37°C. For the invasion assay, the Transwell inserts were precoated with Matrigel (1 mg/ml; BD Biosciences) for 48 h at 37°C. The cells were subsequently fixed with 4% paraformaldehyde for 5 min at 37°C and stained with 0.1% crystal violet for 30 min at 37°C. The numbers of migratory or invading A549 cells were counted in at least three randomly selected fields of view using a phase contrast microscope (Olympus Corp; magnification, ×100). Each experiment was performed at least three times.

Flow cytometry analysis

A549 cells (1×106 cells/well) were cultured until 90% confluence was reached. Apoptosis was assessed after incubation with TF (5 mg/ml) for 24 h at 37°C. A549 cells were subsequently trypsinized, washed in cold PBS, and adjusted to 1×106 cells/ml in PBS. Cells were stained with annexin V-fluorescein isothiocyanate and propidium iodide (Annexin V-FITC kit; cat. no. 556547; BD Biosciences) and analyzed using a flow cytometer (FACScan; BD Biosciences). Cell apoptosis was measured using BD FACSuite software (version 2; BD Biosciences).

Animal study

A total of 60 specific pathogen-free male BALB/c nude mice (age, eight weeks; weight, 32–35 g) were purchased from Shanghai Slack Experimental Animals Co., Ltd. Mice received an inguinal injection of 1×108 A549 cells in 150 µl PBS and were randomly divided into two groups (n=10 per group). Mice received treatment 6 days after tumor implantation when the tumor diameter reached 5–8 mm. Mice were intravenously injected with 20 mg/kg/day TF (dissolved in 40% ethanol) or vehicle (ethanol) as a control (19). The treatment was continued for 10 days (20). The tumor volumes were calculated as previously described using the following equation: Volume (mm3)=axb2/2, where a and b represent the longest and shortest diameters, respectively (21). On day 30, three mice per group were sacrificed for further analysis. The remaining mice were kept to investigate the survival time over a 100-day period. Tumor growth, animal health and behavior including self-centered behavior, motivation, anhedonia, anxiety and despair behavior were monitored every five days as described previously (22). Mice were sacrificed by cervical dislocation when the tumor diameter reached 15 mm. Experiments were performed in triplicate. Tumor inhibition rate was calculated using the following formula: Inhibitory rate (%)=(mean tumor volume in PBS-mean tumor volume in TF)/mean tumor volume in PBS ×100%. The present study was approved by the Ethics Committee of Mudanjiang Medical University.

Immunohistochemistry

Tumor tissues were harvested and fixed using 4% formaldehyde for 30 min at 25°C. Tissues were embedded in paraffin and cut into 4-µm serial sections. Antigen retrieval was performed by incubating the tumor sections with citrate buffer (pH 6.0) for 7 min at 100°C. Tumor sections were blocked with 5% BSA (Sigma-Aldrich; Merck KGaA) overnight at 4°C and incubated with rabbit anti-human COX-2 (1:1,000; ab15191; Abcam), Wnt (1:1,000; ab15251; Abcam) and β-catenin (1:1,000; ab32572; Abcam) primary antibodies overnight at 4°C. Following the primary incubation, sections were incubated with horseradish peroxidase-conjugated polyclonal anti-rabbit IgG secondary antibodies (1:10,000; ab6721; Abcam) for 1 h at room temperature. A Ventana Benchmark automated staining system was used to perform the staining (Bio-Rad Laboratories, Inc.). The staining was observed in six randomly selected fields of view (magnification, ×100) under a Zeiss immunofluorescence microscope (DMI5000M; Carl Zeiss AG). Densitometric quantification of the protein expression was performed using Quantity-One software (version 1.2; Bio-Rad Laboratories, Inc.)

Terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling (TUNEL)

TUNEL staining was used to analyze apoptotic cells in lung tumor tissues. Briefly, paraffin-embedded tumor sections were labeled with BrdU (cat. no. MAB4072; 1:1,000; Sigma-Aldrich; Merck KGaA) as previously described (23) and TUNEL-positive cells were identified using the ApopTag kit (EMD Millipore) according to the manufacturer's instructions. The staining was observed in at least three randomly selected microscopic fields under a fluorescence microscope (Carl Zeiss AG; magnification, ×100). Statistical quantification of TUNEL-positive tumor cells was performed to evaluate the pro-apoptotic effects of TF using six randomly selected fields of view to count the total number of cells and TUNEL-positive cells.

Statistical analysis

All data are expressed as the mean ± SD of three independent experiments and analyzed using the Student's t-test or one-way ANOVA followed by the Tukey post hoc test. The Kaplan-Meier method and the log-rank test were used to evaluate overall survival rate. All data were analyzed using SPSS software (version 19.0; IBM Corp.) and GraphPad Prism software (version 5.0; GraphPad Software, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Evaluation of the inhibitory effects of TF on A549 cells

The inhibitory effects of TF on the proliferation, migration and invasion of A549 cells were analyzed in vitro. Compared with the control group, TF-treated cells exhibited a significant dose-dependent decrease in proliferation (P<0.01; Fig. 1A and B). A concentration of 5 g/ml TF inhibited the proliferation of A549 cells to the greatest extent. Furthermore, treatment with 5 mg/ml TF for 24 h significantly inhibited the migration and invasion of A549 cells compared with the control group (P<0.01; Fig. 1C and D). These results indicated that TF treatment significantly inhibited the proliferation, migration and invasion of A549 cells.

TF treatment induces apoptosis of A549 cells

The effects of TF (5 mg/ml) on the apoptosis of A549 cells were investigated in vitro. TF treatment significantly increased the apoptosis of A549 cells compared with the control (P<0.01; Fig. 2A). RT-qPCR revealed that TF treatment decreased the expression levels of the anti-apoptotic genes BCL2L1 and BCL22 compared with control A549 cells (P<0.01; Fig. 2B). Furthermore, TF treatment increased the expression levels of the pro-apoptotic genes BAX and BAD compared with control A549 cells (P<0.01; Fig. 2C). These results indicated that TF treatment significantly affected apoptosis of A549 cells by regulating the expression of apoptosis-associated genes.

TF treatment suppresses proliferation via the COX-2/Wnt/β-catenin signaling pathway

The potential mechanism underlying the anti-cancer effects of TF was analyzed by investigating the COX-2/Wnt/β-catenin signaling pathway in A549 cells. TF treatment significantly decreased the mRNA expression levels of COX-2, Wnt and β-catenin in A549 cells compared with the control group (P<0.01; Fig. 3A). Furthermore, COX-2 overexpression inhibited the TF-mediated COX-2, Wnt and β-catenin downregulation in A549 cells (P<0.01; Fig. 3B). Additionally, COX-2 overexpression inhibited the TF-mediated decrease in proliferation and apoptosis of A549 cells (P<0.01; Fig. 3C and D). The transfection efficiency was determined by RT-qPCR after 72 h (Fig. S1). These results suggested that TF treatment significantly suppressed proliferation and promoted apoptosis of A549 cells via the COX-2/Wnt/β-catenin signaling pathway.

Inhibitory effect of TF on tumor growth in vivo

The anti-cancer efficacy of TF treatment was investigated in mouse xenograft tumor model. No animals presented with multiple tumors in the present study. As shown in Fig. 4A, TF treatment significantly inhibited tumor growth in mice compared with controls (P<0.01), with a tumor inhibition rate of 64.07%. Results demonstrated that TF treatment increased the percentage of apoptotic cells in tumor tissue compared with control (P<0.01; Fig. 4B). Immunohistochemistry revealed that COX-2, Wnt and β-catenin expression levels in tumor tissues were significantly decreased compared with the controls (P<0.01; Fig. 4C). Furthermore, TF administration significantly prolonged the survival of the animals compared with controls over a 100-day period (P<0.01; Fig. 4D). These results suggested that TF may serve as an efficient anti-cancer agent for lung cancer in vivo.

Discussion

NSCLC has a high incidence and a poor prognosis (24). A previous study revealed that a Chinese medicine consisting of TF isolated from Fructus viticis inhibited the stem-like properties of lung cancer stem-like cells by downregulating the expression of p-AKT (25). In the present study, treatment with TF extracted from Daphne genkwa significantly inhibited the proliferation and migration and invasion abilities of A549 cells in a dose-dependent manner. Furthermore, TF promoted the apoptosis of A549 cells by increasing the expression levels of the pro-apoptotic genes apoptotic peptidase activating factor 1 (APAF1) and BAD and decreasing the expression levels of the anti-apoptotic genes BCL2L2 and BCL2. Further investigation revealed that TF increased apoptosis via the COX-2/Wnt/β-catenin signaling pathway.

TF have been shown to exhibit antitumor activity various human cancer cells (26). A previous study has demonstrated that TF significantly inhibited tumor growth and induced the apoptosis of prostate cancer cells via the PI3K/Akt/phosphatase and tensin homolog signaling pathway, which provided a theoretical basis for the investigation of the anti-cancer effects of TF (2729). Furthermore, TF reduced the proliferation and promoted apoptosis of the human breast cancer cell line MCF-7, by selectively reducing BCL2 and nuclear factor-κB expression and increasing the expression of caspase-3 and 9, suggesting that TF inhibited cellular proliferation by activating the mitochondria-dependent apoptotic pathway (30). The results in the present study revealed that compared with the control, TF treatment promoted apoptosis and increased expression of BAK and BAD in A549 cells. Furthermore, the present study revealed that TF treatment inhibited proliferation, migration and invasion of A549 cells by downregulating the COX-2/Wnt/β-catenin in vitro and in vivo. A previous study demonstrated that the COX-2/Wnt/β-catenin signaling pathway may be involved in the antitumor mechanism of TF isolated from Radix tetrastigmae in hepatocellular carcinoma cells (31).

The Wnt/β-catenin signaling pathway plays an important role in regulating tumor growth, differentiation, oncogenesis, invasion and metastasis (32,33). In the present study, TF treatment inhibited the Wnt/β-catenin signaling pathway, which decreased the proliferation of A549 cells and increased apoptosis. A previous study demonstrated that the inhibition of the Wnt/β-catenin signaling pathway improves the anti-tumor effects of sorafenib in hepatocellular carcinoma (34). Another study demonstrated that downregulation of the Wnt/β-catenin signaling pathway inhibited the proliferation and increased apoptosis in A549 and H460 cells (35).

In conclusion, to the best of the authors' knowledge, the present study was the first to report the therapeutic significance of TF in lung cancer to investigate the potential mechanism. TF inhibited the proliferation and induced apoptosis of A549 cells by downregulating the COX-2/Wnt/β-catenin signaling pathway. The results obtained in the current study suggested that TF may serve as a novel anticancer agent and demonstrated that the COX-2/Wnt/β-catenin signaling pathway may be a potential therapeutic target for the treatment of lung cancer.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

LH and SF performed the experiments. GL and MW contributed to data analysis and experiments. RY designed this study and wrote manuscript.

Ethics approval and consent to participate

The study was approved by the Ethics Committee of Mudanjiang Medical University Affiliated Hongqi Hospital (Mudanjiang, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Loganadane G, Martinetti F, Mercier O, Krhili S, Riet FG, Mbagui R, To H, Le Péchoux C and Levy A: Stereotactic ablative radiotherapy for early stage non-small cell lung cancer: A critical literature review of predictive factors of relapse. Cancer Treat Rev. 50:240–246. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Sun G, Liu B, He J, Zhao X and Li B: Expression of EGFR is closely related to reduced 3-year survival rate in Chinese female NSCLC. Med Sci Monit. 21:2225–2231. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Xiao J, Hu CP, He BX, Chen X, Lu XX, Xie MX, Li W, He SY, You SJ and Chen Q: PTEN expression is a prognostic marker for patients with non-small cell lung cancer: A systematic review and meta-analysis of the literature. Oncotarget. 7:57832–57840. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Li SJ, Huang J, Zhou XD, Zhang WB, Lai YT and Che GW: Clinicopathological and prognostic significance of Oct-4 expression in patients with non-small cell lung cancer: A systematic review and meta-analysis. J Thorac Dis. 8:1587–1600. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Lim JS and Soo RA: Nivolumab in the treatment of metastatic squamous non-small cell lung cancer: A review of the evidence. Ther Adv Respir Dis. 10:444–454. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Chen J, Wu X, Shi T and Kang M: Efficacy of targeted agents in the treatment of elderly patients with advanced non-small-cell lung cancer: A systematic review and meta-analysis. Onco Targets Ther. 9:4797–4803. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Stewart EL, Tan SZ, Liu G and Tsao MS: Known and putative mechanisms of resistance to EGFR targeted therapies in NSCLC patients with EGFR mutations-A review. Transl Lung Cancer Res. 4:67–81. 2015.PubMed/NCBI

8 

Zhou JG, Tian X, Wang X, Tian JH, Wang Y, Wang F, Zhang Y and Ma H: Treatment on advanced NSCLC: Platinum-based chemotherapy plus erlotinib or platinum-based chemotherapy alone? A systematic review and meta-analysis of randomised controlled trials. Med Oncol. 32:4712015. View Article : Google Scholar : PubMed/NCBI

9 

Lange A, Prenzler A, Frank M, Golpon H, Welte T and von der Schulenburg JM: A systematic review of the cost-effectiveness of targeted therapies for metastatic non-small cell lung cancer (NSCLC). BMC Pulm Med. 14:1922014. View Article : Google Scholar : PubMed/NCBI

10 

Zamora-Ros R, Knaze V, Lujan-Barroso L, Romieu I, Scalbert A, Slimani N, Hjartåker A, Engeset D, Skeie G, Overvad K, et al: Differences in dietary intakes, food sources and determinants of total flavonoids between Mediterranean and non-Mediterranean countries participating in the European prospective investigation into cancer and nutrition (EPIC) study. Br J Nutr. 109:1498–1507. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Cheng Y, Tan J, Li H, Kong X, Liu Y, Guo R, Li G, Yang B and Pei M: Cardioprotective effects of total flavonoids from Jinhe Yangxin prescription by activating the PI3K/Akt signaling pathway in myocardial ischemia injury. Biomed Pharmacother. 98:308–317. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Zhang XX, Wu QF, Yan YL and Zhang FL: Inhibitory effects and related molecular mechanisms of total flavonoids in Mosla Chinensis Maxim against H1N1 influenza virus. Inflamm Res. 67:179–189. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Liu Q, Zhang L, Shan Q, Ding Y, Zhang Z, Zhu M and Mao Y: Total flavonoids from Astragalus alleviate endothelial dysfunction by activating the Akt/eNOS pathway. J Int Med Res. 46:2096–2103. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Wang G, Wang J, Du L and Li F: Effect and mechanism of total flavonoids extracted from cotinus coggygria against glioblastoma cancer in vitro and in vivo. Biomed Res Int. 2015:8563492015. View Article : Google Scholar : PubMed/NCBI

15 

Du WJ, Yang XL, Song ZJ, Wang JY, Zhang WJ, He X, Zhang RQ, Zhang CF, Li F, Yu CH, et al: Antitumor activity of total flavonoids from daphne genkwa in colorectal cancer. Phytother Res. 30:323–330. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Wang Y, Cao HJ, Sun SJ, Dai JY, Fang JW, Li QH, Yan C, Mao WW and Zhang YY: Total flavonoid aglycones extract in Radix scutellariae inhibits lung carcinoma and lung metastasis by affecting cell cycle and DNA synthesis. J Ethnopharmacol. 194:269–279. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Xiao S, Wang J and Xiao N: MicroRNAs as noninvasive biomarkers in bladder cancer detection: A diagnostic meta-analysis based on qRT-PCR data. Int J Biol Markers. 31:e276–e285. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Chen L, Wei Y, Zhao S, Zhang M, Yan X, Gao X, Li J and Gao Y, Zhang A and Gao Y: Antitumor and immunomodulatory activities of total flavonoids extract from persimmon leaves in H22 liver tumor-bearing mice. Sci Rep. 8:105232018. View Article : Google Scholar : PubMed/NCBI

20 

Feng Z, Hao W, Lin X, Fan D and Zhou J: Antitumor activity of total flavonoids from Tetrastigma hemsleyanum Diels et Gilg is associated with the inhibition of regulatory T cells in mice. OncoTargets Ther. 7:947–956. 2014.

21 

Zhuang T, Djemil T, Qi P, Magnelli A, Stephans K, Videtic G and Xia P: Dose calculation differences between Monte Carlo and pencil beam depend on the tumor locations and volumes for lung stereotactic body radiation therapy. J Appl Clin Med Phys. 14:40112013. View Article : Google Scholar : PubMed/NCBI

22 

Stepanichev M, Markov D, Pasikova N and Gulyaeva N: Behavior and the cholinergic parameters in olfactory bulbectomized female rodents: Difference between rats and mice. Behav Brain Res. 297:5–14. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Kalyuzhny AE: Combination of TUNEL assay with immunohistochemistry for simultaneous detection of DNA fragmentation and oxidative cell damage. Methods Mol Biol. 682:15–27. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Ren Z, Zhou S, Liu Z and Xu S: Randomized controlled trials of induction treatment and surgery versus combined chemotherapy and radiotherapy in stages IIIA-N2 NSCLC: A systematic review and meta-analysis. J Thorac Dis. 7:1414–1422. 2015.PubMed/NCBI

25 

Cao X, Zou H, Cao J, Cui Y, Sun S, Ren K, Song Z, Li D and Quan M: A candidate Chinese medicine preparation-fructus viticis total flavonoids inhibits stem-like characteristics of lung cancer stem-like cells. BMC Complement Altern Med. 16:3642016. View Article : Google Scholar : PubMed/NCBI

26 

Attoub S, Hassan AH, Vanhoecke B, Iratni R, Takahashi T, Gaben AM, Bracke M, Awad S, John A, Kamalboor HA, et al: Inhibition of cell survival, invasion, tumor growth and histone deacetylase activity by the dietary flavonoid luteolin in human epithelioid cancer cells. Eur J Pharmacol. 651:18–25. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Zhu WB, Xiao N and Liu XJ: Dietary flavonoid tangeretin induces reprogramming of epithelial to mesenchymal transition in prostate cancer cells by targeting the PI3K/Akt/mTOR signaling pathway. Oncol Lett. 15:433–440. 2018.PubMed/NCBI

28 

Adhami VM, Syed DN, Khan N and Mukhtar H: Dietary flavonoid fisetin: A novel dual inhibitor of PI3K/Akt and mTOR for prostate cancer management. Biochem Pharmacol. 84:1277–1281. 2012. View Article : Google Scholar : PubMed/NCBI

29 

He L, Wu Y, Lin L, Wang J, Wu Y, Chen Y, Yi Z, Liu M and Pang X: Hispidulin, a small flavonoid molecule, suppresses the angiogenesis and growth of human pancreatic cancer by targeting vascular endothelial growth factor receptor 2-mediated PI3K/Akt/mTOR signaling pathway. Cancer Sci. 102:219–225. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Wang S, Tian Q and An F: Growth inhibition and apoptotic effects of total flavonoids from Trollius chinensis on human breast cancer MCF-7 cells. Oncol Lett. 12:1705–1710. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Qinglin L, Xin W, Like Z, Fang L, Cao G and Huang P: A study on the anti-tumor mechanism of total flavonoids from radix tetrastigmae against additional cell line based on COX-2-mediated Wnt/β -catenin signaling pathway. Oncotarget. 8:54304–54319. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Schweigert A, Fischer C, Mayr D, von Schweinitz D, Kappler R and Hubertus J: Activation of the Wnt/β-catenin pathway is common in wilms tumor, but rarely through beta-catenin mutation and APC promoter methylation. Pediatr Surg Int. 32:1141–1146. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Ye Y, Long X, Zhang L, Chen J, Liu P, Li H, Wei F, Yu W, Ren X and Yu J: NTS/NTR1 co-expression enhances epithelial-to-mesenchymal transition and promotes tumor metastasis by activating the Wnt/β-catenin signaling pathway in hepatocellular carcinoma. Oncotarget. 7:70303–70322. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Lin HH, Feng WC, Lu LC, Shao YY, Hsu CH and Cheng AL: Inhibition of the Wnt/beta-catenin signaling pathway improves the anti-tumor effects of sorafenib against hepatocellular carcinoma. Cancer Lett. 381:58–66. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Bi X, Xia X, Mou T, Jiang B, Fan D, Wang P, Liu Y, Hou Y and Zhao Y: Anti-tumor activity of three ginsenoside derivatives in lung cancer is associated with Wnt/β-catenin signaling inhibition. Eur J Pharmacol. 742:145–152. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2020
Volume 19 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Han L, Fang S, Li G, Wang M and Yu R: Total flavonoids suppress lung cancer growth via the COX‑2‑mediated Wnt/β‑catenin signaling pathway Retraction in /10.3892/ol.2024.14212. Oncol Lett 19: 1824-1830, 2020
APA
Han, L., Fang, S., Li, G., Wang, M., & Yu, R. (2020). Total flavonoids suppress lung cancer growth via the COX‑2‑mediated Wnt/β‑catenin signaling pathway Retraction in /10.3892/ol.2024.14212. Oncology Letters, 19, 1824-1830. https://doi.org/10.3892/ol.2020.11271
MLA
Han, L., Fang, S., Li, G., Wang, M., Yu, R."Total flavonoids suppress lung cancer growth via the COX‑2‑mediated Wnt/β‑catenin signaling pathway Retraction in /10.3892/ol.2024.14212". Oncology Letters 19.3 (2020): 1824-1830.
Chicago
Han, L., Fang, S., Li, G., Wang, M., Yu, R."Total flavonoids suppress lung cancer growth via the COX‑2‑mediated Wnt/β‑catenin signaling pathway Retraction in /10.3892/ol.2024.14212". Oncology Letters 19, no. 3 (2020): 1824-1830. https://doi.org/10.3892/ol.2020.11271