Open Access

The anti‑migration and anti‑invasion effects of Bruceine D in human triple‑negative breast cancer MDA‑MB‑231 cells

  • Authors:
    • Can Luo
    • Yu Wang
    • Cheng Wei
    • Yuxin Chen
    • Zhaoning Ji
  • View Affiliations

  • Published online on: November 12, 2019     https://doi.org/10.3892/etm.2019.8187
  • Pages: 273-279
  • Copyright: © Luo 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

Brucein D (BD) is a naturally occurring major active quassinoid extracted from the Chinese medicinal herb Brucea javanica, which has been previously demonstrated to exhibit anticancer activities. The present study aimed to investigate the anticancer effects of BD on MDA‑MB‑231 cells, a human triple‑negative breast cancer (TNBC) cell line. An MTT assay was performed to assess cell viability, whilst wound healing and Transwell assay were applied to measure cell migration and invasion, respectively. Western blot analysis was performed to assess the expression of E‑cadherin, vimentin and β‑catenin, which are proteins associated with epithelial‑mesenchymal transformation (EMT), and PI3K, AKT and p‑AKT, which are key components of the PI3K/AKT signaling pathway. BD was indicated to reduce cell viability in a dose‑ and time‑dependent manner, whilst cell invasion and migration were also significantly inhibited in a dose‑dependent manner. Western blot analysis demonstrated that BD treatment significantly upregulated the expression of E‑cadherin and downregulated the expression of vimentin and β‑catenin. Additionally, BD downregulated the expression of PI3K and reduced AKT phosphorylation. In conclusion, BD can inhibit MDA‑MB‑231 cell viability, migration and invasion, suggesting the potential use of BD for the treatment of TNBC.

Introduction

Triple-negative breast cancer (TNBC) is a subtype of breast cancer that is characterized by the lack of estrogen receptor, progesterone receptor and human epidermal growth factor receptor 2 expression (1). In total, TNBC accounts for ~10–25% of all breast cancer cases (1), and is associated with unique clinical and pathophysiological phenotypes, including strong invasiveness, low overall survival rate and poor prognosis (2). Compared with other types of breast cancer, TNBC carries an increased probability of metastasis and recurrence (3) due to metastasis to other organs, including the lungs, liver, and brain via the systemic circulation. Targeted therapy cannot be administered to patients with TNBC due to the lack of hormone receptor expression. Although previous studies have reported that TNBC is more sensitive to chemotherapy compared with other types of breast cancer, TNBC remains to exhibit the worst prognosis compared with other subtypes (4). Therefore, the development of novel anti-metastatic therapeutic strategies is required for the effective treatment of TNBC.

Brucea javanica is the fruit of the Brucea javanica (L.) Merr (Simaroubaceae). It has been used to treat a number of diseases, including cancer, dysentery, malaria and stomach ulcers (5). Bruceine D (BD; Fig. 1A) is a bioactive component that can be isolated from Brucea javanica (6), which has previously been reported to induce apoptosis in pancreatic cancer cell lines PANC1, SW1990 and Capan1 and inhibit hepatoma cell proliferation (710).

Metastasis is an important feature of malignant tumors that impede the clinical treatment of cancer (11). Epithelial-mesenchymal transition (EMT) is an important stage in the metastasis of cancer cells. During this process, epithelial cell polarity is lost, and is coupled with concomitant enhancements in migratory and invasive abilities (12). As a result, the epithelial phenotype disappears, whereas the mesenchymal phenotype gradually develops (12). The PI3K/AKT signaling pathway has been previously indicated to be associated with cancer cell proliferation, differentiation, migration and invasion (13). AKT activation in epithelial cells reduces cell polarity and intercellular adhesion and promotes EMT in cancer cells by altering the expression and distribution of epithelial and mesenchymal markers (14).

In the present study, the TNBC cell line MDA-MB-231 was used to investigate the potential inhibitory effects of BD on cell viability, migration and invasion. In addition, the effect of BD on the EMT process and the PI3K/AKT signaling pathway were evaluated in this cell type.

Materials and methods

Materials

The sample of Bruceine D (≥98% purity) used in the current study was provided by the Institute of Traditional Chinese Medicine and Natural Products, Jinan University (Guangzhou, China). RPMI-1640 medium and FBS were purchased from Gibco; Thermo Fisher Scientific, Inc. The MTT cell proliferation and cytotoxicity assay kits and penicillin/streptomycin solution (PS) were purchased from Nanjing KeyGen Biotech Co., Ltd. The antibody targeting PI3K (cat. no. AF5112) was obtained from Affinity Biosciences. Antibodies against AKT (cat. no. 60203-2-Ig), phosphorylated (p)-AKT (cat. no. 66444-1-Ig), E-cadherin (cat. no. 20874-1-AP) and β-catenin (cat. no. 51067-2-AP) were purchased from Proteintech Group, Inc. Vimentin antibody (cat. no. BS1491) was purchased from Bioworld Technology, Inc. Antibodies against GAPDH (cat. no. ab181602) and horseradish peroxidase (HRP)-conjugated goat anti-rabbit (cat. no. ab6721) immunoglobulin (Ig) G were purchased from Abcam.

Cell culture

Human triple-negative breast cancer MDA-MB-231 cells were donated by Nanjing Pharmaceutical Co., Ltd. Cells were cultured in RPMI 1640 medium supplemented with 10% FBS and 1% PS solution in a humidified atmosphere with 5% CO2 at 37°C. BD was dissolved with DMSO and diluted in complete RPMI-1640 medium to required concentrations (1, 2 and 4 µM). The final DMSO concentration in the culture medium was ≤0.1% and control cells were treated with 0.1% DMSO at 37°C.

Cell viability assay

MTT assay was performed to measure cell viability. Cells were seeded into 96-well plates (5×103 cells/well), cultured overnight and subsequently treated at 37°C with ascending concentrations of BD (100, 50, 25, 12.5, 6.25, 3.13, 1.56, 0.78 and 0.39 µM) for 24, 48 and 72 h. Each well then received 20 µl MTT (5 mg/ml) and the cells were cultured for an additional 4 h at 37°C. Subsequently, cells were rinsed using PBS and each well received 150 µl DMSO. Optical density was then measured at the wavelength of 490 nm using a microplate reader (Mutiskan™ MK3; Thermo Fisher Scientific, Inc.). Data were presented as the percentage of survival rate relative to that of control.

Wound-healing assay

A wound-healing assay was performed to evaluate the migratory ability of MDA-MB-231 cells. Cells in the logarithmic growth phase were inoculated into six-well plates (5×103 cells/well). The following day, when ~100% of the surface was occupied a straight cell-free wound was introduced by scratching the bottom of the plate using a sterile pipette tip. Subsequently, the detached cells were washed twice with PBS and then re-incubated with BD (1, 2 and 4 µM) or 0.1% DMSO dissolved in serum-free RPMI 1640 medium for 24 h at 37°C. The wound images were obtained using a fluorescence inverted microscope (magnification, ×100) at 0 and 24 h, respectively, where the wound distance was measured using the following formula: Migration distance=scratch distance at 0 h-scratch distance at 24 h.

Transwell assay

The invasive capabilities of MDA-MB-231 cells were evaluated using a Transwell® assay (24 wells; Matrigel gel; Corning, Inc.). The cells were first cultured in serum-free RPMI medium for 24 h at 37°C. Matrigel was incubated at 4°C overnight, and melted Matrigel was diluted twice with incomplete medium. A total of 30 µl diluted Matrigel was added to the upper chamber of each Transwell insert, which was then incubated at 37°C for 120 min. The upper chamber of the Transwell contained 100 µl cells (1×104 cells/chamber) suspended in serum-free RPMI medium containing different concentrations of BD (1, 2 and 4 µM) or 0.1% DMSO, and the lower chamber was supplemented with 500 µl RPMI-1640 medium containing 20% FBS. Following incubation for 24 h at 37°C, cells on the upper surface of the membrane were removed using a cotton swab, whereas cells on the bottom surface of insert membrane were fixed with 95% alcohol for 10 min at room temperature and subsequently stained with 0.1% crystal violet for 30 min at room temperature. Invasive cells were photographed and counted in five random fields of view under a fluorescence inverted microscope (magnification, ×200).

Western blot analysis

Cells were first treated with BD (1–4 µM) or 0.1% DMSO for 24 h at 37°C before being washed with cold PBS, lysed using RIPA buffer (Gibco; Thermo Fisher Scientific, Inc.) and centrifuged at 13,000 × g for 15 min at 4°C. The supernatant was collected and stored at −20°C for further use. A BCA protein assay kit (Nanjing KeyGen Biotech Co., Ltd.) was used to quantify the total protein concentration for each sample. Protein samples (30 µg) were subsequently separated by 12% SDS-PAGE (90 min, 100 V) and transferred onto nitrocellulose membranes (90 min; 300 mA). The membranes were then blocked with 5% non-fat dry milk for 1 h at room temperature and washed three times with TBS supplemented with Tween-20 (TBS-T) following which the membranes were incubated with primary antibodies against vimentin (1:1,000), E-cadherin (1:5,000), β-catenin (1:5,000), PI3K (1:1,000), AKT (1:5,000), p-AKT (1:5,000) or GAPDH (1:10,000) overnight at 4°C. The membranes with the primary antibodies were then incubated on a shaker at room temperature for 30 min. After further rinsing with TBS-T three times, the membranes were incubated with HRP-conjugated goat anti-rabbit (1:2,000) IgG secondary antibodies at room temperature for 1 h. The protein bands were then visualized with enhanced chemiluminescent substrates (cat. no. 32106; Thermo Fisher Scientific, Inc.) using a Syngene G:BOX Chemi XR5 imaging system (Syngene International). Gel-Pro Analyzer software (version 4.0; Media Cybernetics, Inc.) was used to perform densitometric analysis on each membrane. GAPDH was used as a loading control.

Statistical analysis

All experimental data were presented as the mean ± standard deviation. All experiments were repeated in triplicate. The data were analyzed using a one-way ANOVA, followed by Tukey's test using GraphPad 7 (GraphPad Software, Inc). P<0.05 was considered to indicate a statistically significant difference.

Results

BD reduces MDA-MB-231 cell viability

MTT assay was performed to evaluate the effect of BD on the viability of MDA-MB-231 cells. The cells were treated with varying concentrations of BD (0–100 µM) for 24, 48 and 72 h at 37°C. BD treatment markedly reduced cell viability in a time- and dose-dependent manner (Fig. 1B). The half maximal inhibitory concentrations (IC50) of BD were calculated to be 40.805, 5.84 and 2.364 µM at 24, 48 and 72 h, respectively.

BD suppresses the migratory and invasive capabilities of MDA-MB-231 cells

Wound healing and Transwell assays were performed to explore the effects of BD on cell migration and invasion, respectively. In this assay, a low dose of BD (1, 2 and 4 µM), which exerted little to no effects on cell viability, was used. BD reduced the wound healing ability of MDA-MB-231 cells in a dose-dependent manner (Fig. 2A). The width of the wounds observed following treatment with BD at 1, 2 and 4 µM were 715.79±0.8, 714.62±1.01 and 718.71±1.01 µm at 0 h, respectively, which were not significantly different compared with the control group (719.01±1.34 µm; Fig. 2A and B). Following treatment with different concentrations of BD for 24 h, the width of the wounds were 425.44±13.82, 460.53±12.16 and 549.12±14.15 µm in the 1, 2 and 4 µM treatment groups, respectively, all of which were significantly higher compared with that in the control group (318.13±11.68 µm; Fig. 2A and B). In addition, BD reduced the invasive capabilities of MDA-MB-231 cells in a dose-dependent manner. The numbers of invasive cells were 148.33±5.53, 110±9.21 and 82.33±5.55 in the 1, 2 and 4 µM treatment groups, respectively (Fig. 2C and D), all of which were significantly lower compared with the control group (192.33±6.08). Altogether, these results indicated that BD suppressed the migratory and invasive capabilities of MDA-MB-231 cells in a dose-dependent manner.

BD reverses the EMT process of MDA-MB-231 cells

EMT is an important physiological process in the migration and invasion of malignant tumor cells (12). Following treatment with 1–4 µM BD or 0.1% DMSO for 24 h, western blot analysis was performed to evaluate the expression of E-cadherin, vimentin and β-catenin, which are proteins associated with EMT. E-cadherin expression in cells treated with BD were found to be significantly higher compared with the control group (Fig. 3A and B). In contrast, the expression of vimentin and β-catenin were significantly lower compared with the control group (Fig. 3A and B). Additionally, the upregulation of E-cadherin expression and the downregulation of vimentin and β-catenin expression appeared to be dependent on the dose of BD applied. These observations suggest that BD reversed the EMT process in MDA-MB-231 cells in a dose-dependent manner.

BD inhibits the activation of the PI3K/AKT pathway in MDA-MB-231 cells

The PI3K/AKT signaling pathway has been previously reported to serve a role in cancer cell migration and invasion (13). Therefore, western blot analysis was performed to determine the expression of PI3K, AKT, and p-AKT, which are key components in the PI3K/AKT signaling pathway. BD significantly reduced PI3K expression and AKT phosphorylation in a dose-dependent manner after 24 h, whilst no significant changes were observed in total AKT levels (Fig. 4). These results indicated that BD inhibited the PI3K/AKT signaling pathway in MDA-MB-231 cells in a dose-dependent manner.

Discussion

BD is a potent quassinoid extracted from the Brucea javanica plant, which has been demonstrated to exhibit anticancer effects in a number of previous studies (15). Xiao et al (10) reported that BD induced apoptosis of hepatoma cells by regulating microRNA-95 expression without affecting the growth of normal hepatocytes. Cheng et al (16) demonstrated that BD inhibited liver cancer proliferation by synergizing with the protein kinase inhibitor sorafenib to induce tumor necrosis and apoptosis. However, since the anticancer effects of BD on TNBC cells remain unclear, the present study examined the potential effects of BD on the MDA-MB-231 cell line. Data from the present study demonstrated that BD reduced the viability of MDA-MB-231 cells in a time- and dose-dependent manner. Additionally, low concentrations of BD (1–4 µM) were used for wound healing and Transwell assays, which indicated that BD significantly inhibited MDA-MB-231 cell migration and invasion in a dose-dependent manner, suggesting that BD exerts a potent anti-migratory effect on MDA-MB-231 cells.

Tumor metastasis is a process in which malignant tumor cells migrate from the primary site to other organs via lymphatic channels, blood vessels and body cavities (11). Previous studies have revealed that the EMT serves a pivotal role in the primary invasion and secondary metastasis of breast, colon and liver cancer (17), where the reduction or loss of E-cadherin expression is a key landmark change during the EMT process (18). E-cadherin is an important protein that is associated with cell-cell adhesion and attachment, and promotes adhesion between cells to maintain structural integrity (19). A number of studies have previously reported that E-cadherin is involved in the metastasis of malignancies, including colorectal, breast and cervical squamous cell carcinoma (2023). Sloan and Anderson (24) confirmed that breast cancer patients with lower E-cadherin expression were associated with higher rates of bone and lung metastasis. Indeed, the intracellular cytoskeletal structure is altered during the EMT, which is mainly characterized by upregulated vimentin expression (25). During the EMT process in breast cancer, the cytoskeleton profile changes from cytokeratin to vimentin, significantly increasing cell viability (26). In contrast, silencing vimentin expression reduces the invasiveness of breast cancer (27,28). β-catenin is a cytoskeletal protein that binds to E-cadherin and α-catenin to form the E-cadherin/catenin complex, which serves an important role in cell adhesion and maintaining the structural integrity of epithelial cells (29,30). However, the downregulation of E-cadherin expression results in the dissociation of this complex, releasing β-catenin into the nucleus to activate the TCF/Lef transcription factor, inducing the transcription of genes that regulate invasion and migration (31). Cheng et al (16) indicated that BD inhibited the expression of β-catenin in hepatoma cells. The results of the present study demonstrated that BD upregulates E-cadherin expression whilst downregulating β-catenin and vimentin expression in a concentration-dependent manner, suggesting that BD effectively reversed EMT in MDA-MB-231 cells.

The PI3K/AKT signaling pathway has been found to be aberrantly activated in a large number of tumor cells (32). PI3K is a specific class of kinases that catalyze the synthesis of phosphatidylinositol lipids (33). Activated PI3K activates its downstream target AKT via a second messenger, phosphatidylinositol 3,4,5-triphosphate [PI(3,4,5)P3] (34). Previous studies have demonstrated that inhibition of PI3K inhibits AKT activation, subsequently downregulating the expression of key regulatory factors of the cytoskeleton by suppressing adhesion, thereby inhibiting EMT (13,35,36). Wang et al (37) reported that inhibition of AKT activation inhibited ovarian cancer cell proliferation and invasion. In another study, Nakanishi et al (38) detected AKT phosphorylation in serum-free cultured Li7 cells, which is a liver cancer cell line, suggesting that AKT activation is associated with intrahepatic hematogenous metastasis. Treatment with a PI3K/AKT blocker LY294002 in the implanted model inhibited intrahepatic metastasis, demonstrating that PI3K/AKT serves an important role in metastasis (38). Lai et al (39) previously confirmed that BD inhibited the PI3K/AKT signaling pathway in pancreatic cancer cells. Similarly, the present study revealed that BD treatment reduced PI3K expression and AKT activation in a concentration-dependent manner, suggesting that BD inhibits the PI3K/AKT signaling pathway in MDA-MB-231 cells, which can serve as the underlying mechanism behind the anti-invasion and anti-migratory effects of BD.

However, it should be noted that the present study only explored the inhibitory effects of BD on the PI3K/AKT signaling pathway. Whether this inhibition was mediated by BD directly and if this leads to the suppression of EMT remains unclear, since multiple signaling pathways are likely to be involved in the regulation of EMT. In addition, whether BD directly regulates the expression of genes downstream of AKT was not been investigated in the present study. Since only one cell line was used, whether BD exerts similar anti-tumor effects on other TNBC cell lines remains to be determined. The present study was conducted in vitro, in vivo studies are required to investigate whether BD exerts similar anti-migratory and anti-invasive effects on TNBC xenografts or human samples. Treatment with 1–4 µM BD exerted anticancer effects in vitro, but whether it can safely reach the amount used in vivo will also need to be studied in the future. Therefore, addressing these questions will be the subject for further research.

In conclusion, the present study demonstrated that BD inhibited MDA-MB-231 cell viability migration and invasion whilst suppressing EMT and PI3K/AKT signaling activation. These results highlighted the use of BD as a therapeutic agent for the treatment of TNBC.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81241102).

Availability of data and materials

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

Authors' contributions

ZJ and CL designed the study. CL was a major contributor in writing the manuscript. CL and YW performed cell culture and MTT assay experiments. CL and CW performed the wound healing and Transwell assay. CL and YC performed western blotting. 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.

Glossary

Abbreviations

Abbreviations:

BD

Bruceine D

EMT

epithelial-mesenchymal transformation

TNBC

triple-negative breast cancer

References

1 

Nakhjavani M, Hardingham JE, Palethorpe HM, Price TJ and Townsend AR: Druggable molecular targets for the treatment of triple negative breast cancer. J Breast Cancer. 22:341–361. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Malla RR, Kumari S, Gavara MM, Badana AK, Gugalavath S, Kumar DK and Rokkam P: A perspective on the diagnostics, prognostics, and therapeutics of microRNAs of triple-negative breast cancer. Biophys Rev. 11:227–234. 2019. View Article : Google Scholar : PubMed/NCBI

3 

He MY, Rancoule C, Rehailia-Blanchard A, Espenel S, Trone JC, Bernichon E, Guillaume E, Vallard A and Magne N: Radiotherapy in triple-negative breast cancer: Current situation and upcoming strategies. Crit Rev Oncol Hematol. 131:96–101. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Bianchini G, Balko JM, Mayer IA, Sanders ME and Gianni L: Triple-negative breast cancer: Challenges and opportunities of a heterogeneous disease. Nat Rev Clin Oncol. 13:674–690. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Yan Z, Guo GF and Zhang B: Research of Brucea javanica against cancer. Chin J Integr Med. 23:153–160. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Dou YX, Zhou JT, Wang TT, Huang YF, Chen VP, Xie YL, Lin ZX, Gao JS, Su ZR and Zeng HF: Self-nanoemulsifying drug delivery system of bruceine D: A new approach for anti-ulcerative colitis. Int J Nanomedicine. 13:5887–5907. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Lau ST, Lin ZX, Liao Y, Zhao M, Cheng CH and Leung PS: Bruceine D induces apoptosis in pancreatic adenocarcinoma cell line PANC-1 through the activation of p38-mitogen activated protein kinase. Cancer Lett. 281:42–52. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Lau ST, Lin ZX and Leung PS: Role of reactive oxygen species in brucein D-mediated p38-mitogen-activated protein kinase and nuclear factor-kappaB signalling pathways in human pancreatic adenocarcinoma cells. Br J Cancer. 102:583–593. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Liu L, Lin ZX, Leung PS, Chen LH, Zhao M and Liang J: Involvement of the mitochondrial pathway in bruceine D-induced apoptosis in Capan-2 human pancreatic adenocarcinoma cells. Int J Mol Med. 30:93–99. 2012.PubMed/NCBI

10 

Xiao Z, Ching Chow S, Han Li C, Chun Tang S, Tsui SK, Lin Z and Chen Y: Role of microRNA-95 in the anticancer activity of Brucein D in hepatocellular carcinoma. Eur J Pharmacol. 728:141–150. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Irani S: Emerging insights into the biology of metastasis: A review article. Iran J Basic Med Sci. 22:833–847. 2019.PubMed/NCBI

12 

Lamouille S, Xu J and Derynck R: Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 15:178–196. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Wang H, Duan L, Zou Z, Li H, Yuan S, Chen X, Zhang Y, Li X, Sun H, Zha H, et al: Activation of the PI3K/Akt/mTOR/p70S6K pathway is involved in S100A4-induced viability and migration in colorectal cancer cells. Int J Med Sci. 11:841–849. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Li Z, Xu X, Li Y, Zou K, Zhang Z, Xu X, Liao Y, Zhao X, Jiang W, Yu W, et al: Synergistic antitumor effect of BKM120 with prima-1met via inhibiting PI3K/AKT/mTOR and CPSF4/hTERT signaling and reactivating mutant P53. Cell Physiol Biochem. 45:1772–1786. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Mao G, Tian Y, Sun Z, Ou J and Xu H: Bruceine D isolated from Brucea javanica (L.) merr. As a systemic feeding deterrent for three major lepidopteran pests. J Agric Food Chem. 67:4232–4239. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Cheng Z, Yuan X, Qu Y, Li X, Wu G, Li C, Zu X, Yang N, Ke X, Zhou J, et al: Bruceine D inhibits hepatocellular carcinoma growth by targeting β-catenin/jagged1 pathways. Cancer Lett. 403:195–205. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Yap AS, Crampton MS and Hardin J: Making and breaking contacts: The cellular biology of cadherin regulation. Curr Opin Cell Biol. 19:508–514. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Wells A, Yates C and Shepard CR: E-cadherin as an indicator of mesenchymal to epithelial reverting transitions during the metastatic seeding of disseminated carcinomas. Clin Exp Metastasis. 25:621–628. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Loh CY, Chai JY, Tang TF, Wong WF, Sethi G, Shanmugam MK, Chong PP and Looi CY: The E-cadherin and N-cadherin switch in epithelial-to-mesenchymal transition: Signaling, therapeutic implications, and challenges. Cells. 8:E1182019. View Article : Google Scholar : PubMed/NCBI

20 

Li B, Shi H, Wang F, Hong D, Lv W, Xie X and Cheng X: Expression of E-, P- and N-cadherin and its clinical significance in cervical squamous cell carcinoma and precancerous lesions. PLoS One. 11:e01559102016. View Article : Google Scholar : PubMed/NCBI

21 

Berx G and Van Roy F: The E-cadherin/catenin complex: An important gatekeeper in breast cancer tumorigenesis and malignant progression. Breast Cancer Res. 3:289–293. 2001. View Article : Google Scholar : PubMed/NCBI

22 

Debies MT and Welch DR: Genetic basis of human breast cancer metastasis. J Mammary Gland Biol Neoplasia. 6:441–451. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Wilmanns C, Grossmann J, Steinhauer S, Manthey G, Weinhold B, Schmitt-Graff A and von Specht BU: Soluble serum E-cadherin as a marker of tumour progression in colorectal cancer patients. Clin Exp Metastasis. 21:75–78. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Sloan EK and Anderson RL: Genes involved in breast cancer metastasis to bone. Cell Mol Life Sci. 59:1491–1502. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Scanlon CS, Van Tubergen EA, Inglehart RC and D'Silva NJ: Biomarkers of epithelial-mesenchymal transition in squamous cell carcinoma. J Dent Res. 92:114–121. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Tanaka K, Tokunaga E, Inoue Y, Yamashita N, Saeki H, Okano S, Kitao H, Oki E, Oda Y and Maehara Y: Impact of expression of vimentin and axl in breast cancer. Clin Breast Cancer. 16:520–526.e2. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Lu R, Zhou Z, Yu W, Xia Y and Zhi X: CPEB4 promotes cell migration and invasion via upregulating vimentin expression in breast cancer. Biochem Biophys Res Commun. 489:135–141. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Zelenko Z, Gallagher EJ, Tobin-Hess A, Belardi V, Rostoker R, Blank J, Dina Y and LeRoith D: Silencing vimentin expression decreases pulmonary metastases in a pre-diabetic mouse model of mammary tumor progression. Oncogene. 36:1394–1403. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Conacci-Sorrell M, Zhurinsky J and Ben-Ze'ev A: The cadherin-catenin adhesion system in signaling and cancer. J Clin Invest. 109:987–991. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Ren L, Chen H, Song J, Chen X, Lin C, Zhang X, Hou N, Pan J, Zhou Z, Wang L, et al: MiR-454-3p-mediated Wnt/β-catenin signaling antagonists suppression promotes breast cancer metastasis. Theranostics. 9:449–465. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Yan X, Lyu T, Jia N, Yu Y, Hua K and Feng W: Huaier aqueous extract inhibits ovarian cancer cell motility via the AKT/GSK3β/β-catenin pathway. PLoS One. 8:e637312013. View Article : Google Scholar : PubMed/NCBI

32 

Liu Y, Liu C, Tan T, Li S, Tang S and Chen X: Sinomenine sensitizes human gastric cancer cells to cisplatin through negative regulation of PI3K/AKT/Wnt signaling pathway. Anticancer Drugs. 30:983–990. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Elmenier FM, Lasheen DS and Abouzid KA: Phosphatidylinositol 3 kinase (PI3K) inhibitors as new weapon to combat cancer. Eur J Med Chem. 183:1117182019. View Article : Google Scholar : PubMed/NCBI

34 

Luo J, Manning BD and Cantley LC: Targeting the PI3K-Akt pathway in human cancer: Rationale and promise. Cancer Cell. 4:257–262. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Larue L and Bellacosa A: Epithelial-mesenchymal transition in development and cancer: Role of phosphatidylinositol 3′ kinase/AKT pathways. Oncogene. 24:7443–7454. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Ning J, Liu W, Zhang J, Lang Y and Xu S: Ran GTPase induces EMT and enhances invasion in non-small cell lung cancer cells through activation of PI3K-AKT pathway. Oncol Res. 21:67–72. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Wang J, Xiao L, Luo CH, Zhou H, Hu J, Tang YX, Fang KN and Zhang Y: Overexpression of TRPM7 is associated with poor prognosis in human ovarian carcinoma. Asian Pac J Cancer Prev. 15:3955–3958. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Nakanishi K, Sakamoto M, Yasuda J, Takamura M, Fujita N, Tsuruo T, Todo S and Hirohashi S: Critical involvement of the phosphatidylinositol 3-kinase/Akt pathway in anchorage-independent growth and hematogeneous intrahepatic metastasis of liver cancer. Cancer Res. 62:2971–2975. 2002.PubMed/NCBI

39 

Lai ZQ, Ip SP, Liao HJ, Lu Z, Xie JH, Su ZR, Chen YL, Xian YF, Leung PS and Lin ZX: Brucein D, a naturally occurring tetracyclic triterpene quassinoid, induces apoptosis in pancreatic cancer through ROS-associated PI3K/Akt signaling pathway. Front Pharmacol. 8:9362017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2020
Volume 19 Issue 1

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
Luo C, Wang Y, Wei C, Chen Y and Ji Z: The anti‑migration and anti‑invasion effects of Bruceine D in human triple‑negative breast cancer MDA‑MB‑231 cells. Exp Ther Med 19: 273-279, 2020
APA
Luo, C., Wang, Y., Wei, C., Chen, Y., & Ji, Z. (2020). The anti‑migration and anti‑invasion effects of Bruceine D in human triple‑negative breast cancer MDA‑MB‑231 cells. Experimental and Therapeutic Medicine, 19, 273-279. https://doi.org/10.3892/etm.2019.8187
MLA
Luo, C., Wang, Y., Wei, C., Chen, Y., Ji, Z."The anti‑migration and anti‑invasion effects of Bruceine D in human triple‑negative breast cancer MDA‑MB‑231 cells". Experimental and Therapeutic Medicine 19.1 (2020): 273-279.
Chicago
Luo, C., Wang, Y., Wei, C., Chen, Y., Ji, Z."The anti‑migration and anti‑invasion effects of Bruceine D in human triple‑negative breast cancer MDA‑MB‑231 cells". Experimental and Therapeutic Medicine 19, no. 1 (2020): 273-279. https://doi.org/10.3892/etm.2019.8187