Lunasin suppresses the migration and invasion of breast cancer cells by inhibiting matrix metalloproteinase-2/-9 via the FAK/Akt/ERK and NF-κB signaling pathways

  • Authors:
    • Qianqian Jiang
    • Yu Pan
    • Yupeng Cheng
    • Huiling Li
    • Dandan Liu
    • Hui Li
  • View Affiliations

  • Published online on: May 9, 2016     https://doi.org/10.3892/or.2016.4798
  • Pages: 253-262
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lunasin is a naturally existing bioactive peptide with an Arg-Gly-Asp (RGD) motif, which competes with integrins to bind with the extracellular matrix (ECM) consequently suppressing the integrin-mediated signaling pathway. Owing to the RGD motif, lunasin has been proven as an effective anti-inflammatory, antitumor and antimetastatic agent in many types of cancer. However, knowledge of its inhibitory effect on metastasis and the related mechanism of action in breast cancer cells is limited. In this study, the inhibitory effect of lunasin on the proliferation, migration and invasion of two typical breast cancer cell lines, ER-negative MDA-MB-231 with αVβ3 expression and ER-positive MCF-7 with αVβ5/α5β1 expression, were examined in vitro as well the related mechanisms. The results demonstrated that lunasin (10-20 µM) effectively inhibited the migration and invasion activity and expression of matrix metalloproteinase (MMP)‑2/-9 in both breast cancer cell lines. Meanwhile, we also found that lunasin inhibited the phosphorylation of focal adhesion kinase (FAK), Src, Akt, ERK and nucleus translocation of NF-κB, which indicates that, possibly via competing with αVβ3 or αVβ5/α5β1 integrin, lunasin suppresses the metastasis of breast cancer cells through integrin-mediated FAK/Akt/ERK and NF-κB signaling pathways followed by downregulation of the activity and expression of MMP-2/-9.

Introduction

Breast cancer is the most frequently diagnosed cancer and the leading cause of cancer-related death among females worldwide (1). In most cases, it is the metastasis of malignant tumor cells, rather than the primary solid tumor, that is the main cause of death (2). Metastasis is a complex process involving cell adhesion, migration, invasion and extracellular matrix (ECM) degradation. Hence, it is urgent to develop therapeutic strategies and agents with the ability to inhibit growth, migration, invasion and proteolytic degradation of ECM in breast cancer cells.

Integrin is a family of ubiquitous transmembrane glycoprotein receptors comprised of 18 α and 8 β subunits with different combinations and ligand specificity (3). They link the ECM to the intracellular actin cytoskeleton and stimulate the intracellular signaling pathway that modulates proliferation, migration and invasion of cells. Compared with normal cells, metastatic tumor cells often overexpress certain integrins. For example, αVβ3, αVβ5 and α5β1, a subfamily of integrins, have been demonstrated to be upregulated in lung, melanoma, breast and brain tumors, which make them ideal pharmacological targets for the development of antitumorigenic and antiangiogenic compounds (4,5). Recently, a tripeptide sequence Arg-Gly-Asp (RGD) which is a conserved motif present in ECM proteins, has been identified as an important tool in targeting drugs and imaging agents because of its high affinity to αVβ3, αVβ5 and α5β1 integrins. Therefore, RGD has been investigated as an ideal promising ligand for the development of antitumor or antimetastatic agents.

Many natural peptides containing the RGD motif have been examined as effective agents with antitumor and anti-metastatic activity (6). Lunasin is such a novel 43-amino acid peptide originally isolated from soybean seed with the sequence: SKWQHQQDSCRKQLQGVNLTPCEKHIMEKIQGRGDDDDDDDDD (7). To date, lunasin and its analogues have been successively identified in barley (8), wheat (9) and other plants, e.g. Amaranth, a plant cultivated in Mexico (10), and a traditional Chinese herb, Solanum nigrum L. (11). There is accumulating evidence showing that the bioactivity of lunasin is highly associated with its unique peptide sequences consisting of three functional regions: a C-terminal tail with 8 aspartic acid residues (poly-D), helping lunasin bind directly to histones affecting the H3 and H4 acelylation/deacelylation process (12); a predicted and structurally conserved helix region targeting lunasin to the chromatin. Moreover, as a cell adhesion motif, RGD potentiates the ability of lunasin to internalize into cells and compete with ECM to interact with integrins, such as αVβ3, αVβ5 or α5β1, which is crucial in angiogenesis, tumor progression and metastasis. It was found that lunasin inhibited cell proliferation and induced apoptosis in breast, colon and leukemia cancers owing to its RGD motif (13). As reported, lunasin inhibited the metastasis of colon cancer cells by direct binding with α5β1 integrin through suppression of focal adhesion kinase (FAK)/ERK/NF-κB signaling in vitro and in a mouse model (14). Lunasin has also been reported to exert anti-inflammatory effects on human macrophages via inhibition of the αVβ3 integrin-mediated Akt/NF-κB pathway (15). Additionally, when combined with other chemopreventive agents anacardic acid or oxaliplatin, lunasin potentiated their antitumor effects on MDA-MB-231 (16), and KM12L4 (14) cells, respectively, indicating that this peptide has a promising role as a co-adjuvant in cancer therapy.

Although research has proven that lunasin is an effective bioactive peptide in many cancer therapies, the effect and mechanism of lunasin in regards to the metastasis of breast cancer cells are largely unknown. In this study, we assessed the potential inhibitory effects of lunasin on the growth, migration, invasion and ECM degradation of breast cancer cells. We also verified that lunasin exerts its antimetastatic effect by decreasing the activity and expression of MMP-2/-9 and inactivating the associated proteins in the FAK/Akt/ERK and NF-κB pathways.

Materials and methods

Cell lines

Human breast cancer cell lines MCF-7 and MDA-MB-231 were purchased from the Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (Shanghai, China). MCF-7 cells were cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco, Grand Island, NY, USA) supplemented with 10% FBS (Biological Industries, Kibbutz Beit-Haemek, Israel), 100 mg/ml streptomycin and 100 U/ml penicillin in a 5% CO2 atmosphere at 37°C. MDA-MB-231 cells were cultured in L15 (Sigma-Aldrich, St. Louis, MO, USA) supplemented with 10% FBS, 100 mg/ml streptomycin and 100 U/ml penicillin (Beyotime Institute of Biotechnology, Shanghai, China) at 37°C.

Reagents

Lunasin, SKWQHQQDSCRKQLQGVNLTPCEKHIMEIGRDDDDDDDDD was synthesized by GL Biochem (Shanghai, China). The purity of the peptide was higher than 95%. Stock solutions of synthetic lunasin (1 mM) were prepared with sterile distilled water and stored at −20°C.

3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and PMSF were obtained from Sigma-Aldrich. BCA protein assay kit, RIPA lysis buffer and nuclear/cytoplasmic protein extraction kit were purchased from Beyotime Institute of Technology. The primary antibodies to phospho-Akt (Ser473) rabbit mAb, Akt rabbit mAb, phospho-Src family (Tyr416) rabbit mAb, Src rabbit mAb, phospho-FAK (Tyr397) rabbit mAb, FAK rabbit mAb, phospho-p44/42 MAPK (Erk1/2) (Thr202/Tyr204) rabbit mAb, p44/42 MAPK (Erk1/2) rabbit mAb, phospho-NF-κB p65 (Ser536) rabbit mAb, IκBα mouse mAb, phospho-IκBα (Ser32) rabbit mAb, NF-κB p65 rabbit mAb, β-actin mouse mAb, Lamin B2 (D8P3U) rabbit mAb, anti-rabbit IgG, HRP-linked antibody, anti-mouse IgG and HRP-linked antibody were all purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA).

Cell viability assay

Cell viability was assessed using an MTT assay. MCF-7 and MDA-MB-231 cells were plated at a density of 1×104 cells/well in a 96-well plate overnight and then treated with various concentrations of lunasin peptide (0–320 µM) for 24 or 48 h. At the end of the treatment, 20 µl of MTT (5 mg/ml) was added and incubated at 37°C for 4 h. The supernatant was aspirated and the formazan crystals that formed were dissolved in 150 µl DMSO for 20 min. The absorbance at 490 nm was measured with a microplate reader (Spectra Max 190; Molecular Devices, LLC, Sunnyvale, CA, USA). The viability was expressed as the percentage of the lunasin-treated group to the control group, considered as 100%. All data were analyzed from three independent experiments with six replicates and the results are expressed as the mean ± SD.

Wound healing assay

The wound healing assay was performed as previously reported with some modifications (17). MCF-7 and MDA-MB-231 cells were seeded into a 6-well plate until growth to 70% confluence with complete medium. A plastic tip (1 mm) was used to make a scratch on the cell monolayer as previously described (18). Then the wound area was washed three times with PBS to remove cell debris and the cells were incubated with lunasin (0–20 µM) for 24 h. The cells were allowed to migrate into the wound surface and the average distance of the migrating cells was observed using inverted microscopy (Leica Microsystems GmbH, Wetzlar, Germany) at different times. The migration rate was expressed as the migrated distance of the cells in the experimental group to that of the control group. All data were analyzed from three independent experiments performed in triplicate and the results are expressed as the mean ± SD.

Invasion assay

The invasion assay was performed using a Transwell chamber (8 µm pore polycarbonate, Corning Costar, Cambridge, MA, USA) coated with the diluted BD Matrigel basement membrane matrix (BD Biosciences, Bedford, MA, USA). MCF-7 and MDA-MB-231 cells treated with lunasin (0–20 µM) for 24 h were trypsinized and suspended at a final concentration of 1×106 cells/ml in serum-free DMEM and L15 medium, respectively. Cell suspension was added into each Transwell upper chamber of and the medium with 5% FBS was applied to the bottom of the chamber as a chemoattractant. The chamber was incubated at 37°C for 24 h. After incubation, the non-invaded cells in the upper chamber were removed from the Transwell membrane with a cotton swab. The invaded cells in the lower chamber were fixed with 100% methanol and then stained with 1% crystal violet in 2% ethanol. The invaded cells were counted and photographed under a microscope at five different fields of the chamber. The data are presented as the average number of invaded cells in the experimental group to the control group. All data were analyzed from three independent experiments performed in triplicate and the results are expressed as the mean ± SD.

Gelatin zymography

The proteolytic activity of MMP-2 and MMP-9 in the supernatant was analyzed by gelatin zymography assay as previously described (19). MCF-7 and MDA-MB-231 cells were treated with lunasin (0–20 µM) in serum-free medium for 24 h. After incubation, the supernatant of the cells was collected and centrifuged at 1,000 × g at 4°C for 10 min. Samples were then loaded on 8% SDS-polyacrylamide gel containing 0.1% gelatin. The electrophoresis was performed at 100 V. When finished, the gel was washed with elution buffer (2.5% Triton X-100, 50 mM Tris-HCl, 5 mM CaCl2 and 1 µM ZnCl2, pH 7.6) and washing buffer (50 mM Tris-HCl, 5 mM CaCl2 and 1 µM ZnCl2, pH 7.6) twice, followed by reaction with incubation solution (50 mM Tris-HCl, 5 mM CaCl2, 1 µM ZnCl2 and 0.02% Brij-35, pH 7.6) for 48 h at 37°C. Finally, the gel was stained with staining buffer (0.05% Coomassie Blue R-250, 30% methanol and 10% acetic acid) and destained with destaining buffer A (30% methanol and 10% acetic acid), B (20% methanol and 10% acetic acid) and C (10% methanol and 50% acetic acid) by turn. The results were photographed and analyzed by Biospectrum Imaging system (UVP, Inc., Upland, CA, USA) and Image J software. The data are presented as the degree of grey in the enzymatic region in the lunasin-treated group vs. the control group. All data were analyzed from three independent experiments performed in triplicate and the results are expressed as the mean ± SD.

Western blot analysis

MCF-7 and MDA-MB-231 cells were cultured in a 6-well plate at a density of 2×105 cells/well. After incubation, the cells were pretreated with different concentrations of lunasin (0–20 µM) for 24 h. Then, the cells were harvested and lysed with the RIPA cell lysis buffer in the presence of a protease inhibitor PMSF. The samples were incubated for 30 min on ice and centrifuged at 8,000 × g for 15 min at 4°C. The cell extracts were collected and stored at −80°C until use in subsequent experiments. Total cellular and nuclear proteins were extracted according to the instructions of the nuclear and cytoplasmic protein extraction kit. The nuclear extracts were used to determine NF-κB protein levels and the cytoplasmic extracts were used to determine IκB levels. The protein concentration was determined using the BCA protein assay kit. Equal amounts of protein were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to a polyvinylidene fluoride (PVDF) membrane. Western blot analysis was carried out as previously described (20). The protein bands were visualized by enhanced chemiluminescence detection reagents (Applygen Technologies Inc., Beijing, China) using a Biospectrum Imaging system.

Statistical analysis

All data are presented as the mean ± standard deviation (SD) of 3 independent experiments performed in triplicate. Statistical analysis was performed by Student's t-test or one-way analysis of variance (ANOVA). In all cases, P<0.05 was considered statistically significant, P<0.01 was considered extremely significant.

Results

Cytotoxic effect of lunasin on MCF-7 and MDA-MB-231 breast cancer cells

To evaluate the cytotoxic effect of lunasin on human breast cancer MCF-7 and MDA-MB-231 cells, the viability was determined by MTT assay. As shown in Fig. 1A and B, lunasin did not exhibit strong cytotoxic effects on MCF-7 and MDA-MB-231 cells until the concentration reached 20 µM. With the increase in lunasin concentration and incubation time, the viability of the cells was markedly decreased, which demonstrated that lunasin may exert its inhibitory effect in concentration- and time-dependent manners. After treatment with lunasin for 24 and 48 h, the IC50 values in the MCF-7 cells were 508.6 µM and 431.9 µM, respectively. The cell viability of the MDA-MB-231 cells following treatment with lunasin (Fig. 1B) was much lower than that of the MCF-7 cells. The IC50 was 224.7 µM at 24 h and 194.9 µM at 48 h. The cytotoxic effect of lunasin was also investigated in human normal breast MCF-10A cells (Fig. 1C). As previosly reported (21), lunasin selectively kills cancer cells without having an effect on normal cells.

Lunasin inhibits the migration and motility of breast cancer cells in vitro

A scratch wound assay was carried out to evaluate the migration and motility of the breast cancer cells following treatment with lunasin. According to the MTT assay results, 0–20 µM lunasin was selected for its non-cytotoxicity to cells. As shown in Fig. 2A and B, lunasin effectively inhibited the migration of MCF-7 cells after a 24-h treatment. Compared with the control group, the migration rate in the lunasin group decreased gradually with an increase in lunasin concentration. The same phenomenon was also observed in the MDA-MB-231 cells following treatment with lunasin (Fig. 2C and D). All the results indicated that lunasin significantly suppressed the migration of MCF-7 and MDA-MB-231 breast cancer cells in a dose-dependent manner.

Lunasin inhibits the invasion of breast cancer cells

The inhibitory effects of lunasin on the invasion of breast cancer cells were investigated by Transwell invasion assay. Fig. 3 shows that after incubation for 24 h, lunasin (10–20 µM) markedly decreased the number of invasive cells in both the MCF-7 and MDA-MB-231 cells. The invasiveness of the MCF-7 and MDA-MB-231 cells became less aggressive with an increase in lunasin concentration. These findings were consistent with the wound scratch assay, which indicated that lunasin suppresses the metastasis of breast cancer cells.

Lunasin reduces the activity and expression of MMP-2/-9 in breast cancer cells

Matrix metalloproteinases (MMPs) are a family of proteins that can degrade the ECM leading to tumor metastasis, apoptosis and carcinogensis (22). Among the MMP proteins, MMP-2 and MMP-9 are highly expressed and correlated with the metastasis of breast tumors (23). Hence, in this study the activity and expression of MMP-2 and MMP-9 were investigated by gelatin zymography and western blot analysis to examine the possible anti-invasive ability of lunasin. As shown in Fig. 4A and B, lunasin (10–20 µM) reduced the activity and expression of MMP-9 in the MCF-7 cells. However, there were no significant changes in the activity and expression of MMP-2. In the MDA-MB-231 cells (Fig. 4C and D), lunasin sharply reduced the activity and expression of MMP-9 and decreased that of MMP-2 gradually. The differences in MMP-2 expression may be attributed to the different characteristics between the two breast cancer cell lines.

Lunasin suppresses the phosphorylation of FAK and Src

FAK, a cytoplasmic protein tyrosine kinase, is crucial for proliferation, migration, adhesion and invasion of cancer cells (24). It has been reported that FAK is activated by integrin-clustering and autophosphorylated at Tyr397 (25). Once phosphorylated, FAK supplies a binding site for Src to form the FAK-Src complex, which recruits more signaling molecules participating in integrin-mediated signaling transduction (26). Therefore, we assessed the expression of phosphorylated FAK and Src in breast cancer cells after treatment with lunasin for 24 h. As shown in Fig. 5A and B, lunasin strongly inhibited the phosphorylation of FAK and Src in both MCF-7 and MDA-MB-231 cell lines. It did not have significant effects on total FAK and Src, which demonstrated that lunasin exerted its antimetastatic effect by preventing the phosphorylation of FAK and Src from forming the FAK-Src complex.

Lunasin suppresses the phosphorylation of ERK and Akt

Studies have found that the Ras/MEK/ERK and the PI3K/Akt pathways, the downstream signaling molecules activated by the FAK-Src complex, are correlated to the survival and metastasis of cancer cells (27). Hence, the phosphorylation of key molecules in these pathways, Akt and ERK, were examined further by western blot analysis. As shown in Fig. 6, the phosphorylation of Akt and ERK was attenuated by lunasin (10–20 µM) in a concentration-dependent manner. There were no significant differences in the total amount of Akt and ERK. The results illustrated that lunasin may inhibit the metastasis of breast cancer cells by blocking the PI3K/Akt/ERK pathway.

Lunasin inactivates the translocation process of NF-κB into the nucleus

NF-κB, an important transcriptional factor, regulates the expression of many genes involved in mammary carcinogenesis, survival and metastasis of breast cancer cells (28). The activation of NF-κB is controlled by the targeted phosphorylation and subsequent degradation of IκB, which prevents NF-κB from translocation into the nucleus. To further explore the underlying mechanisms of lunasin, the expression of the NF-κB inhibitor, IκBα and p-IκBα in the cytoplasm, the translocation of NF-κB, p65 and p-p65 in the nucleus, were all examined in our study using western blot analysis. The results showed that the phosphorylation and degradation of IκBα were markedly inhibited by lunasin (10–20 µM) with enhanced IκB expression (Fig. 7). Accordingly, the translocation of activated NF-κB, p-p65 and p65 in the nucleus, was decreased following treatment with lunasin.

Discussion

Metastasis is a series of complex processes consisting of cancer cell proliferation, cell motility, cell adhesion to ECM and ECM proteolysis. It has been reported that 90% of breast cancer deaths are attributed to the metastasis of cancer cells. Therefore, it is urgent to develop new therapeutic agents to inhibit the metastasis of breast cancer. Lunasin, a 43-amino acid peptide derived from soybeans, is a bioactive peptide which has been demonstrated as an effective chemopreventive and antitumor agent in cancer therapy. However, knowledge of the inhibitory effects of lunasin on the metastasis of breast cancer cells and its underlying mechanism is limited. In this study, we demonstrated that lunasin suppressed the proliferation, migration and invasion of breast cancer cells. Meanwhile, the activity and expression of MMP-2/-9, phosphorylation of FAK, Src, Akt, ERK, p65, IκBα and translocation of p65 were decreased, which indicated that lunasin inhibited the metastasis of breast cancer cells via suppression of the FAK/Akt/ERK and NF-κB pathways.

In this present study, we found that lunasin suppressed the proliferation of MCF-7 and MDA-MB-231 breast cancer cells with IC50 values of 508.6 µM and 224.7 µM, respectively. These values were slightly higher than those of previous research (29), in which the IC50 value of MDA-MB-231 was 181 µM following treatment by synthetic lunasin. In order to examine the antimetastatic potential of the peptide, 10–20 µM lunasin was selected because of its non-cytotoxicity to MCF-7 and MDA-MB-231 breast cancer cells. Migration and invasion are two critical processes during the metastasis of cancer cells. The migratory and invasive abilities of cancer cells enable them to migrate to adjacent tissues finally leading to metastasis. In the present study, the inhibitory effects of lunasin on migration and invasion were observed by scratch wound and Transwell assays. As shown in Figs. 2 and 3, lunasin was able to effectively block the migration and invasion of MCF-7 and MDA-MB-231 cells in a concentration-dependent manner. Furthermore, as we examined, lunasin also inhibited the proteolytic degradation of the ECM. MMPs, a family of zinc-containing endopeptidases that can degrade the ECM, play a vital role in this process (30). Among the MMPs, MMP-2 and MMP-9 have been reported as being highly expressed in aggressive breast tumors and associated with poor prognosis (31,32). In our study, it was found that lunasin strongly inhibited the activity and expression of MMP-9 in both the MCF-7 and MDA-MB-231 cell lines. Nevertheless, the decrease in MMP-2 was much more slight compared with MMP-9 in the MCF-7 and MDA-MB-231 cell lines. The results demonstrated that lunasin plays a more important role in the suppression of MMP-9 rather than MMP-2. In conclusion, the results substantiated the inhibitory ability of lunasin on the proliferation, migration, invasion and ECM degradation, which indicates that lunasin inhibits the metastasis of breast cancer cells.

Integrins, a family of heterodimer transmembrane glycoproteins, consist of 18 α and 8 β subunits to form 24 different subtypes (33). In breast cancer cells, αVβ3, αVβ5 and α5β1 integrins are often highly expressed in comparison to other subtypes, which can bind to ECM proteins such as fibronectin, vitronectin and collagen through a tripeptide motif Arg-Gly-Asp (RGD) to facilitate cell adhesion, migration, invasion and intracellular signaling (34). Hence, peptides containing the RGD motif are considered to be ideal agents to compete with ECM to interact specifically with integrins (35). Previous studies suggest that lunasin possesses an affinity for αVβ3 and α5β1 in macrophages (15) and cancer cells (3638) which enable it to prevent the activation of the integrin-mediated signaling pathway by direct blockade. In this present study, we selected two typical breast cancer cell lines, ER-positive MCF-7 cells and ER-negative MDA-MB-231 cells with expression of different integrin subtypes. Research has confirmed that in MDA-MB-231 cancer cells, αVβ3 is often highly expressed (3941), while MCF-7 cells are αVβ3 negative but with αVβ5 and α5β1 expression (42). Thus, we examined the inhibitory effect on these two breast cancer cells and aimed to ascertain the underlying mechanism involved. The differences in the inhibitory effects of lunasin on these two cell lines may be ascribed to the different integrins and ER expression on their surface.

FAK, a receptor protein-tyrosine kinase, plays a central role in the intracellular signaling of integrin. Once integrin binds to the ECM, FAK is recruited to the clustering integrins and activated by autophosphorylation. Then Src is further phosphorylated to form the FAK-Src complex, which initiates the signal transduction of cell survival, proliferation, migration and invasion. As examined in our study (Fig. 5), lunasin strongly inhibited the phosphorylation of FAK and Src in both MCF-7 and MDA-MB-231 cell lines with no changes in total FAK and Src proteins. The results were consistent with the study conducted by Dia et al (14) in which it was reported, that lunasin suppresses the metastasis of colon cancer cells through inhibition of the FAK/Src pathway.

Previous studies have shown that decreased levels of MMP-9 and MMP-2, in breast cancer cells, are highly related to the inhibition of PI3K/Akt and ERK, both of which are downstream targets of FAK (43,44). PI3K, a lipid kinase, participates in multiple cell signaling pathways through the activation of Akt. Additionally, activated Akt can lead to the invasion and metastasis of cancer cells by stimulating the secretion of MMPs (45,46). It is accepted that the MAP family kinases possibly take part in signaling processes that modulate MMPs, including MMP-9 (47). In order to gain a better understanding of the mechanism by which lunasin inhibits tumor metastasis, the expression of Akt and ERK was examined by western blotting (Fig. 6). The results confirmed that lunasin markedly blocked the phosphorylation of Akt and ERK which may stop the cellular signaling to transcription factors of MMPs.

Moreover, NF-κB is considered to regulate the survival, proliferation, chemoresistance, angiogenesis, cellular invasion and migration of cancer cells. Previous studies have reported that NF-κB may be the transcriptional regulatory factor of MMPs and other genes, while IκB is an inhibitor which prevents NF-κB translocation into the nucleus (48). It was reported that PI3K activates IκB kinases (IKKs) to phosphorylate IκB, leading to its ubiquitination and proteasomal degradation to release the NF-κB/Rel complex. Lunasin has been shown to suppress the FAK/ERK/NF-κB signaling pathway in colon cancer cells (14) and Akt/NF-κB pathway in macrophages during activation of lipopolysaccharide-induced inflammation (49). In our study, we also found that lunasin inhibited the metastasis of breast cancer cells through preventing IκB phosphorylated and enhancing IκB protein expression, which decreased the phosphorylation and translocation of NF-κB.

In summary, lunasin inhibited the cell proliferation, cell migration, cell invasion and the activity and expression of MMP-2 and MMP-9 in breast cancer cells. We proposed that lunasin possibly exerted its inhibitory effect via the suppression of the integrin-mediated FAK/Akt/ERK and NF-κB signaling pathways. These results provide a foundation for future investigation of lunasin as an effective antitumor and antimetastatic agent for breast cancer therapy.

Acknowledgments

This study was supported by the National Natural Science Foundation of China (nos. 8147929 and 81573539), the Natural Science Foundation of Heilongjiang Province (no. H2015042), the Excellent Creative Talents Support Project of Heilongjiang University of Chinese Medicine (no. 2012RCQ12), the Research Foundation of Heilongjiang University of Chinese Medicine (no. 201004), and the Natural Science Foundation of Heilongjiang Province (no. H2015042).

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Weigelt B, Peterse JL and van't Veer LJ: Breast cancer metastasis: Markers and models. Nat Rev Cancer. 5:591–602. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Danen EH: Integrins: regulators of tissue function and cancer progression. Curr Pharm Des. 11:881–891. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Eliceiri BP and Cheresh DA: Adhesion events in angiogenesis. Curr Opin Cell Biol. 13:563–568. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Ruoslahti E: Specialization of tumour vasculature. Nat Rev Cancer. 2:83–90. 2002. View Article : Google Scholar

6 

Temming K, Schiffelers RM, Molema G and Kok RJ: RGD-based strategies for selective delivery of therapeutics and imaging agents to the tumour vasculature. Drug Resist Updat. 8:381–402. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Galvez AF and de Lumen BO: A soybean cDNA encoding a chromatin-binding peptide inhibits mitosis of mammalian cells. Nat Biotechnol. 17:495–500. 1999. View Article : Google Scholar : PubMed/NCBI

8 

Jeong HJ, Lam Y and de Lumen BO: Barley lunasin suppresses ras-induced colony formation and inhibits core histone acetylation in mammalian cells. J Agric Food Chem. 50:5903–5908. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Jeong HJ, Jeong JB, Kim DS, Park JH, Lee JB, Kweon DH, Chung GY, Seo EW and de Lumen BO: The cancer preventive peptide lunasin from wheat inhibits core histone acetylation. Cancer Lett. 255:42–48. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Silva-Sánchez C, de la Rosa AP, León-Galván MF, de Lumen BO, de León-Rodríguez A and de Mejía EG: Bioactive peptides in amaranth (Amaranthus hypochondriacus) seed. J Agric Food Chem. 56:1233–1240. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Jeong JB, Jeong HJ, Park JH, Lee SH, Lee JR, Lee HK, Chung GY, Choi JD and de Lumen BO: Cancer-preventive peptide lunasin from Solanum nigrum L. inhibits acetylation of core histones H3 and H4 and phosphorylation of retinoblastoma protein (Rb). J Agric Food Chem. 55:10707–10713. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Hernández-Ledesma B, Hsieh CC and de Lumen BO: Relationship between lunasin's sequence and its inhibitory activity of histones H3 and H4 acetylation. Mol Nutr Food Res. 55:989–998. 2011. View Article : Google Scholar : PubMed/NCBI

13 

de Lumen BO: Lunasin: a cancer-preventive soy peptide. Nutr Rev. 63:16–21. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Dia VP and Gonzalez de Mejia E: Lunasin potentiates the effect of oxaliplatin preventing outgrowth of colon cancer metastasis, binds to α5β1 integrin and suppresses FAK/ERK/NF-κB signaling. Cancer Lett. 313:167–180. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Cam A and de Mejia EG: RGD-peptide lunasin inhibits Akt-mediated NF-κB activation in human macrophages through interaction with the αVβ3 integrin. Mol Nutr Food Res. 56:1569–1581. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Hsieh CC, Hernandez-Ledesma B and de Lumen B: Cell proliferation inhibitory and apoptosis-inducing properties of anacardic acid and lunasin in human breast cancer MDA-MB-231 cells. Food Chem. 125:630–636. 2011. View Article : Google Scholar

17 

Liang CC, Park AY and Guan JL: In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc. 2:329–333. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Xu L and Deng X: Protein kinase Ciota promotes nicotine-induced migration and invasion of cancer cells via phosphorylation of micro- and m-calpains. J Biol Chem. 281:4457–4466. 2006. View Article : Google Scholar

19 

Li C, Zhao Y, Yang D, Yu Y, Guo H, Zhao Z, Zhang B and Yin X: Inhibitory effects of kaempferol on the invasion of human breast carcinoma cells by downregulating the expression and activity of matrix metalloproteinase-9. Biochem Cell Biol. 93:16–27. 2015. View Article : Google Scholar

20 

Zhou R, Xu L, Ye M, Liao M, Du H and Chen H: Formononetin inhibits migration and invasion of MDA-MB-231 and 4T1 breast cancer cells by suppressing MMP-2 and MMP-9 through PI3K/AKT signaling pathways. Horm Metab Res. 46:753–760. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Hernández-Ledesma B, Hsieh CC and de Lumen BO: Lunasin, a novel seed peptide for cancer prevention. Peptides. 30:426–430. 2009. View Article : Google Scholar

22 

McCawley LJ and Matrisian LM: Matrix metalloproteinases: multifunctional contributors to tumor progression. Mol Med Today. 6:149–156. 2000. View Article : Google Scholar : PubMed/NCBI

23 

Hanemaaijer R, Verheijen JH, Maguire TM, Visser H, Toet K, McDermott E, O'Higgins N and Duffy MJ: Increased gelatinase-A and gelatinase-B activities in malignant vs. benign breast tumors. Int J Cancer. 86:204–207. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Zhang J and Hochwald SN: The role of FAK in tumor metabolism and therapy. Pharmacol Ther. 142:154–163. 2014. View Article : Google Scholar

25 

Toutant M, Costa A, Studler JM, Kadaré G, Carnaud M and Girault JA: Alternative splicing controls the mechanisms of FAK autophosphorylation. Mol Cell Biol. 22:7731–7743. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Schlaepfer DD, Mitra SK and Ilic D: Control of motile and invasive cell phenotypes by focal adhesion kinase. Biochim Biophys Acta. 1692:77–102. 2004. View Article : Google Scholar : PubMed/NCBI

27 

van Nimwegen MJ and van de Water B: Focal adhesion kinase: a potential target in cancer therapy. Biochem Pharmacol. 73:597–609. 2007. View Article : Google Scholar

28 

Wu JT and Kral JG: The NF-kappaB/IkappaB signaling system: a molecular target in breast cancer therapy. J Surg Res. 123:158–169. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Hsieh CC, Hernández-Ledesma B and de Lumen BO: Lunasin, a novel seed peptide, sensitizes human breast cancer MDA-MB-231 cells to aspirin-arrested cell cycle and induced apoptosis. Chem Biol Interact. 186:127–134. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Deryugina EI and Quigley JP: Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Rev. 25:9–34. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Pellikainen JM, Ropponen KM, Kataja VV, Kellokoski JK, Eskelinen MJ and Kosma VM: Expression of matrix metalloproteinase (MMP)-2 and MMP-9 in breast cancer with a special reference to activator protein-2, HER2, and prognosis. Clin Cancer Res. 10:7621–7628. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Jezierska A and Motyl T: Matrix metalloproteinase-2 involvement in breast cancer progression: a mini-review. Med Sci Monit. 15:RA32–40. 2009.PubMed/NCBI

33 

Takada Y, Ye X and Simon S: The integrins. Genome Biol. 8:2152007. View Article : Google Scholar : PubMed/NCBI

34 

Cabodi S, Di Stefano P, Leal MP, Tinnirello A, Bisaro B, Morello V, Damiano L, Aramu S, Repetto D, Tornillo G, et al: Integrins and signal transduction. Adv Exp Med Biol. 674:43–54. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Meyer A, Auernheimer J, Modlinger A and Kessler H: Targeting RGD recognizing integrins: drug development, biomaterial research, tumor imaging and targeting. Curr Pharm Des. 12:2723–2747. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Dia VP and Gonzalez de Mejia E: Lunasin induces apoptosis and modifies the expression of genes associated with extracellular matrix and cell adhesion in human metastatic colon cancer cells. Mol Nutr Food Res. 55:623–634. 2011. View Article : Google Scholar : PubMed/NCBI

37 

de Mejia EG, Wang W and Dia VP: Lunasin, with an arginine-glycine-aspartic acid motif, causes apoptosis to L1210 leukemia cells by activation of caspase-3. Mol Nutr Food Res. 54:406–414. 2010. View Article : Google Scholar

38 

Inaba J, McConnell EJ and Davis KR: Lunasin sensitivity in non-small cell lung cancer cells is linked to suppression of integrin signaling and changes in histone acetylation. Int J Mol Sci. 15:23705–23724. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Meyer T, Marshall JF and Hart IR: Expression of alpha-V integrins and vitronectin receptor identity in breast cancer cells. Br J Cancer. 77:530–536. 1998. View Article : Google Scholar : PubMed/NCBI

40 

Rashidi LH, Homayoni H, Zou X, Liu L and Chen W: Investigation of the strategies for targeting of the afterglow nanoparticles to tumor cells. Photodiagnosis Photodyn Ther. 13:244–254. 2016. View Article : Google Scholar

41 

Shan D, Li J, Cai P, Prasad P, Liu F, Rauth AM and Wu XY: RGD-conjugated solid lipid nanoparticles inhibit adhesion and invasion of αVβ3 integrin-overexpressing breast cancer cells. Drug Deliv Transl Res. 5:15–26. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Zeng F, Luo F, Lv S, Zhang H, Cao C, Chen X, Wang S, Li Z, Wang X, Dou X, et al: A monoclonal antibody targeting neuropilin-1 inhibits adhesion of MCF7 breast cancer cells to fibronectin by suppressing the FAK/p130cas signaling pathway. Anticancer Drugs. 25:663–672. 2014.PubMed/NCBI

43 

Pal S, Moulik S, Dutta A and Chatterjee A: Extracellular matrix protein laminin induces matrix metalloproteinase-9 in human breast cancer cell line MCF-7. Cancer Microenviron. 7:71–78. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Cao W, Zheng W and Chen T: Ruthenium polypyridyl complex inhibits growth and metastasis of breast cancer cells by suppressing FAK signaling with enhancement of TRAIL-induced apoptosis. Sci Rep. 5:91572015. View Article : Google Scholar : PubMed/NCBI

45 

Tian T, Nan KJ, Guo H, Wang WJ, Ruan ZP, Wang SH, Liang X and Lu CX: PTEN inhibits the migration and invasion of HepG2 cells by coordinately decreasing MMP expression via the PI3K/Akt pathway. Oncol Rep. 23:1593–1600. 2010.PubMed/NCBI

46 

Jung CH, Kim EM, Park JK, Hwang SG, Moon SK, Kim WJ and Um HD: Bmal1 suppresses cancer cell invasion by blocking the phosphoinositide 3-kinase-Akt-MMP-2 signaling pathway. Oncol Rep. 9:2109–2113. 2013.

47 

O-charoenrat P, Wongkajornsilp A, Rhys-Evans PH and Eccles SA: Signaling pathways required for matrix metalloproteinase-9 induction by betacellulin in head-and-neck squamous carcinoma cells. Int J Cancer. 111:174–183. 2004. View Article : Google Scholar : PubMed/NCBI

48 

Kane LP, Shapiro VS, Stokoe D and Weiss A: Induction of NF-kappaB by the Akt/PKB kinase. Curr Biol. 9:601–604. 1999. View Article : Google Scholar : PubMed/NCBI

49 

de Mejia EG and Dia VP: Lunasin and lunasin-like peptides inhibit inflammation through suppression of NF-kappaB pathway in the macrophage. Peptides. 30:2388–2398. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2016
Volume 36 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jiang Q, Pan Y, Cheng Y, Li H, Liu D and Li H: Lunasin suppresses the migration and invasion of breast cancer cells by inhibiting matrix metalloproteinase-2/-9 via the FAK/Akt/ERK and NF-κB signaling pathways. Oncol Rep 36: 253-262, 2016
APA
Jiang, Q., Pan, Y., Cheng, Y., Li, H., Liu, D., & Li, H. (2016). Lunasin suppresses the migration and invasion of breast cancer cells by inhibiting matrix metalloproteinase-2/-9 via the FAK/Akt/ERK and NF-κB signaling pathways. Oncology Reports, 36, 253-262. https://doi.org/10.3892/or.2016.4798
MLA
Jiang, Q., Pan, Y., Cheng, Y., Li, H., Liu, D., Li, H."Lunasin suppresses the migration and invasion of breast cancer cells by inhibiting matrix metalloproteinase-2/-9 via the FAK/Akt/ERK and NF-κB signaling pathways". Oncology Reports 36.1 (2016): 253-262.
Chicago
Jiang, Q., Pan, Y., Cheng, Y., Li, H., Liu, D., Li, H."Lunasin suppresses the migration and invasion of breast cancer cells by inhibiting matrix metalloproteinase-2/-9 via the FAK/Akt/ERK and NF-κB signaling pathways". Oncology Reports 36, no. 1 (2016): 253-262. https://doi.org/10.3892/or.2016.4798