Silencing of casein kinase 2 inhibits PKC‑induced cell invasion by targeting MMP‑9 in MCF‑7 cells

  • Authors:
    • Jeong‑Mi Kim
    • Eun‑Mi Noh
    • Hyun‑Kyung Song
    • Yong‑Ouk You
    • Sung Hoo Jung
    • Jong‑Suk Kim
    • Kang‑Beom Kwon
    • Young‑Rae Lee
    • Hyun Jo Youn
  • View Affiliations

  • Published online on: April 13, 2018     https://doi.org/10.3892/mmr.2018.8885
  • Pages: 8397-8402
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Casein kinase 2 (CK2) is a serine/threonine protein kinase that has been considered to represent an important factor in mammary tumorigenesis. Increased expression of matrix metalloproteinase‑9 (MMP‑9) via nuclear factor‑κB (NF‑κB) activation has been demonstrated to promote breast cancer cell invasion. In the present study, the involvement of CK2 in protein kinase C (PKC) induced cell invasion in MCF‑7 breast cancer cells was investigated as well as the underlying molecular mechanisms. The mRNA and protein levels of MMP‑9 in MCF‑7 cells were investigated using reverse transcription‑quantitative polymerase chain reaction, western blot analyses and a zymography assay. Cell invasiveness was investigated using a Matrigel invasion assay, and it was revealed that small interfering RNA specific for CK2 suppressed PKC induced cell invasion by regulating MMP‑9 expression via activation of the p38 kinase/c‑Jun N‑terminal kinase/NF‑κB pathway. In addition, it was demonstrated that CK2 inhibitors [apigenin (20 µM), emodin (20 µM) or 2‑dimethylamino‑4,5,6,7‑tetrabromo‑1H‑benzimidazole (2 µM)] suppressed PKC induced cell invasion and MMP‑9 expression. The results of the present study suggested that CK2 is an important factor involved in the induction of MCF‑7 breast cancer cell invasion by PKC. Therefore, CK2 may represent novel candidates for therapy intended to inhibit invasion in breast cancer.

Introduction

Casein kinase 2 (CK2) is a ubiquitous serine/threonine protein kinase that is involved in numerous cellular processes, including proliferation and apoptosis, and has been revealed to be overexpressed in a number of human cancer cell lines (1,2). A previous study suggested that CK2 overexpression creates a favorable environment for the development of the tumor phenotype (3). CK2 has been associated with mammary tumorigenesis, as the levels of CK2 protein and activity are increased in breast cancer tissue compared with normal breast tissue (1,4). Furthermore, overexpression of CK2 in the mammary glands of transgenic mice can cause hyperplasia and breast carcinoma, thus demonstrating the oncogenic potential of CK2 (4).

Invasion and metastasis, the fundamental properties of cancer cells, are the primary causes of poor prognosis in patients with breast cancer. The mechanisms of cancer cell invasion and metastasis comprise a multistep biophysical process (5). In the early stages, the proteolytic degradation of extracellular matrix (ECM) components is facilitated by matrix metalloproteinases (MMPs) (6,7). Apigenin is abundant in common fruits and vegetables, and has notable anti-inflammatory, anti-oxidant and anti-carcinogenic properties (813). Emodin is an active constituent isolated from Rheum palmatum, a Chinese herb (14) and has been revealed to lead to the inhibition of cell proliferation, cell cycle arrest, inhibition of cell division, and decreased cell motility and invasion (15). TBBz is one of the most efficient inhibitors of CK2 (16), and has been demonstrated to induce apoptosis in cancer cells (17,18). MMP-9 has an important role in the degradation of the ECM during breast cancer cell invasion (19,20). Considering its importance in cancer development and progression, MMP-9 can be suggested to represent an early target in the treatment of breast cancer metastasis (21,22), and the inhibition of MMP-9 expression, activity and/or upstream regulatory activity may represent a potential therapeutic strategy. 12-O-tetradecanoylphorbol-13-acetate (TPA), a selective activator of protein kinase C (PKC) (23), induces breast cancer cell invasion by stimulating MMP-9 synthesis and secretion (21,24) via the activation of transcription factors, including nuclear factor-κB (NF-κB) and activator protein-1 (2527).

Considering that CK2 has been previously revealed to induce nuclear factor-κB (NF-κB) activation, the present study aimed to investigate whether the inhibition of CK2 affected TPA induced invasion and MMP-9 expression in MCF-7 human breast cancer cells and determine the associated underlying molecular mechanisms.

Materials and methods

Cells and materials

Human breast cancer MCF-7 cells were obtained from the American Type Culture Collection (Manassas, VA, USA) and were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS) (cat. no. 16000-044; Gibco; Thermo Fisher Scientific., Inc. Waltham, MA, USA) and 1% Anti-Anti (antibiotic-antimycotic) 100X (cat. no. 15240-062; Gibco; Thermo Fisher Scientific, Inc.) at 37°C in a 5% CO2 incubator. CK2 inhibitors [apigenin, 1,3,8-trihydroxy-6-methylanthraquinone (emodin) and 2-dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazole (TBBz)], dimethyl sulfoxide used as the solvent of the CK2 inhibitors and TPA (cat. no. P1585), and anti-β-actin (cat. no. A5441) antibodies were purchased from Sigma-Aldrich; Merck KGaA (Darmstadt, Germany). Matrigel was obtained from Corning Incorporated (Corning, NY, USA). Primary antibodies against CK2α (cat. no. 2656), p38 kinase (p38; cat. no. 9212), c-Jun N-terminal kinase (JNK; cat. no. 9252), extracellular signal-regulated kinase (ERK; cat. no. 9102), as well as the phosphorylated forms of p38 (cat. no. 9211), JNK (cat. no. 9261) and ERK (cat. no. 9101), were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). Antibodies specific to MMP-9 (cat. no. 12759), transcription factor p50 (cat. no. 7178) and transcription factor p65 (cat. no. 372) and proliferating cell nuclear antigen (PCNA; cat. no. 7907), as well as horseradish peroxidase (HRP)-conjugated secondary immunoglobulin G (IgG; cat. no. SC-2004, SC-2005), were all purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA).

RNA interference

CK2α specific small interfering (si)RNA and negative control siRNA (cat. no. SN-1003) were obtained from Bioneer Corporation (Daejeon, Korea). The siRNA used were as follows: CK2α sense, CAU UUA GUU ACU GGG CAU A(dTdT) and antisense, UAU GCC CAG UAA CUA AAU G(dTdT). Briefly, human breast cancer MCF-7 cells (5×105) were transfected with 100 pmol siRNA using RNAiMAX Transfection Reagent for 24 h at 37°C (Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions.

Determination of cell viability

Cell viability assays were performed using the EZ-Cytox Enhanced Cell Viability Assay kit (DOGEN, Seoul, Korea; www.dogenbio.com/shop/item.php?it_id=1490923054), according to the manufacturer's instructions. Briefly, 3×104 cells/well were seeded into 96-well plates and incubated at 37°C for 24 h to allow the cells to adhere. The cells were then treated with apigenin (20 µM), emodin (20 µM) or TBBz (2 µM) for 24 h at 37°C. Following this, all cells were incubated with EZ-CytoxReagent (10 µl) for 30 min at 37°C. Absorbance at 450 nm was determined using an ELISA plate reader (Sunrise™; Tecan Group, Ltd., Mannedorf, Switzerland).

Western blot analysis

MCF-7 cells (7×105) pretreated with CK2 inhibitors [20 µM apigenin, 20 µM emodin, or 2 µM TBBz] for 1 h, and cells (3.5×105) transfected with CK2 were subsequently incubated with TPA for 24 h at 37°C. The cells were lysed with ice-cold radioimmunoprecipitation assay buffer (Thermo Fisher Scientific, Inc.), and the protein concentration of the resulting lysates was determined using a BioSpec-nano (Shimadzu Corporation, Kyoto, Japan). Total protein samples were resolved via 10% SDS-PAGE analysis and then transferred to polyvinylidene fluoride membranes (GE Healthcare Life Sciences, Little Chalfont, UK). The membranes were blocked with 5% bovine serum albumin (cat. no. 160069; MP biomedicals Inc.,) or 5% skimmed milk in TBS with 0.5% Tween-20 at 4°C for 2 h, and then incubated overnight at 4°C with primary antibodies. All antibodies used were diluted 1:2,000. HRP-conjugated IgGs were used as secondary antibodies and incubated at 4°C for 1 h. Protein expression levels were visualized using a Mini HD6 image analyzer using Alliance 1D software (UVItec Cambridge; Cleaver Scientific Ltd., Rugby, UK).

Gelatin zymography assay

MMP-9 activity in the conditioned medium (serum-free DMEM) was determined by gelatin zymography. MCF-7 cells (7×105) were pretreated with CK2 inhibitors [apigenin (20 µM), emodin (20 µM) or TBBz (2 µM)] for 1 h, and then incubated with TPA for 24 h at 37°C. The conditioned medium was mixed with non-reducing sample buffer (0.5M Tris-HCl (pH 6.8), Glycerol 2 ml, 10% SDS, 0.1% Bromophenol blue, D.W 1 ml up to 10 ml), and subjected to 10% SDS-PAGE analysis containing 0.1% [weight (w)/volume (v)] gelatin. The gel was then incubated in a renaturing buffer (2.5% Triton™ X-100; Sigma-Aldrich; Merck KGaA) with gentle agitation to remove the SDS at room temperature for 30 min. Following this, gels were incubated in a developing buffer (5 mM CaCl2, 0.02% Brij, pH 7.5 and 50 mM Tris-HCl) overnight at 37°C. The gel was stained for 30 min at room temperature with 0.25% (w/v) Coomassie brilliant blue R-250 at room temperature followed by destaining with washing buffer (10% acetic acid and 10% methanol). Proteolysis was visualized as a white zone in a dark blue field using a digital imaging system (FluorChem R by ProteinSimple; Cell Biosciences, Palo Alto, CA, USA).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from cultured cells using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Complementary DNA was synthesized from total RNA using a PrimeScript™ RT Reagent kit (Takara Biotechnology Co., Ltd., Dalian, China). mRNA expression levels were determined by qPCR analysis using the StepOnePlus™ Real-Time PCR System and SYBR®-Green (Applied Biosystems; Thermo Fisher Scientific, Inc.). The primers used were as follows: MMP-9 sense, 5′-CCTGGAGACCTGAGAACCAATCT-3′ and antisense, 5′-CCACCCGAGTGTAACCATAGC-3′; GAPDH (NM_002046) sense, 5′-ATGGAAATCCCATCACCATCTT-3′ and antisense, 5′-CGCCCCACTTGATTTTGG-3′. qPCR was performed with a preliminary incubation at 95°C for 10 min, followed by 40 cycles of 95°C for 15 sec and 60°C for 1 min. MMP-9 mRNA expression levels were quantified relative to GAPDH mRNA expression using the comparative 2−∆∆Cq method (28).

Preparation of nuclear extracts

MCF-7 cells (2×106) were transfected with CK2 siRNA in the presence of 20 nM TPA for 3 h at 37°C and then washed twice with PBS, scraped, resuspended in 1.5 ml ice-cold PBS (pH 7.5), and then centrifuged at 1,500 × g for 4 min at 4°C. Isolation of the nuclear and cytoplasmic extracts was performed using NE-PER Nuclear and Cytoplasmic Extraction Reagents kit (cat. no. 78835; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol.

Invasion assay

Invasion of MCF-7 cells was assessed using 24-well chambers with 8 µm pore membranes coated with 20 µl Matrigel. The Matrigel coating was rehydrated in 0.5 ml DMEM for 2 h at 37°C immediately prior to experiments. Cell growth medium (0.5 ml) with suspensed cells (3×105) and cells (2×105) transfected with CK2 siNRA were added to the upper chambers, and cell growth medium (0.5 ml) with TPA, alone or with CK2 inhibitors, were added to the bottom well. The upper chambers and bottom well were incubated with DMEM supplemented 10% FBS and 1% antibiotic for 24 h. Following incubation, cells on the upper membrane surface were removed using cotton swabs, and cells that had migrated to the lower membrane surface were fixed by formaldehyde solution (3.6%) at room temperature, stained with crystal violet, and counted in five random fields per chamber using a Leica DM ILLED Inverted Lab microscope (Leica, Wetzlar, Germany) used magnification, ×10.

Statistical analysis

Data are expressed as the mean ± standard error. Statistical significance was determined using one-way analysis of variance followed by Scheffe post hoc test in Excel (Microsoft Excel 2013). P<0.005 was considered to indicate a statistically significant difference. All experiments were performed in triplicate.

Results

Inhibition of CK2α inhibits TPA-induced MMP-9 expression in MCF-7 cells

In order to investigate the effects of CK2α inhibition on TPA-induced cell invasion and MMP-9 expression, intracellular CK2α expression was suppressed via transfection with siRNA (Fig. 1A). The siRNA-mediated inhibition of CK2α significantly suppressed the increase in MMP-9 mRNA/protein expression compared with the TPA group (Fig. 1B and C). Furthermore, MCF-7 cells treated with TPA exhibited significantly increased invasion compared with the untreated control cells. However, the inhibition of CK2α significantly suppressed TPA induced MCF-7 cell invasion (Fig. 1D). These results suggest that CK2α may be involved in the underlying mechanism resulting in the TPA-induced increase of cell invasion and MMP-9 expression.

Inhibition of CK2α suppresses TPA-induced mitogen-activated protein kinase (MAPK) and NF-κΒ activation in MCF-7 cells

The role of MAPKs (ERK, p38 and JNK) as upstream modulators of NF-κΒ in the activation of MMP-9 expression has been well established (25,26,29). The effects of CK2α inhibition on the TPA-induced phosphorylation of MAPKs was investigated. Following suppression of CK2α expression via siRNA knockdown, the phosphorylation levels of p38 and JNK, but not of ERK, were significantly suppressed (Fig. 2A). As revealed in Fig. 2B, treatment with TPA significantly enhanced the level of NF-κΒ (p65 and p50 subunits) compared with the control group. However, following suppression of intracellular CK2α expression, the expression levels of NF-κΒ were significantly inhibited (Fig. 2B). These results suggest that CK2α is an upstream regulator of p38 and JNK in the PKC induced MAPK-NF-κΒ signaling pathway responsible for the regulation of MMP-9 expression.

CK2 inhibitors suppress TPA-induced cell invasion and MMP-9 expression in MCF-7 cells

To further elucidate the therapeutic potential of CK2 inhibition for the prevention of PKC induced breast cancer cell invasion, three CK2 inhibitors were investigated. The cytotoxicity of these three CK2 inhibitors was investigated using a EZ-cytox assay. As revealed in Fig. 3A, treatment with 20 µM apigenin, 20 µM emodin and 2 µM TBBz did not cause any significant changes in cell viability. Furthermore, the CK2 inhibitors were revealed to suppress TPA-induced increase in MMP-9 mRNA/protein expression and cell invasion (Fig. 3B-D). These results suggest that CK2 inhibitors have the ability to inhibit cell invasion via regulating MMP-9 expression.

Discussion

The stages of breast cancer are classified as 0–4. Number staging systems usually use the TNM system to divide cancers into stages. The majority of cancer types are classified into 4 stages, numbered from 1 to 4, which are important for diagnosis and treatment (30). From stage 2 onwards, metastases develop and the 5 year survival rate begins to decline (31). Mortality from metastatic breast cancer are on the rise, and thus suppression of metastasis has emerged as a major challenge in breast cancer treatment (32,33). In the early stages of tumor metastasis, individual tumor cells or clusters invade the ECM surrounding the primary tumor (34,35). The ECM consists of type IV collagen and other matrix proteins (19). Type IV collagen is a major component of the basement membrane and type IV collagenase MMP-9 has previously been revealed to be an important molecule in tumor progression and invasion in mammary tumors (36).

Elevated MMP-9 levels are functionally associated with breast cancer metastasis, and MMP-9 serves a role in TPA induced invasion of MCF-7 cells (21,24,37). Therefore, the suppression of MMP-9 expression may represent a novel therapeutic strategy for the prevention of tumor metastasis. TPA upregulates MMP-9 expression in MCF-7 cells; however, the underlying molecular mechanisms have not been fully established. Therefore, the present study aimed to investigate the role of CK2 in the regulation of MMP-9 expression in MCF-7 cells. The results of the present study revealed that the inhibition of CK2α suppressed TPA-induced MMP-9 expression and invasion in MCF-7 cells. Furthermore, the results of the present study revealed that CK2α is a regulator of PKC-induced invasion in MCF-7 cells.

In addition, the inhibition of CK2 was demonstrated to suppress the activation of NF-κB in MCF-7 cells. The MAPK/NF-κB signaling cascade serves a role in PKC-mediated MMP-9 expression in MCF-7 cells (23,24,38,39). Furthermore, the MAPK signaling pathway has been previously demonstrated to be important for the activation of NF-κB (40,41). CK2 has been reported to induce NF-κB activation (27,42). In the present study, it was revealed that CK2α inhibition markedly suppresses TPA-induced activation of p38, JNK and NF-κB. In addition, it was revealed that the treatment of MCF-7 cells with CK2 inhibitors suppressed TPA-induced invasion and MMP-9 expression in MCF-7 cells.

In conclusion, the results of the present study revealed that suppression of CK2 exerts anti-invasive effects in PKC-induction condition via the regulation of MMP-9 expression levels. Therefore, CK2 may represent a novel anti-invasive target for cancer therapy.

Acknowledgements

Not applicable.

Funding

The present study was supported by a National Research Foundation of Korea grant funded by the Korea Government (grant nos. 2011-0030130 and 2013R1A1A2007181).

Availability of data and materials

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

Authors' contributions

JMK and EMN performed most of the experiments. HKS, YOY and KBK analyzed the data and provided comments. SJ and JSK helped with the experiments. HJY designed the project. YRL designed the analysis. KBK, YRL and HJY wrote the manuscript. All authors reviewed the manuscript.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

Authors declare that they have no competing interests.

References

1 

Munstermann U, Fritz G, Seitz G, Lu YP, Schneider HR and Issinger OG: Casein kinase II is elevated in solid human tumours and rapidly proliferating non-neoplastic tissue. Eur J Biochem. 189:251–257. 1990. View Article : Google Scholar : PubMed/NCBI

2 

Ahmed K, Davis AT, Wang H, Faust RA, Yu S and Tawfic S: Significance of protein kinase CK2 nuclear signaling in neoplasia. J Cell Biochem Suppl. Suppl 35:S130–S135. 2000. View Article : Google Scholar

3 

Ruzzene M and Pinna LA: Addiction to protein kinase CK2: A common denominator of diverse cancer cells? Biochim Biophys Acta. 1804:499–504. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Landesman-Bollag E, Romieu-Mourez R, Song DH, Sonenshein GE, Cardiff RD and Seldin DC: Protein kinase CK2 in mammary gland tumorigenesis. Oncogene. 20:3247–3257. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Christofori G: New signals from the invasive front. Nature. 441:444–450. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Chambers AF and Matrisian LM: Changing views of the role of matrix metalloproteinases in metastasis. J Natl Cancer Inst. 89:1260–1270. 1997. View Article : Google Scholar : PubMed/NCBI

7 

Woodhouse EC, Chuaqui RF and Liotta LA: General mechanisms of metastasis. Cancer. 80 8 Suppl:S1529–S1537. 1997. View Article : Google Scholar

8 

Patel D, Shukla S and Gupta S: Apigenin and cancer chemoprevention: Progress, potential and promise (review). Int J Oncol. 30:233–245. 2007.PubMed/NCBI

9 

Shukla S and Gupta S: Apigenin-induced cell cycle arrest is mediated by modulation of MAPK, PI3K-Akt, and loss of cyclin D1 associated retinoblastoma dephosphorylation in human prostate cancer cells. Cell Cycle. 6:1102–1114. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Shukla S and Gupta S: Suppression of constitutive and tumor necrosis factor alpha-induced nuclear factor (NF)-kappaB activation and induction of apoptosis by apigenin in human prostate carcinoma PC-3 cells: Correlation with down-regulation of NF-kappaB-responsive genes. Clin Cancer Res. 10:3169–3178. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Chen D, Landis-Piwowar KR, Chen MS and Dou QP: Inhibition of proteasome activity by the dietary flavonoid apigenin is associated with growth inhibition in cultured breast cancer cells and xenografts. Breast Cancer Res. 9:R802007. View Article : Google Scholar : PubMed/NCBI

12 

Way TD, Kao MC and Lin JK: Degradation of HER2/neu by apigenin induces apoptosis through cytochrome c release and caspase-3 activation in HER2/neu-overexpressing breast cancer cells. FEBS Lett. 579:145–152. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Kim JS, Eom JI, Cheong JW, Choi AJ, Lee JK, Yang WI and Min YH: Protein kinase CK2alpha as an unfavorable prognostic marker and novel therapeutic target in acute myeloid leukemia. Clin Cancer Res. 13:1019–1028. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Zhang L and Hung MC: Sensitization of HER-2/neu-overexpressing non-small cell lung cancer cells to chemotherapeutic drugs by tyrosine kinase inhibitor emodin. Oncogene. 12:571–576. 1996.PubMed/NCBI

15 

Zou J, Luo H, Zeng Q, Dong Z, Wu D and Liu L: Protein kinase CK2α is overexpressed in colorectal cancer and modulates cell proliferation and invasion via regulating EMT-related genes. J Transl Med. 9:972011. View Article : Google Scholar : PubMed/NCBI

16 

Sarno S, Ruzzene M, Frascella P, Pagano MA, Meggio F, Zambon A, Mazzorana M, Di Maira G, Lucchini V and Pinna LA: Development and exploitation of CK2 inhibitors. Mol Cell Biochem. 274:69–76. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Di Maira G, Brustolon F, Bertacchini J, Tosoni K, Marmiroli S, Pinna LA and Ruzzene M: Pharmacological inhibition of protein kinase CK2 reverts the multidrug resistance phenotype of a CEM cell line characterized by high CK2 level. Oncogene. 26:6915–6926. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Pagano MA, Andrzejewska M, Ruzzene M, Sarno S, Cesaro L, Bain J, Elliott M, Meggio F, Kazimierczuk Z and Pinna LA: Optimization of protein kinase CK2 inhibitors derived from 4,5,6,7-tetrabromobenzimidazole. J Med Chem. 47:6239–6247. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Nakajima M, Welch DR, Belloni PN and Nicolson GL: Degradation of basement membrane type IV collagen and lung subendothelial matrix by rat mammary adenocarcinoma cell clones of differing metastatic potentials. Cancer Res. 47:4869–4876. 1987.PubMed/NCBI

20 

Egeblad M and Werb Z: New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2:161–174. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Lin CW, Hou WC, Shen SC, Juan SH, Ko CH, Wang LM and Chen YC: Quercetin inhibition of tumor invasion via suppressing PKC delta/ERK/AP-1-dependent matrix metalloproteinase-9 activation in breast carcinoma cells. Carcinogenesis. 29:1807–1815. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Lee SO, Jeong YJ, Im HG, Kim CH, Chang YC and Lee IS: Silibinin suppresses PMA-induced MMP-9 expression by blocking the AP-1 activation via MAPK signaling pathways in MCF-7 human breast carcinoma cells. Biochem Biophys Res Commun. 354:165–171. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Newton AC: Regulation of protein kinase C. Curr Opin Cell Biol. 9:161–167. 1997. View Article : Google Scholar : PubMed/NCBI

24 

Lee SO, Jeong YJ, Kim M, Kim CH and Lee IS: Suppression of PMA-induced tumor cell invasion by capillarisin via the inhibition of NF-kappaB-dependent MMP-9 expression. Biochem Biophys Res Commun. 366:1019–1024. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Chung TW, Moon SK, Chang YC, Ko JH, Lee YC, Cho G, Kim SH, Kim JG and Kim CH: Novel and therapeutic effect of caffeic acid and caffeic acid phenyl ester on hepatocarcinoma cells: Complete regression of hepatoma growth and metastasis by dual mechanism. FASEB J. 18:1670–1681. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Hong S, Park KK, Magae J, Ando K, Lee TS, Kwon TK, Kwak JY, Kim CH and Chang YC: Ascochlorin inhibits matrix metalloproteinase-9 expression by suppressing activator protein-1-mediated gene expression through the ERK1/2 signaling pathway: Inhibitory effects of ascochlorin on the invasion of renal carcinoma cells. J Biol Chem. 280:25202–25209. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Woo MS, Jung SH, Kim SY, Hyun JW, Ko KH, Kim WK and Kim HS: Curcumin suppresses phorbol ester-induced matrix metalloproteinase-9 expression by inhibiting the PKC to MAPK signaling pathways in human astroglioma cells. Biochem Biophys Res Commun. 335:1017–1025. 2005. View Article : Google Scholar : PubMed/NCBI

28 

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

29 

Ohashi Y, Tsuchiya Y, Koizumi K, Sakurai H and Saiki I: Prevention of intrahepatic metastasis by curcumin in an orthotopic implantation model. Oncology. 65:250–258. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Reyes-Ortiz CA, Eschbach K, Zhang DD and Goodwin JS: Neighborhood composition and cancer among hispanics: Tumor stage and size at time of diagnosis. Cancer Epidemiol Biomarkers Prev. 17:2931–2936. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Howlader N, Noone AM, Krapcho M, Miller D, Bishop K, Kosary CL, Yu M, Ruhl J, Tatalovich Z and Mariotto A: SEER cancer statistics review, 1975–2014. National Cancer Institute; Bethesda, MD: 2017

32 

Polyak K: Heterogeneity in breast cancer. J Clin Invest. 121:3786–3788. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Neophytou C, Boutsikos P and Papageorgis P: Molecular mechanisms and emerging therapeutic targets of triple-negative breast cancer metastasis. Front Oncol. 8:312018. View Article : Google Scholar : PubMed/NCBI

34 

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

35 

Li DM and Feng YM: Signaling mechanism of cell adhesion molecules in breast cancer metastasis: Potential therapeutic targets. Breast Cancer Res Treat. 128:7–21. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Scorilas A, Karameris A, Arnogiannaki N, Ardavanis A, Bassilopoulos P, Trangas T and Talieri M: Overexpression of matrix-metalloproteinase-9 in human breast cancer: A potential favourable indicator in node-negative patients. Br J Cancer. 84:1488–1496. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Park SK, Hwang YS, Park KK, Park HJ, Seo JY and Chung WY: Kalopanaxsaponin A inhibits PMA-induced invasion by reducing matrix metalloproteinase-9 via PI3K/Akt- and PKCdelta-mediated signaling in MCF-7 human breast cancer cells. Carcinogenesis. 30:1225–1233. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Weng CJ, Chau CF, Hsieh YS, Yang SF and Yen GC: Lucidenic acid inhibits PMA-induced invasion of human hepatoma cells through inactivating MAPK/ERK signal transduction pathway and reducing binding activities of NF-kappaB and AP-1. Carcinogenesis. 29:147–156. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Eberhardt W, Huwiler A, Beck KF, Walpen S and Pfeilschifter J: Amplification of IL-1 beta-induced matrix metalloproteinase-9 expression by superoxide in rat glomerular mesangial cells is mediated by increased activities of NF-kappa B and activating protein-1 and involves activation of the mitogen-activated protein kinase pathways. J Immunol. 165:5788–5797. 2000. View Article : Google Scholar : PubMed/NCBI

40 

Yao J, Xiong S, Klos K, Nguyen N, Grijalva R, Li P and Yu D: Multiple signaling pathways involved in activation of matrix metalloproteinase-9 (MMP-9) by heregulin-beta1 in human breast cancer cells. Oncogene. 20:8066–8074. 2001. View Article : Google Scholar : PubMed/NCBI

41 

Madrid LV, Mayo MW, Reuther JY and Baldwin AS Jr: Akt stimulates the transactivation potential of the RelA/p65 Subunit of NF-kappa B through utilization of the Ikappa B kinase and activation of the mitogen-activated protein kinase p38. J Biol Chem. 276:18934–18940. 2001. View Article : Google Scholar : PubMed/NCBI

42 

Saja K, Babu MS, Karunagaran D and Sudhakaran PR: Anti-inflammatory effect of curcumin involves downregulation of MMP-9 in blood mononuclear cells. Int Immunopharmacol. 7:1659–1667. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2018
Volume 17 Issue 6

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kim JM, Noh EM, Song HK, You YO, Jung S, Kim JS, Kwon KB, Lee YR and Youn H: Silencing of casein kinase 2 inhibits PKC‑induced cell invasion by targeting MMP‑9 in MCF‑7 cells. Mol Med Rep 17: 8397-8402, 2018
APA
Kim, J., Noh, E., Song, H., You, Y., Jung, S., Kim, J. ... Youn, H. (2018). Silencing of casein kinase 2 inhibits PKC‑induced cell invasion by targeting MMP‑9 in MCF‑7 cells. Molecular Medicine Reports, 17, 8397-8402. https://doi.org/10.3892/mmr.2018.8885
MLA
Kim, J., Noh, E., Song, H., You, Y., Jung, S., Kim, J., Kwon, K., Lee, Y., Youn, H."Silencing of casein kinase 2 inhibits PKC‑induced cell invasion by targeting MMP‑9 in MCF‑7 cells". Molecular Medicine Reports 17.6 (2018): 8397-8402.
Chicago
Kim, J., Noh, E., Song, H., You, Y., Jung, S., Kim, J., Kwon, K., Lee, Y., Youn, H."Silencing of casein kinase 2 inhibits PKC‑induced cell invasion by targeting MMP‑9 in MCF‑7 cells". Molecular Medicine Reports 17, no. 6 (2018): 8397-8402. https://doi.org/10.3892/mmr.2018.8885