Open Access

Role of NOX1 and NOX5 in protein kinase C/reactive oxygen species‑mediated MMP‑9 activation and invasion in MCF‑7 breast cancer cells

  • Authors:
    • Hyun-Kyung Song
    • Jeong-Mi Kim
    • Eun-Mi Noh
    • Hyun Jo Youn
    • Young-Rae Lee
  • View Affiliations

  • Published online on: August 20, 2024     https://doi.org/10.3892/mmr.2024.13312
  • Article Number: 188
  • Copyright: © Song 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

NADPH oxidases (NOXs) are a family of membrane proteins responsible for intracellular reactive oxygen species (ROS) generation by facilitating electron transfer across biological membranes. Despite the established activation of NOXs by protein kinase C (PKC), the precise mechanism through which PKC triggers NOX activation during breast cancer invasion remains unclear. The present study aimed to investigate the role of NOX1 and NOX5 in the invasion of MCF‑7 human breast cancer cells. The expression and activity of NOXs and matrix metalloprotease (MMP)‑9 were assessed by reverse transcription‑quantitative PCR and western blotting, and the activity of MMP‑9 was monitored using zymography. Cellular invasion was assessed using the Matrigel invasion assay, whereas ROS levels were quantified using a FACSCalibur flow cytometer. The findings suggested that NOX1 and NOX5 serve crucial roles in 12‑O‑tetradecanoylphorbol‑13‑acetate (TPA)‑induced MMP‑9 expression and invasion of MCF‑7 cells. Furthermore, a connection was established between PKC and the NOX1 and 5/ROS signaling pathways in mediating TPA‑induced MMP‑9 expression and cellular invasion. Notably, NOX inhibitors (diphenyleneiodonium chloride and apocynin) significantly attenuated TPA‑induced MMP‑9 expression and invasion in MCF‑7 cells. NOX1‑ and NOX5‑specific small interfering RNAs attenuated TPA‑induced MMP‑9 expression and cellular invasion. In addition, knockdown of NOX1 and NOX5 suppressed TPA‑induced ROS levels. Furthermore, a PKC inhibitor (GF109203X) suppressed TPA‑induced intracellular ROS levels, MMP‑9 expression and NOX activity in MCF‑7 cells. Therefore, NOX1 and NOX5 may serve crucial roles in TPA‑induced MMP‑9 expression and invasion of MCF‑7 breast cancer cells. Furthermore, the present study indicated that TPA‑induced MMP‑9 expression and cellular invasion were mediated through PKC, thus linking the NOX1 and 5/ROS signaling pathways. These findings offer novel insights into the potential mechanisms underlying their anti‑invasive effects in breast cancer.

Introduction

Breast cancer, characterized by malignant solid tumors originating from mammary gland epithelium, is a major cause of mortality among women worldwide (1). Notably, the 5-year overall survival rate remains poor, primarily owing to the metastasis of cancer cells to vital organs such as the lungs, brain, and bones (24). Therefore, elucidating the molecular mechanisms underlying breast cancer metastasis is imperative.

The metastatic cascade involves a complex array of biological processes, including extracellular matrix (ECM) degradation; ECM, a crucial noncellular tissue component, provides structural and biochemical support essential for cellular functions (5,6). Degradation of ECM requires the involvement of several extracellular proteinases, among which matrix metalloproteases (MMPs), a family of zinc-dependent proteinases, play pivotal roles in pathological processes, including breast cancer (7). Notably, MMP-9 is a key player in cancer cell invasion and metastasis. MMP-9 expression is induced through the activation of various intracellular signaling proteins through the stimulation of probol esters, inflammatory cytokines, epidermal growth factors (EGFs), or phorbol esters such as 12-O-tetradecanoylphorbol-13-acetate (TPA) (810).

TPA, a selective activator of protein kinase C (PKC), orchestrates a cascade of events leading to the production of reactive oxygen species (ROS) either upstream or downstream (1113). Consequently, ROS modulate several intracellular signaling pathways, including protein kinase B (AKT) and mitogen-activated protein kinases (MAPKs) (1416), thereby influencing the activation of transcription factors such as activator protein-1 (AP-1) and nuclear factor-kappa B (NF-κB), which are intricately associated with MMP expression during tumor invasion and metastasis (1519).

Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) are a family of membrane proteins involved in intracellular ROS production and facilitate electron transfer across biological membranes. NOXs catalyze superoxide generation at the plasma membrane, subsequently releasing it into the extracellular space, where it is converted into ROS, such as hydrogen peroxide and superoxide anions (12,13). NOXs are closely associated with phosphatidylinositol 3-OH kinase (PI3K) signaling; PKC, a downstream molecule of PI3K, is essential for superoxide generation via NOXs (14,20,21). PKC mediates the activation of NOXs (22); however, the mechanism underlying NOX activation via PKC during breast cancer invasion remains unclear.

In this study, we investigated whether PKC regulates ROS production through NOXs during cell invasion of MCF-7 cells, a human breast cancer cell. Furthermore, we confirm the significance of NOXs in TPA-induced MMP-9 expression and cell invasion in these cells. We believe that our findings would help broaden our understanding of the molecular mechanisms underlying breast cancer metastasis.

Materials and methods

Cells and reagents

MCF-7 cells (cat. no. HTB-22) were procured from the American Type Culture Collection (Manassas, VA, USA). The cells were cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with heat-inactivated 10% fetal bovine serum (FBS) and 1% antibiotics. The cell culture was maintained in a controlled environment with a temperature of 37°C in a 5% CO2 atmosphere in an incubator. β-actin antibody (cat. no. A5441), bovine serum albumin (BSA), skim milk, TPA, diphenyleneiodonium chloride (DPI), and apocynin (APO) were purchased from Sigma-Aldrich (St. Louis, MO, USA). MMP-9 (cat. no. SC-12759) and goat anti-mouse IgG-HRP (cat. no. sc-2005) antibodies were obtained from Santa Cruz Biotechnology (Dallas, TX, USA). GF109203X (PKC inhibitor; GF) was purchased from Abcam (Cambridge, MA, USA). Phosphate-buffered saline (PBS), FBS, and DMEM were purchased from Gibco-BRL (Gaithersburg, MD, USA).

RNA interference

NOX1-5-specific and control siRNAs were purchased from BIONEER (Daejeon, Korea). The siRNA sets used for amplification were as follows: NOX1, sense: 5′-GAGCAUGAAUGAGAGUCAU-3′, antisense: 5′-AUGACUCUCAUUCAUGCUC-3′; NOX2, sense: 5′-GUAAUGUCAGUGGAAGUUA-3′, antisense: 5′-UAACUUCCACUGACAUUAC-3′; NOX3, sense: 5′-CACCAUGUUUUCAUCGUCU-3′, antisense: 5′-AGACGAUGAAAACAUGGUG-3′; NOX4, sense: 5′-CAGAGUUUACCCAGCACAA-3′, antisense: 5′-UUGUGCUGGGUAAACUCUG-3′; NOX5, sense: 5′-GUGACUACUUGUAUCUGAA-3′, antisense: 5′-UUCAGAUACAAGUAGUCAC-3′; control siRNA, sense: 5′-UUCUCCGAACGUGUCACGU-3′, antisense: 5′-ACGUGACACGUUCGGAGAA-3′. The cells were transfected with NOX1 (30 pmol) and NOX3 (30 pmol) siRNAs for 24 h and with NOX2 (30 pmol), NOX4 (100 pmol), and NOX5 (100 pmol) siRNAs for 48 h. Control siRNA transfections were also conducted for each respective time point. All siRNA transfections in MCF-7 cells were performed using Lipofectamine™ RNAiMAX (Invitrogen, San Diego, CA, USA), in accordance with the manufacturer's forward transfection protocol.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was isolated from cells using TRIzol® (Life Technologies, Grand Island, NY, USA) following the manufacturer's protocol. RNA concentration and purity were calculated using BioSpec-nano (Shimadzu, Kyoto, Japan). cDNA was synthesized using 1 µg total RNA using a PrimeScript™RT reagent Kit (cat. no. RR047A; TaKaRa, Shiga, Japan). The RT-qPCR cycling protocol comprised an initial denaturation step at 95°C for 10 min, followed by 40 cycles of amplification consisting of denaturation at 95°C for 15 sec and annealing/extension at 60°C for 1 min. Subsequently, a melting curve analysis was conducted with temperature ramping from 95°C for 15 sec, followed by annealing/extension at 60°C for 1 min, and concluding with a final denaturation step at 95°C for 15 sec. The mRNA expression levels of MMP-9, NOX1, NOX5, and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) were analyzed using the ABI PRISM 7900 sequence detection system and SYBR Green (Applied Biosystems, Foster City, CA, USA). Relative quantitation was performed using the comparative 2−ΔΔCq method (23). The primer sets used for amplification were as follows: MMP-9 (NM 004994), forward primer: 5′-CCTGGAGACCTGAGAACCAATCT-3′, reverse primer: 5′-CCACCCGAGTGTAACCATAGC-3′; GAPDH (NM 002046), forward primer: 5′-ATGGAAATCCCATCACCATCTT-3′, reverse primer: 5′-CGCCCCACTTGATTTTGG-3′; Primers for NOX1 (NM_007052, cat. no. PPH06068A) and NOX5 (NM_024505, cat. no. PPH17569A) were obtained from QIAGEN (Hilden, Germany). To account for variations in mRNA concentration, the expression levels of MMP9, NOX1, and NOX5 were normalized to that of the housekeeping gene, GAPDH. Relative quantification was analyzed using the comparative 2−ΔΔCq method, following the manufacturer's instructions.

Quantification of intracellular ROS

The intracellular ROS was detected using an oxidation-sensitive fluorescent probe dye (CM-H2DCFDA; Invitrogen). MCF-7 cells were stimulated with 20 nM TPA for 24 h, after which the cells were incubated with 10 µM CM-H2DCFDA at 37°C for 30 min. CM-H2DCFDA was oxidized to the green fluorescent dichlorofluorescein (DCF) using hydrogen peroxide. The DCF fluorescence was measured using a FACSCalibur flow cytometer (BD Biosciences, San Diego, CA, USA). ROS production was expressed as the mean fluorescence intensity and analyzed using the CellQuest software (BD Biosciences).

Western blot analysis

The cells (7×105) were incubated with 20 nM TPA for 24 h at 37°C. Following the treatments, cells were lysed with ice-cold radioimmunoprecipitation assay (RIPA) buffer (Thermo Scientific, Rockford, IL, USA) for 30 min on ice, and the protein concentration in the resulting lysates was determined using a Bio Spec-nano (Shimadzu). Subsequently, 20 µg of protein samples were resolved using 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to Hybond™ polyvinylidene fluoride (PVDF) membranes (GE Healthcare Life Sciences, Buckinghamshire, UK) through western blotting. Following the blocking with 5% BSA or skim milk for 2 h at 4°C, the membranes were cropped around 70 kDa. Subsequently, the upper and lower portions of the cropped membranes were incubated overnight at 4°C with the primary antibody (1:2,500) targeting MMP-9 (92 kDa) and β-actin (45 kDa), respectively, HRP-conjugated IgG (1:2,500) served as the secondary antibody for 1 h at 4°C. Protein expression levels were determined through signal analysis using a MINI HD6 image analyzer (UVITEC, Cambridge, UK).

Matrigel invasion assay

The invasion of MCF-7 cells was assessed using a 24-well cell culture insert (8-µm pore size) coated with rehydrated 20 µl Matrigel™ (Corning Life Sciences, Corning, NY, USA) in culture medium for 30 min. Suspended cells (4×105) and chemical attractant, 20 nM of TPA, in 0.5 ml culture medium (supplemented with 10% FBS and 1% antibiotics) were transferred into the upper and lower chambers. Following 24 h incubation in a 5% CO2 incubator at 37°C, the cells on the upper side of the membrane were gently removed using cotton swabs. The cells that invaded the lower chamber through the membrane were fixed with a 10% formalin solution for 30 min at room temperature. Following fixation, they were stained with 0.2% crystal violet for an additional 30 min at room temperature. Subsequently, the invading cells were counted in five random areas of the membrane under a light microscope.

Gelatin zymography assay

MCF-7 cells were pre-treated with DPI (5 µM) or APO (300 µM) for 1 h and then stimulated with TPA (20 nM) for 24 h at 37°C in serum-free DMEM medium. The collected culture medium was suspended in a zymography sample buffer. Electrophoresis was performed under non-reducing conditions using 10% sodium dodecyl sulfate-polyacrylamide gel containing 0.1% (w/v) gelatin. Following electrophoresis, the gel was washed with 2.5% Triton X-100 for 30 min under gentle agitation. Subsequently, the gel was incubated overnight at 37°C in a developing solution (50 mM Tris-HCl, 5 mM CaCl2, 100 mM NaCl, 0.02% Brij-35; pH 7.5). After incubation, the gel was stained with 0.25% Coomassie blue R-250 at room temperature for 30 min and washed with a destaining solution (40% methanol and 7% acetic acid) until the bands were visible. The proteolytic activity of MMP-9 was measured by comparing the transparent bands resulting from the decomposition of gelatin. Bands were visualized using a MINI HD6 image analyzer (UVITEC).

NOX activity assay

Cells were washed twice in ice-cold PBS and then scraped from the plate using the same solution. Subsequently, the cells were centrifuged at 4,000 rpm at 4°C for 10 min, and the resulting pellet was suspended in a buffer containing 20 mM KH2PO4, 1 mM EGTA, 150 mM sucrose, and a protease inhibitor mixture. The cell suspension was lysed on ice for 30 min. For the assay, 20 µl of lysate was mixed with 180 µl of an assay buffer containing 250 mM HEPES (pH 7.4), 250 µM lucigenin, 1.2 mM MgSO4 (7H2O), 120 mM NaCl, 1.75 mM CaCl2 (2H2O), 11 mM glucose, 0.5 mM EDTA, 5.9 mM KCl, and 200 µM NADPH for a duration of 10 sec. Photoemission in terms of RLU was measured using the GloMax®-Multi Jr Detection System (Promega, Madison, Wisconsin, USA) every minute for 15 min.

Membrane fractionation

MCF-7 cells (5×107) were pre-treated with GF for 1 h followed by incubation with TPA for an additional hour at 37°C. Subsequently, the cells were suspended in homogenization buffer (composed of 20 mM Tris-HCl, 2 mM EDTA, 5 mM EGTA, 5 mM DTT, and protease inhibitor; pH 7.5) and homogenized using a sonicator (5 times of 10 sec each at 10% amplitude), followed by a 30-min incubation on ice. The resulting cell lysate was then subjected to centrifugation at 16,000 × g for 15 min at 4°C to separate the soluble (cytosolic) fraction from the pellet (membrane) fraction. The pellet fraction was further treated with solubilization buffer (homogenization buffer supplemented with 1% NP-40) for 30 min on ice, followed by another centrifugation step at 16,000 × g for 15 min at 4°C.

Statistical analyses

Statistical analyses were performed using GraphPad Prism version 8.0 (GraphPad Software, San Diego, CA, USA). Data are presented as the mean and standard error of the mean. An unpaired Student's t-test was used to compare two groups, and one-way analysis of variance with Tukey's post hoc test was used to compare independent multiple groups. P<0.05 was considered to indicate a statistically significant difference. All experiments were performed at least in triplicate.

Results

Inhibition of NOX suppresses TPA-induced cell invasion and MMP-9 expression in MCF-7 cells

In our previous study, we reported that stimulation of MCF-7 cells with 20 nM TPA induces the expression of MMP-9 and cell invasion, which occurs through various intracellular signaling pathways (24,25). In addition, according to previous reports, breast cancer cells are treated with DPI at concentrations from 1 to 10 µM and APO at concentrations from 100 to 500 µM. Therefore, in our study, we used intermediate concentrations of 5 µM DPI and 300 µM APO (2630). To investigate the impact of NOXs on TPA-induced MMP-9 expression and cell invasion, MCF-7 cells were pre-treated with 5 µM DPI and 300 µM APO for 1 h, followed by stimulation with 20 nM TPA for 24 h. Western blotting and zymography revealed that NOX inhibitor treatment effectively inhibited the upregulation of TPA-induced MMP-9 protein expression and exocytosis in MCF-7 cells (Fig. 1A). RT-qPCR revealed that TPA increased MMP9 levels in MCF-7 cells, whereas NOX inhibitors mitigated TPA-induced MMP9 mRNA upregulation (Fig. 1B). Additionally, the Matrigel invasion assay confirmed that pretreatment with NOX inhibitors mitigated the increase in TPA-induced cell invasion (Fig. 1C). Therefore, the inhibition of NOX expression suppresses TPA-induced breast cancer cell invasion by inhibiting MMP-9 expression.

Role of NOX isotypes in TPA-induced expression in MCF-7 cells

NOXs contribute to breast cancer cell invasion by modulating MMP-9 expression and activity (Fig. 1). Among the members of the NOX family, we investigated which isotypes (NOX1-5) were implicated in TPA-induced MMP-9 protein expression in MCF-7 cells. For accurate comparison, we identified conditions wherein the expression of NOX mRNA decreased by approximately 50% following cell transfection with NOX-specific small interfering RNA (siRNA) (Fig. 2A). We also identified a decrease in protein expression by western blot analysis (Fig. S1). However, we could not detect the NOX3 band under our conditions, so we conducted the experiment based on mRNA expression levels. To assess the contribution of NOX isotypes in TPA-induced MMP-9 expression, MCF-7 cells were stimulated with TPA for 24 h following each condition of NOX isotype transfection. Western blot analysis revealed that transfection with NOX1 and NOX5 siRNA attenuated TPA-induced MMP-9 expression in MCF-7 cells (Fig. 2B). Therefore, TPA-induced MMP-9 expression in MCF-7 cells is mediated by NOX1 and NOX5.

Validation of NOX1 and NOX5 roles in cell invasion and ROS regulation in TPA-induced MCF-7 cells

We further validated the roles of NOX1 and NOX5 in cell invasion. Cell invasion was significantly increased in TPA-treated cells compared with that in control cells, and this increase was significantly reduced by the knockdown of NOX1 and NOX5 (Fig. 3). Furthermore, we investigated whether NOX1 and NOX5 knockdown mediates ROS regulation. The 5-(and-6)-chloromethyl-2′,7′-dichlorodihydrofluorescein diacetate (CM-H2DCFDA) fluorescence assay helped confirm that TPA treatment (20 nM) induced ROS production in MCF-7 cells, this effect was mitigated by knockdown of NOX1 and NOX5 (Fig. 4). Therefore, NOX1 and NOX5 inhibit TPA-induced cell invasion by regulating ROS production.

PKC modulation of NOX activity and MMP-9 expression in TPA-induced MCF-7 cells

TPA selectively activates PKC, acting as an upstream or downstream regulator to generate ROS (1113). Therefore, we used GF109203X (GF), a widely used PKC inhibitor, to confirm the involvement of PKC in TPA-induced ROS generation, MMP-9 expression, and MCF-7 cell activation. It is known that the activation of PKC by TPA involves the translocation of PKC isoforms from the cytosol to the plasma membrane (31). Pretreatment with GF was found to inhibit the membrane translocation of the PKC isoforms α, β, and δ, which was induced by TPA stimulation (Fig. S2). DPI was used as a positive control in this experiment as a representative NOX inhibitor (32). We observed that GF effectively inhibited TPA-induced ROS production (Fig. 5A) and MMP-9 expression and activation (Fig. 5B) in MCF-7 cells. Additionally, GF treatment attenuated the increase in TPA-induced NOX activity in MCF-7 cells (Fig. 5C). Therefore, NOX activity is mediated by PKC, which plays a pivotal role in regulating TPA-induced MMP-9 expression and invasion in MCF-7 cells.

Discussion

Breast cancer is a malignant tumor that is the leading cause of mortality among women (33). Metastasis to diverse organs, including bones, lungs, liver, brain, and kidneys, accounts for most breast cancer-related deaths (34). The initial event in cancer cell invasion and migration involves a decrease in the ECM, which poses biochemical and mechanical barriers to cell movement (6,35). The most important factors in ECM degradation are MMP expression and activity, which play pivotal roles in breast cancer (6,36). MMP is a family of proteases that play important roles in the development and progression of cancer; among these, MMP-9 is vital in tumor invasion and metastasis owing to its collagenase activity in ECM degradation (37). Breast cancer cells, stimulated by various factors, including TPA, increase MMP-9 expression by activating several intracellular signaling pathways (36,38). Following MMP-9 expression and activation, ECM loss in blood vessels or lymphatic walls facilitates cancer cell invasion into these systems, leading to metastasis to other organs. Therefore, regulation of MMP-9 expression is pivotal in controlling cancer metastasis.

Mitochondria and the NOX family (NOX1-5 and DUOX1/2) constitute two important sources of ROS production in cancer cells (39). NOX, a protein facilitating electron transport across biological membranes, generates superoxide in the plasma membrane, which is converted into hydrogen peroxide; this facilitates its entry into the cell in the form of superoxide or hydrogen peroxide, thus impacting various intracellular signaling mechanisms (40,41). NOX family members play crucial roles in various human cancer tissues (42,43). However, the role of NOX isotypes (NOX1-5) in TPA-induced breast cancer cell invasion remains unexplored. NOX1 is overexpressed in various human solid tumors, including colon cancer, prostate cancer, and melanoma (44,45), and it contributes to the regulation of cell invasion by regulating MMP-9 production and cell migration (46). NOX5 expression is increased in tumor tissues of patients with breast cancer, and it promotes breast cancer cell proliferation and metastasis (47,48). However, we lack reports on the importance of NOX1 and NOX5 in breast cancer invasion. We confirmed that the previously known NOX inhibitors, DPI and APO, inhibit MMP-9 expression and cell invasion in MCF-7 cells (Fig. 1). Furthermore, we confirmed that the inhibition of NOX1 and NOX5 among the NOXs is involved in MMP-9 expression and cell invasion in MCF-7 cells (Figs. 2 and 3).

TPA induces multiple signaling pathways in a PKC-dependent manner (49); PKC activation promotes tumor development and is associated with special cell functions, such as adhesion, invasion, and metastasis (50). PKC activation in breast cancer is strongly associated with increased invasion through the production and secretion of MMP-9 (24,25,51). Furthermore, ROS generation induces TPA-mediated migration and invasion (52). PKC activates NADPH oxidases, leading to the production of ROS (22,5357). Therefore, we confirmed the inhibitory effects of NOX1 and NOX5 suppression on TPA-induced ROS generation in MCF-7 cells (Fig. 4). Finally, we confirmed the inhibitory effects of PKC inhibitors on TPA-induced ROS production, MMP-9 expression, and NOX activation in MCF-7 cells (Fig. 5).

However, the current study has limitations as it lacks evidence from animal experiments and does not explore the role of DUOX1 and DUOX2 in breast cancer metastasis. Therefore, future studies incorporating animal models and investigating the involvement of additional NOX isoforms could provide a more comprehensive understanding of the mechanisms underlying breast cancer invasion.

In conclusion, our findings confirm that NOX1 and NOX5 mediate TPA-induced invasion of MCF-7 cells by regulating MMP-9 expression and activation; this is achieved mainly by modulating ROS generation via PKC. To the best of our knowledge, this is the first study to demonstrate that TPA-induced PKC-dependent-MCF-7 cell invasion is modulated by NOXs. Despite some limitations in our study, our findings highlight potential strategies for treating breast cancer metastasis via NOX1 and NOX5 regulation.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

This study was supported by Wonkwang University in 2022.

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

YRL and HJY designed and conceptualized the experiments. HKS, EMN and JMK performed the experiments and data collection. HKS wrote the original draft and revised the manuscript. JMK analyzed and generated the figures. YRL and HJY confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publications

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Jemal A, Murray T, Ward E, Samuels A, Tiwari RC, Ghafoor A, Feuer EJ and Thun MJ: Cancer statistics, 2005. CA Cancer J Clin. 55:10–30. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Anne N, Sulger E and Pallapothu R: Primary squamous cell carcinoma of the breast: A case report and review of the literature. J Surg Case Rep. 2019:rjz1822019. View Article : Google Scholar : PubMed/NCBI

3 

Zagelbaum NK, Ward MF II, Okby N and Karpoff H: Invasive ductal carcinoma of the breast with osteoclast-like giant cells and clear cell features: A case report of a novel finding and review of the literature. World J Surg Oncol. 14:2272016. View Article : Google Scholar : PubMed/NCBI

4 

Berman AT, Thukral AD, Hwang WT, Solin LJ and Vapiwala N: Incidence and patterns of distant metastases for patients with early-stage breast cancer after breast conservation treatment. Clin Breast Cancer. 13:88–94. 2013. View Article : Google Scholar : PubMed/NCBI

5 

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

6 

Woessner JF Jr: Matrix metalloproteinases and their inhibitors in connective tissue remodeling. FASEB J. 5:2145–2154. 1991. View Article : Google Scholar : PubMed/NCBI

7 

Mizutani K, Kofuji K and Shirouzu K: The significance of MMP-1 and MMP-2 in peritoneal disseminated metastasis of gastric cancer. Surg Today. 30:614–621. 2000. View Article : Google Scholar : PubMed/NCBI

8 

Zeigler ME, Chi Y, Schmidt T and Varani J: Role of ERK and JNK pathways in regulating cell motility and matrix metalloproteinase 9 production in growth factor-stimulated human epidermal keratinocytes. J Cell Physiol. 180:271–284. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Hozumi A, Nishimura Y, Nishiuma T, Kotani Y and Yokoyama M: Induction of MMP-9 in normal human bronchial epithelial cells by TNF-alpha via NF-kappa B-mediated pathway. Am J Physiol Lung Cell Mol Physiol. 281:L1444–L1452. 2001. View Article : Google Scholar : PubMed/NCBI

10 

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

11 

Lee HB, Yu MR, Song JS and Ha H: Reactive oxygen species amplify protein kinase C signaling in high glucose-induced fibronectin expression by human peritoneal mesothelial cells. Kidney Int. 65:1170–1179. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Wu WS, Tsai RK, Chang CH, Wang S, Wu JR and Chang YX: Reactive oxygen species mediated sustained activation of protein kinase C alpha and extracellular signal-regulated kinase for migration of human hepatoma cell Hepg2. Mol Cancer Res. 4:747–758. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Traore K, Sharma RB, Burek CL and Trush MA: Role of ROS and MAPK in TPA-induced ICAM-1 expression in the myeloid ML-1 cell line. J Cell Biochem. 100:1010–1021. 2007. View Article : Google Scholar : PubMed/NCBI

14 

McCubrey JA, Lahair MM and Franklin RA: Reactive oxygen species-induced activation of the MAP kinase signaling pathways. Antioxid Redox Signal. 8:1775–1789. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Huang L, Lin H, Chen Q, Yu L and Bai D: MPPa-PDT suppresses breast tumor migration/invasion by inhibiting Akt-NF-κB-dependent MMP-9 expression via ROS. BMC Cancer. 19:11592019. View Article : Google Scholar : PubMed/NCBI

16 

Lee GH, Jin SW, Kim SJ, Pham TH, Choi JH and Jeong HG: Tetrabromobisphenol A induces MMP-9 expression via NADPH Oxidase and the activation of ROS, MAPK, and Akt pathways in human breast cancer MCF-7 Cells. Toxicol Res. 35:93–101. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Hsu TC, Young MR, Cmarik J and Colburn NH: Activator protein 1 (AP-1)- and nuclear factor kappaB (NF-kappaB)-dependent transcriptional events in carcinogenesis. Free Radic Biol Med. 28:1338–1348. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Huang Q, Shen HM and Ong CN: Inhibitory effect of emodin on tumor invasion through suppression of activator protein-1 and nuclear factor-kappaB. Biochem Pharmacol. 68:361–371. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Savaraj N, Wei Y, Unate H, Liu PM, Wu CJ, Wangpaichitr M, Xia D, Xu HJ, Hu SX and Tien Kuo M: Redox regulation of matrix metalloproteinase gene family in small cell lung cancer cells. Free Radic Res. 39:373–381. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Karin M: The regulation of AP-1 activity by mitogen-activated protein kinases. J Biol Chem. 270:16483–16486. 1995. View Article : Google Scholar : PubMed/NCBI

21 

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

22 

Brandes RP, Weissmann N and Schroder K: Nox family NADPH oxidases: Molecular mechanisms of activation. Free Radic Biol Med. 76:208–226. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Schmittgen TD and Livak KJ: Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 3:1101–1108. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Kim JM, Park J, Noh EM, Song HK, Kang SY, Jung SH, Kim JS, Park BH, Lee YR and Youn HJ: Bruton's agammaglobulinemia tyrosine kinase (Btk) regulates TPA-induced breast cancer cell invasion via PLCγ2/PKCβ/NF-κB/AP-1-dependent matrix metalloproteinase-9 activation. Oncol Rep. 45:562021. View Article : Google Scholar : PubMed/NCBI

25 

Noh EM, Park YJ, Kim JM, Kim MS, Kim HR, Song HK, Hong OY, So HS, Yang SH, Kim JS, et al: Fisetin regulates TPA-induced breast cell invasion by suppressing matrix metalloproteinase-9 activation via the PKC/ROS/MAPK pathways. Eur J Pharmacol. 764:79–86. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Piszczatowska K, Przybylska D, Sikora E and Mosieniak G: Inhibition of NADPH oxidases activity by diphenyleneiodonium chloride as a mechanism of senescence induction in human cancer cells. Antioxidants (Basel). 9:12482020. View Article : Google Scholar : PubMed/NCBI

27 

Ren G, Luo W, Sun W, Niu Y, Ma DL, Leung CH, Wang Y, Lu JJ and Chen X: Psoralidin induced reactive oxygen species (ROS)-dependent DNA damage and protective autophagy mediated by NOX4 in breast cancer cells. Phytomedicine. 23:939–947. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Nasimian A, Farzaneh P, Tamanoi F and Bathaie SZ: Cytosolic and mitochondrial ROS production resulted in apoptosis induction in breast cancer cells treated with Crocin: The role of FOXO3a, PTEN and AKT signaling. Biochem Pharmacol. 177:1139992020. View Article : Google Scholar : PubMed/NCBI

29 

D'Anneo A, Carlisi D, Emanuele S, Buttitta G, Di Fiore R, Vento R, Tesoriere G and Lauricella M: Parthenolide induces superoxide anion production by stimulating EGF receptor in MDA-MB-231 breast cancer cells. Int J Oncol. 43:1895–1900. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Kaikai S, Lu F, Xie J, Wu M, Cai B, Liu Y, Zhang H, Tan H, Pan Y and Xu H: Cambogin exerts anti-proliferative and pro-apoptotic effects on breast adenocarcinoma through the induction of NADPH oxidase 1 and the alteration of mitochondrial morphology and dynamics. Oncotarget. 7:50596–50611. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Jacobson PB, Kuchera SL, Metz A, Schächtele C, Imre K and Schrier DJ: Anti-inflammatory properties of Gö 6850: A selective inhibitor of protein kinase C. J Pharmacol Exp Ther. 275:995–1002. 1995.PubMed/NCBI

32 

Augsburger F, Filippova A, Rasti D, Seredenina T, Lam M, Maghzal G, Mahiout Z, Jansen-Dürr P, Knaus UG, Doroshow J, et al: Pharmacological characterization of the seven human NOX isoforms and their inhibitors. Redox Biol. 26:1012722019. View Article : Google Scholar : PubMed/NCBI

33 

Siegel R, Ma J, Zou Z and Jemal A: Cancer statistics, 2014. CA Cancer J Clin. 64:9–29. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Redig AJ and McAllister SS: Breast cancer as a systemic disease: A view of metastasis. J Intern Med. 274:113–126. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Jiang WG, Sanders AJ, Katoh M, Ungefroren H, Gieseler F, Prince M, Thompson SK, Zollo M, Spano D, Dhawan P, et al: Tissue invasion and metastasis: Molecular, biological and clinical perspectives. Semin Cancer Biol. 35 (Suppl):S244–S275. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Duffy MJ, Maguire TM, Hill A, McDermott E and O'Higgins N: Metalloproteinases: Role in breast carcinogenesis, invasion and metastasis. Breast Cancer Res. 2:252–257. 2000. View Article : Google Scholar : PubMed/NCBI

37 

Scorilas A, Karameris A, Arnogiannaki N, 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

38 

Yan C and Boyd DD: Regulation of matrix metalloproteinase gene expression. J Cell Physiol. 211:19–26. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Landry WD and Cotter TG: ROS signalling, NADPH oxidases and cancer. Biochem Soc Trans. 42:934–938. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Jiang F, Zhang Y and Dusting GJ: NADPH oxidase-mediated redox signaling: Roles in cellular stress response, stress tolerance, and tissue repair. Pharmacol Rev. 63:218–242. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Bedard K and Krause KH: The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol Rev. 87:245–313. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Lambeth JD: NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol. 4:181–189. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Kamata T: Roles of Nox1 and other Nox isoforms in cancer development. Cancer Sci. 100:1382–1388. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Block K and Gorin Y: Aiding and abetting roles of NOX oxidases in cellular transformation. Nat Rev Cancer. 12:627–637. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Deep G, Kumar R, Jain AK, Dhar D, Panigrahi GK, Hussain A, Agarwal C, El-Elimat T, Sica VP, Oberlies NH and Agarwal R: Graviola inhibits hypoxia-induced NADPH oxidase activity in prostate cancer cells reducing their proliferation and clonogenicity. Sci Rep. 6:231352016. View Article : Google Scholar : PubMed/NCBI

46 

Shinohara M, Adachi Y, Mitsushita J, Kuwabara M, Nagasawa A, Harada S, Furuta S, Zhang Y, Seheli K, Miyazaki H and Kamata T: Reactive oxygen generated by NADPH oxidase 1 (Nox1) contributes to cell invasion by regulating matrix metalloprotease-9 production and cell migration. J Biol Chem. 285:4481–4488. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Juhasz A, Ge Y, Markel S, Chiu A, Matsumoto L, van Balgooy J, Roy K and Doroshow JH: Expression of NADPH oxidase homologues and accessory genes in human cancer cell lines, tumours and adjacent normal tissues. Free Radic Res. 43:523–532. 2009. View Article : Google Scholar : PubMed/NCBI

48 

Dho SH, Kim JY, Lee KP, Kwon ES, Lim JC, Kim CJ, Jeong D and Kwon KS: STAT5A-mediated NOX5-L expression promotes the proliferation and metastasis of breast cancer cells. Exp Cell Res. 351:51–58. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Lim PS, Sutton CR and Rao S: Protein kinase C in the immune system: From signalling to chromatin regulation. Immunology. 146:508–522. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Parekh DB, Ziegler W and Parker PJ: Multiple pathways control protein kinase C phosphorylation. EMBO J. 19:496–503. 2000. View Article : Google Scholar : PubMed/NCBI

51 

Kim JM, Park J, Noh EM, Song HK, Kang SY, Jung SH, Kim JS, Youn HJ and Lee YR: Downregulation of matriptase suppresses the PAR-2/PLCγ2/PKC-mediated invasion and migration abilities of MCF-7 breast cancer cells. Oncol Rep. 46:2472021. View Article : Google Scholar : PubMed/NCBI

52 

Wu WS: The signaling mechanism of ROS in tumor progression. Cancer Metastasis Rev. 25:695–705. 2006. View Article : Google Scholar : PubMed/NCBI

53 

Eid BG, Abu-Sharib AT, El-Bassossy HM, Balamash K and Smirnov SV: Enhanced calcium entry via activation of NOX/PKC underlies increased vasoconstriction induced by methylglyoxal. Biochem Biophys Res Commun. 506:1013–1018. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Cui X, Li X, He Y, Yu J, Fu J, Song B and Zhao RC: Combined NOX/ROS/PKC signaling pathway and metabolomic analysis reveals the mechanism of TRAM34-Induced endothelial progenitor cell senescence. Stem Cells Dev. 30:671–682. 2021. View Article : Google Scholar : PubMed/NCBI

55 

Chen F, Yu Y, Haigh S, Johnson J, Lucas R, Stepp DW and Fulton DJ: Regulation of NADPH oxidase 5 by protein kinase C isoforms. PLoS One. 9:e884052014. View Article : Google Scholar : PubMed/NCBI

56 

Lee TH, Chen JL, Liu PS, Tsai MM, Wang SJ and Hsieh HL: Rottlerin, a natural polyphenol compound, inhibits upregulation of matrix metalloproteinase-9 and brain astrocytic migration by reducing PKC-delta-dependent ROS signal. J Neuroinflammation. 17:1772020. View Article : Google Scholar : PubMed/NCBI

57 

Brandes RP and Schroder K: NOXious phosphorylation: Smooth muscle reactive oxygen species production is facilitated by direct activation of the NADPH oxidase Nox1. Circ Res. 115:898–900. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2024
Volume 30 Issue 4

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
Song H, Kim J, Noh E, Youn H and Lee Y: Role of NOX1 and NOX5 in protein kinase C/reactive oxygen species‑mediated MMP‑9 activation and invasion in MCF‑7 breast cancer cells. Mol Med Rep 30: 188, 2024.
APA
Song, H., Kim, J., Noh, E., Youn, H., & Lee, Y. (2024). Role of NOX1 and NOX5 in protein kinase C/reactive oxygen species‑mediated MMP‑9 activation and invasion in MCF‑7 breast cancer cells. Molecular Medicine Reports, 30, 188. https://doi.org/10.3892/mmr.2024.13312
MLA
Song, H., Kim, J., Noh, E., Youn, H., Lee, Y."Role of NOX1 and NOX5 in protein kinase C/reactive oxygen species‑mediated MMP‑9 activation and invasion in MCF‑7 breast cancer cells". Molecular Medicine Reports 30.4 (2024): 188.
Chicago
Song, H., Kim, J., Noh, E., Youn, H., Lee, Y."Role of NOX1 and NOX5 in protein kinase C/reactive oxygen species‑mediated MMP‑9 activation and invasion in MCF‑7 breast cancer cells". Molecular Medicine Reports 30, no. 4 (2024): 188. https://doi.org/10.3892/mmr.2024.13312