Curcumin inhibits superoxide dismutase-induced epithelial-to-mesenchymal transition via the PI3K/Akt/NF-κB pathway in pancreatic cancer cells

  • Authors:
    • Wei Li
    • Zhengdong Jiang
    • Xue Xiao
    • Zheng Wang
    • Zheng Wu
    • Qingyong Ma
    • Lei Cao
  • View Affiliations

  • Published online on: March 1, 2018     https://doi.org/10.3892/ijo.2018.4295
  • Pages: 1593-1602
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Curcumin is a natural polyphenol compound derived from turmeric. It possesses multiple pharmacological properties, including antioxidant, anti-inflammatory and anti-tumor progression properties. Our recent study demonstrated that superoxide dismutase (SOD)-dependent production of hydrogen peroxide (H2O2) promoted the invasive and migratory activity of pancreatic cancer cells. However, whether curcumin suppresses SOD-induced cancer progression and the related mechanisms remains unclear. Since epithelial‑to-mesenchymal transition (EMT) plays a key role in tumor metastasis, the aim of the present study was to examine whether curcumin intervenes with SOD-induced EMT in pancreatic cancer and the underlying mechanism. The human pancreatic cancer cells BxPC-3 and Panc-1 were exposed to SOD in the presence or absence of curcumin, catalase (CAT, a scavenger of H2O2), or LY 294002 [a phosphoinositide-3 kinase (PI3K) inhibitor]. Intracellular reactive oxygen species (ROS) and H2O2 were evaluated by 2,7-dichlorodihydrofluorecein diacetate and H2O2 assay, respectively. The activation of p-Akt and p-nuclear factor (NF)-κB were examined by western blotting. The migratory and invasive abilities of pancreatic cancer cells were tested by the wound healing and Transwell invasion assays. The expression of E-cadherin, N-cadherin and vimentin (EMT-related genes) were measured by reverse transcription-quantitative polymerase chain reaction and western blotting at the mRNA and protein levels, respectively. The findings of the present study demonstrated that curcumin decreased SOD-induced production of ROS and H2O2 in BxPC-3 and Panc-1 cells. Curcumin was able to suppress SOD-induced invasion and migration, and it also regulated the expression of the above‑mentioned EMT-related genes and cell morphology. SOD-induced cell invasion was also inhibited by catalase and LY 294002. Furthermore, the levels of p-Akt and p-NF-κB caused by SOD could be offset by treatment with curcumin and LY 294002. To summarize, these results demonstrated that curcumin was able to prevent SOD-driven H2O2-induced pancreatic cancer metastasis by blocking the PI3K/Akt/NF-κB signaling pathway. The use of curcumin to inhibit the H2O2/Akt/NF-κB axis may be a promising therapeutic approach to the treatment of patients with pancreatic cancer.

Introduction

Pancreatic cancer is a highly aggressive and lethal human malignancy, with a very low 5-year survival rate worldwide (8%). The low survival rate is partly attributed to the fact that over half of the cases are diagnosed at an advanced stage, for which the 5-year survival may be as low as 3% (1). An estimated 53,670 new pancreatic cancer cases were diagnosed in 2017, which may result in 43,090 cancer-related deaths in the United States (1). In China, pancreatic cancer is the seventh most fatal disease, with an even lower 5-year survival rate (4.1%) (2). The routine treatments include surgery, radiation and chemotherapy (with gemcitabine, 5-fluorouracil or their combination). However, ~80% of pancreatic cancer subjects are diagnosed when the tumor is unresectable and/or has developed distant metastasis, and acquired resistance to chemotherapeutic treatment always develops during the course of the disease (3). Therefore, more comprehensive and effective interventions are urgently required to improve the treatment outcome of pancreatic cancer patients.

Curcumin is a natural polyphenol compound derived from turmeric. It possesses several biological properties, including anti-inflammatory and anti-oxidant properties, and also plays a key role in inhibiting the initiation, progression and metastasis of several tumors (4). Curcumin exerts its anticancer effects by targeting multiple intracellular signaling pathways, including mitogen-activated protein kinase (MAPK), nuclear factor (NF)-κB, Akt and Wnt/β-catenin, among others (57). In addition, we recently demonstrated that curcumin inhibited hypoxia-induced epithelial-to-mesenchymal transition (EMT) in pancreatic cancer cells via blockade of the hedgehog signaling pathway (8).

EMT has been recognized not only as a physiological mechanism in mammalian embryonic development and tissue remodeling, but also as an important phenomenon observed in tumorigenesis and cancer development (9). During EMT, epithelial cells lose their polarity, cell-cell tight junctions and adhesive connections, and acquire mesenchymal characteristics (10). The hallmark of EMT is the loss of E-cadherin expression (an epithelial marker) and the gain of vimentin and N-cadherin expression (mesenchymal markers) (10). We previously demonstrated that several factors could induce EMT in pancreatic cancer cells, including hypoxic (8) and hyperglycemic environment (11), as well as the induction of superoxide dismutase (SOD) (12).

Reactive oxygen species (ROS), including hydrogen peroxide (H2O2), are a group of chemically reactive molecules derived from oxygen. Intrinsic antioxidant enzymes play a crucial role in the regulation of oxidative stress in cells. SOD is one of the primary cellular antioxidants, and it can catalyze the conversion of superoxide anion to H2O2, which is cleared by catalase (CAT) (13). Due to the cytoprotective effects of SOD, its overexpression has been associated with increased incidence of tumor metastasis (14). In our previous study, we verified that the invasive ability and EMT of pancreatic cancer cells was aggravated by SOD-dependent production of ROS via the extracellular signal-regulated kinase (ERK)/NF-κB signaling pathway (12). However, whether curcumin can repress the SOD-induced progression of pancreatic cancer, and the possible underlying mechanisms, have not been fully elucidated.

The aim of the present study was to verify our hypothesis and determine whether curcumin has the potential to suppress SOD-driven H2O2-induced invasive and migratory abilities and inhibit EMT in pancreatic cancer cells. We also aimed to examine the effect of curcumin on SOD-induced activation of the phosphoinositide-3 kinase (PI3K)/Akt signaling pathway and the transcription factor NF-κB, to determine whether using curcumin to inhibit the H2O2/Akt/NF-κB axis may be a promising approach to the treatment of pancreatic carcinoma.

Materials and methods

Cell culture and reagents

The human pancreatic cancer cell lines BxPC-3 and Panc-1 were purchased from the American Type Culture Collection (Manassas, VA, USA). The cells were cultured in Dulbecco's modified Eagle's medium (DMEM), which contains 10% dialyzed heat-inactivated fetal bovine serum (FBS), 100 U/ml penicillin and 100 µg/ml streptomycin in a humidified atmosphere of 5% CO2 at 37°C. The cells exponentially grew in complete medium and treated with 400 U/ml SOD, with or without 20 µM curcumin, 400 U/ml CAT or 10 µM LY 294002 (a PI3K inhibitor) for the indicated time intervals, according to the experimental protocol. DMEM and FBS were purchased from Gibco; Thermo Fisher Scientific (Grand Island, NY, USA). Curcumin, CuZnSOD, CAT and LY294002 were purchased from Sigma-Aldrich; Merck KGaA (St. Louis, MO, USA). The hydrogen peroxide assay kit and the ROS assay kit were obtained from Beyotime (Jinan, China). Millicell culture plate inserts were purchased from Millipore (Bedford, MA, USA). Matrigel was purchased from BD Biosciences (Bedford, MA, USA). Primary antibodies [dilution 1:100 in phosphate-buffered saline (PBS)-Tween-20] against E-cadherin (sc-52328), N-cadherin (sc-53488) and vimentin (sc-66002) were obtained from Santa Cruz Biotechnology (Santa Cruz, CA, USA). The anti-Akt (no. 9272), anti-phospho-Akt (anti-p-Akt, Ser473, no. 4060), anti-NF-κB (no. 6956) and anti-phospho-NF-κB (anti-p-NF-κB, Ser468, no. 3039) antibodies (dilution 1:200 in PBS-Tween-20) were obtained from Cell Signaling Technology (Beverly, MA, USA). Other reagents were purchased from common commercial sources. All drug solutions were freshly prepared on the day of testing.

Measurement of intracellular ROS

The level of intracellular ROS was examined using the ROS assay kit according to the manufacturer's instructions. Briefly, pancreatic cancer cells were incubated with 2,7-dichlorodihydrofluorescein diacetate (DCFDA) for 30 min. After washing 3 times, fluorescence intensity was measured using a fluorometer (Becton-Dickinson, Franklin Lakes, NJ, USA) with excitation at 488 nm and emission at 525 nm.

Hydrogen peroxide assay

The level of intracellular H2O2 was tested using a hydrogen peroxide assay kit. In this kit, H2O2 oxidizes ferrous (Fe2+) to ferric (Fe3+) ions; then, Fe3+ ions combine with the indicator dye xylenol orange to form a complex and produce a visible purple-colored complex, which may be measured using a microplate reader at a wavelength of 560–590 nm (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Transwell Matrigel invasion assay

The 8.0-µm pore inserts were covered with 25 µl Matrigel. BxPC-3 and Panc-1 cells were subjected to 24 h serum starvation and were then suspended in DMEM supplemented with 1% FBS in the upper chamber at a density of 5×104, with or without SOD, SOD with curcumin, CAT or LY 294002. In addition, 500 ml DMEM with 20% FBS were placed in the lower chamber. The Matrigel invasion chamber was placed in a humidified tissue incubator for 48 h. The non-invading cells were cleared away from the top surface using a cotton swab and the filter was rinsed with phosphate-buffered saline, fixed and stained with crystal violet. Finally, the stained cells on the bottom surface were counted to evaluate the invasive capacity of cancer cells. Three random fields were captured at a magnification of ×20 (n=3).

Wound healing assay

Cells were seeded in 24-well plates (1.0×105 cells/500 µl). After the cells had grown to a 90–100% confluence, a sterile pipette tip was used to create a scratch wound. Cellular debris was cleared away and the remaining cells were allowed to grow and migrate in the plate for 24 h. Images were captured at time points 0 and 24 h post-wounding by a Nikon Diaphot TMD inverted microscope (magnification, ×10). The relative distance traveled by the leading edge from 0 to 24 h was assessed using Photoshop software (n=5).

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

Total RNA was extracted from cancer cells using the Fastgen200 RNA isolation system (Fastgen, Shanghai, China). cDNA was reverse-transcribed from RNA using the Fermentas RevertAid™ Kit (MBI Fermentas, Burlington, ON, Canada). The primer sequences were as follows: E-cadherin, F: 5′-ATT CTGATTCTGCTGCTCTTG-3′ and R: 5′-AGTCCTGGTCCTC TTCTCC-3′; N-cadherin, F: 5′-TGTTTGACTATGAAGGCAG TGG-3′ and R: 5′-TCAGTCATCACCTCCACCAT-3′; vimentin, F: 5′-AATGACCGCTTCGCCAAC-3′ and R: 5′-CCGCATCTCC TCCTCGTAG-3′; and β-actin, F: 5′-GACTTAGTTGCGTTACAC CCTTTCT-3′ and R: 5′-GAACGGTGAAGGT GACAGCAGT-3′.

The PCR conditions were as follows: 30 sec at 95°C, followed by 40 cycles at 95°C for 5 sec, 60°C for 30 sec and 72°C for 30 sec. A dissociation curve analysis was applied after each RT-qPCR experiment. The relative gene expression was calculated using the previously described 2−ΔΔCq method (15).

Western blotting

Identical amounts of protein from cancer cells were loaded on a denaturing sodium dodecyl sulfate-polyacrylamide gel. After gel electrophoresis, the protein was transferred onto nitrocellulose membranes. Subsequently, the membranes were initially blocked with 5% non-fat dry milk in Tris-buffered saline (TBS) for 2 h, followed by incubation with different primary antibodies (E-cadherin, N-cadherin, vimentin, Akt, p-Akt, NF-κB, p-NF-κB and β-actin antibodies) at 4°C overnight. The membranes were then incubated with secondary goat anti-mouse or goat anti-rabbit antibodies (Sigma-Aldrich; Merck KGaA) for 2 h at room temperature. Immunopositive bands were developed using an enhanced chemiluminescence (ECL) detection system (Amersham, Piscataway, NJ, USA). All analyses were conducted in triplicate.

Statistical analysis

Statistical analysis was performed using SPSS software, version 17.0 (SPSS Inc., Chicago, IL, USA). Data are presented as the means ± standard error of the mean of three replicate assays. Differences between the groups were analyzed by analysis of variance with Dunnett's post-hoc test. Statistical significance was set at P<0.05. All experiments were repeated independently at least three times.

Results

Curcumin inhibits SOD-induced oxidative stress in pancreatic cancer cells

ROS caused by oxidative stress damage cellular DNA, proteins and lipids, and produce toxic and highly mutagenic metabolites that may modify tumor behavior (16). We previously demonstrated that SOD stimulated the production of H2O2 in pancreatic cancer cells (12). As curcumin possess antioxidant properties, in the present study, we first examined the effects of curcumin on the production of ROS and H2O2 in BxPC-3 and Panc-1 cells following treatment with SOD. Our results revealed that SOD significantly increased the DCFDA staining intensity (Fig. 1) and the level of H2O2 (Fig. 2) in pancreatic cancer cells, while curcumin was able to counterbalance these effects at 24 and 48 h in both cancer cell types.

Curcumin downregulates the activation of the Akt/NF-κB signaling pathway

The PI3K/Akt pathway plays an important role in numerous cellular processes, such as cell metabolism, survival, differentiation, proliferation, motility and angiogenesis (17). It has been proven that cancer cell signaling is mediated by PI3K/Akt via activation of the transcription factor NF-κB, which is associated with cell proliferation, migration and invasion (18).

We previously demonstrated that SOD promotes activation of the ERK/NF-κB signaling pathway (12). The findings of the present study indicated that SOD could also upregulate the Akt/NF-κB signaling pathway, as the phosphorylated levels of both Akt and NF-κB genes were found to be significantly elevated following SOD addition (Fig. 3A). When SOD was added to the cell in culture medium along with curcumin, the expression of p-Akt and p-NF-κB was strongly downregulated (Fig. 3A). Our results also demonstrated that curcumin alone was able to inhibit the expression of p-AKT and p-NF-κB (Fig. 3B). In addition, LY 294002, a PI3K inhibitor, also suppressed the expression of both p-Akt and p-NF-κB, indicating that NF-κB is located downstream of the PI3K/Akt pathway (Fig. 3C).

Curcumin inhibits SOD-induced invasion of pancreatic cancer cells

Malignant tumors are often characterized by metastasis, which is dissemination of cells from a primary site to colonize distant organs. The ability of cancer cells to migrate and invade other tissues are two important steps in the development of metastasis. To verify whether curcumin is able to affect SOD-induced cell invasion in pancreatic cancer cells, a Transwell invasion assay was performed. As shown in Fig. 4, the mean number of cells invading into the lower chamber increased in the presence of SOD after a 48-h incubation, and this increase was inhibited by co-treatment with curcumin. In addition, CAT and LY 294002 also suppressed the effect of SOD, indicating that SOD-induced invasion is associated with the H2O2/Akt axis (Fig. 4).

Curcumin suppresses SOD-induced migration of pancreatic cancer cells

A classic wound healing assay was next conducted to assess the effect of curcumin on SOD-induced cell motility in pancreatic cancer cells. Our results revealed that the migration of cancer cells was significantly promoted by SOD after a 24-h incubation. Delayed wound closure was observed following treatment with curcumin in the two cancer cell types. The increase in cell migration was also inhibited by co-treatment with CAT and LY 294002. Therefore, curcumin likely exerts its inhibitory effects on cancer cell motility through suppression of the H2O2/Akt/NF-κB axis (Fig. 5).

Curcumin inhibits SOD-promoted EMT in pancreatic cancer cells

EMT is considered a prerequisite for cells to adopt a motile and invasive phenotype and eventually become metastatic. EMT includes four important steps: Loss of epithelial cell adhesion, expression of mesenchymal proteins and acquisition of a mesenchymal-like phenotype, degradation of the basement membrane, and enhanced cell migration and invasion, which facilitate tumor cell invasion into the stroma and entrance into the circulation (10). Our previous study demonstrated that SOD-induced H2O2 production is able to promote EMT in pancreatic cancer cells, resulting in increased cell motility and invasion via activation of the ERK signaling pathway (12). In the present study, we demonstrated that cancer cell morphology changed from a typical epithelial phenotype to a mesenchymal phenotype following SOD treatment for 48 h, which was counterbalanced by curcumin (Fig. 6A).

A hallmark of EMT includes a marked decline in E-cadherin expression (a cell-cell adhesion molecule) and increase in vimentin and N-cadherin expression (mesenchymal markers) (11). To further verify the effect of curcumin on SOD-induced EMT, the mRNA and protein expression levels of several EMT-related factors (E-cadherin, N-cadherin and vimentin) were assessed after the cells were treated with SOD in the presence or absence of curcumin. As shown in Fig. 6B, SOD decreased the mRNA level of E-cadherin and increased the mRNA level of N-cadherin and vimentin, whereas curcumin significantly reversed these effects. As shown in Fig. 6C, SOD exposure could modulate the protein levels of EMT-related factors, while curcumin suppressed these effects of SOD.

To summarize the abovementioned results, SOD was able to induce EMT progression and facilitate tumor invasion and migration via the production of H2O2 in BxPC-3 and Panc-1 pancreatic cancer cells, whereas these effects were counterbalanced by curcumin treatment in both types of cells.

Discussion

An increasing volume of evidence suggests that EMT plays a key role in cancer progression, cancer stem cell intravasation, establishment of metastasis and treatment resistance, resulting in a marked increase in disease aggressiveness and poorer disease outcome and overall patient survival (19). Pancreatic cancer, an aggressive and lethal malignant disease, is predicted to become the leading cause of cancer-related mortality in the USA by 2050 (20). The poor prognosis of pancreatic cancer patients is mainly attributed to the metastatic predilection of cancer cells. Although some tumors appear to be resectable, in reality surgery is not curative, due to the microscopic systemic spread that occurs prior to surgical resection (21). Our previous studies demonstrated that several factors may induce EMT in pancreatic cancer cells, thereby promoting tumor progression, including a hypoxic environment (8), a hyperglycemic environment (11), as well as the induction SOD (12). The aim of the present study was to determine whether curcumin has the potential to inhibit SOD-induced cell invasion and migration and EMT in the BxPC-3 and Panc-1 cell lines, and elucidate the possible related mechanism.

Our findings demonstrated that curcumin significantly decreased SOD-induced production of ROS and H2O2 in pancreatic cancer cells. Curcumin, CAT (a scavenger of H2O2) and LY 294002 (a PI3K inhibitor) were able to suppress SOD-induced migration and invasion of BxPC-3 and Panc-1 cells. SOD-modulated cancer cell morphology and the expression of E-cadherin, N-cadherin and vimentin were markedly affected by curcumin. In addition, SOD-induced activation of the PI3K/Akt signaling pathway and the transcription factor NF-κB were also suppressed by curcumin. It is known that H2O2 induces ERK and Akt activation through a number of mechanisms. For example, Nrf2 is a transcription factor that plays a key role in controlling the response to oxidative stress by regulating antioxidant enzymes. The activation of Nrf2 may also be mediated by additional signal transduction pathways, such as ERK, AMPK or PI3K/Akt, exerting antioxidant effects, which mediate enhanced resistance to oxidative stress (22). The thioredoxin/thioredoxin reductase/TXNIP system is also involved in oxidative stress and the ERK pathway, as it was previously proven that hyperglycemia-induced TXNIP expression is involved in diabetes-mediated oxidative stress in pancreatic cancer via the p38, MAPK and ERK pathways (23). Previous studies demonstrated that H2O2 contributes to both copper-zinc SOD (CuZnSOD)- and manganese SOD (MnSOD)-induced cancer progression through activation of Akt and ERK (12,24,25), while curcumin suppresses H2O2-induced migration and invasion of pancreatic cancer cells (26). Our results revealed that curcumin suppresses SOD-induced EMT in pancreatic cancer, which may be associated with the H2O2/Akt/NF-κB axis.

ROS and the activation of redox-sensitive signaling pathways are important participants in the development of neoplasms (27). SOD is a type of primary cellular antioxidant catalyzing the conversion of superoxide to H2O2, which has been proven to favor tumor progression (12). There are three members of the SOD family present in mammals, namely CuZnSOD, MnSOD and extracellular SOD (EC-SOD), which are located in different places inside or outside the cell (28). Epidemiological evidence has indicated that increased levels of SOD were observed in several tumor types along with tumor progression from early-stage (non-invasive) to late-stage (metastatic) (29). SOD polymorphism is associated with increased risk of prostate cancer, esophageal cancer, non-Hodgkin lymphoma, lung cancer and colorectal cancer (30,31). The MnSOD-1221G>A AA genotype carriers exhibited a significantly increased risk of pancreatic cancer among those with a low dietary vitamin E intake (32). In addition, both in vitro and in vivo studies demonstrated that cancer cells containing elevated levels of SOD exhibit a propensity for metastasis, proliferation and resistance to apoptosis. Hart et al (33) revealed that SOD-induced H2O2 sustained the Warburg effect through AMPK-dependent signaling, enabling cancer cell survival. Chronic inflammation is a major activator of the metastatic cascade. Tumor-associated inflammation also participates in the regulation of EMT, which contributes to cancer invasion and metastasis. Yi et al (34) recently reported that SOD may favor inflammation-mediated EMT and migration of tumor cells in AFG1-induced lung adenocarcinoma. We have previously reported that SOD-dependent production of ROS promoted the invasion of pancreatic cancer cells (12). The present study demonstrated that curcumin effectively inhibited SOD-induced EMT in the BxPC-3 and Panc-1 pancreatic cancer cell lines.

Curcumin is a bioactive natural compound, which has been proven to restrain initiation, progression and metastasis of multifarious tumors, including pancreatic cancer (8). More importantly, curcumin is associated with minimal toxicity and it is safe at a high dose, as demonstrated by human clinical trials, in contrast with conventional cytotoxic drugs (35). Curcumin exerts its anticancer effects via multiple signaling pathways, such as Notch, mammalian target of rapamycin (mTOR), MAPK and NF-κB (5,3639). Zhou et al (36) proved that curcumin suppressed pancreatic cancer cell growth, induced apoptosis and cell cycle arrest, weakened clonogenic potential, and inhibited migration and invasion via suppression of YAP/TAZ and Notch signaling. Treatment with curcumin also effectively attenuated tobacco smoke-induced activation of ERK and JNK MAPK pathways, AP-1 proteins and EMT alterations in mouse liver (5). Curcumin was also found to be able to induce autophagy and activate lysosomal function via its inhibitory effects on the Akt-mTOR signaling pathway and via direct targeting and activation of TFEB (37). In addition, curcumin exerted its anticancer effects both alone and in combination with other anticancer drugs. It has been proven that curcumin promotes the anticancer effects of gemcitabine via suppression of cancer cell proliferation, angiogenesis and inhibition of the NF-κB pathway in a pancreatic cancer model (38). A recent study indicated that co-treatment with metformin and curcumin not only induced apoptosis of hepatocellular carcinoma cells through activating the mitochondrial pathways, but also suppressed the invasion and metastasis of cancer cells and angiogenesis of human umbilical vein endothelial cells via suppression of PI3K/Akt/mTOR/NF-κB and EGFR/STAT3 signaling (39). Our recent study demonstrated that curcumin was able to suppress hypoxia-induced pancreatic cancer EMT and metastasis by inhibiting the hedgehog signaling pathway (8). It was also proven that curcumin was able to suppress cell migration and invasion through suppression of the ROS/ERK/NF-κB signaling pathway (26). It was previously demonstrated that curcumin inhibited pancreatic cancer cell migration and invasion and suppressed NEDD4 expression (40). In our previous study, we also demonstrated that LY294002 exposure for 24 h reduced the migration of pancreatic cancer cells without the addition of SOD (41). The present study revealed that curcumin inhibited the effects of SOD in pancreatic cancer through suppression of the PI3K/Akt/NF-κB signaling pathway in pancreatic cancer cells.

The PI3K/Akt signaling pathway is one of the most frequently changing signaling networks in human cancer, and has long been identified as being implicated in cancer metastasis. Akt is hyperactivated in cancer cells through multiple mechanisms, including the loss of phosphatase and tensin homolog, mutations that activate the catalytic subunit of PI3K, mutations that activate Akt isoforms, and amplification of the genes encoding the catalytic subunit of PI3K and Akt (42). Increased activation of PI3K/Akt signaling has been found in ~50% of pancreatic cancers, which is usually associated with a low grade of tumor differentiation and is correlated with a poor prognosis. PI3K/Akt signaling modulates a series of cellular functions, including cell transformation, proliferation, growth, motility and survival (43). Our previous study demonstrated that resveratrol was highly efficient in inhibiting the proliferation, migration and invasion of pancreatic cancer cells in vitro by regulating EMT-related factors via the PI3K/Akt/NF-κB signaling pathway (41). Recent studies demonstrated that both CuZnSOD and MnSOD can activate PI3K/Akt signaling in cancer cells (24,25). The results of the present study revealed that curcumin has great therapeutic potential, as it can restrain SOD-induced activation of p-Akt and p-NF-κB. Following suppression of the PI3K/Akt signaling pathway by LY 249002, the expression of p-Akt and NF-κB decreased, and the invasive and migratory abilities of pancreatic cancer cells were weakened.

In conclusion, the present study demonstrated that curcumin inhibited SOD-induced invasion and migration of pancreatic cancer cells by regulating EMT-related factors via the PI3K/Akt/NF-κB signaling pathway. Therefore, inhibition of the H2O2/Akt/NF-κB axis by curcumin may represent a promising option for the treatment of patients with pancreatic cancer.

Acknowledgments

Not applicable.

Notes

[1] Funding

This study was supported by the Natural Science Basic Research Project of Shaanxi Province (grant no. 2017JM8037).

[2] Availability of data and materials

The analysed data sets generated during the study are available from the corresponding authors on reasonable request.

[3] Author contributions

QM and LC conceived the study; WL, ZWu and LC designed the study; WL, ZJ and XX conducted the experiments; and ZWa and LC performed the data analysis. All authors have read and approved this manuscript.

[4] Ethics approval and consent to participate

Not applicable.

[5] Consent for publication

Not applicable.

[6] Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer Statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Lin QJ, Yang F, Jin C and Fu DL: Current status and progress of pancreatic cancer in China. World J Gastroenterol. 21:7988–8003. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Vincent A, Herman J, Schulick R, Hruban RH and Goggins M: Pancreatic cancer. Lancet. 378:607–620. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Shanmugam MK, Rane G, Kanchi MM, Arfuso F, Chinnathambi A, Zayed ME, Alharbi SA, Tan BK, Kumar AP and Sethi G: The multifaceted role of curcumin in cancer prevention and treatment. Molecules. 20:2728–2769. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Liang Z, Wu R, Xie W, Xie C, Wu J, Geng S, Li X, Zhu M, Zhu W, Zhu J, et al: Effects of curcumin on tobacco smoke-induced hepatic MAPK pathway activation and epithelial-mesenchymal transition in vivo. Phytother Res. 31:1230–1239. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Fu H, Wang C, Yang D, Wei Z, Xu J, Hu Z, Zhang Y, Wang W, Yan R and Cai Q: Curcumin regulates proliferation, autophagy, and apoptosis in gastric cancer cells by affecting PI3K and P53 signaling. J Cell Physiol. Sep 19–2017.Epub ahead of print. View Article : Google Scholar

7 

Liang Z, Lu L, Mao J, Li X, Qian H and Xu W: Curcumin reversed chronic tobacco smoke exposure induced urocystic EMT and acquisition of cancer stem cells properties via Wnt/β-catenin. Cell Death Dis. 8:e30662017. View Article : Google Scholar

8 

Cao L, Xiao X, Lei J, Duan W, Ma Q and Li W: Curcumin inhibits hypoxia-induced epithelial mesenchymal transition in pancreatic cancer cells via suppression of the hedgehog signaling pathway. Oncol Rep. 35:3728–3734. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Bhatia S, Monkman J, Toh AKL, Nagaraj SH and Thompson EW: Targeting epithelial-mesenchymal plasticity in cancer: Clinical and preclinical advances in therapy and monitoring. Biochem J. 474:3269–3306. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Li W, Ma Q, Liu J, Han L, Ma G, Liu H, Shan T, Xie K and Wu E: Hyperglycemia as a mechanism of pancreatic cancer metastasis. Front Biosci (Landmark Ed). 17:1761–1774. 2012. View Article : Google Scholar

11 

Li W, Zhang L, Chen X, Jiang Z, Zong L and Ma Q: Hyperglycemia promotes the epithelial-mesenchymal transition of pancreatic cancer via hydrogen peroxide. Oxid Med Cell Longev. 2016:51903142016. View Article : Google Scholar : PubMed/NCBI

12 

Li W, Cao L, Han L, Xu Q and Ma Q: Superoxide dismutase promotes the epithelial-mesenchymal transition of pancreatic cancer cells via activation of the H2O2/ERK/NF-κB axis. Int J Oncol. 46:2613–2620. 2015. View Article : Google Scholar

13 

Costa A, Scholer-Dahirel A and Mechta-Grigoriou F: The role of reactive oxygen species and metabolism on cancer cells and their microenvironment. Semin Cancer Biol. 25:23–32. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Guo L, Tan K, Wang H and Zhang X: Pterostilbene inhibits hepatocellular carcinoma through p53/SOD2/ROS-mediated mitochondrial apoptosis. Oncol Rep. 36:3233–3240. 2016. View Article : Google Scholar : PubMed/NCBI

15 

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

16 

Martinez-Useros J, Li W, Cabeza-Morales M and Garcia-Foncillas J: Oxidative stress: A new target for pancreatic cancer prognosis and treatment. J Clin Med. 6:E292017. View Article : Google Scholar : PubMed/NCBI

17 

Mayer IA and Arteaga CL: The PI3K/AKT pathway as a target for cancer treatment. Annu Rev Med. 67:11–28. 2016. View Article : Google Scholar

18 

Yu X, Wu Q, Wang L, Zhao Y, Zhang Q, Meng Q, Pawan and Wang S: Silencing of ST6GalNAc I suppresses the proliferation, migration and invasion of hepatocarcinoma cells through PI3K/AKT/NF-κB pathway. Tumour Biol. 37:12213–12221. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Gaianigo N, Melisi D and Carbone C: EMT and treatment resistance in pancreatic cancer. Cancers (Basel). 9:92017. View Article : Google Scholar

20 

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

21 

Castellanos EH, Cardin DB and Berlin JD: Treatment of early-stage pancreatic cancer. Oncology (Williston Park). 25:182–189. 2011.

22 

Claudia L, Jette R, Rudolf L, Ludger AW and Barbara Sr: Hydrogen peroxide - production, fate and role in redox signaling of tumor cells. Cell Commun Signal. 13:392015. View Article : Google Scholar

23 

Li W, Wu Z, Ma Q, Liu J, Xu Q, Han L, Duan W, Lv Y, Wang F, Reindl KM, et al: Hyperglycemia regulates TXNIP/TRX/ROS axis via p38 MAPK and ERK pathways in pancreatic cancer. Curr Cancer Drug Targets. 14:348–356. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Li F, Wang H, Huang C, Lin J, Zhu G, Hu R and Feng H: Hydrogen peroxide contributes to the manganese superoxide dismutase promotion of migration and invasion in glioma cells. Free Radic Res. 45:1154–1161. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Damiano S, Petrozziello T, Ucci V, Amente S, Santillo M and Mondola P: Cu-Zn superoxide dismutase activates muscarinic acetylcholine M1 receptor pathway in neuroblastoma cells. Mol Cell Neurosci. 52:31–37. 2013. View Article : Google Scholar

26 

Cao L, Liu J, Zhang L, Xiao X and Li W: Curcumin inhibits H2O2-induced invasion and migration of human pancreatic cancer via suppression of the ERK/NF-κB pathway. Oncol Rep. 36:2245–2251. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Schieber M and Chandel NS: ROS function in redox signaling and oxidative stress. Curr Biol. 24:R453–R462. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Griess B, Tom E, Domann F and Teoh-Fitzgerald M: Extracellular superoxide dismutase and its role in cancer. Free Radic Biol Med. 112:464–479. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Hempel N, Carrico PM and Melendez JA: Manganese superoxide dismutase (Sod2) and redox-control of signaling events that drive metastasis. Anticancer Agents Med Chem. 11:191–201. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Sun GG, Wang YD, Lu YF and Hu WN: Different association of manganese superoxide dismutase gene polymorphisms with risk of prostate, esophageal, and lung cancers: Evidence from a meta-analysis of 20,025 subjects. Asian Pac J Cancer Prev. 14:1937–1943. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Kang SW: Superoxide dismutase 2 gene and cancer risk: Evidence from an updated meta-analysis. Int J Clin Exp Med. 8:14647–14655. 2015.PubMed/NCBI

32 

Tang H, Dong X, Day RS, Hassan MM and Li D: Antioxidant genes, diabetes and dietary antioxidants in association with risk of pancreatic cancer. Carcinogenesis. 31:607–613. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Hart PC, Mao M, de Abreu AL, Ansenberger-Fricano K, Ekoue DN, Ganini D, Kajdacsy-Balla A, Diamond AM, Minshall RD, Consolaro ME, et al: MnSOD upregulation sustains the Warburg effect via mitochondrial ROS and AMPK-dependent signalling in cancer. Nat Commun. 6:60532015. View Article : Google Scholar : PubMed/NCBI

34 

Yi L, Shen H, Zhao M, Shao P, Liu C, Cui J, Wang J, Wang C, Guo N, Kang L, et al: Inflammation-mediated SOD-2 upregulation contributes to epithelial-mesenchymal transition and migration of tumor cells in aflatoxin G1-induced lung adenocarcinoma. Sci Rep. 7:79532017. View Article : Google Scholar :

35 

Yang C, Su X, Liu A, Zhang L, Yu A, Xi Y and Zhai G: Advances in clinical study of curcumin. Curr Pharm Des. 19:1966–1973. 2013.

36 

Zhou X, Su J, Feng S, Wang L, Yin X, Yan J and Wang Z: Antitumor activity of curcumin is involved in down-regulation of YAP/TAZ expression in pancreatic cancer cells. Oncotarget. 7:79076–79088. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Zhang J, Wang J, Xu J, Lu Y, Jiang J, Wang L, Shen HM and Xia D: Curcumin targets the TFEB-lysosome pathway for induction of autophagy. Oncotarget. 7:75659–75671. 2016.PubMed/NCBI

38 

Kunnumakkara AB, Guha S, Krishnan S, Diagaradjane P, Gelovani J and Aggarwal BB: Curcumin potentiates antitumor activity of gemcitabine in an orthotopic model of pancreatic cancer through suppression of proliferation, angiogenesis, and inhibition of nuclear factor-kappaB-regulated gene products. Cancer Res. 67:3853–3861. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Zhang HH, Zhang Y, Cheng YN, Gong FL, Cao ZQ, Yu LG and Guo XL: Metformin incombination with curcumin inhibits the growth, metastasis, and angiogenesis of hepatocellular carcinoma in vitro and in vivo. Mol Carcinog. 57:44–56. 2018. View Article : Google Scholar

40 

Su J, Zhou X, Yin X, Wang L, Zhao Z, Hou Y, Zheng N, Xia J and Wang Z: The effects of curcumin on proliferation, apoptosis, invasion, and NEDD4 expression in pancreatic cancer. Biochem Pharmacol. 140:28–40. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Li W, Ma J, Ma Q, Li B, Han L, Liu J, Xu Q, Duan W, Yu S, Wang F, et al: Resveratrol inhibits the epithelial-mesenchymal transition of pancreatic cancer cells via suppression of the PI-3K/Akt/NF-κB pathway. Curr Med Chem. 20:4185–4194. 2013. View Article : Google Scholar

42 

Wang Q, Chen X and Hay N: Akt as a target for cancer therapy: More is not always better (lessons from studies in mice). Br J Cancer. 117:159–163. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Baer R, Cintas C, Therville N and Guillermet-Guibert J: Implication of PI3K/Akt pathway in pancreatic cancer: When PI3K isoforms matter? Adv Biol Regul. 59:19–35. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2018
Volume 52 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li W, Jiang Z, Xiao X, Wang Z, Wu Z, Ma Q and Cao L: Curcumin inhibits superoxide dismutase-induced epithelial-to-mesenchymal transition via the PI3K/Akt/NF-κB pathway in pancreatic cancer cells. Int J Oncol 52: 1593-1602, 2018
APA
Li, W., Jiang, Z., Xiao, X., Wang, Z., Wu, Z., Ma, Q., & Cao, L. (2018). Curcumin inhibits superoxide dismutase-induced epithelial-to-mesenchymal transition via the PI3K/Akt/NF-κB pathway in pancreatic cancer cells. International Journal of Oncology, 52, 1593-1602. https://doi.org/10.3892/ijo.2018.4295
MLA
Li, W., Jiang, Z., Xiao, X., Wang, Z., Wu, Z., Ma, Q., Cao, L."Curcumin inhibits superoxide dismutase-induced epithelial-to-mesenchymal transition via the PI3K/Akt/NF-κB pathway in pancreatic cancer cells". International Journal of Oncology 52.5 (2018): 1593-1602.
Chicago
Li, W., Jiang, Z., Xiao, X., Wang, Z., Wu, Z., Ma, Q., Cao, L."Curcumin inhibits superoxide dismutase-induced epithelial-to-mesenchymal transition via the PI3K/Akt/NF-κB pathway in pancreatic cancer cells". International Journal of Oncology 52, no. 5 (2018): 1593-1602. https://doi.org/10.3892/ijo.2018.4295