Pristimerin effectively inhibits the malignant phenotypes of uveal melanoma cells by targeting NF‑κB pathway

  • Authors:
    • Biao Zhang
    • Jing Zhang
    • Jingxuan Pan
  • View Affiliations

  • Published online on: July 25, 2017     https://doi.org/10.3892/ijo.2017.4079
  • Pages: 887-898
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Uveal melanoma (UM) is a highly aggressive intraocular malignancy that lacks any effective targeted-therapy. Neither survival nor prognosis has been improved for the past decades in patients with metastatic UM. NF‑κB pathway is reported to be abnormally activated in UM. However, the role of NF‑κB pathway as a potential therapeutical target in UM remains unclear. Here, the effect of pristimerin, a potent inhibitor of NF‑κB pathway, on UM cells in terms of growth, apoptosis, motility, invasion and cancer stem-like cells (CSCs) was evaluated in vitro. We showed that pristimerin suppressed tumor necrosis factor α (TNFα)-induced IκBα phosphorylation, translocation of p65, and expression of NF‑κB-dependent genes. Moreover, pristimerin decreased cell viability and clonogenic ability of UM cells. A synergistic effect was observed in the treatment of pristimerin combined with vinblastine, a frontline therapeutic agent, in UM. Pristimerin led to a significant increase in the Annexin V+ cell population as measured by flow cytometry. We also observed that pristimerin impaired the abilities of migration and invasion in UM cells. Furthermore, pristimerin eliminated the ALDH+ cells and weakened serial re-plating ability of melanosphere. Collectively, pristimerin shows remarkable anticancer activities in UM cells through inactivating NF‑κB pathway, revealing that pristimerin may be a promising therapeutic agent in UM.

Introduction

Uveal melanoma (UM), originated from melanocyte malignant transformation, is the most common intraocular tumor in adults (1). The yearly incidence is 5–6 cases per million, and the median age at diagnosis is 62 years (2). Hepatic metastasis is the leading cause of death in UM patients, and the average survival time for patients diagnosed with liver metastasis is only 2–14 months (2). Although there are multiple options available for the treatment of primary UM patients, such as surgical resection of diseased eye ball, plaque radiotherapy, transpupillary thermotherapy and chemotherapy (24), the 5-year survival rate displayed no improvement in past decades, which is attributed to lacking effective therapy for metastasis (2). Therefore, there is an urgent demand to develop more efficient agents against both primary and metastatic UM.

Aberrant activation of NF-κB is widely present in diverse malignancies (59). NF-κB pathway regulates the transcription of numerous genes which are involved in diverse cellular functions, including apoptosis, proliferation, angiogenesis, immune response, cell invasion, and cancer stem-like cells (CSCs) (10). More importantly, NF-κB pathway may be a potential molecular target for cancer therapy (11). Previous evidence indicates that constitutive activation of NF-κB can increase UM cellular proliferation and evasion of apoptosis, and is involved in both primary and metastatic UM (12). Thus, pharmacological inhibition of NF-κB may be an effective approach to kill UM cells.

Previous studies demonstrated that pristimerin, a natural product isolated from natural herb plants, is a potent inhibitor of NF-κB pathway (9,13,14) and displays antimicrobial, anti-inflammatory, anti-peroxidation activities (13) and anticancer effects in various human malignancies (1518). Moreover, pristimerin has shown inhibitory effects on proliferation, survival, angiogenesis, metastasis, and CSCs characteristics (16,17,1922). Thus, we wondered whether this compound also had anticancer activity in UM.

In the present study, we showed the inhibitory activity of pristimerin against TNFα-induced NF-κB activation in UM cells. We also found that the downregulation of survivin on mRNA level was critical in pristimerin-inducing apoptosis in UM cells. In addition, pristimerin attenuated the properties of CSCs and the combination of pristimerin and vinblastine, a frontline therapeutic agent, showed a synergistic effect against UM cells. This study suggests that pristimerin is a promising compound for UM treatment.

Materials and methods

Reagents and antibodies

Pristimerin (Fig. 1A, purity >99%, HPLC) was purchased from Paypaytech Inc. (Shenzhen, China), and dissolved in dimethyl sulphoxide (DMSO, Sigma-Aldrich, Shanghai, China) at a stock concentration of 20 mM, and stored at −20°C. Vinblastine was obtained from Selleck (Shanghai, China). Antibodies against survivin, Bcl-XL and PCNA were from Santa Cruz Biotech (Santa Cruz, CA, USA). Antibodies against p65, IκBα, phospho-IκBα (S32), Tubulin, matrix metalloproteinase 2 (MMP2), matrix metalloproteinase 9 (MMP9), Slug, Sox2, Nanog and KLF4 were purchased from Cell Signaling Technology (Beverly, MA, USA). Antibodies against PARP (clone 4C10–5), caspase-3, active caspase-3 (CM1), cytochrome c (clone 6H2.B4), X-linked inhibitor of apoptosis protein (XIAP), Bcl-2, and c-Myc were purchased from BD Biosciences (San Jose, CA, USA). Anti-mouse and anti-rabbit immunoglobulin G fluorescent-conjugated secondary antibodies were purchased from LI-COR Biotechnology (NE, USA). Dual-luciferase assay kit was provided by Promega (Madison, WI, USA). The Aldeflour kit was from StemCell Technologies (Vancouver, Canada). Gelatin (G1890-100G) was obtained from Sigma-Aldrich). Coomassie Brilliant Blue R-250 (44329-7C) was from Farco Chemical Supplies (Hong Kong, China).

Figure 1

Pristimerin suppresses TNFα-induced NF-κB activation in uveal melanoma cells. (A) The structure of pristimerin is shown. (B) Pristimerin inhibited TNFα-induced NF-κB-dependent reporter gene expression in UM cells. Mel 270, Omm 1 and Omm 2.3 cells co-transfected with NF-κB-TATA-Luc reporter plasmid and Renilla luciferase reporter plasmid were treated with various concentrations of pristimerin for 16 h, and then stimulated with TNFα for 8 h; the luciferase activity of cells was detected. The levels of firefly luciferase activity were normalized to Renilla luciferase activity. The results represented the means ± SE of three independent experiments. **P<0.01, ***P<0.001, one-way ANOVA, post hoc intergroup comparisons, Tukey's test. (C and D) Pristimerin blocked the nuclear translocation of p65 in UM cells stimulated by TNFα. 92.1 or Omm 1 cells were pretreated with or without 1.0 µM pristimerin for 12 h, then stimulated with TNFα (10 ng/ml) at the indicated time; cytosolic and nucleus fractionations were subjected to western blot analysis. Tubulin was a cytoplasmic loading control, while PCNA was a nuclear loading control (C). 92.1 and Omm 1 cells were pretreated with medium containing DMSO or 0.5 µM pristimerin for 24 h, and then incubated with TNFα (10 ng/ml) for 15 min, undergoing immunofluorescence analysis of p65. DAPI was used to stain the nuclear (D). Scale bar represents 20 µm. (E) Pristimerin decreased the levels of NF-κB-dependent prosurvival proteins in UM cells. After pre-incubation with pristimerin for 12 h, Omm 1 cells were exposed to 0.1 nM TNFα for the indicated durations, and then western blot analysis was performed.

Cell culture

The UM cells 92.1, Mel 270, Omm 1 and Omm 2.3 were cultured in RPMI-1640 supplemented with 10% fetal bovine serum (FBS), penicillin (100 U/ml), streptomycin (100 µg/ml) and L-glutamine (2 mM), and incubated at 37°C and 5% CO2, as described previously (23). 293T cells were cultured in DMEM supplemented with 10% FBS, penicillin (100 U/ml), streptomycin (100 µg/ml) and incubated at 37°C and 5% CO2.

Dual luciferase reporter assay

Omm 1, 92.1 and Omm 2.3 cells seeded in 24-well plates were co-transfected with NF-κB-TATA-Luc reporter plasmid (0.5 µg) and Renilla luciferase reporter plasmid (10 ng), respectively. After 24 h, these UM cells were exposed to the indicated concentrations of pristimerin for 16 h, then added with TNFα (10 ng/ml) for 8 h and measured with dual-luciferase assay kit as described previously (9,24). The luciferase activities were normalized with NF-κB-dependent firefly luciferase to Renilla luciferase activity.

Western blot analysis

The prepared cell lysates were quantified by Pierce™ BCA Protein assay kit (Thermo Scientific, USA) according to the manufacturer's instructions. Protein samples were subjected to SDS-PAGE and then blotted to nitrocellulose membranes. After blocked by 5% skim milk, membranes were subsequently incubated with the primary antibodies overnight at 4°C before incubation with appropriate secondary antibodies. The immunoblots were recorded with the Odyssey infrared imaging system (LI-COR).

Immunofluorescence staining

After cultured overnight, 92.1 and Omm 1 cells were incubated with or without pristimerin for 24 h, and then the cells were stimulated with TNFα (20 ng/ml) for 15 min before fixed by 4% paraformaldehyde. After permeabilization by 0.2% (v/v) Triton X-100 at room temperature for 15 min, the samples were washed with 500 µl of PBS for 5 min three times. Subsequently, PBS buffer containing 5% bovine serum albumin was used to block non-specifical binding for 1 h and then the cells were incubated with 200 µl of PBS containing the primary antibody for 1 h at room temperature. After washing with PBS containing 0.1% (v/v) NP-40, the cells were incubated with 200 µl of PBS containing the secondary antibody for 1 h at room temperature. Nuclear staining was performed with DAPI for 20 min. After washing with PBS for 5 min, all samples were added with a half drop of anti-fade regent and then sealed by nail polish (9).

Preparation of cytoplasmic and nuclear fractionations

After stimulation with TNFα (10 ng/ml), cells treated with or without pristimerin were collected by centrifugation and washed with PBS. Pellets resuspended in 150 µl cold lysis buffer (10 mM HEPES, 10 mM KCl, 0.1 mM EDTA, 0.4% NP-40, 1 mM DTT, 0.5 mM PMSF, 1 mM sodium orthovanadate and Complete Protease Inhibitor Mix) by pipetting up and down ~10 times were incubated on ice for 10 min, the lysates were centrifuged at 20,000 g (9,25). The supernatants were transferred to a fresh tube as cytoplasmic extracts. After rinsed with the lysis buffer, the pellets were vigorously suspended in nuclear protein extraction buffer (20 mM HEPES, 0.4 mM NaCl, 1 mM EDTA, 1 mM DTT, 0.5 mM phenylmethylsulfonyl fluoride (PMSF), 1 mM sodium orthovanadate and Complete Protease Inhibitor Mix) and incubated for 15 min. After centrifugation with 10 min at 20,000 g, the supernatant was referred to as nuclear fractions (9,25).

Preparation of whole cell lysates and cytosolic fractionations

Control or treated cells were pelleted by centrifugation and then rinsed with PBS. The whole cell lysates were prepared with RIPA buffer (1X PBS, 0.1% SDS, 1% NP-40, 0.5% sodium deoxycholate) supplemented with 1X protease inhibitor cocktail, 10 mM β-glycerophosphate, 1 mM sodium orthovanadate, 10 mM sodium fluoride, and 1 mM PMSF (26). The cytosolic fraction was extracted with digitonin extraction buffer (10 mM PIPES, 0.015% digitonin, 300 mM sucrose, 100 mM NaCl, 3 mM MgCl2, 5 mM EDTA, and 1 mM PMSF) on ice for 10 min and then centrifuged at 20,000 g for 10 min at 4°C. The supernatants were collected as cytosolic fractions (27).

Cell viability assay

The UM cells (5,000 cells/well) seeded into 96-well plates overnight were then exposed to serially diluted pristimerin for 72 h. Optical intensity was measured by microplate reader after addition with 20 µl mixture of MTS and PMS (28).

Colony formation assay

The UM cells were treated with various concentrations of pristimerin for 24 h and rinsed with PBS, then 8,000 cells were seeded to a modified drug-free double layer soft agar system (29). After 10–14 days, the number of colonies composed of >50 cells was counted under a microscope.

Cell cycle analysis by flow cytometry

After pretreated with or without pristimerin for 24 h and washed with PBS, UM cells were fixed with 66% ethanol overnight. Cells rinsed with PBS were pelleted by centrifugation. Subsequently, the pellets were resuspended in PBS and stained with propidium iodide (PI, 50 µg/ml) and RNase (2.5 µg/ml) for 30 min at room temperature. DNA content was analyzed by flow cytometry (9).

Apoptosis analysis by flow cytometry

Annexin V-FITC/PI double staining was used to detect apoptosis by flow cytometry. Cells pre-incubated with or without pristimerin were collected by centrifugation and washed with PBS, and then stained with 0.3% Annexin V-FITC in binding buffer for 15 min at room temperature in the dark. After centrifuged at 250 g and resuspended in 1X binding buffer, the cells were added with PI before flow cytometry analysis (9,29).

Measurement of mitochondrial transmembrane potential

UM cells were incubated with 2.0 µM pristimerin for different durations, and then stained with MitoTracker probes (CMXRos and MTGreen) for 1 h at 37°C. After being centrifuged at 250 g for 10 min, the pellets were raised in PBS and subjected to flow cytometry to detect the changes in mitochondrial transmembrane potential (Δψ) (25,29).

Lentiviral transfection

Lentiviruses were produced by transient transfection in 293T cells with control shRNA (Scramble) or specific shRNA together with the pCMV-dR8.2 packing construct and the pCMV-VSVG envelope construct. 92.1 and Omm 1 cells (1×105 cells/well) were infected twice with virus-containing supernatants. The cells were then incubated in the presence of puromycin (1 µg/ml) for ~5 days. Scramble and specific shRNAs were purchased from Sigma-Aldrich, and the sequences were as follows: PLKO.1-Non-target shRNA: CCG GGC GCG ATA GCG CTA ATA ATT TCT CGA GAA ATT ATT AGC GCT ATC GCG CTT TTT; shsurvivin: CCG GGA AGA ATT AAC CCT TGG TGA ACT CGA GTT CAC CAA GGG TTA ATT CTT CTT TTTG. The Omm 1 cells overexpressed survivin were established with the same methods. Constructs bearing full length human survivin cDNA in pTSB-CMV-MCS-SBP-tRFP-F2A-PuroR and empty vector were provided by Transheep (Shanghai, China). Human baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5, NCBI Reference Sequence ID: NG_029069.1) was cloned into the pTSB-CMV-MCS-SBP-tRFP-F2A-PuroR (lentivirus) plasmid using ClonExpress MultiS One Step Cloning kit, which was provided by Vazyme (Nanjing, China) and clone sites were EcoRI and BamHI. The efficiency of knockdown or overexpression was examined by western blot analysis.

Real-time quantitative PCR

After pre-incubated with various concentrations of pristimerin, total RNA was extracted by TRIzol reagent, and then reverse-transcribed into first-strand complementary DNA with maxima first strand cDNA synthesis kit. GAPDH was used as an endogenous reference. The PCR primers were as follows: survivin forward, 5′-CAT CTC TAC ATT CAA GAA CTG G-3′; reverse, 5′-GGT TAA TTC TTC AAA CTG CTT C-3′ (30). GAPDH forward, 5′-GAT CGA ATT AAA CCT TAT CGT CGT-3′; reverse, 5′-AGC AGC AGA ACT TCC ACT CGG T-3′.

The qRT-PCR reaction was performed in SYBR Premix EX Taq with Bio-Rad CFX96 Real-Time Thermocycler according to the manufacturer's instructions (26,29). In relative quantification, ΔΔCq method was used, as described previously (31).

The scratch wound healing assay

92.1 and Omm 1 cells (5×105/well) were cultured in a 6-well plate supplemented with 10% FBS. The monolayer was gently and slowly scratched with a 200-µl pipette tip across the center of the well at ~90% confluence. The cells were treated with or without pristimerin (0.25 or 0.5 µM), the same wounded area was recorded under a microscope at different time periods (29,32).

Migration and invasion assay

After exposed to vehicle, 0.25 or 0.5 µM pristimerin for 24 h, equal amounts of 92.1 and Omm 1 cells were cultured in the upper of Transwell chamber covered with or without Matrigel and contained with FBS-free medium, while to the lower chamber 20% FBS was added as chemoattractant. After 48 h, the cells on the surface of chamber were removed by a cotton swab, and the migrated or invaded cells were fixed with 4% paraformaldehyde and stained by crystal violet. The cells in three randomly selected low power fields were counted (29).

Gelatin zymography assay

92.1 and Omm 1 cells were seeded into 6-well culture plates. When cells grew to ~80% confluency, were washed by serum-free medium three times. Cells were incubated with serum-free medium with or without pristimerin (0.25 and 0.5 µM) for 24 h. MDA-MB-231 cells were used as positive control (33). Serum-free medium without cells was used as negative control. After incubation, the supernatants were collected and were subjected to SDS-PAGE using 10% acrylamide gels containing 0.1% gelatin. After electrophoresis, the gels were washed for 15 min at room temperature in a buffer (50 mM Tris-Cl pH 7.6, 10 mM CaCl2, 20 mM NaCl, 1 µM ZnCl2 and 2.5% Triton X-100) three times. After incubation with activation buffer (50 mM Tris-Cl pH 7.6, 10 mM CaCl2, 20 mM NaCl, 1 µM ZnCl2) for 48 h at 37°C, gels were stained with 0.25% Coomassie Brilliant Blue R-250 in 40% methanol and 10% acetic acid and then briefly destained in 10% acetic acid and 30% methanol. The locations of gelatinolytic enzymes were visualized as clear bands on the background (34). The experiment was repeated three times and the optical density of each bands was quantitated by image pro plus.

Aldehyde dehydrogenase (ALDH) assay

The Aldefluor kit was used to identify a population of cells with high ALDH enzymatic activity (35). In brief, UM cells (92.1 and Omm 1) pre-incubated with DMSO or 1.0 µM pristimerin for 24 h, and then incubated with 5 µl ALDH reagent in the absence or presence of 5 µl DEAB for 1 h at 37°C. After washed with ALDH assay buffer, the cells were resuspended in ALDH assay buffer. The proportion of ALDH+ cells was defined by flow cytometry (29,35).

Melanosphere culture

92.1 and Omm 1 cells pretreated with DMSO or 1.0 µM pristimerin for 24 h were seeded to 24-well low attachment plates containing 500 µl DMEM/F12 medium supplemented with 1X B27, 10 ng/ml basic fibroblast growth factor (bFGF), 20 ng/ml epidermal growth factor (EGF) each well. After ~14 days, melanospheres with >50 cells were counted as per the universal standard (29,36). The secondary and tertiary rounds of melanosphere cultures were implemented in drug-free culture after collecting the first or second round of tumor sphere culture cells. Representative images were taken by a microscope.

Statistical analysis

All experiments were performed at least thrice, and the results are expressed as the means ± standard error (SE), unless otherwise stated. GraphPad Prism 5.0 software was used for statistical analysis. Comparison between 2 groups used two-tailed Student's t-test, and comparison among multiple groups involved one-way ANOVA with post hoc intergroup comparisons using Tukey's test. P<0.05 was regarded as statistically significant.

Results

Pristimerin blocks TNFα-induced NF-κB activation in UM cells

Our previous report indicated that pristimerin potently inhibits activation of canonical NF-κB pathway in leukemia cells (9). Here, we tested the effect of pristimerin in UM cells. We first examined whether pristimerin exerted the inhibitory effect on TNFα-induced NF-κB-dependent reporter gene transcription. The results showed that the NF-κB dependent luciferase activity was increased after TNFα stimulation in UM cells. This effect was inhibited by pristimerin in a dose-dependent manner (Fig. 1B). Because the ubiquitination-dependent degradation of IκBα protein triggers p65 nuclear translocation, which is a critical step in the activation of the canonical NF-κB pathway (37), we determined the influence of pristimerin on IκBα and p65. Western blot analysis of cytoplasmic fractionations showed that the levels of phosphorylated IκBα in the cytoplasm were appreciably increased after TNFα induction compared to the untreated control, while the total IκBα was coincidently decreased (Fig. 1C). However, the phosphorylation of IκBα was blocked, and the level of total IκBα remained constant in the cells that were pretreated with pristimerin (Fig. 1C). Consistently, the level of p65 in nuclear fractionation was remarkably escalated after TNFα stimulation (Fig. 1C), which was diminished by the presence of pristimerin (Fig. 1C). On the other hand, immunofluorescence staining analysis similarly revealed that nuclear translocation of p65 was prominently increased after TNFα stimulation; whereas pristimerin abrogated its translocation in both 92.1 and Omm 1 cells (Fig. 1D).

We next ascertained the effect of pristimerin on the expression of encoding products of NF-κB-dependent genes involved in cell survival by western blotting (10). The expression of MMP9, c-Myc, Bcl-XL and survivin was increased after stimulation with TNFα (Fig. 1E). Nevertheless, pristimerin blocked such an increase (Fig. 1E).

Taken together, our results suggested that pristimerin represses the activation of the canonical NF-κB pathway.

Pristimerin suppresses the growth of UM cells

Seventy-two-hour cell-based MTS assay revealed that pristimerin significantly inhibited the cell viability of 92.1, Mel 270, Omm 1 and Omm 2.3 cells, and the IC50 values were 1.1, 1.7, 2.2 and 2.9 µM, respectively (Fig. 2A). Given the advantages of colony formation assay in reflecting malignant behavior of tumor cells, we assessed the impact of pristimerin on anchorage-independent growth of UM cells in soft agar. The results indicated that pristimerin significantly inhibited the formation of colony in a dose-dependent manner (Fig. 2B).

To further study the role of pristimerin in the growth of UM cells, cell cycle distribution analysis was performed after UM cells were exposed to different concentrations of pristimerin for 24 h. The results showed that pristimerin caused G1-phase arrest in UM cells (Fig. 2C).

Pristimerin exerts induction of apoptosis in UM cells

We carried out flow cytometry analysis based on Annexin V-FITC/PI double staining and found that pristimerin remarkably induced cell death in UM cells in a dose- and time-dependent manner (Fig. 3A). Western blot analysis revealed that pristimerin induced cleavage of PARP and caspase-3 in UM cells (Fig. 3B), which demonstrated the occurrence of apoptosis induced by pristimerin.

Figure 3

Pristimerin induces intrinsic apoptosis in UM cells. (A-D) UM cells were treated with different concentrations of pristimerin for 24 h, or 2.0 µM pristimerin for the indicated times. (A) Pristimerin induced apoptosis of UM cells in a dose- and time-dependent manner. The representative flow cytometry histograms of Annexin V-FITC/PI double staining are shown (top), statistical chart showed the quantitative analysis of three independent experiments (bottom). The results represent the means ± SE of three independent experiments. *P<0.05, **P<0.01, ***P<0.001, one-way ANOVA, post hoc intergroup comparisons, Tukey's test. (B) Pristimerin decreased the expression of apoptosis-indicative proteins in dose- and time-dependent manner. Western blotting showed the effect of pristimerin on expression of apoptosis-indicative proteins including PARP, caspase-3 and active caspase-3. (C) Pristimerin triggered the mitochondrial depolarization of UM cells. The mitochondrial depolarization of UM cells was detected by flow cytometry after treatment with pristimerin. UM cells pre-incubated with 2.0 µM pristimerin in the indicated time periods and then stained with CMXRos and MTGreen. Mitochondrial membrane potential (Δψ) was analyzed by flow cytometry. The results represented the means ± SE of three independent experiments. *P<0.05, **P<0.01, ***P<0.001, one-way ANOVA, post hoc intergroup comparisons, Tukey's test. (D) Pristimerin induced the release of cytochrome c in UM cells. Western blotting showed the level of cytochrome c in cytoplasm, and tubulin as a loading control for cytosol.

Next, we evaluated the effect of pristimerin on mitochondrial depolarization in UM cells. After being treated with pristimerin, the cell populations with loss of mitochondrial transmembrane potential were significantly increased (Fig. 3C). In parallel, western blot analysis showed that the levels of cytochrome c in the cytosolic fractionation were increased in a time-dependent manner (Fig. 3D). These results suggested that pristimerin might cause damage of mitochondria and trigger the intrinsic pathway of apoptosis in UM cells.

Survivin plays an important role in pristimerin-induced apoptosis

To dissect the mechanism by which pristimerin induced apoptosis in UM cells, we next measured apoptosis-related proteins. The results showed that pristimerin decreased the expression of XIAP and survivin, but not Bcl-2 and Bcl-XL (Fig. 4A). Given that survivin was obviously declined and the anti-apoptotic role of survivin in pristimerin-inducing apoptosis in prostate cancer cells (38), we hypothesized that survivin might be critical in pristimerin-inducing apoptosis in UM cells. The Omm 1 cell ectopic overexpression of survivin by lentiviral construct encoding survivin were more resistant to pristimerin than cells transfected with empty vector (Fig. 4B). On the other hand, 92.1 cells were transfected with scramble or survivin shRNA, and the results showed that knockdown of survivin facilitated the sensitivity of UM cells to pristimerin (Fig. 4C). These data imply that survivin indeed plays a critical role in pristimerin-inducing apoptosis.

We next studied the regulation of survivin by pristimerin. qRT-PCR analysis showed that pristimerin treatment in 92.1 and Omm 1 cells led to a decrease in the mRNA level of BIRC5 (encoding survivin) (Fig. 4D). The results suggested that downregulation of survivin by pristimerin occurs at the level of transcription in UM cells.

Pristimerin inhibits migration and invasion of UM cells

Although multiple therapies are available for primary UM patients, therapeutic options are limited for the metastatic patients with no apparent reduced mortality in the past decades (1,2,4). In order to investigate the effect of pristimerin on the motility of UM cells, wound healing ability was tested in 92.1 and Omm 1 cells. After 72 h of pristimerin treatment, the occlusion of UM cells was remarkably slowed (Fig. 5A). We next examined the inhibitory effect of pristimerin on Transwell migration and invasion and found that pristimerin profoundly decreased the number of migrated (Fig. 5B) and invaded (Fig. 5C) cells. Local invasion is an initial step of metastasis with involvement of matrix metalloproteinases (MMPs) (29,39). We therefore assumed that pristimerin suppressed the invasion of UM cells by decreasing the expression or activity of MMP2 and MMP9. Western blotting results showed that pristimerin decreased the protein levels of MMP2 and MMP9 in a dose-dependent manner (Fig. 5D). Moreover, gelatin zymography assay results indicated that pristimerin inhibited the activity of MMP9 in a dose-dependent manner (Fig. 5E).

Figure 5

Pristimerin impairs the ability of migration and invasion in UM cells. (A) Pristimerin reduced the motility of UM cells. After 92.1 and Omm 1 cells were seeded in 6-well plates overnight, a wound was made by a 200-µl pipette tip. The cells were exposed to vehicle, 0.25 or 0.5 µM pristimerin for the indicated periods, and the wound closure was recorded under microscope. The images were the representative results (top), while the graphs were the quantitative analysis of the percentage of wound closure at different time-points (bottom). Data were shown as the means ± SE. *P<0.05, **P<0.01, one-way ANOVA was used for statistical analysis. Scale bar represents 200 µm, original magnification, ×200. (B) Pristimerin inhibited the Transwell migration ability of UM cells. Left, representative image of migration assay of 92.1 and Omm 1 cells after exposed to vehicle, 0.25 or 0.5 µM pristimerin for 24 h. Right, quantitative analysis of the migration cells was showed. Values were expressed as mean cell numbers in three random fields of view (×200) in three independent experiments. Error bars represented the means ± SE. *P<0.05, **P<0.01, ***P<0.001, one-way ANOVA was used for statistical analysis. (C) Pristimerin suppressed the invasion ability of UM cells. The representative images of invaded cells which were treated with vehicle, 0.25 or 0.5 µM pristimerin for 24 h. Scale bar represents 200 µm (left), and results of the invaded cells of per field (three random field) (right). The results showed as the means ± SE. **P<0.01, ***P<0.001, one-way ANOVA was used for statistical analysis. (D) Pristimerin reduced the expression of MMP2 and MMP9 in a dose-dependent manner. The whole lysates of cells were used to detect the protein level of MMP2 and MMP9 by western blot analysis after Omm 1 and 92.1 cells were treated with pristimerin for 24 h. (E) Pristimerin inhibited the activity of MMP9 in a dose-dependent manner. Omm 1 and 92.1 cells were cultured and treated with or without pristimerin (0.25 and 0.5 µM) for 24 h. The serum-free medium of cells was used to detect the activity of MMP9 by gelatin zymography assay. The serum-free medium of MDA-MB-231 cells and serum-free medium without cells were used as positive control and negative control, respectively. The images (left) were the representative result, while the graphs (right) were the quantitative analysis of MMP9 activity at different concentration of pristimerin (n=3). The results showed as the means ± SE. *P<0.05, **P<0.01, one-way ANOVA was used for statistical analysis.

Pristimerin weakened the properties of cancer stem-like cells in UM cells

CSC theory holds a view that CSC is the main cause for metastasis and therapeutic resistant (40). Several in vitro assays such as ALDH+ assay and tumor sphere formation allow for the study of CSCs (10). Flow cytometry analysis showed that the percentage of ALDH+ cells was significantly decreased in the cells treated with pristimerin compared to that in control group (Fig. 6A). On the other hand, three rounds of serially re-plating culture of melanoshpere showed that the numbers of melanosphere in the pristimerin-treated cells were significantly lower than those in control group (Fig. 6B), implying that pristimerin exerts an inhibitory activity on the self-renewal capacity of CSCs in UM cells. We also examined the universal stemness-related proteins which were also regulated by NF-κB pathway such as Slug, Sox2, Nanog and KLF4 (10), the results showed that the expression of Slug and Sox2 was decreased in the cells treated with the highest concentration (2 µM) of pristimerin (Fig. 6C). The protein levels of Nanog and KLF4 remained constant (Fig. 6C).

Pristimerin enhances the sensitivity of UM cells to vinblastine

Vinblastine, a conventional chemotherapeutic agent, was used in clinical treatment of UM patients (3). We thus exploited whether pristimerin increased sensitivity of UM cells to vinblastine. The results of 72 h of combinational treatment showed a synergistic effect in 92.1 and Omm 1 cells (combinational index, CI<1) (Fig. 7A). Furthermore, detection of PARP cleavage and trypan blue exclusion assay confirmed an enhanced apoptosis-inducing effect in UM cells (Fig. 7B).

Discussion

Although recent technical advances have opened up many new ways in treating primary UM patients, there are no effective therapeutic approaches for those who have metastasized. In this study, our results demonstrated that pristimerin displayed significant anticancer activities by blocking the NF-κB pathway in UM cells. Pristimerin suppressed TNFα-induced IκBα phosphorylation and degradation, translocation of p65, and expression of NF-κB-dependent genes. Pristimerin effectively inhibited the malignant phenotypes such as proliferation, resistance to apoptosis, migration, invasion and CSCs in UM cells. Additionally, pristimerin decreased the expression of survivin at transcriptional level which plays a critical role in pristimerin-inducing apoptosis of UM cells.

It is well-known that cancer-related inflammation is a hallmaker of cancer (41). In physiologic condition, eyes are in a state of immune privilege without existences of immune cells. Once tumor occurs in the eyes, this state is challenged with infiltration with myeloid and T cells which constitute an intraocular inflammatory microenvironment (42). Previous evidence has demonstrated that cytokines and chemokines (e.g., TNFα, IL-6, IL-8, IP-10, MIP-1 and IFN-γ) in the vitreous fluid of eyes with UM are elevated (43). Among them, TNFα can activate the NF-κB pathway (37). Moreover, hyperactivation of NF-κB not only contributes to aberrant local tumor cell survival and growth, but also promotes distant metastasis (12,41,43). Thus, NF-κB inhibition appears to offer a promising strategy in cancer therapy in UM.

The CSC theory has emerged as an important landmark in the understanding of drug resistance and cancer recurrence, and CSCs are regarded as an essential cause of tumor metastasis, recurrence and drug tolerance (40). So far, many classic signal pathways such as Wnt/β-catenin, Hedgehog, and Notch pathway have shown extensive involvement in the coordination of CSCs (40). Additionally, accumulating evidence indicated the existence of links between CSCs and NF-κB pathway (10). Our study showed that pristimerin inhibited the characteristic of CSCs in UM cells, which is consistent with a recent report in prostate cancer cells (20,22). Given the fundamental role of Slug in CSCs in breast cancer (44), the role of Slug in UM CSCs need to be defined in the future.

In order to acquire better therapeutic outcomes, concomitant drugs become a conventional approach in clinical cancer therapy. Therefore, we chose vinblastine as a representative front line agent in our research to explore the potential value of pristimerin in UM clinical combination treatment. This is the first report that pristimerin has synergistic effect with vinblastine in UM cells.

Pristimerin is a compound isolated from nature products and the antitumor activity of this compound is complicated. It is reported that pristimerin modulates other molecular targets such as cyclins, apoptosis-related proteins, proteasome activity, AKT/mTOR and MAPK/ERK pathways contributing to its antitumor activity (14).

Our results indicated that pristimerin effectively blocks the NF-κB pathway, which contributes to the inhibition of malignant phenotypes in UM cells. However, we could not exclude the possibility of this compound to target other molecular pathways in UM cells.

In conclusion, pristimerin not only killed the bulk of tumor cells, but also effectively eliminated CSCs in UM cells. Pristimerin as a lead compound also exerted favorable anticancer effects both in single administration and combination with vinblastine. Thus, further clinical trial of pristimerin against UM may be warranted.

Acknowledgments

This study was supported by grants from National Natural Science Funds (nos. U1301226, and 81373434 to J. Pan), the National Basic Research Program of China (973 program no. 2009CB825506 to J. Pan), the Research Foundation of Education Bureau of Guangdong Province, China (no. cxzd1103 to J. Pan), and Natural Science Foundation of Guangdong province (no. 2015A030312014 to J. Pan).

References

1 

Singh AD, Bergman L and Seregard S: Uveal melanoma: Epidemiologic aspects. Ophthalmol Clin North Am. 18:75–84. viii2005. View Article : Google Scholar : PubMed/NCBI

2 

Singh AD, Turell ME and Topham AK: Uveal melanoma: Trends in incidence, treatment, and survival. Ophthalmology. 118:1881–1885. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Bedikian AY, Johnson MM, Warneke CL, Papadopoulos NE, Kim KB, Hwu WJ, McIntyre S, Rohlfs M, Homsi J and Hwu P: Does complete response to systemic therapy in patients with stage IV melanoma translate into long-term survival? Melanoma Res. 21:84–90. 2011. View Article : Google Scholar

4 

Damato B and Heimann H: Personalized treatment of uveal melanoma. Eye (Lond). 27:172–179. 2013. View Article : Google Scholar

5 

Li B, Li YY, Tsao SW and Cheung AL: Targeting NF-κB signaling pathway suppresses tumor growth, angiogenesis, and metastasis of human esophageal cancer. Mol Cancer Ther. 8:2635–2644. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Sakamoto K and Maeda S: Targeting NF-κB for colorectal cancer. Expert Opin Ther Targets. 14:593–601. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Luedde T and Schwabe RF: NF-κB in the liver - linking injury, fibrosis and hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 8:108–118. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Nogueira L, Ruiz-Ontañon P, Vazquez-Barquero A, Moris F and Fernandez-Luna JL: The NF-κB pathway: A therapeutic target in glioblastoma. Oncotarget. 2:646–653. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Lu Z, Jin Y, Chen C, Li J, Cao Q and Pan J: Pristimerin induces apoptosis in imatinib-resistant chronic myelogenous leukemia cells harboring T315I mutation by blocking NF-κB signaling and depleting Bcr-Abl. Mol Cancer. 9:1122010. View Article : Google Scholar

10 

Rinkenbaugh AL and Baldwin AS: The NF-κB pathway and cancer stem cells. Cells. 5:52016. View Article : Google Scholar

11 

Lee CH, Jeon YT, Kim SH and Song YS: NF-κB as a potential molecular target for cancer therapy. Biofactors. 29:19–35. 2007. View Article : Google Scholar

12 

Dror R, Lederman M, Umezawa K, Barak V, Pe'er J and Chowers I: Characterizing the involvement of the nuclear factor-κB (NF κB) transcription factor in uveal melanoma. Invest Ophthalmol Vis Sci. 51:1811–1816. 2010. View Article : Google Scholar

13 

Dirsch VM, Kiemer AK, Wagner H and Vollmar AM: The triterpenoid quinonemethide pristimerin inhibits induction of inducible nitric oxide synthase in murine macrophages. Eur J Pharmacol. 336:211–217. 1997. View Article : Google Scholar

14 

Yousef BA, Hassan HM, Zhang LY and Jiang ZZ: Anticancer potential and molecular targets of pristimerin: A mini-review. Curr Cancer Drug Targets. 17:100–108. 2017. View Article : Google Scholar

15 

Lee SO, Kim JS, Lee MS and Lee HJ: Anti-cancer effect of pristimerin by inhibition of HIF-1α involves the SPHK-1 pathway in hypoxic prostate cancer cells. BMC Cancer. 16:7012016. View Article : Google Scholar

16 

Xie G, Yu X, Liang H, Chen J, Tang X, Wu S and Liao C: Pristimerin overcomes adriamycin resistance in breast cancer cells through suppressing Akt signaling. Oncol Lett. 11:3111–3116. 2016.PubMed/NCBI

17 

Yousef BA, Hassan HM, Guerram M, Hamdi AM, Wang B, Zhang LY and Jiang ZZ: Pristimerin inhibits proliferation, migration and invasion, and induces apoptosis in HCT-116 colorectal cancer cells. Biomed Pharmacother. 79:112–119. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Zhao H, Wang C, Lu B, Zhou Z, Jin Y, Wang Z, Zheng L, Liu K, Luo T, Zhu D, et al: Pristimerin triggers AIF-dependent programmed necrosis in glioma cells via activation of JNK. Cancer Lett. 374:136–148. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Deng Q, Bai S, Gao W and Tong L: Pristimerin inhibits angiogenesis in adjuvant-induced arthritic rats by suppressing VEGFR2 signaling pathways. Int Immunopharmacol. 29:302–313. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Huang S, He P, Peng X, Li J, Xu D and Tang Y: Pristimerin inhibits prostate cancer bone metastasis by targeting PC-3 stem cell characteristics and VEGF-induced vasculogenesis of BM-EPCs. Cell Physiol Biochem. 37:253–268. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Yousef BA, Guerram M, Hassan HM, Hamdi AM, Zhang LY and Jiang ZZ: Pristimerin demonstrates anticancer potential in colorectal cancer cells by inducing G1 phase arrest and apoptosis and suppressing various pro-survival signaling proteins. Oncol Rep. 35:1091–1100. 2016.PubMed/NCBI

22 

Zuo J, Guo Y, Peng X, Tang Y, Zhang X, He P, Li S, Wa Q, Li J, Huang S, et al: Inhibitory action of pristimerin on hypoxia-mediated metastasis involves stem cell characteristics and EMT in PC-3 prostate cancer cells. Oncol Rep. 33:1388–1394. 2015.PubMed/NCBI

23 

Ma YW, Liu YZ and Pan JX: Verteporfin induces apoptosis and eliminates cancer stem-like cells in uveal melanoma in the absence of light activation. Am J Cancer Res. 6:2816–2830. 2016.

24 

Jin B, Ding K and Pan J: Ponatinib induces apoptosis in imatinib-resistant human mast cells by dephosphorylating mutant D816V KIT and silencing β-catenin signaling. Mol Cancer Ther. 13:1217–1230. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Jin Y, Lu Z, Ding K, Li J, Du X, Chen C, Sun X, Wu Y, Zhou J and Pan J: Antineoplastic mechanisms of niclosamide in acute myelogenous leukemia stem cells: Inactivation of the NF-κB pathway and generation of reactive oxygen species. Cancer Res. 70:2516–2527. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Jin Y, Zhou J, Xu F, Jin B, Cui L, Wang Y, Du X, Li J, Li P, Ren R, et al: Targeting methyltransferase PRMT5 eliminates leukemia stem cells in chronic myelogenous leukemia. J Clin Invest. 126:3961–3980. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Pan J, Quintás-Cardama A, Kantarjian HM, Akin C, Manshouri T, Lamb P, Cortes JE, Tefferi A, Giles FJ and Verstovsek S: EXEL-0862, a novel tyrosine kinase inhibitor, induces apoptosis in vitro and ex vivo in human mast cells expressing the KIT D816V mutation. Blood. 109:315–322. 2007. View Article : Google Scholar

28 

Jin B, Wang C, Li J, Du X, Ding K and Pan J: Anthelmintic niclosamide disrupts the interplay of p65 and FOXM1/β-catenin and eradicates leukemia stem cells in chronic myelogenous leukemia. Clin Cancer Res. 23:789–803. 2017. View Article : Google Scholar

29 

Dai W, Zhou J, Jin B and Pan J: Class III-specific HDAC inhibitor Tenovin-6 induces apoptosis, suppresses migration and eliminates cancer stem cells in uveal melanoma. Sci Rep. 6:226222016. View Article : Google Scholar : PubMed/NCBI

30 

Morrison DJ, Hogan LE, Condos G, Bhatla T, Germino N, Moskowitz NP, Lee L, Bhojwani D, Horton TM, Belitskaya-Levy I, et al: Endogenous knockdown of survivin improves chemotherapeutic response in ALL models. Leukemia. 26:271–279. 2012. View Article : Google Scholar

31 

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

32 

Yarrow JC, Perlman ZE, Westwood NJ and Mitchison TJ: A high-throughput cell migration assay using scratch wound healing, a comparison of image-based readout methods. BMC Biotechnol. 4:212004. View Article : Google Scholar : PubMed/NCBI

33 

Dinicola S, Pasqualato A, Cucina A, Coluccia P, Ferranti F, Canipari R, Catizone A, Proietti S, D'Anselmi F, Ricci G, et al: Grape seed extract suppresses MDA-MB231 breast cancer cell migration and invasion. Eur J Nutr. 53:421–431. 2014. View Article : Google Scholar

34 

Toth M and Fridman R: Assessment of gelatinases (MMP-2 and MMP-9 by gelatin zymography. Methods Mol Med. 57:163–174. 2001.PubMed/NCBI

35 

Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, Brown M, Jacquemier J, Viens P, Kleer CG, Liu S, et al: ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell. 1:555–567. 2007. View Article : Google Scholar

36 

Nonaka M, Yawata T, Takemura M, Higashi Y, Nakai E, Shimizu K and Ueba T: Elevated cell invasion in a tumor sphere culture of RSV-M mouse glioma cells. Neurol Med Chir (Tokyo). 55:60–70. 2015. View Article : Google Scholar

37 

Chen LF and Greene WC: Shaping the nuclear action of NF-κB. Nat Rev Mol Cell Biol. 5:392–401. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Liu YB, Gao X, Deeb D, Brigolin C, Zhang Y, Shaw J, Pindolia K and Gautam SC: Ubiquitin-proteasomal degradation of antiapop-totic survivin facilitates induction of apoptosis in prostate cancer cells by pristimerin. Int J Oncol. 45:1735–1741. 2014.PubMed/NCBI

39 

Huang Q, Lan F, Wang X, Yu Y, Ouyang X, Zheng F, Han J, Lin Y, Xie Y, Xie F, et al: IL-1β-induced activation of p38 promotes metastasis in gastric adenocarcinoma via upregulation of AP-1/c-fos, MMP2 and MMP9. Mol Cancer. 13:182014. View Article : Google Scholar

40 

Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, Visvader J, Weissman IL and Wahl GM: Cancer stem cells-perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res. 66:9339–9344. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Bronkhorst IH and Jager MJ: Inflammation in uveal melanoma. Eye (Lond). 27:217–223. 2013. View Article : Google Scholar

43 

Nagarkatti-Gude N, Bronkhorst IH, van Duinen SG, Luyten GP and Jager MJ: Cytokines and chemokines in the vitreous fluid of eyes with uveal melanoma. Invest Ophthalmol Vis Sci. 53:6748–6755. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Storci G, Sansone P, Mari S, D'Uva G, Tavolari S, Guarnieri T, Taffurelli M, Ceccarelli C, Santini D, Chieco P, et al: TNFalpha up-regulates SLUG via the NF-kappaB/HIF1alpha axis, which imparts breast cancer cells with a stem cell-like phenotype. J Cell Physiol. 225:682–691. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2017
Volume 51 Issue 3

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
Zhang B, Zhang J and Pan J: Pristimerin effectively inhibits the malignant phenotypes of uveal melanoma cells by targeting NF‑κB pathway. Int J Oncol 51: 887-898, 2017
APA
Zhang, B., Zhang, J., & Pan, J. (2017). Pristimerin effectively inhibits the malignant phenotypes of uveal melanoma cells by targeting NF‑κB pathway. International Journal of Oncology, 51, 887-898. https://doi.org/10.3892/ijo.2017.4079
MLA
Zhang, B., Zhang, J., Pan, J."Pristimerin effectively inhibits the malignant phenotypes of uveal melanoma cells by targeting NF‑κB pathway". International Journal of Oncology 51.3 (2017): 887-898.
Chicago
Zhang, B., Zhang, J., Pan, J."Pristimerin effectively inhibits the malignant phenotypes of uveal melanoma cells by targeting NF‑κB pathway". International Journal of Oncology 51, no. 3 (2017): 887-898. https://doi.org/10.3892/ijo.2017.4079