
Psoralen is the parent compound in a family of furanocoumarins and naturally occurs in traditional medicinal plants such as
It is well known that p53 protein is a major tumor suppressor in diverse cancers, playing the role of a cell-cycle checkpoint to maintain genetic stability (Vogelstein
Although many aspects of PUVA therapy have been extensively studied, the effects of 8-MOP on human gastric cancer cells have not been elucidated. In this study, we found for the first time that 8-MOP alone inhibited the viability of the human gastric cancer cell line SNU1 via p53 upregulation and caspase-3 activation. Furthermore, 8-MOP reduces cell mobility to inhibit metastasis in a p53-independent manner. Overall, these results raise the possibility of 8-MOP becoming a part of combination chemotherapy for gastric cancer, in addition to other well-known drugs.
General chemicals and reagents were obtained from Sigma-Aldrich (St. Louis, MO, USA) unless stated otherwise. Gemcitabine and 8-MOP were purchased from Sigma-Aldrich. Inhibitors including Z-VAD-FMK, 3-MA, and necrostatin-1 were purchased from Selleckchem (Houston, TX, USA).
All human cancer cell lines were obtained from the Korean Cell Line Bank (KCLB, Seoul, Korea). Two cell lines, if available, were used for each type of cancer. The human cancer cell lines used in this study were as follows: A549 and H1650 (lung cancer); SNU449 and SK-Hep1 (hepatocellular carcinoma); MDA-MB453 and T47D (breast cancer); SKOV3 (ovarian cancer); PC3 and DU145 (prostate cancer); HCT15 and DLD1 (colorectal cancer); AGS and SNU1 (gastric cancer harboring wild-type p53); and SK-MEL-2 (skin cancer). Cells were cultivated in Roswell Park Memorial Institute (RPMI) medium supplemented with 10% fetal bovine serum (FBS) (Thermo-Fisher Scientific, Waltham, MA, USA) and 1% antibiotic-antimycotic solution at 37°C in 5% CO2 atmosphere, unless stated otherwise.
The cytotoxicity of 8-MOP against SNU1 cells was determined using the Cell Counting Kit-8 (CCK-8) assay kit (Sigma-Aldrich) according to the manufacturer’s instructions. Cells were seeded in 96-well plates and incubated for 18 h, followed by 8-MOP treatment at 2-fold increasing concentrations up to 250 µM for 48 h. Following treatment, the cells were incubated with 20 µL of CCK-8 solution for 1-2 h, and absorbance was measured at 460 nm using a microplate reader (SpectraMaxi3x; Molecular Devices, San Jose, CA, USA).
Cells (1×106) were treated with 100, 200, or 300 µM of 8-MOP for 24 h and then treated with trypsin. The detached cells were fixed and permeabilized with 70% methanol at –20°C overnight. After washing with ice-cold phosphate-buffered saline, the cells were stained with DAPI (4’,6-Diamidino-2-Phenylindole, Thermo-Fisher Scientific for cell cycle phase) or AnnexinV-PI (propidium iodide) (Annexin V apoptosis detection kit, Thermo-Fisher Scientific for apoptosis) in the dark for 10 min at room temperature, according to the manufacturer’s instructions. Flow cytometry was performed using Attune CytPix (Thermo-Fisher Scientific). Ten thousand events per sample were counted in duplicate.
Equal amounts of proteins were loaded on 10-15% SDS-PAGE gels, and the separated bands were transferred onto polyvinylidene fluoride (PVDF) membranes. Membranes were blocked with 5% non-fat milk in Tris-buffered saline and 0.1% Tween-20 buffer. Antibodies used in western blot analysis were purchased from Santa Cruz Biotechnology (Dallas, TX, USA), unless stated otherwise. The blotted membrane was then incubated with antibodies against cleaved caspase-3, poly(ADP-ribose) polymerase (PARP), MMP-2, MMP-9, Snail (Cell Signaling Technology, Danvers, MA, USA), β-actin, and p53 at 4°C overnight. After washing, the blots were incubated with horseradish peroxidase-labeled secondary antibodies at room temperature for 2 h. Bands were visualized by enhanced chemiluminescence using LuminiGraph I (ATTO, Tokyo, Japan).
Cells were transiently transfected with scrambled (sc)- or p53-siRNA using Lipofectamine RNAi MAX reagent (Thermo-Fisher Scientific/Invitrogen) according to the manufacturer’s protocol. Transfected cells were cultivated in the presence or absence of 200 μM 8-MOP for 48 h. The cells were stained with DAPI and subjected to flow cytometry. The p53-siRNA sequence used in this study was 5′-ACUCCACACGCAAAUUUCCTT-3′.
SNU1 cells were seeded in 6-well plates at a density of 5×105 cells/well to obtain a confluent monolayer. After overnight incubation, a straight line was scratched in the middle of the monolayer using a pipette tip. The cells were washed with 1 mL PBS to remove cellular debris, placed in 2 mL medium, and treated with 100, 200, or 300 μM 8-MOP. After 24 h of incubation, the width of the scratched wound was measured and compared to that observed at 0 h. All experiments were carried out in triplicate and repeated three times.
The migration ability of SNU1 cells was measured using Transwell chambers with 8 μm filter inserts (Corning Inc., Corning, NY, USA) without Matrigel (Corning Inc.). Cells were seeded in inserts (1×103 cells/insert) and treated with 100, 200, or 300 μM 8-MOP in RPMI-1640 medium without FBS. After incubation for 24 h, the inserts were fixed with 500 μL of 4% paraformaldehyde for 15 min and stained with a Brilliant Blue solution for 15 min. The inserts were washed several times with distilled water and the migrated cells were counted from an average of five random visual fields under a microscope. All experiments were performed in triplicates and repeated three times.
SNU1 cells were seeded in inserts (1×103 cells/insert) in a Transwell chamber (Corning Inc.) and treated with 100, 200, or 300 μM 8-MOP in RPMI-1640 medium without FBS. The outer membrane of a polycarbonate membrane insert (pore size 8 μm) was coated with 0.5 mg/mL collagen, and the insert was filled with 10 μL Matrigel diluted with 40 μL PBS. After incubation for 24 h at 37°C, the inserts were fixed with 500 μL of 4% paraformaldehyde for 15 min and stained with Brilliant Blue solution for 15 min. The insert was washed several times with distilled water and the migrated cells were counted from an average of five random visual fields under a microscope. All experiments were performed in triplicate and repeated three times.
All experiments were performed in triplicate and repeated at least three times. Data are represented as mean ± SD, unless indicated otherwise. Analysis of variance with Tukey’s test was performed for multiple comparisons among groups using GraphPad Prism 5.0 software (GraphPad, La Jolla, CA, USA). Results with
Following treatment with 8-MOP alone, we observed changes, in a dose-dependent manner, in the viability of diverse human cancer cell lines including lung, liver, breast, ovarian, prostate, colorectal, skin, and gastric cancers. As shown in Fig. 1, among the 14 cancer cell lines only a couple of gastric cancer cell lines showed detectable IC50 values (dotted line), when treated up to the 8-MOP concentration of 250 μM, as the follows: AGS cells, 280.1 μM; and SNU1 cells, 222.5 μM. In contrast, the remaining cell lines did not show detectable IC50 values, although some showed a slight decrease in their viability.
The IC50 analysis revealed, for the first time, that 8-MOP treatment alone, without UVA irradiation, inhibits the viability of human gastric cancer cells. These results prompted us to elucidate the mechanism underlying the anti-proliferative effects of 8-MOP in SNU1 cells, the most 8-MOP sensitive gastric cancer cell line.
As the first step in determining the anti-proliferative effects of 8-MOP in SNU1 cells, the percentile change of G1-arrest or apoptosis in response to treatment with 8-MOP was analyzed by flow cytometry following DAPI or annexin V/PI staining, respectively. As shown in Fig. 2A, the population of cells in the sub-G1 phase increased significantly in a dose-dependent manner following treatment with 8-MOP for 24 h, compared to the untreated cells. When treated at 100 μM 8-MOP, 17.1% of the cells remained in the sub-G1 phase; at 200 μM 8-MOP, 24.8%, and at 300 μM 8-MOP, 35.9%. Similarly, a profound increase (
Next, to confirm the underlying mechanism of 8-MOP-induced G1-arrest and apoptosis in SNU1 cells, western blot analysis was conducted using antibodies against p53, a canonical G1-arrest marker, and against PARP and cleaved caspase-3, typical apoptotic markers. As expected, we found that exposure of gastric cancer cell lines to 8-MOP resulted in a significant increase in the cellular levels of p53, PARP, and cleaved caspase-3 in a dose-dependent manner, compared to that in untreated cells (Fig. 2C). These results suggested that 8-MOP-induced G1-arrest and apoptosis in SNU1 cells were caused by the upregulation of p53 and caspase-3 via PARP, respectively. For further in-depth study, we determined whether 8-MOP-induced apoptosis was attributed to caspase-3, autophagy, or necrosis using inhibitors such as Z-VAD-FMK (pan-caspase inhibitor), 3-MA (autophagy inhibitor), and necrostatin-1 (necrosis inhibitor). As shown in Fig. 2D, pretreatment with 50 µM Z-VAD-FMK almost abrogated the activation of procaspase-3 and PARP induced by treatment with 200 µM 8-MOP in SNU1 cells. In contrast, pretreatment with 5 mM 3-MA or 50 µM necrostatin-1 had no effect. These results indicate that 8-MOP induces caspase-dependent apoptosis in the gastric cancer cell line SNU1.
The above results suggest that 8-MOP-induced G1-arrest and apoptosis are attributed to gain-of-function, such as p53 upregulation and activation of caspase-3, respectively. For further in-depth study, these results were validated through the concept of loss-of-function using specific siRNAs and inhibitors. In this regard, the p53-dependent G1-arrest and caspase-dependent apoptosis in response to 8-MOP treatment were confirmed using p53-siRNA and the apoptosis inhibitor, Z-VAD-FMK. Compared with no significant effects of sc-siRNA on G1-arrest, pretreatment with p53-siRNA significantly abrogated the 200 µM 8-MOP-induced G1-arrest in SNU1 cells via p53 knockdown (Fig. 3A). Similarly, pretreatment with 50 µM Z-VAD-FMK, an apoptosis inhibitor, significantly abrogated the 200 µM 8-MOP-induced G1-arrest in SNU1 cells (Fig. 3B). Consequently, 8-MOP-induced apoptosis did not occur after p53-siRNA pre-treatment, as determined by FACS analysis (data not shown). These results confirm that 8-MOP-induced G1-arrest followed by apoptosis is attributed to the upregulation of p53.
To identify effector proteins involved in 8-MOP-induced apoptosis, we screened the expression changes of several candidates using an immune blotting assay. We found that the expression of metastasis-related proteins, including MMP-2, MMP-9, and Snail, was inhibited despite upregulating the expression of p53 in response to 8-MOP treatment (Fig. 4A, upper part). The signaling pathways involved in 8-MOP-induced apoptosis and/or downregulation of metastasis-related proteins were attributed to the inactivation of well-known signaling MAP kinases, including PI3K, ERK2, and STAT3 (Fig. 4A, lower part). As 8-MOP inhibited the expression of metastasis-related proteins while increasing the expression of p53, we examined the relationship between p53 and metastasis-related proteins. As shown in Fig. 4B, the expression of metastasis-related proteins was only affected by the presence of 8-MOP, irrespective of expression of p53. These results suggest that the expression of metastasis-related proteins is not associated with the expression of p53.
These results prompted us to examine whether 8-MOP affects cell motility, including wound healing, invasion, and migration. As shown in Fig. 5, upper part, treatment with 8-MOP affected wound healing, migration, and invasion abilities in a dose-dependent manner compared to untreated controls. In detail (Fig. 5, lower part), the wound healing rates after 24-h of treatment with 8-MOP concentrations of 100, 200, and 300 µM decreased to 94.7%, 54.3%, and 44.6%, respectively, when compared to those of controls. Cell populations migrating through the membrane in the group after 24-h treatment with 8-MOP concentrations of 100, 200, and 300 µM decreased to 68.5%, 82.3%, and 96.5%, respectively. Similarly, the percentage of invasive cells passing through Matrigel in the group after 24-h treatment with 8-MOP concentrations of 100, 200, and 300 µM was inhibited by 63.4%, 84.5%, and 99.9%, respectively.
Since the combination chemotherapy of 5-FU and a platinum analog has been established as a first-line global standard regimen for treatment of metastatic gastric cancer, similar research is ongoing for clinical trials (Takashima
8-MOP is the parent compound that naturally occurs in traditional medicinal plants from the historical period. Due to the nature of naturally occurring phytochemicals, orally administered 8-MOP is safer for humans, even when used as a food supplement or chemotherapy, with fewer side effects compared to other chemotherapeutic chemicals. To date, 8-MOP has been studied as a component of the PUVA therapy. In contrast, diverse animals and human cancer cells were tested to study their sensitivity to 8-MOP alone without UVA. Recently, research areas have been extended to study other effects of 8-MOP or to find other sensitive tissues on the skin. For example, the antimetastatic effect of 8-MOP has been reported in hepatocellular carcinoma (Xiong
p53 and caspase-3 are well-known genetic guardians and apoptosis effectors. Moreover, downregulation of p53 has been well documented in gastric cancer (Fenoglio-Preiser
In this study, we investigated, for the first time, the mechanisms underlying 8-MOP-induced cytotoxic and anti-metastatic effects on the human gastric cancer cell line, SNU1. Overall, we demonstrated the potential use of the phytochemical 8-MOP in the treatment of human gastric cancer.
This research was supported by the National Research Foundation of Korea (NRF) and funded by the Ministry of Science and ICT of Korea (NRF-2017M3A9B5060881, NRF-2017M3C9A5028693, and NRF-2017M3A9B5060880). Human resources were supported by the Chungnam National University and the Korea Research Institute of Bioscience and Biotechnology (KRIBB).
The authors declare that there are no conflicts of interest.
![]() |
![]() |