Title: EtNBSe-PDT inhibited proliferation and induced autophagy of HNE-1 cells via downregulating the Wnt/β-catenin signaling pathway
Authors: An Liu, Weiwei Zhang, Yihui Chen, Dawei Zhou, Zhen Wang, Jian Kang, Le Wei
PII: S1572-1000(18)30392-2
DOI: https://doi.org/10.1016/j.pdpdt.2019.02.024
Reference: PDPDT 1371
To appear in: Photodiagnosis and Photodynamic Therapy
Received date: 9 November 2018
Revised date: 19 January 2019
Accepted date: 25 February 2019
Please cite this article as: Liu A, Zhang W, Chen Y, Zhou D, Wang Z, Kang J, Wei L, EtNBSe-PDT inhibited proliferation and induced autophagy of HNE-1 cells via downregulating the Wnt/β-catenin signaling pathway, Photodiagnosis and Photodynamic Therapy (2019), https://doi.org/10.1016/j.pdpdt.2019.02.024
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
EtNBSe-PDT inhibited proliferation and induced autophagy of HNE-1 cells via downregulating the Wnt/β-catenin signaling pathway
Authors’ names: An Liu1, Weiwei Zhang2, Yihui Chen2, Dawei Zhou2, Zhen Wang2, Jian Kang2*, Le Wei2*
Authors’ affiliations:
1 Department of Otorhinolaryngology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
2 Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
* Corresponding authors:
Jian Kang, Department of Dermatology, Third Xiangya hospital, Central south University, Tongzipo road #138, Changsha, Hunan 410013, China
Email: [email protected]
Le Wei, Department of Dermatology, Third Xiangya hospital, Central south University, Tongzipo road #138, Changsha, Hunan 410013, China
Email: [email protected]
Highlights
• The study aimed to determine the influence of EtNBSe medicated PDT on autophagy, proliferation and Wnt/β-catenin pathway in human NPC cell line (HNE-1 cells), and further explore the underlying crosstalk between them.
• The EtNBSe explored here is a novel synthetic bipolar photosensitizer with stable chemical structure, high permeability, rapid absorption, and oncotarget accumulation.
• The study demonstrated that EtNBSe-PDT suppressed viability and proliferation, and induced autophagy of HNE-1 cells via downregulating the Wnt/β-catenin pathway.
• The autophagy induced by EtNBSe-PDT further constituted the cytoprotective mechanisms involved in HNE-1 cells.
Abstract
Background: Increasing evidence has suggested that autophagy may play a resistant role during photodynamic therapy (PDT). The Wnt/β-catenin pathway is tightly involved in cell proliferation and autophagy. In this study, we aimed to determine the influence of 5-Ethylamino-9-diethylaminobenzo[a]phenoselenazinium (EtNBSe) mediated PDT (EtNBSe-PDT) on autophagy, proliferation and Wnt/β-catenin pathway in human NPC cell line (HNE-1 cells), and further explore the underlying crosstalk between them.
Methods: Cell viability and proliferation was evaluated by MTT assay. Autophagy and Wnt/β-catenin signaling pathway was analyzed by western blotting and immunofluorescence.
Results: It was revealed that EtNBSe-PDT significantly impeded the viability and proliferation of HNE-1 cells. Meanwhile EtNBSe-PDT could notably induce autophagy in HNE-1 cells accompanied with the inhibition of Wnt/β-catenin pathway. The Wnt/β- catenin pathway activator Wnt agonist was found to partially counteract the inhibitory proliferation of HNE-1 cells and suppress the autophagy induced by EtNBSe-PDT. In addition, pretreatment with the autophagy inhibitor 3-methyladenine (3-MA) or Wnt agonist showed the potential in enhancing the cytotoxic effect of EtNBSe-PDT (cell survival from 50.71±4.16% to 24.53±4.27% and from 52.64±3.54% to 35.74±4.27% respectively).
Conclusion: Taken together, this study demonstrated that EtNBSe-PDT suppressed viability and proliferation, and induced autophagy of HNE-1 cells via downregulating the Wnt/β-catenin pathway. The autophagy further constituted the cytoprotective mechanisms involved in HNE-1 cells, which suggested that the combination of EtNBSe-PDT and autophagy inhibitors may be a promising strategy for the treatment of human NPC.
Key words : PDT, Wnt/β-catenin, autophagy, proliferation, EtNBSe
Introduction
PDT is an effective, minimally invasive and local targeting clinical approach. It involves the systemic or topical administration of a target localizing photoactivatable compound (photosensitizer) and subsequent irradiation with light of a certain wavelength. When illuminated, the photosensitizer becomes a cytotoxin and generates large amounts of reactive oxygen species, thus inducing complicated biological responses which leads to cell death.1 Since PDT causes essentially no effect on connective tissues, the anatomical integrity of hollow organs such as the nasopharynx can be preserved in patients undergoing PDT.2 Therefore, endoscopic PDT with the fiber optic delivery system is especially suitable for NPC with surgical contraindications. In terms of a successful PDT, the damaging stress led by PDT must exceed the cytoprotection threshold. Otherwise, the remaining cells may repopulate the tumor and form a resistance to PDT.3 Accordingly, in order to improve the therapeutic efficacy of PDT, the mechanisms of PDT-induced cell damage as well as possible cell resistance must be further clarified.
Autophagy is a form of stress adaption response when cells are subjected to environmental stress. In proficient autophagy, cells maintain the intracellular homeostasis by recycling their damaged or degenerated components, hence mediating a cytoprotective effect. And previous studies have reported that blockage of autophagy with pharmacological or genetic interventions accelerated the demise of cells responding to stress.4 Nevertheless, in a number of pathological settings, the molecular machinery for autophagy contributes to cellular demise directly or favors the engagement of other regulated cell death modalities.5 Noteworthily, our previous research and numerous other studies have revealed the potential involvement of PDT in triggering autophagy.6 However, the exact role of autophagy in response to PDT is still elusive and the precise molecular mechanisms behind PDT-induced autophagy are not well understood. Wnt/β-catenin signaling pathway is well known to be sensitive to intracelluar stress and has been reported to be affected by PDT.7 Moreover, previous studies reported that certain chemical agents could induce autophagy via Wnt/β-catenin signaling pathway in human tumor cell lines.8,9 However, the detailed mechanism remains unclear.
EtNBSe is a new synthetic bipolar photosensitizer with many advantages including stable chemical structure, high permeability, rapid absorption, and oncotarget accumulation.10 Moreover, it has been substantiated that the photoactivity of EtNBSe is 10,000 times as much as that of photofrin and other photosensitizers in the benzophenoxazinium family.10 Therefore, we explored the effect of EtNBSe-PDT on Wnt/β-catenin pathway, along with its function in NPC cell proliferation and autophagy. In addition, further studies managed to demonstrate autophagic impacts in cell resistance against EtNBSe-PDT of HNE-1 cells.
Materials and methods Reagents
EtNBSe was synthesized by the chemistry department of Central South University (Changsha, China) using the synthetic method reported in our previous study and its chemical structure is shown in Figure 1.10 Fetal bovine serum (FBS) and Dulbecco’s modified Eagle’s medium (DMEM) were purchased from Gibco (Grand Island, NY, USA). Wnt agonist (sc-222416) and 3-methyladenine (3-MA; sc-205596) were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). 4,6-diamidino-2- phenylindole (DAPI; D8417) and Thiazolyl Blue Tetrazolium Bromide (MTT; M2128) were obtained from Sigma-Aldrich (Danvers, MA, USA). The anti-Cyclin-D(sc-753), anti-β-catenin(sc-7199), anti-c-Myc(sc-40), anti-GSK-3β(sc-7291), anti-Beclin-1(sc- 48341), anti-LC3-I(sc-134226), anti-LC3-II(sc-28266), anti-GAPDH (sc-293335) antibodies were purchased from Santa Cruz Biotechnology Inc. (Dallas, TX, USA). Detailed information about the antibodies is listed and characterized in Table 1. The autophagy activator rapamycin (#9004) was purchased from Cell Signaling Technology (Shanghai, People’s Republic of China).
Cell culture
HNE-1 cells were purchased from Shanghai cell bank at the Chinese Academy of Science (Shanghai, China). All cells were cultured in DMEM medium (Gibco, Grand Island, NY, USA) supplemented with 10% FBS, 100 U/ml penicillin, 0.1 mg/ml streptomycin, and maintained at 37 °C under a humidified atmosphere of 5% CO2.
PDT protocol in vitro
In all EtNBSe-PDT groups, HNE-1 cells were seeded in 6-well culture plates at density of 6×105 cells/well and were incubated for 24 h. Then, medium was aspirated from cultures and replaced with PBS containing various concentrations of EtNBSe (100, 200, 400, 600 and 800 nmol/L). The cells were incubated with EtNBSe for 20min in the dark and then irradiated in the EtNBSe-containing PBS. The irradiation was done using an LED with a wavelength of 635 nm and a laser power density of 50 mW/cm2. Furthermore, the total energy densities were adjusted by the various irradiation time (0, 30, 60,120 and 240 s; 0, 1.4, 2.8 and 5.6 J/cm2 light energy density correspondingly). After light irradiation, fresh culture medium was replaced. Then, the treated cells were maintained at 37 °C under an atmosphere of 5% CO2 in the dark for 24 h and the cell viability and protein levels were evaluated. In the EtNBSe or LED alone group, cells were subjected to only EtNBSe or LED light in the same manner as the EtNBSe-PDT group.
MTT analysis
MTT was used to detect cell viability and proliferation. HNE-1 cells at 5 × 103 cells/well were seeded on 96-well plates. After the attachment, the cells were treated according to the requirement of corresponding groups and incubated for 0, 12, 24, 48 or 72h at 37℃. 10μL MTT (5mg/ml in PBS) was added to each well. Following incubation for 2h, the supernatants were removed and 150 µl DMSO was added into each well to dissolve the formazan crystals. After oscillated for 10 min, the 96-well plates were then placed in a Microplate reader (Bio-Rad, Hercules, CA, USA) to assess the absorbance at 490 nm.
Immunofluorescence
HNE-1 cells were grown on coverslips. At 4 h after PDT, HNE-1 cells were washed with PBS for 2 min, fixed with 4% PFA for 10 min and gently rinsed in 0.3% Triton X- 100 for 15 min at room temperature. After washing with PBS for 3 min, the cells were blocked with 5% BSA for 1 h and were then incubated with anti-lamp, anti-LC3-II and anti-β-catenin antibodies (dilution 1:500) overnight at 4°C. Afterwards, Alexa Fluor 577-labeled, Alexa Fluor 647-labeled and Alexa Fluor 647-labeled secondary antibodies (dilution 1:200) were added respectively, to develop the fluorescence staining in a dark chamber. The fluorescence intensity and the co-localization were observed under a fluorescence microscope (BX41 OLYMPOS, Tokyo, Japan) and quantitatively analyzed by Image Pro Plus 6.0.
Western-blotting analysis
HNE-1 cells after corresponding treatments were washed three times with precooled PBS and harvested in sample buffer containing 20 mmol/L Tris-HCl (pH 8.8), 2.5 mmol/L EDTA, 150 mmol/L NaCl, 10% glycerine, 10% SDS, 1% Triton X-100, 10 mmol/L sodium pyrophosphate and 1 mmol/L PMSF for 30min. Then the supernatants from cells were centrifugated at 10000× for 15 min and the sample-loading buffer was added. Equal amount of total protein was loaded and separated by 10% SDS-PAGE and electrophoretically transferred to the polyvinylidene difluoride (PVDF) membrane. The membrane was then blocked for 1 h with 5% bovine serum albumin in Tris-buffered saline and incubated with primary antibodies (listed and characterized in Table 1) overnight at 4°C. Next the membrane was washed three rounds with PBST and was incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies (1:1000) for 2 h at room temperature. Then following another round of washing with TBST, the membrane was developed using electrochemiluminescence, and the signals were detected using Quantity One 1-D Analysis Software (Bio-Rad, Hercules, CA, USA).
Cell cycle analysis
HNE-1 cells were seeded in the 96-well plates at the density of 5 × 103 cells/well and incubated for 24 hours. After corresponding treatments, cells were harvested and fixed with 75% cold ethanol overnight at 4℃. The fixed cells were washed three times with PBS and stained darkly with propidium iodide (PI) for 30 minutes. The stained cells were analyzed by FACS Calibur flow cytometer (BD Biosciences, San Jose, CA, USA). Statistical analysis All quantitative data were presented as the mean ± SD from of triplicate independent experiments. The SPSS 19.0 software package was used to perform all statistical analysis. Comparisons between two groups were performed using the Student’s-t test, and between multiple groups using ANOVA analysis. P-values of <0.05 were considered statistically significant. Results EtNBSe-PDT decreased cell viability in HNE-1 cells In order to investigate the antitumor effect of EtNBSe-PDT on NPC cells, human NPC cell line, HNE-1, was cultured with various concentrations of EtNBSe (0, 100, 200, 400, and 600 nmol/L). And then the treated HNE-1 cells were exposed to LED light with increasing light densities, followed by the measurement of cell viability using MTT. As shown in Figure 2, the viability of HNE-1 cells notably decreased in concentration- and light energy density-dependent manner. And cell viabilities in all EtNBSe-PDT groups treated with 400, 600 and 800 nmol/L EtNBSe and the groups treated with 200nmol/L EtNBSe combined with 2.8 and 5.6 J/cm2 light doses were significantly suppressed when compared with the control group (0 nmol/L EtNBSe, 0 J/cm2). Additionally, the EtNBSe and LED alone groups showed no significant inhibition of cell viability. Furthermore, at a light dose of 2.8 J/cm2, the half maximal inhibitory concentration of EtNBSe was 400 ± 23 nmol/L. The inhibition rate in the group that received 400 nmol/L EtNBSe combined with a light dose of 2.8 J/cm2 was 49.21 ± 4.23 %. Thus, all subsequent experiments were performed using 400 nmol/L EtNBSe combined with 2.8 J/cm2 light energy density. EtNBSe-PDT induced autophagy in HNE-1 cells Previous studies revealed that autophagy could be activated by some photosensitizers mediated PDT. In this study, we investigated whether EtNBSe-PDT could induce autophagy in HNE-1 cells and the well-known autophagy activator Rapamycin was used as the positive control. Firstly, the expression levels of autophagy-related protein LC3-I, LC3-II and Beclin-1 were detected by Western blotting. Upon the induction of autophagy, LC3-I was transformed into LC3-II, which contributed favorably to the formation of autophagosomes. Thus, the LC3-II/LC3-I ratio indicated the level of autophagy. Figure 3A and 3B showed that the ratio of LC3-II/LC3-I and expression of Beclin-1 were significantly increased in 200 and 400 nmol/L EtNBSe combined with a light dose of 2.8 J/cm2 treated groups. In addition, autophagy is characterized by the formation of autophagosomes which then fuse with the lysosome, forming autolysosome. Thus, the relative fluorescence level of LC3-II and Lamp was evaluated to monitor autophagosome and lysosome formation. As shown in Figure 3C and 3D, the staining of LC3-II was distributed evenly throughout the cell in the control group, whereas EtNBSe-PDT resulted in more distinctive LC3-II spots and a significant increase in the relative fluorescence intensity of LC3-II in the HNE-2 cells, which was consistent with our western blotting findings. Meanwhile obvious colocalization between the LC3- and Lamp-positive staining was observed 2 h after EtNBSe-PDT, indicating the formation of autolysosome. Taken together, these results indicated that EtNBSe-PDT induced notable autophagy in HNE-1 cells. EtNBSe-PDT suppressed the Wnt/β-catenin pathway in HNE-1 cells Wnt/β-catenin signaling pathway plays an important oncogenic role in many cancers. Moreover, numerous studies have reported that many antitumor therapies could induce cancer cell death by modulating this signaling pathway. Given that, we investigated the effect of EtNBSe-PDT on Wnt/β-catenin pathway in HNE-1 cells. As shown in Figure 4A and 4B, 200 and 400nmol/L EtNBSe combined with a light dose of 2.8 J/cm2 caused a significant decrease of β-catenin expression. And the expressions of c-Myc and Cyclin-D, well known β-catenin downstream targets were also significantly downregulated by EtNBSe-PDT. Next we explored the impact of EtNBSe-PDT on distribution of β-catenin in HNE-1 cells. And obvious nuclear export of β-catenin was observed in EtNBSe-PDT groups. As shown in Figure 3C, the RFP-β-catenin shuttled from the nucleus into cytoplasm after administration indicating the inhibition of Wnt/β- catenin pathway. Significantly, β-catenin gradually exhibited an evenly cytoplasmic distribution, which represented the formation of β-catenin destruction complex. Meanwhile, western blotting was used to detect the expression of GSK3β which was the component of destruction complex. We found that the expression of GSK3β increased by nearly two times in EtNBSe-PDT groups compared with the control group.Together, these data indicated that EtNBSe-PDT could inhibit the activation of Wnt/β- catenin pathway in HNE-1 cells. EtNBSe-PDT inhibited proliferation via suppressing the Wnt/β-catenin pathway in HNE-1 cells To further elucidate the cytotoxic impact of EtNBSe-PDT, cells were collected at various time points post EtNBSe-PDT and the proliferation of HNE-1 cells was assessed by MTT assay. It was showed that EtNBSe-PDT significantly inhibited the proliferation of HNE-1 cells compared with the control, but the Wnt/β-catenin activator, Wnt agonist partly rescued the suppression in cell proliferation induced by EtNBSe- PDT (Figure 5A). As reported, the inhibition of proliferation maybe partially attributed to cell cycle arrest.11 Subsequently, we investigated the cell cycle profile of HNE-1 cells using PI staining and flow cytometry. As shown in Figure 5B, EtNBSe-PDT caused an obvious accumulation in G1 phase arrest (from 42.57±2.62% to 57.19±3.87%) accompanied by a decreased cell population in G2/M phases (from 34.81±2.35% to 18.19±1.08%) compared with the control group. The percentage of cell entering the G2 and M phases in the combination of EtNBSe-PDT and Wnt agonist group was 29.16±2.17%, which was greatly more than that of EtNBSe-PDT alone. Meanwhile, data showed that the percentage of EtNBSe-PDT treated cells in the G1 phase was 57.19±3.87%. This value considerably declined to 45.29±3.66% in group co-treatment of EtNBSe-PDT and Wnt agonist. These data indicated that EtNBSe-PDT inhibits proliferation of HNE-1 cells by inducing G1 phase cell cycle arrest via suppressing Wnt/β-catenin. Wnt/β-catenin pathway was involved in autophagy induced by EtNBSe-PDT in HNE-1 cells To further confirm the role of cross-talking between autophagy and Wnt/β-catenin pathway in HNE-1 cells, we pretreated some cells with 1μmol/L Wnt agonist which could activate the Wnt/β-catenin signaling pathway for two hours. Then we performed HNE-1 cells with EtNBSe-PDT (400 nmol/L, 2.8 J/cm2) and assessed the level of autophagy. Meanwhile, 3-MA, an autophagy inhibitor was set as the negative control. As shown in Figure 5A and 5B, the expression of Beclin-1 and the ratio of LC3-II/LC3- I were elevated by EtNBSe-PDT and suppressed by Wnt agonist compared with the control group. As expected, the increased expression of Beclin-1 and the ratio of LC3- II/LC3-I were obviously whittled down by the combination of EtNBSe-PDT and 3-MA compared with the individual role of EtNBSe-PDT. Moreover, the promoting effect of EtNBSe-PDT on the level of autophagy was significantly counteracted by the cooperation of Wnt agonist. Thus these results above indicated that the inhibition of Wnt/β-catenin signaling pathway was involved in the EtNBSe-PDT induced autophagy. Inhibition of autophagy enhanced the cytotoxic effect of EtNBSe-PDT in HNE-1 cells. To further clarify the relative role of autophagy in HNE-1 cells escape from EtNBSe- PDT induced inhibitory cell viability, autophagy was inhibited by 3-MA and cell viability was measured using MTT. Meanwhile, the combination of PDT and rapamycin group was set as the positive control. As shown in Figure7, Similar results were observed when autophagy was suppressed by activating Wnt/β-catenin pathway using Wnt agonist. The MTT results showed that activation of Wnt/β-catenin pathway significantly enabled the enhancement of EtNBSe-PDT induced cell death and decreased the cell viability from 52.64±3.54% to 35.74±4.27%. Furthermore, treatment with the autophagy activator rapamycin decreased cytotoxicity in the HNE-1 cells treated with PDT compared with cells that did not receive rapamycin. However, solely using 3-MA, Wnt agonist or rapamycin did not appear to exert an obvious effect on the A-431 cell survival. Collectively, these data showed that induction of autophagy by EtNBSe-PDT protected HNE-1 cells from PDT-induced cell death, which may make HNE-1 cells resistant to EtNBSe-PDT. Discussion NPC is one of the most challenging malignancies arising from the epithelial cells lining the nasopharynx.12 It accounts for over 80% of all head and neck cancers in China. The anatomical position of NPC is adjacent to important nerves and vessels, leading to difficulty in the use of surgery for treatment. Currently, radiotherapy, with or without chemotherapy, has become the primary treatment for NPC. But these therapies produce severe adverse effects and the overall 5 years survival in patients is still only 35%-55% after a standard treatment.13-15 Therefore, it is imperative to develop new antitumor modalities to reduce side effects and to reinforce the effectiveness of therapy in NPC. PDT has emerged as a minimally invasive cancer treatment based on the creation of extreme stress in tumor cells through the excitation of photosensitizer by light.16,17 EtNBSe is a novel synthetic bipolar photosensitizer having a heavier atom in place of the centrally located atom, which has the potential to possess much improved photoactivity.18 Furthermore, our previous studies have shown that EtNBSe-PDT possesses anti-tumor effects on skin squamous cell carcinoma.10,19 However, its related mechanisms in NPC remain largely unknown. In this study, our results showed that EtNBSe-PDT significantly suppressed HNE-1 cell viability in a concentration- and light energy density-dependent manner. In contrast, treatment with EtNBSe or light alone showed no obvious inhibitory viability effect on HNE-1 cells. These results indicated that EtNBSe possessed low dark toxicity and high phototoxicity to HNE-1 cells, making it a promising photosensitizer in photodynamic therapy to NPC. Next, we studied the effects of the combination of 400 nmol/L EtNBSe and a light dose of 2.8J/cm2 on HNE-1 cells. The results suggested that EtNBSe-PDT could inhibit cell proliferation, induce cell cycle arrest and autophagy. Concerning autophagy in cancer, this process can be a pro-death as well as a pro-survival pathway and it is largely dependent on the complicated autophagic regulatory machinery. Accordingly, accumulating studies have also suggested the dual effect of autophagy on PDT.20-22 Ji et al. found that the death of PC12 and CL1-0 cells induced by 5-aminolevulinic acid mediated PDT could be inhibited by an autophagy inhibitor, indicating that the cell death was autophagic in nature.23 In contrast, Michael et al. suggested that autophagy might promote cell adaptation and delayed cell death against PDT at low dose.24 In our experiments, inhibition of autophagy enhanced the cytotoxic effect of EtNBSe-PDT, demonstrating that autophagy contribute to the resistance of HNE-1 cells to EtNBSe- PDT. The detailed mechanisms mediating autophagy were complicated, and may differed with photosensitizer, cellular genotype, and target molecule.25,26 Further research on the mechanisms underlying autophagy and cytotoxic effect of PDT, will be crucial to provide additional efficient targets for therapeutic intervention in PDT. Accumulating evidence has demonstrated that the Wnt/β-catenin signaling pathway plays a decisive role in regulating the cellular fate and is overactivated in several human cancers, including NPC.27 The Wnt/β-catenin pathway is canonical pathway of Wnt families, which is the pleiotropic protein, β-catenin, dependent. It results in the stabilization of a cytoplasmic pool of β-catenin that would otherwise be marked for proteasomal-mediated degradation by a destruction complex, composed of GSK-3β. Upon binding to receptors of the Frizzled family and the co-receptors, extracellular Wnt stimulation transmits a signal to the intracellular adaptor, which inhibits the destruction complex and allows nuclear translocation of stabilized β-catenin to activate gene transcription.28 In this study, the notable decreased expression of β-catenin and obvious cytoplasmic translocation of β-catenin were observed after the EtNBSe-PDT administration. Consistently, the expression levels of c-Myc and Cyclin-D, the proteins synthetized by target genes of β-catenin, were suppressed. All these results suggested that the Wnt/β-catenin pathway was inhibited by EtNBSe-PDT in HNE-1 cells. However, the role of the Wnt/β-catenin pathway in autophagy is still unclear. Yun et al. activated the Wnt/β-catenin signaling pathway by a tumor suppressor, and found promoted autophagy in human NSCLC cells.29 Contrary to this finding, here we provided evidence that the pre-treatment of Wnt/β-catenin pathway activator, Wnt Agonist, significantly decreased the autophagy induced by EtNBSe-PDT, indicating that Wnt/β-catenin pathway played a negative role in the induction of autophagy of HNE-1 cells. Similarly, Li et al. also suggested that autophagy could be induced by the triptolide via repressing Wnt/β-catenin signaling in osteosarcoma treatment.30 Indeed, different tumor mutational status, intracellular microenvironment and complicated crosstalk between signaling pathway may account for these discrepancies. Furthermore, we used pharmacological inhibitor and activating Wnt/β-catenin signaling pathway to downregulate the level of autophagy under our experimental conditions. It was found that both of the two methods could significantly enhance the sensitivity of HNE-1 cells towards EtNBSe-PDT by a cytotoxicity and proliferation assay. In conclusion, we demonstrated that EtNBSe-PDT significantly inhibited the viability of HNE-1 cells. In addition, EtNBSe-PDT could suppress the Wnt/β-catenin signaling pathway, which further promoted autophagy and inhibited proliferation in HNE-1 cells. In particular, inhibition of autophagy enhanced the cytotoxicity of EtNBSe-PDT, which suggests that a combination of EtNBSe-PDT and autophagy inhibitors may be a promising strategy for the treatment of human NPC cell lines. Acknowledgments This work was supported by the New Xiangya Talent Project of the Third Xiangya hospital of Central South University (20170301). Conflict of Interest The authors have declared that no conflicts of interest exist. References 1. Li B, Lin L, Lin H, Wilson BC. Photosensitized singlet oxygen generation and detection: Recent advances and future perspectives in cancer photodynamic therapy. J Biophotonics. 2016;9(11- 12):1314-1325. 2. Wei MF, Chen MW, Chen KC, et al. Autophagy promotes resistance to photodynamic therapy- induced apoptosis selectively in colorectal cancer stem-like cells. Autophagy. 2014;10(7):1179- 1192. 3. Liu K, Yao H, Wen Y, et al. Functional role of a long non-coding RNA LIFR-AS1/miR-29a/TNFAIP3 axis in colorectal cancer resistance to pohotodynamic therapy. Biochim Biophys Acta Mol Basis Dis. 2018;1864(9 Pt B):2871-2880. 4. Kaizhen Y, Ting N, Mengyu L, Lihua T, Ling K. Enhanced cytotoxicity and apoptosis through inhibiting autophagy in metastatic potential colon cancer SW620 cells treated with Chlorin e6 photodynamic therapy. Photodiagnosis Photodyn Ther. 2018; S1572-1000(18):30221-7. 5. Feng X, Shi Y, Xie L, et al. 2-deoxy-D-glucose augments photodynamic therapy induced mitochondrial caspase-independent apoptosis and energy-mediated autophagy. Lasers Surg Med. 2018;58:654-664. 6. Chen J, Huang JH, Wang Z, et al. Endoplasmic reticulum stress-mediated autophagy contributes to 5-ethylamino-9-diethylaminobenzo[a]phenoselenazinium-mediated photodynamic therapy via the PERK-eIF2alpha pathway. Onco Targets Ther. 2018;11:4315-4325. 7. Ye X, Lin J, Lin Z, et al. Axin1 up-regulated 1 accelerates stress-induced cardiomyocytes apoptosis through activating Wnt/beta-catenin signaling. Exp Cell Res. 2017;359(2):441-448. 8. Hou S, Tian T, Qi D, et al. S100A4 promotes lung tumor development through beta-catenin pathway-mediated autophagy inhibition. Cell Death Dis. 2018;9(3):277. 9. Hu P, Ke C, Guo X, et al. Both glypican-3/Wnt/beta-catenin signaling pathway and autophagy contributed to the inhibitory effect of curcumin on hepatocellular carcinoma. Dig Liver Dis. 2018;S159: 799-805. 10. Foley JW, Song X, Demidova TN, Jalil F, Hamblin MR. Synthesis and properties of benzo[a]phenoxazinium chalcogen analogues as novel broad-spectrum antimicrobial photosensitizers. J Med Chem. 2006;49(17):5291-5299. 11. Tomoyasu C, Kikuchi K, Kaneda D, et al. OBP801, a novel histone deacetylase inhibitor, induces Mphase arrest and apoptosis in rhabdomyosarcoma cells. Oncol Rep. 2018;68:153-158. 12. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;136(5):E359-86. 13. Jiong L, Berrino F, Coebergh JW. Variation in survival for adults with nasopharyngeal cancer in Europe, 1978-1989. EUROCARE Working Group. Eur J Cancer. 1998;34(14 Spec No):2162-2166. 14. Lu H, Yao M. The current status of intensity-modulated radiation therapy in the treatment of nasopharyngeal carcinoma. Cancer Treat Rev. 2008;34(1):27-36. 15. Marks JE, Phillips JL, Menck HR. The National Cancer Data Base report on the relationship of race and national origin to the histology of nasopharyngeal carcinoma. Cancer. 1998;83(3):582- 588. 16. Lee HH, Choi MG, Hasan T. Application of photodynamic therapy in gastrointestinal disorders: an outdated or re-emerging technique? Korean J Intern Med. 2017;32(1):1-10. 17. Wen X, Li Y, Hamblin MR. Photodynamic therapy in dermatology beyond non-melanoma cancer:An update. Photodiagnosis Photodyn Ther. 2017;19:140-152. 18. Zhang XF, Zhang I, Liu L. Photophysics of halogenated fluoresceins: involvement of both intramolecular electron transfer and heavy atom effect in the deactivation of excited states. Photochem Photobiol. 2010;86(3):492-498. 19. Akilov OE, Kosaka S, O'Riordan K, et al. The role of photosensitizer molecular charge and structure on the efficacy of photodynamic therapy against Leishmania parasites. Chem Biol. 2006;13(8):839-847. 20. Lin S, Yang L, Shi H, et al. Endoplasmic reticulum-targeting photosensitizer Hypericin confers chemo-sensitization towards oxaliplatin through inducing pro-death autophagy. Int J Biochem Cell Biol. 2017;87:54-68. 21. Ouyang G, Xiong L, Liu Z, et al. Inhibition of autophagy potentiates the apoptosis-inducing effects of photodynamic therapy on human colon cancer cells. Photodiagnosis Photodyn Ther. 2018;21:396-403. 22. Rodriguez ME, Catrinacio C, Ropolo A, Rivarola VA, Vaccaro MI. A novel HIF-1alpha/VMP1- autophagic pathway induces resistance to photodynamic therapy in colon cancer cells. Photochem Photobiol Sci. 2017;16(11):1631-1642. 23. Ji HT, Chien LT, Lin YH, Chien HF, Chen CT. 5-ALA mediated photodynamic therapy induces autophagic cell death via AMP-activated protein kinase. Mol Cancer. 2010;9:91. 24. Dewaele M, Martinet W, Rubio N, et al. Autophagy pathways activated in response to PDT contribute to cell resistance against ROS damage. J Cell Mol Med. 2011;15(6):1402-1414. 25. Domagala A, Stachura J, Gabrysiak M, et al. Inhibition of autophagy sensitizes cancer cells to Photofrin-based photodynamic therapy. BMC Cancer. 2018;18(1):210. 26. Cisarikova A, Barbierikova Z, Janovec L, et al. Acridin-3,6-dialkyldithiourea hydrochlorides as new photosensitizers for photodynamic therapy of mouse leukemia cells. Bioorg Med Chem. 2016;24(9):2011-2022. 27. Zhang J, Wen X, Ren XY, et al. YPEL3 suppresses epithelial-mesenchymal transition and metastasis of nasopharyngeal carcinoma cells through the Wnt/beta-catenin signaling pathway. J Exp Clin Cancer Res. 2016;35(1):109. 28. Pashirzad M, Shafiee M, Khazaei M, et al. Therapeutic potency of Wnt signaling antagonists in the pathogenesis of prostate cancer, current status and perspectives. J Cell Physiol. 2018 ;9(7):245-256. 29. Peng Y, Cao J, Yao XY, et al. TUSC3 induces autophagy in human non-small cell lung cancer cells through Wnt/beta-catenin signaling. Oncotarget. 2017;8(32):52960-52974. 30. Li X, Lu Q, Xie W, Wang Y, Wang G. Anti-tumor effects of triptolide on angiogenesis and cell apoptosis in osteosarcoma cells by inducing autophagy via repressing Wnt/beta-Catenin signaling. Wnt agonist 1 Biochem Biophys Res Commun. 2018;496(2):443-449.