Abstract
Objective(s):
To analyze the potential influences of propofol on the oxidative stress of H2O2-induced human esophageal squamous cell carcinoma (ESCC) Eca109 cell through mediating the Wnt/β-catenin signaling pathway.
Materials and Methods:
Eca109 cells were classified into 5 groups: Control group, H2O2 group, Propofol + H2O2 group, Dkk1 (Dickkopf-1, Wnt/β-catenin pathway antagonist) + H2O2 group, and Propofol + LiCl (Lithium chloride, Wnt/β-catenin pathway agonist) + H2O2 group. Western blotting was performed to determine the protein expressions, flow cytometry to measure the content of ROS, immunofluorescence staining to detect the oxidative DNA damage, as well as MTT, AnnexinV-FITC/PI, Wound-healing, and Transwell assays to test the biological characteristics of Eca109 cells.
Results:
H2O2 resulted in the increased nuclear and cytoplasmatic expression of β-catenin, reduced p-GSK3β expression, up-regulated ROS content, and induced oxidative DNA damage in Eca109 cells. Moreover, Eca109 cells treated with H2O2 alone had enhanced cell proliferation and metastasis but decreased cell apoptosis, as compared with those without any treatment; meanwhile, the declined Cyt C, Bax, and cleaved caspase-3, as well as the elevated Bcl-2 were also observed in Eca109 cells in the H2O2 group, which were reversed by Propofol or Dkk1. Moreover, Propofol could inhibit the effect of LiCl on activating the Wnt/β-catenin signaling pathway in H2O2-induced Eca109 cells.
Conclusion:
Propofol elicits protective effects to inhibit H2O2-induced proliferation and metastasis and promote apoptosis of Eca109 cells via blocking the Wnt/β-catenin pathway, offering a possible therapeutic modality for ESCC.
Key Words: Esophageal neoplasms, Hydrogen peroxide, Propofol, Wnt signaling pathway β-catenin
Introduction
Esophageal cancer (EC), arising from the esophagus, ranks as the sixth most fatal cancer around the world, with the rapidly increasing incidence year by year (1). Pathologically, EC can be divided into esophageal adenocarcinoma (EAC) and esophageal squamous cell carcinoma (ESCC), with the latter being the dominant one mainly found in Asian and African regions and the former usually occurring in western countries (2, 3). Although significant progress has been made in EC treatment, the 5-year overall survival rate of ESCC is only about 14% (4). Numerous studies have reported that the accumulation of DNA damage under the condition of long-term oxidative stress would facilitate the aging process and lead to the occurrence of malignancies and degenerative diseases (5, 6). As reported, hydrogen peroxide (H2O2), as an exogenous active oxygen, can diffuse across membranes to form highly reactive radicals with iron ions in cells through Fenton’s reaction, which is often used to construct various oxidative damage models owing to its stability and easy availability (7, 8). Additionally, the degree of oxidative stress was suggested to affect the occurrence and development of ESCC (9, 10). Hence, it is of great importance and urgency to explore the promising therapeutic target to inhibit the oxidative stress in ESCC.
Propofol (2,6-diisopropylphenol) is commonly accepted as an immediately effective intravenous anesthetic agent in clinical use initially introduced to China in the late 1980s, which can induce and maintain anesthesia in a fast and stable manner, as well as rapid recovery from anesthesia (11). In recent years, propofol has been proven to have various non-anesthetic effects in addition to its multiple advantages as an anesthetic drug (12). For example, propofol can also exert the potential anti-tumor characteristics to affect the proliferation and metastasis of human cancer cells (13, 14). It also has been suggested to have immunomodulatory, antioxidant, analgesic, antiemetic, and neuroprotective properties through its clinical practice(12), highlighting a better option during cancer surgeries when compared to other anesthetic agents (13). Mounting studies have demonstrated the great influence of propofol on cancer development through mediating signaling pathways. For instance, propofol could restrict the proliferation and invasion of HepG2 cells by blocking the Wnt/β-catenin pathway, but facilitate the cell apoptosis, as indicated by Ou et al (15). To our knowledge, Wnt signaling is important during organ development and tissue homeostasis involved in the pathogenesis of several diseases, including cancers (16, 17), which can be broadly diversified into canonical or noncanonical pathways (18). The canonical pathway could regulate the stability of transcription co-activator β-catenin, thus involving in human malignancies (19, 20). An increasing amount of evidence demonstrated Wnt/β-catenin was associated with cancer cell proliferation, migration, invasion, and resistance to drugs, suggesting its potential role as a novel cancer therapeutic target (21, 22). Worth mentioning, there was reported linkage between the Wnt/β-catenin pathway and the carcinogenesis of ESCC (23, 24), which was also related to DNA damage response and oxidative stress (25). Nevertheless, whether propofol can regulate the occurrence and progression of ESCC by mediating the Wnt/β-catenin pathway remains unclear.
As such, we induced the human ESCC cell line Eca109 by H2O2 and then observed the pre-treatment effect of propofol, dickkopf-1 (Dkk1, wnt/β-catenin pathway antagonist), and lithium chloride (LiCl, Wnt/β-catenin pathway agonist) on the oxidative DNA damage and other biological characteristics of Eca109 cells.
Materials and Methods
Treatment and grouping of Eca109 cells
Human ESCC cell line Eca109 (American Type Culture Collection, ATCC, Manassas, VA, USA) were cultured in the RPMI-1640 medium (BioWhittaker and Cambrex, Verviers, Belgium) containing 10% inactivated fetal bovine serum (FBS, Gibco BRL, Grand Island, NY, USA), penicillin (100 units/ml, Sigma, St-Louis, MO, USA), and streptomycin (100 mg/ml, Sigma, St-Louis, MO, USA), and incubated in 5% CO2 at 37 °C. Eca109 cells were classified into Control group (cells without any treatment), H2O2 group (cells treated by 10 μM H2O2 for 3 hr), Propofol + H2O2 group (cells treated with 5 μg/l propofol for 30 min before 3 hr of treatment with 10 μM H2O2), Dkk1 + H2O2 group (cells treated with 20 mmol/l Dkk1 for 30 min before 3 hr of treatment with 10 μM H2O2), and Propofol + LiCl + H2O2 group (cells treated with 5 μg/l propofol and 20 mmol/l LiCl for 30 min before 3 hr of treatment with 10 μM H2O2).
Western blotting
A commercial protein isolation kit (TransGenBiotec, Beijing, China) was used to prepare the protein form cytoplasmic and nuclear fractions, and the concentration was determined according to the bicinchoninic acid (BCA) Kit (Thermo Scientific, IL, USA). The proteins were heated with the addition of the loading buffer for 10 min at 95 °C. Electrophoresis was applied for the separation of proteins (50 μg/well) in sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) gels (Bio-Rad Laboratories, Hercules, California, USA). The resolved proteins were transferred to polyvinylidene difluoride (PVDF) membrane at 120 V for 3 hr, which was placed in 5% bovine serum albumin (BSA) for 1 hr at room temperature, followed by the addition of primary antibodies (Abcam, Cambridge, CA, USA) for overnight incubation at 4 °C, including anti-β-Catenin (ab16051, 0.25 µg/ml dilution), anti-phosphor-GSK3β (ab75745, 1/500 dilution), anti-GSK3β (ab32391, 1/10000 dilution), anti-Cytochrome C (ab13575, 1 µg/ml dilution), anti-Bax (ab32503, 1/2000 dilution), anti-active Caspase-3 (ab2302, 1 µg/ml dilution), anti-Bcl-2 (ab32124, 1/1000 dilution), anti-lamin B1 (ab16048, 1/1000 dilution), and anti-β-actin (ab8226, 1 µg/ml dilution). After the membrane was washed with Tris-buffered saline with Tween 20 (TBST, 3 times/5 min), the corresponding secondary antibodies (Abcam, Cambridge, CA, USA) were added for incubation for 1 hr, followed by another rinse with TBST (3 times/5 min). The development was based on the chemiluminescent (CL) detection. The β-actin and lamin B1 served as cytoplasmic and nuclear protein loading controls, respectively. The experiment was repeated three times to validate the results.
Measurement of ROS content with flow cytometry
When cell confluence reached about 80%, cells were digested using 0.2% collagenase and separately inoculated into the 60 mm2 culture dishes. When the cell confluence was about 70–80%, they were washed with PBS twice, digested by Trypsin for 2 min, mixed with an appropriate volume of culture medium, and transferred to corresponding Eppendorf (EP) tubes for centrifugation (5 min at 800 g). Then, the supernatant was removed followed by the addition of ROS detection solution (500 μl). At last, cells were re-suspended and left for 30 min of reaction at 37 °C in a dark environment, followed by detection in a flowcytometer (Beckman-Coulter, CA, USA). The experiment was done independently three times to test reproducibility.
Immunofluorescence assay
Eca109 cells were incubated in the medium containing 10% normal goat serum at room temperature for 20 min, which were then incubated with anti-8-oxoguanineat 4 °C overnight and the secondary antibody conjugated with tetramethylrhodamine isothiocyanate (TRITC) for 45 min at room temperature. After that, slides were mounted for analysis by Vectashield with 4’-6-diamidino-2-phenylindole (DAPI) (Invitrogen, Molecular Probes, Eugene, Oregon, USA) and placed under a fluorescence microscope for observation and photo-taking. The image analysis was performed by using Image J software (National Institutes of Health, USA), and at least 1000 cells were measured each time. For better visualization, the red fluorescence was converted into pseudo green. As regards fluorescein-labeled avidin (FITC-avidin), the 8-Oxoguanine antibody was replaced by FITC-conjugated avidin (1:200, Santa Cruz), and cells were incubated in the dark environment at 37 °C for another 1 hr. The experiment was repeated three times.
MTT assay
Eca109 cells at the logarithmic growth phase were collected and inoculated into a 96-well plate (5×103 cells/well), which were cultured at 37 °C with 5% CO2 for various time periods (12 hr, 24 hr, 48 hr, and 72 hr). Next, the culture plate was taken out from the incubator followed by the addition of 3-(4,5 Dimethylthiazol -2-1)-2-5-diphenyl tetrazolium bromide (MTT, 50 μl) for another incubation (4 hr). After that, the cells were centrifuged for 10 min at the rate of 2000 rpm, and dimethyl sulfoxide (DMSO, 200 µl) solution was added for 10 min incubation. At last, the optical density (OD) values at 490 nm were detected by a microplate reader (Molecular Devices Corp., Sunnyvale, CA, USA), which was regarded as the indicator for the analysis of cell proliferation. The MTT assay was repeated three times with similar results.
AnnexinV-FITC/PI double staining
Eca109 cells were digested with trypsin, centrifuged, and made into single cell suspension (1×106 cells/ml) by using the buffer solution (calcium-containing PBS). Next, the cell suspension (100 μl) was transferred into a tube, followed by the addition of 5 μl AnnexinV-FITC and 5 μl propidium iodide (PI) for incubation at 4 °C for 30 min. Then, flow cytometry (Beckman-Coulter, CA, USA) was immediately used to detect cell apoptosis after the addition of a binding buffer (400 μl), followed by data analysis using Cell Quest software (Becton Dickinson, CA, USA). This experiment was repeated independently in triple.
Wound-healing assay
Eca109 cells were placed onto the 6-well plate for incubation at 37 °C. When cells covered the bottom of the plate, the scratch lines across the center of the well were drawn by using a sterilized P200 tip, which were photographed and marked as 0 hr under an inverted microscope (Olympus America, Melville, New York, NY, USA). After incubation for 24 hr at 37 °C, cell culture fluid was removed followed by rinsing three times in PBS. Next, serum-free medium was added onto the plate, which was photographed (24 hr). The distance was measured by the Image J software (Scion Corp., MD, USA) and the migration rate was calculated. The assay was repeated in triple to obtain the mean value.
Transwell assay
The upper and lower Transwell chambers (Costar, Boddenheim, Germany) were added with pre-warmed medium at 37 °C for 2 hr. Then, 60–80 μl matrigel was added into each chamber for incubation at 37 °C for 30 min until solidification of matrigel. Then, the complete medium (0.5 ml) was added onto the plate followed by the addition of cell suspension (0.5 ml, 5×l04 cells/ml) into the upper chamber for incubation at 37 °C for 24 hr. Then, the cells on the surface were removed, and the plate was rinsed with PBS. Next, after fixing with cold methanol for 30 min, the cells migrating to the lower Transwell chamber were stained with 0.1% crystal violet solution for 10 min, and the number of cells was observed under an Olympus inverted microscope (Melville, New York, NY, USA) by using six visual fields. The procedure of Transwell invasion assay was repeated three times.
Statistical method
The SPSS software (version 22.0, Chicago, IL, USA) was used to statistically analyze the data in this study. Measurement data were exhibited by mean ± standard deviation (mean±SD). The differences between the two groups were evaluated by using the Student’s t-test. The comparison among more than two independent groups was performed by one-way analysis of variance (ANOVA) with a Tukey’s post hoc test. P<0.05 indicated the significant difference.
Results
Propofol blocked the Wnt/β-catenin pathway in H 2 O 2 -induced Eca109 cells
According to the results of Western blotting demonstrated in Figure 1, H2O2 significantly up-regulated the nuclear and cytoplasmatic expressions of β-catenin, but down-regulated the p-GSK3β expression (all P<0.05). However, propofol could apparently restore β-catenin nuclear accumulation, but elevate p-GSK3β in H2O2-induced Eca109 cells, which was similar to Dkk1 (all P<0.05). Although the nuclear and cytoplasmatic expressions of β-catenin were lower and p-GSK3β was higher in the Dkk1 + H2O2 group (all P<0.05), there was no difference from the propofol + H2O2 group (all P>0.05). In addition, propofol can effectively inhibit the activation of the Wnt/β-catenin pathway caused by LiCl in the H2O2-induced Eca109 cells (P<0.05). Moreover, there was no statistical difference among any groups of cells regarding the expression of total β-catenin and total GSK3β (all P>0.05).
Figure 1.
Effect of propofol on the Wnt/β-catenin signaling pathway in hydrogen peroxide-induced Eca109 cells
Note: A-B, The expression of GSK3β in Eca109 cells detected by Western blotting. C-D, Western blotting analysis determined the nuclear and cytoplasmic expression of β-catenin in Eca109 cells. * P<0.05 compared with the Control group; # P<0.05 compared with H2O2 and the Propofol + LiCl + H2O2 groups
Propofol alleviated oxidative stress and DNA damage in H 2 O 2 -induced Eca109 cells via inhibition of Wnt/β-catenin pathway
H2O2 resulted in increase in the ROS content (all P<0.05), whereas propofol and Dkk1 could effectively inhibit the oxidative stress induced by H2O2 (P<0.05). Besides, in terms of the ROS content in Eca109 cells, the H2O2 group showed no significant difference with the propofol + LiCl + H2O2 group (all P>0.05, Figure 2). Moreover, H2O2 presented an obvious oxidative DNA damage in Eca109 cells when compared with the Control group (all P<0.05). However, propofol and Dkk1 remarkably alleviated the oxidative DNA damage in H2O2-induced Eca109 cells (all P<0.05). Furthermore, there was no statistical difference between the effects of propofol and Dkk1 on oxidative DNA damage in H2O2 - induced Eca109 cells (all P>0.05, Figure 3).
Figure 2.
Effect of propofol-mediated Wnt/β-catenin pathway on reactive oxygen species (ROS) content in hydrogen peroxide-induced Eca109 cells
Note: * P<0.05 compared with the Control group; # P<0.05 compared with H2O2 and the Propofol + LiCl + H2O2 groups
Figure 3.
Effect of propofol-mediated Wnt/β-catenin pathway on the oxidative DNA damage of hydrogen peroxide-induced Eca109 cells
Note: * P<0.05 compared with the Control group; # P<0.05 compared with H2O2 and Propofol + LiCl + H2O2 groups
Effect of propofol-mediated Wnt/β-catenin pathway on the proliferation and apoptosis of H 2 O 2 -induced Eca109 cells
The H2O2 group had increased cell proliferation (24, 48, and 72 hr) and decreased cell apoptosis as compared with the Control group (all P<0.05). After treated with propofol or Dkk1, the proliferation was inhibited while the apoptosis was enhanced in H2O2-induced Eca109 cells (both P<0.05). Meanwhile, Eca109 cells in the propofol + LiCl + H2O2 group did not show any observable difference from cell proliferation and apoptosis in the H2O2 group (all P>0.05, Figures 4A-C). Additionally, we found that H2O2 could effectively suppress the expressions of cytochrome c (Cyt C), Bax, and cleaved caspase-3, but promote the Bcl-2 expression in Eca109 cells (all P<0.05), which were reversed by propofol or Dkk1 (all P<0.05). Moreover, H2O2 and propofol + LiCl + H2O2 groups had no observable differences concerning the expressions of mitochondrial apoptosis-related proteins (all P>0.05, Figures 4 D-E).
Figure 4.
Effect of propofol-mediated Wnt/β-catenin pathway on the proliferation and apoptosis of hydrogen peroxide-induced Eca109 cells
Note: A, Cell proliferation in each group detected by MTT assay; B-C, Cell apoptosis in each group measured by AnnexinV-FITC/PI double staining; D-E, The expression of mitochondrial apoptosis-related proteins (including Cyt C, Bax, cleaved caspase-3, and Bcl-2) in each group determined by Western blotting; * P<0.05 compared with the Control group; # P<0.05 compared with H2O2 and the Propofol + LiCl + H2O2 groups
Propofol inhibited the migration and invasion of H 2 O 2 -induced Eca109 cells through mediating the Wnt/β-catenin pathway
As shown in Figure 5, it is obvious that H2O2 could induce the migration and invasion of Eca109 cells in comparison with Controls (all P<0.05). Besides, both propofol and Dkk1 markedly reduced the metastasis of H2O2-induced Eca109 cells (all P<0.05), and Dkk1 slightly but not statistically decreased the migration and invasion of H2O2-induced Eca109 cells when compared with propofol (all P>0.05). In addition, Propofol + LiCl + H2O2 and H2O2 groups were not statistically different from each other regarding the cell abilities of migration and invasion (all P>0.05).
Figure 5.
Effect of propofol mediating Wnt/β-catenin pathway on the migration and invasion of hydrogen peroxide-induced Eca109 cells
Note: A, C: Cell migration ability in each group evaluated by Wound-healing assay; B, D: Cell invasion ability in each group measured by Transwell assay; * P<0.05 compared with the Control group; # P<0.05 compared with H2O2 and the Propofol + LiCl + H2O2 groups
Discussion
Cells may produce numerous ROS from essential metabolic processes to participate in regulating various physiological and pathological processes, like superoxide anion radical (O2-), hydroxyl radical (·OH), and H2O2 (26, 27). The increased oxidative stress could induce a broad spectrum of damage to biological systems, ultimately leading to different types of diseases, such as cardiovascular diseases, neurodegenerative diseases, and tumors (28). Therefore, better understanding the regulatory mechanism of anti-oxidative stress may contribute to the improvement of the therapeutic effect of ESCC. H2O2, an important relatively stable non-radical ROS, is usually used in the investigation of oxidative stress, since it can diffuse freely to the nucleus to induce DNA base damage and strand breakage (29, 30). As reported, ROS has pro-cancer activities, including promoting cell proliferation, angiogenesis, and metastases, as well as suppressing apoptosis (31). Researchers also found H2O2 could sequentially activate the ERK2 MAPK, NF-kappaB1 p50, and NOX5-S, and enhance the production of ROS, thus increasing the proliferation of EAC cells (32). Besides, the H2O2 -induced migration and invasion could be reversed using curcumin in pancreatic cancer cells by blocking the ROS/ERK/NF-kappaB signaling pathway (33). From our models, we found that H2O2 significantly increased the ROS content and caused oxidative DNA damage of Eca109 cells with elevated cell proliferation and metastasis, as well as decreased cell apoptosis, showing the successful construction of oxidative cell damage models in our research.
Propofol has been widely used clinically for anesthesia and sedation due to its safe and excellent anesthetic effects (34). A large number of studies found the antioxidant properties of propofol. For example, Romuk et al. has confirmed the neuroprotective effects of propofol in the brain by controlling oxidative stress (35). A study by Romuk revealed that the antioxidative protection of propofol in PC12 cells was Ca2+-dependent and was mediated by NADPH oxidase (36). Worth mentioning, our study also revealed that propofol could remarkably alleviate the oxidative stress and DNA damage in Eca109 cells induced by H2O2. Accumulating evidence has pointed out the influence of propofol on the activity of tumor cells. As reported by researchers, propofol could inhibit the transcriptional activity of androgen receptors, as well as cell proliferation, in human prostate cancer cells (37). Ecimovic and his colleague also revealed that propofol could inhibit the breast cancer cell migration, but was reversed by down-regulation of NET1 (38). In our study, propofol could also dramatically suppress the proliferation and metastasis of H2O2-induced Eca109 cells, effectively promote cell apoptosis. Consistently, Ou et al. also demonstrated a similar trend of propofol in Eca109 cells (15). There was a possible explanation that propofol can reduce the production of ROS by the NADPH oxidase enzymatic system, thus act as a pro-apoptotic, anti-proliferative, and anti-metastasis stimulus in ESCC.
Notably, the Wnt/β-catenin pathway has attracted increasing attention from scholars in recent reports concerning the aspect of oxidative stress. Not surprisingly, H2O2 in our study could activate the Wnt/β-catenin pathway in Eca109 cells. As we know, oxidative stress generated by treatment of cells with H2O2 can activate the expressions of Foxo transcription factors (like Foxo1, Foxo3a, and Foxo4), which may interact with β-catenin to inhibit the apoptosis of cancer cells (39). Besides, the Wnt pathway is believed to be one of the major targets of p53 and p53-β-catenin pathways, playing an important role in the regulation of DNA damage and cell apoptosis (40, 41). In addition, it is well documented that the Bcl-2 family members (the arbiters of mitochondrial apoptotic pathway), derived from BH3 domains, which can process with the pro-apoptotic Bcl-2 members (like Bax and Bak) to release Cyt C (42, 43), thereby agglomerating procaspase-9 in cytoplasm and activating downstream Caspase-3, eventually inducing cell apoptosis (44, 45). Actually, our study also observed the promoted proliferation and inhibited apoptosis of Eca109 cells treated by H2O2 with the decreased expression of Cyt C, Bax, and cleaved caspase-3, and the increased expression of Bax. Moreover, consistent with the findings of a previous study (33), H2O2 could facilitate the abilities of migration and invasion of Eca109 cells in our study, which indicated the possibility that H2O2 could activate the Wnt/β-catenin signaling pathway and promote the epithelial-mesenchymal transition (EMT) (46), thereby promoting metastasis of EC cells. Moreover, propofol was found to block the Wnt/β-catenin signaling pathway in H2O2-induced Eca109 cells, which was consistent with the study reported previously (15). Coincidently, another study demonstrated that propofol could inhibit the Wnt/β-catenin pathway and reduce β-catenin expression in metastatic tumor tissues, eventually blocking the pulmonary metastasis of MADB106 tumor cells in tumor-bearing rats (47). Last but not least, propofol in our study was also shown to inhibit the effect of LiCl on activating the Wnt/β-catenin signaling pathway in H2O2-induced Eca109 cells, which suggested that propofol may play a protective role in H2O2-induced ESCC cells by blocking Wnt/β-catenin signaling pathway.
Conclusion
Collectively, our study may suggest that propofol exerts a protective role in H2O2-induced Eca109 cells possibly by blocking the Wnt/β-catenin signaling pathway.
Acknowledgment
The authors appreciate the reviewers for their suggestions in this work.
Conflicts of Interest
The authors declare that there are no conflicts of interest.
References
- 1.Zhang HF, Wu C, Alshareef A, Gupta N, Zhao Q, Xu XE, et al. The PI3K/AKT/c-MYC axis promotes the acquisition of cancer stem-like features in esophageal squamous cell carcinoma. Stem Cells. 2016;34:2040–2051. doi: 10.1002/stem.2395. [DOI] [PubMed] [Google Scholar]
- 2.Agrawal N, Jiao Y, Bettegowda C, Hutfless SM, Wang Y, David S, et al. Comparative genomic analysis of esophageal adenocarcinoma and squamous cell carcinoma. Cancer Discov. 2012;2:899–905. doi: 10.1158/2159-8290.CD-12-0189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Liang Y, Liu JL, Wu Y, Zhang ZY, Wu R. Cyclooxygenase-2 polymorphisms and susceptibility to esophageal cancer: a meta-analysis. Tohoku J Exp Med. 2011;223:137–144. doi: 10.1620/tjem.223.137. [DOI] [PubMed] [Google Scholar]
- 4.Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. Ca A Cancer Journal for Clinicians. 2011;61:69. doi: 10.3322/caac.20107. [DOI] [PubMed] [Google Scholar]
- 5.Fimognari C. Role of oxidative RNA damage in chronic-degenerative diseases. Oxid Med Cell Longev. 2015;2015:358713. doi: 10.1155/2015/358713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bobrovnikova-Marjon E, Grigoriadou C, Pytel D, Zhang F, Ye J, Koumenis C, et al. PERK promotes cancer cell proliferation and tumor growth by limiting oxidative DNA damage. Oncogene. 2010;29:3881–3895. doi: 10.1038/onc.2010.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Garban HJ, Bonavida B. Nitric oxide disrupts H2O2-dependent activation of nuclear factor kappa B Role in sensitization of human tumor cells to tumor necrosis factor-alpha -induced cytotoxicity. J Biol Chem. 2001;276:8918–8923. doi: 10.1074/jbc.M008471200. [DOI] [PubMed] [Google Scholar]
- 8.De Marinis E, Fiocchetti M, Acconcia F, Ascenzi P, Marino M. Neuroglobin upregulation induced by 17beta-estradiol sequesters cytocrome c in the mitochondria preventing H2O2-induced apoptosis of neuroblastoma cells. Cell Death Dis. 2013;4:e508. doi: 10.1038/cddis.2013.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sehitogullari A, Aslan M, Sayir F, Kahraman A, Demir H. Serum paraoxonase-1 enzyme activities and oxidative stress levels in patients with esophageal squamous cell carcinoma. Redox Rep. 2014;19:199–205. doi: 10.1179/1351000214Y.0000000091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zhang X, Lan L, Niu L, Lu J, Li C, Guo M, et al. Oxidative stress regulates cellular bioenergetics in esophageal squamous cell carcinoma cell. Biosci Rep. 2017:37. doi: 10.1042/BSR20171006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zhang L, Wang N, Zhou S, Ye W, Jing G, Zhang M. Propofol induces proliferation and invasion of gallbladder cancer cells through activation of Nrf2. J Exp Clin Cancer Res. 2012;31:66. doi: 10.1186/1756-9966-31-66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Vasileiou I, Xanthos T, Koudouna E, Perrea D, Klonaris C, Katsargyris A, et al. Propofol: a review of its non-anaesthetic effects. Eur J Pharmacol. 2009;605:1–8. doi: 10.1016/j.ejphar.2009.01.007. [DOI] [PubMed] [Google Scholar]
- 13.Yang N, Liang Y, Yang P, Yang T, Jiang L. Propofol inhibits lung cancer cell viability and induces cell apoptosis by upregulating microRNA-486 expression. Braz J Med Biol Res. 2017;50:e5794. doi: 10.1590/1414-431X20165794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Huang X, Teng Y, Yang H, Ma J. Propofol inhibits invasion and growth of ovarian cancer cells via regulating miR-9/NF-kappaB signal. Braz J Med Biol Res. 2016;49:e5717. doi: 10.1590/1414-431X20165717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ou W, Lv J, Zou X, Yao Y, Wu J, Yang J, et al. Propofol inhibits hepatocellular carcinoma growth and invasion through the HMGA2-mediated Wnt/beta-catenin pathway. Exp Ther Med. 2017;13:2501–2506. doi: 10.3892/etm.2017.4253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lim X, Nusse R. Wnt signaling in skin development, homeostasis, and disease. Cold Spring Harb Perspect Biol. 2013:5. doi: 10.1101/cshperspect.a008029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Polakis P. Wnt signaling and cancer. Genes Dev. 2000;14:1837–1851. [PubMed] [Google Scholar]
- 18.Grumolato L, Liu G, Mong P, Mudbhary R, Biswas R, Arroyave R, et al. Canonical and noncanonical Wnts use a common mechanism to activate completely unrelated coreceptors. Genes Dev. 2010;24:2517–2530. doi: 10.1101/gad.1957710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Mohammed MK, Shao C, Wang J, Wei Q, Wang X, Collier Z, et al. Wnt/beta-catenin signaling plays an ever-expanding role in stem cell self-renewal, tumorigenesis and cancer chemoresistance. Genes Dis. 2016;3:11–40. doi: 10.1016/j.gendis.2015.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hoffmeyer K, Raggioli A, Rudloff S, Anton R, Hierholzer A, Del Valle I, et al. Wnt/beta-catenin signaling regulates telomerase in stem cells and cancer cells. Science. 2012;336:1549–1554. doi: 10.1126/science.1218370. [DOI] [PubMed] [Google Scholar]
- 21.Khan NI, Bradstock KF, Bendall LJ. Activation of Wnt/beta-catenin pathway mediates growth and survival in B-cell progenitor acute lymphoblastic leukaemia. Br J Haematol. 2007;138:338–348. doi: 10.1111/j.1365-2141.2007.06667.x. [DOI] [PubMed] [Google Scholar]
- 22.Martins-Neves SR, Paiva-Oliveira DI, Wijers-Koster PM, Abrunhosa AJ, Fontes-Ribeiro C, Bovee JV, et al. Chemotherapy induces stemness in osteosarcoma cells through activation of Wnt/beta-catenin signaling. Cancer Lett. 2016;370:286–295. doi: 10.1016/j.canlet.2015.11.013. [DOI] [PubMed] [Google Scholar]
- 23.Li Y, Zhong D, Lei L, Jia Y, Zhou H, Yang B. Propofol Prevents Renal Ischemia-Reperfusion Injury via Inhibiting the Oxidative Stress Pathways. Cell Physiol Biochem. 2015;37:14–26. doi: 10.1159/000430329. [DOI] [PubMed] [Google Scholar]
- 24.Ueda T, Tsubamoto H, Inoue K, Sakata K, Shibahara H, Sonoda T. Itraconazole modulates hedgehog, WNT/beta-catenin, as well as Akt Signalling, and inhibits proliferation of cervical cancer cells. Anticancer Res. 2017;37:3521–3526. doi: 10.21873/anticanres.11720. [DOI] [PubMed] [Google Scholar]
- 25.Karimaian A, Majidinia M, Bannazadeh Baghi H, Yousefi B. The crosstalk between Wnt/beta-catenin signaling pathway with DNA damage response and oxidative stress: Implications in cancer therapy. DNA Repair (Amst) 2017;51:14–19. doi: 10.1016/j.dnarep.2017.01.003. [DOI] [PubMed] [Google Scholar]
- 26.Chang CL, Lin CS. Phytochemical Composition, Antioxidant Activity, and Neuroprotective Effect of Terminalia chebula Retzius Extracts. Evid Based Complement Alternat Med. 2012;2012:125247. doi: 10.1155/2012/125247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Vallejo MJ, Salazar L, Grijalva M. Oxidative Stress Modulation and ROS-Mediated Toxicity in Cancer: A Review on In Vitro Models for Plant-Derived Compounds. Oxid Med Cell Longev. 2017;2017:4586068. doi: 10.1155/2017/4586068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Umeno A, Biju V, Yoshida Y. In vivo ROS production and use of oxidative stress-derived biomarkers to detect the onset of diseases such as Alzheimer’s disease, Parkinson’s disease, and diabetes. Free Radic Res. 2017;51:413–427. doi: 10.1080/10715762.2017.1315114. [DOI] [PubMed] [Google Scholar]
- 29.Cho MG, Ahn JH, Choi HS, Lee JH. DNA double-strand breaks and Aurora B mislocalization induced by exposure of early mitotic cells to H2O2 appear to increase chromatin bridges and resultant cytokinesis failure. Free Radic Biol Med. 2017;108:129–145. doi: 10.1016/j.freeradbiomed.2017.03.025. [DOI] [PubMed] [Google Scholar]
- 30.Yang L, Mu Y, Cui H, Liang Y, Su X. MiR-9-3p augments apoptosis induced by H2O2 through down regulation of Herpud1 in glioma. PLoS One. 2017;12:e0174839. doi: 10.1371/journal.pone.0174839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Stojnev S, Ristic-Petrovic A, Jankovic-Velickovic L. Reactive oxygen species, apoptosis and cancer. Vojnosanit Pregl. 2013;70:675–678. doi: 10.2298/vsp1307675s. [DOI] [PubMed] [Google Scholar]
- 32.Zhou X, Li D, Resnick MB, Behar J, Wands J, Cao W. Signaling in H2O2-induced increase in cell proliferation in Barrett’s esophageal adenocarcinoma cells. J Pharmacol Exp Ther. 2011;339:218–227. doi: 10.1124/jpet.111.182352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Cao L, Liu J, Zhang L, Xiao X, Li W. Curcumin inhibits H2O2-induced invasion and migration of human pancreatic cancer via suppression of the ERK/NF-kappaB pathway. Oncol Rep. 2016;36:2245–2251. doi: 10.3892/or.2016.5044. [DOI] [PubMed] [Google Scholar]
- 34.Xu YB, Du QH, Zhang MY, Yun P, He CY. Propofol suppresses proliferation, invasion and angiogenesis by down-regulating ERK-VEGF/MMP-9 signaling in Eca-109 esophageal squamous cell carcinoma cells. Eur Rev Med Pharmacol Sci. 2013;17:2486–2494. [PubMed] [Google Scholar]
- 35.Romuk E, Szczurek W, Nowak P, Skowron M, Prudel B, Hudziec E, et al. Effects of propofol on oxidative stress parameters in selected parts of the brain in a rat model of parkinson disease. Postepy Hig Med Dosw (Online) 2016;70:1441–1450. doi: 10.5604/17322693.1227841. [DOI] [PubMed] [Google Scholar]
- 36.Chen XH, Zhou X, Yang XY, Zhou ZB, Lu DH, Tang Y, et al. Propofol protects against H2O2-induced oxidative injury in differentiated PC12 cells via inhibition of Ca(2+)-dependent NADPH oxidase. Cell Mol Neurobiol. 2016;36:541–551. doi: 10.1007/s10571-015-0235-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Tatsumi K, Hirotsu A, Daijo H, Matsuyama T, Terada N, Tanaka T. Effect of propofol on androgen receptor activity in prostate cancer cells. Eur J Pharmacol. 2017;809:242–252. doi: 10.1016/j.ejphar.2017.05.046. [DOI] [PubMed] [Google Scholar]
- 38.Ecimovic P, Murray D, Doran P, Buggy DJ. Propofol and bupivacaine in breast cancer cell function in vitro - role of the NET1 gene. Anticancer Res. 2014;34:1321–1331. [PubMed] [Google Scholar]
- 39.Essers MA, de Vries-Smits LM, Barker N, Polderman PE, Burgering BM, Korswagen HC. Functional interaction between beta-catenin and FOXO in oxidative stress signaling. Science. 2005;308:1181–1184. doi: 10.1126/science.1109083. [DOI] [PubMed] [Google Scholar]
- 40.Lee KH, Li M, Michalowski AM, Zhang X, Liao H, Chen L, et al. A genomewide study identifies the Wnt signaling pathway as a major target of p53 in murine embryonic stem cells. Proc Natl Acad Sci U S A. 2010;107:69–74. doi: 10.1073/pnas.0909734107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Zhang DY, Wang HJ, Tan YZ. Wnt/beta-catenin signaling induces the aging of mesenchymal stem cells through the DNA damage response and the p53/p21 pathway. PLoS One. 2011;6:e21397. doi: 10.1371/journal.pone.0021397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Certo M, Del Gaizo Moore V, Nishino M, Wei G, Korsmeyer S, Armstrong SA, et al. Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2 family members. Cancer Cell. 2006;9:351–365. doi: 10.1016/j.ccr.2006.03.027. [DOI] [PubMed] [Google Scholar]
- 43.Peixoto PM, Ryu SY, Kinnally KW. Mitochondrial ion channels as therapeutic targets. FEBS Lett. 2010;584:2142–2152. doi: 10.1016/j.febslet.2010.02.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Honarpour N, Tabuchi K, Stark JM, Hammer RE, Sudhof TC, Parada LF, et al. Embryonic neuronal death due to neurotrophin and neurotransmitter deprivation occurs independent of Apaf-1. Neuroscience. 2001;106:263–274. doi: 10.1016/s0306-4522(01)00275-5. [DOI] [PubMed] [Google Scholar]
- 45.Allan LA, Clarke PR. Apoptosis and autophagy: Regulation of caspase-9 by phosphorylation. FEBS J. 2009;276:6063–6073. doi: 10.1111/j.1742-4658.2009.07330.x. [DOI] [PubMed] [Google Scholar]
- 46.He J, Zhou M, Chen X, Yue D, Yang L, Qin G, et al. Inhibition of SALL4 reduces tumorigenicity involving epithelial-mesenchymal transition via Wnt/beta-catenin pathway in esophageal squamous cell carcinoma. J Exp Clin Cancer Res. 2016;35 doi: 10.1186/s13046-016-0378-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wang W, Lin C, Zhang Y, Chen Y, Guo P. [Effects of propofol on pulmonary metastasis of intravenously injected MADB106 tumor cells and expression of E-cadherin and beta-catenin in rats] Nan Fang Yi Ke Da Xue Xue Bao. 2015;35:852–856. [PubMed] [Google Scholar]





