Open Access

Pizotifen inhibits the proliferation and invasion of gastric cancer cells

  • Authors:
    • Ying Jiang
    • Wei Wang
    • Xi Wu
    • Jihua Shi
  • View Affiliations

  • Published online on: December 9, 2019     https://doi.org/10.3892/etm.2019.8308
  • Pages: 817-824
  • Copyright: © Jiang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer is the fifth most common malignancy and the third highest cause of cancer‑associated mortality worldwide. Therefore, research on the pathogenesis of gastric cancer is of utmost importance. It has been reported that aberrant activation of the Wnt/β‑catenin signaling pathway is involved in the occurrence and development of gastric cancer. In the present study, it was found that pizotifen could inhibit the viability of gastric cancer cell lines MNK45 and AGS cells in a dose‑dependent manner. Pizotifen treatment suppressed cell migration and invasion in MNK45 and AGS cells, whilst also inducing apoptosis. Western blot analysis demonstrated that pizotifen blocked the expression of Wnt3a, β‑catenin and N‑cadherin, whilst increasing E‑cadherin expression. In addition, BML‑284, a pharmacological Wnt signaling activator, partially reversed the changes in the expression levels of β‑catenin, N‑cadherin and E‑cadherin in MNK45 and AGS cells induced by pizotifen. Collectively, these findings suggested that pizotifen demonstrates potential as a novel anti‑cancer drug for the treatment of gastric cancer by inhibiting the Wnt/β‑catenin pathway.

Introduction

Gastric cancer is currently the third highest cause of cancer mortality among all malignancies worldwide (1). In recent years, the overall incidence and mortality of gastric cancer has been decreasing due to control of risk factors and the development of gastric cancer screening (2). However, the burden of gastric cancer remains critical due to the large population base and an increasingly aging population (3). In China, the incidence of gastric cancer and associated deaths accounts for ~50% of all gastric cancer cases in the world (4). This may be due to the higher proportion of advanced gastric cancer cases upon diagnosis. Therefore, investigations into the pathogenesis of gastric cancer may reveal biomarkers that can be used for early diagnosis and treatment.

The Wnt signaling pathway, named after the wingless and Int-1 genes (5), performs a variety of different functions, including wide involvement in embryonic development, tissue homeostasis and cell carcinogenesis (6). It is typically divided into the canonical and non-canonical signaling pathways, of which the canonical pathway (Wnt/β-catenin) is the more well-known pathway and is highly conserved throughout the evolutionary process (7). Previous studies have demonstrated that the Wnt/β-catenin signaling pathway serves a key role in regulating cell proliferation, maintenance of stem cell characteristics and homeostasis of normal gastric mucosa (810). In particular, it has been reported that activation of the Wnt/β-catenin signaling cascade is found in >30% of gastric cancer cases (11,12). Indeed, evidence is accumulating showing that Wnt3a, one member of the Wnt family, has the ability to either promote or inhibit cancer progression via the canonical Wnt/β-catenin signaling pathway, depending on the type of cancer (13).

β-catenin is a multifunctional protein, which was initially found to bind to E-cadherin-binding protein and participate in cell adhesion (14). E-cadherin is not only a key member of Wnt signaling pathway, but also a critical epithelial marker of epithelial-mesenchymal transition (EMT) (15). EMT refers to the process whereby epithelial cells transform into mesenchymal phenotype cells, resulting in enhanced cell motility and invasiveness. EMT is one of the key initiation events for tumor metastasis and has a crucial role in the invasion and metastasis of cancers, including gastric cancer (1618). Therefore, increasing research has considered EMT as an effective therapeutic target in cancer treatment (19). Research over the past decade has mainly focused on searching for novel anticancer agents and reassessing known drugs that have been used in other diseases to uncover any potential anti-tumor effects (20). One of those, pizotifen, also known as 4-(1-methyl-4-piperidylidine)-9,10-dihydro-4H-benzo-(4,5)-cyclohepta(1,2)-thiophene, is a potent serotonin or 5-hydroxytryptamine (HT)2 receptor antagonist (21). Pizotifen is primarily indicated for the prevention of vascular headaches (22). It has been previously reported that serotonin 5-HT2A receptor antagonists, such as ritanserin, can inhibit cell viability and promote cell apoptosis in colorectal cancer cells (23). However, the effect of pizotifen on cancer growth and metastasis remains unknown.

In the present study, it was found that pizotifen inhibited the viability, migration and invasion of gastric cancer whilst inducing cell apoptosis. The Wnt/β-catenin-epithelial-mesenchymal transition (EMT) signaling pathway may be involved in this process. These results suggested that pizotifen could potentially be a new therapeutic agent for the treatment of gastric cancer and warrants further assessment in future clinical studies.

Materials and methods

Reagents and antibodies

Pizotifen was purchased from MedChemExpress and its molecular structure is shown in Fig. 1A. The primary antibodies against Wnt3a (cat. no. ab28472; dilution, 1:1,000), active-caspase-3 (cat. no. ab2302; dilution, 1:1,000) were purchased from Abcam, and the primary antibodies against Cyclin D-1 (cat. no. 60186-1-lg; dilution, 1:1,000), Bax (cat. no. 50599-2-lg; dilution, 1:1,000), Bcl-2 (cat. no. 12789-1-AP; dilution, 1:1,000) and β-tubulin (cat. no. 10094-1-AP; dilution, 1:1,000) were purchased from ProteinTech Group, Inc. Primary antibodies against β-catenin (cat. no. 9562; dilution, 1:1,000), E-cadherin (cat. no. 14472; dilution, 1:1,000) and N-cadherin (cat. no. 4061; dilution, 1:1,000) were purchased from Cell Signaling Technology, Inc. Both secondary antibodies (horseradish peroxidase conjugated anti-rabbit, cat. no. SA00001-15; anti-mouse, cat. no. SA00001-1; both dilutions, 1:5,000) were purchased from ProteinTech Group, Inc. BML-284, a selective Wnt activator, was purchased from MedChemExpress. Unless specifically indicated, all other reagents were obtained from Sigma-Aldrich (Merck KGaA).

Cell culture and treatment

Human gastric cancer cell lines MNK45 and AGS were obtained from the Type Culture Collection of the Chinese Academy of Sciences. The cell lines were cultured in DMEM containing 10% FBS (Gibco; Thermo Fisher Scientific, Inc.), penicillin (100 U/ml), and streptomycin (100 mg/ml), and were maintained in a humidified atmosphere (95%) at 37°C under 5% CO2. Cells in the logarithmic growth phase were treated with different concentrations of pizotifen (10, 20 and 40 µM) or BML-284 (10 µM) alone or co-treated with pizotifen (20 µM) and BML-284 (10 µM). The cells were treated for varying time periods, including 0, 24, 48 and 72 h, 0.1% DMSO was used as negative control.

Cell viability assay

The Cell Counting Kit-8 (CCK-8) assay was performed according to the manufacturer's protocol (Beijing Solarbio Science & Technology Co., Ltd.). Cells were seeded into 96-well plates at a density of 1×103 cells/well and cultured overnight to allow for adherence. Cultures were subsequently treated with different concentrations of pizotifen (10, 20 and 40 µM). DMSO (0.1%) was used as the negative control. The cells were then cultured for 3 days and measurements were obtained every 24 h. When the cells were measured, CCK-8 solution (10 µl/well) was added to each well and incubated for an additional 1.5 h at 37°C. The final optical density was measured at a wavelength of 450 nm to estimate cell viability in the different treatment groups. Each experiment was performed in triplicate.

Wound healing assay

Cells were first seeded into six-well plates at a density of 5×105 cells/well and incubated overnight at 37°C. The following day, when cells had reached ~95–100%confluency, wounds were generated using pipette tips. After being scratched with the pipettes, the cells were cultured in serum-free medium supplemented with 10 or 20 µM pizotifen or DMSO for 24 h. The wounds were photographed using a light microscope (magnification, ×40) and analyzed using ImageJ software v1.8.0 (National Institutes of Health). The percentage wound closure was calculated using the mathematical equation: Percentage wound closure (%) = (A0-At)/A0 × 100%, where A0 is the initial wound area, and At is the wound area at 0, 24 h post-treatment.

Western blot analysis

Total protein was extracted from the cell cultures according to instructions from radioimmunoprecipitation assay Lysis Buffer (CWBIO Tech). Protein concentration was quantified using an Enhanced Bicinchoninic Acid Protein Assay kit. A total of 20 µg protein from each sample were separated by 10% SDS-PAGE and then transferred to PVDF membranes (Bio-Rad Laboratories, Inc.). The membranes were incubated in blocking buffer [5% non-fat milk dissolved in TBS with Tween-20 (TBST)] at room temperature for 1-h and then probed at 4°C overnight with primary antibodies (dilution, 1:1,000). The membranes were subsequently rinsed with TBST and incubated with horseradish peroxidase-conjugated secondary antibodies (1:5,000) for 1-h at room temperature. The blots were visualized using the enhanced chemiluminescence detection kit (Beyotime Institute of Biotechnology) and analyzed using the Quantity One 1-D software v4.6.7 (Bio-Rad Laboratories, Inc.). Each experiment was repeated at least three times.

Apoptosis assay

Cells (5×105) pretreated with 20 µM pizotifen or 0.1% DMSO were double-stained with fluorescein isothiocyanate-conjugated Annexin V (25 mg/ml) and propidium iodide (PI; 50 mg/ml) at 4°C for 15 min according to the manufacturer's protocol (4A Biotech Co., Ltd.), and then washed and analyzed using a BD FACSCalibur flow cytometry system (BD Biosciences). The obtained data was analyzed using FlowJo software v7.6.1 (FlowJo LLC).

Transwell assays

For the Transwell migration assay, 5×103 cells pretreated with pizotifen or BML-284 were seeded into the upper sections of the non-coated 96-well inserts (pore size, 8 µm; Corning, Inc.). For the invasion assay, Matrigel (BD Biosciences) was thawed at 4°C and diluted in serum-free medium at a ratio of 1:6, then added to the upper chamber of the Transwell chamber and maintained at 37°C for 4–6 h. A total of 1×105 cells were seeded into the top chamber of 24-well inserts with 8 µm pore size and Matrigel-coated membranes. In both assays, cells were diluted in medium without FBS and seeded into the top chamber, whereas medium containing 10% FBS were introduced into the lower chamber in order to establish a chemoattractant gradient. Cells were incubated for 24 h, fixed with 4% paraformaldehyde at room temperature for 30 min, and stained with 0.1% crystal violet solution (Sigma-Aldrich; Merck KGaA) at room temperature for 20 min. Then, 5 microscopic fields were used to count and photograph the stained cells under a light microscope (magnification, ×100).

Statistical analysis

Experimental data are presented as the mean ± SD. All statistical analysis was performed using SPSS 13.5 (SPSS, Inc.). Student's t-test and one-way ANOVA followed by Tukey's posthoc analysis were performed. P<0.05 was considered to indicate a statistically significant difference.

Results

Pizotifen inhibits the viability of gastric cancer MNK45 and AGS cells

A CCK8 assay was carried out to determine the effect of pizotifen on the viability of gastric cancer cell lines MNK45 and AGS. As shown in Fig. 1B, pizotifen treatment of 48h or 72 h significantly inhibited the viability of MNK45 cells in a dose-dependent manner (P<0.05). Similarly, pizotifen also inhibited the viability of AGS cells (Fig. 1C).

Pizotifen inhibits MNK45 and AGS cells migration and invasion

Wound-healing and Transwell assays were performed to investigate whether pizotifen affects the migratory and invasive properties of gastric cancer cells. Since 10 and 20 µM pizotifen both caused appropriate inhibitory effect on MNK45 and AGS cells, whilst 40 µM pizotifen caused increased cell death, 10 and 20 µM of pizotifen were used in the rest experiments. As shown in Fig. 2A, 10 and 20 µM pizotifen both led to a significant decrease in the migratory abilities of MNK45 cells in a dose-dependent manner (P<0.05). A similar inhibitory effect of pizotifen was observed in AGS cells (P<0.05; Fig. 2B). Moreover, the results of the Transwell assay further suggested that the migratory abilities of MNK45 and AGS cells were significantly inhibited following treatment with pizotifen (P<0.05; Fig. 3A-C). In addition, the invasive abilities of MNK45 and AGS cells treated with pizotifen were significantly reduced (P<0.05; Fig. 3D-F). Furthermore, the data showed that BML-284, a selective Wnt signaling activator that has been revealed to induce β-catenin in vitro (24), significantly increased the migration and invasion of both MNK45 and AGS cells (both P<0.05; Fig. 3), and partially restored the migratory and invasive abilities of MNK45 and AGS cells inhibited by pizotifen (P<0.05; Fig. 3).

Pizotifen induces the mitochondrial-mediated apoptosis of MNK45 and AGS cells

Annexin V-FITC/PI assay was performed to investigate whether pizotifen inhibited the growth of gastric cancer cells by triggering apoptosis. As shown in Fig. 4A, pizotifen (20 µM) significantly enhanced cell apoptosis in both MNK45 and AGS cells (both P<0.01) compared with the NC group (Fig. 4A). To further determine the mechanism of pizotifen-induced apoptosis, western blot analysis was performed to detect the expression of proteins associated with the mitochondrial apoptotic pathway in MNK45 and AGS cells treated with pizotifen for 48 h (Fig. 4B). The data demonstrated that the expression of the anti-apoptotic protein Bcl-2 was significantly reduced in pizotifen-treated cells, whereas the expression of pro-apoptotic proteins Bax and active caspase-3 were significantly increased (P<0.05; Fig. 4B). These data indicated that pizotifen may induce the apoptosis of gastric cancer cells via the intrinsic apoptotic pathway, specifically the Bax/Bcl-2 and caspase cascade.

Pizotifen downregulates the Wnt/β-catenin-EMT signaling pathway in MNK45 and AGS cells

The ability of pizotifen to inhibit the Wnt/β-catenin signaling pathway in MNK and AGS cells was investigated using western blot analysis. Pizotifen treatment significantly reduced the expression of Wnt3a and β-catenin in a dose-dependent manner in MNK45 and AGS cells (P<0.05; Fig. 5A). In addition, the expression of the epithelial marker E-cadherin was significantly upregulated by pizotifen treatment in MNK45 and AGS cells, while the expression of the mesenchymal marker N-cadherin was significantly downregulated (P<0.05; Fig 5A), suggesting that pizotifen regulated the expression of EMT markers proteins in a dose-dependent manner. This suggested that pizotifen can suppress the Wnt/β-catenin-EMT signaling pathway in MNK45 and AGS cells.

Wnt/β-catenin-EMT signaling pathway may play a role in the anti-invasive effect of pizotifen

To further investigate the mechanism of action of pizotifen in the inhibition of gastric cancer cell invasion, BML-284 was used. It was found that the expression of β-catenin was significantly upregulated in MNK45 and AGS cells following BML-284 treatment compared with the NC group (Fig. 5B). In the presence of pizotifen, BML-284 treatment also partially restored β-catenin expression compared with the pizotifen-only group (Fig. 5B). In addition, BML-284 treatment partially reversed the effects induced by pizotifen on E-cadherin and N-cadherin expression in MNK45 and AGS cells compared with the pizotifen-treated group (Fig. 5B). Taken together, the results of the present study indicated that the pizotifen-induced inhibitory effects on gastric cancer cell migration and invasion may be due to the inhibition of the Wnt/β-catenin-EMT signaling pathway.

Discussion

Gastric cancer is one of the most serious malignant tumors; its mortality rate is lower only than that of lung cancer and liver cancer worldwide (1). Therefore, it has become a priority to explore the mechanism of the occurrence and development of gastric cancer in order to develop potential novel anti-cancer agents. In the present study, it was found that pizotifen could inhibit the viability, migration and invasion of MNK45 and AGS cells, as well as inducing apoptosis. Pizotifen is a serotonin antagonist that is primarily indicated for the prevention of recurrent migraine headaches. A number of studies have reported that serotonin can promote the growth of several types of cancer, including breast (25), prostate (26), colorectal (27), hepatocellular (28) and small-cell lung cancer (29). In addition, it has been reported that serotonin is involved in cancer cell migration, metastasis and angiogenesis (30). In some cases, antagonists of serotonin receptors have been successfully used to inhibit cancer cell growth (31,32). The present study is the first to report the anti-tumor properties of pizotifen against gastric cancer cells. However, the effects of pizotifen on cell morphology and non-cancerous cells remain unclear, which is a limitation of the present study. In future studies, any potential anti-tumor properties of pizotifen should be further investigated in vivo and in clinical studies.

Aberrant activation of the Wnt signaling pathway is associated with the occurrence and development of tumors, and it has become the focus of studies seeking to therapeutic targets (33,34). Therefore, drug development has been mainly focused on inhibiting the Wnt signaling cascade, and some of these drugs have since entered clinical trials (35,36). Genome sequencing studies have revealed that the mutation of many components of the Wnt pathway are inherited in patients with gastric cancer, such as AXIN1, AXIN2 and β-catenin (37), suggesting that targeting Wnt has potential in the treatment of gastric cancer. These data were verified in a mouse model of gastric cancer (3840). This suggests that Wnt signaling is likely to be a driving factor in the occurrence of gastric cancer.

It was demonstrated in the present study that pizotifen could significantly downregulate the expression of Wnt3a and β-catenin in MNK45 and AGS cells. β-catenin is a multifunctional protein, which was initially found to bind to E-cadherin-binding protein and participate in cell adhesion (14). E-cadherin has been reported to be a key member of the Wnt signaling pathway, with transcriptional regulatory activity (15). It is hypothesized that the disruption of E-cadherin/β-catenin complexes is involved in the malignant transformation of epithelial tumors (41). E-cadherin and N-cadherin are key markers for the EMT process, both of which serve essential roles in tumor metastasis. It was previously demonstrated that the downregulation of E-cadherin in gastric cancer could reduce the expression of the cadherin/catenin complex to increase cancer cell invasiveness (42). In the present study, it was found that pizotifen treatment in gastric cancer cell lines upregulated the expression of E-cadherin and downregulated the expression of N-cadherin and β-catenin, an effect that was reversed by concomitant treatment with BML-284, a selective Wnt signaling activator. It was also observed in the present study that MNK45 and AGS cell migration and invasion, which were inhibited by pizotifen, were restored by simultaneous BML-284 treatment. Taken together, these observations suggested that inhibition of the Wnt/β-catenin-EMT signaling pathway may be responsible for the anti-invasion properties of pizotifen.

In summary, the present study showed that pizotifen could inhibit the growth, migration and invasion of gastric cancer cells and induce cell apoptosis. The anti-cancer activity of pizotifen may be achieved by blocking the Wnt/β-catenin-EMT signaling pathway. Based on these findings, pizotifen could potentially be used in a novel therapeutic approach for the treatment of gastric cancer, a notion which warrants further clinical evaluation. However, the precise mechanism of action of pizotifen will be explored further in a future study.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

YJ, WW and XW designed the study. YJ and WW performed the experiments, and XW and JS analyzed the data. All authors interpreted the results, and produced and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Carlomagno N, Santangelo ML, Amato B, Calogero A, Saracco M, Cremone C, Miranda A, Dodaro C and Renda A: Total colectomy for cancer: Analysis of factors linked to patients' age. Int J Surg. 12 (Suppl 2):S135–S139. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Strand MS, Lockhart AC and Fields R: Genetics of gastric cancer. Surg Clin North Am. 97:345–370. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Hamashima C: Current issues and future perspectives of gastric cancer screening. World J Gastroenterol. 20:13767–13774. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Baker NE: Transcription of the segment-polarity gene wingless in the imaginal discs of drosophila, and the phenotype of a pupal-lethal wg mutation. Development. 102:489–497. 1988.PubMed/NCBI

6 

Croce JC and McClay DR: Evolution of the Wnt pathways. Methods Mol Biol. 469:3–18. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Clevers H and Nusse R: Wnt/β-catenin signaling and disease. Cell. 149:1192–1205. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Rapp J, Jaromi L, Kvell K, Miskei G and Pongracz JE: WNT signaling - lung cancer is no exception. Respir Res. 18:1672017. View Article : Google Scholar : PubMed/NCBI

9 

Byun T, Karimi M, Marsh JL, Milovanovic T, Lin F and Holcombe RF: Expression of secreted Wnt antagonists in gastrointestinal tissues: Potential role in stem cell homeostasis. J Clin Pathol. 58:515–519. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Barker N, Huch M, Kujala P, van de Wetering M, Snippert HJ, van Es JH, Sato T, Stange DE, Begthel H, van den Born M, et al: Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell. 6:25–36. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Wang K, Yuen ST, Xu J, Lee SP, Yan HH, Shi ST, Siu HC, Deng S, Chu KM, Law S, et al: Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer. Nat Genet. 46:573–582. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC, Wong SS, Liu J, Yue YG, Wang J, Yu K, et al: Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 21:449–456. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Nagaoka T, Karasawa H, Turbyville T, Rangel MC, Castro NP, Gonzales M, Baker A, Seno M, Lockett S, Greer YE, et al: Cripto-1 enhances the canonical Wnt/β-catenin signaling pathway by binding to LRP5 and LRP6 co-receptors. Cell Signal. 25:178–189. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Garinis GA, Spanakis NE, Menounos PG, Manolis EN and Peros G: Transcriptional impairment of beta-catenin/E-cadherin complex is not associated with beta-catenin mutations in colorectal carcinomas. Br J Cancer. 88:206–209. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Kikuchi A, Yamamoto H, Sato A and Matsumoto S: New insights into the mechanism of Wnt signaling pathway activation. Int Rev Cell Mol Biol. 291:21–71. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Micalizzi DS, Farabaugh SM and Ford HL: Epithelial-mesenchymal transition in cancer: Parallels between normal development and tumor progression. J Mammary Gland Biol Neoplasia. 15:117–134. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Peng Z, Wang CX, Fang EH, Wang GB and Tong Q: Role of epithelial-mesenchymal transition in gastric cancer initiation and progression. World J Gastroenterol. 20:5403–5410. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Yue H, Tang B, Zhao Y, Niu Y, Yin P, Yang W, Zhang Z and Yu P: MIR-519d suppresses the gastric cancer epithelial-mesenchymal transition via Twist1 and inhibits Wnt/β-catenin signaling pathway. Am J Transl Res. 9:3654–3664. 2017.PubMed/NCBI

19 

Cho ES, Kang HE, Kim NH and Yook JI: Therapeutic implications of cancer epithelial-mesenchymal transition (EMT). Arch Pharm Res. 42:14–24. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Fuchs R, Schwach G, Stracke A, Meier-Allard N, Absenger M, Ingolic E, Haas HS, Pfragner R and Sadjak A: The anti-hypertensive drug prazosin induces apoptosis in the medullary thyroid carcinoma cell line TT. Anticancer Res. 35:31–38. 2015.PubMed/NCBI

21 

Lin OA, Karim ZA, Vemana HP, Espinosa EV and Khasawneh FT: The antidepressant 5-HT2A receptor antagonists pizotifen and cyproheptadine inhibit serotonin-enhanced platelet function. PLoS One. 9:e870262014. View Article : Google Scholar : PubMed/NCBI

22 

Speight TM and Avery GS: Pizotifen(BC-105): A review of its pharmacological properties and its therapeutic efficacy in vascular headaches. Drugs. 3:159–203. 1972. View Article : Google Scholar : PubMed/NCBI

23 

Ahmadi AA, Shadifar M, Ataee R, Vaillancourt C, Ataee A, Oufkir T and Jafari-Sabet M: The serotonin 5-HT2A receptor antagonist ritanserin induces apoptosis in human colorectal cancer and acts in synergy with curcumin. Int Biol Biomed J. 1:56–65. 2015.

24 

Liu J, Wu X, Mitchell B, Kintner C, Ding S and Schultz PG: A small-molecule agonist of the Wnt signaling pathway. Angew Chem Int Ed Engl. 44:1987–1990. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Cheng YY, Yu J, Wong YP, Man EP, To KF, Jin VX, Li J, Tao Q, Sung JJ, Chan FK and Leung WK: Frequent epigenetic inactivation of secreted frizzled-related protein 2 (SFRP2) by promoter methylation in human gastric cancer. Br J Cancer. 97:895–901. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Hanaki H, Yamamoto H, Sakane H, Matsumoto S, Ohdan H, Sato A and Kikuchi A: An anti-Wnt5a antibody suppresses metastasis of gastric cancer cells in vivo by inhibiting receptor-mediated endocytosis. Mol Cancer Ther. 11:298–307. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Ataee R, Ajdary S, Zarrindast M, Rezayat M and Hayatbakhsh MR: Anti-mitogenic and apoptotic effects of 5-HT1B receptor antagonist on HT29 colorectal cancer cell line. J Cancer Res Clin Oncol. 136:1461–1469. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Soll C, Riener MO, Oberkofler CE, Hellerbrand C, Wild PJ, DeOliveira ML and Clavien PA: Expression of serotonin receptors in human hepatocellular cancer. Clin Cancer Res. 18:5902–5910. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Vicentini LM, Cattaneo MG and Fesce R: Evidence for receptor subtype cross-talk in the mitogenic action of serotonin on human small-cell lung carcinoma cells. Eur J Pharmacol. 318:497–504. 1996. View Article : Google Scholar : PubMed/NCBI

30 

Soll C, Jang JH, Riener MO, Moritz W, Wild PJ, Graf R and Clavien PA: Serotonin promotes tumor growth in human hepatocellular cancer. Hepatology. 51:1244–1254. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Dizeyi N, Hedlund P, Bjartell A, Tinzl M, Austild-Taskén K and Abrahamsson PA: Serotonin activates MAP kinase and PI3K/Akt signaling pathways in prostate cancer cell lines. Urol Oncol. 29:436–445. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Siddiqui EJ, Shabbir MA, Mikhailidis DP, Mumtaz FH and Thompson CS: The effect of serotonin and serotonin antagonists on bladder cancer cell proliferation. BJU Int. 97:634–639. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Hirokawa Y, Yip KH, Tan CW and Burgess AW: Colonic myofibroblast cell line stimulates colonoid formation. Am J Physiol Gastrointest Liver Physiol. 306:G547–G556. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Byun MR, Hwang JH, Kim AR, Kim KM, Hwang ES, Yaffe MB and Hong JH: Canonical Wnt signalling activates TAZ through PP1A during osteogenic differentiation. Cell Death Differ. 21:854–863. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Bu G, Lu W, Liu CC, Selander K, Yoneda T, Hall C, Keller ET and Li Y: Breast cancer-derived Dickkopf1 inhibits osteoblast differentiation and osteoprotegerin expression: Implication for breast cancer osteolytic bone metastases. Int J Cancer. 123:1034–1042. 2008. View Article : Google Scholar : PubMed/NCBI

36 

He S, Lu Y, Liu X, Huang X, Keller ET, Qian CN and Zhang J: Wnt3a: Functions and implications in cancer. Chin J Cancer. 34:554–562. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Pan KF, Liu WG, Zhang L, You WC and Lu YY: Mutations in components of the Wnt signaling pathway in gastric cancer. World J Gastroenterol. 14:1570–1574. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Kahn M: Can we safely target the WNT pathway? Nat Rev Drug Discov. 13:513–532. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Mook RA Jr, Wang J, Ren XR, Chen M, Spasojevic I, Barak LS, Lyerly HK and Chen W: Structure-activity studies of Wnt/β-catenin inhibition in the niclosamide chemotype: Identification of derivatives with improved drug exposure. Bioorg Med Chem. 23:5829–5838. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Steinhart Z, Pavlovic Z, Chandrashekhar M, Hart T, Wang X, Zhang X, Robitaille M, Brown KR, Jaksani S, Overmeer R, et al: Corrigendum: Genome-wide CRISPR screens reveal a Wnt-FZD5 signaling circuit as a druggable vulnerability of RNF43-mutant pancreatic tumors. Nat Med. 23:13842017. View Article : Google Scholar : PubMed/NCBI

41 

Kim H, Yoo SB, Sun P, Jin Y, Jheon S, Lee CT and Chung JH: Alteration of the E-cadherin/β-catenin complex is an independent poor prognostic factor in lung adenocarcinoma. Korean J Pathol. 47:44–51. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Cheng XX, Wang ZC, Chen XY, Sun Y, Kong QY, Liu J, Gao X, Guan HW and Li H: Frequent loss of membranous E-cadherin in gastric cancers: A cross-talk with Wnt in determining the fate of beta-catenin. Clin Exp Metastasis. 22:85–93. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2020
Volume 19 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jiang Y, Wang W, Wu X and Shi J: Pizotifen inhibits the proliferation and invasion of gastric cancer cells. Exp Ther Med 19: 817-824, 2020
APA
Jiang, Y., Wang, W., Wu, X., & Shi, J. (2020). Pizotifen inhibits the proliferation and invasion of gastric cancer cells. Experimental and Therapeutic Medicine, 19, 817-824. https://doi.org/10.3892/etm.2019.8308
MLA
Jiang, Y., Wang, W., Wu, X., Shi, J."Pizotifen inhibits the proliferation and invasion of gastric cancer cells". Experimental and Therapeutic Medicine 19.2 (2020): 817-824.
Chicago
Jiang, Y., Wang, W., Wu, X., Shi, J."Pizotifen inhibits the proliferation and invasion of gastric cancer cells". Experimental and Therapeutic Medicine 19, no. 2 (2020): 817-824. https://doi.org/10.3892/etm.2019.8308