Open Access

Cardamonin reduces chemotherapy resistance of colon cancer cells via the TSP50/NF‑κB pathway in vitro

  • Authors:
    • Sen Lu
    • Caizhao Lin
    • Xiaobin Cheng
    • Hanju Hua
    • Tao Xiang
    • Yu Huang
    • Xi Huang
  • View Affiliations

  • Published online on: April 26, 2018     https://doi.org/10.3892/ol.2018.8580
  • Pages: 9641-9646
  • Copyright: © Lu 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

It has previously been reported that cardamonin is able to regulate glycometabolism and vasodilation whilst also exhibiting anti‑inflammatory and antitumor properties. The antitumor effect of cardamonin is multifaceted, and so it is necessary to investigate the antitumor mechanisms of cardamonin at the molecular level. Cardamonin alters chemotherapy‑resistant colon cancer cell growth; however, the underlying mechanism is unknown. The present study was conducted to investigate the effect of cardamonin on chemotherapy‑resistant colon cancer cells and the possible mechanisms of action. Cardamonin significantly suppressed the growth of chemotherapy‑resistant colon cancer cells, induced apoptosis and promoted caspase‑3/9 activity and Bax protein expression in 5‑fluorouracil (5‑FU)‑resistant HCT‑116 cells. Cardamonin significantly suppressed c‑MYC, octamer‑binding transcription factor 4, cyclin E, testes‑specific protease 50 and nuclear factor‑κB protein expression in 5‑FU‑resistant HCT‑116 cells. The findings of the present study demonstrate that cardamonin suppresses chemotherapy‑colon cancer cell via the NF‑κB pathway in vitro.

Introduction

Colon cancer is a common malignant tumor of the gastrointestinal tract (1). Domestic studies following colorectal surgery suggest that the annual survival rate of patients with high risk stage II and stage III colon cancer is ~20–30% (2). The primary cause of mortality in patients with colon cancer is recurrence and metastasis (2). In recent years, the morbidity of colon cancer has increased (3). The main subtypes of colon cancer include adenocarcinoma, mucinous adenocarcinoma and undifferentiated carcinoma (3). The primary therapeutic method for treating colon cancer is surgical resection, which is accompanied by chemotherapy, immunotherapy and traditional Chinese medicine (TCM) (3). Metastasis is a common feature of aggressive colon cancer, with these metastases primarily affecting the liver (4); ~30% of patients exhibit pre- or postoperative liver metastasis (3). However, a minority of patients are suitable for excision and are susceptible to postoperative recurrence (5).

Distant metastasis of colon cancer is a topic that has been well researched in recent years (6). Although there are a large number of novel chemotherapeutics available, the toxic and adverse effects, as well as the tolerance generated by these therapies, mean that they are not completely satisfactory (7). Prior studies have demonstrated that patients with cancer succumb to drug-resistant disease induced by chemotherapeutics, which invalidates treatment (7). There is a lack of novel effective chemotherapeutics that can overcome this resistance (8), as the molecular mechanisms of chemotherapy resistance and the associated genetic changes are complicated and comprise multiple aspects, including drug transport, supersession, repair and regulation of apoptosis (8).

Apoptosis is regulated by a number of factors, including nuclear transcription factor-κB (NF-κB) (9), which combines with the fixed nucleotide sequences of certain gene promoters. NF-κB is activated by multiple stimuli, including cell adhesive attraction (10). The activation of NF-κB regulates cell apoptosis and inflammation (10). However, suppression of NF-κB activity is able to increase the occurrence of spontaneous apoptosis in cancer cells or increase apoptosis induced by cytotoxic drugs (9). On the basis of these data, it has been speculated that cell adhesion may regulate apoptosis genes and changes the sensitivity of cancer cells to drugs by activating NF-κB, to induce multi-cell drug resistance (10).

Cardamonin was originally extracted and separated from black cardamom, of the family Zingiberaceae (the fruit of Amomum subulatum; Fig. 1) in 1976 and is a monomeric alkaloid (11). A previous study revealed that cardamonin serves an important role in cell proliferation by regulating various signal transduction pathways (12). Cardamonin activates the mechanistic target of rapamycin complex 1 downstream target p70 ribosomal S6 kinase and eukaryotic initiation factor 4E binding protein 1 in smooth muscle cells to reverse insulin resistance (13). Furthermore, cardamonin is able to activate mitogen-activated protein kinase and NF-κB signal pathways in mononuclear cells, regulating inflammatory responses mediated by lipopolysaccharides (14). The monomer regulates cell surface receptors and Wnt/β-catenin signal pathways in the inflammatory response (14). The aim of the present study was to investigate the effect of cardamonin on chemotherapy-resistant colon cancer cell and its possible mechanism.

Materials and methods

Cell lines and cell culture

The human colon cancer HCT116 cell line was purchased from Wuhan Boster Biological Technology, Ltd. (Wuhan, China) and cultured for 24 h in Roswell Park Memorial Institute (RPMI)-1640 medium containing 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (all from Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) at 37°C in an atmosphere containing 5% CO2. Fluorouracil (5-FU) was purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). 5-FU-resistant HCT116 cells were generated by continuous exposure to increasing concentrations (0, 1.5, 3.25, 6.5, 12.5, 25 µmol/l) of 5-FU for 6–8 months.

Cell viability assay

HCT116 cells (5×103 cells/well) were cultured in a 96-well plate and treated with cardamonin (0, 10, 20, 40 and 80 µM, Sigma-Aldrich; Merck KGaA) for 24, 48 and 72 h, as described previously (15). A total of 20 µl MTT (Sigma-Aldrich; Merck KGaA; 0.5 mg/ml) was added to each well for 4 h and the medium was removed. Next, 150 µl DMSO was added per well for 20 min to dissolve formazan crystals. Cell viability was measured using an ELISA reader at 492 nm.

Cell apoptosis analysis

HCT116 cells (2×106 cells/well) were cultured in a 6-well plate and treated with cardamonin (0, 20, 40 and 80 µM) for 72 h. Apoptosis was measured using an Annexin V-FITC/PI Cell Apoptosis Double Dye kit (BD Biosciences, Franklin Lakes, NJ, USA). Cells were resuspended with 1X binding buffer (BD Biosciences, Franklin Lakes, NJ, USA). The cell suspension was incubated with 5 µl Annexin V-FITC and 5 µl PI at room temperature for 15 min in the dark. Cell apoptosis was measured using a flow cytometer (C6; BD Biosciences) and analyzed using FlowJo software (version 7.6.1; FlowJo LLC, Ashland, OR, USA).

Caspase activity assay

HCT116 cells (2×106 cells/well) were cultured overnight in a 6-well plate and treated with cardamonin (0, 20, 40 and 80 µM) for 72 h. Cell lysates were prepared by the addition of 100 µl lysis buffer (Beyotime Institute of Biotechnology, Haimen, China) for 15 min. Protein content was determined using a bicinchoninic acid assay. A total of 5–10 µg protein was incubated with caspase-3 colorimetric DEVD-pNA substrate (Beyotime Institute of Biotechnology) and caspase-9 colorimetric LEHD-pNA (Beyotime Institute of Biotechnology) at 37°C for 1 h. Caspase activity was measured using an ELISA reader at 405 nm.

Western blotting

HCT116 cells (2×106 cells/well) were cultured overnight in a 6-well plate and treated with cardamonin (0, 20, 40 and 80 µM) for 72 h. Cell lysates were prepared by the addition of 100 µl lysis buffer for 15 min. Protein content was determined using the BCA method. A total of 50 µg protein per lane was separated by 8–10% SDS-PAGE and electrotransferred onto polyvinylidene difluoride membranes. The membranes were blocked with TBST containing 5% non-fat dried milk at 37°C for 1 h and incubated with specific primary antibodies targeted at B-cell lymphoma-associated X (Bax, sc-6236, 1:1,000), Myc proto-oncogene protein (c-MYC, sc-789, 1:1,000), octamer-binding transcription factor 4 (Oct4, sc-9081, 1:1,000), and cyclin E (sc-481, 1:1,000) (all from Santa Cruz Biotechnology, Inc.), testes-specific protease 50 (TSP50, ab181993, 1:1,000; Abcam), NF-κB (sc-101749, 1:1,000) and GAPDH (sc-25778, 1:1,000) (both from Santa Cruz Biotechnology, Inc.) overnight at 4°C. Following three washes with TBST, membranes were incubated with anti-rabbit or mouse horseradish peroxidase-conjugated secondary antibodies (sc-2004 or sc-2005, 1:5,000; Santa Cruz Biotechnology, Inc.) for 1 h at room temperature. Signals were detected by enhanced chemiluminescence (ECL Plus detection system) and band density was analyzed using Carestream MI software (Carestream Health, Inc., Rochester, NY, USA).

Statistical analysis

Data are presented as the mean ± standard deviation. Statistical analysis was performed using SPSS software (version 19.0; IBM Corp., Armonk, NY, USA). Statistical analysis was performed using one-way analysis of variance analysis, with post-hoc Student-Newman-Keuls test. P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of cardamonin on 5-FU-resistant colon cancer cell growth

To determine the anticancer effect of cardamonin on chemotherapy-resistant colon cancer cell growth, an MTT assay was used to measure the viability of HCT166 cells. As presented in Fig. 2, cardamonin reduced the viability of 5-FU-resistant HCT166 cells in dose- and time-dependent manner. Particularly, 20–80 µM of cardamonin significantly reduced the viability of 5-FU-resistant HCT116 cells following treatment for 24, 48 and 72 h (Fig. 2; P<0.01).

Effect of cardamonin on 5-FU-resistant colon cancer cell death

The apoptosis rate in 5-FU-resistant HCT116 cells was assessed using flow cytometry. Increasing doses of cardamonin was demonstrated to promote 5-FU-resistant HCT166 cell apoptosis in a dose-dependent manner, with 40–80 µM of cardamonin significantly increasing 5-FU-resistant HCT166 cell apoptosis at 72 h (Fig. 3; P<0.01).

The effect of cardamonin on caspase-3/9 activities levels in 5-FU -colon cancer cell

The apoptotic mechanism of 5-FU-resistant HCT166 cells treated with cardamonin was analyzed. The activity of caspase-3 and −9 was measured using commercial kits. Caspase-3/9 activity was significantly increased in 5-FU-resistant HCT166 cells treated with 40–80 µM of cardamonin compared with the control group (Fig. 4; P<0.01).

The effect of cardamonin on Bax, c-MYC and Oct4 protein expression in 5-FU-resistant colon cancer cells

The influence of cardamonin on Bax, c-MYC and Oct4 protein expression in chemotherapy-resistant HCT116 cells was assessed. Compared with the control, 40–80 µM of cardamonin significantly increased Bax protein expression and suppressed that of c-MYC and Oct4 in 5-FU-resistant HCT166 cells (Fig. 5; P<0.01).

The effect of cardamonin on cyclin E, TSP50 and NF-κB protein expression in 5-FU-resistant colon cancer cells

Western blotting was used to investigate the possible mechanism by which cardamonin exhibits its anticancer effect. The results revealed that 40–80 µM of cardamonin significantly suppressed of cyclin E, TSP50 and NF-κB protein expression in 5-FU-HCT166 cell, compared with the control group (Fig. 6; P<0.01).

Discussion

Chemotherapy is an important treatment modality for patients with colon cancer; however, there are a number of associated negative side effects, including myelosuppression, gastrointestinal reaction and neurovirulence (16). As a result, the development of effective treatments without these effects is a frequently studied topic in the field of oncology (17). It has been reported that TCM exhibits favorable effects in colon cancer, improving symptoms and increasing the survival rate and patient quality of life (17). In addition, TCM avoids the side effects of chemotherapy by treating them (18). In the present study, cells were treated with 0, 10, 20, 40 and 80 µM of cardamonin for 24, 48 and 72 h, as previously described (15); the results indicated that cardamonin significantly reduced cell viability and induced apoptosis in 5-FU-resistant HCT116 cells. Jia et al (14) reported that cardamonin reduced chemotherapy-enriched breast cancer.

NF-κB serves an important role in the development and progression of tumors (19). NF-κB activation is associated with oncogenesis, angiogenesis, distant metastasis, anti-apoptosis and chemotherapy resistance (19). The association between NF-κB activation and chemotherapy resistance has previously been demonstrated (19). Cells overexpressing NF-κB were insensitive to the chemotherapeutic bleomycin in B-cell lymphoma (20). In the present study, cardamonin significantly suppressed NF-κB protein expression in 5-FU-resistant HCT166 cells.

Apoptosis is an important physiological and pathological process (21). When external stimulating factors act on pro-apoptotic proteins, including caspase-3 and Bax, apoptotic pathways are activated (22). When external stimulating factors act on anti-apoptotic proteins, such as B-cell lymphoma 2, apoptosis is restrained and cell survival increases (22). Apoptosis is regulated by multiple factors, including NF-κB which is activated by a number of stimuli and suppressed apoptosis by regulating the expression of pro- and anti-apoptotic genes (23). The results of the present study indicate that cardamonin significantly increases caspase-3/9 activity and Bax expression in 5-FU-resistant HCT116 cell.

c-MYC is a nuclear protein, and can bind chromosomal DNA and regulates cell growth, differentiation and malignant transformation (24). In a number of human cancer cell lines, including myelogenous leukemia, retinoblastoma, neuroblastoma, breast cancer and lung cancer, the relevant sequences of c-MYC are amplified (25). Gene amplification of c-MYC is also observed in cell lines of humans' colon cancer (25,26). These findings also indicate that c-MYC has potential as an antitumor target. The expression of c-MYC is associated with cell growth and proliferation and is a potential inducible factor of apoptosis (27). The results of the present study demonstrate that cardamonin significantly suppresses c-MYC protein expression in 5-FU-resistant HCT166 cells. Park et al (28) demonstrated that cardamonin suppresses the proliferation of colon cancer cells by inhibiting the expression of cyclin D1 and c-MYC (28).

Oct4 regulates the cellular function of human embryonic stem cells (29). There are epithelial and melanin stem cells positive for Oct4 expression in human hair follicle tissues which are yet to differentiate into multiple cells (30). A previous study reported that Oct4 was abnormally expressed in malignant colorectal, lung and breast cancers (30). Oct4 expression is associated with the occurrence and development of tumors and the prognosis of patients (30). A prior study reported that Oct4 serves an important role in the drug resistance of malignant tumors, including prostate and liver tumors (30). Oct4 mediates chemotherapy resistance via the Oct4-AKT-ABCG2 pathway (31). Together, these studies suggest that cardamonin suppresses Oct4 protein expression in 5-FU-resistant HCT166 cells.

The expression of TSP50 promotes tumor development (32). TSP50 gene silencing in mouse teratocarcinoma P19 cells has been reported to suppress tumor cell proliferation, colony formation and migration, induce apoptosis and enhance sensitivity to doxorubicin (33). The mechanism by which this occurs is reported to be associated with the n E proteins in 5-FU-resistant HCT116 cells. Mi et al (15) reported that cardamonin inhibited cell viability via blocking the activation of the TSP50-mediated NF-κB signaling pathway in cancer cells (15). It is possible that repression of the NF-κB signaling and cyclin E proteins in 5-FU-resistant HCT116 cells. Mi et al (15) reported that cardamonin inhibited cell viability via blocking the activation of the TSP50-mediated NF-κB signaling pathway in cancer cells (15). It is possible that repression of the NF-κB signaling pathway is the mechanism by which cardamonin elicits its anticancer effect in chemotherapy-resistant colon cancer cells.

In conclusion, the results of the present study demonstrated that cardamonin significantly suppresses chemotherapy-resistant colon cancer cell growth, induces apoptosis and promotes the activation of caspase-3/9 and Bax expression. These data suggest that the anticancer effect of cardamonin in 5-FU-resitant HCT116 cells may be mediated via TSP50/NF-κB protein expression (Fig. 7). Cardamonin may therefore be a potential treatment for chemotherapy-resistant colon cancer and should be researched further.

Competing interests

The authors declare that they have no competing interests.

References

1 

Kobayashi T, Masaki T, Kogawa K, Matsuoka H and Sugiyama M: Efficacy of gum chewing on bowel movement after open colectomy for left-sided colorectal cancer: A randomized clinical trial. Dis Colon Rectum. 58:1058–1063. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Karoui M, Rullier A, Luciani A, Bonnetain F, Auriault ML, Sarran A, Monges G, Trillaud H, Le Malicot K, Leroy K, et al: Neoadjuvant FOLFOX 4 versus FOLFOX 4 with Cetuximab versus immediate surgery for high-risk stage II and III colon cancers: A multicentre randomised controlled phase II trial-the PRODIGE 22-ECKINOXE trial. BMC Cancer. 15:5112015. View Article : Google Scholar : PubMed/NCBI

3 

Nuñez-Sánchez MA, García-Villalba R, Monedero-Saiz T, García-Talavera NV, Gómez-Sánchez MB, Sánchez-Álvarez C, García-Albert AM, Rodríguez-Gil FJ, Ruiz-Marín M, Pastor-Quirante FA, et al: Targeted metabolic profiling of pomegranate polyphenols and urolithins in plasma, urine and colon tissues from colorectal cancer patients. Mol Nutr Food Res. 58:1199–1211. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Kaufman HL, Kim DW, Kim-Schulze S, DeRaffele G, Jagoda MC, Broucek JR and Zloza A: Results of a randomized phase I gene therapy clinical trial of nononcolytic fowlpox viruses encoding T cell costimulatory molecules. Hum Gene Ther. 25:452–460. 2014. View Article : Google Scholar : PubMed/NCBI

5 

André T, de Gramont A, Vernerey D, Chibaudel B, Bonnetain F, Tijeras-Raballand A, Scriva A, Hickish T, Tabernero J, Van Laethem JL, et al: Adjuvant fluorouracil, leucovorin, and oxaliplatin in stage II to III colon cancer: Updated 10-year survival and outcomes according to BRAF mutation and mismatch repair status of the MOSAIC study. J Clin Oncol. 33:4176–4187. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Hussmann M, Janke K, Kranz P, Neumann F, Mersch E, Baumann M, Goepelt K, Brockmeier U and Metzen E: Depletion of the thiol oxidoreductase ERp57 in tumor cells inhibits proliferation and increases sensitivity to ionizing radiation and chemotherapeutics. Oncotarget. 6:39247–39261. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Apostolou P, Toloudi M, Kourtidou E, Mimikakou G, Vlachou I, Chatziioannou M and Papasotiriou I: Use of the comet assay technique for quick and reliable prediction of in vitro response to chemotherapeutics in breast and colon cancer. J Biol Res (Thessalon). 21:142014. View Article : Google Scholar : PubMed/NCBI

8 

Kozovska Z, Gabrisova V and Kucerova L: Colon cancer: Cancer stem cells markers, drug resistance and treatment. Biomed Pharmacother. 68:911–916. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Liang L and Huang J: Oxymatrine inhibits epithelial-mesenchymal transition through regulation of NF-kappaB signaling in colorectal cancer cells. Oncol Rep. 36:1333–1338. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Mladenova D, Pangon L, Currey N, Ng I, Musgrove EA, Grey ST and Kohonen-Corish MR: Sulindac activates NF-kappaB signaling in colon cancer cells. Cell Commun Signal. 11:732013. View Article : Google Scholar : PubMed/NCBI

11 

Wang S, Zhai C, Zhang Y, Yu Y, Zhang Y, Ma L, Li S and Qiao Y: Cardamonin, a novel antagonist of hTRPA1 cation channel, reveals therapeutic mechanism of pathological pain. Molecules. 21:pii: E1145. 2016. View Article : Google Scholar

12 

Niu PG, Zhang YX, Shi DH, Liu Y, Chen YY and Deng J: Cardamonin inhibits metastasis of lewis lung carcinoma cells by decreasing mTOR activity. PLoS One. 10:e01277782015. View Article : Google Scholar : PubMed/NCBI

13 

Liao Q, Shi DH, Zheng W, Xu XJ and Yu YH: Antiproliferation of cardamonin is involved in mTOR on aortic smooth muscle cells in high fructose-induced insulin resistance rats. Eur J Pharmacol. 641:179–186. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Jia D, Tan Y, Liu H, Ooi S, Li L, Wright K, Bennett S, Addison CL and Wang L: Cardamonin reduces chemotherapy-enriched breast cancer stem-like cells in vitro and in vivo. Oncotarget. 7:771–785. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Mi XG, Song ZB, Sun LG, Bao YL, Yu CL, Wu Y and Li YX: Cardamonin inhibited cell viability and tumorigenesis partially through blockade of testes-specific protease 50-mediated nuclear factor-kappaB signaling pathway activation. Int J Biochem Cell Biol. 73:63–71. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Corvaisier M, Bauzone M, Corfiotti F, Renaud F, El Amrani M, Monté D, Truant S, Leteurtre E, Formstecher P, Van Seuningen I, et al: Regulation of cellular quiescence by YAP/TAZ and cyclin E1 in colon cancer cells: Implication in chemoresistance and cancer relapse. Oncotarget. 7:56699–56712. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Ayeka PA, Bian Y, Mwitari PG, Chu X, Zhang Y, Uzayisenga R and Otachi EO: Immunomodulatory and anticancer potential of Gan cao (Glycyrrhiza uralensis Fisch.) polysaccharides by CT-26 colon carcinoma cell growth inhibition and cytokine IL-7 upregulation in vitro. BMC Complement Altern Med. 16:2062016. View Article : Google Scholar : PubMed/NCBI

18 

Dahl O and Pfeffer F: Twenty-five years with adjuvant chemotherapy for colon cancer-a continuous evolving concept. Acta Oncol. 54:1–4. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Kang HW, Kim JM, Cha MY, Jung HC, Song IS and Kim JS: Deguelin, an Akt inhibitor, down-regulates NF-κB signaling and induces apoptosis in colon cancer cells and inhibits tumor growth in mice. Dig Dis Sci. 57:2873–2882. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Stark LA, Reid K, Sansom OJ, Din FV, Guichard S, Mayer I, Jodrell DI, Clarke AR and Dunlop MG: Aspirin activates the NF-kappaB signalling pathway and induces apoptosis in intestinal neoplasia in two in vivo models of human colorectal cancer. Carcinogenesis. 28:968–976. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Wang J, Liu L, Qiu H, Zhang X, Guo W, Chen W, Tian Y, Fu L, Shi D, Cheng J, et al: Ursolic acid simultaneously targets multiple signaling pathways to suppress proliferation and induce apoptosis in colon cancer cells. PLoS One. 8:e638722013. View Article : Google Scholar : PubMed/NCBI

22 

Radhakrishnan EK, Bava SV, Narayanan SS, Nath LR, Thulasidasan AK, Soniya EV and Anto RJ: [6]-Gingerol induces caspase-dependent apoptosis and prevents PMA-induced proliferation in colon cancer cells by inhibiting MAPK/AP-1 signaling. PLoS One. 9:e1044012014. View Article : Google Scholar : PubMed/NCBI

23 

Elkady AI, Hussein RA and El-Assouli SM: Harmal extract induces apoptosis of HCT116 human colon cancer cells, mediated by inhibition of nuclear factor-κB and activator protein-1 signaling pathways and induction of cytoprotective genes. Asian Pac J Cancer Prev. 17:1947–1959. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Ding X, Zhou X, Jiang B, Zhao Q and Zhou G: Triptolide suppresses proliferation, hypoxia-inducible factor-1α and c-Myc expression in pancreatic cancer cells. Mol Med Rep. 12:4508–4513. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Thao NP, Luyen BT, Kim EJ, Kang JI, Kang HK, Cuong NX, Nam NH, Kiem PV, Minh CV and Kim YH: Steroidal constituents from the edible sea urchin Diadema savignyi Michelin induce apoptosis in human cancer cells. J Med Food. 18:45–53. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Takaoka Y, Shimizu Y, Hasegawa H, Ouchi Y, Qiao S, Nagahara M, Ichihara M, Lee JD, Adachi K, Hamaguchi M and Iwamoto T: Forced expression of miR-143 represses ERK5/c-Myc and p68/p72 signaling in concert with miR-145 in gut tumors of Apc(Min) mice. PLoS One. 7:e421372012. View Article : Google Scholar : PubMed/NCBI

27 

Kawasaki Y, Komiya M, Matsumura K, Negishi L, Suda S, Okuno M, Yokota N, Osada T, Nagashima T, Hiyoshi M, et al: MYU, a Target lncRNA for Wnt/c-Myc signaling, mediates induction of CDK6 to promote cell cycle progression. Cell Rep. 16:2554–2564. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Park S, Gwak J, Han SJ and Oh S: Cardamonin suppresses the proliferation of colon cancer cells by promoting β-catenin degradation. Biol Pharm Bull. 36:1040–1044. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Talebi A, Kianersi K and Beiraghdar M: Comparison of gene expression of SOX2 and OCT4 in normal tissue, polyps, and colon adenocarcinoma using immunohistochemical staining. Adv Biomed Res. 4:2342015.PubMed/NCBI

30 

Ng WL, Chen G, Wang M, Wang H, Story M, Shay JW, Zhang X, Wang J, Amin AR, Hu B, et al: OCT4 as a target of miR-34a stimulates p63 but inhibits p53 to promote human cell transformation. Cell Death Dis. 5:e10242014. View Article : Google Scholar : PubMed/NCBI

31 

Amini S, Fathi F, Mobalegi J, Sofimajidpour H and Ghadimi T: The expressions of stem cell markers: Oct4, Nanog, Sox2, nucleostemin, Bmi, Zfx, Tcl1, Tbx3, Dppa4, and Esrrb in bladder, colon, and prostate cancer, and certain cancer cell lines. Anat Cell Biol. 47:1–11. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Song ZB, Bao YL, Zhang Y, Mi XG, Wu P, Wu Y, Yu CL, Sun Y, Zheng LH, Huang YX, et al: Testes-specific protease 50 (TSP50) promotes cell proliferation through the activation of the nuclear factor-κB (NF-κB) signalling pathway. Biochem J. 436:457–467. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Liu YL and Sun YN: Down-regulation of testes-specific protease 50 induces apoptosis in human laryngocarcinoma HEp2 cells in a NF-κB-mediated pathway. Mol Biol Rep. 41:7743–7747. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2018
Volume 15 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lu S, Lin C, Cheng X, Hua H, Xiang T, Huang Y and Huang X: Cardamonin reduces chemotherapy resistance of colon cancer cells via the TSP50/NF‑κB pathway in vitro. Oncol Lett 15: 9641-9646, 2018
APA
Lu, S., Lin, C., Cheng, X., Hua, H., Xiang, T., Huang, Y., & Huang, X. (2018). Cardamonin reduces chemotherapy resistance of colon cancer cells via the TSP50/NF‑κB pathway in vitro. Oncology Letters, 15, 9641-9646. https://doi.org/10.3892/ol.2018.8580
MLA
Lu, S., Lin, C., Cheng, X., Hua, H., Xiang, T., Huang, Y., Huang, X."Cardamonin reduces chemotherapy resistance of colon cancer cells via the TSP50/NF‑κB pathway in vitro". Oncology Letters 15.6 (2018): 9641-9646.
Chicago
Lu, S., Lin, C., Cheng, X., Hua, H., Xiang, T., Huang, Y., Huang, X."Cardamonin reduces chemotherapy resistance of colon cancer cells via the TSP50/NF‑κB pathway in vitro". Oncology Letters 15, no. 6 (2018): 9641-9646. https://doi.org/10.3892/ol.2018.8580