Role of gambogic acid and NaI131 in A549/DDP cells

  • Authors:
    • Jing Huang
    • Xiaoli Zhu
    • Huan Wang
    • Shuhua Han
    • Lu Liu
    • Yan Xie
    • Daozhen Chen
    • Qiang Zhang
    • Li Zhang
    • Yue Hu
  • View Affiliations

  • Published online on: November 25, 2016     https://doi.org/10.3892/ol.2016.5435
  • Pages: 37-44
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Resistance to platinum in tumor tissue is a considerable barrier against effective lung cancer treatment. Radionuclide therapy is the primary adjuvant treatment, however, the toxic side effects limit its dosage in the clinical setting. Therefore, the present study aimed to determine whether an NaI131 radiosensitizer could help reduce the toxic side effects of radionuclide therapy. In vitro experiments were conducted to determine whether NaI131 can inhibit platinum resistance in A549/DDP cells, which are cisplatin‑resistant non-small cell lung cancer cells, and whether gambogic acid (GA) is an effective NaI131 radiosensitizer. Cell proliferation following drug intervention was analyzed using MTT and isobolographic analysis. Apoptosis was assessed by flow cytometry. In addition, the mechanisms of drug intervention were analyzed by measuring the expression of P-glycoprotein (P‑gP), B cell lymphoma 2 (Bcl‑2), Bcl2‑associated X protein (Bax) and P53 using western blot analysis and immunocytochemistry. According to isobolographic analysis, a low concentration of NaI131 combined with GA had a synergistic effect on the inhibition of A549/DDP cell proliferation, which was consistent with an increased rate of apoptosis. Furthermore, the overexpression of Bax, and the downregulation of P-gP, P53 and Bcl‑2 observed demonstrated the potential mechanism(s) of NaI131 and GA intervention. NaI131 may induce apoptosis in A549/DDP cells by regulating apoptosis‑related proteins. A low concentration combination of NaI131 and GA was able to significantly inhibit A549/DDP cell proliferation and induce cell apoptosis. Thus, the two drugs appear to have a synergistic effect on apoptosis of A549/DDP cells.

Introduction

Lung cancer is the leading cause of cancer-associated mortality in the USA. In 2015, there was an estimated 221,200 novel cases of lung and bronchial cancer diagnosed, and 158,040 mortalities were attributed to lung cancer (1). In China, lung cancer-associated mortality accounts for >20% of all cancer mortalities (2). Currently, chemotherapy regimens containing platinum are the primary therapies for advanced non-small cell lung cancer (NSCLC) (3). However, the efficacy of chemotherapy is influenced by the resistance to platinum in the tumor tissue, leading to variations in efficacy between individuals (4). Several mechanisms have been proposed to explain the resistance of cancer cells to chemotherapy (57). It has been demonstrated in previous studies that resistance to platinum in lung cancer cells could be enhanced by P-glycoprotein (P-gP) expression, which is upregulated by the multi-drug resistance-1 gene (8,9). In addition, other studies have demonstrated that an imbalance in the expression of cell apoptosis-related regulatory proteins, such as P53 and Bcl2-associated X protein (Bax), can inhibit tumor cell apoptosis, causing lung cancer cells to become resistant to platinum (10,11).

Radionuclide therapy is an important method of tumor irradiation. The combination of 125I implantation in tumor tissue and chemotherapy has an important role in the clinical treatment of lung cancer, and this regimen has a significantly positive efficacy compared with chemotherapy alone (12). However, to the best of our knowledge, no studies have been performed regarding the therapeutic mechanism of radionuclides in drug-resistant tumor cells. Highly concentrated radioactivity also results in toxic side effects for patients, such as myelosuppression and aplastic anemia (13), limiting the dosage of radionuclide therapy that can be used in the clinic. Therefore, it is critical to identify an effective radiosensitizer to reduce the side effects of radionuclide therapy.

Gambogic acid (GA) is extracted from gamboge and the monomers are purified. The molecular formula of GA is C38H44O9 (molecular weight, 628.34 g/mol) and its chemical structure is known. GA is an effective anti-tumor agent that has been shown to have multiple effects on several types of solid human tumors in vitro and in vivo (14,15). For example, studies have demonstrated that GA can upregulate the expression of Bax and P53, and downregulate the expression of B cell lymphoma-2 (Bcl-2), inhibiting tumor cell apoptosis (16,17). GA has also been implicated in several mechanisms of cisplatin resistance (18). Furthermore, it has exhibited detectable effects on patients with lung cancer, colorectal cancer and renal cell carcinoma (1921). Thus, GA has been approved for evaluation in a phase II clinical trial for NSCLC in China (approval no. 2004L00333) (22).

In vitro experiments were conducted in the current study to determine whether NaI131 is able to inhibit platinum resistance in cisplatin-resistant A549/DDP NSCLC cells and whether GA is an effective NaI131 radiosensitizer during the treatment of A549/DDP cells. The present study also aimed to determine the potential mechanism(s) of GA-associated NaI131 radiosensitization in lung cancer.

Materials and methods

Materials

Human cisplatin-resistant NSCLC cells were provided by Dr Zhibo Hou (Department of Pneumology, Nanjing Chest Hospital, Medical School of Southeast University, Nanjing, China). MTT and mouse monoclonal anti-P-gP (catalog no. ab3366), anti-Bcl-2 (catalog no. ab692), anti-Bax (catalog no. ab77566), anti-P53 (catalog no. ab28), anti-β-actin (catalog no. ab8226) antibodies, and goat anti-mouse IgG secondary antibody (catalog no. ab6789) were acquired from Abcam (Cambridge, MA, USA). The secondary antibody used for immunocytochemistry, goat anti-mouse IgG/horseradish peroxidase-conjugated antibody, was purchased from ZSGB-BIO (Beijing, China).

Cell culture

Cells were cultured in RPMI-l640 medium supplemented 10% fetal bovine serum (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA). Cells were digested with 0.25% EDTA-phosphate-buffered saline (PBS) during the logarithmic proliferation phase. Cell suspensions were transferred to frozen vessels and stored at 4°C for 30 min, −20°C for 1 h, and −80°C overnight in liquid nitrogen.

MTT assay

A549/DDP cells in the logarithmic proliferation phase were randomly assigned to the NaI131 intervention group, GA intervention group or control group. The A549/DDP cells were seeded into 96-well plates (1×104-1×105 cells/well). The NaI131 group was treated with NaI131 5.7, 11.4, 17.1, 22.8, 28.5 or 34.1 MBq; the GA group was treated with GA 0.5, 1.0, 1.5, 2.0 or 3.0 µg/ml; and the control group was treated with equal volumes of PBS. The cells were incubated in a standard cell culture incubator at 37°C with 5% CO2. After 48 h of cell culture, 5 mg/ml MTT (20 µl/well) was added to the media and the cells were additionally incubated for 4 h. Dimethylsulfoxide (150 µl; Sigma-Aldrich, St. Louis, MO, USA) was added to the cells in each of the wells after the media was removed, and the cells were further incubated for 10 min. The optical density (OD) of each well was measured using a microplate reader (Multiskan™ GO Microplate Spectrophotometer; Thermo Fisher Scientific, Inc.) at 560 nm. All experiments were performed in triplicate, and results were analyzed according to the following formula: Cell inhibitory rate (%) = (1- OD test group / OD control group) × 100.

Treatment with 5.7 MBq NaI131 and 0.2 µg/ml GA was also performed; these drug concentrations were empirically determined based on the various doses of NaI131 and GA evaluated, which were then used to measure the half maximal inhibitory concentrations (IC50) of the two drugs. Graphical representations of the isobolographic analysis, performed as previously described (23,24), were used to determine whether NaI131 and GA synergistically inhibit the proliferation of A549/DDP cells.

Apoptosis and protein detection
Cell treatment

A549/DDP cells in the logarithmic proliferation phase were assigned to the NaI131, GA, NaI131 combined with GA or control group. The A549/DDP cells were seeded into 96-well plates (1×104-1×105 cells/well). Based on IC50 values, the NaI131 group was treated with 17.5 MBq NaI131; the GA group was treated with 1.5 µg/ml GA; and the combined treatment group was treated with 17.5 MBq NaI131 and 0.2 µg/ml GA. The control group was treated with an equal volume of PBS.

Apoptosis analysis by flow cytometry

After 48 h of culture (at 37°C with 5% CO2), cell apoptosis was analyzed using Annexin V-fluorescein isothiocyanate (FITC) and propidium iodide (PI) iodide kit (Kaiji Bio, Co., Nanjing, China). Adherent and floating cells were harvested and disaggregated to a single-cell suspension. Staining was performed according to the manufacturer's protocols. The data were analyzed by flow cytometry using CXP2.2 software (Beckman Coulter, Inc., Brea, CA, USA).

Bcl-2, P-gP, Bax and P53 immunocytochemistry

After 48 h of culture (at 37°C with 5% CO2), 5×105 cells/ml were placed on Culture Slides (BD Biosciences, Bedford, MA, USA). The slides were washed with PBS, and fixed using 4% paraformaldehyde, permeabilized with 0.5% Triton X-100, incubated with 3% hydrogen peroxide and then maintained in a blocking solution composed of PBS with 3% albumin from bovine serum (Sigma-Aldrich). The slides were subsequently incubated overnight at 4°C with specific primary antibodies against Bcl-2, P-gP, Bax and P53 (dilution, 1:200). Subsequently, the cells were washed with PBS, and incubated with diluted biotinylated secondary antibody and then with VECTASTAIN® ABC Reagent (Dako Italia, Milan, Italy). Finally, the slides were washed, incubated with peroxidase substrate solution until desired stain intensity developed (Peroxidase/DAB; Dako Italia), rinsed in tap water, counterstained with Mayer's hematoxylin and mounted with BioMount. The results of immunocytochemical staining was negative (−), weak positive (+) and positive (++) and strong positive (+++). Analysis of the OD data was performed using ImageJ software (version 1.37; National Institutes of Health, Bethesda, MD, USA). The results were expressed as the mean ± standard deviation (%).

Western blot analysis

Cell protein was extracted using 500 µl radioimmunoprecipitation assay lysis buffer with 5 µl phenylmethylsulfonyl fluoride and protease inhibitor (Abcam, Cambridge, UK). Total proteins were quantified using the Bicinchoninic Acid Assay (Beyotime Institute of Biotechnology, Shanghai, China), according to the manufacturer's protocols. Total protein (20 µg) was loaded onto an 8% sodium dodecyl sulfate-polyacrylamide gel and transferred to a polyvinylidene difluoride membrane (EMD Millipore, Billerica, MA, USA). The membrane was incubated for 1 h at 25°C in 5% skimmed dried milk and then washed three times for 5 min at 25°C using blocking buffer. Subsequently, the membrane was incubated overnight at 4°C with specific primary antibodies for P-gp, p53, Bax and Bcl-2. All antibodies were diluted to 1:1,000. Subsequent to being washed three times with TBST for 5 min each, the membranes were incubated for 1 h with horseradish peroxidase-conjugated secondary antibodies (dilution, 1:5,000). In order to evaluate protein expression accurately, β-actin was used as an internal standard. Band intensity was analyzed with ImageJ software, and protein expression was presented as the ratio of the protein band intensity to β-actin in the same blot.

Statistical analysis

Every experiment was performed in triplicate, and each group was comprised of three duplicate wells. Statistics were analyzed with SPSS (version 19.0; IBM SPSS, Armonk, NY, USA). All data are presented as the mean ± standard deviation. Graphs were plotted using Microsoft Office Excel 2007 (Microsoft Inc., Redmond, WA, USA). Differences between groups were analyzed by one-way analysis of variance or independent sample t-tests. The SNK test are used at the second stage of the analysis of variance if the null hypothesis was rejected. P<0.05 was considered to indicate a statistically significant difference.

Results

Cell proliferation inhibition rates per group
Inhibitory effect of NaI131 or GA alone on A549/DDP cells

The inhibition of A549/DDP cell proliferation after 48 h of treatment with NaI131 is shown in Table I. The half maximal inhibitory concentration (IC50) value was 17.54±1.86 MBq (Fig. 1).

Table I.

Cell proliferation inhibition ratio of A549/DDP cells following treatment with various doses of NaI131.

Table I.

Cell proliferation inhibition ratio of A549/DDP cells following treatment with various doses of NaI131.

NaI131, MBqInhibition ratio of A549/DDP cells, a
5.718.12±11.16
11.429.31±7.61
17.150.43±9.00
22.864.52±2.32
28.565.82±5.67
34.170.78±2.29

a Data are presented as the mean ± standard deviation.

The cell proliferation inhibition ratio of A549/DDP cells after 48 h of GA treatment is shown in Table II. The IC50 value was 1.46±0.07 µg/ml (Fig. 2).

Table II.

Cell proliferation inhibition ratio of A549/DDP cells following treatment with various doses of GA.

Table II.

Cell proliferation inhibition ratio of A549/DDP cells following treatment with various doses of GA.

GA, µg/mlInhibition ratio of A549/DDP cells, a
0.57.67±1.53
1.029.00±2.65
1.553.33±2.08
2.068.33±2.08
3.078.00±1.00

a Data are presented as the mean ± standard deviation. GA, gambogic acid.

Inhibitory effect of NaI131 combined with GA on A549/DDP cells

NaI131 and GA administered singly would kill a limited number of cells. Preincubation of A549/DDP cells with the usual GA dose (1.46 µg/ml) followed by their exposure to NaI131 (17.54 MBq) results in the rapid death of all cells. Therefore, in principle, a combination consisting of a low dose of GA and appropriately adjusted NaI131 dosage would be effective. Consequently, doses of 5.7 MBq NaI131 and 0.2 µg/ml GA were empirically determined for the combined treatment. The effects of various doses of NaI131 and GA on cell proliferation were determined by the MTT assay, and the proliferation inhibition rates of the two groups were calculated. The proliferation inhibition rates of A549/DDP cells after treatment with NaI131 (5.7 MBq) and GA (various doses) for 48 h are shown in Table III. The IC50 value of GA was 0.22±0.03 µg/ml. The cell proliferation inhibition rates of A549/DDP cells after 48 h of treatment with GA (0.2 µg/ml) and NaI131 (various doses) are indicated in Table IV. The IC50 value of NaI131 was 7.14±0.88 MBq.

Table III.

Cell proliferation inhibition rate of A549/DDP cells following treatment with NaI131 (5.7 MBq) and GA (various doses).

Table III.

Cell proliferation inhibition rate of A549/DDP cells following treatment with NaI131 (5.7 MBq) and GA (various doses).

GA, µg/mlInhibition ratio of A549/DDP cells, a
0.133.10±4.30
0.240.30±6.00
0.352.83±8.56
0.475.16±3.47

a Data are presented as the mean ± standard deviation. GA, gambogic acid.

Table IV.

Cell proliferation inhibition rate of A549/DDP cells following treatment with GA (0.2 µg/ml) and NaI131 (various doses).

Table IV.

Cell proliferation inhibition rate of A549/DDP cells following treatment with GA (0.2 µg/ml) and NaI131 (various doses).

NaI131, MBqInhibition ratio of A549/DDP cells, a
5.744.82±5.52
11.459.89±3.07
17.173.72±3.69

a Data are presented as the mean ± standard deviation. GA, gambogic acid.

Based on the aforementioned data, two composite points were plotted: i) 5.7 MBq NaI131 and 0.22±0.03 µg/ml GA; and ii) 0.2 µg/ml GA and 7.14±0.88 MBq NaI131. In A549/DDP cells, the IC50 of NaI131 was 17.54±1.86 MBq (point A) and the IC50 of GA was 1.46±0.07 µg/ml (point B) (Fig. 3). According to the isobolographic analysis, the straight line connecting A and B is the locus of points (dose pairs) that will produce this effect in a simply additive combination. Two composite points are in the lower left from the hypotenuse, suggesting that NaI131 and GA synergistically inhibit A549/DDP cell proliferation.

Apoptosis detection analysis

Annexin V-FITC and PI flow cytometry revealed that the cell count ratios in the Q4 quadrant were significantly increased in the NaI131, GA and combined treatment groups compared with the control group, indicating an increase in the rate of early apoptosis. Furthermore, compared with the control group, the cell count ratio in the Q2 quadrant (late apoptosis) was significantly increased in the NaI131, GA and combined treatment groups. In addition, the number of cells in early or late apoptosis in the combined treatment group were increased significantly compared with those in the NaI131 and GA alone groups. Total apoptosis rates also showed the same patterns (Table V; Fig. 4).

Table V.

Cell apoptosis was analyzed using flow cytometry following 48 h of cell culture.

Table V.

Cell apoptosis was analyzed using flow cytometry following 48 h of cell culture.

Apoptosis, %

Treatment groupEarlyLateTotal
Control1.74±0.602.21±1.333.95±1.24
NaI131 10.39±1.17a   4.39±0.96a 14.78±1.91a
GA 13.38±1.96a   5.73±1.58a 19.11±1.03a
Combined 20.06±3.02b 11.49±2.34b 31.55±5.32b

{ label (or @symbol) needed for fn[@id='tfn5-ol-0-0-5435'] } Data are presented as the mean ± standard deviation.

a P<0.05 vs. control group

b P<0.01 vs. control NaI131 and GA groups. GA, gambogic acid.

Intracellular P-gP, Bcl-2, Bax and P53 protein expression detected by immunocytochemistry

The P-gP protein was detected by immunostaining with granular coloring observed in the cell membrane and cytoplasm surrounding the nucleus. Expression was strongly positive (+++) in the control cells, positive (+) in the NaI131 and GA cells, and moderate (±) in the combined treatment cells. P53 protein was primarily detected in the nucleus. Its expression was positive (++) in the control group, weakly positive (+) in the NaI131 and GA groups, and moderate (±) in the combined treatment group. Bax protein was detected with cytoplasmic staining. It showed moderate expression (±) in the control group, weakly positive expression (+) in the NaI131 and GA groups, and positive expression (++) in the combined treatment group. Bcl-2 protein was detected by immunostaining with granular coloring primarily observed in the cell membrane and cytoplasm. Its expression was positive (++) in the control group, weakly positive (+) in the NaI131 and GA groups, and moderate (±) in the combined treatment group (Fig. 5).

P-gP, Bcl-2, Bax and P53 protein expression detected by western blot analysis

The protein expression levels of P-gP, Bcl-2 and P53 significantly decreased, while the expression level of Bax protein significantly increased in the combined treatment group compared with the control, NaI131 and GA groups. Similarly, P-gP, Bcl-2 and P53 levels significantly decreased and Bax levels significantly increased compared in the NaI131 and GA alone groups compared with the control group (Table VI; Fig. 6).

Table VI.

Expression of P-gP, Bcl-2, P53 and Bax protein expression as detected by western blot assay.

Table VI.

Expression of P-gP, Bcl-2, P53 and Bax protein expression as detected by western blot assay.

Protein expression level

Treatment groupP-gPBaxBcl-2P53
Control0.83±0.080.15±0.010.89±0.200.67±0.11
NaI131   0.55±0.05a   0.32±0.02a   0.59±0.13a0.54±0.09
GA   0.74±0.07a   0.29±0.02a   0.66±0.15a   0.57±0.09a
Combined   0.34±0.03b   0.85±0.06b   0.28±0.06b   0.36±0.06b

{ label (or @symbol) needed for fn[@id='tfn8-ol-0-0-5435'] } Data are presented as mean ± standard deviation.

a P<0.05 vs. control group

b P<0.01 vs. control NaI131 and GA groups. GA, gambogic acid; P-gP, P-glycoprotein; Bax, Bcl2-associated X protein; Bcl-2, B cell lymphoma-2.

Discussion

Previous studies have demonstrated that cisplatin resistance in NSCLC cells occurs via several mechanisms (811). These mechanisms of resistance primarily involve the abnormal expression of drug transporters and drug metabolizing enzymes in cells (25), increased DNA damage repair (26), abnormal regulation of cell cycle and apoptosis-related protein expression (27).

In general, implanting radioactive particles into tumor tissue is supplemented with chemotherapy (28,29). Radiation can induce the apoptosis of tumor cells through activation of the P53 and Bax genes, and inhibition of the Bcl-2 gene (3033). The current results suggest that NaI131 also induces apoptosis by regulating P53, Bax and Bcl-2 expression in A549/DDP cells. However, due to their lack of specificity and strong binding force, radionuclides can cause extensive damage to non-targeted cells. The presence of nuclear particles in the normal tissue surrounding the tumor lesion commonly causes localized and systemic side effects during nuclide particle therapy (13,3436), which limits their value in the clinic. Several studies have found that GA can promote lung cancer cell apoptosis by multiple mechanisms (3740). Furthermore, GA has been reported to act as a sensitizer for chemotherapy. A previous analysis of the ability of GA to sensitize lung cancer for chemotherapy demonstrated positive results (41). According to the results of the isobolographic analysis performed in the present study, we hypothesize that a combination of GA and I131 may have a synergistic effect on the inhibition of A549/DDP cells. Thus, only a low dose of GA combined with I131 is required to significantly increase the rate of apoptosis in A549/DDP cells.

P-gP is a membrane transport protein that can efflux intracellular anticancer agents out of tumor cells, increasing a patient's resistance to antitumor drugs (42). The current results suggest that GA and I131 alone may decrease the expression of P-gP. In addition, compared with GA or I131 treatment alone, I131 combined with low doses of GA was better able to inhibit the expression of P-gP. Application of this treatment strategy may, therefore, further weaken the capacity of tumor cells to efflux intracellular drugs, partially reversing the multidrug resistance of cells.

It is widely accepted that the protein ratio of Bax and Bcl-2 has an important role in apoptosis. Apoptosis is considered to be promoted in cells with high Bax protein expression and inhibited in cells with high Bcl-2 protein expression. Several previous studies have also confirmed the role of both proteins in lung cancer cells (4345). The current results suggest that in A549/DDP cells, I131 and GA alone are able to regulate the protein expression levels of Bcl-2 and Bax. Compared with the groups treated with each agent alone, the protein expression of Bcl-2 was significantly decreased and that of Bax was significantly increased in the I131 combined with GA group. Thus, we hypothesize that a low dose of GA can enhance the ability of I131 to regulate Bcl-2 and Bax expression, which may promote apoptosis in A549/DDP cells.

Other previous studies of lung cancer have found that wild-type P53 can promote tumor cell apoptosis (46,47), increasing the chemosensitivity of tumor cells (48,49). However, mutant P53 can promote chemotherapeutic tolerance in tumor cells via several factors, such as decreased pro-apoptotic capacity, increased DNA repair function (50,51) and upregulated P-gP expression (9,52). It has been confirmed that the wild-type P53 protein cannot be detected by immunohistochemical techniques due to its instability, rapid hydrolysis and short half-life, as well as for other unknown reasons. However, mutant P53 protein can be detected by immunohistochemical methods reasonably well due to its relative stability and longer half-life (50,53). The results of the current immunocytochemical analysis and western blot assay suggest that A549/DDP cells exhibit high P53 protein expression levels, and treatment with GA or I131 alone was able to reduce this expression. In addition, compared with the groups treated with each agent alone, P53 protein expression was further decreased in the I131 combined with GA group, suggesting that the combination of these two compounds may promote apoptosis in A549/DDP cells by regulating the expression of mutant P53.

In conclusion, the current results suggest that the effect of NaI131 on A549/DDP cells may promote apoptosis by increasing P53 and Bax protein expression, while reducing Bcl-2 protein expression. Treatment with NaI131 combined with a low dose of GA was more capable of promoting apoptosis compared with the two monotherapies. Additional protein detection suggested that the protein expression levels of P-gP, P53 and Bcl-2 were markedly decreased and the protein expression of Bax was significantly increased in the combined treatment group compared with each monotherapy group. These results suggest that low-dose GA may enhance the pro-apoptotic role of NaI131 administered to A549/DDP cells in cisplatin-resistant NSCLC. Therefore, low-dose GA may be a sensitizer to NaI131 radiotherapy, since reducing the dosage of NaI131 used in the clinic would reduce the toxic side effects caused by radiotherapy.

Acknowledgements

The present study was supported by the National Natural Science Foundation of China (grant nos. 81372480 and 81202032).

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Chen ZM, Peto R, Iona A, Guo Y, Chen YP, Bian Z, Yang L, Zhang WY, Lu F, Chen JS, et al: Emerging tobacco-related cancer risks in China: A nationwide, prospective study of 0.5 million adults. Cancer 121 Suppl. 17:3097–3106. 2015. View Article : Google Scholar

3 

Wood DE, Kazerooni E, Baum SL, Dransfield MT, Eapen GA, Ettinger DS, Hou L, Jackman DM, Klippenstein D, Kumar R, et al: Lung cancer screening, version 1.2015: Featured updates to the NCCN guidelines. J Natl Compr Canc Netw. 13:23–34. 2015.PubMed/NCBI

4 

Chang A: Chemotherapy, chemoresistance and the changing treatment landscape for NSCLC. Lung Cancer. 71:3–10. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Bowden NA: Nucleotide excision repair: Why is it not used to predict response to platinum-based chemotherapy. Cancer Lett. 346:163–171. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Yamagishi T, Sahni S, Sharp DM, Arvind A, Jansson PJ and Richardson DR: P-glycoprotein mediates drug resistance via a novel mechanism involving lysosomal sequestration. J Biol Chem. 288:31761–31771. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Selivanova G: Wild type p53 reactivation: From lab bench to clinic. FEBS Lett. 588:2628–2638. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Triller N, Korosec P, Kern I, Kosnik M and Debeljak A: Multidrug resistance in small cell lung cancer: Expression of P-glycoprotein, multidrug resistance protein 1 and lung resistance protein in chemo-naive patients and in relapsed disease. Lung Cancer. 54:235–240. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Podolski-Renić A, Jadranin M, Stanković T, Banković J, Stojković S, Chiourea M, Aljančić I, Vajs V, Tešević V, Ruždijić S, et al: Molecular and cytogenetic changes in multi-drug resistant cancer cells and their influence on new compounds testing. Cancer Chemother Pharmacol. 72:683–697. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Chung SK, Zhu S, Xu Y and Fu X: Functional analysis of the acetylation of human p53 in DNA damage responses. Protein Cell. 5:544–551. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Chakraborty S, Mazumdar M, Mukherjee S, Bhattacharjee P, Adhikary A, Manna A, Chakraborty S, Khan P, Sen A and Das T: Restoration of p53/miR-34a regulatory axis decreases survival advantage and ensures Bax-dependent apoptosis of non-small cell lung carcinoma cells. FEBS Lett. 588:549–559. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Jiang YL, Meng N, Wang JJ, Jiang P, Yuan HSh, Liu C, Qu A and Yang RJ: CT-guided iodine-125 seed permanent implantation for recurrent head and neck cancers. Radiat Oncol. 5:682010.PubMed/NCBI

13 

Schroeder T, Kuendgen A, Kayser S, Kröger N, Braulke F, Platzbecker U, Klärner V, Zohren F, Haase D, Stadler M, et al: Therapy-related myeloid neoplasms following treatment with radioiodine. Haematologica. 97:206–212. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Yi T, Yi Z, Cho SG, Luo J, Pandey MK, Aggarwal BB and Liu M: Gambogic acid inhibits angiogenesis and prostate tumor growth by suppressing vascular endothelial growth factor receptor 2 signaling. Cancer Res. 68:1843–1850. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Gu H, You Q, Liu W, Yang Y, Zhao L, Qi Q, Zhao J, Wang J, Lu N, Ling H, et al: Gambogic acid induced tumor cell apoptosis by T lymphocyte activation in H22 transplanted mice. Int Immunopharmacol. 8:1493–1502. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Wang LH, Li Y, Yang SN, Wang FY, Hou Y, Cui W, Chen K, Cao Q, Wang S, Zhang TY, et al: Gambogic acid synergistically potentiates cisplatin-induced apoptosis in non-small-cell lung cancer through suppressing NF-κB and MAPK/HO-1 signalling. Br J Cancer. 110:341–352. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Li C, Qi Q, Lu N, Dai Q, Li F, Wang X, You Q and Guo Q: Gambogic acid promotes apoptosis and resistance to metastatic potential in MDA-MB-231 human breast carcinoma cells. Biochem Cell Biol. 90:718–730. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Qin Y, Meng L, Hu C, Duan W, Zuo Z, Lin L, Zhang X and Ding J: Gambogic acid inhibits the catalytic activity of human topoisomerase IIalpha by binding to its ATPase domain. Mol Cancer Ther. 6:2429–2440. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Li D, Yang H, Li R, Wang Y, Wang W, Li D, Ma S and Zhang X: Antitumor activity of gambogic acid on NCI-H1993 xenografts via MET signaling pathway downregulation. Oncol Lett. 10:2802–2806. 2015.PubMed/NCBI

20 

Wen C, Huang L, Chen J, Lin M, Li W, Lu B, Rutnam ZJ, Iwamoto A, Wang Z, Yang X and Liu H: Gambogic acid inhibits growth, induces apoptosis, and overcomes drug resistance in human colorectal cancer cells. Int J Oncol. 47:1663–1671. 2015.PubMed/NCBI

21 

Jang JH, Kim JY, Sung EG, Kim EA and Lee TJ: Gambogic acid induces apoptosis and sensitizes TRAIL-mediated apoptosis through downregulation of cFLIPL in renal carcinoma Caki cells. Int J Oncol. 48:376–384. 2016.PubMed/NCBI

22 

Wang X, Deng R, Lu Y, Xu Q, Yan M, Ye D and Chen W: Gambogic acid as a non-competitive inhibitor of ATP-binding cassette transporter B1 reverses the multidrug resistance of human epithelial cancers by promoting ATP-binding cassette transporter B1 protein degradation. Basic Clin Pharmacol Toxicol. 112:25–33. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Wellman PJ, Tow S and McMahon L: Isobolographic assessment of the effects of combinations of phenylpropanolamine and fenfluramine on food intake in rats. Pharmacol Biochem Behav. 50:287–291. 1995. View Article : Google Scholar : PubMed/NCBI

24 

Gessner PK: Isobolographic analysis of interactions: An update on applications and utility. Toxicology. 105:161–179. 1995. View Article : Google Scholar : PubMed/NCBI

25 

Chiou JF, Liang JA, Hsu WH, Wang JJ, Ho ST and Kao A: Comparing the relationship of Taxol-based chemotherapy response with P-glycoprotein and lung resistance-related protein expression in non-small cell lung cancer. Lung. 181:267–273. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Oliver TG, Mercer KL, Sayles LC, Burke JR, Mendus D, Lovejoy KS, Cheng MH, Subramanian A, Mu D, Powers S, et al: Chronic cisplatin treatment promotes enhanced damage repair and tumor progression in a mouse model of lung cancer. Genes Dev. 24:837–852. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Cetintas VB, Kucukaslan AS, Kosova B, Tetik A, Selvi N, Cok G, Gunduz C and Eroglu Z: Cisplatin resistance induced by decreased apoptotic activity in non-small-cell lung cancer cell lines. Cell Biol Int. 36:261–265. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Kroger LA, DeNardo GL, Gumerlock PH, Xiong CY, Winthrop MD, Shi XB, Mack PC, Leshchinsky T and DeNardo SJ: Apoptosis-related gene and protein expression in human lymphoma xenografts (Raji) after low dose rate radiation using 67Cu-2IT-BAT-Lym-1 radioimmunotherapy. Cancer Biother Radiopharm. 16:213–225. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Kemény-Beke A, Berényi E, Facskó A, Damjanovich J, Horváth A, Bodnár A, Berta A and Aradi J: Antiproliferative effect of 4-thiouridylate on OCM-1 uveal melanoma cells. Eur J Ophthalmol. 16:680–685. 2006.PubMed/NCBI

30 

Szostak MJ, Kaur P, Amin P, Jacobs SC and Kyprianou N: Apoptosis and bcl-2 expression in prostate cancer: Significance in clinical outcome after brachytherapy. J Urol. 165:2126–2130. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Oka K, Suzuki Y, Iida H and Nakano T: Radiation therapy induces the p53 (+) p21 (−) expression in squamous cell carcinomas of the uterine cervix. Gynecol Oncol. 93:340–344. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Brantley MA Jr, Worley L and Harbour JW: Altered expression of Rb and p53 in uveal melanomas following plaque radiotherapy. Am J Ophthalmol. 133:242–248. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Harima Y, Nagata K, Harima K, Oka A, Ostapenko VV, Shikata N, Ohnishi T and Tanaka Y: Bax and Bcl-2 protein expression following radiation therapy versus radiation plus thermoradiotherapy in stage IIIB cervical carcinoma. Cancer. 88:132–138. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Nakada K, Ishibashi T, Takei T, Hirata K, Shinohara K, Katoh S, Zhao S, Tamaki N, Noguchi Y and Noguchi S: Does lemon candy decrease salivary gland damage after radioiodine therapy for thyroid cancer? J Nucl Med. 46:261–266. 2005.PubMed/NCBI

35 

Fallahi B, Adabi K, Majidi M, Fard-Esfahani A, Heshmat R, Larijani B and Haghpanah V: Incidence of second primary malignancies during a long-term surveillance of patients with differentiated thyroid carcinoma in relation to radioiodine treatment. Clin Nucl Med. 36:277–282. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Iyer NG, Morris LG, Tuttle RM, Shaha AR and Ganly I: Rising incidence of second cancers in patients with low-risk (T1N0) thyroid cancer who receive radioactive iodine therapy. Cancer. 117:4439–4446. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Li Q, Cheng H, Zhu G, Yang L, Zhou A, Wang X, Fang N, Xia L, Su J, Wang M, et al: Gambogenic acid inhibits proliferation of A549 cells through apoptosis-inducing and cell cycle arresting. Biol Pharm Bull. 33:415–420. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Qi Q, Lu N, Li C, Zhao J, Liu W, You Q and Guo Q: Involvement of RECK in gambogic acid induced anti-invasive effect in A549 human lung carcinoma cells. Mol Carcinog. 54:(Suppl 1). E13–E25. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Zhu X, Zhang H, Lin Y, Chen P, Min J, Wang Z, Xiao W and Chen B: Mechanisms of gambogic acid-induced apoptosis in non-small cell lung cancer cells in relation to transferrin receptors. J Chemother. 21:666–672. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Wu ZQ, Guo QL, You QD, Zhao L and Gu HY: Gambogic acid inhibits proliferation of human lung carcinoma SPC-A1 cells in vivo and in vitro and represses telomerase activity and telomerase reverse transcriptase mRNA expression in the cells. Biol Pharm Bull. 27:1769–1774. 2004. View Article : Google Scholar : PubMed/NCBI

41 

Wang LH, Yang JY, Yang SN, Li Y, Ping GF, Hou Y, Cui W, Wang ZZ, Xiao W and Wu CF: Suppression of NF-κB signaling and P-glycoprotein function by gambogic acid synergistically potentiates adriamycin-induced apoptosis in lung cancer. Curr Cancer Drug Targets. 14:91–103. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Sharom FJ: Complex interplay between the P-glycoprotein multidrug efflux pump and the membrane: Its role in modulating protein function. Front Oncol. 4:412014. View Article : Google Scholar : PubMed/NCBI

43 

Bai L, Chen J, McEachern D, Liu L, Zhou H, Aguilar A and Wang S: BM-1197: A novel and specific Bcl-2/Bcl-xL inhibitor inducing complete and long-lasting tumor regression in vivo. PLoS One. 9:e994042014. View Article : Google Scholar : PubMed/NCBI

44 

Yamaguchi M: The anti-apoptotic effect of regucalcin is mediated through multisignaling pathways. Apoptosis. 18:1145–1153. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Mai CW, Yaeghoobi M, Abd-Rahman N, Kang YB and Pichika MR: Chalcones with electron-withdrawing and electron-donating substituents: Anticancer activity against TRAIL resistant cancer cells, structure-activity relationship analysis and regulation of apoptotic proteins. Eur J Med Chem. 77:378–387. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Rohr UP, Wulf MA, Stahn S, Heyd F, Steidl U, Fenk R, Opalka B, Pitschke G, Prisack HB, Bojar H, et al: Non-small lung cancer cells are prime targets for p53 gene transfer mediated by a recombinant adeno-associated virus type-2 vector. Cancer Gene Ther. 10:898–906. 2003. View Article : Google Scholar : PubMed/NCBI

47 

Cuddihy AR, Jalali F, Coackley C and Bristow RG: WTp53 induction does not override MTp53 chemoresistance and radioresistance due to gain-of-function in lung cancer cells. Mol Cancer Ther. 7:980–992. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Lai SL, Perng RP and Hwang J: p53 gene status modulates the chemosensitivity of non-small cell lung cancer cells. J Biomed Sci. 7:64–70. 2000. View Article : Google Scholar : PubMed/NCBI

49 

Osaki S, Nakanishi Y, Takayama K, Pei XH, Ueno H and Hara N: Alteration of drug chemosensitivity caused by the adenovirus-mediated transfer of the wild-type p53 gene in human lung cancer cells. Cancer Gene Ther. 7:300–307. 2000. View Article : Google Scholar : PubMed/NCBI

50 

Brattström D, Bergqvist M, Lamberg K, Kraaz W, Scheibenflug L, Gustafsson G, Inganäs M, Wagenius G and Brodin O: Complete sequence of p53 gene in 20 patients with lung cancer: Comparison with chemosensitivity and immunohistochemistry. Med Oncol. 15:255–261. 1998. View Article : Google Scholar : PubMed/NCBI

51 

Blandino G, Levine AJ and Oren M: Mutant p53 gain of function: Differential effects of different p53 mutants on resistance of cultured cells to chemotherapy. Oncogene. 18:477–485. 1999. View Article : Google Scholar : PubMed/NCBI

52 

Gu J, Tang Y, Liu Y, Guo H, Wang Y, Cai L, Li Y and Wang B: Murine double minute 2 siRNA and wild-type p53 gene therapy enhances sensitivity of the SKOV3/DDP ovarian cancer cell line to cisplatin chemotherapy in vitro and in vivo. Cancer Lett. 343:200–209. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Levine AJ, Momand J and Finlay CA: The p53 tumour suppressor gene. Nature. 351:453–456. 1991. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2017
Volume 13 Issue 1

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
Huang J, Zhu X, Wang H, Han S, Liu L, Xie Y, Chen D, Zhang Q, Zhang L, Hu Y, Hu Y, et al: Role of gambogic acid and NaI131 in A549/DDP cells. Oncol Lett 13: 37-44, 2017
APA
Huang, J., Zhu, X., Wang, H., Han, S., Liu, L., Xie, Y. ... Hu, Y. (2017). Role of gambogic acid and NaI131 in A549/DDP cells. Oncology Letters, 13, 37-44. https://doi.org/10.3892/ol.2016.5435
MLA
Huang, J., Zhu, X., Wang, H., Han, S., Liu, L., Xie, Y., Chen, D., Zhang, Q., Zhang, L., Hu, Y."Role of gambogic acid and NaI131 in A549/DDP cells". Oncology Letters 13.1 (2017): 37-44.
Chicago
Huang, J., Zhu, X., Wang, H., Han, S., Liu, L., Xie, Y., Chen, D., Zhang, Q., Zhang, L., Hu, Y."Role of gambogic acid and NaI131 in A549/DDP cells". Oncology Letters 13, no. 1 (2017): 37-44. https://doi.org/10.3892/ol.2016.5435