Curcumin inhibits the development of non‑small cell lung cancer by inhibiting autophagy and apoptosis

  • Authors:
    • Aili Wang
    • Jinxiang Wang
    • Shuming Zhang
    • Hongxia Zhang
    • Zhenyang Xu
    • Xiaohui Li
  • View Affiliations

  • Published online on: September 21, 2017     https://doi.org/10.3892/etm.2017.5172
  • Pages: 5075-5080
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Among patients with primary lung cancer, 75‑80% present with non‑small cell lung cancer (NSCLC). However, there is a lack of studies into the potential preventive effects of curcumin against the activation of autophagy in NSCLC. Therefore, the present study primarily focused on the protective role of curcumin in NSCLC. It was demonstrated that curcumin decreased the viability of the human lung cancer cells lines, A549 and H1299, in a time‑and dose‑dependent manner (P<0.05). Treatment with curcumin also suppressed the colony formation capacities of A549 and H1299 cells. Following incubation with 10 µM curcumin for 48 h, cell apoptosis was significantly increased by 2.35‑ and 3.02‑fold in A549 and H1299 cells, respectively, when compared with controls (P<0.01). Furthermore, curcumin treatment markedly increased the number and volume of autophagosomes in A549 and H1299 cells when compared with controls. Treatment with 10 µM curcumin for 48 h also significantly reduced the phosphorylation levels of mechanistic target of rapamycin (mTOR), ribosomal protein S6, phosphoinositide 3‑kinase and AKT (protein kinase B) in A549 and H1299 cells (P<0.05). These data indicated that curcumin enhanced autophagy and apoptosis in NSCLC cells by acting as an mTOR complex1/2 inhibitor.

Introduction

As the most common cause of cancer-related mortality, lung cancer is divided into small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) (1). It is estimated that 75–80% of patients with primary lung cancer present with NSCLC (2,3). At present, surgery is the most effective treatment method for NSCLC. However, the postoperative prognosis remains poor and the 5-year survival rate for NSCLC is reported to be <20% (4). Thus, it is necessary to develop novel prevention and treatment strategies that effectively manage NSCLC.

More recently, autophagy has become established as an important cellular process involved in the development of NSCLC (5,6). Autophagy primarily refers to degradation of cytoplasmic content, superfluous or damaged organelles and engulfment of pathogens (79). The mechanism by which autophagy regulates cancer is complex and principally depends on the tumor type and stage (10). Under homeostatic conditions, autophagy generally suppresses the progression of cancer, and abnormal activation of autophagy may lead to malignancy, as observed for NSCLC (1113). Therefore, insight into the regulation of homeostasis may aid to identify effective therapeutic methods that prevent the progression of NSCLC.

Curcumin is a natural polyphenol that is extracted from the spice turmeric, and is characterized by anti-inflammatory, anti-oxidative, anti-carcinogenic and immuno-regulatory activities (14). In mice, it has been observed that treatment with curcumin decreased Helicobacter pylori infection and suppressed infection-induced gastric damage (15). In a clinical trial, an average curcumin dose of 500 mg for 7 days markedly decreased the level of serum lipid peroxide, as a biomarker of oxidative stress (16). Furthermore, an enhanced therapeutic effect against cancer has been observed when curcumin was used alone or in combination with chemotherapeutic agents (17,18). However, to the best of our knowledge, no previous studies have investigated the potential protective effects of curcumin against the activation of autophagy in NSCLC. This was the focus of the present study.

The present study explored the effects of curcumin on the proliferation and migration of lung cancer cells. The findings indicated that curcumin reduced cell growth and suppressed colony formation capacity in human NSCLC cells. Furthermore, abnormal activation of autophagy was inhibited following curcumin treatment, indicating that curcumin may be a useful anticancer agent for the treatment of NSCLC.

Materials and methods

Lung cancer cell lines and cell culture

The human lung cancer cells lines, A549 and H1299 (American Type Culture Collection, Manassas, VA, USA) were cultured in RPMI-1640 medium and Dulbecco's modified Eagle's medium (DMEM) both supplemented with 10% fetal bovine serum (FBS) (all from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA), respectively. The cells were incubated under humidified conditions at 37°C and 5% CO2.

Cell proliferation assay

Curcumin was purchased from Cayman Chemical Company (Ann Arbor, MI, USA). To determine the effects of curcumin on cell proliferation, A549 and H1299 cells were seeded into 96-well tissue culture plates at a density of 5×103 cells/well. Cells were administered with medium only (containing 0.01% dimethyl sulfoxide as a negative control) or incubated with 0.5, 1, 5, 10 and 20 µM curcumin. Following incubation for 24, 48 and 72 h at 37°C, cell viability was determined with an 3-(4,5-dimethylthiazol-2-yl)-2,5-di-phenyltetrazolium bromide (MTT) assay (Sigma-Aldrich; Merck KGaA; Darmstadt, Germany). Following treatment, the cells were cultured in fresh media containing 0.5 mg/ml MTT for 4 h. Dimethyl sulfoxide (DMSO; Sigma-Aldrich; Merck KGaA) was then added to the wells to dissolve the formazan products and the absorbance was measured spectrophotometrically at a wavelength of 550 nm. Three replicates were performed and analyzed.

Cell apoptosis assay

Following 10 µM curcumin or DMSO control treatment for 48 h, the cells were washed three times with cold phosphate-buffered saline (PBS). An Annexin V-fluorescein isothiocyanate (FITC)-propidium iodide (PI) Apoptosis kit (Invitrogen; Thermo Fisher Scientific, Inc.) was used to measure cell apoptosis. Briefly, cells were washed three times with 1X PBS and suspended at a density of 2–3×106 cells/ml in 1X Annexin V-binding buffer (10 mM HEPES/NaOH, pH 7.4, 140 mM NaCl, 2.5 mM CaCl2). Annexin V-FITC and PI buffer was administered to the cells, which were then incubated for 15 min at room temperature in the dark. Cells lacking treatment with curcumin were used as an internal control. Following incubation, the cells were filtered with a 200-mesh filter screen and analyzed with a FACScan flow cytometer (BD Biosciences, San Jose, CA, USA) within 1 h of staining. Cell apoptosis was analyzed using BD CellQuest Pro software (BD Biosciences). A total of 10,000 cells were evaluated in each sample.

Western blotting

Following 10 µM curcumin or DMSO control treatment for 48 h, cell protein was extracted using radioimmunoprecipitation assay buffer (Solarbio Science & Technology Co., Ltd., Beijing, China) and was collected after centrifugation at 10,000 × g at 4°C for 20 min. A bicinchoninic protein assay kit (Pierce; Thermo Fisher Scientific, Inc.) was used to determine the protein concentration. A total of 15 µg protein was loaded per lane and separated by 10% SDS-PAGE and transferred to polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). The membranes were blocked with 8% nonfat dry milk at 4°C overnight. Following three washes with PBS with Tween-20 (5 min/wash), the membranes were incubated with primary antibodies at 4°C overnight. The blots were then incubated with horseradish peroxidase (HRP)-conjugated anti-immunoglobulin G (all 1:5,000; Zhongshan Gold Bridge Biological Technology Co., Beijing, China) for 2 h at room temperature and then washed. Protien detection was performed with enhanced chemiluminescent substrate (EMD Millipore). Primary antibodies against microtubule-associated protein 1 light chain 3 II/I (LC3II/I; L8918, 1:1,000, Sigma-Aldrich; Merck KGaA), beclin-1 (cat no. 3495; 1:1,000), mechanistic target of rapamycin (mTOR; cat no. 2983; 1:1,000), phosphorylated (p)-mTOR (cat no. 5536; 1:1,000), ribosomal protein S6 (cat no. 2217; 1:1,000), p-S6 (cat no. 4858; 1:1,000), phosphoinositide 3-kinase (PI3K; cat no. 4249; 1:1,000), p-PI3K (cat no. 4228; 1:1,000), AKT (protein kinase B; cat no. 9840; 1:1,000), p-AKT (cat no. 8200; 1:1,000) and β-actin (cat no. 4970; 1:1,000) (all from Cell Signaling Technology, Inc., Boston, MA, USA). β-actin was used as an internal control. ImageJ software (National Institutes of Health, Bethesda, MD, USA) was used for density analysis.

Colony formation assay

Cells were suspended in 0.3% agar (Sigma-Aldrich; Merck KGaA) in RPMI-1640 or DMEM with or without 10 µM curcumin treatment for 48 h and plated at a density of 1×105 cells/dish into a 10-cm dish, which was preloaded with a thin layer of 1.0% agar. Cells were maintained in RPMI-1640 or DMEM supplemented with 10% FBS during the assay and monitored for colony formation. After culturing for 7 days, the colony formation was observed. The clones were stained with trypan blue (Sigma-Aldrich; Merck KGaA) at room temperature for 15 min to evaluate colony formation.

Electron microscopy

Following 10 µM curcumin or DMSO control treatment for 48 h, cells were centrifuged at 800 × g for 10 min at room temperature and the cell pellets were fixed at room temperature in 2.3% glutaraldehyde for 1 h, postfixed in 2% osmium tetroxide (O5500; Sigma-Aldrich; Merck KGaA) at 4°C for 30 min and 0.5% uranyl acetate (EMD Millipore) at room temperature for 15 min, dehydrated and embedded in Spurr epoxy resin (Shanghai Huake Co., Ltd., Shanghai, China) at 4°C overnight. Ultrathin sections (90 nm) were cut and double-stained with 3% uranyl acetate and lead citrate (Solarbio, Science & Technology Co., Ltd.) at room temperature for 20 min, and viewed with a Philips CM10 transmission electron microscope (Phillips Electronics, Amsterdam, The Netherlands).

Statistical analysis

Data are presented as the mean ± standard deviation for the indicated number of separate experiments. Three independent experiments were performed for each study. Data were statistically evaluated with an unpaired Student's t-test using GraphPad Prism software (version 6.0; GraphPad Software, Inc., LaJolla, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Curcumin decreases human lung cancer cell viability in a time- and dose-dependent manner

A549 and H1299 cells were treated with 0.5, 1, 5, 10 and 20 µM curcumin for 48 h and cell viability was determined using an MTT assay. As depicted in Fig. 1A and B, treatment with 5, 10 and 20 µM curcumin significantly suppressed cell proliferation when compared with control cells (for A549 cells: P<0.05 for 5 µM curcumin and P<0.01 for 10 and 20 µM curcumin; for H1299 cells: P<0.05 for 5 and 10 µM curcumin and P<0.01 for 20 µM curcumin). Furthermore, when A549 and H1299 cells were incubated with 10 µM curcumin for 24, 48, and 72 h, cell proliferation was significantly reduced by 27 and 36% for A549 cells, and 24 and 32% for H1299 cells at 48 and 72 h, respectively (Fig. 1C and D). These data suggested that curcumin decreased the viability of A549 and H1299 cells in a time- and dose-dependent manner.

Curcumin inhibits colony formation and promotes apoptosis

A549 and H1299 cells were subsequently treated with 10 µM curcumin for 48 h and the colony formation capacity of cells was determined. As depicted in Fig. 2A, treatment with curcumin suppressed the colony formation capacities of both A549 and H1299 cells. Cell apoptosis was also measured using flow cytometry. Following incubation with 10 µM curcumin for 48 h, cell apoptosis was significantly increased by 2.35- and 3.02-fold in A549 and H1299 cells, respectively, when compared with controls (P<0.01; Fig. 2B).

Curcumin enhances autophagy in human lung cancer cells

It was also determined whether curcumin induced cell autophagy in lung cancer cells. Following treatment with 10 µM curcumin for 48 h, western bolt analysis demonstrated that the ratio of LC3-II to LC3-I and protein level of beclin-1 were significantly increased in A549 and H1299 cells (P<0.05; Fig. 3A). Electronic microphotography was also used to assess the autophagosomes in each group. As depicted in Fig. 3B, curcumin markedly increased the number and volume of autophagosomes in curcumin-treated cells when compared with controls, indicating that curcumin may induce cell autophagy in human lung cancer cells.

Curcumin suppresses PI3K/mTOR activation

It has been documented that P13K/mTOR signaling serves a key inhibitory role in the autophagy of various types of human cancer (19). Therefore, the activation of P13K/mTOR signaling following curcumin treatment was evaluated. As depicted in Fig. 4, treatment with 10 µM curcumin for 48 h markedly reduced the phosphorylation levels of mTOR, S6, PI3K and AKT in A549 (all P<0.05) and H1299 (P<0.05 for S6 and AKT, P<0.01 for mTOR and PI3K) cells.

Discussion

Lung cancer has been reported as the most common cause of cancer-related mortality (20). Therefore, detailed studies into the oncological mechanisms of lung cancer are required for the development of improved therapeutics. The majority of lung carcinomas present as NSCLC (20). In signaling pathways related to cell growth and proliferation, the PI3K/AKT/mTOR pathway serves a key role (21,22). Aberrant regulation of the PI3K/AKT/mTOR pathway has been identified in various types of tumor, including prostate, breast, lung and liver cancer (22,23). Recent studies have investigated the potential of mTOR targeting as a molecular-targeting therapy for the treatment of human cancers (24,25).

The present study focused on curcumin, as a natural polyphenol isolated from the spice turmeric (26). Previous studies have demonstrated that curcumin suppresses cytotoxicity and DNA injury by suppressing oxidative damage caused by reactive oxygen species (27,28). The current study observed that curcumin decreased the viability of human lung cancer cells in an apparent dose- and time-dependent manner. Furthermore, cell apoptosis was increased and colony formation capacity was inhibited by curcumin treatment, as determined by annexin V-PI staining and evaluation of colony numbers, respectively. These results indicated that curcumin reduced the viability of lung cancer cells by promoting cell apoptosis and reducing colony formation capacity.

Autophagy is a complex process and is under fine regulation, which depends upon the physiological and pathological conditions of the cellular environment (29). As such, autophagy has become established as a potential anti-cancer therapeutic (24). The process of autophagy may stimulate the degradation of cytoplasmic content in the lysosomal compartment of cells at a cellular level (30). The maintenance of autophagy at a steady level is a possible therapeutic target for anti-cancer therapy. In the process of autophagy, the conversion of LC3-I into LC3-II form is regarded as a hallmark of autophagy, and prompts the formation of the autophagosome (29). In the present study, it was observed that curcumin enhanced autophagy in lung cancer cells, potentially by acting as an mTOR complex 1/2 inhibitor.

In conclusion, the present findings demonstrated novel data that curcumin suppressed mTOR/PI3K/AKT signaling, thereby inducing lung cancer cell apoptosis and autophagy. Thus, curcumin is a potential therapeutic for the treatment of human NSCLC. However, the present study lacks in vivo experiments and further clinical investigation is required.

References

1 

Clark PA: Puberty: When it comes too soon guidelines for the evaluation of sexual precocity. J Ky Med Assoc. 96:440–447. 1998.PubMed/NCBI

2 

Lv X, Liu F, Shang Y and Chen SZ: Honokiol exhibits enhanced antitumor effects with chloroquine by inducing cell death and inhibiting autophagy in human non-small cell lung cancer cells. Oncol Rep. 34:1289–1300. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Zhao R, Chen M, Jiang Z, Zhao F, Xi B, Zhang X, Fu H and Zhou K: Platycodin-D induced autophagy in non-small cell lung cancer cells via PI3K/Akt/mTOR and MAPK signaling pathways. J Cancer. 6:623–631. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Lu W, Zhang H, Niu Y, Wu Y, Sun W, Li H, Kong J, Ding K, Shen HM, Wu H, et al: Long non-coding RNA linc00673 regulated non-small cell lung cancer proliferation, migration, invasion and epithelial mesenchymal transition by sponging miR-150-5p. Mol Cancer. 16:1182017. View Article : Google Scholar : PubMed/NCBI

5 

Choi JY, Hong WG, Cho JH, Kim EM, Kim J, Jung CH, Hwang SG, Um HD and Park JK: Podophyllotoxin acetate triggers anticancer effects against non-small cell lung cancer cells by promoting cell death via cell cycle arrest, ER stress and autophagy. Int J Oncol. 47:1257–1265. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Giatromanolaki A, Kalamida D, Sivridis E, Karagounis IV, Gatter KC, Harris AL and Koukourakis MI: Increased expression of transcription factor EB (TFEB) is associated with autophagy, migratory phenotype and poor prognosis in non-small cell lung cancer. Lung Cancer. 90:98–105. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Hwang KE, Kim YS, Jung JW, Kwon SJ, Park DS, Cha BK, Oh SH, Yoon KH, Jeong ET and Kim HR: Inhibition of autophagy potentiates pemetrexed and simvastatin-induced apoptotic cell death in malignant mesothelioma and non-small cell lung cancer cells. Oncotarget. 6:29482–29496. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Izdebska M, Klimaszewska-Wiśniewska A, Hałas M, Gagat M and Grzanka A: Green tea extract induces protective autophagy in A549 non-small lung cancer cell line. Postepy Hig Med Dosw (Online). 69:1478–1484. 2015.PubMed/NCBI

9 

Lee JG, Shin JH, Shim HS, Lee CY, Kim DJ, Kim YS and Chung KY: Autophagy contributes to the chemo-resistance of non-small cell lung cancer in hypoxic conditions. Respir Res. 16:1382015. View Article : Google Scholar : PubMed/NCBI

10 

Liu JT, Li WC, Gao S, Wang F, Li XQ, Yu HQ, Fan LL, Wei W, Wang H and Sun GP: Autophagy inhibition overcomes the antagonistic effect between gefitinib and cisplatin in epidermal growth factor receptor mutant non-small-cell lung cancer cells. Clin Lung Cancer. 16:e55–e66. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Wei J, Ma Z, Li Y, Zhao B, Wang D and Jin Y and Jin Y: miR-143 inhibits cell proliferation by targeting autophagy-related 2B in non-small cell lung cancer H1299 cells. Mol Med Rep. 11:571–576. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Xu JH, Yang HP, Zhou XD, Wang HJ, Gong L and Tang CL: Autophagy accompanied with bisdemethoxycurcumin-induced apoptosis in non-small cell lung cancer cells. Biomed Environ Sci. 28:105–115. 2015.PubMed/NCBI

13 

Zhang L, Dai F, Sheng PL, Chen ZQ, Xu QP and Guo YQ: Resveratrol analogue 3,4,4′-trihydroxy-trans-stilbene induces apoptosis and autophagy in human non-small-cell lung cancer cells in vitro. Acta Pharmacol Sin. 36:1256–1265. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Sankar P, Telang AG, Ramya K, Vijayakaran K, Kesavan M and Sarkar SN: Protective action of curcumin and nano-curcumin against arsenic-induced genotoxicity in rats in vivo. Mol Biol Rep. 41:7413–7422. 2014. View Article : Google Scholar : PubMed/NCBI

15 

De R, Kundu P, Swarnakar S, Ramamurthy T, Chowdhury A, Nair GB and Mukhopadhyay AK: Antimicrobial activity of curcumin against Helicobacter pylori isolates from India and during infections in mice. Antimicrob Agents Chemother. 53:1592–1597. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Soni KB and Kuttan R: Effect of oral curcumin administration on serum peroxides and cholesterol levels in human volunteers. Indian J Physiol Pharmacol. 36:273–275. 1992.PubMed/NCBI

17 

Betts JW, Sharili AS, La Ragione RM and Wareham DW: In vitro antibacterial activity of curcumin-polymyxin B combinations against multidrug-resistant bacteria associated with traumatic wound infections. J Nat Prod. 79:1702–1706. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Zhu DJ, Huang YF, Chen XW, Luo ZT, Wang GX, Liu CC, Zhang WJ and Ouyang MZ: Curcumin partly ameliorates irinotecan-induced diarrhea and synergistically promotes apoptosis in colorectal cancer through mediating oxidative stress. Oncotarget. Jul 14–2016.(Epub ahead of print).

19 

Bjelogrlić SK, Srdić T and Radulović S: Mammalian target of rapamycin is a promising target for novel therapeutic strategy against cancer. J BUON. 11:267–276. 2006.PubMed/NCBI

20 

Micke P, Mattsson JS, Djureinovic D, Nodin B, Jirström K, Tran L, Jönsson P, Planck M, Botling J and Brunnström H: The impact of the fourth edition of the WHO classification of lung tumours on histological classification of resected pulmonary NSCCs. J Thorac Oncol. 11:862–872. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Laplante M and Sabatini DM: mTOR signaling in growth control and disease. Cell. 149:274–293. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Efeyan A and Sabatini DM: mTOR and cancer: Many loops in one pathway. Curr Opin Cell Biol. 22:169–176. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Salmena L, Carracedo A and Pandolfi PP: Tenets of PTEN tumor suppression. Cell. 133:403–414. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Benjamin D, Colombi M, Moroni C and Hall MN: Rapamycin passes the torch: A new generation of mTOR inhibitors. Nat Rev Drug Discov. 10:868–880. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Baehrecke EH: Autophagy: Dual roles in life and death? Nat Rev Mol Cell Biol. 6:505–510. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Shi J, Wang Y, Jia Z, Gao Y, Zhao C and Yao Y: Curcumin inhibits bladder cancer progression via regulation of b-catenin expression. Tumour Biol. 39:10104283177025482017. View Article : Google Scholar : PubMed/NCBI

27 

Granados-Castro LF, Rodríguez-Rangel DS, Fernández-Rojas B, León-Contreras JC, Hernández-Pando R, Medina-Campos ON, Eugenio-Pérez D, Pinzón E and Pedraza-Chaverri J: Curcumin prevents paracetamol-induced liver mitochondrial alterations. J Pharm Pharmacol. 68:245–256. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Trujillo J, Molina-Jijón E, Medina-Campos ON, Rodríguez-Muñoz R, Reyes JL, Loredo ML, Barrera-Oviedo D, Pinzón E, Rodríguez-Rangel DS and Pedraza-Chaverri J: Curcumin prevents cisplatin-induced decrease in the tight and adherens junctions: Relation to oxidative stress. Food Funct. 7:279–293. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T, Noda T, Kominami E, Ohsumi Y and Yoshimori T: LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 19:5720–5728. 2000. View Article : Google Scholar : PubMed/NCBI

30 

Huang S, Shu L, Dilling MB, Easton J, Harwood FC, Ichijo H and Houghton PJ: Sustained activation of the JNK cascade and rapamycin-induced apoptosis are suppressed by p53/p21(Cip1). Mol Cell. 11:1491–1501. 2003. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 14 Issue 5

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
Wang A, Wang J, Zhang S, Zhang H, Xu Z and Li X: Curcumin inhibits the development of non‑small cell lung cancer by inhibiting autophagy and apoptosis. Exp Ther Med 14: 5075-5080, 2017
APA
Wang, A., Wang, J., Zhang, S., Zhang, H., Xu, Z., & Li, X. (2017). Curcumin inhibits the development of non‑small cell lung cancer by inhibiting autophagy and apoptosis. Experimental and Therapeutic Medicine, 14, 5075-5080. https://doi.org/10.3892/etm.2017.5172
MLA
Wang, A., Wang, J., Zhang, S., Zhang, H., Xu, Z., Li, X."Curcumin inhibits the development of non‑small cell lung cancer by inhibiting autophagy and apoptosis". Experimental and Therapeutic Medicine 14.5 (2017): 5075-5080.
Chicago
Wang, A., Wang, J., Zhang, S., Zhang, H., Xu, Z., Li, X."Curcumin inhibits the development of non‑small cell lung cancer by inhibiting autophagy and apoptosis". Experimental and Therapeutic Medicine 14, no. 5 (2017): 5075-5080. https://doi.org/10.3892/etm.2017.5172