
2022 Impact Factor
Lung cancer is the leading cause of cancer-related deaths in the world, comprising small cell lung carcinoma and non-small cell lung carcinoma (NSCLC) which accounts for approximately 85% of lung cancer (Han and Roman, 2010). NSCLC is caused by smoking, secondhand smoke, radon, other toxic substances, and family history (Gridelli
Cisplatin (
Gallic acid (3,4,5-trihydroxybenzoic acid, GA) (Fig. 1) is a naturally occurring polyphenol phytochemical found in nuts, teas and grapes (Badhani
The PI3K/Akt pathway plays a critical role in regulating diverse normal cellular processes. PI3K is activated by various extracellular stimuli such as growth factors, cytokines and hormones,
The object of this study was to confirm the anti-cancer effects and possible molecular mechanisms of GA compared with CDDP in NSCLC cells and tumor xenograft mouse model. These findings suggest the potential of GA as a PI3K/Akt pathway targeted therapeutic agent for NSCLC treatment.
CDDP and GA (Fig. 1) were purchased from Sigma-Aldrich (St. Louis, MO, USA). CDDP was dissolved in sterile phosphate-buffered saline (PBS), and GA was dissolved in dimethyl sulfoxide (DMSO) used as a negative control (NC) or vehicle. The final concentrations of sterile PBS and DMSO were 1.5% and 0.1%, respectively.
The NSCLC cell line A549, and normal human diploid lung fibroblast cell line WI-26, were purchased from the Korean Cell Line Bank (Seoul, Korea). A549 cells were cultured in RPMI-1640 (Gibco, Gaithersburg, MD, USA) supplemented with 10% fetal bovine serum (FBS; RMBIO, Missoula, MT, USA) and 2% Antibiotic- Antimycotic (Gibco), and WI-26 cells were cultured in DMEM (HyClone Laboratories, Chicago, IL, USA) supplemented with 10% FBS (RMBIO), 100 U/mL penicillin G/100 μg/mL (Biowest, San Marcos, TX, USA) and 1% HEPES (Gibco); Both cell lines were maintained at 37°C in a humidified atmosphere of 5% CO2.
A549 and WI-26 cells were seeded at a density of 5×103 cells per well in 96-well culture plates, and incubated for 24 h. Cells were then exposed to varying concentrations of CDDP (0-50 μM) and GA (0-200 μM), and incubated for 48 h. Cell viability was determined using the EZ-Cytox cell viability assay kit (iTSBiO, Seoul, Korea). Briefly, the medium was removed and EZ-Cytox was added to all wells, incubated for 1 h, with subsequent measurement of absorbance of each well at 450 nm using a microplate spectrophotometer (Epoch, BioTek, Winnoski, VT, USA).
A549 cells were seeded at a density of 500 cells per well in 6-well plates, and incubated for 24 h, followed by exposure to NC (0.1% DMSO), CDDP (12 μM) and GA (75 μM) and incubation for 10 days. The medium containing the appropriate chemical was replaced every 3 days. After 10 days, the cells were washed with Dulbecco’s phosphate-buffered saline (DPBS; Welgene, Daegu, Korea), fixed with 4% formaldehyde (Sigma-Aldrich) for 10 min, and permeabilized with methanol (Sigma-Aldrich) for 10 min. The colonies formed by single cells were subsequently stained with 0.5% crystal violet solution and washed with DPBS. The stained colonies were counted by the Image J program (National Institutes of Health, Bethesda, MD, USA).
A549 cells in medium containing NC (0.1% DMSO), CDDP (12 μM) and GA (75 μM) were seeded at 500 cells/25 μL/droplet on the lid of petri dish using an 8-channel pipette, and incubated for 6 days. After 6 days, each droplet was gently collected with medium and transferred to 6-well plates, and photographed using the IX-73 inverted microscope (Olympus, Tokyo, Japan). The size of tumor spheroids was measured by the Image J program (National Institutes of Health).
The Annexin V assay was performed using Alexa Fluor 488 or FITC Annexin V/Dead Cell Apoptosis kit (Invitrogen, Carlsbad, CA, USA). Briefly, A549 cells were seeded in cell culture dishes at a density of 8×104 cells, incubated for 24 h, and subsequently treated with NC (0.1% DMSO), CDDP (12 μM) and GA (75 μM) for 48 h. The cells were then detached with trypsin and centrifuged at 2,000 rpm for 3 min. The collected cells were suspended in 1X annexin-binding buffer and stained with Alexa Fluor 488 or FITC and PI for 15 min at room temperature in the dark. A total of 3×104 stained cells were analyzed by flow cytometry (SH800 Cell sorter, Sony, Tokyo, Japan).
TUNEL assay was performed using the DeadEndTM Fluorometric TUNEL System (Promega, Madison, WI, USA), according to the instructions described in the kit. Briefly, A549 cells were seeded at a density of 3×104 cells per well in 24-well plates and incubated for 24 h. After treatment with NC (0.1% DMSO), CDDP (12 μM) and GA (75 μM) for 48 h, the cells were fixed with 4% paraformaldehyde (Biosesang Inc., Gyeonggi, Korea) for 25 min at 4°C, followed by two washes with DPBS. Cells were permeabilized by adding 0.1% Triton X-100 for 10 min, and subsequently washed with DPBS. Permeabilized cells were then stained with TUNEL mix solution, and the nuclei were counterstained with 4’,6-diamidino-2-phenylindole (DAPI; Invitrogen). DNA fragmentation by CDDP or GA was visualized using the IX-73 inverted microscope (Olympus) and quantified using the Image J program (National Institutes of Health).
After treatment of A549 cells with NC (0.1% DMSO), CDDP (12 μM) and GA (75 μM) for 48 h, the total proteins were extracted using the EzRIPA Lysis Kit (ATTO, Tokyo, Japan) and quantified by bicinchoninic acid (BCA; Sigma-Aldrich), mixed with a copper II sulfate solution (Sigma-Aldrich) at a ratio of 50:1. Total cell proteins (50 μg) were separated in 10% or 15% SDS-PAGE gel and transferred to a polyvinylidene difluoride (PVDF) membrane (Bio-Rad, Hercules, CA, USA). The transferred membrane was then blocked by 5% skim milk (Blotting-Grade Blocker; Bio-Rad) for an hour at 4°C on a shaker. The blocked membrane was then incubated overnight at 4°C on a shaker, with 5% bovine serum albumin (BSA, RMBIO) containing following primary antibodies: mouse monoclonal anti-GAPDH (1:10,000; Abcam, Cambridge, UK), mouse monoclonal anti-Cyclin D1 (1:2,000; Abcam), rabbit monoclonal anti-p27 (1:2,000; Abcam or 1:1000; Santa Cruz Biotechnology, Dallas, TX, USA), mouse monoclonal anti-Cyclin E1 (1:1,000; Santa Cruz Biotechnology), mouse monoclonal anti-p53 (1:1,000; Santa Cruz Biotechnology), mouse monoclonal anti-Bax (1:1,000; Online, Atlanta, GA, USA), mouse monoclonal anti-Bcl-2 (1:500; Bio Legend, San Diego, CA, USA), rabbit polyclonal anti-cleaved caspase-3 (1:2,000; Cell Signaling Technology, Danvers, MA, USA), rabbit monoclonal anti-p-Akt (Ser473, 1:2,000; Cell Signaling Technology), rabbit polyclonal anti-p-Akt (Thr308, 1:1,000; Bioss, Woburn, MA, USA), rabbit polyclonal anti-Akt (1:1,000; Bioss), and rabbit polyclonal anti-PI3K (1:300; Bioss). Primary antibody binding was detected by combining with secondary antibodies with horse radish peroxidase-conjugated anti-mouse IgG or anti-rabbit IgG (1:2,000; Thermo Scientific, Waltham, MA, USA) for 2 h at 4°C on shaker. Finally, the signals indicating expression levels of target proteins were detected by applying the SuperSignalTM West Pico PLUS Chemiluminescent Substrate (Thermo Scientific) and Lumino graph II (ATTO). All protein expression levels were normalized and quantified using CSAnalyzer4 (ATTO).
BALB/c nude mice (males, 4 weeks old) were purchased from Orient Bio (Gyeonggi, Korea). The animal experiment was approved by the Animal Care Committee of Chungbuk National University (Cheongju, Korea) (CBNUA-1284-19-01). The mice were housed in the CBNU Laboratory Animal Research Center at 24°C under a 12 h light-dark cycle, and were acclimated to the environment for 3 weeks before the experiment. A549 cells (3×106 cells/10% FBS) mixed with Matrigel (BD Biosciences, Bedford, MA, USA) in a 1:1 ratio, were subcutaneously (SC) injected into both flanks of each mouse. The A549 cells-derived tumor volume was measured three times a week using a caliper (Mitutoyo, Gyeonggi, Korea), and calculated as 0.5× (length×width2) (mm3). The body weight was also measured at the same time as the tumor size measurement. Once the tumor size reached 100 mm3, vehicle (0.5% DMSO in corn oil; n=7), CDDP (7 mg/kg; n=7) and GA (50 mg/kg; n=7) dissolved in 0.5% DMSO and corn oil (Sigma-Aldrich) were administered by intraperitoneal (IP) injection at 150 μL for 4 weeks (three times a week for vehicle (0.5% DMSO in corn oil) and GA, and once a week for CDDP). The mice were sacrificed after 4 weeks, and the tumors were subsequently harvested from the vehicle (0.5% DMSO in corn oil), GA or CDDP treated mice and fixed in 4% paraformaldehyde. The fixed tumor mass was dehydrated and embedded in paraffin for histological analysis.
Paraffin-embedded xenograft tumors were sliced into 5 μm sections using a microtome (Leica RM2145, Leica Microsystems, Nussloch, Germany) and collected on the microscope slides. The tissue sections were deparaffinized in xylene for 10 min, and gradually rehydrated in 100, 90, 80, and 70% ethanol and gently running tap water, each for 5 min. Next, the sections were stained with hematoxylin (Merck Millipore, Burlington, MA, USA) and eosin (Merck Millipore) and washed in gently running tap water for 5 min. The tissue sections were subsequently dehydrated in 70, 80, 90 and 100% ethanol, followed by soaking in xylene for 5 min, and then mounted using a Cytoseal (Invitrogen). The tumor mass structure was imaged using the IX-73 inverted microscope (Olympus).
Paraffin-embedded xenograft tumors were sliced into 5 μm sections using a microtome (Leica Microsystems) and collected on the microscope slides. After deparaffinization (as stated above), antigen retrieval was accomplished by boiling the tumor section slides with 0.01 M citrate buffer (pH 6.0) for 20 min in a microwave, and subsequent cooling at room temperature. The slides were blocked with 5% BSA plus 10% goat serum in PBS plus Tween-20 for an hour, followed by treatment with 0.3% H2O2 (Sigma-Aldrich) for 30 min. The slides were then incubated overnight at 4°C with the following primary antibodies: mouse monoclonal anti-proliferating cell nuclear antigen (PCNA; 1:500; Santa Cruz Biotechnology), rabbit polyclonal anti-cleaved caspase-3 (1:200), and rabbit monoclonal anti-p-Akt (1:200). Subsequently, the probed slides were incubated with biotinylated mouse or rabbit anti-goat IgG (1:500; Vector Laboratories, Burlingame, CA, USA) secondary antibodies for 30 min at 37°C. The sections were then incubated with avidin-biotin complex (ABC; Vector Laboratories) for 30 min and diaminobenzidine tetrahydrochloride hydrate (DAB; Sigma-Aldrich) for 2 min. Finally, the sections were dehydrated and mounted using a Cytoseal (Invitrogen). The expression of each protein in the tumor mass was observed and visualized using the IX-73 inverted microscope (Olympus).
All
To evaluate the effect of GA or CDDP on cell viability, the water soluble tetrazolium salt assay was performed using the NSCLC cell line A549 and normal human lung fibroblast cell line WI-26. Both cell lines were exposed to stepwise concentrations of CDDP (0-50 μM) and GA (0-200 μM) for 48 h. The culture medium containing 0.1% DMSO was applied as the NC. Although GA and CDDP were dissolved in 0.1% DMSO and 1.5% sterile PBS, respectively, no difference was obtained in cell viability when comparing 0.1% DMSO and 1.5% sterile PBS (data not shown). Cell viability of both cell lines was inhibited in a dose-dependent manner when exposed to CDDP (Fig. 2A). Exposure to GA significantly decreased cell viability of A549 from 50 to 200 μM in a dose-dependent manner. No statistical change was observed at 50 and 100 μM GA, but significant inhibition was observed from 150 to 200 μM following treatment with GA in WI-26 cells (Fig. 2B). For A549 and WI-26 cells, the inhibitory concentration 50 (IC50) values achieved for CDDP were 12.13 and 15.80 μM, respectively, and for GA were 74.19 and 148.1 μM, respectively. Based on these results, 12 μM CDDP and 75 μM GA were used for subsequent experiments.
The cell survival ability after GA or CDDP exposure was assessed
Tumor spheroid formation assay, in which tumor cells aggregate as 3D structures mimicking the
Western blot assay was performed to confirm the effects of GA or CDDP on the expression of cell cycle-related proteins (Fig. 3C). Exposure to GA or CDDP downregulated the expressions of Cyclin D1 and E1 required for progression of the cell cycle, but the expressions of p21 and p27 associated with cell cycle arrest were upregulated by GA or CDDP treatment in A549 cells (Fig. 3D). These results suggested that GA inhibited the cell proliferation by negatively regulating cell cycle progression.
To evaluate the apoptotic effects of GA or CDDP on A549 cells, changes in the distribution of early or late apoptotic cells and DNA fragmentation in late apoptosis in A549 cells treated with GA or CDDP were observed through Annexin V assay and TUNEL assay. Flow cytometric analysis of Annexin V assay revealed significantly increased distribution of apoptotic cells, and decreased number of live cells in A549 exposed to GA or CDDP, indicating that treatment with GA or CDDP induced cell apoptosis (Fig. 4A). TUNEL assay showed that exposure to GA or CDDP caused DNA fragmentation, representing late apoptosis. TUNEL-positive cells (green fluorescence) were increased when treated with GA or CDDP. Since most treated cells were non-viable, very few DAPI-stained cells (blue fluorescence) were attached and observed in the GA or CDDP treatment groups. However, based on the percentage of TUNEL-stained cells to normalization of DAPI-stained cells, GA or CDDP significantly induced DNA fragmentation in cell nuclei (Fig. 4B). These results indicated that GA induced considerable apoptosis of A549 cells, comparable to CDDP.
The expression of apoptosis related proteins after GA or CDDP treatments was detected by Western blot assay (Fig. 4C). Similar to results achieved in the above experiments, upregulation in the expression of p53 (tumor suppressor protein) was observed only in the CDDP treatment group, but the expression of Bax (pro-apoptotic protein) was significantly upregulated, and the expression of Bcl-2 (anti-apoptotic protein) was downregulated in GA and CDDP treatment groups (Fig. 4D). These results indicated that GA induced cell apoptosis
To explore the molecular mechanism of GA-mediated cell cycle arrest and apoptosis compared to CDDP, the expressions of protein associated with PI3K/Akt pathway were examined by Western blot assay (Fig. 5A). Exposure to GA or CDDP significantly decreased the levels of PI3K, and completely inhibited Akt by phosphorylation of Akt at Ser473 and Thr308. No significant difference was observed in the expression of total Akt. These data demonstrated that GA suppressed the PI3K/Akt pathway in A549 cells (Fig. 5B).
To confirm the
The anti-tumor effect of GA was histologically examined in the A549 cells-derived tumor mass of xenograft mice. H&E staining was performed to observe the structure of xenograft tumors from mice groups treated with GA or CDDP. The tumor mass was characterized by disordered arrangement and nest-like distribution. As shown in Fig. 7, Most cells in the tumor mass of the vehicle (0.5% DMSO in corn oil) were arranged with complete and atypical structures. In the GA and CDDP treatment groups, there were incomplete cell membranes and low tumor density, as compared to vehicle (0.5% DMSO in corn oil). IHC was performed to confirm the expressions of PCNA, cleaved caspase-3 and p-Akt in the tumor mass. It was determined that administration of GA or CDDP downregulated the expressions of PCNA and p-Akt, and upregulated the expression of cleaved caspase-3, as compared to the vehicle (0.5% DMSO in corn oil) (Fig. 7). These findings indicated that IP injection of GA inhibited the tumor growth, which was consistent with
GA, as a naturally occurring polyphenol compound, especially exerts its anti-cancer effects by acting on the molecular mechanisms of cell cycle, apoptotic processes, angiogenesis, invasion and metastasis (Verma
GA or CDDP significantly inhibited the viability of A549 cells, a cell line of NSCLC that accounts for 85% of lung cancer. We found that GA dose-dependently affected the viability of human lung cancer cells. The IC50 value for GA was 74.15 μM in A549 cells. This value differs from these previous studies in some cancer cell lines (Moghtaderi
The effects of GA or CDDP on the colony and tumor spheroid forming abilities of A549 cells were confirmed as a series of cell viability assays. Both results were consistent with the results of cell viability assay; exposure to GA or CDDP considerably inhibited the formation of colonies and tumor spheroids of A549 cells. The colony formation assay reflects cell survival and proliferation against the effectiveness of cytotoxic agents (Franken
The above results were ultimately related to cell cycle progression. Western blot assay proved that GA or CDDP perturbed the protein expressions of cell cycle related genes such as Cyclin D1, Cyclin E1, p21 and p27. In A549 cells, GA or CDDP treatment downregulated the expressions of Cyclin D1 and E1. Cyclin D1 and E1 are responsible for the initial and terminal G1 phase, respectively, and these cyclins play a role in G1/S transition (Gao
Induction of apoptosis in A549 cells following GA or CDDP treatment was identified by the Annexin V assay and TUNEL assay. Results of the Annexin V assay revealed that exposure to GA or CDDP increased the distribution of early apoptotic cells and late apoptotic cells, with a reduction in the number of live A549 cells. Along with the increased distribution of apoptotic cells, the TUNEL assay revealed that treatment with GA or CDDP significantly induced DNA fragmentation of A549 lung cancer cells. In accordance with these findings, the levels of apoptosis related proteins were found to be altered after GA or CDDP exposure in A549 cells; the expressions of Bax and cleaved caspase-3 were upregulated, whereas Bcl-2 was downregulated. However, only CDDP, and not GA, upregulated the expression of p53. p53 activation can be accomplished independently of changes in protein levels, indicating that accumulation of p53 is not always necessary, or even required, for the employment of p53 downstream responses. In other words, even if no increase in p53 levels is observed, p53 can be activated by a different manner, in example, post-translationally (Bellamy, 1997). The p53 and Bcl-2 family proteins such as Bcl-2, Bcl-xl, Bax and Bad are known as key regulators of apoptosis. In the intrinsic pathway, p53 shifts the balance of Bcl-2 family proteins which are located in the mitochondria, leading to permeabilization of mitochondria and apoptosis (Hemann and Lowe, 2006; Xu
It was studied to some extent in the previous studies that GA inhibited the growth of cancer cells by inactivating the PI3K/Akt signaling pathway (Zhao and Hu, 2013), which is in line with our results. Also, there are accumulating evidences that the downregulation of phosphorylated Akt plays a critical role in the growth and survival of various cancer cells by inducing apoptosis. It was found that GA affected the cell viability, proliferation, invasion and angiogenesis through the inactivation of PI3K/Akt and Ras/MAPK signaling pathway in human glioma cells (Lu
To further explore the therapeutic potential of GA in lung cancer, a mouse xenograft tumor model was constructed. In agreement with
In conclusion, the findings of the present study determined that GA, a common dietary phenolic compound present in many plant-derived foods, exerted its anti-cancer activities on A549 cells by interrupting cell cycle progression and inducing apoptosis through suppression of the PI3K/Akt pathway (Fig. 8). These results indicated that GA would be an emerging novel and effective therapeutic agent targeting the PI3K/Akt pathway in NSCLC and possibly in other PI3K/Akt-dependent cancers.
This work was supported by the Basic Science Research Program (2020R1A2C2006060) and the Global Research and Development Center (GRDC) Program (2017K1A4A3014959) through the National Research Foundation (NRF) of Korea funded by the Ministry of Science and ICT. In addition, this work was also supported by Korea Institute of Planning and Evaluation for Technology in Food, Agriculture, Forestry and Fisheries (IPET) through Agriculture, Food and Rural Affairs Convergence Technologies Program for Educating Creative Global Leader, funded by Ministry of Agriculture, Food and Rural Affairs (MAFRA; grant number: 320005-4).
The authors do not have any conflicts of interest to declare.
![]() |
![]() |