Profiling of apoptosis- and autophagy-associated molecules in human lung cancer A549 cells in response to cisplatin treatment using stable isotope labeling with amino acids in cell culture

  • Authors:
    • Zongqiang Wang
    • Guifeng Liu
    • Jinlan Jiang
  • View Affiliations

  • Published online on: January 18, 2019     https://doi.org/10.3892/ijo.2019.4690
  • Pages: 1071-1085
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cis‑diammine‑dichloro‑platinum II‑based adjuvant chemotherapy provides an alternative therapy to improve the survival of patients with lung tumors, especially those with non‑small cell lung cancer (NSCLC). However, drug resistance is a large clinical problem and its underlying mechanism remains unclear. In the present study, NSCLC A549 cells were treated with a low concentration of cisplatin in order to observe and determine the development of chemoresistance, via growth curves, colony forming assays and apoptosis assays. Then the induction of autophagy was examined in the cisplatin‑treated A549 cells with a fluorescence reporter. Profiled proteins in the cisplatin‑treated A549 cells were also assessed using the stable isotope labeling by amino acids in cell culture (SILAC) method, and then the differentially expressed molecules were verified. The results demonstrated that A549 cells became less sensitive to cisplatin [resistant A549 cells (A549R)] following 20 passages in the medium containing a low concentration of cisplatin, with less apoptotic cells post‑cisplatin treatment. A549R cells grew more efficiently in the cisplatin medium, with more colony formation and more cells migrating across the baseline. In addition, NSCLC results demonstrated that more autophagy‑related proteins (ATGs) were expressed in the A549R cells. Furthermore, the western blotting results confirmed this upregulation of ATGs in A549R cells. In addition, two repeated SILAC screening experiments recognized 15 proteins [glucose‑regulated protein, 78 kDa (GRP78), heat shock protein 71, pre‑mRNA processing factor 19, polypyrimidine tract binding protein 1, translationally controlled tumor protein, Cathepsin D, Cytochrome c, thioredoxin domain containing 5, MutS homolog (MSH) 6, Annexin A2 (ANXA2), BRCA2 and Cyclin dependent kinase inhibitor 1A interacting protein, MSH2, protein phosphatase 2A 55 kDa regulatory subunit Bα, Rho glyceraldehyde‑3‑phosphate‑dissociation inhibitor 1 and ANXA4] that were upregulated by >1.5‑fold in heavy (H)‑ and light (L)‑labeled A549R cells. In addition, 16 and 14 proteins were downregulated by >1.5‑fold in the H‑ and L‑labeled A549R cells, respectively. The majority of the downregulated proteins were associated with apoptosis. In conclusion, the present study isolated a cisplatin‑resistant human lung cancer A549 cell clone, with reduced apoptosis and high levels of autophagy, in response to cisplatin treatment. In cisplatin‑resistant A549R cells, SILAC proteomics recognized the high expression of GRP78 and other proteins that are associated with anti‑apoptosis and/or autophagy promotion.

Introduction

Lung cancer leads to high levels of cancer morbidity and cancer-associated mortality worldwide (1). Lung cancers are clinically classified as non-small-cell lung cancer (NSCLC; accounts for 80-85% of cases) and small cell lung cancer (SCLC) (2). Surgical resection is the most potentially curative therapeutic modality for this disease. Cis-diammine-dichloro-platinum II-based adjuvant chemotherapy significantly improves the prognosis of patients with advanced NSCLC (3), particularly in those with Stage II-IIIA (4,5). However, innate non-sensitiveness to or acquired resistance to cisplatin is a major challenge in the management of patients with lung cancer (6,7). Therefore, the identification of mechanisms underlining cisplatin chemoresistance in NSCLC is urgently required.

Advances in technology, including DNA sequencing, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and microarray methods, enable the discovery of predictive markers and the identification of significant expression at the transcriptional level of chemoresistance-associated genes (6,7). In particular, the profiling by microarray screening is highly effective in predicting chemotherapeutic sensitivity, with thousands of genes being simultaneously evaluated (8-10). However, the relatively low sensitivity and poor lower thresholds of microarray detection reduce its accuracy; thus, follow-up quantitative methods are required to confirm the results.

Stable isotope labeling by amino acids in cell culture (SILAC) is effective in distinguishing the protein profiling from one group to the other (11-13). Five passages would transform ~97% of 12C-labeled amino acids in A549 cells into 13C-labeled amino acids [1-(1/2)5 = 97%] and thus, cells only contain 'heavy' proteins (11,13). The incorporation of stable isotopes facilitates the quantitative recognition of the differences in expression profiles by tandem mass spectrometry between the 12C- and 13C-labeled A549 cells. SILAC has also been useful in the identification of cancer biomarkers, and chemoresistance-associated biomarkers in hepatocellular carcinoma (14), breast cancer (15) and lung cancer (16,17).

In the present study, a cisplatin-resistant A549 cell clone (A549R) was isolated from A549 cells post-serial passages under cisplatin pressure. The differences in proliferation, apoptosis and autophagy were investigated between A549R and A549 cells under cisplatin treatment. Then the SILAC method was utilized to profile A549R specific proteomics under cisplatin treatment. The results implied that autophagy may be an important mechanism underlining the cisplatin resistance of NSCLC A549 cells.

Materials and methods

Reagents, cell culture, cisplatin-resistant clone selection and treatment

Human NSCLC A549 cells were purchased from American Type Culture Collection (Manassas, VA, USA) and were cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Invitrogen; Thermo Fisher Scientific, Inc.) at 37°C, under 5% CO2. For the selection of cisplatin-resistant clone, A549 (A549R) cells were seeded in 12-well plates (Corning Incorporated, Corning, NY, USA), with <200 cells per well, and were then incubated at 37°C for 3-5 days with 1 µM cisplatin (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). The larger cell colonies were picked and propagated with DMEM + 10% FBS. Another 9 passages of selection were performed via colony forming assays with 1 µM cisplatin treatment, which were followed by a further 10 passages of selection with 2 µM cisplatin treatment. For A549R selection, A549 cells were cultured with 1 µM cisplatin for 5 passages (without selection/purification of larger colonies), and then larger colonies were isolated after each passage for a further 5 passages with 1 µM cisplatin. A similar selection process was performed for the isolation of colonies following treatment with 2 µM cisplatin. For the stability examinations, A549R cells were cultured for an additional 20 passages in DMEM without cisplatin, then the colony forming and growth assays were performed; A549R cells prior to serial passaging were used as the control cells.

For heavy (H)- or light (L)-Lysine labeling experiments, A549 or A549R cells were cultured serially for 5 passages in SILAC™ DMEM (Thermo Fisher Scientific, Inc.) supplemented with 10% FBS without Lysine, which was then respectively supplemented with 13C6H14N2O2-Lysine-HCL (H-labeled) or 12C6H14N2O2-Lysine-HCL (L-labeled). A total of 10 µM cisplatin was added to each group of cells, which were incubated for 24 h at 37°C with 5% CO2 in a T75 cell flask. For SILAC proteomics analysis, ~1×107 H- or L-labeled A549R/A549 cells with 80-90% confluence were collected for further analysis.

L- or H-labeled A549/A549R cells were collected and washed four times with 10 ml ice-cold phosphate-buffered saline (PBS) and counted

A total of 1×107 L- or H-labeled cells were lysed with 0.5% 4-Nonylphenol Ethoxylate (Santa Cruz Biotechnology, Inc., Dallas, TX, USA) containing 1.1 µM pepstatin A (Sigma-Aldrich; Merck KGaA) on ice for 30 min. Nuclei and other organelles were removed following centrifugation at 5,000 × g for 10 min at 4°C. The supernatant protein samples were transferred to fresh tubes and then the protein concentration was quantified with a Bicinchoninic Acid protein assay (Thermo Fisher Scientific, Inc.). For SILAC analysis, the H- and L-labeled protein samples were mixed in a ratio of 1:1; the remaining samples were stored at −80°C prior to subsequent use.

Colony forming, cell proliferation and migration assays

For the colony forming assay, ~200 A549 or A549R cells were seeded in 12-well plates and incubated at 37°C with DMEM containing 0 or 10 µM cisplatin for 3-5 days. The cell colonies were stained at room temperature with 0.005% crystal violet for 10 min and observed with a UVP imaging system (UVP; LLC, Phoenix, AZ, USA). The colony number and size were quantified, respectively. To generate the growth curves of A549 or A549R cells, 103 cells were incubated with DMEM containing 0 or 10 µM cisplatin for 0, 24, 48 or 72 h at 37°C, under 5% CO2. Then the cell number in each group was counted with the Olympus BX60 light microscope (Olympus Corporation, Tokyo, Japan). For the cell migration assay, A549 or A549R cells were cultured in 25 cm cell dishes with DMEM + 10% FBS to ~85% confluence, and were then scratched with a cell scratcher (Costar; Corning Incorporated). Cells were cultured for a further 24 h at 37°C with DMEM + 10% FBS, containing 2 µM cisplatin. The number of cells that crossed the baseline was then counted as the number of migrating cells using the Olympus BX60 light microscope (Olympus Corporation).

Fluorescence-activated cell sorting (FACS) can flow analysis of apoptotic cells

A total of 1×106 A549 or A549R cells were treated at 37°C with or without 10 µM cisplatin for 24 h; then cells in each group were collected for flow cytometry analysis with a Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) Apoptosis Detection kit (Abcam, Cambridge, UK). A549 or A549R cells were trypsinized with 0.125% trypsin and then suspended in 1 ml binding buffer, to which 10 µl Annexin V-FITC and 10 µl PI were added successively for incubation at room temperature in the dark for 15 min. The number of apoptotic cells was then determined using a FACScan flow cytometer (Bio-Rad Laboratories, Inc., Hercules, CA, USA) and analyzed using FlowJo version 10 (FlowJo LLC, Ashland, OR, USA).

Imaging of autophagic puncta with green fluorescence protein (GFP)-light chain (LC)-3 reporter

For the imaging of autophagic vesicles (puncta), A549 or A549R cells were transfected with a GFP-LC3 reporter plasmid (1 µg per well of a 12-well plate; Biovector Science Laboratory, Beijing, China) for 6 h using Lipofectamine 3000™ (Invitrogen; Thermo Fisher Scientific, Inc.). Fresh DMEM containing 2% FBS was added to the cells, which were then treated with or without 10 µM cisplatin at 37°C for 24 h. Treatment with 3 µM Rapamycin (Sigma-Aldrich; Merck KGaA) was taken as the positive autophagy induction control, and blank A549 or A549R cells (cells transfected with the GFP-LC3 reporter plasmid only) with fresh DMEM containing 2% FBS was used as the blank control. A total of 5 nM 3-methyladenine (3MA; an autophagy inhibitor; Sigma-Aldrich; Merck KGaA) was utilized to inhibit cisplatin-induced autophagy in A549 or A549R cells via treatment for 24 h at 37°C. Autophagic puncta were imaged and counted by confocal laser microscopy, and analyzed using FluoView software version 5.0 (both from Olympus Corporation).

Protein digestion, identification and quantification

The mixed H-/L-labeled protein sample was added into sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) loading buffer and incubated in pre-boiled water for 3 min. Proteins were then separated by electrophoresis with 12% SDS-PAGE (as described below) and stained with Coomassie Brilliant Blue at 26°C for 3 h. The whole gel lane was sliced into 40 pieces according to Sun et al (18). The excised sections were homogenized and de-stained twice with a 1:1 ratio of 50 mM Tris acetonitrile and 50 mM ammonium bicarbonate solution (both from Sigma-Aldrich; Merck KGaA). The extraction of tryptic peptides from the gel was sequentially performed with 5% Trifluoroacetate (Beijing Chemical Co., Ltd., Beijing, China) in the microwave oven at 750 W for 8 min, and with 2.5% Trifluoroacetate and with 50% Tris acetonitrile in the microwave oven at 750 W for 8 min. The extracts were pooled and dried completely by centrifugal lyophilization.

A mobile phase of 90 min at a flow rate of 300 nl/min was performed to separate each peptide mixture sample from the sliced gel, which were then subjected analysis with a Linear Trap Quadruple-Fourier Transform (LTQ-FT) mass spectrometer (Thermo Fisher Scientific, Inc.), which was equipped with a nanospray source and Agilent 1100 high-performance liquid chromatography system (Agilent Technologies, Inc., Santa Clara, CA, USA). The peptide eluent was introduced directly to an LTQ-FT mass spectrometer via electrospray ionization. Positively identified proteins were considered when at least two reliable peptides were matched and a protein score >64 was observed. The false positive rate of identified peptides was calculated as the ratio of total peptide hits in the reverse database to the number of peptide hits in the forward database above the same threshold. Identified proteins were quantified by SILAC-specific software (MSQuant 1.4.1; msquant.sourceforge.net) and inspected manually. Peptide abundances were calculated as ratios of the areas of the mono-isotopic peaks of the H-labeled versus the L-labeled peptides, and the protein ratios were calculated from the average of all quantified peptides of it.

Western blotting

Nuclear and cytosol fractions of the protein samples were isolated from A549 or A549R cells using a Nuclear/Cytosol Fractionation kit (BioVision, Inc., Milpitas, CA, USA) and then a protease inhibitor (Sigma-Aldrich; Merck KGaA) was added. The concentration of each protein sample was determined using a BCA Protein Assay Reagent kit (Pierce; Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions. Proteins (8 µg/lane) were separated by 12% SDS-PAGE and transferred to a nitrocellulose membrane (EMD Millipore, Billerica, MA, USA) in order to separate the proteins in each sample by molecular weight. Then the membrane was blocked with 2% bovine serum albumin (Sigma-Aldrich; Merck KGaA) at 4°C overnight, and then incubated with the rabbit or mouse anti-human LC3 (cat. no. sc-28266; 1:500), autophagy-related protein (Atg) 7 (cat. no. sc-517310; 1:500) or β-actin primary antibodies (cat. no. sc-517582; 1:1,000; all from Santa Cruz Biotechnology, Inc.) for 2 h at room temperature (26°C). Membranes were then incubated with horseradish peroxidase (HRP)-conjugated anti-rabbit secondary antibodies [bovine anti-rabbit immunoglobulin G (IgG)-HRP: cat. no. sc-2379, 1:1,000; or bovine anti-mouse IgG-HRP: cat. no. sc-2380, 1:1,000; Santa Cruz Biotechnology, Inc.] for 1 h at room temperature (26°C). Membranes were washed 4 times with 1X PBS-Tween-20 (0.1% final concentration) prior to each incubation. The antigen-antibody binding was visualized with Enhanced chemiluminescence (Thermo Fisher Scientific, Inc.) using the UVP BioSpectrum 500 imaging system (UVP, LLC, Phoenix, AZ, USA) and ImageJ version 1.43b (National Institutes of Health, Bethesda, MD, USA).

Gene ontology (GO) analysis

GO analyses were performed using DAVID 6.7 (david.ncifcrf.gov/). Apoptosis- and autophagy-associated genes were selected for analysis when the P-value of the correlation was <0.05.

Statistical analysis

SPSS 16.0 software (SPSS, Inc., Chicago, IL, USA) was utilized for statistical analysis. Quantitative results were presented as the mean ± standard error of 3 or 4 repeated experiments. Statistical differences were analyzed with Student's t-test or one-way analysis of variance with Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Acquisition of cisplatin resistance in human lung cancer A549 cells following serial passages with cisplatin treatment

Cisplatin-resistant human lung cancer A549 cells (A549R cells) were obtained following serial passages under 1 µM cisplatin (5 blind passages, then purification for another 5 passages) and then 2 µM cisplatin (5 blind passages, then purification for another 5 passages) treatment via colony forming assays. As indicated in Fig. 1A, a phenotype with a larger colony size of A549 cells was obtained (2.75±0.48 vs. 1.32±0.26; P<0.001). The level of proliferation in A549R cells was significantly higher than that of A549 cells, under treatment with 10 µM cisplatin for 24, 48 or 72 h (P<0.05, P<0.01 or P<0.001; Fig. 1B). Colony formation results also confirmed the difference in the level of proliferation between A549R and A549 cells (Fig. 1C). The colony number (Fig. 1D) and colony size (Fig. 1E) were greater in A549R cells post-treatment with 10 µM cisplatin (P<0.05 or P<0.001). In addition, a migration assay was performed for A549 and A549R cells in the presence of 2 µM cisplatin. It was demonstrated in Fig. 1F and G that more cells crossed the baseline in the A549R cell group (P<0.01). Taken together, the results indicate that cisplatin resistance was acquired in A549 cells post 10 passages under cisplatin treatment.

In addition, A549R cells were cultured in DMEM without cisplatin for an additional 20 passages. It was indicated in Fig. 2A-C that there was no marked difference in growth efficiency between A549R and A549 cells.

Reduced apoptosis induction by cisplatin in the cisplatin-resistant A549R cells

To confirm the difference in the sensitivity to cisplatin between A549R and A549 cells, apoptosis induction of either A549R or A549 cells, post-treatment with 10 µM cisplatin for 24 h was examined by flow cytometry analysis following staining with the Annexin V-FITC/PI Apoptosis Detection kit. As presented in Fig. 3A-D, in contrast to the A549 (Fig. 3A) or A549R (Fig. 3B) cells without cisplatin treatment, treatment with 10 µM cisplatin for 24 h induced significantly high levels of apoptosis in A549 and A549R cells (P<0.001; Fig. 3C-E). Furthermore, there were less apoptotic cells in the A549R group (Fig. 3D and E) than in the A549 group following 10 µM cisplatin treatment (P<0.05; Fig. 3C and E). Therefore, these results confirmed resistance in A549R cells to cisplatin.

Autophagy induction by cisplatin in A549R cells

Autophagy has been supported by more studies as one of mechanisms underlining the chemoresistance of lung cancer cells (19,20). Firstly in the present study, autophagy-specific acidic vesicular organelles (AVOs) in A549R or A549 cells were observed under a fluorescence microscope with a GFP-LC3 reporter. When compared with the blank A549 or A549R cells, 10 µM rapamycin induced significantly high levels of AVOs (P<0.01 or P<0.001; Fig. 4A and B). Notably, the 10 µM cisplatin treatment also induced significant levels of AVOs in A549 and A549R cells (P<0.01 or P<0.001). In addition, this induction could be inhibited by the autophagy inhibitor 3MA in the two types of cells (P<0.01). Furthermore, more AVOs were induced by cisplatin in A549R cells, than in A549 cells (P<0.01; Fig. 4B).

Western blotting was also performed to examine the expression of autophagy-associated genes in the cisplatin-treated A549 or A549R cells. Fig. 4C demonstrated that rapamycin and cisplatin induced a high level of LC3-I to LC3-II conversion and a high expression of Atg7 in A549 and A549R cells, both of which were inhibited by 3MA treatment. In addition, a greater LC3-II/LC3-I ratio and increased Atg7 expression were observed in the cisplatin-treated A549R cells when compared with the cisplatin-treated A549 cells.

General proteomics information by SILAC in the cisplatin-treated A549R cells

To recognize the discriminating protein profile underlining cisplatin resistance in A549R cells, a SILAC method was adopted to quantify the cellular response to cisplatin treatment in either A549 or A549R cells. The general technological process of SILAC is presented in Fig. 5A. The 12C6H14N2O2-Lysine-HCL (L-labeled) A549R cells (Fig. 5B) or the 13C6H14N2O2-Lysine-HCL (H-labeled) A549R cells (Fig. 5C) were respectively utilized to quantify the responsive protein profile to cisplatin, with H-labeled or L-labeled A549 cells as control. To examine the quality of each procedure, cellular proteins were separated by 12% SDS-PAGE. As shown in Fig. 6A, protein bands were equally distributed in the H- or L-labeled A549R or A549 cells. The general difference in proteomics between A549R and A549 cells were summarized in Fig. 6B: Total of 1,161±152 quantitative peptides, and 357±36 proteins were induced by cisplatin (10 µM) between A549R and A549 cells.

Upregulation of anti-apoptosis and autophagy-associated proteins in cisplatin-treated A549R cells via SILAC screening

Among the upregulated proteins in the H-labeled A549R cells, there were 23 proteins with expression that was >1.5-fold greater than in the L-labeled A549 cells (Fig. 6C). In particular, 15 proteins, including glucose-regulated protein, 78 kDa (GRP78), heat shock protein 71 (HSP71), heterogeneous nuclear ribonucleoprotein A1 (ROA1) and pre-mRNA processing factor 19 (PRP19), had increased expression by >2-fold in the H-labeled A549R cells when compared with the L-labeled A549 cells (Table I). In another repeated experiment with L-labeled A549R cells and H-labeled A549 cells, there were 18 proteins recognized also with expression that was >1.5 fold greater (Fig. 6D; Table II). GO analysis indicated that the majority of the upregulated proteins were involved in anti-apoptosis, DNA repair and autophagy (Tables I and II). In addition, the two repeated experiments demonstrated that 15 proteins [GRP78, HSP71, PRP19, polypyrimidine tract binding protein 1 (PTBP1), translationally controlled tumor protein (TCTP), Cathepsin D (CATD), Cytochrome c (CYC), thioredoxin domain containing 5 (TXND5), MutS homolog 6 (MSH6), Annexin A2 (ANXA2), RCA2 and Cyclin dependent kinase inhibitor 1A interacting protein (BCCIP), MSH2, protein phosphatase 2A 55 kDa regulatory subunit Bα (PP2AB), Rho glyceraldehyde-3-phosphate-dissociation inhibitor 1 (GDIR1) and ANXA4)] were repeatedly upregulated by >1.5-fold greater in H- and L-labeled A549R cells (Tables I and II).

Table I

Proteins with >1.5-fold change (H/L) in expression levels in A549R cells when compared with A549 cells.

Table I

Proteins with >1.5-fold change (H/L) in expression levels in A549R cells when compared with A549 cells.

Accession no.Uniprot IDProtein nameFold (H/L)GO term name in biological pathway
P11021*GRP7878 kDa glucose-regulated protein (bip)3.57Anti-apoptosis, calcium ion binding, enzyme binding, misfolded protein binding
Q9UMS4*PRP19Pre-mRNA-processing factor 193.28DNA repair, ubiquitin-ubiquitin ligase activity
P13693*TCTP Translationally-controlled tumor protein (TCTP)3.12Anti-apoptosis in response to DNA damage
P08107*HSP71Heat shock 70 kDa protein 1A/1B2.75Anti-apoptosis, cellular response to oxidative stress, negative regulation of cell death
P09651ROA1hnRNP core protein A1 (hnRNP A1)2.7mRNA processing, negative regulation of telomere maintenance via telomerase
P26599*PTBP1Polypyrimidine tract-binding protein 1 (hnRNP I)2.63mRNA processing, mRNA splicing, via spliceosome
P08758ANXA5Annexin A5 (Annexin V)2.58Anti-apoptosis, calcium-dependent phospholipid binding
P07339*CATDCathepsin D2.56Autophagic vacuole assembly, protein catabolic process, proteolysis
Q04760LGULLactoylglutathione lyase (Aldoketomutase)2.43Anti-apoptosis, regulation of transcription by RNA polymerase II
Q01081U2AF1Splicing factor U2AF 35 kDa subunit2.37mRNA processing, mRNA splicing, via spliceosome, RNA export from nucleus
P99999*CYCCytochrome C2.31Cellular respiration, cellular response to oxidative stress, intrinsic apoptotic signaling pathway
Q8NBS9*TXND5Thioredoxin domain-containing protein 52.25Anti-apoptosis, protein folding, response to endoplasmic reticulum stress
P14625ENPLHeat shock protein 90 kDa β member 1 (GRP-94)2.12Anti-apoptosis, RNA binding, unfolded protein binding
P52701*MSH6DNA mismatch repair protein Msh6 (hmsh6)2.1DNA damage response, methylated histone binding, mismatched DNA binding
P07355*ANXA2Annexin A2 (Annexin II)2.01Skeletal system development, phosphatidylinositol-4,5-bisphosphate binding, phospholipase A2 inhibitor activity
Q9P287*BCCIPBRCA2 and CDKN1A-interacting protein1.94DNA repair, regulation of cyclin-dependent protein serine/threonine kinase activity
P43246*MSH2DNA mismatch repair protein Msh2 (hmsh2)1.83Mismatch repair, intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediator
Q16531DDB1DNA damage-binding protein 1 (DDB p127 subunit)1.74Nucleotide-excision repair, DNA damage response, detection of DNA damage
P13073COX41Cytochrome C oxidase subunit 4 isoform 1 (COX IV-1)1.71Response to nutrient, mitochondrial electron transport, Cytochrome C to oxygen
P62714*PP2AB Serine/threonine-protein phosphatase 2A catalytic subunit β isoform (PP2A-β)1.69Protein amino acid dephosphorylation, apoptotic mitochondrial changes, negative regulation of Ras protein signal, response to endoplasmic reticulum stress
P52565*GDIR1Rho GDP-dissociation inhibitor 1 (Rho GDI 1)1.67Anti-apoptosis, regulation of Rho protein signal transduction and of small GTPase mediated signal transduction
P62805H4Histone H41.56Negative regulation of megakaryocyte differentiation
P09525*ANXA4Annexin A4 (Annexin IV)1.51Anti-apoptosis, negative regulation of NF-κB transcription factor activity
Q15833STXB2Syntaxin-binding protein 20.66Cellular response to interferon-gamma, protein transport
Q9NZJ7*MTCH1Mitochondrial carrier homolog 10.66Activation of cysteine-type endopeptidase activity, apoptotic process
O60218AK1BAAldo-keto reductase family 1 member B100.64Aldo-keto reductase (NADP) activity, geranylgeranyl reductase activity
Q0WX57*U17LO Ubiquitin-specific-processing protease 170.63Apoptotic process, protein deubiquitination involved in ubiquitin-dependent protein catabolic process
P11498PYCPyruvate carboxylase, mitochondrial0.63Biotin binding, identical protein binding, biotin metabolic process
P10909CLUSClusterin0.61Chaperone binding, positive regulation of apoptotic process
Q16513*PKN2 Serine/threonine-protein kinase N20.59Apoptotic process, cell adhesion, cell cycle and cell division
Q9HBU6EKI1Ethanolamine kinase 10.56ATP binding, ethanolamine kinase activity, phosphatidylethanolamine biosynthetic process
Q9GZU2PEG3 Paternally-expressed gene 3 protein0.53Apoptotic process, nucleic acid binding
O95140MFN2Mitofusin-20.53GTP binding, apoptotic process, macroautophagy
O14763*TR10BTRAIL receptor 20.52Receptor for the cytotoxic ligand TNFSF10/TRAIL, the adapter molecule FADD recruits caspase-8 to the activated receptor
Q96S44PRPKEKC/KEOPS complex subunit TP53RK0.47ATP binding,p53 binding, protein serine/threonine kinase activity
I0J062PANO1Proapoptotic nucleolar protein 10.47Positive regulation of apoptotic process, regulation of protein stability
Q15464*SHBSH2 domain-containing adapter protein B0.46Apoptotic process, cell differentiation, SH3/SH2 adaptor activity
Q96FX8PERPP53 apoptosis effector related to PMP-220.45Notch signaling pathway, positive regulation of proteolysis, regulation of apoptotic process
P26447S10A4Protein S100-A40.38Epithelial to mesenchymal transition, positive regulation of I-κB kinase/NF-κB signaling

[i] Accession numbers marked with an asterisks (*) are those that are affected by either up- or downregulation. GO, Gene Ontology; H, heavy-labeled; L, light-labeled.

Table II

Proteins with >1.5-fold change (L/H) in expression levels in A549R cells when compared with A549 cells.

Table II

Proteins with >1.5-fold change (L/H) in expression levels in A549R cells when compared with A549 cells.

Accession no.Uniprot IDProtein nameFold (L/H)GO term name biological pathway
P11021*GRP7878 kDa glucose-regulated protein (bip)2.87Anti-apoptosis, calcium ion binding, enzyme binding, misfolded protein binding
P43246*MSH2DNA mismatch repair protein Msh2 (hmsh2)2.75Mismatch repair, intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediator
P26599*PTBP1Polypyrimidine tract-binding protein 1 (hnRNP I)2.7mRNA processing, mRNA splicing, via spliceosome
P07339*CATDCathepsin D2.67Autophagic vacuole assembly, protein catabolic process, proteolysis
Q8NBS9*TXND5Thioredoxin domain-containing protein 52.6Anti-apoptosis, protein folding, response to endoplasmic reticulum stress
P13693*TCTP Translationally-controlled tumor protein (TCTP)2.58Anti-apoptosis in response to DNA damage
Q9Y3F4STRAPSerine-threonine kinase receptor-associated protein2.52mRNA processing, negative regulation of pathway-restricted SMAD protein phosphorylation
Q9UMS4*PRP19Pre-mRNA-processing factor 192.46DNA repair, ubiquitin-ubiquitin ligase activity
P61224RAP1BRas-related protein Rap-1b2.03Cell proliferation, positive regulation of ERK1 and ERK2 cascade
P08107*HSP71Heat shock 70 kDa protein 1A/1B1.89Anti-apoptosis, cellular response to oxidative stress, negative regulation of cell death
P52701*MSH6DNA mismatch repair protein Msh6 (hmsh6)1.76DNA damage response, methylated histone binding, mismatched DNA binding
P09525*ANXA4Annexin A4 (Annexin IV)1.73Anti-apoptosis, negative regulation of NF-κB transcription factor activity
P99999*CYCCytochrome C1.72Cellular respiration, cellular response to oxidative stress, intrinsic apoptotic signaling pathway
P62714*PP2AB Serine/threonine-protein phosphatase 2A catalytic subunit β isoform (PP2A-β)1.69Protein amino acid dephosphorylation, apoptotic mitochondrial changes, negative regulation of Ras protein signal, response to endoplasmic reticulum stress
P52565*GDIR1Rho GDP-dissociation inhibitor 1 (Rho GDI 1)1.67Anti-apoptosis, regulation of Rho protein signal transduction and of small GTPase mediated signal transduction
Q01130SFRS2 Serine/arginine-rich splicing factor 2 (Protein PR264)1.58mRNA processing, mitotic cell cycle, mRNA export from nucleus
P07355*ANXA2Annexin A2 (Annexin II)1.53Skeletal system development, phosphatidylinositol-4,5-bisphosphate binding, phospholipase A2 inhibitor activity
Q9P287*BCCIPBRCA2 and CDKN1A-interacting protein1.51DNA repair, regulation of cyclin-dependent protein serine/threonine kinase activity
O00194RB27BRas-related protein Rab-27B0.66GTP binding, myosin V binding, protein domain specific binding
Q0WX57*U17LO Ubiquitin-specific-17 processing protease0.66Apoptotic process, protein deubiquitination involved in ubiquitin-dependent protein catabolic process
O94804STK10 Serine/threonine-protein kinase 100.64Regulation of apoptotic process, regulation of mitotic cell cycle, signal transduction by protein phosphorylation
O60656UD19 UDP-glucuronosyltransferase 1-90.62 Glucuronosyltransferase activity, negative regulation of cellular glucuronidation
Q16513*PKN2 Serine/threonine-protein kinase N20.62Apoptotic process, cell adhesion, cell cycle and cell division
Q16222UAP1 UDP-N-acetylhexosamine pyrophosphorylase0.6Carbohydrate binding, UDP-N-acetylglucosamine diphosphorylase activity
Q16539MK14Mitogen-activated protein kinase 140.59Apoptotic process, DNA damage checkpoint, intracellular signal transduction
P62308RUXGSmall nuclear ribonucleoprotein G0.58RNA binding, histone mRNA metabolic process, mRNA splicing, RNA splicing
Q07812BAXApoptosis regulator BAX0.58Apoptotic mitochondrial changes, apoptotic process, DNA damage response
Q9NZJ7*MTCH1Mitochondrial carrier homolog 10.56Activation of cysteine-type endopeptidase activity, apoptotic process
Q15464*SHBSH2 domain-containing adapter protein B0.55Apoptotic process, cell differentiation, SH3/SH2 adaptor activity
O14763*TR10BTRAIL receptor 20.53Receptor for the cytotoxic ligand TNFSF10/TRAIL, the adapter molecule FADD recruits caspase-8 to the activated receptor
Q16890TPD53Tumor protein D530.49G2/M transition of mitotic cell cycle source, positive regulation of apoptotic signaling pathway and of JNK cascade
Q9H4P4RNF41E3 ubiquitin-protein ligase NRDP10.47Autophagy, extrinsic apoptotic signaling pathway, negative regulation of cell proliferation

[i] Accession numbers marked with an asterisks (*) are those that are affected by either up- or downregulation. GO, Gene Ontology; H, heavy-labeled; L, light-labeled.

Downregulation of apoptosis-associated proteins in cisplatin-treated A549R cells

In addition, there were 26 and 22 proteins that were downregulated >1.5-fold in the H- and L-labeled A549R cells, respectively, when compared with the L- and H-labeled A549 cells (Tables I and II). It was indicated in Table I that there were 16 proteins that were downregulated by >1.5-fold in H-labeled A549R cells when compared with L-labeled A549 cells. In another experiment, 14 proteins were revealed to be downregulated in L-labeled A549R cells when compared with the H-labeled A549 cells (Table II). Notably, the majority of the downregulated proteins were associated with apoptosis. In particular, 5 proteins [tumor necrosis factor receptor superfamily member 10B (TR10B), ubiquitin specific peptidase 17 (U17LO), SHB, PKN2, MTCH1) were downregulated in H- and L-labeled A549R cells; all of these proteins were involved in apoptotic processes or signaling. Therefore, apoptosis-associated proteins were downregulated in cisplatin-treated A549R cells.

Discussion

Cisplatin-based combinations of cytotoxic chemotherapy are the primary form of lung cancer chemotherapy as it significantly improves lung cancer patient outcomes (3,4,21,22). Approximately 30% of patients with stage IV NSCLC are responsive to cisplatin-based, two-drug combination treatment, and >95% patients live >3 years (22,23). Even for patients with SCLC, their initial response rates to cisplatin combination are higher, at 50-80%. However, almost all lung cancers are either initially or ultimately resistant to the current chemotherapy drugs, including cisplatin (22,23). In the present study, a NSCLC cell clone, A549, was chosen as a cell model to evaluate the sensitivity/resistance of lung cancer cells to cisplatin. Notably, serial passages of A549 cells under 1-2 µM cisplatin treatment gave rise to the cisplatin-resistant phenotype of A549 cells. A549 colonies with a larger size were manually enriched via colony forming assay. The results of growth curve, colony formation and migration assays confirmed that the A549R cells with the cisplatin-resistant phenotype grew and migrated more efficiently under cisplatin treatment than wild-type A549 cells. In addition, cisplatin-induced apoptosis was significantly decreased in A549R cells when compared with A549 cells. Taken together, A549R cells were less responsive to cisplatin.

Marked improvements have been achieved in the past few decades in our understanding of lung cancer biology (24,25). The identification of driver oncogenes in lung cancers has led to a change in cancer treatments. Some studies have provided greater understanding regarding the mechanisms underlying chemotherapy sensitivity/resistance and the associated biomarkers of lung cancer (26-30). Deregulated mesenchymal-epithelial transition (31) and reduced apoptosis induction (32) have been indicated to underlie the chemoresistance in lung cancer. Autophagy is a self-protective mechanism to guarantee basic energy supply under nutrition-deficient conditions, such as starvation (33). The cytoprotective mechanism of autophagy against chemotherapy has also been recognized in lung cancer cells (33) and other types of cancers (9,15,34,35). Thus, autophagy has been highlighted as one of the mechanisms underlying the chemoresistance of NSCLC. In the present study, autophagy induction by cisplatin was observed in A549 and A549R cells, and could be inhibited by the autophagy inhibitor, 3MA. Notably, a significantly higher level of autophagy was observed in A549R cells when compared with A549 cells. This implies that autophagy may contribute to the cisplatin-resistance phenotype of A549R cells.

Proteomics has been widely utilized to profile, screen and identify specific phenotype- or genotype-associated biomarkers (36,37). In recent years, SILAC has stood out when distinguishing the proteomics from one group to another, such as in cancer biomarker discovery (11,13) and in the identification of chemoresistance-associated biomarkers (16). In the present study, two rounds of SILAC procedures were performed with paired groups of H-labeled A549R cells and L-labeled A549 cells, or with paired groups of L-labeled A549R cells and H-labeled A549 cells. A total of 1,161±152 quantitative peptides and 357±36 proteins were induced by cisplatin (10 µM) between A549R and A549 cells. A total of 344±21 proteins were confirmed by two or more peptides. In addition, among the 23 proteins with 1.5-fold greater expression in the H-labeled A549R cells and the 18 proteins with 1.5-fold greater expression in the L-labeled A549R cells, there were 15 proteins that were repeated in the 2 rounds of experiments. On the other hand, there were 17 and 15 proteins that were downregulated in H- and L-labeled A549R cells, respectively. Particularly, the downregulation of apoptosis-associated proteins, including TR10B, U17LO, SHB, PKN2 and MTCH1, was observed in the two types of labeling experiments. These downregulated proteins may be involved in mitochondrial dysfunction, the cell response to stress, nuclear acid damage and finally in apoptosis induction. Exposure of any of the two proapoptotic domains of MTCH1 on the surface of mitochondria is sufficient for the induction of apoptosis in a B-cell lymphoma-2 (Bcl-2)-associated X/Bcl-2 antagonist/killer-independent manner (38). SH2 domain-containing adapter protein B (SHB) has been indicated to be involved in the Fyn related Src family tyrosine kinase-SHB signaling pathway, and regulates cell survival, differentiation and proliferation (39). The possible roles of U17LO, PKN2 and TR10B were not clear up until now.

GO analysis indicated that the majority of the proteins regulated apoptosis (26,28,29,40-43), DNA damage repairing (43-46) and other biological pathways. The effect of anti-apoptosis and autophagy promotion was also identified for these proteins in human lung cancer cells and other types of cells. GRP78 antagonizes apoptosis and positively regulates autophagy in human NSCLC cells via the adenosine monophosphate-activated protein kinase-mammalian target of rapamycin signaling pathway (28,29). TCTP also inhibits apoptosis by binding to p53 in lung carcinoma cells (26,47,48). The anti-apoptosis effect of HSP71 was also recognized in azacytidine-treated myeloma cells (49). Given the importance of anti-apoptosis and autophagy in chemotherapy resistance in cancer, the present study summarized the involvement of all of the 15 upregulated proteins in H- and L-labeled A549R cells in anti-apoptosis and/or autophagy promotion (Table III). It was indicated that the majority of these proteins were closely associated with anti-apoptosis and/or autophagy promotion in lung cancers or in other types of cancers. In addition, the majority of proteins that directly regulate autophagy were upregulated by <1.5-fold, though autophagy was significantly different in the two groups of A549 cells, which may be due to the difference in sensitivity among SILAC and other methods.

Table III

Involvement of upregulated proteins in anti-apoptosis and autophagy promotion in human lung cancer and other types of cells.

Table III

Involvement of upregulated proteins in anti-apoptosis and autophagy promotion in human lung cancer and other types of cells.

UniProt IDAnti-apoptosisAutophagy promotion
Author, yearCell typeRef.Author, yearCell typeRef.
GRP78Sun et al, 2012; Ahmad et al, 2014Lung cancer(28,29)Kim et al, 2012; Xie et al, 2016Lung cancer(27,46)
PRP19Lu et al, 2007Other(39)NR/
TCTPDu et al, 2017; Rho et al, 2011Lung cancer(26,43)Chen et al, 2014; Bonhoure et al, 2017Other(44,47)
HSP71Tian et al, 2013Other(45)NR/
PTBP1Takai et al, 2017Other(48)Li et al, 2016; Takai et al, 2017Other(33,48)
CATDWille et al, 2004; Li et al, 2004Lung cancer(37,49)Oliveira et al, 2015; Hah et al, 2012Other(50,51)
CYCChen et al, 2015; Moravcikova et al, 2014; Lee et al, 2015Lung cancer(52-54)Li et al, 2016; Kaminskyy et al, 2012Lung cancer(38,55)
TXND5Lee et al, 2010Other(34)NR/
MSH6Yu et al, 2017; Habiel et al, 2017Lung cancer(30,56)Knizhnik et al, 2013; Zanotto-Filo et al, 2015Other(40,57)
ANXA2Dassah et al, 2014; Wang et al, 2017Lung cancer(36,58)Wang et al, 2017; Chen et al, 2017Lung cancer(58,59)
BCCIPXu et al, 2017Lung cancer(60)NR/
MSH2Zhang et al, 2016; Terry et al, 2015Lung cancer(41,61)Zeng et al, 2007; Zeng et al, 2007Other(42,62)
PP2ABHuang et al, 2004; Shtrichman et al, 2000Other(63,64)Banreti et al, 2012; Ogura et al, 2010Other(65,66)
GDIR1NR/NR/
ANXA4Yao et al, 2016; Nagappan et al, 2016Lung cancer(67,68)NR/

[i] 'Other', indicates different types of cells to lung cancer cells; NR, not reported; GRP78, glucose-regulated protein, 78 kDa; HSP71, heat shock protein 71; PRP19, pre-mRNA processing factor 19; PTBP1, polypyrimidine tract binding protein 1; TCTP, translationally controlled tumor protein; CATD, Cathepsin D; CYC, Cytochrome c; TXND5, thioredoxin domain containing 5; MSH2/6, MutS homolog 2/6; ANXA2/4, Annexin A2/4; BCCIP, RCA2 and Cyclin dependent kinase inhibitor 1A interacting protein; PP2AB, protein phosphatase 2A 55 kDa regulatory subunit Bα; GDIR1, Rho glyceraldehyde-3-phosphate-dissociation inhibitor 1.

However, the detailed signaling pathways underlying such chemoresistance in A549R cells were not clear. In particular, though proteins such as GRP78, HSP71, PRP19, PTBP1, TCTP, CATD, CYC, TXND5, MSH6, ANXA2, BCCIP, MSH2, PP2AB, GDIR1 and ANXA4 were significantly deregulated in A549R cells, the association of each molecule with autophagy or directly with chemoresistance requires further investigation.

In conclusion, the present study isolated a cisplatin-resistant human lung cancer A549 cell clone, with reduced apoptosis and high levels of autophagy, in response to cisplatin treatment. SILAC proteomics recognized the high expression of GRP78 and other proteins that were associated with anti-apoptosis and/or autophagy promotion in cisplatin-resistant A549R cells.

Funding

The present study was supported by grants from the National Nature Science Foundation of China (grant no. 80151459), the Development Project from Science and Technology Department of Jilin Province (grant no. 140520020JH) and the Thirteen Five Science and Technology Research Project of Jilin Province Department of Education (grant no. 2016-467).

Availability of data and materials

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

Authors' contributions

ZW and GL designed the experiments. ZW, GL and JJ performed the experiments. ZW conducted the statistical analysis. GL wrote the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Fidler MM, Soerjomataram I and Bray F: A global view on cancer incidence and national levels of the human development index. Int J Cancer. 139:2436–2446. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Shen C, Wang X, Tian L, Zhou Y, Chen D, Du H, Wang W, Liu L and Che G: 'Different trend' in multiple primary lung cancer and intrapulmonary metastasis. Eur J Med Res. 20:172015. View Article : Google Scholar

3 

Kalemkerian GP: Advances in pharmacotherapy of small cell lung cancer. Expert Opin Pharmacother. 15:2385–2396. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Scagliotti GV, Parikh P, von Pawel J, Biesma B, Vansteenkiste J, Manegold C, Serwatowski P, Gatzemeier U, Digumarti R, Zukin M, et al: Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer. J Clin Oncol. 26:3543–3551. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Socinski MA, Smit EF, Lorigan P, Konduri K, Reck M, Szczesna A, Blakely J, Serwatowski P, Karaseva NA, Ciuleanu T, et al: Phase III study of pemetrexed plus carboplatin compared with etoposide plus carboplatin in chemotherapy-naive patients with extensive-stage small-cell lung cancer. J Clin Oncol. 27:4787–4792. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Kim ES: Chemotherapy resistance in lung cancer. Adv Exp Med Biol. 893:189–209. 2016. View Article : Google Scholar

7 

Willers H, Azzoli CG, Santivasi WL and Xia F: Basic mechanisms of therapeutic resistance to radiation and chemotherapy in lung cancer. Cancer J. 19:200–207. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Zembutsu H, Ohnishi Y, Tsunoda T, Furukawa Y, Katagiri T, Ueyama Y, Tamaoki N, Nomura T, Kitahara O, Yanagawa R, et al: Genome-wide cDNA microarray screening to correlate gene expression profiles with sensitivity of 85 human cancer xenografts to anticancer drugs. Cancer Res. 62:518–527. 2002.PubMed/NCBI

9 

Kihara C, Tsunoda T, Tanaka T, Yamana H, Furukawa Y, Ono K, Kitahara O, Zembutsu H, Yanagawa R, Hirata K, et al: Prediction of sensitivity of esophageal tumors to adjuvant chemotherapy by cDNA microarray analysis of gene-expression profiles. Cancer Res. 61:6474–6479. 2001.PubMed/NCBI

10 

Beltran H, Yelensky R, Frampton GM, Park K, Downing SR, MacDonald TY, Jarosz M, Lipson D, Tagawa ST, Nanus DM, et al: Targeted next-generation sequencing of advanced prostate cancer identifies potential therapeutic targets and disease heterogeneity. Eur Urol. 63:920–926. 2013. View Article : Google Scholar :

11 

Hoedt E, Zhang G and Neubert TA: Stable isotope labeling by amino acids in cell culture (SILAC) for quantitative proteomics. Adv Exp Med Biol. 806:93–106. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Patella F, Neilson LJ, Athineos D, Erami Z, Anderson KI, Blyth K, Ryan KM and Zanivan S: In-depth proteomics identifies a role for autophagy in controlling reactive oxygen species mediated endothelial permeability. J Proteome Res. 15:2187–2197. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Lanucara F and Eyers CE: Mass spectrometric-based quantitative proteomics using SILAC. Methods Enzymol. 500:133–150. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Yeh CC, Hsu CH, Shao YY, Ho WC, Tsai MH, Feng WC and Chow LP: Integrated stable isotope labeling by amino acids in cell culture (SILAC) and isobaric tags for relative and absolute quantitation (iTRAQ) quantitative proteomic analysis identifies galectin-1 as a potential biomarker for predicting dorafenib resistance in liver cancer. Mol Cell Proteomics. 14:1527–1545. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Wu X, Zahari MS, Renuse S, Nirujogi RS, Kim MS, Manda SS, Stearns V, Gabrielson E, Sukumar S and Pandey A: Phosphoproteomic analysisidentifies focal adhesion kinase2 (FAK2) as a potential therapeutic target for tamoxifen resistance in breast cancer. Mol Cell Proteomics. 14:2887–2900. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Xu H, Dephoure N, Sun H, Zhang H, Fan F, Liu J, Ning X, Dai S, Liu B, Gao M, et al: Proteomic profiling of paclitaxel treated cells identifies a novel mechanism of drug resistance mediated by PDCD4. J Proteome Res. 14:2480–2491. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Bosse K, Haneder S, Arlt C, Ihling CH, Seufferlein T and Sinz A: Mass spectrometry-based secretome analysis of non-small cell lung cancer cell lines. Proteomics. 16:2801–2814. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Sun P, Feng LX, Zhang DM, Liu M, Liu W, Mi T, Wu WY, Jiang BH, Yang M, Hu LH, et al: Bufalin derivative BF211 inhibits proteasome activity in human lung cancer cells in vitro by inhibiting β1 subunit expression and disrupting proteasome assembly. Acta Pharmacol Sin. 37:908–918. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Liu G, Pei F, Yang F, Li L, Amin AD, Liu S, Buchan JR and Cho WC: Role of autophagy and apoptosis in non-small-cell lung cancer. Int J Mol Sci. 18:182017.

20 

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

21 

Liu M, Ma S, Liu M, Hou Y, Liang B, Su X and Liu X: Synergistic killing of lung cancer cells by cisplatin and radiation via autophagy and apoptosis. Oncol Lett. 7:1903–1910. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Pisters KM, Evans WK, Azzoli CG, Kris MG, Smith CA, Desch CE, Somerfield MR, Brouwers MC, Darling G, Ellis PM, et al Cancer Care Ontario; American Society of Clinical Oncology: Cancer Care Ontario and American Society of Clinical Oncology adjuvant chemotherapy and adjuvant radiation therapy for stages I-IIIA resectable non small-cell lung cancer guideline. J Clin Oncol. 25:5506–5518. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Paz-Ares L, Mezger J, Ciuleanu TE, Fischer JR, von Pawel J, Provencio M, Kazarnowicz A, Losonczy G, de Castro G Jr, Szczesna A, et al INSPIRE investigators: Necitumumab plus pemetrexed and cisplatin as first-line therapy in patients with stage IV non-squamous non-small-cell lung cancer (INSPIRE): An open-label, randomised, controlled phase 3 study. Lancet Oncol. 16:328–337. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Lemjabbar-Alaoui H, Hassan OU, Yang YW and Buchanan P: Lung cancer: Biology and treatment options. Biochim Biophys Acta. 1856:189–210. 2015.PubMed/NCBI

25 

Sánchez-Céspedes M: Lung cancer biology: A genetic and genomic perspective. Clin Transl Oncol. 11:263–269. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Du J, Yang P, Kong F and Liu H: Aberrant expression of translationally controlled tumor protein (TCTP) can lead to radioactive susceptibility and chemosensitivity in lung cancer cells. Oncotarget. 8:101922–101935. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Kim KM, Yu TK, Chu HH, Park HS, Jang KY, Moon WS, Kang MJ, Lee DG, Kim MH, Lee JH, et al: Expression of ER stress and autophagy-related molecules in human non-small cell lung cancer and premalignant lesions. Int J Cancer. 131:E362–E370. 2012. View Article : Google Scholar

28 

Sun Q, Hua J, Wang Q, Xu W, Zhang J, Zhang J, Kang J and Li M: Expressions of GRP78 and Bax associate with differentiation, metastasis, and apoptosis in non-small cell lung cancer. Mol Biol Rep. 39:6753–6761. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Ahmad M, Hahn IF and Chatterjee S: GRP78 up-regulation leads to hypersensitization to cisplatin in A549 lung cancer cells. Anticancer Res. 34:3493–3500. 2014.PubMed/NCBI

30 

Yu W, Lu W, Chen G, Cheng F, Su H, Chen Y, Liu M and Pang X: Inhibition of histone deacetylases sensitizes EGF receptor-TK inhibitor-resistant non-small-cell lung cancer cells to erlotinib in vitro and in vivo. Br J Pharmacol. 174:3608–3622. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Mortimore GE, Miotto G, Venerando R and Kadowaki M: Autophagy. Subcell Biochem. 27:93–135. 1996. View Article : Google Scholar : PubMed/NCBI

32 

Notte A, Ninane N, Arnould T and Michiels C: Hypoxia counteracts taxol-induced apoptosis in MDA-MB-231 breast cancer cells: Role of autophagy and JNK activation. Cell Death Dis. 4:e6382013. View Article : Google Scholar : PubMed/NCBI

33 

Li C, Zhao Z, Zhou Z and Liu R: Linc-ROR confers gemcitabine resistance to pancreatic cancer cells via inducing autophagy and modulating the miR-124/PTBP1/PKM2 axis. Cancer Chemother Pharmacol. 78:1199–1207. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Lee WL, Wen TN, Shiau JY and Shyur LF: Differential proteomic profiling identifies novel molecular targets of paclitaxel and phytoagent deoxyelephantopin against mammary adenocar-cinoma cells. J Proteome Res. 9:237–253. 2010. View Article : Google Scholar

35 

Liang S, Xu Z, Xu X, Zhao X, Huang C and Wei Y: Quantitative proteomics for cancer biomarker discovery. Comb Chem High Throughput Screen. 15:221–231. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Dassah M, Almeida D, Hahn R, Bonaldo P, Worgall S and Hajjar KA: Annexin A2 mediates secretion of collagen VI, pulmonary elasticity and apoptosis of bronchial epithelial cells. J Cell Sci. 127:828–844. 2014. View Article : Google Scholar :

37 

Wille A, Gerber A, Heimburg A, Reisenauer A, Peters C, Saftig P, Reinheckel T, Welte T and Bühling F: Cathepsin L is involved in cathepsin D processing and regulation of apoptosis in A549 human lung epithelial cells. Biol Chem. 385:665–670. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Li YR, Li S, Ho CT, Chang YH, Tan KT, Chung TW, Wang BY, Chen YK and Lin CC: Tangeretin derivative, 5-acetyloxy-6,7,8,4'-tetramethoxyflavone induces G2/M arrest, apoptosis and autophagy in human non-small cell lung cancer cells in vitro and in vivo. Cancer Biol Ther. 17:48–64. 2016. View Article : Google Scholar

39 

Lu X and Legerski RJ: The Prp19/Pso4 core complex undergoes ubiquitylation and structural alterations in response to DNA damage. Biochem Biophys Res Commun. 354:968–974. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Knizhnik AV, Roos WP, Nikolova T, Quiros S, Tomaszowski KH, Christmann M and Kaina B: Survival and death strategies in glioma cells: Autophagy, senescence and apoptosis triggered by a single type of temozolomide-induced DNA damage. PLoS One. 8:e556652013. View Article : Google Scholar : PubMed/NCBI

41 

Zhang M, Hu C, Tong D, Xiang S, Williams K, Bai W, Li GM, Bepler G and Zhang X: Ubiquitin-specific peptidase 10 (USP10) deubiquitinates and stabilizes MutS homolog 2 (MSH2) to regulate cellular sensitivity to DNA damage. J Biol Chem. 291:10783–10791. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Zeng X and Kinsella TJ: A novel role for DNA mismatch repair and the autophagic processing of chemotherapy drugs in human tumor cells. Autophagy. 3:368–370. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Rho SB, Lee JH, Park MS, Byun HJ, Kang S, Seo SS, Kim JY and Park SY: Anti-apoptotic protein TCTP controls the stability of the tumor suppressor p53. FEBS Lett. 585:29–35. 2011. View Article : Google Scholar

44 

Chen K, Huang C, Yuan J, Cheng H and Zhou R: Long-term artificial selection reveals a role of TCTP in autophagy in mammalian cells. Mol Biol Evol. 31:2194–2211. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Tian E, Tang H, Xu R, Liu C, Deng H and Wang Q: Azacytidine induces necrosis of multiple myeloma cells through oxidative stress. Proteome Sci. 11:242013. View Article : Google Scholar : PubMed/NCBI

46 

Xie WY, Zhou XD, Yang J, Chen LX and Ran DH: Inhibition of autophagy enhances heat-induced apoptosis in human non-small cell lung cancer cells through ER stress pathways. Arch Biochem Biophys. 607:55–66. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Bonhoure A, Vallentin A, Martin M, Senff-Ribeiro A, Amson R, Telerman A and Vidal M: Acetylation of translationally controlled tumor protein promotes its degradation through chaperone-mediated autophagy. Eur J Cell Biol. 96:83–98. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Takai T, Yoshikawa Y, Inamoto T, Minami K, Taniguchi K, Sugito N, Kuranaga Y, Shinohara H, Kumazaki M, Tsujino T, et al: A Novel combination RNAi toward Warburg effect by replacement with miR-145 and silencing of PTBP1 induces apoptotic cell death in bladder cancer cells. Int J Mol Sci. 18:182017. View Article : Google Scholar

49 

Li X, Rayford H, Shu R, Zhuang J and Uhal BD: Essential role for cathepsin D in bleomycin-induced apoptosis of alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol. 287:L46–L51. 2004. View Article : Google Scholar : PubMed/NCBI

50 

Oliveira CS, Pereira H, Alves S, Castro L, Baltazar F, Chaves SR, Preto A and Côrte-Real M: Cathepsin D protects colorectal cancer cells from acetate-induced apoptosis through autophagy-independent degradation of damaged mitochondria. Cell Death Dis. 6:e17882015. View Article : Google Scholar : PubMed/NCBI

51 

Hah YS, Noh HS, Ha JH, Ahn JS, Hahm JR, Cho HY and Kim DR: Cathepsin D inhibits oxidative stress-induced cell death via activation of autophagy in cancer cells. Cancer Lett. 323:208–214. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Chen H, Liang ZW, Wang ZH, Zhang JP, Hu B, Xing XB and Cai WB: Akt activation and inhibition of cytochrome C release: mechanistic insights into leptin-promoted survival of type II Alveolar Epithelial Cells. J Cell Biochem. 116:2313–2324. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Moravcikova E, Krepela E, Prochazka J, Benkova K and Pauk N: Differential sensitivity to apoptosome apparatus activation in non-small cell lung carcinoma and the lung. Int J Oncol. 44:1443–1454. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Lee J, Yeganeh B, Ermini L and Post M: Sphingolipids as cell fate regulators in lung development and disease. Apoptosis. 20:740–757. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Kaminskyy VO, Piskunova T, Zborovskaya IB, Tchevkina EM and Zhivotovsky B: Suppression of basal autophagy reduces lung cancer cell proliferation and enhances caspase-dependent and -independent apoptosis by stimulating ROS formation. Autophagy. 8:1032–1044. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Habiel DM, Camelo A, Espindola M, Burwell T, Hanna R, Miranda E, Carruthers A, Bell M, Coelho AL, Liu H, et al: Divergent roles for clusterin in lung injury and repair. Sci Rep. 7:154442017. View Article : Google Scholar : PubMed/NCBI

57 

Zanotto-Filho A, Braganhol E, Klafke K, Figueiró F, Terra SR, Paludo FJ, Morrone M, Bristot IJ, Battastini AM, Forcelini CM, et al: Autophagy inhibition improves the efficacy of curcumin/temozolomide combination therapy in glioblastomas. Cancer Lett. 358:220–231. 2015. View Article : Google Scholar

58 

Wang K, Zhang T, Lei Y, Li X, Jiang J, Lan J, Liu Y, Chen H, Gao W, Xie N, et al: Identification of ANXA2 (annexin A2) as a specific bleomycin target to induce pulmonary fibrosis by impeding TFEB-mediated autophagic flux. Autophagy. 14:269–282. 2018. View Article : Google Scholar :

59 

Chen YD, Fang YT, Cheng YL, Lin CF, Hsu LJ, Wang SY, Anderson R, Chang CP and Lin YS: Exophagy of annexin A2 via RAB11, RAB8A and RAB27A in IFN-γ-stimulated lung epithelial cells. Sci Rep. 7:56762017. View Article : Google Scholar

60 

Xu XT, Hu WT, Zhou JY and Tu Y: Celecoxib enhances the radiosensitivity of HCT116 cells in a COX-2 independent manner by up-regulating BCCIP. Am J Transl Res. 9:1088–1100. 2017.PubMed/NCBI

61 

Terry MR, Arya R, Mukhopadhyay A, Berrett KC, Clair PM, Witt B, Salama ME, Bhutkar A and Oliver TG: Caspase-2 impacts lung tumorigenesis and chemotherapy response in vivo. Cell Death Differ. 22:719–730. 2015. View Article : Google Scholar :

62 

Zeng X, Yan T, Schupp JE, Seo Y and Kinsella TJ: DNA mismatch repair initiates 6-thioguanine - induced autophagy through p53 activation in human tumor cells. Clin Cancer Res. 13:1315–1321. 2007. View Article : Google Scholar : PubMed/NCBI

63 

Huang S, Shu L, Easton J, Harwood FC, Germain GS, Ichijo H and Houghton PJ: Inhibition of mammalian target of rapamycin activates apoptosis signal-regulating kinase 1 signaling by suppressing protein phosphatase 5 activity. J Biol Chem. 279:36490–36496. 2004. View Article : Google Scholar : PubMed/NCBI

64 

Shtrichman R, Sharf R and Kleinberger T: Adenovirus E4orf4 protein interacts with both Balpha and B' subunits of protein phosphatase 2A, but E4orf4-induced apoptosis is mediated only by the interaction with Balpha. Oncogene. 19:3757–3765. 2000. View Article : Google Scholar : PubMed/NCBI

65 

Bánréti Á, Lukácsovich T, Csikós G, Erdélyi M and Sass M: PP2A regulates autophagy in two alternative ways in Drosophila. Autophagy. 8:623–636. 2012. View Article : Google Scholar : PubMed/NCBI

66 

Ogura K, Okada T, Mitani S, Gengyo-Ando K, Baillie DL, Kohara Y and Goshima Y: Protein phosphatase 2A cooperates with the autophagy-related kinase UNC-51 to regulate axon guidance in Caenorhabditis elegans. Development. 137:1657–1667. 2010. View Article : Google Scholar : PubMed/NCBI

67 

Yao H, Sun C, Hu Z and Wang W: The role of Annexin A4 in cancer. Front Biosci. 21:949–957. 2016. View Article : Google Scholar

68 

Nagappan A, Venkatarame Gowda Saralamma V, Hong GE, Lee HJ, Shin SC, Kim EH, Lee WS and Kim GS: Proteomic analysis of selective cytotoxic anticancer properties of flavonoids isolated from Citrus platymamma on A549 human lung cancer cells. Mol Med Rep. 14:3814–3822. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 54 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Z, Liu G and Jiang J: Profiling of apoptosis- and autophagy-associated molecules in human lung cancer A549 cells in response to cisplatin treatment using stable isotope labeling with amino acids in cell culture. Int J Oncol 54: 1071-1085, 2019
APA
Wang, Z., Liu, G., & Jiang, J. (2019). Profiling of apoptosis- and autophagy-associated molecules in human lung cancer A549 cells in response to cisplatin treatment using stable isotope labeling with amino acids in cell culture. International Journal of Oncology, 54, 1071-1085. https://doi.org/10.3892/ijo.2019.4690
MLA
Wang, Z., Liu, G., Jiang, J."Profiling of apoptosis- and autophagy-associated molecules in human lung cancer A549 cells in response to cisplatin treatment using stable isotope labeling with amino acids in cell culture". International Journal of Oncology 54.3 (2019): 1071-1085.
Chicago
Wang, Z., Liu, G., Jiang, J."Profiling of apoptosis- and autophagy-associated molecules in human lung cancer A549 cells in response to cisplatin treatment using stable isotope labeling with amino acids in cell culture". International Journal of Oncology 54, no. 3 (2019): 1071-1085. https://doi.org/10.3892/ijo.2019.4690