Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Apr 20.
Published in final edited form as: Proteomics. 2014 Nov 20;14(0):2750–2759. doi: 10.1002/pmic.201400378

Proteomic signatures associated with p53 mutational status in lung adenocarcinoma

Ayumu Taguchi 1,*, Oliver Delgado 2, Müge Çeliktaş 2, Hiroyuki Katayama 2, Hong Wang 2, Adi F Gazdar 3, Samir M Hanash 2
PMCID: PMC4403731  NIHMSID: NIHMS676459  PMID: 25331784

Abstract

p53 is commonly mutated in lung adenocarcinoma. Mutant p53 loses wild-type function and some missense mutations further acquire oncogenic functions, while p53 wild-type may also induce pro-survival signaling. Therefore identification of signatures based on p53 mutational status has relevance to our understanding of p53 signaling pathways in cancer and identification of new therapeutic targets. To this end, we compared proteomic profiles of three cellular compartments (whole cell extract (WCE), cell surface, and media) from 28 human lung adenocarcinoma cell lines that differ based on p53 mutational status. In total, 11,598, 11,569, and 9,090 protein forms were identified in WCE, cell surface, and media, respectively. Bioinformatic analysis revealed that representative pathways associated with epithelial adhesion, immune and stromal cells, and mitochondrial function were highly significant in p53 missense mutations, p53 loss and wild-type p53 cell lines, respectively. Of note, mRNA levels of PGC1-α, a transcription co-activator that promotes mitochondrial oxidative phosphorylation and mitochondrial biogenesis, was substantially higher in p53 wild-type cell lines compared to either cell lines with p53 loss or with missense mutation. siRNA targeting PGC1-α inhibited cell proliferation in p53 wild-type cell lines, indicative of PGC1-α and its downstream molecules as potential therapeutic targets in p53 wild-type lung adenocarcinoma.

Keywords: lung adenocarcinoma, p53, PGC-1α, proteomics, transcriptomics

1. INTRODUCTION

Tumor suppressor p53 plays crucial and diverse roles in physiological processes [1]. p53 is commonly mutated in human cancer [2], and a large body of evidence indicates that mutant p53 loses wild-type functions and some missense mutations further acquire oncogenic functions [3, 4]. p53 mutation is not required for tumor development as many cancers emerge with wild-type p53. Approximately 50% of lung adenocarcinomas do not harbor p53 mutation [2, 5]. In these tumors, p53 is often inactivated by two major mechanisms: One is overexpression of MDM2, which ubiquitinates p53 leading to its proteasomal degradation [6]. MDM2 is detectable by immunohistochemistry in 30%-60% of lung adenocarcinomas while its amplification occurs in less than 10% [7-10]. The other mechanism is inactivation of p14ARF, which occurs in ~40% of lung adenocarcinomas, as a result of homozygous deletion, hypermethylation of promoter CpG islands, or point mutations [11]. p14ARF promotes degradation of MDM2, leading to stabilization and accumulation of p53 [12]. Thus, restoration of p53 function has been an attractive strategy for cancer therapy. However recent findings have indicated that p53 wild-type in cancer cells may induce pro-survival signaling, such as induction of various genes associated with DNA repair, cell cycle regulation, oxidative stress response, and MAPK signaling, which leads to activation of anti-apoptotic pathways [13, 14]. In addition, p53 wild-type is indeed overexpressed in a subset of non-small cell lung cancers with low level expression of MDM2 protein and upregulated expression of p14ARF protein [15, 16], further supporting the potential relevance of p53 wild-type to cancer cell survival.

On this basis, identification of signatures based on p53 mutational status has relevance to our understanding of p53 signaling pathways in cancer and identification of new therapeutic targets. To this end, we compared proteomic profiles of three cellular compartments from 28 human lung adenocarcinoma cell lines that differ based on p53 mutational status.

2. MATERIALS AND METHODS

Cell culture and transfection

All lung adenocarcinoma cell lines used in this study were cultured in RPMI1640 containing 10% FBS and 1% penicillin/streptomycin cocktail (Gibco). For the siRNA transfection experiments, three small interfering RNAs (siRNAs) targeting PPARGC1A (Sigma; #1: SASI_Hs01_00063323, #2: SASI_Hs01_00063324, and #3: SASI_Hs01_00063325) and negative control siRNA (Life Technologies; Cat # 4390844) were used. Cells were transfected at a final concentration of 50nM siRNA using Lipofectamine RNAiMAX (Life Technologies) according to the manufacturer's instructions. 72-hours post-transfection, protein was harvested using RIPA buffer for western blotting and MTS assay (CellTiter 96® AQueous One Solution Cell Proliferation Assay, Promega) was performed according to the manufacturer's protocol. Three independent experiments for MTS assay were done in triplicate.

Gene expression analysis

Total RNA was extracted from lung adenocarcinoma cell lines using RNeasy Plus Mini Kit (QIAGEN). RNA quality and concentration were checked by the Experion Automated Electrophoresis System (Bio-Rad) according to manufacturer's protocol. Expression data were obtained using the Illumina Human WG-6 v3.0 Expression BeadChips (Illumina) at the Genomics Core at UT Southwestern. Bead-summarized data were obtained using the Illumina BeadStudio software, and pre-processed using the R package MBCB (Model-based Background Correction for Beadarray) for background correction and probe summarization. Pre-processed data were the quantile-normalized and log transformed.

Mass spectrometry analysis

Proteomic analysis was performed as previously described [17, 18]. All cell lines were grown in RPMI1640 (Pierce) containing 10% of dialyzed FBS (Invitrogen), 1% penicillin/streptomycin cocktail and 13C-lycine instead of regular lysine, for seven passages according to the standard SILAC protocol [19]. To obtain whole cell extracts, ~2×107 cells were lysed in 1 ml of PBS containing the detergent octyl-glucoside (OG) (1% w/v) and protease inhibitors (complete protease inhibitor cocktail, Roche Diagnostics), followed by sonication and centrifugation at 20,000×g with collection of the supernatant, and filtration through a 0.22 μm filter. The secreted proteins were obtained directly from the cell conditioned media after 48 h of culture. Cells and debris were removed by centrifugation at 5000×g and filtration through a 0.22 μm filter. To isolate cell surface proteins, ~2×108 cells were biotinylated in the culture plate with 10 ml of 0.25 mg/ml of Sulfo-NHS-SS-BIOTIN (Thermo Scientific) in PBS at room temperature (23–24°C) for 10 min, after extensive PBS rinsing. The residual biotinylation reagent was quenched with 10mM Lysine. Protein extraction was performed in a solution containing 2% (v/v) Igepal CA-630 (Sigma-Aldrich) with cell disruption by sonication followed by centrifugation at 20,000×g. Biotinylated proteins were chromatographically isolated by affinity chromatography using 1 ml of NeutrAvidin Plus UltraLink Resin (Pierce) according to the manufacturer's instructions. Proteins bound to the column were recovered by reduction of the biotinylation reagent with 5 ml of a solution containing 65 μmol of DTT and 1% octyl-glucoside (OG) detergent for overnight at 4°C. Two mg of whole cell extracts, 1mg of conditioned media, and 0.5mg of cell surface proteins were reduced in DTT and alkylated with iodoacetamide before fractionation with reverse-phase high performance liquid chromatography (RP-HPLC). A total of 84 fractions were collected at a rate of 3 fractions/minute. The mobile phase A was: H2O:Acetonitrile (95:5, v/v) with 0.1% of trifluoracetic acid (TFA); mobile phase B was: Acetonitrile:H2O (95:5)with 0.1% of TFA. The collected fractions from RP-HPLC were dried by lyophilization and subjected to in-solution digestion with trypsin (Trypsin Gold, Mass Spectrometry Grade, Promega). Trypsin powder was dissolved in the Digestion Buffer (100 mM amino bicarbonate, 2% Acetonitrile) to make 8ng/μL of Trypsin solution. 50μL of Trypsin solution was added to each of the dried fractions, capped and mixed thoroughly, followed by digestion for 5 hours at 37°C. The digestion was quenched by adding 10μL of quench solution (1%TFA in H2O). Based on chromatogram profile, 84 fractions were grouped into 24-pool fractions for LC-MS/MS analysis by RPLC-MS/MS using a nanoflow LC system (Eksigent) coupled on-line with Mass spectrometry (MS) Separations were performed using 75μm inner diameter × 360μm outer diameter × 25cm long fused silica capillary column (New Objective) slurry packed in house with 5μm, 200Å pore size C18 silica-bonded stationary pgase (Magic C18 AQ, New Objective). Following injection of ~2μg of protein digest onto a C18 trap column (Waters, 180μmID × 20mmL), the LC column was wash for 5min with mobile phase A (2% acetonitrile, o.1% formic acid) at a flow rate of 10μL/min. Peptides were eluted using a linear gradient of 0.35% mobile phase B (0.1 formic acid in acetonitrile)/minute for 90 min, then to 95% B in an additional 10 min, all at a constant flow rate of 300nL/minEluted peptides were analyzed by MS using an LTQ Orbitrap mass spectrometer (Thermo Scientific) in data dependent acquisition (DDA) mode. Each full MS scan (m/z 400-1800) was followed by 10 MS/MS scans (normalized collision energy of 35%) for the 10 most abundant precursor ions in a ~1.5s of duty cycle. Dynamic exclusion was enabled to minimize redundant selection of peptides previously selected for MS/MS analysis. Parameters for MS1 were 60,000 for resolution, 1 × 106 for automatic gain control (AGC) target, and 150 ms for maximum injection time. MS/MS was done by collision-induced dissociation (CID) fragmentation with 3 × 104 for AGC, 10 ms for maximum injection time, 35 for normalized collision energy (NCE), 2.0 m/z for isolation width, 0.25 for activation q-value, and 10 ms for activation time. MS/MS spectra were searched against the IPI proteome database (IPI human, v3.57 and IPI bovine, v3.43) using SEQUEST through the Computational Proteomics Analysis System, CPAS (LabKey Software Foundation, https://www.labkey.org/Project/home/CPAS/begin.view) with one fixed modification of carbamidomethylation at Cysteine (57.04304 Da) and two variable modifications, oxidation at Methionine (15.9949 Da) and SILAC at Lysine (6.0201 Da). Peptides were considered legitimately identified if they achieved specific charge state and proteolytic cleavage-dependent cross-correlation (Xcorr) scores of 1.9 for [M+H]1+, 2.2 FOR [M+2H]2+, and 3.5 for [M+3H]3+, and a minimum delta correlation score (ΔCn) of 0.08. A false protein discovery rate of approximately 5% was determined by searching the primary tandem MS data using the same criteria against a decoy database wherein the protein sequences are reversed. To eliminate the possible contamination of bovine proteins, any unlabeled lysine containing peptides with bovine homology were discarded. The total number of spectral counts for each protein group was normalized to total spectral counts of the samples which were set as 50,000, and used for the differential analysis among different samples. Total spectral counts were normalized to 50,000 total events for each cell line.

Western blot analysis

Anti-PGC-1α antibodies were obtained from Cell Signaling Technology used for Western blot analysis. Anti-GAPDH antibody (Abcam) and β-tubulin (Cell Signaling Technology) were used as loading control. Signal intensities of PGC-1α protein bands were quantified using ImageJ (http://rsbweb.nih.gov/ij/) and normalized to β-tubulin signal intensities.

3. RESULTS

Proteomic profiling of lung adenocarcinoma cell lines

Three cellular protein compartments (whole cell extract (WCE), cell surface, and media) of 28 human lung adenocarcinoma cell lines, including 23 with mutant p53 and 5 with wild type p53, were analyzed by mass spectrometry (Supporting Information Tables 1, 2, and 3). p53-mutant cell lines were further classified into two groups: 1- with missense mutation and 2- with p53-loss, as p53 missense mutations have the potential to gain oncogenic functions (Table 1) [3, 4]. p53 mutational status was not associated with phenotypic characteristics such as cell morphology or invasion propensity nor with the mutational status of EGFR or KRAS, which are major oncogenic drivers in lung adenocarcinoma. In total, 11,598, 11,569, and 9,090 protein forms were identified in WCE, cell surface, and media, respectively. Average number of identified proteins in each compartment (Mean ± Standard Deviation (SD)) was 3380.7 ± 726.1 in WCE, 3,291.4 ± 694.3 in surface, and 1,987 ± 451.3 in media (Table 1). Next we assessed the enrichment of membrane proteins in the cell surface compartment and of extracellular proteins in the media compartment. Localization of proteins was predicted using the Ingenuity Pathways Analysis (IPA; http://www.ingenuity.com/) and protein abundance was estimated based on normalized MS/MS counts [17]. The subcellular localization demonstrated by mass spectrometric analysis and predictions from database analysis were concordant, particularly for abundant proteins (Supporting Information Figure 1), indicating enrichment of cell surface or extracellular proteins based on cellular compartment. Interestingly, cytoplasmic proteins that lack a signal peptide were also enriched in the conditioned media, suggesting potential release of cytoplasmic proteins via extracellular vesicles in these lung adenocarcinoma cells [20].

Table 1.

Characteristic of lung adenocarcinoma cell lines and number of identified proteins.

Characteristics Mutational Status Number of identified proteins
Group Cell line Age Gender Site Smoking EGFR Kras Mutation WCE Surface Media
Missense H1355 53 M M Y Wild Mutant E285K 3364 3275 1973
H1373 56 M NA Y Wild Mutant P47L 3380 3557 2147
H1437 60 M M Y Wild Wild R267P 4087 3063 1965
H1573 35 F M N Wild Mutant R248L 3282 3076 2405
H1651 71 M P NA Wild Wild C176Y 3495 3837 2378
H1793 52 F P NA Wild Wild R209*, R273H 3286 4267 2058
H1838 NA F P NA Wild Wild R273L 3330 3571 1679
H1975 NA F P N Mutant Wild R273H 2715 3128 1150
H1993 47 F M Y Wild Wild C242W 3734 1842 1669
H2009 68 F M Y Wild Mutant R273L 2143 2766 1927
H2030 NA M M N Wild Mutant G262V 2787 3063 1395
H2122 46 F M Y Wild Mutant Q16L, C176F 4358 4165 2193
H2291 NA M M N Wild Mutant G154V 4360 3958 2333
H23 51 M P Y Wild Mutant M246I 1463 1880 1390
H2342 55 M P NA Wild Wild Y220C 4095 4060 2008
H2405 47 M M Y Wild Wild R273H 3724 3441 2632
H650 NA M M N Wild Mutant K164N 3310 2692 1884
H820 53 M M NA Mutant Wild E284K 4517 3894 3179
HCC2279 NA F NA N Mutant Wild Y234C 3446 3259 2311
Loss H2228 NA F P N Wild Wild Q331* 3475 3324 2088
H838 59 M M Y Wild Wild E62* 2147 2502 2034
H522 60 M P Y Wild Wild P191fs*56 4072 4200 2745
H1650 27 M M Y Mutant Wild c.673-2A>G; Splicing Frameshift 3580 3242 1739
Wild H1385 49 F M Y Wild Mutant None 4187 4100 1769
H1395 55 F P Y Wild Wild None 3645 3719 1548
H1563 NA M P NA Wild Wild None 2800 3442 1986
H1568 NA F M Y Wild Wild None 3555 3185 1933
H1944 NA F M Y Wild Mutant None 2323 1652 1133
Total 11598 11569 9090
Mean 3380.7 3291.4 1987.5
SD 726.1 694.3 451.3

Proteomic characterization of lung adenocarcinoma cell lines based on p53 mutational status

Protein abundance based on normalized MS/MS counts was compared among 19 cell lines with p53 missense mutations, four cell lines with p53 loss, and five p53 wild-type cell lines. The following criteria were used: (1) a protein is identified in 50% or more cell lines in the group, (2) ratio of normalized MS/MS counts between two groups > 2, and (3) average of MS/MS counts in the group > 2. A total of 118, 66, and 112 proteins were found to be increased in WCE, cell surface, and media of cell lines with missense mutations compared to cell lines with p53 loss and p53 wild-type cell lines, respectively (Supporting Information Figure 2A). Similarly, 289 (WCE), 338 (surface), and 351 (media) proteins were elevated in cell lines with p53 loss, and 167 (WCE), 180 (surface), and 82 (media) proteins were elevated in p53 wild-type cell lines (Supporting Information Figure 2B and C). Next, we performed pathway analysis of these elevated proteins using IPA. Canonical pathways associated with epithelial adhesion (Epithelial Adherens Junction Signaling and Remodeling of Epithelial Adherens Junction) were highly significant in all three compartments of cell lines with p53 missense mutations (Figure 1A). These two pathways include 13 proteins (ARPC1B, ARPC2, ARPC5L, CDH1, CTNND1, JUP, LMO7, MET, PARD3, RAB7A, RAP1B, TGFB2, TUBB4B) (Supporting Information Table 4). Among them, protein expression levels of TGFB2, RAB7A and LMO7 in media and ARPC5L in WCE were significantly elevated in cell lines with p53 missense mutations (P < 0.05, Mann–Whitney U test) (Figure 2A). For cell lines with p53 loss, pathways associated with immune and stromal cells (Antiproliferative Role of TOB in T Cell Signaling, T Helper Cell Differentiation Role of Macrophages, Fibroblasts and Endothelial Cells in Rheumatoid Arthritis Interestingly) were highly significant in WCE and cell surface (Figure 1B). CALML5, CD40, CDKN1B, CSF1, CUL1, IL18, LRP1, MMP1, NGFR, PRSS1, PRSS3, RAC1, ROR2, SKP1, STAT4, and TGFB1 were included in these pathways and protein expression levels of LRP1, MMP1 and ROR2 in the cell surface compartment were significantly elevated in cell lines with p53 loss (P < 0.05, Mann–Whitney U test) (Figure 2B). Four of five top significant canonical pathways in TCE of p53 wild-type cell lines were involved in mitochondrial function: Oxidative Phosphorylation, TCA Cycle II (Eukaryotic), Branched-chain α-keto acid Dehydrogenase Complex, and Mitochondrial Dysfunction (Figure 1C). Of note, p53 was predicted as one of the top significantly activated upstream regulators in all three cellular compartments in p53 wild-type cell lines, suggesting functional roles of p53 signaling pathways in p53 wild-type lung adenocarcinoma cell lines (Table 2). To further investigate key molecules associated with p53 wild-type signatures, we compared mRNA expression among the 28 lung adenocarcinoma cell lines profiled. Fifty-one probes were elevated in p53 wild-type cell lines compared to cell lines with p53 loss according to the following criteria: ratio of log2 intensity of wild-type vs p53 loss > 2 with P value of less than 0.01 (unpaired t test). Similarly, mRNA expression levels of 109 probes were significantly higher in p53 wild-type cell lines compared to cell lines with p53 missense mutations (Figure 3A).

Figure 1. Top 5 significant canonical pathways for elevated proteins in three cellular compartments predicted by Ingenuity Pathway Analysis.

Figure 1

Top 5 significant canonical pathways for cell lines with p53 missense mutation (A), cell lines with p53 loss (B), and p53 wild-type cell lines (C). P values were calculated by Fisher's exact test.

Figure 2. Signature proteins significantly elevated in mutant p53 lung adenocarcinoma cell lines.

Figure 2

A. Protein expression levels of ARPC5L in WCE, and LMO7, RAB7A, and TGFB2 in media. B. Protein expression levels of LRP1, MMP1 and ROR2 in cell surface. P values were calculated by Mann–Whitney U test.

Table 2.

Top 5 upstream regulators predicted by Ingenuity Pathway Analysis.

WCE Surface Media
Upstream Regulator Activation z-score p-value of overlap Upstream Regulator Activation z-score p-value of overlap Upstream Regulator Activation z-score p-value of overlap
Missense GLI1 2.646 4.56E-
05
miR-16-
5p
−1.982 2.02E-
03
IL6 2.881 5.84E-03
Vegf 2.630 5.20E-
03
RAF1 1.980 2.55E-
03
NFE2L2 2.797 6.59E-04
PPARG 2.570 4.49E-
03
HGF 1.956 1.07E-
02
IL1B 2.729 5.44E-03
MGEA5 −2.449 1.05E-
03
OSM 1.531 1.37E-
02
miR-30c-
5p
−2.630 9.81E-07
miR-124-
3p
−2.449 1.08E-
03
HNF4A 1.114 1.68E-
02
IFNG 2.626 2.76E-03
Loss NFE2L2 3.132 1.66E-
02
TNF 4.453 2.93E-
06
TP53 3.159 1.64E-05
miR-16-
5p
−2.800 2.17E-
03
NFkB
(complex)
3.804 8.51E-
04
miR-16-
5p
−2.985 1.85E-03
TP73 2.596 1.46E-
02
IFNG 3.602 7.09E-
06
ANGPT2 2.813 1.50E-03
CD40LG 2.596 6.06E-
03
Vegf 3.410 1.59E-
03
miR-1 −2.772 7.42E-03
HIF1A 2.558 4.96E-
03
FOXO1 3.286 1.41E-
03
TP73 2.763 1.43E-05
Wild TP53 3.417 5.00E-
11
IFNG 3.092 8.90E-
03
TP53 3.340 5.95E-07
INSR 2.596 2.84E-
06
NFE2L2 3.082 5.99E-
04
FGF2 2.599 1.13E-04
IL2 2.581 6.92E-
03
TNF 3.026 1.68E-
02
Vegf 2.404 1.98E-03
IL4 2.577 2.72E-
02
TP53 2.879 3.02E-
10
HRAS 2.177 4.02E-04
Map4k4 −2.449 1.30E-
04
TGFB1 2.825 5.32E-
06
CEBPA 2.158 2.47E-05

Figure 3. PGC-1α as a potential target of p53 wild-type lung adenocarcinoma.

Figure 3

A. Venn diagrams of genes elevated in p53 wild-type lung adenocarcinoma cell lines. B. mRNA expression of PPARGC1A in lung adenocarcinoma cell lines. Bars indicate mean and standard deviation of log2 intensity. P value was calculated by unpaired t test. C. Knockdown of PGC1-α with siRNA in H1944 cells. GAPDH was served as a loading control. D. MTS assay of H1944 cells treated with negative control siRNA or siRNAs against PGC1-α. Columns indicate the average of triplicate samples from a representative experiment, and bars indicate standard deviation.

We found that the mRNA expression level of PPARGC1A is substantially higher in p53 wild-type cell lines compared to either cell lines with p53 loss (P = 0.0035, unpaired t test) or with missense mutation (P = 0.0003, unpaired t test) (Figure 3B). PGC1-α, encoded by PPARGC1A, is a transcription co-activator that promotes mitochondrial oxidative phosphorylation and mitochondrial biogenesis [21], thus suggestive of PGC1-α as a key molecule associated with p53 wild-type function. Protein expression of PGC1-α was significantly associated with PPARGC1A mRNA expression levels in 10 lung adenocarcinoma cell lines (Spearman r = 0.81, P = 0.0072) (Supporting Information Figure 3A-C). We therefore investigated the effect of suppressing PGC1-α. Indeed, cell proliferation was inhibited with treatment of PGC1-α siRNA in H1944 cells (Fig. 3C and D), indicating PGC1-α as a potential therapeutic target in p53 wild-type lung adenocarcinoma. We also sought to identify PGC1-α-associated proteins in p53 wild-type cell lines. Based on IPA, 368 proteins have either direct or indirect interaction with PGC1-α. Among elevated proteins in p53 wild-type cell lines determined as above (Supporting Information Figure 2C), 19, 16, and 9 proteins were PGC1-α-interacting proteins in WCE, cell surface, and media of p53 wild-type cell lines respectively (Supporting Information Table 5).

4. DISCUSSION

In this study, we profiled the proteome and transcriptome of 28 human lung adenocarcinoma cell lines to identify inherent signatures associated with p53 mutational status. Protein signatures associated with p53 missense mutations were comprised of proteins involved in epithelial adhesion. Interestingly, this includes a general trend for higher surface localization of the epithelial marker CDH1 and the proto-oncogene MET. Detection of CDH1 indicates an epithelial phenotype in the context of epithelial-to-mesenchymal transition (EMT) [22], while activation of the MET pathway by HGF is known to induce EMT leading to the suppression of CDH1 [23]. Missense p53 mutations can induce EMT by several mechanisms such as influencing TGFβ pathway signaling or modulating ZEB1 expression. TGFβ2 was indeed significantly elevated in the media of cell lines containing missense p53 mutations. Recently, it has been reported that TGFB2 is upregulated by mutant p53 through its gain-of-function ability to bind to and sequester p63, which indirectly represses several genes associated with EMT, including TGFβ2 [24]. TGFβ pathway activation induces EMT in a variety of cell lines [22, 25]. TGFβ signaling requires the binding of TGFβ first to the type I and II TGFβ receptor (TGFBR2) [25]. Expression levels of TGFBR1 and TGFBR2 were very low in the missense p53 cell lines profiled (data not shown), suggesting that TGFβ2 secreted in the media is not sufficient to induce EMT in these cell lines. Missense p53 mutations can also potentiate EMT induction by MET by regulating its expression and/or enhancing MET cell surface recycling [3, 4]. The detection of both CDH1 and MET on the surface, however, suggests that these cells are poised to engage EMT driven by MET, but retain an epithelial phenotype in the absence of autocrine HGF stimulation as HGF was not identified in the secretome from lung adenocarcinoma cell lines profiled in this study (data not shown). Therefore additional molecular programs might be required to induce full EMT in these cell lines.

For cell lines with p53 loss, ROR2 is of particular interest as it is a receptor type tyrosine kinase. ROR2 regulates Wnt signaling pathways, and overexpression and oncogenic functions of ROR2 has been reported in various types of cancer [26-31]. ROR2 was highly expressed in H2291 cell line which harbors p53 missense mutation. However levels of basal p53 levels were very low in H2201 cell lines (data not shown), so p53 G154V mutation occurred in H2291 cells might be loss of function mutation.

Proteomic signatures associated with p53 wild-type are associated with mitochondrial function. p53 regulates various metabolic pathways including mitochondrial respiration [32, 33]. We found that the mRNA expression level of PPARGC1A, which encodes PGC-1α, is substantially higher in p53 wild-type cell lines compared to either cell lines with p53 loss or with missense mutation. Although PGC-1α is a known transcriptional target of p53, it is context-dependent whether PGC-1α would be induced or repressed [34, 35]. The main physiological functions of PGC-1α is control of energy metabolism by increasing oxidative metabolism, and in particular mitochondrial oxidative phosphorylation (OxPhos) in muscle, brain, liver and adipose tissue and also inhibit apoptosis [21]. Although the role of PGC-1α in cancer is still controversial, recent papers have indicated the oncogenic roles of PGC-1α in cancer [21, 36-39]. In addition PGC-1α activates proarrest and metabolic target genes by modulating p53 transactivation function and promote cell survival under metabolic stress [40]. Furthermore, mitochondrial function is required for transformation and tumor growth [41, 42]. Therefore p53-PGC-1α axis would be relevant to induce cancer-associated metabolic changes in p53 wild-type lung adenocarcinoma. Therapeutic targets for p53 wild-type cancer have not been well studied: miR-197 has been identified as a potential therapeutic target in p53 wild-type non-small cell lung cancer [43], and Nutlin-3, a MDM2 antagonist, preferentially sensitizes p53 wild-type cancer to Death Receptor 5-induced apoptosis in p53 wild-type cancer [44]. Unfortunately, PGC-1α itself is not currently druggable [21]. Thus identification of key transcriptional partners or downstream molecules of PGC-1α would enable us to inhibit specific pathways regulated by PGC-1α in cancer. Bioinformatic analysis of our proteomic profiling identified many potential therapeutic targets which are associated with PGC-1α (Supporting Information Table 5), such as SOD1 and SOD2. SOD inhibitor, 2-methoxyestradiol, is currently undergoing clinical trials for treatment of a variety of cancers [45-47].

In conclusion, proteomic characterization of lung adenocarcinoma cell lines based on p53 mutational status, integrating transcriptome and bioinformatic analysis, identified molecular pathways and potential therapeutic targets associated with p53 missense mutations, p53 loss and wild-type p53.

Supplementary Material

Suppl Figs
Suppl Tables

ACKNOWLEDGEMENTS

This work was supported by the Canary Foundation; the Lungevity Foundation; the National Cancer Institute Early Detection Research Network; The Department of Defense W81XWH-09-LCRP-CTRA; MD Anderson's Moon Shots Program. The authors have declared no conflict of interest.

Abbreviations

PGC-1α

Peroxisome proliferator-activated receptor gamma coactivator 1-alpha

WCE

whole cell extract

IPA

Ingenuity Pathways Analysis

EMT

epithelial-tomesenchymal transition

Footnotes

Publisher's Disclaimer: This article has been accepted for publication and undergone full peer review but has not been through the copyediting, typesetting, pagination and proofreading process, which may lead to differences between this version and the Version of Record. Please cite this article as doi: 10.1002/pmic.201400378.

REFERENCES

  • 1.Vousden KH, Prives C. Blinded by the Light: The Growing Complexity of p53. Cell. 2009;137:413–431. doi: 10.1016/j.cell.2009.04.037. [DOI] [PubMed] [Google Scholar]
  • 2.Kandoth C, McLellan MD, Vandin F, Ye K, et al. Mutational landscape and significance across 12 major cancer types. Nature. 2013;502:333–339. doi: 10.1038/nature12634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Muller PA, Vousden KH. Mutant p53 in Cancer: New Functions and Therapeutic Opportunities. Cancer cell. 2014;25:304–317. doi: 10.1016/j.ccr.2014.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Muller PA, Vousden KH. p53 mutations in cancer. Nature cell biology. 2013;15:2–8. doi: 10.1038/ncb2641. [DOI] [PubMed] [Google Scholar]
  • 5.Imielinski M, Berger AH, Hammerman PS, Hernandez B, et al. Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell. 2012;150:1107–1120. doi: 10.1016/j.cell.2012.08.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Wade M, Li YC, Wahl GM. MDM2, MDMX and p53 in oncogenesis and cancer therapy. Nature reviews. Cancer. 2013;13:83–96. doi: 10.1038/nrc3430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Xue Q, Sano T, Kashiwabara K, Saito M, et al. Aberrant expression of pRb, p16, p14ARF, MDM2, p21 and p53 in stage I adenocarcinomas of the lung. Pathology international. 2002;52:103–109. doi: 10.1046/j.1440-1827.2002.01321.x. [DOI] [PubMed] [Google Scholar]
  • 8.Eymin B, Gazzeri S, Brambilla C, Brambilla E. Mdm2 overexpression and p14(ARF) inactivation are two mutually exclusive events in primary human lung tumors. Oncogene. 2002;21:2750–2761. doi: 10.1038/sj.onc.1205359. [DOI] [PubMed] [Google Scholar]
  • 9.Higashiyama M, Doi O, Kodama K, Yokouchi H, et al. MDM2 gene amplification and expression in non-small-cell lung cancer: immunohistochemical expression of its protein is a favourable prognostic marker in patients without p53 protein accumulation. British journal of cancer. 1997;75:1302–1308. doi: 10.1038/bjc.1997.221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Momand J, Jung D, Wilczynski S, Niland J. The MDM2 gene amplification database. Nucleic acids research. 1998;26:3453–3459. doi: 10.1093/nar/26.15.3453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Tam KW, Zhang W, Soh J, Stastny V, et al. CDKN2A/p16 inactivation mechanisms and their relationship to smoke exposure and molecular features in non-small-cell lung cancer. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2013;8:1378–1388. doi: 10.1097/JTO.0b013e3182a46c0c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Zhang Y, Xiong Y, Yarbrough WG. ARF promotes MDM2 degradation and stabilizes p53: ARF-INK4a locus deletion impairs both the Rb and p53 tumor suppression pathways. Cell. 1998;92:725–734. doi: 10.1016/s0092-8674(00)81401-4. [DOI] [PubMed] [Google Scholar]
  • 13.Mandinova A, Lee SW. The p53 pathway as a target in cancer therapeutics: obstacles and promise. Science translational medicine. 2011;3:64rv61. doi: 10.1126/scitranslmed.3001366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Janicke RU, Sohn D, Schulze-Osthoff K. The dark side of a tumor suppressor: anti-apoptotic p53. Cell death and differentiation. 2008;15:959–976. doi: 10.1038/cdd.2008.33. [DOI] [PubMed] [Google Scholar]
  • 15.Wang YC, Lin RK, Tan YH, Chen JT, Chen CY. Wild-type p53 overexpression and its correlation with MDM2 and p14ARF alterations: an alternative pathway to non-small-cell lung cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2005;23:154–164. doi: 10.1200/JCO.2005.03.139. [DOI] [PubMed] [Google Scholar]
  • 16.Koga T, Hashimoto S, Sugio K, Yoshino I, et al. Heterogeneous distribution of P53 immunoreactivity in human lung adenocarcinoma correlates with MDM2 protein expression, rather than with P53 gene mutation. International journal of cancer. Journal international du cancer. 2001;95:232–239. doi: 10.1002/1097-0215(20010720)95:4<232::aid-ijc1040>3.0.co;2-5. [DOI] [PubMed] [Google Scholar]
  • 17.Taguchi A, Politi K, Pitteri SJ, Lockwood WW, et al. Lung Cancer Signatures in Plasma Based on Proteome Profiling of Mouse Tumor Models. Cancer cell. 2011;20:289–299. doi: 10.1016/j.ccr.2011.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Faca VM, Ventura AP, Fitzgibbon MP, Pereira-Faca SR, et al. Proteomic analysis of ovarian cancer cells reveals dynamic processes of protein secretion and shedding of extra-cellular domains. PloS one. 2008;3:e2425. doi: 10.1371/journal.pone.0002425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ong SE, Mann M. A practical recipe for stable isotope labeling by amino acids in cell culture (SILAC). Nat. Protoc. 2006;1:2650–2660. doi: 10.1038/nprot.2006.427. [DOI] [PubMed] [Google Scholar]
  • 20.Inal JM, Kosgodage U, Azam S, Stratton D, et al. Blood/plasma secretome and microvesicles. Biochimica et biophysica acta. 2013;1834:2317–2325. doi: 10.1016/j.bbapap.2013.04.005. [DOI] [PubMed] [Google Scholar]
  • 21.Girnun GD. The diverse role of the PPARgamma coactivator 1 family of transcriptional coactivators in cancer. Seminars in cell & developmental biology. 2012;23:381–388. doi: 10.1016/j.semcdb.2012.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–890. doi: 10.1016/j.cell.2009.11.007. [DOI] [PubMed] [Google Scholar]
  • 23.Ogunwobi OO, Liu C. Hepatocyte growth factor upregulation promotes carcinogenesis and epithelial-mesenchymal transition in hepatocellular carcinoma via Akt and COX-2 pathways. Clinical & experimental metastasis. 2011;28:721–731. doi: 10.1007/s10585-011-9404-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Neilsen PM, Noll JE, Mattiske S, Bracken CP, et al. Mutant p53 drives invasion in breast tumors through up-regulation of miR-155. Oncogene. 2013;32:2992–3000. doi: 10.1038/onc.2012.305. [DOI] [PubMed] [Google Scholar]
  • 25.Ikushima H, Miyazono K. TGFbeta signalling: a complex web in cancer progression. Nature reviews. Cancer. 2010;10:415–424. doi: 10.1038/nrc2853. [DOI] [PubMed] [Google Scholar]
  • 26.O'Connell MP, Fiori JL, Xu M, Carter AD, et al. The orphan tyrosine kinase receptor, ROR2, mediates Wnt5A signaling in metastatic melanoma. Oncogene. 2010;29:34–44. doi: 10.1038/onc.2009.305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ren DY, Minami Y, Nishita M. Critical role of Wnt5a-Ror2 signaling in motility and invasiveness of carcinoma cells following Snail-mediated epithelial-mesenchymal transition. Genes Cells. 2011;16:304–315. doi: 10.1111/j.1365-2443.2011.01487.x. [DOI] [PubMed] [Google Scholar]
  • 28.Morioka K, Tanikawa C, Ochi K, Daigo Y, et al. Orphan receptor tyrosine kinase ROR2 as a potential therapeutic target for osteosarcoma. Cancer Sci. 2009;100:1227–1233. doi: 10.1111/j.1349-7006.2009.01165.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Edris B, Espinosa I, Muhlenberg T, Mikels A, et al. ROR2 is a novel prognostic biomarker and a potential therapeutic target in leiomyosarcoma and gastrointestinal stromal tumour. J Pathol. 2012;227:223–233. doi: 10.1002/path.3986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Li CG, Chen HY, Hu LY, Xing YM, et al. Ror2 modulates the canonical Wnt signaling in lung epithelial cells through cooperation with Fzd2. Bmc Mol Biol. 2008:9. doi: 10.1186/1471-2199-9-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Yamamoto H, Oue N, Sato A, Hasegawa Y, et al. Wnt5a signaling is involved in the aggressiveness of prostate cancer and expression of metalloproteinase. Oncogene. 2010;29:2036–2046. doi: 10.1038/onc.2009.496. [DOI] [PubMed] [Google Scholar]
  • 32.Vousden KH, Ryan KM. p53 and metabolism. Nature reviews. Cancer. 2009;9:691–700. doi: 10.1038/nrc2715. [DOI] [PubMed] [Google Scholar]
  • 33.Matoba S, Kang JG, Patino WD, Wragg A, et al. p53 regulates mitochondrial respiration. Science. 2006;312:1650–1653. doi: 10.1126/science.1126863. [DOI] [PubMed] [Google Scholar]
  • 34.Aquilano K, Baldelli S, Pagliei B, Cannata SM, et al. p53 orchestrates the PGC-1alpha-mediated antioxidant response upon mild redox and metabolic imbalance. Antioxidants & redox signaling. 2013;18:386–399. doi: 10.1089/ars.2012.4615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Sahin E, Colla S, Liesa M, Moslehi J, et al. Telomere dysfunction induces metabolic and mitochondrial compromise. Nature. 2011;470:359–365. doi: 10.1038/nature09787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.D'Errico I, Salvatore L, Murzilli S, Lo Sasso G, et al. Peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC1alpha) is a metabolic regulator of intestinal epithelial cell fate. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:6603–6608. doi: 10.1073/pnas.1016354108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Bhalla K, Hwang BJ, Dewi RE, Ou L, et al. PGC1alpha promotes tumor growth by inducing gene expression programs supporting lipogenesis. Cancer research. 2011;71:6888–6898. doi: 10.1158/0008-5472.CAN-11-1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Vazquez F, Lim JH, Chim H, Bhalla K, et al. PGC1alpha expression defines a subset of human melanoma tumors with increased mitochondrial capacity and resistance to oxidative stress. Cancer cell. 2013;23:287–301. doi: 10.1016/j.ccr.2012.11.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Klimcakova E, Chenard V, McGuirk S, Germain D, et al. PGC-1alpha promotes the growth of ErbB2/Neu-induced mammary tumors by regulating nutrient supply. Cancer research. 2012;72:1538–1546. doi: 10.1158/0008-5472.CAN-11-2967. [DOI] [PubMed] [Google Scholar]
  • 40.Sen N, Satija YK, Das S. PGC-1alpha, a key modulator of p53, promotes cell survival upon metabolic stress. Molecular cell. 2011;44:621–634. doi: 10.1016/j.molcel.2011.08.044. [DOI] [PubMed] [Google Scholar]
  • 41.Funes JM, Quintero M, Henderson S, Martinez D, et al. Transformation of human mesenchymal stem cells increases their dependency on oxidative phosphorylation for energy production. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:6223–6228. doi: 10.1073/pnas.0700690104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Weinberg F, Hamanaka R, Wheaton WW, Weinberg S, et al. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proceedings of the National Academy of Sciences of the United States of America. 2010;107:8788–8793. doi: 10.1073/pnas.1003428107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Fiori ME, Barbini C, Haas TL, Marroncelli N, et al. Antitumor effect of miR-197 targeting in p53 wild-type lung cancer. Cell death and differentiation. 2014;21:774–782. doi: 10.1038/cdd.2014.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Meijer A, Kruyt FA, van der Zee AG, Hollema H, et al. Nutlin-3 preferentially sensitises wild-type p53-expressing cancer cells to DR5-selective TRAIL over rhTRAIL. British journal of cancer. 2013;109:2685–2695. doi: 10.1038/bjc.2013.636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kulke MH, Chan JA, Meyerhardt JA, Zhu AX, et al. A prospective phase II study of 2-methoxyestradiol administered in combination with bevacizumab in patients with metastatic carcinoid tumors. Cancer chemotherapy and pharmacology. 2011;68:293–300. doi: 10.1007/s00280-010-1478-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Harrison MR, Hahn NM, Pili R, Oh WK, et al. A phase II study of 2-methoxyestradiol (2ME2) NanoCrystal(R) dispersion (NCD) in patients with taxane-refractory, metastatic castrate-resistant prostate cancer (CRPC). Investigational new drugs. 2011;29:1465–1474. doi: 10.1007/s10637-010-9455-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zhou Q, Gustafson D, Nallapareddy S, Diab S, et al. A phase I dose-escalation, safety and pharmacokinetic study of the 2-methoxyestradiol analog ENMD-1198 administered orally to patients with advanced cancer. Investigational new drugs. 2011;29:340–346. doi: 10.1007/s10637-009-9383-9. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Suppl Figs
Suppl Tables

RESOURCES