Orlistat, a novel potent antitumor agent for ovarian cancer: proteomic analysis of ovarian cancer cells treated with Orlistat

  • Authors:
    • Hui-Qiong Huang
    • Jing Tang
    • Sheng-Tao Zhou
    • Tao Yi
    • Hong-Ling Peng
    • Guo-Bo  Shen
    • Na Xie
    • Kai Huang
    • Tao Yang
    • Jin-Hua Wu
    • Can-Hua Huang
    • Yu-Quan Wei
    • Xia  Zhao
  • View Affiliations

  • Published online on: May 8, 2012     https://doi.org/10.3892/ijo.2012.1465
  • Pages: 523-532
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Orlistat is an orally administered anti-obesity drug that has shown significant antitumor activity in a variety of tumor cells. To identify the proteins involved in its antitumor activity, we employed a proteomic approach to reveal protein expression changes in the human ovarian cancer cell line SKOV3, following Orlistat treatment. Protein expression profiles were analyzed by 2-dimensional polyacrylamide gel electrophoresis (2-DE) and protein identification was performed on a MALDI-Q-TOF MS/MS instrument. More than 110 differentially expressed proteins were visualized by 2-DE and Coomassie brilliant blue staining. Furthermore, 71 proteins differentially expressed proteins were positively identified via mass spectrometry (MS)/MS analysis. In particular, PKM1/2, a key enzyme involved in tumorigenesis, was found to be significantly downregulated in SKOV3 cells following treatment with Orlistat. Moreover, PKM1/2 was proved to be downregulated in SKOV3 cells by western blot analysis after treatment with Orlistat. Taken together, using proteomic tools, we identified several differentially expressed proteins that underwent Orlistat-induced apoptosis, particularly PKM2. These changes confirmed our hypothesis that Orlistat is a potential inhibitor of ovarian cancer and can be used as a novel adjuvant antitumor agent.

Introduction

In the 1920s, the Nobel Prize winner Otto Warburg observed a marked increase in glycolysis and enhanced lactate production in tumor cells even when maintained in conditions of high oxygen tension (termed Warburg effect), leading to widespread concerns about the metabolic changes in human types of cancer (1). Either as a consequence or as a cause, alterations of cancer cell-intrinsic metabolism have been considered as essential hallmarks of cancer. Among these metabolic changes, de novo fatty acid biosynthesis was found elevated in the majority of human types of cancer, such as prostate (2), colorectal (3), ovarian (4), bladder (5), esophageal (6), gastric (7), lung (8), endometrial (9), breast (10) and soft tissue sarcomas (11). Fatty acid synthase (FASN) is regarded as a key regulator of de novo fatty acid synthesis and was widely found upregulated in a wide variety of human malignancies and their pre-neoplastic lesions. Recent studies also reveal that FASN is associated with the stage of cancer and indicate a poor prognosis (12). Thus, FASN could be considered as a reliable predictor of recurrence and disease-free survival along with neo-plastic stage (13). In vivo treatment with inhibitors of FASN has been proven to lead to markedly decreased survival in human cancer xenografts (14) and silencing of the FASN gene by siRNA also inhibits cancer cell growth and ultimately induces cancer cell apoptosis (15). Therefore, agents that inhibit FASN and the de novo fatty-acid synthesis pathways could be considered as novel antitumor strategies.

Orlistat, an anti-obesity drug approved by the US Food and Drug Administration, which possesses extremely low oral bio-availability (16), exhibits anti-proliferative and anti-tumor properties against prostate and breast cancer cells due to its ability to block the lipogenic activity of FASN (16), by acting on the 2.3-A-resolution crystal structure of the thioesterase domain of FASN (17). Orlistat negatively influences FASN activity and has a significant effect on the antitumor activity by inducing remarkable diversification such as a complete G2-M phase loss, S-phase accumulation and the emerging sub-G1 (apoptotic) cell increase, and repression of the promoter activity of Her2/neu gene (18).

Ovarian cancer is the most common malignancy of the female reproductive tract and is the leading cause of death from gynecologic types of cancer; it is currently the fifth leading cause of female cancer-related mortality (19). Finding a novel therapeutic approach is essential since the 5-year survival rate of women with ovarian cancer is low, despite the fact that significant progress has been made in the therapy of this disease (20).

2-DE based proteomics has been shown to be a powerful tool in rapidly profiling differentially expressed proteins associated with a number of diseases (2123). In our study, we aimed to investigate the differential expression in Orlistat-treated SKOV3 cells using a 2DE-MS-based proteomics approach, in order to better understand the molecular mechanisms underlying Orlistat-induced tumor repression. In total, more than 110 differentially expressed proteins were found altered between Orlistat-treated and untreated SKOV3 cells, and subsequently 71 proteins were identified by MS analysis. Furthermore, we showed that PKM2 was significantly down-regulated in Orlistat-treated SKOV3 cells, which confirmed the antitumor properties of Orlistat, indicating that Orlistat can be used as a novel adjuvant antitumor agent for ovarian cancer patients.

Materials and methods

Cell culture and treatment

The human epithelial serous cystadenocarcinoma cell line SKOV3 was obtained from the American Type Culture Collection (ATCC, Rockville, MD). Cells were grown in Dulbecco’s-modified Eagle’s medium (DMEM, Gibco, USA) containing 10% fetal calf serum (Hyclone, USA), penicillin (107 U/l) and streptomycin (10 mg/l) at 37°C in a humidified chamber containing 5% CO2. Orlistat was dissolved in dimethyl sulphoxide (DMSO). When the cells reached 50–70% confluency, the medium was replaced by a fresh culture medium containing Orlistat. Control cells were cultured in a medium containing an equal amount of DMSO instead of Orlistat. For 2-DE analysis, SKOV3 cells were treated with 20 mM Orlistat for 4 days and the media were changed every day. Cells were washed twice by centrifugation in phosphate buffered saline (PBS) and transferred to sterile plastic tubes for storage at −80°C prior to use.

Cell proliferation assay

Cell growth and viability were assessed using an MTT cell proliferation kit (Roche Applied Science). The cells were seeded on 96-well microplates at 2.0x103/well. At 48, 72 and 96 h, the cells were treated with different concentrations of Orlistat and incubated at 37°C in 5% CO2. The cells were subsequently incubated with 10 μl of MTT for 4 h, then the media were removed and 150 μl DMSO were added. We put the plate in a shaker before reading absorbance at 490-nm using a microplate reader (3550-UV, Bio-Rad, USA), after 20 min of incubation. The procedure was repeated three times with similar results. The following formula was used to calculate the inhibition rate of SKOV3 cell proliferation: (1-experimental group OD value/negative control OD value) x 100%. Media-only treated (untreated) cells were considered as the negative control group.

2-DE and image analysis

Cells (1.3x108) were lysed in 1 ml lysis buffer (7 M urea, 2 M thiourea, 4% CHAPS, 100 mM DTT, 0.2% pH 3.0–10.0 ampholyte, Bio-Rad, USA) containing protease inhibitor cocktail 8340 (Sigma, St. Louis, MO, USA). Samples were then kept on ice and sonicated for six cycles of 10 sec, with each cycle consisting of 5 sec sonication, followed by a 10 sec break. After centrifugation at 14,000 rpm for 1 h at 4°C, the supernatant was collected and the protein concentrations determined using the DC Protein Assay Kit (Bio-Rad). Protein samples (3 mg) were applied to an immobilized pH gradient (IPG) strip (17 cm, pH 3.0–10.0 NL, Bio-Rad) using a passive rehydration method. After 12–16 h of rehydration, the strips were transferred to an isoelectric focusing (IEF) cell (Bio-Rad) and focused for a total of 60,000 Vh. The second dimension was performed using 12% equilibration. The gels were stained using CBB R-250 (Merck, Germany) and scanned with a Bio-Rad GS-800 scanner. Triplicate samples were analyzed at each time point of treatment to ensure the reproducibility of analyses. The maps were analyzed by PDQuest software Version 6.1 (Bio-Rad). Each gel spot was normalized as a percentage of the total quantity of all spots in that gel and evaluated in terms of OD. Only those spots that changed consistently and significantly (>2.0-fold) were selected for MS analysis.

In-gel digestion

In-gel digestion of proteins was carried out using MS-grade Trypsin Gold (Promega, Madison, WI, USA), according to the manufacturer’s instructions. Briefly, spots were cut out of the gel (1–2 mm diameter) using a razor blade, and destained twice with 100 mM NH4HCO3/50% acetonitrile (ACN) at 37°C for 45 min in each treatment. Following dehydration and drying, the gels were pre-incubated in 10–20 μl trypsin solution for 1 h. Samples were then added in adequate digestion buffer (40 mM NH4HCO3/10% ACN) to cover the gels and incubated overnight at 37°C. Tryptic digests were extracted using MiliQ water initially, followed by extraction twice with 50% ACN/5% trifluoroacetic acid (TFA) for 1 h each time. The combined extracts were dried in a vacuum concentrator at room temperature. The samples were then subjected to MS analysis.

MALDI-Q-TOF analysis and protein identification

Mass spectra were acquired using a quadrupole time-of-flight (Q-TOF) mass spectrometer (Micromass, Manchester, UK) with a matrix-assisted laser desorption ionization (MALDI) source (Micromass). Tryptic digests were dissolved in 5 μl of 70% ACN/0.1% TFA, and then 1 μl of the digestion was mixed with 1 μl saturated α-cyano-4-hydroxy-cinnamic acid (CHCA) in 50% ACN/0.5% TFA and spotted onto a 96-well target plate. MS/MS was performed in a data-dependent mode in which the top ten most abundant ions for each MS scan were selected for MS/MS analysis. The MS/MS data were acquired and processed using the MassLynx software (Micromass) and MASCOT was used to search the database. Database searches were carried out using the following parameters: database, Swiss-Prot; taxonomy, Homo sapien; enzyme, trypsin; and allowance of one missed cleavage. Carbamidomethylation was selected as a fixed modification and oxidation of methionine was allowed to be variable. The peptide and fragment mass tolerance were at 1 and 0.2 Da, respectively. The data format selected was Micromass PKL and the instrument selected was MALDI-Q-TOF. Proteins with probability-based MOWSE scores exceeding their threshold (P<0.05) were considered to be positively identified.

Western blot analysis

Collected cells were lysed in RIPA buffer (50 mM Tris-base, 1.0 mM EDTA, 150 mM NaCl, 0.1% SDS, 1% Triton X-100, 1% sodium deoxycholate, 1 mM PMSF) to extract all the proteins and quantified by the DC protein assay Kit (Bio-Rad). Samples were separated by 12% SDS-PAGE and transferred to polyvinylidene difluoride (PVDF) membranes (Amersham Biosciences). The membranes were blocked overnight with PBS containing 0.1% Tween 20 in 5% skimmed milk at 4°C, and subsequently probed by the primary antibodies: rabbit anti-PKM2 (diluted 1:500, Abcam, UK). Blots were incubated with secondary antibody conjugated to horseradish peroxidase for 2 h at room temperature. Target proteins were detected by enhanced chemiluminescence reagents (Amersham Pharmacia Biotech, Piscataway, USA), and β-actin was used as an internal control.

Statistics

All quantitative data are recorded as the means ± SD. Comparisons between two groups were performed by Student’s t-test. Differences among multiple groups were assessed by one-way ANOVA analysis. Relevance analysis of ordinal data was performed by cross χ2 test. A statistically significant difference was defined as p<0.05.

Results

Proliferation activity of Orlistat-treated SKOV3 cells

The proliferation activity of Orlistat-treated SKOV3 cells was examined using the MTT assays. MTT results showed that the proliferation activity was suppressed by Orlistat in both a dose- and duration-dependent manner, and the proliferation ratio was decreased to 60% of the control value 96 h after treatment with Orlistat when the drug concentration was 20 mM, as shown in Fig. 1.

Proteomic analysis of Orlistat-treated SKOV3 cell protein expression compared with the parental SKOV3 cells

To explore the molecular mechanisms underlying the Orlistat-induced antitumor activity of SKOV3 cells, 2-DE based proteomics was used to profile differentially expressed proteins in SKOV3 cells treated with or without Orlistat. Image analysis was performed using PDQuest 7.1 software. Representative 2-DE maps are shown in Fig. 2. Approximately 1000–1100 protein spots were detected by CBB R-250 staining in a single 2-DE gel. Each protein spot was normalized as a percentage of the total intensity of all spots in the gel. By comparing 2-DE patterns, differentially expressed proteins were defined as statistically meaningful (p<0.05) based on both of the following two criteria: i) intensity alterations of >2.0-fold (t-test, p<0.05) and ii) observed in at least 3 individual experiments. According to these criteria, a total of 111 spots were selected and analyzed using MALDI-Q-TOF tandem mass spectrometry. A total of 71 proteins from the 111 spots were identified (Fig. 2). As different isoforms of a protein might have distinct functions, each isoform/spot was considered to be a single protein for analysis in our study.

Protein identification and bioinformatics analysis

In total, 71 spots with differential expression levels were subjected to MS/MS analysis. The MS/MS data were queried using the search algorithm MASCOT against the Expasy protein sequence database. Proteins were identified based on a number of criteria including PI, MW, the number of matched-peptides and MOWSE score (Table I and II).

Table I

Protein spots identified by MALDI-Q-TOF.

Table I

Protein spots identified by MALDI-Q-TOF.

Spot no.Accession no.bProtein nameaGene nameMwcPIcNo. of peptideCoverage (%)Scored
Upregulated
1P15311EzrinEZR69,4845.9456133
2P68104Elongation factor 1-α 1EEF1A150,4519.101528179
3P06733α-enolasENO147,4817.0181857
4P04264Keratin, type II cytoskeletal 1KRT166,1708.153368
5P50453Serpin B9SERPINB943,0045.612746372
6O00429Dynamin-1-like proteinDNM1L82,3396.373439
7P23526 S-adenosyl-L-homocysteine hydrolaseAHCY48,2555.92162486
8P04075 Fructose-bisphosphate aldolase AALDOA39,8518.305668723
9P04406 Glyceraldehyde-3-phosphate dehydrogenaseGAPDH36,2018.572755400
10P2262660S acidic ribosomal protein P0RPLP037,4648.971241238
11P45880Voltage-dependent anion-selective channel protein 2VDAC232,0607.49102991
12P63244Guanine nucleotide-binding protein subunit β-2-like 1GNB2L135,5117.601252101
13Q15056Eukaryotic translation initiation factor 4HEIF4H27,4256.67728125
14P04083Annexin A1Annexin I38,9186.5771245
15P52907F-actin-capping protein subunit α-1CAPZA133,0735.45848176
16Q16740Putative ATP-dependent
Clp protease proteolytic subunit, mitochondrial
CLPP30,4468.26210116
17P07355Annexin A2ANXA238,8087.574554885
18Q99497Protein DJ-1PARK720,0506.33133097
19P30048 Thioredoxin-dependent peroxide reductase, mitochondrialPRDX328,0177.67920115
20P04792Heat shock protein β-1HSPB122,8265.9863748
21P62158CalmodulinCALM116,8274.0963084
22P60981DestrinDSTN18,9508.0683391
23P00441Superoxide dismutaseSOD116,1545.701432140
24P30044Peroxiredoxin-5, mitochondrialPRDX522,3018.932856281
25Q04837Single-stranded DNA-binding protein, mitochondrialSSBP117,2499.5983276
26P07737Profilin-1PFN115,2168.44957135
27P61088 Ubiquitin-conjugating enzyme E2 NUBE2N17,1846.1373642
Downregulated
28P11142Heat shock cognate 71 kDa proteinHSPA871,0825.375028562
29P35232ProhibitinPHB29,8435.572951440
30P17987T-complex protein 1 subunit αTCP160,8195.802837170
31P30101Protein disulfide-isomerase A3PDIA357,1463.4771269
32P78371T-complex protein 1 subunit βCCT257,7946.013649446
33P14618Pyruvate kinase isozymes M1/M2PKM258,4707.962636309
34Q9BWD1Acetyl-CoA acetyltransferaseACAT241,8386.47124186
35P34949Mannose-6-phosphate isomeraseMPI47,1965.622738
36P09972 Fructose-bisphosphate aldolase CALDOC39,8306.411315205
37P68363Tubulin α-1BTUBA1B50,8044.943541513
38P50213Isocitrate dehydrogenase (NAD) subunit α, mitochondrialIDH3A40,0226.472249152
39P37837TransaldolaseTALDO137,6886.362337363
40P07195L-lactate dehydrogenase B chainLDHB36,9005.71124186
41O00764Pyridoxal kinasePDXK35,3085.751842175
42O0048726S proteasome non-ATPase regulatory subunit 14PSMD1434,7266.06146262
43P11177Pyruvate dehydrogenase E1 component subunit β, mitochondrialPDHB39,5506.201416186
44O00154Cytosolic acyl coenzyme A thioester hydrolaseACOT742,4548.851325177
45P15121Aldose reductaseAKR1B136,2306.51401162
46P00338L-lactate dehydrogenase A chainLDHA36,9508.444052319
47P31937 3-hydroxyisobutyrate dehydrogenase, mitochondrialHIBADH35,7058.381439
48P12004Proliferating cell nuclear antigenPCNA29,0924.574554559
49Q06830 Peroxiredoxin-1PRDX122,3248.2721860
50Q13162 Peroxiredoxin-4PRDX430,7495.8631429
51P00491Purine nucleoside phosphorylasePNP32,3256.454535
52P30086 Phosphatidylethanolamine-binding protein 1PEBP121,1587.013662432
53P60174Triosephosphate isomeraseTPI126,9386.451557136
54P49720Proteasome subunit β type-3PSMB323,2196.1473050
55Q15185Prostaglandin E synthase 3PTGES318,9714.3561890
56Q997143-hydroxyacyl-CoA dehydrogenase type-2HSD17B1027,1347.664273632
57P62826GTP-binding nuclear protein RanRAN24,5797.0141819
58P42126Enoyl-CoA δ isomerase 1, mitochondrialECI133,0808.805860
59P15531Nucleoside diphosphate kinase ANME117,3095.831959192
60P62937Peptidyl-prolyl cis-trans isomerase APPIA18,2297.682261327
61P16949StathminSTMN117,2925.761824608
62P24666Low molecular weight phosphotyrosine protein phosphataseACP118,4876.3042227
63Q9UHV9Prefoldin subunit 2PFDN216,6956.201946
64P62942Peptidyl-prolyl cis-trans isomeraseFKBP1A12,0007.8832726
65Q01469Fatty acid-binding protein, epidermalFABP515,4976.6093790
66O60925Prefoldin subunit 1PFDN114,2026.322946
67P35080Profilin-2PFN215,3786.551830179
68Q99584Protein S100-A13S100A1311,4645.9122228
69P04264Keratin, type II cytoskeletal 1KRT166,1708.1581128
70P31949Protein S100-A11S100A1111,8476.5641536
71P14174Macrophage migration inhibitory factorMIF12,6397.741617366

a For several proteins, a few isoforms were identified in the same individual;

b accession numbers were derived from the ExPASy database;

c theoretical molecular mass (kDa) and PI from the ExPASy database;

d probability-based MOWSE (molecular weight search) scores.

Table II

Proteins identified to be involved in the metabolic process.

Table II

Proteins identified to be involved in the metabolic process.

Spot no.Accession no.Protein nameAverage ratioSubcellular locationMain function
3P06733α-enolas10.54Cell membraneGlycolysis
7P23526 S-adenosyl-L-homocysteine hydrolase8.91CytoplasmControl of methylations
8P04075 Fructose-bisphosphate aldolase A23.57CytoplasmGlycolysis and gluconeogenesis
9P04406 Glyceraldehyde-3-phosphate dehydrogenase4.72CytoplasmGlycolysis
31P30101Protein disulfide-isomerase A30.23CytoplasmCysteine-type endopeptidase activity
33P14618Pyruvate kinase isozymes M1/M20.08CytoplasmGlycolysis
34Q9BWD1Acetyl-CoA acetyltransferase0.22CytoplasmAcetyl-CoA C-acetyltransferase activity
35P34949Mannose-6-phosphate isomerase0.73CytoplasmMannose-6-phosphate isomerase activity
36P09972 Fructose-bisphosphate aldolase C0.19CytoplasmGlycolysis and gluconeogenesis
38P50213Isocitrate dehydrogenase (NAD) subunit α, mitochondrial0.17MitochondrionTricarboxylic acid cycle
39P37837Transaldolase0.43CytoplasmPentose-phosphate pathway
40P07195L-lactate dehydrogenase B chain0.34Cytoplasm(S)-lactate + NAD+= pyruvate + NADH.
43P11177Pyruvate dehydrogenase E1 component subunit β, mitochondrial0.32MitochondrionPyruvate dehydrogenase (acetyl-transferring) activity
44O00154Cytosolic acyl coenzyme A thioester hydrolase0.37MitochondrionFatty-acyl-CoA binding
45P15121Aldose reductase0.31CytoplasmCatalytic efficiencies
46P00338L-lactate dehydrogenase A chain0.23CytoplasmL-lactate dehydrogenase activity
47P31937 3-hydroxyisobutyrate dehydrogenase, mitochondrial0.33Mitochondrion 3-hydroxyisobutyrate dehydrogenase activity
51P00491Purine nucleoside phosphorylase0.27CytoplasmImmune response
53P60174Triosephosphate isomerase0.47CytoplasmTriose-phosphate isomerase activity
55Q15185Prostaglandin E synthase 30.35CytoplasmMolecular chaperone
56Q997143-hydroxyacyl-CoA dehydrogenase type-20.41Mitochondrion 3-hydroxy-2-methylbutyryl-CoA dehydrogenase activity
58P42126Enoyl-CoA delta isomerase 1, mitochondrial0.38MitochondrionDodecenoyl-CoA delta-isomerase activity
65Q01469Fatty acid-binding protein, epidermal0.27CytoplasmHigh specificity for fatty acids

The identified proteins were divided into various groups based on their biological functions and subcellular localization. This implicated roles in metabolism (32%), protein folding (8%), translation (5%), protein modification (4%), cell proliferation (15%), apoptosis (10%), signal transduction (14%) and cell cytoskeleton (12%). The proteins were found to be located in the cytoplasm (57%), nucleus (11%), mitochondrion (15%), cell membrane (10%) or were secreted (12%) (Fig. 2). For a macroscopic presentation, cluster maps and protein interaction and function networks were generated using Cluster or the KEGG-based software tool Cytoscape, respectively. Twenty-three proteins, accounting for 32% of the proteins identified, were found to be associated with metabolism regulation. The metabolism-regulating proteins were grouped in different clusters. Pyruvate kinase isozymes M1/M2 were found to show one of the most significant differences in expression between SKOV3 cells treated with or without Orlistat. It was downregulated more than 10-fold in SKOV3 cells treated with Orlistat compared to those without Orlistat, and MS/MS analysis revealed 15 matched peptides with 36% sequence coverage and a MOWSE score of 309 (Fig. 3).

Proteomic validation of identified proteins

The expression level of PKM2 was further validated by western blotting. Consistent with the observations in 2-DE analysis, PKM2 was downregulated in the Orlistat-treated SKOV3 cells compared with the parental SKOV3 cells. A similar change in the expression level of FASN was detected in SKOV3 cells treated with Orlistat (Fig. 5).

Discussion

Altered expression of lipid metabolic enzymes is a feature of various types of cancer, including those that develop in ovarian tissues (24). Highly proliferating cancer cells need to synthesize fatty acids de novo to continually provide lipids for membrane production. The synthesized fatty acids are also used for energy production through β-oxidation and lipid modification of proteins (Fig. 4). FASN, one of the key enzymes involved in de novo fatty-acid synthesis, was found to be overexpressed in various human types of cancer, including prostate, ovary, colon, and lung (25). FASN has been found to be essential for ovarian cancer cell survival and inhibition of FASN activity has been shown to have potential chemo-preventive (26) and therapeutic applications (27).

In this study, we found that treatment with Orlistat, an inhibitor of FASN, promoted the apoptosis of SKOV3 cells (Fig. 1). We confirm the inhibitory effect of Orlistat on FASN by western blot analysis using the ovarian cancer cells (SKOV3) as a model, and we found that FASN was 2-fold downregulated after treatment with Orlistat. As shown in Fig. 5, we employed a 2-DE-based proteomics approach to annotate the altered proteins in the SKOV3 cells prior to and following treatment with Orlistat. Our proteomic analysis revealed a total of 71 differentially expressed proteins, which were associated with cell metabolism, proliferation and/or apoptosis.

Among them, Profilin 1, a member of the profilin family, also known as PFN1, was ubiquitous and upregulated more than 10-fold in SKOV3 cells after treatment with Orlistat. PFN1 was found to be involved in multiple cell behaviors, such as cell adhesion, growth, proliferation and signal transduction (34,35). Moreover, 23 proteins were found differentially expressed related to metabolism. Among them, pyruvate kinase (PK), a rate-limiting enzyme during glycolysis, catalyzes the production of pyruvate and adenosine 5′-triphosphate (ATP) from phosphoenolpyruvate (PEP) and adenosine 5′-diphosphate (ADP) (28). Four mammalian PK isoenzymes (M1, M2, L and R) were found in normal adult cells. By contrast, PKM2 is found predominantly in the fetus as well as in tumor cells, where the abundance of other isoforms of PK is low. PKM2 can exist in either active tetramers or inactive dimers, but in tumor cells it predominantly occurs in dimers with low activity (29). Cancer cells universally express the M2 isoform of the glycolytic enzyme pyruvate kinase (PKM2), and previous studies have demonstrated that PKM2 expression is necessary for aerobic glycolysis and cell proliferation in vivo (28,30). Knockdown of PKM2 using RNA interference significantly impairs cell growth in tissue culture, inhibition of PKM2 with peptide aptamers inhibits cell proliferation, and PKM2 expression is necessary for both aerobic glycolysis and tumor growth in vivo (31,32). It has been proven that the downregulation of PKM2 activity in cancer cells aids in shunting key glycolytic intermediates toward pathways where they, in turn, are utilized as precursors for lipid, amino acid and nucleic acid synthesis. Therefore, the downregulation of PKM2 activity provides a purposeful divergence from catabolic metabolism aimed at energy production toward an anabolic state aimed at providing the needed resources for rapid cellular construction (33). Research has also shown that PKM2 plays a general role in caspase- and Bcl-independent apoptosis, thereby validating PKM2 as a promising, generally relevant target for the development of anticancer therapies with broad efficacy (34). In our study, PKM was downregulated more than 10-fold, confirming our hypothesis that Orlistat has antitumor abilities. Furthermore, significant downregulation of PKM2 after treatment with Orlistat was confirmed in the ovarian cancer cell line SKOV3 cells by western blot analysis.

In conclusion, using proteomic tools, we identified 71 differentially expressed proteins following Orlistat treatment of ovarian cancer cells. The functions of the differentially expressed proteins were correlated to apoptosis and/ or anti-proliferation cellular processes. These results support the hypothesis that Orlistat is a potential inhibitor of ovarian cancer and can be used as a novel assistant antitumor agent, combined with conventional surgical resection and chemotherapy.

Abbreviations:

2-DE

two-dimensional polyacrylamide gel electrophoresis;

MALDI-Q-TOF

matrix-assisted laser desorption ionization quadrupole time-of-flight;

MOWSE

molecular weight search;

ALODA

aldolase A;

LDHA

L-lactate dehydrogenase A chain;

KPYM

pyruvate kinase muscle isozyme;

MS

mass spectrometry;

MTT

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide;

PI

propidium iodide;

CAPS

calcyphosine;

FAS

fatty-acid synthase

Acknowledgements

This work was supported by the National Key Basic Research Program (973 Program) of China (2011CB910703).

References

1 

Clemens MJ: Targets and mechanisms for the regulation of translation in malignant transformation. Oncogene. 23:3180–3188. 2004. View Article : Google Scholar : PubMed/NCBI

2 

Bull JH, Ellison G, Patel A, et al: Identification of potential diagnostic markers of prostate cancer and prostatic intraepithelial neoplasia using cDNA microarray. Br J Cancer. 84:1512–1519. 2001. View Article : Google Scholar : PubMed/NCBI

3 

Rashid A, Pizer ES, Moga M, et al: Elevated expression of fatty acid synthase and fatty acid synthetic activity in colorectal neoplasia. Am J Pathol. 150:201–208. 1997.PubMed/NCBI

4 

Gansler TS, Hardman W III, Hunt DA, et al: Increased expression of fatty acid synthase (OA-159) in ovarian neoplasms predicts shorter survival. Hum Pathol. 18:686–692. 1997. View Article : Google Scholar : PubMed/NCBI

5 

Gansler TS, Hardman W III, Hunt DA, et al: Immunohistochemical expression and prognostic significance of FAS and GLUT1 in bladder carcinoma. Anticancer Res. 23:335–339. 2003.PubMed/NCBI

6 

Nemoto T, Terashima S, Kogure M, et al: Overexpression of fatty acid synthase in oesophageal squamous cell dysplasia and carcinoma. Pathobiology. 69:297–303. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Kusakabe T, Nashimoto A, Honma K and Suzuki T: Fatty acid synthase is highly expressed in carcinoma, adenoma and in regenerative epithelium and intestinal metaplasia of the stomach. Histopathology. 40:71–79. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Piyathilake CJ, Frost AR, Manne U, et al: The expression of fatty acid synthase (FAS) is an early event in the development and progression of squamous cell carcinoma of the lung. Hum Pathol. 31:1068–1073. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Pizer ES, Lax SF, Kuhajda FP, Pasternack GR and Kurman RJ: Fatty acid synthase expression in endometrial carcinoma: correlation with cell proliferation and hormone receptors. Cancer. 83:528–537. 1998. View Article : Google Scholar : PubMed/NCBI

10 

Alò PL, Visca P, Marci A, Mangoni A, Botti C and Di Tondo U: Expression of fatty acid synthase (FAS) as a predictor of recurrence in stage I breast carcinoma patients. Cancer. 77:474–482. 1996.PubMed/NCBI

11 

Pizer ES, Wood FD, Heine HS, Romantsev FE, Pasternack GR and Kuhajda FP: Inhibition of fatty acid synthase delays disease progression in a xenograft model of ovarian cancer. Cancer Res. 56:189–193. 1996.PubMed/NCBI

12 

Alò PL, Visca P, Botti C, et al: Immunohistochemical expression of human erythrocyte glucose transporter and fatty acid synthase in infiltrating breast carcinomas and adjacent typical/ atypical hyperplastic or normal breast tissue. Am J Clin Pathol. 116:129–134. 2001.PubMed/NCBI

13 

Alò PL, Visca P, Framarino ML, et al: Immunohistochemical study of fatty acid synthase in ovarian neoplasms. Oncol Rep. 7:1383–1388. 2000.PubMed/NCBI

14 

Costello LC and Franklin RB: ‘Why do tumour cells glycolyse?’: from glycolysis through citrate to lipogenesis. Mol Cell Biochem. 280:1–8. 2005.

15 

De Schrijver E, Brusselmans K, Heyns W, Verhoeven G and Swinnen JV: RNA interference-mediated silencing of the fatty acid synthase gene attenuates growth and induces morphological changes and apoptosis of LNCaP prostate cancer cells. Cancer Res. 63:3799–3804. 2003.PubMed/NCBI

16 

Menendez JA, Vellon L and Lupu R: Orlistat: From antiobesity drug to anticancer agent in Her-2/neu (erbB-2)-overexpressing gastrointestinal tumors? Exp Biol Med (Maywood). 230:151–154. 2005.PubMed/NCBI

17 

Pemble CW IV, Johnson LC, Kridel SJ and Lowther WT: Crystal structure of the thioesterase domain of human fatty acid synthase inhibited by Orlistat. Nat Struct Mol Biol. 14:704–709. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Knowles LM, Axelrod F, Browne CD and Smith JW: A fatty acid synthase blockade induces tumor cell-cycle arrest by down-regulating Skp2. J Biol Chem. 279:30540–30545. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T and Thun MJ: Cancer statistics. CA Cancer J Clin. 58:71–96. 2008.

20 

Raki M, Rein DT, Kanerva A and Hemminki A: Gene transfer approaches for gynecological diseases. Mol Ther. 14:154–163. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Liu R, Li Z, Bai S, et al: Mechanism of cancer cell adaptation to metabolic stress: proteomics identification of a novel thyroid hormone-mediated gastric carcinogenic signaling pathway. Mol Cell Proteomics. 8:70–85. 2009. View Article : Google Scholar

22 

Tong A, Wu L, Lin Q, et al: Proteomics analysis of cellular protein alterations using a hepatitis B virus-producing cellular model. Proteomics. 8:2012–2023. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Liu R, Wang K, Yuan K, et al: Integrative oncoproteomics strategies for anticancer drug discovery. Expert Rev Proteomics. 7:411–429. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Medes G, Thomas A and Weinhouse S: Metabolism of neoplastic tissue. IV: A study of lipid synthesis in neoplastic tissue slices in vitro. Cancer Res. 13:27–29. 1953.PubMed/NCBI

25 

Witkowski A, Joshi AK and Smith S: Coupling of the de novo fatty acid biosynthesis and lipoylation pathways in mammalian mitochondria. J Biol Chem. 282:14178–14185. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Chirala SS and Wakil SJ: Structure and function of animal fatty acid synthase. Lipids. 39:1045–1053. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Kuhajda FP, Jenner K, Wood FD, Hennigar RA, Jacobs LB, Dick JD and Pasternack GR: Fatty acid synthesis: a potential selective target for antineoplastic therapy. Proc Natl Acad Sci USA. 91:6379–6383. 1994. View Article : Google Scholar : PubMed/NCBI

28 

Christofk HR, Vander Heiden MG, Harris MH, et al: The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature. 452:230–233. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Guminska M, Ignacak J, Kedryna T and Stachurska MB: Tumor-specific pyruvate kinase isoenzyme M2 involved in biochemical strategy of energy generation in neoplastic cells. Acta Biochim Pol. 44:711–724. 1997.PubMed/NCBI

30 

Mazurek S, Boschek CB, Hugo F and Eigenbrodt E: Pyruvate kinase type M2 and its role in tumor growth and spreading. Semin Cancer Biol. 15:300–308. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Dombrauckas JD, Santarsiero BD and Mesecar AD: Structural basis for tumor pyruvate kinase M2 allosteric regulation and catalysis. Biochemistry. 44:9417–9429. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Spoden GA, Mazurek S, Morandell D, et al: Isotype-specific inhibitors of the glycolytic key regulator pyruvate kinase subtype M2 moderately decelerate tumor cell proliferation. Int J Cancer. 123:312–321. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Steták A, Veress R, Ovádi J, Csermely P, Kéri G and Ullrich A: Nuclear translocation of the tumor marker pyruvate kinase M2 induces programmed cell death. Cancer Res. 67:1602–1608. 2007.PubMed/NCBI

34 

Janke J, Schlüter K, Jandrig B, et al: Suppression of tumorigenicity in breast cancer cells by the microfilament protein profilin 1. J Exp Med. 191:1675–1686. 2000. View Article : Google Scholar : PubMed/NCBI

35 

Wu N, Zhang W, Yang Y, et al: Profilin 1 obtained by proteomic analysis in all-trans retinoic acid-treated hepatocarcinoma cell lines is involved in inhibition of cell proliferation and migration. Proteomics. 6:6095–6106. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August 2012
Volume 41 Issue 2

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
Huang H, Tang J, Zhou S, Yi T, Peng H, Shen G, Xie N, Huang K, Yang T, Wu J, Wu J, et al: Orlistat, a novel potent antitumor agent for ovarian cancer: proteomic analysis of ovarian cancer cells treated with Orlistat. Int J Oncol 41: 523-532, 2012
APA
Huang, H., Tang, J., Zhou, S., Yi, T., Peng, H., Shen, G. ... Zhao, X. (2012). Orlistat, a novel potent antitumor agent for ovarian cancer: proteomic analysis of ovarian cancer cells treated with Orlistat. International Journal of Oncology, 41, 523-532. https://doi.org/10.3892/ijo.2012.1465
MLA
Huang, H., Tang, J., Zhou, S., Yi, T., Peng, H., Shen, G., Xie, N., Huang, K., Yang, T., Wu, J., Huang, C., Wei, Y., Zhao, X."Orlistat, a novel potent antitumor agent for ovarian cancer: proteomic analysis of ovarian cancer cells treated with Orlistat". International Journal of Oncology 41.2 (2012): 523-532.
Chicago
Huang, H., Tang, J., Zhou, S., Yi, T., Peng, H., Shen, G., Xie, N., Huang, K., Yang, T., Wu, J., Huang, C., Wei, Y., Zhao, X."Orlistat, a novel potent antitumor agent for ovarian cancer: proteomic analysis of ovarian cancer cells treated with Orlistat". International Journal of Oncology 41, no. 2 (2012): 523-532. https://doi.org/10.3892/ijo.2012.1465