You are on page 1of 10

Journal of Proteomics 151 (2017) 2–11

Contents lists available at ScienceDirect

Journal of Proteomics

journal homepage: www.elsevier.com/locate/jprot

Proteomic analysis of ovarian cancer cells during


epithelial-mesenchymal transition (EMT) induced by epidermal growth
factor (EGF) reveals mechanisms of cell cycle control
Mariana Lopes Grassi a,b, Camila de Souza Palma a,b, Carolina Hassibe Thomé a,b, Guilherme Pauperio Lanfredi a,b,
Aline Poersch a, Vitor Marcel Faça a,b,⁎
a
Dept. Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
b
Cell-Based Therapy Center, Ribeirão Preto Blood Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil

a r t i c l e i n f o a b s t r a c t

Article history: Epithelial to mesenchymal transition (EMT) is a well-orchestrated process that culminates with loss of epithelial
Received 29 January 2016 phenotype and gain of a mesenchymal and migratory phenotype. EMT enhances cancer cell invasiveness and
Received in revised form 20 May 2016 drug resistance, favoring metastasis. Dysregulation of transcription factors, signaling pathways, miRNAs and
Accepted 7 June 2016
growth factors including EGF, TGF-beta and HGF can trigger EMT. In ovarian cancer, overexpression of the
Available online 6 July 2016
EGFR family is associated with more aggressive clinical behavior. Here, the ovarian adenocarcinoma cell line
Keywords:
Caov-3 was induced to EMT with EGF in order to identify specific mechanisms controlled by this process.
Epithelial to mesenchymal transition Caov-3 cells induced to EMT were thoroughly validated and a combination of subcellular proteome enrichment,
Epidermal growth factor GEL-LC-MS/MS and SILAC strategy allowed consistent proteome identification and quantitation. Protein network
Cell cycle analysis of differentially expressed proteins highlighted regulation of metabolism and cell cycle. Activation of rel-
Ovarian cancer evant signaling pathways, such as PI3K/Akt/mTOR and Ras/Erk MAPK, in response to EGF-induced EMT was val-
idated. Also, EMT did not affected the proliferation rate of Caov-3 cells, but led to cell cycle arrest in G1 phase
regulated by increased levels of p21Waf1/Cip1, independently of p53. Furthermore, a decrease in G1 and G2
checkpoint proteins was observed, supporting the involvement of EGF-induced EMT in cell cycle control.
Biological significance: Cancer is a complex multistep process characterized by accumulation of several hallmarks
including epithelial to mesenchymal transition (EMT), which promotes cellular and microenvironmental chang-
es resulting in invasion and migration to distant sites, favoring metastasis. EMT can be triggered by different ex-
tracellular stimuli, including growth factors such as EGF. In ovarian cancer, the most lethal gynecological cancer,
overexpression of the EGFR family is associated with more aggressive clinical behavior, increasing mortality rate
caused by metastasis. Our proteomic data, together with specific validation of specific cellular mechanisms dem-
onstrated that EGF-induced EMT in Caov-3 cells leads to important alterations in metabolic process (protein syn-
thesis) and cell cycle control, supporting the implication of EGF/EMT in cancer metastasis, cancer stem cell
generation and, therefore, poor prognosis for the disease.
© 2016 Elsevier B.V. All rights reserved.

1. Introduction inducing the acquisition of a more motile mesenchymal phenotype


that progresses to detachment from the main epithelial tumor, invasion
Cancer is a complex multistep process characterized by accumula- of the surrounding stroma and culminating with cells entering in the
tion of several genetic and epigenetic alterations that stimulate in ma- circulatory system to finally metastasize to distant sites [2].
lignant cells a number of hallmarks that promote uncontrolled Initially described as a distinct cell differentiation process in embryo-
proliferation, migration and invasion [1]. During malignant tumor pro- genesis, EMT involves the loss of epithelial markers - such as the adher-
gression, epithelial to mesenchymal transition (EMT) arises as a crucial ents junction proteins E-cadherin and β-catenin, and the tight junction
mechanism to enhance cancer cell invasiveness and drug resistance, proteins Claudin and Occludin - cytoskeleton reorganization and gain of
mesenchymal markers, including fibroblast-like cell morphology and
other proteins such as N-cadherin, vimentin, fibronectin and matrix me-
⁎ Corresponding author at: Department of Biochemistry and Immunology, Ribeirão
Preto Medical School, University of São Paulo, Av. Bandeirantes, 3900 - Monte Alegre,
talloproteinase [3,4]. These morphogenetic changes are well-orches-
CEP 14049-900 Ribeirão Preto, SP, Brazil. trated by a number of master transcription regulators that involve
E-mail address: vitor.faca@fmrp.usp.br (V.M. Faça). three families of transcription factors: the zinc finger proteins Snail

http://dx.doi.org/10.1016/j.jprot.2016.06.009
1874-3919/© 2016 Elsevier B.V. All rights reserved.
M.L. Grassi et al. / Journal of Proteomics 151 (2017) 2–11 3

and Slug, the zinc finger and homeodomain proteins Zeb1 and Zeb2 and 2.3. Western Blot
the helix-loop-helix proteins Twist (TWIST1 and TWIST2). All together,
these transcription factors repress epithelial markers, specially the ex- Caov-3 cells were washed in cold PBS and lysed with CelLytic
pression of E-cadherin, as well as enhance the expression of mesenchy- (Sigma-Aldrich) containing the 5% (v/v) protease inhibitor cocktail
mal genes [5] (Sigma-Aldrich). Three sonication cycles were carried out for 5 min
Several studies have shown that dysregulation of transcription fac- each in an ultrasonic bath (Unique, São Paulo, SP, Brazil) with cooled
tors, signaling pathways, miRNAs and growth factors can trigger the ini- water. Lysates were centrifuged at 20,000 ×g for 30 min at 4 °C, to ob-
tiation of EMT. Growth factors such as transforming growth factor tain the total cell lysates. The protein concentration was determined ac-
(TGF), hepatocyte growth factor (HGF) and epidermal growth factor cording to the Bradford method (Bio-Rad, Hercules, CA, USA). Proteins
(EGF) have been widely associated with EMT induction, since these were submitted to SDS-PAGE and transferred to polyvinylidene fluoride
stimuli can be directly produced by tumor-associated stroma cells. In membranes (GE Healthcare, Life Sciences, Pittsburgh, PA, USA). Mem-
addition to stimulation by growth factors, the effective implementation branes were blocked with 3% bovine serum albumin (BSA) in 0.05%
of EMT is also dependent on a variety of signal transduction pathways, Tween-TBS and incubated with the specific antibodies. Rabbit anti-
including PI3K, MAPK, Wnt/β-catenin and Notch signaling [6,7]. Vimentin (#5741), rabbit anti-E-cadherin (#3195), rabbit anti-N-
EGF has contributed effectively as a potent EMT inducer in different cadherin (#4061), mouse anti-Snail (#3895), rabbit anti-EGFR
types of solid tumors, including gynecological cancer such as endome- (#2646), rabbit anti-phospho-EGFR (Tyr1068) (#3777), rabbit anti-
trial, ovarian and cervical cancer, significantly favoring the poor progno- p21Waf1/Cip1 (#2947), rabbit anti-p53 (#2527), rabbit anti-phospho-Rb
sis and increasing mortality rate caused by metastasis. In ovarian cancer, (Ser807/811) (#9308), mouse anti-cdc2 (#9116), rabbit anti-cyclin
the most lethal gynecological cancer, overexpression of the EGFR family D1 (#2978), rabbit anti-phospho-cyclin D1 (#3300), rabbit anti-mTOR
has been associated with more aggressive clinical behavior [8,9]. (#2983), rabbit anti-phospho-Erk 1/2 (Thr202/Tyr204) (#4370), rabbit
In addition, it has been shown that EGF stimulation can induce EMT anti-Akt (#9272), rabbit anti-phospho-Akt (Ser 473) (#4060), rabbit
in ovarian surface epithelium (OSE) through inhibition of keratin ex- anti-GAPDH (#2118), and horseradish peroxidase-conjugated second-
pression and enhancing of pro-MMP 2/9 expression, with activation of ary antibody goat anti-mouse IgG were purchased from Cell Signaling
ERK and integrin-linked kinase (ILK) pathways. Moreover, EGF binds (Beverly, MA, USA). The protein-antibody complex was detected using
to the EGF receptor (EGFR), inducing EMT through phosphorylation of the ECL™ Prime Western Blotting Detection Reagents (GE Healthcare,
the Janus kinase/signal transducer and activator of transcription (JAK/ Life Sciences) and chemiluminescent was detected using a CCD-camera
STAT3) pathway with involvement of ERK, increasing the based system (ImageQuant 350, GE Healthcare, Uppsala, Sweden).
chemoresistance in ovarian carcinoma cell lines [10,11].
In order to obtain a proteomic perspective about the complex molec- 2.4. Multiple reaction monitoring (MRM) analysis
ular mechanisms of EMT, we performed a multiple-compartment anal-
ysis of the proteomic alterations during EMT induced by EGF in the Aliquots of 100 μg of proteins from total cell lysates, quantitated by
ovarian adenocarcinoma cell line Caov-3. Here, we report that EGF-in- Bradford assay, were diluted in 50 μl Urea 8 M, Tris-HCl 0.1 M, pH 8.5.
duced EMT activated signaling pathways relevant for growth and cell Protein cysteine residues were reduced with 50 μg dithiothreitol at
proliferation, represented by increased phosphorylation of Akt, ERK1/2 37 °C for 30 min and then alkylated with 250 μg iodoacetamide for
and S6 ribosomal protein, which leads to alterations in the cell prolifer- 30 min, protected from light. Protein solution was then diluted in
ation pattern. We also show that the underlying mechanisms of EMT in- 740 μl Tris-HCl 0.2 M, pH 8.0 to reduce urea concentration (b1.5 M)
volve cell cycle arrest through activation of p21Waf1/Cip1 independent of and in-solution trypsin digestion was performed by adding 4 μg trypsin
p53 with decreased phosphorylation status of checkpoint proteins such (Promega), incubated overnight at 37 °C. Samples were then desalted
as cyclin D1, Rb and CDK1/cdc2. Altogether, these results not only con- using solid-phase extraction columns Oasis HLB (Waters) according to
tribute for broadening the comprehension of the molecular basis of the manufacturer's instructions. Peptides were eluted in 52.5% ACN/
EMT, but also highlight important pathways that can serve as targets 0.1% formic acid and dried in SpeedVac (Thermo Scientific, Marietta,
to control cancer progression and metastasis. OH, USA). Before MRM analysis, samples were reconstituted in 50 μl of
5% acetonitrile/0.1% formic acid solution, agitated thoroughly, centri-
fuged for 20 min at 20,000 ×g and then transferred to injection vials.
2. Material and methods Each sample was injected in triplicate through the LC-MS/MS Xevo
TQs system (Waters). Chromatographic separation was performed in a
2.1. Cell culture UPLC (I-class, Waters) using a C18 column (1.8 μm particle size, 100 Å
pore size, 1 mm × 150 mm, Waters) in a linear gradient of 5 to 30% ace-
Human ovarian adenocarcinoma cell line Caov-3 was acquired from tonitrile over 15 min at 100 μl/min in a formic acid:acetonitrile:water
the ATCC and cultured in Dulbecco's Modified Eagle Medium (DMEM, solvent system. Detection of proteotypic peptides was programmed in
Life Technologies, Carlsbad, CA, USA) supplemented with 10% fetal bo- a scheduled method using 3–5 transitions per peptide and monitoring
vine serum (FBS, Thermo Scientific, Marietta, OH, USA), 100 U/ml peni- windows of 2 min. Detailed information about proteotypic peptides,
cillin and 100 μg/ml streptomycin (Life Technologies). Cells were transitions, collision energies and dwell times are presented in Supple-
maintained at 37 °C in a humidified incubator in an atmosphere of 5% mentary Table 1. Both method development and data analysis were
CO2. conducted using Skyline software [12].

2.5. Immunofluorescence microscopy


2.2. EMT induction
Caov-3 cells grown on glass coverslips were rinsed briefly in PBS,
Caov-3 cells were seeded at 1.5 × 106 cells on a 100 mm plate in fixed in PBS containing 3% formaldehyde, and incubated in a solution
DMEM supplemented with 10% FBS. Cells were washed in pre-warmed containing 0.3% triton x-100, 22.52 mg/ml glycine and 1% BSA in PBS
PBS and the media was replaced by DMEM without FBS. After 24 h, for 1 h at room temperature. Cells were incubated overnight with spe-
DMEM without FBS and containing 10 ng/ml EGF (R&D Systems) was cific antibodies. Rabbit anti-E-cadherin (#3195) and rabbit anti-
added and cells were maintained in culture for 96 h to induce EMT. Vimentin (#9854) were purchased from Cell Signaling (Beverly, MA,
Each 24 hour media was replaced by a new DMEM solution containing USA). Mouse anti-N-cadherin (#33-3900) was purchased from Thermo
10 ng/ml EGF was replaced. Fisher Scientific (Waltham, MA, USA). Secondary antibody anti-rabbit
4 M.L. Grassi et al. / Journal of Proteomics 151 (2017) 2–11

IgG (Alexa Fluor® 488-conjugated) (#4412) was purchased from Cell reduce disulfide bonds, and alkylated with iodoacetamide (5 mg/mg
Signaling (Beverly, MA, USA). Images were captured on Leica CTR of total protein). Samples were then separated using a 10% SDS-PAGE
6000 Fluorescence, with sequential fluorophore excitation. (Bio-Rad, Hercules, CA, USA). Gel lanes corresponding to each fraction
were cut into 5 pieces of approximately 1.2 cm each, washed, and
2.6. RTK activity array digested with trypsin as described elsewhere [15]. Tryptic peptides
were successively extracted with 0.1% formic acid and 50% acetonitrile,
Caov-3 cells were washed in cold PBS and lysed with buffer from and then 70% acetonitrile, and dried by SpeedVac. Peptide mixtures
PathScan® RTK Signaling Antibody Array kit (Cell Signaling) containing were dissolved in 10 μl 5% acetonitrile/0.1% formic and desalted using
5% (v/v) protease inhibitor cocktail (Sigma-Aldrich). To extract proteins, ZipTip columns (Supelco, Sigma-Aldrich) according to the
three sonication cycles were carried out for 5 min each in an ultrasonic manufacturer's instructions. Samples were eluted in 52.5% acetoni-
bath (Unique, São Paulo, SP, Brazil) with cooled water. Lysates were trile/water in 0.1% formic acid, dried again, and re-suspended in 15 μl
centrifuged at 20,000 ×g for 30 min at 4 °C, and protein concentration 5% acetonitrile/0.1% formic acid for LC-MS analysis. Samples were ana-
was determined according to the Bradford method (Bio-Rad, Hercules, lyzed by LC-MS/MS using an LTQ-Orbitrap Velos mass spectrometer
CA, USA). 50 μg aliquots of total cell extracts were used, according to coupled with a nano-LC (EASY-nLC II, ThermoScientific). Chromatogra-
the manufacturer's instructions. The protein-antibody complex was de- phy was performed using an in-house packed 75-μm inner diameter
tected using the ECL Western Blotting Detection Reagents (GE (New Objective) × 25-cm long C18 column packed with Magic C18
Healthcare, Life Sciences) and signals were detected using a CCD-Cam- resin, at 600 nl/min with 90 min linear gradients from 5 to 40% acetoni-
era (Image Quant LAS 4000 mini, Uppsala, Sweden). trile in 0.1% formic acid. MS/MS scans of the ten most abundant doubly
or triply charged ions in the FT-MS scan were selected for data-depen-
2.7. SILAC labeling dent analysis in the linear ion-trap. Peptides and proteins were identi-
fied with a local installation of the Labkey Server (2013.3), using the
Caov-3 cells were cultured in SILAC DMEM (SILAC™ Advanced X!Tandem search engine (2013.2.01 release) [16], and PeptideProphet
DMEM, Thermo Fisher Scientific) containing heavy lysine (13C6-L-ly- [17] and ProteinProphet [18] algorithms for statistical validation of
sine) (0.1 mg/ml) and supplemented with 10% dialyzed FBS, 2 mM L- data and protein grouping. MS data were searched against a human pro-
glutamine, 4.5 mg/ml D-glucose and 0.01% (w/v) penicillin/streptomy- teome database (Uniprot April 2014 - 68,561 entries). Search parame-
cin. To incorporate heavy lysine, cells were incubated for more than ters for tryptic peptides included up to two miss-cleavages, mass
five passages and the incorporation rate was measured by a MRM meth- tolerance of 0.5 Da for fragment ions, fixed cysteine modification with
od that quantifies 13C6-L-lysine peptides of actin-beta (ACTB) protein. carbamidomethylation (+ 57.02146), variable methionine oxidation
Incorporation was superior to 96%. (+15.99491), and variable lysine modification (+6.020129) to account
for both heavy and light SILAC labels. Only peptides with a
2.8. Subcellular fractionation PeptideProphet score above 0.9 were considered for protein quantifica-
tion. The list of proteins was generated with a ProteinProphet cut-off
Nuclear, cytoplasmic and membrane fractions were obtained by value of 0.9, representing an overall protein false discovery rate of ap-
using a successive detergent extraction method as described by Adam proximately 1% based on the ProteinProphet estimate. Proteins were
et al. [13] with modifications. Caov-3 (1 × 107) cells were washed quantitated as previously described, using the Q3 algorithm (1.22a re-
twice with PBS and lysed with 100 μl hypotonic buffer M (50 mM lease) to measure SILAC peak intensities [19,20].
HEPES pH 7.4, 10 mM NaCl, 5 mM MgCl2, 0.1 mM EDTA, 1 mM
Na3VO4, 1 mM NaF and 1 mM Na4P2O7 ∙ 10H2O) containing 5% (v/v) 2.10. Proteomic data analysis
protease inhibitor cocktail (product number P8340; Sigma-Aldrich).
Samples were vortexed for 20 s, passed through a 25-gauge needle The proteomic data was evaluated and correlated with several dif-
(20 times) and homogenized (D-130 tissue homogenizer, Biosystems, ferent bioinformatics tools. Protein-protein interactions and networks
São José dos Pinhais, PR, Brazil) for 1 min. At this point, equal amounts were evaluated using STRING-DB (string-db.org) [21]. Gene ontology
of protein from Caov-3 cells induced to EMT/non-induced and Caov-3 annotation was obtained using PANTHER tool (phanterdb.org) [22].
cells cultured in SILAC containing heavy lysine (13C6-L-lysine) were Protein expression information based on immunohistochemistry (IHC)
mixed. The nuclear pellet was obtained by centrifugation at 500 × g for normal and cancer tissues was obtained from ProteinAtlas
for 20 min at 4 °C. Supernatant was centrifuged at 16,000 × g for (proteinatlas.org) [23].
15 min at 4 °C, collected, and designated as the cytoplasmic fraction.
The pellet was treated with buffer A (25 mM 2-(N-morpholino)- 2.11. Cell cycle and proliferation rate
ethanesulfonic acid, 150 mM NaCl, pH 6.5) and combined with equal
volume of buffer A containing 2% Triton X-100 (v/v) with protease Cell cycle analysis was assessed using the DNA-binding dye
and phosphatase inhibitors. After 60 min of incubation, samples were propidium iodide (PI) (Sigma-Aldrich) and cell proliferation rate was
centrifuged at 16,000 ×g for 20 min at 4 °C. The supernatant was collect- measured by monitoring the incorporation of 5-bromo-2′-deoxyuridine
ed and designated as the membrane fraction. Protein extraction of the (BrdU) (Invitrogen, Thermo Fisher Scientific), according to the
nuclear pellet was performed with lysis solution containing 8 M urea, manufacturer's instructions. Both assays were analyzed by flow cytom-
2% CHAPS and protease inhibitor cocktail. Three sonication cycles etry (BD FACSCanto™, BD Biosciences).
were carried out for 5 min each in an ultrasonic bath (Unique, São
Paulo, SP, Brazil) with cooled water. Samples were centrifuged at 3. Results
20,000 ×g for 30 min at 4 °C and supernatant was collected and stored
at −80 °C. Western Blot analysis was performed to determine the en- 3.1. EGF receptor (EGFR) activation in Caov-3 cells
richment efficiency.
Since studies have reported a significant association between in-
2.9. In-gel digestion and LC-MS/MS analysis creased EGFR expression/phosphorylation and advanced stages of
OvCa in patients, we aimed to evaluate the activation of EGFR specifical-
50 μg of the cytoplasmic and nuclear fractions and 30 μg of the mem- ly in Caov-3 ovarian cancer cells using initially a brief stimulation with
brane fraction were dissolved in 30 μl Laemmli sample buffer [14] con- EGF. Although Caov-3 cells exhibited a basal phosphorylation of EGFR,
taining dithiothreitol (1 mg/mg of total protein), boiled for 5 min to the stimulation with EGF was able to increase the EGFR phosphorylation
M.L. Grassi et al. / Journal of Proteomics 151 (2017) 2–11 5

after 15 min even at the lowest concentration of 1 ng/ml, while the total images showed the dynamic change of Vimentin filaments extended
amount of EGFR remained unchanged (Fig. 1A). On the other hand, throughout the entire spindle-shaped cell after EGF-induced EMT,
MRM analysis demonstrated that only after 96 h of stimulation with with higher fluorescent intensity than the control cells, as well as a
10 ng/ml EGF, Caov-3 cells showed a significant increase of EGFR abun- weaker and discontinuous distribution of E-cadherin near the cell mem-
dance (Fig. 1B). Concomitantly, the proteins Ephrins A2, A3 and B2, brane with a lesser intensity than the control cells (Fig. 2C).
which compose the largest subfamily of receptor protein-tyrosine ki-
nases (RTK), together with the proto-oncogene tyrosine-protein kinase 3.3. Proteomic profile of EGF-induced EMT in Caov-3 cells
Src, also showed high levels of phosphorylation in response to EGF ex-
posure in Caov-3 cells, based on our RTK activity array analysis (Fig. After proper confirmation that EGF induces molecular and cellular
1C). Overall, the phosphorylation profiles of these proteins indicate changes associated with EMT in Caov-3 cells, an in-depth quantitative
that the Caov-3 cell line is functional and responsive to the growth fac- proteomic analysis was performed to elucidate in more detail the coor-
tor EGF. dinated protein changes as well as cellular process altered during the
EMT. Initially, conventional SILAC strategy, containing dialyzed fetal bo-
3.2. Morphological and functional evaluation of EMT induced by EGF in vine serum, was adopted for quantitative analysis. However Caov-3 cells
Caov-3 cells grown in SILAC media had a significantly different pattern EMT when
compared to those induced to EMT in regular DMEM media. Therefore,
In order to ensure EMT induction by EGF in Caov-3 ovarian cancer we designed a strategy based on mixing Caov-3 cells induced to EMT (or
cells, specific tools were applied to characterize the changes observed the corresponding control not induced to EMT) grown in regular DMEM
during this process. Contrast-phase microscopy showed clearly mor- media containing natural (“light”) lysine, with reference Caov-3 cells
phological alterations indicated by the acquisition of a spindle-shaped grown in heavy SILAC media (13C6-lysine).
and loss of cell-cell contact after EGF stimulation in Caov-3 cells (Fig. After mixing equal amounts (300 μg) of control or EMT-induced
2A). MRM analysis demonstrated that the newly acquired mesenchy- Caov-3 cells with Caov-3 heavy labeled cells, samples were fractionated
mal morphology was supported by alterations in several proteins in- by differential centrifugation to obtain subcellular extracts enriched in
volved in EMT. Increasing levels of Vimentin (VIM), N-cadherin cytoplasmic, nuclear or membrane proteins. Initially, the quality of sub-
(CDH2) and Annexin A2 (ANXA2) proteins followed by the EMT-related cellular fractionation was confirmed by Western Blotting, using anti-
transcription factor SNAIL (SNAI1) were observed after 96 h of EMT in- bodies to detect proteins exclusive to each subcellular compartment
duction by EGF. As expected, significant reduction of E-cadherin (CDH1) (data not shown). Each subcellular fraction was further fractionated
epithelial marker abundance was observed (Fig. 2B). Changes were not by SDS-PAGE and gel lanes were cut into five bands, generating a total
detected for the SLUG (SNAI2), another EMT-related transcription of 30 fractions. A schematic description of our experimental workflow
factor. is presented in Supplemental Fig. 1. Each fraction was subjected to in-
In addition, changes in the levels of Vimentin and E-cadherin pro- gel trypsin digestion and individually analyzed by LC-MS/MS. Around
teins were also evaluated by immunofluorescence microscopy to con- 450,000 MS/MS scans were collected, providing confident identification
firm EMT induction. In agreement with MRM analysis, fluorescence for 3018 proteins represented by unique gene names with b1% FDR

Fig. 1. Profile of EGF receptor (EGFR) activation in ovarian adenocarcinoma cell line Caov-3. A) Western blotting shows the activation of EGFR (Tyr1068) in Caov-3 cells after 15 min of
stimulation with two different doses of EGF: 1 and 10 ng/ml. GAPDH protein was used as endogenous control. B) MRM analysis shows that Caov-3 cells stimulated with EGF maintain
high levels of EGFR when compared to untreated cells, after 96 h of stimulation. Bars indicate the sum of the means of each transition of two independent replicates. Statistical
significance was assessed by Student's t-test. *p b 0.05. Detailed method parameters are shown in Supplementary Table 1. C) RTK activity array analysis using the PathScan® RTK
Signaling Antibody Array kit (cell signaling), showed differential phosphorylation in important pathways in response to EGF treatment.
6 M.L. Grassi et al. / Journal of Proteomics 151 (2017) 2–11

Fig. 2. Characterization of EMT induction in Caov-3 cells by EGF. A) Morphological changes observed by contrast phase microscopy in Caov-3 cells induced to EMT by EGF 10 ng/ml after
96 h. B) EGF-induced EMT increased levels of known mesenchymal related proteins (VIM, CDH2, ANX2, SNAI1), as well as decreased CDH1, but not changed levels of the transcription
factor Slug (SNAI2), base on MRM analysis after 96 h of induction. Bars indicate the sum of the means of each transition of two independent experiments. Statistical significance was
assessed by Student's t-test. *p b 0.05. Detailed method parameters are shown in Supplementary Table 1. C) Immunofluorescence microscopy of Caov-3 cells induced to EMT shows a
higher fluorescent intensity of the dynamic change of Vimentin filaments extended throughout the cells, while E-cadherin fluorescence intensity appears weaker than the control cells.
Caov-3 CTRL: non-induced; Caov-3 EGF: induced with EGF 10 ng/ml during 96 h.

(False Discovery Rate). The complete list of protein identification is pre- processes were significantly changed: 1) metabolism (63.3%), specifi-
sented in Supplementary Table 2. The subsets of proteins identified in cally protein synthesis, and 2) cellular processes (38.9%) including cell
each subcellular compartment were complementary, since proteins de- cycle and cell-cell communication (Fig. 3B). In fact, metabolic
tected in a single subcellular compartment contributed to 13% to 19% reprogramming is one of the most important hallmarks of cancer and
additional identifications and only 28% of all proteins were identified very favorable not only for tumor survival and growth but also for inva-
in all three subcellular compartments. sion and metastasis. In addition, cell cycle regulation has been closely
Our quantitative analysis elected 206 differentially expressed pro- linked to EMT since Snail transcription factor family members can in-
teins, based on a stringent criteria where only proteins that were ob- duce cell cycle arrest in G1 phase and the main cell cycle regulation
served with at least two peptides with associated quantitative pathways seem to be dependent on cell-cell adhesion [24–29].
information and changes N 1.5-fold in at least one subproteome were In order to validate if the 72 regulated proteins from our PPI network
considered for further evaluation. The full list of regulated proteins is are relevant for ovarian cancer, we relied on ProteinAtlas IHC data to
presented in Supplementary Table 3. verify expression in normal ovarian and/or ovarian cancer tissue. As ob-
Of note, AnxA6 (Annexin A6) was downregulated in nuclear and served in Fig. 3C, most proteins were upregulated by EMT in at least one
membrane fractions and upregulated in the cytoplasm, PSMA5 (protea- subcellular compartment and several of these proteins appeared highly
some subunit alpha type 5) was downregulated in membrane fraction stained in most of ovarian cancer tumor tissues analyzed by
and upregulated in cytoplasm and ITGA2 (integrin alpha 2) was upreg- ProteinAtlas, such as P4HA1 (Prolyl 4-hydroxylase subunit alpha-1),
ulated only in membrane and nuclear fractions. These differences show HNRNPA1 (heterogeneous nuclear ribonucleoprotein A1), RAB7A
the important feature of analyzing multiple subcellular compartments (Ras-related protein Rab-7a), RPL24 (60S ribosomal protein L24) and
in parallel, potentially highlighting protein translocation processes EPS15 (epidermal growth factor receptor substrate 15). Moreover, sever-
such those mentioned above. al proteins downregulated in our proteomic data are highly expressed in
normal ovarian tissues, suggesting that these proteins could potentially
3.4. Protein-protein interaction (PPI) network analysis and validation of represent metastasis suppressors XRCC6 (X-ray repair cross-
proteomic findings using ProteinAtlas complementing protein 6), EWSR1 (RNA-binding protein EWS), PSMC2
(26S protease regulatory subunit 7), RAD23B (UV excision repair protein
Initially, using STRING, we performed a PPI network analysis with RAD23 homolog B) and EFTUD2 (116 kDa U5 small nuclear ribonucleo-
the 206 differentially expressed proteins during EMT induced by EGF. protein component). In this context, information available in the Protein
From that set of regulated protein, we selected a subset composed of Atlas database represented an external validation for our data and was
72 proteins (Fig. 3A) for which STRING established a PPI network. highly relevant for future investigation of proteins potentially related to
After, using PANTHER, we identified major cellular processes arising EMT and metastasis, since both up- and downregulation of proteins
from EMT induced by EGF in Caov-3 cells. Two distinct cellular might represent important processes for ovarian cancer progression.
M.L. Grassi et al. / Journal of Proteomics 151 (2017) 2–11 7

Fig. 3. Proteins regulated by EGF-induced EMT in Caov-3 cells. A) From a total of 206 differentially expressed proteins, 72 were able to establish a PPI network based on STRING analysis. The
network indicates a major hub composed specially by ribosomal proteins (RPs). B) PANTHER analysis indicated that these 72 proteins present in the PPI network are involved in metabolic
(protein synthesis) and cellular processes (cell cycle and cell-cell adhesion). Dotted red circle indicates EGF growth factor added together with the 206 regulated proteins data to connect
these proteins with EMT process induction. C) The heat map shows the distribution of the 72 differentially expressed proteins among the three different subcellular compartments (left
panel). Color scale represents each protein log2 ratio from EMT-induced/control experiments. Immunohistochemical data from ovarian cancer and normal tissues obtained from The
Human Protein Atlas database (right panel) was used as an external indicator of relevance of regulated proteins identified in our study. The scale (0–100%) refers to the percentage of
immunohistochemically stained breast cancer samples and the color gradient represents the staining intensity: high (dark blue), medium (blue), low (light blue), not detected (white)
or not available.

Fig. 4. Analysis of Ras/Erk MAPK and PI3K/Akt/mTOR signaling pathways and cell proliferation during EGF-induced EMT in Caov-3 cells. A) EMT induced by EGF in Caov-3 cells activates the
Ras/Erk MAPK pathway, with high levels of Erk 1/2 (Thr202/Tyr204), and the PI3K/Akt pathway indicated by high levels of Akt (Ser473) favored by deactivation of PTEN (Ser308), the main
PI3K/Akt phosphatase inhibitor, while mTOR level did not change. GAPDH protein was used as endogenous control. B) Analysis by flow cytometry of BrdU incorporation indicates no
significant change of proliferation rate for Caov-3 cells induced to EMT by EGF. Bars indicate the mean ± standard deviation of two independent experiments. Statistical significance
was assessed by Student's t-test. ns = no significant.
8 M.L. Grassi et al. / Journal of Proteomics 151 (2017) 2–11

3.5. Induction of EMT by EGF in Caov-3 cells implicates in Akt and ERK 1/2 our results from PPI-network analysis, indicating that EGF-induced
activation without changing proliferation pattern EMT indeed affects cell cycle control.

Based on the PPI network obtained from proteomic data, the induc- 4. Discussion
tion of EMT in Caov-3 cells by EGF promotes regulation of several ribo-
somal proteins (RPs) and proteins related to cell cycle control. To test The role of EMT in cancer invasion and metastasis is strongly sup-
whether these molecular changes promoted in Caov-3 cells by EMT ported by several cellular models. However, monitoring this process
would result in modulation of the cell growth and proliferation patterns, during cancer progression is still difficult since cells undergoing EMT
we evaluated the activation of key signaling pathways, such as PI3K/ share many molecular and morphological characteristics with the sur-
Akt/mTOR and ERK 1/2 MAP kinase. We also measured the cellular pro- rounding stromal cells. Here we show an effective in vitro induction of
liferation rate by BrdU incorporation. Fig. 4A indicates that AKT activa- EMT by EGF treatment in ovarian cancer cells. The concentration of
tion was stronger in EMT-induced Caov-3 cells compared to control EGF used in this study (10 ng/ml) is considerably below what is found
cells, with increased levels of total and phosphorylated Akt (Ser473) in general literature for EMT induction, usually N 50 ng/ml, but greater
and reduced levels of PTEN, the major negative regulator of this kinase. than the know regular concentration of EGF in circulation (40pM–
Independent of Akt activation, a decreasing level of mTOR was ob- 240 pg/ml) [30]. In fact, EGF stimulation in ovarian surface epithelial
served, followed by high levels in ERK 1/2 (Thr202/Tyr204) phosphory- cells has demonstrated to be involved in matrix remodeling mediated
lation (Fig. 4A). Although EMT induction resulted in evident by ERK and ILK/GSK-3β pathways [11] and combined targeting of
phosphorylation of specific kinases involved in cell proliferation, BrdU endothelin and EGF receptors has shown to enhance antitumor activity
incorporation unexpectedly demonstrated that Caov-3 cells prolifera- in ovarian cancers [31].
tion rate did not change after EGF-induced EMT (Fig. 4B). In our model, EGF-stimulated Caov-3 cells showed high EGFR activ-
ity followed by high phosphorylation levels of Ephrins A2, A3 and B2
3.6. EGF-induced EMT promotes cell cycle arrest in Caov-3 cells (Fig. 1C). These RTKs have played crucial roles in cell morphology, adhe-
sion, migration and invasion by modifying the actin cytoskeleton reor-
Since Caov-3 ovarian cancer cells showed no significant change in ganization and regulating the activities of integrins and cell-cell
proliferation rate after EGF-induced EMT, we performed Western Blot- adhesion molecules. Several studies correlating the role of EPH recep-
ting analysis for proteins relevant to cell cycle regulation. We also per- tors in EMT process have emerged. In hepatocellular carcinoma, for ex-
formed cell cycle analysis using flow cytometry. As demonstrated in ample, the EPHA receptor favored EMT through the activation of HGF
Fig. 5, EGF-induced EMT had a measurable impact over Caov-3 cell signaling pathway, since HGF induced EMT-like morphological changes
cycle. The remarkable increase in p21 protein levels indicates accumula- as well as upregulation of SNAIL and N-cadherin [32]. Notably, EPHA2
tion of cells in G1 phase and consequent blockage of S phase, while p53 overexpression was able to upregulate EMT molecular markers N-
protein was not detected (Fig. 5A and B). Concomitantly, we verified a cadherin and Snail, and downregulate E-cadherin in gastric cancer
reduction in the phosphorylation levels of cyclin D1, retinoblastoma cells [33]. Hence, these evidences suggest that elevated expression of
protein (Rb) and cyclin-dependent kinase 1 (CDK1)/cdc2 protein in re- EPH/Ephrin may be correlated with increased invasiveness in different
sponse to EMT induced by EGF (Fig. 5C). Altogether this data supports types of aggressive tumors, including ovarian cancer.

Fig. 5. EGF-induced EMT in Caov-3 cells promote cell cycle arrest through regulation of checkpoint proteins. A) Cell cycle analysis of Caov-3 cells by PI incorporation shows that EMT
induced by EGF blocks G1/S transition phase increasing the G1 cell population. Bars indicate the mean ± standard deviation of three independent experiments. Statistical significance
was assessed by two way ANOVA. ***p b 0.0001. B) Regulation of G1 cell-cycle arrest in Caov-3 cells was mediated by high levels of the cyclin-dependent kinase inhibitor p21Cip1/Waf1,
an inhibitor of the G1-to-S phase, independent of p53 expression. C) Decreasing or non-alteration in phosphorylation levels of G1/S checkpoint proteins cyclin D1 (Thr286) and Rb
(Ser807/811) followed by a decrease in G2/M checkpoint protein CDK1/cdc2 supports the effect of EGF-induced EMT in Caov-3 cells. GAPDH protein was used as endogenous control.
M.L. Grassi et al. / Journal of Proteomics 151 (2017) 2–11 9

Morphological and molecular changes, such as the acquisition of a translation through eIF4E phosphorylation by Erk-activated MNK1 and
spindle shaped morphology and increased levels of EMT-related pro- MNK2, favoring translation and tumor development [49]. In this con-
teins (Vimentin, N-cadherin, Annexin A2, Snail), support the efficient text, although these signaling pathways appear activated by EGF during
EMT induction with EGF (Fig. 2). These molecular changes observed in the induction of EMT in Caov-3 cells, it seems that decreasing mTOR
our EMT model are relevant for a metastatic context, since in high- levels either is sufficient to maintain the cell proliferation pattern un-
grade serous ovarian carcinomas, the low N-cadherin mRNA expression changed or is controlling autophagy, once mTOR inhibition promotes
has been associated with worse survival and the transcription factor autophagy, which is required for the management of metabolic alter-
SNAIL is commonly overexpressed in later stage ovarian tumors (III ations and can limit tumor progression [50]. Together these alterations
and IV) compared [34–36]. suggest a remarkable connection between EMT process and translation-
Based on this valid model for EMT, proteomic and bioinformatics al control in ovarian cancer since these events share the same classical
analysis of the three subcellular fractions from EGF-induced Caov-3 signaling pathways.
cells revealed several differentially expressed proteins. The organization In addition to the role of ribosomal stabilization and maintenance of
of such data into functional classes highlighted groups of proteins previ- the efficiency and accuracy of translation, the RPs have also demonstrat-
ously linked to cellular metabolism, tumor progression and EMT (Fig. 3; ed secondary functions, or extra-ribosomal functions, related to other
Supplementary Table 3). In fact, metabolic reprogramming is one of the cellular processes including proliferation, cell cycle and gene expression
most important hallmarks of cancer and very beneficial not only for [51–53]. Overexpression of RP L41 can arrest the cell cycle at the G1
tumor survival and growth but also invasion and metastasis. phase in human lung carcinoma H1299 cells through an increase in
Several proteins differentially expressed in our Caov-3 model have p21Cip1/Waf1 levels, while constitutive overexpression of RP L7 in Jurkat
been already associated with EMT and metastasis. AnxA6 depletion in T-lymphoma cells leads to G1 arrest via the modulation of cell cycle pro-
the invasive BT-549 breast cancer cells results in strong inhibition of gression related proteins [54,55]. In our data, EGF-induced EMT was
cell motility and invasiveness, contributing to cancer progression [37, able to impair the transition from G1 to S phase by maintaining high
38]. A recent study showed that ITGA2, when modulated by the miR- levels of p21Cip1/Waf1, which block the G1/S transition, independent of
128, suppressed cell migration, invasion and EMT in MG-63 osteosarco- p53 modulation (Fig. 5). Clearly, the loss or inhibition of p53 protein
ma cells [39]. In addition, NT5E (5′-nucleotidase or CD73), PRMT1 (pro- prevents cellular senescence, by controlling cell cycle checkpoints, and
tein arginine methyl transferase 1) and Ostf1b (osmotic stress apoptosis. In fact, relevant cell cycle proteins such as cyclin D1, Rb and
transcription factor 1b) proteins have been considered as new regula- CDK1/cdc2 had a significant decrease after EGF-induced EMT, and its ex-
tors of EMT by favoring the tumor progression in patients with gallblad- pression has also been predictive of poor clinical outcome in patients
der cancer, enhancing TWIST1 methylation in non-small cell lung with ovarian cancer [56]. About half of all human cancers, the p53
cancer and promoting cell migration and cytoskeleton rearrangement tumor suppressor is either lost or mutated, resulting in the expres-
in HEK293 cells, respectively [40–42]. sion of a transcriptionally inactive mutant protein. Curiously, studies
It is also important to highlight that most of the proteins observed as showed that a point mutation in TP53 gene (resulting in a chain ter-
regulated in our study are detected as moderate or intense in tumor mination signal likely to truncate the p53 peptide at amino acid 135)
samples, according ProteinAtlas external data validation. More interest- and substantial copy-number changes in Caov-3 cell line are key
ing, some of the proteins that are downregulated were strongly stained characteristics related to high-grade serous ovarian cancer, although
in normal tissue. Based on these observations, one could speculate that it also can reflect the difficulty for detecting the p53 protein using
proteins such as XRCC6, EWSRS1, PSMC2, RAD23B and EFTUD2 can act antibodies [57,58].
as tumor suppressors. Indeed, loss of the canonical non-homologous EGF has the ability to stimulate differentiation and proliferation of
end-joining (cNHEJ) composed by XRCC6 and RB1 has been recently several cell types by binding to its transmembrane receptor EGFR [59].
implicated in genomic instability and cancer progression [43]. EWS-fu- Surprisingly, the presence of EGF as an EMT inducer did not affect the
sion-proteins (EFPS) play a role in different tumorigenic processes and proliferation rate of Caov-3 cells during the acquisition of the mesen-
is directly associated with mesenchymal stem cell phenotype [44]. chymal phenotype and suggests a new response profile very similar to
Thus, this set of downregulated proteins seems extremely interesting TGF-β anti-proliferative effect discussed in different studies [60–62].
in terms of targets for tumor control. Furthermore, the conversion to a mesenchymal phenotype, which
Recently, alterations in translational control and cancer etiology denotes a dramatic actin cytoskeleton reorganization, may be in-
have outlined a new perspective in cancer research since it has been compatible with a highly proliferative state, as we previously report-
known that genetic alterations in several components of the transla- ed in a recent study where MCF7 cells exhibited reduced capacity of
tional apparatus (translation factors, ribosomes, RPs) and cancer are in- cell proliferation after EMT induction by SNAIL-overexpressing,
terconnected, favoring increased cancer susceptibility [45]. As we show supporting the idea that cell proliferation is not essential for tumor
in Fig. 3B, the great number of regulated RPs creates a dense network in malignancy [26,29].
our data set, suggesting a link between the EMT process induced by EGF
and ribosome biogenesis/protein translational regulation in ovarian
cancer. Following a traditional view, most of the studies have reported 5. Conclusions
that dysregulation of protein synthesis during cancer development
merely results in cell growth and proliferation. However, protein dys- Overall, our study demonstrated that EGF, a potent growth factor
regulation can be one of the mechanisms that favor cancer develop- that is tightly connected to cancer development and progression, in-
ment, since important oncogenic signaling pathways are intrinsically duced EMT in Caov-3 cells as well as important alterations in metabolic
related to the translational machinery at almost every stage of cancer process (protein synthesis) and cell cycle control. Although these
initiation [46]. changes result from complex reprograming of protein expression, our
Akt and ERK1/2 phosphorylation status indicates the activation of data contribute to support the implication of the EMT highlighted path-
PI3K/Akt/ERK1/2 pathway after EGF-induced EMT in Caov-3 cells. ways that are likely linked to in vivo cancer metastasis and cancer stem
PI3K/Akt pathway controls ribosome biogenesis by modulating the cell generation. Further clinical studies that correlate RP expression,
transcription of Pol I and III [47], and the loss of PTEN causes upregula- translational control and tumor progression are therefore worthwhile
tion of this pathway (Fig. 4). Moreover, the Ras/Erk MAPK pathway, as to uncover new therapeutic targets for cancer treatment and improved
well PI3K/Akt, regulates several downstream effector pathways, includ- patient outcome.
ing mTORC1, by Erk- and p90RSK-dependent inhibitory phosphoryla- Supplementary data to this article can be found online at http://dx.
tion of tuberous sclerosis 2 (TSC2) [48]. Ras pathway also stimulates doi.org/10.1016/j.jprot.2016.06.009.
10 M.L. Grassi et al. / Journal of Proteomics 151 (2017) 2–11

Acknowledgments [23] M. Uhlén, E. Björling, C. Agaton, C.A. Szigyarto, B. Amini, E. Andersen, et al., A human
protein atlas for normal and cancer tissues based on antibody proteomics, Mol. Cell.
Proteomics 4 (2005) 1920–1932.
We thank São Paulo Research Foundation (FAPESP, grant EMU- [24] R.A. Cairns, I.S. Harris, T.W. Mak, Regulation of cancer cell metabolism, Nat. Rev. Can-
2004/09448-5), the Centre of Flow Cytometry of Faculty of Pharmaceu- cer 11 (2011) 85–95.
[25] T. Han, D. Kang, D. Ji, X. Wang, W. Zhan, M. Fu, et al., How does cancer cell
tical Sciences of Ribeirão Preto – University of Sao Paulo, for providing metabolism affect tumor migration and invasion? Cell Adhes. Migr. 7 (2013)
access to the equipment flow cytometer BD FACSCanto I and the Central 395–403.
de Análises Químicas Instrumentais –Instituto de Química de São Carlos [26] S. Vega, A.V. Morales, O.H. Ocaña, F. Valdés, I. Fabregat, M.A. Nieto, Snail blocks the
cell cycle and confers resistance to cell death, Genes Dev. 18 (2004) 1131–1143.
– University of São Paulo, for providing access to the LTQ-Orbitrap Velos [27] F.E. Turner, S. Broad, F.L. Khanim, A. Jeanes, S. Talma, S. Hughes, et al., Slug regulates
Instrument. This research was supported by FAPESP (Young Scientist integrin expression and cell proliferation in human epidermal keratinocytes, J. Biol.
Grant – Proc. No. 2011/0947-1), CNPq (Proc. No. 308561/2014-7), Chem. 281 (2006) 21321–21331.
[28] J.L. Walker, A.K. Fournier, R.K. Assoian, Regulation of growth factor signaling and cell
Center for Cell Based Therapy - CTC-CEPID (Proc. FAPESP 2013/08135-
cycle progression by cell adhesion and adhesion-dependent changes in cellular ten-
2) and CISBi-NAP. M.L.G, C.S.P. and C.H.T. received fellowships from sion, Cytokine Growth Factor Rev. 16 (2005) 395–405.
FAPESP Proc. Nos. 2013/08755-0, 2012/09682-4 and 2013/07675-3 re- [29] C.S. Palma, M.L. Grassi, C.H. Thomé, G.A. Ferreira, D. Albuquerque, M.T. Pinto, et al.,
spectively. A.P. received fellowship from CAPES. C.S.P., G.P.P. and Proteomic analysis of epithelial to mesenchymal transition reveals crosstalk be-
tween SNAIL and HDAC1 in breast cancer cells, Mol. Cell. Proteomics (2016).
V.M.F. received fellowships from CNPq, Proc. Nos. 154138/2014-2, [30] B. Marquèze-Pouey, S. Mailfert, V. Rouger, J.M. Goaillard, D. Marguet, Physiological
130142/2015-8 and 308561/2014-7 respectively. epidermal growth factor concentrations activate high affinity receptors to elicit cal-
cium oscillations, PLoS One 9 (2014) e106803.
[31] L. Rosanò, V. Di Castro, F. Spinella, G. Tortora, M.R. Nicotra, P.G. Natali, et al., Com-
References bined targeting of endothelin A receptor and epidermal growth factor receptor in
ovarian cancer shows enhanced antitumor activity, Cancer Res. 67 (2007)
[1] D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation, Cell 144 6351–6359.
(2011) 646–674. [32] T. Nagai, T. Arao, K. Furuta, K. Sakai, K. Kudo, H. Kaneda, et al., Sorafenib inhibits the
[2] R. Kalluri, R.A. Weinberg, The basics of epithelial-mesenchymal transition, J. Clin. In- hepatocyte growth factor-mediated epithelial mesenchymal transition in hepatocel-
vest. 119 (2009) 1420–1428. lular carcinoma, Mol. Cancer Ther. 10 (2011) 169–177.
[3] E.D. Hay, An overview of epithelio-mesenchymal transformation, Acta Anat. (Basel) [33] J. Huang, D. Xiao, G. Li, J. Ma, P. Chen, W. Yuan, et al., EphA2 promotes epithelial-
154 (1995) 8–20. mesenchymal transition through the Wnt/β-catenin pathway in gastric cancer
[4] J.M. Lee, S. Dedhar, R. Kalluri, E.W. Thompson, The epithelial-mesenchymal transi- cells, Oncogene 33 (2014) 2737–2747.
tion: new insights in signaling, development, and disease, J. Cell Biol. 172 (2006) [34] B. Davidson, C.G. Tropé, R. Reich, Epithelial-mesenchymal transition in ovarian car-
973–981. cinoma, Front. Oncol. 2 (2012) 33.
[5] A. Puisieux, T. Brabletz, J. Caramel, Oncogenic roles of EMT-inducing transcription [35] L. Quattrocchi, A.R. Green, S. Martin, L. Durrant, S. Deen, The cadherin switch in ovar-
factors, Nat. Cell Biol. 16 (2014) 488–494. ian high-grade serous carcinoma is associated with disease progression, Virchows
[6] M.A. Huber, N. Kraut, H. Beug, Molecular requirements for epithelial-mesenchymal Arch. 459 (2011) 21–29.
transition during tumor progression, Curr. Opin. Cell Biol. 17 (2005) 548–558. [36] S. Elloul, M.B. Elstrand, J.M. Nesland, C.G. Tropé, G. Kvalheim, I. Goldberg, et al., Snail,
[7] L. Larue, A. Bellacosa, Epithelial-mesenchymal transition in development and can- Slug, and Smad-interacting protein 1 as novel parameters of disease aggressiveness
cer: role of phosphatidylinositol 3′ kinase/AKT pathways, Oncogene 24 (2005) in metastatic ovarian and breast carcinoma, Cancer 103 (2005) 1631–1643.
7443–7454. [37] C. Enrich, C. Rentero, S.V. de Muga, M. Reverter, V. Mulay, P. Wood, et al., Annexin
[8] H. Niikura, H. Sasano, S. Sato, A. Yajima, Expression of epidermal growth factor-re- A6-linking Ca(2+) signaling with cholesterol transport, Biochim. Biophys. Acta
lated proteins and epidermal growth factor receptor in common epithelial ovarian 2011 (1813) 935–947.
tumors, Int. J. Gynecol. Pathol. 16 (1997) 60–68. [38] A.M. Sakwe, R. Koumangoye, B. Guillory, J. Ochieng, Annexin A6 contributes to the
[9] J.M. Bartlett, S.P. Langdon, B.J. Simpson, M. Stewart, D. Katsaros, P. Sismondi, et al., invasiveness of breast carcinoma cells by influencing the organization and localiza-
The prognostic value of epidermal growth factor receptor mRNA expression in pri- tion of functional focal adhesions, Exp. Cell Res. 317 (2011) 823–837.
mary ovarian cancer, Br. J. Cancer 73 (1996) 301–306. [39] X. Liu, Z. Liang, K. Gao, H. Li, G. Zhao, S. Wang, et al., MicroRNA-128 inhibits EMT of
[10] P. Yue, X. Zhang, D. Paladino, B. Sengupta, S. Ahmad, R.W. Holloway, et al., Hyperac- human osteosarcoma cells by directly targeting integrin α2, Tumour Biol. (2015).
tive EGF receptor, Jaks and Stat3 signaling promote enhanced colony-forming abil- [40] L. Xiong, Y. Wen, X. Miao, Z. Yang, NT5E and FcGBP as key regulators of TGF-1-in-
ity, motility and migration of cisplatin-resistant ovarian cancer cells, Oncogene 31 duced epithelial-mesenchymal transition (EMT) are associated with tumor progres-
(2012) 2309–2322. sion and survival of patients with gallbladder cancer, Cell Tissue Res. 355 (2014)
[11] N. Ahmed, S. Maines-Bandiera, M.A. Quinn, W.G. Unger, S. Dedhar, N. Auersperg, Molec- 365–374.
ular pathways regulating EGF-induced epithelio-mesenchymal transition in human [41] S. Avasarala, M. Van Scoyk, M.K. Karuppusamy Rathinam, S. Zerayesus, X. Zhao, W.
ovarian surface epithelium, Am. J. Physiol. Cell Physiol. 290 (2006) C1532–C1542. Zhang, et al., PRMT1 is a novel regulator of epithelial-mesenchymal-transition in
[12] B. MacLean, D.M. Tomazela, N. Shulman, M. Chambers, G.L. Finney, B. Frewen, et al., non-small cell lung cancer, J. Biol. Chem. 290 (2015) 13479–13489.
Skyline: an open source document editor for creating and analyzing targeted prote- [42] K.P. Lai, A.Y. Law, M.C. Lau, Y. Takei, W.K. Tse, C.K. Wong, Osmotic stress transcrip-
omics experiments, Bioinformatics 26 (2010) 966–968. tion factor 1b (Ostf1b) promotes migration properties with the modulation of epi-
[13] R.M. Adam, W. Yang, D. Di Vizio, N.K. Mukhopadhyay, H. Steen, Rapid preparation of thelial mesenchymal transition (EMT) phenotype in human embryonic kidney
nuclei-depleted detergent-resistant membrane fractions suitable for proteomics cell, Int. J. Biochem. Cell Biol. 45 (2013) 1921–1926.
analysis, BMC Cell Biol. 9 (2008) 30. [43] R. Cook, G. Zoumpoulidou, M.T. Luczynski, S. Rieger, J. Moquet, V.J. Spanswick, et al.,
[14] U.K. Laemmli, Cleavage of structural proteins during the assembly of the head of Direct involvement of retinoblastoma family proteins in DNA repair by non-homol-
bacteriophage T4, Nature 227 (1970) 680–685. ogous end-joining, Cell Rep. 10 (2015) 2006–2018.
[15] C.H. Thomé, G.A. dos Santos, G.A. Ferreira, P.S. Scheucher, C. Izumi, A.M. Leopoldino, [44] F. Tirode, K. Laud-Duval, A. Prieur, B. Delorme, P. Charbord, O. Delattre, Mesenchy-
et al., Linker for activation of T-cell family member2 (LAT2) a lipid raft adaptor pro- mal stem cell features of Ewing tumors, Cancer Cell 11 (2007) 421–429.
tein for AKT signaling, is an early mediator of alkylphospholipid anti-leukemic activ- [45] D. Silvera, S.C. Formenti, R.J. Schneider, Translational control in cancer, Nat. Rev.
ity, Mol. Cell. Proteomics 11 (2012) 1898–1912. Cancer 10 (2010) 254–266.
[16] B. MacLean, J.K. Eng, R.C. Beavis, M. McIntosh, General framework for developing [46] J.P. Le Quesne, K.A. Spriggs, M. Bushell, A.E. Willis, Dysregulation of protein synthesis
and evaluating database scoring algorithms using the TANDEM search engine, Bio- and disease, J. Pathol. 220 (2010) 140–151.
informatics 22 (2006) 2830–2832. [47] M.J. James, J.C. Zomerdijk, Phosphatidylinositol 3-kinase and mTOR signaling path-
[17] A.I. Nesvizhskii, A. Keller, E. Kolker, R. Aebersold, A statistical model for identifying ways regulate RNA polymerase I transcription in response to IGF-1 and nutrients,
proteins by tandem mass spectrometry, Anal. Chem. 75 (2003) 4646–4658. J. Biol. Chem. 279 (2004) 8911–8918.
[18] A. Keller, A.I. Nesvizhskii, E. Kolker, R. Aebersold, Empirical statistical model to esti- [48] X.M. Ma, J. Blenis, Molecular mechanisms of mTOR-mediated translational control,
mate the accuracy of peptide identifications made by MS/MS and database search, Nat. Rev. Mol. Cell Biol. 10 (2009) 307–318.
Anal. Chem. 74 (2002) 5383–5392. [49] S. Pyronnet, H. Imataka, A.C. Gingras, R. Fukunaga, T. Hunter, N. Sonenberg, Human
[19] V. Faca, M. Coram, D. Phanstiel, V. Glukhova, Q. Zhang, M. Fitzgibbon, et al., Quanti- eukaryotic translation initiation factor 4G (eIF4G) recruits mnk1 to phosphorylate
tative analysis of acrylamide labeled serum proteins by LC-MS/MS, J. Proteome Res. eIF4E, EMBO J. 18 (1999) 270–279.
5 (2006) 2009–2018. [50] R. Mathew, V. Karantza-Wadsworth, E. White, Role of autophagy in cancer, Nat. Rev.
[20] V.M. Faca, K.S. Song, H. Wang, Q. Zhang, A.L. Krasnoselsky, L.F. Newcomb, et al., A Cancer 7 (2007) 961–967.
mouse to human search for plasma proteome changes associated with pancreatic [51] H. Naora, Involvement of ribosomal proteins in regulating cell growth and apopto-
tumor development, PLoS Med. 5 (2008) e123. sis: translational modulation or recruitment for extraribosomal activity? Immunol.
[21] D. Szklarczyk, A. Franceschini, S. Wyder, K. Forslund, D. Heller, J. Huerta-Cepas, et al., Cell Biol. 77 (1999) 197–205.
STRING v10: protein-protein interaction networks, integrated over the tree of life, [52] M. Thapa, A. Bommakanti, M. Shamsuzzaman, B. Gregory, L. Samsel, J.M. Zengel,
Nucleic Acids Res. 43 (2015) D447–D452. et al., Repressed synthesis of ribosomal proteins generates protein-specific cell
[22] H. Mi, A. Muruganujan, P.D. Thomas, PANTHER in 2013: modeling the evolution of cycle and morphological phenotypes, Mol. Biol. Cell 24 (2013) 3620–3633.
gene function, and other gene attributes, in the context of phylogenetic trees, [53] M.S. Lindström, Emerging functions of ribosomal proteins in gene-specific transcrip-
Nucleic Acids Res. 41 (2013) D377–D386. tion and translation, Biochem. Biophys. Res. Commun. 379 (2009) 167–170.
M.L. Grassi et al. / Journal of Proteomics 151 (2017) 2–11 11

[54] M.J. Kim, Y.A. Yoo, H.J. Kim, S. Kang, Y.G. Kim, J.S. Kim, et al., Mitochondrial ribosomal [59] Y. Yarden, M.X. Sliwkowski, Untangling the ErbB signalling network, Nat. Rev. Mol.
protein L41 mediates serum starvation-induced cell-cycle arrest through an in- Cell Biol. 2 (2001) 127–137.
crease of p21(WAF1/CIP1), Biochem. Biophys. Res. Commun. 338 (2005) [60] L. Levy, C.S. Hill, Smad4 dependency defines two classes of transforming growth fac-
1179–1184. tor {beta} (TGF-{beta}) target genes and distinguishes TGF-{beta}-induced epitheli-
[55] F. Neumann, U. Krawinkel, Constitutive expression of human ribosomal protein L7 al-mesenchymal transition from its antiproliferative and migratory responses, Mol.
arrests the cell cycle in G1 and induces apoptosis in Jurkat T-lymphoma cells, Exp. Cell. Biol. 25 (2005) 8108–8125.
Cell Res. 230 (1997) 252–261. [61] J. Seoane, Escaping from the TGFbeta anti-proliferative control, Carcinogenesis 27
[56] A. Bali, P.M. O'Brien, L.S. Edwards, R.L. Sutherland, N.F. Hacker, S.M. Henshall, Cyclin (2006) 2148–2156.
D1, p53, and p21Waf1/Cip1 expression is predictive of poor clinical outcome in se- [62] A. Klotzsche-von Ameln, A. Muschter, S. Mamlouk, J. Kalucka, I. Prade, K. Franke,
rous epithelial ovarian cancer, Clin. Cancer Res. 10 (2004) 5168–5177. et al., Inhibition of HIF prolyl hydroxylase-2 blocks tumor growth in mice through
[57] S. Domcke, R. Sinha, D.A. Levine, C. Sander, N. Schultz, Evaluating cell lines as tumour the antiproliferative activity of TGFβ, Cancer Res. 71 (2011) 3306–3316.
models by comparison of genomic profiles, Nat. Commun. 4 (2013) 2126.
[58] Y. Yaginuma, H. Westphal, Abnormal structure and expression of the p53 gene in
human ovarian carcinoma cell lines, Cancer Res. 52 (1992) 4196–4199.

You might also like