ova66 increases cell growth, invasion and survival via regulation of

9
© The Author 2014. Published by Oxford University Press. All rights reserved. For Permissions, please email: [email protected] Carcinogenesis vol.35 no.7 pp.1573–1581, 2014 doi:10.1093/carcin/bgu070 Advance Access publication March 25, 2014 OVA66 increases cell growth, invasion and survival via regulation of IGF-1R–MAPK signaling in human cancer cells Wei Rao , Haowen Li , Feifei Song, Renfeng Zhang, Qinqin Yin, Ying Wang, Yebin Xi and Hailiang Ge* School of Medicine, Shanghai Institute of Immunology, Shanghai Jiaotong University, 280 South Chongqing Road, Shanghai 200025, PR China *To whom correspondence should be addressed. Tel: +86 21 64452974; Fax: +86 21 63846383; Email: [email protected]. Correspondence may also be addressed to Wei Rao. Tel: +86 21 63846590- 776795; Fax: +86 21 63846383; Email: [email protected]. Ovarian cancer-associated antigen 66 (OVA66), also known as CML66 (GenBank Accession No. AF283301), was first identified in an ovarian carcinoma complementary DNA (cDNA) expression library and was shown to play a role in tumorigenesis. Here, we find that OVA66 influences tumorigenesis by regulating the type I insulin-like growth factor receptor (IGF-1R) signaling pathway. Stable knockdown of OVA66 in cancer cells attenuated phos- phorylation of IGF-1R and extracellular signal-regulated kinase 1/2 (ERK1/2)–Hsp27; similarly, a higher level of p-IGF-1R and ERK1/2–Hsp27 signaling was also detected after OVA66 over- expression in HO8910 cells. In vivo knockdown of OVA66 both reduced tumor burden in nude mice and decreased phosphoryla- tion of IGF-1R, ERK1/2 and hsp27. We blocked IGF-1R function both by small interfering RNA (siRNA) and with the chemical inhibitor Linsitinib (OSI-906). By either method, tumorigenesis was inhibited regardless of OVA66 expression; thus, mechanisti- cally, IGF-1R, probably, lies downstream of OVA66 in cancer cells. We also found that OVA66 regulates expression of murine double minute 2 (MDM2); this attenuates ubiquitination of IGF-1R in response to IGF-1 stimulation and promotes active ERK1/2 sign- aling. Thus, we propose that combined overexpression of OVA66 and MDM2 promotes oncogenesis by enhancing activation of the IGF-1R–ERK1/2 signaling pathway. Introduction Much work has been performed to identify cancer testis (CT) anti- gens as potential therapeutic and prognostic targets in tumors. CT antigens are promising targets because they are highly immunogenic and because their expression is highly restricted to cancer cells. To date, over 1000 serological analysis of recombinant cDNA expres- sion library-defined antigens have been identified (1). However, the exact functions of many of these antigens are unknown; instead, they have been ascribed general functions, such as cell signaling, tran- scription, translation and chromosomal recombination (2,3). CT antigens potentially represent defined targets for antigen-based diag- nosis, vaccination and antigen-directed immunotherapy; as a result, these CT antigens are now a primary focus in the area of tumorim- munology (4,5). Using the approach of serological analysis of recombinant cDNA expression libraries, which is based on immunoscreening of tumor cDNA expression libraries with the autologous sera from tumor patients (6), one gene was identified named OVA66, which is homolo- gous to human chronic myelocytic leukemia antigen CML66. We also showed OVA66 was capable of eliciting both humoral and cellular immune responses (7,8). Silencing of ovarian cancer-associated anti- gen 66 (OVA66) in HeLa cells inhibited tumorigenesis both in vitro and in vivo (9). Thus, we hypothesize that OVA66 is a novel CT anti- gen that may be a valuable target for cancer therapy. Although we have proved that OVA66 functioned as a novel CT antigen in can- cer, the mechanisms of OVA66 in process of tumorigenesis was still unclear. We examined expression of OVA66 in several cancer cell lines and found that it was generally overexpressed in almost all the cancer cells, compared with normal cell lines such HEK293 and NIH3T3 cells. The result was exactly consistent with our previous studies on expression patterns of OVA66 in cell lines and cancer tissues (7), and the results of tissue expression of CML66 gene (8). Stable knockdown of OVA66 in the ovarian cancer cell line SKOV3 led to decreased tumor growth, potentially via inhibition of type I insulin-like growth factor receptor (IGF-1R)–extracellular signal-regulated kinase 1/2 (ERK1/2) signaling. The findings that IGF-1R-ERK1/2 signaling was regulated by OVA66 were also seen in the OVA66 gene stably trans- fected ovarian carcinoma cell line HO8910 cells, as well as OVA66 gene transiently transfected HEK293 cells. Furthermore, inhibition of IGF-IR, either with Linsitinib (OSI-906) or small interfering RNA (siRNA), attenuated the OVA66-mediated increase in proliferation, invasion and survival in HO8910 cells. Although IGF-IR inhibition attenuated these characteristics in control SKOV3 cells, it had no effect in cells with OVA66 knockdown. Taken together, these data support that OVA66 and IGF-IR signaling pathway may be signifi- cantly correlated and functioning in a common pathway. In addition, IGF-1R has been shown to interact with murine double minute 2 (MDM2) and be critical for tumorigenesis (10,11). The work we pre- sent here links OVA66 to these important factors and elucidates a new role for OVA66 in tumorigenesis. IGF-IR has recently emerged as a bona fide clinical target for cancer treatment (12,13). Some studies have also shown that IGF-IR inhibi- tion is especially useful in attenuating the growth of tumors harboring KRAS mutations (14). Several therapies to inhibit IGF-IR signaling, including antibodies and small molecule tyrosine kinase inhibitors, have been developed (13), but the outcomes, especially in phase III clinical trials, have been largely disappointing (15). Although the exact mechanisms of therapeutic resistance in each case are unclear, several explanations have been proposed, including insulin receptor- mediated resistance (16,17), downstream ERK1/2 signaling activation and IGF-IR receptor downregulation (18). We previously reported that the oncogene MDM2 was downregulated after silencing OVA66 in HeLa cells (9). Additionally, OVA66 affects IGF-1R–ERK1/2 sign- aling, probably via its regulation of MDM2, which functions as an E3 ubiquitin ligase for IGF-1R (19–21). Here, we focus on the role of the novel oncoprotein OVA66 in regulating cancer cell proliferation, invasion and survival. Additionally, we propose that OVA66 is a new diagnostic and potentially therapeutic target in cancer. Materials and methods Cell culture and establishment of OVA66 stable knock down cells The cervical carcinoma cell lines HeLa and Siha; ovarian carcinoma cell lines SKOV3 and HO8910; breast carcinoma cell line MDA-MB-231, HHCC smmc-7721 (7721) and PLC/PRF5; human gastric cancer cell lines BGC-823, SGC-7901 and MKN-45; and several normal cell lines such as HEK293, Wish, Chang liver and NIH3T3 were purchased from the Shanghai Institutes for Abbreviations: ASK1, apoptosis signal-regulating kinase 1; cDNA, com- plementary DNA; CT, cancer testis; DMEM, Dulbecco’s modified Eagle’s medium; ERK1/2, extracellular signal-regulated kinase 1/2; FBS, fetal bovine serum; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; IGF-1R, type I insulin-like growth factor receptor; IHC, immunohistochemistry; MAPK, mitogen-activated protein kinase; MDM2, murine double minute 2; mRNA, messenger RNA; OVA66, ovarian cancer-associated antigen 66; PBS, phos- phate-buffered saline; RTK, receptor tyrosine kinase; shRNA, short hairpins RNA; siRNA, small interfering RNA; WB, western blot. These authors contributed equally to this work. 1573 Downloaded from https://academic.oup.com/carcin/article-abstract/35/7/1573/379228 by guest on 09 April 2018

Upload: hoangdang

Post on 11-Feb-2017

216 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: OVA66 increases cell growth, invasion and survival via regulation of

© The Author 2014. Published by Oxford University Press. All rights reserved. For Permissions, please email: [email protected]

Carcinogenesis vol.35 no.7 pp.1573–1581, 2014doi:10.1093/carcin/bgu070Advance Access publication March 25, 2014

OVA66 increases cell growth, invasion and survival via regulation of IGF-1R–MAPK signaling in human cancer cells

Wei Rao†, Haowen Li†, Feifei Song, Renfeng Zhang, Qinqin Yin, Ying Wang, Yebin Xi and Hailiang Ge*

School of Medicine, Shanghai Institute of Immunology, Shanghai Jiaotong University, 280 South Chongqing Road, Shanghai 200025, PR China

*To whom correspondence should be addressed. Tel: +86 21 64452974; Fax: +86 21 63846383; Email: [email protected] may also be addressed to Wei Rao. Tel: +86 21 63846590-776795; Fax: +86 21 63846383; Email: [email protected].

Ovarian cancer-associated antigen 66 (OVA66), also known as CML66 (GenBank Accession No. AF283301), was first identified in an ovarian carcinoma complementary DNA (cDNA) expression library and was shown to play a role in tumorigenesis. Here, we find that OVA66 influences tumorigenesis by regulating the type I insulin-like growth factor receptor (IGF-1R) signaling pathway. Stable knockdown of OVA66 in cancer cells attenuated phos-phorylation of IGF-1R and extracellular signal-regulated kinase 1/2 (ERK1/2)–Hsp27; similarly, a higher level of p-IGF-1R and ERK1/2–Hsp27 signaling was also detected after OVA66 over-expression in HO8910 cells. In vivo knockdown of OVA66 both reduced tumor burden in nude mice and decreased phosphoryla-tion of IGF-1R, ERK1/2 and hsp27. We blocked IGF-1R function both by small interfering RNA (siRNA) and with the chemical inhibitor Linsitinib (OSI-906). By either method, tumorigenesis was inhibited regardless of OVA66 expression; thus, mechanisti-cally, IGF-1R, probably, lies downstream of OVA66 in cancer cells. We also found that OVA66 regulates expression of murine double minute 2 (MDM2); this attenuates ubiquitination of IGF-1R in response to IGF-1 stimulation and promotes active ERK1/2 sign-aling. Thus, we propose that combined overexpression of OVA66 and MDM2 promotes oncogenesis by enhancing activation of the IGF-1R–ERK1/2 signaling pathway.

Introduction

Much work has been performed to identify cancer testis (CT) anti-gens as potential therapeutic and prognostic targets in tumors. CT antigens are promising targets because they are highly immunogenic and because their expression is highly restricted to cancer cells. To date, over 1000 serological analysis of recombinant cDNA expres-sion library-defined antigens have been identified (1). However, the exact functions of many of these antigens are unknown; instead, they have been ascribed general functions, such as cell signaling, tran-scription, translation and chromosomal recombination (2,3). CT antigens potentially represent defined targets for antigen-based diag-nosis, vaccination and antigen-directed immunotherapy; as a result, these CT antigens are now a primary focus in the area of tumorim-munology (4,5).

Using the approach of serological analysis of recombinant cDNA expression libraries, which is based on immunoscreening of tumor cDNA expression libraries with the autologous sera from tumor patients (6), one gene was identified named OVA66, which is homolo-gous to human chronic myelocytic leukemia antigen CML66. We also showed OVA66 was capable of eliciting both humoral and cellular immune responses (7,8). Silencing of ovarian cancer-associated anti-gen 66 (OVA66) in HeLa cells inhibited tumorigenesis both in vitro and in vivo (9). Thus, we hypothesize that OVA66 is a novel CT anti-gen that may be a valuable target for cancer therapy. Although we have proved that OVA66 functioned as a novel CT antigen in can-cer, the mechanisms of OVA66 in process of tumorigenesis was still unclear.

We examined expression of OVA66 in several cancer cell lines and found that it was generally overexpressed in almost all the cancer cells, compared with normal cell lines such HEK293 and NIH3T3 cells. The result was exactly consistent with our previous studies on expression patterns of OVA66 in cell lines and cancer tissues (7), and the results of tissue expression of CML66 gene (8). Stable knockdown of OVA66 in the ovarian cancer cell line SKOV3 led to decreased tumor growth, potentially via inhibition of type I insulin-like growth factor receptor (IGF-1R)–extracellular signal-regulated kinase 1/2 (ERK1/2) signaling. The findings that IGF-1R-ERK1/2 signaling was regulated by OVA66 were also seen in the OVA66 gene stably trans-fected ovarian carcinoma cell line HO8910 cells, as well as OVA66 gene transiently transfected HEK293 cells. Furthermore, inhibition of IGF-IR, either with Linsitinib (OSI-906) or small interfering RNA (siRNA), attenuated the OVA66-mediated increase in proliferation, invasion and survival in HO8910 cells. Although IGF-IR inhibition attenuated these characteristics in control SKOV3 cells, it had no effect in cells with OVA66 knockdown. Taken together, these data support that OVA66 and IGF-IR signaling pathway may be signifi-cantly correlated and functioning in a common pathway. In addition, IGF-1R has been shown to interact with murine double minute 2 (MDM2) and be critical for tumorigenesis (10,11). The work we pre-sent here links OVA66 to these important factors and elucidates a new role for OVA66 in tumorigenesis.

IGF-IR has recently emerged as a bona fide clinical target for cancer treatment (12,13). Some studies have also shown that IGF-IR inhibi-tion is especially useful in attenuating the growth of tumors harboring KRAS mutations (14). Several therapies to inhibit IGF-IR signaling, including antibodies and small molecule tyrosine kinase inhibitors, have been developed (13), but the outcomes, especially in phase III clinical trials, have been largely disappointing (15). Although the exact mechanisms of therapeutic resistance in each case are unclear, several explanations have been proposed, including insulin receptor-mediated resistance (16,17), downstream ERK1/2 signaling activation and IGF-IR receptor downregulation (18). We previously reported that the oncogene MDM2 was downregulated after silencing OVA66 in HeLa cells (9). Additionally, OVA66 affects IGF-1R–ERK1/2 sign-aling, probably via its regulation of MDM2, which functions as an E3 ubiquitin ligase for IGF-1R (19–21). Here, we focus on the role of the novel oncoprotein OVA66 in regulating cancer cell proliferation, invasion and survival. Additionally, we propose that OVA66 is a new diagnostic and potentially therapeutic target in cancer.

Materials and methods

Cell culture and establishment of OVA66 stable knock down cellsThe cervical carcinoma cell lines HeLa and Siha; ovarian carcinoma cell lines SKOV3 and HO8910; breast carcinoma cell line MDA-MB-231, HHCC smmc-7721 (7721) and PLC/PRF5; human gastric cancer cell lines BGC-823, SGC-7901 and MKN-45; and several normal cell lines such as HEK293, Wish, Chang liver and NIH3T3 were purchased from the Shanghai Institutes for

Abbreviations: ASK1, apoptosis signal-regulating kinase 1; cDNA, com-plementary DNA; CT, cancer testis; DMEM, Dulbecco’s modified Eagle’s medium; ERK1/2, extracellular signal-regulated kinase 1/2; FBS, fetal bovine serum; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; IGF-1R, type I  insulin-like growth factor receptor; IHC, immunohistochemistry; MAPK, mitogen-activated protein kinase; MDM2, murine double minute 2; mRNA, messenger RNA; OVA66, ovarian cancer-associated antigen 66; PBS, phos-phate-buffered saline; RTK, receptor tyrosine kinase; shRNA, short hairpins RNA; siRNA, small interfering RNA; WB, western blot.

†These authors contributed equally to this work.

1573Downloaded from https://academic.oup.com/carcin/article-abstract/35/7/1573/379228by gueston 09 April 2018

Page 2: OVA66 increases cell growth, invasion and survival via regulation of

Wei Rao et al.

Biological Sciences, Chinese Academy of Sciences, routinely maintained in the laboratory and cultured in Dulbecco’s modified Eagle’s medium (HyClone) supplemented with 10% fetal bovine serum (FBS, Invitrogen). Cells were transduced either with retrovirus harboring OVA66-specific short hairpins RNA (OVA66-shRNA) or control shRNA (NC-shRNA) in the presence of 4 ug/ml polybrene. For selecting stably transfected cells, cells were cultured in medium with an appropriate concentration of puromycin (Invitrogen) for 3 days. Puromycin-resistant cells were isolated, cultured in medium contain-ing half the original concentration of antibiotic and expanded for subsequent study.

Plasmid construction and establishment of OVA66-overexpressing HO8910 cellsThe plasmid pIRESpuro-OVA66 was constructed by inserting OVA66 gene made by PCR amplification of HeLa cDNA into the pIRESpuro3 vector (Invitrogen) at the EcoRI and NotI sites. HO8910 cells were then transfected with 4 ug recombinant plasmid pIRESpuro-OVA66 or empty vector using Lipofectamine 2000 (Invitrogen) according to the manufacturer’s instructions. Forty-eight hours later, to select stably transfected cells, 1.5 ug/ml puromycin (Invitrogen) was added to the medium and maintained for 3 days, at which point the puromycin-resistant cells were isolated and expanded in medium containing 0.7 ug/ml puromycin for another week. The plasmid pIRESpuro3 and pIRESpuro-OVA66 were also used to transiently transfect HEK293 cells using Lipofectamine 2000 according to instructions. The expression of OVA66 in these stably or transiently transfected cells was determined by western blot.

Cell Counting Kit-8 assay and cell invasion assays

We measured proliferation by performing a Cell Counting Kit-8 assay. Cells (5 × 103 per well) were plated in 96-well plates and cultured in medium sup-plemented with 0.5% FBS for 24 h. Then, at the indicated times, 10 μl of the Cell Counting Kit-8 (Dojindo, Kumamoto, Japan) reagent was added to each well for 2-h incubation at 37°C in a humidified CO2 incubator before meas-uring optical density at 450 nm with a microplate reader (SpectraMax M5, Molecular Devices). All experiments were performed in triplicate.

For invasion assays, 2.5 × 105 cells suspended in 500 µl of Dulbecco’s modi-fied Eagle’s medium (DMEM) or DMEM containing 1 uM Linsitinib (IGF-1R inhibitor) or the vehicle dimethyl sulfoxide without serum were seeded in the upper chambers coated with BD Matrigel Basement Membrane Matrix (BD BioCoat Matrigel™ Invasion Chambers, BD #354480). The chambers were then inserted into a 24-well plate filled with 750  µl of medium containing 10% FBS and incubated for 16 h (PLC/PRF/5, SKOV3, HO8910) or 12 h (MDA-MB-231) at 37°C in a humidified CO2 incubator. Then, non-invading cells remaining on the upper surface of the membranes were removed by wip-ing with a cotton swab. Cells attached to the lower surface were fixed with 4% formaldehyde and stained in a dye solution containing 0.05% crystal violet. Cells on the lower surface of the filter were counted in a total of six random fields under a light microscope (Olympus, Shanghai China) and three random fields were photographed under an inverted microscope (Axio Observer, Zeiss) at ×20 magnification (for PLC, MDA231) or ×10 magnification (for SKOV3, HO8910).

Tumor growth in nude miceFive-week-old BALB/c nude mice (Shanghai SLAC Laboratory Animal Co. Ltd, Shanghai, China) were used to examine the tumorigenicity of SKOV3 cells stably transfected with either a control hairpin (NC) or with shRNA tar-geting OVA66. SKOV3-NC-shRNA and SKOV3-OVA66-shRNA cells sus-pended in physiological saline were subcutaneously injected into the bilateral axillary fossa of nude mice (3 × 106 cells per side) separated into two groups (four mice per group), “Con group” inoculated with SKOV3-NC-shRNA cells and “KD group” inoculated with SKOV3-OVA66-shRNA cells. Tumor size was monitored every 5 days using a caliper during a month and the width (the short diameter) and length (the long diameter) were recorded. The volume was then calculated by the formula, V = width2 × length/2 (mm3). After then, the mice were killed by Euthanasia approach of Cervical Dislocation and tumors were removed, weighed, either frozen in liquid nitrogen or fixed in formalin and embedded in paraffin before being sectioned for subsequent analysis by immunohistochemistry (IHC) and hematoxylin and eosin staining. All ani-mal protocols were reviewed and approved by the China Institutional Ethics Review Committee for Animal Experimentation.

Samples from SKOV3-NC-shRNA and SKOV3-OVA66-shRNA cells xenografted tumors were used for immunohistochemical staining of OVA66, p-IGF-1R, p-ERK1/2 and p-Hsp27ser82 expression. Antibodies used to detect p-IGF-1R and p-ERK1/2 were from Cell Signalling technology; p-Hsp27ser82 were from Epitomics (Abcam Company) and OVA66 antibody for IHC detec-tion was prepared by our own lab (22). The secondary antibodies against mouse or rabbit immunoglobin G were supplied in an IHC kit (#GK600505) from Gene Tech Company Limited (Shanghai, China).

Receptor tyrosine kinase and mitogen-activated protein kinase signaling antibody arrayThese experiments were performed by Wayen Biotechnologies (Shanghai), using the FullMoon BioSystems Antibody Microarray (PST228 and PMK185; Full Moon BioSystems). Briefly, equal numbers (2 × 106) of SKOV3-NC-shRNA cells and SKOV3-OVA66-shRNA cells were cultured in serum-free medium for 16 h and treated with 10% FBS for 1 h. Cells were then washed with cold phosphate-buffered saline (PBS) and lysed in 300 μl of extraction buffer. Proteins from whole-cell extracts were biotinylated before being spotted on a microscope slide chamber that had various antibodies bound to its surface. Cy3-streptavidin was added, and fluorescence was detected with a microar-ray scanner (GenePix 4000B, Axon Instruments). The RayBio Human RTK Phosphorylation Antibody Array 1 (RayBiotech) was also used for identifying the relative levels of phosphorylation of 71 different human receptor tyrosine kinases (RTKs) in cell lysates of HeLa-NC-shRNA and HeLa-OVA66-shRNA cell lines treated similarly as above. All experiments were performed accord-ing to the manufacturer’s instructions. The signal intensities were quantified with Photoshop CS 2 software. Positive and negative controls were used to normalize the results from the two membranes being compared.

Cell stimulation and cell lysate preparationCells were cultured in regular growth medium containing the appropriate con-centration of puromycin. Cells were washed with PBS and serum-deprived for 16 h. Medium was then replaced with serum-free medium containing the indicated concentration of IGF-1 or FBS for the times indicated in each figure. Cells were washed twice with ice-cold PBS and lysed in M-PER (Pierce) reagent containing 1× Protease Inhibitor Cocktail and 1× Phosphatase Inhibitor Cocktail (Pierce).

Western blotWhole-cell lysates were prepared in M-PER (Pierce). The total protein con-centration was determined using a BCA Protein Assay Reagent Kit (Pierce) according to the manufacturer’s instructions. Thirty micrograms of protein were separated on a 10% sodium dodecyl sulfate–polyacrylamide gel electro-phoresis gel and transferred onto a polyvinylidene difluoride membrane (Bio-Rad). Membranes were blocked for 1 h at room temperature and incubated overnight with primary antibodies at 4ºC. The membrane was then washed 3 times with Tris-buffered saline with Tween (1× Tris-buffered saline buffer + 0.1% Tween-20), incubated with horseradish peroxidase-conjugated second-ary antibody (KPL company) for 1 h at room temperature and detected with enhanced chemiluminescence reagent (Millipore). The following primary antibodies were used: monoclonal anti-OVA66 (1B10/C5; prepared by our lab); p-IGF-1R (#3024), phospho-p44/42 mitogen-activated protein kinase (MAPK; Erk1/2; #4370), p44/42 MAPK (Erk1/2; #4695), p-Akt (Ser473; #4060) and Akt (#4685; Cell Signaling Technology); IGF-IRβ (#sc-9038), anti-ubiquitin (#sc-8017), anti-MDM2 (#sc-965) and anti-p53 (#sc-126; Santa Cruz); p-Hsp27 (pS82) (#7351-1; Epitomics); monoclonal anti-glyceralde-hyde 3-phosphate dehydrogenase (GAPDH; #G8795; Sigma) and β-ACTIN (Catalog #3598-100, BioVision) antibody.

IGF-1R-siRNA and real-time PCRWe used three siRNAs to target IGF-1R; sequences are siRNA-1: 5′-GGAGA GAACUGUCAUUUCUTT-3′ and 5′-AGAAAUGACAGUUCUCUCCTT-3′, siRNA-2: 5′-GGAGUCUUCACCACUUACUTT-3′ and 5′-AGUAAGUGGUG AAGACUCCTT-3′, and siRNA-3: 5′-GCGGUGUCCAAUAACUACATT-3′ and 5′-UGUAGUUAUUGGACACCGCTT-3′, which were purchased from GenePharma (Shanghai, China). The transfection procedures were performed with lipofectamine 2000 reagent (Invitrogen) according to the manufacturer’s specification. The following real-time PCR primers were used in our studies: OVA66: 5′-TGCTATTGAGCCTGATGG-3′ and 5′-CTGGAAGCCGTATGG TTA-3′; GAPDH: 5′-GAAGGTGAAGGTCGGAGTC-3′ and 5′-GAAGATGG TGATGGGATTTC-3′; MDM2: 5′-GAATCATCGGACTCAGGTACATC-3′ and 5′-TCTGTCTCACTAATTGCTCTCCT-3′; p53: 5′-GAGGTTGGCTCTGA CTGTACC-3′ and 5′-TCCGTCCCAGTAGATTACCAC-3′; IGF-1R: 5′-GCCG CCACTACTACTATGC-3′ and 5′-CAGAAGTCACGGTCCACA-3′. First-strand cDNA was synthesized using the RevertAid First Strand cDNA Synthesis Kit (Fermentas). Real-time PCR was performed using SYBR Premix Ex Taq kit (TaKaRa, Dalian, China) and run in an ABI 7500 Fast system (ABI). The real-time PCR results were analyzed using delta CT method. Expression of GAPDH was used as an internal control.

Flow cytometry analysis of apoptosisThe extent of apoptosis was measured using an AnnexinV-Cy5 apoptosis detection kit (BD Corporation) according to the manufacturer’s instructions. Cells were treated as indicated in the figure legends. After being trypsinized, cells were washed twice with cold PBS and gently resuspended in 400 μl binding buffer. Next, we added 5 μl of Annexin V-Cy5. Samples were gen-tly vortexed, and cells were incubated in the dark for 10 min. We next added

1574Downloaded from https://academic.oup.com/carcin/article-abstract/35/7/1573/379228by gueston 09 April 2018

Page 3: OVA66 increases cell growth, invasion and survival via regulation of

OVA66 increases cell growth, invasion and survival

3 μl of propidium iodide to each tube and incubated for another 5 min in the dark. Finally, samples were analyzed using an Accuri C6 flow cytometer (BD Corporation). Cells in the right quadrant represented apoptotic cells.

ImmunoprecipitationTreated cells were lysed as described previously. To 200 μg of protein from whole-cell extracts, 20 μl of protein G agarose beads (Pierce) and 1 μg of IGF-1R antibody were added. After overnight incubation at 4ºC on a rotator platform, the immunoprecipitates were collected by centrifugation at 3000g for 1 min, and the supernatant was discarded. The beads were then washed 5 times with lysis buffer and dissolved in loading buffer for western blot analysis.

StatisticsResults are given as mean ± SEM. Data were analyzed by two-tailed Student’s t-test. P values < 0.05 were considered to be statistically significant. Calculation was performed using GraphPad Prism software version 5.01.

Results

OVA66 increases proliferation and invasiveness in several cancer cell lines in vitroThe novel CT antigen OVA66 was identified in an ovarian cancer cDNA expression library using the serological analysis of recom-binant cDNA expression library approach. OVA66 has been shown previously to be overexpressed at both the messenger RNA (mRNA) and the protein levels in many tumor types (7). To confirm this in our own studies, we measured OVA66 protein expression in a panel of cancer cell lines using a mouse monoclonal antibody developed in our own lab (named 1B10C5), which is proved to be applicable in detect-ing the expression of OVA66 protein (Supplementary Figure S1A, available at Carcinogenesis Online). Most of the cancer cell lines we examined had elevated levels of OVA66 compared with the normal

cell lines such as peripheral blood mononuclear cells (Supplementary Figure S1B, available at Carcinogenesis Online), HEK293 and NIH3T3 cells although Wish and Chang Liver cells also expressed OVA66 (Figure  1A). Then, we used two shRNA to stably knock-down OVA66 in HeLa, PLC/PRF/5, MDA-MB-231 and SKOV3 cells lines that expressed a high level of OVA66 protein (Figure 1B). For the PLC/PRF/5 and MDA231 cell lines examined, the two OVA66 knocked down cells exhibited a decreased proliferation and inva-siveness (Figure 1C), which is consistent with the findings in HeLa cells (9). As the two shRNA-transfected cell lines showed the simi-lar consequences, we then studied shRNA-1-transfected SKOV3 cells and another ovarian cancer cell line HO8910 transfected with OVA66 gene due to its relatively lower expression level of OVA66 compared with SKOV3. Consistent with our findings in PLC/PRF/5 and MDA-MD-231, OVA66 promoted SKOV3 and HO8910 cell pro-liferation and invasiveness in vitro (Figure 1D). These data support a role of OVA66 as a novel oncogene regulating the process of tumor-genesis in several cancer cell lines.

Knockdown of OVA66 attenuates both FBS- and IGF-1-mediated activations of the IGF-1R–MAPK signaling pathwayTo examine the mechanism by which OVA66 promotes the tumorigen-esis in cervical carcinoma cell line HeLa and ovarian carcinoma cell line SKOV3, we performed a phospho-proteomics-based study using a phospho-antibody microarray, which provides a high-throughput protein analysis. We tested platform for assessing the phosphorylation status of the two cell lines (SKOV3 and HeLa) by this method. Lysates from both control and OVA66-shRNA-1 cells were analyzed on the RTK Pathway Phosphorylation Antibody Array Moon Bio-Systems and RayBiotech Human RTK Phosphorylation Antibody Array (Full Array 1).

Fig. 1. OVA66 is overexpressed in cancer cells and able to promote cell proliferative and invasive potential. (A) Cell lysates were prepared from multiple malignant and normal cell lines and then analyzed by western blot (WB) for OVA66 expression. GAPDH was used as a loading control. (B) Cancer cells HeLa, PLC/PRF/5, MDA-MB-231, and SKOV3 were stably transfected with control shRNA or two OVA66-shRNAs. WB was performed to determine expression levels in each cell line. (C) OVA66 increases PLC/PRF/5 and MDA-MB-231 cancer cell proliferation and invasion in vitro. We examined the effect of stable knockdown of OVA66 in PLC/PRF/5 and MDA231 cells both on cell viability using a Cell Counting Kit-8 assay (left) and on cell invasive potential using transwell invasion assay (right); comparisons were made to control cells. (D) Cell Counting Kit-8 and transwell invasion assays were performed using OVA66-shRNA-1-transfected SKOV3 cells and OVA66 gene-transfected HO8910 cells. WB analysis and growth curves are shown on the left, whereas cell invasion assays are shown on the right side. Representative images from the invasion assay are shown. The values shown are expressed as the means ± SEM. *P ≤ 0.05.

1575Downloaded from https://academic.oup.com/carcin/article-abstract/35/7/1573/379228by gueston 09 April 2018

Page 4: OVA66 increases cell growth, invasion and survival via regulation of

Wei Rao et al.

As shown in Figure  2A, both Hela-OVA66-shRNA and SKOV3-OVA66-shRNA cells showed a reduction in phosphorylated IGF-1R, compared with their respective controls. We confirmed these find-ings by western blot for phospho-IGF-IR following shRNA-mediated knockdown of OVA66 in HeLa, MDA-MB-231, SKOV3 and OVA66 gene-transfected HO8910 cells. Both of the shRNA-transfected HeLa cells, MDA-MB-231 and SKOV3, showed a reduction in phosphoryla-tion of IGF-1R and ERK1/2 under IGF-1 stimulation, whereas OVA66 overexpressed HO8910 cells showed an increased level of p-IGF-1R and p-ERK1/2 (Figure  2B). We then used the shRNA-1-mediated OVA66 knocked down cells to study if OVA66 could also influence the FBS-mediated IGF-1R–ERK1/2 activation in these cells. Our data showed that under FBS condition OVA66 was able to influence the level of p-IGF-1R and p-ERK1/2 (Figure 2C). Besides, OVA66 over-expressed HO8910 cells also showed an upregulated level of p-IGF-1R and p-ERK1/2 under FBS stimuli (Supplementary Figure S2, avail-able at Carcinogenesis Online). Based on these data, we hypothesize that OVA66 influences tumorigenesis, at least in part, via the IGF-1R–ERK1/2–MAPK signaling pathway.

Silencing OVA66 in SKOV3 cells inhibits tumor growth in vivoBecause depletion of OVA66 in SKOV3 cells decreased proliferation in vitro, we wanted to examine if the same was true in vivo. We per-formed xenograft studies in nude mice separated into two groups (n = 4 per group) by injecting the SKOV3 control cells or OVA66-shRNA

(shRNA-1) cells into the bilateral axillary fossa of nude mice before monitoring tumor growth for 30 days. Both the average tumor vol-ume over 30  days of measurements and the average tumor weight of four tumors between the two groups were recorded. Tumors from mice inoculated with SKOV3-OVA66-shRNA cells were significantly smaller than tumors from mice injected with control cells (Figure 3A and B). Mice were killed 30 days after injection, and paraffin-embed-ded tumor sections were examined histopathologically (Figure  3C) and immunohistochemically (Figure 3D). Tumors from the SKOV3-OVA66-shRNA group were practically negative for OVA66 expres-sion, confirming successful knockdown; cells from this group also showed decreased levels of phosphorylated IGF-1R and ERK1/2 (Figure  3D). Thus, our IHC results validated our in vitro findings showing that knockdown of OVA66 in SKOV3 cells inhibited IGF-1R and ERK1/2–MAPK signaling.

MAPK antibody array shows decreased pHsp27 in SKOV3-OVA66-shRNA cellsOur initial findings showed that OVA66 regulates the ERK1/2–MAPK signaling pathway. Therefore, we performed a secondary anti-body microarray to detect the phosphorylation of proteins involved in MAPK signaling (Full Moon Bio-systems Antibody microarray). We identified proteins whose phosphorylation levels were decreased by at least 1.5-fold when OVA66 was stably knocked down in SKOV3 cells. Many of the proteins meeting this cutoff, including Elk-1,

Fig. 2. Stable knockdown of OVA66 in HeLa or SKOV3 cells decreases phosphorylation and activation of the IGF-1R signaling pathway. (A) Two RTK signaling phospho-antibody microarrays from RayBiotech and Moon Bio-Systems were examined for the effect of OVA66 silencing on full proteins of HeLa and SKOV3 cell lines. The extent of protein phosphorylation was normalized within each slide, and comparisons were made between OVA66 stable knockdown cells and control cells. Both HeLa and SKOV3 cells depleted of OVA66 showed decreased phosphorylation of IGF-1R. (B) Stable knockdown of OVA66 by two OVA66-shRNAs in HeLa (cervical carcinoma cell), MDA-MB-231 (breast cancer cell) and SKOV3 (ovarian carcinoma cell) showed decreased phosphorylation of IGF-1R and ERK1/2, whereas overexpression of OVA66 in ovarian carcinoma HO8910 cells showed increased phosphorylation level of IGF-1R and ERK1/2 in response to 10 ng/ml IGF-1 stimuli (for the indicated time after serum deprived for 16 h). (C) OVA66 activates the IGF-1R and ERK1/2–MAPK signaling pathway in SKOV3, HeLa and MDA-MB-231 cells. OVA66-depleted SKOV3 cells were starved for 16 h and then treated with 10% FBS. The treatments were maintained for the indicated times. Total cell lysates were resolved by sodium dodecyl sulfate–polyacrylamide gel electrophoresis and analyzed by WB using the indicated antibodies. Detection of GAPDH was used as a loading control. Total cell lysates of OVA66-depleted HeLa and MDA231 cells normally cultivated in 10% FBS were extracted and analyzed by WB. β-actin was detected as a loading control.

1576Downloaded from https://academic.oup.com/carcin/article-abstract/35/7/1573/379228by gueston 09 April 2018

Page 5: OVA66 increases cell growth, invasion and survival via regulation of

OVA66 increases cell growth, invasion and survival

Hsp27, Pyk2, apoptosis signal-regulating kinase 1 (ASK1), c-Kit, c-jun N-terminal kinase, activating transcription factor 2 and ERK1/2, are well known to be involved various tumorigenic processes, such as invasion, metastasis and apoptosis (Figure 4A). We performed immu-noblotting to confirm that knockdown of OVA66 in SKOV3 cells resulted in decreased IGF-1-mediated phosphorylation of ERK1/2 and Hsp27 (Figure 4B).

Consistent with our knockdown findings, overexpression of OVA66 in either HO8910 or HEK293 cells increased phosphorylation of ERK1/2 and Hsp27 (Figure 4B and C). These findings were also con-firmed by IHC for p-Hsp27 (Ser82) level of tumor sections from nude mice inoculated with either SKOV3 control cells or SKOV3/OVA66-shRNA cells (Figure 4D). These data suggest that OVA66 may func-tion through affecting the phosphorylated level of ERK1/2–Hsp27 to regulate tumorigenesis.

Inhibition of IGF-1R by siRNA and the inhibitor Linsitinib both inhibited the OVA66 upregulated and downregulated cell growth, invasive and antiapoptotic capacitiesWe next sought to determine if IGF-IR is a critical downstream medi-ator of OVA66 function. We successfully depleted IGF-IR protein lev-els using three different siRNAs (Supplementary Figure S3, available at Carcinogenesis Online); siRNA-1 appeared to be the most effective of the three. The siRNA-mediated knockdown of IGF-IR significantly inhibited IGF-1-mediated activation of ERK1/2 and Hsp27 in SKOV3 cells (Supplementary Figure S4, available at Carcinogenesis Online). Similarly, inhibition of IGF-1R with the small molecule inhibitor

Linsitinib (23) also reduced phosphorylation level of ERK1/2 and Hsp27 (Figure 5A). OVA66 increased cellular proliferation of both SKOV3 and HO8910 cells under normal cultivated or control condi-tions by adding same volume of dimethyl sulfoxide. However, this effect was attenuated in the presence of 1 uM Linsitinib (Figure 5B). OVA66-mediated cell invasion was also inhibited by both IGF-1R-siRNA and Linsitinib (Figure 5C and Supplementary Figure S5, avail-able at Carcinogenesis Online). We also found that OVA66 promotes resistance of both cell lines to apoptosis induced by multiple stimuli, including nutrient deprivation (Supplementary Figure S6, available at Carcinogenesis Online) and 5-fluorouracil; treatment with Linsitinib was able to attenuate this effect, regardless of OVA66 expression (Figure 5D and Supplementary Figure S7, available at Carcinogenesis Online). These data support the fact that OVA66-mediated SKOV3 and HO8910 cell proliferation, invasion and survival are dependent on activation of IGF-1R and possibly downstream ERK1/2-Hsp27 signaling.

OVA66 and MDM2 are coexpressed in cancer cells and regulate ubiquitination of IGF-1RMDM2 is an ubiquitin ligase of IGF-1R (19), and IGF-1R signaling is, probably, controlled by ubiquitination of the receptor (20). Previous reports have shown that OVA66-depleted HeLa cells also have decreased mRNA levels of MDM2 (9). Thus, we examined mRNA expression of both MDM2 and IGF-IR in the several cell lines we used for our experiments. The level of MDM2 transcripts were cor-related with mRNA level of OVA66 in both HeLa and SKOV3 cells,

Fig. 3. ShRNA-1-mediated stable knockdown of OVA66 in SKOV3 cells inhibits tumor formation in nude mice. (A) SKOV3-OVA66-shRNA or SKOV3 control cells (3 million each) were subcutaneously injected into the two flanks of 5-week-old nude mice (N = 4). Images are of representative mice from each injection group and all the eight tumors of two xenografted groups. (B) Tumor volumes (mm3) were monitored every 5 days for 30 days and tumor weights (g) were measured. Values represent means ± SEM, *P ≤ 0.05. (C) Representative photomicrographs of hematoxylin and eosin-stained tumor tissues from control and OVA66-shRNA SKOV3 cells (original magnification: ×100). (D) Representative images of IHC performed for expression of OVA66, p-IGF-1R (Y1135/1136) and p-ERK1/2 in tumor xenografts from control and OVA66 knockdown SKOV3 cells (original magnification: ×200); images show reduced expression of OVA66, p-IGF-1R and p-ERK1/2 in OVA66 knocked down SKOV3 xenografted tumor tissues.

1577Downloaded from https://academic.oup.com/carcin/article-abstract/35/7/1573/379228by gueston 09 April 2018

Page 6: OVA66 increases cell growth, invasion and survival via regulation of

Wei Rao et al.

as well as OVA66 overexpressed HO8910 cells, whereas IGF-1R transcript showed no difference (Figure 6A). Furthermore, p53 pro-tein levels were increased following OVA66 knockdown in HeLa and SKOV3 cells and decreased following overexpression of OVA66 in HO8910 and HEK293 (Supplementary Figure S8A, available at Carcinogenesis Online). There were no changes in p53 mRNA levels (Supplementary Figure S8B, available at Carcinogenesis Online), sug-gesting that the regulation occurs at the protein level. Similar results were obtained with SKOV3-NC/OVA66-shRNA and HO8910-pIRES-alone/OVA66 cells treated with a high concentration of 5-fluorouracil (Supplementary Figure S9, available at Carcinogenesis Online). Based on these findings, we hypothesized that OVA66 and MDM2 are, proba-bly, coexpressed in cancer cells. We next performed immunoprecipita-tion experiments to assess the ubiquitination status of IGF-1R in cells stimulated with its ligand IGF-1. In HeLa and SKOV3 cells, knock-down of OVA66 reduced IGF-1-mediated ubiquitination of IGF-1R; phospho-IGF-1R, ERK1/2 and Hsp27 were also significantly reduced (Figure 6B). Similarly, overexpression of OVA66 in both HO8910 and HEK293 cells enhanced IGF-1R ubiquitination following IGF-1 treat-ment (Figure 6C). Our data suggest a role for OVA66 and MDM2 in regulation of IGF-IR–ERK1/2 signaling in these cells.

Discussion

OVA66 was first identified in an ovarian cancer cDNA expression library (7). It functions as an oncoprotein in HeLa cells, promoting

proliferation, invasion and survival (9). Here, we find that OVA66 also regulates tumorigenesis in two ovarian cancer cell lines, SKOV3 and HO8910. Stable knockdown of OVA66 in SKOV3 cells inhibited cell growth both in vitro and in vivo; invasion was also attenuated, and apoptosis in response to either nutrient deprivation or 5-fluorouracil treatment was increased. Overexpression of OVA66 in HO8910 led to consistent findings. We next investigated the signaling molecules that might act downstream of OVA66 using high-throughput RTK and MAPK antibody arrays. Following OVA66 depletion by shRNA, we identified several molecules whose phosphorylation was reduced, including IGF-1R, ERK1/2, Hsp27, ELK-1, Pyk2, ASK1, c-Kit, c-jun N-terminal kinase and activating transcription factor 2. Thus, OVA66 is associated with activation of several signaling molecules implicated in tumor development.

As is currently reported, active IGF-1R and its downstream signal-ing play a critical role in cell growth, metastasis and resistance to chemotherapy and targeted drugs (10,24,25). High levels of Hsp27 have been reported in many cancers, including breast (26), ovar-ian(27), and prostate(28). Additionally, phosphorylation of Hsp27 is associated with tumor cell migration, invasion and survival (29–32). The transcription factor ELK-1 can be phosphorylated by ERK1/2 in response to serum or growth factors and mediates expression of a number of genes, including the proto-oncogene c-fos (33,34). Protein tyrosine kinase Pyk2 is part of the MAPK signaling pathway and plays a critical role in regulating cancer cell motility and invasion (35–37). ASK1 plays a causal role in cell death induced by a number

Fig. 4. OVA66 enhances phosphorylation of proteins in the MAPK signaling pathway. (A) Full Moon BioSystems MAPK signaling phospho-specific antibody array was performed. The experiment included six replicates (vertical columns) of both phospho-specific and total antibodies, which were assessed against both control and OVA66-depleted SKOV3 cells. The graph on the right lists proteins whose level of phosphorylation was ~1.5-fold decreased in OVA66-depleted cells compared with control cells. (B) OVA66 enhances IGF-1R-mediated activation of ERK1/2 and Hsp27. SKOV3 (both control and OVA66-shRNA cells) and HO8910 (both empty vector and OVA66-overexpressing cells) were maintained in serum-free medium for 16 h before being treated with 10 ng/ml IGF-1 for the times indicated. Protein was then extracted and analyzed by WB using the indicated antibodies. (C) HEK293 cells were transiently transfected with different amount of pIRES-puro-OVA66 or empty vector. After 48 h, cells were plated and maintained in serum-free medium for 16 h before being treated with 10 ng/ml IGF-1 for the indicated times. Whole-cell lysates were prepared and probed with indicated antibodies. GAPDH was used as a loading control. Phosphorylation of IGF-1R, ERK1/2 and Hsp27 was increased with OVA66 overexpression; total IGF-1R protein showed unchanged. (D) IHC was performed to validate the reduction of phosphorylated Hsp27 (Ser82) in tumor xenografts of OVA66-depleted SKOV3 cells compared with control cells. Representative photomicrographs are shown at magnifications of ×200 and ×400.

1578Downloaded from https://academic.oup.com/carcin/article-abstract/35/7/1573/379228by gueston 09 April 2018

Page 7: OVA66 increases cell growth, invasion and survival via regulation of

OVA66 increases cell growth, invasion and survival

of stimuli (38,39); however, phosphorylation of ASK1 at serine 83 attenuates ASK1-induced apoptosis and promotes cell survival (40). The c-Kit proto-oncogene encodes a transmembrane tyrosine kinase receptor; it is aberrantly expressed in several cancers and correlates with poor prognosis (41). Additionally, c-Kit activation protects can-cer cells from apoptosis and enhances their invasive potential (42,43). Activating transcription factor 2 has been shown to be oncogenic by increasing expression of target genes, such as cyclinD1, MMP-2, cytokines, E-selectin and bcl-2 (44,45). In this study, our experimental results unmasked a critical role of OVA66 in influencing the IGF-1R–ERK1/2–Hsp27 signaling in two ovarian cancer cell lines SKOV3 and HO8910, by methods of RTK and MAPK antibody arrays and addi-tional analyses of phosphorylation of IGF-1R, ERK1/2 and Hsp27 regulated by OVA66 in both ovarian cancer cell lines examined. We subsequently blocked the IGF-1R signaling and observed that OVA66 was retarded in promoting proliferative, invasive and stress resistant potential in both ovarian carcinoma cells SKOV3 and HO8910, espe-cially the effect of both OVA66 knockdown and IGF-1R blocking was quite similar to merely inhibiting the IGF-1R signaling, suggesting that OVA66 functioning was at least partially dependent on the activa-tion of IGF-1R signaling. Based on these, we proposed that OVA66 was not only able to influence the IGF-1R–ERK1/2–Hsp27 signal-ing but also relying on the activation of IGF-1R signaling to promote tumorigenesis in two ovarian cancer cell line SKOV3 and HO8910.

Tumorigenesis arises from combined deregulation of proliferation and apoptosis (46). In this study, we identified a novel oncoprotein, OVA66, which enhances proliferative, invasive and antiapoptotic capacity of two ovarian cancer cell lines, SKOV3 and HO8910. We also identified the correlation between OVA66 expression and IGF-1R signaling, proposing that OVA66 played an upstream role in influenc-ing IGF-1R–ERK1/2–Hsp27 signaling and promoted tumorigenesis via IGF-1R signaling. Although, it is reported that therapies to inhibit IGF-1R action seemed to be clinically useful for the treatment of can-cer (13–15), we suggested in this study that another tumor associ-ated protein OVA66 may play a crucial part in regulating the IGF-1R hyperactivation in at least the two ovarian cancer cells SKOV3 and HO8910. However, our results of antibody arrays also identified several other signaling molecules that were inhibited after OVA66 knockdown, suggesting other pathways might be involved in OVA66 promoting the process of tumorigenesis. To further elucidate the tumorigenic role and mechanisms of OVA66 and the potentiality of OVA66 as a therapeutic target in cancer, more detailed investigations must be performed and our research on such as identifying networks of OVA66 interacting proteins (recent findings showed some heat shock proteins such as hsp47 could directly bind to OVA66), clini-cal correlations between expression of OVA66 and other biomarkers such as IGF-1R, other siganling pathways that might be influenced by OVA66 in different types of cancer (recent study suggested the

Fig. 5. Inhibition of IGF-1R signaling decreases proliferation, invasion and resistance to apoptosis. (A) Both SKOV3-NC/OVA66-shRNA and HO8910-pIRESpure-vector/OVA66 cells were starved for 16 h and then pretreated with 1 uM Linsitinib or dimethyl sulfoxide for 1 h. Next, cells were treated with 10 ng/ml IGF-1 for the indicated times. Whole-cell lysates were extracted, resolved by sodium dodecyl sulfate–polyacrylamide gel electrophoresis and probed with antibodies indicated. Detection of GAPDH served as a loading control. (B) Normally, OVA66 was able to promote cell growth in SKOV3 and HO8910 cells. Nevertheless, OVA66-mediated cell viability of both SKOV3 and HO8910 cells was then abolished in the presence of the IGF-1R inhibitor Linsitinib. Cell growth was monitored for the indicated times using Cell Counting Kit-8 reagents. In Linsitinib-treated cell proliferative assays, proliferation values are displayed as percentage of untreated cells plated on the 1st day. (C) Transwell invasion assays from SKOV3-NC/OVA66-shRNA and HO8910-pIRESpuro-vector/OVA66 cells treated with 1 uM Linsitinib. Representative images are shown on the left, and the quantification of >3 selected fields is shown on the right. (D) Apoptosis was assessed by Annexin V-Cy5 staining following treatment with 200 ug/ml 5--fluorouracil for 48 h in addition to treatment with dimethyl sulfoxide or 1 uM Linsitinib. Representative fluorescence activated cell sorter images are shown. All data are expressed as means ± SEM of three independent experiments. *P ≤ 0.05, **P ≤ 0.01.

1579Downloaded from https://academic.oup.com/carcin/article-abstract/35/7/1573/379228by gueston 09 April 2018

Page 8: OVA66 increases cell growth, invasion and survival via regulation of

Wei Rao et al.

VEGFA-KDR signaling was regulated by OVA66 in gastric cancer cells) etc. are now ongoing.

Supplementary material

Supplementary Figures S1–S9 can be found at http://carcin.oxford-journals.org/

Funding

National Natural Science Foundation of China (81272328, 30972786); State National Science Foundation of China (J50207); Doctoral Innovation Foundation of Shanghai Jiao Tong University School of Medicine (BXJ201205).

Conflict of Interest Statement: None declared.

References

1. Caballero,O.L. et al. (2009) Cancer/testis (CT) antigens: potential targets for immunotherapy. Cancer Sci., 100, 2014–2021.

2. Zendman,A.J. et  al. (2003) Cancer/testis-associated genes: identifica-tion, expression profile, and putative function. J. Cell. Physiol., 194, 272–288.

3. Scanlan,M.J. et al. (2004) The cancer/testis genes: review, standardization, and commentary. Cancer Immun., 4, 1.

4. Suri,A. (2006) Cancer testis antigens –their importance in immuno-therapy and in the early detection of cancer. Expert Opin Biol Ther, 6, 379–389.

5. Simpson,A.J. et al. (2005) Cancer/testis antigens, gametogenesis and can-cer. Nat. Rev. Cancer, 5, 615–625.

6. Sahin,U. et al. (1997) Serological identification of human tumor antigens. Curr. Opin. Immunol., 9, 709–716.

7. Jin,S. et al. (2008) Humoral immune responses against tumor-associated antigen OVA66 originally defined by serological analysis of recombinant cDNA expression libraries and its potentiality in cellular immunity. Cancer Sci., 99, 1670–1678.

8. Yang,X.F. et  al. (2001) CML66, a broadly immunogenic tumor antigen, elicits a humoral immune response associated with remission of chronic myelogenous leukemia. Proc. Natl Acad. Sci. USA, 98, 7492–7497.

9. Wang,Q. et al. (2008) RNA interference targeting CML66, a novel tumor antigen, inhibits proliferation, invasion and metastasis of HeLa cells. Cancer Lett., 269, 127–138.

10. Larsson,O. et al. (2005) Role of insulin-like growth factor 1 receptor sig-nalling in cancer. Br. J. Cancer, 92, 2097–2101.

11. Fürstenberger,G. et  al. (2002) Insulin-like growth factors and cancer. Lancet Oncol., 3, 298–302.

12. Yee,D. (2012) Insulin-like growth factor receptor inhibitors: baby or the bathwater? J. Natl Cancer Inst., 104, 975–981.

13. Gualberto,A. et  al. (2009) Emerging role of insulin-like growth factor receptor inhibitors in oncology: early clinical trial results and future direc-tions. Oncogene, 28, 3009–3021.

14. Klinakis,A. et al. (2009) IGF1R as a therapeutic target in a mouse model of basal-like breast cancer. Proc. Natl Acad. Sci. USA, 106, 2359–2364.

Fig. 6. OVA66 is coexpressed with MDM2 and regulates ubiquitination of IGF-1R and activation of downstream signaling. (A) Depletion of OVA66 in HeLa and SKOV3 cells reduces the mRNA level of MDM2. Overexpression of OVA66 in HO8910 cells increases MDM2 mRNA levels; analyses were performed by real-time PCR. The expression of target genes was normalized to GAPDH, and the fold change was calculated by the 2−ΔΔCt method. *P ≤ 0.05, **P ≤ 0.01 (B) HeLa and SKOV3 cells stably transfected with control or OVA66-shRNA were incubated in serum-free medium for 16 h and then treated with 10 ng/ml IGF-1 for 0, 30 and 60 min. Protein lysates were analyzed by WB. GAPDH served as a loading control. IGF-1R was then immunoprecipitated from the same lysates, which were prepared from cells treated with IGF-1 for 30 and 60 min. The immunoprecipitates were analyzed by WB for ubiquitination, p-IGF-1R and total IGF-1R (t-IGF-1R). (C) HO8910-pIRESpuro-vector/OVA66 cells were starved for 16 h before treatment with 10 ng/ml IGF-1 for the indicated times. Whole-cell lysates were analyzed by WB and IP as indicated above. HEK293 cells were transiently transfected with pIRESpuro-vector, as well as 1 ug and 2 ug pIRESpuro-OVA66 vectors. Forty-eight hours later, cells were incubated in serum-free medium for 16 h and then treated with 10 ng/ml IGF-1 for 60 min. Proteins were harvested and analyzed by both WB and IP as indicated above.

1580Downloaded from https://academic.oup.com/carcin/article-abstract/35/7/1573/379228by gueston 09 April 2018

Page 9: OVA66 increases cell growth, invasion and survival via regulation of

OVA66 increases cell growth, invasion and survival

15. Pollak,M. (2012) The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nat. Rev. Cancer, 12, 159–169.

16. Avnet,S. et al. (2009) Insulin receptor isoform A and insulin-like growth factor II as additional treatment targets in human osteosarcoma. Cancer Res., 69, 2443–2452.

17. Ulanet,D.B. et al. (2010) Insulin receptor functionally enhances multistage tumor progression and conveys intrinsic resistance to IGF-1R targeted therapy. Proc. Natl Acad. Sci. USA, 107, 10791–10798.

18. Zheng,H. et al. (2012) β-Arrestin-biased agonism as the central mechanism of action for insulin-like growth factor 1 receptor-targeting antibodies in Ewing’s sarcoma. Proc. Natl Acad. Sci. USA, 109, 20620–20625.

19. Girnita,L. et al. (2003) Mdm2-dependent ubiquitination and degradation of the insulin-like growth factor 1 receptor. Proc. Natl Acad. Sci. USA, 100, 8247–8252.

20. Sehat,B. et al. (2007) Role of ubiquitination in IGF-1 receptor signaling and degradation. PLoS One, 2, e340.

21. Girnita,L. et al. (2007) Beta-arrestin and Mdm2 mediate IGF-1 receptor-stimulated ERK activation and cell cycle progression. J. Biol. Chem., 282, 11329–11338.

22. You,Q. et al. (2003) [Cloning, prokaryotic expression of CML66 and prep-aration of its polyclonal antibody]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi, 19, 582–584.

23. Fox,E.M. et al. (2011) A kinome-wide screen identifies the insulin/IGF-I receptor pathway as a mechanism of escape from hormone dependence in breast cancer. Cancer Res., 71, 6773–6784.

24. Rosenzweig,S.A. et al. (2010) Defining the pathway to insulin-like growth factor system targeting in cancer. Biochem. Pharmacol., 80, 1115–1124.

25. Knowlden,J.M. et  al. (2008) Insulin receptor substrate-1 involvement in epidermal growth factor receptor and insulin-like growth factor receptor signalling: implication for Gefitinib (‘Iressa’) response and resistance. Breast Cancer Res. Treat., 111, 79–91.

26. Conroy,S.E. et al. (1998) Antibodies to heat-shock protein 27 are associ-ated with improved survival in patients with breast cancer. Br. J. Cancer, 77, 1875–1879.

27. Arts,H.J. et al. (1999) Heat-shock-protein-27 (hsp27) expression in ovar-ian carcinoma: relation in response to chemotherapy and prognosis. Int. J. Cancer, 84, 234–238.

28. Cornford,P.A. et al. (2000) Heat shock protein expression independently predicts clinical outcome in prostate cancer. Cancer Res., 60, 7099–7105.

29. Shin,K.D. et al. (2005) Blocking tumor cell migration and invasion with biphenyl isoxazole derivative KRIBB3, a synthetic molecule that inhibits Hsp27 phosphorylation. J. Biol. Chem., 280, 41439–41448.

30. Shiota,M. et al. (2013) Hsp27 regulates epithelial mesenchymal transition, metastasis, and circulating tumor cells in prostate cancer. Cancer Res., 73, 3109–3119.

31. Mehlen,P. et al. (1996) Small stress proteins as novel regulators of apopto-sis. Heat shock protein 27 blocks Fas/APO-1- and staurosporine-induced cell death. J. Biol. Chem., 271, 16510–16514.

32. Garrido,C. et  al. (1997) HSP27 as a mediator of confluence-dependent resistance to cell death induced by anticancer drugs. Cancer Res., 57, 2661–2667.

33. Cavigelli,M. et  al. (1995) Induction of c-fos expression through JNK-mediated TCF/Elk-1 phosphorylation. EMBO J., 14, 5957–5964.

34. Kortenjann,M. et al. (1994) Inhibition of v-raf-dependent c-fos expression and transformation by a kinase-defective mutant of the mitogen-activated protein kinase Erk2. Mol. Cell. Biol., 14, 4815–4824.

35. Watson,J.M. et  al. (2001) Inhibition of the calcium-dependent tyrosine kinase (CADTK) blocks monocyte spreading and motility. J. Biol. Chem., 276, 3536–3542.

36. Genua,M. et al. (2012) Proline-rich tyrosine kinase 2 (Pyk2) regulates IGF-I-induced cell motility and invasion of urothelial carcinoma cells. PLoS One, 7, e40148.

37. Behmoaram,E. et  al. (2008) Focal adhesion kinase-related proline-rich tyrosine kinase 2 and focal adhesion kinase are co-overexpressed in early-stage and invasive ErbB-2-positive breast cancer and cooperate for breast cancer cell tumorigenesis and invasiveness. Am. J.  Pathol., 173, 1540–1550.

38. Chang,H.Y. et al. (1998) Activation of apoptosis signal-regulating kinase 1 (ASK1) by the adapter protein Daxx. Science, 281, 1860–1863.

39. Chen,Z. et al. (1999) ASK1 mediates apoptotic cell death induced by geno-toxic stress. Oncogene, 18, 173–180.

40. Kim,A.H. et al. (2001) Akt phosphorylates and negatively regulates apop-tosis signal-regulating kinase 1. Mol. Cell. Biol., 21, 893–901.

41. Sammarco,I. et al. (2004) Expression of the proto-oncogene c-KIT in nor-mal and tumor tissues from colorectal carcinoma patients. Int. J. Colorectal Dis., 19, 545–553.

42. Bellone,G. et al. (2001) Aberrant activation of c-kit protects colon carci-noma cells against apoptosis and enhances their invasive potential. Cancer Res., 61, 2200–2206.

43. Attoub,S. et al. (2002) The c-kit tyrosine kinase inhibitor STI571 for colo-rectal cancer therapy. Cancer Res., 62, 4879–4883.

44. Lau,E. et al. (2012) ATF2 - at the crossroad of nuclear and cytosolic func-tions. J. Cell Sci., 125(Pt 12), 2815–2824.

45. Vlahopoulos,S.A. et  al. (2008) The role of ATF-2 in oncogenesis. Bioessays, 30, 314–327.

46. Green,D.R. et  al. (2002) A matter of life and death. Cancer Cell, 1, 19–30.

Received September 26, 2013; revised February 16, 2014; accepted February 19, 2014

1581Downloaded from https://academic.oup.com/carcin/article-abstract/35/7/1573/379228by gueston 09 April 2018