Overexpression of secretory phospholipase A2-IIa supports cancer stem cell phenotype via HER/ERBB-elicited signaling in lung and prostate cancer cells

  • Authors:
    • Shan Lu
    • Zhongyun Dong
  • View Affiliations

  • Published online on: April 19, 2017     https://doi.org/10.3892/ijo.2017.3964
  • Pages: 2113-2122
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Resistance to conventional chemotherapies remains a significant clinical challenge in treatment of cancer. The cancer stem cells (CSCs) have properties necessary for tumor initiation, resistance to therapy, and progression. HER/ERBB‑elicited signaling supports CSC properties. Our previous studies revealed that secretory phospholipase A2 group IIa (sPLA2‑IIa) is overexpressed in both prostate and lung cancer cells, leading to an aberrant high level in the interstitial fluid, i.e., tumor microenvironment and blood. HER/ERBB-PI3K-Akt-NF-κB signaling stimulates sPLA2‑IIa overexpression, and in turn, sPLA2‑IIa activates EGFR family receptors and HER/ERBB-elicited signaling and stimulates sPLA2‑IIa overexpression in a positive feedback manner. The present study determined the molecular mechanisms of sPLA2‑IIa in stimulating HER/ERBB-elicited signaling and supporting CSC properties. We found that sPLA2‑IIa binds both EGFR and HER3 demonstrated by co-immunoprecipitation experiments and also indirectly interacts with HER2, suggesting that sPLA2‑IIa functions as a ligand for both EGFR and HER3. Furthermore, both side population CSCs from non-small cell lung cancer (NSCLC) A549 and H1975 cells and ALDH1‑high CSCs from castration-resistant prostate cancer (CRPC) 22Rv1 cells overexpress sPLA2‑IIa and produce tumors when inoculated into subcutis of nude mice. Given an aberrant high level of sPLA2‑IIa in the tumor microenvironment that should be much higher than that in the blood, our findings support the notion that sPLA2‑IIa functions as a ligand for EGFR family receptors and supports CSC properties via HER/ERBB-elicited signaling, which may contribute to resistance to therapy and cancer progression.

Introduction

The cancer stem cells (CSCs) are a rare population of tumor cells and enable them to simultaneously self-perpetuate with a consistently maintained CSC subpopulation and to generate differentiated progeny via asymmetrical cell division, giving rise to heterogenic tumors (13). CSCs are relatively quiescent and have the properties necessary for tumor initiation, resistance to therapy, and progression (47). The CSC population expands during periods of stress, such as radiation (8,9), chemotherapy (5), and castration (10), which are likely the initiating cells of chemoresistant cancer relapses and metastases. Based on CSC markers, a number of CSC populations have been identified. For instance, the side population (SP) cells, which express high levels of ABCG2 and ABCB1 multi-drug efflux pumps that may contribute to chemoresistance, possess the stem cell-like properties (2,3,11). The aldehyde dehydrogenases (ALDH) are cytosolic isoenzymes responsible for oxidizing intracellular aldehydes. The ALDH1-high cell subpopulations show distinct stem-like characteristics and are highly resistant to chemotherapeutic agents commonly used as first-line therapy, such as cisplatin and docetaxel (2,3). The current anticancer therapies fail to destroy, but tend to favor the selection and expansion of resistant CSCs in tumors, resulting in poor responses and outcomes. The elimination of CSCs is of utmost importance at the time of therapeutic intervention in order to prevent CSC expansion and subsequent tumor recurrence, relapse, and metastasis.

Secretory phospholipase A2 group IIa (sPLA2-IIa) is distributed in trace amounts in a variety of normal mammalian tissues, but found at high levels in various inflamed tissues and some cancers. sPLA2-IIa, a NF-κB target gene (12,13), has traditionally been associated with their enzymatic activity and participates in biosynthesis of potent biologically active lipid mediators, particularly arachidonic acid-derived eicosanoids, which promote inflammation, angiogenesis, and tumorigenesis (14,15). Independent of its enzymatic activity, sPLA2-IIa can also serve as a ligand for cell membrane receptors and stimulate integrin activation, COX-2 expression, and secretion of cytokines (1618). sPLA2-IIa binds to integrin avβ3 at KD of 0.2 mM, and induces cell proliferation (16). sPLA2-IIa also weakly interacts with M-type receptor in human cells and induces pro-inflammatory signaling (15,19,20).

sPLA2-IIa is associated with the pathology of several types of malignancies, including cancers of the colon, breast, stomach, esophagus, ovary, lung, and prostate (21,22). sPLA2-IIa is overexpressed in almost all human prostate cancer tissues and elevated levels are associated with advanced tumor grades (13,2326). sPLA2-IIa remains elevated in androgen-independent prostate cancers (27) and is significantly increased in metastatic lesions (28). We are the first to demonstrate that cancer cells overexpress and secrete sPLA2-IIa into the interstitial fluids, i.e., tumor microenvironment and blood, in patients with both prostate and lung cancers (13,29,30). High levels of plasma sPLA2-IIa are associated with poor prognosis, advanced cancer stage, and short cancer survival. We confirmed that tumors secret sPLA2-IIa into the circulation to a detectable level in the mouse model of human cancer (29). The most recent report by others supports our finding in that high levels of plasma sPLA2-IIa are associated with poor prognosis of cancers (31). More importantly, we revealed that elevated HER/ERBB-PI3K-Akt-NF-κB signaling induces sPLA2-IIa overexpression and secretion in both lung and prostate cancer cells; in turn, sPLA2-IIa stimulates its overexpression via HER/ERBB-elicited signaling in the positive feedback regulation manner (26,32). sPLA2-IIa induces phosphorylation of HER2 and HER3 in a dose-dependent manner in NSCSC A549 and H1975 cells (32). However, the underlying molecular mechanisms of sPLA2-IIa at an aberrant high level in the tumor microenvironment in stimulating cancer progression and metastasis remains to be elucidated.

The HER/ERBB-elicited signaling is essential for cell growth and survival. Recent studies further demonstrate that this signaling pathway is of critical importance in supporting CSC properties (3335). We determined a role of sPLA2-IIa in CSCs and revealed that sPLA2-IIa is overexpressed in both side population (SP) CSCs from NSCSC cells and ALDH1-high CSCs from CRPC cells. Furthermore, sPLA2-IIa directly interacts with and activates EGFR family receptors, suggesting that sPLA2-IIa is a ligand for both EGFR and HER3. These findings, together with our previous data (13,29,30,32), support the notion that high levels of sPLA2-IIa in the tumor micro-environment support CSC phenotype via HER/ERBB-elicited signaling, and sPLA2-IIa is a novel therapeutic target against cancer and CSCs.

Materials and methods

Reagents

RPMI-1640 medium was purchased from Invitrogen (Gaithersburg, MD, USA). Fetal bovine serum (FBS) and charcoal/dextran-treated FBS were purchased from Hyclone Laboratories (Logan, UT, USA). sPLA2-IIa antibodies were obtained from Life Span BioSciences (Seattle, WA, USA) and Cayman Chemical (Ann Arbor, MI, USA). Recombinant human sPLA2-IIa was obtained from BioVendor (Candler, NC, USA). EGFR, HER2, and HER3 antibodies were from Cell Signaling Technology (Danvers, MA, USA) and Santa Cruz Biotechnology (Santa Cruz, CA, USA). RT-PCR primers were customly synthesized by Genscript (Piscataway, NJ, USA). Plasmid sPLA2-IIa(−800)-Luc was constructed as we described previously (13).

Cell culture

The human prostate adenocarcinoma cell lines LNCaP and 22Rv1, and lung cancer adenocarcinoma H1975 were obtained from ATCC (Rockville, MD, USA) and maintained in RPMI-1640 medium supplemented with 10% FBS (complete medium) at 37°C in 5% CO2. LNCaP-AI cells were generated by us previously (36) and maintained in RPMI-1640 medium supplemented with 10% charcoal/dextran-treated FBS (stripped medium). Lung cancer adenocarcinoma A549 cells were obtained from ATCC and maintained in MEM medium supplemented with 5% FBS (complete medium) at 37°C in 5% CO2.

3D cell culture

A 96-well plate was coated with 100 µl/well of medium containing 0.9% agarose. SP CSCs and non-SP control cells in medium with 10% FBS on ice were mixed with a same volume of cold (4°C) medium containing 10% FBS and 10% growth factor-reduced Matrigel matrix (BD Biosciences, Bedford, MA, USA) and seeded into the agarose plate at 100 µl/well. The cells were cultured at 37°C for 14 days.

CSC sorting

The ALDH1-high CSCs from 22Rv1 cells was isolated using the Aldefluor assay kit (StemCell Technologies, Vancouver, Canada) and BD FACSAria II cell sorters. ALDH1 cleaves boron-dipyrromethene-aminoacetaldehyde (BAAA) to release fluorescent dye, which can be blocked by diethylaminobenzaldehyde (DEAB). After gating with DEAB control, the fluorescent cells were sorted out as ALDH1-high CSCs.

SP CSCs were isolated as previously described (37). Briefly, A549 and H1975 cells were resuspended at 1×106/ml in prewarmed DMEM with 2% FCS and 10 mmol/l HEPES buffer. Hoechst 33342 dye was added at a final concentration of 5 µg/ml in the absence or presence of reserpine (50 µmol/l; Sigma) and the cells were incubated at 37°C for 90 min with intermittent shaking. At the end of the incubation, the cells were washed with ice-cold HBSS with 2% FCS and 10 mmol/l HEPES, centrifuged down at 4°C, and resuspended in ice-cold HBSS containing 2% FCS and 10 mmol/l HEPES. Propidium iodide at a final concentration of 2 µg/ml was added to gate viable cells. Gated by control cells treated with reserpine that blocks Hoechst 33342 transporter, SP CSCs were sorted using BD FACSAria II cell sorters. SP CSCs were defined as the missing region in the presence of reserpine.

Tumorigenesis in mice

One thousand of CSCs or control cells in 50 µl PBS were mixed with 50 µl of cold Matrigel (Thermo Fisher Scientific) and inoculated subcutaneously into nude mice. Tumor incidence and sizes were measured using calipers and recorded twice a week. The mice were maintained in a facility approved by the American Association for Accreditation of Laboratory Animal Care (AAALAC) and in accordance with current regulations and standards of the U.S. Department of Agriculture, U.S. Department of Health and Human Services, and NIH. The animal studies were approved by the Institutional Animal Care and Use Committee (IACUC) and executed according to IACUC guidelines.

RT-PCR

RNAs from both CSCs and control cells were isolated using RNeasy Plus Universal Mini kit (Qiagen, Germany). RNA samples were then treated with DNase using DNA-free™ kit (Thermo Fisher Scientific) and subjected to reverse transcription reactions using High Capacity cDNA Reverse Transcription kits (Applied Biosystems, Thermo Fisher Scientific). Real-time RT-PCR was performed using the Fast SYBR® Green Master Mix and 7300 Real-Time PCR system (Applied Biosystems, Thermo Fisher Scientific).

Co-immunoprecipitation assay

Cell extracts from LNCaP-AI cells were prepared. Co-immunoprecipitation was performed in a modified RIPA buffer (PBS, 0.1% NP-40, 0.1% sodium deoxycholate, 150 mM NaCl, 1 mM EDTA, 1 mM dithiothreitol, 1 mM phenylmethylsulfonyl fluoride, and 1X protease inhibitor cocktail). Antibody was first incubated with 1 mg of cell extract at 4°C on Rocker platform for 1–2 h of Protein G plus/protein A agarose beads (50 µl) (Calbiochem, Thermo Fisher Scientific) was then added, and the samples were incubated at 4°C on Rocker platform overnight. The beads were washed with the modified RIPA buffer four times and boiled in SDS loading buffer. The protein samples were subjected to SDS-PAGE and western blot analysis.

Western blot analysis

Western blot analysis was performed as previously described (13). Briefly, aliquots of samples with the same amount of protein, determined using the DC Protein assay kit (Bio-Rad, Hercules, CA, USA), were mixed with loading buffer (final concentrations of 62.5 mM Tris-HCl, pH 6.8, 2.3% SDS, 100 mM dithiothreitol, and 0.005% bromophenol blue), boiled, fractionated in an SDS-PAGE, and transferred onto a 0.45-mm nitrocellulose membrane (Bio-Rad). The membranes were blocked with 2% fat-free milk in PBS, and probed with first antibody in PBS containing 0.01% Tween-20 (PBST) and 1% fat-free milk. The membranes were then washed four times in PBS and incubated with IRDye 800CW secondary antibody (LI-COR Biosciences, Lincoln, NE, USA) in PBST containing 1% fat-free milk for 30 min. After washing four times in PBS, the membranes were visualized using Odyssey imaging system (LI-COR).

Reporter assay

Cells (105/well) were seeded in 12-well tissue culture plates. The next day, Lipofectamine 3000 reagent was used for the transient transfection assay according to the protocol provided by Invitrogen/Life Technologies, Inc. The cells were then treated for 24 h. Subsequently, the cell extracts were prepared and luciferase activity was assessed in a Berthold Detection system (Titertek-Berthold, Pforzheim, Germany) using a Luciferase assay kit (Promega, Madison, WI, USA) according to the manufacturer's instructions. For each assay, cell extract (20 µl) was used and the reaction was started by injection of 50 µl of luciferase substrate. Each reaction was measured for 10 sec in the luminometer. Luciferase activity was defined as light units/mg protein.

Results

sPLA2-IIa stimulates HER/ERBB-elicited signaling in cancer cells

We reported previously that treatment of NSCLC A549 and H1975 cells with recombinant human sPLA2-IIa induces phosphorylation of HER2 and HER3 within 2 h in a dose-dependent manner (32). The reporter assay revealed that sPLA2-IIa enhances the promoter activities of NF-κB and sPLA2-IIa genes in a dose-dependent manner in lung cancer cells. For validation, we determined the effects of sPLA2-IIa in prostate cancer cells. As shown in Fig. 1A and B, recombinant human sPLA2-IIa induces phosphorylation of HER2 within 2 h in a dose- and time-dependent manner in CRPC LNCaP-AI cells (36). sPLA2-IIa also enhances the promoter activities of NF-κB (Fig. 1C) and sPLA2-IIa (Fig. 1D) genes in a dose-dependent manner in the cells. The stimulatory effects of sPLA2-IIa on sPLA2-IIa promoter activity were abolished by antibodies against sPLA2-IIa (Fig. 1E). These data implicated that sPLA2-IIa enhances HER/ERBB-elicited signaling in prostate cancer cells.

sPLA2-IIa functions as a ligand for EGFR family receptors

A structural analysis we performed previously suggests that the sPLA2-IIa β hairpin shares significant similarity with the EGF hairpin and could be displaced to provide additional contacts with EGFR, and protein docking computation shows that sPLA2-IIa directly interacts with the extracellular domain (ECD) of EGFR in such a way as to stabilize EGFR in its active conformation (32). For validation of the interactions of sPLA2-IIa with EGFR family receptors, co-immunoprecipitation (co-IP) was performed using cell extracts from LNCaP-AI cells. As shown in Fig. 2A, EGFR, HER2, and HER3 were detected in the co-IP complex pulled down by an antibody to sPLA2-IIa. Reciprocally, sPLA2-IIa was detected in the co-IP complexes pulled down by an antibody to EGFR (Fig. 2B). These data reveal that sPLA2-IIa activates EGFR family receptors and complexes with EGFR, HER2, and HER3, strongly suggesting that sPLA2-IIa is a novel ligand for EGFR and HER3 (32).

Overexpression of sPLA2-IIa in CSCs

SP CSCs express high levels of ABCG2 and/or ABCB1 multidrug efflux pump proteins and possess stem cell-like properties (37). We isolated SP CSCs from NSCLC cells using flow cytometry and Hoechst 33342 dye efflux assay (37) and found ~1% of SP CSCs in A549 cells and 0.5% of SP CSCs in H1975 cells (Fig. 3A). ALDH1 is a universal functional marker for CSCs (3844) and is involved in cellular responses to oxidative stress (42) and drug resistance (41). Overexpression of ALDH1 correlates with poor prognosis in prostate cancer (38). ALDH1-based approach has been used to successfully isolate CSCs from many cell lines of diverse cancer types (45,46), including CRPC 22Rv1 and PC-3 cells (39,44). Consistent with data reported in the literature (44), we found ~2.1% of ALDH1-high CSCs in 22Rv1 cells (Fig. 3B).

The real-time RT-PCR was performed to determine expression of CSC marker genes in the CSCs. As shown in (Tables I and II), the CSC markers, including ABCG2, ALDH1, CD133, CD44, CD56, Focx2, MDR1, Nanog, NKX3.1, NSE, Slug, Snail, Sox2, and Twist (2,3,47), are heterogeneously overexpressed in SP CSCs relative to non-SP control cells or in ALDH1-high CSCs relative to ALDH1-low control cells. In addition, HER2, HER3, or AR gene is also moderately increased in these CSCs. Interestingly, sPLA2-IIa levels in SP and ALDH1-high CSCs are elevated 41-fold (H1975), 4.3-fold (A549), and 6-fold (22Rv1), respectively, suggesting that sPLA2-IIa is a novel CSC marker and supports CSC properties (Table II).

Table I

Primers used for RT-PCR.

Table I

Primers used for RT-PCR.

ABCG2Upper primer 5′-AAACCTGGTCTCAACGCCATCC-3′
Lower primer 5′-TGCCCATCACAACATCATCTTG-3′
ALDH1Upper primer 5′-CTTACCTGTCCTACTCACCGATTTG-3′
Lower primer 5′-CCTTGTCAACATCCTCCTTATCTCC-3′
ARUpper primer 5′-GTCTTCGGAAATGTTATGAAGCA-3′
Lower primer 5′-ACGATCGAGTTCCTTGATGTAG-3′
CD133Upper primer 5′-TGAGACCCAAGACTCCCATAAAGC-3′
Lower primer 5′-GGACACAGCATAGAATAATCCCTGC-3′
CD44Upper primer 5′-CGTGGAGAAAAATGGTCGCTAC-3′
Lower primer 5′-TACTGGGAGGTGTTGGATGTGAGG-3′
CD56Upper primer 5′-CGGCATTTACAAGTGTGTGG-3′
Lower primer 5′-GACATCTCGGCCTTTGTGTT-3′
Focx2Upper primer 5′-AGAATTACTACCGGGCTGCG-3′
Lower primer 5′-TGAGCGCGATGTAGCTGTAG-3′
HER2Upper primer 5′-AATGGAGACCCGCTGAACAATAC-3′
Lower primer 5′-CACAAAATCGTGTCCTGGTAGCAG-3′
HER3Upper primer 5′-TACGAGAGGTGTGAGGTGGTGATG-3′
Lower primer 5′-GGAGGTTGGGCAATGGTAGAGTAG-3′
MDR1Upper primer 5′-TGGGAAGAGCACAACAGTCCAG-3
Lower primer 5′-CGTGGTGGCAAACAATACAGGTTC-3′
NanogUpper primer 5′-CCAGTCCCAAAGGCAAACAAC-3′
Lower primer 5′-TGGAGGCTGAGGTATTTCTGTCTC-3′
NKX3.1Upper primer 5′-AAAGGCACTTGGGGTCTTATCTG-3′
Lower primer 5′-CTTCTGATGGCTGAACTTCCTCTC-3′
NSEUpper primer 5′-GTCCCACGTGTCTTCCACTT-3′
Lower primer 5′-CCCAAGTCAGGCCAGTTTTA-3′
SlugUpper primer 5′-GAGCATTTGCAGACAGGTCA-3′
Lower primer 5′-GCTTCGGAGTGAAGAAATGC-3′
SnailUpper primer 5′-ACCCCACATCCTTCTCACTG-3′
Lower primer 5′-TACAAAAACCCACGCAGACA-3′
SOX2Upper primer 5′-GCACCGCTACGACGTGA-3′
Lower primer 5′-TGCGAGTAGGACATGCTGTAGG-3′
sPLA2-IIaUpper primer 5′-TTGACGACAGGAAAGGAAGCCG-3′
Lower primer 5′-TCTGCTCCCCGAGTTGCTAAAC-3′
TwistUpper primer 5′-GGAGTCCGCAGTCTTACGAG-3′
Lower primer 5′-CCAGCTTGAGGGTCTGAATC-3′
VimentinUpper primer 5′-AATCCAAGTTTGCTGACCTCTCTG-3′
Lower primer 5′-CTCTTCCATTTCACGCATCTGG-3′

Table II

CSCs express a high level of sPLA2-IIa.

Table II

CSCs express a high level of sPLA2-IIa.

H1975 cellsNone-SP controlSP CSCsFold increase
ABCG22.26E-043.05E-0313.53
ALDH12.01E-064.00E-0519.92
CD1331.01E-064.53E-0245033.82
Focx24.95E-052.46E-044.97
HER31.20E-045.07E-044.21
MDR13.43E-077.19–05209.32
Nanog4.59E-057.80E-051.7
NKX3.14.31E-032.83E-026.57
Slug1.26E-026.91E-025.5
Snail5.60E-021.83E-013.27
sPLA2-IIa7.66E-083.15E-0641.1
Twist7.66E-083.33E-05435.24
A549 cells
 SOX25.05E-042.22E-034.39
sPLA2-IIa1.83E-047.91E-044.31
 Twist3.64E-041.15E-033.16

22RV1 cellsALDH1-low controlALDH1-high CSCsFold increase

ALDH12.80E-041.81E-0264.63
AR3.72E-059.44E-052.54
CD1331.26E-073.56E-05283.24
CD443.40E-033.11E-029.16
CD565.99E-062.33E-053.89
Focx25.17E-051.79E-043.47
HER21.01E-021.87E-021.85
HER31.08E-022.95E-021.56
Nanog4.77E-047.85E-041.6
NSE1.24E-028.63E-026.44
Slug3.31E-061.09E-052.25
Snail2.19E-042.23E-0310.2
SOX21.93E-056.20E-053.21
sPLA2-IIa3.93E-062.34E-055.95

[i] The mRNA expression levels are determined relative to that of β-actin.

Characterization of CSCs in 3D cell culture and in mice

We performed 3D cell culture to validate CSCs properties and found that SP CSCs from A549 cells generate more and much larger spheroids than those by non-SP control cells (Fig. 4). Similar observation was also made in ALDH1-high CSCs from 22RV1 cells and SP CSCs from H1975 cells (data not shown).

To further characterize the tumor initiating properties of CSCs in mice, a subcutaneous inoculation of 1,000 cells/mouse of SP CSCs, but not non-SP control cells, into nude mice produced visible tumors on day 20. The tumors reached ~100 mm3 on day 30. A subcutaneous inoculation of 1,000 cells/mouse of ALDH1-high CSCs, but not ALDH1-low control cells, into nude mice produced tumors on day 25. The tumors reached ~100 mm3 on day 35.

Overexpression of CSC markers in CRPC LNCaP-AI cells and taxane-resistant 22Rv1-R cells

CRPC LNCaP-AI cells were established by culturing androgen-dependent LNCaP cells in the stripped medium as we described previously (36). CRPC 22Rv1 cells express constitutively active androgen receptor (48,49). We cultured 22Rv1 cells in the presence of increasing concentrations of paclitaxel to derive 22Rv1-R cells. As shown in Table III, 22Rv1-R cells are resistant to all taxenes.

Table III

Establishment of taxane-resistant 22Rv1-R cell line.

Table III

Establishment of taxane-resistant 22Rv1-R cell line.

GenepTxdTxcTx
22Rv1211
22Rv1-R32730515
R index16430515

[i] IC50 (nM) of the cells to pTx, dTx, and cTx in a 4-day MTT assay. Resistance (R) index = IC50 of 22Rv1-R/IC50 of 22Rv1.

We found that CSC markers, including ABCG2, ALDH1, CD44, CD56, Focx2, MDR1, Nanog, NSE, Snail, Sox2, and Vimentin, were heterogeneously overexpressed in LNCaP-AI cells relative to parental LNCaP cells (Table IV) and in taxane-resistant 22Rv1-R cells relative to parental 22Rv1 cells (Table V). Strikingly, MDR1 level is increased by 1,380-fold in 22Rv1-R cells and 66-fold in LNCaP-AI cells. The expression of sPLA2-IIa in LNCaP-AI cells relative to LNCaP cells is increased 60-fold (Table IV), confirming the finding reported in our previous study (13). sPLA2-IIa is not overexpressed in 22Rv1-R cells relative to 22Rv1 cells, but is overexpressed in CSCs from 22Rv1 cells (Table II), indicating the heterogeneity of gene expression in cancer cells. These findings suggest that CSC population is elevated in LNCaP-AI and 22Rv1-R cells, supporting the notion that the current anticancer therapies favor the selection and expansion of CSCs.

Table IV

Overexpression of CSC markers in CRPC LNCaP-AI cells.

Table IV

Overexpression of CSC markers in CRPC LNCaP-AI cells.

GeneLNCaP cellsLNCaP-AI cellsFold increase
ALDH15.60E-052.77E-044.94
CD448.96E-051.19E-042.44
CD561.20E-068.10E-0567.74
MDR14.49E-062.97E-0466.2
Nanog2.95E-045.54E-041.88
NSE2.64E-038.11E-033.07
SOX22.40E-071.26E-068
sPLA2-IIa9.19E-035.50E-0159.9
Vimentin2.35E-038.55E-0236.4

[i] The mRNA expression levels are determined relative to that of β-actin.

Table V

Overexpression of CSC markers in taxane-resistant CRPC 22RV1-R cells.

Table V

Overexpression of CSC markers in taxane-resistant CRPC 22RV1-R cells.

Gene22RV1 cells22RV1-R cellsFold increase
ABCG21.98E-031.17E-025.89
ALDH11.35E-035.63E-034.18
CD442.30E-036.56E-032.85
Focx26.70E-063.29E-054.91
MDR11.56E-042.15E-011,380.42
Nanog3.02E-041.03E-033.4
NSE3.19E-029.42E-023
Snail5.56E-043.80E-039.9
SOX21.04E-042.52E-042.57

[i] The mRNA expression levels are determined relative to that of β-actin.

Discussion

We demonstrated that cancer cells overexpress and secrete sPLA2-IIa into the interstitial fluid, i.e., tumor microenvironment and blood (13,29,30). Plasma sPLA2-IIa continuously increased with prostate cancer progression and reached as high as 18 ng/ml at the late stage in metastatic prostate cancer (29). High levels of plasma sPLA2-IIa, based on the optimum cutoff value of 2.0 ng/ml, significantly predicted advanced stage and high Gleason score in prostate cancer (13,29). We further showed that sPLA2-IIa is overexpressed in almost all lung cancers and is significantly elevated in the blood of lung cancer patients (30). High levels of plasma sPLA2-IIa, at the optimum cutoff value of 2.4 ng/ml, are significantly associated with advanced lung cancer stage and decreased overall cancer survival (30). The most recent report by others supports our finding in that high levels of plasma sPLA2-IIa are associated with poor prognosis of cancers (31).

The ultimate cause of cancer treatment failure is that tumor cells evolve and develop multiple mechanisms to escape the cytotoxic effects of anticancer drugs, including enhancement of cell survival pathway, impaired apoptotic machinery, increased DNA repair mechanisms, and multidrug resistance phenotype by overexpression of drug-efflux pump proteins P-glycoprotein (MDR1) and ABCG-2 (5052). These mechanisms may also drive CSC properties and tumor progression. ATP-binding cassette (ABC) transporters, such as P-glycoprotein (MDR1), multidrug resistant associated protein (MRP1), and ATP-binding cassette membrane transporter G2 [ABCG2, also known as breast cancer resistance protein 1 (BCRP1)], are membrane transporters that can pump cytotoxic chemotherapeutic drugs out of the cell. CSCs express high levels of ABC transporters leading to low intracellular drug concentrations and conferring multidrug resistance to many anticancer drugs. The efflux capacity of SP CSCs, determined by high ABCG2 activity, is associated with tumor growth, progression, and metastasis. The aldehyde dehydrogenase (ALDH) family members are cytosolic isoenzymes responsible for oxidizing intracellular aldehydes. ALDH1-high CSCs are highly resistant to chemotherapeutic agents commonly used as first-line therapy in the clinical setting, such as cisplatin, gemcitabine, doxorubicin, vinorelbine and docetaxel, whereas ALDH1-low cells are sensitive to the cytotoxic activity of these drugs. In the present study, we found that both SP CSCs from NSCLC cells and ALDH1-high CSCs from CRPC cells overexpress a number of CSC markers, supporting the notion that multiple mechanisms contribute to CSC phenotype (Table II). More importantly, we found that both SP CSCs and ALDH1-high CSCs overexpress sPLA2-IIa. It was reported that ALDH1-high CSCs from lung cancer cells also overexpress sPLA2-IIa (53). These findings strongly suggest that sPLA2-IIa is a marker for CSCs and may support CSC properties.

Gene amplification, overexpression, and mutations in EGFR family receptors have been well described in various cancers, including breast, head and neck, prostate, and NSCLC, and contribute to resistance to therapy and cancer progression (5459). EGF is a preferable ligand for EGFR/EGFR homodimer or EGFR/HER2 heterodimer, while heregulin-a is a preferable ligand for HER2/HER3 heterodimer (34,60,61). HER2 has no ligand and HER3 has no tyrosine kinase activity, and they function by forming heterodimer with other HER receptor. Because HER3 has six tyrosine containing binding sites for p85, the regulatory subunit of PI3K, HER2/HER3 complex is much more effective than EGFR/HER2 in activation of the PI3K/Akt pathway. HER3, which signaling function cannot be inhibited by tyrosine kinase inhibitor (TKI), provides a focal point in resistance to TKI therapy (62). Increasing body of evidence highlight the role of HER3 in lung cancer, which has not been successfully addressed in the targeted therapy to date. By coupling to numerous signaling pathways, such as the RAS-ERK and PI3K-Akt pathways, and multiple feedback regulatory loops, HER/ERBB-elicited signaling propels the clonal expansion of CSCs (3335). One such positive feedback loop is stimulation of sPLA2-IIa overexpression via HER/ERBB-PI3K-Akt-NF-κB signaling (13,29,30). We uncovered that elevated HER/ERBB-PI3K-Akt-NF-κB signaling induces sPLA2-IIa overexpression and secretion in both lung and prostate cancer cells, and in turn, sPLA2-IIa activates EGFR family receptors and HER/ERBB-elicited signaling and stimulates sPLA2-IIa overexpression in a positive feedback manner (13,29,30).

We further investigated the molecular action of sPLA2-IIa. Given the potential ligand activity of sPLA2-IIa, we hypothesized that sPLA2-IIa functions as a ligand for EGFR family receptors, leading to an enhanced HER/ERBB-elicited signaling. Indeed, we found that sPLA2-IIa enhances HER/ERBB-PI3K-Akt-NF-κB signaling in both prostate cancer cells (Figs. 1 and 2) and lung cancer cells (32). Our protein docking analysis and co-immunoprecipitation experiments revealed that sPLA2-IIa interacts with EGFR family receptors (Fig. 2) (32). sPLA2-IIa directly or indirectly interacts with EGFR, HER2, and HER3, suggesting that it may be a ligand for both EGFR and HER3. Given that both SP and ALDH1-high CSCs overexpress sPLA2-IIa (Table II), sPLA2-IIa in the tumor microenvironment may function as a ligand for EGFR family receptors, stimulates HER/ERBB- elicited signaling, and promotes the clonal expansion of CSCs and cancer progression (3335).

sPLA2-IIa stimulates growth of prostate cancer cells (13,27,63), colon cancer cells (64), and brain tumor cells (65,66), which may be via the EGFR-, MAPK-, PI3K/Akt-, NF-κB-mediated cell growth and survival signaling pathways (6770). sPLA2-IIa abrogates TNF-α-induced apoptosis and compromises immune surveillance function (71). In the transgenic adenocarcinoma of the mouse prostate (TRAMP) model, sPLA2-IIa contributes to aggressive phenotypes, androgen-independent growth, and metastasis (72). sPLA2-IIa binds to integrins and induces proliferation of monocytic cells in an integrin-dependent manner (16). We showed that sPLA2-IIa is overexpressed in CSCs, which may support CSC properties. On the other hand, knocking down expression of sPLA2-IIa reduces lung cancer growth (73). Our previous and current studies revealed the underlying mechanisms of sPLA2-IIa action, in which sPLA2-IIa functions as a ligand for EGFR family receptors, leading to sPLA2-IIa overexpression via HER/ERBB-PI3K-Akt-NF-κB signaling in a positive feedback manner. Aberrant high levels of sPLA2-IIa in the tumor microenvironment support CSC properties and contribute to cancer progression and metastasis. sPLA2-IIa is a novel therapeutic target against cancer.

It has been shown that the treatment with taxol and cisplatin may not affect growth, but can even stimulate growth in CSCs (74,75). Similarly, castration has been shown to induce epithelial-mesenchymal transition, promote growth of CSCs in prostate cancer, and lead to castration-resistance and metastasis (10). Consistent with these observations, we found that CRPC LNCaP-AI cells, selected in the stripped medium (36), and multidrug resistant 22Rv1-R cells, derived from CRPC 22Rv1 cells selected in the medium containing paclitaxel, overexpress several CSC markers and drug efflux pump proteins (Tables IV and V). Therefore, LNCaP-AI and 22Rv1-R cells provide valuable tools for studying CSCs and determine the roles of sPLA2-IIa in tumor progression.

Acknowledgments

This study was supported in part by the Millennium Scholar funds of University of Cincinnati Cancer Center and a pilot grant from the Department of Internal Medicine, University of Cincinnati College of Medicine.

References

1 

O'Flaherty JD, Barr M, Fennell D, Richard D, Reynolds J, O'Leary J and O'Byrne K: The cancer stem-cell hypothesis: Its emerging role in lung cancer biology and its relevance for future therapy. J Thorac Oncol. 7:1880–1890. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Leon G, MacDonagh L, Finn SP, Cuffe S and Barr MP: Cancer stem cells in drug resistant lung cancer: Targeting cell surface markers and signaling pathways. Pharmacol Ther. 158:71–90. 2016. View Article : Google Scholar

3 

MacDonagh L, Gray SG, Breen E, Cuffe S, Finn SP, O'Byrne KJ and Barr MP: Lung cancer stem cells: The root of resistance. Cancer Lett. 372:147–156. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Liu T, Xu F, Du X, Lai D, Liu T, Zhao Y, Huang Q, Jiang L, Huang W, Cheng W, et al: Establishment and characterization of multidrug resistant, prostate carcinoma-initiating stem-like cells from human prostate cancer cell lines 22RV1. Mol Cell Biochem. 340:265–273. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Fang DD, Cao J, Jani JP, Tsaparikos K, Blasina A, Kornmann J, Lira ME, Wang J, Jirout Z, Bingham J, et al: Combined gemcitabine and CHK1 inhibitor treatment induces apoptosis resistance in cancer stem cell-like cells enriched with tumor spheroids from a non-small cell lung cancer cell line. Front Med. 7:462–476. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Vlashi E and Pajonk F: The metabolic state of cancer stem cells - a valid target for cancer therapy? Free Radic Biol Med. 79:264–268. 2015. View Article : Google Scholar

7 

Pfeiffer MJ and Schalken JA: Stem cell characteristics in prostate cancer cell lines. Eur Urol. 57:246–254. 2010. View Article : Google Scholar

8 

Lagadec C, Vlashi E, Della Donna L, Dekmezian C and Pajonk F: Radiation-induced reprogramming of breast cancer cells. Stem Cells. 30:833–844. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Ghisolfi L, Keates AC, Hu X, Lee DK and Li CJ: Ionizing radiation induces stemness in cancer cells. PLoS One. 7:e436282012. View Article : Google Scholar : PubMed/NCBI

10 

Li P, Yang R and Gao WQ: Contributions of epithelial-mesen-chymal transition and cancer stem cells to the development of castration resistance of prostate cancer. Mol Cancer. 13:552014. View Article : Google Scholar

11 

Brown MD, Gilmore PE, Hart CA, Samuel JD, Ramani VA, George NJ and Clarke NW: Characterization of benign and malignant prostate epithelial Hoechst 33342 side populations. Prostate. 67:1384–1396. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Antonio V, Brouillet A, Janvier B, Monne C, Bereziat G, Andreani M and Raymondjean M: Transcriptional regulation of the rat type IIA phospholipase A2 gene by cAMP and interleukin-1beta in vascular smooth muscle cells: Interplay of the CCAAT/enhancer binding protein (C/EBP), nuclear factor-kappaB and Ets transcription factors. Biochem J. 368:415–424. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Dong Z, Liu Y, Scott KF, Levin L, Gaitonde K, Bracken RB, Burke B, Zhai QJ, Wang J, Oleksowicz L, et al: Secretory phospholipase A2-IIa is involved in prostate cancer progression and may potentially serve as a biomarker for prostate cancer. Carcinogenesis. 31:1948–1955. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Cummings BS: Phospholipase A2 as targets for anti-cancer drugs. Biochem Pharmacol. 74:949–959. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Triggiani M, Granata F, Giannattasio G and Marone G: Secretory phospholipases A2 in inflammatory and allergic diseases: Not just enzymes. J Allergy Clin Immunol. 116:1000–1006. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Saegusa J, Akakura N, Wu CY, Hoogland C, Ma Z, Lam KS, Liu FT, Takada YK and Takada Y: Pro-inflammatory secretory phospholipase A2 type IIA binds to integrins alphavbeta3 and alpha4beta1 and induces proliferation of monocytic cells in an integrin-dependent manner. J Biol Chem. 283:26107–26115. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Triggiani M, Granata F, Balestrieri B, Petraroli A, Scalia G, Del Vecchio L and Marone G: Secretory phospholipases A2 activate selective functions in human eosinophils. J Immunol. 170:3279–3288. 2003. View Article : Google Scholar : PubMed/NCBI

18 

Tada K, Murakami M, Kambe T and Kudo I: Induction of cyclooxygenase-2 by secretory phospholipases A2 in nerve growth factor-stimulated rat serosal mast cells is facilitated by interaction with fibroblasts and mediated by a mechanism independent of their enzymatic functions. J Immunol. 161:5008–5015. 1998.PubMed/NCBI

19 

Cupillard L, Mulherkar R, Gomez N, Kadam S, Valentin E, Lazdunski M and Lambeau G: Both group IB and group IIA secreted phospholipases A2 are natural ligands of the mouse 180-kDa M-type receptor. J Biol Chem. 274:7043–7051. 1999. View Article : Google Scholar : PubMed/NCBI

20 

Nicolas JP, Lambeau G and Lazdunski M: Identification of the binding domain for secretory phospholipases A2 on their M-type 180-kDa membrane receptor. J Biol Chem. 270:28869–28873. 1995. View Article : Google Scholar : PubMed/NCBI

21 

Scott KF, Sajinovic M, Hein J, Nixdorf S, Galettis P, Liauw W, de Souza P, Dong Q, Graham GG and Russell PJ: Emerging roles for phospholipase A2 enzymes in cancer. Biochimie. 92:601–610. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Meyer AM, Dwyer-Nield LD, Hurteau GJ, Keith RL, O'Leary E, You M, Bonventre JV, Nemenoff RA and Malkinson AM: Decreased lung tumorigenesis in mice genetically deficient in cytosolic phospholipase A2. Carcinogenesis. 25:1517–1524. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Kallajoki M, Alanen KA, Nevalainen M and Nevalainen TJ: Group II phospholipase A2 in human male reproductive organs and genital tumors. Prostate. 35:263–272. 1998. View Article : Google Scholar : PubMed/NCBI

24 

Jiang J, Neubauer BL, Graff JR, Chedid M, Thomas JE, Roehm NW, Zhang S, Eckert GJ, Koch MO, Eble JN, et al: Expression of group IIA secretory phospholipase A2 is elevated in prostatic intraepithelial neoplasia and adenocarcinoma. Am J Pathol. 160:667–671. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Graff JR, Konicek BW, Deddens JA, Chedid M, Hurst BM, Colligan B, Neubauer BL, Carter HW and Carter JH: Expression of group IIa secretory phospholipase A2 increases with prostate tumor grade. Clin Cancer Res. 7:3857–3861. 2001.PubMed/NCBI

26 

Dong Z, Liu Y, Levin L, Oleksowicz L, Wang J and Lu S: Vav3 oncogene is involved in regulation of secretory phospholipase A2-IIa expression in prostate cancer. Oncol Rep. 25:1511–1516. 2011.PubMed/NCBI

27 

Sved P, Scott KF, McLeod D, King NJ, Singh J, Tsatralis T, Nikolov B, Boulas J, Nallan L, Gelb MH, et al: Oncogenic action of secreted phospholipase A2 in prostate cancer. Cancer Res. 64:6934–6940. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Mirtti T, Laine VJ, Hiekkanen H, Hurme S, Rowe O, Nevalainen TJ, Kallajoki M and Alanen K: Group IIA phospholipase A as a prognostic marker in prostate cancer: Relevance to clinicopathological variables and disease-specific mortality. APMIS. 117:151–161. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Oleksowicz L, Liu Y, Bracken RB, Gaitonde K, Burke B, Succop P, Levin L, Dong Z and Lu S: Secretory phospholipase A2-IIa is a target gene of the HER/HER2-elicited pathway and a potential plasma biomarker for poor prognosis of prostate cancer. Prostate. 72:1140–1149. 2012. View Article : Google Scholar :

30 

Kupert E, Anderson M, Liu Y, Succop P, Levin L, Wang J, Wikenheiser-brokamp K, Chen P, Pinney SM, Macdonald T, et al: Plasma secretory phospholipase A2-IIa as a potential biomarker for lung cancer in patients with solitary pulmonary nodules. BMC Cancer. 11:5132011. View Article : Google Scholar : PubMed/NCBI

31 

Menschikowski M, Hagelgans A, Schuler U, Froeschke S, Rosner A and Siegert G: Plasma levels of phospholipase A2-IIA in patients with different types of malignancies: Prognosis and association with inflammatory and coagulation biomarkers. Pathol Oncol Res. 19:839–846. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Dong Z, Meller J, Succop P, Wang J, Wikenheiser-Brokamp K, Starnes S and Lu S: Secretory phospholipase A2-IIa upregulates HER/HER2-elicited signaling in lung cancer cells. Int J Oncol. 45:978–984. 2014.PubMed/NCBI

33 

Mimeault M, Hauke R, Mehta PP and Batra SK: Recent advances in cancer stem/progenitor cell research: Therapeutic implications for overcoming resistance to the most aggressive cancers. J Cell Mol Med. 11:981–1011. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Schneider MR and Yarden Y: The EGFR-HER2 module: A stem cell approach to understanding a prime target and driver of solid tumors. Oncogene. 35:2949–2960. 2016. View Article : Google Scholar :

35 

Singh S, Trevino J, Bora-Singhal N, Coppola D, Haura E, Altiok S and Chellappan SP: EGFR/Src/Akt signaling modulates Sox2 expression and self-renewal of stem-like side-population cells in non-small cell lung cancer. Mol Cancer. 11:732012. View Article : Google Scholar : PubMed/NCBI

36 

Lu S, Tsai SY and Tsai MJ: Molecular mechanisms of androgen- independent growth of human prostate cancer LNCaP-AI cells. Endocrinology. 140:5054–5059. 1999. View Article : Google Scholar : PubMed/NCBI

37 

Ho MM, Ng AV, Lam S and Hung JY: Side population in human lung cancer cell lines and tumors is enriched with stem-like cancer cells. Cancer Res. 67:4827–4833. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Li T, Su Y, Mei Y, Leng Q, Leng B, Liu Z, Stass SA and Jiang F: ALDH1A1 is a marker for malignant prostate stem cells and predictor of prostate cancer patients' outcome. Lab Invest. 90:234–244. 2010. View Article : Google Scholar

39 

Doherty RE, Haywood-Small SL, Sisley K and Cross NA: Aldehyde dehydrogenase activity selects for the holoclone phenotype in prostate cancer cells. Biochem Biophys Res Commun. 414:801–807. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Marcato P, Dean CA, Giacomantonio CA and Lee PW: Aldehyde dehydrogenase: Its role as a cancer stem cell marker comes down to the specific isoform. Cell Cycle. 10:1378–1384. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Januchowski R, Wojtowicz K and Zabel M: The role of aldehyde dehydrogenase (ALDH) in cancer drug resistance. Biomed Pharmacother. 67:669–680. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Singh S, Brocker C, Koppaka V, Chen Y, Jackson BC, Matsumoto A, Thompson DC and Vasiliou V: Aldehyde dehydrogenases in cellular responses to oxidative/electrophilic stress. Free Radic Biol Med. 56:89–101. 2013. View Article : Google Scholar :

43 

Wu A, Luo W, Zhang Q, Yang Z, Zhang G, Li S and Yao K: Aldehyde dehydrogenase 1, a functional marker for identifying cancer stem cells in human nasopharyngeal carcinoma. Cancer Lett. 330:181–189. 2013. View Article : Google Scholar

44 

Nishida S, Hirohashi Y, Torigoe T, Kitamura H, Takahashi A, Masumori N, Tsukamoto T and Sato N: Gene expression profiles of prostate cancer stem cells isolated by aldehyde dehydrogenase activity assay. J Urol. 188:294–299. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Nishida S, Hirohashi Y, Torigoe T, Inoue R, Kitamura H, Tanaka T, Takahashi A, Asanuma H, Masumori N, Tsukamoto T, et al: Prostate cancer stem-like cells/cancer-initiating cells have an autocrine system of hepatocyte growth factor. Cancer Sci. 104:431–436. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Jiang F, Qiu Q, Khanna A, Todd NW, Deepak J, Xing L, Wang H, Liu Z, Su Y, Stass SA, et al: Aldehyde dehydrogenase 1 is a tumor stem cell-associated marker in lung cancer. Mol Cancer Res. 7:330–338. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Mimeault M and Batra SK: Recent progress on tissue-resident adult stem cell biology and their therapeutic implications. Stem Cell Rev. 4:27–49. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Hu R, Dunn TA, Wei S, Isharwal S, Veltri RW, Humphreys E, Han M, Partin AW, Vessella RL, Isaacs WB, et al: Ligand-independent androgen receptor variants derived from splicing of cryptic exons signify hormone-refractory prostate cancer. Cancer Res. 69:16–22. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Dehm SM, Schmidt LJ, Heemers HV, Vessella RL and Tindall DJ: Splicing of a novel androgen receptor exon generates a constitutively active androgen receptor that mediates prostate cancer therapy resistance. Cancer Res. 68:5469–5477. 2008. View Article : Google Scholar : PubMed/NCBI

50 

Munoz M, Henderson M, Haber M and Norris M: Role of the MRP1/ABCC1 multidrug transporter protein in cancer. IUBMB Life. 59:752–757. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Modok S, Mellor HR and Callaghan R: Modulation of multidrug resistance efflux pump activity to overcome chemoresistance in cancer. Curr Opin Pharmacol. 6:350–354. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Signore M, Ricci-Vitiani L and De Maria R: Targeting apoptosis pathways in cancer stem cells. Cancer Lett. 332:374–382. 2013. View Article : Google Scholar

53 

Bennett DT, Deng XS, Yu JA, Bell MT, Mauchley DC, Meng X, Reece TB, Fullerton DA and Weyant MJ: Cancer stem cell phenotype is supported by secretory phospholipase A2 in human lung cancer cells. Ann Thorac Surg. 98:439–445; discussion 445–436. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Di Lorenzo G, Tortora G, D'Armiento FP, De Rosa G, Staibano S, Autorino R, D'Armiento M, De Laurentiis M, De Placido S, Catalano G, et al: Expression of epidermal growth factor receptor correlates with disease relapse and progression to androgen-independence in human prostate cancer. Clin Cancer Res. 8:3438–3444. 2002.PubMed/NCBI

55 

Shi Y, Brands FH, Chatterjee S, Feng AC, Groshen S, Schewe J, Lieskovsky G and Cote RJ: Her-2/neu expression in prostate cancer: High level of expression associated with exposure to hormone therapy and androgen independent disease. J Urol. 166:1514–1519. 2001. View Article : Google Scholar : PubMed/NCBI

56 

Osman I, Scher HI, Drobnjak M, Verbel D, Morris M, Agus D, Ross JS and Cordon-Cardo C: HER-2/neu (p185neu) protein expression in the natural or treated history of prostate cancer. Clin Cancer Res. 7:2643–2647. 2001.PubMed/NCBI

57 

Signoretti S, Montironi R, Manola J, Altimari A, Tam C, Bubley G, Balk S, Thomas G, Kaplan I, Hlatky L, et al: Her-2-neu expression and progression toward androgen independence in human prostate cancer. J Natl Cancer Inst. 92:1918–1925. 2000. View Article : Google Scholar : PubMed/NCBI

58 

Yeh S, Lin HK, Kang HY, Thin TH, Lin MF and Chang C: From HER2/Neu signal cascade to androgen receptor and its coacti-vators: A novel pathway by induction of androgen target genes through MAP kinase in prostate cancer cells. Proc Natl Acad Sci USA. 96:5458–5463. 1999. View Article : Google Scholar

59 

Craft N, Shostak Y, Carey M and Sawyers CL: A mechanism for hormone-independent prostate cancer through modulation of androgen receptor signaling by the HER-2/neu tyrosine kinase. Nat Med. 5:280–285. 1999. View Article : Google Scholar : PubMed/NCBI

60 

Schulze WX, Deng L and Mann M: Phosphotyrosine interactome of the ErbB-receptor kinase family. Mol Syst Biol. 1:2005 00082005. View Article : Google Scholar

61 

Hsieh AC and Moasser MM: Targeting HER proteins in cancer therapy and the role of the non-target HER3. Br J Cancer. 97:453–457. 2007. View Article : Google Scholar : PubMed/NCBI

62 

Tsao MS, Sakurada A, Cutz JC, Zhu CQ, Kamel-Reid S, Squire J, Lorimer I, Zhang T, Liu N, Daneshmand M, et al: Erlotinib in lung cancer - molecular and clinical predictors of outcome. N Engl J Med. 353:133–144. 2005. View Article : Google Scholar : PubMed/NCBI

63 

Patel MI, Singh J, Niknami M, Kurek C, Yao M, Lu S, Maclean F, King NJ, Gelb MH, Scott KF, et al: Cytosolic phospholipase A2-alpha: A potential therapeutic target for prostate cancer. Clin Cancer Res. 14:8070–8079. 2008. View Article : Google Scholar : PubMed/NCBI

64 

Belinsky GS, Rajan TV, Saria EA, Giardina C and Rosenberg DW: Expression of secretory phospholipase A2 in colon tumor cells potentiates tumor growth. Mol Carcinog. 46:106–116. 2007. View Article : Google Scholar

65 

Hernández M, Martín R, García-Cubillas MD, Maeso- Hernández P and Nieto ML: Secreted PLA2 induces proliferation in astrocytoma through the EGF receptor: Another inflammation-cancer link. Neuro-oncol. 12:1014–1023. 2010. View Article : Google Scholar : PubMed/NCBI

66 

Martín R, Hernández M, Ibeas E, Fuentes L, Salicio V, Arnés M and Nieto ML: Secreted phospholipase A2-IIA modulates key regulators of proliferation on astrocytoma cells. J Neurochem. 111:988–999. 2009. View Article : Google Scholar : PubMed/NCBI

67 

Valentin E and Lambeau G: Increasing molecular diversity of secreted phospholipases A(2) and their receptors and binding proteins. Biochim Biophys Acta. 1488:59–70. 2000. View Article : Google Scholar : PubMed/NCBI

68 

Lambeau G and Lazdunski M: Receptors for a growing family of secreted phospholipases A2. Trends Pharmacol Sci. 20:162–170. 1999. View Article : Google Scholar : PubMed/NCBI

69 

Hernández M, Burillo SL, Crespo MS and Nieto ML: Secretory phospholipase A2 activates the cascade of mitogen-activated protein kinases and cytosolic phospholipase A2 in the human astrocytoma cell line 1321N1. J Biol Chem. 273:606–612. 1998. View Article : Google Scholar : PubMed/NCBI

70 

Park DW, Kim JR, Kim SY, Sonn JK, Bang OS, Kang SS, Kim JH and Baek SH: Akt as a mediator of secretory phospholipase A2 receptor-involved inducible nitric oxide synthase expression. J Immunol. 170:2093–2099. 2003. View Article : Google Scholar : PubMed/NCBI

71 

Ibeas E, Fuentes L, Martín R, Hernández M and Nieto ML: Inflammatory protein sPLA(2)-IIA abrogates TNFalpha-induced apoptosis in human astroglioma cells: Crucial role of ERK. Biochim Biophys Acta. 1793:1837–1847. 2009. View Article : Google Scholar : PubMed/NCBI

72 

Morgenbesser SD, McLaren RP, Richards B, Zhang M, Akmaev VR, Winter SF, Mineva ND, Kaplan-Lefko PJ, Foster BA, Cook BP, et al: Identification of genes potentially involved in the acquisition of androgen-independent and metastatic tumor growth in an autochthonous genetically engineered mouse prostate cancer model. Prostate. 67:83–106. 2007. View Article : Google Scholar

73 

Yu JA, Mauchley D, Li H, Meng X, Nemenoff RA, Fullerton DA and Weyant MJ: Knockdown of secretory phospholipase A2 IIa reduces lung cancer growth in vitro and in vivo. J Thorac Cardiovasc Surg. 144:1185–1191. 2012. View Article : Google Scholar : PubMed/NCBI

74 

Larzabal L, El-Nikhely N, Redrado M, Seeger W, Savai R and Calvo A: Differential effects of drugs targeting cancer stem cell (CSC) and non-CSC populations on lung primary tumors and metastasis. PLoS One. 8:e797982013. View Article : Google Scholar : PubMed/NCBI

75 

Lopez-Ayllon BD, Moncho-Amor V, Abarrategi A, Ibañez de Cáceres I, Castro-Carpeño J, Belda-Iniesta C, Perona R and Sastre L: Cancer stem cells and cisplatin-resistant cells isolated from non-small-lung cancer cell lines constitute related cell populations. Cancer Med. 3:1099–1111. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2017
Volume 50 Issue 6

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lu S and Lu S: Overexpression of secretory phospholipase A2-IIa supports cancer stem cell phenotype via HER/ERBB-elicited signaling in lung and prostate cancer cells. Int J Oncol 50: 2113-2122, 2017
APA
Lu, S., & Lu, S. (2017). Overexpression of secretory phospholipase A2-IIa supports cancer stem cell phenotype via HER/ERBB-elicited signaling in lung and prostate cancer cells. International Journal of Oncology, 50, 2113-2122. https://doi.org/10.3892/ijo.2017.3964
MLA
Lu, S., Dong, Z."Overexpression of secretory phospholipase A2-IIa supports cancer stem cell phenotype via HER/ERBB-elicited signaling in lung and prostate cancer cells". International Journal of Oncology 50.6 (2017): 2113-2122.
Chicago
Lu, S., Dong, Z."Overexpression of secretory phospholipase A2-IIa supports cancer stem cell phenotype via HER/ERBB-elicited signaling in lung and prostate cancer cells". International Journal of Oncology 50, no. 6 (2017): 2113-2122. https://doi.org/10.3892/ijo.2017.3964