Anticancer effect of luteolin is mediated by downregulation of TAM receptor tyrosine kinases, but not interleukin-8, in non-small cell lung cancer cells

  • Authors:
    • Youn Ju Lee
    • Taeho Lim
    • Min Su Han
    • Sun-Hwa Lee
    • Suk‑Hwan Baek
    • Hong-Yan Nan
    • Chuhee Lee
  • View Affiliations

  • Published online on: December 23, 2016     https://doi.org/10.3892/or.2016.5336
  • Pages: 1219-1226
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

TAM receptor tyrosine kinases (RTKs), Tyro3, Axl and MerTK, transduce diverse signals responsible for cell survival, growth, proliferation and anti-apoptosis. In the present study, we demonstrated the effect of luteolin, a flavonoid with antioxidant, anti-inflammatory and anticancer activities, on the expression and activation of TAM RTKs and the association with its cytotoxicity in non-small cell lung cancer (NSCLC) cells. We observed the cytotoxic effect of luteolin in parental A549 and H460 cells as well as in cisplatin-resistant A549/CisR and H460/CisR cells. Exposure of these cells to luteolin also resulted in a dose‑dependent decrease in clonogenic ability. Next, luteolin was found to decrease the protein levels of all three TAM RTKs in the A549 and A549/CisR cells in a dose‑dependent manner. In a similar manner, in H460 and H460/CisR cells, the protein levels of Axl and Tyro3 were decreased following luteolin treatment. In addition, Axl promoter activity was decreased by luteolin, indicating that luteolin suppresses Axl expression at the transcriptional level. We next found that luteolin abrogated Axl phosphorylation in response to growth arrest-specific 6 (Gas6), its ligand, implying the inhibitory effect of luteolin on Gas6-induced Axl activation. Ectopic expression of Axl was observed to attenuate the antiproliferative effect of luteolin, while knockdown of the Axl protein level using a gold nanoparticle-assisted gene delivery system increased its cytotoxicity. In contrast to the inhibitory effect of luteolin on the expression of TAM RTKs, interleukin-8 (IL-8) production was not decreased by luteolin in H460 and H460/CisR cells, while IL-8 production/cell was increased. Collectively, our data suggest that TAM RTKs, but not IL-8, are promising therapeutic targets of luteolin to abrogate cell proliferation and to overcome chemoresistance in NSCLC cells.

Introduction

Platinum-based chemotherapy including cisplatin is the first-line therapy for the treatment of NSCLC (1). However, NSCLC remains the leading cause of cancer-related deaths which are attributed to late diagnosis and development of chemoresistance. Many mechanisms and several molecules involved in chemoresistance have been investigated, but therapeutic outcomes are still unsatisfactory.

The TAM receptor tyrosine kinase (RTK) family, consisting of Tyro3, Axl, MerTK is a subfamily of RTK, which has been reported to be involved in cell growth, proliferation, metastasis and resistance to chemotherapy (2) in various types of cancers. TAM RTKs also play important roles in immune regulation, since they function as phagocytic receptors in normal tissues (3,4). In addition, structural features of TAM RTKs in the extracellular, transmembrane and cytoplasmic kinase domains are very similar (3), which allow them to share various ligands including growth arrest-specific 6 (Gas6), protein S, tubby and tulip. Gas6 has been known to bind to all three TAM RTKs with higher affinity for Axl than the others (5). Upon activation of Axl by Gas6 binding, tyrosine residues of the cytoplasmic tyrosine kinase domain are autophosphorylated (6), which in turn activate several signaling pathways mediating cell growth, survival, proliferation, migration and inhibition of apoptosis (3,7).

Axl, also referred to Ark, Tyro7 or Ufo, was first isolated from chronic myelogenous leukemia cells (8). Since then, Axl overexpression has been reported in a multitude of cancers including acute leukemia (9), breast (10), colon (11), esophageal, lung (12), ovarian (13), prostate (14) and thyroid cancer (15). The overexpression of Axl has been shown to be associated with epithelial-to-mesenchyaml transition (EMT), anticancer drug resistance and angiogenesis (1618), whereas Axl inhibition decreased cancer cell growth and migration and increased sensitivity to anticancer drugs (19). The overexpression of Axl is observed in ~50% of clinical specimens of lung cancer cases and 60% of NSCLC cell lines and is associated with increased invasion and poor prognosis (20,21). Moreover, Axl expression mediates resistance to anti-EGF receptor (EGFR) therapy including gefitinib and erlotinib in NSCLC (22,23). The inhibition of Axl with Axl-specific siRNA or monoclonal antibodies has been reported to decrease the proliferation of NSCLC cells in vitro and in vivo in a tumor xenograft model (24,25) and depletion of Rac1, a downstream effector of Axl has been shown to result in enhanced sensitivity to anticancer drugs (26,27).

Interleukin (IL)-8 is a proinflammatory cytokine overexpressed in many types of cancer including colon carcinoma, melanoma (28), ovarian (29) and prostate cancer (30), and several studies recently demonstrated that upregulation of IL-8 expression was associated with the acquired resistance against various chemotherapeutic drugs such as cisplatin (31,32), paclitaxel (33), and a receptor tyrosine kinase (RTK) inhibitor, erlotinib which specifically targets epidermal growth factor receptor (EGFR), in ovarian (33), lung (34,35) and head and neck cancer (HNC) cells (36), respectively. Particularly, in erlotinib-resistant HNC cells, the expression of a panel of genes including IL-8, EGFR and Axl was found to be increased (36). In addition, Gong et al reported that in gastric cancer, ErbB2, a member of the EGFR family, is activated and that an ErbB2-targeting agent, trastuzumab, decreased the expression of IL-8 (37). Therefore, IL-8 has received a lot of attention as a potent therapeutic target to control cancer progression as well as chemoresistance and it appears to be quite beneficial to examine whether auto- and/or paracrine regulation of IL-8 production is associated with the expression of some RTKs such as EGFR and Axl or vice versa.

Luteolin, 3′,4′,5,7-tetrahydroxyflavone, is a non-toxic flavonoid widely found in various plants and has many biological activities including antioxidant, anti-inflammatory and anticancer effects. Recent studies have shown that the luteolin induced sensitization of many different cancers to therapeutic drugs (3840), and its anticancer effects were mediated by diverse signaling pathways involved in cell proliferation, angiogenesis, metastasis and apoptosis (41). However, the effect of luteolin has not been studied yet in the expression and activation of TAM RTKs and the association with its cytotoxicity.

In the present study, we tested the association of TAM RTKs in the anticancer effect of luteolin in parental and cisplatin-resistant NSCLC cells to provide a potent therapeutic target to inhibit cell proliferation and overcome chemoresistance.

Materials and methods

Reagents and antibodies

Luteolin was obtained from Sigma-Aldrich (St. Louis, MO, USA). A549 and H460 cells were purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA). Both control shRNA and Axl shRNA which were annealed to gold nanoparticles were synthesized by the domestic company, Bioneer Corp. (Daejeon, Korea). Lipofectamine 2000 and G418 were obtained from Roche Diagnostics Corp. (Indianapolis, IN, USA) and Gibco-BRL (Gaithersburg, MD, USA), respectively. The plasmid, pGL3-basic vector, and the Dual-Glo luciferase assay kit were purchased from Promega Corp. (Madison, WI, USA). For western blot analysis, specific antibodies against Axl, phosphor-Axl, MerTK, Tyro3 and GAPDH, as well as secondary antibodies were obtained from Santa Cruz Biotechnology (Dallas, TX, USA).

Cell culture and establishment of cisplatin-resistant cells

The A549 and H460 cells were grown in RPMI-1640 medium (Gibco-BRL) containing 10% fetal bovine serum (FBS), 2 mM L-glutamine, 10 U/ml penicillin and 10 g/ml streptomycin at 37°C in 5% CO2 in a water-saturated atmosphere. Cisplatin-resistant cells, A549/CisR and H460/CisR, were established by stepwise exposure of parental cells to escalating concentrations of cisplatin (ranging from 0.5 to 2 µM).

Reverse transcription PCR (RT-PCR)

Cells (3×105) were seeded in a 60 mm culture dish and grown overnight. They were then treated with the indicated concentrations of luteolin for 24 h. Total RNA was extracted using TRI reagent and subjected to cDNA synthesis and PCR. The specific primers were as follows: Axl sense, 5′-AACCTTCAACTCCTGCCTTCTCG-3′ and antisense, 5′-CAGCTTCTCCTTCAGCTCTTCAC-3′; GAPDH sense, 5′-GGAGCCAAAAGGGTCATCAT-3′ and antisense, 5′-GTGATGGCATGGACTGTGGT-3′.

Promoter activity assessment

The promoter reporter plasmid, pGL3-Axl, which contains the Axl promoter region ranging from −887 to +7 bp of the transcriptional start site was amplified by PCR and subcloned into the pGL3-basic vector, the luciferase reporter plasmid. The constructed promoter-reporter plasmid was co-transfected into cells (3×105 cells in a 60-mm dish) with Renilla luciferase vectors, pRL-SV40, as an internal control. Luciferase activity was asessed using a Dual-Glo luciferase assay system.

Western blot analysis

Total cell lysates were prepared from the parental or chemoresistant cells treated with the indicated concentrations (0, 20, 40 and 80 µM) of luteolin using lysis buffer [1% Triton X-100, 50 mM Tris (pH 8.0), 150 mM NaCl, 1 mM phenylmethylsulphonyl fluoride (PMSF), 1 mM Na3VO4 and protease inhibitor cocktail]. Untreated cells were used as controls. Protein concentrations were determined using Bio-Rad protein assays. Proteins from the cell lysates (20–40 µg) were separated by 12% SDS-PAGE, and electrotransferred onto nitrocellulose membranes. The membranes were blocked for 30 min at room temperature in Tris-buffered saline with 0.05% Tween-20 (TTBS) containing 5% non-fat dry milk, and then incubated with TTBS containing a primary antibody for 4 h at room temperature. After 3×10 min washes in TTBS, the membranes were incubated with a peroxidase-conjugated secondary antibody for 1 h. Following three additional 10-min washes with TTBS, the protein bands of interest were visualized using an enhanced chemiluminescence detection system (Amersham™ ECL™ Prime Western Blotting Detection Reagent; GE Healthcare, Piscataway, NJ, USA). The density of each protein level was measured by LAS-3000 FujiFilm Image Reader and Multi-Gauge 3.0 software and the Axl protein level was normalized with that of GAPDH.

Cell viability measurement

To assess cell viability, the number of viable cells was counted after trypan blue staining. Briefly, 3×103 cells were seeded into a 60-mm culture dish, grown overnight and then treated with the indicated concentrations (0, 20, 40 and 80 µM) of luteolin for 24 h. After luteolin treatment, cells were harvested and stained with 0.4% trypan blue solution. Dye-excluding viable cells were counted under the microscope. Cell viability was also expressed as a percentage of the viable cells with respect to the untreated control cells. Additionally, the viability of cells was assessed using Cell Counting Kit-8 (CCK-8) assay kit (Dojindo Laboratories, Kumamoto, Japan). Cells (1×103 cells/well) were seeded in 96-well plates and grown overnight, and then treated with the indicated concentrations of luteolin with or without 4 µM cisplatin for the 24 h. At the end of the treatment, 10 µl of CCK-8 solution was added and further incubated for 4 h. The absorbance at 450 nm was measured using a microplate reader (Model 680 microplate reader; Bio-Rad Laboratories, Hercules, CA, USA). Values are expressed as the mean ± SD for triplicate wells and normalized to that of the control group to determine the % of viability.

Colony formation assay

Cells were seeded into 24-well plates (1×102 cells/well) and treated with the indicated concentrations (0, 20, 40 and 80 µM) of luteolin for 24 h. Luteolin-treated cells were then cultured for the next 7–10 days to form colonies. Colonies of >50 cells were stained with crystal violet (in 60% methanol; Junsei Chemical Co., Ltd., Tokyo, Japan) and images were acquired using the RAS-3000 Image Analysis System (FujiFilm, Tokyo, Japan).

Ectopic expression of Axl

To ectopically express Axl, the recombinant plasmid, pcDNA3-Axl, was constructed by cloning the Axl cDNA into the EcoRI and BamHI sites of the pcDNA3 vector and 2 µg of purified plasmids were transfected into the A549 or A549/Cis cells (3×105 cells in a 100-mm dish) using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA). To establish stable cell lines, which constitutively express Axl, the transfected cells were cultured in the presence of 400 µg/ml of G418. The RPMI-1640 medium containing G418 was refreshed every three days. After three to four weeks, the Axl-expressing cells were enriched and the Axl expression in these cells was analyzed by western blot analysis.

Gold nanoparticle-assisted gene delivery system for Axl silencing

Chemically functionalized gold nanoparticles (AuNPs) were annealed with the shRNA targeting Axl gene (5′-TAATACGACTCACTATAGGGAAGAUUUGGAGAACACACUGA-3′) and used as a gene delivery system (GDS) to decrease Axl expression. Briefly, cells (3×105) were seeded in 60-mm culture dishes, grown overnight and then incubated with 10 nM AuNPs-Axl or control AuNPs. The cells were harvested for 24 and 48 h after transfection and used to evaluate protein expression and cell proliferation, respectively.

Statistical analysis

Data are expressed as the means ± SD of triplicate samples or at least three independent experiments. To determine statistical significance, the Student's t-test was used with a P-value threshold of <0.05.

Results

Luteolin inhibits proliferation of both parental and cisplatin-resistant non-small lung cancer cells

The antiproliferative effect of luteolin was first examined in parental non-small cell lung cancer (NSCLC) A549 and H460 cells, as well as cisplatin-resistant A549/CisR and H460/CisR cells. As shown in Fig. 1A, the viability of these cells was found to be decreased in a dose-dependent manner. Next, a clonogenic assay was further performed to confirm the cytotoxicity of luteolin in A549, H460, A549/CisR and H460/CisR cells. These cells were treated with the indicated concentrations of luteolin, and then allowed to grow for the next 10 days. Luteolin treatment resulted in the dose-dependent decrease in the colony formation (Fig. 1B). Notably, H460 and H460/CisR cells that were incubated with 80 µM luteolin failed to form visible colonies, suggesting that luteolin seems to be more cytotoxic to H460 cells than to A549 cells. We also examined the effect of co-treatment of luteolin and cisplatin on cell proliferation using H460/CisR cells. While 4 µM cisplatin alone decreased the viability of the H460/CisR cells to 61% and luteolin decreased that to 67, 41 and 27% in proportion to the concentration of luteolin, co-treatment of cells with luteolin and cisplatin reduced that to 36, 27 and 18%, respectively (Fig. 1C). The results demonstrated that in the presence of cisplatin, the cytotoxicity of luteolin was additively increased in the cisplatin-resistant cells.

Figure 1.

Luteolin inhibits cell proliferation of parental and cisplatin-resistant non-small lung cancer cells. (A) A549, H460 and their cisplatin-resistant cells (A549/CisR and H460/CisR) were seeded into 60 mm dishes (3×105 cells/dish) and grown overnight. Cells were treated with 20, 40 and 80 µM of luteolin for 24 h, and then cells were harvested and stained with trypan blue. The number of viable cells was counted. Data are expressed as the means ± SD of triplicate samples conducted in three independent experiments. The asterisks indicate the significant difference compared to the control value (*P<0.05, vs. the untreated group). (B) Cells (2×103 cells/well) were seeded into a 24-well culture plate, grown overnight, exposed to 20, 40 and 80 µM of luteolin, and allowed to grow for the next 7–10 days. The colonies were visualized by crystal violet staining. The data shown are representative of at least three independent experiments. (C) H460/CisR cells (1×103 cells/well) were seeded in 96-well plates, grown overnight and then treated with the indicated concentrations of luteolin in the absence or presence of 4 µM cisplatin for the 24 h. To assess the cell viability, CCK-8 solution was added into each well and the absorbance at 450 nm was assessed. Values are normalized by the absorbance of the control group and expressed as the mean ± SD for triplicate wells. The asterisks indicate the significant difference compared to the value of cells without cisplatin treatment (*P<0.05).

Luteolin downregulates the expression of RTKs and inhibits Axl activation upon its ligand binding

Since TAM RTKs have been reported to be associated with oncogenesis, proliferation, survival and anti-apoptosis (9,10,42,43), we next assessed the effect of luteolin on the expression of TAM RTKs. Cells were incubated with the indicated concentrations of luteolin for 24 h and the protein levels of each TAM RTK was measured by western blot analysis. We found that in the A549 and A549/CisR cells, luteolin treatment dose-dependently decreased the expression levels of all three TAM RTKs, Tyro3, Axl and MerTK. Similarly in the H460 and H460/CisR cells, the expression levels of both Axl and Tyro3 were decreased by luteolin (Fig. 2A). Notably, in the H460 cells, MerTK was undetectable by western blot analysis.

Figure 2.

Luteolin downregulates the expression of TAM RTKs and inhibits Axl activation upon Gas6 stimulation in non-small lung cancer cells. (A) A549, H460, A549/CisR and H460/CisR cells (3×105 cells/dish) were seeded onto 60-mm dishes, grown overnight, treated with 20, 40 and 80 µM of luteolin for 24 h, and then harvested. The total cell lysates were prepared to determine Tyro3, Axl and MerTK protein levels by western blot analysis. GAPDH was used as a loading control and the results shown are a representative of at least three independent experiments. (B) For RT-PCR, H460 cells (3×105 cells) were seeded into 60 mm culture dishes, grown overnight and treated with the indicated concentrations of luteolin for 24 h. Total RNAs from the cells were isolated and used to determine Axl mRNA levels. GAPDH mRNA was also amplified by RT-PCR as an internal control. The data shown are a representative of three independent experiments The asterisks indicate a significant difference compared to the control value (*P<0.05, **P<0.01, vs. the untreated group). (C) To assess the effect of luteolin on Axl promoter activity, the H460 cells (3×104 cells) were transfected with pGL3-Axl using Lipofectamine 2000. The cells were then incubated with 20, 40 and 80 µM of luteolin for 30, 60, 120 and 240 min and total cell lysates were used to measure luciferase activity. Data are expressed as the means ± SD of triplicate samples conducted in three independent experiments. (D) H460 cells (3×105 cells/dish) were pre-incubated with 40 µM of luteolin for 60 min and stimulated with Gas6 (250 ng/ml) for 20 min. Phospho- and total Axl protein levels were determined by western blot analysis. Total Axl protein level was used as a loading control. Results shown are representative of three independent experiments.

Consistent with the western blot results, the inhibitory effect of luteolin on the Axl gene expression was further demonstrated by RT-PCR and assessment of Axl promoter activity. As shown in Fig. 2B, the mRNA level of Axl was decreased by luteolin treatment in a dose-dependent manner. Additionally, H460 cells transfected with pGL3-Axl, the Axl promoter-luciferase reporter plasmid, were treated with 0, 20, 40 and 80 µM of luteolin for the indicated time periods and luciferase activity was found to be dose-dependently decreased by luteolin treatment (Fig. 2C), indicating that luteolin suppresses Axl expression at the transcriptional level.

Since growth arrest-specific gene 6 (Gas6) binds to all three TAM RTKs and is highly specific to Axl (5), we next observed the effect of luteolin on Axl activation upon ligand binding. Serum-starved H460 cells were pre-treated with luteolin and then stimulated with Gas6. As illustrated in Fig. 2D, Gas6-induced Axl phosphorylation was significantly inhibited by luteolin, indicating that luteolin suppresses tyrosine kinase activity of Axl which is mediated by autophosphorylation of tyrosine residues in the intracellular kinase domain in response to ligand binding (6).

Luteolin affects the expression of Axl, but not IL-8, to exert its antiproliferative effect

To validate the association of Axl in the antiproliferative effects of luteolin, we examined its cytotoxicity toward the H460 cells in which the Axl protein was overexpressed or knocked down, respectively. As shown in Fig. 3A, cells which were transfected with pcDNA3-Axl containing Axl cDNA for ectopic expression of Axl were found to be a less sensitive to luteolin than cells transfected with the pcDNA3 vector (Fig. 3A). Colony formation assay results also showed that clonogenicity of Axl-overexpressing cells was less affected by luteolin treatment compared with the cells transfected with pcDNA3, since luteolin treatment led H460/pc-DNA3 cells to form less colonies and the size of each colony was smaller than that of the H460/pc-DNA3-Axl cells (Fig. 3B). Consistent with the cell viability and colony formation assays, the Axl protein level in the H460/pcDNA3-Axl cells was found to be higher than that in the control cells even after luteolin treatment (Fig. 3C). These results point to the fact that overexpression of Axl attenuates the antiproliferative effect of luteolin.

Next, we examined whether knockdown of Axl increases the antiproliferative effect of luteolin. To decrease Axl expression, we used gold nanoparticle (AuNP)-assisted gene delivery systems (GDS) (44,45). H460 cells were incubated with AuNP GDS conjugates which were annealed with Axl-specific shRNA, or control shRNA. As shown in Fig. 4A, AuNP-GDS-Axl conjugates resulted in the additional decrease in Axl expression following luteolin treatment. The cell viability assay also showed that the AuNP-GDS-Axl conjugates increased cytotoxicity of luteolin (Fig. 4B). These results indicate that the amount of Axl protein was tightly correlated with cell viability and imply that luteolin exerts an antiproliferative effect via the downregulation of Axl expression. Collectively, these results indicate that luteolin inhibits Axl expression, concomitantly induces the protein level of p21, and subsequently abrogates cell proliferation.

Since IL-8, a multifunctional inflammatory cytokine, has been reported to be associated with cell survival, growth, angiogenesis and metastasis in various types of cancers such as melanoma, lung cancer, nasopharyngeal, hepatocellular, ovarian, colorectal and prostate cancer (33,4649), we observed the effect of luteolin on IL-8 expression. ELISA results showed that the IL-8 production in cells treated with 80 µM luteolin was decreased, whereas 20 and 40 µM luteolin rather increased IL-8 level (Fig. 5A). Notably, IL-8 expression/cell was found to be fairly increased by luteolin treatment (Fig. 5B), indicating that the antiproliferative effect of luteolin was not associated with dysregulation of IL-8 expression.

Discussion

A series of recent studies have shown the positive correlation between the expression levels of Axl and the resistance to anticancer drugs, invasiveness and poor outcome in several types of cancer. Moreover, Axl silencing was found to significantly reduce the cell proliferation in NSCLC cells. Therefore, Axl has received more and more attention as a promising therapeutic target in the restriction of disease (50).

In the present study, luteolin was found to suppress expression of all three TAM receptor tyrosine kinases (RTKs) (Fig. 2A). We also observed the inhibitory effect of luteolin on Axl expression (Fig. 2B and C) and Gas6-induced activation of Axl (Fig. 2D). In addition, our data demonstrated that overexpression of Axl by ectopic expression of the Axl gene or knockdown of the Axl protein levels by gold nanoparticle-assisted gene delivery system led to the attenuation or increase of luteolin-induced cytotoxicity, respectively (Figs. 3A and B, and 4B). These results strongly indicate that luteolin targets TAM RTKs, particularly Axl, to function as an anticancer drug. Notably, some increase in the protein level of Axl was found in cisplatin-resistant A549/CisR and H460/CisR cells (Fig. 2A), which appears to be a strategy in order to survive in the presence of cisplatin and/or a mechanism for the acquisition of cisplatin-resistance. However, cisplatin-resistant cells were still sensitive enough to luteolin in spite of the elevated Axl protein level of these cells, suggesting that luteolin can potentially overcome chemoresistance and that there must be other molecules including Tyro3 and MerTK which are affected by luteolin and contribute to its cytotoxicity.

Increasing evidence indicates that cancer-related inflammation of which cytokines are key constituents promotes survival and proliferation of malignant cells (47), supports angiogenesis and metastasis (5153), and evokes epithelial-to-mesenchymal transition (EMT) (54,55) and chemoresistance against anticancer agents (33,56,57). Interleukin-8 (IL-8) has been reported to be overexpressed in tumor tissue and associated with advanced stage disease and poor prognosis (48). Consistent with previous studies (48,58), we found that the level of IL-8 was increased in the cisplatin-resistant H460/CisR cells compared to the parental H460 cells. However, luteolin did not appear to be useful in modulating the levels of IL-8, since only a high concentration of luteolin (80 µM) was found to decrease the expression IL-8 and relatively low concentrations of luteolin (20 and 40 µM) were observed to elevate the levels of IL-8. In parallel, we also found that IL-8 production from a single cell was increased by luteolin. Our results indicate that an adjuvant therapy which decreases the levels of IL-8 or blocks IL-8-mediated signaling pathways may result in a synergistic outcome in cancer treatment. Therefore, IL-8 is a noteworthy target for improved cancer treatment.

In summary, we demonstrated that luteolin suppressed the expression of TAM RTKs, but not IL-8, and activation of Axl upon Gas6 binding, facilitated the inhibitory effect of luteolin on cell proliferation in both the parental and cisplatin-resistant NSCLC cells. Thus, our data imply that TAM RTKs may be potent therapeutic targets of luteolin by which it exerts its anticancer activity, particularly to circumvent chemoresistance in NSCLC cells.

Acknowledgements

The present study was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2014R1A1A2056565), Korea.

Glossary

Abbreviations

Abbreviations:

AuNP

gold nanoparticle

IL-8

interleukin-8

Gas6

growth arrest-specific 6

GDS

gene delivery system

NSCLC

non-small cell lung cancer

RTK

receptor tyrosine kinase

References

1 

Stinchcombe TE and Socinski MA: Treatment paradigms for advanced stage non-small cell lung cancer in the era of multiple lines of therapy. J Thorac Oncol. 4:243–250. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Lemke G: Biology of the TAM receptors. Cold Spring Harb Perspect Biol. 5:a0090762013. View Article : Google Scholar : PubMed/NCBI

3 

Lemke G and Rothlin CV: Immunobiology of the TAM receptors. Nat Rev Immunol. 8:327–336. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Lemke G and Burstyn-Cohen T: TAM receptors and the clearance of apoptotic cells. Ann NY Acad Sci. 1209:23–29. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Nagata K, Ohashi K, Nakano T, Arita H, Zong C, Hanafusa H and Mizuno K: Identification of the product of growth arrest-specific gene 6 as a common ligand for Axl, Sky, and Mer receptor tyrosine kinases. J Biol Chem. 271:30022–30027. 1996. View Article : Google Scholar : PubMed/NCBI

6 

Braunger J, Schleithoff L, Schulz AS, Kessler H, Lammers R, Ullrich A, Bartram CR and Janssen JW: Intracellular signaling of the Ufo/Axl receptor tyrosine kinase is mediated mainly by a multi-substrate docking-site. Oncogene. 14:2619–2631. 1997. View Article : Google Scholar : PubMed/NCBI

7 

Sasaki T, Knyazev PG, Clout NJ, Cheburkin Y, Göhring W, Ullrich A, Timpl R and Hohenester E: Structural basis for Gas6-Axl signalling. EMBO J. 25:80–87. 2006. View Article : Google Scholar : PubMed/NCBI

8 

O'Bryan JP, Frye RA, Cogswell PC, Neubauer A, Kitch B, Prokop C, Espinosa R III, Le Beau MM, Earp HS and Liu ET: axl, a transforming gene isolated from primary human myeloid leukemia cells, encodes a novel receptor tyrosine kinase. Mol Cell Biol. 11:5016–5031. 1991. View Article : Google Scholar : PubMed/NCBI

9 

Rochlitz C, Lohri A, Bacchi M, Schmidt M, Nagel S, Fopp M, Fey MF, Herrmann R and Neubauer A: Axl expression is associated with adverse prognosis and with expression of Bcl-2 and CD34 in de novo acute myeloid leukemia (AML): Results from a multicenter trial of the Swiss Group for Clinical Cancer Research (SAKK). Leukemia. 13:1352–1358. 1999. View Article : Google Scholar : PubMed/NCBI

10 

Berclaz G, Altermatt HJ, Rohrbach V, Kieffer I, Dreher E and Andres AC: Estrogen dependent expression of the receptor tyrosine kinase axl in normal and malignant human breast. Ann Oncol. 12:819–824. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Craven RJ, Xu LH, Weiner TM, Fridell YW, Dent GA, Srivastava S, Varnum B, Liu ET and Cance WG: Receptor tyrosine kinases expressed in metastatic colon cancer. Int J Cancer. 60:791–797. 1995. View Article : Google Scholar : PubMed/NCBI

12 

Nemoto T, Ohashi K, Akashi T, Johnson JD and Hirokawa K: Overexpression of protein tyrosine kinases in human esophageal cancer. Pathobiology. 65:195–203. 1997. View Article : Google Scholar : PubMed/NCBI

13 

Rankin EB, Fuh KC, Taylor TE, Krieg AJ, Musser M, Yuan J, Wei K, Kuo CJ, Longacre TA and Giaccia AJ: AXL is an essential factor and therapeutic target for metastatic ovarian cancer. Cancer Res. 70:7570–7579. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Sainaghi PP, Castello L, Bergamasco L, Galletti M, Bellosta P and Avanzi GC: Gas6 induces proliferation in prostate carcinoma cell lines expressing the Axl receptor. J Cell Physiol. 204:36–44. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Ito T, Ito M, Naito S, Ohtsuru A, Nagayama Y, Kanematsu T, Yamashita S and Sekine I: Expression of the Axl receptor tyrosine kinase in human thyroid carcinoma. Thyroid. 9:563–567. 1999. View Article : Google Scholar : PubMed/NCBI

16 

Byers LA, Diao L, Wang J, Saintigny P, Girard L, Peyton M, Shen L, Fan Y, Giri U, Tumula PK, et al: An epithelial-mesenchymal transition gene signature predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhibitor resistance. Clin Cancer Res. 19:279–290. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Asiedu MK, Beauchamp-Perez FD, Ingle JN, Behrens MD, Radisky DC and Knutson KL: AXL induces epithelial-to-mesenchymal transition and regulates the function of breast cancer stem cells. Oncogene. 33:1316–1324. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Wu F, Li J, Jang C, Wang J and Xiong J: The role of Axl in drug resistance and epithelial-to-mesenchymal transition of non-small cell lung carcinoma. Int J Clin Exp Pathol. 7:6653–6661. 2014.PubMed/NCBI

19 

Brand TM, Iida M, Stein AP, Corrigan KL, Braverman CM, Coan JP, Pearson HE, Bahrar H, Fowler TL, Bednarz BP, et al: AXL is a logical molecular target in head and neck squamous cell carcinoma. Clin Cancer Res. 21:2601–2612. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Wimmel A, Glitz D, Kraus A, Roeder J and Schuermann M: Axl receptor tyrosine kinase expression in human lung cancer cell lines correlates with cellular adhesion. Eur J Cancer. 37:2264–2274. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Shieh YS, Lai CY, Kao YR, Shiah SG, Chu YW, Lee HS and Wu CW: Expression of axl in lung adenocarcinoma and correlation with tumor progression. Neoplasia. 7:1058–1064. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Zhang Z, Lee JC, Lin L, Olivas V, Au V, LaFramboise T, Abdel-Rahman M, Wang X, Levine AD, Rho JK, et al: Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat Genet. 44:852–860. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Rho JK, Choi YJ, Kim SY, Kim TW, Choi EK, Yoon SJ, Park BM, Park E, Bae JH, Choi CM, et al: MET and AXL inhibitor NPS-1034 exerts efficacy against lung cancer cells resistant to EGFR kinase inhibitors because of MET or AXL activation. Cancer Res. 74:253–262. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Ye X, Li Y, Stawicki S, Couto S, Eastham-Anderson J, Kallop D, Weimer R, Wu Y and Pei L: An anti-Axl monoclonal antibody attenuates xenograft tumor growth and enhances the effect of multiple anticancer therapies. Oncogene. 29:5254–5264. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Li Y, Ye X, Tan C, Hongo JA, Zha J, Liu J, Kallop D, Ludlam MJ and Pei L: Axl as a potential therapeutic target in cancer: Role of Axl in tumor growth, metastasis and angiogenesis. Oncogene. 28:3442–3455. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Chen QY, Xu LQ, Jiao DM, Yao QH, Wang YY, Hu HZ, Wu YQ, Song J, Yan J and Wu LJ: Silencing of Rac1 modifies lung cancer cell migration, invasion and actin cytoskeleton rearrangements and enhances chemosensitivity to antitumor drugs. Int J Mol Med. 28:769–776. 2011.PubMed/NCBI

27 

Gastonguay A, Berg T, Hauser AD, Schuld N, Lorimer E and Williams CL: The role of Rac1 in the regulation of NF-κB activity, cell proliferation, and cell migration in non-small cell lung carcinoma. Cancer Biol Ther. 13:647–656. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Scheibenbogen C, Möhler T, Haefele J, Hunstein W and Keilholz U: Serum interleukin-8 (IL-8) is elevated in patients with metastatic melanoma and correlates with tumour load. Melanoma Res. 5:179–181. 1995. View Article : Google Scholar : PubMed/NCBI

29 

Browne A, Sriraksa R, Guney T, Rama N, Van Noorden S, Curry E, Gabra H, Stronach E and El-Bahrawy M: Differential expression of IL-8 and IL-8 receptors in benign, borderline and malignant ovarian epithelial tumours. Cytokine. 64:413–421. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Kim SJ, Uehara H, Karashima T, Mccarty M, Shih N and Fidler IJ: Expression of interleukin-8 correlates with angiogenesis, tumorigenicity, and metastasis of human prostate cancer cells implanted orthotopically in nude mice. Neoplasia. 3:33–42. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Wang Y, Qu Y, Niu XL, Sun WJ, Zhang XL and Li LZ: Autocrine production of interleukin-8 confers cisplatin and paclitaxel resistance in ovarian cancer cells. Cytokine. 56:365–375. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Stronach EA, Cunnea P, Turner C, Guney T, Aiyappa R, Jeyapalan S, de Sousa CH, Browne A, Magdy N, Studd JB, et al: The role of interleukin-8 (IL-8) and IL-8 receptors in platinum response in high grade serous ovarian carcinoma. Oncotarget. 6:31593–31603. 2015.PubMed/NCBI

33 

Duan Z, Feller AJ, Penson RT, Chabner BA and Seiden MV: Discovery of differentially expressed genes associated with paclitaxel resistance using cDNA array technology: Analysis of interleukin (IL) 6, IL-8, and monocyte chemotactic protein 1 in the paclitaxel-resistant phenotype. Clin Cancer Res. 5:3445–3453. 1999.PubMed/NCBI

34 

Fernando RI, Hamilton DH, Dominguez C, David JM, McCampbell KK and Palena C: IL-8 signaling is involved in resistance of lung carcinoma cells to erlotinib. Oncotarget. 7:42031–4204. 2016.PubMed/NCBI

35 

Liu YN, Chang TH, Tsai MF, Wu SG, Tsai TH, Chen HY, Yu SL, Yang JC and Shih JY: IL-8 confers resistance to EGFR inhibitors by inducing stem cell properties in lung cancer. Oncotarget. 6:10415–10431. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Giles KM, Kalinowski FC, Candy PA, Epis MR, Zhang PM, Redfern AD, Stuart LM, Goodall GJ and Leedman PJ: Axl mediates acquired resistance of head and neck cancer cells to the epidermal growth factor receptor inhibitor erlotinib. Mol Cancer Ther. 12:2541–2558. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Gong J, Morishita A, Kurokohchi K, Tani J, Kato K, Miyoshi H, Inoue H, Kobayashi M, Liu S, Murota M, et al: Use of protein array to investigate receptor tyrosine kinases activated in gastric cancer. Int J Oncol. 36:101–106. 2010.PubMed/NCBI

38 

Tu SH, Ho CT, Liu MF, Huang CS, Chang HW, Chang CH, Wu CH and Ho YS: Luteolin sensitises drug-resistant human breast cancer cells to tamoxifen via the inhibition of cyclin E2 expression. Food Chem. 141:1553–1561. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Chian S, Li YY, Wang XJ and Tang XW: Luteolin sensitizes two oxaliplatin-resistant colorectal cancer cell lines to chemotherapeutic drugs via inhibition of the Nrf2 pathway. Asian Pac J Cancer Prev. 15:2911–2916. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Qu Q, Qu J, Guo Y, Zhou BT and Zhou HH: Luteolin potentiates the sensitivity of colorectal cancer cell lines to oxaliplatin through the PPARγ/OCTN2 pathway. Anticancer Drugs. 25:1016–1027. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Lin Y, Shi R, Wang X and Shen HM: Luteolin, a flavonoid with potential for cancer prevention and therapy. Curr Cancer Drug Targets. 8:634–646. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Hutterer M, Knyazev P, Abate A, Reschke M, Maier H, Stefanova N, Knyazeva T, Barbieri V, Reindl M, Muigg A, et al: Axl and growth arrest-specific gene 6 are frequently overexpressed in human gliomas and predict poor prognosis in patients with glioblastoma multiforme. Clin Cancer Res. 14:130–138. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Gustafsson A, Martuszewska D, Johansson M, Ekman C, Hafizi S, Ljungberg B and Dahlbäck B: Differential expression of Axl and Gas6 in renal cell carcinoma reflecting tumor advancement and survival. Clin Cancer Res. 15:4742–4749. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Ryou SM, Kim JM, Yeom JH, Hyun S, Kim S, Han MS, Kim SW, Bae J, Rhee S and Lee K: Gold nanoparticle-assisted delivery of small, highly structured RNA into the nuclei of human cells. Biochem Biophys Res Commun. 416:178–183. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Rosi NL, Giljohann DA, Thaxton CS, Lytton-Jean AK, Han MS and Mirkin CA: Oligonucleotide-modified gold nanoparticles for intracellular gene regulation. Science. 312:1027–1030. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Araki S, Omori Y, Lyn D, Singh RK, Meinbach DM, Sandman Y, Lokeshwar VB and Lokeshwar BL: Interleukin-8 is a molecular determinant of androgen independence and progression in prostate cancer. Cancer Res. 67:6854–6862. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Luppi F, Longo AM, de Boer WI, Rabe KF and Hiemstra PS: Interleukin-8 stimulates cell proliferation in non-small cell lung cancer through epidermal growth factor receptor transactivation. Lung Cancer. 56:25–33. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Waugh DJ and Wilson C: The interleukin-8 pathway in cancer. Clin Cancer Res. 14:6735–6741. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Cheng D, Kong H and Li Y: Prognostic value of interleukin-8 and MMP-9 in nasopharyngeal carcinoma. Eur Arch Otorhinolaryngol. 271:503–509. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Levin PA, Brekken RA, Byers LA, Heymach JV and Gerber DE: Axl receptor axis: A new therapeutic target in lung cancer. J Thorac Oncol. 11:1357–1362. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Shi J and Wei PK: Interleukin-8: A potent promoter of angiogenesis in gastric cancer. Oncol Lett. 11:1043–1050. 2016.PubMed/NCBI

52 

Lin Y, Huang R, Chen L, Li S, Shi Q, Jordan C and Huang RP: Identification of interleukin-8 as estrogen receptor-regulated factor involved in breast cancer invasion and angiogenesis by protein arrays. Int J Cancer. 109:507–515. 2004. View Article : Google Scholar : PubMed/NCBI

53 

Kubo F, Ueno S, Hiwatashi K, Sakoda M, Kawaida K, Nuruki K and Aikou T: Interleukin 8 in human hepatocellular carcinoma correlates with cancer cell invasion of vessels but not with tumor angiogenesis. Ann Surg Oncol. 12:800–807. 2005. View Article : Google Scholar : PubMed/NCBI

54 

Long X, Ye Y, Zhang L, Liu P, Yu W, Wei F, Ren X and Yu J: IL-8, a novel messenger to cross-link inflammation and tumor EMT via autocrine and paracrine pathways (Review). Int J Oncol. 48:5–12. 2016.PubMed/NCBI

55 

Fu XT, Dai Z, Song K, Zhang ZJ, Zhou ZJ, Zhou SL, Zhao YM, Xiao YS, Sun QM, Ding ZB, et al: Macrophage-secreted IL-8 induces epithelial-mesenchymal transition in hepatocellular carcinoma cells by activating the JAK2/STAT3/Snail pathway. Int J Oncol. 46:587–596. 2015.PubMed/NCBI

56 

Duan Z, Lamendola DE, Penson RT, Kronish KM and Seiden MV: Overexpression of IL-6 but not IL-8 increases paclitaxel resistance of U-2OS human osteosarcoma cells. Cytokine. 17:234–242. 2002. View Article : Google Scholar : PubMed/NCBI

57 

Maheshwari A, Lu W, Guida WC, Christensen RD and Calhoun DA: IL-8/CXC ligand 8 survives neonatal gastric digestion as a result of intrinsic aspartyl proteinase resistance. Pediatr Res. 57:438–444. 2005. View Article : Google Scholar : PubMed/NCBI

58 

Zuco V, Cassinelli G, Cossa G, Gatti L, Favini E, Tortoreto M, Cominetti D, Scanziani E, Castiglioni V, Cincinelli R, et al: Targeting the invasive phenotype of cisplatin-resistant non-small cell lung cancer cells by a novel histone deacetylase inhibitor. Biochem Pharmacol. 94:79–90. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2017
Volume 37 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lee YJ, Lim T, Han MS, Lee S, Baek SH, Nan H and Lee C: Anticancer effect of luteolin is mediated by downregulation of TAM receptor tyrosine kinases, but not interleukin-8, in non-small cell lung cancer cells. Oncol Rep 37: 1219-1226, 2017
APA
Lee, Y.J., Lim, T., Han, M.S., Lee, S., Baek, S., Nan, H., & Lee, C. (2017). Anticancer effect of luteolin is mediated by downregulation of TAM receptor tyrosine kinases, but not interleukin-8, in non-small cell lung cancer cells. Oncology Reports, 37, 1219-1226. https://doi.org/10.3892/or.2016.5336
MLA
Lee, Y. J., Lim, T., Han, M. S., Lee, S., Baek, S., Nan, H., Lee, C."Anticancer effect of luteolin is mediated by downregulation of TAM receptor tyrosine kinases, but not interleukin-8, in non-small cell lung cancer cells". Oncology Reports 37.2 (2017): 1219-1226.
Chicago
Lee, Y. J., Lim, T., Han, M. S., Lee, S., Baek, S., Nan, H., Lee, C."Anticancer effect of luteolin is mediated by downregulation of TAM receptor tyrosine kinases, but not interleukin-8, in non-small cell lung cancer cells". Oncology Reports 37, no. 2 (2017): 1219-1226. https://doi.org/10.3892/or.2016.5336