IL-5-induced migration via ERK1/2-mediated MMP-9 expression by inducing activation of NF-κB in HT1376 cells

  • Authors:
    • Eo-Jin Lee
    • Sung-Soo  Park
    • Wun-Jae Kim
    • Sung-Kwon Moon
  • View Affiliations

  • Published online on: June 12, 2012     https://doi.org/10.3892/or.2012.1857
  • Pages: 1084-1090
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Interleukin-5 (IL-5) plays an important role in the growth and differentiation of human B cells and eosinophils. However, little is known about the effect of IL-5 on cancer cells. In this study, we investigated the molecular mechanisms involved in the IL-5-induced migration of HT1376 bladder cancer cells. Our results indicated that IL-5 significantly enhanced migration and MMP-9 expression in HT1376 cells. We also found that IL-5 induces transcriptional activation of the binding of NF-κB and AP-1, which are two important nuclear transcription factors that are linked to MMP-9 expression in HT1376 cells. In subsequent experiments, we found activation of ERK1/2 in IL-5-treated HT1376 cells. To examine the involvement of the ERK1/2 signaling pathway on IL-5-induced cell responses, we pretreated HT1376 cells with the ERK1/2 inhibitor U0126 followed by IL-5 treatment. The results showed that U0126 treatment inhibited migration of IL-5-treated HT1376 cells. Moreover, IL-5-stimulated MMP-9 expression was suppressed by the addition of U0126. Inhibition of ERK1/2 function consistently rescued transcriptional activity of NF-κB, without altering AP-1 activation, in IL-5-treated cells. Finally, inhibition of the IL-5-specific receptor IL-5Rα by small interfering RNA (siRNA) suppressed migration, ERK1/2 activation, MMP-9 expression and binding activation of NF-κB in IL-5-treated HT1376 cells. The results of the present study indicate that the IL-28A/IL-28AR1 dyad induces cell migration through ERK1/2-mediated expression of MMP-9 by binding activation of NF-κB in bladder cancer cells. In conclusion, these novel findings indicate that binding of IL-5 to IL-5Rα plays a critical role in MMP-9 expression, which may be involved in the migration of bladder cancer.

Introduction

Bladder cancer is the most common form of cancer in developed countries. The vast majority of malignant tumors found in the urinary bladder are transitional cell carcinoma (TCC) (1), which is a type that is intricately associated with metastasis (2). The knowledge of the cellular and molecular mechanisms of metastasis in TCC of the bladder is essential for the potential application of effective treatment.

The matrix metalloproteinase (MMP) family of extracellular proteinases plays a central role in the migration and invasion of tumor cells (3,4). Tumor cell migration requires a degradation of the extracellular matrix (ECM) and basement membrane by proteases such as MMP-2 (72 kDa) and MMP-9 (92 kDa) (3,4). Previous results demonstrated that MMP-9 expression is associated directly with tumor grade, invasion, migration, and metastasis in the progression of bladder cancer (510). MMP-9 is induced by several growth factors and cytokines in different cell types (1113). Accumulative studies showed that the identification of the transcription factors, including NF-κB, Sp-1 and AP-1, was essential for the induction of MMP-9 in cancer cells (1113).

IL-5 is a T-cell replacing factor (TRF) that stimulates the differentiation of B cells (14). Previous studies have demonstrated the regulatory roles involved in the activation, proliferation and survival of eosinophils (15). IL-5 binds at the cell surface of a receptor made up of heterodimer complexes composed of 2 chains: ligand-specific receptor IL-5Rα and the accessory receptor β-subunit (βc) (14,15). The binding of IL-5 to IL-5Rα resulted in the activation of Jak/Stat, MAPK and PI3K in B cells and eosinophils (14,15). Although many studies have demonstrated the biological role of IL-5, its exact regulatory mechanism in the process of tumor cell migration remains unknown.

Here, we show the molecular and cellular mechanism involved in cytokine IL-5-induced cell migration. In the present study, the expression of both IL-5 and its receptor IL-5Rα was observed in bladder cancer HT-1376 cells. This study is the first to show the potent induction of cell migration by IL-5 in bladder cancer cells. In addition, our results demonstrated the essential role of ERK1/2-mediated MMP-9 expression in IL-5-induced migration of bladder cancer cells.

Materials and methods

Materials

Polyclonal antibodies to ERK, phospho-ERK, p38MAPK, phospho-p38MAPK, JNK and phospho-JNK were obtained from Cell Signaling (Danvers, MA). U0126 was obtained from Calbiochem (San Diego, CA). The polyclonal MMP-9 antibody was obtained from Chemicon. Small interfering RNA (siRNA) oligonucleotides targeting IL-5Rα (5′-GCAGAACGACCACTCACTA-3′) and scramble (5′-CUGUCAGUCAGUCGUAGUAUU-3′) were designed and synthesized by Genolution (Seoul, Korea).

Cell cultures

A human bladder carcinoma cell line (HT1376) was obtained from the American Type Culture Collection. The cells were maintained in DMEM (4.5 g glucose/liter) supplemented with 10% fetal calf serum, L-glutamine and antibiotics (Biological Industries, Beit Haemek, Israel) at 37°C in a 5% CO2 humidified incubator.

RNA extraction

Total RNA was isolated from tissue using TRIzol reagent (Life Technologies, Grand Island, NY), according to the manufacturer’s protocol. The quality and integrity of the RNA was confirmed by agarose gel electrophoresis and ethidium bromide staining, followed by visual examination under ultraviolet light.

Real-time PCR

Real-time PCR assays using a Rotor-Gene 3000 PCR system (Corbett Research, Mortlake, Australia) were performed in the original and independent cohorts. GAPDH was analyzed in parallel as an internal control. Real-time-PCR reactions containing primers and SYBR Premix EX Taq (Takara Bio Inc., Otsu, Japan) were carried out in micro-reaction tubes (Corbett Research). Spectral data were captured and analyzed using Rotor-Gene Real-Time Analysis software 6.0 Build 14 (Corbett Research). For amplification, IL-5 sense (5′-CATCCAGTGCTACTTGTGTT-3′), IL-5 anti-sense (5′-ACTTCAGGTCGAAGTCAATC-3′), IL-5Rα sense (5′-GCAGAACGACCACTCACTA-3′), and IL-5Rα anti-sense (5′-GGTGCAGTGAAGGGAAACT-3′) primers were used. GAPDH was analyzed in parallel as an endogenous RNA reference gene, and data were normalized to the expression of GAPDH.

Immunoblot

Growth-arrested cells were treated with IL-5 in the absence of 10% FBS for various durations at 37°C. The cells were then washed twice with cold PBS and freeze-thawed in 250 μl lysis buffer [containing, in mmol/l, HEPES (pH 7.5) 50, NaCl 150, EDTA 1, EGTA 2.5, DTT 1, β-glycerophosphate 10, NaF 1, Na3VO4 0.1, and phenylmethylsulfonyl fluoride 0.1 and 10% glycerol, 0.1% Tween-20, 10 μg/ml of leupeptin and 2 μg/ml of aprotinin), and then scraped into 1.5-ml tubes. The lysates were placed on ice for 15 min and then centrifuged at 12,000 rpm for 20 min at 4°C. The protein concentration of the supernatant was determined using a Bradford reagent method (Bio-Rad Laboratories). Equal amounts of cellular proteins were resolved by electrophoresis on a 0.1% SDS-10% polyacrylamide gel (SDS-PAGE) under denaturing conditions. The proteins were transferred electrophoretically to nitrocellulose membranes (Hybond; Amersham Corp). After blocking in 10 mmol/l Tris-HCl (pH 8.0), 150 mmol/l NaCl and 5% (wt/vol) non-fat dry milk, the membranes were treated with primary antibodies for 90 min, followed by incubation with peroxidase-conjugated secondary antibodies for 45 min. The immunocomplexes were detected using a chemiluminescence reagent kit (Amersham Corp). For the immunoblotting studies, the experiments were repeated at least 3 times.

Wound healing migration assay

Cells were plated on 6-well dishes and grown to 90% confluence in 2 ml of growth medium. The cells were damaged using a 2-mm-wide tip and were then treated with IL-5. They were allowed to migrate, and photographs were taken through an inverted microscope (original magnification, ×40).

Zymography

Conditioned medium was electrophoresed in a polyacrylamide gel containing 1 mg/ml gelatin. The gel was then washed at room temperature for 2 h with 2.5% Triton X-100 and subsequently at 37°C overnight in a buffer containing 10 mM CaCl2, 150 mM NaCl and 50 mM Tris-HCl, pH 7.5. The gel was stained with 0.2% Coomassie blue and photographed on a light box. Proteolysis was detected as a white zone in a dark blue field (16).

Transfection

Cells were transfected with siRNA using Lipofectamine 2000 transfection reagent according to the manufacturer’s protocols (Invitrogen). After the indicated incubation with IL-5, the cells were studied via immunoblot, zymography, EMSA and wound healing migration.

Nuclear extracts and electrophoretic mobility shift assay (EMSA)

Cultured cells were collected by centrifugation, washed and suspended in a buffer containing 10 mM HEPES (pH 7.9), 10 mM KCl, 0.1 mM EDTA, 0.1 mM EGTA, 1 mM DTT and 0.5 mM PMSF. After 15 min on ice, the cells were vortexed in the presence of 0.5% Nonidet NP-40. The nuclear pellet was then collected by centrifugation and extracted in a buffer containing 20 mM HEPES pH 7.9, 0.4 M NaCl, 1 mM EDTA, 1 mM EGTA, 1 mM DTT and 1 mM PMSF for 15 min at 4°C.

The nuclear extract (10–20 μg) was preincubated at 4°C for 30 min with the 100-fold excess of an unlabeled oligonucleotide spanning the -79 MMP-9 cis-element of interest. The sequences were as follows: AP-1, CTGACCCCTGAGTCAGC ACTT; NF-κB, CAGTGGAATTCCCCAGCC; Sp-1, GCCCA TTCCTTCCGCCCCCAGATGAAGCAG. The reaction mixture was then incubated at 4°C for 20 min in a buffer (25 mM HEPES buffer pH 7.9, 0.5 mM EDTA, 0.5 mM DTT, 0.05 M NaCl and 2.5% glycerol) with 2 μg of poly dI/dC and 5 fmol (2×104 cpm) of a Klenow end-labeled [32P-ATP] 30-mer oligonucleotide, which spanned the DNA binding site in the MMP-9 promoter. The reaction mixture was electrophoresed at 4°C in a 6% polyacrylamide gel using a TBE (89 mM Tris, 89 mM boric acid and 1 mM EDTA) running buffer. The gel was rinsed with water, dried and exposed to X-ray film overnight (16).

Statistical analysis

Where appropriate, data were expressed as the mean ± SE. Data were analyzed by factorial ANOVA and Fisher’s least significant difference test where appropriate. Statistical significance was set at P<0.05.

Results

Expression of IL-5 and its receptor IL-5Rα in bladder cancer HT1376 cells

To investigate whether the IL-5 and its receptor IL-5Rα is expressed in bladder cancer HT1376 cells, we analyzed IL-5 and IL-5Rα mRNA expression. Real-time PCR analysis showed detection of mRNA expression of IL-5 in HT1376 cells (Fig. 1A). Expression of IL-5Rα mRNA was also observed in HT1376 cells (Fig. 1A). Moreover, both IL-5 and IL-5Rα mRNA expressions were strongly enhanced by the addition of 10% FBS (Fig. 1A). These results indicate that expression of IL-5 and its receptor IL-5Rα can be found in bladder cancer HT1376 cells.

IL-5 induces wound healing migration and MMP-9 expression in HT1376 cells

Previous studies have demonstrated the involvement of cell migration in the development of bladder cancer (1,2). We next examined the effect of IL-5 on bladder cancer cell migration using a wound healing migration assay. Treatment of HT1376 cells with IL-5 for 24 h induced the capacity of migration, as compared with the control (Fig. 1B). To explore the relationship between cell migration and MMP expression, we performed gelatin zymographic assay in IL-5-treated HT1376 cells. IL-5 was added to HT1376 cells at various concentrations, in order to determine the optimal dose. IL-5 significantly induced both MMP-2 and MMP-9 expression at 100 ng/ml, as compared with the control in HT1376 cells (Fig. 1C). In addition, both MMP-2 and MMP-9 levels were increased in IL-5-treated HT1376 cells in a time-dependent manner (Fig. 1C). The effect of IL-5 was confirmed by immunoblot. Increase in expression levels of both MMP-2 and MMP-9 were observed in HT1376 cells induced by IL-5 (Fig. 1C).

IL-5-induced MMP-9 expression is involved in binding activities of NF-κB and AP-1 in HT1376 cells

Since the expression of MMP-9 is deeply associated with the development of bladder cancer (510), we focused our investigation on IL-5-induced MMP-9 expression. In order to define transcription factors involved in the IL-5 induction of MMP-9 in HT1376 cells, we performed a gel shift assay (EMSA) using nuclear extract obtained from cells cultured for 24 h in the presence or absence of IL-5 (100 ng/ml). IL-5 significantly increased nuclear binding to NF-κB and AP-1 binding sites (Fig. 2). However, no significant Sp-1 levels were observed in IL-5-treated HT1376 cells (Fig. 2). These results suggest that IL-5-induced MMP-9 expression might be mediated through the binding activation of NF-κB and AP-1.

ERK1/2 inhibitor, U0126, decreases the IL-5-induced migration in HT1376 cells

To examine the MAPK signaling pathway in IL-5-treated HT1376 cells, we carried out immunoblot experiments. Treatment with IL-5 resulted in a significant induction of ERK1/2 activation in HT1376 cells (Fig. 3A). IL-5-induced activation of ERK1/2 was suppressed by U0126, ERK1/2-specific inhibitor (Fig. 3B). In contrast, treatment of HT1376 cells with IL-5 did not lead to activation of JNK and p38MAPK (Fig. 3A). To further investigate the role of ERK1/2 signaling in IL-5-treated HT1376 cells, cells were pretreated with U0126. Blockage of ERK1/2 signaling significantly reduced the migration of HT1376 cells induced by IL-5 (Fig. 4A). These results indicate that ERK1/2 signaling plays an important role in the IL-5-induced migration of bladder cancer cells.

Inhibition of ERK1/2 signaling abolishes IL-5-mediated MMP-9 expression and NF-κB activation in HT1376 cells

The results of the present study showed that IL-5 regulates cell migration and MMP-9 expression (Fig. 1B and C). In addition, IL-5-mediated migration of bladder cancer cells was suppressed by an ERK1/2 specific inhibitor U0126 (Fig. 4A). Thus, to define the role of ERK1/2 signaling in IL-5-induced MMP-9 expression, HT1376 cells were pretreated with U0126 for 40 min, followed by IL-5 treatment for 24 h. U0126 effectively blocked increased MMP-9 expression (Fig. 4B). To verify the possible implication of ERK1/2 signaling in transcription factors NF-κB and AP-1, which is associated with IL-5-induced MMP-9 expression, we next performed a gel shift assay. As shown in Fig. 4C, NF-κB DNA binding activity was almost abolished by the addition of U0126. In contrast, the inhibition of ERK1/2 had no effect on the IL-5-induced binding activity of AP-1. These results suggest that the ERK1/2 signaling pathway must be involved in IL-5-induced MMP-9 expression via activation of NF-κB in HT1376 cells.

Knockdown of IL-5Rα, ligand-specific IL-5 receptor, reduces IL-5-induced migration of HT1376 cells

To determine the regulatory mechanism of IL-5, we used the siRNA-mediated knockdown of IL-5Rα in IL-5-treated HT1376 cells. The cells were transfected with si-IL-5Rα and scramble siRNA, respectively, followed by treatment with IL-5. To evaluate the transfection efficiency of siRNA in HT1376 cells, expression of IL-5Rα was examined by immunoblotting. IL-5Rα was inhibited by transfection of HT1376 cells with a specific siRNA against IL-5Rα (si-IL-5Rα) (Fig. 5E). In addition, transfection of scramble siRNA into cells remained unchanged for protein levels of IL-5Rα (Fig. 5E). The results suggested that the siRNA molecule was specific and effective. The effect of si-IL-5Rα was then determined on IL-5-induced migration. Our results showed that the inhibition of IL-5Rα significantly reduced the migration of HT1376 cells induced by IL-5, compare to siRNA control groups (Fig. 5A). These data suggest that IL-5 induced the migration of bladder cancer cells through its specific IL-5 receptor, IL-5Rα.

Inhibition of IL-5Rα knockdown decreases ERK1/2 activation, MMP-9 expression, and binding activity of NF-κB in IL-5-treated HT1376 cells

We sequentially further investigated the effects of si-IL-5Rα on the induction of ERK1/2 activation, MMP-9 expression, and binding activity of NF-κB in IL-5-treated HT1376 cells. As shown in Fig. 5B, transfection of si-IL-5Rα significantly suppressed the activation of ERK1/2 in response to IL-5. In addition, the blockade of IL-5Rα reversed MMP-9 expression and NF-κB binding activity to control levels in IL-5-treated HT1376 cells (Fig. 5C and D). These results suggest that IL-5 enhanced ERK1/2 activation, MMP-9 expression, and binding activity of NF-κB via IL-5Rα receptor in HT1376 cells.

Discussion

Although the role of cytokine IL-5 in the biological responses of immune cells is well established, the role and mechanism involved in IL-5-induced migration of tumor cells remains to be investigated. Previous studies proposed that IL-5 is a regulatory cytokine supporting the growth and differentiation of activated B cells (14). Subsequent studies have shown the essential roles of IL-5 in the growth, activation and survival of eosinophils (15). Some studies have indicated that IL-5 has an antitumor effect in mouse B cell lymphoma and colon tumor cells (17,18). In contrast, the results of the present study from our results showed that IL-5 plays a pivotal role in the migration of bladder cancer cells.

The inflammatory process may be responsible for the development and progression of cancer (19). Inflammation in the bladder is the result of several pathological processes, which involve the accumulation of immune cells and the release of cytokines (20,21). We hypothesize that increased production of inflammatory cytokines may contribute to an altered microenvironment in the bladder, which leads to the progression of bladder cancer. In the first stage, real-time PCR analysis revealed that the mRNA expression of IL-5 and its specific subunit of receptor IL-5Rα were detected in bladder cancer HT1376 cells. These results suggest that IL-5 is constitutively expressed in bladder cancer cells and might be an important regulatory cytokine associated with the progression of bladder cancer.

We next investigated the molecular regulation involved in the development and progression of bladder cancer. In the present study, we demonstrated that IL-5 enhanced wound healing migration of bladder cancer cells. In addition, both MMP-2 and MMP-9 expressions were induced by IL-5 treatment. MMPs have been implicated in cell migration through the degradation of extracellular matrix components (3,4). It is well accepted that MMP-2 and MM-9 are particularly important factors in cell migration (3,4). The importance and role of MMP-9 in bladder tumor has been demonstrated in in vivo orthotopic xenograft models (22,23), preclinical evidence (59), and in the study of polymorphisms (24). Therefore, we investigated the transcription factors binding to human MMP-9 promoter regions. Several proximal binding sites, including NF-κB, AP-1 and Sp-1, that regulate MMP-9 promoter have been identified in tumor cells (1113). In 5637 bladder cancer cells stimulated with TNF-α, NF-κB was shown to be essential for MMP-9 expression, but AP-1 and Sp-1 was not affected (25). Our EMSA results indicated that IL-5 increased the binding activity of NF-κB and AP-1, known binding sites in the MMP-9 promoter, without detecting inducible levels of Sp-1. We concluded that NF-κB and AP-1 sites may be responsible for the IL-5-induced transcriptional activation of the MMP-9 in HT1376 bladder cancer cells.

Several signaling pathways have been identified in the MMP-9 regulation of various cells (16,2629). Studies of MMP-9 in bladder cancer cells have focused mainly on investigating the signaling pathways in response to various factors. The ERK1/2 pathway mediates induction of MMP-9 via TNF-α in HT1376 cells (30). The involvement of p38MAPK has been associated with the regulation of MMP-9 expression in bladder cancer HTB9, HTB5, 5637 and HT1376 cells (10,25,30). In a recent study, Ras-induced MMP-9 expression was inhibited by RhoA inhibitor Y-27632 (31). However, the issue of the signaling pathways underlying the induction of MMP-9 in IL-5-treated cancer cells has not yet been clarified. In the present study, our result showed that IL-5 treatment induced activation of ERK1/2 in HT1376 cells. Our data also show that U0126 (ERK1/2 inhibitor) decreases IL-5-induced wound healing migration and MMP-9 expression via reductions in NF-κB binding without altering AP-1 activation in HT1376 cells. The results of the present study clearly show that ERK1/2 mediates MMP-9 expression via activation of NF-κB binding, which results in enhanced cell migration in bladder cancer HT1376 cells. It is possible that NF-κB-mediated expression of MMP-9, regulated by ERK1/2, may cooperate in the migration of bladder cancer cells.

IL-5 transduces signals through heterodimer receptor complexes, a unique IL-5Rα and a common β-subunit (βc), leading to the activation of different signaling pathways on target cells (14,15). Previous reports have suggested that the cytoplasmic region of the IL-5Rα subunit is critical for IL-5 signaling (14,15,32). We therefore investigated whether IL-5Rα contributes to the IL-5-mediated cell responses using IL-5Rα-specific siRNA (si-IL-5Rα) in bladder cancer cells. The increased migration, activation of ERK1/2, MMP-9 expression, and NF-κB binding activity in response to IL-5 was suppressed by abolition of IL-5Rα, suggesting that IL-5Rα is indispensable in the transmission of the migratory cell signal of IL-5.

Our results disagree with those of previous reports showing an antitumor effect of IL-5 on mouse B cell lymphoma and colon tumor cells. Considering these opposing results, this may explain the differences in cell responses by IL-5 within tumor cell type species. Although an emerging amount of attention is being paid to the opposing functions of inflammatory cytokines as extrinsic suppressors of tumors and as pro-tumor growth stimulators, the model remains to be controversial (19,33). In the present study, we showed that IL-6 released by bladder cancer cells could play a crucial role in tumor growth and development. Further studies are required to investigate the role of IL-5 in tumor growth using animal models.

In summary, our results are consistent with 3 major results: i) both IL-5 and IL-5Rα are produced by bladder cancer HT1376 cells; ii) IL-5-induced migration regulates ERK1/2-mediated MMP-9 expression via the binding activity of NF-κB in HT1376 cells; and iii) IL-5Rα receptor is essential for the migration of bladder cancer HT1376 cells by IL-5, which may be mediated in part by regulating the ERK1/2-associated MMP-9 expression via activation of NF-κB binding. Collectively, these novel results point to the potential use of IL-5 in future potential molecular therapy for bladder cancer.

Acknowledgements

This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF), funded by the Ministry of Education, Science and Technology (2010-0001736) and the Korea Healthcare Technology R&D Project, Ministry of Health & Welfare, Republic of Korea (A100651-1011-0000200).

References

1 

Jemal A, Siegel R, Ward E, Murray T, Xu J and Thun MJ: Cancer statistics, 2007. CA Cancer J Clin. 57:43–66. 2007. View Article : Google Scholar

2 

Black PC and Dinney CP: Bladder cancer angiogenesis and metastasis - translation from murine model to clinical trial. Cancer Metastasis Rev. 26:623–634. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Matrisian LM: Metalloproteinases and their inhibitors in matrix remodeling. Trends Genet. 6:121–125. 1990. View Article : Google Scholar : PubMed/NCBI

4 

Liotta LA: Tumor invasion and metastasis-role of extracellular matrix: Rhoads Memorial Award Lecture. Cancer Res. 46:1–7. 1986.PubMed/NCBI

5 

Sier CF, Casetta G, Verheijen JH, et al: Enhanced urinary gelatinase activities (matrix metalloproteinases 2 and 9) are associated with early-stage bladder carcinoma: a comparison with clinically used tumor markers. Clin Cancer Res. 6:2333–2340. 2000.

6 

Davies B, Waxman J, Wasan H, et al: Levels of matrix metalloproteases in bladder cancer correlate with tumor grade and invasion. Cancer Res. 53:5365–5369. 1993.PubMed/NCBI

7 

Nutt JE, Durkan GC, Mellon JK and Lunec J: Matrix metalloproteinases (MMPs) in bladder cancer: the induction of MMP9 by epidermal growth factor and its detection in urine. BJU Int. 91:99–104. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Moses MA, Wiederschain D, Loughlin KR, Zurakowski D, Lamb CC and Freeman MR: Increased incidence of matrix metalloproteinases in urine of cancer patients. Cancer Res. 58:1395–1399. 1998.PubMed/NCBI

9 

Di Carlo A, Terracciano D, Mariano A and Macchia V: Urinary gelatinase activities (matrix metalloproteinases 2 and 9) in human bladder tumors. Oncol Rep. 15:1321–1326. 2006.PubMed/NCBI

10 

Kumar B, Koul S, Petersen J, et al: p38 mitogen-activated protein kinase-driven MAPKAPK2 regulates invasion of bladder cancer by modulation of MMP-2 and MMP-9 activity. Cancer Res. 70:832–841. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Mook OR, Frederiks WM and Van Noorden CJ: The role of gelatinases in colorectal cancer progression and metastasis. Biochim Biophys Acta. 1705:69–89. 2004.PubMed/NCBI

12 

Visse R and Nagase H: Matrix metalloproteinases and tissue inhibitors of metalloproteinases: structure, function, and biochemistry. Circ Res. 92:827–839. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Busti C, Falcinelli E, Momi S and Gresele P: Matrix metalloproteinases and peripheral arterial disease. Intern Emerg Med. 5:13–25. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Takatsu K and Nakajima H: IL-5 and eosinophilia. Curr Opin Immunol. 20:288–294. 2008. View Article : Google Scholar

15 

Adachi T and Alam R: The mechanism of IL-5 signal transduction. Am J Physiol. 275:C623–C633. 1998.PubMed/NCBI

16 

Moon SK, Cha BY and Kim CH: ERK1/2 mediates TNF-alpha-induced matrix metalloproteinase-9 expression in human vascular smooth muscle cells via the regulation of NF-kappaB and AP-1: involvement of the ras dependent pathway. J Cell Physiol. 198:417–427. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Wu HK, Hirai H, Inamori K, Kitamura K and Takaku F: Anti-tumor effects of interleukin-4 and interleukin-5 against mouse B cell lymphoma and possible mechanisms of their action. Jpn J Cancer Res. 83:200–210. 1992. View Article : Google Scholar : PubMed/NCBI

18 

Masuda Y, Mita S, Sakamoto K, Ishiko T and Ogawa M: Suppression of in vivo tumor growth by the transfection of the interleukin-5 gene into colon tumor cells. Cancer Immunol Immunother. 41:325–330. 1995. View Article : Google Scholar : PubMed/NCBI

19 

Coussens LM and Werb Z: Inflammation and cancer. Nature. 420:860–867. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Tyagi P, Barclay D, Zamora R, et al: Urine cytokines suggest an inflammatory response in the overactive bladder: a pilot study. Int Urol Nephrol. 42:629–635. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Michaud DS: Chronic inflammation and bladder cancer. Urol Oncol. 25:260–268. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Dinney CP, Fishbeck R, Singh RK, et al: Isolation and characterization of metastatic variants from human transitional cell carcinoma passaged by orthotopic implantation in athymic nude mice. J Urol. 154:1532–1538. 1995. View Article : Google Scholar : PubMed/NCBI

23 

Mian BM, Dinney CP, Bermejo CE, et al: Fully human anti-interleukin 8 antibody inhibits tumor growth in orthotopic bladder cancer xenografts via down-regulation of matrix metalloproteases and nuclear factor-kappaB. Clin Cancer Res. 9:3167–3175. 2003.PubMed/NCBI

24 

Kader AK, Liu J, Shao L, et al: Matrix metalloproteinase polymorphisms are associated with bladder cancer invasiveness. Clin Cancer Res. 13:2614–2620. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Lee SJ, Park SS, Lee US, Kim WJ and Moon SK: Signaling pathway for TNF-alpha-induced MMP-9 expression: mediation through p38 MAP kinase, and inhibition by anti-cancer molecule magnolol in human urinary bladder cancer 5637 cells. Int Immunopharmacol. 8:1821–1826. 2008. View Article : Google Scholar

26 

Cho A, Graves J and Reidy MA: Mitogen-activated protein kinases mediate matrix metalloproteinase-9 expression in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 20:2527–2532. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Wiehler S, Cuvelier SL, Chakrabarti S and Patel KD: p38 MAP kinase regulates rapid matrix metalloproteinase-9 release from eosinophils. Biochem Biophys Res Commun. 315:463–470. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Yao J, Xiong S, Klos K, et al: Multiple signaling pathways involved in activation of matrix metalloproteinase-9 (MMP-9) by heregulin-beta1 in human breast cancer cells. Oncogene. 20:8066–8074. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Genersch E, Hayess K, Neuenfeld Y and Haller H: Sustained ERK phosphorylation is necessary but not sufficient for MMP-9 regulation in endothelial cells: involvement of Ras-dependent and -independent pathways. J Cell Sci. 113:4319–4330. 2003.PubMed/NCBI

30 

Lee SJ, Park SS, Cho YH, et al: Activation of matrix metalloproteinase-9 by TNF-alpha in human urinary bladder cancer HT1376 cells: the role of MAP kinase signaling pathways. Oncol Rep. 19:1007–1013. 2008.PubMed/NCBI

31 

Chang HR, Huang HP, Kao YL, et al: The suppressive effect of Rho kinase inhibitor, Y-27632, on oncogenic Ras/RhoA induced invasion/migration of human bladder cancer TSGH cells. Chem Biol Interact. 183:172–180. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Takaki S, Murata Y, Kitamura T, Miyajima A, Tominaga A and Takatsu K: Reconstitution of the functional receptors for murine and human interleukin 5. J Exp Med. 177:1523–1529. 1993. View Article : Google Scholar : PubMed/NCBI

33 

Dranoff G: Cytokines in cancer pathogenesis and cancer therapy. Nat Rev Cancer. 4:12–22. 2004. View Article : Google Scholar

Related Articles

Journal Cover

September 2012
Volume 28 Issue 3

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 E, Park S, Kim W and Moon S: IL-5-induced migration via ERK1/2-mediated MMP-9 expression by inducing activation of NF-κB in HT1376 cells. Oncol Rep 28: 1084-1090, 2012
APA
Lee, E., Park, S., Kim, W., & Moon, S. (2012). IL-5-induced migration via ERK1/2-mediated MMP-9 expression by inducing activation of NF-κB in HT1376 cells. Oncology Reports, 28, 1084-1090. https://doi.org/10.3892/or.2012.1857
MLA
Lee, E., Park, S., Kim, W., Moon, S."IL-5-induced migration via ERK1/2-mediated MMP-9 expression by inducing activation of NF-κB in HT1376 cells". Oncology Reports 28.3 (2012): 1084-1090.
Chicago
Lee, E., Park, S., Kim, W., Moon, S."IL-5-induced migration via ERK1/2-mediated MMP-9 expression by inducing activation of NF-κB in HT1376 cells". Oncology Reports 28, no. 3 (2012): 1084-1090. https://doi.org/10.3892/or.2012.1857