Open Access

A novel uPAg-KPI fusion protein inhibits the growth and invasion of human ovarian cancer cells in vitro

  • Authors:
    • Li-Ping Zhao
    • Tian-Min Xu
    • Mu-Jie Kan
    • Ye-Chen Xiao
    • Man-Hua Cui
  • View Affiliations

  • Published online on: March 24, 2016     https://doi.org/10.3892/ijmm.2016.2540
  • Pages: 1310-1316
  • Copyright: © Zhao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Urokinase-type plasminogen activator (uPA) acts by breaking down the basement membrane and is involved in cell proliferation, migration and invasion. These actions are mediated by binding to the uPA receptor (uPAR) via its growth factor domain (GFD). The present study evaluated the effects of uPAg-KPI, a fusion protein of uPA-GFD and a kunitz protease inhibitor (KPI) domain that is present in the amyloid β-protein precursor. Using SKOV-3 cells, an ovarian cancer cell line, we examined cell viability, migration, invasion and also protein expression. Furthermore, we examined wound healing, and migration and invasion using a Transwell assay. Our data showed that uPAg-KPI treatment reduced the viability of ovarian cancer SKOV-3 cells in both a concentration and time-dependent manner by arresting tumor cells at G1/G0 phase of the cell cycle. The IC50 of uPAg-KPI was 0.5 µg/µl after 48 h treatment. At this concentration, uPAg-KPI also inhibited tumor cell colony formation, wound closure, as well as cell migration and invasion capacity. At the protein level, western blot analysis demonstrated that uPAg-KPI exerted no significant effect on the expression of total extracellular signal-regulated kinase (ERK)1/ERK2 and AKT, whereas it suppressed levels of phosphorylated ERK1/ERK2 and AKT. Thus, we suggest that this novel uPAg-KPI fusion protein reduced cell viability, colony formation, wound healing and the invasive ability of human ovarian cancer SKOV-3 cells in vitro by regulating ERK and AKT signaling. Further studies using other cell lines will confirm these findings.

Introduction

Ovarian cancer is a significant health problem worldwide, accounting for more than 200,000 new cases of cancer and 125,000 cancer-related deaths each year. Furthermore, ovarian cancer is ranked as the sixth most common cancer and seventh most common cause of mortality for women (1). Despite recent advancements in the prevention, detection and treatment of ovarian cancer, it remains the cause of the highest number of gynecological-related mortalities in Western countries. This is thought to be due to late diagnosis, thereby reducing treatment options (stages III–IV) (2). As a result of this situation, novel treatments and earlier diagnostic tools are urgently required in order to effectively target ovarian cancer.

In ovarian cancer, similar to other cancers, tumor metastasis has been shown to be responsible for a significant number of mortalities, and tumor cell invasion is dependent on finely regulated extracellular proteolytic activity, which allows tumor cells to invade the extracellular matrix (3). Urokinase-type plasminogen activator (uPA) breaks down the basement membrane upon binding to the uPA receptor (uPAR) through the respective growth factor domain (GFD) (4). Thus, the activated uPA/uPAR/plasmin proteolytic cascade plays a key role in tumor invasion and the dissemination of various malignancies (5,6). In addition to its recognized function in extracellular matrix degradation, uPA signaling has been implicated in tumor cell proliferation, migration and the invasion of local tissues (4). However, it has been hypothesized that downregulation of uPA and uPAR, blockade of uPA activity using an antibody or small molecule inhibitor, or interfering with uPA binding to its receptor could suppress the promotion of cell mobility and growth. This approach is considered to be a novel method in treating tumors. Indeed, certain of these potential approaches have demonstrated potent anticancer effects and are currently being further evaluated in clinical trials (710) (http://www.wilex.de/portfolio/mesupron/phase-i-mit-wx-uk1/).

In cases of ovarian cancer, the expression of uPA and uPAR has been associated with epithelial ovarian cancer progression (11,12). Previously, it has been demonstrated that high levels of expression of uPA are associated with residual tumor progression in ovarian cancer patients, suggesting that targeting uPA signaling helps clinicians to stratify patients (13). Thus, in the present study, we constructed and expressed a fusion protein, uPAg-kunitz protease inhibitor (KPI). This involved fusing uPA GFD with KPI, a kunitz protease inhibitor domain that is found in amyloid beta-protein precursor (APP). Incorporation of the KPI-amyloid protein precursor allowed for inhibition of serine protease hydrolysis (14). Subsequently, we assessed uPAg-KPI antitumor activity in ovarian cancer cells in vitro. Our previous study expressed uPAg-KPI in Pichia pastoris, and used ion exchange chromatography to purify the target protein. Renaturalization was carried out using ammonium sulfate, and biological analysis demonstrated that this fusion protein inhibited serine protease activity (15). The present study provides insightful information in relation to the effect of uPAg-KPI regulation on ovarian cancer cell viability, migration and invasion in vitro.

Materials and methods

Chemicals and reagents

3-(4,5-dimethylthiazol-2-yl)-2,5-di-phenyltetrazolium bromide (MTT) and dimethyl sulfoxide (DMSO) were both purchased from Sigma (St. Louis, MO, USA), and the BrdU Flow kit and Biocoat Matrigel™ invasion inserts were both purchased from BD Biosciences (San Jose, CA, USA). The Transwell chamber was from Corning (Corning, NY, USA), and Iscove's modified Dulbecco's medium (IMDM) and fetal bovine serum (FBS) were both from HyClone (Logan, UT, USA). Crystal violet, Trypan blue, penicillin-streptomycin and trypsin-EDTA were all purchased from Solarbio (Beijing, China), and the primary antibodies used for western blot analysis of p-extracellular signal-regulated kinase (ERK; 4695), p-ERK (9101), AKT (9272) and p-AKT (4058) were all purchased from Cell Signaling Technology (Danvers, MA, USA). uPAg-KPI was constructed and prepared in the College of Pharmacy, Biological Engineering Laboratory (Jilin University, Changchun, China), it was expressed in Pichia pastoris, by renaturation and ion exchange chromatography; biological analysis demonstrated that the fusion protein inhibited serine protease activity, as previously described (15).

Human ovarian adenocarcinoma cell line and culture

The human ovarian adenocarcinoma cell line SKOV-3 was obtained from Biochemical Teaching and Research Section, Basic Medical Institute, Jilin University (Changchun, China) and cultured in IMDM supplemented with 10% FBS, 100 U/ml penicillin, and 100 µg/ml streptomycin in a humidified incubator with 5% CO2 and 95% air at 37°C.

Cell viability MTT assay

SKOV-3 cells were seeded and grown on a 96-well plate with a flat bottom at a density of 5,000 cells/well in complete medium supplemented with 10% FBS for 24 h. Following incubation, the wells were treated with 0, 0.05, 0.1, 0.5 and 1.0 µg/µl of uPAg-KPI, respectively for up to 72 h. At the end of each experiment, 20 µl MTT solution (5 mg/ml) was added and cells were further incubated for 4 h at 37°C. Following this, cell medium was replaced with 100 µl/well DMSO and optical density (A490 nm) was read on an automated microplate reader (BioTek, Winooski, VT, USA). The data were calculated as the means ± SD.

Flow cytometric cell cycle distribution assay

To analyze cell cycle distribution, cells were cultured overnight in serum-free IMDM overnight and in IMDM-containing 10% FBS for treatment with 0.5 µg/µl concentration of uPAg-KPI for 48 h. BrdU was then added (10 µl/ml) to cell cultures for an additional 0.5 h. Cells were then collected by trypsinization, fixed in a Cytofix/Cytoperm buffer and Cytoperm-plus buffer in the dark, and then washed in a Perm/Wash buffer, and digested with RNase (all from BD Biosciences) for 1 h at 37°C. The resulting solution was resuspended in 1 ml of Perm/Wash buffer and stained with 0.01 mg/ml of propidium iodide in the dark for 30 min at room temperature. Cells were analyzed using a flow cytometer (BD Accuri C6; BD Biosciences).

Colony formation assay

Cells at a density of 200 cells/well were added to 6-well plates and cultured in IMDM containing 10% FBS in triplicate for 12 h and then treated with uPAg-KPI (0.5 µg/µl) at 37°C for 14 days. Following treatment, cells were washed twice with phosphate-buffered saline (PBS; Solarbio) and fixed with 4% paraformaldehyde for 30 min and then stained with 0.1% crystal violet hydrate solution for 20 min. The number of colonies with ≥50 cells was counted under an inverted microscope (Nikon, Tokyo, Japan) and calculated as: plate clone formation efficiency = number of colonies/number of cells inoculated ×100.

In vitro 'wound' closure assay

SKOV-3 cells were plated in 6-well plates and treated with 0.5 µg/µl uPAg-KPI until cells reached confluence. A 'wound' was then created by scratching the surface of the well with a pipette tip. Following injury, cells were further cultured in serum-free IMDM with or without uPAg-KPI (0.5 µg/µl) for 24 and 48 h. Cells were then photographed using a phase-contrast microscope (×100) and the wound closure was measured as previously described (16).

Tumor cell migration and invasion assay

In the present study, SKOV-3 cells were treated in the presence or absence of 0.5 µg/µl uPAg-KPI for two days, and tumor cell migration and invasion capacity were measured using the Transwell system (Corning) with or without Matrigel precoating (BD Biocoat™ Matrigel Invasion chamber with an 8-µm filter), respectively. In brief, cells were trypsinized and resuspended in serum-free IMDM and placed in the upper chamber of a Transwell plate at a density of 2×104 cells/well, containing 0.5 µg/µl of uPAg-KPI or control loading solution (PBS). The bottom chambers contained IMDM and 20% FBS. Cells were incubated for 48 h in a humidified atmosphere with 95% air and 5% CO2 at 37°C. Non-migrated or invasive cells in the upper chamber were removed by wiping the upper side of the membrane with a cotton swab. Migrated or invading cells on the reverse side of the membrane were fixed with 4% paraformaldehyde and stained with 0.1% crystal violet for 15 min and counted under a light microscope at a magnification of ×200.

Protein extraction and western blot analysis

SKOV-3 cells were plated onto a 10-cm tissue culture dish and grown for 24 h. uPAg-KPI (0.5 µg/µl) was then added to each dish and PBS (the solvent) was used as a vehicle control. Cells were incubated at 37°C for 0, 12, 24 and 48 h and harvested and resuspended in ice-cold PBS (pH 7.4) containing 2 mM EDTA and 0.1% Triton X-100. Collected cells were sonicated and centrifuged at 12,000 rpm for 20 min at 4°C in order to remove cell debris. The supernatant was collected for determination of total protein concentration using a Pierce™ protein BCA assay kit (Thermo Fisher Scientific, Waltham, MA, USA) with bovine serum albumin (BSA) as a standard. Briefly, samples were electrophoresed and then transferred to polyvinylidene difluoride membrane using a semidry transfer system. Non-specific binding sites were blocked for 1 h in Tris-buffered saline, pH 7.6, containing 5% dried skimmed milk. Blots were incubated with ERK1/2, p-ERK1/2, AKT and p-AKT antibodies overnight at 4°C, then diluted with secondary antibody for 1 h. After several washes, reactive bands were visualized using an ECL chemiluminescence detection kit (Thermo Fisher Scientific) (17).

Statistical analysis

In the present study, all data are represented as the means ± SD of three independent experiments. Statistical difference was evaluated using the Student's t-test, and a P-value <0.05 was considered to indicate a statistically significant difference. All statistical analyses were performed using SPSS, version 19 software (SPSS, Chicago, IL, USA) to compare the experimental versus control groups.

Results

Effects of uPAg-KPI on inhibition of SKOV-3 cell viability

In order to assess the effect of uPAg-KPI on the inhibition of ovarian cancer cell growth, we treated cells with various concentrations of uPAg-KPI for up to 72 h and performed the cell viability MTT assay. Our data demonstrated that uPAg-KPI markedly reduced cell viability in a dose- and time-dependent manner in SKOV-3 cells (Fig. 1A). The IC50 value of uPAg-KPI was 0.5 µg/µl in SKOV-3 cells following 48 h treatment. We thus selected 0.5 µg/µl uPAg-KPI for 48 h treatment for subsequent experiments. A reduction in cell viability by uPAg-KPI was also confirmed by Trypan blue dye exclusion (data not shown).

Following the aforementioned experiments, flow cytometric analysis (cell cycle distribution assay) demonstrated that cells treated with uPAg-KPI (0.5 µg/µl) for 48 h accumulated at the G1 phase of cell cycle (52.7 vs. 36.6% of control) (Fig. 1B and C). Moreover, the number of cells in the S and G2/M phases declined (21.6 vs. 24.8% and 15.0 vs. 23.5% of control, respectively). These results demonstrated that uPAg-KPI inhibited SKOV-3 cell proliferation (Fig. 1B and C).

Effects of uPAg-KPI on the inhibition of tumor cell colony formation

Considering uPAg-KPI has the propensity to affect cell proliferation in vitro, a plate colony formation assay was performed. The efficiency of plate colony formation indicated that there was a significant difference between cells treated with 0.5 µg/µl of uPAg-KPI compared to untreated control cells. The number of colonies after treatment was notably fewer compared to the controls (Fig. 2). The result, to an extent, further confirmed that treatment with uPAg-KPI reduced the proliferation of ovarian cancer cells.

Effects of uPAg-KPI on the inhibition of SKOV-3 cell migration and invasive capacity

We analyzed the effects of uPAg-KPI on the migration and invasive capacity of SKOV-3 cells using the previously mentioned wound healing assay. As shown in Fig. 3, relative wound closure was decreased following treatment with uPAg-KPI (0.5 µg/µl) for 24 or 48 h compared to the controls.

Following this, we performed Transwell tumor cell migration and invasion assays and demonstrated that uPAg-KPI had a significant inhibitory effect on cell migration and invasion at the concentration 0.5 µg/µl (Fig. 4).

Effects of uPAg-KPI on the regulation of ERK1/2 and AKT activity in SKOV-3 cells

After establishing the ability of uPAg-KPI to inhibit cell migration, we assessed the underlying molecular mechanisms of uPAg-KPI. Using western blot analysis, we demonstrated that treatment with uPAg-KPI suppressed p-ERK and p-AKT activity. However, no marked effects on the total levels of ERK1/2 and AKT proteins were noted (Fig. 5).

Discussion

Ovarian cancer is a highly metastatic type of cancer, and identification of the underlying molecular signaling pathways will help effectively control disease progression (18). Generally, degradation of the extracellular matrix is considered to be the first step of tumor invasion or metastasis. Activation of the uPA/uPAR/plasmin proteolytic signaling network plays a key role in extracellular matrix degradation. Previous studies using natural and synthetic uPA inhibitors have demonstrated abrogated tumor cell growth, invasion and metastasis due to inhibition of uPA serine protease activities (19,20) and the ability of certain uPA antagonists (2123) to block binding to uPAR on the tumor cell surface.

In our previous study, we constructed uPAg-KPI, which contained uPA GFD conjugated with a kunitz protease inhibitor (KPI) domain that is present in the amyloid beta-protein precursor (APP). Our previous data demonstrated that this fusion protein inhibited serine protease activity in vitro (15). Thus, in the present study, we assessed the effects of this fusion protein uPAg-KPI on the regulation of ovarian cancer cell phenotypes and protein expression. We found that uPAg-KPI treatment reduced the viability of ovarian cancer cells in a concentration and time-dependent manner and arrested tumor cells at the G1/G0 phase of the cell cycle. The IC50 of uPAg-KPI was 0.5 µg/µl after 48 h treatment, and treatment of ovarian cancer cells with this concentration inhibited tumor cell colony formation, wound healing, and tumor cell migration and invasive capacity. Molecularly, uPAg-KPI suppressed the activity of ERK1/2 and AKT proteins. Thus, we demonstrated that this novel uPAg-KPI fusion protein exerted antitumor activity in ovarian cancer SKOV-3 cells in vitro by regulation of ERK and AKT signaling.

uPA was originally isolated from human urine and is present in the bloodstream and the extracellular matrix (24). The primary physiological substrate of uPA is plasminogen, and activation of plasmin triggers a proteolytic cascade to promote thrombolysis or extracellular matrix degradation. Altered expression or altered activity of uPA is linked to a variety of vascular diseases and cancers (25,26). Extracellular matrix degradation, following plasminogen activation has been shown to induce tumor cell tissue invasion and metastasis, whereas inhibition of uPA activity or expression has been used as an anticancer agent (27,28). Indeed, Mesupron®, a small molecule serine protease inhibitor developed by WILEX, has been used in clinical trials (http://www.wilex.de/portfolio/mesupron/phase-i-ii-mit-mesupron/). Studies have suggested that the drug appears to be safe when combined with chemotherapy in cases of breast cancer (http://www.wilex.de/portfolio/mesupron/phase-i-ii-mit-mesupron/).

In the present study, we found that the fusion protein uPAg-KPI not only demonstrated the ability to inhibit tumor cell growth, but also inhibited tumor cell invasion and metastasis. It is envisioned that futire in vivo studies will assess the effectiveness of this fusion protein uPAg-KPI in animals before clinical trials. However, the uPA signal transduction pathway is complex, and there is a plethora of combining pathways. For example, previous studies have shown that the uPA/uPAR signaling cascade may be at the intersection of multiple tumor invasion and metastasis-related signaling molecules or pathways (2932). In addition to activating extracellular matrix degradation, the uPA/uPAR system also activates Src, Raf, FAK, ERK or MAPK signaling pathways, which play an important role in tumor progression (3335). With respect to the induction of tumor cell proliferation, previous studies have shown that uPA induced a cascade of several cell proliferation signaling pathways, such as the signal transducer and activator of transcription (Stat3) pathway, ERK1/2 pathway and the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway (3639). In order to investigate the possible mechanisms by which uPAg-KPI induced cell growth arrest and inhibition of tumor cell invasion, the present study detected the level of ERK, p-ERK, AKT and p-AKT proteins and found that uPAg-KPI suppressed the expression of phosphorylated ERK1/ERK2 and AKT. These two pathways have previously been shown to regulate cell growth and invasion (40,41). Thus, the data obtained from the present study suggest that uPAg-KPI binds to membrane-anchored uPAR and restrains plasminogen activation on the tumor cell surface. This blocks the ERK and AKT signaling pathways and thus significantly decreases tumor growth and invasion. However, further investigation is required in order to elucidate how exactly uPAg-KPI suppresses phosphorylation and the activity of ERK1/ERK2 and AKT proteins.

Acknowledgments

This study was supported in part by grants from the National Natural Science Foundation of China (nos. 81302242 and 81272875), the Jilin Provincial Science and Technology Funds (nos. 20150204007YY, 20130102094JC, 20140204022YY, 20150204041YY and 20130727039YY), the Jilin provincial development and Reform Commission Funds (no. 2013C026-3).

References

1 

Stewart BW and Kleihues P: World Cancer Report. IARC press; Lyon: 2003

2 

Mao Y, Xu J, Li Z, Zhang N, Yin H and Liu Z: The role of nuclear β-catenin accumulation in the Twist2-induced ovarian cancer EMT. PLoS One. 8:e782002013. View Article : Google Scholar

3 

Fazioli F and Blasi F: Urokinase-type plasminogen activator and its receptor: new targets for anti-metastatic therapy? Trends Pharmacol Sci. 15:25–29. 1994. View Article : Google Scholar : PubMed/NCBI

4 

Mekkawy AH, Pourgholami MH and Morris DL: Involvement of urokinase-type plasminogen activator system in cancer: an overview. Med Res Rev. 34:918–956. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Andreasen PA, Kjøller L, Christensen L and Duffy MJ: The urokinase-type plasminogen activator system in cancer metastasis: a review. Int J Cancer. 72:1–22. 1997. View Article : Google Scholar : PubMed/NCBI

6 

Kwaan HC: The plasminogen-plasmin system in malignancy. Cancer Metastasis Rev. 11:291–311. 1992. View Article : Google Scholar : PubMed/NCBI

7 

Berkenblit A, Matulonis UA, Kroener JF, Dezube BJ, Lam GN, Cuasay LC, Brünner N, Jones TR, Silverman MH and Gold MA: A6, a urokinase plasminogen activator (uPA)-derived peptide in patients with advanced gynecologic cancer: a phase I trial. Gynecol Oncol. 99:50–57. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Ghamande SA, Silverman MH, Huh W, Behbakht K, Ball G, Cuasay L, Würtz SO, Brunner N and Gold MA: A phase 2, randomized, double-blind, placebo-controlled trial of clinical activity and safety of subcutaneous A6 in women with asymptomatic CA125 progression after first-line chemotherapy of epithelial ovarian cancer. Gynecol Oncol. 111:89–94. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Gold MA, Brady WE, Lankes HA, Rose PG, Kelley JL, De Geest K, Crispens MA, Resnick KE and Howell SB: A phase II study of a urokinase-derived peptide (A6) in the treatment of persistent or recurrent epithelial ovarian, fallopian tube, or primary peritoneal carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol. 125:635–639. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Heinemann V, Ebert MP, Laubender RP, Bevan P, Mala C and Boeck S: Phase II randomised proof-of-concept study of the urokinase inhibitor upamostat (WX-671) in combination with gemcitabine compared with gemcitabine alone in patients with non-resectable, locally advanced pancreatic cancer. Br J Cancer. 108:766–770. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Chen H, Hao J, Wang L and Li Y: Coexpression of invasive markers (uPA, CD44) and multiple drug-resistance proteins (MDR1, MRP2) is correlated with epithelial ovarian cancer progression. Br J Cancer. 101:432–440. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Wang L, Madigan MC, Chen H, Liu F, Patterson KI, Beretov J, O'Brien PM and Li Y: Expression of urokinase plasminogen activator and its receptor in advanced epithelial ovarian cancer patients. Gynecol Oncol. 114:265–272. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Dorn J, Harbeck N, Kates R, Gkazepis A, Scorilas A, Soos-aipillai A, Diamandis E, Kiechle M, Schmalfeldt B and Schmitt M: Impact of expression differences of kallikrein-related peptidases and of uPA and PAI-1 between primary tumor and omentum metastasis in advanced ovarian cancer. Ann Oncol. 22:877–883. 2011. View Article : Google Scholar

14 

Salameh MA, Robinson JL, Navaneetham D, Sinha D, Madden BJ, Walsh PN and Radisky ES: The amyloid precursor protein/protease nexin 2 Kunitz inhibitor domain is a highly specific substrate of mesotrypsin. J Biol Chem. 285:1939–1949. 2010. View Article : Google Scholar :

15 

Zhao L and Yan W: Expression and identification of recombinant humanuPA17-KPI in Pichia pastoris. Chin J Lab Diagn. 14:1685–1688. 2010.

16 

Lai KC, Huang AC, Hsu SC, Kuo CL, Yang JS, Wu SH and Chung JG: Benzyl isothiocyanate (BITC) inhibits migration and invasion of human colon cancer HT29 cells by inhibiting matrix metalloproteinase-2/-9 and urokinase plasminogen (uPA) through PKC and MAPK signaling pathway. J Agric Food Chem. 58:2935–2942. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Lin Y, Cui M, Xu T, Yu W and Zhang L: Silencing of cyclooxy-genase-2 inhibits the growth, invasion and migration of ovarian cancer cells. Mol Med Rep. 9:2499–2504. 2014.PubMed/NCBI

18 

Vergara D, Merlot B, Lucot JP, Collinet P, Vinatier D, Fournier I and Salzet M: Epithelial-mesenchymal transition in ovarian cancer. Cancer Lett. 291:59–66. 2010. View Article : Google Scholar

19 

Evans DM and Lin PL: Suppression of pulmonary metastases of rat mammary cancer by recombinant urokinase plasminogen activator inhibitor. Am Surg. 61:692–696; discussion 696–697. 1995.PubMed/NCBI

20 

Praus M, Wauterickx K, Collen D and Gerard RD: Reduction of tumor cell migration and metastasis by adenoviral gene transfer of plasminogen activator inhibitors. Gene Ther. 6:227–236. 1999. View Article : Google Scholar : PubMed/NCBI

21 

Guo Y, Higazi AA, Arakelian A, Sachais BS, Cines D, Goldfarb RH, Jones TR, Kwaan H, Mazar AP and Rabbani SA: A peptide derived from the nonreceptor binding region of urokinase plasminogen activator (uPA) inhibits tumor progression and angiogenesis and induces tumor cell death in vivo. FASEB J. 14:1400–1410. 2000. View Article : Google Scholar : PubMed/NCBI

22 

Guo Y, Mazar AP, Lebrun JJ and Rabbani SA: An antiangiogenic urokinase-derived peptide combined with tamoxifen decreases tumor growth and metastasis in a syngeneic model of breast cancer. Cancer Res. 62:4678–4684. 2002.PubMed/NCBI

23 

Ploug M, Østergaard S, Gårdsvoll H, Kovalski K, Holst-Hansen C, Holm A, Ossowski L and Danø K: Peptide-derived antagonists of the urokinase receptor. Affinity maturation by combinatorial chemistry, identification of functional epitopes, and inhibitory effect on cancer cell intravasation. Biochemistry. 40:12157–12168. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Zaravinos A: The regulatory role of MicroRNAs in EMT and cancer. J Oncol. 2015(865816)2015. View Article : Google Scholar : PubMed/NCBI

25 

Chandrasekar N, Mohanam S, Gujrati M, Olivero WC, Dinh DH and Rao JS: Downregulation of uPA inhibits migration and PI3k/Akt signaling in glioblastoma cells. Oncogene. 22:392–400. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Wang Q, Wang Y, Zhang Y, Zhang Y and Xiao W: The role of uPAR in epithelial-mesenchymal transition in small airway epithelium of patients with chronic obstructive pulmonary disease. Respir Res. 14(67)2013. View Article : Google Scholar

27 

Jankun J and Skrzypczak-Jankun E: Molecular basis of specific inhibition of urokinase plasminogen activator by amiloride. Cancer Biochem Biophys. 17:109–123. 1999.

28 

Matthews H, Ranson M and Kelso MJ: Anti-tumour/metastasis effects of the potassium-sparing diuretic amiloride: an orally active anti-cancer drug waiting for its call-of-duty? Int J Cancer. 129:2051–2061. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Lester RD, Jo M, Montel V, Takimoto S and Gonias SL: uPAR induces epithelial-mesenchymal transition in hypoxic breast cancer cells. J Cell Biol. 178:425–436. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Liu SQ, Huang JA, Qin MB, Su YJ, Lai MY, Jiang HX and Tang GD: Sphingosine kinase 1 enhances colon cancer cell proliferation and invasion by upregulating the production of MMP-2/9 and uPA via MAPK pathways. Int J Colorectal Dis. 27:1569–1578. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Chou RH, Hsieh SC, Yu YL, Huang MH, Huang YC and Hsieh YH: Fisetin inhibits migration and invasion of human cervical cancer cells by down-regulating urokinase plasminogen activator expression through suppressing the p38 MAPK-dependent NF-κB signaling pathway. PLoS One. 8:e719832013. View Article : Google Scholar

32 

Jo M, Lester RD, Montel V, Eastman B, Takimoto S and Gonias SL: Reversibility of epithelial-mesenchymal transition (EMT) induced in breast cancer cells by activation of urokinase receptor-dependent cell signaling. J Biol Chem. 284:22825–22833. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Blasi F and Sidenius N: The urokinase receptor: focused cell surface proteolysis, cell adhesion and signaling. FEBS Lett. 584:1923–1930. 2010. View Article : Google Scholar

34 

Eastman BM, Jo M, Webb DL, Takimoto S and Gonias SL: A transformation in the mechanism by which the urokinase receptor signals provides a selection advantage for estrogen receptor-expressing breast cancer cells in the absence of estrogen. Cell Signal. 24:1847–1855. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Nowicki TS, Zhao H, Darzynkiewicz Z, Moscatello A, Shin E, Schantz S, Tiwari RK and Geliebter J: Downregulation of uPAR inhibits migration, invasion, proliferation, FAK/PI3K/Akt signaling and induces senescence in papillary thyroid carcinoma cells. Cell Cycle. 10:100–107. 2011. View Article : Google Scholar :

36 

Gogineni VR, Gupta R, Nalla AK, Velpula KK and Rao JS: uPAR and cathepsin B shRNA impedes TGF-β1-driven proliferation and invasion of meningioma cells in a XIAP-dependent pathway. Cell Death Dis. 3:e4392012. View Article : Google Scholar

37 

Shetty S, Rao GN, Cines DB and Bdeir K: Urokinase induces activation of STAT3 in lung epithelial cells. Am J Physiol Lung Cell Mol Physiol. 291:L772–L780. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Stewart AG, Xia YC, Harris T, Royce S, Hamilton JA and Schuliga M: Plasminogen-stimulated airway smooth muscle cell proliferation is mediated by urokinase and annexin A2, involving plasmin-activated cell signalling. Br J Pharmacol. 170:1421–1435. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Ulisse S, Baldini E, Sorrenti S and D'Armiento M: The urokinase plasminogen activator system: a target for anti-cancer therapy. Curr Cancer Drug Targets. 9:32–71. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Santarpia L, Lippman SM and El-Naggar AK: Targeting the MAPK-RAS-RAF signaling pathway in cancer therapy. Expert Opin Ther Targets. 16:103–119. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Romano G: The role of the dysfunctional akt-related pathway in cancer: establishment and maintenance of a malignant cell phenotype, resistance to therapy, and future strategies for drug development. Scientifica (Cairo). 2013:3171862013.

Related Articles

Journal Cover

May-2016
Volume 37 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao L, Xu T, Kan M, Xiao Y and Cui M: A novel uPAg-KPI fusion protein inhibits the growth and invasion of human ovarian cancer cells in vitro. Int J Mol Med 37: 1310-1316, 2016
APA
Zhao, L., Xu, T., Kan, M., Xiao, Y., & Cui, M. (2016). A novel uPAg-KPI fusion protein inhibits the growth and invasion of human ovarian cancer cells in vitro. International Journal of Molecular Medicine, 37, 1310-1316. https://doi.org/10.3892/ijmm.2016.2540
MLA
Zhao, L., Xu, T., Kan, M., Xiao, Y., Cui, M."A novel uPAg-KPI fusion protein inhibits the growth and invasion of human ovarian cancer cells in vitro". International Journal of Molecular Medicine 37.5 (2016): 1310-1316.
Chicago
Zhao, L., Xu, T., Kan, M., Xiao, Y., Cui, M."A novel uPAg-KPI fusion protein inhibits the growth and invasion of human ovarian cancer cells in vitro". International Journal of Molecular Medicine 37, no. 5 (2016): 1310-1316. https://doi.org/10.3892/ijmm.2016.2540