Journal Search Engine
Search Advanced Search Adode Reader(link)
Download PDF Export Citaion korean bibliography PMC previewer
ISSN : 1225-1577(Print)
ISSN : 2384-0900(Online)
The Korean Journal of Oral and Maxillofacial Pathology Vol.44 No.1 pp.9-18
DOI : https://doi.org/10.17779/KAOMP.2020.44.1.002

RNAi Transfection Effect of the uPAR and uPA on Salivary Gland Tumor Cell Line

Min Koo Oh, Chong Heon Lee*
Department of Oral Pathology, College of Dentistry, Oral Aging Research Center, Dankook University
Correspondence: Chong Heon Lee, Department of Oral Pathology, Dental College, Dankook University, Aseodong San 29, Cheonan, Chungnam, 330-714, Korea Tel: +82-41-550-1946 E-mail: chleeop@naver.com
January 6, 2020 January 17, 2020 February 7, 2020

Abstract


Despite existing chemotherapy and surgical resection strategies, salivary gland adenocarcinoma(AdCa NOS) is one of the major causes of mortality among malignant salivary gland tumors. New therapeutic measure are needed to improve the outcome for patients with AdCa. Overexpression of urokinase-type plasminogen activator receptor/urokinase-type plasminogen activator(uPAR-uPA) has been implicated in progression and metastasis of oral cancer. RNA interference(RNAi) which has emerged as an effective method to target specific genes for silencing has provided new opportunities for cancer therapy. But there has been rarely reported using RNAi-uPAR/uPA transfection in salivary gland AdCa. The purpose of this study were to examine the specific inhibition of uPAR/uPA mRNA and protein expression by RNAi transfection of uPAR/uPA through RT-PCR and Immunoslot blot, and to study tumor cell proliferation activity, adhesion, invasion and migration of SGT cell line in vitro compared to the controls. In adhesion assay, cells transfected with RNAi-uPAR/uPA inhibited markedly adhesion to vitronectin compared to parental cells. Angiogenic assays revealed a significant decrease in the angiogenic potential of SGT cells downregulated by both uPAR and uPA. In migration assay, suppressing uPAR and uPA inhibited the capacity of the cells to migrate compared to parental cells. In invasion assay, cells transfected with RNAi-uPAR/uPA showed the maximum decrease in invasion when compared to all other treatment conditions. RNAi expressing plasmids efficiently downregulated mRNA and protein expression of uPAR and uPA. Cell cycle analysis showed that the simultaneous downregulation of uPAR and uPA caused the accumulation of cells in the sub-G0/G1 phase in SGT cells. Immunoslot blot analysis revealed that downregulation of uPAR and uPA caused the prominent activation of caspase 8. It suggested that the RNAi targeting of the uPAR/uPA system could have a therapeutic potentiality for malignant salivary gland tumors.



uPAR 및 uPA의 RNAi 형질감염이 타액선 종양세포주에 미치는 영향

오 민구, 이 종헌*
단국대학교 치과대학 구강병리학교실, 구강노화연구소

초록


    Ⅰ. INTRODUCTION

    Adenocarcinoma NOS(AdCa NOS) is the third most common malignant tumor of the salivary glands which is characterized by unique clinical features and behavior, including a relatively unfavorable prognosis with high frequencies of recurrence and distant metastasis1-2). Despite advances in diagnosis and therapy, long-term survival of AdCa patients has only moderately improved during the past 20 years. Although the standard treatment for major malignant salivary gland tumor is surgical resection, a substantial risk of localregional recurrence and distant metastasis exists36) because of the ability of malignant salivary gland tumor cells to invade surrounding tissues.

    Although the molecular mechanisms underlying metastasis are complex, it is clear that migration of tumor cell can invade local tissues and metastasize to distal sites through ECM. Migrating cells use both adhesion molecules and proteolytic enzymes to regulate their interaction with and response to the ECM. One protease system intimately connected to integrins is the urokinase-type plasminogen activator(uPA)/uPA receptor(uPAR)/ plasmin system7) in inhibiting the characteristic feature of malignant tumor. Recently it is presented that the involvement of the uPA/uPAR system in chemotaxis and in the activation of intracellular signalling pathways lead to increased cellular proliferation, adhesion, and migration8-11). Therefore, uPAR/uPA system is a potential target for therapy in patients with malignant salivary gland tumor. Although adjuvant chemotherapy and radiation has been used in previous studies in order to prevent the local-regional and distant metastatic failure rate12-14), the high rate of survival rates has not been improved by these therapy.

    The recent discovery of RNA interference(RNAi), a more powerful tool for the inhibition of gene expression, has provided new opportunities for cancer therapy15-16). It was thought that RANi-uPAR/uPA could play an important role in the downregulation mechanisms to characteristic features of AdCa. Unfortunately previous studies about the transfection effect of RNAi-uPAR/uPA on AdCa have been rarely reported.

    The purpose of this study were to examine downregulation of uPAR/uPA expression through RNAi transfection of uPAR/uPA in SGT cell line and to investigate the inhibition of proliferation activity, adhesion, migration and invasion potentiality of SGT cell line in vitro.

    Ⅱ. MATERIALS AND METHODS

    1. Cell Culture, RNAi-expressing Plasmid Construction and Transfections

    SGT cells were grown in Dulbecco’s modified Eagle’'s medium (DMEM, Hyclone USA) supplemented with 1% glutamine, 10% fetal bovine serum(pH 7.2–7.4) in a humidified atmosphere containing 5% CO2 at 37°C. Construction of short hairpin RNA-expressing plasmids were constructed as well described17). Transfections were performed with Lipofectaminet 2000 reagent(Invitrogen, USA). using 1-2 mg of expression vector/ml serum-free medium as described by the manufacturer. After 5–6hrs of transfection, the medium was replaced by serumcontaining medium and incubated for a further 48hrs. SGT cells were transfected with plasmids expressing RNAi targeting uPAR(RNAi-uPAR), uPA (RNAi-uPA), and uPAR–uPA simultaneously (RNAi-uPAR/uPA).

    2. Cell Adhesion Assay

    A standard static adhesion assay was carried out using a 96-well microtiter plate coated with 10mg/ml of type IV collagen or BSA. A suspensions(2X104cells/well) of parental and RNAi-uPAR-, RNAi-uPA-, RNAi- uPAR/uPA-transfected cells, respectively, were added to the wells and allowed to adhere at 37°C for 60 min. The wells were washed three times with PBS at room temperature. Attached cells were fixed and stained with 0.2% crystal violet in 10% ethanol and lysed in solubilization buffer. The absorbance was measured at 590 nm with a microplate reader.

    4. Cell Migration Assay

    A modified Boyden chamber was used for the cell migration assay, as described previously with slight modifications. The chamber(Falcon, USA) consisted of upper and lower compartments separated by a polyethylene terephthalate track-etched filter (6.4mm diameter) with 8mm pores. The lower side of the filter was coated overnight with 10mg/ml of VN coating or BSA and then blocked with 3% BSA for 1h at room temperature. The lower compartment of the chamber was filled with 700ml of serum free medium containing 0.1% BSA and 10mg/ml of type IV collagen. A 200ml aliquot of suspended cells including parental and RNAi-uPAR-, RNAi-uPA-, RNAiuPAR/ uPA-transfected cells(1x105 cells in serum-free medium containing 0.1% BSA) was placed in the upper compartment of the chamber. After a 12hrs incubation at 37°C, cells that migrated to the lower side of the filter were quantified by light microscopy under a high power field(x200). For each of the triplicate experiments, cells in five randomly chosen fields were counted, and averaged.

    5. Cell Invasion Assay

    The in vitro invasiveness of SGT cells in the presence of the vector expressing siRNA for uPAR and uPA was assessed using a modified Boyden chamber assay. A cell invasion assay was conducted with QCM96-well cell invasion assay kit(Corning Costar Fisher Scientific, USA). This well invasion plate is based on the Boyden chamber principle. This plate contains 96 inserts; each containing an 8um pore size polycarbonate membrane coated with a layer of ECMatrix. Briefly, parental and RNAi-uPAR-, RNAi-uPA-, RNAi-uPAR/ uPA-transfected cells, respectively, were resuspended in serum free DMEM, and 5x104 cells were seeded into the ECM layer, which had been previously rehydrated at room temperature for 1–2hrs. 150μl DMEM containing 10% FBS was added to the lower chamber as chemoattractant. Cells were incubated for 24hrs at 37°C in 5% CO2 incubator. Invaded cells on the bottom of the insert membrane were dissociated from the membrane by incubation with cell detachment buffer and subsequently lysed and detected by a fluorescence plate leader. The fluorescence was quantified with a fluorescence plate reader using a 485/535-nm filter set.

    5. in vitro Angiogenic Assay

    SGT cells(2x106/well) were seeded and transfected with RNAi-uPAR, RNAi-uPA, or RNAi-uPAR/uPA. After a 24-hour incubation period, the conditioned medium was removed and added to a 4x104 human dermal endothelial cell in 8-well chamber slides. Cells were allowed to grow for 72 hours. Cells were then fixed in 3.7% formaldehyde and H&E stained to visualize capillary network formation. The degree of angiogenesis was quantified based on the numerical value for the product of the number of branches and number of branch points as an average of 10 fields. The results are graphically represented.

    6. Cell Cycle Analysis

    SGT cells (1x106/ml) were transfected with RNAi-uPAR, RNAi-uPA, or RNAi-uPAR/uPA for 48 hours. Cells were trypsinized, and fixed in chilled 70% ethanol for 12 h. After washed with PBS for 3x5 min, and resuspended in 200 ul PBS. RNase A(100 ul of 0.1% solution) was added to the cell suspensions followed by a 15-min incubation at 37°C. Cells were treated with 50 mg/mL propidium iodide(0.001% RNAse A solution for 15 min) as per standard protocol. The cells were sorted on a fluorescence-activated cell sorter and quantified (10,000 cells sorted per treatment condition with 3 replications).

    7. RT-PCR

    Total RNA was extracted from parental and RNAi-uPAR-, RNAi-uPA-, RNAi-uPAR/uPA-transfected cells with the TRIzol reagent(Life Technologies, USA) according to the manufacturer’'s instructions. To avoid DNA contamination, total RNA was treated with RNase-free DNase I(Takara, Japan) for 60min at 37°C, and extracted with the TRIzol reagent again. Takara Ex Taq DNA polymerase(Takara, Japan) were used for the PCR. Then the RNA was reverse transcribed at 42°C for 60min in a 20μl reaction volume using a First Strand cDNA synthesis kit(Clontech Lab, USA), according to the manufacturer’ instructions. cDNA was incubated at 95°C for five minutes to inactivate the reverse transcriptase, and used as template DNA in the PCR amplification of the uPAR region. The conditions comprised an initial denaturation step at 94°C for 1min, then 45 cycles at 94°C for 10s, 55°C for 30s and 72°C for 1min and finally a extension step at 72°C for 5min with 32P-dCTP labeling using PCR machine. As an internal control of each sample, Human β-actin gene was used for standardization, and the amplification was quantified in duplicate. The PCR primer and probes sequences used in this study was uPAR(sense : TTACCTCGAATGCATTTCCT, antisense : TTG CACAGCC TCTTACCATA), uPA(sense : TGCGTCCTGGTCG TGAGCGA, antisense : CAAGCGTGTCAGCGCT GTAG) and Human β-actin(sense : GTGGGGCGC CCCAGGCACCA, antisense : CTCCTTAATG TCACGCACGATTTC). Each PCR product was electrophoresed on an agarose gel to confirm that there was only one band with the expected size for the target gene. The quantification of mRNA expression was determined by densitometric analysis. All the PCR products were analyzed comparatively to the amount of human β-actin detected in the same mRNA sample. mRNA expression of uPAR was used to prepare a standard graph. Data were representative of three experiments. For each experiment, triplicate cultures were used; the results were the means of three independent experiments. The uPAR and uPA gene expression levels were evaluated semi-quantitatively.

    8. Immunoslot blot

    SGT cells were transfected with RNAi-uPA, RNAi-uPAR, or RNAi-uPAR/uPA for 72 hours. Then, cells were collected, and total cell lysates were prepared in 0.1M Tris-acetate(pH 7.5), 1mM EDTA, protease inhibitors. Protein amount of those extracts was quantified at 495nm with spectrophotometer, serially diluted in the range of 10μg to 1μg, and applied into slot chambers containing nitrocellulose membrane. Slot chambers were twice washed with TBS. The membrane was processed as the following western blotting methods. The membrane was treated with blocking serum, incubated with primary antibodies and secondary antibodies. The antibodies for the antigens were purchased from the following sources: are uPAR and uPA as per standard protocols(Santa Cruz Biotechnology), and cleaved caspase 8(Cell Signaling Technology) commercially available. This was developed by an enhanced chemino luscence(ECL) method, and examined by densitometer in triplicate. The protein concentrations were determined by Bio-Rad Protein Assay.

    Ⅲ. RESULTS

    In adhesion assay, cells transfected with RNAi-uPAR/uPA inhibited markedly adhesion to vitronectin compared to parental cells, and showed about 10% adhesion activity compared to parental cells, while cells transfected with RNAi-uPA showed about 30%, and cells transfected with RNAi-uPAR showed about 20%(Fig. 1).

    For invasion assay, no significant difference was observed in control cells. Quantitative analysis revealed that invasion decreased by 33% in SGT cells after RNAi-uPAR transfection. Cells transfected with RNAi-uPA showed similar decreases in invasion(31%). Cells transfected with RNAi-uPAR/uPA showed the maximum decrease in invasion(60%) when compared to all other treatment conditions (Fig. 2).

    In a migration assay, cells transfected with RNAi-uPAR/ uPA showed a significantly lower capability of cells to migrate to vitronectin as compared to parental cells(Fig. 3), and showed about 25% migration activity compared to parental cells(Fig. 3), while cells transfected with RNAi-uPA showed about 75%, and cells transfected with RNAi-uPAR showed about 60%. It suggested that suppressing uPAR and uPA could inhibit the capacity of the cells to migrate on vitronectin.

    Endothelial cells formed dense capillary-like structures when cultured with conditioned media from untransfected SGT cells, while endothelial cells grown in conditioned media of SGT cells transfected with RNAi-uPAR or RNAi-uPA showed a decrease in the density and completeness of the capillary network structure(broken and rudimentary network formation) as compared to the controls. Quantitative analysis indicated that transfection with RNAi-uPAR(46%) or RNAi-uPA(38%) decreased capillary network formation(controls considered as 100% angiogenic). Transfection with RNAi-uPAR/uPA(73%) caused the most significant decrease in endothelial cell network formation. It suggested that suppression of uPAR and uPA expression in SGT cells could retard their angiogenic potential in vitro.

    To verify whether the uPAR and uPA gene was silencing by vector construct expressing RNAi, uPAR and uPA mRNA expression of parental and RNAi-uPAR transfected cells were detected by RT-PCR. Using specific primers for uPAR, uPA and human β-actin in SGT cells, there was no reduction in the mRNA levels of uPAR. Cells transfected with RNAiuPAR and RNAi-uPA showed significantly reduced uPAR and uPA mRNA expression(Fig. 5,6), Figure 2A shows a 1/4 fold decrease in the mRNA levels of uPAR in RNAi-uPAR transfected cells and a significant(1/2 fold) decrease in the mRNA levels of uPA in RNAi-uPA, while 1/8 fold of uPAR mRNA level and 1/4 fold of uPA mRNA level in RNAi-uPAR/ uPA-transfected cells when compared to control cells, respectively. It suggested that the uPAR and uPA gene might be silenced by RNA inference.

    uPAR protein expression decreased by about 1/2 fold in RNAi-uPAR-transfected cells and about 1/5 fold in RNAiuPAR/ uPA-transfected cells as compared to the controls(Fig. 7,8). Cells transfected with RNAi-uPA had decreased about 0.4 fold uPA activity but about 1/4 fold in uPA expression levels in RNAi-uPAR/uPA-transfected cells(Fig. 2B). It suggested that plasmids expressing RNAi targeting uPAR and uPA could suppress uPAR and uPA protein levels in SGT cells.

    From the results of FACS analysis, simultaneous downregulation of uPAR and uPA caused a higher accumulation of cells in the sub-G0/G1 phase in SGT cells as compared to downregulation of either uPAR or uPA alone. Controls cells had very little accumulation of cells in the sub-G0/G1 phase(Fig. 3B), while quantitative analysis indicated that SGT cells transfected with RNAi-uPAR(35%) or RNAi-uPA(29%) had increased accumulation of cells in the sub-G0/G1 phase. Cell death in RNAi-uPAR/ uPA-transfected cells(57%) was found to be more than 50% (Fig. 4). SGT cells were transfected with RNAi expressing plasmids and proteins were isolated. Control cells did not have increased cleavage of caspase 8 by immunoslot blot(Fig. 9,10). In contrast, cells transfected with RNAi-uPAR, RNAi-uPA and RNAi-uPAR/uPA had increased caspase 8 cleavage (about 1.5, 2.5, 3.5 folds, respectively)(Fig. 9,10). It suggested that suppression of uPAR and uPA expression in SGT cells could cause the accumulation of cells in the sub-G0/1 phase, and activates caspase 8.

    Ⅳ. DISCUSSION

    Recent studies have suggested that the expression levels of urokinase-type plasminogen activator (uPA) and fibroblastic uPA receptor (uPAR) are correlated with invasiveness and metastasis of malignant tumor18,19). Malignant tumors have the capacity to degrade the extracellular matrix(ECM) by controlled proteolysis. One proteolytic system involved in these processes is the uPA system, which consists of uPA, the uPAR, and uPA inhibitors 1 and 2, plasminogen activator inhibitor (PAI) 1 and 220). By binding uPA, uPAR localizes proteolytic activity to the leading edge of cells, thereby facilitating cellular migration and penetration across tissue boundaries21-22). uPAR also binds directly to vitronectin and associates with integrins within the plasma membrane, which alters the strength of cellular adhesion10). There are 3 basic steps involved in invasion and intracellular signaling: (1) uPAR–uPA promotes extracellular proteolysis by regulating plasminogen activation, (2) uPAR–uPA regulates cell/ECM interactions as an adhesion receptor for vitronectin(Vn) and through its capacity to modulate integrin function, and (3) uPAR–uPA regulates cell migration as a signal transduction molecule and by its intrinsic chemotactic activity18,19).

    In the present study, we have attempted to retard the invasive ability and angiogenic potential of SGT cells in vitro. This study showed that both uPAR and uPA were highly expressed in SGT cells, and the expression of uPAR was always correlated with the expression of its ligand uPA. Researchers have shown that activation of the uPAR/uPA system is an early event in the development of cancer and that uPAR gene amplifications identify a subgroup of particularly aggressive tumors, thereby making the uPAR/uPA system a critical and highly promising target for therapeutic interventions23).

    To target the uPAR/uPA system, we used plasmids expressing shRNA targeting uPAR and uPA24,25-29). In this study, SGT cells transfected with RNAi-uPAR displayed markedly inhibited adhesion to vitronectin compared to control, while cells transfected with RNAi-uPAR compared to parental cells did not show difference in the ability to adhere to types I and IV collagen and laminin30). The direct function of uPAR might be to mediate the initial binding of cells to vitronectin as a matrix substrate, which was used for adhesion assay in this study. Thus, these results showed that inhibition of uPAR via RNA interference might also contribute to regulate the adhesion activity of SGT cells. From the Matrigel invasion assay, we observed that the simultaneous downregulation of uPAR and uPA caused a significant decrease in invasive potential as compared to cells downregulated for either uPAR or uPA. Recent research provide new functional evidence of the uPAR/uPA system's role in cancer invasion/ metastasis31). Researchers have also shown that inhibiting the activity of the uPAR and associated molecules like integrins32) causes a significant decrease of migration and intracellular signaling in cancer cells. The uPAR/uPA system is known to be localized at the invasive front of tumor cells, activating plasmin to plasminogen and further enabling the activation of various proteases33). Previous studies have shown that uPAR interacts with the extracellular domain of integrins allowing association with the cytoskeleton, thereby mediating cell adhesion and migration34,35). These findings confirmed that downregulation of uPAR via RNAi might decrease cell migration, possibly due to cytoskeletal alterations. Several reports indicated that expression of uPAR was essential components of the invasion process30,36,37). In the present study, RNAi-uPAR/uPA inhibited prominently the invasiveness of SGT cells.

    The classical role of the uPAR/uPA system is the activation of plasmin, and plasmin-overproducing cancer cells are also known to overexpress tPA, uPA, and uPAR. Plasmin is also known to promote angiogenesis and metastasis38). The mechanisms by which plasmin promotes angiogenesis and metastasis remain poorly understood. It was thought that suppressing the plasmin activating system could essentially inhibit angiogenic activation. Suppression of both uPAR and uPA retarded angiogenic activation under in vitro conditions, which meant that interference with the uPAR/uPA system retarded angiogenesis. And also simultaneous downregulation of uPAR and uPA could suppress angiogenin, MCP-1 and ENA-78 as a proangiogenic molecule, but upregulated the activity of RANTES in cancer cells39-42). It was presented that upregulation of RANTES and downregulation of MCP-1 play a role in boosting clinical antitumor immunity41). Quantitative analysis of neovasculature from the in vivo angiogenesis assay indicated that controls and control cells had similar increases in vasculature, whereas RNAi-uPAR treated cells and RNAiuPA- treated cells had decreased angiogenesis. RNAi-uPAR/ uPA treated cells exhibited little increase in vasculature.

    Interestingly, the downregulation of the uPAR/uPA system also caused the activation of proapoptotic molecules like caspase 8. From FACS analysis, we did observe the accumulation of cells in the sub-G0/G1 phase in cells downregulated for uPAR and uPA either alone or in combination. In immunoslot blotting, caspase 8 activation showed that cells simultaneously downregulated for uPAR and uPA had strong caspase 8 activation, indicating that both uPAR and uPA are necessary for maintenance of a viable invasive phenotype. Immunoslot blot analysis suggests this decrease may be due to the overall shut down of cellular processes and an indicator of apoptosis. The simultaneous downregulation of uPAR and uPA caused the activation of DNA damage as shown by TUNEL assay in vivo study43). The tumor tissues had decreased tumor cell density and fragmented nuclei, which are both indicative of apoptosis. By immunohistochemistry in vivo, uPA expression was seen uniformly throughout the tumor tissue with little expression in the large excretory ducts, whereas uPAR expression was observed in the tumor tissues as well as in the excretory ducts43).

    Taken together, it suggested that the targeting of the uPAR/uPA system has significant therapeutic potential for the treatment of AdCa. Because uPAR/uPA system may have certain unknown regulatory functions, further study to determine if this has any regulatory function is recommended.

    Figure

    KAOMP-44-1-9_F1.gif

    Adhesion Assay of Control, RNAi-uPAR, RNAi-uPA and RNAi-uPAR/uPA to Collagen Ⅳ

    KAOMP-44-1-9_F2.gif

    Invasion Assay of Control, RNAi-uPAR, RNAi-uPA and RNAi-uPAR/uPA

    KAOMP-44-1-9_F3.gif

    Migration Assy of Control, RNAi-uPAR, RNAi-uPA and RNAi-uPAR/uPA to Vitronectin

    KAOMP-44-1-9_F4.gif

    Cell Cycle Analysis of Control, RNAi-uPAR, RNAi-uPA and RNAi-uPAR/uPA by Flow Cytometry

    KAOMP-44-1-9_F5.gif

    uPAR m RNA expression in Control, RNAi-uPAR, RNAi-uPA and RNAi-uPAR/uPA

    KAOMP-44-1-9_F6.gif

    uPAR and uPA mRNA expression Measurement in Control, RNAi-uPAR, RNAi-uPA and RNAi-uPAR/uPA by Densitometer

    KAOMP-44-1-9_F7.gif

    uPAR and uPA Protein Concentration of Control, RNAi-uPAR, RNAi-uPA and RNAi-uPAR/uPA by Immunoslot blotting

    KAOMP-44-1-9_F8.gif

    uPAR and uPA Protein Concentration of Control, RNAiuPAR, RNAi-uPA and RNAi-uPAR/uPA by Immunoslot blotting with Densitometer

    KAOMP-44-1-9_F9.gif

    Caspase 8 Protein Concentration of Control, RNAi-uPAR, RNAi-uPA and RNAi-uPAR/uPA by Immunoslot blotting

    KAOMP-44-1-9_F10.gif

    Caspase 8 Protein Concentration of Control, RNAi-uPAR, RNAi-uPA and RNAi-uPAR/uPA by Immunoslot blotting with Densitometer

    Table

    Reference

    1. Ellis GL, Auclair PL, Gnepp DR: Surgical pathology of the salivary glands. Philadelphia, WB Saunders CO., 1991, p129- 134, p 318-332.
    2. Neville BW: Oral and Maxillofacial Pathology. Saunders CO., 2009, p498-499.
    3. Barnes L: Surgical Pathology of the Head and Neck. Marcel Dekker Inc., 1986. p533-646.
    4. Spiro RH: Salivary neoplasms: overview of a 35-year experience with 2807 patients. Head Neck Surg 1986;8:177–184.
    5. Spiro RH, Armstrong J, Strong EW: Carcinoma of major salivary glands. Recent trends. Arch Otolaryngol Head Neck Surg 1989;115:316–321.
    6. Kane WJ, McCaffrey TV, Lewis JE: Primary parotid malignancies. A clinical and pathologic review. Arch Otolaryngol Head Neck Surg 1991;117:307–315.
    7. Sidenius N, Blasi F: The uPA system in cancer: Recent advances and implication for prognosis and therapy. Cancer Metastasis Rev 2003;22:205-222.
    8. Jo M, Thomas KS, Somlyo AV: Cooperativity between the Ras-ERK and Rho-Rho kinase pathways in urokinase-type plasminogen activator- stimulated cell migration. J Biol Chem 2002;277:12479–12485.
    9. Preissner KT, May AE: Urokinase receptor: a molecular organizer in cellular communication. Curr Opin Cell Biol 2000;12:621 -628.
    10. Blasi F, Carmeliet P: uPAR: a versatile signalling orchestrator. Nat Rev Mol Cell Biol 2002;3:932–943.
    11. Pillay V, Dass CR, Choong PF: The urokinase plasminogen activator receptor as a gene therapy target for cancer. Trends Biotechnol 2007;25:33-39.
    12. North CA, Lee DJ, Johns ME: Carcinoma of the major salivary glands treated by surgery or surgery plus postoperative radiotherapy. Int J Radiat Oncol Biol Phys 1990;18:1319– 1326.
    13. Lee BH, Lee CH: Manumycin A Induced apoptotic effect on SGT cell line by caspase activation. Kor J Oral Maxiilofac 2007;31:327-336.
    14. Hocwald E, Korkmaz H, Jacobs JR: Prognostic factors in major salivary gland cancer. Laryngoscope 2001;111:1434– 1439.
    15. Elbashir SM, Harborth J, Weber K, Tuschl T: Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001;24411:494-498.
    16. Westhof E, Filipowicz W: From RNAi to epigenomes: how RNA rules the world. Chem Biochem 2005;6:441–443.
    17. Kiyan J, Kiyan R, Haller H, Dumler I: Urokinase-induced signaling in human vascular smooth muscle cells is mediated by PDGFR-beta. EMBO J 2005;24:1787–1797.
    18. Sidenius N, Blasi F: The urokinase plasminogen activator system in cancer: recent advances and implication for prognosis and therapy. Cancer Metastasis Rev 2003;22:205–222.
    19. Kugler MC, Wei Y, Chapman HA: Urokinase receptor and integrin interactions. Curr Pharm Des 2003;9:1565–1574.
    20. Rao JS. Molecular mechanisms of glioma invasiveness: the role of proteases. Nat Rev Cancer 2003;3:489–501.
    21. Nguyen DHD, Webb DJ, Catling AD: uPA stimulates the Ras/extracellular signal-regulated kinase(ERK) signaling pathway and MCF-7 cell migration by a mechanism that requires focal adhesion kinase, Src, and Shc. J Biol Chem 2000;275:19382- 19388.
    22. Chapman HA, Wei Y: Protease crosstalk with integrins: the urokinase receptor paradigm. Thromb Haemost 2001;86:124 –129.
    23. Hildenbrand R, Marx A, Belharazem D, Schleger C: Amplification of the urokinase-type plasminogen activator receptor(uPAR) gene in ductal pancreatic carcinomas identifies a clinically high-risk group. Am J Pathol 2009;174:2246–2253.
    24. Gondi CS, Lakka SS, Gujrati M, Rao JS: Intraperitoneal injection of an hpRNA-expressing plasmid targeting uPAR and uPA retards angiogenesis and inhibits intracranial tumor growth in nude mice. Clin Cancer Res 2007;13:4051–4060.
    25. Gondi CS, Rao JS: Therapeutic potential of siRNA-mediated targeting of urokinase plasminogen activator, its receptor, and matrix metalloproteinases. Methods Mol Biol 2009;487: 267–281.
    26. Kargiotis O, Chetty C, Pulukuri SM, Kyritsis AP: uPA/uPAR downregulation inhibits radiation-induced migration, invasion and angiogenesis in IOMM-Lee meningioma cells and decreases tumor growth in vivo. Int J Oncol 2008;33:937–947.
    27. Kondraganti S, Gondi CS, Gujrati M, Rao JS: RNAi-mediated downregulation of urokinase plasminogen activator and its receptor in human meningioma cells inhibits tumor invasion and growth. Int J Oncol 2006;28:1353–1360.
    28. Kunigal S, Gondi CS, Estes N, Rao JS: RNAi-mediated downregulation of urokinase plasminogen activator receptor and matrix metalloprotease-9 in human breast cancer cells results in decreased tumor invasion, angiogenesis and growth. Int J Cancer 2007;121:2307–2316.
    29. Subramanian R, Gondi CS, Lakka SS, Rao JS. siRNAmediated simultaneous downregulation of uPA and its receptor inhibits angiogenesis and invasiveness triggering apoptosis in breast cancer cells. Int J Oncol 2006;28:831–839.
    30. Xinhua Liang, Xiaoqin Yang, Lin Xiao, Zuyi Mao: RNAimediated downregulation of urokinase plasminogen activator receptor inhibits proliferation, adhesion, migration and invasion in oral cancer cells. Oral Oncology 2008;44:1172– 1180.
    31. Nowicki TS, Kummer NT, Schaefer S, Schantz S: Inhibition of uPAR and uPA reduces invasion in papillary thyroid carcinoma cells. Laryngoscope 2010;120:1383–90.
    32. Duriseti KS, Goetz DH, Wei Y, Craik CS: Antagonstic antiupar antibodies significantly inhibit uPAR-mediated cellular signaling and migration. J Biol Chem 2010. [Epub ahead of print].
    33. Andreasen PA, Egelund R, Petersen HH: The plasminogen activation system in tumor growth, invasion, and metastasis. Cell Mol Life Sci 2000;57:25–40.
    34. Planus E, Rogers RA, Ingber N, Wang N: Binding of urokinase to PAI-1 mediates cell adhesion and spreading. J Cell Sci 1997;110(Pt 9):1091–1098.
    35. Lindberg P, Larsson A, Nielsen BS: Expression of PAI-1, urokinase receptor and laminin gamma-2 chain is an early coordinated event in incipient oral squamous cell carcinoma. Int J Cancer 2006;118:2948–2956.
    36. Yamamoto E, Kohama G, Iwai M, Hiratsuka H: Mode of invasion, bleomycin sensitivity, and clinical course in squamous cell carcinoma of the oral cavity. Cancer 1983;51:2175–2180.
    37. Nozaki S, Endo Y, Kawashiri S, Yonemura Y: Immunohistochemical localization of a urokinase-type plasminogen activator system in squamous cell carcinoma of the oral cavity: association with mode of invasion and lymph node metastasis. Oral Oncol 1998;34:58–62.
    38. Sharma MR, Ownbey RT, Sharma MC: Angiogenesis-associated protein annexin II in breast cancer: selective expression in invasive breast cancer and contribution to tumor invasion and progression. Exp Mol Pathol 2006;81:146–156.
    39. Kim CK, Hong SH, Joe YA, Lee SK, Hong YK: The recombinant kringle domain of urokinase plasminogen activator inhibits in vivo malignant glioma growth. Cancer Sci 2007;98:253–258.
    40. Montuori N, Ragno P: Multiple activities of a multifaceted receptor: roles of cleaved and soluble uPAR. Front Biosci 2009;14:2494–503.
    41. Persson T, Bjartell A, Malm J, Calafat J: Expression of the neutrophil-activating CXC chemokine ENA-78/ CXCL5 by human eosinophils. Clin Exp Allergy 2003;33:531–537.
    42. Lapteva N, Huang XF: CCL5 as an adjuvant for cancer immunotherapy. Expert Opin Biol Ther 2010;10:725–33.
    43. Bharathi Gorantla, Swapna Asuthkar, Christopher S Gondi: Suppression of the uPAR–uPA System Retards Angiogenesis, Invasion, and In Vivo Tumor Development in Pancreatic Cancer Cells. Mol Cancer Res;9;377–89.
    오늘하루 팝업창 안보기 닫기