dipeptidyl peptidase inhibits malignant phenotype of prostate.pdf

Upload: alfox2000

Post on 14-Apr-2018

221 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/29/2019 Dipeptidyl Peptidase Inhibits Malignant Phenotype of Prostate.pdf

    1/11

    2005;65:1325-1334.Cancer ResUmadevi V. Wesley, Michelle McGroarty and Asal HomoyouniFactor Signaling PathwayProstate Cancer Cells by Blocking Basic Fibroblast GrowthDipeptidyl Peptidase Inhibits Malignant Phenotype of

    Updated version

    http://cancerres.aacrjournals.org/content/65/4/1325Access the most recent version of this article at:

    Cited Articles

    http://cancerres.aacrjournals.org/content/65/4/1325.full.html#ref-list-1This article cites by 45 articles, 14 of which you can access for free at:

    Citing articles

    http://cancerres.aacrjournals.org/content/65/4/1325.full.html#related-urlsThis article has been cited by 11 HighWire-hosted articles. Access the articles at:

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected] at

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications

    Permissions

    [email protected] at

    To request permission to re-use all or part of this article, contact the AACR Publications

    Research.on June 23, 2013. 2005 American Association for Cancercancerres.aacrjournals.orgDownloaded from

    http://cancerres.aacrjournals.org/content/65/4/1325http://cancerres.aacrjournals.org/content/65/4/1325http://cancerres.aacrjournals.org/content/65/4/1325.full.html#ref-list-1http://cancerres.aacrjournals.org/content/65/4/1325.full.html#ref-list-1http://cancerres.aacrjournals.org/content/65/4/1325.full.html#related-urlshttp://cancerres.aacrjournals.org/content/65/4/1325.full.html#related-urlshttp://cancerres.aacrjournals.org/cgi/alertshttp://cancerres.aacrjournals.org/cgi/alertsmailto:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/mailto:[email protected]:[email protected]://cancerres.aacrjournals.org/cgi/alertshttp://cancerres.aacrjournals.org/content/65/4/1325.full.html#related-urlshttp://cancerres.aacrjournals.org/content/65/4/1325.full.html#ref-list-1http://cancerres.aacrjournals.org/content/65/4/1325
  • 7/29/2019 Dipeptidyl Peptidase Inhibits Malignant Phenotype of Prostate.pdf

    2/11

    Dipeptidyl Peptidase Inhibits Malignant Phenotype of Prostate

    Cancer Cells by Blocking Basic Fibroblast Growth

    Factor Signaling Pathway

    Umadevi V. Wesley, Michelle McGroarty, and Asal Homoyouni

    Department of Microbiology and Molecular Genetics, Vermont Cancer Center, University of Vermont, Burlington, Vermont

    Abstract

    Dipeptidyl peptidase IV (DPPIV) is a serine protease with

    tumor suppressor function. It regulates the activities of

    mitogenic peptides implied in cancer development. Progres-

    sion of benign prostate cancer to malignant metastasis is

    linked to increased production of basic fibroblast growth

    factor (bFGF), a powerful mitogen. In this study, using in vitro

    model system we show that DPPIV loss is associated with

    increased bFGF production in metastatic prostate cancer cells.

    DPPIV reexpression in prostate cancer cells blocks nuclear

    localization of bFGF, reduces bFGF levels, inhibits mitogen-activated protein kinase (MAPK)-extracellular signal-regulated

    kinase (ERK)1/2 activation, and decreases levels of urokinase-

    type plasminogen activator, known downstream effectors of

    bFGF signaling pathway. These molecular changes were

    accompanied by induction of apoptosis, cell cycle arrest,

    inhibition of in vitro cell migration, and invasion. Silencing

    of DPPIV by small interfering RNA resulted in increased

    bFGF levels and restoration of mitogen-activated protein

    kinase (MAPK)-extracellular signal-regulated kinase (ERK)1/2

    activation. These results indicate that DPPIV inhibits the

    malignant phenotype of prostate cancer cells by blocking

    bFGF signaling pathway. (Cancer Res 2005; 65(4): 1325-34)

    Introduction

    Prostate cancer is the second leading causeof cancer deaths among

    men. Recent evidence suggest that the development of androgen-

    independent, metastatic prostate cancer is associated with increased

    activities of basic fibroblast growth factor (bFGF), a powerful mitogen

    and an angiogenic inducer (16). Although, the role of bFGF in cancer

    progression is fairly well understood, very little is known about the

    mechanisms that regulate bFGF signaling during the transition from

    benign to highly malignant prostate cancer.

    Dipeptidyl peptidase IV (DPPIV), a membrane glycoprotein,

    regulates the activities of mitogenic growth factors and neuro-

    peptides. Its proteolytic activity leads to inactivation or degrada-

    tion of these peptides (7, 8). DPPIV is involved in diverse biologicalprocesses, including cell differentiation, adhesion, immunomodu-

    lation, and apoptosis, functions that are critical for controlling

    neoplastic transformation (713). DPPIV is expressed in normal

    epithelial cells and melanocytes (1115). Its expression is lost in

    various cancers, suggesting loss of DPPIV as an important event

    during cancer progression (11, 12, 1418). We have previously

    shown that reexpression of DPPIV in melanoma and lung cancers

    leads to reemergence of dependence on exogenous growth factors

    and suppresses their tumorigenic potential (11, 12).

    bFGF, a multifunctional molecule regulates cell proliferation,

    migration, cell survival, wound healing, angiogenesis, and tumor

    progression (19, 20). It is expressed as a low molecular weight

    cytoplasmic isoform (18 kDa) and high molecular weight nuclear

    isoforms (21, 2.5, and 24 kDa). The high molecular weight isoforms

    possess the NH2-terminal nuclear localization signal and are

    exclusively localized to the nucleus (1921). Recent data indicate

    that increased expression of bFGF promotes tumorigenic and

    metastatic properties of various malignancies including prostate

    cancer and is associated with inferior survival of prostate cancer

    patients (2, 3, 19, 2126). Furthermore, loss of even one allele of

    bFGF in the transgenic adenocarcinoma of the mouse prostate

    model increases their survival by suppressing metastasis (27).

    Several studies have linked bFGF signaling to activation of the

    mitogen-activated protein kinase (MAPK)-extracellular signal-

    regulated kinase (ERK)1/2 pathway that promotes cancer progres-

    sion (2831). During cellular migration and angiogenesis, activation

    of ERK1/2 by bFGF increases the production of urokinase-type

    plasminogen activator (uPA), a serine protease that converts

    plasminogen to plasmin. Plasmin, in turn promotes cancer

    metastasis by breaking down the extracellular matrix, including

    fibronectin and laminin (32, 33). Notably, the malignant phenotypeof prostatic tumor cells correlates with high expression of both uPA

    and its receptor (uPAR), and plasma uPA has been identified as a

    biomarker for prostate cancer progression in Dunning rats and

    prostate cancer patients (3436).

    In this study, we investigated the mechanism by which DPPIV

    suppresses the malignant phenotype of prostate cancer cells. We

    establish a correlation between DPPIV loss, increased bFGF

    production, and malignant phenotype of prostate cancer cells.

    We show that reexpression of DPPIV in metastatic prostate cancer

    cells (DU-145), blocks nuclear accumulation of bFGF, decreases

    bFGF leading to inhibition of MAPK-ERK1/2 activation and uPA

    production, known downstream effectors of bFGF signaling

    pathway. We also show that DPPIV expression is associated withinhibition of in vitro cell migration, invasion, induction of

    apoptosis, and cell cycle arrest. These data identify a novel

    mechanism for regulation of bFGF activity by DPPIV in prostate

    cancer cells and provide new directions for therapeutic inter-

    ventions aimed at blocking the mitogenic activities of cancer

    promoting growth factors.

    Materials and Methods

    Antibodies and Reagents. The antibodies to bFGF, uPA, p27, and actin

    were obtained from Santa Cruz Biotechnology (Santa Cruz, CA); the

    polyclonal antibody against ERK1/2 from Cell Signaling Technologies

    Requests for reprints: Umadevi V. Wesley, Department of Microbiology andMolecular Genetics Vermont Cancer Center, University of Vermont, 320B, StaffordHall, Burlington, VT 05405. Phone: 802-652-2164; Fax: 802-656-8749; E-mail:[email protected] American Association for Cancer Research.

    www.aacrjournals.org 1325 Cancer Res 2005; 65: (4). February 15, 2005

    Research Article

    Research.on June 23, 2013. 2005 American Association for Cancercancerres.aacrjournals.orgDownloaded from

    http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/
  • 7/29/2019 Dipeptidyl Peptidase Inhibits Malignant Phenotype of Prostate.pdf

    3/11

    (Beverly, MA); CY-3 goat anti-rabbit (red) secondary antibody, horseradish

    peroxidase-sheep anti-mouse, goat anti-rabbit IgG were from Jackson

    laboratories (Bar Harbor, ME). The mouse monoclonal antibody to DPPIV

    was purified from the culture media of S27 hybridomas obtained from

    American Type Culture Collection (Manassas, VA). The cyanine dimer

    nucleic acid stain, YOYO-1 iodide, and Oregon Green were from Molecular

    probes, (Eugene, OR); LipofectAMINE reagent from Invitrogen, (Grand

    Island, NY); 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

    assay kit was from American Type Culture Collection. Cell invasion assay kit

    was from Chemicon International, Inc. (Temecula, CA); APOPTAG kit wasfrom Intergen (Gaithersburg, MD); and pSilencer small interfering RNA

    (siRNA) expression vectors were from Ambion, Inc. (Austin, TX).

    Cells. The normal prostate epithelial cells (NPrEC) were obtained from

    Cambrex Bio Science, Inc. (Baltimore, MD). RWPE-1, LNCaP, and DU-145

    cells were obtained from the American Type Culture Collection. DU-145

    and LNCaP cells were grown in RPMI 1640 supplemented with 10% fetal

    bovine serum, 0.1 mmol/L nonessential amino acids, 100 units/mL

    penicillin, and 0.1 mg/mL streptomycin. NPrEC cells were grown in media

    provided by the supplier. RWPE-1 cells were grown in keratinocyte

    medium containing bovine pituitary extract (0.05 mg/mL) and epidermal

    growth factor (0.5 ng/mL; Invitrogen).

    Flow Cytometry. Flow cytometric analysis was done using FACScan

    (Becton Dickinson, San Jose, CA). Cells were stained using the S27

    monoclonal antibody (anti-DPPIV) as the primary antibody. FITC-

    conjugated rabbit anti-mouse IgG was used as the secondary antibody.

    Cells stained with secondary antibody alone were used as controls.

    DPPIV Enzyme Activity. DPPIV peptidase activities were measured in

    presence and absence of doxycycline by colorimetric assay using Gly-Pro

    nitroanilide as the substrate, following previously described procedures (11).

    Total RNA Isolation and Northern Blot Analysis. Total RNAs were

    isolated from cells using RNAeasy extraction kit (Qiagen, Valencia, CA).

    Total RNA (20 Ag) from each cell line was size fractionated and transferred

    to nylon membrane. The mRNA levels of DPPIV, glyceraldehyde-3-

    phosphate dehydrogenase, uPA, and h-actin were assessed using the

    probes generated by PCR. The primers used in PCR reactions are as follows:

    5V-tcatatgacatttatgattta-3V and 5V-caaaatgaggaggcaagatcata-3V (800-bp

    DPPIV); 5V-atcttccaggagcgagatcc-3V and 5V-accactgacacgttggcagt-3V (502-bp

    glyceraldehyde-3-phosphate dehydrogenase); 5V-tcccggactatacagaccat-3V

    and 5V-tctcttccttggtgtgactg-3V (387-bp uPA); 5V-gggaaatcgtgcgtaacattaag-3Vand 5V-tgtgttggcgtacaggtctttg-3V (275-bp h-actin). Membranes were

    washed and visualiz ed by autoradio graphy.

    Establishment of Prostate Cancer Cell Line DU-145 Reexpressing

    DPPIV. Tetracycline inducible expression vectors carrying full-length

    DPPIV (pTRE-DPPIV) was constructed as described (11). DU-145 cell line

    was cotransfected at 60% confluency with pTRE-DPPIV or control vector,

    along with the plasmid pTET-on using LipofectAMINE. Stably transfected

    cells were selected using G418 (600 Ag/mL). To determine the growth rate,

    cells were plated at a density of 104 cells per well in triplicate in 24-well

    plates. Viable cells were counted daily for 10 days using trypan blue. Time of

    doubling was determined from a least-squares regression fit of cell number

    versus time during the logarithmic growth phase.

    Immunofluorescence Microscopy. For assessing bFGF, uPA, fibronec-

    tin, and ERK1/2 expression, cells grown on chamber slides for 48 hours,

    without or with doxycycline (1 Ag/mL) were stained with the respectivemonoclonal antibodies and incubated with either Oregon green or CY-3

    conjugated goat anti-rabbit (red) secondary antibody. Cells were counter

    stained with nuclear DNA-specific stain, Yo-Yo 1 iodide. Stained cells were

    viewed with a inverted microscope (model- ECLIPSE TE2000-U from Nikon,

    Melville, NY) connected to a RT Slider Spot Digital camera (Diagnostic

    Instruments, Sterling Heights, MI). Images were acquired and merged

    images were generated using the software SPOT version 3.2.

    Immunoprecipitation and Western Blot Analysis. For metabolic

    labeling of proteins, cells were cultured in medium containing [35S]

    methionine for 18 hours. Total cell lysates were immunoprecipitated with

    the DPPIV-specific monoclonal antibody S27. The protein samples were

    analyzed by SDS-PAGE and visualized by autoradiography. For Western

    blot analysis, equal amounts of protein lysates (50 Ag) were separated by

    SDS-PAGE, transferred to nitrocellulose membranes, and probed with the

    respective antibodies, over night at 4C. Signals were developed with

    chemiluminescence.

    MTT Cell Proliferation Assay. Colorimetric MTT assays were done

    using MTT assay kit on days 1, 2, 4, 6, 8, and 10. Cells (n = 5 103) per well

    were plated in triplicates on 96-well plates and cultured in absence or

    presence of doxycycline (1 Ag/mL). The absorbance of released purple

    formazan was measured at 570 nm in a microtiter plate reader. The

    experiment was repeated twice and the data are presented as mean F SD

    of triplicates.Wound Induced Cell Migration Assay and Quantitative Cell Invasion

    Assay. Cells were plated at 60% confluence and cell monolayers were

    wounded 24 hours after plating, by scraping with a sterile micropipette tip.

    Each well was then washed and daily fed with normal medium. Cells were

    photographed at 2 and 24 hours after wounding using a phase contrast

    microscope. Additionally, a quantitative cell invasion assay was done using

    an invasion assay kit following the manufacturers instructions. Absorbance

    values correlating with cell invasion were plotted. The experiment was

    repeated twice. The data are presented as the mean of triplicate plates.

    In situ Apoptotic Cell Detection by Terminal Deoxynucleotidyl

    TransferaseMediated Nick End Labeling Assay. For apoptosis assays,

    cells were grown (without or with doxycycline, 1 Ag/mL) in serum-free

    media for 3 days. Fixed cells were washed and measurements of DNA nicks

    were carried out by terminal deoxynucleotidyl transferasemediated nick

    end labeling assay using the APOPTAG kit. Percent apoptosis was

    calculated by FACScan analysis.

    Cell Cycle Analysis. Equal number of DU-145, vector, and DPPIV

    transfected DU-145 cells (2 106) were washed, resuspended in PBS

    containing 2% fetal bovine serum, 5 mmol/L EDTA, and were stained with

    20 mg/mL propidium iodide for 2 hours at room temperature. The cells

    were then examined by flow cytometry (Becton Dickinson FACS Scanner,

    Franklin Lakes, NJ) to determine cell cycle distribution. The percentage of

    cells present in G0-G1, S, and G2-M phases were determined using ModFit

    software package (Verity Software House, Topsham, ME).

    RNA Interference Assay. Expression of DPPIV was silenced using the

    vector, psilencer 3.1-H1 hygro. The 60-mer complementary oligonucleotides

    encoding 19-mer hairpin sequences specific to the DPPIV (NM001935)

    mRNA (nucleotide 1,219 to 1,237, AGTCATCGGGATAGAAGCT), a loop

    sequence (TTCAAGAGA) separating the two complementary domains, anda polythymidine tract (TTTTTT) to terminate transcription were synthe-

    sized and ligated into the psilencer 3.1-H1 hygro vector. Two sequential

    transfections (days 1 and 4) of LNCaP and DU-145 cells reexpressing DPPIV

    with 1 Ag of siRNA plasmid were carried out using the lipofectamine

    reagent. Stably transfected cells were selected for further analysis by adding

    hygromycin (200 Ag/mL) to culture media.

    Results

    DPPIV Loss Is Associated with Increased bFGF Production

    in Metastatic Prostate Cancer Cells (DU-145). We analyzed the

    expression pattern of DPPIV and bFGF in (a) NPrEC; (b)

    nontumorigenic in vitro transformed RWPE-1 cells (37); (c)

    androgen-sensitive, LNCaP cells that grow slow and are weaklytumorigenic (38, 39), and (d) androgen-independent, tumorigenic,

    and moderately metastatic prostate cancer cell line DU-145 (40).

    Flow cytometric analysis indicated high levels of cell surface

    expression of DPPIV on NPrEC cells with decreased expression to

    moderate levels on RWPE-1 and LNCaP cells, and very low levels on

    DU-145 cells (Fig. 1A). Whereas DPPIV enzyme activities in NPrEC

    cells ranged from 180 to 220 and 100 to 120 pmol per g protein per

    minute in RWPE-1 and LNCaP cells, it was reduced to 20 to 40

    pmol per g protein per minute in DU-145 cells (Fig. 1B). Northern

    blot analysis showed that decreased or loss of DPPIV expression in

    prostate cancer cells occurs at RNA level (Fig. 1C). Western blot

    analysis showed low levels of bFGF in NPrEC, RWPE-1, and LNCaP

    Cancer Research

    Cancer Res 2005; 65: (4). February 15, 2005 1326 www.aacrjournals.org

    Research.on June 23, 2013. 2005 American Association for Cancercancerres.aacrjournals.orgDownloaded from

    http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/
  • 7/29/2019 Dipeptidyl Peptidase Inhibits Malignant Phenotype of Prostate.pdf

    4/11

    cells, but high levels in DU-145 cells (Fig. 1D). These observationsindicate an inverse correlation between DPPIV and bFGF

    expression during the emergence of malignant phenotype in

    prostate cancer cells.

    DPPIV Blocks Nuclear Localization and Reduces Expression

    Levels of bFGF in Metastatic Prostate Cancer, DU-145 Cells. To

    understand the role of DPPIV in prostate cancer progression, we

    reexpressed DPPIV in DU-145 cells using the tetracycline inducible

    expression system. Two independent clones expressing DPPIV

    were randomly selected for further studies. Untransfected and

    vector-transfected DU-145 cells were used as controls. The levels of

    DPPIV enzyme activities in presence of doxycycline (1 Ag/mL)

    ranged between 180 and 225 pmol per minute per Ag protein

    in DPPIV transfected cells and were comparable to the levels inNPrEC (200-220 pmol per minute per Ag protein; Fig. 2A). The

    DPPIV enzyme activities in DPPIV transfected cells ranged from

    30 to 60 pmol per minute per Ag protein in the absence of

    doxycycline, similar to control DU-145 cells (20-40 pmol per minute

    per g protein; Fig. 2A). Furthermore, immunoprecipitation analysis

    showed the expected 110-kDa protein corresponding to DPPIV

    with its natural post-translational glycosylation in DPPIV trans-

    fected DU-145 cells in presence of doxycycline (Fig. 2B).

    Because DPPIV and bFGF levels were inversely correlated in

    normal and metastatic prostate cancer cells, we investigated the

    effect of DPPIV on bFGF levels in DU-145 cells. Western blot

    analysis showed high levels of both nuclear (22, 22.5, and 24 kDa)

    and cytoplasmic isoforms (18 kDa) of bFGF in DU-145 and vectortransfected DU-145 cells. Reexpression of DPPIV in DU-145 cells

    greatly decreased the levels of nuclear bFGF isoforms; the levels of

    low molecular weight cytoplasmic isoform were also decreased

    (Fig. 2C). In the absence of DPPIV induction, the same DU-145 cells

    showed bFGF levels comparable to control DU-145 cells, demon-

    strating the specific effect of DPPIV on bFGF levels (Fig. 2D).

    Consistent with Western blot results, immunofluorescence staining

    revealed high levels of both nuclear and cytoplasmic form of bFGF

    in control DU-145 cells (Fig. 2E, a and d). In DPPIV reexpressing

    DU-145 cells, the nuclear accumulation of bFGF was greatly

    reduced; most of bFGF was observed in a punctate pattern around

    the perinuclear region (Fig. 2E, g and j). We confirmed altered

    nuclear localization by costaining the cells with nuclear-specificYo-Yo 1 iodide (Fig. 2E, b , e , h , k; see also merged images in c, f, i, l).

    The staining patterns of bFGF in individual cells are shown in

    Fig. 2F. These observations indicated that DPPIV negatively

    regulates bFGF expression and disrupts its subcellular localization.DPPIV Inhibits Activation of MAPK-ERK1/2, Decreases uPA

    Expression, and Restores Fibronectin Matrix Assembly.

    Expression of bFGF is associated with constitutive activation of

    the MAPK-ERK1/2 signaling pathway. Interestingly, the levels of

    phosphorylated p44/42 were greatly reduced in DPPIV expressing

    DU-145 cells, compared with the control DU-145 cells (Fig. 3A). In

    absence of doxycycline, levels of phosphorylated p44/42 were not

    affected, indicating specific effects of DPPIV on ERK 1/2 kinase

    Figure 1. DPPIV loss and increased bFGF productionare correlated in metastatic PCa cell lines. A, cell surfaceexpression of DPPIV in NPrEC and prostate cancer cells(RWPE-1, LNCaP, and DU-145) as determined by flowcytometry. Cells were stained with the DPPIV-specific mAbS27. Y-axis, relative cell number; X-axis, log of relativefluorescence intensity. Solid black line, DPPIV expression;filled histogram, control IgG1 antibody. B, DPPIVenzymatic activity in total cell lysates as measured bycolorimetric assay using p-nitroanilide as a substrate.Columns, mean values of DPPIV activity of triplicateexperiments; F1 SD. C, DPPIV mRNA levels as assessedby Northern blot analysis using the PCR-amplified cDNAfrangment of DPPIV as probe. GAPDH probe was usedto assess relative levels of total RNA in all samples. D,bFGF protein levels as assessed by Western blot analysis

    using the antibody against bFGF. The monoclonal antibodyagainst actin was used to assess relative levels of totalprotein in all lanes.

    Dipeptidyl Peptidase Blocks bFGF Signaling

    www.aacrjournals.org 1327 Cancer Res 2005; 65: (4). February 15, 2005

    Research.on June 23, 2013. 2005 American Association for Cancercancerres.aacrjournals.orgDownloaded from

    http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/
  • 7/29/2019 Dipeptidyl Peptidase Inhibits Malignant Phenotype of Prostate.pdf

    5/11

    activities (Fig. 3A). The levels of total ERK1/2 remained the same in

    all cell lines in the presence and absence of doxycycline (Fig. 3A).

    Immunofluorescence staining showed the nuclear localization of

    phosphorylated ERK1/2 in control DU-145 cells (Fig. 3B, a and b),

    whereas in DPPIV-transfected cells, nuclear ERK1/2 were barely

    detectable in the presence of doxycycline (Fig. 3B, c and d).

    During prostate cancer progression, high levels of bFGF and

    ERK1/2 activation are associated with increased uPA expression.

    Our current study shows that control DU-145 cells express high

    levels of uPA and reexpression of DPPIV leads to undetectable

    levels of uPA mRNA in these cells (Fig. 3C). Immunofluorescence

    staining further confirmed high levels of uPA in control DU-145

    Figure 2. DPPIV blocks nuclear localization and reduces expression levels of bFGF. A, DPPIV enzyme activity in NPrEC, DU-145 clones transfected with vector,and DPPIV in the absence or presence of doxycycline (1 Ag/mL). Columns, mean values of triplicates; bars, F1 SD. B, immunoprecipitation analysis of DPPIVexpression in DU-145 cells transfected with DPPIV and one vector in the absence and presence of doxycycline (1 Ag/mL). C and D, Western blot analysis of the levelsof nuclear and cytoplasmic isoforms of bFGF. DU-145 and vector control cells showed very high levels of nuclear and moderate levels of cytoplasmic isoform of bFGFin untransfected DU-145 cells and DU-145 cells transfected with either vector or DPPIV in the presence of doxycycline (1 Ag/mL; C) or in the absence of doxycycline(D). E, immunofluorescence staining showing bFGF expression (red) in untransfected, vector, and DPPIV transfected Du-145 cells in the presence of doxyccline.Cells were counter stained with nuclear specific Yo-Yo 1 iodide (green). Stained cells were viewed with an inverted microscope. Merged images show either yellow(red and green) or green nucleus. (Magnification 400). F, staining patterns of bFGF in single cells for each group are shown (Magnification 600). Mergedimages (yellow) represent colocalization of bFGF (red) and nuclear-specific Yo-Yo 1 iodide (green).

    Cancer Research

    Cancer Res 2005; 65: (4). February 15, 2005 1328 www.aacrjournals.org

    Research.on June 23, 2013. 2005 American Association for Cancercancerres.aacrjournals.orgDownloaded from

    http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/
  • 7/29/2019 Dipeptidyl Peptidase Inhibits Malignant Phenotype of Prostate.pdf

    6/11

    cells (Fig. 3D, 1 and 2) and low levels in DPPIV reexpressing DU-

    145 cells. (Fig. 3D, 3 and 4). Western blot analysis showed increased

    levels of fibronectin proteins in DPPIV expressing DU-145 cells

    compared with the control DU-145 cells (Fig. 3E). Immunofluores-

    cence staining confirmed decreased levels of fibronectin matrix on

    the control DU-145 cells but with increased assembly of fibronectin

    matrix that distributed in a fibrilar array on DU-145 cells

    reexpressing DPPIV (Fig. 3F).DPPIV Induces Contact Inhibition and Inhibits Cellular

    Proliferation, Migration, and Invasion of Prostate Cancer DU-

    145 Cells. Restoration of DPPIV expression resulted in profound

    morphologic changes in DU-145 cells. DU-145 cells and vector

    transfected DU-145 cells showed small, rounded morphology and

    grew in disorganized clumps. These cells grew at higher saturation

    densities (Fig. 4A , 1 and 2). DU-145 cells reexpressing DPPIV in

    presence of doxycycline were enlarged, assumed flat epithelial

    morphology, and grew in an organized manner, and exhibited

    contact inhibition (Fig. 4A , 3 and 4). The growth pattern of DU-145

    cells expressing DPPIV was comparable to NPrEC (Fig. 4A , e) with

    no change in absence of doxycycline (shown in insets of Fig. 4A ,

    3 and 4). The alteration of cell shape and growth pattern supports

    previous reports that DPPIV is a cell adhesion molecule and may

    Figure 3. DPPIV decreases activation of ERK1/2, decreases uPA expression and increases cell surface FN matrix assembly. A, Western blot analysis of the levels oftotal and phosphorylated ERK1/2 proteins in control- or DPPIV-transfected DU-145 cells, in the presence or absence of doxycycline. B, immunofluorescence stainingof nuclear translocation of phosphorylated ERK1/2 in DU-145- and vector-transfected DU-145 cells ( a and b) and in DPPIV-transfected DU-145 cells (c and d). C,Northern blot analysis of uPA mRNA levels in untransfected, vector control, and DPPIV-transfected DU-145 cells. The blot was reprobed with 275-bp PCR fragment ofh-actin to assess for equal amount of RNA loaded from all samples. D, immunofluorescence staining of expression of uPA protein in DU-145- ( 1 ), vector- (2), andDPPIV-transfected DU-145 cells (3 and 4). E, Western blot analysis showing the levels of fibronectin in DU-145-, vector-, and DPPIV-transfected DU-145 cells. F,immunoflourescence staining showing very low levels of fibronectin matrix assembly on the cell surface of DU-145 and vector transfected DU-145 cells ( 1 and 2) andincreased assembly of fibronectin matrix on the cell surfaces of DPPIV-transfected DU-145 cells (3 and 4). Data shown in B-F are in the presence of doxycycline.

    Dipeptidyl Peptidase Blocks bFGF Signaling

    www.aacrjournals.org 1329 Cancer Res 2005; 65: (4). February 15, 2005

    Research.on June 23, 2013. 2005 American Association for Cancercancerres.aacrjournals.orgDownloaded from

    http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/
  • 7/29/2019 Dipeptidyl Peptidase Inhibits Malignant Phenotype of Prostate.pdf

    7/11

    play an important role in regulating cell motility. The doubling

    time for DU-145 cells reexpressing DPPIV was increased to 48 from

    36 hours. DPPIV expression also inhibited cell proliferation up to

    50% of the control DU-145 cells in presence of doxycycline without

    much change in the absence of doxycycline as indicated by MTT

    assay (Fig. 4B).In vitro cell migration and invasion are indirect measures of the

    in vivo metastatic potential of a transformed cell and these

    functions are facilitated by bFGF and ERK1/2 activation. The effect

    of DPPIV on prostate cancer cell migration was evaluated

    qualitatively by a wound induced migration assay. At 2 hours,

    DU-145, DPPIV expressing DU-145 cells, and control DU-145 cells

    showed no visible migration (Fig. 4C, 1 , 2, 3, and 4). After 24 hours,

    DU-145 and vector control cells migrated to fill the wounded area

    completely, indicating their strong migratory potential (Fig. 4C, 5

    and 6). However, the migratory potential of DU-145 cells

    Figure 4. DPPIV expression results in contact inhibition and inhibits cellular proliferation, in vitro cell migration, and invasion of prostate cancer cells (DU-145).A, bright field photographs of morphologic changes in DU-145 cells transfected with either vector or DPPIV. Untransfected DU-145 (1) and vector-transfectedDU-145 (2) grew in a disorganized array of cells with clumps indicating lack of contact inhibition. DU-145 cells transfected with DPPIV ( 3 and 4) showed flatepitheloid and dendritic morphology with contact inhibited growth in the presence of doxycycline, similar to normal prostate epithelial cells ( 5). Morphology of DPPIVtransfected DU-145 cells remained similar to parental DU-145 cells in the absence of doxycycline ( insets 3a and 4a). Original magnification, 200. B, the ratesof cellular proliferation of DU-145, either vector or DPPIV transfected DU-145 cells were assessed in absence (A) or presence (B) of doxycycline by MTT assay,as described in Materials and Methods. The experiment was repeated twice. Columns, absorbance expressed as mean of triplicates; bars, F1 SD. C, woundhealing assay showing migration of DU-145, vector or DPPIV transfected DU-145 cells in the presence of doxycycline, after 2 and 24 hours of scraping.(Magnification 100). D, quantitative cell invasion assay as done using Boyden chamber cell invasion assay kit. Absorbances correlating with the number ofcells invaded were read at 540 nm. Columns, mean values of triplicates; bars, F1 SD.

    Cancer Research

    Cancer Res 2005; 65: (4). February 15, 2005 1330 www.aacrjournals.org

    Research.on June 23, 2013. 2005 American Association for Cancercancerres.aacrjournals.orgDownloaded from

    http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/
  • 7/29/2019 Dipeptidyl Peptidase Inhibits Malignant Phenotype of Prostate.pdf

    8/11

    reexpressing DPPIV was greatly reduced as indicated by theunfilled wounded area (Fig. 4C, 7 and 8). Furthermore, the ability of

    cells to invade through ECM coated filters was assessed

    quantitatively by the Boyden Chamber assay. The number

    of invading DU-145 cells reexpressing DPPIV in presence of

    doxycycline was greatly reduced with inhibition up to 50% to 60%

    when compared with control DU-145 cells (Fig. 4D). These results

    indicated that DPPIV inhibits in vitro invasive and migratory

    potential of prostate cancer cells.DPPIV Induces G2-M Arrest and Cell Death in Prostate

    Cancer Cells (DU-145). To further understand the mechanism by

    which DPPIV inhibits proliferation of DU-145 cells, we did cell

    cycle analysis. The percentage of DPPIV expressing DU-145 cells

    in G2-M stage increased to 24% to 34 % compared with the 9%to 10% in control DU-145 cells (Fig. 5A). Transformed cells are

    typically released from dependence on exogenous growth factor

    for survival during tumor progression. Parental-, vector-, and

    DPPIV-transfected DU-145 cells were serum starved with or

    without induction of DPPIV by doxycycline. Control DU-145 cells

    continue to grow with low levels of detectable apoptotic cell

    death (about 5% of cells showed DNA fragmentation over 3

    days). However, apoptosis was evident at day 3, in 24% to 27% of

    DU-145 cells reexpressing DPPIV in presence of doxycycline

    (Fig. 5B). We further found out that DPPIV up-regulates the

    expression of cycline D kinase inhibitor, p27(kip1) by 4- to 6-folds in

    DPPIV transfected cells in the presence of doxycycline, compared

    with the parental and vector transfected DU-145 cells (Fig. 5C).These results link DPPIV expression with the blocking of bFGF

    function that is shown to decrease p27 levels, thereby increasing

    proliferative and survival potential of cancer cells.Silencing of DPPIV by siRNA Restores bFGF Expression and

    Increased ERK1/2 Phosphorylation. To confirm the direct

    involvement of DPPIV in regulating bFGF expression and ERK1/2

    activation, we used RNA interference (siRNA) to silence DPPIV

    expression in LNCaP and DU-145 cells reexpressing DPPIV (DU-

    DPPIV-1). The siRNA plasmid transfected LNCaP and DU-DPPIV-1

    cells were CHANGE analyzed for DPPIV, bFGF, ERK1/2 levels, and

    phenotypic changes. Flow cytometric analysis showed decreased

    cell surface expression of DPPIV in siRNA transfected DU-DPPIV-1

    cells (Fig. 6A). Furthermore, immunoprecipitation and enzymeactivity analysis confirmed the decreased levels of DPPIV protein

    and enzyme activities in siRNA transfected DU-DPPIV-1 cells as

    shown in Fig. 6B and C. The morphology and growth pattern of cells

    transfected with DPPIV-specific siRNA reverted to malignant

    phenotype and started to grow in disorganized colonies, indicating

    loss of contact inhibition similar to the vector transfected DU-145

    cells (Fig. 6D). Knock down of DPPIV expression resulted in

    increased expression of bFGF (Fig. 6E) and restoration of increased

    levels of phosphorylated ERK1/2 MAPK (Fig. 6F). Similar results

    were obtained when DPPIV was silenced in LNCaP cells (Fig. 6G).

    These results suggested that the inhibitory effects of DPPIV on the

    malignant phenotype of prostate cancer cells are at least partly

    Figure 5. DPPIV Induces G2-M cellcycle arrest and cell death in prostatecancer cells (DU-145). A, cell cycle analysiswas done by flow cytometry using DPPIVtransfected DU-145 cells in the presenceof doxycycline (1 Ag/mL). Untransfectedand vector transfected DU-145 cellswere analyzed in parallel. The results aredepicted as a cascade plot. Inset, %cells in various stages of cell cycle. B,% apoptosis of parental-, vector-, andDPPIV-transfected DU-145 cells inserum-free condition as indicated byterminal deoxynucleotidyl transferasemediated nick end labeling assay.Apoptosis was assessed in the absence or

    presence of doxycycline at days 1 and 3.Columns, mean of triplicates; bars, F1 SD.C, Western blot analysis showing theexpression of cycline D kinase inhibitorp27 (kip1) and actin in DU-145-,vector-, and DPPIV-transfected DU-145cells in the presence and absence ofdoxycycline.

    Dipeptidyl Peptidase Blocks bFGF Signaling

    www.aacrjournals.org 1331 Cancer Res 2005; 65: (4). February 15, 2005

    Research.on June 23, 2013. 2005 American Association for Cancercancerres.aacrjournals.orgDownloaded from

    http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/
  • 7/29/2019 Dipeptidyl Peptidase Inhibits Malignant Phenotype of Prostate.pdf

    9/11

    mediated through the negative regulation of the bFGF-MAPK

    signaling pathway.

    Discussion

    The results of our experiments show that DPPIV loss,

    increased bFGF production and in vitro metastatic potential of

    prostate cancer cells are correlated. DPPIV suppresses the

    malignant phenotype of prostate cancer cells by decreasing the

    levels and blocking nuclear localization of bFGF, thereby

    resulting in reduced bFGF signaling as indicated by decreased

    MAPK-ERK1/2 activation and uPA levels. This is the first

    demonstration that bFGF, a powerful mitogen and an angiogenic

    inducer is a target for a protease regulated growth suppressor

    activity in prostate cancer cells.Progression of benign prostate cancer to fatal hormone

    refractory disease is associated with over expression of growth

    factors that mediate alternative mitogenic signaling (15). The cell

    surface protease, DPPIV controls normal cellular growth and

    differentiation by regulating the activity of peptide factors (7, 8).

    Figure 6. Knockdown of DPPIV expression by siRNA restores bFGF expression and increases ERK1/2 phosphorylation. A, flow cytometric analysis showingcell surface expression of DPPIV in DU-145 cells transfected with vector or DPPIV (DU+DPPIV-1) with and without DPPIV specific siRNA treatment. Y-axis, relativecell number; X-axis, log of relative fluorescence intensity. Solid black line, DPPIV expression; filled histogram, control IgG1 antibody. B, immunoprecipitation analysisof the levels of DPPIV and actin protein in control and DU+DPPIV-1 cells with and without siRNA treatment. C, colorimetric enzyme assay of the levels of DPPIVenzyme activities in control and DU+DPPIV-1 cells with and without siRNA treatment. Columns, mean of triplicates; bars, F1 SD. D, morphology of DU-145, vector,and DU+DPPIV-1 cells before and after siRNA treatment. DU-145 cells transfected with control vector did not show any morphologic changes after transfectionwith DPPIV-specific siRNA (1 and 3). The DPPIV transfected DU-145 cells showed flat epitheliod morphology with contact inhibited growth ( 2). The morphologyand growth pattern of the DU-DPPIV-1 cells transfected with DPPIV-specific siRNA reverted back to malignant phenotype and grew in a disorganized coloniesindicating loss of contact inhibition similar to parental DU-145 cells ( 4). E and F, Western blot analysis of the effects of DPPIV silencing on levels of bFGF andphosphorylated ERK1/2. Knock down of DPPIV expression by siRNA resulted in increased expression of bFGF ( E) and increased levels of phosphorylated ERK1/2 (F).G, treatment of LNCaP cells with DPPIV specific siRNA resulted in decreased levels of DPPIV protein as indicated by immunoprecipitation analysis. Silencing of

    DPPIV in LNCaP cells lead to increased levels of bFGF and phosphorylated ERK1/2 as shown by Western blot analysis.

    Cancer Research

    Cancer Res 2005; 65: (4). February 15, 2005 1332 www.aacrjournals.org

    Research.on June 23, 2013. 2005 American Association for Cancercancerres.aacrjournals.orgDownloaded from

    http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/
  • 7/29/2019 Dipeptidyl Peptidase Inhibits Malignant Phenotype of Prostate.pdf

    10/11

    The present studies have shown that DPPIV is expressed at high

    levels in NPrEC that were established from normal prostatic tissue

    and its expression is decreased to moderate levels in RWPE-1 and

    LNCaP cells, and to almost undetectable levels in DU-145 cells. The

    three cell lines, RWPE-1, LNCaP, and DU-145 cells vary widely in

    their phenotypic characteristics. The HPV-18 transformed and

    immortalized RWPE-1 cells represent early stage of prostate cancer

    progression because these cells neither grow in agar nor form

    tumors when injected into nude mice. Although RWPE-1 cells

    express cytokeratins 8 and 18, which are characteristic of luminal

    prostatic epithelial cells, they also coexpress basal cell cytokeratins

    (37). The DU-145 cell lines is AR negative and poorly differentiated,

    whereas the LNCaP cell line that exhibit less aggressive phenotype

    has functional AR, grows less rapidly, and is less tumorigenic than

    the DU-145 cell line. Metastasis formation by LNCaP cells is

    observed only when the cells were injected orthotopicaly into

    prostate glands (38, 39, 41, 42). Our data suggest that loss of DPPIV

    may be an important event during progression of prostate cancer

    cells to aggressive phenotype. This observation is supported by

    other studies conducted with clinical specimens, reporting loss of

    DPPIV expression in >50% of the metastatic and poorly differen-tiated advanced prostate cancer samples (16, 17).

    Several studies have also shown that bFGF is up-regulated in

    prostate cancer cell lines and tumors tissues with increased me-

    tastatic and invasive potential (16). Thus, the mutually exclusive

    expression patterns of DPPIV and bFGF in metastatic cell lines

    probably reflect their role in dedifferentiation from an epithelial to a

    malignant phenotype. Supporting this notion, our data show that

    reexpression of DPPIV in DU-145 cells induces the normal epithelial

    phenotype, cell-cell contact, decreased cell proliferation rate, and

    reduced anchorage-independent growth ability. We have previously

    shown that DPPIV induces similar effects and abrogates the

    tumorigenic potential of melanomas and lung cancer cells (11,

    12). These data support the idea that DPPIV is associated withnonproliferative phenotype of normal prostate cells.

    Although elevated levels of bFGF are often associated with

    prostate cancer progression, the underlying mechanism leading

    to dysregulated bFGF signaling is not known. Our studies

    suggest that this dysregulation could be in part due to loss of

    DPPIV leading to high levels of bFGF in the nucleus that

    disrupts the normal cellular transcriptional program. We

    speculate that in normal cells DPPIV cleaves the NH2 terminal

    nuclear localization signal thereby confining bFGF to the

    perinuclear region. This initial cleavage may be the first step

    in the degradation of bFGF. Alternatively, it is possible that

    DPPIV directly associates with bFGF and interferes with post-

    translational modifications of bFGF such as NH2-terminalmethylation shown to affect its nuclear accumulation (1921).

    DPPIV may also affect the stability of bFGF resulting in its

    reduced levels. Indeed, high levels of bFGF are shown to be

    required for promoting malignant transformation (13, 27).Loss of expression of other membrane associated peptidases

    such as CD10/NEP, CD13/APN, and BP-1/6C3/APA has been

    reported in several types of human cancers (4345). Recently,

    neutral endopeptidase has been shown to interact directly with the

    tumor suppressor gene, pTEN and block FAK signaling pathway

    thereby functioning as a tumor suppressor gene for prostate cancer

    cells (45). Thus, cell surface proteases seem to play important roles

    in regulating the prostate cancer progression.

    Several studies have linked bFGF to activation of the MAPK-

    ERK1/2 signaling pathway that plays a critical role in survival and

    migration of tumor cells (28, 29). Indeed, bFGF-deficient endothelial

    cells lack ERK1/2 activation and loose their migratory potential

    (28). Activation of ERK1/2 is shown to be associated with

    progression of prostate cancer to the lethal androgen-independent

    state (30, 31). However, the causal role of these downstream kinases

    in androgen-independent prostate cancer is not completely

    understood. Our data show that DPPIV expression leads todecreased levels of phosphorylated ERK1/2 resulting in inhibition

    of migratory and invasive potential of prostate cancer cells (DU-

    145). This effect was abrogated upon DPPIV silencing by siRNA

    treatment of the same cells demonstrating the specific effects of

    DPPIV on bFGF mediated ERK1/2 activation. Interestingly, silencing

    of DPPIV in LNCaP cells also resulted in increased bFGF expression

    and activation of ERK1/2. This indicates that regardless of androgen

    receptor expression, DPPIV regulates bFGF and ERK1/2 signaling in

    prostate cancer cells. This observation is of importance because

    most clinical samples of hormone refractory disease still express

    androgen receptor and continue to survive and proliferate probably

    through activating alternative survival and proliferative signaling

    pathways (15). There is evidence to show that, among otherfactors, polypeptide growth factors that are associated with

    activation of MAP kinase signaling pathways play a critical role in

    this phenomenon (4). Our present studies link DPPIV with blocking

    of bFGF-mediated signaling pathway implicated in prostate cancer

    progression.

    DPPIV expression in DU-145 cells lead to an increase in the

    expression of a cyclin D kinase inhibitor, P27 (kip1) leading to an

    arrest in the G2-M phase, and induced apoptosis upon serum

    withdrawal, indicating the emergence of dependence on exoge-

    nous growth factors for their survival. It is shown that bFGF

    promotes cell proliferation and protection from apoptosis by

    decreasing p27 expression (46, 47). Also, disruption of FGF

    signaling by the dominant-negative FGFR1 is shown to result in

    cell cycle arrest of prostate cancer cells in G2-M stage (48). Takentogether, these observations suggest that DPPIV blocks the

    signaling pathway coordinated by bFGF and ERK1/2 in prostate

    cancer cells.

    Tumor cell migration and invasion depends largely on an

    enzymatic cascade of proteolysis that involves uPA and its receptor

    (uPAR). This cascade is an essential step for malignant cell

    transport. Growth factors including bFGF and activated ERK1/2

    have been shown to promote uPA production in various tumoral

    and normal cell types (3235). Elevated levels of uPA are found in

    patients with advanced stages of prostate cancer and it is a strong

    indicator of poor prognosis (3436). It is interesting to note that

    uPA activates plasminogen to generate plasmin that degrades

    extracellular matrix components including fibronectin, a requiredevent for malignant cell metastasis. We have shown that DPPIV

    dramatically decreases uPA mRNA production and increases the

    fibronectin assembly on the cell surface. Thus, increase in uPA

    expression seen in DU-145 cells, may result from the cooperative

    action of signaling provided by bFGF. Disruption of these signaling

    events by DPPIV is probably involved in the assembly of fibronectin

    matrix and increase in cell adhesion that serves as a barrier for cell

    motility. This possibility is supported by data showing DPPIV

    binds to fibronectin and functions as an adhesion molecule (10),

    and that fibronectin on the cell surface disappears upon oncogenic

    transformation. Furthermore, overexpression of fibronectin leads

    to suppression of the transformed phenotype (49, 50). Thus, loss of

    Dipeptidyl Peptidase Blocks bFGF Signaling

    www.aacrjournals.org 1333 Cancer Res 2005; 65: (4). February 15, 2005

    Research.on June 23, 2013. 2005 American Association for Cancercancerres.aacrjournals.orgDownloaded from

    http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/http://cancerres.aacrjournals.org/
  • 7/29/2019 Dipeptidyl Peptidase Inhibits Malignant Phenotype of Prostate.pdf

    11/11

    DPPIV may render tumor cells less adhesive thereby promotingdetachment of cells from primary tumors initiating metastasis.However, in vivo studies will further strengthen the role of DPPIVin blocking metastatic potential of prostate cancer cells.

    Given the ability of DPPIV to regulate the cell proliferation,differentiation, apoptosis, and cell motility through its regulationof bFGF provides a new and potentially significant approach forbetter understanding prostate cancer progression and develop-

    ment of treatment strategies aimed at blocking mitogenic andangiogenic signaling.

    Acknowledgments

    Received 5/26/2004; revised 11/15/2004; accepted 12/3/2004.Grant support: Department of Microbiology and Molecular Genetics, Lake

    Champlain Cancer Research Organization, Vermont Cancer Center at the University ofVermont.

    The costs of publication of this article were defrayed in part by the payment of pagecharges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.

    We thank Emma H ewes and Scott Tighe at shared Core Facility o f Vermont CancerCenter for technical help in construction of siRNA plasmids and flow cytometric

    analysis and Drs. David Nanus, Cedric Wesley, Alan Houghton, Nick Heintz,Mark Plante, Brooke Mossman, and Susan Wallace for helpful discussions.

    References1. Ware JL. Growth factor network disruption in prostate

    cancer progression. Cancer Metastasis Rev 19981999;17:4437.

    2. Giri D, Ropiquet F, Ittmann M. Alterations inexpression of basic fibroblast growth factor (FGF) 2and its receptor FGFR-1 in human prostate cancer. ClinCancer Res 1999;5:106371.

    3. Dow JK, deVere White RW. Fibroblast growth factor 2:its structure and property, paracrine function, tumorangiogenesis, and prostate-related mitogenic and on-

    cogenic functions. Urology 2000;55:8006.4. Isaacs JT, Isaacs WB. Androgen receptor outwits

    prostate cancer drugs. Nat Med 2004;10:267.

    5. Feldman BJ, Feldman D. The development of andro-gen-independent prostate cancer. Nat Rev Cancer 2001;1:3445.

    6. Nakamoto T, Chang CS, Li AK, Chodak GW. Basicfibroblast growth factor in human prostate cancer cells.Cancer Res 1992;52:5717.

    7. Boonacker E, Van Noorden CJ. The multifunctional ormoonlighting protein CD26/DPPIV. Eur J Cell Biol 2003;82:5373.

    8. Proost P, Struyt S, Schols D, et al. Truncation ofMacrophage-derived Chemokines by CD26/dipeptidylpeptidase IV beyond its predicted cleavage site affectschemotactic activity and CC chemokine receptor 4interaction. J Biol Chem 1999;274:398893.

    9. Bauvois BA. Collagen-binding glycoprotein on the

    surface of mouse fibroblasts is identified as dipeptidylpeptidase IV. Biochem J 1988;252:72331.

    10. Piazza GA, Callanan HM, Mowery J, Hixson DC.Evidence for a role of dipeptidyl peptidase IV infibronectin-mediated interactions of hepatocytes withextracellular matrix. Biochem J 1989;262:32734.

    11. Wesley UV, Albino AP, Tiwari S, Houghton AN. A rolefor dipeptidyl peptidase IV in suppressing the malig-nant phenotype of melanocytic cells. J Exp Med 1999;190:31122.

    12. Wesley UV, Tiwari S, Houghton AN. Role fordipeptidyl peptidase IV in tumor suppression of humannon small cell lung carcinoma cells. Int J Cancer 2004;109:85566.

    13. Sedo A, Kraml J. Dipeptidyl peptidase IV in cellproliferation and differentiation. Sb Lek 1994;95:2858.

    14. Houghton AN, Albino AP, Cordon-Cardo C, Davis LJ,Eisinger M. Cell surface antigens of human melanocytes

    and melanoma. Expression of adenosine deaminasebinding protein is extinguished with melanocytetransformation. J Exp Med 1988;167:197212.

    15. Abbott CA, Gorrell MD, Levy MT, McCaughan GW.Molecular analyses of human and rat dipeptidylpeptidase IV. Adv Exp Med Biol 1997;421:1619.

    16. Bogenrieder T, Finstad CL, Freeman RH, et al.Expression and localization of aminopeptidase A,aminopeptidase N, and dipeptidyl peptidase IV inbenign and malignant human prostate tissue. Prostate1997;33:22532.

    17. Dingens WNM, TenKate J, Kirch JM, et al. Adenosinedeaminase complexing protein (ADCP) expression andmetastatic potential in prostatic adenocarcinomas.J Pathol 1999;160:195201.

    18. Sakamoto J, Watanabe T, Teramukai S, et al.Distribution of adenosine deaminase binding protein

    in normal and malignant tissues of the gastrointestinaltract studied by monoclonal antibodies. J Surg Oncol1993;52:12434.

    19. Bikfalvi A, Klein S, Pintucci G, Rifkin DB. Biologicalroles of fibroblast growth factor-2. Endocr Rev 1997;18:2645.

    20. Delrieu I. The high molecular weight isoforms ofbasic fibroblast growth factor (FGF-2): an insightinto an intracrine mechanisms. FEBS Lett 2000;18:468:610.

    21. Bugler B, Amalric F, Prats H. Alternative initiation oftranslation determines cytoplasmic or nuclear localiza-

    tion of basic fibroblast growth factor. Mol Cell Biol1991;11:5737.

    22. Okada-Ban M, Moens G, Thiery JP, Jouanneau J.Nuclear 24 kD fibroblast growth factor (FGF)-2 confersmetastatic properties on rat bladder carcinoma cells.Oncogene 1999;18:671924.

    23. Fukui S, Otani N, Nawashiro H, et al. Nuclearaccumulation of basic fibroblast growth factor as apredictor for the recurrence of pituitary adenomas.J Neurooncol 2002;57:2219.

    24. Meyer GE, Yu E, Siegal JA, Petteway JC, Blumenstein BA,Brawar MK. Serum basic fibroblast growth factor inmen with and without prostate carcinoma. Cancer1995;76:230411.

    25. Bok RA, Halabi S, Fei DT, et al. Vascular endothelialgrowth factor and basic fibroblast growth factor urinelevels as predictors of outcome in hormone-refractoryprostate cancer patients: a cancer and leukemia group

    B study. Cancer Res 2001;61:25336.26. Cronauer MV, Hittmair A, Eder IE, et al. Basicfibroblast growth factor levels in cancer cells a and insera of patients suffering from proliferative disorders ofthe prostate. Prostate 1997;31:22333.

    27. Polnaszek N, Kwabi-Addo B, Peterson LE, et al.Fibroblast growth factor 2 promotes tumor progressionin an autochthonous mouse model of prostate cancer.Cancer Res 2003;63:575460.

    28. Pintucci G, Moscatelli D, Saponara F, et al. Lack ofERK activation and cell migration in FGF-2-deficientendothelial cells. FASEB J 2002;16:598600.

    29. Gaubert F, Escaffit F, Bertrand C, et al. Expression ofthe high molecular weight fibroblast growth factor-2isoform of 210 amino acids is associated withmodulation of protein kinases Cy and epsilon andERK activation. J Biol Chem 2001;276:154554.

    30. Price DT, Rocca GD, Guo C, Ballo MS, Schwinn DA,Luttrell LM. Activation of extracellular signal-regulated

    kinase in human prostate cancer. J Urol 1999;162:153742.31. Gioeli D, Mandell JW, Petroni GR, Frierson HF Jr,

    Weber MJ. Activation of mitogen-activated proteinkinase associated with prostate cancer progression.Cancer Res 1999;15;59:27984.

    32. Rabbani SA, Mazar AP. The role of the plasminogenactivation system in angiogenesis and metastasis. SurgOncol Clin N Am 2001;10:393415.

    33. Giuliani R, Bastaki M, Coltrini D, Presta M. Role ofendothelial cell extracellular signal-regulated kinase1/2in urokinase-type plasminogen activator upregulationand in vitro angiogenesis by fibroblast growth factor-2.J Cell Sci 1999;112:2597606.

    34. Hienert G, Kirchheimer JC, Pfluger H, Binder BR.Urokinase-type plasminogen activator as a marker forthe formation of distant metastases in prostaticcarcinomas. J Urol 1988;140:14669.

    35. Hall CL, Tsan R, Mugnai G, Mazar A, Radinsky R,Pettaway CA. Enhanced invasion of hormone refractoryprostate cancer cells through hepatocyte growth factor(HGF) induction of urokinase-type plasminogen activa-tor (u-PA). Prostate 2004;59:16776.

    36. Forbes K, Gillette K, Kelley LA, Sehgal I. Increasedlevels of urokinase plasminogen activator receptor inprostate cancer cells derived from repeated metastasis.World J Urol 2004;22:6771.

    37. Webber MM, Bello D, Quader S. Immortalized andtumorigenic adult human prostatic epithelial cell lines:characteristics and applications Part 2. Tumorigenic

    cell lines. Prostate 1997;30:5864.38. Horoszewicz JS, Leong SS, Kawinski E, et al. LNCaP

    model of human prostatic carcinoma. Cancer Res 1983;43:180918.

    39. Wu HC, Hsieh JT, Gleave ME, Brown NM, Pathak S,Chung LW. Derivation of androgen-independent humanLNCaP prostatic cancer cell sublines: role of bonestromal cells. Int J Cancer 1994;57:40612;

    40. Stone KR, Mickey DD, Wunderli H, Mickey GH,Paulson DF. Isolation of a human prostate carcinomacell line (DU 145). Int J Cancer 1978;15;21:27481.

    41. Wang X, An Z, Geller J, Hoffman RM. High-malignancy orthotopic nude mouse model of humanprostate cancer LNCaP. Prostate 1999;15;39:1826.

    42. Rembrink K, Romijn JC, van der Kwast TH, Rubben H,Schroder FH. Orthotopic implantation of humanprostate cancer cell lines: a clinically relevant animalmodel for metastatic prostate cancer. Prostate 1997;

    15;31:16874.43. Ganju RK, Sunday M, Tsarwhas DG, Card A, Shipp MA.

    CD10/NEP in non-small cell lung carcinomas. Rela-tionship to cellular proliferation. J Clin Invest 1994;94:178491.

    44. Cohen AJ, Bunn PA, Franklin W, et al. Neutralendopeptidase: variable expression in human lung,inactivation in lung cancer, and modulation ofpeptide-induced calcium flux. Cancer Res 1996;15;56:8319.

    45. Sumitomo M, Iwase A, Zheng R, et al. Synergyin tumor suppression by direct interaction ofneutral endopeptidase with PTEN. Cancer Cell 2004;5:6778.

    46. Shankland SJ, Pippin J, Flanagan M, et al. Mesangialcell proliferation mediated by PDGF and bFGF isdetermined by levels of the cyclin kinase inhibitorp27Kip1. Kidney Int 1997;51:108899.

    47. Hiromura K, Pippin JW, Fero ML, Roberts JM,Shankland SJ. Modulation of apoptosis by the cyclin-dependent kinase inhibitor p27(Kip1). J Clin Invest1999;103:597604.

    48. Ozen M, Giri D,Ropiquet F, Mansukhani A,Ittmann M.Role of fibroblast growth factor receptor signaling inprostate cancer cel l survival. J Natl Cancer Inst 2001 Dec5;93:178390.

    49. Akamatsu H, Ichihara-Tanaka K, Ozono K, Kamiike W,Matsuda H, Sekiguchi K. Suppression of transformedphenotypes of human fibrosarcoma cells by over-expression of recombinant fibronectin. Cancer Res1996;56:45416.

    50. Brenner KA, Corbett SA, Schwarzbauer JE. Regulationof fibronectin matrix assembly by activated Ras intransformed cells. Oncogene 2000 Jun 29;19:15663.

    Cancer Research

    Cancer Res 2005; 65: (4). February 15, 2005 1334 www.aacrjournals.org