14
Transactivation of the Urokinase-type Plasminogen Activator Receptor Gene through a Novel Promoter Motif Bound with an Activator Protein-2a-related Factor* (Received for publication, June 25, 1998, and December 8, 1998) Heike Allgayer, Heng Wang, Yao Wang‡, Markus M. Heiss§, Reinhard Bauer, Okot Nyormoi, and Douglas Boydi From the Department of Cancer Biology and Department of Biochemistry, M.D. Anderson Cancer Center, Houston, Texas 77030, the Department of Renal Medicine, Westmead Hospital, Westmead, University of Sydney, Westmead, New South Wales, Australia, and the §Department of Surgery, Klinikum Grosshadern, Ludwig-Maximilians University, D-81377 Munich, Germany The urokinase receptor overexpressed in invasive cancers promotes laminin degradation. The current study was undertaken to identify cis elements and trans-acting factors activating urokinase receptor ex- pression through a footprinted (2148/2124) region of the promoter containing putative activator protein-2- and Sp1-binding motifs. Mobility shifting experiments using nuclear extract from a high urokinase receptor- expressing cell line (RKO) indicated that Sp1, Sp3, and a factor similar to, but distinct from, activator protein-2a bound to this region. Mutations preventing the binding of the activator protein 2a-related factor diminished urokinase receptor promoter activity. In RKO cells, the expression of a negative regulator of activator protein-2 function diminished urokinase receptor promoter activ- ity, protein, and laminin degradation. Conversely, urokinase receptor promoter activity in low urokinase receptor-expressing GEO cells was increased by activa- tor protein-2aA expression. Although using GEO nu- clear extract, little activator protein-2a-related factor bound to the footprinted region, phorbol 12-myristate 13-acetate treatment, which induces urokinase receptor expression, increased complex formation. Mutations preventing the activator protein-2a-related factor and Sp1/Sp3 binding reduced urokinase receptor promoter stimulation by this agent. Thus, the constitutive and phorbol 12-myristate 13-acetate-inducible expression of the urokinase receptor is mediated partly through trans-activation of the promoter via a sequence (2152/ 2135) bound with an activator protein-2a-related factor. The urokinase-type plasminogen activator (urokinase) is a serine protease that converts the inert zymogen plasminogen into plasmin, a protease with broad substrate specificity lead- ing to extracellular matrix degradation and tumor invasion (1–3). Urokinase can bind specifically and with high affinity (K D ;0.5 nM) to a 45– 60-kDa heavily glycosylated cell surface receptor (u-PAR) 1 (4, 5) composed of three similar repeats approximately 90 residues each (6, 7). The amino-terminal domain binds the plasminogen activator with the carboxyl ter- minus domain serving to anchor the binding protein to the cell surface via a glycosyl-phosphatidylinositol chain (6, 7). The u-PAR has multiple functions. First, urokinase bound to the u-PAR activates plasminogen at a much faster rate than fluid phase plasminogen activator (8, 9), and this contributes to type IV collagenase activation (10). Second, the binding site clears urokinase-inhibitor complexes from the extracellular space (11) via a a 2 -macroglobulin receptor-dependent mecha- nism (12). Third, the u-PAR interacts with the extracellular domain of integrins to connect to the cytoskeleton, thereby mediating cell adhesion and migration (13–15). Fourth, the u-PAR is chemotactic for human monocytes and mast cells, and this may require the cleavage of the binding site between domains 1 and 2 (16, 17). The u-PAR gene is 7 exons long and is located on chromo- some 19q13 (18, 19). Transcription of the u-PAR gene yields a 1.4-kilobase mRNA or an alternatively spliced variant lacking the membrane attachment peptide sequence (20, 21). The amounts of u-PAR are controlled mainly at the transcriptional level, but altered message stability and receptor recycling may represent other means of controlling the amount of this gene product at the cell surface (22–25). The transcriptional regulation of the u-PAR gene is still poorly understood. Soravia et al. (26) reported that the basal expression of the gene was regulated via Sp1 motifs proximal and upstream of the transcriptional start site. Our laboratory showed that both the constitutive and PMA-inducible expres- sion of the gene required a footprinted region (2190/2171) of the promoter containing an AP-1 motif (22). We also observed a second footprinted region of the promoter (2148/2124), and deletion of this region caused a dramatic reduction in the constitutive u-PAR promoter activity in a colon cancer cell line characterized by its high expression of this gene. Interestingly, this region of the promoter contained noncanonical AP-2 (2142/2134) and Sp1 (2147/2138) motifs overlapping with each other as well as nonconsensus polyomavirus activator 3 (2133/2127) motifs. The u-PAR has been implicated in a number of physiological and pathological processes involving tissue remodeling, al- * This work was supported by National Institutes of Health Grants R01 CA58311, R01 DE10845, and P01 DE11906; a Physician’s Referral Service grant (to D. B.); and fellowships from the Dr. Mildred Scheel Cancer Foundation (Deutsche Krebshilfe, Bonn, Germany) and the Deutsche Forschungsgemeinschaft (Bonn, Germany) (to H. A. and R. B., respectively). The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. This paper is in tribute to Barbara Young for her dedicated work. i To whom all correspondence should be addressed: Dept. of Cancer Biology, Box 108, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030. Tel.: 713-792-8953; Fax: 713-794-0209; E-mail: [email protected]. 1 The abbreviations used are: u-PAR, urokinase-type plasminogen activator receptor; AP-1 and -2, activator protein-1 and -2, respectively; CAT, chloramphenicol acetyltransferase; EMSA, electrophoretic mobil- ity shift assay; PMA, phorbol 12-myristate 13-acetate; RSV, Rous sar- coma virus. THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 274, No. 8, Issue of February 19, pp. 4702–4714, 1999 © 1999 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A. This paper is available on line at http://www.jbc.org 4702 by guest on March 25, 2018 http://www.jbc.org/ Downloaded from

Transactivation of the Urokinase-type Plasminogen Activator

Embed Size (px)

Citation preview

Page 1: Transactivation of the Urokinase-type Plasminogen Activator

Transactivation of the Urokinase-type Plasminogen ActivatorReceptor Gene through a Novel Promoter Motif Bound with anActivator Protein-2a-related Factor*

(Received for publication, June 25, 1998, and December 8, 1998)

Heike Allgayer, Heng Wang, Yao Wang‡, Markus M. Heiss§, Reinhard Bauer¶, Okot Nyormoi,and Douglas Boydi

From the Department of Cancer Biology and ¶Department of Biochemistry, M.D. Anderson Cancer Center,Houston, Texas 77030, the ‡Department of Renal Medicine, Westmead Hospital, Westmead, University of Sydney,Westmead, New South Wales, Australia, and the §Department of Surgery, Klinikum Grosshadern,Ludwig-Maximilians University, D-81377 Munich, Germany

The urokinase receptor overexpressed in invasivecancers promotes laminin degradation. The currentstudy was undertaken to identify cis elements andtrans-acting factors activating urokinase receptor ex-pression through a footprinted (2148/2124) region ofthe promoter containing putative activator protein-2-and Sp1-binding motifs. Mobility shifting experimentsusing nuclear extract from a high urokinase receptor-expressing cell line (RKO) indicated that Sp1, Sp3, and afactor similar to, but distinct from, activator protein-2abound to this region. Mutations preventing the bindingof the activator protein 2a-related factor diminishedurokinase receptor promoter activity. In RKO cells, theexpression of a negative regulator of activator protein-2function diminished urokinase receptor promoter activ-ity, protein, and laminin degradation. Conversely,urokinase receptor promoter activity in low urokinasereceptor-expressing GEO cells was increased by activa-tor protein-2aA expression. Although using GEO nu-clear extract, little activator protein-2a-related factorbound to the footprinted region, phorbol 12-myristate13-acetate treatment, which induces urokinase receptorexpression, increased complex formation. Mutationspreventing the activator protein-2a-related factor andSp1/Sp3 binding reduced urokinase receptor promoterstimulation by this agent. Thus, the constitutive andphorbol 12-myristate 13-acetate-inducible expression ofthe urokinase receptor is mediated partly throughtrans-activation of the promoter via a sequence (2152/2135) bound with an activator protein-2a-related factor.

The urokinase-type plasminogen activator (urokinase) is aserine protease that converts the inert zymogen plasminogeninto plasmin, a protease with broad substrate specificity lead-ing to extracellular matrix degradation and tumor invasion(1–3). Urokinase can bind specifically and with high affinity

(KD ;0.5 nM) to a 45–60-kDa heavily glycosylated cell surfacereceptor (u-PAR)1 (4, 5) composed of three similar repeatsapproximately 90 residues each (6, 7). The amino-terminaldomain binds the plasminogen activator with the carboxyl ter-minus domain serving to anchor the binding protein to the cellsurface via a glycosyl-phosphatidylinositol chain (6, 7).

The u-PAR has multiple functions. First, urokinase bound tothe u-PAR activates plasminogen at a much faster rate thanfluid phase plasminogen activator (8, 9), and this contributes totype IV collagenase activation (10). Second, the binding siteclears urokinase-inhibitor complexes from the extracellularspace (11) via a a2-macroglobulin receptor-dependent mecha-nism (12). Third, the u-PAR interacts with the extracellulardomain of integrins to connect to the cytoskeleton, therebymediating cell adhesion and migration (13–15). Fourth, theu-PAR is chemotactic for human monocytes and mast cells, andthis may require the cleavage of the binding site betweendomains 1 and 2 (16, 17).

The u-PAR gene is 7 exons long and is located on chromo-some 19q13 (18, 19). Transcription of the u-PAR gene yields a1.4-kilobase mRNA or an alternatively spliced variant lackingthe membrane attachment peptide sequence (20, 21). Theamounts of u-PAR are controlled mainly at the transcriptionallevel, but altered message stability and receptor recycling mayrepresent other means of controlling the amount of this geneproduct at the cell surface (22–25).

The transcriptional regulation of the u-PAR gene is stillpoorly understood. Soravia et al. (26) reported that the basalexpression of the gene was regulated via Sp1 motifs proximaland upstream of the transcriptional start site. Our laboratoryshowed that both the constitutive and PMA-inducible expres-sion of the gene required a footprinted region (2190/2171) ofthe promoter containing an AP-1 motif (22). We also observed asecond footprinted region of the promoter (2148/2124), anddeletion of this region caused a dramatic reduction in theconstitutive u-PAR promoter activity in a colon cancer cell linecharacterized by its high expression of this gene. Interestingly,this region of the promoter contained noncanonical AP-2(2142/2134) and Sp1 (2147/2138) motifs overlapping witheach other as well as nonconsensus polyomavirus activator 3(2133/2127) motifs.

The u-PAR has been implicated in a number of physiologicaland pathological processes involving tissue remodeling, al-

* This work was supported by National Institutes of Health GrantsR01 CA58311, R01 DE10845, and P01 DE11906; a Physician’s ReferralService grant (to D. B.); and fellowships from the Dr. Mildred ScheelCancer Foundation (Deutsche Krebshilfe, Bonn, Germany) and theDeutsche Forschungsgemeinschaft (Bonn, Germany) (to H. A. andR. B., respectively). The costs of publication of this article were defrayedin part by the payment of page charges. This article must therefore behereby marked “advertisement” in accordance with 18 U.S.C. Section1734 solely to indicate this fact.

This paper is in tribute to Barbara Young for her dedicated work.i To whom all correspondence should be addressed: Dept. of Cancer

Biology, Box 108, M.D. Anderson Cancer Center, 1515 Holcombe Blvd.,Houston, TX 77030. Tel.: 713-792-8953; Fax: 713-794-0209; E-mail:[email protected].

1 The abbreviations used are: u-PAR, urokinase-type plasminogenactivator receptor; AP-1 and -2, activator protein-1 and -2, respectively;CAT, chloramphenicol acetyltransferase; EMSA, electrophoretic mobil-ity shift assay; PMA, phorbol 12-myristate 13-acetate; RSV, Rous sar-coma virus.

THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 274, No. 8, Issue of February 19, pp. 4702–4714, 1999© 1999 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.

This paper is available on line at http://www.jbc.org4702

by guest on March 25, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 2: Transactivation of the Urokinase-type Plasminogen Activator

though it is not critical for mouse development (27). In cancer,several experimental and clinical findings support the viewthat the u-PAR plays a prominent role in tumor cell invasionand metastasis. For example, the u-PAR mRNA is expressed inthe tumor cells of invasive colon cancers (28, 29), and a highu-PAR protein level is predictive of short survival times forpatients with this disease (30). Further, earlier studies haveshown that the overexpression of a human u-PAR cDNA in-creased the ability of human osteosarcoma cells to invade intoan extracellular matrix-coated porous filter (31). Conversely,down-regulating u-PAR levels using antisense expression con-structs, oligonucleotides, or synthetic compounds reduced theability of divergent invasive cancers to invade in vitro and invivo (32–36). Since the u-PAR is a key factor in promotingtumor-associated proteolysis, down-regulation of its expressioncould be a promising strategy for inhibiting cancer invasionand metastasis. We therefore undertook a study with two ob-jectives: (a) to identify cis-elements and trans-acting factorsregulating constitutive and PMA-inducible u-PAR gene expres-sion via the footprinted region spanning nucleotides 2148/2124 and (b) to determine the effect of interfering with tran-scription factors binding to this region on u-PAR-directedlaminin degradation.

EXPERIMENTAL PROCEDURES

Vectors and Antibodies—The u-PAR CAT reporter consisted of 449base pairs of sequence (37) stretching from 2398 to 151 (relative to thetranscription start site) cloned into the XbaI site of the pCAT-Basicvector (Promega, Madison, WI). Reporter constructs regulated by trun-cated u-PAR promoter fragments were as described previously (22). Theurokinase CAT reporter consisted of 2345 base pairs of 59-flankingregion fused directly to the reporter (38). Antibodies to Sp1, Sp2, Sp3,and AP-2 isoforms were purchased from Santa Cruz Biotechnology, Inc.(Santa Cruz, CA). The Jun-D expression construct was described else-where (22). Oligonucleotides were purchased from Genosys Biotechnol-ogies (The Woodlands, TX). Recombinant AP-2aA and Sp1 (full-lengthhuman proteins) were obtained from Promega (Madison, WI). Expres-sion vectors for AP-2aA, AP-2aB, and AP-2 antisense (39) consisted ofthe cloned sequences inserted into the EcoRI site of pSG5 (Stratagene,La Jolla, CA) and were kindly provided by Dr. Michael Tainsky. TheAP-2 pBLCAT2 reporter construct contained three consensus AP-2motifs 59 of the pBLCAT2 reporter (40). For the generation of the R2CAT reporter construct, an oligonucleotide spanning nucleotides 2154/2128 was cloned into the XbaI site of pCATbasic (Promega).

Preparation of Nuclear Extracts and EMSA—Nuclear extracts andEMSA were carried out as described elsewhere (22). EMSA was carriedout using nuclear extract (8 mg), 0.6 mg of poly(dI/dC), and 2 3 104 cpmof a T4 polynucleotide kinase-labeled [g-32P]ATP oligonucleotide. Thesequences of the AP-2 and Sp1 consensus oligonucleotides were: 59-GATCGA ACT GAC CGC CCG CGG CCC GT-39 (Santa Cruz Biotechnologycatalog no. sc-2513) and 59-ATT CGA TCG GGG CGG GGC GAG C-39(Santa Cruz Biotechnology catalog no. sc-2502), respectively. The se-quence of the mutated (underlined nucleotides) AP-2 consensus-con-taining oligonucleotide was 59-GAT CGA ACT GAC CGC TTG CGGCCC GT-39 (Santa Cruz Biotechnology catalog no. sc-2516).

Site-directed Mutagenesis—This was performed according to the pro-tocol of the Site-Directed Mutagenesis Kit (5Z701) of CLONTECH (PaloAlto, CA). For generation of the Sp1/Sp3mt u-PAR CAT, pCATbasic(Promega) regulated by 398 base pairs of the u-PAR promoter (37)served as a template. A mutation primer substituting T for G at posi-tions 2148, 2147, 2144, and 2142 and its corresponding selectionprimer (59-CTTATCATGTCTGGTACCCCCGGAATTC-39) convertingthe BamHI site of pCATbasic to a KpnI site were annealed to thedenatured template plasmid, and the plasmid was amplified accordingto the protocol. Remaining wild-type plasmid was eliminated by twoBamHI digestions for 4 h, each of them followed by transformation ofnondigested DNA into BMH 71–18 mutS cells (CLONTECH, Palo Alto,CA). DNA of selected clones was isolated and sequenced using theAmersham Pharmacia Biotech T7-Sequenase 2.0 Kit.

For generation of the AP-2/Sp1/Sp3mt u-PAR CAT construct, theSp1/Sp3mt u-PAR CAT plasmid served as a template. The secondmutation primer substituted A for C at positions 2146, 2145, 2142,and 2141 of the u-PAR promoter, and the selection primer (59-CTTAT-CATGTCTGGATCCCCCGGAATTC-39) changed the KpnI site gener-

ated above back to BamHI. Selection for AP-2/Sp1/3-mutated plasmidswas done by KpnI digestion. The procedure was continued as describedabove.

CAT Assays—Cells were transfected at 60% confluency using poly-L-ornithine as described previously (41). All transient transfectionswere performed in the presence of a luciferase expression vector (4 mg),and transfection efficiencies were determined by assaying for luciferaseactivity. CAT activity was measured as described previously (22). Theamount of acetylated [14C]chloramphenicol was determined using aStorm 840 PhosphorImager (Molecular Dynamics, Sunnyvale, CA) us-ing ImageQuant software. Student’s t test analysis was performedtwo-sided using the SPSS for Windows statistical software (release6.1.3) (SPSS Inc., Chicago, IL). Statistical significance was defined asp # 0.05.

Magnetic Separation of Transfected and Nontransfected Cells—Transfected cells were enriched by the MACS-Select method of MiltenyiBiotech (Auburn, CA). RKO cells were co-transfected with the AP-2aBexpression construct and a plasmid encoding a mutated CD4 molecule(pMACS 4) in a 3:1 ratio. Cells were harvested after 42 h in 320 ml ofPBE buffer (phosphate-buffered saline, 0.5% bovine serum albumin, 5mM EDTA) and incubated for 15 min with 80 ml of a magnetic bead-conjugated antibody directed against the mutated CD4 molecule. Thecell suspension was then run through VS1 separation columns usingthe VarioMACS magnet according to the manufacturer’s protocol.

Western Blotting and Enzyme-linked Immunosorbent Assay for u-PAR Protein—RKO cells were extracted into a buffer (10 mM Tris, pH7.4, 0.15 M NaCl, 1% Triton X-100, 0.5% Nonidet P-40, 20 mg/ml apro-tinin, 1 mM phenylmethylsulfonyl fluoride, 1 mM EGTA, 1 mM EDTA)for 10 min at 4 °C. Insoluble material was removed by centrifugation,and 750 mg protein of cell extract was immunoprecipitated at 4 °C for16 h with 0.25 mg of a polyclonal anti-u-PAR antibody and proteinA-agarose beads. The polyclonal antibody (kindly provided by Dr. An-drew Mazar, Angstrom Pharmaceuticals, San Diego, CA) was raised inrabbits against amino acids 1–281 of the human u-PAR and purified ona Sepharose-immobilized u-PAR column. The immunoprecipitated ma-terial was subjected to standard Western blotting (42), and the blot wasprobed with 5 mg/ml of an anti-u-PAR monoclonal antibody (catalog no.3931, American Diagnostica, Greenwich, CT) and a horseradish perox-idase-conjugated goat anti-mouse IgG. Bands were visualized by en-hanced chemiluminescence (Amersham Pharmacia Biotech).

For the determination of u-PAR by enzyme-linked immunosorbentassay, resected tissue was prepared as described by the manufacturer(American Diagnostica).

Laminin Degradation Assays—These were carried out as describedpreviously (43). RKO cells were harvested with 3 mM EDTA/phosphate-buffered saline, washed twice, and seeded (500,000 cells) on radioactivelaminin-coated (2 mg/dish) dishes. The cells were allowed to attachovernight. Subsequently, cell surface u-PARs were saturated by incu-bating the cells at 37 °C for 30 min with 5 nM urokinase, and unboundplasminogen activator was removed by washing. The cells were thenreplenished with serum-free medium with or without 10 mg/ml plas-minogen (final concentration). After varying times at 37 °C, aliquots ofthe culture medium were withdrawn and counted for radioactivity.Solubilized laminin represents the degraded glycoprotein (43).

RESULTS

Region II of the u-PAR Promoter Footprinted by NuclearExtract from a High u-PAR-expressing Cell Line Is Bound withSp1, Sp3, and an AP-2a-related Factor—We previously re-ported (22) that nuclear extract from a high (3 3 105 bindingsites/cell) u-PAR-expressing colon cancer cell line (RKO) foot-printed a region (referred to as region II) of the u-PAR promoter(nucleotides 2148/2124). As a first step to identifying tran-scription factor(s) bound to this region, EMSA was carried outusing an oligonucleotide spanning nucleotides 2154/2128 (Fig.1A). The oligonucleotide was not extended to the 39-end (2124)of the footprinted region II, since preliminary EMSA utilizing aprobe that included sequences 39 of 2128 had failed to revealany specific binding complexes. Employing the 2154/2128 ol-igonucleotide, three slower migrating bands (indicated by abrace, arrow, and asterisk) were apparent (Fig. 1B, lanes 2 and8) with a 100-fold excess of the nonradioactive oligonucleotideeliminating (lane 3) all of these bands. Computer analysis ofthis region of the u-PAR promoter revealed the presence ofputative AP-2- (2142/2134) and Sp1/Sp3- (2147/2138) bind-

Regulation of u-PAR Expression by an AP-2a-related Factor 4703

by guest on March 25, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 3: Transactivation of the Urokinase-type Plasminogen Activator

ing motifs all bearing one mismatch with the correspondingcanonical (AP-2, GCCNNNGGC; Sp1, RYYCCGCCCM) se-quences. The addition of a 100-fold excess of a consensus AP-2-containing oligonucleotide (Fig. 1B, lane 7 and 11) from the

human metallothionein IIa promoter sequence (44) eliminatedone of the shifted bands (*). In contrast, substitution of thisoligonucleotide at the AP-2 motif (CC to TT) prevented it fromcompeting for the shifted bands (Fig. 1B, lane 10). Increasing

FIG. 1. Binding of Sp1, Sp3, and an AP-2-related factor to footprinted region II of the u-PAR promoter. A, schematic representationof the footprinted region II of the u-PAR promoter (22) and the oligonucleotide used in EMSA in this figure. B, RKO nuclear extract (8 mg) wasincubated at 21 °C for 20 min with an end-labeled oligonucleotide (2154/2128 u-PAR) in the presence or absence of a 100-fold excess of competitorsequences. After this time, 2 mg of the indicated antibodies were added, and complexes were subsequently analyzed by gel electrophoresis. C, EMSAwas carried out as described for B with the exception that the amount of the Sp1 consensus sequence was varied. D and E, RKO nuclear extractwas incubated where indicated with an anti-AP-2aA, an anti-AP-2b, or an anti-AP-2g antibody (2 mg) or an equivalent amount of an unspecific IgGfor 50 min at 4 °C followed by protein A-agarose beads. Beads were subsequently removed by centrifugation, and treated and untreated nuclearextract was analyzed by EMSA as described for B. The data are typical of duplicate experiments.

Regulation of u-PAR Expression by an AP-2a-related Factor4704

by guest on March 25, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 4: Transactivation of the Urokinase-type Plasminogen Activator

amounts of a nonradioactive oligonucleotide bearing a consen-sus Sp1 motif caused a dose-dependent decrease in the inten-sity of two of the shifted bands (indicated with a brace andarrow) (Fig. 1C) while having little effect on the band (*)competed with the AP-2 motif-containing oligonucleotide.

The ability of the consensus Sp1 motif to compete for thebinding of nuclear-extracted proteins to the u-PAR promoterfootprinted region II oligonucleotide suggested that transcrip-tion factors recognized by these motifs were bound to the u-PAR promoter. To examine this possibility, “supershifting” ex-periments were carried out. The addition of an anti-Sp1-specific antibody to the RKO nuclear extract resulted (Fig. 1B,lane 4) in a slower migrating band (indicated with a line) witha concomitant decreased intensity of the complex (indicated bya brace) competed with the Sp1 consensus sequence. On theother hand, the addition of an antibody against Sp2 had noeffect (lane 5) on the migration pattern, while an antibodydirected at the DNA-binding domain of Sp3 completely abol-ished (Fig. 1B, lane 6) the shifted band with the intermediatemobility (arrow). These data suggested that the region of theu-PAR promoter footprinted with nuclear extract from a highu-PAR-expressing cell line (22) is bound with Sp1 and Sp3.

Since the fastest migrating complex evident in the EMSA(indicated by an asterisk) was competed with an AP-2 consen-sus motif (Fig. 1B), we hypothesized that the bound protein wasan AP-2 isoform. To test this hypothesis, two sets of experi-ments were carried out. First, RKO nuclear extract was mixedwith 2 mg of an anti-AP-2a antibody or an equal amount of IgGand subsequently with protein A-agarose beads. The superna-tant (depleted of AP-2a-immunoreactive proteins) was thenused in band shifting experiments. The fastest migrating band(*), which was competed with an excess of the AP-2 consensus,was practically abolished (Fig. 1D) by treatment of the nuclearextract with the anti-AP-2a antibody, whereas the bands rec-ognized by the Sp1 (brace) and Sp3 (arrow) antibodies wereunaffected. Further, the IgG control had no effect on the inten-sity or mobility of any of the shifted bands and antibodiesspecific for AP-2b, or AP-2g failed to deplete the RKO nuclearextract of the binding factor (Fig. 1E). Second, we determined ifauthentic AP-2a could bind to the u-PAR promoter region II. InEMSA using an oligonucleotide spanning 2154/2128, authen-tic AP-2a (Promega) gave rise to a shifted band (line) (Fig. 2),which could be competed with an excess of either the u-PARregion II oligonucleotide (2154/2128) or a consensus AP-2motif. The authentic AP-2a bound to the 2154/2128 probe hada different mobility from the retarded band (which was re-moved by immunoprecipitation with the anti-AP-2a antibody;Fig. 1D) observed using RKO nuclear extract (*).

To delineate the minimal sequence of footprinted region IIrequired for the binding of Sp1/Sp3 and the factor recognizedby the anti-AP-2a antibody, band-shifting experiments werecarried out with the u-PAR promoter oligonucleotide truncatedfrom either the 59- or 39-end (Fig. 3A). The removal of two basepairs at the 59 terminus (generating oligonucleotide 2152/2128) had little effect on the intensities of the Sp1- (brace) andSp3- (arrow) bound complexes when compared with probe2154/2128. However, a severe attenuation in the binding ofthese factors was apparent with further truncation from the59-end as evident with probe 2150/2128. These data suggestedthat nucleotides 59 of the nonconsensus Sp1 motif (2147/2138)are required for the optimal binding of Sp1 and Sp3 to region IIof the u-PAR promoter. On the other hand, the fastest migrat-ing band (*), which is recognized by the anti-AP-2a antibody,was unaffected by the removal of 59 nucleotides with bindingmaintained with probe 2143/2128. These data rule out thepossibility that an AP-2-like motif located further upstream

(2151/2143) is mediating the binding of this factor to region IIof the u-PAR promoter. While the binding of Sp1/Sp3 and theAP-2a antibody-reactive factor demonstrated different 59 re-quirements, the binding of these transcription factors showedidentical requirements for 39 sequences. Thus, the removal ofup to 5 base pairs from the 39 terminus of oligonucleotide2154/2130 (generating oligonucleotide 2154/2135) had littleeffect on transcription factor binding. However, the deletion ofan additional 3 base pairs from the 39-end (oligonucleotide2154/2138) completely abolished the binding of these threetranscription factors. Thus, sequences in the u-PAR promoterextending 39 to 2135 are required for the optimal binding ofSp1, Sp3, and the factor recognized by the anti-AP-2a antibody.

While the anti-AP-2a antibody “supershifted” (arrow) au-thentic AP-2a bound to the u-PAR promoter oligonucleotide2154/2128 (Fig. 2), in contrast, we were unable to detect a“supershift” of the fastest migrating band (using RKO nuclearextract) with this antibody (data not shown). However, thiscould be due to the presence of multiple complexes evident inthe EMSA masking a “supershifted” band. Thus, we repeatedthe EMSA using an oligonucleotide spanning 2147/2128 (Fig.3C). This probe is unable to bind Sp1/Sp3 as demonstratedfrom Fig. 3A. The addition of RKO nuclear extract to this probeyielded a slower migrating band (indicated with an asterisk),which was abolished with an excess of the unlabeled AP-2consensus oligonucleotide. However, the addition of the anti-AP-2a antibody, effective in supershifting authentic AP-2abound to the 2147/2128 probe (arrow), failed to produce a“supershifted” complex. Likewise, antibodies to other AP-2aisoforms did not yield a “supershift” of the RKO-derived nu-clear factor. Another possible explanation for the lack of a

FIG. 2. Footprinted region II of the u-PAR promoter bindsauthentic AP-2a. RKO nuclear extract (8 mg) or purified AP-2a (10 ng)was incubated at 21 °C for 20 min with radioactive oligonucleotidescorresponding to nucleotides 2154/2128 of the u-PAR promoter in thepresence or absence of a 100-fold excess of the indicated unlabeledcompetitor sequences. After this time, 2 mg of an anti-AP-2a antibody oran equivalent amount of an unspecific IgG was added to the reactionmixture where indicated. Binding complexes were subsequently ana-lyzed by EMSA. The data are typical of duplicate experiments.

Regulation of u-PAR Expression by an AP-2a-related Factor 4705

by guest on March 25, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 5: Transactivation of the Urokinase-type Plasminogen Activator

“supershift” using RKO nuclear extract is that there may be aninhibitor present in the extract. However, this is unlikely, sincethe addition of nuclear extract from this cell line to authenticAP-2a did not prevent a supershift of the latter in EMSAemploying the 2154/2128 probe (data not shown). These datasuggest that a DNA-binding factor related to AP-2a (hereafterreferred to as AP-2a-related factor), derived from RKO nuclearextract, is bound to region II of the u-PAR promoter sequence.

Binding of Authentic Sp1 and AP-2a to the u-PAR PromoterRegion II Is Mutually Exclusive—There is substantial overlapbetween the Sp1/Sp3 (2147/2138) and AP-2 motifs (2142/2134) in the footprinted region II of the u-PAR promoter. Thus,we considered it unlikely that Sp1 and the AP-2a-related factorwere simultaneously binding to the u-PAR oligonucleotideprobe. Nevertheless, to examine this possibility, the u-PARoligonucleotide spanning 2154/2128 was incubated with au-thentic Sp1 and authentic AP-2a proteins alone or in combina-tion. Binding of the oligonucleotide with the individual purifiedproteins resulted in slower migrating complexes (Fig. 4). How-ever, the simultaneous addition of both purified proteins tooligonucleotide 2154/2128 did not yield a ternary complex andin fact resulted in a decrease in binding of either protein. Thetopmost band represents material remaining in the well and

failed to resolve as a third band even with continuous electro-phoresis. These data would suggest that the binding of Sp1 andAP-2a to the u-PAR promoter sequence spanning 2154/2128 ismutually exclusive. However, it should be emphasized that thisconclusion is based on the use of authentic AP-2a rather thanthe AP-2a-related factor present in RKO nuclear extract.

Effect of Inhibiting Sp1/Sp3 Binding to Region II on u-PARPromoter Activation—To determine whether u-PAR promoteractivation was dependent on the binding of Sp1/Sp3 to regionII, G nucleotides at positions 2148, 2147, 2144, and 2142(within the Sp1-like motif) were substituted for T (Fig. 5A).EMSA using this substituted oligonucleotide indicated that theslower migrating bands identified as Sp1 (brace) and Sp3 (ar-row) by their reactivity with specific antibodies (see Fig. 1B)were markedly diminished in their intensity when comparedwith an oligonucleotide corresponding to the wild type u-PARpromoter sequence (Fig. 5B). In contrast, the intensity of thecomplex (*) recognized with the anti-AP-2a antibody (see Fig.1D) was not decreased by this change. RKO cells were thentransfected with a CAT reporter driven by either the wild typeu-PAR promoter (u-PAR CAT) or the promoter harboring themutations that reduced Sp1 and Sp3 binding to region II. Induplicate experiments, the activity of the mutated u-PAR pro-

FIG. 3. Identification of minimal sequences of footprinted region II required for binding of nucleus-derived transcription factorsfrom RKO cells. A, RKO nuclear extract (8 mg) was incubated at 21 °C for 20 min with end-labeled oligonucleotides with (1), or without (2) a100-fold excess of the indicated competitor or an oligonucleotide containing a consensus AP-2 motif (AP-2 consensus). Binding complexes wereresolved by EMSA as described in the legend to Fig. 1. The experiments were carried out three times. B, the nucleotide sequence of the u-PARpromoter, including the footprinted region II is shown. Underlined and overlined nucleotides indicate putative motifs identified by the GeneticsComputer Group program (Madison, WI). C, RKO nuclear extract (8 mg) or AP-2a (10–20 ng) was incubated at 21 °C for 20 min with a radioactiveoligonucleotide corresponding to nucleotides 2147/2128 of the u-PAR promoter in the presence or absence of a 100-fold excess of the unlabeledcompetitor sequences. After this time, 2 mg of the indicated antibody was added to the reaction mixture where indicated. Binding complexes weresubsequently analyzed by EMSA. The data are typical of duplicate experiments.

Regulation of u-PAR Expression by an AP-2a-related Factor4706

by guest on March 25, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 6: Transactivation of the Urokinase-type Plasminogen Activator

moter was similar to that achieved with the wild type u-PARpromoter (Fig. 5C).

Elimination of Sp1/Sp3 and AP-2a-related Factor Bindingto u-PAR Region II Abolishes Constitutive Promoter Activa-tion—We were unable to interfere with the binding of theAP-2a-related factor to region II of the u-PAR promoter with-out having a deleterious effect on Sp1/Sp3 binding, and thiswas consistent with the large overlap of these motifs and bandshifting experiments (see Fig. 3A), which revealed identical 39nucleotide requirements. We therefore determined the effect ofeliminating the binding of these three transcription factors onconstitutive u-PAR promoter activity. Nucleotide substitutions(Fig. 6A) of the Sp1-like and AP-2-like motifs, which prevented(Fig. 6B) these transcription factors binding to oligonucleotide2154/2128, were engineered into the u-PAR promoter CATreporter construct. The reporter construct was then comparedwith the wild type promoter for activation in the u-PAR-over-expressing RKO cells. We found in duplicate experiments thatthe activity of the mutated u-PAR promoter was reduced byover 90% in comparison with the wild type promoter construct(Fig. 6C). These data, combined with the observation that themutation of region II, which prevented the binding of Sp1/Sp3(but not the AP-2a-related factor), had minimal effect on theactivity of the u-PAR promoter (see Fig. 5C), implies thatbinding of the AP-2a-related factor to this region is critical forthe constitutive activity of this promoter in RKO cells.

Down-regulation of u-PAR Promoter Activity by the Co-ex-pression of either a Dominant Negative or an Antisense AP-2—As indicated above, any conclusions drawn as to the role ofthe AP-2a-related factor in the regulation of u-PAR promoteractivity were confounded by the fact that nucleotide substitu-tions of region II affecting the binding of this factor also dis-rupted the binding of Sp1 and Sp3. To circumvent this problem,we determined the effect of interfering with AP-2a on u-PARpromoter activity. RKO cells were transiently transfected withan AP-2aB expression construct. AP-2aB is an alternativelyspliced product of AP-2a and acts as a negative regulator ofAP-2 transcriptional activity (39). In three separate experi-ments, increasing amounts of the AP-2aB expression construct

caused a dose-dependent repression of u-PAR promoter activity(Fig. 7). An input of 0.5 mg of the dominant negative expressionconstruct brought about an 81 6 4% inhibition of u-PAR pro-moter activity compared with the empty expression vector(pSG5). Increasing the amount of AP-2aB DNA to 5 mg wasmarginally more effective, resulting in a 93 6 5% inhibition ofu-PAR promoter activity.

As an alternative to using the dominant negative expressionconstruct, parallel experiments were also carried out using anantisense AP-2a expression vector. Similar to the resultsachieved with the AP-2aB, the antisense expression constructcaused an inhibition of u-PAR promoter activity (data notshown), with 10 mg of DNA yielding a 90 6 14% repression ofu-PAR promoter activity when compared with the vector back-bone (pSG5). To rule out the possibility that interfering withAP-2a activity was having a general suppressive effect on tran-scription, RKO cells were transiently co-transfected with theAP-2aB expression construct and a CAT reporter driven by theurokinase promoter. The AP-2aB expression construct failed torepress the urokinase reporter construct using 1 mg of theeffector an amount that inhibited u-PAR promoter activity byover 90 6 7% (data not shown). Thus, it is unlikely that theeffect of AP-2aB on u-PAR promoter activity is due to a generalshut down of transcription.

To confirm that the repressive effect of AP-2aB on u-PARpromoter activity was mediated via the footprinted region II,two experiments were carried out. First, RKO cells were tran-siently co-transfected with an AP-2aB expression constructand a CAT reporter regulated by either 148 or 105 base pairs of59-flanking sequence of the u-PAR gene. Based on band shiftingexperiments (see Fig. 3A) the 2148 u-PAR promoter fragmentcan, via region II, bind the AP-2a-related factor, whereas theu-PAR promoter regulated by only 105 base pairs of upstream

FIG. 4. The binding of authentic AP-2a and Sp1 to region II ofthe u-PAR promoter is mutually exclusive. Authentic AP-2aA (10ng) and Sp1 (10 ng) were incubated alone or in combination at 21 °C for20 min with an end-labeled oligonucleotide (2154/2128) with or with-out a 100-fold excess of the unlabeled oligonucleotide. After this time,binding complexes were resolved by EMSA as described in the legend toFig. 1. The data are representative of two separate experiments.

FIG. 3—continued

Regulation of u-PAR Expression by an AP-2a-related Factor 4707

by guest on March 25, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 7: Transactivation of the Urokinase-type Plasminogen Activator

sequence cannot. It should be noted that Sp1/Sp3 binding ispreserved in the 2148 u-PAR construct due to the contributionof plasmid sequences. The AP-2aB expression construct causeda marked reduction (80%) in promoter activity using the re-porter construct flanked by 148 base pairs of upstream se-quence while bringing about only a modest repression (20%) ofthe reporter construct regulated by 105 base pairs of the u-PARregulatory sequence (data not shown). Second, RKO cells wereco-transfected with the AP-2aB expression construct and aCAT reporter regulated by either an oligonucleotide (2154/2128) spanning region II (R2 CAT) of the u-PAR promoter or398 base pairs of 59-flanking sequence (u-PAR CAT). The CATreporter regulated by region II (R2 CAT) was activated (Fig. 8)in RKO cells, albeit to a lesser extent than that achieved with398 base pairs of flanking sequence (u-PAR CAT). In two sep-arate experiments, the activity of the R2 CAT reporter con-struct was inhibited by over 75% by the co-expression of theAP-2aB when compared with an equivalent amount of theempty expression construct (pSG5). This inhibition was quan-titatively similar to that achieved using the u-PAR CAT re-porter. Taken together, these data suggest that the u-PARpromoter activity is indeed regulated by an AP-2a-related fac-tor and possibly Sp1/Sp3 in RKO cells and that this occurs viaregion II of the promoter.

Nuclear Extract from u-PAR-rich Cells Demonstrate In-creased Binding of the AP-2a-related Factor to the u-PAR Pro-moter Region II Compared with Nuclear Extract from Low

u-PAR-expressing Cells—Considering the evidence implicatingthe AP-2a-related factor in the regulation of u-PAR expressionin RKO cells, we speculated that nuclear extract derived fromcells rich in u-PAR protein would contain more of this tran-scription factor bound to region II of the u-PAR promoter com-pared with nuclear extract derived from cells characterized bytheir low u-PAR protein. Toward this end, we made use ofanother colon cancer cell line (GEO), which displays 10-foldfewer u-PARs compared with RKO as a consequence of reducedtranscription of the gene (22). Nuclear extracts were generatedfrom each cell line, and equal protein amounts were incubatedwith the radioactive oligonucleotide 2154/2128 in the presenceor absence of excess oligonucleotide competitors (Fig. 9). Bind-ing complexes were then analyzed by electrophoresis. Nuclearextract from GEO cells gave rise to a retarded band with amobility similar to that identified as the AP-2a-related factor(*) using RKO nuclear extract. An excess of the AP-2 consensussequence reduced the intensity of this band, consistent with thenotion that it represents an AP-2a-related factor-bound com-plex. Interestingly, the intensity of the complex (*) was reducedin nuclear extract from the low u-PAR-expressing GEO cellswhen compared with RKO cells, the latter of which display over10-fold more u-PAR. Equally important, treatment of GEOcells with PMA, which increases u-PAR gene transcription inthis cell line (22, 45), brought about a dramatic increase in theamount of this complex (*). In contrast, the intensity of theretarded band, which was indistinguishable from Sp3 (arrow),

FIG. 5. Effect of interfering with the binding of Sp1/Sp3 to footprinted region II on the constitutive activity of the u-PAR promoter.A, substituted nucleotides in the u-PAR promoter region II are indicated with asterisks. Underlined and overlined nucleotides indicate putativemotifs identified by the Genetics Computer Group program. B, RKO nuclear extract (8 mg) was incubated with an oligonucleotide spanningnucleotides 2154/2128 of the u-PAR promoter (2154/2128) or the corresponding sequence that had been substituted (G to T) at the indicatednucleotides. EMSA was performed as described in the legend to Fig. 1. C, RKO cells were transiently transfected with the wild type (u-PAR CAT)or mutated (Sp1/Sp3mt u-PAR CAT) u-PAR promoter (see A for nucleotide substitutions). After 48 h, the cells were harvested, lysed, and assayedfor CAT activity. Parallel dishes were transfected with pRSV CAT and pSV0CAT as positive and negative controls, respectively. The data arerepresentative of duplicate experiments.

Regulation of u-PAR Expression by an AP-2a-related Factor4708

by guest on March 25, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 8: Transactivation of the Urokinase-type Plasminogen Activator

was decreased by PMA treatment of GEO cells.To determine if the altered amounts of the region II-bound

factors were required for the stimulation of u-PAR promoteractivity by PMA, GEO cells were co-transfected with a CATreporter flanked by the wild type or mutated u-PAR promoterand a vector bearing AP-2aB and subsequently treated withthe phorbol ester. Treatment of the cells with PMA caused astrong (10–20-fold) induction of the wild type (u-PAR CAT)u-PAR promoter, which was prevented in cells made to co-express AP-2aB (Fig. 10). Mutation of the u-PAR promoter toprevent Sp1/Sp3 binding (Sp1/Sp3mt u-PAR CAT) reduced thestimulation by PMA. This stimulation was further reducedwhen nucleotide substitutions of the u-PAR promoter wereundertaken to abolish the binding of the AP-2a-related factoras well as Sp1/Sp3 (AP-2/Sp1/Sp3mt u-PAR CAT). Interest-ingly, the ability of the AP-2aB expression construct to repressu-PAR promoter activity was diminished by mutations thatprevented the binding of Sp1/Sp3 (Sp1/Sp3mt u-PAR CAT).Taken together, these data would suggest that PMA stimula-

tion of u-PAR gene expression in GEO cells requires, at least inpart, the binding of the AP-2a-related factor as well as Sp1/Sp3to region II of the promoter.

Stimulation of u-PAR Promoter Activity in the Low u-PAR-expressing GEO Cell Line by the Expression of Exogenous AP-2aA—While the above experiments suggested a requirementfor the AP-2a-related factor in the stimulation of u-PAR geneexpression by phorbol ester, it was not clear as to whether thistranscription factor alone was sufficient to augment u-PARpromoter activity. To address this issue, GEO cells, which havelow u-PAR protein, were co-transfected on three separate oc-casions with a u-PAR promoter-regulated CAT reporter and anexpression vector bearing the full-length form of AP-2a (AP-2aA). The activity of the promoter alone was below the detec-tion limit of the assay (Fig. 11), consistent with the low expres-sion of the u-PAR gene in this cell line. However, the co-transfection of AP-2aA into these cells caused a dose-dependent increase in u-PAR promoter activity with up to 0.05mg of the effector plasmid. This amount of the expression con-

FIG. 6. Interfering with the binding of the AP-2-related factor and Sp1/Sp3 to region II of the u-PAR promoter abolishes itsconstitutive activity in RKO cells. A, substituted nucleotides in region II of the u-PAR promoter sequence are indicated with asterisks.Underlined and overlined nucleotides indicate putative motifs identified by the Genetics Computer Group program. B, EMSA was carried out usingoligonucleotides corresponding to the wild type u-PAR sequence (nucleotides 2154/2128) or the mutated sequence (AP-2/Sp1/Sp3 mutant) andRKO nuclear extract. C, RKO cells were, at 60% confluence, transfected with a RSV-luciferase expression construct and a CAT reporter regulatedby the wild type or mutated u-PAR (as indicated in A) promoter sequence. Both promoter sequences were in the context of 398 base pairs of59-flanking sequence. After 48 h, the cell were harvested, lysed, and assayed for CAT activity after normalization for transfection efficiency. Theamount of acetylated chloramphenicol was determined using a Storm 840 PhosphorImager. The data are typical of duplicate experiments.

Regulation of u-PAR Expression by an AP-2a-related Factor 4709

by guest on March 25, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 9: Transactivation of the Urokinase-type Plasminogen Activator

struct brought about over a 13-fold stimulation of promoteractivity. Higher amounts of the AP-2aA diminished u-PARpromoter stimulation, presumably as a consequence of squelch-ing (46). The AP-2aA-dependent stimulation of the u-PAR pro-moter was greater than that achieved with 2 mg of a JunDexpression vector, which we had shown previously (22) to stim-ulate u-PAR expression via a separate footprinted region(2190/2171) of the promoter containing a classical AP-1 motif.The transfection of GEO cells with JunD and AP-2aA togetherstimulated u-PAR promoter activity to an extent greater thanthe sum of the individual expression constructs, indicative ofsynergism. Thus, it is likely that that these transcription fac-tors cooperate with each other to regulate u-PAR expression.Notwithstanding these observations, our data indicate that theexpression of AP-2aA is sufficient to up-regulate u-PAR geneexpression in GEO cells.

Expression of AP-2aB Decreases Endogenous u-PAR ProteinAmount in RKO Cells—We then determined if the expression ofAP-2aB, which acts as a negative regulator of AP-2 function,reduces the expression of the endogenous u-PAR gene in RKOcells. Cells were co-transfected with an expression vector en-coding a mutated CD4 and varying amounts of an expressionconstruct bearing AP-2aB. Cells were harvested 48 h later, andtransfected cells were enriched with magnetic beads coatedwith an anti-mutated CD4 antibody (which is non-cross-reac-

tive with wild type CD4) and assayed for u-PAR protein byWestern blotting (Fig. 12A). A band whose molecular mass wasindistinguishable from that of u-PAR (55 kDa) (47) was de-tected in the immunoblot. Increasing amounts of the AP-2aBexpression vector caused a dose-dependent decrease in theamount of this protein. The reduced amount of u-PAR proteinwas associated with the attenuated activity of a CAT reporterregulated by three tandem AP-2 motifs upstream of a thymi-dine kinase minimal promoter (Fig. 12B). Thus, the expressionof a negative regulator of AP-2 reduces the expression of theendogenous u-PAR gene in RKO cells.

Inhibition of u-PAR-directed Laminin Degradation in RKOCells Made to Express AP-2aB—One of the functions of theu-PAR is to accelerate plasminogen-dependent proteolysis (8),and this is a requirement for the invasive potential of a diver-gent set of cancers (31, 32, 48). We were therefore interested indetermining whether interfering with the transcriptional acti-vation of the gene leading to reduced u-PAR synthesis woulddiminish extracellular matrix degradation. RKO cells in se-rum-free medium (RKO-SF) demonstrated minimal solubiliza-tion of laminin (Fig. 12C). However, the addition of plasmino-gen to the RKO cells transfected with the vector backbone(RKO-pSG5-Pl-SF) resulted in a strong time-dependent in-crease in laminin degradation, indicating plasmin-dependentproteolysis. After a 2-h incubation in the presence of zymogen,

FIG. 7. The expression of a negative regulator of AP-2a down-regulates u-PAR promoter activity in RKO cells. RKO cells weretransiently transfected with a CAT reporter regulated by 398 base pairs of upstream sequence of the urokinase receptor promoter (u-PAR CAT)and the indicated amounts of an expression vector (pSG5) bearing AP-2aB (AP-2aB pSG5). Parallel dishes of RKO cells were transfected with pRSVCAT or pSV0CAT as positive and negative transfection controls. CAT assays were carried out as described in the legend to Fig. 5. Representativedata of triplicate experiments are shown. Significant differences are indicated as follows: *, p , 0.05; **, p , 0.01 for comparison of CAT activitiesachieved with the AP-2aB when compared with the equivalent amount of the empty expression construct.

Regulation of u-PAR Expression by an AP-2a-related Factor4710

by guest on March 25, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 10: Transactivation of the Urokinase-type Plasminogen Activator

nearly 500,000 cpm of solubilized laminin was evident in theculture supernatant. In contrast, RKO cells transfected withthe AP-2aB expression construct (RKO-AP-2B-Pl-SF) showedmarkedly reduced plasminogen-dependent degradation (80%reduction) of this glycoprotein after a 2-h incubation. Thesedata suggest that reduced u-PAR synthesis brought about by

interfering with the AP-2a-dependent transcriptional activa-tion of the u-PAR gene in RKO cells attenuates plasminogen-dependent proteolysis.

DISCUSSION

The u-PAR plays a critical role in extracellular matrix deg-radation and tumor invasion (34, 49). We have identified animportant regulatory region in the u-PAR promoter (2152/2135) that is trans-activated by an AP-2a-related factor. Fur-ther, our finding that the expression of AP-2aB, which inter-feres with the transcriptional activation of the u-PAR gene bythe AP-2a-related factor and Sp1/Sp3, diminishes u-PAR pro-tein amounts and laminin degradation serves to illustrate thattranscriptional studies of a target gene can be utilized to bringabout a more indolent phenotype of cancer at least in an ex-perimental setting.

Interestingly, in a previous report, Soravia et al. (26), usingHeLa nuclear extracts, failed to detect, by EMSA, the bindingof AP-2 to the urokinase receptor promoter. The reason for thisdifference is unclear at the present time. One possibility is thaturokinase receptor expression is regulated differently in theseparate cell lines used in the two studies.

The identity of the AP-2a-related factor bound to the u-PARpromoter region II is unknown at the present time. Certainly,it is strongly related to AP-2a, since depletion of nuclear ex-tract with an anti-AP-2a antibody and protein A-agarose beadsseverely diminished the intensity of the retarded band inEMSA and since the u-PAR promoter region II can clearly bindauthentic AP-2a. On the other hand, in EMSA it had a distinctmobility from authentic AP-2a, and the anti-AP-2a antibodywas unable to supershift the RKO nuclear factor bound to aregion II oligonucleotide while effecting a supershift with au-thentic AP-2a bound to this oligonucleotide. We were also un-able to supershift or immunodeplete the AP-2a-related factorfrom RKO nuclear extract using recently available antibodies(Santa Cruz Biotechnology) to AP-2b or AP-2g, making it un-likely that the RKO-derived nuclear factor was one of theseisoforms. Lee et al. (50) also noted an AP-2a-related factor inTHP-1 monocytes that regulated the expression of the B2 sub-unit of the V-ATPase. Similar to our study, the THP-1-derivedAP-2a-related factor was not supershifted in EMSA and had a

FIG. 8. Repression of u-PAR pro-moter activity by AP-2aB is mediatedthrough footprinted region II. RKOcells were transiently co-transfected witha CAT reporter regulated by either 398base pairs of flanking sequence (u-PARCAT) or by a sequence of the u-PAR pro-moter spanning nucleotides 2154/2128(R2 CAT) and the indicated amount of anexpression vector (pSG5) bearing AP-2aB(AP-2aB pSG5). CAT assays were per-formed as stated in the legend to Fig. 5.RKO cells were transfected with pRSVCAT and pSV0CAT as positive and nega-tive controls, respectively. The CAT assayof a representative experiment is shownwith the range of chloramphenicol conver-sions of duplicate experiments indicatedat the top.

FIG. 9. Nuclear extract from cells expressing high u-PAR lev-els intrinsically (RKO) or as a consequence of PMA treatment(GEO) contains more AP-2a-related factor bound to the u-PARfootprinted region II compared with nuclear extract from theuntreated GEO cells, which have few u-PAR. Nuclear extract (8mg) from 90% confluent cells treated with, or without, PMA (100 nM) for60 min was incubated with a radioactive oligonucleotide spanning nu-cleotides 2154 to 2128 of the u-PAR promoter. Where indicated, a100-fold excess of nonradioactive oligonucleotide competitors was in-cluded. Binding complexes were then analyzed by EMSA. The data arerepresentative of duplicate experiments.

Regulation of u-PAR Expression by an AP-2a-related Factor 4711

by guest on March 25, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 11: Transactivation of the Urokinase-type Plasminogen Activator

different size from authentic AP-2a based on its mobility in gelretardation studies. It remains to be determined whether theAP-2a-related factor found in the current study and in thestudy by Lee et al. (50) are the same.

In addition to the AP-2a-related factor recognized by foot-printed region II of the u-PAR promoter, both Sp1 and Sp3were also bound, as evident in EMSA using nuclear extractfrom the high u-PAR-expressing cell line RKO. However, wefound that mutations of the u-PAR promoter that preventedthe binding of either of these transcription factors had a neg-ligible effect on promoter activation, arguing against a role forthese factors in regulating at least the constitutive u-PARexpression in RKO cells.

The requirement of the u-PAR promoter region II for thePMA-dependent elevation of u-PAR gene expression (53) mer-its discussion. We had shown previously (22) that the inductionby this phorbol ester required an intact AP-1 motif (at 2184) ina separately footprinted region (2190/2171) of the u-PAR pro-moter. However, several observations in the current study

would indicate that the induction of the u-PAR promoter byPMA requires, in addition, other transcription factor bindingsites including the motif bound with the AP-2a-related factor.Thus, nucleotide substitutions of the u-PAR promoter thatprevented the binding of the AP-2a-related factor substantiallyreduced the stimulation of the promoter by the phorbol ester.Further, the co-expression of AP-2aB, which is a negative reg-ulator of AP-2 function, completely ablated the stimulation ofthe u-PAR promoter by PMA. Finally, an increased amount ofthe AP-2a-related factor bound to the u-PAR promoter region IIwas evident using nuclear extract from PMA-treated GEO cells(compared with nuclear extract from untreated GEO cells). Ourresults are reminiscent of other studies in which the stimula-tion of the PAC-1 (phosphatase of activated cells) phosphataseand neuropeptide tyrosine genes by PMA was shown to bemediated partly through an AP-2-related site and coincidedwith induced DNA binding of AP-2 (54, 55). It was also clearthat interfering with the binding of Sp1/Sp3 (but not affectingthe binding of the AP-2a-related factor) to region II of the

FIG. 10. The binding of the AP-2a-related factor and Sp1/Sp3 to regionII are required for the stimulation ofu-PAR promoter activity by PMA inGEO cells. GEO cells were co-transfectedwith a CAT reporter driven by the wildtype u-PAR promoter (u-PAR CAT) or thepromoter that had nucleotide substitu-tions in region II to prevent the binding ofSp1/Sp3 (Sp1/Sp3mt u-PAR CAT) or AP-2/Sp1/Sp3 (AP-2/Sp1/Sp3mt u-PAR CAT)and an expression vector (pSG5) encodingAP-2aB (AP-2aB pSG5). Following trans-fection, cells were replenished with freshmedium supplemented with or withoutPMA and cultured for an additional 2days. Cells were then harvested, lysed,and assayed for CAT activity as describedin the legend to Fig. 5. The data are typ-ical of two separate experiments.

FIG. 11. Stimulation of u-PAR pro-moter activity in GEO cells made toco-express AP-2aA. GEO cells weretransiently co-transfected with a u-PARpromoter-regulated CAT and an expres-sion vector (pSG5) bearing either JunD orAP-2aA. Parallel cultures were trans-fected with pSV0CAT or pRSV CAT asnegative and positive controls, respec-tively. After 48 h, the cells were lysed andassayed for CAT activity as described inthe legend to Fig. 5. The data are repre-sentative of triplicate experiments. Chlor-amphenicol conversions are shown as av-erage values 6 S.D. for the threeexperiments. An asterisk indicates thatdifferences in chloramphenicol conver-sions achieved by the expression of theJunD and/or the AP-2aA when comparedwith the empty expression construct(pSG5) were statistically significant (p ,0.05).

Regulation of u-PAR Expression by an AP-2a-related Factor4712

by guest on March 25, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 12: Transactivation of the Urokinase-type Plasminogen Activator

u-PAR promoter had a deleterious effect on the activation ofthe u-PAR promoter by PMA. A similar requirement for Sp1has been reported for the stimulation of thromboxane receptorgene expression by PMA (56), although, in contrast to thatstudy, we did not detect increased binding of this transcriptionfactor. Taken together, it is likely that the stimulation of theu-PAR gene expression by phorbol ester is complex, requiringthe interactions of multiple transcription factors with theircognate binding sites in the upstream sequence of this gene.

The mechanism by which the AP-2aB alters u-PAR promoteractivity deserves comment. Initially, we assumed that this wasthrough a direct antagonism of the AP-2a-like factor bound to

the u-PAR promoter region II, presumably as a consequence ofheterodimerization of the two proteins (39). However, one ex-periment suggested that this proposal might not be entirelyaccurate. Thus, the ability of the AP-2aB to counter the stim-ulation of u-PAR promoter activity by PMA was diminished bymutations in the u-PAR promoter that prevented the binding ofSp1/Sp3. These data suggested that the expressed AP-2aB wasmediating its effect, at least in part, through the Sp1/Sp3-binding motif.

Our data based on mobility shift assays using purified pro-teins strongly suggested that the binding of at least AP-2aAand Sp1 to the u-PAR promoter region II is mutually exclusive,

FIG. 12. Reduced amount of u-PAR protein and laminin degradation by RKO cells made to express AP-2aB. A, RKO cells wereco-transfected with the indicated amount of the expression vector encoding AP-2aB or the vector backbone (pSG5) alone along with an expressionconstruct bearing a mutated CD4. After 48 h, the cells were harvested and assayed for u-PAR protein by Western blotting. 2ve Control, no cellextract. B, RKO cells were co-transfected with an expression vector (pSG5) bearing AP-2aB (AP-2aB) along with a CAT reporter regulated eitherby three AP-2 tandem repeats upstream of a thymidine kinase minimal promoter (AP-2 pBLCAT2) or the minimal promoter alone (pBLCAT2).After 48 h, the cells were lysed and assayed for CAT activity. C, RKO cells transfected and harvested as described for A were plated in serum-freemedium on radioactive laminin-coated dishes. After cell attachment, surface u-PAR were saturated with exogenous urokinase, and the cultureswere washed extensively and then replenished with serum-free medium supplemented with or without plasminogen. Aliquots of the culturesupernatant were withdrawn at the indicated times thereafter and counted for radioactivity. After 2 h, cells were enumerated. The data are typicalof duplicate experiments. l, serum-free medium only; -, serum-free medium plus plasminogen; Œ, RKO-serum-free medium; f, RKO-pSG5-plasminogen-serum-free medium; Ô, RKO-AP-2B-serum-free medium; ●, RKO-AP-2B-plasminogen-serum-free medium.

Regulation of u-PAR Expression by an AP-2a-related Factor 4713

by guest on March 25, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 13: Transactivation of the Urokinase-type Plasminogen Activator

and this is consistent with the considerable (about 50%) over-lap of the Sp1/Sp3 and AP-2 motifs. Thus, it is unlikely that theAP-2a-related factor and Sp1 are physically interacting on theu-PAR promoter region II sequence. The biological significanceof having two overlapping transcription factor binding motifs inthe u-PAR promoter region II, which can be bound with onlyone transcription factor at any one time, can only be speculatedon. Certainly, for the regulation of keratin 3 gene expression incorneal epithelial cells, overlapping AP-2 and Sp1 motifs act asa switch to up-regulate expression of this gene in response tothe latter transcription factor (57). For the u-PAR promoter,since this motif can be bound with Sp3, which for some genes isa trans-repressor (58), it may be that this represents a mech-anism for either up- or down-regulating u-PAR expression.

In conclusion, we have shown that the constitutive and PMA-inducible expression of the u-PAR gene requires, at least inpart, the trans-activation of a region of the promoter spanning2152/2135 by an AP-2a-related factor. A high u-PAR proteinlevel in colon cancer, which promotes the invasive phenotype(31, 32) and portends a poor patient outcome (30), is probablypartly a consequence of trans-activation of this gene by agreater amount and/or binding activity of the AP-2a-relatedfactor. Further, interfering with the trans-activation of thepromoter by the AP-2a-related factor reduced endogenous u-PAR expression and diminished laminin degradation. Thesefindings raise the exciting possibility that interfering with theAP-2a-related factor-dependent trans-activation of the u-PARgene may represent a novel means of diminishing extracellularmatrix degradation and consequently reducing colon cancerinvasion and metastasis.

Acknowledgments—Expression constructs encoding AP-2aA,AP-2aB, the antisense AP-2, and the AP-2 CAT reporter were kindlyprovided by Dr. Michael Tainsky (Department of Tumor Biology, M.D.Anderson Cancer Center, Houston, TX). The urokinase CAT reporterwas kindly provided by Dr. Francesco Blasi (University of Milan, Italy).We thank Drs. Michael Tainsky, Ernest Lengyel (Department of Gyn-ecology and Obstetrics, Technical University of Munich, Germany),Christian Simon (Department of Ears, Nose, and Throat, University ofTubingen, Germany), and Jose Juarez (Department of Urology, Univer-sity of California, Davis School of Medicine) for excellent intellectualand technical input. We thank Prof. F. W. Schildberg (Department ofSurgery, Klinikum Grosshadern, Ludwig-Maximilians University ofMunich, Germany) for input. Finally, we are grateful to Drs. TimSchaeffer and Francois Claret for critical appraisal of the manuscript.

REFERENCES

1. Nielsen, L., Hansen, J., Skriver, L., Wilson, E., Kaltoft, K., Zenthen, J., andDano, K. (1982) Biochemistry 21, 6410–6415

2. Robbins, K. C., Summaria, L., Hsieh, B., and Shah, R. (1967) J. Biol. Chem.242, 2333–2342

3. Tryggvason, K., Hoyhtya, M., and Salo, T. (1987) Biochim. Biophys. Acta 907,191–217

4. Stoppelli, M. P., Tacchetti, C., Cubellis, M., Corti, A., Hearing, V., Cassani, G.,Appella, E., and Blasi, F. (1986) Cell 45, 675–684

5. Vassalli, J., Baccino, D., and Belin, D. (1985) J. Cell Biol. 100, 86–926. Behrendt, N., Ploug, M., Patthy, L., Houen, G., Blasi, F., and Dano, K. (1991)

J. Biol. Chem. 266, 7842–78477. Riittinen, L., Limongi, P., Crippa, M. P., Conese, M., Hernandez-Marrero, L.,

Fazioli, F., and Blasi, F. (1996) FEBS Lett. 381, 1–68. Ellis, V., Behrendt, N., and Dano, K. (1991) J. Biol. Chem. 266, 12752–127589. Higazi, A., Cohen, R., Henkin, J., Kniss, D., Schwartz, B. S., and Cines, D.

(1995) J. Biol. Chem. 270, 17375–1738010. Ginestra, A., Monea, S., Seghezzi, G., Dolo, V., Nagase, H., Mignatti, P., and

Vittorelli, M. (1997) J. Biol. Chem. 272, 17216–1722211. Cubellis, M., Wun, T., and Blasi, F. (1990) EMBO J. 9, 1079–108512. Conese, M., Olson, D., and Blasi, F. (1994) J. Biol. Chem. 269, 17886–1789213. Wei, Y., Lukashev, M., Simon, D. I., Bodary, S. C., Rosenberg, S., Doyle, M. V.,

and Chapman, H. A. (1996) Science 273, 1551–155514. Yebra, M., Parry, G. C. N., Stromblad, S., Mackman, N., Rosenberg, S.,

Mueller, B. M., and Cheresh, D. A. (1996) J. Biol. Chem. 271, 29393–2939915. Bohuslav, J., Horejsi, V., Hansmann, C., Stockl, J., Weidle, U. H., Majdic, O.,

Bartke, I., Knapp, W., and Stockinger, H. (1995) J. Exp. Med. 181,1381–1390

16. Resnati, M., Guttinger, M., Valcamonica, S., Sidenius, N., Blasi, F., andFazioli, F. (1996) EMBO J. 15, 1572–1582

17. Sillaber, C., Baghestanian, M., Hofbauer, R., Virgolini, I., Bankl, H. C.,Fureder, W., Agis, H., Willheim, M., Leimer, M., Scheiner, O., Binder, B. R.,Kiener, H. P., Bevec, D., Fritsch, G., Majdic, O., Kress, H. G., Gadner, H.,Lechner, K., and Valent, P. (1997) J. Biol. Chem. 272, 7824–7832

18. Vagnarelli, P., Raimondi, E., Mazzieri, R., De Carli, L., and Mignatti, P. (1992)Cytogenet. Cell Genet. 60, 197–199

19. Casey, J. R., Petranka, J. G., Kottra, J., Fleenor, D. E., and Rosse, W. F. (1994)Blood 84, 1151–1156

20. Roldan, A., Cubellis, M., Masucci, M., Behrendt, N., Lund, L., Dano, K.,Appella, E., and Blasi, F. (1990) EMBO J. 9, 467–474

21. Pyke, C., Eriksen, J., Solberg, H., Schnack, B., Nielsen, S., Kristensen, P.,Lund, L. R., and Dano, K. (1993) FEBS Lett. 326, 69–74

22. Lengyel, E., Wang, H., Stepp, E., Juarez, J., Wang, Y., Doe, W., Pfarr, C. M.,and Boyd, D. (1996) J. Biol. Chem. 271, 23176–23184

23. Shetty, S., Kumar, A., and Idell, S. (1997) Mol. Cell. Biol. 17, 1075–108324. Lund, L. R., Ellis, V., Ronne, E., Pyke, C., and Dano, K. (1995) Biochem. J. 310,

345–35225. Nykjaer, A., Conese, M., Cremona, O., Gliemann, J., and Blasi, F. (1997)

EMBO J. 16, 2610–262026. Soravia, E., Grebe, A., De Luca, P., Helin, K., Suh, T. T., Degen, J. L., and

Blasi, F. (1995) Blood 86, 624–63527. Carmeliet, P., Moons, L., Dewerchin, M., Rosenberg, S., Herbert, J., Lupu, F.,

and Collen, D. (1998) J. Cell Biol. 140, 233–24528. Pyke, C., Salo, S., Ralfkiaer, E., Romer, J., Dano, K., and Tryggvason, K. (1995)

Cancer Res. 55, 4132–413929. Pyke, C., Kristensen, P., Ralfkiaer, E., Grondahl-Hansen, J., Eirksen, J., Blasi,

F., and Dano, K. (1991) Am. J. Pathol. 138, 1059–106730. Ganesh, S., Sier, C. F. M., Heerding, M. M., Griffioen, G., Lamers, C. B. H., and

Verspaget, H. W. (1994) Lancet 344, 401–40231. Kariko, K., Kuo, A., Boyd, D., Okada, S., Cines, D., and Barnathan, E. (1993)

Cancer Res. 53, 3109–311732. Kook, Y. H., Adamski, J., Zelent, A., and Ossowski, L. (1994) EMBO J. 13,

3983–399133. Xing, R. H., Mazar, A., Henkin, J., and Rabbani, S. A. (1997) Cancer Res. 57,

3585–359334. Quattrone, A., Fibbi, G., Anichini, E., Pucci, M., Zamperini, A., Capaccioli, S.,

and Del Rosso, M. (1995) Cancer Res. 55, 90–9535. Min, H. Y., Doyle, L. V., Vitt, C. R., Zandonella, C. L., Stratton-Thomas, J. R.,

Shuman, M. A., and Rosenberg, S. (1996) Cancer Res. 56, 2428–243336. Wilhelm, O., Weilde, U., Rettenberger, S., Schmitt, M., and Graeff, H. (1994)

FEBS Lett. 337, 131–13437. Wang, Y., Dang, J., Johnson, L. K., Selhamer, J. J., and Doe, W. F. (1995) Eur.

J. Biochem. 227, 116–12238. Verde, P., Boast, S., Franze, A., Robbiati, F., and Blasi, F. (1988) Nucleic Acids

Res. 16, 10699–1071539. Buettner, R., Kannan, P., Imhof, A., Bauer, R., Yim, S. O., Glockshuber, R.,

Van Dyke, M. W., and Tainsky, M. A. (1993) Mol. Cell. Biol. 13, 4174–418540. Luckow, B., and Schutz, G. (1987) Nucleic Acids Res. 15, 5490–549041. Nead, M. A., and McCance, D. J. (1995) J. Invest. Dermatol. 105, 668–67142. Burnette, W. (1981) Anal. Biochem. 112, 195–20343. Schlechte, W., Murano, G., and Boyd, D. D. (1989) Cancer Res. 49, 6064–606944. Williams, T., Admon, A., Luscher, B., and Tjian, R. (1988) Genes Dev. 2,

1557–156945. Wang, H., Skibber, J., Juarez, J., and Boyd, D. (1994) Int. J. Cancer 58,

650–65746. Kannan, P., Buettner, R., Chiao, P. J., Yim, S. O., Sarkiss, M., and Tainsky,

M. A. (1994) Genes Dev. 8, 1258–126947. Nielsen, L., Kellerman, G. M., Behrendt, N., Picone, R., Dano, K., and Blasi, F.

(1988) J. Biol. Chem. 263, 2358–236348. Bruckner, A., Filderman, A. E., Kirchheimer, J. C., Binder, B. R., and Remold,

H. G. (1992) Cancer Res. 52, 3043–304749. Ossowski, L. (1992) Cancer Res. 52, 6754–676050. Lee, B. S., Krits, I., Crane-Zelkovic, M. K., and Gluck, S. L. (1997) J. Biol.

Chem. 272, 174–18151. Zhao, L., and Chang, L.-S. (1997) J. Biol. Chem. 272, 4869–488252. Long, F., Sonenshein, G. E., and Linsenmayer, T. F. (1998) J. Biol. Chem. 273,

6542–654953. Lund, L., Ronne, E., Roldan, A., Behrendt, N., Romer, J., Blasi, F., and Dano,

K. (1991) J. Biol. Chem. 266, 5177–518154. Grumont, R. J., Rasko, J. E. J., Strasser, A., and Gerondakis, S. (1996) Mol.

Cell. Biol. 16, 2913–292155. Andersson, G., Pahlman, S., Parrow, V., Johansson, I., and Hammerling, U.

(1994) Cell Growth Differ. 5, 27–3656. D’Angelo, D. D., Oliver, B. G., Davis, M. G., McCluskey, T. S., and Dorn, G. W.,

II (1996) J. Biol. Chem. 271, 19696–1970457. Chen, T.-T., Wu, R.-L., Castro-Munozledo, F., and Sun, T.-T. (1997) Mol. Cell.

Biol. 17, 3056–306458. Hagen, G., Muller, S., Beato, M., and Suske, G. (1994) EMBO J. 13, 3843–3851

Regulation of u-PAR Expression by an AP-2a-related Factor4714

by guest on March 25, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 14: Transactivation of the Urokinase-type Plasminogen Activator

Nyormoi and Douglas BoydHeike Allgayer, Heng Wang, Yao Wang, Markus M. Heiss, Reinhard Bauer, Okot

-related Factorαthrough a Novel Promoter Motif Bound with an Activator Protein-2Transactivation of the Urokinase-type Plasminogen Activator Receptor Gene

doi: 10.1074/jbc.274.8.47021999, 274:4702-4714.J. Biol. Chem. 

  http://www.jbc.org/content/274/8/4702Access the most updated version of this article at

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

  http://www.jbc.org/content/274/8/4702.full.html#ref-list-1

This article cites 58 references, 38 of which can be accessed free at

by guest on March 25, 2018

http://ww

w.jbc.org/

Dow

nloaded from