The Promyelocytic Leukemia Protein Interacts with Sp1 and Inhibits Its ...

3 downloads 75 Views 2MB Size Report
Jun 7, 1998 - target promoters. The promyelocytic leukemia gene, PML, was first identified at the breakpoint of the t(15;17) translocation in acute promy-.
MOLECULAR AND CELLULAR BIOLOGY, Dec. 1998, p. 7147–7156 0270-7306/98/$04.0010 Copyright © 1998, American Society for Microbiology. All Rights Reserved.

Vol. 18, No. 12

The Promyelocytic Leukemia Protein Interacts with Sp1 and Inhibits Its Transactivation of the Epidermal Growth Factor Receptor Promoter SADEQ VALLIAN,1 KHEW-VOON CHIN,2

AND

KUN-SANG CHANG1*

Division of Laboratory Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030,1 and University of Medicine and Dentistry of New Jersey, Piscataway, New Jersey 088542 Received 14 April 1998/Returned for modification 7 June 1998/Accepted 19 August 1998

The promyelocytic leukemia protein (PML) is a nuclear phosphoprotein with growth- and transformationsuppressing ability. Having previously shown it to be a transcriptional repressor of the epidermal growth factor receptor (EGFR) gene promoter, we have now shown that PML’s repression of EGFR transcription is caused by inhibition of EGFR’s Sp1-dependent activity. On functional analysis, the repressive effect of PML was mapped to a 150-bp element (the sequences between 2150 and 216, relative to the ATG initiation site) of the promoter. Transient transfection assays with Sp1-negative Drosophila melanogaster SL2 cells showed that the transcription of this region was regulated by Sp1 and that the Sp1-dependent activity of the promoter was suppressed by PML in a dose-dependent manner. Coimmunoprecipitation and mammalian two-hybrid assays demonstrated that PML and Sp1 were associated in vivo. In vitro binding by means of the glutathione S-transferase (GST) pull-down assay, using the full-length and truncated GST-Sp1 proteins and in vitrotranslated PML, showed that PML and Sp1 directly interacted and that the C-terminal (DNA-binding) region of Sp1 and the coiled-coil (dimerization) domain of PML were essential for this interaction. Analysis of the effects of PML on Sp1 DNA binding by electrophoretic mobility shift assay (EMSA) showed that PML could specifically disrupt the binding of Sp1 to DNA. Furthermore, cotransfection of PML specifically repressed Sp1, but not the E2F1-mediated activity of the dihydrofolate reductase promoter. Together, these data suggest that the association of PML and Sp1 represents a novel mechanism for negative regulation of EGFR and other Sp1 target promoters.

The promyelocytic leukemia gene, PML, was first identified at the breakpoint of the t(15;17) translocation in acute promyelocytic leukemia (APL) (10, 14, 19, 35, 37). PML encodes a nuclear phosphoprotein that functions as a transcriptional regulator (9, 50, 58) and belongs to the RING family of proteins, which share a cysteine-rich motif at the N terminus. This motif is divided into a RING finger (C3C4 zinc binding) motif and two B-box (B1 and B2) motifs (18). This region is followed by a predicted a-helical coiled-coil (dimerization) domain, which allows PML to homodimerize and form heterodimer complexes with the APL fusion protein PMLRARa and the promyelocytic leukemia zinc finger (PLZF) protein (37, 40). PML localizes to distinct domains in the nucleus called PML nuclear bodies, or PML oncogenic domains (PODs) (16, 60). In addition to PML, there are several other POD-associated factors, including SP100, the ubiquitin-like protein PIC1, and the interferon-stimulated 20-kDa gene product called ISG20 (3, 6, 20). PODs are frequently targeted and/or reorganized by viral proteins, such as the herpes simplex virus type 1 (HSV-1) gene product Vmw110 (17), the adenoviral proteins E1A and E4ORF3 (8), the Epstein-Barr virus-encoded nuclear antigen EBNA-5 (53), and the human cytomegalovirus major immediate-early proteins IE1 and IE2 (1). PMLRARa, which retains the cysteine-rich motif and the dimerization domain of PML and the DNA-binding and ligand-binding domains of retinoic acid receptor a (RARa), has

been shown to play a direct role in POD morphology and hence APL leukemogenesis in vitro (16, 37, 60). Treatment of APL cells with all-trans-retinoic acid (ATRA) restores PMLcontaining PODs, apparently by degradation of the PMLRARa fusion protein and hence induces terminal differentiation (16, 60). Furthermore, in line with the in vitro studies, transgenic results have shown that the expression of PMLRARa plays a critical role in the development of leukemia in mice (7, 22, 26). The results from our studies and others have shown that PML functions as a growth suppressor (2, 25, 39, 43, 47, 50), presumably by inducing G1 cell cycle arrest and apoptosis (43). Interestingly, the domains of PML that mediate its association with PODs have also been found to be involved in its growth suppression function (44). Recently, the growth suppressor function of PML was conclusively demonstrated by PML gene knockout (59). Although the mechanism through which PML suppresses cellular growth and transformation is unknown, recent studies have shown that PML is involved in regulating transcription of certain genes in either a positive or negative manner. In particular, we have demonstrated previously that PML can repress transcription of the epidermal growth factor receptor (EGFR) and multidrug resistance 1 (MDR1) promoters (50, 58). Analysis of transcriptional repression of PML, by means of the GAL4 fusion assay, localized the repressive effects of PML mainly to the coiled-coil (dimerization) domain (58). PML has also been reported to enhance the transactivation properties of the progesterone receptor (24). Recently, we have found that PML is associated with the AP-1 complex and is able to upregulate Fos-mediated transcriptional activity. Although no direct interaction between PML and Fos was detected, it was found that the stimulation of transcriptional

* Corresponding author. Mailing address: The University of Texas M. D. Anderson Cancer Center, Division of Laboratory Medicine, Houston, TX 77030. Phone: (713) 792-2581. Fax: (713) 794-1800. E-mail: [email protected]. 7147

7148

VALLIAN ET AL.

activity of Fos required the RING finger and the B1-box motifs of PML and the C-terminal domain of Fos (57). Moreover, PML was recently shown to interact with the retinoblastoma protein (pRb) in vivo and in vitro (2). Functional analysis of this PML-Rb interaction revealed that PML can inhibit Rbmediated transactivation of the glucocorticoid receptor transcription, providing further evidence for the involvement of PML in regulation of transcription. Our previous study demonstrated that PML suppresses the promoter activity of the EGFR gene (50). This promoter element is GC rich, contains multiple Sp1-binding sites, and lacks both TATA and CAAT boxes (28, 31). The promoter activity is regulated by a number of factors, including epidermal growth factor (EGF), cyclic AMP, and 12-O-tetradecanoylphorbol 13-acetate (27). Several transcription factors (e.g., p53 and Sp1) activate the promoter (13, 34), whereas others such as the ligand-activated thyroid hormone (T3R) and retinoic acid receptors (RARs) repress it (61). EGFR overexpression is associated with several malignancies (e.g., breast, colon, ovarian, and head and neck cancers), suggesting an important role for EGFR expression in growth and differentiation (48, 54). For the present study, the 59 proximal region of the EGFR promoter was characterized to search for factors that mediate transcriptional repression of the EGFR promoter activity by PML. Results from this study demonstrated that the repressive effects of PML are mapped to the sequences between 2150 and 216 of the EGFR promoter, which comprise the majority of the basal promoter activity mediated mainly by the transcription factor Sp1 through the Sp1-binding sites (31, 34). We found that this Sp1-dependent activity was inhibited by PML in a dose-dependent manner. In vitro and in vivo binding assays demonstrated that PML and Sp1 are associated through specific domains. Furthermore, our study indicated that interaction of PML with Sp1 disrupted its ability to bind DNA. Thus, repression of EGFR promoter activity by PML is most likely caused by inhibition of its Sp1-dependent activity, which could represent a novel mechanism for negative regulation of the EGFR promoter. MATERIALS AND METHODS Plasmids. Full-length and mutant GST-Sp1 in a pGex2TKMSC expression vector and pRCE2F1 were kindly provided by E. Wintersberger (Universita¨t Wien, Vienna, Austria) (36). The pCDNA3/Sp1 expression vector used for in vitro translation was constructed by subcloning an XhoI-SmaI fragment from pGex2TKMCS/Sp1 into an XhoI-EcoRV-digested pCDNA3 plasmid (Invitrogen, San Diego, Calif.). The pPac0, pPac-Sp1, and pPac-b-gal vectors, which contain the Drosophila b-actin promoter and polyadenylation sequences, were obtained from R. Tjian (33). pPac-PML was constructed by subcloning a BamHIBglII fragment containing the full-length PML cDNA from the pFM211 vector (50) into the BamHI site in pPac0. pPac-E2F1 was generated by subcloning the full-length E2F1 cDNA as a BamHI-SalI (partial digestion) fragment from pVP16/E2F1 into the BamHI-XhoI site in pPac0. pVP16/E2F1 was a kind gift from H. Rotheneder (Universita¨t Wien). PMLRARa cDNA expressed from a pSG5 plasmid under the control of the simian virus 40 (SV40) early promoter and enhancer was a gift from P. Chambon (37). PML mutants used in in vitro translations were constructed as described previously and were also expressed from pSG5 plasmid (44). The construction of EGFR promoter deletion mutants was described previously (34). The pDHFR-CAT reporter plasmid, which contains the hamster dihydrofolate reductase (DHFR) gene promoter linked to the chloramphenicol acetyltransferase (CAT) gene, was obtained from D. G. Johnson (5, 32). The pCMV-CAT reporter construct contains the cytomegalovirus minimal promoter in front of the CAT gene in a pCDNA3 vector (50). The p4x(UAS)-Luc vector, which contains four GAL4 DNA-binding sites (upstream activation sequences) in front of the thymidine kinase minimal promoter linked to the firefly luciferase gene, was obtained from M.-J. Tsai (Department of Cell Biology, Baylor College of Medicine, Houston, Tex.). The expression vectors used in two-hybrid assays were pK3VP16/PML and pHKG/Sp1. pK3VP16/PML, which expresses a VP16-PML fusion protein, was constructed by in-frame subcloning of PML cDNA as a BamHI-BglII fragment from the pFM211 vector (described above) into the BamHI site in pK3Vp16 vector, downstream of the sequences of the activation domain of the VP16 transcription factor. pHKG/Sp1 was constructed by subcloning the Sp1 cDNA as an XhoI-SmaI fragment (see

MOL. CELL. BIOL. above) into the SalI-XbaI (blunt ended) site in the pHKG4 vector, downstream of the DNA-binding domain of GAL4 transcription factor. pK3Vp16 and pHKG4 vectors were kindly provided by T. Kouzarides (Cambridge University, Cambridge, United Kingdom) (4). Cell culture. SW13 human adenocortical carcinoma and U2OS human osteosarcoma cells were maintained in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 100 U of penicillin per ml, and 100 mg of streptomycin per ml (GIBCO/BRL, Gaithersburg, Md.) in 5% CO2 at 37°C. Drosophila melanogaster SL2 cells were cultured at 24°C in Schneider’s Drosophila medium (GIBCO/BRL) supplemented with 10% heatinactivated FBS and antibiotics as described above. Transfection, mammalian two-hybrid assays, and CAT assays. For gene transfection experiments, SL2 cells were seeded at approximately 5 3 106 per 60mm-diameter dish 24 h before transfection. Cells were transfected by a calcium phosphate coprecipitation method as described previously (58). The quantities of plasmids used for transfections were as indicated in the legends to the figures. The total amount of DNA was adjusted to 15 mg with sheared and denatured salmon sperm DNA and sufficient amounts of pPac0 plasmid (containing the Drosophila b-actin promoter) to maintain a constant promoter level. The pPacb-gal plasmid (100 ng) (b-galactosidase expression vector) was included in each experiment to monitor transfection efficiency. Twenty-four hours after transfection, cells were harvested by pipetting the medium up and down several times, transferred into 15-ml tubes, centrifuged at 1,000 3 g for 5 min, washed two times in phosphate-buffered saline, transferred into Eppendorf tubes, and resuspended in 150 ml of 0.25 M Tris-HCl (pH 7.8). Cells were lysed by three cycles of freeze-thawing, and the clarified supernatants were used for b-galactosidase and CAT assays as described previously (58). SW13 and U2OS cells were transfected at semiconfluence (50 to 70%) by calcium phosphate coprecipitation with the quantities of plasmids indicated in the legends to the figures. The total amounts of transfected DNA were adjusted to 20 mg with salmon sperm DNA and sufficient amounts of pSG5 vector. To monitor transfection efficiency, 1 mg of pCMV–b-galactosidase expression plasmid was included in each transfection. Approximately 16 h after transfection, the precipitate was washed. Cells were then fed with fresh medium for an additional 24 h, harvested, and lysed. The amounts of extracts used for CAT assays were then normalized with respect to the b-galactosidase activity as described previously (58). The CAT activities were quantitated with a PhosphorImager (Bio-Rad Laboratories, Hercules, Calif.). In all experiments, each transfection was repeated at least twice. The CAT assay results presented are from typical experiments. In mammalian two-hybrid assays, U2OS cells were cotransfected by calcium phosphate coprecipitation with the quantities of plasmids indicated in the legend to Fig. 3. In each transfection, 1 mg of pCMV–b-galactosidase expression plasmid was included to monitor the transfection efficiency. The luciferase assay was performed with the Promega (Madison, Wis.) luciferase assay system according to the supplier’s instructions. The luciferase activity was measured with a Luminometer (Turner Design, Sunnyvale, Calif.) and normalized against b-galactosidase activity. Immunoprecipitation and immunoblotting. In immunoprecipitation experiments, approximately 400 mg of nuclear proteins from HeLa cells transiently overexpressing PML was diluted to 1 mg/ml in radioimmunoprecipitation assay (RIPA) buffer (140 mM NaCl, 27 mM KCl, 10 mM Na2HPO4, 18 mM KH2PO4, 1% Triton X-100, 13 mM sodium deoxycholate, 0.1% sodium dodecyl sulfate [SDS], 1 mM EDTA, 1 mM phenylmethylsulfonyl fluoride, 2 mg of leupeptin per ml, 1 mg of aprotinin per ml) and incubated with a polyclonal anti-PML antibody at 4°C overnight. The immunocomplexes were absorbed to protein A-Sepharose (Santa Cruz Biotechnology, Inc., Santa Cruz, Calif.) for 2 h at 4°C and washed three times in 1 ml of RIPA buffer. Associated proteins were then eluted in SDS sample buffer by boiling for 4 min. The released proteins were then resolved by 8% SDS-polyacrylamide gel electrophoresis (PAGE), transferred to nitrocellulose membranes (Schleicher & Schuell, Inc., Keene, N.H.), probed with anti-Sp1 antibody (Santa Cruz), and detected with the ECL (enhanced chemiluminescence) system (Amersham Life Sciences, Inc., Arlington Heights, Ill.). Immunoblot analyses of PML and PML mutants translated in vitro were performed by denaturing the proteins in SDS sample buffer and resolving by SDS-PAGE as described above. In vitro transcription and translation. For in vitro translation experiments, the PML, PML mutants, E2F1, and cyclin A plasmids were transcribed in vitro with T7 RNA polymerase, while Sp1 plasmid was transcribed with SP6 RNA polymerase from the appropriate expression vectors (described above). The products were labeled with [35S]methionine (NEN, Boston, Mass.) using the TNT coupled transcription-translation system (Promega Corp.). In the in vitro translation reactions, empty expression vectors were used as a control. GST fusion proteins and GST pull-down assay. Full-length and mutated glutathione S-transferase (GST)-Sp1 proteins were expressed from pGex-2TKMCS plasmids (described above), and the GST-PML protein was expressed from pGex3X plasmid as described previously (14). The GST fusion proteins produced in host bacteria were purified by standard procedures. The GST pull-down assays were performed essentially as described previously (57). Briefly, similar quantities of GST or GST fusion proteins immobilized on glutathione-Sepharose beads were washed in NETN buffer (100 mM NaCl, 1 mM EDTA, 20 mM Tris-HCl [pH 8], 0.5% Nonidet P-40) and incubated with 5 ml of proteins translated in vitro and labeled with [35S]methionine in 200 ml of NETN buffer for

VOL. 18, 1998

INHIBITION OF Sp1-MEDIATED TRANSCRIPTION BY PML

7149

2 h at 4°C on a rocker. Bound proteins were then washed three times in 0.5 ml of NETN buffer and eluted in SDS sample buffer by boiling for 5 min. The eluted proteins were subjected to SDS-PAGE as described above. The quantity and expression of GST and GST fusion proteins were determined by SDS-PAGE followed by Coomassie blue staining. EMSAs. Nuclear proteins were isolated from HeLa cells and used in electrophoretic mobility shift assays (EMSAs) as described previously (58). ProteinDNA binding assays were performed by first preincubating 5 mg of HeLa nuclear proteins in a binding buffer (4% Ficoll 400, 20 mM HEPES [pH 7.9], 2 mM MgCl2, 1 mg of salmon sperm DNA with the final concentration of KCl in the reaction mixture adjusted to 100 mM) for 10 min at room temperature in a total volume of 19 ml. In each reaction mixture, 10 fmol (1 ml) of the 32P-labeled probe was then added, and the reaction mixture was then incubated at room temperature for an additional 30 min. Double-stranded Sp1 and E2F oligonucleotide probes were made by annealing the complementary strands of the following oligonucleotides: Sp1, 59-CATTCGATCGGGGCGGGGCGAGC-39; and E2F, 59-TCCGTAGTTTTCGCGCTTAAATTTGAGAAAGGGCGCGAAACTAG TC-39. For reactions analyzing the effects of PML or PML mutants on Sp1 DNA binding, the purified Sp1 protein or the HeLa nuclear extracts were preincubated with the labeled Sp1 probe for 10 min in the binding buffer as described above. The in vitro-translated proteins were then added, and the reaction mixtures were incubated for an additional 30 min. Similar amounts of the reticulocyte lysate from the control in vitro translation reactions (see above) were used in control reactions. In supershift assays, 1 mg of anti-Sp1 antibody (Santa Cruz) or 1 ml of the preimmune serum was preincubated with the extracts for 10 min prior to the addition of the labeled Sp1 probe. In competition assays, 100- and 200-fold molar excesses of Sp1 and E2F double-stranded oligonucleotides, respectively, were preincubated with the extracts for 10 min before addition of the labeled probes. The protein-DNA complexes were resolved on a 4% native polyacrylamide gel in 0.25 TBE (44.5 mM Tris-HCl, 44.5 mM boric acid, 1 mM EDTA) and visualized by autoradiography.

RESULTS Identification of the EGFR promoter domains responsive to PML. We have previously shown that PML can suppress the transcriptional activity of several promoters, including the EGFR and human MDR1 genes (50, 58). To identify the region or specific sequences responsive to the repressive activity of PML, the effects of PML on a series of deletion constructs of the EGFR promoter spanning the sequences from 21109 to 216 (the initiator ATG being 11) and linked to the CAT gene were examined (Fig. 1). The constructs were cotransfected into SW13 cells with or without PML. As expected, expression of PML significantly suppressed the activity of the EGFR promoter ERCAT(21109) (Fig. 1B). Deletion of sequences between 21109 and 2150, although it reduced basal promoter activity, did not affect the repression of promoter activity by PML. Indeed, PML suppressed the activity of both ERCAT(2150), containing sequences from 2150 to 216, and the full-length construct ERCAT(21109) to a similar extent. This suggested that the region between 2150 and 216 conferred most of the repressive effects of PML. Given the very low basal activity of ERCAT(2105) and ERCAT(2167/ 2105), it was not feasible to further investigate the effects of PML on these deletion constructs. Under similar conditions, and in contrast to PML, the PMLRARa fusion protein caused a small inhibition (up to 20%) of ERCAT(21109) and ERCAT(2911) activity but had little or no effect on EGFR mutants spanning sequences between 2850 and 216 (Fig. 1B). The fact that the PMLRARa fusion protein had an altered transcriptional activity compared with that of the wild-type PML protein indicated the specificity of PML’s effects. Inhibition of Sp1-mediated transcription of EGFR by PML. The study described above demonstrated that the 150-bp 59 element of the EGFR promoter confers repressive effects on PML. The transcriptional activity of the EGFR promoter is efficiently regulated by the transcription factor Sp1 through at least four Sp1 binding sites, and the 150-bp 59 element has been shown to be sufficient for the promoter activity (31, 34, 61). To examine whether the repression of EGFR transcription

FIG. 1. Functional analysis of the effects of PML and PMLRARa on EGFR promoter. (A) Schematic representation of EGFR deletion mutants linked to the CAT gene. The numbers indicate the sequences of the EGFR promoter relative to the translation start site (ATG 11). (B) Relative CAT activity in SW13 cells cotransfected with 5 mg of different deletion mutants of the EGFR promoterCAT constructs as shown in panel A, together with 10 mg of PML and PMLRARa expression plasmids. The CAT activity of the longest mutant of the EGFR promoter, ERCAT(21109), in the absence of PML or PMLRARa, was arbitrarily set at 100, and the activities of the other constructs were calculated relatively. The mean CAT activity from three independent experiments is shown for each construct.

by PML was caused by inhibition of its Sp1-mediated activity, the effects of PML on Sp1-mediated activity of EGFR were examined in Drosophila melanogaster SL2 cells, which lack endogenous Sp1. To achieve sufficient protein expression in transient transfection assays, PML and Sp1 were expressed under the control of the Drosophila b-actin promoter from pPac vector (provided by R. Tjian) (33). SL2 cells were transfected with ERCAT(2150) alone or together with pPacSp1 and increasing amounts of pPacPML expression plasmids. As expected, cotransfection of ERCAT(2150) with 250 ng of Sp1 expression vector resulted in about a 50-fold induction of promoter activity (Fig. 2B). Strikingly, cotransfection of the pPacPML expression vector at different concentrations (1, 2.5, and 5 mg) inhibited the Sp1-mediated stimulation of the promoter activity in a dose-dependent manner (Fig. 2B, lanes 3 to 5). When the expression of both Sp1 and PML in the transfected cells was examined by immunostaining, no significant changes in the expression of Sp1 were observed in the presence of different concentrations of PML. Similar to the case in mammalian cells, expression of PML in SL2 cells in the presence or absence of Sp1 produced the normal PML nuclear speckled pattern (15). In control transfections, PML had no significant effect on the basal promoter activity derived from a pCMV-CAT construct, indicating that the inhibition of Sp1

7150

VALLIAN ET AL.

FIG. 2. Inhibition of Sp1-mediated transactivation of the EGFR promoter by PML. (A) Schematic representation of Sp1 binding sites in the 150-bp element of the EGFR promoter (28). (B) Effects of different concentrations of PML on Sp1-stimulated activity of the 150-bp region of EGFR. Drosophila SL2 cells were transfected with 3 mg of the ERCAT(2150) reporter plasmid, pPacSp1, and increasing concentrations of pPacPML expression plasmids as indicated. In each transfection, 100 ng of pPac-b-gal (b-galactosidase expression plasmid) was used to monitor transfection efficiency. The cells were harvested 24 h after transfection, and CAT activities were measured. The bar graph shows the fold CAT activity of each transfection relative to the activity exhibited by the ERCAT(2150) reporter construct in the absence of the effector plasmids. (C) Effect of increasing concentrations of pPacPML on the basal activity of the pCMVCAT vector in SL2 cells. In each transfection, cells were transfected with 5 mg of pCMV-CAT and increasing concentrations of pPacPML as indicated.

activity by PML was specific (Fig. 2C). Together, these results suggested that the suppression of EGFR promoter activity by PML was caused, at least in part, by inhibition of its Sp1mediated activity. Association of PML and Sp1 in vivo. Given PML’s specific inhibition of the Sp1-mediated activity of the EGFR promoter, experiments were done to see if an interaction between PML and Sp1 were possible. First, their association in vivo was examined by immunoprecipitation. For this, nuclear extracts prepared from HeLa cells transiently overexpressing the PML protein were used. In brief, the HeLa nuclear extracts were immunoprecipitated with a polyclonal anti-PML antibody, after which the immunocomplexes were absorbed to protein A-agarose, washed extensively, analyzed by SDS-PAGE, and detected by anti-Sp1 antibody (Santa Cruz). The results, as shown in Fig. 3A, indicated that PML and Sp1 did indeed associate in vivo. In control experiments, an unrelated antibody (anti-GAL4) produced no signal. The presence of Sp1 in the nuclear extracts was also confirmed by analyzing a sample of the extracts in a parallel lane by SDS-PAGE (Fig. 3A). The possible cross-reactivity of anti-PML antibody and Sp1 protein was ruled out, because the anti-PML antibody failed to precipitate either the purified or in vitro-translated Sp1 protein; likewise, the anti-Sp1 antibody was unable to detect PML pro-

MOL. CELL. BIOL.

FIG. 3. In vivo analysis of the association of PML and Sp1. (A) Immunoprecipitation. Nuclear extracts from HeLa cells transiently overexpressing PML were subjected to immunoprecipitation with anti-PML antibody (a-PML) or anti-GAL4 antibody (a-GAL4). The immunocomplexes were absorbed to protein A-Sepharose, washed, resolved by SDS-PAGE (8% polyacrylamide), transferred to nitrocellulose membranes, and probed with anti-Sp1 antibody. In a parallel lane, a sample of nuclear extracts (lysate) was analyzed to indicate the position of the Sp1 band. (B) Mammalian two-hybrid assay. U2OS cells were cotransfected with 1 mg of GAL4-Sp1 and/or 3 mg of VP16-PML expression plasmids, as indicated, together with 4 mg of the luciferase reporter plasmid [4 3 (UAS)-Luc]. In each transfection, cells were also transfected with 0.5 mg of pCMV-b-gal (b-galactosidase expression plasmid) for monitoring transfection efficiency and normalization of luciferase activity. The luciferase activity of each sample was measured, was calculated relative to the activity exhibited by cells transfected with empty expression vector (control), and is shown as the fold increase.

tein in extracts from cells transiently overexpressing PML or the PML protein translated in vitro (not shown). PML could not be immunoprecipitated with the Sp1 antibody, but whether this was due to the antibody or to the nature of the Sp1-PML interaction, in which Sp1 epitopes detected by Sp1 antibody might be masked, was unclear. The in vivo association of PML and Sp1 was further investigated by using a mammalian two-hybrid assay. cDNAs of PML and Sp1 were fused in frame to the sequences of the activation domain of VP16 activator protein (VP16-PML) and the DNA binding region of the GAL4 transcription factor (GAL4-Sp1), respectively. The VP16-PML and GAL4-Sp1 fusion vectors were then cotransfected into U2OS cells together with a luciferase target reporter plasmid bearing four GAL4binding sites [p4x(UAS)-Luc] in front of the firefly luciferase gene. As shown in Fig. 3B, cotransfection of VP16-PML or GAL4-Sp1 with the GAL4-responsive target reporter did not induce significant luciferase activity. Strikingly, cotransfection of VP16-PML and GAL4-Sp1 together resulted in a marked (about 65-fold) induction of luciferase activity from the target promoter, indicating the presence of a physical interaction between the PML and Sp1 moieties of the fusion constructs (Fig. 3B). As a positive control, PML-PML interaction was investigated by cotransfection of GAL4-PML and VP16-PML constructs together with the GAL4-responsive luciferase promoter. The level of luciferase activity consequently stimulated by PML-PML interaction was lower than that induced by PML-Sp1 interaction. One explanation could be that GAL4PML had the suppressive effects on basal transcriptional ac-

VOL. 18, 1998

INHIBITION OF Sp1-MEDIATED TRANSCRIPTION BY PML

FIG. 4. Analysis of in vitro binding of PML and Sp1. (A) Schematic diagram of Sp1 and Sp1 deletion mutants used in the GST pull-down assay. The letters A to D indicate different domains of the Sp1 protein. Three zinc finger DNA binding motifs of Sp1 present in the C-terminal region are shown. The numbers in parentheses indicate the amino acids of Sp1 linked to the GST protein. (B) GST and GST-Sp1 (full-length and deletion mutants) immobilized on glutathione-Sepharose beads were incubated with in vitro-translated, 35S-labeled PML. Bound proteins were then eluted and analyzed by SDS-PAGE (8% polyacrylamide) as described in Materials and Methods. As a control, a 1/10 volume of labeled PML protein used in the in vitro binding assays was resolved by SDSPAGE. (C) GST and GST-Sp1 proteins were incubated with in vitro-translated 35 S-labeled E2F protein as described for panel B. (D) GST and GST-PML proteins bound to glutathione-Sepharose beads were incubated with in vitrotranslated Sp1 and resolved by SDS-PAGE (8% polyacrylamide). (E) In vitrotranslated 35S-labeled PML, E2F1, and cyclin A (CA) were incubated with GST-PML protein as described for panel D.

tivity reported previously (58). The basal luciferase activity was consistently reduced 50 to 70% in the presence of GAL4-PML. In control transfections, cotransfection of VP16 and/or the GAL4 DNA-binding domain did not activate the GAL4responsive target promoter (data not shown). PML-Sp1 interaction in vitro. The direct interaction of PML and Sp1 in solution was examined with the GST pull-down assay. In this assay, the in vitro-translated 35S-labeled PML was incubated with the recombinant full-length protein or various truncated GST-Sp1 proteins immobilized on the glutathioneSepharose beads, and the associated proteins were washed and analyzed by SDS-PAGE. As shown in Fig. 4B, the PML protein bound the full-length GST-Sp1 as well as the C-terminal Sp1 mutant GST-Sp1(622–788), but not the N-terminal Sp1 mutant GST-Sp1(1–293) or GST-Sp1(1–621). Little or no background binding with PML was detected in control experiments with the GST protein alone. Also, in assays run with control in vitro translation reaction mixtures (including those

7151

run with no DNA or empty expression vectors), no binding with the GST-Sp1 proteins was detected (not shown). In a parallel experiment, the binding of E2F1 to GST-Sp1 was examined as a positive control (36, 46). As expected, GST-Sp1 showed strong binding to E2F1 protein translated in vitro (Fig. 4C). Notably, the intensity of PML-Sp1 binding was comparable to the interaction observed between E2F1 and Sp1 when examined under the same conditions. The in vitro association of PML with Sp1 was further confirmed by incubation of the in vitro-translated Sp1 protein with the full-length GST-PML protein. As can be seen in Fig. 4D, the GST-PML fusion protein, but not the GST protein alone, efficiently bound Sp1. Since PML has been shown to form homodimer complexes both in vitro and in vivo (37, 60), the interaction of GST-PML and in vitro-translated 35S-labeled PML in this experiment therefore served as a positive control (Fig. 4E). Moreover, since the in vitro association of E2F1 and Sp1 has been shown to be mediated through the C-terminal domain of Sp1 (36, 46), and because the same region in Sp1 involves interaction with PML (see above), the possible interaction of PML and E2F1 was also examined. The 35S-labeled E2F1 translated in vitro was incubated with the GST or GSTPML protein, and their binding was analyzed as above. In this experiment, the association of the cyclin A protein with PML was also tested. As expected, GST-PML efficiently bound PML, whereas cyclin A and E2F1 could not (Fig. 4E). This study further confirmed the specificity of the association between PML and Sp1. Mediation of PML-Sp1 interaction by the coiled-coil domain. To identify domains of PML involved in its association with Sp1, several deletion mutants of the PML protein (Fig. 5A) were translated in vitro, labeled with [35S]methionine, and subjected to the GST pull-down assay by using the full-length GST-Sp1 fusion protein. As shown in Fig. 5B, mutants of PML lacking the N-terminal proline-rich region (PMLpro2), the proline-rich region plus the RING finger domain (PMLpr2), or the C-terminal serine/proline-rich region (PMLsp2) bound the Sp1 protein, whereas the deletion mutant of PML lacking the coiled-coil (dimerization) domain (PMLdim2) could not. In control experiments, the incubation of the in vitro-translated, 35S-labeled PML mutants with the GST protein alone produced no significant binding (not shown). Together, these results showed that the coiled-coil domain of PML was required for the association of PML with Sp1 in vitro. Disruption of Sp1-DNA binding by PML. The finding that PML and Sp1 could associate and that the C-terminal region of Sp1 and the coiled-coil domain of PML were involved in this association, combined with the knowledge that the C-terminal region of Sp1 mediates its binding to target DNA through three zinc finger motifs (33), raised the possibility that the binding of PML to Sp1 may interfere with its DNA-binding activity, thus providing a mechanism for the suppression of the EGFR promoter activity by PML. To examine the possible effects of PML on Sp1 DNA binding, EMSAs were done with nuclear extracts from HeLa cells incubated with labeled oligonucleotide probes bearing an Sp1 binding site in the presence of different concentrations of in vitro-translated PML protein. As shown in Fig. 6A, addition of in vitro-translated PML protein (1, 3, and 5 ml in lanes 8 to 10, respectively) to the EMSA reaction mixtures resulted in the disruption of low-mobility DNA-protein complexes corresponding to Sp1 (see below) in a manner dependent on PML concentration (compare lanes 8 to 10 with lane 5 in Fig. 6A). In control reactions, addition of rabbit reticulocyte lysate had no effect on Sp1 or other DNAprotein complexes retarded by the labeled Sp1 probe (lanes 11 and 12). An immunoblot analysis of the PML protein trans-

7152

VALLIAN ET AL.

FIG. 5. Identification of domains of PML involved in its association with Sp1. (A) Schematic representation of PML and PML deletion mutants. Pro, prolinerich region; R, RING finger; B, B-box motifs (B1 and B2); coiled-coil, PML dimerization domain; Serine, serine/threonine-rich region. PMLpro2, PMLpr2, PMLdim2, and PMLsp2, respectively, lack the proline-rich region, the prolinerich region plus the RING finger, the dimerization domain, and the serine/ proline-rich domain, as shown. (B) GST-Sp1 protein bound to glutathioneSepharose beads was incubated with in vitro-translated, 35S-labeled PML and PML mutants. Bound proteins were then washed and analyzed by SDS-PAGE (8% polyacrylamide). (C) The in vitro-translated proteins (input) used in panel B (a 1/10 volume) were resolved by SDS-PAGE (8% polyacrylamide).

lated in vitro, representing amounts similar to those used in EMSA, was shown in Fig. 6C. In EMSA reactions, the specificity of the Sp1 complex was examined by oligonucleotide competition assay with a specific anti-Sp1 antibody not crossreactive with Sp2, Sp3, or Sp4 (see Materials and Methods). As shown in Fig. 6A, in the presence of a 100-fold molar excess of nonlabeled probe, the specific DNA-protein complexes could be competed out (lanes 2 and 6). Incubation of extracts with anti-Sp1 antibody, but not nonimmune serum, resulted in an Sp1 supershift (lanes 4 and 7) that indicated the presence of Sp1 in the complexes disrupted by PML. To confirm that the effects of PML on DNA binding were specific to Sp1 site and not a general effect of PML on DNAbinding activity, the EMSA was repeated with the HeLa nuclear extracts by using oligonucleotides containing the binding site for the transcription factor E2F as probe, in the presence or absence of amounts of PML similar to those used in the Sp1 binding assays (see above). As shown in Fig. 6B, neither PML nor the control lysate affected the E2F binding complex, thus indicating that the disruption of the Sp1 complex by PML was specific. Together, these results indicated that PML could disrupt the Sp1 DNA binding, presumably by forming a non-DNA-binding complex with the Sp1 protein. Involvement of the PML coiled-coil domain in Sp1 DNA binding. To analyze the effects of PML on Sp1 DNA binding

MOL. CELL. BIOL.

FIG. 6. The effects of PML on Sp1 DNA binding. (A) EMSA of HeLa nuclear proteins. HeLa nuclear proteins were incubated with 32P-labeled Sp1 probe, and the Sp1-DNA complexes were resolved on a 4% native polyacrylamide gel. In lanes 2 and 5, specific complexes were competed out with 100-fold molar excesses of nonlabeled (cold) Sp1 probe to examine the specificity of the retarded complexes. The presence of Sp1 protein in the complexes was also tested with anti-Sp1 antibody (a-Sp1) (lanes 4 and 7) and preimmune serum (lane 3). In lanes 8 to 10, the nuclear extracts were treated with 1, 3, and 5 ml of in vitro-translated PML protein, respectively. In lanes 11 and 12, 3 and 5 ml, respectively, of the lysate from the control reaction mixtures were used. (B) E2F DNA binding. In lane 1, HeLa nuclear proteins were incubated with 32P-labeled E2F probe, and the E2F-DNA complexes were subjected to electrophoresis as described for panel A. Lane 2 shows that the specific retarded complexes were competed out for binding by a 200-fold molar excess of nonlabeled E2F probe. In lanes 3 and 4, the extracts were treated with 3 and 5 ml of in vitro-translated PML protein, respectively, as described for panel A. In lane 5, nuclear proteins were treated with 5 ml of control lysate. (C) Analysis of the expression of the in vitro-translated PML proteins shown in panels A and B. In vitro-translated PML proteins (1, 3, and 5 ml) and control lysate (5 ml) were subjected to SDS-PAGE (8% polyacrylamide) and detected with anti-PML antibody.

more directly, a purified Sp1 protein (0.25 footprinting unit/ reaction; Promega) was used in EMSA reactions (Fig. 7A). As in the experiments containing HeLa nuclear extracts, the addition of the PML protein at increasing concentrations disrupted the Sp1 DNA-binding complex in a dose-dependent manner (Fig. 7A, lanes 4 to 6). In control reactions, addition of rabbit reticulocyte lysate had no effect on the complex (lane 7). These results further confirmed the specificity of the effect of PML on Sp1 DNA binding. Because the coiled-coil region of PML was found to mediate the association of PML with Sp1, it was of interest to examine whether the same region was involved in the effects of PML on Sp1 DNA binding. Deletion mutants of PML similar to those used in the GST pull-down assays were translated in vitro and

VOL. 18, 1998

INHIBITION OF Sp1-MEDIATED TRANSCRIPTION BY PML

7153

FIG. 7. Effects of PML and PML deletion mutants on the DNA-binding activity of purified Sp1 protein. (A) Purified Sp1 protein (0.25 footprinting unit/reaction) was incubated with 32P-labeled Sp1 oligonucleotide probe, and the DNA-protein complexes were analyzed by electrophoresis as described for Fig. 6. Lane 1 shows the binding of Sp1 protein to the labeled Sp1 probe. The specificity of the Sp1 complex was examined by oligonucleotide competition assay using the nonlabeled Sp1 probe (lane 2) and anti-Sp1 antibody (lane 3). In lanes 4 to 6, 1, 3, and 5 ml of PML protein, respectively, were added to the Sp1-binding reaction mixtures. Lane 7 contains 5 ml of the control lysate. See text for details. (B) In vitro-translated PML and PML deletion mutants (3 ml) were incubated with labeled Sp1 probe, and the DNA-protein complexes were analyzed by electrophoresis as described for panel A. (C) HeLa nuclear extracts were incubated with PML deletion mutants as in panel B. In each of panels A to C, the Sp1 complex is indicated by an arrow. (D) The expression of PML and PML deletion mutants was then analyzed by immunoblotting. The abbreviations are as given for Fig. 5.

used in EMSA reactions with the purified Sp1 protein. Consistent with the results presented above, addition of the fulllength PML protein significantly reduced the binding of purified Sp1 to the labeled Sp1 oligonucleotide probe (Fig. 7B, lane 7). Similar to the full-length PML, addition of PMLpro2, PMLpr2, PMLnls2, and PMLsp2 to the EMSA reaction mixtures reduced the Sp1 DNA binding (Fig. 7B). Strikingly, PMLdim2 (which lacks the coiled-coil domain of PML) lost a significant part, but not all, of PML’s ability to inhibit Sp1 DNA binding (lane 4 in Fig. 7B). Similar experiments were repeated with HeLa nuclear extracts, and comparable results were obtained (Fig. 7C). Analysis of the in vitro-translated proteins by immunoblotting showed that all proteins were expressed at similar levels, indicating that the loss of the activity in PMLdim2 was not due to the lack of protein expression (Fig. 7D). Together, these results showed that the coiled-coil domain of PML, which mediates PML’s interaction with Sp1, was also involved in its effects on Sp1 DNA binding. Therefore, the disruption of Sp1 DNA binding by PML was likely caused by their physical interaction. Abrogation of Sp1-mediated DHFR promoter activity by PML. Given the specific inhibition of Sp1 DNA-binding activity and transactivation by PML, it was important to examine whether PML could repress transcription from another Sp1-

regulated promoter. Most of the growth-regulated promoters contain binding sites for Sp1 and E2F. Among these promoters, thymidine kinase and DHFR gene promoters have recently been shown to be regulated by Sp1 (36, 46). We therefore investigated the effects of PML on the Sp1-dependent activity of DHFR in SL2 cells, in which the activity of DHFR is dependent on exogenous Sp1 (46). For this purpose, the hamster DHFR promoter driving expression of the CAT gene (DHFR-CAT) was transfected into SL2 cells either alone or together with pPacSp1 and different concentrations of the pPacPML expression vectors. The hamster DHFR promoter contains four Sp1 binding sites and two overlapping binding sites for the transcription factor E2F1 (5). As expected, the DHFR target promoter was almost silent in SL2 cells, and its activity depended upon the Sp1 expression (Fig. 8A, compare lanes 1 and 2). Transfection of 250 ng of pPacSp1 resulted in a marked (35-fold) induction of CAT expression from the DHFR target promoter. Similar to the effects of PML on Sp1-mediated activity of the EGFR target promoter (Fig. 2A), cotransfection of pPacPML suppressed Sp1-stimulated activity of the DHFR reporter gene in a dose-dependent manner (Fig. 8A). Moreover, PML could suppress the transcriptional activity of DHFR and thymidine kinase in mammalian cells (unpublished data). In control transfection experiments, expres-

7154

VALLIAN ET AL.

MOL. CELL. BIOL.

FIG. 8. Inhibition of Sp1-mediated transactivation of DHFR promoter by PML. (A) Dose-dependent inhibition of Sp1-mediated activity of DHFR by PML. Drosophila SL2 cells were transfected with 3 mg of DHFR-CAT, promoter pPacSp1, and increasing concentrations of pPacPML as indicated. In each transfection, cells were also transfected with 100 ng of pPac-b-gal (b-galactosidase expression plasmid) for monitoring of transfection efficiency and normalization of CAT activities. (B) Effect of PML on E2F activity of DHFR promoter. SL2 cells were transfected as in panel A, except that in lanes 2 and 3, pPacE2F1 was included as indicated. In each panel, the CAT activity exhibited by the DHFR-CAT in the absence of Sp1 or E2F1 was set to 1, and the activities in other experiments were calculated in relation to that setting.

sion of PML did not significantly affect the E2F1-stimulated activity of DHFR target promoter, indicating the specificity of PML’s effect (Fig. 8B, lanes 2 and 3). Together, these results showed that the Sp1-mediated transcriptional activity of the DHFR promoter could be inhibited by PML, suggesting that DHFR is another target of Sp1-PML. DISCUSSION We report here that the PML growth suppressor interacts with Sp1 and inhibits the Sp1-mediated transcriptional activity of the EGFR gene promoter. This finding is based on the observations that (i) PML repressed the transcription of the EGFR promoter by targeting a 150-bp 59 element of the promoter that is driven by the transcription factor Sp1; (ii) PML and Sp1 interacted both in vitro and in vivo through specific domains; (iii) PML specifically disrupted the binding of Sp1 to target DNA; and (iv) expression of exogenous PML inhibited Sp1-mediated transcription of Sp1 target promoters in Sp1negative Drosophila melanogaster SL2 cells. The inhibition of Sp1-mediated transcriptional activity by PML may therefore represent a novel mechanism for inhibiting EGFR promoter activity. Furthermore, cotransfection of PML inhibited the Sp1-mediated activity of the DHFR promoter, indicating that the repressive effects of PML are not limited to EGFR and that PML may function as an inhibitor of Sp1-targeted promoters. The repressive effects of PML on EGFR transcription are

specific because the 150-bp element 59 to the initiation site is responsible for this activity. Under similar conditions, the PMLRARa fusion protein failed to inhibit the promoter activity at this site. One possible explanation could be its altered nuclear localization from a speckled pattern (PODs) to a diffused pattern (16, 60). The recent findings that PODs are possible sites for transcription support this notion (29, 42). Moreover, PMLRARa, which contains most of the functional domains of PML and RARa, has also been shown to have different functional properties compared with those of PML and RARa (14, 35, 58). The EGFR promoter is GC rich and lacks both TATA and CAAT boxes (28). Several binding sites for Sp1 have been identified in the EGFR promoter, four of which have been shown to bind Sp1 (34). Accordingly, in Drosophila SL2 cells, which are negative for Sp1, the activity of the EGFR promoter is entirely dependent on the exogenous Sp1 (61) (Fig. 2A). In mammalian cells, several transcription factors have been shown to exert their effects on the promoter activity directly or indirectly through interaction with Sp1. For example, T3R and RAR have been shown to inhibit the EGFR promoter activity by competing with Sp1 to bind an overlapping binding site present in the proximal 36-bp segment (between 2112 and 277) of the promoter (27, 61). Moreover, p53, which functions as an activator of EGFR promoter, has been shown to form complexes with Sp1 and to stimulate its binding to DNA (13, 23). These findings support the notion that Sp1 functions as a

VOL. 18, 1998

INHIBITION OF Sp1-MEDIATED TRANSCRIPTION BY PML

crucial regulator of EGFR promoter activity. Moreover, the present study showed that the inhibition of EGFR transcription by PML is caused, in part, by inhibition of its Sp1-mediated activity and indicated that PML could form complexes with Sp1 in vivo and in vitro and efficiently repress the Sp1mediated activity of the EGFR promoter in Sp1-negative SL2 cells. The physical interaction between PML and Sp1 provides further direct support for the inhibition of EGFR activity by PML. In particular, our in vitro binding assays showed that the C-terminal region of Sp1 is required for interaction with PML, which contains three zinc fingers that mediate Sp1 DNA-binding activity. Our EMSAs demonstrated that addition of PML protein disrupted the binding of Sp1 to target DNA, regardless of whether the crude nuclear extracts or the purified Sp1 protein was used. Furthermore, our attempts to detect PML-Sp1 complexes that bind DNA were unsuccessful (not shown). Together, these data suggest that PML interferes with Sp1 DNAbinding activity most likely by forming complexes that do not bind DNA. An increasing number of transcription factors have been found that interact with Sp1. One set of these factors includes E2F1, GATA1, and YY1, which act synergistically with Sp1 on DNA to increase transcriptional activity (21, 36, 45, 46). Another set of Sp1-interacting transcription factors that impair Sp1-mediated transcriptional activity includes the von HippleLindau (VHL) tumor suppressor protein, p107, and Sp1-I. The VHL interacts with Sp1 and inhibits its activation of the vascular endothelial growth factor (VEGF) promoter (51). Strikingly similar to PML in activity, VHL was originally found to inhibit VEGF promoter activity through a GC-rich element whose activity is regulated by Sp1. This suggests that VEGF may also be an Sp1-PML target. The cell cycle-regulatory protein p107, a member of the Rb family, also associates with Sp1 and inhibits its transcriptional activity (12), but the mechanism by which it does so is unclear. However, as with PML, no Sp1-p107 complex that bound DNA was detected, suggesting that p107 may also interfere with Sp1 DNA binding. Sp1-I, a 20-kDa Rb-associated factor (11), impairs Sp1 transcriptional activity by association with the pocket domain of Rb. Rb has been reported to synergistically stimulate transcriptional activity of Sp1 (55, 56). Because no direct interaction between Rb and Sp1 has been detected, it has been hypothesized that Rb expression sequesters or liberates inhibitory factors associated with Sp1, such as Sp1-I. It has recently been reported that Rb interacts with PML both in vitro and in vivo through its pocket domain (2). The Rb pocket has also been shown to mediate its stimulation of Sp1-mediated transcriptional activity (38). It is therefore tempting to hypothesize that PML may be another Sp1-associated inhibitor that is targeted by Rb. Therefore, it would be interesting to examine whether Sp1 activity inhibited by PML can be restored by Rb in a set of squelching experiments. Several studies from our laboratory and others have shown that PML is a growth and transformation suppressor (24, 41, 43, 46, 48, 49, 59). We have recently found that overexpression of PML can significantly suppress the growth and tumorigenicity of breast cancer cells by inducing G1 arrest and apoptosis (43). Accordingly, ectopic expression of PML in normal human fibroblasts results in the induction of G1 arrest (unpublished results). Furthermore, we have recently shown that the stable expression of PML in HeLa cells can lengthen the G1 phase of the cell cycle (49). Together, these studies further demonstrate that the effect of PML on G1 cell cycle progression correlates with an alteration of Rb phosphorylation and expression of a number of cell cycle-related proteins, such as cyclin E, cyclin

7155

D1, Cdk2, p27, p21, and p53 (43, 49). These findings in turn suggest that PML most likely exerts its suppressive effects on cell growth by targeting the protein factors involved at the G1/S transition checkpoint. A large group of genes whose products are involved in cell growth (e.g., DHFR and thymidine kinase) are activated during progression through G1/S and contain binding sites for Sp1 as well as E2F in their promoter regions (5, 46). In this study, we have shown that the repressive effects of PML on Sp1 activity are not limited to EGFR and that PML efficiently suppresses the Sp1-stimulated activity of the DHFR promoter in Drosophila SL2 cells. The region of Sp1 implicated in inhibition by PML also mediates Sp1-E2F1 interaction (36, 46). Thus, the present findings imply that PML inactivation of Sp1 during the G1-S transition could be a major mechanism for rendering PML’s growth suppressor function. Recently, it has been demonstrated that Sp1 functions as a critical factor in cell growth and control of DHFR expression during cell cycle progression after serum stimulation of quiescent cells (52). In addition, detailed analysis of the DHFR promoter has revealed that the Sp1 sites, but not the E2F site, of the promoter mediate its transcriptional activity during the late G1/S phase of the cell cycle (30). Together, these studies strongly support the importance of Sp1 in the regulation of cell growth and cell cycle progression. In this light, investigations into the effect of PML on Sp1-mediated transcription of other genes involved in the G1/S checkpoint are now under way in our laboratory. ACKNOWLEDGMENTS We are grateful to E. Wintersberger, H. Rotheneder, R. Tjian, P. Chambon, T. Kouzarides, D. Johnson, and M.-J. Tsai for providing the vectors and plasmids used in these studies. We are also grateful to J. Richard for critically reading the manuscript. This work was supported by grant CA-55577 from the National Institutes of Health to K.S.C. REFERENCES 1. Ahn, J.-H., and G. S. Hayward. 1997. The major immediate-early proteins IE1 and IE2 of human cytomegalovirus colocalize with and disrupt PMLassociated nuclear bodies at very early times in infected permissive cells. J. Virol. 71:4599–4613. 2. Alcalay, M., L. Tomassoni, E. Colombo, S. Stoldt, F. Grignani, M. Fagioli, L. Szekely, K. Helin, and P. G. Pelicci. 1998. The promyelocytic leukemia gene product (PML) forms stable complexes with the retinoblastoma protein. Mol. Cell. Biol. 18:1084–1093. 3. Ascoli, C. A., and G. G. Maul. 1991. Identification of a novel nuclear domain. J. Cell Biol. 112:785–795. 4. Bannister, A. J., and T. Kouzarides. 1995. CBP-induced stimulation of c-Fos activity is abrogated by E1A. EMBO J. 14:4758–4762. 5. Blake, M. C., and J. C. Azizkhan. 1989. Transcriptional factor E2F is required for efficient expression of the hamster dihydrofolate reductase gene in vitro and in vivo. Mol. Cell. Biol. 9:4994–5002. 6. Boddy, M. N., K. Howe, L. D. Etkin, E. Solomon, and P. S. Freemont. 1996. PIC 1, a novel ubiquitin-like protein which interacts with the PML component of a multiprotein complex that is disrupted in acute promyelocytic leukaemia. Oncogene 13:971–982. 7. Brown, D., S. Kogan, E. Lagasse, I. Weissman, M. Alcalay, P. G. Pelicci, S. Atwater, and J. M. Bishop. 1997. A PMLRARalpha transgene initiates murine acute promyelocytic leukemia. Proc. Natl. Acad. Sci. USA 94:2551– 2556. 8. Carvalho, T., J.-S. Seeler, K. Ohman, P. Jordan, U. Pettersson, G. Akusjarvi, M. Carmo-Fonseca, and A. Dejean. 1995. Targeting of adenovirus E1A and E4-ORF3 proteins to nuclear matrix-associated PML bodies. J. Cell Biol. 131:45–56. 9. Chang, K. S., Y. H. Fan, M. Andreeff, J. X. Liu, and Z. M. Mu. 1995. The PML gene encodes a phosphoprotein associated with the nuclear matrix. Blood 85:3646–3653. 10. Chang, K. S., S. A. Stass, D.-T. Chu, L. L. Deaven, J. M. Trujillo, and E. J. Freireich. 1992. Characterization of a fusion cDNA (RARA/myl) transcribed from the t(15;17) translocation breakpoint in acute promyelocytic leukemia. Mol. Cell. Biol. 12:800–810. 11. Chen, L. I., T. Nishinaka, K. Kwan, I. Kitabayashi, K. Yokoyama, Y.-H. Fu, S. Gru ¨nwald, and R. Chiu. 1994. The retinoblastoma gene product RB

7156

12. 13. 14.

15. 16. 17. 18. 19. 20. 21.

22.

23. 24.

25. 26.

27. 28. 29. 30. 31. 32. 33. 34. 35.

36. 37.

VALLIAN ET AL.

stimulates Sp1-mediated transcription by liberating Sp1 from a negative regulator. Mol. Cell. Biol. 14:4380–4389. Datta, P. K., P. Raychaudhuri, and S. Bagchi. 1995. Association of p107 with Sp1: genetically separable regions of p107 are involved in regulation of E2Fand Sp1-dependent transcription. Mol. Cell. Biol. 15:5444–5452. Ded, S. P., R. M. Munoz, D. R. Brown, M. A. Subler, and D. Sumitra. 1994. Wild-type human p53 activates the human epidermal growth factor receptor promoter. Oncogene 9:1341–1349. de The, H., C. Lavau, A. Marchio, C. Chomienne, L. Degos, and A. Dejean. 1991. The PML-RARalpha fusion mRNA generated by the t(15;17) translocation in acute promyelocytic leukemia encodes a functionally altered RAR. Cell 66:675–684. Doucas, V., and R. M. Evans. 1996. The PML nuclear component and cancer. Biochim. Biophys. Acta 1288:M25–M29. Dyck, J. A., G. G. Maul, W. H. Miller, J. D. Chen, A. Kakizuka, and R. M. Evans. 1994. A novel macromolecular structure is a target of the promyelocyte-retinoic acid receptor oncoprotein. Cell 76:333–343. Everett, R. D., and G. G. Maul. 1994. HSV-1 IE protein Vmw110 causes redistribution of PML. EMBO J. 13:5062–5069. Freemont, P. S., I. M. Hanson, and J. Trowsdale. 1991. A novel cystein-rich sequence motif. Cell 64:483–484. Goddard, A. D., J. Borrow, P. S. Freemont, and E. Solomon. 1991. Characterization of a zinc finger gene disrupted by the t(15;17) in acute promyelocytic leukemia. Science 254:1371–1374. Gongora, D., G. David, L. Pintard, C. Tissot, T. D. Hua, A. Dejean, and N. Mechti. 1997. Molecular cloning of a new interferon-induced PML nuclear body-associated protein. J. Biol. Chem. 272:19457–19463. Gregory, R. C., D. J. Taxman, D. Seshasayee, M. H. Kensinger, J. J. Bieker, and D. M. Wojchowski. 1996. Functional interaction of GATA1 with erythroid Kruppel-like factor and Sp1 at defined erythroid promoters. Blood 87:1793–1801. Grisolano, J. L., R. L. Wesselschmidt, P. G. Pelicci, and T. J. Ley. 1997. Altered myeloid development and acute leukemia in transgenic mice expressing PMLRARa under control of catapsin G regulatory sequences. Blood 89:376–387. Gualberto, A., and A. S. Baldwin, Jr. 1995. p53 and Sp1 interact and cooperate in the tumor necrosis factor-induced transcriptional activation of the HIV-1 long terminal repeat. J. Biol. Chem. 270:19680–19683. Guiochon-Mantel, A., F. J. Savouret, F. Quignon, K. Delaber, E. Milgrom, and H. De The. 1995. Effect of PML and PML-RAR on the transactivation properties and subcellular distribution of steroid hormone receptors. Mol. Endocrinol. 9:1791–1803. He, D., Z.-M. Mu, X. Le, J.-T. Hsieh, R.-C. Pong, L. W. K. Chung, and K.-S. Chang. 1997. Adenovirus-mediated expression of PML suppresses growth and tumorigenicity of prostate cancer cells. Cancer Res. 57:1868–1872. He, L. Z., C. Tribioli, R. Rivi, D. Peruzzi, P. G. Pelicci, V. Soares, G. Cattoretti, and P. P. Pandolphi. 1997. Acute leukemia with promyelocytic features in PMLRARalpha transgenic mice. Proc. Natl. Acad. Sci. USA 94:5302–5307. Hudson, L. G., J. B. Santon, and G. N. Gill. 1989. Regulation of epidermal growth factor receptor gene expression. Mol. Endocrinol. 3:400–408. Ishii, S., Y.-H. Xu, R. H. Stratton, B. A. Roe, G. T. Merlino, and I. Pastan. 1985. Characterization and sequence of the promoter region of the human epidermal growth factor receptor gene. Proc. Natl. Acad. Sci. USA 82:4920–4924. Ishov, A. M., R. Stenberg, and G. G. Maul. 1997. Human cytomegalovirus immediate early interaction with host nuclear structures: definition of an immediate transcript environment. J. Cell Biol. 138:5–16. Jensen, D., A. R. Black, A. Swick, and J. C. Azizkhan. 1997. Distinct role for Sp1 and E2F sites in the growth/cell cycle regulation of the DHFR promoter. J. Cell. Biochem. 67:24–31. Johnson, A. C., S. Ishii, Y. Jinno, I. Pastan, and G. T. Merlino. 1988. Epidermal growth factor receptor gene promoter. J. Biol. Chem. 263:5693–5699. Johnson, D., J. Schwarz, W. Cress, and J. Nevins. 1993. Expression of transcription factor E2F1 induces quiescent cells to enter S phase. Nature 365:349–352. Kadonaga, J. T., A. J. Courey, J. Ladika, and R. Tjian. 1988. Distinct regions of Sp1 modulate DNA binding and transcriptional activation. Science 242: 1566–1570. Kageyama, R., G. T. Merlino, and I. Pastan. 1988. Epidermal growth factor (EGF) receptor gene transcription. J. Biol. Chem. 263:6329–6336. Kakizuka, A., W. H. J. Miller, K. Umesono, R. P. J. Warrell, S. R. Frankel, V. V. V. S. Murty, E. Dmitrovsky, and R. M. Evans. 1991. Chromosomal translocation t(15;17) in human acute promyelocytic leukemia fuses RARalpha with a novel putative transcription factor, PML. Cell 66:663–674. Karlseder, J., H. Rotheneder, and E. Wintersberger. 1996. Interaction of Sp1 with the growth- and cell cycle-regulated transcription factor E2F. Mol. Cell. Biol. 16:1659–1667. Kastner, P., A. Perez, Y. Lutz, C. Rochette Egly, M. P. Gaub, B. Durand, M. Lanotte, R. Berger, and P. Chambon. 1992. Structure, localization and transcriptional properties of two classes of retinoic acid receptor alpha fusion proteins in acute promyelocytic leukemia (APL): structural similarities with a new family of oncoproteins. EMBO J. 11:629–642.

MOL. CELL. BIOL. 38. Kennett, S. B., A. J. Udvadia, and J. M. Horowitz. 1997. Sp3 encodes multiple proteins that differ in their capacity to stimulate or repress transcription. Nucleic Acids Res. 25:3110–3117. 39. Koken, M. H. M., G. Linares Cruz, F. Quignon, A. Viron, M. K. Chelbi Alix, J. Sobczak Thepot, L. Juhlin, L. Degos, F. Calvo, and H. De The´. 1995. The PML growth-suppressor has an altered expression in human oncogenesis. Oncogene 10:1315–1324. 40. Koken, M. H. M., A. Reid, F. Quignon, M. K. Chelbi-Alix, J. M. Davies, J. H. S. Kabarowski, J. Zhu, S. Dong, S.-J. Chen, Z. Chen, C. C. Tan, J. Licht, S. Waxman, H. De The´, and A. Zelent. 1997. Leukemia-associated retinoic acid receptor alpha fusion partners, PML and PLZF, heterodimerize and colocalize to nuclear bodies. Proc. Natl. Acad. Sci. USA 94:10255–10260. 41. Kumar, A. P., and A. P. Butler. 1997. Transcription factor Sp3 antagonizes activation of the ornithine decarboxylase promoter by Sp1. Nucleic Acids Res. 25:2012–2019. 42. LaMorte, V. J., J. A. Dyck, R. L. Ochs, and R. M. Evans. 1998. Localization of nascent RNA and CREB binding protein with the PML-containing nuclear body. Proc. Natl. Acad. Sci. USA 95:4991–4996. 43. Le, X.-F., S. Vallian, Z.-M. Mu, M.-C. Hung, and K.-S. Chang. 1998. Recombinant PML adenovirus suppresses growth and tumorigenicity of human breast cancer cells by inducing G1 cell cycle arrest and apoptosis. Oncogene 16:1839–1849. 44. Le, X. F., P. Yang, and K. S. Chang. 1996. Analysis of the growth and transformation suppressor domains of promyelocytic leukemia gene, PML. J. Biol. Chem. 271:130–135. 45. Lee, J.-S., K. M. Galvin, and Y. Shi. 1993. Evidence for physical interaction between the zinc-finger transcription factor YY1 and Sp1. Proc. Natl. Acad. Sci. USA 90:6145–6149. 46. Lin, S.-Y., A. R. Black, D. Kostic, S. Pajovic, C. N. Hoover, and J. C. Azizkhan. 1996. Cell cycle-regulated association of E2F1 and Sp1 is related to their functional interaction. Mol. Cell. Biol. 16:1668–1675. 47. Liu, J. H., Z. M. Mu, and K. S. Chang. 1995. PML suppresses oncogenic transformation of NIH/3T3 cells by activated neu. J. Exp. Med. 181:1965–1973. 48. Morishige, K.-I., H. Kurachi, K. Amemiya, Y. Fujita, T. Yamamoto, A. Miyake, and O. Tanizawa. 1991. Evidence for the involvement of transforming growth factor alpha and epidermal growth factor receptor autocrine growth mechanism in primary human ovarian cancers in vitro. Cancer Res. 51:5322–5328. 49. Mu, Z.-M., X.-F. Le, S. Vallian, A. B. Glassman, and K.-S. Chang. 1997. Stable overexpression of PML alters regulation of cell cycle progression in HeLa cells. Carcinogenesis 18:2063–2069. 50. Mu, Z.-M., K.-V. Chin, J.-H. Liu, G. Lozano, and K.-S. Chang. 1994. PML, a growth suppressor disrupted in acute promyelocytic leukemia. Mol. Cell. Biol. 14:6858–6867. 51. Mukhopadhyay, D., B. Knebelmann, H. T. Cohen, S. Ananth, and V. P. Sukhatme. 1997. The von Hippel-Lindau tumor suppressor gene product interacts with Sp1 to repress vascular endothelial growth factor promoter activity. Mol. Cell. Biol. 17:5629–5639. 52. Noe, V., C. Chen, C. Alemany, M. Nicolas, I. Caragol, L. Chasin, and C. Ciudad. 1997. Cell-growth regulation of the hamster dihydrofolate reductase gene promoter by transcription factor Sp1. Eur. J. Biochem. 249:13–20. 53. Szekely, L., K. Pokrovskaja, W.-Q. Jiang, H. de The´, N. Ringertz, and G. Klein. 1996. The Epstein-Barr virus-encoded nuclear antigen EBNA-5 accumulates in PML-containing bodies. J. Virol. 70:2562–2568. 54. Toi, M., A. Osaki, H. Yamada, and T. Toge. 1991. Epidermal growth factor receptor expression as a prognostic indicator in breast cancer. Eur. J. Cancer 27:977–980. 55. Udvadia, A. J., K. T. Rogers, P. D. Higgins, Y. Murata, K. H. Martin, P. A. Humphrey, and J. M. Horowitz. 1993. Sp-1 binds promoter elements regulated by the RB protein and Sp-1-mediated transcription is stimulated by RB coexpression. Proc. Natl. Acad. Sci. USA 90:3265–3269. 56. Udvadia, A. J., D. J. Templeton, and J. M. Horowitz. 1995. Functional interactions between the retinoblastoma (Rb) protein and Sp-family members: superactivation by Rb requires amino acids necessary for growth suppression. Proc. Natl. Acad. Sci. USA 92:3953–3957. 57. Vallian, S., A. J. Gaken, E. B. Gingold, T. Kouzarides, K.-S. Chang, and F. Farzaneh. 1998. Modulation of Fos-mediated AP1 transcription by the promyelocytic leukemia protein. Oncogene 16:2843–2853. 58. Vallian, S., A. J. Gaken, I. D. Trayner, E. B. Gingold, T. Kouzarides, K.-S. Chang, and F. Farzaneh. 1997. Transcriptional repression by the promyelocytic leukemia protein, PML. Exp. Cell Res. 237:371–382. 59. Wang, Z. G., L. Delva, M. Gaboli, R. Rivi, M. Giorgio, C. Cordon-Cardo, F. Grosveld, and P. P. Pandolfi. 1998. Role of PML in cell growth and the retinoic acid pathway. Science 279:1547–1551. 60. Weis, K., S. Rambaud, C. Lavau, J. Jansen, T. Carvalho, M. Carmo Fonseca, A. Lamond, and A. Dejean. 1994. Retinoic acid regulates aberrant nuclear localization of PML-RARalpha in acute promyelocytic leukemia cells. Cell 76:345–356. 61. Xu, J., K. L. Thompson, L. B. Shephard, L. G. Hudson, and G. N. Gill. 1993. T3 receptor suppression of Sp1-dependent transcription from the epidermal growth factor receptor promoter via overlapping DNA-binding sites. J. Biol. Chem. 268:16065–16073.