The Role of Hepatocyte Nuclear Factor-3α (Forkhead Box A1) and Androgen Receptor in Transcriptional Regulation of Prostatic Genes (original) (raw)

Journal Article

,

1Department of Cell and Developmental Biology (N.G., J.Z., R.J.M.), Vanderbilt University Medical Center

2Department of Urologic Surgery (N.G., J.Z., T.C.C., J.M., Y.W., R.J., A.G., R.J.M.), and the Vanderbilt Prostate Cancer Center, Vanderbilt University

Search for other works by this author on:

,

1Department of Cell and Developmental Biology (N.G., J.Z., R.J.M.), Vanderbilt University Medical Center

2Department of Urologic Surgery (N.G., J.Z., T.C.C., J.M., Y.W., R.J., A.G., R.J.M.), and the Vanderbilt Prostate Cancer Center, Vanderbilt University

Search for other works by this author on:

,

4The Prostate Centre at Vancouver General Hospital (M.A.R., P.S.R.), Vancouver, British Columbia V6H 3Z6, Canada

Search for other works by this author on:

,

2Department of Urologic Surgery (N.G., J.Z., T.C.C., J.M., Y.W., R.J., A.G., R.J.M.), and the Vanderbilt Prostate Cancer Center, Vanderbilt University

3Department of Cancer Biology (T.C.C., J.M., Y.W., R.J., A.G., R.J.M.), and the Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232

Search for other works by this author on:

,

2Department of Urologic Surgery (N.G., J.Z., T.C.C., J.M., Y.W., R.J., A.G., R.J.M.), and the Vanderbilt Prostate Cancer Center, Vanderbilt University

3Department of Cancer Biology (T.C.C., J.M., Y.W., R.J., A.G., R.J.M.), and the Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232

Search for other works by this author on:

,

2Department of Urologic Surgery (N.G., J.Z., T.C.C., J.M., Y.W., R.J., A.G., R.J.M.), and the Vanderbilt Prostate Cancer Center, Vanderbilt University

3Department of Cancer Biology (T.C.C., J.M., Y.W., R.J., A.G., R.J.M.), and the Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232

Search for other works by this author on:

,

2Department of Urologic Surgery (N.G., J.Z., T.C.C., J.M., Y.W., R.J., A.G., R.J.M.), and the Vanderbilt Prostate Cancer Center, Vanderbilt University

3Department of Cancer Biology (T.C.C., J.M., Y.W., R.J., A.G., R.J.M.), and the Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232

Search for other works by this author on:

,

2Department of Urologic Surgery (N.G., J.Z., T.C.C., J.M., Y.W., R.J., A.G., R.J.M.), and the Vanderbilt Prostate Cancer Center, Vanderbilt University

3Department of Cancer Biology (T.C.C., J.M., Y.W., R.J., A.G., R.J.M.), and the Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232

Search for other works by this author on:

,

4The Prostate Centre at Vancouver General Hospital (M.A.R., P.S.R.), Vancouver, British Columbia V6H 3Z6, Canada

Search for other works by this author on:

1Department of Cell and Developmental Biology (N.G., J.Z., R.J.M.), Vanderbilt University Medical Center

2Department of Urologic Surgery (N.G., J.Z., T.C.C., J.M., Y.W., R.J., A.G., R.J.M.), and the Vanderbilt Prostate Cancer Center, Vanderbilt University

3Department of Cancer Biology (T.C.C., J.M., Y.W., R.J., A.G., R.J.M.), and the Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232

*Address all correspondence and requests for reprints to: Robert J. Matusik, Ph.D., Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee 37232.

Search for other works by this author on:

Received:

20 January 2003

Published:

01 August 2003

Cite

Nan Gao, Jianfeng Zhang, Mira A. Rao, Thomas C. Case, Janni Mirosevich, Yongqing Wang, Renjie Jin, Aparna Gupta, Paul S. Rennie, Robert J. Matusik, The Role of Hepatocyte Nuclear Factor-3α (Forkhead Box A1) and Androgen Receptor in Transcriptional Regulation of Prostatic Genes, Molecular Endocrinology, Volume 17, Issue 8, 1 August 2003, Pages 1484–1507, https://doi.org/10.1210/me.2003-0020
Close

Navbar Search Filter Mobile Enter search term Search

Abstract

Androgens and mesenchymal factors are essential extracellular signals for the development as well as the functional activity of the prostate epithelium. Little is known of the intraepithelial determinants that are involved in prostatic differentiation. Here we found that hepatocyte nuclear factor-3α (HNF-3α), an endoderm developmental factor, is essential for androgen receptor (AR)-mediated prostatic gene activation. Two HNF-3 **cis-regulatory elements were identified in the rat probasin (PB) gene promoter, each immediately adjacent to an androgen response element. Remarkably, similar organization of HNF-3 and AR binding sites was observed in the prostate-specific antigen (PSA) gene core enhancer, suggesting a common functional mechanism. Mutations that disrupt these HNF-3 motifs significantly abolished the maximal androgen induction of PB and PSA activities. Overexpressing a mutant HNF-3α deleted in the C-terminal region inhibited the androgen-induced promoter activity in LNCaP cells where endogenous HNF-3α is expressed. Chromatin immunoprecipitation revealed in vivo that the occupancy of HNF-3α on PSA enhancer can occur in an androgen-depleted condition, and before the recruitment of ligand-bound AR. A physical interaction of HNF-3α and AR was detected through immunoprecipitation and confirmed by glutathione-S**-transferase pull-down. This interaction is directly mediated through the DNA-binding domain/hinge region of AR and the forkhead domain of HNF-3α. In addition, strong HNF-3α expression, but not HNF-3β or HNF-3γ, is detected in both human and mouse prostatic epithelial cells where markers (PSA and PB) of differentiation are expressed. Taken together, these data support a model in which regulatory cues from the cell lineage and the extracellular environment coordinately establish the prostatic differentiated response.

THE PROSTATE IS a male accessory sex organ that is found exclusively in mammals. This organ produces various components found in the semen. Androgens are required to initiate prostatic development, to establish prostatic morphogenesis, and to begin secretory activities. In the mouse, the developing fetal testis begins to produce androgens at embryonic d 12.5–13 (E12.5–13) with peak production at E17–18, during which the earliest signs of prostatic formation are observed (1, 2). Ablation or surgical removal of fetal testes during the ambisexual period inhibits the development of prostate and other male internal sex organs (3). Androgens act directly through androgen receptor (AR) to elicit their effects (4). Testicular feminization mice, which express a nonfunctional AR, never develop a prostate, even though their testes produce adequate amounts of testosterone (3, 5). In addition, prostatic epithelial cells that do not express functional AR, also do not express characteristic secretory markers (6). These observations demonstrated the essential requirement of androgens and AR for prostate development and function. Interestingly, tissue recombination experiments have shown that the urogenital epithelium from testicular feminization mice develops normal prostate gland histology when combined with wild-type urogenital mesenchyme (UGM), although these urogenital epithelium contain a defective AR (7). Also, it has been observed that the induction of prostatic epithelial buds occurs before AR expression in wild-type prostatic epithelial cells (810). These results argue that the epithelial AR is not required for prostatic determination, although it is essential for the production of androgen-dependent secretory proteins at later stages (6).

Extensive tissue recombination studies have proven that mesenchymal-epithelial interactions play a key role in directing prostate development (3, 11). Epithelial cells from other urogenital sinus derivatives (bladder, urethra, and vagina) can be instructively induced by UGM to give rise to prostatic tissue (3, 1215). Similar results have been obtained using human bladder epithelial cells (16). The ductal-acinar structures that form in such experiments histologically resemble prostatic epithelium and produce prostate-specific antigen (PSA) (16). Nevertheless, the response of epithelia to inductive mesenchyme is limited by the developmental repertoire of the germ layer origin of the epithelium (17). For example, mesodermally derived seminal vesicle epithelium responds to either urogenital sinus or seminal vesicle mesenchyme by generating seminal vesicle. In contrast, the endodermally derived epithelia of the prostate, bladder, or urethra respond to the same inductive mesenchyme by generating prostatic tissue. Therefore, it is apparent that the cell developmental program is determined by both epithelial and mesenchymal factors. Although androgens and mesenchymal factors are critical for prostatic development, epithelial factors that directly participate in this process are still unknown.

Hepatocyte nuclear factor-3 proteins (newly named as forkhead box A proteins) are a group of endoderm-related developmental factors (HNF-3α, HNF-3β, and HNF-3γ) that belong to the forkhead box transcription factor family (1820). In mice, HNF-3β is first expressed in the endoderm progenitor during gastrulation (E6.5), immediately followed by the expression of HNF-3α (before organogenesis, E7–8) and HNF-3γ (19, 21). The targeted null mutation of HNF-3β gene results in embryonic lethality due to the absence of endodermal progenitor cells (22), whereas homozygous mutation of HNF-3α gene leads to perinatal death due to pancreatic defects (23). All three HNF-3 genes are restrictively expressed in the endoderm-derived organs in the adult and are involved in endodermal differentiation (19). In vivo footprinting using mouse embryo liver cells showed that HNF-3 factors, among a few of the earliest transcription factors, bind to the enhancer of liver-specific albumin gene before this gene is activated (19, 24). This binding of HNF-3 is believed to provide developmental competence to the target gene, which becomes activated during hepatic induction when additional liver- enriched transcription factors are recruited (19, 25, 26). Although HNF-3 proteins were first discovered in liver (27), the highest HNF-3α mRNA level was detected in the prostate gland by comparison of 16 human tissues (28). A novel expression pattern of HNF-3α in the epithelium of the bladder, urethra, and prostate has also been reported (28, 29). However, in the urogenital system, target genes that are regulated by HNF-3α have not been identified. The role of HNF-3α in the urogenital system remains to be defined.

In the adult prostate, fully differentiated luminal epithelial cells secrete androgen-dependent prostate-specific proteins, such as probasin (PB) in the rat and PSA in the human. Activation of these characteristic genes in the epithelium signifies that prostatic differentiation has occurred. The transcription factors that regulate these genes may also be involved in the process of prostate development. Therefore, a better understanding of the transcriptional regulation of these prostate-specific genes is likely to provide insight into organ development. In the present study, characterization of the PB promoter has led to the identification of two HNF-3α _cis_-regulatory motifs, which are immediately adjacent to functional AR binding sites (ARBSs). We have observed a similar organization of the androgen response element (ARE) adjacent to HNF-3α sites in the PSA gene enhancer and the prostatic acid phosphatase (PAP) gene promoter/enhancer, suggesting that a common functional mechanism is involved. DNA mutation studies showed that these HNF-3 sites are essential for maximal androgen induction of both PB and PSA gene. Chromatin immunoprecipitation (ChIP) assay confirmed that HNF-3α occupies the PSA enhancer in vivo. In addition, we detected a direct protein-protein interaction between HNF-3α and AR. These data suggest that an endodermal transcription factor HNF-3α and a steriod receptor AR may coordinately participate in the assembly of a nucleoprotein complex, which directs prostatic differentiated function. The regulatory mechanism between HNF-3α and AR may also apply to the transcriptional control of other prostatic epithelial cell-specific genes.

RESULTS

Two HNF-3 Binding Sites Are Identified on the PB Promoter

The rat PB is a prostate-specific and androgen-regulated gene, which has been extensively characterized (3038). It has been shown that a 12-kb fragment of the PB 5′-flanking sequence, a −426/+28-bp fragment as well as an artificial PB promoter, androgen-response region (ARR)2PB, which contains an extra copy of −244/−96 bp fused in front of the −286/+28-bp region, is able to specifically target genes to transgenic mouse prostate (31, 33, 35, 36, 3941). Therefore, the minimal fragment that confers prostate specificity was mapped to the 315-bp region (−286/+28) (36), which contains two functional ARBSs (ARBS-1 at −236/−223 and ARBS-2 at −140/−117) (30). Based on the hypothesis that additional key _cis_-acting elements remain to be uncovered, an extensive mutation analysis was performed. In this study, we report two critical HNF-3 motifs that are essential for promoter activity.

The first _cis_-acting element, designated as R1, is located at −253/−237 bp, immediately upstream to ARBS-1. The second element, designated as R2, is located at −126/−107 bp, downstream and slightly overlapping ARBS-2 (Fig. 1A). To identify putative transcription factor (TF)-binding sites, we employed web-based search engines TESS (www.cbil.upenn.edu/tess) and TFSEARCH (www.cbrc.jp/research/db/TFSEARCH.html). The search was restricted to a maximum of 20% mismatch within an element length of six nucleotides (nt) or greater. Either a 9-nt sequence (−252/−244 bp) located in R1 or a 9-nt sequence (−121/−113 bp) located in R2 highly matched with the HNF-3 consensus binding sequence (5′-TRTTTRYTY-3′) (20). The first 7-nt 5′-T(A/G)TT(T/G)(G/A)(T/C)-3′, extracted from various known HNF-3-regulated gene promoters (20), perfectly matched with the TATTTGT motif in R2 (Fig. 1A). Figure 1B shows a typical EMSA using nuclear extract prepared from LNCaP human prostate cancer cells, which express HNF-3α (Fig. 1C>). The TTRs (Fig. 1B) is a strong HNF-3 binding unit on the liver transthyretin (TTR) gene promoter and was originally used to affinity purify HNF-3 proteins (27, 42, 43). Strong complexes were formed when the TTRs consensus HNF-3 binding site was used (Fig. 1B, lane 2), and these complexes were reduced with the addition of antibodies against HNF-3α or HNF-3β (lanes 3 and 4). RT-PCR using primers specific for HNF-3β failed to detect the expression of HNF-3β in LNCaP cells (data not shown), indicating the effect caused by HNF-3β (M20) antibody may be due to the cross-reactivity of this antibody because HNF-3α and HNF-3β have highly homologous C termini. Four complexes, designated as a, b, c, and d, were formed with radiolabeled PB-127/-102, which contains R2 (Fig. 1B, lane7). Three complexes, designated as A, B, and C, were formed with PB-257/-232, which contains R1 (Fig. 1B, lane 20). HNF-3α antibody distinctly abolished the binding of two R2 complexes (lane 8, b and c) and R1 complex B (lane 21), whereas mock antibodies had no visible effect (lanes 10–16 and 23). A 200-fold molar excess of cold TTRs oligonucleotide completely competed off the complexes b and c (lane 17), indicating binding specificity. An increasing competitor (400- to 600-fold) further affected complex a (lanes 18 and 19), suggesting that a higher-order HNF-3α complex might be contained in complex a. Because complex a was not affected by HNF-3α antibody (lane 8), it indicates that this complex may have higher stability and the epitope in this complex may not be accessible to HNF-3α antibody. Western blot in Fig. 1C shows that HNF-3α protein was strongly detected in LNCaP and PC3, but weakly detected in DU145 cells in comparison with the positive control HepG2 cells, and the negative control Hela or Cos-1 cells. The expression of HNF-3α mRNA in these three prostatic cell lines (LNCaP, PC3, and DU145) was confirmed by RT-PCR (data not shown). Similar EMSA results in Fig. 1B were obtained when PC3 nuclear extract was used (data not shown).

Identification of Two HNF-3 Binding Sites in the PB Promoter A, Location of HNF-3 motifs. Mutation study revealed two key cis-acting elements R1 and R2 (dashed lines). Each contains a potential HNF-3 binding motif (bold arrows). The direction of the arrow indicates the sense or the antisense strand that matched the consensus sequence. Two adjacent ARBSs (ARBS-1 and ARBS-2) are underlined. Sequences in italic indicate the nucleotide replacements in mutation assays. B, EMSA. LNCaP nuclear extracts were incubated with radiolabeled TTRs (consensus HNF-3 binding sequence), PB−127/−102, or PB−257/−232 probes. Strong complexes formed with TTRs (lane 2). HNF-3α or HNF-3β antibody reduced the corresponding band (lanes 3 and 4). Four complexes, a, b, c and d, were formed with R2 (lane 7). Complexes b and c were specifically removed by HNF-3α or HNF-3β antibody (lanes 8 and 9), but not other mock antibodies (lanes 10–16). molar excess (200-fold) of cold TTRs oligonucleotides completely competed off complexes b and c (lane 17), whereas 400- and 600-fold molar excess of TTRs also affected complex a (lanes 18 and 19). Three complexes, A, B, and C, were formed with R1 (lane 18). Complex B was specifically eliminated by HNF-3α antibody (lane 19). C, Western blot. Whole-cell lysates (20 μg) from HepG2, Hela, Cos-1, LNCaP, PC3, and DU145 cells were resolved on SDS-PAGE and probed with HNF-3α antibody. D, A radiolabled oligonucleotide (ARBS2/R2, Table 1) containing both ARBS-2 and R2 was used in EMSA with 10 μg LNCaP nuclear extract. With the presence of an ARBS, two HNF-3α complexes (arrows) still formed with R2, because both complexes were reduced or removed by HNF-3α antibody (lane 1 vs. 2), or eliminated by mutating the HNF-3α motif in ARBS2/R2-M (lane 7, Table 1). Both complexes were not affected by 100-, 200-, and 500-fold molar excess of cold ARBS-2 site (lanes 4–6), whereas several other complexes (asterisk) were sensitive to the competition. E, Southwestern blot. Nuclear extracts from LNCaP and PC-3 cells were separated on SDS-PAGE, transblotted to nitrocellulose membranes, and probed with radiolabeled 2×R2 (lanes 1 and 3), 2×mR2 (lanes 2 and 4) or 2×AR-con probes (lane 5). A Western blot using HNF-3α antibody was performed as a parallel control. Arrows indicate migration of proteins bound to the respective probes or antibody.

Identification of Two HNF-3 Binding Sites in the PB Promoter A, Location of HNF-3 motifs. Mutation study revealed two key cis-acting elements R1 and R2 (dashed lines). Each contains a potential HNF-3 binding motif (bold arrows). The direction of the arrow indicates the sense or the antisense strand that matched the consensus sequence. Two adjacent ARBSs (ARBS-1 and ARBS-2) are underlined. Sequences in italic indicate the nucleotide replacements in mutation assays. B, EMSA. LNCaP nuclear extracts were incubated with radiolabeled TTRs (consensus HNF-3 binding sequence), PB−127/−102, or PB−257/−232 probes. Strong complexes formed with TTRs (lane 2). HNF-3α or HNF-3β antibody reduced the corresponding band (lanes 3 and 4). Four complexes, a, b, c and d, were formed with R2 (lane 7). Complexes b and c were specifically removed by HNF-3α or HNF-3β antibody (lanes 8 and 9), but not other mock antibodies (lanes 10–16). molar excess (200-fold) of cold TTRs oligonucleotides completely competed off complexes b and c (lane 17), whereas 400- and 600-fold molar excess of TTRs also affected complex a (lanes 18 and 19). Three complexes, A, B, and C, were formed with R1 (lane 18). Complex B was specifically eliminated by HNF-3α antibody (lane 19). C, Western blot. Whole-cell lysates (20 μg) from HepG2, Hela, Cos-1, LNCaP, PC3, and DU145 cells were resolved on SDS-PAGE and probed with HNF-3α antibody. D, A radiolabled oligonucleotide (ARBS2/R2, Table 1) containing both ARBS-2 and R2 was used in EMSA with 10 μg LNCaP nuclear extract. With the presence of an ARBS, two HNF-3α complexes (arrows) still formed with R2, because both complexes were reduced or removed by HNF-3α antibody (lane 1 vs. 2), or eliminated by mutating the HNF-3α motif in ARBS2/R2-M (lane 7, Table 1). Both complexes were not affected by 100-, 200-, and 500-fold molar excess of cold ARBS-2 site (lanes 4–6), whereas several other complexes (asterisk) were sensitive to the competition. E, Southwestern blot. Nuclear extracts from LNCaP and PC-3 cells were separated on SDS-PAGE, transblotted to nitrocellulose membranes, and probed with radiolabeled 2×R2 (lanes 1 and 3), 2×mR2 (lanes 2 and 4) or 2×AR-con probes (lane 5). A Western blot using HNF-3α antibody was performed as a parallel control. Arrows indicate migration of proteins bound to the respective probes or antibody.

Fig. 1.

Identification of Two HNF-3 Binding Sites in the PB Promoter A, Location of HNF-3 motifs. Mutation study revealed two key _cis_-acting elements R1 and R2 (dashed lines). Each contains a potential HNF-3 binding motif (bold arrows). The direction of the arrow indicates the sense or the antisense strand that matched the consensus sequence. Two adjacent ARBSs (ARBS-1 and ARBS-2) are underlined. Sequences in italic indicate the nucleotide replacements in mutation assays. B, EMSA. LNCaP nuclear extracts were incubated with radiolabeled TTRs (consensus HNF-3 binding sequence), PB−127/−102, or PB−257/−232 probes. Strong complexes formed with TTRs (lane 2). HNF-3α or HNF-3β antibody reduced the corresponding band (lanes 3 and 4). Four complexes, a, b, c and d, were formed with R2 (lane 7). Complexes b and c were specifically removed by HNF-3α or HNF-3β antibody (lanes 8 and 9), but not other mock antibodies (lanes 10–16). molar excess (200-fold) of cold TTRs oligonucleotides completely competed off complexes b and c (lane 17), whereas 400- and 600-fold molar excess of TTRs also affected complex a (lanes 18 and 19). Three complexes, A, B, and C, were formed with R1 (lane 18). Complex B was specifically eliminated by HNF-3α antibody (lane 19). C, Western blot. Whole-cell lysates (20 μg) from HepG2, Hela, Cos-1, LNCaP, PC3, and DU145 cells were resolved on SDS-PAGE and probed with HNF-3α antibody. D, A radiolabled oligonucleotide (ARBS2/R2, Table 1) containing both ARBS-2 and R2 was used in EMSA with 10 μg LNCaP nuclear extract. With the presence of an ARBS, two HNF-3α complexes (arrows) still formed with R2, because both complexes were reduced or removed by HNF-3α antibody (lane 1 vs. 2), or eliminated by mutating the HNF-3α motif in ARBS2/R2-M (lane 7, Table 1). Both complexes were not affected by 100-, 200-, and 500-fold molar excess of cold ARBS-2 site (lanes 4–6), whereas several other complexes (asterisk) were sensitive to the competition. E, Southwestern blot. Nuclear extracts from LNCaP and PC-3 cells were separated on SDS-PAGE, transblotted to nitrocellulose membranes, and probed with radiolabeled 2×R2 (lanes 1 and 3), 2×mR2 (lanes 2 and 4) or 2×AR-con probes (lane 5). A Western blot using HNF-3α antibody was performed as a parallel control. Arrows indicate migration of proteins bound to the respective probes or antibody.

Because ARBS-2 slightly overlaps with the HNF-3α motif in R2, it is necessary to determine 1) whether HNF-3α still binds R2 when the ARBS-2 site is occupied by AR, 2) whether the HNF-3α/R2 interaction is dependent on adjacent AR binding. To address these questions in vitro, we used a radiolabeled probe (Table 1, ARBS2/R2) containing both ARBS-2 and R2. Figure 1D shows that, among the multiple complexes formed with LNCaP nuclear extract, two complexes were specifically reduced or removed by HNF-3α antibody (lane 1 vs. 2), whereas both complexes were disrupted by mutating the HNF-3 motif in ARBS2/R2 string (lane 3 vs. 7 and Table 1, ARBS2/R2-M1). Competition assay, using 100-, 200-, and 500-fold molar excess of cold ARBS-2 site (lane 3 vs. 4–6), showed that several complexes (indicated as asterisks) were sensitive to the competitor, whereas none of the HNF-3α complexes were competed off. These results demonstrated in vitro that HNF-3α/R2 binding can occur concomitantly with AR/ARBS-2 interaction, but HNF-3α binding is independent of AR/ARBS-2 interaction.

Table 1.

Oligonucleotides Used in this Study

Name Sequence Purpose Restriction Site Added
TTRs 5′-TGACTAAGTCAATAATCAGAATCAG-3′ EMSA, Competition
PB-127/−102 5′-AGAACCTATTTGTATACTAGATGACA-3′ EMSA
PB-257/−232 5′-CAGTTAAGAAAATATGATAGCATCTT-3′ EMSA
ARBS2/R2 5′-GCCTAGTAAAGTACTCCAAGAACCTATTTGTATACTAGA-3′ EMSA
ARBS2/R2-M1 5′-GCCTAGTAAAGTACTCCAAGAACCTAgaaGTATACTAGA-3′ EMSA
ARBS2/R2-M2 5′-GCCTAGTAAAGTACTCCAAGAACCTATTTGatgACTAGA-3′ EMSA
ARBS1/R1 5′-CAGTTAAGAAAATATGATAGCATCTTGTTCTTAGTCTTTTT-3′ EMSA
ARBS1/R1-M3 5′-CAGTTAActAAATatGAatGCATCTTGTTCTTAGTCTTTTT-3′ EMSA
PB−127/−102-M1-F 5′-AGAACCTAgaaGTATACTAGATGACA-3′ EMSA, Mutagenesis
PB−127/−102-M1-R 5′-TGTCATCTAGTATACttcTAGGTTCT-3′ EMSA, Mutagenesis
PB−127/−102-M2-F 5′-AGAACCTATTTGatgACTAGATGACA-3′ EMSA, Mutagenesis
PB−127/−102-M2-R 5′-TGTCATCTAGTcatCAAATAGGTTCT-3′ EMSA, Mutagenesis
PB−257/−232M-F 5′-CAGTTAActAAATtaGAatGCATCTT-3′ Mutagenesis
PB−257/−232M-R 5′-AAGATGCatTCtaATTTagTTAACTG-3′ Mutagenesis
2 × R2 5′-ACCTATTTGTATACTAACCTATTTGTATACTAGATGACA-3′ Southwestern
2 × mR2 5′-ACCTAgaaGTATACTAACCTAgaaGTATACTAGATGACA-3′ Southwestern
PB-Primer 5′-TGTCATCTAG-3′ Southwestern
2 × AR-consensus 5′-GTCTGGTACAGGGTGTTCTTTTTGGTACAGGGTGTTCTTTTTG Southwestern
AR-Primer 5′-CAAAAAGAAC-3′ Southwestern
PSA1 5′-CCTACTCTGGAGGAACATATTGTATTGATTGTCCTTGACAGTA AACAAATCTGTT-3′ EMSA
PSA2 5′-GGATGCCTGCTTTACAAACATCCTTGAAAC-3′ EMSA
mPSA1 5′-CCTACTCTGGAGGAACATATTGTATTGATTGTCCTTGACAGT AcACAcATCTGTT-3′ EMSA
mPSA2 5′-GGATGCCTGCTcTcCAcACATCCTTGAAAC-3′ EMSA
hPAP 5′-TTCTTTTGTTTGTTTTTTGTTGTTGTTTGTTTGTTTTGCTG-3′ EMSA
rPAP1 5′-TGGTTTTGTTTTCTTTTTAATGTTTGTTAT-3′ EMSA
rPAP2 5′-GCCAATCTCTTGATTAAATAGGCACTTCCC-3′ EMSA
rPAP3 5′-TCTTGACAGAACAGGAAGCCGAGAGTGAGC-3′ EMSA
PBPC1-1 5′-ATTGACTGAATGTTTATTTAATTTCTCCTT-3′ EMSA
PBPC1-2 5′-GAATTAGCAAATAATTTCTCTTATAAAAAT-3′ EMSA
PSA-P-F 5′-CCCGGGTTGGATTTTGAAATGCTA-3′ PSA-EPLuc _Sma_I
PSA-P-R 5′-CTCGAGAAGCTTGGGGCTGG-3′ PSA-EPLuc _Xho_I
PSA-E-F 5′-GAGCTCCTGCAGAGAAATTA-3′ PSA-EPLuc _Sac_I
PSA-E-R 5′-CCCGGGCCATGGTTCTGTCA-3′ PSA-EPLuc _Sma_I
mPSA1-F 5′-TGACAGTAcACAcATCTGTTGTAAG-3′ Mutagenesis
mPSA2-F 5′-TGCTcTcCAcACATCCTTGAAACAA-3′ Mutagenesis
WT (1–466)-F 5′-CACCGAATTCATGTTAGGGACTGTGAAGAT-3′ WT construct _Eco_RI
WT (1–466)-R 5′-AGCTCGAGCGGAAGTATTTAGCACGGGTCT-3′ WT construct _Xho_I
NT (1–180)-R 5′-AGCTCGAGCGGTGATGAGCGAGATGTAGGA-3′ NT construct _Xho_I
FH (141–294)-F 5′-CACCGAATTCATGGCGTACGCTCCGTCCAA-3′ FH construct _Eco_RI
FH (141–294)-R 5′-AGCTCGAGCTGAGGGGTCTTTGCGGTTTTC-3′ FH construct _Xho_I
CT (295–466)-F 5′-CACCGAATTCATGGGCCCGGTTAACCCCAGTGC-3′ CT construct _Eco_RI
Δ23 (1–385)-R 5′-AGCTCGAGCGGCCCCTTTCAGGTGCAGCTG-3′ Δ23 construct _Xho_I
Δ3 (1–420)-R 5′-AGCTCGAGCGTACTGCAGTGCCTGCTGATA-3′ Δ3 construct _Xho_I
DN (59–345)-F 5′-CACCGAATTCATGACCCCGGCTTCCTTCAA-3′ DN construct _Eco_RI
DN (59–345)-R 5′-AGCTCGAGCCAACTCCGAACCGCCCCCTGT-3′ DN construct _Xho_I
AR-LBD-F 5′-TCCGGATCCAAGGCTATGAATGTCAACCT-3′ GST-AR-LBD _Bam_HI
AR-LBD-R 5′-TCCGGATCCTCACTGTGTGTGGAAATAGAT-3′ GST-AR-LBD _Bam_HI
AR-NT-F 5′-TTTGGATCCCCATGGAGGTGCAGTTAGGGCTG-3′ GST-AR-NTD _Bam_HI
AR-NT-R 5′-TTTGGATCCTCACATGTCCCCATAAGGTCCGGA-3′ GST-AR-NTD _Bam_HI
Name Sequence Purpose Restriction Site Added
TTRs 5′-TGACTAAGTCAATAATCAGAATCAG-3′ EMSA, Competition
PB-127/−102 5′-AGAACCTATTTGTATACTAGATGACA-3′ EMSA
PB-257/−232 5′-CAGTTAAGAAAATATGATAGCATCTT-3′ EMSA
ARBS2/R2 5′-GCCTAGTAAAGTACTCCAAGAACCTATTTGTATACTAGA-3′ EMSA
ARBS2/R2-M1 5′-GCCTAGTAAAGTACTCCAAGAACCTAgaaGTATACTAGA-3′ EMSA
ARBS2/R2-M2 5′-GCCTAGTAAAGTACTCCAAGAACCTATTTGatgACTAGA-3′ EMSA
ARBS1/R1 5′-CAGTTAAGAAAATATGATAGCATCTTGTTCTTAGTCTTTTT-3′ EMSA
ARBS1/R1-M3 5′-CAGTTAActAAATatGAatGCATCTTGTTCTTAGTCTTTTT-3′ EMSA
PB−127/−102-M1-F 5′-AGAACCTAgaaGTATACTAGATGACA-3′ EMSA, Mutagenesis
PB−127/−102-M1-R 5′-TGTCATCTAGTATACttcTAGGTTCT-3′ EMSA, Mutagenesis
PB−127/−102-M2-F 5′-AGAACCTATTTGatgACTAGATGACA-3′ EMSA, Mutagenesis
PB−127/−102-M2-R 5′-TGTCATCTAGTcatCAAATAGGTTCT-3′ EMSA, Mutagenesis
PB−257/−232M-F 5′-CAGTTAActAAATtaGAatGCATCTT-3′ Mutagenesis
PB−257/−232M-R 5′-AAGATGCatTCtaATTTagTTAACTG-3′ Mutagenesis
2 × R2 5′-ACCTATTTGTATACTAACCTATTTGTATACTAGATGACA-3′ Southwestern
2 × mR2 5′-ACCTAgaaGTATACTAACCTAgaaGTATACTAGATGACA-3′ Southwestern
PB-Primer 5′-TGTCATCTAG-3′ Southwestern
2 × AR-consensus 5′-GTCTGGTACAGGGTGTTCTTTTTGGTACAGGGTGTTCTTTTTG Southwestern
AR-Primer 5′-CAAAAAGAAC-3′ Southwestern
PSA1 5′-CCTACTCTGGAGGAACATATTGTATTGATTGTCCTTGACAGTA AACAAATCTGTT-3′ EMSA
PSA2 5′-GGATGCCTGCTTTACAAACATCCTTGAAAC-3′ EMSA
mPSA1 5′-CCTACTCTGGAGGAACATATTGTATTGATTGTCCTTGACAGT AcACAcATCTGTT-3′ EMSA
mPSA2 5′-GGATGCCTGCTcTcCAcACATCCTTGAAAC-3′ EMSA
hPAP 5′-TTCTTTTGTTTGTTTTTTGTTGTTGTTTGTTTGTTTTGCTG-3′ EMSA
rPAP1 5′-TGGTTTTGTTTTCTTTTTAATGTTTGTTAT-3′ EMSA
rPAP2 5′-GCCAATCTCTTGATTAAATAGGCACTTCCC-3′ EMSA
rPAP3 5′-TCTTGACAGAACAGGAAGCCGAGAGTGAGC-3′ EMSA
PBPC1-1 5′-ATTGACTGAATGTTTATTTAATTTCTCCTT-3′ EMSA
PBPC1-2 5′-GAATTAGCAAATAATTTCTCTTATAAAAAT-3′ EMSA
PSA-P-F 5′-CCCGGGTTGGATTTTGAAATGCTA-3′ PSA-EPLuc _Sma_I
PSA-P-R 5′-CTCGAGAAGCTTGGGGCTGG-3′ PSA-EPLuc _Xho_I
PSA-E-F 5′-GAGCTCCTGCAGAGAAATTA-3′ PSA-EPLuc _Sac_I
PSA-E-R 5′-CCCGGGCCATGGTTCTGTCA-3′ PSA-EPLuc _Sma_I
mPSA1-F 5′-TGACAGTAcACAcATCTGTTGTAAG-3′ Mutagenesis
mPSA2-F 5′-TGCTcTcCAcACATCCTTGAAACAA-3′ Mutagenesis
WT (1–466)-F 5′-CACCGAATTCATGTTAGGGACTGTGAAGAT-3′ WT construct _Eco_RI
WT (1–466)-R 5′-AGCTCGAGCGGAAGTATTTAGCACGGGTCT-3′ WT construct _Xho_I
NT (1–180)-R 5′-AGCTCGAGCGGTGATGAGCGAGATGTAGGA-3′ NT construct _Xho_I
FH (141–294)-F 5′-CACCGAATTCATGGCGTACGCTCCGTCCAA-3′ FH construct _Eco_RI
FH (141–294)-R 5′-AGCTCGAGCTGAGGGGTCTTTGCGGTTTTC-3′ FH construct _Xho_I
CT (295–466)-F 5′-CACCGAATTCATGGGCCCGGTTAACCCCAGTGC-3′ CT construct _Eco_RI
Δ23 (1–385)-R 5′-AGCTCGAGCGGCCCCTTTCAGGTGCAGCTG-3′ Δ23 construct _Xho_I
Δ3 (1–420)-R 5′-AGCTCGAGCGTACTGCAGTGCCTGCTGATA-3′ Δ3 construct _Xho_I
DN (59–345)-F 5′-CACCGAATTCATGACCCCGGCTTCCTTCAA-3′ DN construct _Eco_RI
DN (59–345)-R 5′-AGCTCGAGCCAACTCCGAACCGCCCCCTGT-3′ DN construct _Xho_I
AR-LBD-F 5′-TCCGGATCCAAGGCTATGAATGTCAACCT-3′ GST-AR-LBD _Bam_HI
AR-LBD-R 5′-TCCGGATCCTCACTGTGTGTGGAAATAGAT-3′ GST-AR-LBD _Bam_HI
AR-NT-F 5′-TTTGGATCCCCATGGAGGTGCAGTTAGGGCTG-3′ GST-AR-NTD _Bam_HI
AR-NT-R 5′-TTTGGATCCTCACATGTCCCCATAAGGTCCGGA-3′ GST-AR-NTD _Bam_HI

For EMSA probes, only the sense strand sequences are shown here. Underscored sequences represent the binding sites for HNF-3 or AR. Sequences in bold lowercase characters are nucleotide replacements in mutagenesis studies.

Table 1.

Oligonucleotides Used in this Study

Name Sequence Purpose Restriction Site Added
TTRs 5′-TGACTAAGTCAATAATCAGAATCAG-3′ EMSA, Competition
PB-127/−102 5′-AGAACCTATTTGTATACTAGATGACA-3′ EMSA
PB-257/−232 5′-CAGTTAAGAAAATATGATAGCATCTT-3′ EMSA
ARBS2/R2 5′-GCCTAGTAAAGTACTCCAAGAACCTATTTGTATACTAGA-3′ EMSA
ARBS2/R2-M1 5′-GCCTAGTAAAGTACTCCAAGAACCTAgaaGTATACTAGA-3′ EMSA
ARBS2/R2-M2 5′-GCCTAGTAAAGTACTCCAAGAACCTATTTGatgACTAGA-3′ EMSA
ARBS1/R1 5′-CAGTTAAGAAAATATGATAGCATCTTGTTCTTAGTCTTTTT-3′ EMSA
ARBS1/R1-M3 5′-CAGTTAActAAATatGAatGCATCTTGTTCTTAGTCTTTTT-3′ EMSA
PB−127/−102-M1-F 5′-AGAACCTAgaaGTATACTAGATGACA-3′ EMSA, Mutagenesis
PB−127/−102-M1-R 5′-TGTCATCTAGTATACttcTAGGTTCT-3′ EMSA, Mutagenesis
PB−127/−102-M2-F 5′-AGAACCTATTTGatgACTAGATGACA-3′ EMSA, Mutagenesis
PB−127/−102-M2-R 5′-TGTCATCTAGTcatCAAATAGGTTCT-3′ EMSA, Mutagenesis
PB−257/−232M-F 5′-CAGTTAActAAATtaGAatGCATCTT-3′ Mutagenesis
PB−257/−232M-R 5′-AAGATGCatTCtaATTTagTTAACTG-3′ Mutagenesis
2 × R2 5′-ACCTATTTGTATACTAACCTATTTGTATACTAGATGACA-3′ Southwestern
2 × mR2 5′-ACCTAgaaGTATACTAACCTAgaaGTATACTAGATGACA-3′ Southwestern
PB-Primer 5′-TGTCATCTAG-3′ Southwestern
2 × AR-consensus 5′-GTCTGGTACAGGGTGTTCTTTTTGGTACAGGGTGTTCTTTTTG Southwestern
AR-Primer 5′-CAAAAAGAAC-3′ Southwestern
PSA1 5′-CCTACTCTGGAGGAACATATTGTATTGATTGTCCTTGACAGTA AACAAATCTGTT-3′ EMSA
PSA2 5′-GGATGCCTGCTTTACAAACATCCTTGAAAC-3′ EMSA
mPSA1 5′-CCTACTCTGGAGGAACATATTGTATTGATTGTCCTTGACAGT AcACAcATCTGTT-3′ EMSA
mPSA2 5′-GGATGCCTGCTcTcCAcACATCCTTGAAAC-3′ EMSA
hPAP 5′-TTCTTTTGTTTGTTTTTTGTTGTTGTTTGTTTGTTTTGCTG-3′ EMSA
rPAP1 5′-TGGTTTTGTTTTCTTTTTAATGTTTGTTAT-3′ EMSA
rPAP2 5′-GCCAATCTCTTGATTAAATAGGCACTTCCC-3′ EMSA
rPAP3 5′-TCTTGACAGAACAGGAAGCCGAGAGTGAGC-3′ EMSA
PBPC1-1 5′-ATTGACTGAATGTTTATTTAATTTCTCCTT-3′ EMSA
PBPC1-2 5′-GAATTAGCAAATAATTTCTCTTATAAAAAT-3′ EMSA
PSA-P-F 5′-CCCGGGTTGGATTTTGAAATGCTA-3′ PSA-EPLuc _Sma_I
PSA-P-R 5′-CTCGAGAAGCTTGGGGCTGG-3′ PSA-EPLuc _Xho_I
PSA-E-F 5′-GAGCTCCTGCAGAGAAATTA-3′ PSA-EPLuc _Sac_I
PSA-E-R 5′-CCCGGGCCATGGTTCTGTCA-3′ PSA-EPLuc _Sma_I
mPSA1-F 5′-TGACAGTAcACAcATCTGTTGTAAG-3′ Mutagenesis
mPSA2-F 5′-TGCTcTcCAcACATCCTTGAAACAA-3′ Mutagenesis
WT (1–466)-F 5′-CACCGAATTCATGTTAGGGACTGTGAAGAT-3′ WT construct _Eco_RI
WT (1–466)-R 5′-AGCTCGAGCGGAAGTATTTAGCACGGGTCT-3′ WT construct _Xho_I
NT (1–180)-R 5′-AGCTCGAGCGGTGATGAGCGAGATGTAGGA-3′ NT construct _Xho_I
FH (141–294)-F 5′-CACCGAATTCATGGCGTACGCTCCGTCCAA-3′ FH construct _Eco_RI
FH (141–294)-R 5′-AGCTCGAGCTGAGGGGTCTTTGCGGTTTTC-3′ FH construct _Xho_I
CT (295–466)-F 5′-CACCGAATTCATGGGCCCGGTTAACCCCAGTGC-3′ CT construct _Eco_RI
Δ23 (1–385)-R 5′-AGCTCGAGCGGCCCCTTTCAGGTGCAGCTG-3′ Δ23 construct _Xho_I
Δ3 (1–420)-R 5′-AGCTCGAGCGTACTGCAGTGCCTGCTGATA-3′ Δ3 construct _Xho_I
DN (59–345)-F 5′-CACCGAATTCATGACCCCGGCTTCCTTCAA-3′ DN construct _Eco_RI
DN (59–345)-R 5′-AGCTCGAGCCAACTCCGAACCGCCCCCTGT-3′ DN construct _Xho_I
AR-LBD-F 5′-TCCGGATCCAAGGCTATGAATGTCAACCT-3′ GST-AR-LBD _Bam_HI
AR-LBD-R 5′-TCCGGATCCTCACTGTGTGTGGAAATAGAT-3′ GST-AR-LBD _Bam_HI
AR-NT-F 5′-TTTGGATCCCCATGGAGGTGCAGTTAGGGCTG-3′ GST-AR-NTD _Bam_HI
AR-NT-R 5′-TTTGGATCCTCACATGTCCCCATAAGGTCCGGA-3′ GST-AR-NTD _Bam_HI
Name Sequence Purpose Restriction Site Added
TTRs 5′-TGACTAAGTCAATAATCAGAATCAG-3′ EMSA, Competition
PB-127/−102 5′-AGAACCTATTTGTATACTAGATGACA-3′ EMSA
PB-257/−232 5′-CAGTTAAGAAAATATGATAGCATCTT-3′ EMSA
ARBS2/R2 5′-GCCTAGTAAAGTACTCCAAGAACCTATTTGTATACTAGA-3′ EMSA
ARBS2/R2-M1 5′-GCCTAGTAAAGTACTCCAAGAACCTAgaaGTATACTAGA-3′ EMSA
ARBS2/R2-M2 5′-GCCTAGTAAAGTACTCCAAGAACCTATTTGatgACTAGA-3′ EMSA
ARBS1/R1 5′-CAGTTAAGAAAATATGATAGCATCTTGTTCTTAGTCTTTTT-3′ EMSA
ARBS1/R1-M3 5′-CAGTTAActAAATatGAatGCATCTTGTTCTTAGTCTTTTT-3′ EMSA
PB−127/−102-M1-F 5′-AGAACCTAgaaGTATACTAGATGACA-3′ EMSA, Mutagenesis
PB−127/−102-M1-R 5′-TGTCATCTAGTATACttcTAGGTTCT-3′ EMSA, Mutagenesis
PB−127/−102-M2-F 5′-AGAACCTATTTGatgACTAGATGACA-3′ EMSA, Mutagenesis
PB−127/−102-M2-R 5′-TGTCATCTAGTcatCAAATAGGTTCT-3′ EMSA, Mutagenesis
PB−257/−232M-F 5′-CAGTTAActAAATtaGAatGCATCTT-3′ Mutagenesis
PB−257/−232M-R 5′-AAGATGCatTCtaATTTagTTAACTG-3′ Mutagenesis
2 × R2 5′-ACCTATTTGTATACTAACCTATTTGTATACTAGATGACA-3′ Southwestern
2 × mR2 5′-ACCTAgaaGTATACTAACCTAgaaGTATACTAGATGACA-3′ Southwestern
PB-Primer 5′-TGTCATCTAG-3′ Southwestern
2 × AR-consensus 5′-GTCTGGTACAGGGTGTTCTTTTTGGTACAGGGTGTTCTTTTTG Southwestern
AR-Primer 5′-CAAAAAGAAC-3′ Southwestern
PSA1 5′-CCTACTCTGGAGGAACATATTGTATTGATTGTCCTTGACAGTA AACAAATCTGTT-3′ EMSA
PSA2 5′-GGATGCCTGCTTTACAAACATCCTTGAAAC-3′ EMSA
mPSA1 5′-CCTACTCTGGAGGAACATATTGTATTGATTGTCCTTGACAGT AcACAcATCTGTT-3′ EMSA
mPSA2 5′-GGATGCCTGCTcTcCAcACATCCTTGAAAC-3′ EMSA
hPAP 5′-TTCTTTTGTTTGTTTTTTGTTGTTGTTTGTTTGTTTTGCTG-3′ EMSA
rPAP1 5′-TGGTTTTGTTTTCTTTTTAATGTTTGTTAT-3′ EMSA
rPAP2 5′-GCCAATCTCTTGATTAAATAGGCACTTCCC-3′ EMSA
rPAP3 5′-TCTTGACAGAACAGGAAGCCGAGAGTGAGC-3′ EMSA
PBPC1-1 5′-ATTGACTGAATGTTTATTTAATTTCTCCTT-3′ EMSA
PBPC1-2 5′-GAATTAGCAAATAATTTCTCTTATAAAAAT-3′ EMSA
PSA-P-F 5′-CCCGGGTTGGATTTTGAAATGCTA-3′ PSA-EPLuc _Sma_I
PSA-P-R 5′-CTCGAGAAGCTTGGGGCTGG-3′ PSA-EPLuc _Xho_I
PSA-E-F 5′-GAGCTCCTGCAGAGAAATTA-3′ PSA-EPLuc _Sac_I
PSA-E-R 5′-CCCGGGCCATGGTTCTGTCA-3′ PSA-EPLuc _Sma_I
mPSA1-F 5′-TGACAGTAcACAcATCTGTTGTAAG-3′ Mutagenesis
mPSA2-F 5′-TGCTcTcCAcACATCCTTGAAACAA-3′ Mutagenesis
WT (1–466)-F 5′-CACCGAATTCATGTTAGGGACTGTGAAGAT-3′ WT construct _Eco_RI
WT (1–466)-R 5′-AGCTCGAGCGGAAGTATTTAGCACGGGTCT-3′ WT construct _Xho_I
NT (1–180)-R 5′-AGCTCGAGCGGTGATGAGCGAGATGTAGGA-3′ NT construct _Xho_I
FH (141–294)-F 5′-CACCGAATTCATGGCGTACGCTCCGTCCAA-3′ FH construct _Eco_RI
FH (141–294)-R 5′-AGCTCGAGCTGAGGGGTCTTTGCGGTTTTC-3′ FH construct _Xho_I
CT (295–466)-F 5′-CACCGAATTCATGGGCCCGGTTAACCCCAGTGC-3′ CT construct _Eco_RI
Δ23 (1–385)-R 5′-AGCTCGAGCGGCCCCTTTCAGGTGCAGCTG-3′ Δ23 construct _Xho_I
Δ3 (1–420)-R 5′-AGCTCGAGCGTACTGCAGTGCCTGCTGATA-3′ Δ3 construct _Xho_I
DN (59–345)-F 5′-CACCGAATTCATGACCCCGGCTTCCTTCAA-3′ DN construct _Eco_RI
DN (59–345)-R 5′-AGCTCGAGCCAACTCCGAACCGCCCCCTGT-3′ DN construct _Xho_I
AR-LBD-F 5′-TCCGGATCCAAGGCTATGAATGTCAACCT-3′ GST-AR-LBD _Bam_HI
AR-LBD-R 5′-TCCGGATCCTCACTGTGTGTGGAAATAGAT-3′ GST-AR-LBD _Bam_HI
AR-NT-F 5′-TTTGGATCCCCATGGAGGTGCAGTTAGGGCTG-3′ GST-AR-NTD _Bam_HI
AR-NT-R 5′-TTTGGATCCTCACATGTCCCCATAAGGTCCGGA-3′ GST-AR-NTD _Bam_HI

For EMSA probes, only the sense strand sequences are shown here. Underscored sequences represent the binding sites for HNF-3 or AR. Sequences in bold lowercase characters are nucleotide replacements in mutagenesis studies.

In Fig. 1E, Southwestern blot was used to determine the relative molecular masses of R2-interacting proteins. LNCaP and PC-3 cell nuclear extracts were resolved on sodium dodecyl sulfate (SDS)-polyacrylamide gels and transferred to nitrocellulose membranes. The blots were probed with 32P-labeled oligonucleotides containing two tandem copies of wild-type R2 (2×R2). For both LNCaP and PC-3 cells, two proteins with molecular masses of approximately 52 kDa and 46 kDa were detected (lanes 1 and 3). Similar blots probed with 2×mR2 (containing two copies of mutant R2, see Table 1 for sequence) showed that the binding of both proteins was distinctly reduced in LNCaP cells or completely blocked in PC-3 cells (lanes 2 and 4), indicating that the binding is sequence specific. A radiolabeled oligonucleotide containing two tandem copies of AR consensus binding site (Table 1), serving as a positive control, was hybridized specifically to a 110-kDa protein (the size of human AR), but not the low molecular mass proteins (lane 5). A parallel Western blot using HNF-3α antibody showed a specific band around 52 kDa (lanes 6 and 7), suggesting that the 52-kDa protein detected by Southwestern blot was HNF-3α. The 46-kDa protein detected in both LNCaP and PC3 might be an HNF-3α minor degradation product that escaped antibody detection but still retained DNA binding activity. However, the presence of a higher-order protein complex on R2 (complex a in Fig. 1B, lane 7) suggests that additional DNA-binding protein on R2 may exist.

In Vitro Synthesized HNF-3α Binds R1 and R2

Because crude nuclear extracts were used in EMSA and Southwestern experiments, it was necessary to confirm the results with HNF-3α synthesized by in vitro transcription/translation (TNT). Figure 2 shows that synthesized HNF-3α specifically bound to the radiolabeled R2 in EMSA (Fig. 2A, lane 3), in contrast to the TNT blank control (lane 2), which only formed a nonspecific band. Three mutant HNF-3α proteins deleted in the N-terminal (ΔN), C-terminal (ΔC), or both terminal regions [FH (forkhead)] still showed binding activities (lanes 4–6), because all mutants still contain the FH DNA-binding domain (DBD). The binding of HNF-3α was completely removed by an antibody against the HNF-3α C terminus (Fig. 2B, lane 2 vs. 3). The same antibody could not eliminate the complex formed by the mutant HNF-3α deleted in the C terminus (lane 5 vs. 6). In addition, a mutant probe (mPB−127/−102 bp) containing a mutated HNF-3α motif also affected the specific binding (lane 2 vs. 4). Similar results were observed when using R1 (PB−257/−232 bp) sequence. As shown in Fig. 2C, HNF-3α/R1 interaction is relatively weaker in contrast to HNF-3α/R2 (Fig. 2C, lane 2 vs. 4), because R1 has a 1-bp mismatch with HNF-3 cognate binding site, whereas R2 is a perfect match. The presence of multiple HNF-3 sites with different affinities has been proposed to be relevant for transcriptional modulation (44). These in vitro experiments confirmed the identification of two HNF-3α _cis_-regulatory elements adjacent to ARBSs in the PB proximal promoter.

In Vitro Synthesized HNF-3α Binds R1 and R2 A, Wild-type HNF-3α and three mutant proteins (ΔN, FH, and ΔC) were synthesized by in vitro TNT, and incubated with radiolabeled PB−127/−102 probe. In contrast to a nonspecific band (arrowhead) formed with the TNT blank control (lane 2), wild- type HNF-3α and HNF-3αΔC formed a strong complex (lanes 3 and 6), whereas ΔN and FH (arrowhead) showed weak binding ability (lanes 4 and 5, overexposure). B, The binding of HNF-3α with PB−127/−102 was eliminated by an antibody raised against the HNF-3α C terminus (lane 3 vs. 2). The same antibody did not affect the complex formed by HNF-3αΔC, which has a deletion in the C terminus (lane 6 vs. 5). The HNF-3α binding was also affected by mutating the TRTTTGY motif (lane 4 vs. 2). C, Similar results but weaker binding affinity was observed when PB−257/−232 was used as a probe (lane 4 vs. 2).

Fig. 2.

In Vitro Synthesized HNF-3α Binds R1 and R2 A, Wild-type HNF-3α and three mutant proteins (ΔN, FH, and ΔC) were synthesized by in vitro TNT, and incubated with radiolabeled PB−127/−102 probe. In contrast to a nonspecific band (arrowhead) formed with the TNT blank control (lane 2), wild- type HNF-3α and HNF-3αΔC formed a strong complex (lanes 3 and 6), whereas ΔN and FH (arrowhead) showed weak binding ability (lanes 4 and 5, overexposure). B, The binding of HNF-3α with PB−127/−102 was eliminated by an antibody raised against the HNF-3α C terminus (lane 3 vs. 2). The same antibody did not affect the complex formed by HNF-3αΔC, which has a deletion in the C terminus (lane 6 vs. 5). The HNF-3α binding was also affected by mutating the TRTTTGY motif (lane 4 vs. 2). C, Similar results but weaker binding affinity was observed when PB−257/−232 was used as a probe (lane 4 vs. 2).

HNF-3α Is Expressed in Prostate Epithelial Cells

Two previous studies have reported the mRNA expression profile of HNF-3 in the mouse and rat prostates by using in situ hybridization and Northern blot (28, 29). We examined the HNF-3 protein expression pattern in the human and mouse prostate by immunohistochemical and Western blot analyses. In Fig. 3, immunohistochemistry showed a clear nuclear staining of HNF-3α in both mouse (panels A and B) and human (panels C and D) prostate luminal epithelial cells. HNF-3β and HNF-3γ were not detected (data not shown). Consistently, Western blot showed that HNF-3α is expressed in all mouse prostate lobes, with highest levels of expression in the ventral prostate (VP). This pattern is consistent with a previous study that showed that HNF-3α mRNA levels in the VP are 14 times more abundant than in the liver (28). Results from RT-PCR using primers specific for HNF-3α, HNF-3β, or HNF-3γ are consistent with immunohistochemistry and Western blot (data not shown). The absence of HNF-3β expression in prostate has been reported before (29), and this is different from the expression patterns in other organs where HNF-3α and HNF-3β are often coexpressed (42).

HNF-3α Protein Is Expressed in Mouse and Human Prostate A–D, Immunohistochemistry. Nuclear staining of HNF-3α protein was restrictively detected in both mouse (A and B) and human luminal epithelial cells (C and D). E, Western blot. Fresh mouse prostate tissue lysates from different lobes (VP, ventral prostate; DLP, dorsal-lateral prostate; AP, anterior prostate; NP, whole normal prostate) were separated by SDS-PAGE and detected with HNF-3α (C20), HNF-3β (P19), and HNF-3γ (N19) antibodies.

Fig. 3.

HNF-3α Protein Is Expressed in Mouse and Human Prostate A–D, Immunohistochemistry. Nuclear staining of HNF-3α protein was restrictively detected in both mouse (A and B) and human luminal epithelial cells (C and D). E, Western blot. Fresh mouse prostate tissue lysates from different lobes (VP, ventral prostate; DLP, dorsal-lateral prostate; AP, anterior prostate; NP, whole normal prostate) were separated by SDS-PAGE and detected with HNF-3α (C20), HNF-3β (P19), and HNF-3γ (N19) antibodies.

HNF-3α Binding Sites Are Essential for Maximum Androgenic Induction

The mechanism of HNF-3α transcriptional regulation, in some part, involves nucleosome disruption (24, 45). Although transiently transfected DNA cannot assemble the same higher-order chromatin structure as genomic DNA, certain levels of nucleosome-mediated regulation can be observed on transiently introduced DNA (46, 47). Studies using transient transfection have demonstrated that HNF-3α relieves nucleosome-mediated transcriptional repression of a liver specific α-Fetoprotein gene (47). To examine the biological role of the two HNF-3α binding motifs, we compared the activities of mutant PB promoters with wild-type promoter in transfection assays. PCR-based site-directed mutagenesis was used to generate point mutations in HNF-3α binding sites (R1 and R2). As seen in Fig. 4A, three mutant promoters were generated. Two of them, M1 and M2, contain mutant HNF-3 motif in R2. The third one, M3, contains mutant HNF-3 motif in R1. EMSA in Fig. 4B demonstrated that M1 completely abolished HNF-3α complexes a, b, and c. M2 slightly affected complexes b and c, but distinctly removed complex a. Addition of HNF-3α antibody further removed complex b and c, indicating that M2 affects the formation of a higher-order complex, which was competed by the HNF-3 consensus sequence (Fig. 1B, lanes 18 and 19). All mutant, as well as a wild-type PB reporter, constructs were transfected into LNCaP cells and luciferase activities were compared. Experiments were repeated at least six times. Figure 4C is a representative experiment showing that the maximal androgen induction of PB promoter activity was significantly affected in either mutant R1 or mutant R2. In contrast to R1 motif, R2 may have a greater biological relevance because both M1 and M2 mutations almost completely abolished PB activity (P < 0.001). The significant decrease in M2 activity suggests that complex a (Fig. 4B), which is in a higher order, is biologically important. The reduction in M3 activity is consistent with a previous study (48) conducted by another group, who demonstrated a significant loss in PB activity by deleting a region overlapping with R1. Similar results were obtained when these transfection experiments were performed in PC3 cells (data not shown). In addition, EMSA in Fig. 4D demonstrated in vitro that a purified GST-AR-DBD fusion protein bound to these mutant ARBS-1/R1 or ARBS-2/R2 strings (Table 1 for sequences) with similar intensity as to the wild-type sequence, indicating that these mutations generated in HNF-3 motifs do not directly abrogate the AR/ARBS interaction. Thus, the activity loss in PB promoter (Fig. 4C) is not directly due to the loss of AR/ARBS binding. However, such mutations may destabilize the formation of a complete nucleoprotein complex in these regions, and this will be further discussed in this paper. These data strongly suggested that two HNF-3 motifs are required for the maximal androgenic induction in PB gene.

Two HNF-3α Motifs Are Essential for Maximum Androgenic Induction in PB A, Wild-type PB promoter sequences containing boxed HNF-3α motifs were mutated (underlined lowercase characters) as indicated in M1–M3. B, EMSA was conducted using 10 μg LNCaP cell extracts incubated with either wild-type PB−127/−102 or mutant R2 probes (lanes 1–3). M1 disrupted the binding of three complexes (lane 2, a, b, and c), whereas only the higher-order complex a was eliminated by M2 (lane 3). Addition of HNF-3α antibody further removed complexes b and c (lane 4). C, LNCaP cells were transiently transfected with luciferase reporter constructs (0.2 μg/well) containing either wild-type or mutant (M1–M3) PB promoters. Each well also received 0.0125 μg of Renilla vector. Cells were treated with or without 10−8m DHT for 24 h before harvest. Background activities of cell lysates with no DNA transfection were subtracted from the data obtained in the experimental group, before the normalization with Renilla activities. Results are presented as relative luciferase activity. Data shown here are a representative from at least six independent experiments in triplicate. Error bars indicate sd values. Asterisk indicates where P < 0.01 as compared with the androgen-induced activity in wild-type (WT) reporter. D, Probes (ARBS2/R2-M1, -M2, and ARBS1/R1-M3, Table 1) containing mutant HNF-3α motifs were used in EMSA to determine their interactions with a purified GST-AR-DBD protein described in Materials and Methods.

Fig. 4.

Two HNF-3α Motifs Are Essential for Maximum Androgenic Induction in PB A, Wild-type PB promoter sequences containing boxed HNF-3α motifs were mutated (underlined lowercase characters) as indicated in M1–M3. B, EMSA was conducted using 10 μg LNCaP cell extracts incubated with either wild-type PB−127/−102 or mutant R2 probes (lanes 1–3). M1 disrupted the binding of three complexes (lane 2, a, b, and c), whereas only the higher-order complex a was eliminated by M2 (lane 3). Addition of HNF-3α antibody further removed complexes b and c (lane 4). C, LNCaP cells were transiently transfected with luciferase reporter constructs (0.2 μg/well) containing either wild-type or mutant (M1–M3) PB promoters. Each well also received 0.0125 μg of Renilla vector. Cells were treated with or without 10−8m DHT for 24 h before harvest. Background activities of cell lysates with no DNA transfection were subtracted from the data obtained in the experimental group, before the normalization with Renilla activities. Results are presented as relative luciferase activity. Data shown here are a representative from at least six independent experiments in triplicate. Error bars indicate sd values. Asterisk indicates where P < 0.01 as compared with the androgen-induced activity in wild-type (WT) reporter. D, Probes (ARBS2/R2-M1, -M2, and ARBS1/R1-M3, Table 1) containing mutant HNF-3α motifs were used in EMSA to determine their interactions with a purified GST-AR-DBD protein described in Materials and Methods.

Overexpressing a Mutant HNF-3α Inhibits PB Activity

It has been suggested that both the N- and C-terminal regions of HNF-3 proteins have transactivation activities (49, 50). Recent studies using in vitro nucleosome assembly showed that the transcriptional modulation by HNF-3α is mediated through its C-terminal region (24). We generated two mutant recombinant HNF-3α proteins (ΔN141−466, ΔC1−294) with deletions in either the N- or C-terminal regions. These mutant HNF-3α proteins, as well as a wild-type HNF-3α expression vector (0.2 μg/well), were cotransfected into LNCaP cells with the ARR2PB-Luc reporter construct. Luciferase activities were measured and compared. As shown in Fig. 5A, overexpression of wild-type HNF-3α did not significantly increase the androgen induction of PB activity, indicating that the endogenous HNF-3α level is sufficient for the maximal promoter response. In contrast to wild-type and ΔN141−466 proteins, overexpressing ΔC1−294 significantly inhibited the total PB activity (Fig. 5A, P < 0.01), and this inhibition was dose responsive (Fig. 5B). In contrast, transfection of same amount of ΔC1−294 did not significantly inhibit the activity of the mutant PB promoter (Fig. 5C), which contains a nonfunctional HNF-3 site (M1 in Fig. 4C). Finally, Western blot showed that transient expression of these HNF-3α proteins in LNCaP cells did not result in a detectable change in the endogenous AR protein levels (Fig. 5D), suggesting that the reduction in PB promoter activity was not due to reduced AR levels. These results strongly indicated a dominant negative mechanism for ΔC1−294, and further supported the regulatory role of HNF-3α C-terminal region in transcriptional modulation.

Overexpressing a Mutant HNF-3α Inhibits PB Activity A, An empty vector as well as expression vectors corresponding to wild-type HNF-3α, ΔN141−466, or ΔC1−294 (0.2 μg per well) were cotransfected with a PB luciferase reporter into LNCaP cells. Cells were treated with or without 10−8m R1881 for 24 h followed by luciferase assays. B, Different amounts of ΔC1−294 (0.05 μg, 0.15 μg, or 0.25 μg per well) were cotransfected into LNCaP cells with PB reporter. Luciferase activities were determined after 24 h incubation in the presence or absence of androgen. C, In contrast to panel B, ΔC1−294 (0.05 μg, 0.15 μg, or 0.25 μg per well) was cotransfected into LNCaP cells with a mutant PB reporter (M1, in Fig. 4). Error bars indicate sd values. Asterisk indicates P < 0.01 in comparison with androgen-induced activities in control wells, which were transfected with empty vector. D, No significant change in AR protein level was detected by Western blot in cells transiently transfected with wild-type HNF-3α, ΔN141−466, or ΔC1−294.

Fig. 5.

Overexpressing a Mutant HNF-3α Inhibits PB Activity A, An empty vector as well as expression vectors corresponding to wild-type HNF-3α, ΔN141−466, or ΔC1−294 (0.2 μg per well) were cotransfected with a PB luciferase reporter into LNCaP cells. Cells were treated with or without 10−8m R1881 for 24 h followed by luciferase assays. B, Different amounts of ΔC1−294 (0.05 μg, 0.15 μg, or 0.25 μg per well) were cotransfected into LNCaP cells with PB reporter. Luciferase activities were determined after 24 h incubation in the presence or absence of androgen. C, In contrast to panel B, ΔC1−294 (0.05 μg, 0.15 μg, or 0.25 μg per well) was cotransfected into LNCaP cells with a mutant PB reporter (M1, in Fig. 4). Error bars indicate sd values. Asterisk indicates P < 0.01 in comparison with androgen-induced activities in control wells, which were transfected with empty vector. D, No significant change in AR protein level was detected by Western blot in cells transiently transfected with wild-type HNF-3α, ΔN141−466, or ΔC1−294.

Identification of Two Functional HNF-3α Binding Sites in PSA Core Enhancer

The results obtained for rat PB gene prompted us to examine whether HNF-3α is also required for the regulation of human PSA gene. It has been reported that the core enhancer region (−4.2/−3.8 kb) of the PSA gene, but not the proximal promoter, is essential and sufficient for androgen regulation and prostate specificity (5154). Six androgen response elements (AREs) are located in this region (55). Among these AREs, ARE III (at −4154/−4132 bp) shows the highest AR affinity and biological activity, because mutations in ARE III significantly abolished PSA enhancer activity (55). Interestingly, we identified two strong HNF-3 binding motifs in this enhancer region (Fig. 6A). The first site, designated as PSA1, is located at −4122/−4109 bp, immediately downstream of the ARE III. The second site, designated as PSA2, is located at −4028/−4005 bp, between ARE IIIA and ARE IIIB (Fig. 6A). In vitro synthesized HNF-3α bound to both elements (Fig. 6B, lane 2 vs. 3 and 6E, lane 3 vs. 4). Similar to the results for PB, mutant HNF-3α proteins (ΔN, ΔC, and FH) containing the FH domain also showed binding activity (Fig. 6B, lanes 4–6). In addition, the binding of HNF-3α was supershifted by HNF-3α antibody (lane 3 vs. 7). Therefore, both elements were confirmed to be authentic HNF-3α sites by these in vitro experiments.

Identification of HNF-3α Binding Motifs in the PSA Core Enhancer A, Two HNF-3α binding sequences (bold arrow) were identified in the PSA core enhancer (−4.2/−3.9 kb) region. The first element (−4122/−4109), designated as PSA1, is adjacent to ARE III (underlined). The second one (−4028/−4005), designated as PSA2, is located in the middle of ARE IIIA and ARE IIIB (underlined). B, EMSA. In vitro synthesized wild-type HNF-3α and mutant proteins (ΔN, FH, and ΔC) were able to bind PSA1 (lanes 3–6) as compared with the TNT blank control (lane 2). The HNF-3α binding was supershifted by the HNF-3α antibody (lane 3 vs. 7). C, Concomitant DNA binding of AR and HNF-3α. Radiolabeled oligonucleotide containing both ARE III and PSA1 was incubated with a constant amount (0.1 μg) of a purified GST AR-DBD protein. A slow migrating ternary complex (AR/HNF-3α/DNA) (lanes 5–8) was formed as addition of increasing amounts (1, 2, 3, and 4 μl) of in vitro synthesized HNF-3α, in contrast to HNF-3α alone (lane 3) or AR alone (lane 4), indicating that HNF-3α and AR can occupy DNA concomitantly. The ternary complex was disrupted by addition of HNF-3α antibody (lane 9). D, EMSA using LNCaP nuclear extract shows the formation of HNF-3α/PSA1 complexes (lane 2), which were disrupted by HNF-3α antibody (lane 3). The probe PSA1 (see Table 1 for sequence) contains binding sites for both AR and HNF-3α. E, In vitro synthesized wild-type HNF-3α also interacts with PSA2 (lane 4).

Fig. 6.

Identification of HNF-3α Binding Motifs in the PSA Core Enhancer A, Two HNF-3α binding sequences (bold arrow) were identified in the PSA core enhancer (−4.2/−3.9 kb) region. The first element (−4122/−4109), designated as PSA1, is adjacent to ARE III (underlined). The second one (−4028/−4005), designated as PSA2, is located in the middle of ARE IIIA and ARE IIIB (underlined). B, EMSA. In vitro synthesized wild-type HNF-3α and mutant proteins (ΔN, FH, and ΔC) were able to bind PSA1 (lanes 3–6) as compared with the TNT blank control (lane 2). The HNF-3α binding was supershifted by the HNF-3α antibody (lane 3 vs. 7). C, Concomitant DNA binding of AR and HNF-3α. Radiolabeled oligonucleotide containing both ARE III and PSA1 was incubated with a constant amount (0.1 μg) of a purified GST AR-DBD protein. A slow migrating ternary complex (AR/HNF-3α/DNA) (lanes 5–8) was formed as addition of increasing amounts (1, 2, 3, and 4 μl) of in vitro synthesized HNF-3α, in contrast to HNF-3α alone (lane 3) or AR alone (lane 4), indicating that HNF-3α and AR can occupy DNA concomitantly. The ternary complex was disrupted by addition of HNF-3α antibody (lane 9). D, EMSA using LNCaP nuclear extract shows the formation of HNF-3α/PSA1 complexes (lane 2), which were disrupted by HNF-3α antibody (lane 3). The probe PSA1 (see Table 1 for sequence) contains binding sites for both AR and HNF-3α. E, In vitro synthesized wild-type HNF-3α also interacts with PSA2 (lane 4).

Because PSA1 is closely adjacent to ARE III, it was intriguing to ask whether HNF-3α and AR can bind to DNA concomitantly. Thus, an oligonucleotide (Table 1) containing both ARE III and PSA1 was radiolabeled and incubated with constant amounts of a purified GST-AR-DBD fusion protein. A specific AR-DBD/DNA complex was formed (Fig. 6C, lane 4). As increasing amounts of HNF-3α proteins were added to the reaction, a specific HNF-3α/DNA complex as well as a slow-migrating band (AR/HNF-3α/DNA ternary complex) became stronger (lanes 5–8). Addition of HNF-3α antibody disrupted the complex (lane 9). Similarly, in EMSA using LNCaP nuclear extract, HNF-3α antibody disrupted a strong complex that formed with this oligonucleotide (Table 1, PSA1) containing both HNF-3α and AR binding sites (Fig. 6D, lane 2 vs. 3). These data indicate that AR and HNF-3α can bind to respective DNA binding sites concomitantly. Thus, the binding of one protein does not negatively affect the adjacent binding of another protein. However, the dislocation of HNF-3α from the target binding site (Fig. 6C, lane 9) may destabilize a higher-order protein complex associated with this entire regulatory region, which may include AR.

A similar mutation assay was performed to determine the biological relevance of these HNF-3α motifs in PSA enhancer. Figure 7A shows two mutant PSA reporter constructs, mPSA1-EP and mPSA2-EP, which were generated by PCR and transfected into LNCaP cells. Primers used for PCR are shown in Table 1. Luciferase activities were measured and compared with wild-type construct. Figure 7B shows that point mutations that abolish HNF-3α binding in either PSA1 (Fig. 6C, lane 2) or PSA2 (lane 4) significantly affected the maximal androgen-induced PSA activities (Fig. 7B, P < 0.01). A 95% reduction of activity in the PSA enhancer has been reported previously by deleting the region corresponding to PSA1 (53). Similarly, the direct interaction between AR and ARE III remained intact when mutant oligonucleotide (Table 1, mPSA1), containing both ARE III and mutant HNF-3 motif, was used in EMSA (Fig. 7D), indicating that the loss of PSA activity is not directly due to the loss of AR/ARE III binding. These results suggest that both HNF-3α motifs are essential for maximal PSA induction by androgen.

Mutations in HNF-3α Sites Inhibited Maximal Androgen Induction of PSA A, Two mutant PSA reporters (mPSA1-EP and mPSA2-EP) were generated by introducing point mutations to HNF-3α motifs in PSA1 and PSA2, individually. B, LNCaP cells were transiently transfected with wild-type and mutant PSA-EP luciferase constructs. Luciferase activities were determined after 24 h incubation in the presence or absence of 10−8m R1881. Three independent experiments were performed in triplicate. Background activity of untransfected cell lysate was subtracted from the results before normalization with Renilla activities. Error bars indicate sd values. Asterisk indicates where P < 0.01 in comparison with the androgen-induced activity in wild-type reporter. C, Oligonucleotides containing mutant HNF-3α sites (mPSA1 and mPSA2, Table 1) were used in EMSA to determine the HNF-3α binding. D, An oligonucleotide (mPSA1, Table 1 for sequence) containing both ARE III and a mutant HNF-3α motif shows similar binding intensity with a purified GST AR-DBD protein in EMSA.

Fig. 7.

Mutations in HNF-3α Sites Inhibited Maximal Androgen Induction of PSA A, Two mutant PSA reporters (mPSA1-EP and mPSA2-EP) were generated by introducing point mutations to HNF-3α motifs in PSA1 and PSA2, individually. B, LNCaP cells were transiently transfected with wild-type and mutant PSA-EP luciferase constructs. Luciferase activities were determined after 24 h incubation in the presence or absence of 10−8m R1881. Three independent experiments were performed in triplicate. Background activity of untransfected cell lysate was subtracted from the results before normalization with Renilla activities. Error bars indicate sd values. Asterisk indicates where P < 0.01 in comparison with the androgen-induced activity in wild-type reporter. C, Oligonucleotides containing mutant HNF-3α sites (mPSA1 and mPSA2, Table 1) were used in EMSA to determine the HNF-3α binding. D, An oligonucleotide (mPSA1, Table 1 for sequence) containing both ARE III and a mutant HNF-3α motif shows similar binding intensity with a purified GST AR-DBD protein in EMSA.

HNF-3α Binds PSA Enhancer in Vivo

Because LNCaP cells express endogenous PSA gene, ChIP was performed to investigate the in vivo association of HNF-3α with the PSA enhancer. Figure 8A is a schematic diagram of the PSA regulatory region. Five previously described DNA fragments (56) corresponding to the distal region, ARE III, middle region, ARE II, and ARE (2) were tested in the experiment. The ARE III region (−4170/−3978 bp) is a 192-bp fragment that covers both HNF-3α motifs we identified in this study (Fig. 8A). To determine the occupancy of HNF-3α on active vs. inactive PSA chromatin status, LNCaP cells were initially grown in RPMI medium 1640 supplemented with 5% charcoal/dextran-treated fetal bovine serum. After 3 d of cultivation, cells were either treated with 10−8m dihydrotestosterone (DHT) or maintained in androgen-depleted medium. Soluble chromatin was prepared after formaldehyde treatment of cells. Specific antibodies against HNF-3α or AR were used to immunoprecipitate antigen-bound genomic DNA fragments. The DNAs were amplified by PCR using specific primers (56) spanning the tested regions. After 48 h of DHT treatment, PSA expression was induced as compared with the undetectable level in untreated cells (Fig. 8B). In contrast, the expression levels of HNF-3α protein were almost identical in androgen-treated and untreated cells. In agreement with a previous study (56), DHT induced the recruitment of AR onto multiple AREs, but not control regions (Fig. 8D). In contrast to AR, HNF-3α constantly occupies the ARE III region independently of DHT treatment (Fig. 8D). These ChIP assays provided in vivo evidence for the association of HNF-3α with PSA enhancer and also indicated that the occupancy of HNF-3α alone does not result in the transactivation of PSA gene.

HNF-3α Occupies PSA Enhancer in Vivo A, Schematic diagram of PSA gene 5′-upstream region. Six DNA fragments corresponding to the distal, ARE III, middle, ARE II, and ARE (2 ) regions were amplified in ChIP assays using primers described previously (56 ). B, LNCaP cells were initially grown in RPMI 1640 with 5% charcoal/dextran-treated fetal bovine serum for 3 d. Cells were then incubated for another 48 h with or without 10−8m DHT treatment. Western blot was performed to determine the expression levels of PSA and HNF-3α. C, Formaldehyde-cross-linked chromatins were sheared by sonication into an average size of 0.5–1.0 kb. D, After an overnight IP with AR or HNF-3α antibodies, formaldehyde cross-linking was reversed, and DNA fragments were extracted, followed by PCR amplification (Materials and Methods).

Fig. 8.

HNF-3α Occupies PSA Enhancer in Vivo A, Schematic diagram of PSA gene 5′-upstream region. Six DNA fragments corresponding to the distal, ARE III, middle, ARE II, and ARE (2 ) regions were amplified in ChIP assays using primers described previously (56 ). B, LNCaP cells were initially grown in RPMI 1640 with 5% charcoal/dextran-treated fetal bovine serum for 3 d. Cells were then incubated for another 48 h with or without 10−8m DHT treatment. Western blot was performed to determine the expression levels of PSA and HNF-3α. C, Formaldehyde-cross-linked chromatins were sheared by sonication into an average size of 0.5–1.0 kb. D, After an overnight IP with AR or HNF-3α antibodies, formaldehyde cross-linking was reversed, and DNA fragments were extracted, followed by PCR amplification (Materials and Methods).

HNF-3α Interacts with AR

Steroid receptors play a pivotal role in transcriptional control of numerous genes. In addition to functioning through binding specific steroid response DNA elements, steroid receptors interact with many coregulators as well as other DNA-binding transcription factors. It has been reported that glucocorticoid receptor (GR)-mediated activation of multiple liver-specific genes requires HNF-3β (5762). Recent studies have shown that estrogen receptor (ER) interacts with three FH transcription factors, FKHR, FKHRL1, and AFX (63, 64). AR is a member of the steroid receptor family and shares many similarities with GR and ER in terms of structure and mechanism of function. The observation of close proximity of AR and HNF-3α binding sites prompted us to examine whether these two proteins can interact with each other. For this purpose, IP was performed in an AR-Hela cell line (55), in which a flag-tagged full-length AR was stably integrated. Because Hela cells do not express HNF-3α (Fig. 1C), we transfected an HNF-3α mammalian expression vector (pcDNA3.1D/V5-HNF-3α) and a control LacZ expression vector (pcDNA3.1D/V5-LacZ) into these cells. Western blot using anti-V5 confirmed the expression of V5-tagged LacZ and HNF-3α proteins in transfected cells (Fig. 9A, lanes 3 and 4 vs. 1 and 2). Cells were grown in medium with 10−8m DHT. All cell lysates were immunoprecipitated with an anti-flag M2 affinity gel in the presence of ethidium bromide (EB, 0–100 μg/ml), which was used to disrupt DNA-protein interactions (65). The gel was washed before Western blot analysis. As seen in Fig. 9B, flag-AR was detected in all AR-Hela precipitates (lanes 6–10) in contrast to normal Hela precipitate (lane 5). HNF-3α (lanes 14–16) but not LacZ (lane 13) was detected in the AR-Hela precipitates, indicating that flag-AR specifically immunoprecipitates with HNF-3α. It is significant that this interaction was resistant to the presence of EB (lanes 15 and 16), ruling out the possibility that it was mediated through DNA. Next, IP was performed in LNCaP cells to examine this interaction in a physical context in which AR and HNF-3α proteins express at endogenous levels. LNCaP cells were either grown in androgen-depleted medium for at least 3 d or grown in the presence of 10−8m DHT. Cell lysates (1 mg per IP reaction) were immunoprecipitated with anti-AR- conjugated protein G-Sepharose beads in the presence of EB. Similar IPs were performed using HNF-3α antibody instead of anti-AR. Each reaction was performed in the presence of 1% NP-40 and 1 mg BSA to quench nonspecific binding. Figure 9B unambiguously demonstrated a physical interaction of AR and HNF-3α in DHT-treated cells in contrast to the androgen- depleted condition. This interaction was resistant to the presence of EB at a concentration of 100 μg/ml, demonstrating a protein-protein interaction (Fig. 9B). These IP experiments strongly suggested that AR and HNF-3α might be involved in the assembly of a multinucleoprotein complex under a physical context.

HNF-3α Directly Interacts with AR A, AR-Hela cells, with stably integrated flag-AR, were transfected with an HNF-3α expression vector (pcDNA3.1D/V5-HNF-3α) and a control LacZ expression vector (pcDNA3.1D/V5-LacZ). Western blot using anti-V5 antibody detected the V5-labeled LacZ (lane 3) and HNF-3α (lane 4) proteins in transfected cells, but not in nontransfected cells (lanes 1 and 2). All cell lysates were immunoprecipitated by an anti-flag M2 affinity gel in the presence of EB. The gel was washed before Western blot analysis. Anti-flag detected flag-AR in all AR-Hela cell precipitates but not in normal Hela cells (lanes 5 vs. 6–10). Anti-V5 detected HNF-3α (lanes 14–16) but not LacZ (lane 13) in AR-Hela precipitates. The AR/HNF-3α interaction was not affected in the presence of 25 μg/ml (lane 15) and 100 μg/ml (lane 16) EB. B, LNCaP cell lysates (1 mg per IP) from DHT-treated or untreated cells were immunoprecipitated with HNF-3α or AR antibody in the presence of EB (0–100 μg/ml) as indicated. Western blot was performed using individual antibodies as indicated. C, A schematic diagram showing a series of AR and HNF-3α subdomains used in in vitro GST pull-down assays. The AR DBD/hinge region and the HNF-3α FH domain are highlighted in black. Two conserved HNF-3αC-terminal domains designated as region 2 and 3 are in striped boxes. D, Five purified GST-AR fusion proteins (20 μg for each) were bound to glutathione agarose beads, followed by incubation with V5-labled HNF-3α protein synthesized in vitro. Western blot was performed to determine the HNF-3α-interacting domain in AR. E, The GST AR fusion proteins DBD/Hinge (50 ng), NT/DBD (250 ng), and DBD/LBD (150 ng) were used in EMSA to determine their DNA binding activities with ARBS-1 on a molar basis. F, The expression of eight V5-labeled HNF-3α subdomains as well as a LacZ protein was confirmed in Western blot (lanes 1–9). These proteins were in vitro synthesized and incubated with the GST-bound AR NT/DBD, DBD/Hinge, and DBD/LBD to determine the AR-interacting region in HNF-3α. G, The interaction of AR-DBD/Hinge and HNF-3α FH domain was resistant to the presence of 25–150 μg/ml of EB. LBD, Ligand-binding domain; NT, N-terminal domain.

HNF-3α Directly Interacts with AR A, AR-Hela cells, with stably integrated flag-AR, were transfected with an HNF-3α expression vector (pcDNA3.1D/V5-HNF-3α) and a control LacZ expression vector (pcDNA3.1D/V5-LacZ). Western blot using anti-V5 antibody detected the V5-labeled LacZ (lane 3) and HNF-3α (lane 4) proteins in transfected cells, but not in nontransfected cells (lanes 1 and 2). All cell lysates were immunoprecipitated by an anti-flag M2 affinity gel in the presence of EB. The gel was washed before Western blot analysis. Anti-flag detected flag-AR in all AR-Hela cell precipitates but not in normal Hela cells (lanes 5 vs. 6–10). Anti-V5 detected HNF-3α (lanes 14–16) but not LacZ (lane 13) in AR-Hela precipitates. The AR/HNF-3α interaction was not affected in the presence of 25 μg/ml (lane 15) and 100 μg/ml (lane 16) EB. B, LNCaP cell lysates (1 mg per IP) from DHT-treated or untreated cells were immunoprecipitated with HNF-3α or AR antibody in the presence of EB (0–100 μg/ml) as indicated. Western blot was performed using individual antibodies as indicated. C, A schematic diagram showing a series of AR and HNF-3α subdomains used in in vitro GST pull-down assays. The AR DBD/hinge region and the HNF-3α FH domain are highlighted in black. Two conserved HNF-3αC-terminal domains designated as region 2 and 3 are in striped boxes. D, Five purified GST-AR fusion proteins (20 μg for each) were bound to glutathione agarose beads, followed by incubation with V5-labled HNF-3α protein synthesized in vitro. Western blot was performed to determine the HNF-3α-interacting domain in AR. E, The GST AR fusion proteins DBD/Hinge (50 ng), NT/DBD (250 ng), and DBD/LBD (150 ng) were used in EMSA to determine their DNA binding activities with ARBS-1 on a molar basis. F, The expression of eight V5-labeled HNF-3α subdomains as well as a LacZ protein was confirmed in Western blot (lanes 1–9). These proteins were in vitro synthesized and incubated with the GST-bound AR NT/DBD, DBD/Hinge, and DBD/LBD to determine the AR-interacting region in HNF-3α. G, The interaction of AR-DBD/Hinge and HNF-3α FH domain was resistant to the presence of 25–150 μg/ml of EB. LBD, Ligand-binding domain; NT, N-terminal domain.

Fig. 9.

HNF-3α Directly Interacts with AR A, AR-Hela cells, with stably integrated flag-AR, were transfected with an HNF-3α expression vector (pcDNA3.1D/V5-HNF-3α) and a control LacZ expression vector (pcDNA3.1D/V5-LacZ). Western blot using anti-V5 antibody detected the V5-labeled LacZ (lane 3) and HNF-3α (lane 4) proteins in transfected cells, but not in nontransfected cells (lanes 1 and 2). All cell lysates were immunoprecipitated by an anti-flag M2 affinity gel in the presence of EB. The gel was washed before Western blot analysis. Anti-flag detected flag-AR in all AR-Hela cell precipitates but not in normal Hela cells (lanes 5 vs. 6–10). Anti-V5 detected HNF-3α (lanes 14–16) but not LacZ (lane 13) in AR-Hela precipitates. The AR/HNF-3α interaction was not affected in the presence of 25 μg/ml (lane 15) and 100 μg/ml (lane 16) EB. B, LNCaP cell lysates (1 mg per IP) from DHT-treated or untreated cells were immunoprecipitated with HNF-3α or AR antibody in the presence of EB (0–100 μg/ml) as indicated. Western blot was performed using individual antibodies as indicated. C, A schematic diagram showing a series of AR and HNF-3α subdomains used in in vitro GST pull-down assays. The AR DBD/hinge region and the HNF-3α FH domain are highlighted in black. Two conserved HNF-3αC-terminal domains designated as region 2 and 3 are in striped boxes. D, Five purified GST-AR fusion proteins (20 μg for each) were bound to glutathione agarose beads, followed by incubation with V5-labled HNF-3α protein synthesized in vitro. Western blot was performed to determine the HNF-3α-interacting domain in AR. E, The GST AR fusion proteins DBD/Hinge (50 ng), NT/DBD (250 ng), and DBD/LBD (150 ng) were used in EMSA to determine their DNA binding activities with ARBS-1 on a molar basis. F, The expression of eight V5-labeled HNF-3α subdomains as well as a LacZ protein was confirmed in Western blot (lanes 1–9). These proteins were in vitro synthesized and incubated with the GST-bound AR NT/DBD, DBD/Hinge, and DBD/LBD to determine the AR-interacting region in HNF-3α. G, The interaction of AR-DBD/Hinge and HNF-3α FH domain was resistant to the presence of 25–150 μg/ml of EB. LBD, Ligand-binding domain; NT, N-terminal domain.

Responsible Domains for AR/HNF-3α Interaction

An in vitro GST pull-down assay was performed to confirm AR/HNF-3α interaction as well as to determine the interacting regions. A full-length HNF-3α protein labeled with a C-terminal V5 epitope was synthesized in vitro. Five GST-AR fusion proteins containing different AR subdomains (Fig. 9C) were purified as described previously (66). Figure 9D is a GST pull-down experiment showing that the AR DBD/hinge region (524–649 amino acids) alone is sufficient to mediate the interaction with HNF-3α. Experiments were repeated at least five times. In our experiments, the ARNT/DBD showed a weaker interaction with HNF-3α as compared with ARDBD/Hinge or ARDBD/LBD (Fig. 9D), suggesting that the AR N terminus might have a negative effect on the interaction. Although the biological relevance of this effect is currently unknown, this effect was obviously not due to the diminished activity of ARNT/DBD protein, because it bound to the ARBS-1 sequence (on PB promoter) to a similar extent as ARDBD/Hinge or ARDBD/LBD on a molar basis (Fig. 9E). In Fig. 9, C and F, in vitro synthesized HNF-3α fragments were used to map the AR-interacting region(s) in the HNF-3α protein. Most interestingly, the FH domain (amino acids 141–294) alone was sufficient to mediate a strong interaction with AR (Fig. 9F, lanes 13, 22, and 31), whereas the N- or C-terminal domain alone did not bind with AR. Figure 9G demonstrated that the interaction between ARDBD/Hinge and HNF-3α FH domain was insensitive to the presence of EB (0–150 μg/ml), supporting a direct protein-protein interaction. The ligand effect on this interaction was also examined (data not shown) and the results suggested that, under these in vitro conditions, the interaction between ARDBD/LBD and HNF-3α could occur without ligand. This was not surprising because in vitro purified GST-ARDBD/LBD fragments may not act identically as in vivo wild-type AR, in terms of protein folding. Although these in vitro binding experiments cannot exactly reflect the physical conditions, they identified regions responsible for AR/HNF-3α interaction.

DISCUSSION

In this study, we report that an extracellular signal mediator, AR, and a tissue-restricted FH protein, HNF-3α, coordinately participate in prostatic gene regulation. HNF-3/FH proteins are a group of transcription factors that share remarkable sequence similarity over their DBD, termed a FH or winged-helix domain (19, 20, 67, 68). The HNF-3/FH proteins are involved in the differentiation of endoderm-derived tissues. This feature has been strikingly conserved among the metazoans (19). The Drosophila homeotic protein, FH, which is equivalent to HNF-3, was found to be essential for the development of the terminal region including the foregut and hindgut (67); the PHA-4 protein, which is a Caenorhabditis elegans HNF-3 equivalent protein, is also required for the formation of terminal gut structures in the worm (44). In mammals, HNF-3 proteins (HNF-3α, HNF-3β, and HNF-3γ) were originally discovered as liver-enriched factors, because of their ability to bind to the TTR gene promoter (27, 42). In early embryo development, HNF-3 proteins are believed to provide developmental competence to numerous liver-specific genes through their high-affinity DNA elements. This mechanism is essential for the activation of these target genes in later stages (19, 26). The role that HNF-3 and some other developmental factors play in organ differentiation has been proposed as genetic potentiation (19, 26, 69).

We observed strong nuclear staining of HNF-3α in both human and mouse prostate epithelial cells (Fig. 3, C–F). The prostate epithelium, like the epithelium of other urogenital sinus derivatives (bladder and urethra), is derived from the endodermal layer of the embryo. In contrast, Wolffian duct derivatives (epididymis, ductus, deferens, and seminal vesicle) are derived from the mesoderm (17, 70). Understanding the role of HNF-3α in the prostate may provide an insight into the differences in epithelial patterning and developmental programs among prostate and Wolffian-derived organs, which are all androgen dependent (3, 70). The cell type-dependent expression of HNF-3α and other endodermal factors could easily provide an explanation why prostatic differentiation markers such as PSA can be instructively induced in bladder epithelial cells under proper extracellular signals (16), whereas such differentiation, under the same signals, cannot occur in a mesoderm-derived context (3).

Using in vitro gel shift and mutation assays, functional HNF-3α sites were identified in both PSA and PB gene-regulatory regions (Figs. 1, 2, and 6). ChIP assay unambiguously demonstrated that HNF-3α binds to PSA enhancer in vivo (Fig. 8). In our experiments, HNF-3α binding sites were further identified in other prostate-specific gene-regulatory regions. As shown in Fig. 10A, the proximal promoter of the rat prostatic acid phosphatase gene (rPAP) contains an HNF-3α binding sequence at −113/−103 bp. This sequence, as well as an upstream ARE, is well conserved in its human homolog hPAP gene promoter (71). In the hPAP upstream enhancer (−1258/−779 bp), a G/T-rich region (−1183/−1151 bp) contains two HNF-3α binding sites with a putative ARE in the middle. This region also exists in the rPAP enhancer (Fig. 10A), except that only one HNF-3α binding site was found. Importantly, the hPAP enhancer (−1258/−779 bp) is related to the cell-specific expression of hPAP gene (48). In addition, two HNF-3α motifs (at −184/−173 bp and −141/126 bp, Fig. 10A) were found in the rat prostatic steroid-binding protein C1 (PBP C1) gene promoter (72). We confirmed all of these HNF-3α sites experimentally by EMSA (data not shown). The sequences used in EMSA are shown in Table 1. Figure 10B is a schematic diagram showing a striking similarity in the organization of binding motifs for HNF-3α and AR in these highly prostate-specific genes, which suggests that a common functional mechanism may apply.

Similar Organization of HNF-3 and AR Binding Motifs in Prostate Gene-Regulatory Regions A, A summary of HNF-3α binding motifs identified in this study. B, Schematic diagram showing remarkable similarity in the organization of AR and HNF-3α binding motifs in different prostate gene regulatory regions.

Fig. 10.

Similar Organization of HNF-3 and AR Binding Motifs in Prostate Gene-Regulatory Regions A, A summary of HNF-3α binding motifs identified in this study. B, Schematic diagram showing remarkable similarity in the organization of AR and HNF-3α binding motifs in different prostate gene regulatory regions.

To develop a complex eukaryote requires differential expression of more than 30,000 genes/proteins in precise spatial and temporal patterns (73). An organism uses transcriptional synergy to achieve such diversity, to maintain cell specificity, and to respond to the environment dynamically (73). A combinatory use of a subgroup of general transcription factors, signal-dependent factors, and cell-specific factors could lead to large number of regulatory possibilities, ensuring that every gene is precisely controlled (74). In the prostate, AR is a signal-dependent nuclear receptor mediating extracellular signals, whereas HNF-3α is a cell type-limited genetic potentiator (19). A synergy between these classes of factors has been suggested in other studies (74), and HNF-3α plus AR may be involved in prostatic differentiation. In an in vitro DNA footprinting study, a prostate-specific protein was reported to bind a DNA sequence in PB promoter (48). Interestingly, this region overlaps the R1 site we reported in Fig. 1A. Similarly, previous gel shift experiments have discovered several cell-specific factor-binding elements in the PSA core enhancer (51). Among these elements, two of them completely cover the two HNF-3α motifs shown in Fig. 6A (PSA1 and PSA2), whereas others overlap with binding sites for GATA proteins (75), which belong to another endodermal transcription factor family (19). Consistently, the PSA1 site in Fig. 6A, which is adjacent to ARE III, was protected by a cell-specific protein in a previous DNA footprinting study (53). Interestingly, the PB promoter (−286/+28 bp), the PSA enhancer (−4.2/−3.8 kb), and the hPAP enhancer (−1278/−779 bp), where HNF-3α binding sites are located, have all been implicated in prostate-specific regulation (36, 51, 54, 76). These observations strongly suggest that HNF-3α and AR may coordinately participate in the transcriptional regulation of other prostate epithelial cell-specific genes.

X-ray crystallography revealed that the structure of the HNF-3 FH domain resembles the globular domain of linker histone H1 (77). In vitro nucleosome assembly experiments further showed that HNF-3α is able to open compacted chromatin by displacing linker histone in an ATP-independent manner (24, 45). This mechanism of HNF-3α action is mediated through its high-affinity DNA-binding sites as well as the binding of its C terminus to histones H3 and H4 (24). We used transient transfection experiments to examine the role of HNF-3α in prostatic gene regulation. Although transiently transfected DNA does not appear to assemble the same higher-order chromatin structure observed with genomic DNA, certain aspects of nucleosome-mediated regulation can be observed on transiently expressed DNA (46, 47). This has been shown by the studies of Crowe et al. (47), who demonstrated that HNF-3α relieves nucleosome-mediated transcription repression of a liver-specific gene in transient transfection experiments. In our study, mutations of these HNF-3α binding motifs significantly abolished PB and PSA gene activities (Figs. 4 and 7). Overexpression of a mutant HNF-3α protein (ΔC1−294) suppressed PB promoter activity in a dose-responsive and binding site-dependent manner (Fig. 5). These results are consistent with the assumption that some level of nucleosome assembly occurs on the introduced plasmid. Also, these data are in agreement with previous studies showing that overproduction of mutant HNF-3 proteins deleted in a similar region suppressed multiple liver gene expressions (78, 79). However, in contrast to AR, which binds to ARE in a ligand-dependent manner, HNF-3α occupies the PSA enhancer in the absence of androgen (Fig. 8). The DNA binding of HNF-3α does not seem to be directly associated with PSA activation, which eventually requires the recruitment of ligand-bound AR onto the multiple AREs (Fig. 8). Thus, HNF-3α, by itself, is not a strong transactivator; however, the occupancy of HNF-3α in the regulatory region may confer certain transcriptional potential to target prostatic genes that is required for the later recruitment of additional factors as well as activation by a strong and rate-limiting activator (ligand-bound AR) during prostatic differentiation.

Transcriptional regulation depends not only on the interactions between DNA-binding proteins and their respective _cis_-regulatory elements but also on the interactions among these proteins and with other components of the transactivation machinery. In addition to interacting with various type I or type II coregulators, steroid receptors also make contact with other DNA binding proteins, resulting in modulation of transcriptional activity (80). In the present work, AR was found to directly interact with HNF-3α (Fig. 9), because such interaction was resistant to the presence of a DNA intercalator, EB (Fig. 9, A, B, and G), which serves as a good indicator of DNA-dependent and DNA-independent protein associations (65). In fact, AR has been found to interact with a number of DNA binding transcription factors including activator protein 1 (AP-1) (81), Sma- and mad-related protein 3 (82, 83), nuclear factor κB (NFκB) (84), sex-determining region Y (SRY) (85), prostate-derived ets factor (86), and other steroid receptors (80). The phenomenon of interactions of AR and these distinct transcription factors was called cross-modulation, which was illustrated by well documented AR interactions with AP-1 and NFκ B (81, 84). Such cross-talks between different signaling pathways may increase regulatory diversity and provide opportunities for cell-specific response. For example, a testis-expressed protein encoded by the SRY interacts with AR and plays a role in germ cell development (85); a newly discovered prostatic epithelium-specific Ets transcription factor PDEF directly contacts AR and activates PSA gene expression. Here, the adjacent binding of HNF-3α and a direct contact with AR may establish an efficient molecular mechanism for ligand-bound AR to rapidly target correct gene sequences. Crystallographic study of HNF-3γ/DNA revealed a bend of DNA of about 13° upon HNF-3γ binding (77), which may provide favorable DNA conformation for adjacent factors, because chromatin-associated proteins such as high-mobility group box-containing proteins have been well demonstrated for their ability to enhance DNA binding of steroid receptors by generating a sharp bend in DNA (87). A similar mechanism may apply to the current model. It has been shown dynamically that the physical binding of GR to a glucocorticoid response element (GRE) was facilitated by the adjacent HNF-3β binding (58). This appears to be true for several other liver-specific genes (57, 61). In addition, HNF-3 proteins cooperate with ER to activate vitellogenin B1 gene transcription (88). Thus, cooperation between steroid receptors and HNF-3 proteins may be a general mechanism for the control of various tissue-specific genes.

Strong evidence suggests that HNF-3α, like AP-1 (81), SRY (85), and PDEF (86), is another AR-DBD-interacting transcription factor (Fig. 9, D and F). Even though the DBDs of steroid receptors appear to be mainly involved in DNA binding and homodimerization of receptor monomers, mounting evidence suggests that this domain also serves as an interaction interface for other proteins. Among these interacting partners are coactivators (8992), factors of basal transcription machinery (93), and other transcription factors (81, 85, 86). Most of these interactions are biologically relevant and result in transcriptional activation or repression. In the present work, IP experiments in LNCaP cells demonstrated that the interaction of endogenous AR and HNF-3α occurs in the presence of androgen (Fig. 9B), which suggests the physical relevance of this interaction. In vitro DNA binding experiments showed that both proteins can adjacently and concomitantly bind respective _cis_-regulatory elements (Figs. 1D and 6, C and D); these results suggest that such AR/HNF-3α interaction does not inhibit the DNA binding of either protein and that these interaction events are mediated through separate motifs in AR-DBD. The identification of an AR/HNF-3α interaction may extend our understanding of the role of DBD in androgen action and provide a general insight into the regulatory mechanism between HNF-3 proteins and steroid receptors.

In summary, we report multiple HNF-3α _cis_-acting motifs in prostatic gene regulatory regions and provide evidence that HNF-3α is essential for maximal gene activation by AR. The interaction of HNF-3α and AR on closely positioned regulatory sequences may be a part of a complex that is important for prostate gene transactivation. The expression of HNF-3α in bladder (and urethra) epithelium (28, 29) and the emergence of AR expression in bladder epithelium in tissue recombination (with UGM) (16, 17) further support the involvement of these two factors in prostatic differentiation. We predict that the cooperation between AR and HNF-3α may be an extensive mechanism involving the establishment of the prostatic lineage and differentiated function.

MATERIALS AND METHODS

Cell Culture and Transfection Assays

The human prostate carcinoma cell lines DU145, LNCaP, and PC-3, the human cervical adenocarcinoma cell line Hela, the human hepatocellular carcinoma cell line HepG2, and the monkey kidney cell line COS-1 were obtained from American Type Culture Collection (Manassas, VA). All cell lines were cultured as recommended by American Type Culture Collection. The AR-Hela cells expressing stably integrated flag-tagged full-length AR were acquired from Dr. Michael Carey (UCLA School of Medicine, Los Angeles, CA) (55). Transient transfection assays were performed using lipofectin reagent (4 μl/well) for LNCaP cells and lipofectamine 2000 for other cell lines. The pRL-CMV containing the Renilla luciferase reporter gene (Promega Corp., Madison, WI) was used to optimize transfection efficiencies for each cell line. Optimal volumes of liposome and transfection durations were obtained and used to get the highest transfection efficiencies. Briefly, cells were seeded at an initial density of 8–10 × 104 per well in 24-well plates 1 d before transfection. The following morning, cells were transfected with plasmid DNA and lipofectin or lipofectamine 2000 in Opti-MEM I-reduced serum medium (31985; Life Technologies, Inc., Gaithersburg, MD). In transfection experiments where AR expression was required, 0.2 μg of rat AR expression vector was transfected for each well. The total amount of plasmid DNA was normalized to 0.8–1 μg/well by the addition of pVZ-1 plasmid. In addition, all samples received 12.5 ng/well of pRL-CMV reporter plasmid. After 4–6 h of transfection, the medium was replaced by MEM (11090; Life Technologies, Inc.) with 5% charcoal/dextran-treated fetal bovine serum (HyClone, Logan, UT) in the presence or absence of 10−8m R1881 or DHT. Cells were harvested and lysed with 80 μl passive lysis buffers after 24 h of incubation. The luciferase activity was determined using the dual luciferase reporter assay system (E1960; Promega Corp.) and LUMIstar (BMG Lab. Technologies, Inc., Durham, NC). Background activity of the cell lysate with no DNA transfection was subtracted from the activities obtained from the experimental group. All values were normalized by Renilla activity to correct for the transfection efficiency. Results are presented as relative luciferase activities. Each experiment was repeated at least three separate times in triplicate. In promoter mutagenesis experiments, mean normalized values from samples showing the highest inhibition of promoter activity were compared with those from samples transfected with the wild-type promoter by using Student’s paired t test.

Reporter Plasmids and Expression Vectors

ARR2PBLuc reporter contains an additional androgen- response region (−244 to −96 nt) fused upstream of the −286/+28-bp PB promoter to enhance the androgen response (36). A 621-bp fragment of PSA minimal promoter (−610 to +11 nt) was amplified by PCR and cloned at the _Sma_I and _Xho_I sites of pGL3-basic luciferase vector (Promega Corp.), after which an 823-bp upstream enhancer fragment (−4758 to −3935 bp) containing the −4.1/−3.9 kb PSA core enhancer region was obtained by PCR and inserted upstream of the PSA promoter at _Sac_I and _Sma_I sites, resulting in PSA-EPLuc reporter construct (EP stands for enhancer/promoter). For promoter mutagenesis studies, three mutant PB promoter fragments and two mutant PSA enhancer fragments were generated using PCR-based site-directed mutagenesis. All mutant and wild-type fragments were cloned into pGL3-basic vector. The primers used for PCR are listed in Table 1.

Mammalian HNF-3α expression vector pRB-HNF-3α (25) was kindly provided by Dr. Kenneth S. Zaret (Fox Chase Cancer Center, Philadelphia, PA). Full-length wild-type HNF-3α cDNA and eight cDNAs encoding differently truncated HNF-3α fragments were PCR amplified and directionally cloned into pcDNA3.1D/V5/His-TOPO expression vector (K4900; Invitrogen, San Diego, CA) in frame with the carboxy-terminal V5 epitope and 6×His tag. The resulting expression vectors were pHNF-3α-WT1−466, pHNF-3α-ΔNT141−466, pHNF-3α-FH141−294, pHNF-3α-ΔCT1−294, pHNF-3α-Δ231−385, pHNF-3α-Δ31−420, pHNF-3α-NT1−180, pHNF-3α-CT295−466, and pHNF-3α-DN59−345. The pcDNA3.1D/V5/His/lacZ is an expression control vector included in the pcDNA3.1 Directional TOPO Expression Kit. The rat AR expression vector (36) and GST-ARNT/DBD, GST-ARDBD, and GST-ARDBD/LBD vectors (66) have been described previously. GST-ARNT was amplified by PCR and cloned into pRC/CMV (Invitrogen) as a _Bam_HI fragment and subcloned into the _Bam_HI site of pGEX-3X vector (Pharmacia Biotech, Piscataway, NJ) in frame with the GST fusion. GST-ARLBD was cloned using pCMV/AR6 as the template (66), and the PCR product was digested with _Bam_HI and cloned into the _Bam_HI site of pGEX-3X vector. All reporter and expression constructs were confirmed by sequencing.

EMSAs

Nuclear extract for PC-3 cells was prepared as described previously. Nuclear extract for LNCaP cells was purchased from Geneka Biotechnology, Inc. (Montreal, Quebec, Canada). Recombinant wild-type and truncated HNF-3α proteins were synthesized in vitro using the TNT T7 Quick Coupled Transcription/Translation System (L1170; Promega Corp.). GST-AR fusion proteins were purified as described previously (66). All nuclear extracts and purified proteins were stored in buffers containing 1× concentration of complete protease inhibitor cocktail (Roche Molecular Biochemicals, Mannheim, Germany). All oligonucleotides for EMSAs were synthesized by Integrated DNA Technologies, Inc. (Coralville, IA). The probes were end labeled with T4 polynucleotide kinase (New England Biolabs, Beverly, MA), [γ-32P]ATP and purified by 15% PAGE. A typical binding reaction involved a 10-min preincubation with 10 μg of nuclear extract, 1 μg of the nonspecific competitor poly (dI-dC), and buffer D [20 mm HEPES-NaOH (pH 7.9); 100 mm KCl; 0.2 mm EDTA; 1.5 mm MgCl2; 1 mm dithiothreitol; 20% glycerol; and 1 mm phenylmethylsulfonyl fluoride (PMSF)], followed by a 15-min incubation with 200,000 cpm of radiolabeled probe in a total volume of 20 μl. In oligonucleotide competitions, 200- to 600-fold molar excess of cold, double-stranded oligonucleotide was added to the preincubation mix. In experiments where in vitro synthesized HNF-3α proteins were used, 1–5 μl of 50 μl products from the TNT reaction system were added to the preincubation mix. In supershift analyses, antibodies were added after the binding reaction and incubated for an additional 20 min on ice before electrophoresis. All supershift antibodies (AR, C-19: sc-815X; AR, N-20: sc-816X; HNF-3α, C-20: sc-6553X; HNF-3β, M-20: sc-6554X; HNF3-γ, N-19: sc-5361X; p-c-Jun: sc-822X; c-Fos: sc-447X; NF-1, H-300: sc-5567X; Oct-1: sc-8024X) were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA). The concentration of antibody in each EMSA reaction was 0.2 μg/μl. Complexes were resolved by electrophoresis for 2.5 h at 160 V on a 5% native polyacrylamide gel, which was later dried and processed for autoradiography.

Southwestern and Western Blot Analysis

An oligonucleotide containing two tandem copies of wild-type −124/−109 bp (2×R2: 5′-ACCTATTTGTATACTAACCTATTTGTATACTAGATGACA-3′), and another oligonucleotide containing two mutant sites (2×mR2: 5′-ACCTAgaaGTATACTAACCTAgaaGTATACTAGATGACA-3′) were radiolabeled by extension of an annealed 10-bp primer, 5′-TGTCATCTAG-3′. (Underscored letters represent the nucleotide replacements in the wild-type TATTTGTAT motif.) The primed oligonucleotide probes were radiolabeled with the Klenow fragment of Escherichia coli DNA polymerase, deoxynucleotide triphosphates, [α-32P]dATP, and [α-32P]dTTP. An oligonucleotide containing two copies of consensus ARBS (see Table 1) was radiolabeled in the same condition and used for a positive control. Nuclear extracts (30 μ g) of PC-3 and LNCaP cells were heated at 70 C for 10 min in 1× lithium dodecyl sulfate loading buffer (Invitrogen), resolved on 4% stacking, 12% resolving SDS-polyacrylamide gel, and electrophoretically transferred to nitrocellulose membranes (Bio-Rad, Hercules, CA). Membranes were soaked in PBS for 15 min, blocked for 4 h at room temperature with buffer A [20 mm HEPES-NaOH (pH 7.8), 50 mm NaCl, 12.5 mg/ml skim milk, 2.5 mg/ml BSA, 200 μ_g_/ml native salmon sperm DNA, 5 μ_g_/ml poly dI-dC, and 50 ng/ml single-stranded DNA], and incubated overnight at room temperature in 2.5 ml of buffer A plus ≥107 cpm of radiolabeled probe. Membranes were washed three times for 15 min each at room temperature in a washing buffer containing 20 mm HEPES-NaOH, 50 mm NaCl, 1 mg/ml of skim milk, and 0.025% Nonidet P-40. Membranes were dried completely at room temperature before autoradiography.

For Western blot analysis, cell pellets and freshly dissected mouse tissues were collected, sonicated, and centrifuged in cold RIPA buffer [1× PBS (pH 7.4), 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, 1 mm PMSF, and 1× concentration of complete protease inhibitor cocktail]. After the transfer to polyvinylidine difluoride membrane (Invitrogen), membranes were blocked in Tris-buffered saline containing 0.1% Tween 20 and 5% skim milk, and incubated with primary antibody (1:1000 dilution: anti-HNF-3α, anti-HNF-3β, anti-HNF-3γ, anti-AR, anti-PSA and anti-flag) for 1 h with shaking at room temperature. Anti-flag M2 monoclonal antibody (F3165) was purchased from Sigma (St. Louis, MO). In cases where anti-V5-horseradish peroxidase antibody (R96125; Invitrogen) was used to detect the recombinant V5-tagged HNF-3α proteins, a dilution of 1:5000 was used according to the protocol of the manufacturer. The signal was visualized by enhanced chemiluminescence assay (Amersham Pharmacia Biotech, Arlington Heights, IL).

Immunohistochemical Assays

The individual prostate lobes (anterior prostate, dorsal prostate, lateral prostate, and ventral prostate) were dissected from 10-wk-old CD-1 mice and fixed in 10% buffered formalin. Human prostate tissues were obtained from the Department of Pathology of Vanderbilt University Medical Center (Nashville, TN). After processing and embedding in paraffin, 5-μm sections were cut for immunohistochemical detection of HNF-3α proteins. Sections were deparaffinized, rehydrated, and placed in 1 m urea. Antigenic sites were exposed by microwaving the sections for 30 min at 95−99 C before the removal of endogenous peroxidase activity with DAKO peroxidase blocking reagent (DAKO Corp., Carpinteria, CA). Nonspecific binding was blocked by incubating the sections with Vectastain rabbit normal serum (Vectastain ABC Kit, PK-6105, Vector Laboratories, Inc., Burlingame, CA) for 30 min. The sections were incubated with anti-HNF-3α (C-20) antibody overnight at 4 C (1:1500 dilution), washed in PBS (pH 7.4), and incubated with Vectastain biotinylated antigoat IgG at room temperature for 30 min. The sections were washed with PBS and incubated for 30 min with Vectastain streptavidin. After additional washes with PBS, peroxidase activity was detected using 3′,3′-diaminobenzidine tetrahydrochlorate (Liquid DAB Substrate-Chromagen System, DAKO Corp.). The reaction was terminated in distilled water, and the sections were counterstained with Harris hematoxylin (Surgipath, Richmond, VA), dehydrated, and permanently mounted with cytoseal XYL (Stephens Scientific, Kalamazoo, MI).

ChIP

The procedure and PCR primers used in this study were described previously (56). LNCaP cells were initially grown in RPMI 1640 with 5% charcoal/dextran-treated fetal bovine serum (HyClone). After 3 d of cultivation, cells were either treated with 10−8m DHT or continued to grow in the androgen-depleted medium. After 48 h of treatment, cells were washed with PBS and cross-linked with 1% formaldehyde at 37 C for 10 min. Cells were scraped into conical tube, pelleted for 4 min at 2000 rpm at 4 C, resuspended in SDS lysis buffer [1% SDS, 10 mm EDTA, 50 mm Tris-HCl (pH 8.1), 1× proteinase inhibitor cocktail] for 200 μl per 106 cells, and sonicated with four to five sets of 10-sec pulses at an 80% maximum power (Fisher Sonic Dismembrator, model 50; Fisher Scientific, Pittsburgh, PA). After centrifugation for 10 min, supernatants were collected and diluted 1:10 in ChIP dilution buffer [0.01% SDS, 1.1% Triton X-100, 1.2 mm EDTA, 16.7 mm Tris-HCl (pH 8.1), 167 mm NaCl], followed by preclearing for 30 min with 3 μg of sonicated salmon sperm DNA with protein A agarose (80 μl of 50% slurry in 10 mm Tris-HCl, 1 mm EDTA). IP was performed overnight at 4 C with specific antibodies. Protein A agarose (60 μl) with salmon sperm DNA was added for 1 h with rotation to collect the complex. Beads were sequentially washed for 5 min each with low-salt wash buffer (0.1% SDS, 1% Triton X-100, 2 mm EDTA, 20 mm Tris-HCl, 150 mm NaCl), high-salt wash buffer (0.1% SDS, 1% Triton X-100, 2 mm EDTA, 20 mm Tris-HCl, 500 mm NaCl), LiCl wash buffer (0.25 m LiCl, 1% Nonidet P-40, 1% deoxycholate, 1 mm EDTA, 10 mm Tris-HCl) and twice with TE buffer (10 mm Tris-HCl, 1 mm EDTA). The complex was eluted twice with 250 μl of elution buffer (1% SDS, 0.1 m NaHCO3) and eluates were pooled. The formaldehyde cross-linking was reversed by adding 20 μl 5 m NaCl and incubating for 6–8 h at 65 C. Eluates were incubated for an additional 1 h at 45 C with 2 μl of 10 mg/ml Proteinase K. DNA was extracted using QIAquick Spin Column (Qiagen) and 3–5 μl of extracted DNA was used in each PCR amplification.

IP Analysis

DHT-treated or untreated LNCaP cells (0.5–1×107) were washed three times with cold PBS and lysed with 1 ml of nondenaturing lysis buffer (50 mm Tris, 150 mm NaCl, 10 mm EDTA, 0.02% NaN3, 50 mm NaF, 1 mm Na3VO4, 1% NP-40, 1 mm PMSF, 0.5 mm dithiothreitol, and 1× concentration of complete protease inhibitor cocktail). After sonication and centrifugation, 1 mg of total cell lysate for each reaction was incubated at 4 C for 3 h with 20 μl (dry volume) protein G-Sepharose beads (Amersham Biotech), which were conjugated with 1 μg experimental antibody or mock antibody. BSA (1 mg) was added to quench the nonspecific binding. IPs were performed in the presence of EB (0–100 μg/ml as indicated in Results) to disrupt DNA-protein interaction (65). Beads were washed four times with lysis buffer and once with PBS for 5 min each with rotation, which was followed by Western blot. The anti-AR (441) mouse monoclonal IgG (sc-7305; Santa Cruz Biotechnology, Inc.) was used for immunoprecipitate AR, and the anti-HNF-3α (C-20) goat polyclonal IgG (sc-6553; Santa Cruz Biotechnology, Inc.) was used to immunoprecipitate HNF-3α. For IP experiments in AR-Hela cells, anti-Flag M2 affinity gel was used to immunoprecipitate the flag-tagged AR, according to the protocol provided in the Flag Tagged Protein Immunoprecipitation Kit (FLAGIPT-1; Sigma).

In Vitro Translation of HNF-3α Proteins and GST Pull-Down Assay

HNF-3α expression vectors with a T7 promoter were transcribed and translated in vitro using the TNT T7 Quick Coupled Transcription/Translation System (L1170; Promega Corp.). A standard reaction involved a 90-min incubation at 30 C with 40 μl of TNT Quick Master, 2 μl of cold 2 mm methionine, and 2 μg of plasmid DNA in a final volume of 50 μl. A LacZ expression vector was translated using the same conditions as a control. In vitro translated recombinant HNF-3α and LacZ proteins were labeled with a C-terminal V5-epitope and were used immediately for in vitro binding reactions.

GST-AR fusion proteins were purified as described previously (66). For GST pull-down assays, 50 μl swelled glutathione agarose beads (G-4510; Sigma) were incubated with 20 μg GST or GST-AR fusion proteins for each reaction. GST-bound beads were equilibrated with PBS-T binding buffer [1× PBS (pH 7.4), 1% Tween 20, and protease inhibitors] and incubated for 2 h at 4 C with 5–10 μl products from the TNT reactions. Complexes were washed four times with 1.5 ml of cold binding buffer, heated for 10 min at 70 C in 1× LDS loading buffer, and separated by SDS-PAGE, after which V5-horseradish peroxidase antibody was used in a standard Western blot to detect proteins that interact with AR in vitro.

Acknowledgments

We thank Dr. Michael Carey for providing the AR-Hela cells, Dr. Scott Shappell for providing human prostate tissues, and Dr. Kenneth S. Zaret for providing the HNF-3α expression vector. We are grateful to Dr. Marie-Claire Orgebin-Crist, Dr. Susan Kasper, and Dr. Simon W. Hayward for reading the manuscript. We thank Manik Paul for technical assistance. We are also indebted to Dr. Sunil K. Halder, Dr. Chaitanya Nirodi, and Dr. William Tu for their experimental advice.

This work was supported by NIH Grants R01-DK-55748 and R01-CA76142, the Frances Williams Preston Laboratories of the T. J. Martell Foundation and the grant from the Terry Fox Foundation (to P.S.R.).

Abbreviations:

1

Pointis

G

,

Latreille

MT

,

Mignot

TM

,

Janssens

Y

,

Cedard

L

1979

Regulation of testosterone synthesis in the fetal mouse testis.

J Steroid Biochem

11

:

1609

1612

2

Pointis

G

,

Latreille

MT

,

Cedard

L

1980

Gonado-pituitary relationships in the fetal mouse at various times during sexual differentiation.

J Endocrinol

86

:

483

488

3

Cunha

GR

,

Donjacour

AA

,

Cooke

PS

,

Mee

S

,

Bigsby

RM

,

Higgins

SJ

,

Sugimura

Y

1987

The endocrinology and developmental biology of the prostate.

Endocr Rev

8

:

338

362

4

Gelmann

EP

2002

Molecular biology of the androgen receptor.

J Clin Oncol

20

:

3001

3015

5

Quigley

CA

, De

Bellis

A

,

Marschke

KB

, el

Awady

MK

,

Wilson

EM

,

French

FS

1995

Androgen receptor defects: historical, clinical, and molecular perspectives.

Endocr Rev

16

:

271

321

6

Donjacour

AA

,

Cunha

GR

1993

Assessment of prostatic protein secretion in tissue recombinants made of urogenital sinus mesenchyme and urothelium from normal or androgen-insensitive mice.

Endocrinology

132

:

2342

2350

7

Cunha

GR

,

Chung

LW

1981

Stromal-epithelial interactions. I. Induction of prostatic phenotype in urothelium of testicular feminized (Tfm/y) mice.

J Steroid Biochem

14

:

1317

1324

8

Takeda

H

,

Mizuno

T

,

Lasnitzki

I

1985

Autoradiographic studies of androgen-binding sites in the rat urogenital sinus and postnatal prostate.

J Endocrinol

104

:

87

92

9

Cooke

PS

,

Young

P

,

Cunha

GR

1991

Androgen receptor expression in developing male reproductive organs.

Endocrinology

128

:

2867

2873

10

Hayward

SW

,

Baskin

LS

,

Haughney

PC

,

Cunha

AR

,

Foster

BA

,

Dahiya

R

,

Prins

GS

,

Cunha

GR

1996

Epithelial development in the rat ventral prostate, anterior prostate and seminal vesicle.

Acta Anat (Basel)

155

:

81

93

11

Cunha

GR

,

Alarid

ET

,

Turner

T

,

Donjacour

AA

,

Boutin

EL

,

Foster

BA

1992

Normal and abnormal development of the male urogenital tract. Role of androgens, mesenchymal-epithelial interactions, and growth factors.

J Androl

13

:

465

475

12

Cunha

GR

1975

Age-dependent loss of sensitivity of female urogenital sinus to androgenic conditions as a function of the epithelia-stromal interaction in mice.

Endocrinology

97

:

665

673

13

Cunha

GR

,

Lung

B

,

Reese

B

1980

Glandular epithelial induction by embryonic mesenchyme in adult bladder epithelium of BALB/c mice.

Invest Urol

17

:

302

304

14

Neubauer

BL

,

Chung

LW

,

McCormick

KA

,

Taguchi

O

,

Thompson

TC

,

Cunha

GR

1983

Epithelial-mesenchymal interactions in prostatic development. II. Biochemical observations of prostatic induction by urogenital sinus mesenchyme in epithelium of the adult rodent urinary bladder.

J Cell Biol

96

:

1671

1676

15

Cunha

GR

,

Fujii

H

,

Neubauer

BL

,

Shannon

JM

,

Sawyer

L

,

Reese

BA

1983

Epithelial-mesenchymal interactions in prostatic development. I. Morphological observations of prostatic induction by urogenital sinus mesenchyme in epithelium of the adult rodent urinary bladder.

J Cell Biol

96

:

1662

1670

16

Aboseif

S

, El

Sakka

A

,

Young

P

,

Cunha

G

1999

Mesenchymal reprogramming of adult human epithelial differentiation.

Differentiation

65

:

113

118

17

Hayward

SW

2002

Approaches to modeling stromal-epithelial interactions.

J Urol

168

:

1165

1172

18

Kaestner

KH

,

Knochel

W

,

Martinez

DE

2000

Unified nomenclature for the winged helix/forkhead transcription factors.

Genes Dev

14

:

142

146

19

Zaret

K

1999

Developmental competence of the gut endoderm: genetic potentiation by GATA and HNF3/fork head proteins.

Dev Biol

209

:

1

10

20

Kaufmann

E

,

Knochel

W

1996

Five years on the wings of fork head.

Mech Dev

57

:

3

20

21

Sasaki

H

,

Hogan

BL

1993

Differential expression of multiple fork head related genes during gastrulation and axial pattern formation in the mouse embryo.

Development

118

:

47

59

22

Weinstein

DC

,

Altaba

A

,

Chen

WS

,

Hoodless

P

,

Prezioso

VR

,

Jessell

TM

, Darnell

Jr

JE

1994

The winged-helix transcription factor HNF-3β is required for notochord development in the mouse embryo.

Cell

78

:

575

588

23

Kaestner

KH

,

Katz

J

,

Liu

Y

,

Drucker

DJ

,

Schutz

G

1999

Inactivation of the winged helix transcription factor HNF3α affects glucose homeostasis and islet glucagon gene expression in vivo.

Genes Dev

13

:

495

504

24

Cirillo

LA

,

Lin

FR

,

Cuesta

I

,

Friedman

D

,

Jarnik

M

,

Zaret

KS

2002

Opening of compacted chromatin by early developmental transcription factors HNF3 (FoxA) and GATA-4.

Mol Cell

9

:

279

289

25

Jackson

DA

,

Rowader

KE

,

Stevens

K

,

Jiang

C

,

Milos

P

,

Zaret

KS

1993

Modulation of liver-specific transcription by interactions between hepatocyte nuclear factor 3 and nuclear factor 1 binding DNA in close apposition.

Mol Cell Biol

13

:

2401

2410

26

Zaret

KS

2002

Regulatory phases of early liver development: paradigms of organogenesis.

Nat Rev Genet

3

:

499

512

27

Lai

E

,

Prezioso

VR

,

Smith

E

,

Litvin

O

,

Costa

RH

, Darnell

Jr

JE

1990

HNF-3A, a hepatocyte-enriched transcription factor of novel structure is regulated transcriptionally.

Genes Dev

4

:

1427

1436

28

Peterson

RS

,

Clevidence

DE

,

Ye

H

,

Costa

RH

1997

Hepatocyte nuclear factor-3α promoter regulation involves recognition by cell-specific factors, thyroid transcription factor-1, and autoactivation.

Cell Growth Differ

8

:

69

82

29

Kopachik

W

,

Hayward

SW

,

Cunha

GR

1998

Expression of hepatocyte nuclear factor-3α in rat prostate, seminal vesicle, and bladder.

Dev Dyn

211

:

131

140

30

Rennie

PS

,

Bruchovsky

N

,

Leco

KJ

,

Sheppard

PC

,

McQueen

SA

,

Cheng

H

,

Block

ME

,

MacDonald

BS

,

Nickel

BE

,

Chang

C

,

Liao

S

,

Cattini

PA

,

Matusik

RJ

1993

Characterization of two cis-acting elements involved in the androgen regulation of the probasin gene.

Mol Endocrinol

7

:

23

36

31

Greenberg

NM

,

DeMayo

FJ

,

Sheppard

PC

,

Barrios

R

,

Lebovitz

M

,

Finegold

M

,

Angelopoulou

R

,

Dodd

JG

,

Duckworth

ML

,

Rosen

JM

,

Matusik

RJ

1994

The rat probasin gene promoter directs hormonally and developmentally regulated expression of a heterologous gene specifically to the prostate in transgenic mice.

Mol Endocrinol

8

:

230

239

32

Kasper

S

,

Rennie

PS

,

Bruchovsky

N

,

Sheppard

PC

,

Cheng

H

,

Lin

L

,

Shiu

RPC

,

Snoek

R

,

Matusik

RJ

1994

Cooperative binding of androgen receptors to two DNA sequences is required for androgen induction of the probasin gene.

J Biol Chem

269

:

31763

31769

33

Yan

Y

,

Sheppard

PC

,

Kasper

S

,

Lin

L

,

Hoare

S

,

Kapoor

A

,

Dodd

JG

,

Duckworth

ML

,

Matusik

RJ

1997

A large fragment of the probasin promoter targets high levels of transgene expression to the prostate of transgenic mice.

The Prostate

32

:

129

139

34

Kasper

S

,

Rennie

PS

,

Bruchovsky

N

,

Lin

L

,

Cheng

H

,

Snoek

R

,

Dahlman-Wright

K

,

Gustafsson

JA

,

Shui

R

,

Sheppard

PC

,

Matusik

RJ

1999

Selective activation of the probasin androgen responsive region by steroid hormones.

J Mol Endocrinol

22

:

313

325

35

Kasper

S

,

Sheppard

PC

,

Yan

Y

,

Pettigrew

N

,

Borowsky

AD

,

Prins

GS

,

Dodd

JG

,

Duckworth

ML

,

Matusik

RJ

1998

Development, progression and androgen-dependence of prostate tumors in transgenic: a model for prostate cancer.

Lab Invest

78

:

319

334

36

Zhang

ZF

,

Thomas

TZ

,

Kasper

S

,

Matusik

RJ

2000

A small composite probasin promoter confers high levels of prostate-specific gene expression through regulation by androgens and glucocorticoid in vitro and in vivo.

Endocrinology

141

:

4698

4710

37

Masumori

N

,

Thomas

TZ

,

Case

T

,

Paul

M

,

Kasper

S

,

Chaurand

P

,

Caprioli

RM

,

Tsukamoto

T

,

Shappell

SB

,

Matusik

RJ

2001

A probasin-large T antigen transgenic mouse line develops prostate adeno and neuroendocrine carcinoma with metastatic potential.

Cancer Res

61

:

2239

2249

38

Kasper

S

,

Matusik

RJ

2000

Rat probasin: structure and function of an outlier lipocalin.

Biochim Biophys Acta

1482

:

249

258

39

Greenberg

NM

,

DeMayo

FJ

,

Finegold

MJ

,

Medina

D

,

Tilley

WD

,

Aspinall

JO

,

Cunha

GR

,

Donjacour

AA

,

Matusik

RJ

,

Rosen

JM

1995

Prostate cancer in a transgenic mouse.

Proc Natl Acad Sci USA

92

:

3439

3443

40

Barrios

R

,

Lebovitz

RM

,

Wiseman

AL

,

Weisoly

DL

,

Matusik

RJ

,

DeMayo

F

,

Lieberman

MW

1996

RasT24 driven by a probasin promoter induces prostatic hyperplasia in transgenic mice.

Transgenics

2

:

23

28

41

Green

JE

,

Greenberg

NM

,

Ashendel

CL

,

Barrett

JC

,

Boone

CW

,

Getzenberg

RH

,

Henkin

J

,

Matusik

RJ

,

Janus

TJ

,

Scher

HI

1998

Transgenic and reconstitution models of prostate cancer.

The Prostate

36

:

59

63

42

Lai

E

,

Prezioso

VR

,

Tao

WF

,

Chen

WS

, Darnell

Jr

JE

1991

Hepatocyte nuclear factor 3α belongs to a gene family in mammals that is homologous to the Drosophila homeotic gene fork head.

Genes Dev

5

:

416

427

43

Costa

RH

,

Grayson

DR

, Darnell

Jr

JE

1989

Multiple hepatocyte-enriched nuclear factors function in the regulation of transthyretin and α1-antitrypsin genes.

Mol Cell Biol

9

:

1415

1425

44

Gaudet

J

,

Mango

SE

2002

Regulation of organogenesis by the Caenorhabditis elegans FoxA protein PHA-4.

Science

295

:

821

825

45

Cirillo

LA

,

McPherson

CE

,

Bossard

P

,

Stevens

K

,

Cherian

S

,

Shim

EY

,

Clark

KL

,

Burley

SK

,

Zaret

KS

1998

Binding of the winged-helix transcription factor HNF3 to a linker histone site on the nucleosome.

EMBO J

17

:

244

254

46

Smith

CL

,

Hager

GL

1997

Transcriptional regulation of mammalian genes in vivo. A tale of two templates.

J Biol Chem

272

:

27493

27496

47

Crowe

AJ

,

Sang

L

,

Li

KK

,

Lee

KC

,

Spear

BT

,

Barton

MC

1999

Hepatocyte nuclear factor 3 relieves chromatin-mediated repression of the α-fetoprotein gene.

J Biol Chem

274

:

25113

25120

48

Patrikainen

L

,

Shan

J

,

Porvari

K

,

Vihko

P

1999

Identification of the deoxyribonucleic acid-binding site of a regulatory protein involved in prostate-specific and androgen receptor-dependent gene expression.

Endocrinology

140

:

2063

2070

49

Pani

L

,

Overdier

DG

,

Porcella

A

,

Qian

X

,

Lai

E

,

Costa

RH

1992

Hepatocyte nuclear factor 3β contains two transcriptional activation domains, one of which is novel and conserved with the Drosophila fork head protein.

Mol Cell Biol

12

:

3723

3732

50

Qian

X

,

Costa

RH

1995

Analysis of hepatocyte nuclear factor-3 β protein domains required for transcriptional activation and nuclear targeting.

Nucleic Acids Res

23

:

1184

1191

51

Schuur

ER

,

Henderson

GA

,

Kmetec

LA

,

Miller

JD

,

Lamparski

HG

,

Henderson

DR

1996

Prostate-specific antigen expression is regulated by an upstream enhancer.

J Biol Chem

271

:

7043

7051

52

Cleutjens

KB

, van der

Korput

HA

, van

Eekelen

CC

, van

Rooij

HC

,

Faber

PW

,

Trapman

J

1997

An androgen response element in a far upstream enhancer region is essential for high, androgen-regulated activity of the prostate-specific antigen promoter.

Mol Endocrinol

11

:

148

161

53

Farmer

G

, Connolly

Jr

ES

,

Mocco

J

,

Freedman

LP

2001

Molecular analysis of the prostate-specific antigen upstream gene enhancer.

Prostate

46

:

76

85

54

Pang

S

,

Dannull

J

,

Kaboo

R

,

Xie

Y

,

Tso

CL

,

Michel

K

,

deKernion

JB

,

Belldegrun

AS

1997

Identification of a positive regulatory element responsible for tissue- specific expression of prostate-specific antigen.

Cancer Res

57

:

495

499

55

Huang

W

,

Shostak

Y

,

Tarr

P

,

Sawyers

C

,

Carey

M

1999

Cooperative assembly of androgen receptor into a nucleoprotein complex that regulates the prostate-specific antigen enhancer.

J Biol Chem

274

:

25756

25768

56

Shang

Y

,

Myers

M

,

Brown

M

2002

Formation of the androgen receptor transcription complex.

Mol Cell

9

:

601

610

57

Espinas

ML

,

Roux

J

,

Pictet

R

,

Grange

T

1995

Glucocorticoids and protein kinase A coordinately modulate transcription factor recruitment at a glucocorticoid-responsive unit.

Mol Cell Biol

15

:

5346

5354

58

Stafford

JM

,

Wilkinson

JC

,

Beechem

JM

,

Granner

DK

2001

Accessory factors facilitate the binding of glucocorticoid receptor to the phosphoenolpyruvate carboxykinase gene promoter.

J Biol Chem

276

:

39885

39891

59

Wang

JC

,

Stromstedt

PE

,

Sugiyama

T

,

Granner

DK

1999

The phosphoenolpyruvate carboxykinase gene glucocorticoid response unit: identification of the functional domains of accessory factors HNF3 β (hepatic nuclear factor-3 β) and HNF4 and the necessity of proper alignment of their cognate binding sites.

Mol Endocrinol

13

:

604

618

60

O’Brien

RM

,

Noisin

EL

,

Suwanichkul

A

,

Yamasaki

T

,

Lucas

PC

,

Wang

JC

,

Powell

DR

,

Granner

DK

1995

Hepatic nuclear factor 3- and hormone-regulated expression of the phosphoenolpyruvate carboxykinase and insulin-like growth factor-binding protein 1 genes.

Mol Cell Biol

15

:

1747

1758

61

Wang

JC

,

Stromstedt

PE

,

O’Brien

RM

,

Granner

DK

1996

Hepatic nuclear factor 3 is an accessory factor required for the stimulation of phosphoenolpyruvate carboxykinase gene transcription by glucocorticoids.

Mol Endocrinol

10

:

794

800

62

Christoffels

VM

,

Grange

T

,

Kaestner

KH

,

Cole

TJ

,

Darlington

GJ

,

Croniger

CM

,

Lamers

WH

1998

Glucocorticoid receptor, C/EBP, HNF3, and protein kinase A coordinately activate the glucocorticoid response unit of the carbamoylphosphate synthetase I gene.

Mol Cell Biol

18

:

6305

6315

63

Schuur

ER

,

Loktev

AV

,

Sharma

M

,

Sun

Z

,

Roth

RA

,

Weigel

RJ

2001

Ligand-dependent interaction of estrogen receptor-α with members of the forkhead transcription factor family.

J Biol Chem

276

:

33554

33560

64

Zhao

HH

,

Herrera

RE

,

Coronado-Heinsohn

E

,

Yang

MC

,

Ludes-Meyers

JH

,

Seybold-Tilson

KJ

,

Nawaz

Z

,

Yee

D

,

Barr

FG

,

Diab

SG

,

Brown

PH

,

Fuqua

SA

,

Osborne

CK

2001

Forkhead homologue in rhabdomyosarcoma functions as a bifunctional nuclear receptor-interacting protein with both coactivator and corepressor functions.

J Biol Chem

276

:

27907

27912

65

Lai

JS

,

Herr

W

1992

Ethidium bromide provides a simple tool for identifying genuine DNA-independent protein associations.

Proc Natl Acad Sci USA

89

:

6958

6962

66

Snoek

R

,

Rennie

PS

,

Kasper

S

,

Matusik

RJ

,

Bruchovsky

N

1996

Induction of cell-free, in vitro transcription by recombinant androgen receptor peptides.

J Steroid Biochem Mol Biol

59

:

243

250

67

Weigel

D

,

Jackle

H

1990

The fork head domain: a novel DNA binding motif of eukaryotic transcription factors?

Cell

63

:

455

456

68

Lai

E

,

Clark

KL

,

Burley

SK

, Darnell

Jr

J

1993

Hepatocyte nuclear factor 3/fork head or “winged helix” proteins: a family of transcription factors of diverse biologic function.

Proc Natl Acad Sci USA

90

:

10421

10423

69

Gualdi

R

,

Bossard

P

,

Zheng

M

,

Hamada

Y

,

Coleman

JR

,

Zaret

KS

1996

Hepatic specification of the gut endoderm in vitro: cell signaling and transcriptional control.

Genes Dev

10

:

1670

1682

70

Thomson

AA

2001

Role of androgens and fibroblast growth factors in prostatic development.

Reproduction

121

:

187

195

71

Virkkunen

P

,

Hedberg

P

,

Palvimo

JJ

,

Birr

E

,

Porvari

K

,

Ruokonen

M

,

Taavitsainen

P

,

Janne

OA

,

Vihko

P

1994

Structural comparison of human and rat prostate- specific acid phosphatase genes and their promoters: identification of putative androgen response elements.

Biochem Biophys Res Commun

202

:

49

57

72

Claessens

F

,

Dirckx

L

,

Delaey

B

,

Decourt

JL

,

Hemschoote

K

,

Peeters

B

,

Rombauts

W

1989

The androgen-dependent rat prostatic binding protein: comparison of the sequences in the 5′ part and upstream region of the C1 and C2 genes and analysis of their transcripts.

J Mol Endocrinol

3

:

93

103

73

Carey

M

1998

The enhanceosome and transcriptional synergy.

Cell

92

:

5

8

74

Brivanlou

AH

, Darnell

Jr

JE

2002

Signal transduction and the control of gene expression.

Science

295

:

813

818

75

Perez-Stable

CM

,

Pozas

A

,

Roos

BA

2000

A role for GATA transcription factors in the androgen regulation of the prostate-specific antigen gene enhancer.

Mol Cell Endocrinol

167

:

43

53

76

Zelivianski

S

,

Igawa

T

,

Lim

S

,

Taylor

R

,

Lin

MF

2002

Identification and characterization of regulatory elements of the human prostatic acid phosphatase promoter.

Oncogene

21

:

3696

3705

77

Clark

KL

,

Halay

ED

,

Lai

E

,

Burley

SK

1993

Co-crystal structure of the HNF-3/fork head DNA-recognition motif resembles histone H5.

Nature

364

:

412

420

78

Vallet

V

,

Antoine

B

,

Chafey

P

,

Vandewalle

A

,

Kahn

A

1995

Overproduction of a truncated hepatocyte nuclear factor 3 protein inhibits expression of liver-specific genes in hepatoma cells.

Mol Cell Biol

15

:

5453

5460

79

Wang

JC

,

Waltner-Law

M

,

Yamada

K

,

Osawa

H

,

Stifani

S

,

Granner

DK

2000

Transducin-like enhancer of split proteins, the human homologs of Drosophila groucho, interact with hepatic nuclear factor 3β.

J Biol Chem

275

:

18418

18423

80

Heinlein

CA

,

Chang

C

2002

Androgen receptor (AR) coregulators: an overview.

Endocr Rev

23

:

175

200

81

Sato

N

,

Sadar

MD

,

Bruchovsky

N

,

Saatcioglu

F

,

Rennie

PS

,

Sato

S

,

Lange

PH

,

Gleave

ME

1997

Androgenic induction of prostate-specific antigen gene is repressed by protein-protein interaction between the androgen receptor and AP-1/c-Jun in the human prostate cancer cell line LNCaP.

J Biol Chem

272

:

17485

17494

82

Kang

HY

,

Lin

HK

,

Hu

YC

,

Yeh

S

,

Huang

KE

,

Chang

C

2001

From transforming growth factor-β signaling to androgen action: identification of Smad3 as an androgen receptor coregulator in prostate cancer cells.

Proc Natl Acad Sci USA

98

:

3018

3023

83

Kang

HY

,

Huang

KE

,

Chang

SY

,

Ma

WL

,

Lin

WJ

,

Chang

C

2002

Differential modulation of androgen receptor-mediated transactivation by Smad3 and tumor suppressor Smad4.

J Biol Chem

277

:

43749

43756

84

Palvimo

J

,

Reinikainen

P

,

Ikonen

T

,

Kallio

P

,

Moilanen

A

,

Janne

OA

1996

Mutual transcription interference between RelA and androgen receptor.

J Biol Chem

271

:

24151

24156

85

Yuan

X

,

Lu

ML

,

Li

T

,

Balk

SP

2001

SRY interacts with and negatively regulates androgen receptor transcriptional activity.

J Biol Chem

276

:

46647

46654

86

Oettgen

P

,

Finger

E

,

Sun

Z

,

Akbarali

Y

,

Thamrongsak

U

,

Boltax

J

,

Grall

F

,

Dube

A

,

Weiss

A

,

Brown

L

,

Quinn

G

,

Kas

K

,

Endress

G

,

Kunsch

C

,

Libermann

TA

2000

PDEF, a novel prostate epithelium-specific ets transcription factor, interacts with the androgen receptor and activates prostate-specific antigen gene expression.

J Biol Chem

275

:

1216

1225

87

Boonyaratanakornkit

V

,

Melvin

V

,

Prendergast

P

,

Altmann

M

,

Ronfani

L

,

Bianchi

ME

,

Taraseviciene

L

,

Nordeen

SK

,

Allegretto

EA

,

Edwards

DP

1998

High-mobility group chromatin proteins 1 and 2 functionally interact with steroid hormone receptors to enhance their DNA binding in vitro and transcriptional activity in mammalian cells.

Mol Cell Biol

18

:

4471

4487

88

Robyr

D

,

Gegonne

A

,

Wolffe

AP

,

Wahli

W

2000

Determinants of vitellogenin B1 promoter architecture. HNF3 and estrogen responsive transcription within chromatin.

J Biol Chem

275

:

28291

28300

89

Aarnisalo

P

,

Palvimo

JJ

,

Janne

OA

1998

CREB-binding protein in androgen receptor-mediated signaling.

Proc Natl Acad Sci USA

95

:

2122

2127

90

Moilanen

AM

,

Poukka

H

,

Karvonen

U

,

Hakli

M

,

Janne

OA

,

Palvimo

JJ

1998

Identification of a novel RING finger protein as a coregulator in steroid receptor-mediated gene transcription.

Mol Cell Biol

18

:

5128

5139

91

Moilanen

AM

,

Karvonen

U

,

Poukka

H

,

Yan

W

,

Toppari

J

,

Janne

OA

,

Palvimo

JJ

1999

A testis-specific androgen receptor coregulator that belongs to a novel family of nuclear proteins.

J Biol Chem

274

:

3700

3704

92

Moilanen

AM

,

Karvonen

U

,

Poukka

H

,

Janne

OA

,

Palvimo

JJ

1998

Activation of androgen receptor function by a novel nuclear protein kinase.

Mol Biol Cell

9

:

2527

2543

93

Schwerk

C

,

Klotzbucher

M

,

Sachs

M

,

Ulber

V

,

Klein-Hitpass

L

1995

Identification of a transactivation function in the progesterone receptor that interacts with the TAFII110 subunit of the TFIID complex.

J Biol Chem

270

:

21331

21338

Copyright © 2003 by The Endocrine Society

Citations

Views

Altmetric

Metrics

Total Views 2,932

1,999 Pageviews

933 PDF Downloads

Since 1/1/2017

Month: Total Views:
January 2017 3
February 2017 14
March 2017 19
April 2017 20
May 2017 10
June 2017 4
July 2017 12
August 2017 10
September 2017 11
October 2017 15
November 2017 12
December 2017 15
January 2018 21
February 2018 22
March 2018 30
April 2018 17
May 2018 25
June 2018 34
July 2018 33
August 2018 14
September 2018 35
October 2018 26
November 2018 30
December 2018 28
January 2019 17
February 2019 40
March 2019 42
April 2019 106
May 2019 27
June 2019 33
July 2019 37
August 2019 32
September 2019 21
October 2019 24
November 2019 14
December 2019 20
January 2020 24
February 2020 30
March 2020 38
April 2020 59
May 2020 17
June 2020 21
July 2020 34
August 2020 18
September 2020 41
October 2020 34
November 2020 31
December 2020 19
January 2021 27
February 2021 38
March 2021 33
April 2021 22
May 2021 11
June 2021 27
July 2021 21
August 2021 23
September 2021 29
October 2021 16
November 2021 34
December 2021 16
January 2022 26
February 2022 23
March 2022 37
April 2022 25
May 2022 42
June 2022 44
July 2022 19
August 2022 27
September 2022 45
October 2022 70
November 2022 37
December 2022 43
January 2023 29
February 2023 24
March 2023 33
April 2023 36
May 2023 41
June 2023 63
July 2023 27
August 2023 39
September 2023 41
October 2023 37
November 2023 34
December 2023 36
January 2024 59
February 2024 77
March 2024 42
April 2024 84
May 2024 63
June 2024 55
July 2024 29
August 2024 52
September 2024 48
October 2024 9

Citations

210 Web of Science

×

Email alerts

Citing articles via

More from Oxford Academic