12
Selective Role of an NH 2 -Terminal WxxLF Motif for Aberrant Androgen Receptor Activation in Androgen Depletion– Independent Prostate Cancer Cells Scott M. Dehm, Kevin M. Regan, Lucy J. Schmidt, and Donald J. Tindall Departments of Urology and Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota Abstract Systemic prostate cancer therapy requires androgen ablation, which inhibits the production or action of androgens. Prostate cancer ultimately relapses during androgen ablation, and an androgen depletion-independent (ADI) phenotype emerges. Aberrant androgen receptor (AR) activation underlies therapy resistance at this stage of the disease, and mounting evidence implicates the large and highly disordered AR NH 2 -terminal domain (NTD) as a key mediator of this activity. In this study, we investigated the role of the NTD transactivation unit 5 (TAU5) domain in mediating AR transcriptional activity in cell-based models of prostate cancer progression. AR replace- ment and Gal4-based promoter tethering experiments revealed that AR TAU5 had a dichotomous function, inhibiting ligand-dependent AR activity in androgen-dependent prostate cancer cells, while enhancing ligand-independent AR activity in ADI prostate cancer cells. Molecular dissection of TAU5 showed that a WxxLF motif was fully responsible for its ligand- independent activity. Mechanistically, WxxLF did not rely on an interaction with the AR ligand-binding domain to mediate ligand-independent AR activity. Rather, WxxLF functioned as an autonomous transactivation domain. These data show that ligand-dependent and ligand-independent AR activation rely on fundamentally distinct mechanisms, and define WxxLF as the major transactivation motif within the AR TAU5 domain. [Cancer Res 2007;67(20):10067–77] Introduction Prostate cancer is the most common cancer affecting North American men (1). Activity of the androgen receptor (AR), a steroid-regulated nuclear transcription factor, is critical for the growth and survival of normal and cancerous prostate tissue (2, 3). Therefore, androgen ablation, a systemic treatment for locally advanced or metastatic prostate cancer, initially results in tumor regression. However, an untreatable manifestation of the disease, termed androgen depletion–independent (ADI) prostate cancer (4), develops after a median of 18 to 33 months (5). Although ADI prostate cancer is resistant to androgen ablation, a wealth of evidence supports the current paradigm that this stage of the disease remains AR dependent through various mechanisms of aberrant AR activation (3, 5, 6). Characterizing these modes of AR activation is of prime importance for developing new prostate cancer therapies. The AR shares a modular organization with other steroid receptors, having an NH 2 -terminal domain (NTD) harboring AR transcriptional activation function (AF)-1, a central DNA binding domain (DBD), and a COOH-terminal ligand-binding domain (LBD), which also harbors the AF-2 coactivator binding surface (7). Data from our laboratory and others have shown that the AR displays ligand-independent activity in a cell-based model of prostate cancer progression (8–10). Previously, we have shown that ligand-independent AR activity is resistant to antiandrogens such as bicalutamide due to a mechanism of activation that is independent of the AR LBD/AF-2 module (8). These observations suggest that the AR NTD could play a key role in mediating aberrant AR activity in ADI prostate cancer cells. This hypothesis is supported by the finding that a decoy peptide representing the entire AR NTD can inhibit AR activity and prostate cancer tumor growth, and thereby delay emergence of an ADI phenotype in a xenograft-based model of prostate cancer progression (11). The AR NTD is highly flexible and displays intrinsic disorder in solution, which has hampered elucidation of its three-dimensional structure (7, 12–14). Moreover, specific NTD domains, through which coregulatory proteins may bind and regulate ligand- dependent or ligand-independent transcriptional activation, have not been fully described. Deletion analysis of ectopic AR in AR-null cell lines has shown that AF-1 in the AR NTD consists of two large domains, termed transactivation unit (TAU) 1 and TAU5, which participate in transcriptional activation (15). The core domain mediating TAU1 activity has been mapped to a discrete 178 LKDIL 182 motif, and its importance has been confirmed in various cell models (8, 15–17). However, the role or precise identity of AR TAU5 has not been established. The purpose of this study was to elucidate the role of TAU5 in mediating ligand-dependent and ligand-independent AR activation in cell-based models of prostate cancer progression. We show that TAU5 plays fundamen- tally different roles in androgen-dependent and ADI prostate cancer cells. In addition, we define the core sequence 435 WHTLF 439 as a novel AR transactivation motif that mediates TAU5 activity. Blocking the function of AR TAU5, via targeting 435 WHTLF 439 , may represent a novel strategy to selectively inhibit aberrant AR activity in ADI prostate cancer cells. Materials and Methods Cell lines and culture conditions. LNCaP and ADI 22Rv1 cell lines were purchased from the American Type Culture Collection. The C4-2 cell line was purchased from UroCor. LNCaP, C4-2, and 22Rv1 cells were grown in RPMI 1640 supplemented with 10% fetal bovine serum. For androgen response experiments, cells were grown in serum-free medium for 48 h. Growth medium was replaced with serum-free medium containing 1 nmol/L mibolerone (Biomol), 10 Amol/L bicalutamide (Casodex, AstraZeneca), Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Requests for reprints: Donald J. Tindall, Departments of Urology, Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, 200 First Street Southwest, Rochester, MN 55905. Phone: 507-284-8139; Fax: 507-284-2384; E-mail: [email protected]. I2007 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-07-1267 www.aacrjournals.org 10067 Cancer Res 2007; 67: (20). October 15, 2007 Research Article Research. on June 16, 2018. © 2007 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Selective Role of an NH -Terminal WxxLF Motif for …cancerres.aacrjournals.org/content/67/20/10067.full.pdfment and Gal4-based promoter tethering experiments ... -1, a central DNA

Embed Size (px)

Citation preview

Selective Role of an NH2-Terminal WxxLF Motif for Aberrant

Androgen Receptor Activation in Androgen Depletion–

Independent Prostate Cancer Cells

Scott M. Dehm, Kevin M. Regan, Lucy J. Schmidt, and Donald J. Tindall

Departments of Urology and Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota

Abstract

Systemic prostate cancer therapy requires androgen ablation,which inhibits the production or action of androgens. Prostatecancer ultimately relapses during androgen ablation, and anandrogen depletion-independent (ADI) phenotype emerges.Aberrant androgen receptor (AR) activation underlies therapyresistance at this stage of the disease, and mounting evidenceimplicates the large and highly disordered AR NH2-terminaldomain (NTD) as a key mediator of this activity. In this study,we investigated the role of the NTD transactivation unit 5(TAU5) domain in mediating AR transcriptional activity incell-based models of prostate cancer progression. AR replace-ment and Gal4-based promoter tethering experimentsrevealed that AR TAU5 had a dichotomous function, inhibitingligand-dependent AR activity in androgen-dependent prostatecancer cells, while enhancing ligand-independent AR activityin ADI prostate cancer cells. Molecular dissection of TAU5showed that a WxxLF motif was fully responsible for its ligand-independent activity. Mechanistically, WxxLF did not rely onan interaction with the AR ligand-binding domain to mediateligand-independent AR activity. Rather, WxxLF functioned asan autonomous transactivation domain. These data show thatligand-dependent and ligand-independent AR activation relyon fundamentally distinct mechanisms, and define WxxLF asthe major transactivation motif within the AR TAU5 domain.[Cancer Res 2007;67(20):10067–77]

Introduction

Prostate cancer is the most common cancer affecting NorthAmerican men (1). Activity of the androgen receptor (AR), asteroid-regulated nuclear transcription factor, is critical for thegrowth and survival of normal and cancerous prostate tissue (2, 3).Therefore, androgen ablation, a systemic treatment for locallyadvanced or metastatic prostate cancer, initially results in tumorregression. However, an untreatable manifestation of the disease,termed androgen depletion–independent (ADI) prostate cancer (4),develops after a median of 18 to 33 months (5). Although ADIprostate cancer is resistant to androgen ablation, a wealth ofevidence supports the current paradigm that this stage of thedisease remains AR dependent through various mechanisms ofaberrant AR activation (3, 5, 6). Characterizing these modes of AR

activation is of prime importance for developing new prostatecancer therapies.

The AR shares a modular organization with other steroidreceptors, having an NH2-terminal domain (NTD) harboring ARtranscriptional activation function (AF)-1, a central DNA bindingdomain (DBD), and a COOH-terminal ligand-binding domain(LBD), which also harbors the AF-2 coactivator binding surface(7). Data from our laboratory and others have shown that the ARdisplays ligand-independent activity in a cell-based model ofprostate cancer progression (8–10). Previously, we have shown thatligand-independent AR activity is resistant to antiandrogens suchas bicalutamide due to a mechanism of activation that isindependent of the AR LBD/AF-2 module (8). These observationssuggest that the AR NTD could play a key role in mediatingaberrant AR activity in ADI prostate cancer cells. This hypothesis issupported by the finding that a decoy peptide representing theentire AR NTD can inhibit AR activity and prostate cancer tumorgrowth, and thereby delay emergence of an ADI phenotype in axenograft-based model of prostate cancer progression (11).

The AR NTD is highly flexible and displays intrinsic disorder insolution, which has hampered elucidation of its three-dimensionalstructure (7, 12–14). Moreover, specific NTD domains, throughwhich coregulatory proteins may bind and regulate ligand-dependent or ligand-independent transcriptional activation, havenot been fully described. Deletion analysis of ectopic AR in AR-nullcell lines has shown that AF-1 in the AR NTD consists of two largedomains, termed transactivation unit (TAU) 1 and TAU5, whichparticipate in transcriptional activation (15). The core domainmediating TAU1 activity has been mapped to a discrete178LKDIL182 motif, and its importance has been confirmed invarious cell models (8, 15–17). However, the role or precise identityof AR TAU5 has not been established. The purpose of this studywas to elucidate the role of TAU5 in mediating ligand-dependentand ligand-independent AR activation in cell-based models ofprostate cancer progression. We show that TAU5 plays fundamen-tally different roles in androgen-dependent and ADI prostatecancer cells. In addition, we define the core sequence 435WHTLF439

as a novel AR transactivation motif that mediates TAU5 activity.Blocking the function of AR TAU5, via targeting 435WHTLF439, mayrepresent a novel strategy to selectively inhibit aberrant AR activityin ADI prostate cancer cells.

Materials and Methods

Cell lines and culture conditions. LNCaP and ADI 22Rv1 cell lines werepurchased from the American Type Culture Collection. The C4-2 cell line was

purchased from UroCor. LNCaP, C4-2, and 22Rv1 cells were grown in RPMI

1640 supplemented with 10% fetal bovine serum. For androgen responseexperiments, cells were grown in serum-free medium for 48 h. Growth

medium was replaced with serum-free medium containing 1 nmol/L

mibolerone (Biomol), 10 Amol/L bicalutamide (Casodex, AstraZeneca),

Note: Supplementary data for this article are available at Cancer Research Online(http://cancerres.aacrjournals.org/).

Requests for reprints: Donald J. Tindall, Departments of Urology, Biochemistryand Molecular Biology, Mayo Clinic College of Medicine, 200 First Street Southwest,Rochester, MN 55905. Phone: 507-284-8139; Fax: 507-284-2384; E-mail:[email protected].

I2007 American Association for Cancer Research.doi:10.1158/0008-5472.CAN-07-1267

www.aacrjournals.org 10067 Cancer Res 2007; 67: (20). October 15, 2007

Research Article

Research. on June 16, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

ethanol (vehicle control), or combinations of these compounds. Cells werecultured for an additional 24 h and harvested. For 22Rv1 small interfering

RNA (siRNA) transfection experiments, culture medium was replaced

24 h posttransfection with serum-free, phenol red-free RPMI. After 48 h of

growth, cells were harvested.Plasmid constructs. Mouse mammary tumor virus (MMTV)-Luc was

provided by Dr. Frank Claessens (Catholic University of Leuven, Leuven,

Belgium). The p5HBhAR-A plasmid was provided by Dr. Frank French

(University of North Carolina, Chapel Hill, NC). The backbone for the

enhanced green fluorescent protein (EGFP)–expressing AR replacement

plasmid, pCMS4-H1p-EGFP, was provided by Dr. Dan Billadeau (Mayo

Clinic, Rochester, MN). The SV40-Renilla luciferase and pG5-LUC reporter

vectors were purchased from Promega and Clontech, respectively.

PSAenh(ARE)-LUC, PSAenh(GAL4)-LUC, hARGal4, hARGal4DTAU5, and

NTDGal4 have been described (8, 18). Plasmid construction details are

included as Supplementary Materials and Methods.

Transient transfections. For siRNA transfections, 3 � 106 22Rv1 cells

were mixed with 80 pmol of AR-targeted siRNA (AR siRNA1 sense: 5¶-CAAGGGAGGUUACACCAAAUU; ARsiRNA2 sense: 5¶-GAAAUGAUUGCA-

CUAUUGAUU) or a nontargeted control siRNA (Dharmacon). The cell/

siRNA mixture was transferred to electroporation cuvettes with a 4-mm

gap-width (BTX) and subjected to a 350 V electrical pulse for 10 ms using aBTX ElectroSquare Electroporator. Following a 15-min recovery, cells were

seeded in RPMI 1640 + 5% charcoal-stripped, steroid-depleted serum (CSS).

For siRNA transfections coupled with rescue by hARsr, 1 � 105 cells were

seeded in 24-well dishes and transfected the following day with 375 ng ofMMTV-LUC, 125 ng SV40-Renilla, 10 pmol ARsiRNA1 (or control siRNA),

and 11.75 ng ARsr, in complex with 2 AL of LipofectAMINE 2000 (Invitrogen)

according to the manufacturer’s protocol. Following 6 h of transfection, themedium was aspirated and replaced with the appropriate medium

containing 5% CSS. In addition to the LipofectAMINE 2000–based protocol,

electroporation was also used as a mode of transfection for siRNA rescue

experiments with hARsr. For this approach, amounts of plasmids wereincreased by a factor of 24 from the LipofectAMINE 2000–based protocol.

Therefore, for a standard electroporation, 3 � 106 cells from exponentially

growing cultures were suspended in 350 AL RPMI + 5% CSS and mixed with

50 AL of a DNA/siRNA mixture containing 9 Ag MMTV-LUC, 3 Ag SV40-Renilla, 80 pmol ARsiRNA1 (or control siRNA), and 282 ng of hARsr. The

cell/DNA mixture was subjected to a 305 V (LNCaP, C4-2) or 350 V (22Rv1)

pulse for 10 ms. Following a 15-min recovery, cells were seeded in RPMI1640 + 5% CSS.

hARGal4-based tethering assays were done in LNCaP and C4-2 cells as

described (8) with minor modification. Briefly, 1 � 105 cells were seeded in

24-well dishes and transfected the following day using 2.5 AL of Superfect(Qiagen) mixed with 375 ng of either PSAenh(ARE)-LUC or PSAenh(GAL4)-

LUC as a reporter construct, 125 ng of SV40-Renlla, and 23.4 ng of hARGal4.

Transfections were done in the presence of RPMI + 5% CSS. In addition to

the Superfect-based protocol, electroporation was also used as a mode oftransfection for hARGal4-based tethering assays. For this approach, amounts

of plasmids were increased by a factor of 24 from the Superfect-based

protocol. Therefore, for a standard electroporation, 3 � 106 cells were

suspended in 350 AL RPMI + 5% CSS and mixed with 50 AL of a DNAmixture containing 9 Ag of a promoter-LUC reporter, 3 Ag of SV40-Renilla,

and 562 ng of hARGal4. The cell/DNA mixture was electroporated at 305 V

for 10 ms. Following a 15-min recovery, cells were seeded in RPMI 1640 +5% CSS.

Overall transfection efficiency was optimized for each experiment by

transfecting cells with GFP and assessing the number of fluorescent cells 24

h posttransfection. Transfection efficiencies consistently ranged from 60%to 80%. For all reporter-based experiments, 24 h posttransfection, medium

was aspirated and replaced with serum-free, phenol red–free RPMI 1640

containing 1 nmol/L mibolerone, 2 pmol/L to 1 nmol/L dihydrotestosterone

(Sigma Aldrich), or ethanol (vehicle control). Cells were harvested after anadditional 24 h and processed in a lysis buffer provided with a Dual

Luciferase Assay Kit (Promega). Activities of the firefly and Renilla luciferase

reporters were assayed in 96-well plates via a Dual Luciferase Assay Kit anddetected with a Molecular Devices LMax luminometer. Transfection

efficiency was addressed by dividing firefly luciferase activity by Renillaluciferase activity of samples cultured in the absence of androgens. Data

presented represent the mean F SE from at least three independent

experiments, each done in duplicate.

Cell sorting. C4-2 cells were electroporated with AR replacement vectorsexpressing combinations of EGFP, AR-targeted short hairpin RNA (shRNA),

and siRNA-resistant wild-type, DTAU5, or AHTAA mutant AR. EGFP-

positive cells were collected using a FACSVantage SE (Becton Dickson).

RNA extraction, Northern blot, and quantitative reverse transcrip-tion-PCR analysis. Total cellular RNA was isolated via acid-guanidinium

phenol/chloroform extraction as described (19). For Northern blots, equal

amounts of RNA (15 Ag per lane) were fractionated on 1% denaturing

formaldehyde-agarose gels. RNA was transferred to Hybond nylonmembranes (Amersham), UV cross-linked, and hybridized with cDNA

probes specific for prostate-specific antigen (PSA) or glyceraldehyde-3-

phosphate dehydrogenase (GAPDH) labeled with [a32P]dCTP using aRadPrime labeling kit (Invitrogen). Autoradiography was done at �80jCusing an intensifier screen (Kodak). Quantitative real-time PCR was done

exactly as described (20) using primers specific for PSA (5¶-AGGCCTTCCCTGTACACCAA and 5¶-GTCTTGGCCTGGTCATTTCC) andGAPDH (Applied Biosystems). Relative quantitation was used to determine

fold change in expression levels by the comparative C t method using the

formula 2�DDC t where C t is the threshold cycle of amplification.

Chromatin immunoprecipitation. Cells were electroporated withsPSADALL-Gal4 along with empty vector or wild-type, DTAU5, or AHTAA

mutant versions of hARGal4 and cultured in RPMI medium containing 5%

CSS for 48 h. Cells were switched to serum-free, phenol red–free RPMImedium for an additional 24 h. Formaldehyde was added to a final

concentration of 1%, and cells were incubated for 10 min. Cells were

harvested, lysed, and immunoprecipitations were done using a chromatin

immunoprecipitation (ChIP) kit (Upstate Biotechnology) and antibodiesspecific for Gal4 (Santa Cruz Biotechnology), or nonspecific IgG.

Immunoprecipitated DNA fragments were detected via PCR using forward

and reverse primers specific for sPSADALL-Gal4 ( forward: 5¶-AGTG-

CAGGTGCCAGAACATTTC; reverse: 5¶-TTTGTAAAGCAGGCATCCTTGC).Western blot analysis. Cells were harvested directly in a loading buffer

containing 65 mmol/L Tris-HCl (pH 7.0), 2% (w/v) SDS, 5% h-mercaptoe-

thanol, 10% (v/v) glycerol, and 0.5% (w/v) bromophenol blue. Proteinconcentrations were determined using a kit based on a modified Lowry

assay (Bio-Rad RC/DC assay). Equal amounts (typically 30 Ag per lane) of

protein were resolved in 10% SDS-polyacrylamide gels (Invitrogen), followed

by transfer to nitrocellulose and membrane blocking. Blots were incubatedwith antibodies specific for AR (Santa Cruz Biotechnology, C-19 or N-20),

extracellular signal-regulated kinase-2 (ERK-2; Santa Cruz Biotechnology,

D-2), or Gal4 (Santa Cruz Biotechnology, RK5C1) at a final concentration

of 100 ng/mL, washed, and probed with the appropriate secondary antibodyconjugated to horseradish peroxidase (Santa Cruz Biotechnology) diluted

1:10,000. Membranes were immersed in chemiluminescence reagents

(Pierce) and exposed to Kodak XAR film for signal detection.

Results

Differential requirement of TAU5 for ligand-dependent andligand-independent AR activity. Our laboratory has beenstudying the LNCaP/C4-2 model of prostate cancer progressionto study the mechanisms of AR transcriptional regulation inandrogen-dependent and ADI prostate cancer cells (8, 21). The ADIC4-2 cell line was derived from the androgen-dependent LNCaPcell line through serial xenografting in castrated hosts (22). A keyfeature of this model is that LNCaP cells display ligand-dependentmRNA expression of the AR-regulated PSA gene, whereas C4-2 cellsdisplay constitutive PSA mRNA expression that can be furtherenhanced by androgens (Fig. 1A). Experiments using siRNA-mediated AR knock down and ChIP have shown that constitutivePSA expression is due to constitutive, ligand-independent ARtranscriptional activity on the PSA promoter in C4-2 cells (9, 10).

Cancer Research

Cancer Res 2007; 67: (20). October 15, 2007 10068 www.aacrjournals.org

Research. on June 16, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

We recently described a promoter tethering system to study thestructural and functional domains required by the AR to mediatethis ligand-independent activity (8). This strategy involves swap-ping androgen response elements (ARE) in a PSA-based promoterreporter construct [PSAenh(ARE)-LUC] with binding sites for theyeast Gal4 transcription factor [PSAenh(GAL4)-LUC] and concur-rently swapping the zinc-finger AR DBD with the zinc-finger DBDof Gal4 to create chimeric hARGal4 (Fig. 1B, left). We have shownthat these constructs serve as a sensitive monitoring system of ARactivity in AR-expressing prostate cancer cells, which functionsindependent of the endogenous AR. Using this system, we showed

that constitutive AR activity is independent of the AR AF-2 domainin C4-2 cells (8). Due to these observations, we have focused ourattention on transcriptional activation domains within theunstructured AR NTD that may be responsible for mediatingligand-independent AR activity in ADI prostate cancer cells. Aregion of the AR NTD, encompassing amino acids 361 to 490, waspreviously termed TAU5 by virtue of its importance for full ligand-induced activity of ectopic AR in AR-null HeLa cells (15). To assessthe role of TAU5 in mediating AR activity, we tethered wild-typeand TAU5-deleted (DTAU5) versions of hARGal4 to PSAenh(GAL4)-LUC. GAL4 substitution of AREs in the PSA-based reporter

Figure 1. AR TAU5 plays a differential role in mediating ligand-dependent and ligand-independent AR activity in a cell-based model of prostate cancer progression.A, LNCaP and C4-2 cells were treated for 24 h with 1 nmol/L mibolerone (Mib ) or vehicle control (EtOH, ethanol) in serum-free medium. PSA mRNA levels weredetermined by Northern blot. GAPDH mRNA levels and ethidium bromide staining of rRNA subunits are shown as controls. AR protein expression was concurrentlydetermined by Western blot. ERK-2 protein expression is shown as a control. B, left, schematic of a Gal4-based AR tethering system to allow AR functionalstudies in AR-expressing cells. Modularity of the AR protein is indicated, as well as the location of TAU5. The hinge (h ) region between the DBD and LBD is indicated;all other domains are described in the text. Right, LNCaP and C4-2 cells were transfected with ARE-driven or GAL4-driven, PSA-based reporter constructsdepicted at left along with wild-type (WT ) ARGal4 or ARGal4 DTAU5 as indicated. Cells were grown in serum-free medium and treated with 1 nmol/L mibolerone or ethanolfor 24 h. Luciferase activity was determined. Columns, mean from at least three independent experiments, each done in duplicate; bars, SE. Values are shownrelative to the activity of the GAL4-based reporter construct in the absence of androgens and transactivator, which was arbitrarily set to 1. C, LNCaP and C4-2cells were transfected with MMTV-Luc, nontargeted control (ctrl ) or AR-targeted siRNAs, and wild-type or TAU5-deleted (DTAU5) versions of AR harboring silentmutations in the siRNA target site (denoted ARsr) as indicated. Luciferase activity was determined as described for B . Lysates were analyzed by Western blot forAR protein expression. ERK-2 protein expression is shown as a control. The ligand-dependent (LNCaP) and ligand-independent (C4-2) activity of endogenous ARon the MMTV promoter was arbitrarily set to 100%. D, 22Rv1 cells were transfected as described for C . Luciferase activity, AR protein expression, and ERK-2protein expression were determined as described for C . Values are shown relative to the ligand-independent activity of hARsr, which was arbitrarily set to 100%.

WxxLF Mediates Ligand-Independent AR Activity

www.aacrjournals.org 10069 Cancer Res 2007; 67: (20). October 15, 2007

Research. on June 16, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

construct completely abolished androgen-induced promoter activ-ity in both LNCaP and C4-2 cells (Fig. 1B). Importantly, GAL4substitution of AREs also inhibited androgen-independent PSApromoter activity 4-fold, only in C4-2 cells (Fig. 1B ; Supplementary

Fig. S1). In LNCaP cells, hARGal4 DTAU5 displayed over 2-foldhigher androgen-induced transcriptional activity than wild-type hARGal4 (Fig. 1B). Conversely, DTAU5 hARGal4 displayed over2-fold lower androgen-independent transcriptional activity when

Figure 2. TAU5 is required for ligand-hypersensitive AR activity in ADI prostate cancer cells. A, LNCaP and C4-2 cells were transfected with wild-type andDTAU5 versions of ARGal4 in conjunction with PSAenh(GAL4)-LUC. Cells were treated under serum-free conditions with 2 pmol/L to 0.2 nmol/L dihydrotestosterone(DHT ) for 24 h. Luciferase activity was determined. Points, mean from at least three independent experiments, each done in duplicate; bars, SE. The activity ofwild-type hARGal4 in response to 0.2 nmol/L dihydrotestosterone was arbitrarily set to 100%. B, endogenous AR expression was knocked down in LNCaP and C4-2cells via AR-targeted siRNA. Cells were cotransfected with constructs encoding wild-type and DTAU5 versions of siRNA-resistant (ARsr) along with MMTV-LUC asindicated. Cells were treated and luciferase activity was determined as described for A . C, LNCaP and C4-2 cells were transfected with PSAenh(GAL4)-LUC along withwild-type, CTD-deleted (NTDGal4), or CTD/TAU5-deleted (NTDGal4 DTAU5) versions of ARGal4 as indicated and treated with 1 nmol/L mibolerone or vehiclecontrol (ethanol) for 24 h. Luciferase activity was determined as described for A . Values are shown relative to the activity of the GAL4-based reporter construct inthe absence of androgens and transactivator, which was arbitrarily set to 1. Lysates were analyzed by Western blot for AR protein expression. The discreteprotein species recognized by the NTD-targeted AR antibody are indicated.

Cancer Research

Cancer Res 2007; 67: (20). October 15, 2007 10070 www.aacrjournals.org

Research. on June 16, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

compared with wild-type hARGal4 in C4-2 cells (Fig. 1B). Despitethis observation, deletion of TAU5 had no effect on the full level ofligand-induced hARGal4 activity in C4-2 cells (Fig. 1B). These resultsshow that TAU5 deletion selectively impairs ligand-independenthARGal4 transcriptional activity in ADI C4-2 cells.

To ensure that these findings did not represent an artifact ofchimeric hARGal4, and were not restricted to a PSA-based reporterconstruct, we used an AR replacement strategy. To this end, wegenerated wild-type and DTAU5 forms of wild-type hAR that wereresistant to an AR-targeted siRNA (hARsr) and tested their abilitiesto stimulate a MMTV promoter in LNCaP and C4-2 cells (Fig. 1C).AR-targeted siRNA completely attenuated androgen responsivenessof the MMTV promoter in LNCaP cells, but had no effect on ligand-independent MMTV promoter activity (Fig. 1C). Consistent withthis finding, expression of hARsr restored androgen-dependent

MMTV activity in LNCaP cells (Fig. 1C). Conversely, AR-targetedsiRNA inhibited ligand-independent MMTV promoter activity byover 60% in C4-2 cells, which was restored upon expression ofhARsr (Fig. 1C). In addition, as observed in Gal4 tetheringexperiments (Fig. 1B), TAU5 deletion impaired ligand-independent,but not ligand-dependent hARsr activity in C4-2 cells (Fig. 1C ;Supplementary Fig. S2).

To examine whether TAU5 plays a role in mediating aberrant ARactivity in other, non–LNCaP-based models of ADI prostate cancer,we used the ADI 22Rv1 prostate cancer cell line, which was derivedfrom a CWR22 prostate cancer xenograft that relapsed after hostcastration-induced regression (23). In contrast to the T877Amutant AR expressed in LNCaP-derived cells, 22Rv1 cells harboran AR with an in-frame duplication of exon 3, resulting in an extrasecond zinc finger in the AR DBD (24). Similar to C4-2 cells, we

Figure 3. A 435WHTLF439 motif within the TAU5 domain is selectively required for ligand-independent AR activity in ADI prostate cancer cells. A, amino acidsequence of AR TAU5. Propensity for a-helical (h ) or h-sheet (e) secondary structure was determined using the nnpredict algorithm. B, C4-2 cells were transfectedwith wild-type, D393/420, and D420/449 versions of ARGal4 in conjunction with PSAenh(GAL4)-LUC. Cells were maintained 48 h in the absence of androgens.Luciferase activity was determined. Columns, mean from at least three independent experiments, each done in duplicate; bars, SE. Values are shown relative tothe activity of the GAL4-based reporter construct in the absence of androgens and transactivator, which was arbitrarily set to 1. C, C4-2 cells were transfectedwith PSAenh(GAL4)-LUC and wild-type ARGal4 or versions of ARGal4 harboring single, double, or triple alanine substitutions at W435, L438, and F439 as indicated.The relative locations of W425, L438, and F439 are depicted in the context of a helical wheel. Luciferase activity was determined as described for B . Values areshown relative to the activity of wild-type hARGal4 in the absence of androgens and transactivator, which was arbitrarily set to 100% (*P V 0.01; **P V 0.0001). D, LNCaPand C4-2 cells were transfected with MMTV-Luc and nontargeted control or AR-targeted siRNAs as indicated. Wild-type or 435AHTAA439 versions of AR were expressedfrom vectors harboring silent mutations in the siRNA target site (denoted ARsr) as indicated. Cells were treated under serum-free conditions with 1 nmol/L miboleroneor ethanol for 24 h. Luciferase activity was determined as described for B . The ligand-dependent (LNCaP) and ligand-independent (C4-2) activity of hARsr was arbitrarilyset to 100%. Lysates were analyzed by Western blot for AR protein expression. ERK-2 protein expression is shown as a control.

WxxLF Mediates Ligand-Independent AR Activity

www.aacrjournals.org 10071 Cancer Res 2007; 67: (20). October 15, 2007

Research. on June 16, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

determined that 22Rv1 cells displayed constitutively high PSAmRNA expression that resulted from ligand- and AF-2–indepen-dent AR activity (Supplementary Fig. S3). Indeed, AR-targetedsiRNA inhibited androgen-independent MMTV activity in 22Rv1cells (Fig. 1D). Moreover, similar to C4-2 cells, hARsr TAU5 deletionimpaired androgen-independent MMTV activity f50% (Fig. 1D).Together, our findings show that TAU5 plays a selective role inmediating ligand-independent AR activity in ADI prostate cancercells.TAU5 is required for AR ligand hypersensitivity in ADI

prostate cancer cells. Our data indicate that TAU5 could play a

previously unrecognized role in mediating ligand-independent ARactivation in ADI prostate cancer cells. However, although acomplete androgen-depleted environment can be achieved in vitro ,such a situation may not arise in vivo . For example, data from arapid autopsy study showed that, despite androgen ablation,intraprostatic androgen levels in prostate cancer patients persist atlevels sufficient to weakly transactivate the AR (25). Therefore, weexplored the role of TAU5 in the AR transcriptional response tocastrate levels of its natural ligand, dihydrotestosterone. In LNCaPcells, the transcriptional activity of hARGal4 DTAU5 was higherthan hARGal4 at dihydrotestosterone concentrations higher than

Figure 4. The AR TAU5 domain and 435WHTLF439

motif are required for ligand-independent transcriptionalactivation of the endogenous PSA gene in C4-2cells. A, schematic of the plasmid used for ARreplacement experiments. Key features of thisplasmid include EGFP expression regulated by theSV40 promoter, AR shRNA expression regulated bythe histone H1 promoter, and hARsr expressionregulated by the cytomegalovirus (CMV ) promoter.B, C4-2 cells were transfected with versions of theAR replacement plasmid containing the indicatedfeatures. Cells were maintained under serum-freeconditions for 48 h, and a sample was lysed foranalysis of AR expression by Western blot. ERK-2protein expression is shown as a control. EGFP-positivecells were collected by fluorescence-activated cellsorting, reseeded, and cultured in androgen-freemedium for an additional 48 h. Cells were harvestedand total RNA was isolated. RNA was subjected totwo-step quantitative reverse transcription-PCR usingprimers specific for PSA and GAPDH. Results wereanalyzed using the DC t method. Columns, mean fromthree experiments; bars, SE.

Cancer Research

Cancer Res 2007; 67: (20). October 15, 2007 10072 www.aacrjournals.org

Research. on June 16, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Figure 5. The AR 435WHTLF439 motif mediatesligand-independent TAU5 transcriptional activity.A, C4-2 cells were transfected with wild-type,DTAU5, and AHTAA mutant versions of hARGal4,as well as DTAU5 versions of hARGal4 with21-amino-acid inserts encompassing wild-type orAHTAA mutant versions of the 435WHTLF439

domain as indicated. PSAenh(GAL4)-LUC wasused as a reporter for these experiments. Cellswere maintained 48 h in the absence ofandrogens. Luciferase activity was determined.Columns, mean from at least three independentexperiments, each done in duplicate; bars, SE.Values are shown relative to the activity of theGAL4-based reporter construct in the absence ofandrogens and transactivator, which wasarbitrarily set to 1. Lysates were analyzed byWestern blot for hARGal4 protein expression withGal4-specific antibodies. B, C4-2 cells weretransfected with wild-type, DTAU5, or AHTAAversions of hARGal4 along with PSAenh(GAL4)-LUC. Cells were grown under androgen-freeconditions, cross-linked, lysed, and subjected toChIP with antibodies specific for Gal4 ornonspecific IgG. Immunoprecipitated DNAfragments were amplified by PCR using primersspecific for the PSAenh(GAL4)-LUC reporteras indicated in the schematic on the right. C,C4-2 cells were transfected with wild-type orDTAU5 versions of hARGal4, or DTAU5 versionsof hARGal4 harboring one, two, or three copiesof a 21-amino-acid insert encompassing the ARWHTLF motif. PSAenh(GAL4)-LUC was used asa reporter for these experiments. Assays weredone as described for A . D, LNCaP cells weretransfected with hARGal4-based constructs asdescribed for A and B . Cells were treated underserum-free conditions with 1 nmol/L miboleroneor ethanol for 24 h (*P V 0.05).

WxxLF Mediates Ligand-Independent AR Activity

www.aacrjournals.org 10073 Cancer Res 2007; 67: (20). October 15, 2007

Research. on June 16, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

0.1 nmol/L (Fig. 2A). Conversely, in C4-2 cells, hARGal4 DTAU5transcriptional activity was f50% lower than wild-type hARGal4

transcriptional activity over all dihydrotestosterone concentrationsstudied (Fig. 2A). When these experiments were done using an ARreplacement strategy and MMTV-LUC as a reporter, a similarrelationship was observed (Fig. 2B). In LNCaP cells, DTAU5 hARsr

activity was higher than wild-type hARsr at dihydrotestosteroneconcentrations higher than 0.1 nmol/L (Fig. 2B). However, in C4-2cells, hARsr displayed higher activity than hARsr DTAU5 at

dihydrotestosterone concentrations lower than 10 pmol/L(Fig. 2B). At concentrations higher than 0.1 nmol/L dihydrotestos-terone, TAU5 activity was not apparent in C4-2 cells (Fig. 2B).Together, these results suggest that TAU5 plays an important rolein mediating AR activation, only under conditions of no/lowandrogens in ADI prostate cancer cells.

A consistent observation was that treatment of C4-2 cells with1 nmol/L androgens abolished TAU5 activity in C4-2 cells (Figs. 1Band 2B ; Supplementary Fig. S2), suggesting that full AR activity

Figure 6. AR 435WHTLF439 represents anovel ligand-independent transactivationmotif. A, C4-2 cells were transfectedwith wild-type or AF-2 mutant (V716R,K720A, E897K) versions of hARGal4,DTAU5hARGal4, and DTAU5 hARGal4 with a21-amino-acid insert encompassing the ARWHTLFmotif and a wild-type or mutant AF-2(V716R, K720A, E897K) domain asindicated. PSAenh(GAL4)-LUC was usedas a reporter for these experiments. Cellswere maintained for 48 h in the absence ofandrogens. Luciferase activity wasdetermined. Columns, mean from at leastthree independent experiments, eachdone induplicate; bars, SE. Values are shownrelative to the activity of the GAL4-basedreporter construct in the absence ofandrogens and transactivator, which wasarbitrarily set to 1. Lysates were analyzed byWestern blot for hARGal4 protein expressionwith Gal4-specific antibodies. B, LNCaP andC4-2 cells were transfected with fusionsbetween the Gal4 DBD and one, two,or three copies of a 21-amino-acid insertencompassing wild-type or AHTAA mutantversions of the 435WHTLF439 domain asindicated. pG5-LUC was used as a reporterfor these experiments. Cellswere transfectedand luciferase activity determined asdescribed for A .

Cancer Research

Cancer Res 2007; 67: (20). October 15, 2007 10074 www.aacrjournals.org

Research. on June 16, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

does not require TAU5. Previous studies have shown that deletionof the AR COOH-terminal domain (CTD) results in full ARactivation through relieved inhibition of the AR NTD (8, 15–17).Therefore, we assessed the effect of TAU5 deletion in the context ofa truncated version of hARGal4 (NTDGal4; Fig. 2C). In both LNCaPand C4-2 cells, NTDGal4 was constitutively active and did notrespond to androgens (Fig. 2C). Deletion of TAU5 further increasedNTDGal4 activity in both cell lines, confirming that full ARactivation does not require TAU5 activity (Fig. 2C). Together, thesedata show that TAU5 is inactive or inhibitory when the AR is fullyactivated.NTD 435WHTLF439 is an important TAU5 motif that

selectively mediates ligand-independent AR activity. Ourresults suggest the existence of a transactivation domain withinAR TAU5 that could account for f50% of the ligand-independentand/or ligand-hypersensitive AR activity in ADI prostate cancercells. We hypothesized that patches of rigid secondary structurewithin this highly disordered domain could serve as importantprotein interaction sites. The neural network nnpredict secondarystructure prediction algorithm (26) was therefore applied to theamino acid sequence representing AR TAU5. Two potential sites ofextended secondary structure were identified (Fig. 3A). To testwhether either of these regions of putative secondary structure wasimportant for ligand-independent AR activity in C4-2 cells, eachwas independently deleted from hARGal4. Deletion of amino acids420 to 449, but not amino acids 393 to 420, inhibited ligand-independent hARGal4 transcriptional activity to a similar degree asdeletion of the entire TAU5 domain (Fig. 3B). Within this 420 to 449region, a span of predicted secondary structure encompassed435WHTLF439 (Fig. 3A), which was originally described as a shorthelical motif that can mediate androgen-dependent interactionwith AF-2, the COOH-terminal AR coactivator binding surface(27–29). X-ray crystallography studies have shown that peptidescontaining a core WxxLF sequence adopt a helical conformation(29), which, as shown by a helical wheel (Fig. 3C, inset), wouldresult in a hydrophobic surface created by the nonpolar side chainsof W435, L438, and F439. To test the potential role of thesehydrophobic residues within AR 435WHTLF439 in regulating ligand-independent AR transcriptional activity in ADI C4-2 cells, thehARGal4-based tethering system was used (Fig. 3C). When thenonpolar W435, L438, and F439 residues were individuallysubstituted with alanine, no effect on androgen-independenthARGal4 transcriptional activity was observed (Fig. 3C). However,a W435A/L438A/F439A compound mutation, which eliminated allbulky hydrophobic side chains within 435WHTLF439, elicited anapproximate 40% drop in androgen-independent hARGal4 tran-scriptional activity (Fig. 3C). These results suggest that AR435WHTLF439 could mediate selective TAU5 activity in ADI prostatecancer cells. To further test this possibility, we assessed the ligand-dependent and ligand-independent activities of wild-type andAHTAA mutant hARsr in AR replacement experiments. WhereashARsr ATHAA exhibited reduced androgen-independent activity inC4-2 cells, there was no effect on ligand-dependent activity inLNCaP cells (Fig. 3D).

To test whether deletion of the TAU5 domain or mutation of theWHTLF motif also impaired ligand-independent AR activity in thecontext of chromatin, we transfected C4-2 cells with an ARreplacement vector encoding EGFP, AR-targeted shRNA, and wild-type, deletion, or mutant versions of hARsr (Fig. 4A). This approachled to effective replacement of the endogenous AR with ectopicversions of hARsr in transfected C4-2 cells (Fig. 4B). Importantly, in

EGFP-positive cells, DTAU5 and AHTAA mutant versions of hARsr

both displayed a 40% to 50% reduction in ligand-independenttranscriptional activation of the endogenous PSA gene comparedwith wild-type hARsr (Fig. 4C), thus confirming reporter-basedfindings.

To directly assess the relationship between TAU5 and435WHTLF439, the transcriptional activities of DTAU5 and AHTAAversions of hARGal4 were compared via promoter tethering assay inC4-2 cells (Fig. 5A). As shown previously, both TAU5 deletion andAHTAA mutation impaired hARGal4 ligand-independent transcrip-tional activity f50% (Fig. 5A). Importantly, a 21-amino-acidpeptide containing the core 435WHTLF439, but not AHTAA, rescuedfull ligand-independent transcriptional activity of hARGal4DTAU5 inC4-2 cells (Fig. 5A). ChIP analysis confirmed that wild-type, DTAU5,and AHTAA versions of ARGal4 effectively engaged with theircognate GAL4 DNA binding sites in these assays (Fig. 5B). Theseresults show that the AR 435WHTLF439 motif plays a direct role inmediating TAU5 activity in ADI C4-2 cells.The AR 435WHTLF439 motif functions as a transcriptional

activation domain. Our results suggest that AR WHTLF couldrepresent a novel NH2-terminal AR transactivation domain.Alternatively, aberrant AR activation could be facilitated bystabilization of the AR in the absence of ligand via an aberrantN/C interaction between 435WHTLF439 and AF-2. Indeed, structuraland functional studies have shown that ligand-bound AR can bestabilized by an interaction between the AR AF-2 domain and23FQNLF27 or 435WHTLF439 motifs in the AR NTD (27, 29–33). Todifferentiate between these scenarios, we added one, two, or three435WHTLF439-containing peptides to hARGal4 DTAU5. Stepwiseincreases in ligand-independent hARGal4 transcriptional activitywere observed in C4-2 cells with each 435WHTLF439-containingpeptide inserted (Fig. 5C). When we did similar experiments inLNCaP cells, we were surprised to observe that insertion of two orthree 435WHTLF439-containing peptides in hARGal4 DTAU5 resultedin ligand-dependent superactivation of the AR (Fig. 5D).

Although these results were consistent with a transactivationrole for 435WHTLF439, they did not rule out the possibility thatmultiple copies of 435WHTLF439 were mediating more efficientinteractions with AF-2, resulting in stronger N/C stabilization andsubsequent higher levels of AR activity. Therefore, we nextabolished AF-2 function by incorporating three separate mutationsin V716, K720, and E897, which are AR residues that comprise theconserved nuclear receptor AF-2 charge-clamp and mediatebinding with AR 435WHTLF439 (28, 29). Mutations in thesecharge-clamp residues impair ligand-dependent AR transcriptionalactivity in LNCaP as well as other cell lines (8, 28, 30, 32, 34).However, in C4-2 cells, mutations in these charge-clamp residuesdid not disrupt the ligand-independent activity of wild-typehARGal4, nor hARGal4 DTAU5 with a 21-amino-acid insert harboringthe AR 435WHTLF439 motif (Fig. 6A). These data argue against arole for 435WHTLF439 in mediating an aberrant N/C interaction inADI prostate cancer cells, and strongly suggest that 435WHTLF439

represents a novel AR transactivation domain.Finally, to directly assess a role for the AR 435WHTLF439 motif in

transcriptional activation, one, two, or three copies of an AR-derived peptide with a core 435WHTLF439 sequence were tetheredto the Gal4 DBD and tested for their ability to stimulatetranscription from a GAL4-regulated luciferase reporter. In bothLNCaP and C4-2 cells, a Gal4 DBD containing two 435WHTLF439

peptides mediated transcriptional activation (Fig. 6B). Of particularnote, three tandem copies of 435WHTLF439 peptides activated

WxxLF Mediates Ligand-Independent AR Activity

www.aacrjournals.org 10075 Cancer Res 2007; 67: (20). October 15, 2007

Research. on June 16, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

transcription of the GAL4-driven promoter 600-fold in both celllines. We thus conclude that the AR 435WHTLF439 motif is a noveltranscriptional activation domain. Although this motif has thepotential to stimulate transcription in both LNCaP and C4-2 cells(Figs. 5C and 6B), our data indicate that in the context of full-length AR, 435WHTLF439 selectively mediates transcriptionalactivation under no/low androgen conditions in ADI prostatecancer cells.

Discussion

The structures of the LBD and AF-2 folds of the AR have beendetermined by X-ray crystallography studies of peptides contain-ing the AR CTD (29, 30, 35–37). However, the structure of the ARNTD is unknown, and the precise identity of functional domainswithin this region of the AR is currently limited. The AR NTDpossesses potent transactivation potential, which is generallyreferred to by the all-encompassing term AF-1 (14). Conversely,AR AF-2 in isolation is a relatively weak transcriptional activationdomain (32, 34, 38). Therefore, AR AF-1 is thought to be the majordomain responsible for mediating AR transcriptional activity. Theprimary role for AF-2 may be to regulate this dominant AF-1activity, in part by mediating a direct intramolecular interactionwith FxxLF and/or WxxLF motifs in the NTD (27, 28, 31, 33, 39).TAU1 and TAU5 were identified originally as transactivation unitswithin the AR NTD after functional deletion analysis of wild-typeand CTD-deleted AR in AR-null COS-1 and HeLa cells (15).Subsequent studies defined the core sequence 178LKDIL182,resident within a discrete NTD helix, as the motif that activelymediates AR TAU1 transcriptional activity (16, 17). Theimportance of TAU5 has not been firmly established, due toconflicting reports describing TAU5 as either necessary (15–17)or dispensable (40, 41) for full AR transcriptional activity.Notably, the majority of these conflicting studies have been donein AR-null cell lines of nonhuman origin.

This study represents the first examination of TAU5 function inAR-dependent prostate cancer cells, where the AR is a disease-relevant therapeutic target. Using different promoters as reportersfor a Gal4-based promoter tethering strategy as well as an ARreplacement strategy, we showed a striking dichotomy of TAU5function; TAU5 deletion enhanced ligand-induced AR transcrip-tional activity in androgen-dependent prostate cancer cells, butinhibited ligand-independent AR transcriptional activity in ADIprostate cancer cells. We further showed that TAU5 deletioninhibited AR transcriptional activation over a range of castratedihydrotestosterone concentrations in ADI C4-2 cells. However, fullactivation of the AR, via saturation with synthetic androgen orCOOH-terminal deletion, abolished this TAU5 requirement.Through deletion and mutagenesis analysis, we identified a short435WHTLF439 sequence as a putative transcriptional activationmotif within TAU5. Indeed, when tested in functional assays,mutation of this motif to AHTAA selectively inhibited ligand-independent AR activity in C4-2 cells, but not ligand-dependent ARactivity in LNCaP cells. Moreover, short 21-amino-acid peptidesencompassing this 435WHTLF439 motif, but not a mutant AHTAAmotif, could mediate strong transcriptional activation in isolationas well as rescue impaired ligand-independent transcriptionalactivity caused by TAU5 deletion in ADI C4-2 cells. These studiesthus provide a comprehensive molecular dissection of the AR TAU5domain and show that 435WHTLF439 is a novel AR transactivationmotif. In light of studies that have shown the 435WHTLF439 motif

can mediate an N/C interaction with AF-2 (27–29), a dual-functionrole exists for this domain of the AR. Interestingly, a similar dual-function role has been shown for the NTD AR 23FQNLF27 motif. Forexample, in addition to being the primary motif that binds AF-2(29, 33, 37), AR 23FQNLF27 is also able to bind directly to theX chromosome–linked melanoma antigen gene product MAGE-11 ,an androgen-dependent AR coactivator (42). Interestingly, the ARcoactivator GRIP1/TIF-2/SRC-2 has been shown via yeast two-hybrid assay to bind the AR TAU5 domain, which may serve tobridge an interaction between the NH2 and COOH termini of theAR (43, 44). However, our preliminary study has shown that siRNA-mediated knock down of GRIP1 has no effect on ligand-independent AR activity in C4-2 cells, which argues against a rolefor GRIP1 as a WHTLF-binding protein (data not shown).

In a previous study, we showed that the activity of hARGal4 inC4-2 cells was truly ligand independent, as mutations thatabolished LBD or AF-2 function did not significantly affect basaltranscriptional activity in C4-2 cells (8). Using this system, we haveconsistently observed that ligand-independent AR activity in ADIprostate cancer cells is much lower than AR activity stimulated bysaturating levels of androgens (3- to 4-fold versus >100-fold).However, comprehensive ChIP analysis has shown that the AR doestransiently localize to the PSA locus at low levels in the absence ofandrogens in C4-2 versus LNCaP cells (10). This transient ligand-independent localization triggers major locus-wide histone acety-lation and methylation, which significantly amplifies the overalltranscriptional output of the PSA gene (10). It has also beenproposed that a positive feedback loop exists between the AR andits target loci under ligand-free conditions in ADI prostate cancercells, whereby AR-dependent alterations in the chromatin statefacilitate subsequent AR binding and transcriptional activation(10). These mechanisms are likely to underlie AR ligandhypersensitivity as well (10), a property that we have shown toalso require TAU5 (Fig. 2). Although our reporter-based studies donot take into account effects at the chromatin level (i.e., the‘‘amplification’’), they do provide a sensitive measure of ARtranscriptional activity (i.e., the ‘‘trigger’’). This is confirmed byour observation that DTAU5 and AHTAA mutant versions of theAR were impaired in their ability to mediate ligand-independenttranscriptional activation of the endogenous PSA gene in C4-2 cellscompared with wild-type AR (Fig. 4). Our current data thus addsubstantially to previous studies using this cell-based system ofprostate cancer progression, and shows that aberrant 435WHTLF439

activity could be critical for AR-mediated triggering of AR targetgenes in ADI prostate cancer cells.

An important question arising from these studies is how ligand-independent 435WHTLF439 transcriptional activity is prevented inLNCaP cells. Indeed, short peptides containing 435WHTLF439

displayed a similar level of autonomous transcriptional activity inLNCaP and C4-2 cells (Fig. 5B). The finding that TAU5 deletionresulted in nearly 2-fold higher ligand-dependent AR activity inLNCaP cells indicates that negative regulatory element(s) existwithin this domain, and may be dominant over the transactivatingactivity of the 435WHTLF439 motif (Fig. 1B and C). We are currentlyexploring the role and identity of this putative domain in regulatingAR activity in prostate cancer cells. Our results also highlight thepossibility of negative regulatory domain(s) existing outside ofTAU5, because in the absence of TAU5, a single 435WHTLF439-containing peptide was not able to significantly elevate ligand-dependent AR transcriptional activity in a promoter tetheringassay in LNCaP cells (Fig. 4C). However, multiple copies of

Cancer Research

Cancer Res 2007; 67: (20). October 15, 2007 10076 www.aacrjournals.org

Research. on June 16, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

435WHTLF439-containing peptides inserted into TAU5-deleted ARresulted in ligand-dependent superactivation in LNCaP cells(Fig. 4C). These data indicate that multiple negative regulatoryelements, both inside and outside of TAU5, function to preventaberrant 435WHTLF439 activity in the absence of ligand. Therefore,we propose that the overall mechanism of AR activation in ADIprostate cancer is dependent on the adaptable and unorderednature of the NTD (13, 14), and a complex series of derepressionand activation events are necessary for the AR to achieve a criticalthreshold of transcriptional activity in ADI prostate cancer cells.Overall, our data show that the AR TAU5 domain in general, andthe 435WHTLF439 motif in particular, play an important role in thismechanism, accounting for f50% of aberrant AR activity observed

in ADI prostate cancer cells. Thus, 435WHTLF439 could represent anovel interface through which aberrant AR activity could betargeted for therapy of ADI prostate cancer.

Acknowledgments

Received 4/6/2007; revised 7/2/2007; accepted 8/1/2007.Grant support: NIH grants CA121277, DK065236, and CA91956 and the TJ Martell

Foundation (D.J. Tindall). S.M. Dehm is a research fellow of the Terry Fox Foundationthrough a grant from the National Cancer Institute of Canada.

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

We thank Dr. Frank Claessens for providing MMTV-LUC and Dr. Dan Billadeau forproviding pCMS4-H1p-EGFP, and Drs. Hannelore Heemers and Kaustubh Datta for thecritical evaluation of the manuscript.

WxxLF Mediates Ligand-Independent AR Activity

www.aacrjournals.org 10077 Cancer Res 2007; 67: (20). October 15, 2007

References1. Jemal A, Siegel R, Ward E, et al. Cancer statistics, 2006.CA Cancer J Clin 2006;56:106–30.2. Scherr D, Swindle PW, Scardino PT. National Com-prehensive Cancer Network guidelines for the manage-ment of prostate cancer. Urology 2003;61:14–24.3. Heinlein CA, Chang C. Androgen receptor in prostatecancer. Endocr Rev 2004;25:276–308.4. Roy-Burman P, Tindall DJ, Robins DM, et al. Andro-gens and prostate cancer: are the descriptors valid?Cancer Biol Ther 2005;4:4–5.5. Debes JD, Tindall DJ. Mechanisms of androgen-refractory prostate cancer. N Engl J Med 2004;351:1488–90.6. Agoulnik IU, Weigel NL. Androgen receptor action inhormone-dependent and recurrent prostate cancer.J Cell Biochem 2006;99:362–72.7. Bain DL, Heneghan AF, Connaghan-Jones KD, MiuraMT. Nuclear receptor structure: implications for func-tion. Annu Rev Physiol 2006;69:201–20.8. Dehm SM, Tindall DJ. Ligand-independent androgenreceptor activity is activation function-2-independentand resistant to antiandrogens in androgen refractoryprostate cancer cells. J Biol Chem 2006;281:27882–93.9. Yeung F, Li X, Ellett J, Trapman J, Kao C, Chung LW.Regions of prostate-specific antigen (PSA) promoterconfer androgen-independent expression of PSA inprostate cancer cells. J Biol Chem 2000;275:40846–55.10. Jia L, Shen HC, Wantroba M, et al. Locus-widechromatin remodeling and enhanced androgen recep-tor-mediated transcription in recurrent prostate tumorcells. Mol Cell Biol 2006;26:7331–41.11. Quayle SN, Mawji NR, Wang J, Sadar MD. Androgenreceptor decoy molecules block the growth of prostatecancer. Proc Natl Acad Sci U S A 2007;104:1331–6.12. Lavery DN, McEwan IJ. Structure and function ofsteroid receptor AF1 transactivation domains: inductionof active conformations. Biochem J 2005;391:449–64.13. Lavery DN, McEwan IJ. The human androgenreceptor AF1 transactivation domain: interactions withtranscription factor IIF and molten-globule-like struc-tural characteristics. Biochem Soc Trans 2006;34:1054–7.14. McEwan IJ. Molecular mechanisms of androgenreceptor-mediated gene regulation: structure-functionanalysis of the AF-1 domain. Endocr Relat Cancer 2004;11:281–93.15. Jenster G, van der Korput HA, Trapman J, BrinkmannAO. Identification of two transcription activation unitsin the N-terminal domain of the human androgenreceptor. J Biol Chem 1995;270:7341–6.16. Callewaert L, Van Tilborgh N, Claessens F. Interplaybetween two hormone-independent activation domainsin the androgen receptor. Cancer Res 2006;66:543–53.17. Chamberlain NL, Whitacre DC, Miesfeld RL.Delineation of two distinct type 1 activation functions

in the androgen receptor amino-terminal domain. J BiolChem 1996;271:26772–8.18. Debes JD, Comuzzi B, Schmidt LJ, Dehm SM, Culig Z,Tindall DJ. p300 regulates androgen receptor-indepen-dent expression of prostate-specific antigen in prostatecancer cells treated chronically with interleukin-6.Cancer Res 2005;65:5965–73.19. Chomczynski P, Sacchi N. Single-step method of RNAisolation by acid guanidinium thiocyanate-phenol-chlo-roform extraction. Anal Biochem 1987;162:156–9.20. Murillo H, Schmidt LJ, Karter M, et al. Prostatecancer cells use genetic and epigenetic mechanisms forprogression to androgen independence. Genes Chromo-somes Cancer 2006;45:702–16.21. Zegarra-Moro OL, Schmidt LJ, Huang H, Tindall DJ.Disruption of androgen receptor function inhibitsproliferation of androgen-refractory prostate cancercells. Cancer Res 2002;62:1008–13.22. Wu HC, Hsieh JT, Gleave ME, Brown NM, Pathak S,Chung LW. Derivation of androgen-independent humanLNCaP prostatic cancer cell sublines: role of bonestromal cells. Int J Cancer 1994;57:406–12.23. Sramkoski RM, Pretlow TG II, Giaconia JM, et al. Anew human prostate carcinoma cell line, 22Rv1. In VitroCell Dev Biol Anim 1999;35:403–9.24. Tepper CG, Boucher DL, Ryan PE, et al. Character-ization of a novel androgen receptor mutation in arelapsed CWR22 prostate cancer xenograft and cell line.Cancer Res 2002;62:6606–14.25. Mohler JL, Gregory CW, Ford OH III, et al. Theandrogen axis in recurrent prostate cancer. Clin CancerRes 2004;10:440–8.26. Kneller DG, Cohen FE, Langridge R. Improvements inprotein secondary structure prediction by an enhancedneural network. J Mol Biol 1990;214:171–82.27. He B, Kemppainen JA, Wilson EM. FXXLF andWXXLF sequences mediate the NH2-terminal interac-tion with the ligand binding domain of the androgenreceptor. J Biol Chem 2000;275:22986–94.28. He B, Lee LW, Minges JT, Wilson EM. Dependence ofselective gene activation on the androgen receptor NH2-and COOH-terminal interaction. J Biol Chem 2002;277:25631–9.29. Hur E, Pfaff SJ, Payne ES, Gron H, Buehrer BM,Fletterick RJ. Recognition and accommodation at theandrogen receptor coactivator binding interface. PLoSBiol 2004;2:E274.30. Estebanez-Perpina E, Moore JM, Mar E, et al. Themolecular mechanisms of coactivator utilization inligand-dependent transactivation by the androgenreceptor. J Biol Chem 2005;280:8060–8.31. Ikonen T, Palvimo JJ, Janne OA. Interaction betweenthe amino- and carboxyl-terminal regions of the ratandrogen receptor modulates transcriptional activityand is influenced by nuclear receptor coactivators. J BiolChem 1997;272:29821–8.

32. He B, Kemppainen JA, Voegel JJ, Gronemeyer H,Wilson EM. Activation function 2 in the humanandrogen receptor ligand binding domain mediatesinterdomain communication with the NH(2)-terminaldomain. J Biol Chem 1999;274:37219–25.33. He B, Bowen NT, Minges JT, Wilson EM. Androgen-induced NH2- and COOH-terminal interaction inhibitsp160 coactivator recruitment by activation function 2.J Biol Chem 2001;276:42293–301.34. Alen P, Claessens F, Verhoeven G, Rombauts W,Peeters B. The androgen receptor amino-terminaldomain plays a key role in p160 coactivator-stimulatedgene transcription. Mol Cell Biol 1999;19:6085–97.35. Matias PM, Donner P, Coelho R, et al. Structuralevidence for ligand specificity in the binding domainof the human androgen receptor. Implications forpathogenic gene mutations. J Biol Chem 2000;275:26164–71.36. Sack JS, Kish KF, Wang C, et al. Crystallographicstructures of the ligand-binding domains of theandrogen receptor and its T877A mutant complexedwith the natural agonist dihydrotestosterone. Proc NatlAcad Sci U S A 2001;98:4904–9.37. He B, Gampe RT, Jr., Kole AJ, et al. Structuralbasis for androgen receptor interdomain and coac-tivator interactions suggests a transition in nuclearreceptor activation function dominance. Mol Cell2004;16:425–38.38. Bevan CL, Hoare S, Claessens F, Heery DM, ParkerMG. The AF1 and AF2 domains of the androgenreceptor interact with distinct regions of SRC1. MolCell Biol 1999;19:8383–92.39. Klokk TI, Kurys P, Elbi C, et al. Ligand-specificdynamics of the androgen receptor at its responseelement in living cells. Mol Cell Biol 2006;5:1823–43.40. Christiaens V, Bevan CL, Callewaert L, et al.Characterization of the two coactivator-interactingsurfaces of the androgen receptor and their relativerole in transcriptional control. J Biol Chem 2002;277:49230–7.41. Metzger E, Muller JM, Ferrari S, Buettner R, Schule R.A novel inducible transactivation domain in theandrogen receptor: implications for PRK in prostatecancer. EMBO J 2003;22:270–80.42. Bai S, He B, Wilson EM. Melanoma antigen geneprotein MAGE-11 regulates androgen receptor functionby modulating the interdomain interaction. Mol CellBiol 2005;25:1238–57.43. Irvine RA, Ma H, Yu MC, Ross RK, Stallcup MR,Coetzee GA. Inhibition of p160-mediated coactivationwith increasing androgen receptor polyglutaminelength. Hum Mol Genet 2000;9:267–74.44. Shen HC, Buchanan G, Butler LM, et al. GRIP1mediates the interaction between the amino- andcarboxyl-termini of the androgen receptor. Biol Chem2005;386:69–74.

Research. on June 16, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

2007;67:10067-10077. Cancer Res   Scott M. Dehm, Kevin M. Regan, Lucy J. Schmidt, et al.   Independent Prostate Cancer Cells

−Androgen Receptor Activation in Androgen Depletion -Terminal WxxLF Motif for Aberrant2Selective Role of an NH

  Updated version

  http://cancerres.aacrjournals.org/content/67/20/10067

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2007/10/09/67.20.10067.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/67/20/10067.full#ref-list-1

This article cites 44 articles, 25 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/67/20/10067.full#related-urls

This article has been cited by 12 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected] at

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

  Permissions

  Rightslink site. (CCC)Click on "Request Permissions" which will take you to the Copyright Clearance Center's

.http://cancerres.aacrjournals.org/content/67/20/10067To request permission to re-use all or part of this article, use this link

Research. on June 16, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from