SOX9 Is Expressed in Normal Prostate Basal Cells and Regulates Androgen Receptor Expression in...

10
SOX9 Is Expressed in Normal Prostate Basal Cells and Regulates Androgen Receptor Expression in Prostate Cancer Cells Hongyun Wang, 1 Nicole C. McKnight, 1 Tao Zhang, 1 Michael L. Lu, 2 Steven P. Balk, 1 and Xin Yuan 1 1 Cancer Biology Program, Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School and 2 Urology Research Laboratory, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts Abstract SOX9 is a member of the SOX [S ry-related high-mobility group (HMG) box ] family of HMG DNA-binding domain transcription factors and is required for the development and differentiation of multiple cell lineages. This report shows that basal epithelial cells express SOX9 in normal prostate, with no detectable expression in luminal epithelial cells. In contrast, SOX9 is expressed in primary prostate cancers in vivo , at a higher frequency in recurrent prostate cancer and in prostate cancer cell lines (LNCaP, CWR22, PC3, and DU145). SOX9 message and protein levels in prostate cancer cells were increased by treatment with glycogen synthase kinase 3B inhibitor (SB415286), and SOX9 was reduced when B-catenin was down- regulated by small interfering RNA (siRNA), indicating that SOX9 expression in prostate cancer is regulated by Wnt/B- catenin signaling. SOX9 bound specifically to androgen receptor (AR) DNA-binding domain glutathione S -transferase fusion proteins, and this interaction was dependent on a short peptide immediately COOH-terminal to the DNA-binding domain (the C-terminal extension), which is required for interactions between steroid hormone receptors and the architectural HMG proteins. Exogenous SOX9 expressed at high nonphysiologic levels decreased AR expression and activity; however, at lower levels, SOX9 increased AR protein expression. Significantly, down-regulation of SOX9 by siRNA in prostate cancer cells reduced endogenous AR protein levels, and cell growth indicating that SOX9 contributes to AR regulation and decreased cellular proliferation. These results indicate that SOX9 in prostate basal cells supports the development and maintenance of the luminal epithelium and that a subset of prostate cancer cells may escape basal cell requirements through SOX9 expression. [Cancer Res 2007;67(2):528–36] Introduction The human prostate is composed of prostatic glands with well-defined basal and luminal epithelial cell layers, which are embedded in a fibromuscular stroma. Cells within the luminal epithelium have a very low rate of proliferation and express high levels of androgen receptor (AR). These cells are differentiated with a primary function of seminal fluid synthesis, a process that is strongly androgen dependent, and undergo atrophy or apoptosis in response to androgen withdrawal. In contrast, basal cells have a higher rate of proliferation, express low or undetectable levels of AR, and are not androgen dependent (1–3). The precise functions of basal cells are not clear, but accumulating evidence indicates that they play critical roles in prostate organogenesis and homeostasis. Basal cells form a continuous single-cell layer between the fibromuscular stroma and the luminal epithelium, thus providing support and a barrier for the luminal cells. The basal cell layer is also believed to contain prostatic stem cells, which can differentiate into basal, luminal, and neuroendocrine cells (4–10). Knockout of p63, a p53 family protein expressed selectively in basal cells, disrupts formation of a basal cell layer and development of prostate glands (11, 12). Moreover, p63-expressing basal cells are capable of repopulating the luminal cell layer (13). The basal cell layer becomes discontinuous in prostatic intra- epithelial neoplasia, which is believed to be a precancerous lesion. The complete loss of the basal cell layer is a defining feature of prostate cancer. Prostate cancer cells resemble luminal epithelial cells in their high-level expression of AR and androgen dependence, but they also express many protein markers characteristic of basal cells and clearly resemble basal cells with respect to increased proliferative capacity. One group of genes that may regulate aspects of prostate development and function are members of the SOX [S ry-related high-mobility group (HMG) box ] family. SOX proteins are a large family of transcription factors that share a homologous HMG DNA- binding domain and are key regulators of many developmental and tissue-specific processes (14). The HMG DNA-binding domain binds to DNA in the minor groove, and the architectural HMG proteins (HMG-1 and HMG-2) have been found to enhance steroid hormone receptor binding to DNA by locally altering DNA con- formation and through direct protein-protein interactions (15–17). We have similarly identified direct interactions between AR and the sequence-specific HMG proteins SRY and T-cell factor 4 (18, 19). SOX9 in the developing gonad plays a critical role in male sex determination by stimulating expression of anti-Mullerian hormone (AMH, also known as Mullerian-inhibiting substance), a transforming growth factor h–like hormone that causes regression of the female Mullerian ducts (20). SOX9 interacts with steroidogenic factor 1 on the Amh promoter to directly stimulate AMH expression. Moreover, loss of SOX9-mediated AMH produc- tion contributes to XY sex reversal, whereas increased SOX9 expression in XX male (Odsex) mice or transgenic mice is sufficient to cause a female to male reversal even in the absence of Sry (21). Conditional SOX9 knockout in the developing gonad shows that SOX9, expressed by Sertoli cells, is also essential for Sertoli cell differentiation and seminiferous tubule formation (22, 23). Requests for reprints: Xin Yuan, Hematology/Oncology Division, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215. Phone: 617-667- 5937; Fax: 617-667-0610; E-mail: [email protected]. I2007 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-06-1672 Cancer Res 2007; 67: (2). January 15, 2007 528 www.aacrjournals.org Research Article Research. on January 22, 2015. © 2007 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Transcript of SOX9 Is Expressed in Normal Prostate Basal Cells and Regulates Androgen Receptor Expression in...

SOX9 Is Expressed in Normal Prostate Basal Cells

and Regulates Androgen Receptor Expression

in Prostate Cancer Cells

Hongyun Wang,1Nicole C. McKnight,

1Tao Zhang,

1Michael L. Lu,

2

Steven P. Balk,1and Xin Yuan

1

1Cancer Biology Program, Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess MedicalCenter and Harvard Medical School and 2Urology Research Laboratory, Brigham and Women’s Hospital andHarvard Medical School, Boston, Massachusetts

Abstract

SOX9 is a member of the SOX [Sry-related high-mobility group(HMG) box] family of HMG DNA-binding domain transcriptionfactors and is required for the development and differentiationof multiple cell lineages. This report shows that basal epithelialcells express SOX9 in normal prostate, with no detectableexpression in luminal epithelial cells. In contrast, SOX9 isexpressed in primary prostate cancers in vivo, at a higherfrequency in recurrent prostate cancer and in prostate cancercell lines (LNCaP, CWR22, PC3, and DU145). SOX9 message andprotein levels in prostate cancer cells were increased bytreatment with glycogen synthase kinase 3B inhibitor(SB415286), and SOX9 was reduced when B-catenin was down-regulated by small interfering RNA (siRNA), indicating thatSOX9 expression in prostate cancer is regulated by Wnt/B-catenin signaling. SOX9 bound specifically to androgenreceptor (AR) DNA-binding domain glutathione S-transferasefusion proteins, and this interaction was dependent on a shortpeptide immediately COOH-terminal to the DNA-bindingdomain (the C-terminal extension), which is required forinteractions between steroid hormone receptors and thearchitectural HMG proteins. Exogenous SOX9 expressed at highnonphysiologic levels decreased AR expression and activity;however, at lower levels, SOX9 increased AR protein expression.Significantly, down-regulation of SOX9 by siRNA in prostatecancer cells reduced endogenous AR protein levels, and cellgrowth indicating that SOX9 contributes to AR regulation anddecreased cellular proliferation. These results indicate thatSOX9 in prostate basal cells supports the development andmaintenance of the luminal epithelium and that a subset ofprostate cancer cells may escape basal cell requirementsthrough SOX9 expression. [Cancer Res 2007;67(2):528–36]

Introduction

The human prostate is composed of prostatic glands withwell-defined basal and luminal epithelial cell layers, which areembedded in a fibromuscular stroma. Cells within the luminalepithelium have a very low rate of proliferation and express highlevels of androgen receptor (AR). These cells are differentiatedwith a primary function of seminal fluid synthesis, a process thatis strongly androgen dependent, and undergo atrophy or apoptosis

in response to androgen withdrawal. In contrast, basal cells havea higher rate of proliferation, express low or undetectable levelsof AR, and are not androgen dependent (1–3). The precisefunctions of basal cells are not clear, but accumulating evidenceindicates that they play critical roles in prostate organogenesisand homeostasis. Basal cells form a continuous single-cell layerbetween the fibromuscular stroma and the luminal epithelium,thus providing support and a barrier for the luminal cells. Thebasal cell layer is also believed to contain prostatic stem cells,which can differentiate into basal, luminal, and neuroendocrinecells (4–10). Knockout of p63, a p53 family protein expressedselectively in basal cells, disrupts formation of a basal cell layer anddevelopment of prostate glands (11, 12). Moreover, p63-expressingbasal cells are capable of repopulating the luminal cell layer (13).The basal cell layer becomes discontinuous in prostatic intra-epithelial neoplasia, which is believed to be a precancerous lesion.The complete loss of the basal cell layer is a defining feature ofprostate cancer. Prostate cancer cells resemble luminal epithelialcells in their high-level expression of AR and androgen dependence,but they also express many protein markers characteristic of basalcells and clearly resemble basal cells with respect to increasedproliferative capacity.One group of genes that may regulate aspects of prostate

development and function are members of the SOX [Sry-relatedhigh-mobility group (HMG) box] family. SOX proteins are a largefamily of transcription factors that share a homologous HMG DNA-binding domain and are key regulators of many developmental andtissue-specific processes (14). The HMG DNA-binding domainbinds to DNA in the minor groove, and the architectural HMGproteins (HMG-1 and HMG-2) have been found to enhance steroidhormone receptor binding to DNA by locally altering DNA con-formation and through direct protein-protein interactions (15–17).We have similarly identified direct interactions between AR andthe sequence-specific HMG proteins SRY and T-cell factor 4(18, 19). SOX9 in the developing gonad plays a critical role in malesex determination by stimulating expression of anti-Mullerianhormone (AMH, also known as Mullerian-inhibiting substance),a transforming growth factor h–like hormone that causesregression of the female Mullerian ducts (20). SOX9 interacts withsteroidogenic factor 1 on the Amh promoter to directly stimulateAMH expression. Moreover, loss of SOX9-mediated AMH produc-tion contributes to XY sex reversal, whereas increased SOX9expression in XX male (Odsex) mice or transgenic mice is sufficientto cause a female to male reversal even in the absence of Sry (21).Conditional SOX9 knockout in the developing gonad shows thatSOX9, expressed by Sertoli cells, is also essential for Sertoli celldifferentiation and seminiferous tubule formation (22, 23).

Requests for reprints: Xin Yuan, Hematology/Oncology Division, Beth IsraelDeaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215. Phone: 617-667-5937; Fax: 617-667-0610; E-mail: [email protected].

I2007 American Association for Cancer Research.doi:10.1158/0008-5472.CAN-06-1672

Cancer Res 2007; 67: (2). January 15, 2007 528 www.aacrjournals.org

Research Article

Research. on January 22, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

SOX9 is expressed in multiple other tissues during embryogen-esis, including cartilage, neural crest, notochord, kidney, pancreas,and endocardial cushions of the heart. Heterozygous SOX9mutations are the cause of the human disease campomelicdysplasia, a form of dwarfism characterized by extreme cartilageand bone malformation, which is frequently associated with XY sexreversal and other anomalies (24–27). Homozygous knockout ofSOX9 in mice results in embryonic lethality, whereas SOX9heterozygous knockouts exhibit the same skeletal anomalies ascampomelic dysplasia patients (28, 29). The tissue-specificinactivation of SOX9 in limbs results in the absence of cartilageand bone formation, whereas overexpression of SOX9 induceschondrogenic cell differentiation, revealing that SOX9 is requiredfor cartilage development (28–30). Consistent with these defects,major SOX9 target genes in cartilage cells include type II collagen(Col2a1), type XI collagen (Col11a2), and aggrecan, which are allimportant components of cartilage (29, 31).SOX9 knockout in neural stem cells results in defects in

specification of oligodendrocytes and astrocytes, indicating thatthe switch from neurogenesis to gliogenesis fails to take place(32–34). In the intestine, SOX9 is expressed in the crypt, which ispopulated by progenitor/stem cells. Moreover, SOX9 expression inintestinal crypt cells is regulated by the Wnt/h-catenin signalingpathway, which is required to maintain the stem cell compartment(35, 36). SOX9 is also found in the outer root sheath compartmentof hair follicles, where its expression is regulated by sonic hedgehogsignaling (37). SOX9 conditional knockout results in hair loss andabsence of the stem cell compartment, further supporting a criticalrole for SOX9 in maintaining stem cells.In this study, we show that SOX9 in normal adult prostate is

expressed in basal epithelial cells, with no detectable expressionin luminal epithelium. SOX9 was also expressed in a subset ofprimary prostate cancer in vivo , at increased frequency in recurrentprostate cancer, and in prostate cancer cell lines, and its expressionin prostate cancer cells was regulated by the Wnt/h-cateninpathway. Similarly to other HMG proteins, SOX9 interacted with theAR and this interaction was dependent on the C-terminal extension(CTE) in the AR DNA-binding domain. Transient transfections anddoxycycline-inducible expression of SOX9 in LNCaP prostate cancercells showed that SOX9 expressed at very high nonphysiologic levelscould suppress AR protein expression, but SOX9 expressed at lowerlevels enhanced AR protein expression. Moreover, small interferingRNA (siRNA)–mediated down-regulation of SOX9 in prostate cancercells caused a decline in AR protein levels and suppressed cellgrowth. Taken together, these observations indicate that SOX9expressed in normal prostate basal cells may play roles inmaintaining a committed stem cell/progenitor cell compartment,or in regulating the expression of factors that support the luminalepithelium. Moreover, the expression of SOX9 protein in prostatecancer cells may be critical to maintain proliferative potential andgrowth independently of basal cells.

Materials and Methods

Plasmids, reagents, and cell lines. The pcDNA3-SOX9 plasmid, whichexpresses a Flag-tagged SOX9, was a gift from Dr. P. Berta (Human

Molecular Genetics Group, Institut de Genetique Humane, Montpellier,

France; ref. 20). The SOX9-regulated luciferase reporter (4x48-p89-luciferase)was provided by Dr. B. de Crombrugghe (Department of Molecular

Genetics, University of Texas MD Anderson Cancer Center, Houston, TX;

ref. 38). AR expression vectors (pSVARo), ARE4-luciferase reporter, and

Renilla luciferase control vector (pRL-CMV, Promega, Madison, WI) have

been described previously (18). pcDNA6/TR and pcDNA4/TO/myc-His werepurchased from Invitrogen (Carlsbad, CA). pcDNA4/TO/SOX9 was gener-

ated by inserting a BamHI-BamHI fragment from pcDNA3-SOX9 into the

BamHI site of pcDNA4/TO/myc-His.

SB415286, a glycogen synthase kinase 3h (GSK3h) inhibitor, was

purchased from Tocris Cookson (Avonmouth, Bristol, United Kingdom).

Antibodies were from the following sources: anti-SOX9 (H90, C20 from

Santa Cruz Biotechnology, Santa Cruz, CA, and 09-1, a kind gift from

Dr. M. Wegner, Institut fur Biochemie, Emil-Fischer-Zentrum, Universitat

Erlangen, Erlangen, Germany; ref. 32), anti–prostate-specific antigen (PSA;

Biodesign, Saco, ME), anti–h-tubulin (Chemicon, Temecula, CA), anti–h-catenin (Sigma, St. Louis, MO), anti-AR (Upstate Biotechnology, Lake Placid,

NY), anti-p27 (Santa Cruz Biotechnology), and anti-glyceraldehyde-3-

phosphate dehydrogenase (GAPDH; Abcam, Cambridge, MA). Secondary

anti-mouse and anti-rabbit antibodies were from Promega.CV-1, LNCaP, PC3, and C3H10T cells were obtained from American Type

Culture Collection (Manassas, VA). LNCaP cells were maintained in RPMI

with 10% fetal bovine serum (FBS). PC3 and C3H10T cells were maintained

in DMEM with 10% FBS. CWR22R3 cells were established by our laboratory

from a CWR22 xenograft that relapsed after castration and bicalutamide

treatment, and have been cultured long term (>2 years) in DMEM supple-

mented with 10% charcoal-dextran–treated FBS (39). CWR22Rv1 cells

were from Dr. R.M. Sramkoski (Cancer Research Center, Case Western

Reserve University, Cleveland, OH) and were maintained in DMEMwith 10%

FBS (40). RCS cells, a rat chondrosarcoma cell line, were a gift from Dr. de

Crombrugghe and were maintained in DMEMwith 10% FBS. C4-2 was a gift

from Dr. L. Chung (Department of Urology, Emory, University School of

Medicine, Atlanta, GA) andweremaintained in Tmedium (Life Technologies,

Inc., Gaithersburg, MD) supplemented with 5% FBS. Primary epithelial cells

were purchased from Clonetics Corporation (San Diego, CA) and were

maintained using the suggested prostate epithelial cell growth medium.

Immunohistochemistry. Five-micrometer sections from paraffin-

embedded tissue blocks were deparaffinized, rehydrated, and underwent

antigen retrieval by microwaving at high setting for 30 min in 10 mmol/Lcitrate buffer (pH 6.2). After cooling to room temperature, the tissue

sections were blocked using 5% goat serum and avidin blocking solution

(Vector, Burlingame, CA). Primary antibodies were then added andincubated overnight at 4jC. The anti-AR or anti-SOX9 antibodies were

used at 1:50. After four washes in PBST (PBS with 0.05% Tween 20), the

antibodies were detected using biotinylated goat anti-rabbit antibody at

1:400 followed by streptavidin-horseradish peroxidase (HRP) at 1:400(Vector). After an additional four washes with PBST, slides were developed

with 3,3¶-diaminobenzidine and counterstained with hematoxylin.Immunoblotting. Prostatectomy samples were excised and minced in

PBS into 1-mm3 pieces, homogenized with a glass Dounce homogenizer,and sonicated in radioimmunoprecipitation assay (RIPA) lysis buffer

[50 mmol/L Tris-HCl (pH 8.0), 150 mmol/L sodium chloride, 1.0% NP40,

0.5% sodium deoxycholate, 0.1% SDS, 1 mmol/L EDTA, and 1 mmol/LEGTA] containing protease and phosphatase inhibitors. Cultured cells were

directly lysed with RIPA buffer containing protease and phosphatase

inhibitors (antipain hydrochloride, 100 Amol/L; aprotinin, 0.2 Ag/mL;AEBSF, 1 mmol/L; E-64, 10 Amol/L; leupeptin hemisulfate, 100 Amol/L;pepstatin, 1 Amol/L; glycerol phosphate, 1 mmol/L; NaPPO4, 1 mmol/L; andsodium vanadate, 1 mmol/L). Protein quantity was determined by Bradford

assay (Bio-Rad Laboratories, Inc., Hercules, CA). Proteins were separated

by SDS-PAGE under reducing conditions and then transferred to 0.45-Amnitrocellulose membranes by electroblotting. The membranes were blocked

with 5% nonfat powdered milk in PBS and then probed with primary

antibodies at a 1:1,000 dilution in TBS containing 0.2% Tween 20 (TBST)with 5% milk. The membranes were then washed extensively with TBST and

probed with HRP-conjugated secondary antibodies at 1:2,000 dilutions in

TBST with 5% milk. After further washing in TBST, the membranes were

developed with enhanced chemiluminescence Western blotting detectionsystem (Pierce Biotech, Rockford, IL). The Image J program (Wayne

Rasband, NIH, Bethesda, MD) was used to quantify protein band densities

on immunoblots according to author’s instructions, which were normalized

to h-tubulin or GAPDH and expressed as relative density.

SOX9 in Human Prostate

www.aacrjournals.org 529 Cancer Res 2007; 67: (2). January 15, 2007

Research. on January 22, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Dephosphorylation with calf intestinal phosphatase. CWR22Rv1 cellswere plated in a six-well plate the day before collection. At the time of

collection, the cells were washed once with cold PBS and scraped with a

rubber policeman into passive lysis buffer (Promega) supplemented with

protease inhibitors (1 Ag/mL aprotinin, 1 Ag/mL pepstatin, 0.5 Ag/mLleupeptin, 0.2 mg/mL AEBSF, and 1.6 mg/mL iodoacetamide). Lysates

were incubated with rotation for 20 min at room temperature and cleared

by centrifugation for 15 min in a microcentrifuge at 4jC. Forty microlitersof cleared cell lysates with or without added calf intestinal phosphatase(40 units, New England Biolabs) were incubated at 37jC for 5 h. The treatedlysates were subsequently immunoblotted for SOX9 protein.

Transfection. One day before transfection, cells were plated into 24- or48-well plates at a density of 70% to 80%. The cells were transfected withmixtures of DNA and LipofectAMINE 2000 (Invitrogen) for 24 h, according

to the manufacturer’s recommendations. Cells were then switched to fresh

medium containing various treatment reagents for another 24 h and thenlysed with passive lysis buffer and analyzed for luciferase activity using

the Dual-luciferase measurement system (Promega). The siRNAs were

similarly transfected at 40 pmol/mL using LipofectAMINE 2000, and the

cells were studied at 48 to 72 h posttransfection. The following siRNAs werepurchased from Dharmacon (Lafayette, CO). h-Catenin siRNAs were a

mixture of four different siRNA duplexes (M-003482-00). SOX9 siRNAs were

as follows (positive strands): A, 5¶-GCAGCGACGUCAUCUCCAAUU-3¶; B,5¶-CAACGAGUUUGACCAGUACUU-3¶; and control siRNA (siCONTROL Non-Targeting siRNA 1, D-001210-01).

Real-time reverse transcription-PCR. Total RNA was isolated from

cultured cells using the RNeasy protect mini kit (Qiagen, Valencia, CA). Theamount of total RNA was determined by spectrophotometer, and 100 ng of

total RNA from each sample were used to determine the specific RNA level

by TaqMan real-time reverse transcription-PCR (RT-PCR) using an ABI

Prism 7000 (Applied Biosystems, Foster City, CA). The primer sequences forhuman SOX9 were as follows: 5¶-TATGACTGGACCCTGGTG-3¶ ( forward);5¶-TGTGGCTTGTTCTTGCTGG-3¶ (reverse) and 5¶-FAM-TGCCGGTG-CGCGTCAACG-3¶ (probe). The primers for human AR were as follows:

5¶-GGAATTCCTGTGCATGAAA-3¶ ( forward); 5¶-CGAAGTTCATCAAAGA-ATT-3¶ (reverse); and 5¶-FAM-CTTCAGCATTATTCCAGTG-3¶ (probe). Thevalidated 18S human rRNA assay primer/probe set was purchased from

Applied Biosystems. Relative quantitation with the comparative thresholdcycle (C t) method was done as recommended by ABI. The C t is the

fractional cycle number at which the amplified target reaches a fixedthreshold. The amount of target gene normalized to an endogenous

reference gene (18S rRNA) is given by 2�DC t, where DC t is C (target gene) �C t (reference gene).

Establishment of LNCaP cell lines expressing inducible SOX9. TheT-REx system (Invitrogen) was used to generate LNCaP cells that can be

induced to express human SOX9 or SOX9-specific short hairpin RNA

(shRNA). Two LNCaP clones that constitutively express high levels of

tetracycline repressor were first established. Each cell line showed similarresponsiveness as their parental cells to dihydrotestosterone (DHT), as

indicated by the PSA level changes after androgen withdrawal or

stimulation. SOX9-inducible lines were subsequently established by

selecting clones that carry the pcDNA4/TO/SOX9. SOX9 shRNA–induciblelines were established by transduction with SOX9 shRNA in the tetracycline-

regulated pSuperior vector (pSuperior-SOX9i, generated by cloning the

above SOX9 siRNA A sequence into pSuperior), followed by puromycinselection.

Glutathione S-transferase pull down. DNA sequences encoding

various AR protein fragments were generated by PCR and cloned into

the pGEX-2T vectors. Glutathione S-transferase (GST)–AR fusion werepurified on glutathione-agarose beads and used to pull down 35S-labeled,

in vitro transcribed and translated SOX9 or SRY proteins, as previously

described (18).

Cell proliferation and cell cycle analysis. Cells were rinsed in PBS,trypsinized, washed thrice with PBS, and fixed with 95% ethanol at 4jC for30 min. The cells were then treated with propidium iodide and RNase A at

37jC for 30 min and subsequently analyzed by flow cytometry. The resultswere analyzed using CellQuest-Pro software. Cell proliferation was studied

with the cell proliferation ELISA, bromodeoxyuridine (BrdU) (colorimetric)

kit from Roche Applied Science, according the manufacturer’s recommen-

dations. Briefly, CWR22Rv1 or C4-2 cells were plated in 96-well plates at 5 �103/well and transfected with 5 pmol of SOX9 siRNAs (A or B) or control

siRNA. At the 48 h posttransfection, cells were labeled with 100 Amol/LBrdUrd for 24 h. After labeling, the cells were fixed with FixDenat solution at

room temperature for 30 min and then incubated with anti–BrdU-PODantibody for 90 min. The cells were subsequently washed thrice with

washing solution and incubated with substrate solution at room

temperature for 5 to 10 min before the plate was read in an ELISA readerat 490 nm.

Figure 1. SOX9 protein is expressed innormal prostate basal cells and in prostatecancer. A, prostatectomy samples wereimmunostained with anti-SOX9 (H90 orO9-1), or anti-AR antibodies, as indicated.Bottom right, mouse testis stained withH90 anti-SOX9 antibody. B, left,immunoblot of protein extracts from threeprostatectomy samples (samples 1–3 ) withthe C20 anti-SOX9 antibody; extract fromRCS (a rat chondrosarcoma cell line) wasblotted as a positive control and GAPDHwas blotted for a protein loading control.Right, immunoblot of primary epithelialcells (PrEC ) with three different anti-SOX9antibodies (09-1, H90, and C20),with RCS as a positive control andGAPDH for a protein loading control.C, immunohistochemical staining of threeprimary prostate cancers from radicalprostatectomies with H90 anti-SOX9or anti-AR antibodies, as indicated.D, sections from two locally recurrentprostate cancers stained for SOX9.

Cancer Research

Cancer Res 2007; 67: (2). January 15, 2007 530 www.aacrjournals.org

Research. on January 22, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Results

SOX9 protein is expressed by basal cells in normal prostate.Transcripts encoding a number of SOX proteins, such SRY, SOX7,and SOX9, have been reported in prostate, with further dataindicating that SRY loss may contribute to prostate cancer (41–46).However, our immunohistochemical studies have not identified cellsthat express detectable levels of SRY protein in adult humanprostate, despite the use of several anti-SRY antibodies and positivestaining of SRY in human testis (data not shown). In contrast,immunohistochemistry with two independent anti-SOX9 antibodiesshowed strong nuclear and weaker cytoplasmic expression of SOX9in the basal cell layer of normal prostate glands, with no staining inluminal epithelial cells (Fig. 1A). This SOX9 expression pattern isopposite that of AR, which is highly expressed in the luminalepithelium and is weakly expressed or absent in basal epithelial cells(Fig. 1A). SOX9 expression in testis was predominantly in the Sertolicells, consistent with its previously reported pattern and supportingthe specificity of the SOX9 immunohistochemistry (Fig. 1A).We next carried out immunoblotting to confirm that SOX9

protein was expressed in adult prostate. As shown in Fig. 1B (left),extracts from three independent histologically normal frozenhuman radical prostatectomy blocks clearly contained a proteinthat migrated as a doublet with the same mobility as SOX9 fromRCS cells (a rat chondrosarcoma cell line expressing high levelsof SOX9). To further assess SOX9 expression in normal prostatebasal cells, we examined primary epithelial cells cultured shortterm from human prostate. These cells are phenotypically basalcells that express a basal cell pattern of cytokeratins and arenegative for AR and PSA proteins (47). Immunoblotting againshowed a doublet that migrated identically to the RCS SOX9protein (Fig. 1B, right). Significantly, three independent SOX9antibodies against different regions of the protein recognized thisprotein, which supports the conclusion that it is SOX9.SOX9 is expressed in prostate cancer in vivo and in prostate

cancer cell lines. Although SOX9 expression was not detected byimmunohistochemistry in normal prostate luminal epithelium, itcould be clearly observed in prostate cancer samples (Fig. 1C). Thestaining in prostate cancer was predominantly nuclear and variedin intensity and distribution from patchy to diffusely positive. In aseries of randomly collected primary prostate cancer samples fromradical prostatectomies (n = 29), tumor cells with clear nuclearSOX9 expression were readily found in 17 cases. The variation inSOX9 staining did not seem to reflect differences in fixation, asthe negative tumor samples still showed basal cell SOX9 staining inthe adjacent normal glands (Fig. 1C, bottom left) and strong nuclearAR staining in the tumor (Fig. 1C, bottom right). Interestingly, SOX9

expression was further increased in a series transurethral resectionof the prostate samples from patients with recurrent prostatecancer after androgen deprivation therapy (n = 37), with >90% ofthese tumors having SOX9-positive tumor cells (Fig. 1D). Moreover,a larger fraction of the tumor cells were SOX9 positive in theserecurrent tumors versus the initial primary tumors, and thisdifference was statistically significant (Table 1). These results showthat SOX9 is expressed by a large fraction of primary prostatecancer, and indicate that there is positive selection for increasedSOX9 expression in prostate cancer that relapse subsequent toandrogen deprivation therapy.Based on these results, SOX9 expression in a series of prostate

cancer cell lines was next assessed by immunoblotting. Signifi-cantly, each of the cell lines examined (CWR22R3, CWR22Rv1,C4-2, LNCaP, and PC3) expressed readily detectable levels of SOX9,which migrated identically to SOX9 in RCS cells and C3H10T cells(a mouse mesenchymal line reported to express SOX9; Fig. 2A).As an approach to further confirm SOX9 expression in the prostatecancer cells, we used siRNA to down-regulate SOX9. Expressionof SOX9, as assessed by immunoblotting in CWR22Rv1 cells, wasmarkedly reduced by both of the siRNAs against SOX9, but not bya control siRNA (Fig. 2A, bottom left). Finally, the migration ofSOX9 as a doublet on most gels suggested that it was phos-phorylated, as has been previously reported (48). Consistent withthis hypothesis, SOX9 ran as a faster migrating single band aftertreatment with calf intestinal phosphatase (Fig. 2A, bottom right).Taken together, these results show that tumor cells express SOX9in a large fraction of prostate cancer. However, SOX9 expression isconfined to basal cells in normal prostate.SOX9 expression in prostate cancer cells is modulated by

Wnt/B-catenin signaling. Recent studies indicate that SOX9expression in intestinal crypts and hair follicles is required tomaintain stem cell compartments, and that SOX9 expression inthese sites is regulated by Wnt or Shh signaling, which are theimportant pathways for stem cell maintenance (35, 37). Althoughthe identity of stem cells for prostate epithelium remains unclear,the prostate basal cell layer seems to contain precursor cells thatcan give rise to luminal epithelial cells. These observationssuggested that SOX9 expression may be similarly regulated bythe Wnt/h-catenin pathway and may contribute to maintainingproliferative potential in normal prostate and in prostate cancer.Therefore, we next examined whether the Wnt/h-catenin pathwayregulates SOX9 expression in prostate cancer cells.Wnt signaling inhibits GSK3h-mediated h-catenin phosphoryla-

tion, reducing h-catenin ubiquitylation and proteasome degrada-tion and resulting in an accumulation of h-catenin in the nucleus

Table 1. SOX9 expression in primary and locally recurrent prostate cancer

Prostate cancer (n) No. tumors with the indicated % of tumor cells staining positive for SOX9

0–25% 26–50% 51–75% 76–100%

Primary (29) 17 1 3 8

Recurrent (39) 4 6 16 13

NOTE: The percentages of positive cells (tumor cells positive for SOX9 nuclear staining) as a continuous variable for each tumor were compared usingWilcoxon rank sum test. The percentages of positive cells were significantly lower in primary prostate cancer than locally recurrent prostate cancer

(median 25% versus 75%, respectively, P = 0.0006). Of the 17 primary tumors with 0% to 25% staining, 12 were negative for SOX9 expression.

SOX9 in Human Prostate

www.aacrjournals.org 531 Cancer Res 2007; 67: (2). January 15, 2007

Research. on January 22, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

and transactivation of its target genes (49). Akt also inhibitsGSK3h, so that h-catenin is stabilized in PTEN-deficient prostatecancer cells (including PC3, LNCaP, and C4-2, which were derivedfrom LNCaP) due to activation of the phosphatidylinositol 3-kinase/Akt pathway (50, 51). To stimulate the Wnt/h-cateninpathway in prostate cancer cells with intact PTEN, we treated theDU145 prostate cancer cell line with a direct GSK3h inhibitor,SB415286, which caused a marked and dose-dependent increasein h-catenin protein levels (Fig. 2B). Significantly, SB415286 alsostrongly increased the expression of SOX9 protein, consistent withWnt/h-catenin regulation of SOX9 expression. Similarly, inhibitionof GSK3h by SB415286 in CWR22Rv1 cells led to an increase inh-catenin and SOX9 protein levels (Fig. 2B).We next used quantitative real-time RT-PCR to determine

whether SOX9 message levels were increased in response to GSK3hinhibition by SB415286. As shown in Fig. 2C , SB415286 caused anincrease in SOX9 message levels at 6 h, with a further markedincrease at 24 h, which indicates that Wnt signaling increases SOX9transcription. Finally, as GSK3h inhibition can modulate manyproteins in addition to h-catenin, we used h-catenin siRNA todetermine whether SOX9 expression was regulated by h-catenin.Transfection with h-catenin siRNA caused a marked reduction of h-catenin protein levels in CWR22Rv1 cells compared with controlsiRNA (Fig. 2D). Significantly, there was a corresponding decrease inSOX9 protein, which indicates that SOX9 was positively regulated byh-catenin. Taken together, these results show that SOX9 is a Wnt/h-catenin pathway–regulated gene in prostate cancer cells.SOX9 directly interacts with AR. We previously reported that

SRY and AR could interact through their respective DNA-bindingdomains, and that SRY could suppress AR transcriptional activity(18). Given the homology between the HMG DNA-binding domainsof SRY and SOX9, we tested whether there was also an interactionbetween SOX9 and AR by using a series of GST-AR DNA-binding

domain fusion proteins. As shown in Fig. 3A , in vitro transcribed/translated SOX9 could bind specifically to the GST-AR DNA-binding domain fusion proteins. Significantly, SOX9 binding toGST-AR DNA-binding domain was markedly reduced when eightamino acids immediately after the second zinc finger in AR DNA-binding domain were absent (removing amino acids 629–636).This eight-amino-acid sequence was previously identified as a partof the CTE that is critical for steroid hormone receptors, such asAR and progesterone receptor, to interact with the HMG-1 andHMG-2 proteins (Fig. 3C ; refs. 15–17, 52). The observation thatSOX9 binding was abrogated by deletion of this CTE from the ARDNA-binding domain fusion protein indicates that SOX9 interactswith AR by a mechanism similar to the HMG proteins. In contrast,SRY binding to GST-AR DNA-binding domain was more efficientand was not affected by removing the eight amino acids of CTE,suggesting that additional sites on SRY might be involved in ARbinding (Fig. 3B).SOX9 interaction with AR was next assessed functionally in

transient transfections experiments. SOX9 is a transcription factorwith a COOH-terminal transactivation domain, and SOX9 trans-fection stimulated expression of a reporter gene regulated by amultimerized SOX9 binding site (Fig. 4A). In contrast, SOX9transfection inhibited DHT-stimulated AR transactivation of anandrogen-responsive element–regulated reporter gene (Fig. 4B).This inhibition was not a nonspecific effect, as DHT-independentactivity or activity of an internal control cytomegalovirus-regulatedRenilla luciferase reporter were not repressed (Fig. 4B and data notshown). Significantly, immunoblotting showed that AR proteinexpression was decreased at the high levels of SOX9 that repressedAR transcriptional activity (>0.8 ng), whereas lower amounts ofSOX9 seemed to moderately enhance AR protein expression(0.16–0.8 ng; Fig. 4C). In contrast, SOX9 had no effect on a controltransfected protein [green fluorescent protein (GFP)]. Additional

Figure 2. SOX9 expression in prostate cancer cell lines and modulation by Wnt/h-catenin signaling. A, extracts from prostate cancer cell lines were blotted withC20 anti-SOX9 antibody, with extracts from RCS and C3H10T cells as positive controls and GAPDH as a protein loading control. Left bottom, lysates from CWR22Rv1blotted with the O9-1 anti-SOX9 antibody at 72 h posttransfection with control (C ) or two SOX9-specific siRNAs (SOX9i A and B). Right bottom, SOX9 immunoblotof lysates from control or calf intestinal phosphatase (CIP )–treated CWR22Rv1 cells. B, DU145 or CWR22Rv1 cells treated with 50 Amol/L SB415286 (SB415 )or carrier (DMSO) for 24 h were immunoblotted for h-catenin and SOX9. C, SOX9 message levels in CWR22Rv1 cells after 6 or 24 h of SB415286 treatmentwere analyzed by quantitative real-time RT-PCR, with the levels presented as the fold change (relative level) compared with cells without SB415286 treatment.D, CWR22Rv1 cells were transfected with a control (C ) or h-catenin–targeted siRNAs for 72 h, and cell lysates were blotted for h-catenin and SOX9. GAPDH was usedas a protein loading control. Columns, ratios of h-catenin and SOX9 to GAPDH.

Cancer Research

Cancer Res 2007; 67: (2). January 15, 2007 532 www.aacrjournals.org

Research. on January 22, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

transfection experiments confirmed this biphasic response, withlower levels of transfected SOX9 causing an increase in AR proteinexpression (Fig. 4D). These results support the hypothesis that ARand SOX9 can interact, although the down-regulation of AR activityat very high levels of SOX9 (due to decreased AR protein or possiblysquelching) is probably not a physiologic function.SOX9 positively regulates endogenous AR expression in

prostate cancer cells. Based on the above transfection results,we next examined the effect of SOX9 on endogenous AR proteinin prostate cancer cells. For these experiments, we first establishedLNCaP cell lines expressing tetracycline-inducible SOX9. Theinduction of SOX9 expression was very rapid, with substantialamounts of SOX9 detected by 2 to 3 h after doxycycline treatment(Fig. 5A). Only the much lower level of endogenous SOX9 wasdetected when cells were not treated with doxycycline, or in thecontrol cell line that was similarly established with a control vector.Consistent with the transient transfection results, AR proteinwas modestly reduced after 3 to 6 h when the cells expressed highSOX9 levels, but then recovered after 18 h despite the continuedhigh levels of SOX9 (Fig. 5A). We next used real-time RT-PCRto determine whether SOX9 was regulating AR message levels. Asshown in Fig. 5A (bottom), AR message levels did not decline inresponse to SOX9 induction, indicating that the decrease in ARprotein was due to decreased translation or increased degradation.Interestingly, AR message levels increased after f18 h, and thiscorresponded to the recovery in AR protein levels.Although these findings confirmed that very high levels of SOX9

could decrease AR, the level of SOX9 in these cells after induction isclearly nonphysiologic. Therefore, to determine whether SOX9 atphysiologic levels is a regulator of AR expression in prostate cancercells, we tested whether SOX9 down-regulation by shRNA had aneffect on AR levels in LNCaP cells. For this experiment, we usedLNCaP cells expressing the tetracycline repressor and stablytransduced them with pSuperior-SOX9i, expressing doxycycline-inducible SOX9 shRNA. As shown in Fig. 5B , doxycycline induction

resulted in a marked decrease (f10-fold) in the level of SOX9protein. Significantly, there was also a decrease in AR expression(f40%) and a corresponding decrease in expression of the AR-regulated PSA protein (Fig. 5B).We similarly examined the CWR22Rv1 prostate cancer cell line,

which expresses AR and higher levels of SOX9 than LNCaP cells(although it does not express detectable levels of PSA protein).Transfections with two different SOX9 siRNAs (A and B), but nota control siRNA (C), caused a marked decline in SOX9 protein, witha corresponding decline of f40% to 80% in AR protein expression(Fig. 5C). Finally, real-time RT-PCR was used to determine whetherAR message levels were decreased in response to the SOX9knockdowns. Significantly, no decline in AR message was observedusing either of the SOX9 siRNAs (Fig. 5D). Taken together, theseresults indicate that endogenous SOX9 enhances AR proteinexpression in prostate cancer cell lines through translational orposttranslational mechanisms.SOX9 down-regulation inhibits prostate cancer cell prolif-

eration. The persistent expression of SOX9 in prostate cancer cell

Figure 4. Exogenous SOX9 modulates AR protein levels in prostate cancercells. A, SOX9 is transcriptionally active. CV-1 cells were transfected with4x48-p89-luc reporter (SOX-Luc reporter; 50 ng), pRL-CMV (0.1 ng), andpcDNA3-SOX9 as indicated and cultured for 48 h posttransfection. Columns,luciferase activities [expressed as relative light units (RLU )] normalized for theinternal control Renilla activity. B, SOX9 suppresses AR transcriptional activity.CV-1 cells were transfected with pSVARo (50 ng), ARE4-Luc reporter (50 ng),pRL-CMV (0.1 ng), and pcDNA3-SOX9 as indicated. At 24 h, the cells wereswitched to DMEM with 10% CDS-FBS with (+) or without (�) 10 nmol/L DHTand cultured for an additional 24 h. Columns, luciferase activities (expressedas relative light units) normalized for the internal control Renilla activity.C, CV-1 cells were transfected as in (B ), except that each transfectionmixture also included 5 ng of GFP-expressing vector. The cells were lysed at48 h posttransfection and blotted for AR, SOX9, or GFP. D, CV-1 cells weresimilarly analyzed as in (C ) except that transfection was done with a lower rangeof SOX9 expression vector. h-Tubulin was blotted for a protein loading control.Columns, AR and SOX9 normalized to h-tubulin for each sample.

Figure 3. SOX9 directly interacts with AR. 35S-labeled in vitro transcribed/translated SOX9 (A ) or SRY (B ) were incubated with glutathione-agarose beadsbound to GST or GST-AR DNA-binding domain fusion proteins (DBD ; numbersabove the blot, AR fragments used). Top, autoradiographs of GST pulldown;bottom, Coomassie blue–stained protein gels to quantitate the GST/GST-fusionproteins. *, the expected full-length fusion proteins. Columns, the amount ofSOX9/SRY pulled down by each protein normalized to the input. RD, relativedensity. C, schematic of the AR regions expressed as GST fusion proteins.N-ter, portion of AR NH2 terminus; Hinge, portion of 3¶-hinge.

SOX9 in Human Prostate

www.aacrjournals.org 533 Cancer Res 2007; 67: (2). January 15, 2007

Research. on January 22, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

lines indicated that it might play a role in supporting cell growthin vitro as well as in vivo . As an initial approach to assess theimportance of SOX9, we determined whether transient SOX9 down-regulation by siRNA had effects on proliferation. Significantly,SOX9 siRNAs caused a decrease in proliferation in both C4-2 and

CWR22Rv1 cells, as assessed by BrdU incorporation (Fig. 6Aand B). Consistent with the decreased BrdU incorporation, cellcycle analysis in CWR22Rv1 cells showed an increase in the G0-G1population at 48 and 72 h, and a corresponding decrease in theS-G2-M fraction (Fig. 6C). Moreover, immunoblotting showed asubstantial increase in expression of the p27 cyclin–dependentkinase inhibitor (Fig. 6D), consistent with a block at G0-G1 andfurther supporting the hypothesis that SOX9 enhances prostatecancer cell proliferation.

Discussion

Previous studies have established critical roles for SOX9 in thedevelopment of multiple tissues and in maintaining the stem cellcompartments in adult tissues. In this study, we found that SOX9was primarily expressed in basal epithelial cells in adult prostate,with no detectable expression in luminal epithelium. In contrast,immunohistochemistry showed that SOX9 was expressed in asubset of primary prostate cancer and was expressed at higherfrequency in recurrent prostate cancer. The expression of SOX9 inprostate cancer cells was stimulated by suppression of GSK3h andrepressed by h-catenin silencing, indicating that the Wnt/h-cateninpathway regulates SOX9 in prostate cancer. Similar to other HMGproteins, SOX9 interacted with the AR DNA-binding domain.

Figure 5. SOX9 modulates endogenous AR protein expression in prostatecancer cell lines. A, inducible SOX9-expressing (SOX9 ) and nonexpressingcontrol (Control ) LNCaP cells were treated with 0.5 Ag/mL doxycycline for theindicated times, and lysates were blotted for AR or SOX9, with h-tubulin as aprotein loading control. Bottom, AR message levels measured by real-timeRT-PCR, with the levels presented as the fold change compared with cellswithout doxycycline treatment. *, significant differences. B, LNCaP cellsexpressing inducible SOX9 shRNA were cultured for 72 h (� or + doxycycline)and lysates were then immunoblotted for SOX9, AR, PSA, and h-tubulin.Columns, relative density of each band (normalized to h-tubulin). C, CWR22Rv1cells were transfected with two SOX9-targeted siRNAs (SOX9i A and B ) or acontrol siRNA (C ). Cell lysates were collected at 72 h posttransfection andimmunoblotted for AR, SOX9, and h-tubulin. Columns, relative density of AR andSOX9 versus h-tubulin. D, CWR22Rv1 cells were transfected as above andtotal RNA was collected at the indicated times for assessment of AR messagelevels by quantitative real-time RT-PCR. Figure 6. SOX9 down-regulation blocks prostate cancer cell cycle progression.

A and B, C4-2 and CWR22Rv1 cells transfected for 48 h with two SOX9-targetedsiRNAs (SOX9i A or B) or a control siRNA (C ) as indicated were assessedfor BrdU uptake over 24 h by ELISA. *, significant differences. C, CWR22Rv1cells transfected as in (B ) were collected at the indicated time pointsposttransfection and analyzed for cell cycling by propidium iodide staining andflow cytometry. D, cell lysates were collected at 72 h posttransfection as in(C ) and immunoblotted for SOX9 and p27. Columns, SOX9 and AR normalizedfor h-tubulin.

Cancer Research

Cancer Res 2007; 67: (2). January 15, 2007 534 www.aacrjournals.org

Research. on January 22, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Although a high level of overexpression of SOX9 in transienttransfections or in LNCaP prostate cancer cells could suppress ARprotein expression, SOX9 expressed at lower physiologic levelsenhanced AR protein expression. More significantly, SOX9 siRNAcaused a decline in AR protein levels without decreasing ARmessage levels, indicating that SOX9 can regulate AR expressionthrough a posttranscriptional mechanism. Finally, siRNA down-regulation of SOX9 caused a decrease in proliferation and anincrease in p27 expression, indicating that SOX9 has a role insupporting prostate cancer cell growth. Taken together, theseobservations indicate that SOX9 in normal prostate may play rolesin maintaining the proliferative potential of basal cells or insupporting the luminal epithelium, and that these functions maybe important for prostate cancer.Although SOX9 is critical for maintaining the stem cell

compartment in other tissues and may function similarly inprostate epithelium, it is clearly not just a marker of prostate stemcells because stem cells are present at very low frequency in adultprostate, whereas SOX9 is expressed diffusely in the basal celllayer. Instead, the relatively uniform expression of SOX9 by mostprostate basal cells indicates that it may be critical for broaderfunctions and regulate one or more basal cell–specific proteins.These proteins might include growth factors, cell surface proteins,or extracellular matrix components that are critical for stem/progenitor cell maintenance, or for supporting the overlyingluminal epithelium. In either case, we postulate that SOX9-regulated genes expressed by prostatic basal cells are critical tosupport the generation or survival of luminal epithelial cells andthat SOX9 expression in prostate cancer allows the tumor cellsto maintain their proliferative potential and survival in the absenceof basal cell support.The spectrum of targets regulated by SOX9 in basal cells remains

to be determined, but our data indicate that AR is one of the SOX9-regulated proteins in prostate cancer cells. Consistent with previousstudies of other HMG proteins, we found that AR could interactdirectly with SOX9 and that the interactionwas dependent on a shortpeptide at the carboxyl terminus of ARDNA-binding domain, termedthe CTE. This CTE is present in other steroid hormone receptors,suggesting that SOX9 may similarly interact with these receptors(15–17, 52). The AR-SOX9 interaction may serve to stabilize bindingto a subset of genes coregulated by AR and SOX9, but such geneshave not yet been identified and we have not observed SOX9enhancement of AR activity using standard AR reporter genes. High-level overexpression of transfected SOX9 could suppress AR proteinlevels, possibly reflecting degradation of AR-SOX9 complexes;however, this is not physiologic. More importantly, lower levels ofexogenous SOX9 could enhance AR expression, and SOX9 siRNAcaused a decrease in endogenous AR protein expression withoutdecreasing AR message level, indicating that SOX9 at physiologiclevels in prostate cancer cells enhances AR translation or stability.Interestingly, AR message levels are generally increased in

response to treatments that cause AR protein reduction (such asandrogen withdrawal). Therefore, the failure of AR message levels toincrease after SOX9 down-regulation and AR protein reduction

indicates that SOX9 may, in fact, also positively regulate AR messagelevels. A previous study found that SOX9 expression in SV40 Tantigen–transformed prostate epithelial cells (M12 cells) could beinduced by insulin-like growth factor binding protein–relatedprotein, which is expressed at increased levels in senescent humanprostate epithelial cell cultures (46). Significantly, overexpression oftransfected SOX9 in the M12 cells was found to induce expression ofmessage for AR, PSA, and N-cadherin, although cellular proliferationin these cells was reduced by SOX9 overexpression. This latterinhibitory effect on proliferation may reflect differences in the cellsor be due to higher expression of transfected SOX9.Although SOX9 can interact with and regulate AR expression in

prostate cancer cells, which express high levels of AR, thesignificance of SOX9-AR interaction in normal prostate remainsto be determined. Indeed, luminal epithelial cells express highlevels of AR and are SOX9 negative, clearly demonstrating thatSOX9 is not required for AR expression. Conversely, basal epithelialcells express only low levels of AR message and protein, demon-strating that SOX9 alone is not sufficient for high-level ARexpression. Our interpretation of these observations is thatSOX9 may initiate low-level AR gene expression in cells thatare precursors to luminal epithelial cells, and other transcriptionfactors subsequently take over and stimulate high-level ARexpression in fully differentiated luminal epithelial cells. SOX9expression and its regulation of AR in prostate cancer cells wouldthen be consistent with the hypothesis that SOX9-positive prostatecancer cells represent an intermediate developmental stagebetween basal and luminal epithelial cells.In summary, this study shows that SOX9 protein is expressed in

adult prostate basal epithelium and may play roles in maintainingthe committed stem cell compartment, differentiation, and/orsupporting the overlying luminal epithelium. Expression of SOX9in prostate cancer cells indicates that SOX9-regulated genes maysimilarly play critical roles in supporting prostate cancer growthindependently of basal cells. SOX9 expression in prostate cancercells is Wnt/h-catenin regulated, and AR is one identified down-stream target, although the precise mechanisms by which SOX9regulates AR remain to be determined. The further identification ofSOX9-regulated genes should provide new insights into mecha-nisms that regulate the development of normal prostate andprostate cancer, as well as provide new therapeutic targets.

Acknowledgments

Received 5/8/2006; revised 9/9/2006; accepted 10/25/2006.Grant support: NIH grants R01CA65647 (S.P. Balk) and K01DK64739 (X. Yuan),

Department of Defense grant DAMD17-03-0164 (X. Yuan), and the Hershey FamilyProstate Cancer Research Fund.The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.We thank Drs. P. Berta, B. de Crombrugghe, R.M. Sramkoski, and M. Wegner for

providing reagents; Drs. R. Rittmaster and M. Gleave (Prostate Centre, VancouverGeneral Hospital, Vancouver, British Columbia, Canada); Drs. P.A. Abrahamsson, A.Bjartell, and N. Dizeyi (Department of Urology, Malmo University Hospital, LundUniversity, Malmo, Sweden) for providing paraffin sections of locally recurrentprostate cancers; M. Regan for statistical analyses; and Balk laboratory members forhelpful discussions.

SOX9 in Human Prostate

www.aacrjournals.org 535 Cancer Res 2007; 67: (2). January 15, 2007

References

1. Evans GS, Chandler JA. Cell proliferation studies in ratprostate. I. The proliferative role of basal and secretory

epithelial cells during normal growth. Prostate 1987;10:163–78.2. Evans GS, Chandler JA. Cell proliferation studies in therat prostate: II. The effects of castration and androgen-

induced regeneration upon basal and secretory cellproliferation. Prostate 1987;11:339–51.3. English HF, Santen RJ, Isaacs JT. Response of glandularversus basal rat ventral prostatic epithelial cells to

Research. on January 22, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Cancer Research

Cancer Res 2007; 67: (2). January 15, 2007 536 www.aacrjournals.org

androgen withdrawal and replacement. Prostate 1987;11:229–42.4. Collins AT, Habib FK, Maitland NJ, Neal DE. Identifi-cation and isolation of human prostate epithelial stemcells based on a(2)h(1)-integrin expression. J Cell Sci2001;114:3865–72.5. Foster CS, Dodson A, Karavana V, Smith PH, Ke Y.Prostatic stem cells. J Pathol 2002;197:551–65.6. Hudson DL, O’Hare M, Watt FM, Masters JR. Prolifer-ative heterogeneity in the human prostate: evidence forepithelial stem cells. Lab Invest 2000;80:1243–50.7. Uzgare AR, Xu Y, Isaacs JT. In vitro culturing andcharacteristics of transit amplifying epithelial cells fromhuman prostate tissue. J Cell Biochem 2004;91:196–205.8. van Leenders GJ, Schalken JA. Stem cell differentiationwithin the human prostate epithelium: implications forprostate carcinogenesis. BJU Int 2001;88 Suppl 2:35–42.9. Wang Y, Hayward S, Cao M, Thayer K, Cunha G. Celldifferentiation lineage in the prostate. Differentiation2001;68:270–9.10. Rizzo S, Attard G, Hudson DL. Prostate epithelialstem cells. Cell Prolif 2005;38:363–74.11. Kurita T, Medina RT, Mills AA, Cunha GR. Role of p63and basal cells in the prostate. Development 2004;131:4955–64.12. Signoretti S, Waltregny D, Dilks J, et al. p63 is aprostate basal cell marker and is required for prostatedevelopment. Am J Pathol 2000;157:1769–75.13. Signoretti S, Pires MM, Lindauer M, et al. p63regulates commitment to the prostate cell lineage. ProcNatl Acad Sci U S A 2005;102:11355–60.14. Schepers GE, Teasdale RD, Koopman P. Twenty pairsof sox: extent, homology, and nomenclature of themouse and human sox transcription factor genefamilies. Dev Cell 2002;3:167–70.15. Boonyaratanakornkit V, Melvin V, Prendergast P, et al.High-mobility group chromatin proteins 1 and 2 func-tionally interact with steroid hormone receptorsto enhance their DNA binding in vitro and transcrip-tional activity in mammalian cells. Mol Cell Biol 1998;18:4471–87.16. Verrijdt G, Haelens A, Schoenmakers E, Rombauts W,Claessens F. Comparative analysis of the influence of thehigh-mobility group box 1 protein on DNA binding andtranscriptional activation by the androgen, glucocorti-coid, progesterone and mineralocorticoid receptors.Biochem J 2002;361:97–103.17. Melvin VS, Harrell C, Adelman JS, Kraus WL,Churchill M, Edwards DP. The role of the C-terminalextension (CTE) of the estrogen receptor a and h DNAbinding domain in DNA binding and interaction withHMGB. J Biol Chem 2004;279:14763–71.18. Yuan X, Lu ML, Li T, Balk SP. SRY interacts with andnegatively regulates androgen receptor transcriptionalactivity. J Biol Chem 2001;276:46647–54.19. Amir AL, Barua M, McKnight NC, Cheng S, Yuan X,Balk SP. A direct h-catenin-independent interactionbetween androgen receptor and T cell factor 4. J BiolChem 2003;278:30828–34.20. De Santa BP, Bonneaud N, Boizet B, et al. Directinteraction of SRY-related protein SOX9 and steroido-

genic factor 1 regulates transcription of the human anti-Mullerian hormone gene. Mol Cell Biol 1998;18:6653–65.21. Bishop CE, Whitworth DJ, Qin Y, et al. A transgenicinsertion upstream of sox9 is associated with dominantXX sex reversal in the mouse. Nat Genet 2000;26:490–4.22. Chaboissier MC, Kobayashi A, Vidal VI, et al.Functional analysis of Sox8 and Sox9 during sexdetermination in the mouse. Development 2004;131:1891–901.23. Gao F, Maiti S, Alam N, et al. The Wilms tumor gene,Wt1, is required for Sox9 expression and maintenance oftubular architecture in the developing testis. Proc NatlAcad Sci U S A 2006;103:11987–92.24. Foster JW, Dominguez-Steglich MA, Guioli S, et al.Campomelic dysplasia and autosomal sex reversalcaused by mutations in an SRY-related gene. Nature1994;372:525–30.25. Foster JW. Mutations in SOX9 cause both autosomalsex reversal and campomelic dysplasia. Acta PaediatrJpn 1996;38:405–11.26. Piper K, Ball SG, Keeling JW, Mansoor S, Wilson DI,Hanley NA. Novel SOX9 expression during humanpancreas development correlates to abnormalities incampomelic dysplasia. Mech Dev 2002;116:223–6.27. Wagner T, Wirth J, Meyer J, et al. Autosomal sexreversal and campomelic dysplasia are caused bymutations in and around the SRY-related gene SOX9.Cell 1994;79:1111–20.28. Akiyama H, Chaboissier MC, Martin JF, Schedl A, deCrombrugghe B. The transcription factor Sox9 hasessential roles in successive steps of the chondrocytedifferentiation pathway and is required for expression ofSox5 and Sox6. Genes Dev 2002;16:2813–28.29. de Crombrugghe B, Lefebvre V, Behringer RR, Bi W,Murakami S, Huang W. Transcriptional mechanisms ofchondrocyte differentiation. Matrix Biol 2000;19:389–94.30. Bi W, Deng JM, Zhang Z, Behringer RR, deCrombrugghe B. Sox9 is required for cartilage forma-tion. Nat Genet 1999;22:85–9.31. Bridgewater LC, Lefebvre V, de Crombrugghe B.Chondrocyte-specific enhancer elements in the Col11a2gene resemble the Col2a1 tissue-specific enhancer. J BiolChem 1998;273:14998–5006.32. Stolt CC, Lommes P, Sock E, Chaboissier MC, SchedlA, Wegner M. The Sox9 transcription factor determinesglial fate choice in the developing spinal cord. GenesDev 2003;17:1677–89.33. Wegner M, Stolt CC. From stem cells to neurons andglia: a Soxist’s view of neural development. TrendsNeurosci 2005;28:583–8.34. Sakai D, Suzuki T, Osumi N, Wakamatsu Y.Cooperative action of Sox9, Snail2 and PKA signalingin early neural crest development. Development 2006;133:1323–33.35. Blache P, van de WM, Duluc I, et al. SOX9 is anintestine crypt transcription factor, is regulated by theWnt pathway, and represses the CDX2 and MUC2 genes.J Cell Biol 2004;166:37–47.36. Korinek V, Barker N, Moerer P, et al. Depletion ofepithelial stem-cell compartments in the small intestineof mice lacking Tcf-4. Nat Genet 1998;19:379–83.

37. Vidal VP, Chaboissier MC, Lutzkendorf S, et al. Sox9is essential for outer root sheath differentiation and theformation of the hair stem cell compartment. Curr Biol2005;15:1340–51.38. Murakami S, Kan M, McKeehan WL, de Crom-brugghe B. Up-regulation of the chondrogenic Sox9 geneby fibroblast growth factors is mediated by the mitogen-activated protein kinase pathway. Proc Natl Acad SciU S A 2000;97:1113–8.39. Yuan X, Li T, Wang H, et al. Androgen receptorremains critical for cell-cycle progression in androgen-independent CWR22 prostate cancer cells. Am J Pathol2006;169:682–96.40. Sramkoski RM, Pretlow TG, Giaconia JM, et al. A newhuman prostate carcinoma cell line, 22Rv1. In Vitro CellDev Biol Anim 1999;35:403–9.41. Takash W, Canizares J, Bonneaud N, et al. SOX7transcription factor: sequence, chromosomal localisa-tion, expression, transactivation and interference withWnt signalling. Nucleic Acids Res 2001;29:4274–83.42. Dasari VK, Goharderakhshan RZ, Perinchery G, et al.Expression analysis of Y chromosome genes in humanprostate cancer. J Urol 2001;165:1335–41.43. Lau YF, Zhang J. Expression analysis of thirty one Ychromosome genes in human prostate cancer. MolCarcinog 2000;27:308–21.44. Perinchery G, Sasaki M, Angan A, Kumar V, Carroll P,Dahiya R. Deletion of Y-chromosome specific genes inhuman prostate cancer. J Urol 2000;163:1339–42.45. Jordan JJ, Hanlon AL, Al Saleem TI, Greenberg RE,Tricoli JV. Loss of the short arm of the Y chromosome inhuman prostate carcinoma. Cancer Genet Cytogenet2001;124:122–6.46. Drivdahl R, Haugk KH, Sprenger CC, Nelson PS,Tennant MK, Plymate SR. Suppression of growth andtumorigenicity in the prostate tumor cell line M12 byoverexpression of the transcription factor SOX9. Onco-gene 2004;23:4584–93.47. Garraway LA, Lin D, Signoretti S, et al. Intermediatebasal cells of the prostate: in vitro and in vivocharacterization. Prostate 2003;55:206–18.48. Malki S, Nef S, Notarnicola C, et al. Prostaglandin D2induces nuclear import of the sex-determining factorSOX9 via its cAMP-PKA phosphorylation. EMBO J 2005;24:1798–809.49. Polakis P. Wnt signaling and cancer. Genes Dev 2000;14:1837–51.50. Persad S, Troussard AA, McPhee TR, Mulholland DJ,Dedhar S. Tumor suppressor PTEN inhibits nuclearaccumulation of h-catenin and T cell/lymphoid enhancerfactor 1-mediated transcriptional activation. J Cell Biol2001;153:1161–74.51. Sharma M, Chuang WW, Sun Z. Phosphatidylinositol3-kinase/Akt stimulates androgen pathway throughGSK3h inhibition and nuclear h-catenin accumulation.J Biol Chem 2002;277:30935–41.52. Schoenmakers E, Alen P, Verrijdt G, et al. DifferentialDNA binding by the androgen and glucocorticoidreceptors involves the second Zn-finger and a C-terminal extension of the DNA- binding domains.Biochem J 1999;341:515–21.

Research. on January 22, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

2007;67:528-536. Cancer Res   Hongyun Wang, Nicole C. McKnight, Tao Zhang, et al.   Cancer CellsRegulates Androgen Receptor Expression in Prostate SOX9 Is Expressed in Normal Prostate Basal Cells and

  Updated version

  http://cancerres.aacrjournals.org/content/67/2/528

Access the most recent version of this article at:

   

   

  Cited Articles

  http://cancerres.aacrjournals.org/content/67/2/528.full.html#ref-list-1

This article cites by 51 articles, 21 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/67/2/528.full.html#related-urls

This article has been cited by 14 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected] at

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

  Permissions

  [email protected] at

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

Research. on January 22, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from