The R273H p53 mutation can facilitate the androgen ... - Nature

0 downloads 0 Views 353KB Size Report
Jan 23, 2006 - gene assay to demonstrate that H2 relaxin can induce the expression of prostate-specific antigen via an androgen receptor-mediated pathway.
Oncogene (2006) 25, 2082–2093

& 2006 Nature Publishing Group All rights reserved 0950-9232/06 $30.00 www.nature.com/onc

ORIGINAL ARTICLE

The R273H p53 mutation can facilitate the androgen-independent growth of LNCaP by a mechanism that involves H2 relaxin and its cognate receptor LGR7 RL Vinall1, CG Tepper2, X-B Shi1, LA Xue1, R Gandour-Edwards3 and RW de Vere White1 1

Department of Urology, Davis, School of Medicine and Cancer Center, University of California, Davis, Sacramento, CA, USA; Department of Biological Chemistry, Davis, School of Medicine and Cancer Center, University of California, Sacramento, CA, USA and 3Department of Pathology, Davis, School of Medicine and Cancer Center, University of California, Sacramento, CA, USA

2

Mutations in p53 occur at a rate of approximately 70% in hormone-refractory prostate cancer (CaP), suggesting that p53 mutations facilitate the progression of CaP to androgen-independent (AI) growth. We have previously reported that transfection of p53 gain of function mutant alleles into LNCaP, an androgen-sensitive cell line, allows for AI growth of LNCaP in vitro. We herein confirm the in vivo relevance of those findings by demonstrating that the R273H p53 mutation (p53R273H) facilitates AI growth in castrated nude mice. In addition, we demonstrate that H2 relaxin is responsible for facilitating p53R273Hmediated AI CaP. H2 relaxin is overexpressed in the LNCaP-R273H subline. Downregulation of H2 relaxin expression results in significant inhibition of AI growth, whereas addition of recombinant human H2 relaxin to parental LNCaP promotes AI growth. Inhibition of AI growth was also achieved by blocking expression of LGR7, the cognate receptor of H2 relaxin. Chromatin immunoprecipitation analysis was used to demonstrate that p53R273H binds directly to the relaxin promoter, further confirming a role for H2 relaxin signaling in p53R273H-mediated AI CaP. Lastly, we used a reporter gene assay to demonstrate that H2 relaxin can induce the expression of prostate-specific antigen via an androgen receptor-mediated pathway. Oncogene (2006) 25, 2082–2093. doi:10.1038/sj.onc.1209246; published online 23 January 2006 Keywords: mutant p53; prostate cancer; relaxin

Introduction Prostate cancer (CaP) initially presents as a hormonedependent cancer that requires androgen to develop, grow and differentiate (Huggins and Hodges, 1972). Correspondence: Dr R de Vere White, Department of Urology, UCDMC, 4860 Y Street, Suite 3500, Sacramento, CA 95817, USA. E-mail: [email protected] Received 29 August 2005; revised 3 October 2005; accepted 4 October 2005; published online 23 January 2006

High cure rates for localized disease are achieved using either surgery or radiotherapy. Once CaP becomes metastatic, however, no therapy is curative. The usual treatment for patients with metastatic disease is androgen ablation therapy. While in the short term this treatment results in a reduction in tumor burden by growth arrest and the induction of apoptosis (Kyprianou et al., 1990), this response does not endure and the recurrent tumors no longer require androgen to progress (Gittes, 1991). At this point, the only treatment option available is chemotherapy whose effect is to increase patient survival time by an average of only 2 months (Kasamon and Dawson, 2004; Petrylak et al., 2004; Tannock et al., 2004). The p53 tumor suppressor gene is one of the most commonly mutated genes in human cancers (Olivier et al., 2002). Clinical studies have demonstrated that p53 mutations play a key role in the progression of CaP (Navone et al., 1993; Heidenberg et al., 1995). Our group has reported the frequency of p53 mutations to be about 39–45% in primary CaPs, depending on the approach used for the detection of p53 mutations, rising to 71% in CaP metastases to bone (Shi et al., 2004a). The p53 gene encodes a homotetrameric transcription factor that is activated in response to cellular stress. Its activation results in a global transcriptional response that either blocks cell proliferation or induces apoptosis (Vogelstein et al., 2000). The majority of p53 mutations are missense mutations located in its central DNAbinding domain. These mutations inactivate the normal functions of p53 by diminishing gene transactivation. While the large majority of such mutations result in loss of normal p53 function (LOF), they may also result in a concurrent acquisition of gain-of-function (GOF) growth characteristics. GOF often occurs by the aberrant binding of mutant p53 to atypical promoters, for example, MDR-1 and PCNA, resulting in the transcription of genes known to be involved in cancer progression (Roemer, 1999; Nesslinger et al., 2003; Scian et al., 2004). In published in vitro studies, we have demonstrated that of 16 common p53 mutations analysed, 11 showed various GOF growth characteristics when transfected into CaP cell lines (Shi et al., 2002). In addition, we found a direct link between the

Mutant p53 and H2 relaxin mediate androgen-independent growth RL Vinall et al

2083

expression of four of these GOF mutants (G245S, R248W, R273H and R273C) and the occurrence of AI CaP (Nesslinger et al., 2003). In the current study, a dominant role for p53 mutants in mediating progression of AI CaP in vivo was implicated by the ability of LNCaP-R273H cells to establish xenograft tumors in castrated nude mice. In addition, we identified H2 relaxin as a downstream effector of R273H p53 mutation (p53R273H). The data we present here suggest that H2 relaxin promotes AI CaP by directly affecting the androgen receptor (AR) signaling pathway.

Results p53 GOF mutations facilitate the androgen-independent (AI) growth of LNCaP in vivo We have previously demonstrated that LNCaP transfected with the G245S, R248W, R273H and R273C p53 mutant alleles are able to grow in the absence of androgen in vitro (Nesslinger et al., 2003). We now demonstrate that one of these sublines, LNCaP-R273H, is also able to grow in vivo in the absence of androgen. Five of five castrated nude mice injected with cells of the LNCaP-R273H subline grew tumors, as compared to only one of five injected with LNCaP-vector cells (data not shown). Furthermore, the LNCaP-R273H xenograft tumors were noticeable as early as 16 days postinjection. In contrast, the single LNCaP-vector tumor did not appear until 50 days postinjection. Subsequent histological analysis confirmed the presence of tumors in all the above cases (Figure 1a). Immunohistochemical analysis of LNCaP-R273H tumors demonstrated that p53 was stabilized in >25% of tumor cells (Figure 1b), confirming that these tumors express mutant p53 and were derived from injection of the LNCaP-R273H subline. These data demonstrate that p53 mutant alleles are capable of conferring AI growth of CaP cells in vivo.

LNCaP transfected with p53 GOF mutations secrete a factor(s) that stimulates AI growth of LNCaP A number of cytokines and growth factors have been implicated in the AI growth of CaP cells. To determine whether a secreted factor may be responsible for the AI growth conferred by these p53 GOF mutants, we used a combination of conditioned media experiments and microarray data mining. Our experiments demonstrated that androgen-free medium conditioned by LNCaPR273H for 3 days (charcoal-stripped serum (CSS)R273H) facilitated a sharp increase in growth of LNCaP-vector (Figure 2a). In comparison, addition of nonconditioned androgen-free media (CSS media) or conditioned media taken from the P151S non-GOF subline (CSS-P151S) did not increase growth of LNCaPvector. The P151S p53 mutation results in the loss of p53 function but no gain of function (GOF), that is, it cannot grow in the absence of androgen, and thus is a non-GOF mutant used here as an additional control. Conditioned media taken from the R248W subline (CSS-R248W) facilitated a minimal increase in AI growth of LNCaP-vector (Figure 2b; Po0.05 on day 5). Conditioned media taken from the R273C and G245S sublines (CSS-R273C and CSS-G245S, respectively) did not increase AI growth of LNCaP-vector above that seen with CSS-P151S conditioned media or nonconditioned media. In summary, these experiments suggest that stimulation of AI growth via autocrine/ paracrine signaling is a novel GOF for some p53 GOF mutants, especially the R273H mutant. LNCaP transfected with p53 GOF mutants express H2 relaxin mRNA To identify potential candidates for the secreted factor(s) that mediated AI growth of LNCaP-vector cells, we reanalysed microarray data obtained in a previous study (Tepper et al., 2005). This study was designed to identify genes common to all four GOF sublines that are differentially expressed when subjected

Figure 1 p53 GOF mutations facilitate androgen-independent growth of LNCaP in vivo. Subcutaneous injection of LNCaP-R273H into castrated nude mice resulted in tumor formation in five of five mice, whereas injection of LNCaP-vector resulted in tumor formation in only one of five mice (data not shown). Mice injected with LNCaP-R273H formed tumors as early as 16 days. In contrast, the single LNCaP-vector tumor did not form until 50 days postinjection. All injected mice were observed for a total of 12 weeks postinjection). Histological analysis confirmed that the mice did form tumors (a, H&E stain,  40 magnification). Immunohistochemical analysis confirmed that >25% of cell nuclei in LNCaP-R273H tumors stained positively for p53, that is, expressed mutant p53 (b,  40 magnification) (arrow indicates a representative cell nucleus stained positively for p53). * ¼ tumor. Oncogene

Mutant p53 and H2 relaxin mediate androgen-independent growth RL Vinall et al

a

a 2.4 2.2

CSS-R273H

2

CSS-P151S

1.8 1.6

1.2

b

1 0.8 d1

d3

d5

Fold Change in Cell Number

b 2.4 CSS-media

2.2

CSS-G245S

2

CSS-R248W

1.8

CSS-R273C

Vector G245S R248W R273H R273C P151S

500 400 300 200 100 0

RLN2 RLN1 MEST IL7 ANGPT2 NMU Gene Symbol

600

Vector G245S R248W R273H R273C P151S

500 400 300 200 100 0

Vector G245S R248W R273H R273C P151S

LNCaP-p53 Mutant Subline

1.6

c

1.4

Vector

G245S

R248W

R273H

R273C

P151S

FBS CSS FBS CSS FBS CSS FBS CSS FBS CSS FBS CSS -ve

Relaxin H2

1.2

Ras

1 0.8 d1

d3

d5

Figure 2 LNCaP transfected with p53 GOF mutations secrete a factor(s) that allows for androgen-independent growth of LNCaP. Conditioned media taken from LNCaP-R273H grown in media containing charcoal-stripped serum (CSS-R273H) facilitated the androgen-independent growth of LNCaP-vector cells (a, Po0.005 on day 5 for vector versus R273H). LNCaP-vector cells were unable to grow in CSS conditioned media taken from a p53 nonGOF mutant cell line (LNCaP-P151S) or in nonconditioned media (CSS-media). CSS conditioned media taken from one of the other p53 GOF mutants (R248W) also facilitated the androgenindependent growth of LNCaP-vector, but to a lesser extent compared to the LNCaP-R273H mutant (b, Po0.05 on day 5 for vector versus R248W).

to conditions of androgen deprivation. In the current study, comparison analysis was performed to highlight expression changes conferred by stable expression of the individual mutant alleles. The resulting lists were mined for the overexpression of transcripts encoding secreted proteins having known or putative growth factor properties. The analysis focused upon LNCaP-R273H at the outset, as the conditioned medium derived from this line exhibited the most potent AI-inducing effect. This analysis revealed that LNCaP transfected with p53 GOF alleles express higher levels of five genes that encode secreted proteins when compared to LNCaPvector and LNCaP-P151S. These genes were H2 relaxin, MEST, IL-7, ANGPT2 and NMU (Figure 3a). Expression of one of these genes, H2 relaxin, was particularly high in LNCaP-R273H (a 4.6-fold increase in expression compared to the LNCaP-vector cells) (Figure 3b). Relaxin H1 expression was also elevated in two of the Oncogene

600

1.4

Normalized Signal Intensity

Fold Change in Cell Number

CSS-media

Normalized Signal Intensity

2084

Figure 3 The expression of five genes encoding secreted proteins are upregulated in LNCaP transfected with p53 GOF mutations compared to LNCaP transfected with vector alone. The mRNA expression of H2 relaxin, MEST, IL-7, ANGPT2 and NMU, genes that encode for secreted proteins with putative growth factor properties, is upregulated in all of the p53 GOF mutants (a). Expression of relaxin H1 was upregulated in two of the GOF mutants, G245S and R273H. The LNCaP-R273H mutant was shown to express particularly high levels of H2 relaxin (b). Upregulation of H2 relaxin expression in the p53 GOF mutant cell lines was confirmed via RT–PCR analysis (c). Again, significant upregulation in expression of H2 relaxin mRNA was observed in the LNCaP-R273H mutant (CSS ¼ charcoal stripped serum).

p53 GOF mutant sublines (G245S and R273H), but not to the same extent as H2 relaxin. While relaxin H1 mRNA is expressed in a number of tissues, relaxin H1 protein expression patterns have not been described (Ivell et al., 1989; Garibay-Tupas et al., 2000; Ivell and Einspanier, 2002; Samuel et al., 2003). The microarray findings were validated by RT–PCR analysis, which confirmed that H2 relaxin mRNA is expressed by all of the p53 GOF mutant cells lines (G245S, R248W, R273H, R273C) in the absence of androgen (Figure 3c). In agreement with the microarray data, the R273H mutant expressed particularly high levels of H2 relaxin mRNA. Interestingly, H2 relaxin mRNA expression appeared to be induced in response to androgen withdrawal. In the absence of androgen, a small increase in H2 relaxin mRNA expression was also observed in the vector and P151S non-GOF sublines (Figure 3c). Based on these combined data, in the present study we elected to study the role of H2 relaxin in mediating the progression to androgen independence

Mutant p53 and H2 relaxin mediate androgen-independent growth RL Vinall et al

2085

in the LNCaP-R273H model. Analysis of the other secreted factors that are upregulated in the p53 GOF sublines is ongoing. LNCaP transfected with the R273H p53 GOF mutant express high levels of H2 relaxin protein ELISA analyses revealed that conditioned media taken from the LNCaP-R273H subline contains high levels of

ng/ml relaxin

a 1000

Relaxin ELISA

100 10

or

ct

ve

ve

ct

or

-F

BS G -C 24 SS 5 G S-F 24 5 BS R S-C 24 8 SS R W24 C 8 SS R W-C 27 3 SS R H-F 27 3 BS R H-C 27 3 SS R C-F 27 3 BS P1 C-C 51 SS P1 S-F 51 BS SC SS

1

LNCaP -vector

b

LNCaP -R273H Pro-relaxin (18 kD)

β-actin

Relative Change in Cell Number

c

2.5

** 2

*

1.5

*

1 0.5 0 control

rh rlx (50ng/ml) rh rlx (100ng/ml) R1881 (1nM)

Figure 4 LNCaP transfected with the R273H p53 GOF mutant express high levels of H2 relaxin protein. Recombinant human H2 relaxin can facilitate androgen-independent growth of LNCaP. ELISA analysis revealed that conditioned media taken from LNCaP-R273H contains high levels of H2 relaxin (a). In comparison, conditioned media taken from LNCaP-vector and the other p53 GOF mutants contained insignificant amounts of relaxin that were equal to or even less than that found in unconditioned media (relaxin level in 5% FBS media were 2.5 mg/ ml and in 5% CSS media were 1.2 mg/ml, data not shown). Western blot analysis of treated cells confirmed that higher levels of H2 relaxin are expressed by LNCaP-R273H (b) compared to LNCaPvector cells. The presence of the 18 kDa pro-form of H2 relaxin but not the mature 6 kDa form indicates that cleavage probably occurs after secretion. Addition of recombinant human (rh) relaxin to LNCaP facilitated AI growth of LNCaP (c). Addition of 50 and 100 ng/ml rh relaxin caused a 1.35- and 1.45-fold increase in growth, respectively. R1881, a synthetic androgen, caused a twofold increase in growth. *Po0.05, **Po0.005 and ***Po0.0005.

relaxin protein (Figure 4a). In comparison, conditioned media taken from LNCaP-vector cells and the other p53 GOF mutants contained insignificant amounts of H2 relaxin protein, equal to or less than that found in unconditioned media (the H2 relaxin levels in 5% FBS media were 2.5 ng/ml and in 5% CSS media were 1.2 ng/ ml, data not shown). This result was somewhat surprising as while all of the GOF sublines expressed higher levels of H2 relaxin mRNA compared to the vector and P151S controls, by ELISA analysis only the R273H subline expressed higher expression of relaxin protein. It should be noted that the ELISA used in this study detects both H1 and H2 relaxin. While expression of H1 relaxin protein has not been described in prostate, it cannot be ruled out that the ELISA is measuring a combination of the H1 and H2 relaxin isoforms. All other reagents used in this study specifically detect/ downregulate the H2 relaxin isoform. The discrepancies observed in relaxin protein and mRNA expression levels could also be due to differences in H2 relaxin translation and/or secretion between the sublines. Further studies would be needed to confirm this. A decrease in the relative level of relaxin protein present in conditioned media taken from LNCaP-R273H cultured in 5% CSS versus 5% FBS was observed. Again, this result was somewhat surprising as our mRNA data demonstrated an increase in the expression of H2 relaxin by the GOF sublines in the absence of androgen. It is possible that the decrease in relaxin protein expression is 5% CSS could be due to cell number differences, as LNCaPR273H grow more slowly in 5% CSS media than in 5% FBS. Differences in cell number can not explain the increased expression of relaxin protein by LNCaPR273H relative to the other p53 GOF sublines; however, as we have previously demonstrated, all these sublines grow at a similar rate in both FBS and CSS media (Nesslinger et al., 2003). Western blot analysis of cell lysates confirmed that higher levels of H2 relaxin protein are expressed by LNCaP-R273H (Figure 4b) compared to LNCaP-vector cells. The presence of the 18 kDa proform of H2 relaxin in the cells but not the mature 6 kDa form suggests that cleavage probably occurs after secretion. Gels were run under both reducing and nonreducing conditions to ensure the 6 kDa form was not simply being lost due to disruption of the A and B chains (Figure 4b shows H2 relaxin run under nonreducing conditions, reducing conditions not shown). Interestingly, it has been reported that both the pro- and mature forms of H2 relaxin display the same biological activity (Silvertown et al., 2003a). Taken together, these data confirm that LNCaP-R273H express high levels of H2 relaxin protein. Recombinant human (rh) H2 relaxin stimulates AI growth of LNCaP Treatment of parental LNCaP cells with rh H2 relaxin facilitated their AI growth (Figure 4c). The addition of 50 or 100 ng/ml H2 relaxin, levels similar to those expressed by LNCaP-R273H, to the vector subline cells caused a 1.35–1.45-fold increase in growth by day 5 of Oncogene

Mutant p53 and H2 relaxin mediate androgen-independent growth RL Vinall et al

2086

a

untreated control relaxin siRNA siRNA

-ve

H2 relaxin Ras

b 50 40 ng/ml relaxin

culture (Po0.05), although no increase in growth was observed on days 1 and 3 of culture (data not shown). Combined with our other data, this result suggests that H2 relaxin may be the factor, or one of the factors, in the conditioned medium from R273H cells that allows for the AI growth of LNCaP cells. R273H conditioned media caused a greater increase in AI growth of LNCaP cells, namely a 1.8-fold increase by day 5 of culture (Figure 2a). The difference in growth resulting from rh H2 relaxin versus that from R273H conditioned media may be due to the latter containing factors other than H2 relaxin that are growth stimulatory under conditions of androgen withdrawal. In support of this, our microarray data mining indicated that several secreted proteins with putative growth factor properties are expressed by LNCaP-R273H cells.

30

untreated control siRNA

20

H2 rlx siRNA

*

**

***

48 hours

72 hours

10 0 24 hours

The H2 relaxin receptor, LGR7, is expressed in parental LNCaP and in LNCaP-p53GOF sublines To determine whether LNCaP-R273H cells can use H2 relaxin as part of an autocrine signaling loop that potentially facilitates AI growth of CaP cells, we examined the expression of LGR7, the H2 relaxin receptor. We determined that LGR7 is expressed by both the LNCaP-vector and LNCaP-p53GOF mutant sublines (Figure 6a), confirming that H2 relaxinmediated signaling is possible in these cells. Interestingly, LGR7 expression levels increased significantly when LNCaP transfected with p53 GOF mutants were subjected to androgen withdrawal (i.e., cultured in CSS) (Figure 6a). In contrast, this phenomenon was not observed in the LNCaP subline harboring the P151S Oncogene

Change Fold in Cell Number

c

2 1.8 1.6 1.4 1.2 1 0.8 0.6 0.4 0.2 0

Control siRNA Relaxin siRNA

d1

d Number of colonies >50 cells

RNA interference (RNAi)-mediated H2 relaxin gene silencing diminishes the ability of LNCaP-R273H to grow in the absence of androgen and to form colonies in soft agar Collectively, the above data suggest that H2 relaxin plays an important role as a downstream effector of the p53R273H mutant in mediating AI growth of CaP. To test this hypothesis, we used siRNAs to evaluate the effects of H2 relaxin knockdown upon growth in androgen-free media and in soft agar. Marked downregulation of H2 relaxin mRNA and protein was achieved within 24 h following transfection of LNCaP-R273H with H2 relaxin-specific SMARTPool siRNA (Figure 5a and b). Downregulation of relaxin protein was still observed 72 h following transfection (Figure 5b; Po0.0005). Transfection efficiency neared 100% at 5 h post-transfection (data not shown). Suppression of H2 relaxin expression resulted in a threefold decrease in AI growth after 5 days (Po0.0001) and a twofold decrease after 3 days of treatment (Po0.0005) (Figure 5c). In addition, treatment of LNCaP-R273H cells with H2 relaxinspecific siRNA before culturing in soft agar resulted in a 50% reduction in colony formation (Po0.05) (Figure 5d). These data indicate that H2 relaxin is an important factor in mediating the aggressive growth properties of LNCaP-R273H, namely AI and anchorage-independent growth and survival.

d3

d5

1000 800 600

*

400 200 0 Control siRNA

Relaxin siRNA

Figure 5 RNA interference technology can be used to downregulate expression of H2 relaxin. Downregulation of H2 relaxin in the LNCaP-R273H results in a significant decrease in androgenindependent growth and a decreased ability to form colonies in soft agar. RT–PCR analysis revealed that treatment of LNCaP-R273H with siRNA (100 nM) designed to specifically degrade H2 relaxin mRNA resulted in a decrease in H2 relaxin mRNA expression within 24 h (a). Downregulation of relaxin protein following siRNA treatment was confirmed using ELISA analysis (b). Downregulation of H2 relaxin expression in LNCaP-R273H resulted in a threefold decrease in androgen-independent growth by day 5 of culture (c, Po0.0001 on day 5). In addition, treatment of LNCaP-R273H with H2 relaxin-specific siRNA before culturing in soft agar resulted in a 50% reduction in colony formation (d). *Po0.05, **Po0.005 and ***Po0.0005).

LOF allele. These data suggest that induction of LGR7 expression may represent a component of an autocrine signaling survival mechanism that is activated in p53 GOF mutant cell lines when androgen is removed.

Mutant p53 and H2 relaxin mediate androgen-independent growth RL Vinall et al

2087

a

Vector

G245S

R248W

R273H

R273C

P151S

FBS CSS FBS CSS FBS CSS FBS CSS FBS CSS FBS CSS -ve

LGR7 Ras

b

Fold Change in Cell Number

d

c

Control LGR7 siRNA siRNA

Control LGR7 siRNA siRNA

LGR7

LGR7

Ras

β-actin

LNCaP-R273H

3 2.5 2

Control siRNA LGR7 siRNA

1.5 1 0.5 0 d1

In LNCaP, the expression of p53R273H and H2 relaxin are linked. H2 relaxin is expressed by other AI CaP cell lines Transient transfection of LNCaP with tet-inducible p53R273H revealed that the expression of p53R273H and of H2 relaxin are linked. Increased expression of p53R273H resulted in increased expression of H2 relaxin mRNA (Figure 7a). As expected, transient transfection of p53R273H resulted in AI growth of LNCaP (Figure 7b; Po0.005 on day 5 of culture). Expression of H2 relaxin mRNA was confirmed in a number of AI CaP cell lines besides p53R273H (data not shown). These included CWR22Rv1, DU145 and two CaP cell lines selected for androgen independence in our laboratory by growing LNCaP in CSS for >1 year (Cds1 and 2) (Shi et al., 2004b). The RT–PCR and ELISA methods described in this paper were not able to detect H2 relaxin mRNA or protein in PC3 cells, an AI cell line that lacks p53 expression (data not shown). The PC3 cells were cultured in 5% CSS for 5 days before analysis.

d5

Figure 6 LNCaP and LNCaP transfected with p53 GOF mutations express the H2 relaxin receptor LGR7. Downregulation of LGR7 in LNCaP-R273H by using RNAi results in a decrease in androgen-independent growth. RT–PCR analysis revealed that LNCaP-vector and LNCaP transfected with p53 mutants all express the H2 relaxin receptor, LGR7 (a). Expression of LGR7 mRNA was significantly increased in the LNCaP transfected with p53 GOF mutants in response to androgen withdrawal. This increase was not observed in LNCaP-vector or in the p53 nonGOF mutant LNCaP-P151S. Treatment of LNCaP-R273H with siRNA (100 nM) designed to specifically degrade LGR7 mRNA resulted in a decrease in LGR7 mRNA and protein expression within 72 h (b and c, respectively). This downregulation led to a 1.8fold decrease in the androgen-independent growth of LNCaPR273H by day 5 of culture (d, Po0.005 on day 5).

Downregulation of LGR7 compromises the ability of LNCaP-R273H to grow in the absence of androgen Downregulation of LGR7 mRNA and protein was achieved within 72 h following treatment with SMARTPool siRNA (Figure 6b and c, respectively). Transfection efficiency neared 100% at 5 h post-transfection (data not shown). While control siRNA-treated cells exhibited a 2.5-fold increase in cell growth over the 5-day duration of the experiment, treatment of LNCaP-R273H with the LGR7specific siRNA effectively restrained AI proliferation, permitting only a 0.4-fold increase in cell growth (Figure 6d). While the magnitude of growth inhibition is less than observed with the H2 relaxin siRNA treatment, it is still significant (for control versus LGR7 siRNA treatments on day 5, Po0.005). This lesser growth inhibition induced by LGR7-specific siRNA could stem from slower kinetics induced by the LGR7 message or it could result from H2 relaxin signaling via other receptors. The combined data demonstrate that H2 relaxin and its cognate receptor, LGR7, are involved in facilitating AI growth mediated by the R273H p53 GOF mutant-transfected cells.

p53R273H can bind directly to the H2 relaxin promoter Chromatin immunoprecipitation (ChIP) analysis demonstrated that p53R273H can bind directly to the H2 relaxin promoter (Figure 7c), confirming that H2 relaxin is a downstream effector of p53R273H. The data demonstrate that other p53 mutants can also bind to the H2 relaxin promoter, but to a lesser extent. This result agrees with our other data demonstrating that H2 relaxin expression is considerably higher in the LNCaPR273H subline. A minimal amount of background binding was observed for the LNCaP-vector samples. The human telomerase (hTERT) promoter was used as a positive control in this experiment, as it has been previously demonstrated that mutant p53 can bind the hTERT promoter (Scian et al., 2004). Analysis of the binding of wild-type p53 to the p21 promoter in response to increasing doses of irradiation confirmed that the assay was working (Figure 7d). H2 relaxin can activate an AR-mediated PSA promoter construct To determine whether H2 relaxin may allow for AI CaP by influencing the AR pathway, we used an ARmediated PSA reporter gene assay (Figure 8a). DU145 CaP cells were used for these experiments because they lack expression of the AR. EGF was used as a positive control (Culig et al., 1994). The addition of rh H2 relaxin to DU145 cells, transiently transfected with pPSA-LUC plasmids and AR-expression vectors in the absence of androgens, resulted in a dose-dependent increase in the induction of luciferase expression (Figure 8a). In combination with data that demonstrate LNCaP-R273H express relatively high levels of AR in the absence of androgen (Figure 8b), and that link increased expression of p53R273H in LNCaP to increased expression of PSA (Figure 8c), this result strongly suggests that p53R273H/H2 relaxin are able to influence the AR signaling pathway. Oncogene

Mutant p53 and H2 relaxin mediate androgen-independent growth RL Vinall et al

2088

a

Dox. µg/ml

a

12000 10000

0.25

0.5

1.0

Luciferase activity

0

p53R273H

relaxin

dox+ dox-

2 1

0 pcDNA3.1+ pcDNA3.1+-AR pPSA-Luc EGF (ng/ml)

– – – –

+ – + –

– + + 50

– + + –

– + + –

– + + –

relaxin (ng/ml)







0

25

50

b

0

0

1 3 Time (day)

* 4000

ve ct o G r 24 5S R 24 8W R 27 3H R 27 3C P1 51 S

Growth (relative to day 0)

4 3

6000

2000

ras

b

* 8000

5

AR

R 17 5H

R 27 3H

P1

51

S

p5 3 W T

R 28 4W

β-actin

c

c RLN

hTERT

LNCaP + 0

– 0

LNCaP/R273H – 0

– 0.25

– 0.5

Androgen Dox (µg/ml) PSA ras

Input

d

0

2

10

Gy p21

Input Figure 7 R273H p53 mutation can bind directly to relaxin. Transient transfection of LNCaP with the R273H mutant resulted in a dose-dependent increase in H2 relaxin mRNA expression (a), and confirmed the R273H mutant facilitates AI growth of LNCaP (b, Po0.005 on day 5 of culture). ChIP analysis revealed that p53R273H can bind directly to the relaxin promoter (RLN) (c), confirming that relaxin is a downstream effector of p53R273H. Other p53 mutants can bind to the relaxin promoter, but to a much lesser extent. Human telomerase (hTERT) was used as a positive control. Analysis of the binding of wild-type p53 to the p21 promoter in response to increasing doses of irradiation confirmed that the assay was working (d).

Discussion In the present study, we demonstrate that the p53R273H mutant plays a critical role in the progression to AI CaP. To our knowledge, this is the first study to demonstrate that a p53 GOF mutant can induce AI growth of CaP in vivo. Five of five castrated nude mice established xenograft tumors following subcutaneous injection with LNCaP-R273H compared to only one of five injected Oncogene

Figure 8 H2 relaxin can activate an AR-mediated PSA promoter. Addition of recombinant human (rh) relaxin to DU145 (an ARnegative CaP cell line) cells transiently transfected with the pPSALUC plasmid, and the AR-expression vector in the absence of androgens resulted in a dose-dependent increase in the activation of the reporter construct (a). In the absence of androgen, LNCaPR273H expressed relatively high levels of AR protein (b). Increasing expression of p53R273H in LNCaP is linked to increased expression of PSA mRNA (c). The combined data suggest that p53R273H/H2 relaxin are able to influence the androgen receptor signaling pathway. *Po0.05.

with LNCaP-vector. In addition, four of five of the p53R273H xenograft tumors formed much more rapidly compared to the vector control, demonstrating the aggressive nature of the p53R273H mutant allele. Combined with our previously published in vitro data (Nesslinger et al., 2003), this result shows that p53 GOF mutations can induce the progression of CaP to its lethal, AI form. A number of cytokines and growth factors have been implicated in the AI growth of CaP cells (Hanahan and Weinberg, 2000; Welsh et al., 2003; Lee et al., 2004). Our conditioned medium experiments and subsequent microarray data mining confirmed that secreted factors play a role in p53R273H-mediated AI CaP. Conditioned media taken from LNCaP-R273H was a particularly strong inducer of AI growth. Microarray data mining identified five candidate genes: H2 relaxin, MEST, IL-7,

Mutant p53 and H2 relaxin mediate androgen-independent growth RL Vinall et al

2089

ANGPT2 and NMU. We decided to initially focus on H2 relaxin as it was the most highly upregulated gene in the LNCaP-R273H subline, the most aggressive of the p53 GOF mutants studied. The relaxin H1 and H2 isoforms share 90% homology, and display very similar and overlapping biological activities. (Ivell and Einspanier, 2002; Sherwood, 2004). Both isoforms are expressed in prostate; however, to date only the H2 isoform has been shown to be translated and secreted (Ivell et al., 1989; GaribayTupas et al., 2000; Samuel et al., 2003; Welsh et al., 2003). Our data demonstrate that H2 relaxin is highly upregulated at both the message and protein level in the p53R273H mutant. This result was confirmed using a tetinducible system in which increasing expression of p53R273H correlated with increased expression of H2 relaxin. While H2 relaxin mRNA was also upregulated in all of the other p53 GOF mutants (G245S, R248W and R273C), relaxin protein was not. The data suggest that there is a disconnect between relaxin transcription and translation and/or secretion, although further studies would be needed to confirm this. The data also suggest that H2 relaxin is not a key player in all p53 mutant-mediated AI CaP. In support of different pathways mediating the attainment of p53 mutantmediated AI CaP, our microarray studies show that the p53 GOF mutant cell lines have very different gene expression profiles. These data are consistent with a recent report by Shah et al. (2004), which underscores the fact that like many other cancers, AI CaP can be achieved by the activation of multiple different pathways. As expression of the R273H mutation is frequent in AI CaP patients (Dinjens et al., 1994), H2 relaxin would be an appropriate therapeutic target in the subset of patients having this mutation. A disconnect between H2 relaxin mRNA and H2 relaxin protein expression was also seen in LNCaP-R273H cultured in CSS. H2 relaxin mRNA expression was elevated in CSS, although a small decrease in relaxin protein was observed under the same conditions. These disconnects could indicate control of H2 relaxin occurs at or after translation. The data also suggest that while H2 relaxin expression allows for AI growth of LNCaP-R273H, relaxin protein expression is not upregulated as a result of androgen withdrawal. Our rh H2 relaxin and siRNA experiments confirmed that H2 relaxin plays a direct and functional role in R273H p53 GOF mutant-facilitated AI CaP. Addition of rh H2 relaxin allowed for AI growth of LNCaP to occur. Downregulation of H2 relaxin using RNAi resulted in a significant reduction in the ability of the LNCaP-R273H subline to grow in the absence of androgen and to form colonies in soft agar. These data show that relaxin plays a role in p53R273H-facilitated AI CaP. Interestingly, H2 relaxin mRNA has been shown to be upregulated during neuroendocrine differentiation of LNCaP (Figueiredo et al., 2005). This group reported that induction of neuroendocrine differentiation using epinephrine and interleukin-6 resulted in a 1.5-fold increase in relaxin mRNA expression. In addition, it has been recently reported that strong overexpression of H2

relaxin increases PC3 prostate xenograft growth and angiogenesis (Silvertown et al., 2006). These data support our finding that increased H2 relaxin expression is associated with AI growth of CaP. A functional role for H2 relaxin in the progression of other cancers has also been demonstrated (Stemmermann et al., 1994; Tashima et al., 1994). Treatment of a breast cancer cell line, MCF-7, with H2 relaxin increased both cell proliferation and invasiveness (Tashima et al., 1994). Other studies have demonstrated that H2 relaxin plays a role in processes associated with cancer progression. For example, H2 relaxin can increase blood flow and can influence collagen synthesis and neoangiogenesis (Palejwala et al., 2002; Silvertown et al., 2003b). These data imply that H2 relaxin may play a role in modulation of the tissue microenvironment in AI CaP. The ability of p53 GOF mutants to increase blood flow and influence collagen synthesis would promote CaP tumorigenesis. A role for secreted H2 relaxin in p53R273H-mediated AI CaP was further validated by our finding that LGR7, the H2 relaxin receptor, is expressed in LNCaP-R273H. LGR7 mRNA expression was also detected in the LNCaP-vector and the other p53 mutant cell lines. These data further demonstrate that H2 relaxin autocrine signaling is possible in LNCaP-R273H, and validate the likelihood that H2 relaxin is the major factor in LNCaP-R273H CSS conditioned media facilitating AI growth of LNCaP-vector. LGR7 is a Gprotein-coupled receptor (GPCR), and has considerable homology (60%) to the insulin-like peptide 3 receptor, LGR8 (Hsu et al., 2002). Like other GPCRs, the main signaling pathway activated by ligand binding is the adenylate cyclase/cAMP/PKA pathway (Hsu et al., 2000; Bathgate et al., 2003). Crosstalk between LGR7 and receptor tyrosine kinases has been demonstrated in other cell lines, suggesting that H2 relaxin signaling networks with other pathways that are known to facilitate AI CaP, such as the IGF/IGFR pathway (Chan et al., 1998; Bartsch et al., 2001; Raj et al., 2002; Zhang et al., 2002). Other GPCRs are known to be involved in the progression to AI CaP (Lee et al., 2004). Our finding that LGR7 siRNA treatment results in the inhibition of AI growth in LNCaP-R273H further supports a role for H2 relaxin signaling in R273H p53 mutant-mediated AI CaP. Using RNAi, we achieved downregulation of LGR7 expression by 72 h. Downregulation of LGR7 mRNA and protein expression was minimal at 24 and 48 h time points. The reason for this delay in downregulation of expression is currently unclear, but it may explain why a smaller decrease in AI growth of LNCaP-R273H was observed when LGR7 was downregulated compared to when H2 relaxin itself was downregulated using RNAi. ChIP analysis demonstrated that p53R273H is able to bind directly to the H2 relaxin promoter. These data further implicate H2 relaxin as a downstream effector of p53R273H. In keeping with our data showing that other p53 mutants express H2 relaxin mRNA but at lower levels than p53R273H, these other p53 mutants were also able to bind to the H2 relaxin promoter, but not as strongly as p53R273H. The binding of mutant p53 to novel Oncogene

Mutant p53 and H2 relaxin mediate androgen-independent growth RL Vinall et al

2090

promoters has been documented in many cancers and is considered a GOF characteristic (van Oijen and Slootweg, 2000; El-Hizawi et al., 2002; Shi et al., 2002; Nesslinger et al., 2003; Scian et al., 2004). In CaP, examples of such p53 GOF activity include upregulation of hTERT, MDR-1 and PCNA, and the ability to grow in soft agar (Chin et al., 1992; Cadwell and Zambetti, 2001; Scian et al., 2004). In other cancers, p53 GOF activities include upregulation of c-myc, IGF-1 and EGFR (Deb et al., 1992, 1994; Lanyi et al., 1998). Our ChIP data, combined with data obtained in our previous studies (Shi et al., 2002), show that GOF activity plays a critical role in p53 mutant-mediated AI CaP in our experimental system. Our data point to a connection between the p53R273H/ H2 relaxin signaling pathway and the AR signaling pathway. As almost all hormone-refractory CaPs express AR, and >70% of them have p53 mutations, we hypothesize that p53R273H-induced H2 relaxin expression could activate the AR in a ligand-independent manner. Using a reporter gene assay, we demonstrated that rh H2 relaxin was able to activate an AR-mediated PSA promoter in DU145 cells in the absence of androgen. These data support our hypothesis. Experiments to further confirm and define the intersection of these signaling pathways are ongoing. It is probable that binding of H2 relaxin to LGR7 results in the activation of the cAMP/PKA pathway and/or the MAPK pathway. These signaling pathways have been reported to play pivotal roles in CaP progression to AI status (Amorino and Parsons, 2004; Koul et al., 2004; Culig et al., 2005). In conclusion, we have demonstrated that p53R273H strongly enhances tumorigenicity in castrated nude mice. We have confirmed that this mutant plays a role in AI CaP, and have demonstrated that H2 relaxin is the critical autocrine/paracrine downstream effector of p53R273H-mediated AI CaP. Our data also suggest that the p53R273H/H2 relaxin signaling pathway interacts with the AR signaling pathway. Elucidating the mechanisms by which AI CaP occurs is critical to the development of new treatments for this disease. Our future studies will focus on further delineating the exact pathway by which H2 relaxin signaling mediates p53R273H-mediated AI CaP.

Materials and methods Cell culture Parental LNCaP (ATCC, Manassas, VA, USA) and LNCaP transfected with mutant p53 (LNCaP-G245S, LNCaPR248W, LNCaP-R273H, LNCaP-R273C, LNCaP-P151S) or vector alone (LNCaP-vector) were routinely maintained in RPMI 1640 (Invitrogen, Carlsbad, CA, USA) supplemented with 5% FBS (Omega Scientific, Tarzana, CA, USA). Transfected LNCaP sublines were grown in the presence of G418 (500 mg/ml) (Invitrogen). The P151S p53 mutation causes LOF but no GOF, that is, it cannot grow in the absence of androgen, and is therefore termed a non-GOF mutant, and is used here as a negative control. The other four p53 mutants cause both LOF and GOF, and have been shown to facilitate Oncogene

AI growth in vitro. We have previously shown that expression levels of p53 are higher in LNCaP transfected with mutant p53 cells (Nesslinger et al., 2003). In vivo mouse studies Four-week-old male athymic nude mice (Harlan, Indianapolis, IN, USA) were injected subcutaneously with 5  106 LNCaPR273H or 5  106 LNCaP-vector cells (five mice per group) suspended in 30% matrigel (BD Biosciences, Bedford, MA, USA): 70% RPMI 1640 supplemented with 5% FBS. Mice were castrated 1-week postinjection. Tumor growth was then monitored for 12 weeks postcastration. The mice were euthanized when tumors reached approximately 1 cm3 in size or at the end of the 12-week period. At the time of harvest, tumors were excised and a portion of each sent for pathological analysis. Histology and immunohistochemistry For pathological analysis of tumors, 5 mm paraffin sections were stained using H&E. To detect mutant p53 expression, 5 mm paraffin sections were stained using a monoclonal mouse anti-human p53 antibody (DO7, Novocastra Labs, Burlingame, CA, USA). A paraffin section of DU145 CaP cell line was used as a positive control. The negative control utilized the same section exposed to all conditions, except for the substitution of phosphate-buffered saline (PBS) for the primary antibody. Briefly, deparaffinized sections were exposed to microwave heating for uniform antigen retrieval. The primary antibody at 1:200 dilution was applied for 60 min followed by appropriate rinsing and application of a secondary antibody of biotinylated horse anti-mouse IgG (Vector, Burlingame, CA, USA) at 1:200 dilution. After rinsing, the Vector Elite ABC reagent was applied at a 1:50 dilution. After rinsing, DAB was applied and color development was monitored by light microscopy. The sections were scored for percentage of cells having positive nuclear staining. Growth curves Parental LNCaP, LNCaP-vector and/or LNCaP sublines transfected with p53 mutant alleles (G245S, R248W, R273H, R273C, P151S) were plated at 5000 cells/well in 96-well plates in the presence of RPMI 1640 containing 5% FBS. Each experimental group consisted of five replicate wells, and all experiments were repeated at least three times. For the conditioned media experiments, cells were allowed to attach overnight before switching media to conditioned media taken from LNCaP-vector or LNCaP transfected with p53 mutants that had been cultured for 3 days in RPMI 1640 supplemented with 5% charcoal-stripped FBS (Omega). For the rh relaxin experiments, parental LNCaP were plated in 0.5% CSS overnight and then 50 or 100 ng/ml rh relaxin (National Hormone and Peptide Program, Torrance, CA, USA) was added. Similar cultures treated with the synthetic androgen, R1881 (1 nM) (Sigma, Sigma, St Louis, MA, USA), were used as positive controls. MTT (3-[4,5-dimethylthiazol-2-yl]-2,5diphenyltetrazolium bromide; thiazolyl blue) (Sigma, St Louis, MA, USA) assays were performed to assess growth (Mosmann, 1983). MTT is converted to a colored formazan by mitochondrial dehydrogenases in viable cells and is a dependable correlate of cell proliferation. MTT (0.5 mg/ml in PBS, pH 7.4) was added to each well (10% (v/v)) and incubated for 3 h at 371C/5% CO2. After 3 h, media were aspirated and 175 ml of DMSO (Sigma) added to lyse cells and solubilize the formazan crystals. Plates were then placed on an orbital shaker for 15 min and read at 570 nm using a microplate reader.

Mutant p53 and H2 relaxin mediate androgen-independent growth RL Vinall et al

2091 Microarray studies Genome-wide expression profiling of LNCaP-p53GOF stable sublines was performed as previously described and according to standard protocols (Tepper et al., 2005; Affymetrix GeneChip Expression Analysis Technical Manual). Briefly, three separate clones of each LNCaP-p53GOF subline (G245S, R248W, R273C, R273H) and LNCaP-P151S (loss-of-function only) were subjected to acute androgen deprivation by culture in androgen-depleted medium (RPMI 1640 supplemented with 5% CSS) for 5 days. RNA was then extracted using TRIzol Reagent (Invitrogen, Carlsbad, CA, USA) as per the manufacturer’s instructions. Microarray analysis was performed by the UC Davis Cancer Center Gene Expression Resource using Affymetrix Human Genome U95Av2 (HGU95Av2) GeneChip arrays. These arrays permit expression analysis of 12 599 RNA transcripts. Data analysis (signal scaling and normalization) for each chip was performed with GeneChip Operating System and DNA-Chip Analyzer (Li and Wong, 2001) software algorithms. Scanned images were scaled to an average hybridization intensity of 125 corresponding to approximately two to three transcripts per cell. Comparison analysis was performed using perfect match-only modeling. For the identification of genes specifically upregulated in the LNCaP-R273H cell line, the data from triplicate clones was filtered to select for transcripts having a positive normalized probe intensity value and a mean fold change in expression of X1.8 compared to that of the LNCaP-vector cell line. Following this, the gene list generated was manually inspected for those probe sets detecting genes encoding known or putative polypeptide growth factors. Western blotting Protein was extracted from cells grown for 5 days in RPMI supplemented with 5% CSS using RIPA buffer (150 mM NaCl, 10 mM Tris-HCl, pH 8.0, 5 mM EDTA, 1% Triton X-100) containing 10 mg/ml leupeptin, 0.1 M aprotinin, 0.1 M PMSF and 0.1 M NaVO4 (Sigma). Following extraction, protein samples were sonicated, then nuclei and cellular debris were removed by centrifugation at 14 000 g for 15 min at 41C. Samples were stored at –701C before use. Protein concentration was determined using the BCA Protein Assay Reagent (Pierce, Rockford, IL, USA). Protein (50 mg) was separated on 15% SDS–PAGE minigels and transferred to nitrocellulose membranes. Gels were run under both reducing and nonreducing conditions. Membranes were blocked using 5% blotto (5% nonfat dried milk/Tris-buffered saline/0.1% Tween-20) and then probed with monoclonal antibodies that specifically recognize H2 relaxin (1:1000 dilution; ImmunDiagnostik, Bensheim, Germany), LGR7 (1:1000 dilution; Novus Biologicals, Littleton, CO, USA) and B-actin (1:10 000 dilution; Sigma). After washing with TBST, membranes were incubated with a horseradish peroxidase-linked anti-mouse secondary antibody (1:5000 dilution; Promega, Madison, WI, USA), washed again, and then incubated with enhanced chemiluminescence reagent (Amersham-Pharmacia, Piscataway, NJ, USA). Signals were detected using X-ray film (Kodak, Rochester, NY, USA). All experiments were repeated at least three times, representative blots are shown in figures. RT–PCR RNA was extracted from cells grown in RPMI 1640 containing 5% FBS or 5% CSS for 5 days using TRIzol Reagent (Invitrogen) as per the manufacturers directions. First-strand cDNA synthesis was performed using M-MLV Reverse Transcriptase (Applied Biosystems, Foster City, CA, USA) and a total of 1 mg total RNA. The following primers (IDT,

Coralville, IA, USA) were used to amplify H2 relaxin and LGR7: H2 relaxin – forward, 50 -CGGACTCATGGATGGA GGAAG, reverse, 50 -AACCAACATGGCAACATTTATTA GC; LGR7–forward, 50 -GTGGAGACAACAATGGATGG, reverse, 50 -AAGAAACCGATGGAACAGC. Amplification was carried out in an MJ PTC-100 thermal cycler (Bio-Rad, San Francisco, CA, USA). The following amplification conditions were used: an initial denaturation for 5 min at 941C, 30 cycles of 941C for 30 s, annealing for 45 s at 601C (relaxin)/551C (LGR7), extension at 721C for 45 s, followed by a polishing step for 10 min at 721C. At least three independent experiments were completed for each analysis, and blots representative of the combined data are shown in figures. Relaxin ELISA A commercial ELISA kit (Immundiagnostik) was used to measure the concentration of relaxin in conditioned media. It should be noted that this ELISA detects both H1 and H2 relaxin; however, to date expression of H1 relaxin protein has not been described in prostate. Conditioned media were collected from 1  105 cells, plated in triplicate, cultured in 24 well plates for 3 days in RPMI 1640 containing 5% FBS or 5% CSS. The concentration of relaxin in unconditioned media were also assessed. The kit, highly selective for human relaxin, has a detection limit of 0.4 pg/ml. RNAi Cells were plated overnight in the absence of antibiotics. Following the manufacturer’s instructions, Lipofectamine 2000 (Invitrogen) was used to transfect cells with SMARTPool siRNA (Dharmacon, Lafayette, CO, USA) that specifically downregulate H2 relaxin or LGR7 expression. SMARTPool siRNA comprises four individual siRNAs, each of which is designed to target a different area of the gene in question. A SMARTPool nonsilencing control was also used. The final concentration of SMARTPool siRNA added to cultures was 100 nM. To minimize toxicity of the transfection reagent, the medium was replaced 5 h following transfection. A nonsilencing FITC-conjugated control siRNA was used to assess transfection efficiency. To monitor downregulation of mRNA, RT–PCR analysis was performed 24 and 72 h after transfection. Soft agar assay Soft agar plates were prepared using 35 mm tissue culture dishes. A measure of 1.5 ml of 0.6% Noble agar (Sigma) in 2  RPMI 1640/5% FBS was poured into each plate to form a base. Cells (5000) were diluted 1:1 in 0.5 ml of the 0.6% Noble agar to give a final concentration of 0.3%. The cell mixture was poured on top of the hardened agar base and allowed to solidify. The soft agar cultures were placed at 371C in a 5% CO2 humidified atmosphere for 14–21 days before the frequency of colony formation in soft agar was calculated by dividing the total number of colonies formed by the number of cells plated. In some cases, cells used in this assay were pretreated with siRNA. Before culturing in soft agar, these cells were plated in monolayer culture and treated with H2 relaxin or control siRNA at a concentration of 100 nM for 24 h. Cells were then trypsinized and counted using a hemocytometer before being placed in soft agar. ChIP analysis LNCaP-vector and LNCaP-R273H cells were plated on 150 mm dishes and grown to 90% confluency in 5% CSS. The cells were then crosslinked for 8 min with final concentration of 1% formaldehyde in 10 mM NaCl, 0.1 mM EDTA and Oncogene

Mutant p53 and H2 relaxin mediate androgen-independent growth RL Vinall et al

2092 5 mM HEPES, pH 7.5. Cells were washed 2  with ice-cold PBS, harvested with a cell scraper and pelleted. Cell pellets were resuspended in ice-cold lysis-sonication buffer (10 mM EDTA, 50 mM Tris-HCl, pH 8, 0.5% SDS supplemented with complete protease inhibitor cocktail (Roche) and 0.2 mg/ml AEBSF) and sonicated using a Macrotip at power 1.6, duty cycle 33%, for 30 pulses, three times with 2 min on ice between each cycle. Suspensions were diluted fivefold with ice-cold dilution buffer (150 mM NaCl, 2 mM EDTA, 20 mM Tris-HCl, pH 8, 1% Triton X-100, complete protease inhibitor cocktail, 0.2 mg/ml AEBSF) and 200 ml was removed for input control. At this point, the samples were divided into two to allow for comparison between input versus p53-specific promoter binding. For the analysis of p53-specific promoter binding, complexes were immunoprecipitated with anti-p53 antibody (p53 Ab6; Oncogene) overnight at 41C, after which 2 mg sonicated salmon sperm was added. The immunocomplexes were captured by adding protein A–sepharose 4B beads (Zymed) and incubating for 2 h at 41C. Beads were pelleted and resuspended in TSE (0.1% SDS, 1% Triton X-100, 2 mM EDTA, 20 mM Tris-HCl, pH 8) þ 150 mM NaCl. Beads were washed in succession with TSE þ 150 mM NaCl, TSE þ 500 mM NaCl, Buffer III (0.25 M LiCl, 1% NP-40, 1% NaDOC, 1 mM EDTA, 10 mM Tris-HCl, pH 8) and then TE. Complexes were eluted with 1% SDS, 0.1 M NaHCO3 and reverse crosslinked at 651C overnight. Proteins were digested with Proteinase K for 1 h at 371C and the DNA fragments purified using the QIAquick PCR purification kit from Qiagen. In all, 10% of the ChIP product or 1% of the input control was used in each PCR reaction. PCR was performed using the following parameters: 1 min at 951C; 30 cycles of 1 min at 951C, 1 min at 551C, 1 min at 721C; and 5 min at 721C. Primers spanning the relaxin promoter region are forward 50 -CTGCTACTTGTAAGAGACACT-30 and reverse 50 -TGCTA AGGATCACAGGCAAAT-30 . Primers for the hTERTpositive control (the hTERT promoter has been reported to bind to mutant p53; Scian et al., 2004) are forward 50 -CTTGG CTTTCAGGATGGAGTAGCA-30 and reverse 50 -GGCTTCA AGGCTGGGAGGAAC-30 . Primers for the p21 promoterpositive control (p21 is known to bind to wild-type p53 and was used as a positive control to ensure the assay was working) are forward 50 -TATTGTGGGGCTGTTCTGCA-30 and reverse 50 -CTGTTAGAATGAGCCCCCTTT-30 .

Transfection and luciferase assay DU145 CaP cells (8000/well) were cultured in 24-well plates in CSS medium. DU145 CaP cells were used for these experiments because they lack expression of the AR. After 24 h, cells were transfected with the PSA promoter-firefly luciferase reporter plasmid and the pRL-SV40 Renilla luciferase plasmid (Promega, Madison, WI, USA) using the FuGene 6 transfection reagent. The next day, cells were fed with fresh CSS medium containing 0, 25 or 50 ng/ml rh H2 relaxin (National Hormone and Peptide Program) or with 50 ng/ml EGF (positive control; Culig et al., 1994). After 24 h, cells were harvested and lysated using passive lysis buffer (Promega). Luciferase activity was measured using a dual luciferase reporter assay (Promega) in an EG & G Berthold LB96V MicrolumatPlus microplate luminometer (Perkin-Elmer-Wallac Inc., Gaithersburg, MD, USA). Statistical analysis At least three independent experiments were completed for each analysis described in this paper. When appropriate, Student’s t-test was conducted to determine whether the treatment group was statistically significant compared to the control. *Po0.05, **Po0.005 and ***Po0.0005.

Abbreviations AI, androgen independent; CaP, prostate cancer; ChIP, chromatin immunoprecipitation; R273H p53 mutation, p53R273H; CSS, charcoal-stripped serum; rh, recombinant human; GOF, gain of function; LOF, loss of function; RNAi, RNA interference. Acknowledgements This study was funded by NCI RO1 CA77612 (RLV, XBS, RGE, DVW) and NCI RO192069 (XBS, RGE, DVW) grants. We thank Dr Philip Mack for his useful suggestions during this study, and Dr Arline Deitch for her careful reading of the manuscript. We also thank Dr Parlow of the National Hormone and Peptide Program for providing us with the recombinant human H2 relaxin.

References Affymetrix GeneChip Expression Analysis Technical Manual. Santa Clara, CA. Amorino GP, Parsons SJ. (2004). Crit Rev Eukaryot Gene Expr 14: 287–300. Bartsch O, Bartlick B, Ivell R. (2001). Mol Hum Reprod 7: 799–809. Bathgate RA, Samuel CS, Burazin TC, Gundlach AL, Tregear GW. (2003). Trends Endocrinol Metab 14: 207–213. Cadwell C, Zambetti GP. (2001). Gene 277: 15–30. Chan JM, Stampfer MJ, Giovannucci E, Gann PH, Ma J, Wilkinson P et al. (1998). Science 279: 563–566. Chin KV, Ueda K, Pastan I, Gottesman MM. (1992). Science 255: 459–462. Culig Z, Hobisch A, Cronauer MV, Radmayr C, Trapman J, Hittmair A et al. (1994). Cancer Res 54: 5474–5478. Culig Z, Steiner H, Bartsch G, Hobisch A. (2005). Endocr Relat Cancer 12: 229–244. Deb S, Jackson CT, Subler MA, Martin DW. (1992). J Virol 66: 6164–6170. Oncogene

Deb SP, Munoz RM, Brown DR, Subler MA, Deb S. (1994). Oncogene 9: 1341–1349. Dinjens WN, van der Weiden MM, Schroeder FH, Bosman FT, Trapman J. (1994). Int J Cancer 56: 630–633. El-Hizawi S, Lagowski JP, Kulesz-Martin M, Albor A. (2002). Cancer Res 62: 3264–3270. Figueiredo KA, Palmer JB, Mui AL, Nelson CC, Cox ME. (2005). Ann NY Acad Sci 1041: 320–327. Garibay-Tupas JL, Bao S, Kim MT, Tashima LS, BryantGreenwood GD. (2000). J Mol Endocrinol 24: 241–252. Gittes RF. (1991). N Engl J Med 324: 236–245. Hanahan D, Weinberg RA. (2000). Cell 100: 57–70. Heidenberg HB, Sesterhenn IA, Gaddipati JP, Weghorst CM, Buzard GS, Moul JW et al. (1995). J Urol 154: 414–421. Hsu SY, Kudo M, Chen T, Nakabayashi K, Bhalla A, van der Spek PJ et al. (2000). Mol Endocrinol 14: 1257–1271. Hsu SY, Nakabayashi K, Nishi S, Kumagai J, Kudo M, Sherwood OD et al. (2002). Science 295: 671–674. Huggins C, Hodges CV. (1972). CA Cancer J Clin 22: 232–240.

Mutant p53 and H2 relaxin mediate androgen-independent growth RL Vinall et al

2093 Ivell R, Einspanier A. (2002). Trends Endocrinol Metab 13: 343–348. Ivell R, Hunt N, Khan-Dawood F, Dawood MY. (1989). Mol Cell Endocrinol 66: 251–255. Kasamon KM, Dawson NA. (2004). Curr Opin Urol 14: 185–193. Koul HK, Maroni PD, Meacham RB, Crawford D, Koul S. (2004). Ann NY Acad Sci 1030: 243–252. Kyprianou N, English HF, Isaacs JT. (1990). Cancer Res 50: 3748–3753. Lanyi A, Deb D, Seymour RC, Ludes-Meyers JH, Subler MA, Deb S. (1998). Oncogene 16: 3169–3176. Lee LF, Louie MC, Desai SJ, Yang J, Chen HW, Evans CP et al. (2004). Oncogene 23: 2197–2205. Li C, Wong WH. (2001). Proc Natl Acad Sci USA 98: 31–36. Mosmann T. (1983). J Immunol Methods 65: 55–63. Navone NM, Troncoso P, Pisters LL, Goodrow TL, Palmer JL, Nichols WW et al. (1993). J Natl Cancer Inst 85: 1657–1669. Nesslinger NJ, Shi XB, deVere White RW. (2003). Cancer Res 63: 2228–2233. Olivier M, Eeles R, Hollstein M, Khan MA, Harris CC, Hainaut P. (2002). Hum Mutat 19: 607–614. Palejwala S, Tseng L, Wojtczuk A, Weiss G, Goldsmith LT. (2002). Biol Reprod 66: 1743–1748. Petrylak DP, Tangen CM, Hussain MH, Lara Jr PN, Jones JA, Taplin ME et al. (2004). N Engl J Med 351: 1513–1520. Raj GV, Barki-Harrington L, Kue PF, Daaka Y. (2002). J Urol 167: 1458–1463. Roemer K. (1999). Biol Chem 380: 879–887. Samuel CS, Tian H, Zhao L, Amento EP. (2003). Lab Invest 83: 1055–1067. Scian MJ, Stagliano KE, Deb D, Ellis MA, Carchman EH, Das A et al. (2004). Oncogene 23: 4430–4443.

Shah RB, Mehra R, Chinnaiyan AM, Shen R, Ghosh D, Zhou M et al. (2004). Cancer Res 64: 9209–9216. Sherwood OD. (2004). Endocr Rev 25: 205–234. Shi XB, Gandour-Edwards R, Beckett LA, Deitch AD, de Vere White RW. (2004a). BJU Int 94: 996–1002. Shi XB, Ma AH, Tepper CG, Xia L, Gregg JP, GandourEdwards R et al. (2004b). Prostate 60: 257–271. Shi XB, Nesslinger NJ, Deitch AD, Gumerlock PH, deVere White RW. (2002). Prostate 51: 59–72. Silvertown JD, Geddes BJ, Summerlee AJ. (2003a). Endocrinology 144: 3683–3691. Silvertown JD, Ng J, Sato T, Summerlee AJ, Medin JA. (2006). Int J Cancer 118: 62–73. Silvertown JD, Summerlee AJ, Klonisch T. (2003b). Int J Cancer 107: 513–519. Stemmermann GN, Mesiona W, Greenwood FC, BryantGreenwood GD. (1994). J Endocrinol 140: 321–325. Tannock IF, de Wit R, Berry WR, Horti J, Pluzanska A, Chi KN et al. (2004). N Engl J Med 351: 1502–1512. Tashima LS, Mazoujian G, Bryant-Greenwood GD. (1994). J Mol Endocrinol 12: 351–364. Tepper CG, Gregg JP, Shi XB, Vinall RL, Baron CA, Ryan PE et al. (2005). Prostate 65: 375–389. van Oijen MG, Slootweg PJ. (2000). Clin Cancer Res 6: 2138– 2145. Vogelstein B, Lane D, Levine AJ. (2000). Nature 408: 307–310. Welsh JB, Sapinoso LM, Kern SG, Brown DA, Liu T, Bauskin AR et al. (2003). Proc Natl Acad Sci USA 100: 3410–3415. Zhang Q, Liu SH, Erikson M, Lewis M, Unemori E. (2002). J Cell Biochem 85: 536–544.

Oncogene