In humans and rodents, the prostate gland develops from the embryonic urogenital sinus (UGS). The androgen receptor (AR) is thought to control the expression of morphogenetic genes in inductive UGS mesenchyme, which promotes proliferation and cytodifferentiation of the prostatic epithelium. However, the nature of the AR-regulated morphogenetic genes and the mechanisms whereby AR controls prostate development are not understood. Glial cell line-derived neurotrophic factor (GDNF) binds GDNF family receptor α1 (GFRα1) and signals through activation of RET tyrosine kinase. Gene disruption studies in mice have revealed essential roles for GDNF signaling in development; however, its role in prostate development is unexplored. Here, we establish novel roles of GDNF signaling in mouse prostate development. Using an organ culture system for prostate development and Ret mutant mice, we demonstrate that RET-mediated GDNF signaling in UGS increases proliferation of mesenchyme cells and suppresses androgen-induced proliferation and differentiation of prostate epithelial cells, inhibiting prostate development. We also identify Ar as a GDNF-repressed gene and Gdnf and Gfrα1 as androgen-repressed genes in UGS, thus establishing reciprocal regulatory crosstalk between AR and GDNF signaling in prostate development.

The prostate gland is formed from the embryonic urogenital sinus (UGS), where the pelvic urethra joins the bladder. Morphogenesis of the mouse prostate begins in the pelvic urethra of the UGS around embryonic day (E) 16.5 and is dependent on the inductive properties of the urethral mesenchyme (UrM) (Georgas et al., 2015). In rodents, the UrM is composed of discrete spatial regions with different properties. The peri-urethral mesenchyme (PUM) surrounds the urethral epithelium (UrE) and newly formed prostatic buds. The inductive UrM, which includes the ventral mesenchymal pad (VMP), is peripheral to the PUM and is crucial for prostatic induction (Thomson et al., 2002; Timms et al., 1995, 1994). During induction, mesenchymal cells at the interface of the PUM and VMP differentiate into smooth muscle cells, which surround the prostate epithelial (PrE) ducts as they elongate in the multilobed rodent prostate (Hayward et al., 1998; Thomson et al., 2002; Timms et al., 1995, 1994).

Androgen receptor (AR) signaling is required for prostate development (Cunha and Chung, 1981; Cunha and Lung, 1978; Donjacour and Cunha, 1988; Lasnitzki and Mizuno, 1980), and disruption of AR-mediated prostate development predisposes humans and rodents to prostate neoplasia by altering the phenotype of the gland early in life (Anway and Skinner, 2008; Gupta, 2000; Ho et al., 2006; Hu et al., 2012; Prins et al., 2014, 2008; Ramos et al., 2001; Timms et al., 2005). Testosterone and its potent metabolite 5α-dihydrotestosterone (DHT) activate the AR during prostate development. AR action in the inductive UrM is thought to induce formation of PrE progenitors/buds in the UrE through paracrine signaling (Cunha and Chung, 1981; Cunha and Lung, 1978; Lasnitzki and Mizuno, 1980). Androgen-dependent signaling also stimulates outgrowth, cytodifferentiation and branching morphogenesis of the PrE buds as they give rise to a network of prostatic ducts (Cunha and Chung, 1981; Cunha and Lung, 1978; Donjacour and Cunha, 1988; Sugimura et al., 1986).

AR signaling acts in concert with other signaling pathways to regulate prostate development. Outgrowth of the PrE is dictated by time-specific and cell-specific expression of morphogenetic genes (Prins and Putz, 2008; Thomson and Marker, 2006), including secreted signaling ligands of the hedgehog (Berman et al., 2004; Doles et al., 2006; Freestone et al., 2003; Lamm et al., 2002; Podlasek et al., 1999; Pu et al., 2004; Wang et al., 2003), Wnt (Allgeier et al., 2008; Huang et al., 2009; Joesting et al., 2008; Keil et al., 2012), FGF (Alarid et al., 1994; Donjacour et al., 2003; Sugimura et al., 1996; Thomson and Cunha, 1999) and TGFβ (Cancilla et al., 2001; Cook et al., 2007; Grishina et al., 2005; Itoh et al., 1998; Lamm et al., 2001; Tanji et al., 1994; Tomlinson et al., 2004) gene super-families. These morphogens are thought to communicate autocrine and paracrine signals through their cognate receptors and downstream signaling pathways. However, the nature of the AR-regulated morphogenetic genes is not understood.

Glial cell line-derived neurotrophic factor (GDNF) is a member of the TGFβ protein super-family and regulates spermatogenesis, neuronal survival and differentiation, and kidney development (Arenas et al., 1995; Davies et al., 1995; Henderson et al., 1994; Lin et al., 1993; Meng et al., 2000; Pepicelli et al., 1997; Pichel et al., 1996; Sainio et al., 1997; Vega et al., 1996). Cellular responses to GDNF are mediated by a multicomponent receptor complex consisting of GDNF family receptor α1 (GFRα1) and RET, a receptor tyrosine kinase (Jing et al., 1996; Trupp et al., 1996; Vega et al., 1996). GDNF signaling plays crucial roles in development; however, its role in prostate development has not been explored previously.

We recently reported the localization of GDNF, GFRα1 and RET in the murine UGS. Prior to prostatic induction, GDNF and GFRα1 are expressed in the urethral mesenchyme (UrM) and epithelium (UrE), whereas RET is restricted to the UrM, including the VMP (Park and Bolton, 2015). In addition, we demonstrated that GDNF increases proliferation of UrM cells and UrE cells in naïve UGSs, which were not androgen induced (Park and Bolton, 2015). Our previous findings suggest that GDNF signaling may influence additional aspects of UGS development. Based on our published data and on the requirement of AR signaling in prostatic induction and development, we explore the intriguing hypothesis that the AR and GDNF signaling pathways may interact and influence androgen-induced prostate development.

Here, we report that GDNF signaling antagonizes androgen-induced prostate morphogenesis. In support of this, we show that androgen induces PrE budding, proliferation and cytodifferentiation in the developing prostate, whereas GDNF signaling inhibits these androgen-dependent effects. Using Ret-disrupted mice, we further demonstrate that RET-mediated signaling in the UrM is responsible for the inhibitory effects of GDNF on prostate development, including suppression of androgen-induced proliferation and differentiation of PrE cells. In addition to Ar being GDNF-repressed by RET-mediated signaling, Gdnf and Gfra1 are androgen repressed during prostate development, thus establishing reciprocal regulatory crosstalk between AR and GDNF signaling in androgen-induced prostate development.

GDNF signaling inhibits androgen-induced prostate development in UGS organ culture

We investigated the functional roles of AR and GDNF signaling in androgen-induced prostate development using UGS organ culture, which recapitulates the earliest stages of prostate development in vivo (Doles et al., 2005; Ghosh et al., 2011; Kuslak and Marker, 2007). As AR activation is necessary for prostate development in vivo and ex vivo (Allgeier et al., 2010; Cunha and Chung, 1981; Cunha and Lung, 1978; Doles et al., 2005; Marker et al., 2003), we cultured UGSs from E15.5 male embryos in defined organ culture medium supplemented with 10 nM DHT for 7 days. These UGSs were also co-treated with 10-100 ng/ml GDNF or vehicle as a control. As expected, DHT induced PrE budding in UGSs cultured in the absence of GDNF (Fig. 1A), which is consistent with previous reports of prostate development ex vivo (Allgeier et al., 2010; Doles et al., 2005; Marker et al., 2003). UGSs co-treated with GDNF displayed thicker UrM (Fig. 1A), which is consistent with the effect of GDNF in naïve UGSs (Park and Bolton, 2015). In addition, the co-treated UGSs showed striking changes in morphology, including significant decreases in the number and length of PrE buds that emerged from the UrE (Fig. 1B,C).

Fig. 1.

GDNF signaling inhibits androgen-induced prostate development in UGS organ culture. (A) UGSs from E15.5 male mouse embryos were cultured with 0-100 ng/ml GDNF for 7 days in the absence or presence of 10 nM DHT, which induces prostate development. DHT induced prostatic epithelial budding (black arrowheads) compared with vehicle controls. PrE budding deficiency and thicker UrM were observed in DHT-induced UGSs co-treated with 10-100 ng/ml GDNF. Below the images, the boundary of the UrM has been traced and labeled to distinguish it from the UrE, PrE and seminal vesicles (SV). Quantification of the average bud number (B) and length (C) demonstrates significant, dose-dependent decreases in DHT-induced UGSs co-treated with 10-100 ng/ml GDNF. Data are mean±s.e.m. (n=5 or 6). A significance level of 0.05 was applied during data analysis using ANOVA, and different significance levels have been indicated using different lowercase letters. (D) Immunohistochemistry reveals the altered morphology of the ventral and lateral prostatic epithelium (VPE, LPE) in DHT-induced UGSs co-treated with 100 ng/ml GDNF compared with vehicle for 7 days. Immunodetection of epithelial keratins (green) identifies UrE and PrE cells, whereas vimentin localization (red) identifies mesenchymal cells. Cell nuclei are stained with DAPI (blue). Boundaries between the peri-urethral mesenchyme (PUM) and the UrE/PrE have been traced (dashed white line), and the ventral mesenchymal pad (VMP) has been labeled. Scale bars: 500 µm in A; 100 µm in D.

Fig. 1.

GDNF signaling inhibits androgen-induced prostate development in UGS organ culture. (A) UGSs from E15.5 male mouse embryos were cultured with 0-100 ng/ml GDNF for 7 days in the absence or presence of 10 nM DHT, which induces prostate development. DHT induced prostatic epithelial budding (black arrowheads) compared with vehicle controls. PrE budding deficiency and thicker UrM were observed in DHT-induced UGSs co-treated with 10-100 ng/ml GDNF. Below the images, the boundary of the UrM has been traced and labeled to distinguish it from the UrE, PrE and seminal vesicles (SV). Quantification of the average bud number (B) and length (C) demonstrates significant, dose-dependent decreases in DHT-induced UGSs co-treated with 10-100 ng/ml GDNF. Data are mean±s.e.m. (n=5 or 6). A significance level of 0.05 was applied during data analysis using ANOVA, and different significance levels have been indicated using different lowercase letters. (D) Immunohistochemistry reveals the altered morphology of the ventral and lateral prostatic epithelium (VPE, LPE) in DHT-induced UGSs co-treated with 100 ng/ml GDNF compared with vehicle for 7 days. Immunodetection of epithelial keratins (green) identifies UrE and PrE cells, whereas vimentin localization (red) identifies mesenchymal cells. Cell nuclei are stained with DAPI (blue). Boundaries between the peri-urethral mesenchyme (PUM) and the UrE/PrE have been traced (dashed white line), and the ventral mesenchymal pad (VMP) has been labeled. Scale bars: 500 µm in A; 100 µm in D.

To assess further the PrE-budding defect in the developing prostate rudiments caused by GDNF, we used immunohistochemistry to visualize the outgrowth of the ventral and lateral PrE buds. Immunodetection of epithelial keratins has been shown to identify the epithelial cells of the UrE and PrE buds and ducts, whereas vimentin localization identifies mesenchymal cells. Consistent with our PrE-budding data (Fig. 1B,C), the number and length of PrE buds that elongated from the UrE were dramatically decreased in DHT-induced UGSs co-treated with GDNF compared with vehicle (Fig. 1D). Thicker UrM were also evident in UGSs co-treated with GDNF (Fig. 1D). Taken together, these results demonstrate that GDNF signaling inhibits androgen-induced prostate development.

GDNF signaling stimulates proliferation of UrM cells and inhibits androgen-induced proliferation of PrE cells in the developing prostate

In a previous study, we demonstrated that UrM thickness in the naïve UGS is selectively regulated by GFRα1 and RET kinase activity in response to GDNF (Park and Bolton, 2015). Here, we investigated the effects of androgen and GDNF on proliferation of mesenchymal cells and epithelial cells in the developing prostate by quantifying bromodeoxyuridine (BrdU) incorporation in the DNA of dividing cells. Androgen-supplemented E15.5 UGSs were cultured with 100 ng/ml GDNF or vehicle for 2-7 days, and BrdU was added before tissue processing for immunohistochemistry.

Immunodetection of BrdU revealed an increase in BrdU-positive UrM cells and a decrease in BrdU-positive PrE cells in androgen-induced UGSs co-treated with GDNF for 2 days (Fig. 2A). Quantification of the percentage of BrdU-positive cells in the UrM and the PrE outgrowths from the UrE indeed demonstrates a robust increase in proliferation of UrM cells and a substantial decrease in proliferation of PrE cells in DHT-induced UGSs co-treated with GDNF compared with vehicle (Fig. 2B). Notably, GDNF induced proliferation to a greater extent in the peripheral, inductive UrM compared with the PUM (Fig. 2A).

Fig. 2.

GDNF signaling stimulates proliferation of UrM cells and inhibits androgen-induced proliferation of PrE cells in the developing prostate. (A-D) E15.5 UGSs were cultured with 10 nM DHT and 100 ng/ml GDNF or vehicle for (A,B) 2 days and (C,D) 7 days. BrdU (10 µM) was added 2 h before fixation. (A,C) Representative images are shown for immunodetection of BrdU-positive cells (green), vimentin-positive mesenchymal cells (red) and DAPI-stained nuclei (blue). DHT-induced UGSs co-treated with GDNF display thicker UrM than those co-treated with vehicle. (B,D) Quantification of the percentage of BrdU-positive cells in vimentin-positive UrM and vimentin-negative PrE revealed a 3.7-fold increase in proliferation of UrM cells in DHT-induced UGSs co-treated with GDNF for 2 days compared with UGSs treated with DHT alone (n=5). In contrast, proliferation of PrE cells decreases 64% in DHT-induced UGSs co-treated with GDNF compared with UGSs treated with DHT alone (n=5). UGSs cultured in the absence of DHT and GDNF show baseline proliferation (n=4). (C) The UGSs co-treated with DHT and GDNF displayed thicker UrM compared with those treated with DHT alone; (D) however, the stimulatory effect of GDNF on UrM proliferation dissipated by day 7 (n=5). In contrast, suppression of DHT-induced PrE proliferation in response to GDNF persisted, albeit to a lesser extent, on day 7 (n=5). A significance level of 0.05 was applied during data analysis using ANOVA, and different significance levels have been indicated using different lowercase letters. Scale bars: 100 µm.

Fig. 2.

GDNF signaling stimulates proliferation of UrM cells and inhibits androgen-induced proliferation of PrE cells in the developing prostate. (A-D) E15.5 UGSs were cultured with 10 nM DHT and 100 ng/ml GDNF or vehicle for (A,B) 2 days and (C,D) 7 days. BrdU (10 µM) was added 2 h before fixation. (A,C) Representative images are shown for immunodetection of BrdU-positive cells (green), vimentin-positive mesenchymal cells (red) and DAPI-stained nuclei (blue). DHT-induced UGSs co-treated with GDNF display thicker UrM than those co-treated with vehicle. (B,D) Quantification of the percentage of BrdU-positive cells in vimentin-positive UrM and vimentin-negative PrE revealed a 3.7-fold increase in proliferation of UrM cells in DHT-induced UGSs co-treated with GDNF for 2 days compared with UGSs treated with DHT alone (n=5). In contrast, proliferation of PrE cells decreases 64% in DHT-induced UGSs co-treated with GDNF compared with UGSs treated with DHT alone (n=5). UGSs cultured in the absence of DHT and GDNF show baseline proliferation (n=4). (C) The UGSs co-treated with DHT and GDNF displayed thicker UrM compared with those treated with DHT alone; (D) however, the stimulatory effect of GDNF on UrM proliferation dissipated by day 7 (n=5). In contrast, suppression of DHT-induced PrE proliferation in response to GDNF persisted, albeit to a lesser extent, on day 7 (n=5). A significance level of 0.05 was applied during data analysis using ANOVA, and different significance levels have been indicated using different lowercase letters. Scale bars: 100 µm.

DHT-induced UGSs co-treated with GDNF for 7 days displayed thicker UrM (Fig. 2C). However, the percentage of BrdU-positive UrM cells was similar in UGSs co-treated with GDNF or vehicle (Fig. 2D). In comparison with the robust increase of BrdU-positive UrM cells in UGSs co-treated with DHT and GDNF on day 2 (Fig. 2B), the results on day 7 indicate that GDNF-induced proliferation of UrM cells is time-sensitive during prostate development (compare Fig. 2B,D). Quantification of BrdU-positive cells in the PrE outgrowths from the UrE showed prolonged suppression of DHT-induced proliferation in UGSs co-treated with GDNF for 7 days (Fig. 2D). Although PrE growth suppression was less pronounced on day 7 compared with day 2 (compare Fig. 2B,D). These findings indicate that elevated levels of GDNF induce morphological changes in the developing prostate that are driven by enhanced proliferation of UrM cells and inhibited proliferation of PrE cells.

GDNF signaling upregulates RET expression and localization in the VMP of the developing prostate

To begin to investigate the cellular and molecular mechanism responsible for the effects of GDNF on prostate development, we examined the expression and localization of RET, which forms a co-receptor complex with GFRα1 and mediates GDNF signaling (Jing et al., 1996; Trupp et al., 1996; Vega et al., 1996). Using real-time quantitative polymerase chain reaction (QPCR) analysis, we determined the extent of RET regulation by GDNF signaling in androgen-induced prostate development. Ret mRNA increased substantially in DHT-induced UGSs co-treated with GDNF compared with vehicle (Fig. S1A). Transcripts coding for sprouty 1 (Spry1), an antagonistic regulator of RET-mediated signaling, also increased in co-treated UGSs (Fig. S1B). Consistent with the increase in Ret mRNA, GDNF exposure increased RET protein expression (Fig. S1C). Immunohistochemistry analysis revealed a robust increase in the number of UrM cells expressing RET and a significant increase in RET expression per cell in the VMP of DHT-induced UGSs co-treated with GDNF compared with vehicle (Fig. S1D,E). In contrast, RET localization was absent from the UrE and PrE (Fig. S1D). The increases in RET expression and localization suggest that downstream targets of GDNF signaling in the UrM may regulate transcription of the Ret gene.

In addition, we examined RET localization in the prostate of postnatal day (P) 0 neonates. Immunodetection of fibroblast growth factor 10 (FGF10), which is expressed and secreted from the inductive UrM cells of the VMP (Thomson and Cunha, 1999), and RET revealed robust expression and co-localization of RET and FGF10 in inductive UrM cells of the VMP (Fig. S1F). RET localization was absent from UrE and PrE regions in P0 prostate (Fig. S1F). The colocalization of RET and FGF10 in the VMP demonstrates that RET is indeed expressed in inductive UrM cells during prostate development.

Smooth muscle actin expression and localization are unaffected by GDNF signaling in the developing prostate

During prostatic induction, mesenchymal cells at the interface of the PUM and VMP undergo smooth muscle differentiation, which has been suggested to influence PrE budding and outgrowth from the UrE (Hayward et al., 1998; Thomson et al., 2002; Timms et al., 1995, 1994). To determine whether GDNF signaling affects smooth muscle differentiation in androgen-supplemented UGSs, we examined the localization and expression of smooth muscle actin (SMA), a marker of smooth muscle cells. Immunohistochemistry revealed layers of smooth muscle cells encircle the ventral PrE in DHT-induced E15.5 UGSs regardless of whether they were co-treated with GDNF or vehicle for 7 days (Fig. S2A). Immunoblot analysis of DHT-induced UGSs also demonstrates that smooth muscle actin expression was unaffected by GDNF exposure for 2-7 days (Fig. S2B). Additional immunohistochemistry analysis revealed similar numbers of UrM cells expressing SMA and no significant difference in SMA expression per cell at sites of prostatic budding in DHT-induced UGSs co-treated with GDNF compared with vehicle (Fig. S2C,D). These results indicate that smooth muscle differentiation during prostate development is unaffected by GDNF signaling.

GDNF signaling inhibits androgen-induced PrE cytodifferentiation in the developing prostate

Androgen-dependent signaling during prostate development stimulates outgrowth and cytodifferentiation of PrE buds as they give rise to a network of prostatic ducts (Cunha and Chung, 1981; Cunha and Lung, 1978; Donjacour and Cunha, 1988; Sugimura et al., 1986). Since GDNF antagonizes androgen-induced PrE budding, we sought to determine whether GDNF influences cytodifferentiation of PrE buds formed in the presence of GDNF. We examined PrE cytodifferentiation based on expression and localization of Keratins 8 and 18 (KRT8 and KRT18), which are abundantly expressed in luminal epithelial cells of the prostate (Achtstätter et al., 1985; Hsieh et al., 1992; Soeffing and Timms, 1995; Verhagen et al., 1988). In baseline experiments, we induced prostate development in E15.5 UGSs with DHT for 0-7 days and assessed expression and localization of KRT5 (a basal epithelial keratin) and KRT8 (a luminal epithelial keratin) using immunohistochemistry (Fig. 3A). As expected, colocalization of KRT5 and KRT8 (orange) was observed in the UrE throughout the time course, and KRT5 localization was abundant in PrE bud cells that emerged from the UrE (Fig. 3A). By day 7 in DHT-induced UGSs, abundant KRT8 localization was evident in the central epithelial cells of elongated prostatic buds, and KRT5 localization persisted (Fig. 3A).

Fig. 3.

GDNF signaling inhibits androgen-induced PrE cytodifferentiation in the developing prostate. (A) E15.5 UGSs were androgen induced with 10 nM DHT, and PrE cytodifferentiation was assessed using immunohistochemistry on days 0, 2, 4 and 7. Hematoxylin and Eosin-stained sections of the developing prostate are shown. Immunodetection of KRT8 (green) and KRT5 (red), and DAPI-stained nuclei (blue) show androgen-induced outgrowth of KRT8-negative PrE buds from the KRT8-positive UrE on day 2 and day 4. By day 7, KRT8-positive luminal epithelial cells are present at the midline of the elongating PrE bud and along the luminal surfaces of the developing duct. (B) E15.5 UGSs were cultured for 7 days with 10 nM DHT and 100 ng/ml GDNF or vehicle. KRT8-positive luminal epithelial cells are absent from PrE buds in DHT-induced UGSs co-treated with GDNF compared with UGSs treated with DHT alone. (C) QPCR analysis of UGS cDNA shows that Krt8 and Krt18 mRNA levels were significantly decreased, and Krt14 and Krt5 mRNA levels were increased in UGSs co-treated with GDNF compared with those treated with DHT alone. Data are mean±s.e.m. (n=5) in log2 scale, *P<0.05 and **P<0.01. Scale bars: 200 µm in A (top panels); 50 µm in A (middle and bottom panels) and B.

Fig. 3.

GDNF signaling inhibits androgen-induced PrE cytodifferentiation in the developing prostate. (A) E15.5 UGSs were androgen induced with 10 nM DHT, and PrE cytodifferentiation was assessed using immunohistochemistry on days 0, 2, 4 and 7. Hematoxylin and Eosin-stained sections of the developing prostate are shown. Immunodetection of KRT8 (green) and KRT5 (red), and DAPI-stained nuclei (blue) show androgen-induced outgrowth of KRT8-negative PrE buds from the KRT8-positive UrE on day 2 and day 4. By day 7, KRT8-positive luminal epithelial cells are present at the midline of the elongating PrE bud and along the luminal surfaces of the developing duct. (B) E15.5 UGSs were cultured for 7 days with 10 nM DHT and 100 ng/ml GDNF or vehicle. KRT8-positive luminal epithelial cells are absent from PrE buds in DHT-induced UGSs co-treated with GDNF compared with UGSs treated with DHT alone. (C) QPCR analysis of UGS cDNA shows that Krt8 and Krt18 mRNA levels were significantly decreased, and Krt14 and Krt5 mRNA levels were increased in UGSs co-treated with GDNF compared with those treated with DHT alone. Data are mean±s.e.m. (n=5) in log2 scale, *P<0.05 and **P<0.01. Scale bars: 200 µm in A (top panels); 50 µm in A (middle and bottom panels) and B.

Next, we cultured androgen-supplemented E15.5 UGSs with GDNF or vehicle for 7 days, and assessed expression and localization of KRT5 and KRT8 (Fig. 3B). In DHT-induced UGSs co-treated with vehicle, abundant localization of KRT5 and KRT8 was evident in the central epithelial cells of the prostatic buds. In contrast, the small prostatic buds formed in UGSs co-treated with GDNF displayed abundant KRT5 localization, but they lacked KRT8 (Fig. 3B). Consistent with our immunohistochemistry data, QPCR analysis revealed significant downregulation of luminal Krt8 and Krt18 mRNAs, and upregulation of basal Krt14 and Krt5 mRNAs in DHT-induced UGSs co-treated with GDNF compared with vehicle (Fig. 3C). Taken together, these results suggest that GDNF signaling inhibits AR-mediated cytodifferentiation of luminal PrE cells.

RET-mediated GDNF signaling inhibits androgen-induced prostate development

In addition to RET-mediated signaling by GDNF, RET-independent mechanisms of GDNF action have also been reported (Cao et al., 2008; Panicker et al., 2003; Paratcha et al., 2003; Poteryaev et al., 1999; Trupp et al., 1999). To determine the functional role of RET-mediated signaling in androgen-induced prostate development, we assessed RET protein expression and examined PrE budding using UGSs from wild-type (Ret WT) and Ret-null (Ret KO) embryos. Notably, Ret KO mice die at birth due to complications associated with renal agenesis (Enomoto et al., 2001; Jain et al., 2004; Schuchardt et al., 1994). The Ret WT and mutant genotypes of embryos were distinguished using PCR (Fig. 4A). Immunoblot analysis of DHT-induced E15.5 UGSs confirmed the presence of RET in Ret WT UGSs and an absence of RET in Ret KO UGSs (Fig. 4B).

Fig. 4.

RET-mediated GDNF signaling inhibits androgen-induced prostate development. (A) Genotypes of Ret wild-type and Ret mutant embryos were distinguished using PCR. The wild-type allele produces a 451 bp amplicon and the Ret-null allele produces a 350 bp amplicon. (B-F) E15.5 UGSs from Ret wild-type (Ret WT) males and Ret-null (Ret KO) males were cultured with 10 nM DHT and 100 ng/ml GDNF or vehicle for 7 days. (B) Immunoblots using lysates from Ret WT prostate rudiments demonstrate RET expression, which is absent from Ret KO rudiments. (C) DHT induced PrE budding (black arrowheads) in Ret WT UGSs and Ret KO UGSs co-treated with vehicle. PrE budding deficiency and thicker UrM were observed in Ret WT UGSs co-treated with GDNF; however, these effects were absent from Ret KO UGSs co-treated with GDNF. Quantification of the average bud number (D) and length (E) demonstrates significant decreases in Ret WT UGSs co-treated with DHT and GDNF compared with those treated with DHT alone. In contrast, PrE bud number and length remained unchanged in DHT-induced Ret KO UGSs, regardless of GDNF exposure. Data are mean±s.e.m. (n=5-7). A significance level of 0.05 was applied during data analysis using ANOVA, and different significance levels have been indicated using different lowercase letters. (F) Immunohistochemistry reveals the altered morphology of the ventral and lateral prostatic epithelium (VPE, LPE) in Ret WT UGSs co-treated with DHT and GDNF compared with those treated with DHT alone for 7 days. Immunodetection of epithelial keratins (green) identifies UrE and PrE cells, whereas RET localization (red) identifies the VMP of Ret WT prostate rudiments. Scale bars: 500 µm in C; 100 µm in F.

Fig. 4.

RET-mediated GDNF signaling inhibits androgen-induced prostate development. (A) Genotypes of Ret wild-type and Ret mutant embryos were distinguished using PCR. The wild-type allele produces a 451 bp amplicon and the Ret-null allele produces a 350 bp amplicon. (B-F) E15.5 UGSs from Ret wild-type (Ret WT) males and Ret-null (Ret KO) males were cultured with 10 nM DHT and 100 ng/ml GDNF or vehicle for 7 days. (B) Immunoblots using lysates from Ret WT prostate rudiments demonstrate RET expression, which is absent from Ret KO rudiments. (C) DHT induced PrE budding (black arrowheads) in Ret WT UGSs and Ret KO UGSs co-treated with vehicle. PrE budding deficiency and thicker UrM were observed in Ret WT UGSs co-treated with GDNF; however, these effects were absent from Ret KO UGSs co-treated with GDNF. Quantification of the average bud number (D) and length (E) demonstrates significant decreases in Ret WT UGSs co-treated with DHT and GDNF compared with those treated with DHT alone. In contrast, PrE bud number and length remained unchanged in DHT-induced Ret KO UGSs, regardless of GDNF exposure. Data are mean±s.e.m. (n=5-7). A significance level of 0.05 was applied during data analysis using ANOVA, and different significance levels have been indicated using different lowercase letters. (F) Immunohistochemistry reveals the altered morphology of the ventral and lateral prostatic epithelium (VPE, LPE) in Ret WT UGSs co-treated with DHT and GDNF compared with those treated with DHT alone for 7 days. Immunodetection of epithelial keratins (green) identifies UrE and PrE cells, whereas RET localization (red) identifies the VMP of Ret WT prostate rudiments. Scale bars: 500 µm in C; 100 µm in F.

To determine the role of RET in PrE budding, androgen-supplemented E15.5 UGSs from Ret WT males and Ret KO males were co-treated with GDNF or vehicle for 7 days (Fig. 4C). DHT induced PrE budding in Ret WT UGSs and Ret KO UGSs co-treated with vehicle, indicating that RET-mediated signaling is not required for prostatic induction and PrE budding (Fig. 4C). PrE-budding deficiency and thicker UrM were observed in DHT-induced Ret WT UGSs co-treated with GDNF (Fig. 4C). Further, quantification of PrE budding showed significant decreases in PrE bud number and length in Ret WT UGSs co-treated with GDNF compared with vehicle (Fig. 4D,E). However, these effects of GDNF were absent from DHT-induced Ret KO UGSs exposed to GDNF (Fig. 4C-E).

Using immunohistochemistry, we also compared the outgrowth of PrE buds from Ret WT UGSs and Ret KO UGSs (Fig. 4F). Of the androgen-induced UGSs from Ret WT males and Ret KO males, only Ret WT UGSs co-treated with GDNF displayed thicker UrM (Fig. 4F). Consistent with our PrE-budding data (Fig. 4D,E), the number and length of PrE buds that elongated from the UrE decreased substantially in DHT-induced Ret WT UGSs co-treated with GDNF compared with vehicle. In contrast, PrE budding and UrM thickness of Ret KO UGSs were unaffected by GDNF exposure (Fig. 4F). Taken together, these results demonstrate that RET-mediated GDNF signaling inhibits androgen-induced PrE budding.

RET-mediated GDNF signaling inhibits androgen-induced proliferation and cytodifferentiation of PrE cells in the developing prostate

The role of RET signaling in proliferation of UrM and PrE cells was examined by immunohistochemistry analysis of androgen-supplemented E15.5 UGSs from Ret WT males and Ret KO males co-treated with GDNF or vehicle for 7 days (Fig. 5). Of the DHT-induced UGSs from Ret WT males and Ret KO males, only Ret WT UGSs co-treated with GDNF displayed thicker UrM (Fig. 5A), which is consistent with results in Fig. 4C,F. Quantification of BrdU-positive UrM cells revealed similar proportions of proliferating cells in Ret WT UGSs and Ret KO UGSs regardless of GDNF exposure (Fig. 5B), which is consistent with GDNF-induced proliferation of UrM cells being time-sensitive during prostate development (Fig. 2C,D). The percentage of BrdU-positive cells in the PrE outgrowths from the UrE showed prolonged suppression of DHT-induced proliferation in Ret WT UGSs co-treated with GDNF for 7 days (Fig. 5C). However, the inhibitory effect of GDNF on DHT-induced PrE proliferation was abolished in Ret KO UGSs (Fig. 5C). These data confirm that RET-mediated GDNF signaling is a positive regulator of UrM cell proliferation and a negative regulator of DHT-induced PrE cell proliferation in the developing prostate.

Fig. 5.

RET-mediated GDNF signaling inhibits androgen-induced proliferation and cytodifferentiation of PrE cells in the developing prostate. E15.5 UGSs were cultured with 10 nM DHT and 100 ng/ml GDNF or vehicle for 7 days. (A,B) BrdU (10 µM) was added 2 h before fixation. (A) Immunodetection and (B) quantification of BrdU-positive cells (green), vimentin-positive mesenchymal cells (red) and DAPI-stained nuclei (blue) revealed similar proportions of BrdU-positive cells in the UrM of Ret WT UGSs and Ret KO UGSs co-treated with DHT and GDNF compared with those treated with DHT alone for 7 days (n=5). In contrast, co-treatment with GDNF decreased the number of BrdU-positive cells in the vimentin-negative PrE of DHT-induced Ret WT UGSs but not Ret KO UGSs (n=5). (A) Thicker UrM were observed solely in DHT-induced Ret WT UGSs co-treated with GDNF. (B) A significance level of 0.05 was applied during data analysis using ANOVA, and different significance levels have been indicated using different lowercase letters. (C) With regard to PrE cytodifferentiation, KRT8-positive luminal epithelial cells are absent from PrE buds of Ret WT UGSs co-treated with DHT and GDNF compared with those treated with DHT alone. In contrast, KRT8-positive luminal epithelial cells are present at the midline of PrE buds in DHT-induced Ret KO UGSs regardless of GDNF exposure. Scale bars: 100 µm in A; 20 µm in C.

Fig. 5.

RET-mediated GDNF signaling inhibits androgen-induced proliferation and cytodifferentiation of PrE cells in the developing prostate. E15.5 UGSs were cultured with 10 nM DHT and 100 ng/ml GDNF or vehicle for 7 days. (A,B) BrdU (10 µM) was added 2 h before fixation. (A) Immunodetection and (B) quantification of BrdU-positive cells (green), vimentin-positive mesenchymal cells (red) and DAPI-stained nuclei (blue) revealed similar proportions of BrdU-positive cells in the UrM of Ret WT UGSs and Ret KO UGSs co-treated with DHT and GDNF compared with those treated with DHT alone for 7 days (n=5). In contrast, co-treatment with GDNF decreased the number of BrdU-positive cells in the vimentin-negative PrE of DHT-induced Ret WT UGSs but not Ret KO UGSs (n=5). (A) Thicker UrM were observed solely in DHT-induced Ret WT UGSs co-treated with GDNF. (B) A significance level of 0.05 was applied during data analysis using ANOVA, and different significance levels have been indicated using different lowercase letters. (C) With regard to PrE cytodifferentiation, KRT8-positive luminal epithelial cells are absent from PrE buds of Ret WT UGSs co-treated with DHT and GDNF compared with those treated with DHT alone. In contrast, KRT8-positive luminal epithelial cells are present at the midline of PrE buds in DHT-induced Ret KO UGSs regardless of GDNF exposure. Scale bars: 100 µm in A; 20 µm in C.

Using immunohistochemistry, we also determined the role of RET signaling in PrE cytodifferentiation during the outgrowth of PrE buds from DHT-supplemented Ret WT UGSs and Ret KO UGSs. KRT8-positive luminal epithelial cells were absent from the PrE buds of DHT-induced Ret WT UGSs co-treated with GDNF (Fig. 5C), which is consistent with GDNF inhibiting DHT-induced PrE cytodifferentiation (Fig. 3B). In contrast, KRT8-positive luminal epithelial cells were present at the midline of the PrE buds of Ret KO prostate rudiments regardless of GDNF exposure (Fig. 5C). These results demonstrate that RET-mediated GDNF signaling is a negative regulator of androgen-induced PrE cytodifferentiation.

Crosstalk between AR and RET-mediated GDNF signaling pathways in the UGS and developing prostate

As GDNF signaling inhibits androgen-induced prostate development, we hypothesized that GDNF signaling may influence Ar gene expression. We first assessed AR and RET protein expression in E15.5 UGSs, which were not cultured. AR protein is expressed in the UrM and to a lesser extent in the UrE of E15.5 UGSs, whereas RET expression is limited to the UrM (Fig. 6A). We then cultured androgen-supplemented E15.5 UGSs with GDNF or vehicle for 2-7 days, and assessed AR expression. In comparison to vehicle as a control, GDNF exposure decreased the expression of AR mRNA (Fig. 6B) and AR protein (Fig. 6C). Immunohistochemistry analysis revealed robust AR expression and nuclear localization in nearly all UrM cells of UGSs treated with DHT alone and the Ret-negative UrM cells of UGSs co-treated with DHT and GDNF (Fig. 6D). In contrast, decreases in AR expression and nuclear localization were evident in RET-positive VMP cells of UGSs co-treated with DHT and GDNF (Fig. 6D). These results indicate that GDNF signaling downregulates AR expression in androgen-induced UGSs, establishing a novel regulatory interaction between GDNF signaling and the AR.

Fig. 6.

Crosstalk between AR and RET-mediated GDNF signaling pathways in the UGS and developing prostate. (A) UrM and UrE were separated from E15.5 male UGSs, and immunoblot analysis shows that AR expression is robust in the UrM and modest in the UrE. RET expression is localized to the UrM. The clean separation of the UrM and PrE is supported by mesenchymal vimentin and epithelial keratins. (B,C) E15.5 UGSs were cultured with 10 nM DHT and 100 ng/ml GDNF or vehicle for 2-7 days. (B) QPCR analysis demonstrates that AR transcripts decreased 43% in UGSs co-treated with DHT and GDNF compared with those treated with DHT alone on day 2. Data are mean±s.e.m. (n=5) in log2 scale. (C) Immunoblot analysis of lysates from DHT-induced UGSs reveals that AR expression decreased 44% and 47% in UGSs co-treated with GDNF compared with vehicle on day 2 and day 7, respectively. Relative AR to GAPDH protein ratios are shown using the mean±s.e.m. (n=5). (D) Immunohistochemistry reveals the localization of AR (green) and RET (red) in UGSs co-treated with DHT and GDNF compared with those treated with DHT alone for 2 days. Robust AR expression and nuclear localization are present in nearly all UrM cells of UGSs treated with DHT alone and the Ret-negative UrM cells of UGSs co-treated with DHT and GDNF. In contrast, decreased AR expression and nuclear localization are evident in the RET-positive VMP cells of UGSs co-treated with DHT and GDNF. (E,F) E15.5 UGSs from Ret WT males and Ret KO males were cultured with 10 nM DHT for 7 days. (E) AR mRNA increased 90% in Ret KO UGSs compared with Ret WT UGSs (n=9). (F) Immunoblot analysis of DHT-induced prostate rudiments reveals that AR expression increased 60% in Ret KO UGSs compared with Ret WT UGSs (n=5). (G) UGSs from E15.5 wild-type males were cultured with 10 nM DHT or vehicle for 7 days. QPCR analysis shows that Gdnf and Gfrα1 mRNAs are androgen-repressed in UGSs treated with DHT compared with vehicle. Data are mean±s.e.m. (n=4) in log2 scale. *P<0.05, **P<0.01. Scale bars: 50 µm.

Fig. 6.

Crosstalk between AR and RET-mediated GDNF signaling pathways in the UGS and developing prostate. (A) UrM and UrE were separated from E15.5 male UGSs, and immunoblot analysis shows that AR expression is robust in the UrM and modest in the UrE. RET expression is localized to the UrM. The clean separation of the UrM and PrE is supported by mesenchymal vimentin and epithelial keratins. (B,C) E15.5 UGSs were cultured with 10 nM DHT and 100 ng/ml GDNF or vehicle for 2-7 days. (B) QPCR analysis demonstrates that AR transcripts decreased 43% in UGSs co-treated with DHT and GDNF compared with those treated with DHT alone on day 2. Data are mean±s.e.m. (n=5) in log2 scale. (C) Immunoblot analysis of lysates from DHT-induced UGSs reveals that AR expression decreased 44% and 47% in UGSs co-treated with GDNF compared with vehicle on day 2 and day 7, respectively. Relative AR to GAPDH protein ratios are shown using the mean±s.e.m. (n=5). (D) Immunohistochemistry reveals the localization of AR (green) and RET (red) in UGSs co-treated with DHT and GDNF compared with those treated with DHT alone for 2 days. Robust AR expression and nuclear localization are present in nearly all UrM cells of UGSs treated with DHT alone and the Ret-negative UrM cells of UGSs co-treated with DHT and GDNF. In contrast, decreased AR expression and nuclear localization are evident in the RET-positive VMP cells of UGSs co-treated with DHT and GDNF. (E,F) E15.5 UGSs from Ret WT males and Ret KO males were cultured with 10 nM DHT for 7 days. (E) AR mRNA increased 90% in Ret KO UGSs compared with Ret WT UGSs (n=9). (F) Immunoblot analysis of DHT-induced prostate rudiments reveals that AR expression increased 60% in Ret KO UGSs compared with Ret WT UGSs (n=5). (G) UGSs from E15.5 wild-type males were cultured with 10 nM DHT or vehicle for 7 days. QPCR analysis shows that Gdnf and Gfrα1 mRNAs are androgen-repressed in UGSs treated with DHT compared with vehicle. Data are mean±s.e.m. (n=4) in log2 scale. *P<0.05, **P<0.01. Scale bars: 50 µm.

To determine whether RET mediates downregulation of AR, we quantified AR expression in Ret WT UGSs and Ret KO UGSs cultured with DHT for 7 days to induce prostate development (Figs 3A, 4C). AR mRNA and protein expression were increased significantly in Ret KO prostate rudiments compared with Ret WT rudiments (Fig. 6E,F), thus implicating RET-dependent signaling in the downregulation of AR in the inductive UrM.

As GDNF signaling regulates Ar gene expression, we examined the responsiveness of Gdnf, Gfra1 and Ret to androgen in the embryonic UGS. Using QPCR analysis, we found that Gdnf and Gfra1 mRNAs were androgen-repressed in UGS culture, whereas Ret mRNA levels were unresponsive to androgen (Fig. 6G). These results suggest that expression of GDNF and its receptor, GFRα1, are regulated by AR signaling in DHT-induced UGSs. Taken together, our gene expression results establish reciprocal regulatory crosstalk between AR and GDNF signaling in androgen-induced prostate development (Fig. 7).

Fig. 7.

Proposed mechanisms for crosstalk between AR and GDNF signaling and inhibition of prostate development by RET-mediated GDNF signaling. In androgen-induced prostate development, AR signaling in the UrM orchestrates prostate development by regulating expression of morphoregulatory proteins, such as GDNF and GFRα1. With regard to crosstalk between AR and GDNF signaling, we propose that reciprocal downregulation of AR and GDNF signaling may tune prostate development. AR-mediated transcriptional regulation of GDNF and GFRα1 in the UrM may serve to downregulate GDNF signaling in the developing prostate because RET-mediated GDNF signaling is a negative regulator of PrE proliferation and cytodifferentiation. In the UrM, GDNF binds and activates the GFRα1/RET co-receptor complex, and subsequent RET-mediated signaling activates the MEK-ERK pathway, which leads to increases in RET expression and proliferation of UrM cells, including inductive VMP cells. In addition, we propose that crosstalk between RET-mediated GDNF signaling and AR signaling in the inductive UrM may decrease AR signaling and alter expression of a putative paracrine signal, thus interfering with AR-induced proliferation and cytodifferentiation of PrE cells. In the PrE and UrE, which lack RET expression, GDNF may bind and activate another co-receptor complex consisting of GFRα1 and a putative co-receptor signaling component (coRSC). RET-independent signaling increases proliferation of UrE cells.

Fig. 7.

Proposed mechanisms for crosstalk between AR and GDNF signaling and inhibition of prostate development by RET-mediated GDNF signaling. In androgen-induced prostate development, AR signaling in the UrM orchestrates prostate development by regulating expression of morphoregulatory proteins, such as GDNF and GFRα1. With regard to crosstalk between AR and GDNF signaling, we propose that reciprocal downregulation of AR and GDNF signaling may tune prostate development. AR-mediated transcriptional regulation of GDNF and GFRα1 in the UrM may serve to downregulate GDNF signaling in the developing prostate because RET-mediated GDNF signaling is a negative regulator of PrE proliferation and cytodifferentiation. In the UrM, GDNF binds and activates the GFRα1/RET co-receptor complex, and subsequent RET-mediated signaling activates the MEK-ERK pathway, which leads to increases in RET expression and proliferation of UrM cells, including inductive VMP cells. In addition, we propose that crosstalk between RET-mediated GDNF signaling and AR signaling in the inductive UrM may decrease AR signaling and alter expression of a putative paracrine signal, thus interfering with AR-induced proliferation and cytodifferentiation of PrE cells. In the PrE and UrE, which lack RET expression, GDNF may bind and activate another co-receptor complex consisting of GFRα1 and a putative co-receptor signaling component (coRSC). RET-independent signaling increases proliferation of UrE cells.

RET-mediated GDNF signaling serves as a negative regulator of prostate development

GDNF signaling plays crucial roles in development; however, its role in prostate development has not been explored previously. Here, we show for the first time that elevated levels of GDNF in the UGS inhibit androgen-induced prostate development. In addition to promoting the survival and differentiation of neurons in the nervous system (Arenas et al., 1995; Henderson et al., 1994; Lin et al., 1993), GDNF dose in the urogenital system regulates cell fate decisions of germline progenitors in testis and ureteric branching morphogenesis in kidney development. In mammalian seminiferous tubules, Sertoli cells dictate the microenvironment of the spermatogonial stem cell niche through release of GDNF. At low GDNF levels, the spermatogonial stem cells differentiate, whereas at high levels, the stem cells self-renew and are unable to differentiate (Meng et al., 2000). During formation of the permanent kidney, GDNF, which originates from the metanephric mesenchyme adjacent to the nephric duct epithelium, acts as a mesenchyme-derived paracrine signal to promote RET-mediated ureteric bud outgrowth and branching (Hellmich et al., 1996; Pachnis et al., 1993; Pepicelli et al., 1997; Pichel et al., 1996; Sainio et al., 1997; Vega et al., 1996). Somewhat analogous to the role of GDNF in spermatogonial stem cell self-renewal, our results demonstrate that RET-mediated GDNF signaling functions as a positive regulator of inductive UrM cell self-renewal and a negative regulator of PrE cytodifferentiation in androgen-induced prostate rudiments. The inhibitory effects of GDNF signaling on PrE proliferation and cytodifferentiation are also intriguing because they differ dramatically from the stimulatory effect that GDNF signaling has on ureteric bud outgrowth and branching during kidney morphogenesis.

Molecular effectors of proliferation in the UGS during androgen-induced prostate development

GDNF-induced proliferation of UrM cells in androgen-induced UGSs (Fig. 2) is consistent with our previous report of cell-specific regulation of UrM proliferation by RET-mediated activation of the MEK-ERK pathway (Park and Bolton, 2015). Curiously, we found that GDNF-induced proliferation of UrM cells is time sensitive during androgen-induced prostate development (Fig. 2), despite persistent upregulation of RET (Fig. S1). Hence, we propose that chronic GDNF-induced proliferation of inductive UrM cells in the VMP is dependent on persistent activation of RET and the MEK-ERK pathway (Fig. 7). By day 7, an inhibitory feedback signal is likely activated, as Spry1 mRNA increased substantially in DHT-induced UGSs co-treated with GDNF (Fig. S1). Studies by Basson et al. have demonstrated that Spry1 is an antagonistic regulator of RET-mediated GDNF signaling during kidney morphogenesis (Basson et al., 2005, 2006). Although the mechanism whereby sprouty proteins exert their function is unclear, most studies support a role for them as antagonists of MEK-ERK pathway activation.

Here, we determined that proliferation decreased in the PrE outgrowths from the UrE of DHT-induced UGSs co-treated with GDNF compared with vehicle (Fig. 2). As the negative effect of GDNF on androgen-induced proliferation of PrE cells is time sensitive and dependent on RET (Figs 2 and 5), we propose that an inhibitory feedback signal is likely activated. The upregulation of Spry1 by day 7 is consistent with inhibitory feedback on RET-mediated signaling in the UrM (Fig. 7). Previously, we have reported that a RET-independent mechanism is responsible for GDNF-induced proliferation of UrE cells in naïve UGSs (Park and Bolton, 2015) (Fig. 7). These differences in epithelial cell proliferation are likely due to intrinsic differences between the PrE cells and UrE cells that are bestowed during androgen-dependent prostatic induction and formation of PrE progenitors/buds (Cunha and Chung, 1981; Cunha and Lung, 1978; Lasnitzki and Mizuno, 1980).

Molecular crosstalk between AR and GDNF signaling in androgen-induced prostate development

GDNF and AR signaling components are expressed in the UGS; however, their interaction during prostate development has not been explored previously. GDNF, GFRα1, RET and AR are expressed in the UrM of the embryonic UGS and developing prostate (Park and Bolton, 2015) (Fig. 6), and AR signaling in the UrM is required for prostate development (Cunha and Chung, 1981; Cunha and Lung, 1978; Lasnitzki and Mizuno, 1980). Here, we have demonstrated that RET-mediated GDNF signaling downregulates AR expression and nuclear localization in the inductive UrM of androgen-induced UGSs (Fig. 6). These results suggest that AR downregulation by GDNF signaling may lead to partial androgen insensitivity in the developing prostate. This is an intriguing hypothesis because disruption of AR-mediated prostate development predisposes humans and rodents to prostate neoplasia by altering the phenotype of the gland early in life (Anway and Skinner, 2008; Gupta, 2000; Ho et al., 2006; Hu et al., 2012; Prins et al., 2014, 2008; Ramos et al., 2001; Timms et al., 2005). Consistent with Ar being a GDNF-repressed gene, and Gdnf and Gfra1 being androgen-repressed genes in the UGS and developing prostate (Fig. 6), we propose that these genes and their gene products mediate reciprocal regulatory crosstalk to tune prostate development (Fig. 7). The reciprocal regulatory crosstalk in such a circuit may serve to balance GDNF-induced proliferation of inductive UrM cells with androgen-induced PrE proliferation and cytodifferentiation, which are mediated by AR signaling in the inductive UrM cells. If GDNF and/or GFRα1 are not downregulated early in prostate development, elevated levels of GDNF may lead to partial androgen-insensitivity and disruption of AR-mediated prostate development. Additional studies are needed to elucidate the extent of crosstalk between the AR and GDNF signaling pathways.

Paracrine signaling between the inductive UrM and PrE during prostate development

We have determined that GDNF signaling through RET in UrM cells inhibits DHT-induced proliferation and differentiation of PrE cells in the developing prostate (Figs 4 and 5). We also interrogated smooth muscle differentiation, which may influence paracrine signaling between the inductive UrM and PrE during prostate development (Hayward et al., 1998; Thomson et al., 2002; Timms et al., 1995, 1994). We found that GDNF signaling in the developing prostate does not affect smooth muscle differentiation (Fig. S2), indicating that RET-mediated GDNF signaling in the UrM may selectively affect PrE proliferation and cytodifferentiation. These findings raise an interesting question: how does GDNF signaling in the UrM suppress androgen-induced proliferation and cytodifferentiation in the PrE?

We propose that these effects of GDNF signaling are mediated indirectly on PrE cells by a putative paracrine signal, originating from RET-expressing UrM cells in the VMP (Fig. 7). This mechanism is consistent with previous reports of paracrine signaling for secreted ligands of the WNT (Allgeier et al., 2008; Huang et al., 2009; Joesting et al., 2008; Keil et al., 2012), FGF (Alarid et al., 1994; Donjacour et al., 2003; Sugimura et al., 1996; Thomson and Cunha, 1999) and TGFβ (Cancilla et al., 2001; Cook et al., 2007; Grishina et al., 2005; Itoh et al., 1998; Lamm et al., 2001; Tanji et al., 1994; Tomlinson et al., 2004) protein super-families. For example, regulation of Wnt paracrine signaling from the inductive UrM to the adjacent epithelial cells has been proposed to mediate the effects of TGFβ signaling in the UrM (Li et al., 2009). Given that GDNF family ligands are members of the TGFβ protein super-family, RET-mediated GDNF signaling may regulate aspects of prostate development through communication between inductive UrM cells and PrE cells via a paracrine mechanism.

In conclusion, we have determined that RET-mediated GDNF signaling in the UGS increases proliferation of UrM cells and suppresses androgen-induced PrE proliferation and cytodifferentiation, altering prostate morphogenesis. We also determined that Ar is GDNF repressed by RET-mediated signaling, and Gdnf and Gfra1 are androgen repressed in the developing prostate. These findings reveal that RET-mediated GDNF signaling serves as a negative regulator of prostate development via downregulation of AR expression, and they establish novel crosstalk between AR and GDNF signaling in the cellular and molecular mechanism of androgen-induced prostate development. Therefore, future studies should interrogate the crosstalk mechanism between these signaling pathways and explore whether AR-mediated prostate development and growth in vivo are regulated by other selective GDNF family receptors and activation of RET tyrosine kinase.

Animal maintenance and genotyping

C57BL/6J mice were purchased from Jackson Laboratories. Heterozygous Ret mutant (Ret GFP/+) mice have a null Ret knock-in allele that has been described previously (a gift from Dr Sanjay Jain, Washington University, St Louis, MO, USA) (Hoshi et al., 2012). All animal experiments were approved by the University of Illinois at Urbana-Champaign Animal Care and Use Committee. Timed-pregnant females were generated by pairing males with females overnight. We define midday on the day of plug identification as E0.5. We mated Ret GFP/+ mice to produce Ret GFP/GFP (Ret-null, Ret KO), Ret GFP/+ and Ret +/+ control (Ret wild type, Ret WT) embryos.

Genotyping of mice was performed as described for the Ret locus (Hoshi et al., 2012; Jain et al., 2004) and fetal sex (Park and Bolton, 2015). PCR reactions were performed using wild-type Ret primers (5′-CAGCGCAGGTCTCTCATCAGTACCGCA-3′ and 5′-CAGCTACCCGCAGCGACCCGGTTC-3′ produce a 451 bp amplicon) and mutant Ret-GFP primers (5′-CAGCGCAGGTCTCTCATCAGTACCGCA-3′ and 5′-GCCGTTTACGTCGCCGTCCAGCTCGACCAG-3′ produce a 350 bp amplicon). The PCR was achieved in the presence of 1 M betaine using the following method: a denaturation and polymerase activation step at 94°C for 1 min and then 40 cycles consisting of 94°C for 10 s, 62°C for 10 s and 72°C for 20 s.

Urogenital sinus (UGS) and prostate organ culture

UGSs were dissected from E15.5 male embryos and prostates were collected from P0 neonates in DMEM/F12 (1:1) medium (21041-025, Invitrogen) and cultured on 0.4 µm Millicell CM filters (PICM01250, EMD Millipore) floating on organ culture medium [DMEM/F12 supplemented with 1% insulin, transferrin and selenium (51500-056, Invitrogen), and gentamicin and amphotericin B (R-015-10, Invitrogen)] in a 5% CO2 atmosphere, as described previously (Park and Bolton, 2015). Additional supplements included 0.1% ethanol control, 10 nM DHT (A8380, Sigma Aldrich) in ethanol, 0.1% PBS control and 1-100 ng/ml recombinant GDNF (212-GD, R&D Systems) in PBS. Culture medium was changed every other day.

Morphological changes in UGS whole mounts were imaged using a Leica M205A stereoscope. The prostatic epithelial buds were counted from bright-field images and the length of each bud was measured by scaled reference measurement using ImageJ software. The mean bud number and length were quantified using n=4 or more UGSs for each condition. For cell proliferation experiments, UGSs were cultured in organ culture medium with the previously mentioned supplements for 2-7 days and 10 µM bromodeoxyuridine (BrdU, B5002, Sigma Aldrich) was added 2 h before fixation (Park and Bolton, 2015).

Immunohistochemistry and cell proliferation

UGSs and developing prostates were processed for immunohistochemistry as described previously (Park and Bolton, 2015). Gill Method Hematoxylin Stain 3 (CS4021D, Fisher Scientific) and Eosin Y solution (HT110332, Sigma Aldrich) were used. Immunohistochemistry controls included pre-incubation of primary antibody with antigenic peptide, when available, prior to incubation with secondary antibodies and incubation in the absence of primary antibodies (Park and Bolton, 2015). Primary antibodies against the following proteins were used: vimentin (1:200, 5741S) and pan-keratin (1:200, 4545S) from Cell Signaling Technology; BrdU (1:200, 347580, BD Biosciences); AR (1:200, 06-680, EMD Millipore); RET (5 µg/ml, AF482, R&D Systems); alpha smooth muscle actin (1:1000, ab5694, Abcam); and CK5 (1:300, PRB-160P) and CK8 (1:500, MMS-162P) from Covance. The following secondary antibodies from Molecular Probes were used: AlexaFluor-594 anti-mouse (1:300, A-21203), AlexaFluor-594 anti-rabbit (1:300, A-21207), AlexFluor-594 anti-goat (1:300, A-11058), AlexaFluor-488 anti-mouse (1:300, A-21202) and AlexaFluor-488 anti-rabbit (1:300, A-21206). Tissue sections were then incubated with 0.6 µM 6-diamidino-2-pheylindole (DAPI), washed with PBS and overlaid with mounting medium (10981, Sigma Aldrich) and coverslips. Fluorescence microscopy images were collected and analyzed as described previously (Park and Bolton, 2015).

In cell proliferation experiments, the BrdU-positive cells were detected using BrdU antibody (1:200, 347580, BD Biosciences) and counted in the UrM, UrE and regions where PrE outgrowth occurred. The UrM, and UrE and PrE cells were defined by immunostaining using antibodies for detection of mesenchymal vimentin (5741S, Cell Signaling Technology) and epithelial keratins (4545S, Cell Signaling Technology), respectively. Percentages of proliferating cells in each UGS tissue was calculated as described previously (Park and Bolton, 2015).

Separation of embryonic UGS mesenchyme and epithelium and immunoblot analysis

UGSs from E15.5 male embryos were separated into UrM and UrE components, and lysates were prepared and used for immunoblot analysis as described previously (Park and Bolton, 2015). PVDF membranes were blocked in TBST [20 mM Tris-HCl (pH 7.5), 150 mM NaCl and 0.1% Tween-20] containing 1% BSA. Membranes were incubated overnight with primary antibodies at the indicated dilutions in TBST+1% BSA. Primary antibodies included: RET (0.1 µg/ml, AF482, R&D Systems); α smooth muscle actin (1:2000, ab5694, Abcam); GAPDH (1:1000, 2118L), vimentin (1:1000, 5741S) and pan-keratin (1:1000, 4545S) from Cell Signaling Technology; and AR (1:800, 06-680, EMD Millipore). Membranes were washed in TBST and incubated for 1 h with 1:2000 dilutions of horseradish peroxidase-linked anti-rabbit or anti-goat IgG (Jackson ImmunoResearch Laboratories) in TBST+1% BSA. Immunoblots were visualized, and ImageJ software was used to quantify protein expression relative to GAPDH control.

RNA isolation, reverse transcription and QPCR

UGSs from E15.5 male embryos were cultured in organ culture medium containing 0-10 nM DHT and 0-100 ng/ml GDNF for 2-7 days. RNA was extracted from homogenized UGSs and cDNA was analyzed by QPCR using a StepOnePlus Real-Time PCR System (Applied Biosystems) and validated primers as described previously (Park and Bolton, 2015). Primers included: Ret sense, 5′-TGACATCAGCAAGGATCTGG-3′; Ret antisense, 5′-AGAGCCCATCGTCATACAGC-3′; Pygo2 sense, 5′-CCCAGTCAACCCTTCAACC-3′; Pygo2 antisense, 5′-GAGATCATGGGACCAAATCC-3′; Krt18 sense, 5′-GACGCTGAGACCACACTCAC-3′; Krt18 antisense, 5′-TCCATCTGTGCCTTGTATCG-3′; Krt8 sense, 5′-GACACGTCTGTGGTGCTGTC-3′; Krt8 antisense, 5′-CAGGGTCTGCAATTCCTCAT-3′; Krt14 sense, 5′-GTATTGGTGATGGGCTCCTG-3′; Krt14, antisense, 5′-CACCTCCAGTTCAGTGTTGG-3′; Krt5 sense, 5′-TGCTGCCTACATGAACAAGG-3′; Krt5 antisense, 5′-GCTCCGCATCAAAGAACATC-3′; AR sense, 5′-GGCGGTCCTTCACTAATGTC-3′; AR antisense, 5′-GACAGGTGCCTCATCCTCAC-3′. Data were analyzed using the comparative threshold cycle (Ct) method (Schmittgen and Livak, 2008) and Pygo2 control gene, which is unresponsive to GDNF and DHT (Park and Bolton, 2015). Following normalization to Pygo2 cDNA levels, which is reflected in the ΔCt values, the relative quantification (RQ) of the fold change for each treatment compared with reference control was determined using the following equation: RQ=2(–ΔCt)/2(–ΔCt reference). The mean of the log2 RQ and standard error of the mean (s.e.m.) are plotted.

Statistical analysis

Significance differences between two treatment groups were determined using Student's t-test, whereas differences among multiple groups or time points were determined by analysis of variance (ANOVA), followed by Tukey's honest significant difference test. Significance levels of 0.05 and 0.01 were applied during data analysis using Student's t-test, and different significance levels are indicated (*P<0.05 and **P<0.01). A significance level of 0.05 was applied during data analysis using ANOVA, and different significance levels have been indicated using different lowercase letters.

We thank Hee Jung Chung, Lori Raetzman and Congcong Chen for insightful comments on the manuscript, and Congcong Chen for technical assistance.

Author contributions

Conceptualization: H.-J.P., E.C.B.; Methodology: H.-J.P., E.C.B.; Validation: H.-J.P., E.C.B.; Formal analysis: H.-J.P., E.C.B.; Investigation: H.-J.P., E.C.B.; Resources: E.C.B.; Writing - original draft: H.-J.P., E.C.B.; Writing - review & editing: E.C.B.; Visualization: H.-J.P., E.C.B.; Supervision: E.C.B.; Project administration: E.C.B.; Funding acquisition: E.C.B.

Funding

This work was supported by an award from the National Institute of Diabetes and Digestive and Kidney Diseases (National Institutes of Health) (R56DK109149), by the University of Illinois at Urbana-Champaign start-up funds and by two Arnold O. Beckman Research Awards (RB14084 and RB16156 to E.C.B.). Deposited in PMC for release after 12 months.

Achtstätter
,
T.
,
Moll
,
R.
,
Moore
,
B.
and
Franke
,
W. W.
(
1985
).
Cytokeratin polypeptide patterns of different epithelia of the human male urogenital tract: immunofluorescence and gel electrophoretic studies
.
J. Histochem. Cytochem.
33
,
415
-
426
.
Alarid
,
E. T.
,
Rubin
,
J. S.
,
Young
,
P.
,
Chedid
,
M.
,
Ron
,
D.
,
Aaronson
,
S. A.
and
Cunha
,
G. R.
(
1994
).
Keratinocyte growth factor functions in epithelial induction during seminal vesicle development
.
Proc. Natl. Acad. Sci. USA
91
,
1074
-
1078
.
Allgeier
,
S. H.
,
Lin
,
T.-M.
,
Vezina
,
C. M.
,
Moore
,
R. W.
,
Fritz
,
W. A.
,
Chiu
,
S.-Y.
,
Zhang
,
C.
and
Peterson
,
R. E.
(
2008
).
WNT5A selectively inhibits mouse ventral prostate development
.
Dev. Biol.
324
,
10
-
17
.
Allgeier
,
S. H.
,
Lin
,
T.-M.
,
Moore
,
R. W.
,
Vezina
,
C. M.
,
Abler
,
L. L.
and
Peterson
,
R. E.
(
2010
).
Androgenic regulation of ventral epithelial bud number and pattern in mouse urogenital sinus
.
Dev. Dyn.
239
,
373
-
385
.
Anway
,
M. D.
and
Skinner
,
M. K.
(
2008
).
Transgenerational effects of the endocrine disruptor vinclozolin on the prostate transcriptome and adult onset disease
.
Prostate
68
,
517
-
529
.
Arenas
,
E.
,
Trupp
,
M.
,
Åkerud
,
P.
and
Ibáñez
,
C. F.
(
1995
).
GDNF prevents degeneration and promotes the phenotype of brain noradrenergic neurons in vivo
.
Neuron
15
,
1465
-
1473
.
Basson
,
M. A.
,
Akbulut
,
S.
,
Watson-Johnson
,
J.
,
Simon
,
R.
,
Carroll
,
T. J.
,
Shakya
,
R.
,
Gross
,
I.
,
Martin
,
G. R.
,
Lufkin
,
T.
,
McMahon
,
A. P.
, et al. 
(
2005
).
Sprouty1 is a critical regulator of GDNF/RET-mediated kidney induction
.
Dev. Cell
8
,
229
-
239
.
Basson
,
M. A.
,
Watson-Johnson
,
J.
,
Shakya
,
R.
,
Akbulut
,
S.
,
Hyink
,
D.
,
Costantini
,
F. D.
,
Wilson
,
P. D.
,
Mason
,
I. J.
and
Licht
,
J. D.
(
2006
).
Branching morphogenesis of the ureteric epithelium during kidney development is coordinated by the opposing functions of GDNF and Sprouty1
.
Dev. Biol.
299
,
466
-
477
.
Berman
,
D. M.
,
Desai
,
N.
,
Wang
,
X.
,
Karhadkar
,
S. S.
,
Reynon
,
M.
,
Abate-Shen
,
C.
,
Beachy
,
P. A.
and
Shen
,
M. M.
(
2004
).
Roles for Hedgehog signaling in androgen production and prostate ductal morphogenesis
.
Dev. Biol.
267
,
387
-
398
.
Cancilla
,
B.
,
Jarred
,
R. A.
,
Wang
,
H.
,
Mellor
,
S. L.
,
Cunha
,
G. R.
and
Risbridger
,
G. P.
(
2001
).
Regulation of prostate branching morphogenesis by activin A and follistatin
.
Dev. Biol.
237
,
145
-
158
.
Cao
,
J.-P.
,
Yu
,
J.-K.
,
Li
,
C.
,
Sun
,
Y.
,
Yuan
,
H.-H.
,
Wang
,
H.-J.
and
Gao
,
D.-S.
(
2008
).
Integrin beta1 is involved in the signaling of glial cell line-derived neurotrophic factor
.
J. Comp. Neurol.
509
,
203
-
210
.
Cook
,
C.
,
Vezina
,
C. M.
,
Allgeier
,
S. H.
,
Shaw
,
A.
,
Yu
,
M.
,
Peterson
,
R. E.
and
Bushman
,
W.
(
2007
).
Noggin is required for normal lobe patterning and ductal budding in the mouse prostate
.
Dev. Biol.
312
,
217
-
230
.
Cunha
,
G. R.
and
Chung
,
L. W. K.
(
1981
).
Stromal-epithelial interactions–I. Induction of prostatic phenotype in urothelium of testicular feminized (Tfm/y) mice
.
J. Steroid Biochem.
14
,
1317
-
1324
.
Cunha
,
G. R.
and
Lung
,
B.
(
1978
).
The possible influence of temporal factors in androgenic responsiveness of urogenital tissue recombinants from wild-type and androgen-insensitive (Tfm) mice
.
J. Exp. Zool.
205
,
181
-
193
.
Davies
,
J.
,
Lyon
,
M.
,
Gallagher
,
J.
and
Garrod
,
D.
(
1995
).
Sulphated proteoglycan is required for collecting duct growth and branching but not nephron formation during kidney development
.
Development
121
,
1507
-
1517
.
Doles
,
J. D.
,
Vezina
,
C. M.
,
Lipinski
,
R. J.
,
Peterson
,
R. E.
and
Bushman
,
W.
(
2005
).
Growth, morphogenesis, and differentiation during mouse prostate development in situ, in renal grafts, and in vitro
.
Prostate
65
,
390
-
399
.
Doles
,
J.
,
Cook
,
C.
,
Shi
,
X.
,
Valosky
,
J.
,
Lipinski
,
R.
and
Bushman
,
W.
(
2006
).
Functional compensation in Hedgehog signaling during mouse prostate development
.
Dev. Biol.
295
,
13
-
25
.
Donjacour
,
A. A.
and
Cunha
,
G. R.
(
1988
).
The effect of androgen deprivation on branching morphogenesis in the mouse prostate
.
Dev. Biol.
128
,
1
-
14
.
Donjacour
,
A. A.
,
Thomson
,
A. A.
and
Cunha
,
G. R.
(
2003
).
FGF-10 plays an essential role in the growth of the fetal prostate
.
Dev. Biol.
261
,
39
-
54
.
Enomoto
,
H.
,
Crawford
,
P. A.
,
Gorodinsky
,
A.
,
Heuckeroth
,
R. O.
,
Johnson
,
E. M.
, Jr.
and
Milbrandt
,
J.
(
2001
).
RET signaling is essential for migration, axonal growth and axon guidance of developing sympathetic neurons
.
Development
128
,
3963
-
3974
.
Freestone
,
S. H.
,
Marker
,
P.
,
Grace
,
O. C.
,
Tomlinson
,
D. C.
,
Cunha
,
G. R.
,
Harnden
,
P.
and
Thomson
,
A. A.
(
2003
).
Sonic hedgehog regulates prostatic growth and epithelial differentiation
.
Dev. Biol.
264
,
352
-
362
.
Georgas
,
K. M.
,
Armstrong
,
J.
,
Keast
,
J. R.
,
Larkins
,
C. E.
,
McHugh
,
K. M.
,
Southard-Smith
,
E. M.
,
Cohn
,
M. J.
,
Batourina
,
E.
,
Dan
,
H.
,
Schneider
,
K.
, et al. 
(
2015
).
An illustrated anatomical ontology of the developing mouse lower urogenital tract
.
Development
142
,
1893
-
1908
.
Ghosh
,
S.
,
Lau
,
H.
,
Simons
,
B. W.
,
Powell
,
J. D.
,
Meyers
,
D. J.
,
De Marzo
,
A. M.
,
Berman
,
D. M.
and
Lotan
,
T. L.
(
2011
).
PI3K/mTOR signaling regulates prostatic branching morphogenesis
.
Dev. Biol.
360
,
329
-
342
.
Grishina
,
I. B.
,
Kim
,
S. Y.
,
Ferrara
,
C.
,
Makarenkova
,
H. P.
and
Walden
,
P. D.
(
2005
).
BMP7 inhibits branching morphogenesis in the prostate gland and interferes with Notch signaling
.
Dev. Biol.
288
,
334
-
347
.
Gupta
,
C.
(
2000
).
Reproductive malformation of the male offspring following maternal exposure to estrogenic chemicals
.
Proc. Soc. Exp. Biol. Med.
224
,
61
-
68
.
Hayward
,
S. W.
,
Haughney
,
P. C.
,
Rosen
,
M. A.
,
Greulich
,
K. M.
,
Weier
,
H.-U. G.
,
Dahiya
,
R.
and
Cunha
,
G. R.
(
1998
).
Interactions between adult human prostatic epithelium and rat urogenital sinus mesenchyme in a tissue recombination model
.
Differentiation
63
,
131
-
140
.
Hellmich
,
H. L.
,
Kos
,
L.
,
Cho
,
E. S.
,
Mahon
,
K. A.
and
Zimmer
,
A.
(
1996
).
Embryonic expression of glial cell-line derived neurotrophic factor (GDNF) suggests multiple developmental roles in neural differentiation and epithelial-mesenchymal interactions
.
Mech. Dev.
54
,
95
-
105
.
Henderson
,
C. E.
,
Phillips
,
H. S.
,
Pollock
,
R. A.
,
Davies
,
A. M.
,
Lemeulle
,
C.
,
Armanini
,
M.
,
Simmons
,
L.
,
Moffet
,
B.
,
Vandlen
,
R. A.
,
Simpson
,
L. C. c. t. S. L.
, et al. 
(
1994
).
GDNF: a potent survival factor for motoneurons present in peripheral nerve and muscle
.
Science
266
,
1062
-
1064
.
Ho
,
S.-M.
,
Tang
,
W. Y.
,
Belmonte de Frausto
,
J.
and
Prins
,
G. S.
(
2006
).
Developmental exposure to estradiol and bisphenol A increases susceptibility to prostate carcinogenesis and epigenetically regulates phosphodiesterase type 4 variant 4
.
Cancer Res.
66
,
5624
-
5632
.
Hoshi
,
M.
,
Batourina
,
E.
,
Mendelsohn
,
C.
and
Jain
,
S.
(
2012
).
Novel mechanisms of early upper and lower urinary tract patterning regulated by RetY1015 docking tyrosine in mice
.
Development
139
,
2405
-
2415
.
Hsieh
,
J. T.
,
Zhau
,
H. E.
,
Wang
,
X. H.
,
Liew
,
C. C.
and
Chung
,
L. W.
(
1992
).
Regulation of basal and luminal cell-specific cytokeratin expression in rat accessory sex organs. Evidence for a new class of androgen-repressed genes and insight into their pairwise control
.
J. Biol. Chem.
267
,
2303
-
2310
.
Hu
,
W.-Y.
,
Shi
,
G.-B.
,
Hu
,
D.-P.
,
Nelles
,
J. L.
and
Prins
,
G. S.
(
2012
).
Actions of estrogens and endocrine disrupting chemicals on human prostate stem/progenitor cells and prostate cancer risk
.
Mol. Cell. Endocrinol.
354
,
63
-
73
.
Huang
,
L.
,
Pu
,
Y.
,
Hu
,
W. Y.
,
Birch
,
L.
,
Luccio-Camelo
,
D.
,
Yamaguchi
,
T.
and
Prins
,
G. S.
(
2009
).
The role of Wnt5a in prostate gland development
.
Dev. Biol.
328
,
188
-
199
.
Itoh
,
N.
,
Patel
,
U.
,
Cupp
,
A. S.
and
Skinner
,
M. K.
(
1998
).
Developmental and hormonal regulation of transforming growth factor-beta1 (TGFbeta1), -2, and -3 gene expression in isolated prostatic epithelial and stromal cells: epidermal growth factor and TGFbeta interactions
.
Endocrinology
139
,
1378
-
1388
.
Jain
,
S.
,
Naughton
,
C. K.
,
Yang
,
M.
,
Strickland
,
A.
,
Vij
,
K.
,
Encinas
,
M.
,
Golden
,
J.
,
Gupta
,
A.
,
Heuckeroth
,
R.
,
Johnson
,
E. M.
Jr
 et al.  (
2004
).
Mice expressing a dominant-negative Ret mutation phenocopy human Hirschsprung disease and delineate a direct role of Ret in spermatogenesis
.
Development
131
,
5503
-
5513
.
Jing
,
S.
,
Wen
,
D.
,
Yu
,
Y.
,
Holst
,
P. L.
,
Luo
,
Y.
,
Fang
,
M.
,
Tamir
,
R.
,
Antonio
,
L.
,
Hu
,
Z.
,
Cupples
,
R.
, et al. 
(
1996
).
GDNF-induced activation of the ret protein tyrosine kinase is mediated by GDNFR-alpha, a novel receptor for GDNF
.
Cell
85
,
1113
-
1124
.
Joesting
,
M. S.
,
Cheever
,
T. R.
,
Volzing
,
K. G.
,
Yamaguchi
,
T. P.
,
Wolf
,
V.
,
Naf
,
D.
,
Rubin
,
J. S.
and
Marker
,
P. C.
(
2008
).
Secreted frizzled related protein 1 is a paracrine modulator of epithelial branching morphogenesis, proliferation, and secretory gene expression in the prostate
.
Dev. Biol.
317
,
161
-
173
.
Keil
,
K. P.
,
Mehta
,
V.
,
Branam
,
A. M.
,
Abler
,
L. L.
,
Buresh-Stiemke
,
R. A.
,
Joshi
,
P. S.
,
Schmitz
,
C. T.
,
Marker
,
P. C.
and
Vezina
,
C. M.
(
2012
).
Wnt inhibitory factor 1 (Wif1) is regulated by androgens and enhances androgen-dependent prostate development
.
Endocrinology
153
,
6091
-
6103
.
Kuslak
,
S. L.
and
Marker
,
P. C.
(
2007
).
Fibroblast growth factor receptor signaling through MEK-ERK is required for prostate bud induction
.
Differentiation
75
,
638
-
651
.
Lamm
,
M. L. G.
,
Podlasek
,
C. A.
,
Barnett
,
D. H.
,
Lee
,
J.
,
Clemens
,
J. Q.
,
Hebner
,
C. M.
and
Bushman
,
W.
(
2001
).
Mesenchymal factor bone morphogenetic protein 4 restricts ductal budding and branching morphogenesis in the developing prostate
.
Dev. Biol.
232
,
301
-
314
.
Lamm
,
M. L. G.
,
Catbagan
,
W. S.
,
Laciak
,
R. J.
,
Barnett
,
D. H.
,
Hebner
,
C. M.
,
Gaffield
,
W.
,
Walterhouse
,
D.
,
Iannaccone
,
P.
and
Bushman
,
W.
(
2002
).
Sonic hedgehog activates mesenchymal Gli1 expression during prostate ductal bud formation
.
Dev. Biol.
249
,
349
-
366
.
Lasnitzki
,
I.
and
Mizuno
,
T.
(
1980
).
Prostatic induction: interaction of epithelium and mesenchyme from normal wild-type mice and androgen-insensitive mice with testicular feminization
.
J. Endocrinol.
85
,
423
-
428
.
Li
,
X.
,
Wang
,
Y.
,
Sharif-Afshar
,
A.-R.
,
Uwamariya
,
C.
,
Yi
,
A.
,
Ishii
,
K.
,
Hayward
,
S. W.
,
Matusik
,
R. J.
and
Bhowmick
,
N. A.
(
2009
).
Urothelial transdifferentiation to prostate epithelia is mediated by paracrine TGF-beta signaling
.
Differentiation
77
,
95
-
102
.
Lin
,
L. F.
,
Doherty
,
D. H.
,
Lile
,
J. D.
,
Bektesh
,
S.
and
Collins
,
F.
(
1993
).
GDNF: a glial cell line-derived neurotrophic factor for midbrain dopaminergic neurons
.
Science
260
,
1130
-
1132
.
Marker
,
P. C.
,
Donjacour
,
A. A.
,
Dahiya
,
R.
and
Cunha
,
G. R.
(
2003
).
Hormonal, cellular, and molecular control of prostatic development
.
Dev. Biol.
253
,
165
-
174
.
Meng
,
X.
,
Lindahl
,
M.
,
Hyvonen
,
M. E.
,
Parvinen
,
M.
,
de Rooij
,
D. G.
,
Hess
,
M. W.
,
Raatikainen-Ahokas
,
A.
,
Sainio
,
K.
,
Rauvala
,
H.
,
Lakso
,
M.
, et al. 
(
2000
).
Regulation of cell fate decision of undifferentiated spermatogonia by GDNF
.
Science
287
,
1489
-
1493
.
Pachnis
,
V.
,
Mankoo
,
B.
and
Costantini
,
F.
(
1993
).
Expression of the c-ret proto-oncogene during mouse embryogenesis
.
Development
119
,
1005
-
1017
.
Panicker
,
A. K.
,
Buhusi
,
M.
,
Thelen
,
K.
and
Maness
,
P. F.
(
2003
).
Cellular signalling mechanisms of neural cell adhesion molecules
.
Front. Biosci.
8
,
d900
-
d911
.
Paratcha
,
G.
,
Ledda
,
F.
and
Ibáñez
,
C. F.
(
2003
).
The neural cell adhesion molecule NCAM is an alternative signaling receptor for GDNF family ligands
.
Cell
113
,
867
-
879
.
Park
,
H.-J.
and
Bolton
,
E. C.
(
2015
).
Glial cell line-derived neurotrophic factor induces cell proliferation in the mouse urogenital sinus
.
Mol. Endocrinol.
29
,
289
-
306
.
Pepicelli
,
C. V.
,
Kispert
,
A.
,
Rowitch
,
D. H.
and
McMahon
,
A. P.
(
1997
).
GDNF induces branching and increased cell proliferation in the ureter of the mouse
.
Dev. Biol.
192
,
193
-
198
.
Pichel
,
J. G.
,
Shen
,
L.
,
Sheng
,
H. Z.
,
Granholm
,
A.-C.
,
Drago
,
J.
,
Grinberg
,
A.
,
Lee
,
E. J.
,
Huang
,
S. P.
,
Saarma
,
M.
,
Hoffer
,
B. J.
, et al. 
(
1996
).
Defects in enteric innervation and kidney development in mice lacking GDNF
.
Nature
382
,
73
-
76
.
Podlasek
,
C. A.
,
Barnett
,
D. H.
,
Clemens
,
J. Q.
,
Bak
,
P. M.
and
Bushman
,
W.
(
1999
).
Prostate development requires Sonic hedgehog expressed by the urogenital sinus epithelium
.
Dev. Biol.
209
,
28
-
39
.
Poteryaev
,
D.
,
Titievsky
,
A.
,
Sun
,
Y. F.
,
Thomas-Crusells
,
J.
,
Lindahl
,
M.
,
Billaud
,
M.
,
Arumäe
,
U.
and
Saarma
,
M.
(
1999
).
GDNF triggers a novel ret-independent Src kinase family-coupled signaling via a GPI-linked GDNF receptor alpha1
.
FEBS Lett.
463
,
63
-
66
.
Prins
,
G. S.
and
Putz
,
O.
(
2008
).
Molecular signaling pathways that regulate prostate gland development
.
Differentiation
76
,
641
-
659
.
Prins
,
G. S.
,
Tang
,
W.-Y.
,
Belmonte
,
J.
and
Ho
,
S.-M.
(
2008
).
Perinatal exposure to oestradiol and bisphenol A alters the prostate epigenome and increases susceptibility to carcinogenesis
.
Basic Clin. Pharmacol. Toxicol.
102
,
134
-
138
.
Prins
,
G. S.
,
Hu
,
W.-Y.
,
Shi
,
G.-B.
,
Hu
,
D.-P.
,
Majumdar
,
S.
,
Li
,
G.
,
Huang
,
K.
,
Nelles
,
J. L.
,
Ho
,
S.-M.
,
Walker
,
C. L.
, et al. 
(
2014
).
Bisphenol A promotes human prostate stem-progenitor cell self-renewal and increases in vivo carcinogenesis in human prostate epithelium
.
Endocrinology
155
,
805
-
817
.
Pu
,
Y.
,
Huang
,
L.
and
Prins
,
G. S.
(
2004
).
Sonic hedgehog-patched Gli signaling in the developing rat prostate gland: lobe-specific suppression by neonatal estrogens reduces ductal growth and branching
.
Dev. Biol.
273
,
257
-
275
.
Ramos
,
J. G.
,
Varayoud
,
J.
,
Sonnenschein
,
C.
,
Soto
,
A. M.
,
Muñoz de Toro
,
M.
and
Luque
,
E. H.
(
2001
).
Prenatal exposure to low doses of bisphenol A alters the periductal stroma and glandular cell function in the rat ventral prostate
.
Biol. Reprod.
65
,
1271
-
1277
.
Sainio
,
K.
,
Suvanto
,
P.
,
Davies
,
J.
,
Wartiovaara
,
J.
,
Wartiovaara
,
K.
,
Saarma
,
M.
,
Arumae
,
U.
,
Meng
,
X.
,
Lindahl
,
M.
,
Pachnis
,
V.
, et al. 
(
1997
).
Glial-cell-line-derived neurotrophic factor is required for bud initiation from ureteric epithelium
.
Development
124
,
4077
-
4087
.
Schmittgen
,
T. D.
and
Livak
,
K. J.
(
2008
).
Analyzing real-time PCR data by the comparative C(T) method
.
Nat. Protoc.
3
,
1101
-
1108
.
Schuchardt
,
A.
,
D'Agati
,
V.
,
Larsson-Blomberg
,
L.
,
Costantini
,
F.
and
Pachnis
,
V.
(
1994
).
Defects in the kidney and enteric nervous system of mice lacking the tyrosine kinase receptor Ret
.
Nature
367
,
380
-
383
.
Soeffing
,
W. J.
and
Timms
,
B. G.
(
1995
).
Localization of androgen receptor and cell-specific cytokeratins in basal cells of rat ventral prostate
.
J. Androl.
16
,
197
-
208
.
Sugimura
,
Y.
,
Cunha
,
G. R.
and
Donjacour
,
A. A.
(
1986
).
Morphogenesis of ductal networks in the mouse prostate
.
Biol. Reprod.
34
,
961
-
971
.
Sugimura
,
Y.
,
Foster
,
B. A.
,
Hom
,
Y. K.
,
Lipschutz
,
J. H.
,
Rubin
,
J. S.
,
Finch
,
P. W.
,
Aaronson
,
S. A.
,
Hayashi
,
N.
,
Kawamura
,
J.
and
Cunha
,
G. R.
(
1996
).
Keratinocyte growth factor (KGF) can replace testosterone in the ductal branching morphogenesis of the rat ventral prostate
.
Int. J. Dev. Biol.
40
,
941
-
951
.
Tanji
,
N.
,
Tsuji
,
M.
,
Terada
,
N.
,
Takeuchi
,
M.
and
Cunha
,
G. R.
(
1994
).
Inhibitory effects of transforming growth factor-beta 1 on androgen-induced development of neonatal mouse seminal vesicles in vitro
.
Endocrinology
134
,
1155
-
1162
.
Thomson
,
A. A.
and
Cunha
,
G. R.
(
1999
).
Prostatic growth and development are regulated by FGF10
.
Development
126
,
3693
-
3701
.
Thomson
,
A. A.
and
Marker
,
P. C.
(
2006
).
Branching morphogenesis in the prostate gland and seminal vesicles
.
Differentiation
74
,
382
-
392
.
Thomson
,
A. A.
,
Timms
,
B. G.
,
Barton
,
L.
,
Cunha
,
G. R.
and
Grace
,
O. C.
(
2002
).
The role of smooth muscle in regulating prostatic induction
.
Development
129
,
1905
-
1912
.
Timms
,
B. G.
,
Mohs
,
T. J.
and
Didio
,
L. J.
(
1994
).
Ductal budding and branching patterns in the developing prostate
.
J. Urol.
151
,
1427
-
1432
.
Timms
,
B. G.
,
Lee
,
C. W.
,
Aumüller
,
G.
and
Seitz
,
J.
(
1995
).
Instructive induction of prostate growth and differentiation by a defined urogenital sinus mesenchyme
.
Microsc. Res. Tech.
30
,
319
-
332
.
Timms
,
B. G.
,
Howdeshell
,
K. L.
,
Barton
,
L.
,
Bradley
,
S.
,
Richter
,
C. A.
and
vom Saal
,
F. S.
(
2005
).
Estrogenic chemicals in plastic and oral contraceptives disrupt development of the fetal mouse prostate and urethra
.
Proc. Natl. Acad. Sci. USA
102
,
7014
-
7019
.
Tomlinson
,
D. C.
,
Freestone
,
S. H.
,
Grace
,
O. C.
and
Thomson
,
A. A.
(
2004
).
Differential effects of transforming growth factor-beta1 on cellular proliferation in the developing prostate
.
Endocrinology
145
,
4292
-
4300
.
Trupp
,
M.
,
Arenas
,
E.
,
Fainzilber
,
M.
,
Nilsson
,
A.-S.
,
Sieber
,
B.-A.
,
Grigoriou
,
M.
,
Kilkenny
,
C.
,
Salazar-Grueso
,
E.
,
Pachnis
,
V.
and
Arumäe
,
U.
(
1996
).
Functional receptor for GDNF encoded by the c-ret proto-oncogene
.
Nature
381
,
785
-
789
.
Trupp
,
M.
,
Scott
,
R.
,
Whittemore
,
S. R.
and
Ibanez
,
C. F.
(
1999
).
Ret-dependent and -independent mechanisms of glial cell line-derived neurotrophic factor signaling in neuronal cells
.
J. Biol. Chem.
274
,
20885
-
20894
.
Vega
,
Q. C.
,
Worby
,
C. A.
,
Lechner
,
M. S.
,
Dixon
,
J. E.
and
Dressler
,
G. R.
(
1996
).
Glial cell line-derived neurotrophic factor activates the receptor tyrosine kinase RET and promotes kidney morphogenesis
.
Proc. Natl. Acad. Sci. USA
93
,
10657
-
10661
.
Verhagen
,
A. P. M.
,
Aalders
,
T. W.
,
Ramaekers
,
F. C. S.
,
Debruyne
,
F. M. J.
and
Schalken
,
J. A.
(
1988
).
Differential expression of keratins in the basal and luminal compartments of rat prostatic epithelium during degeneration and regeneration
.
Prostate
13
,
25
-
38
.
Wang
,
B. E.
,
Shou
,
J.
,
Ross
,
S.
,
Koeppen
,
H.
,
de Sauvage
,
F. J.
and
Gao
,
W.-Q.
(
2003
).
Inhibition of epithelial ductal branching in the prostate by sonic hedgehog is indirectly mediated by stromal cells
.
J. Biol. Chem.
278
,
18506
-
18513
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information