Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

ORIGINAL RESEARCH

GPR30 Promotes Prostate Stromal Cell Activation via


Suppression of ER␣ Expression and Its Downstream
Signaling Pathway

Bona Jia, Yu Gao, Mingming Li, Jiandang Shi, Yanfei Peng, Xiaoling Du,

Downloaded from https://academic.oup.com/endo/article/157/8/3023/2422352 by biomedicas user on 06 February 2024


Helmut Klocker, Natalie Sampson, Yongmei Shen, Mengyang Liu, and Ju Zhang
Department of Biochemistry and Molecular Biology (B.J., Y.G., M.L., J.S., X.D., Y.S., J.Z.), College of Life
Sciences, Bioactive Materials Key Lab of the Ministry of Education, Nankai University, Tianjin 300071,
China; School of Integrative Medicine (Y.P.), Tianjin University of Traditional Chinese Medicine, Tianjin
300193, China; Department of Urology (H.K., N.S.), Division of Experimental Urology, Medical University
of Innsbruck, A-6020 Innsbruck, Austria; and Department of Nutrition and Food Science (M.L.), Texas
A&M University, College Station, Texas 77843

Cancer-associated fibroblasts (CAFs) play a vital role in malignant transformation and progression
of prostate cancer (PCa), and accumulating evidence suggests an enhancing effect of estrogens on
PCa. The present study aimed to investigate the possible origin of prostate CAFs and the effects of
estrogen receptors, G protein-coupled receptor 30 (GPR30) and estrogen receptor (ER)-␣, on stro-
mal cell activation. High expression of fibroblast activation protein (FAP), CD44, and nonmuscle
myosin heavy chain B (SMemb) accompanied by low expression of smooth muscle differentiation
markers was found in the stromal cells of PCa tissues and in cultured human prostate CAFs. Ad-
ditionally, SMemb expression, which is coupled to cell phenotype switching and proliferation, was
coexpressed with FAP, a marker of activated stromal cells, and with the stem cell marker CD44 in
the stromal cells of PCa tissue. Prostate CAFs showed high GPR30 and low ER␣ expression. More-
over, GPR30 was coexpressed with FAP, CD44, and SMemb. Furthermore, the study demonstrated
that the overexpression of GPR30 or the knockdown of ER␣ in prostate stromal cells induced the
up-regulation of FAP, CD44, Smemb, and the down-regulation of smooth muscle markers. The
conditioned medium from these cells promoted the proliferation and migration of LNCaP and PC3
PCa cells. GPR30 knockdown or ER␣ overexpression showed opposite effects. Finally, we present
a novel mechanism whereby GPR30 limits ER␣ expression via inhibition of the cAMP/protein kinase
A signaling pathway. These results suggest that stem-like cells within the stroma are a possible
source of prostate CAFs and that the negative regulation of ER␣ expression by GPR30 is centrally
involved in prostate stromal cell activation. (Endocrinology 157: 3023–3035, 2016)

lthough the vast majority of adenocarcinomas stem shown to induce human benign prostatic hyperplasia ep-
A from the epithelial compartment, there is evidence
indicating that tumorigenesis is not merely determined by
ithelial cell line (BPH-1) to form tumors (2), and CAFs
show a vital role in extracellular matrix remodeling and
the malignant cancer cells themselves but also by the tu- promoting cancer cell survival, proliferation and invasion
mor stroma (1–3). Carcinomas with abundant stroma (6 – 8).
show higher malignant degrees (4, 5), and cancer-associ- CAFs are broadly defined as all fibroblast cells within
ated fibroblasts (CAFs), which constitute most tumor the tumor-associated stroma, thus including fibroblasts
stroma, are prominent modifiers of cancer initiation and
progression. In a tissue recombination model, CAFs es- Abbreviations: AR, androgen receptor; CAF, cancer-associated fibroblast; CM, conditioned
medium; DAPI, 4⬘,6⬘-diamino-2-phenylindole; DMSO, dimethylsulfoxide; E2, estradiol; ER,
tablished from prostate cancer (PCa) tissue have been estrogen receptor; FAP, fibroblast activation protein; FBS, fetal bovine serum; FSK, fors-
kolin; GPR30, G protein-coupled receptor 30; HE, hematoxylin and eosin; hTERT, human
ISSN Print 0013-7227 ISSN Online 1945-7170 telomerase reverse transcriptase; IF, immunofluorescence; IHC, immunohistochemical;
Printed in USA MFI, mean fluorescence intensity; MMP, matrix metalloproteinase; NAF, normal fibroblast;
Copyright © 2016 by the Endocrine Society NP, normal prostate; PCa, prostate cancer; PKA, protein kinase A; P/S, penicillin/strepto-
Received January 19, 2016. Accepted May 3, 2016. mycin; RFP, red fluorescence protein; siRNA, small interfering RNA; SM, smooth muscle;
First Published Online May 10, 2016 ␣-SMA, ␣-smooth muscle actin; SMC, SM cell; SMemb, nonmuscle myosin heavy chain B.

doi: 10.1210/en.2016-1035 Endocrinology, August 2016, 157(8):3023–3035 press.endocrine.org/journal/endo 3023


3024 Jia et al GPR30 Promotes Prostate Stromal Cell Activation Endocrinology, August 2016, 157(8):3023–3035

and myofibroblasts (9). The origin of CAFs has been ex- from prostate tumor tissue were obtained from the Department
tensively investigated, and they have been shown to orig- of Urology, Medical University of Innsbruck (Innsbruck, Aus-
inate from different components. The most immediate tria). The primary stromal cells were isolated from prostate can-
cer patients undergoing radical prostatectomy after diagnosis of
source are resident fibroblasts (9). Smooth muscle cells
PCa according to the procedure described previously (26, 27).
(SMCs) are a plausible second source because the tumor
The use of prostate tissue samples was approved by the Ethics
stroma undergoes a progressive loss of SMCs in parallel to Committee of the Medical University of Innsbruck. Three pairs
the appearance of CAFs (10, 11). According to other re- of primary NAFs/CAFs and one pair of human telomerase re-
ports, CAFs originate from mesenchymal stem cells or verse transcriptase (hTERT)-immortalized NAFs/CAFs (11)
other mesenchymal cell types within the tumor such as were used in the experiments. The NAFs/CAFs were routinely
vascular smooth muscle cells, pericytes, or adipocytes, maintained in DMEM phenol red-free medium (Sigma-Aldrich)

Downloaded from https://academic.oup.com/endo/article/157/8/3023/2422352 by biomedicas user on 06 February 2024


which transdifferentiate into CAFs (12–14). Finally, epi- supplemented with 100 U/mL penicillin/streptomycin (P/S; In-
thelial cells, which have undergone epithelial-to-mesen- vitrogen) and 10% fetal bovine serum (FBS; Invitrogen) at 37°C
chymal transition, may also serve as a source of CAFs (4). under 5% CO2. The human prostate stromal cell line WPMY-1,
which was derived from normal stromal cells of the peripheral
Inconsistencies between these reports call for greater ef-
zone in the adult prostate and defined as myofibroblasts (28),
forts in elucidating the source and inductors of CAFs.
was obtained from the American Type Culture Collection. These
In addition, the factor(s) responsible for stromal cell cells were maintained in DMEM phenol red-free medium sup-
activation are not completely known. In prostatic carci- plemented with P/S and 5% FBS. WPMY-1 cells were treated
nogenesis sex hormones play a pivotal role (15, 16), and with vehicle (dimethylsulfoxide [DMSO]) or 1 ␮M of the ad-
accumulating evidence suggests that androgens and also enylyl cyclase agonist forskolin (FSK; Beyotime) for 24 hours.
estrogens participate in PCa progression (2, 17). The bi- Then cells were collected and lysed for protein extraction. The
ological effects of estrogens are mainly mediated through human PCa cell lines LNCaP and PC3 were obtained from the
estrogen receptors. Estrogen receptor (ER)-␣ and ER␤ are American Type Culture Collection. All PCa cell lines were grown
ligand-inducible transcription factors belonging to the nu- in RPMI 1640 phenol red-free medium (Sigma) supplemented
with P/S and 10% FBS.
clear receptor superfamily and mediate classical estrogen
actions. In addition, G protein-coupled receptor 30
Plasmids and small interfering RNA (siRNA)
(GPR30), also called G protein-coupled estrogen receptor,
transfection
an orphan receptor, has been recently discovered to be
Human cDNA fragments encoding GPR30 and ER␣ were
involved in mediating some estrogen actions, in particular
PCR amplified from reverse transcribed WPMY-1 cell total
the nongenomic ones (18). Estrogen actions reported in RNA. Red fluorescence protein (RFP) cDNA was amplified from
stromal cells are mainly mediated by ER␣ and GPR30. plasmid pDsRed-N1. The resulting cDNA fragments were
Previous studies reported that estradiol (E2) regulates ma- cloned into the PLenti6.4-R4R2-V5-Dest vector to generate
trix metalloproteinase (MMP)-2 expression and enolase 1 RFP, GPR30, and ER␣ expression constructs. The three expres-
secretion through ER␣ in prostate stromal WPMY-1 cells sion plasmids were transfected into WPMY-1 cells using Lipo-
(19, 20). Another study showed ER␣ regulates thrombos- fectamine 3000 reagent (Invitrogen). Transfected cells, labeled
pondin 2 expression in CAFs and thereby decreases the as WPMY1-RFP/GPR30/ER␣, were harvested 72 hours after
invasive capability of PCa cells (21). In breast cancer transfection for RNA or protein extraction.
siRNAs targeting GPR30 or ER␣ and a negative control siRNA
CAFs, E2, and the GPR30-selective ligand G-1 up-regulated
that did not match any known human cDNA were purchased from
connective tissue growth factor and vascular endothelial
GenePharma. The sequences used were 5-GCUGUACAUUGAG
growth factor in a GPR30-dependent manner (22, 23). Both, CAGAAATT-3 (forward) and 5-UUUCUGCUCAAUGUA
ER␣ and GPR30 have been reported to be involved in SMC CAGCTT-3 (reverse) for GPR30, 5-UCAUCGCAUUCCUUG
differentiation in several tissue types (24, 25). CAAATT-3 (forward) and 5-UUUGCAAGGAAUGCGAU
In this study, we investigated the possible origin of GATT-3 (reverse) for ER␣, and 5-UUCUCCGAACGUGU
CAFs in PCa. We studied the contribution of GPR30 and CACGUTT-3 (forward) and 5-ACGUGACACGUUCG
ER␣ on phenotypic changes of stromal cells and on PCa GAGAATT-3 (reverse) for the negative control siRNA. A second
cell proliferation and migration. Furthermore, we dem- pair of siRNAs was used to exclude off-target effects (see Supple-
mental Materials for these siRNA sequences and Supplemental
onstrated the interconnected regulation between GPR30
Figure 1 for the knockdown verification). WPMY-1 cells and im-
and ER␣ and described the molecular mechanism thereof.
mortalized NAFs/CAFs were transfected with the corresponding
siRNA using Lipofectamine 3000 reagent (Invitrogen) according
Materials and Methods to the manufacturer’s instructions. For treatment, the transfected
cells were incubated with 2 ␮M protein kinase A (PKA) inhibitor
Cell culture and treatment H89 (Beyotime) in DMSO or an equivalent amount of the solvent.
Primary human prostate normal fibroblasts (NAFs) estab- Cells were collected for RNA or protein extraction 72 hours after
lished from normal prostate tissue and prostate CAFs established transfection.
doi: 10.1210/en.2016-1035 press.endocrine.org/journal/endo 3025

Preparation of conditioned medium (CM) Cell proliferation assay


Prostate stromal cells were transfected with expression plas- For quantitative proliferation assay, 20 000 PCa cells sus-
mids or siRNA in DMEM with 5% FBS for 24 hours. Afterward pended in complete medium were seeded in 24-well plates to-
the media were changed to DMEM with 2% FBS. After a further gether with an equal volume of CM. All treatments were repeated
48 hours, media were collected, centrifuged at 400 ⫻ g for 5 three times. For the determination of cell numbers after 24, 48,
minutes to remove the floating cells, and used immediately or 72, and 96 hours, respectively, cells were trypsinized and
frozen at ⫺20°C until use. The CM volumes were normalized to counted using a hemacytometer.
cell numbers.
Transwell migration assay
Real-time RT-PCR assay PCa cells were seeded in Transwell inserts with 8 ␮m pore size
(BD Biosciences) at 20 000 cells per well in DMEM/F12 serum-

Downloaded from https://academic.oup.com/endo/article/157/8/3023/2422352 by biomedicas user on 06 February 2024


Total RNA was extracted using Trizol reagent (Invitrogen).
free medium with 10 ng/mL mitomycin C to inhibit cell growth.
Reverse transcription was performed as described previously
CMs were added to the bottom chamber. After 24 hours, cells on
(24). SYBR Green I-based real-time quantitative PCR was per-
the upper surface of the filter were removed using a cotton swab.
formed on an MJ Research DNA engine Opticon continuous
Cells that had migrated through the filter to the lower surface
fluorescence detection system (Opticon Monitor II; MJ Re-
were fixed with 4% paraformaldehyde and stained with 0.5%
search, Inc). Specific primer pairs are listed in Table 1. The rel-
crystal violet. Cells were counted from five randomly selected
ative gene expression was determined using the comparative CT fields per chamber.
method and normalized to the housekeeping gene hypoxanthine
phosphoribosyltransferase 1 (HPRT).
Wound-healing assay
PCa cells were seeded in equal numbers in six-well plates.
Western blot assay Once 90% confluent, the medium was changed to DMEM/F12
Cells were lysed with radioimmunoprecipitation assay buffer serum-free medium for 24 hours. Then three vertical wounds per
to extract the total protein. Protein was quantified using the well were scratched using a 10-␮L pipette tip and cells incubated
bicinchoninic acid method. Equal amounts of proteins were sep- with the indicated CM from prostate stromal cells supplemented
arated by SDS-PAGE and then transferred to a polyvinyl diflu- with 10 ng/mL mitomycin C to inhibit cell growth. Images were
oride membrane. The membrane was blotted with primary an- collected at the designated time thereafter at 100-fold magnifi-
tibodies and secondary antibodies (goat antirabbit or goat cation to assess wound closure.
antimouse horseradish peroxidase conjugated, 1:5000; Bio-Rad
Laboratories). The primary antibody dilutions used are given in Histological and immunohistochemical studies
Table 2. After the treatment with enhanced chemiluminescence PCa (seven patients who underwent radical prostatectomy
reagent (Pierce), the membrane was exposed to an x-ray film for prostate carcinoma, Gleason 4⫹3 ⫽ 7) and normal prostate
(Kodak) and ImageJ software (National Institutes of Health, (six autopsy cases of patients who died due to illness other than
Bethesda, Maryland) used for densitometry quantification. malignancy) tissues were obtained from the Department of Pa-

Table 1. Nucleotide Sequence of Primers Used in Real-Time RT-PCR


Gene Primer Sequences Annealing Temperature, Product
Name (5ⴕ–3ⴕ) °C Size, bp
␣-SMA Forward, GGGACATCAAGGAGAAACTG 56 219
Reverse, TGATGCTGTTGTAGGTGGTT
Calponin Forward, TTGAGGCCAACGACCTGTTT 56 137
Reverse, TTTCCGCTCCTGCTTCTCTG
CD44 Forward, TCCAACACCTCCCAGTATG 58 149
Reverse, TTCTGGACATAGCGGGTG
ER␣ Forward, GGACCATATCCACCGAGTCCTG 56 348
Reverse, GCCTCCCCCGTGATGTAATAC
FAP Forward, CCCTGCGTATGTAGGTCC 60 175
Reverse, TGTCTGCCAGTCTTCCCT
GPR30 Forward, TTGTGGGCAACATCCTGA 56 252
Reverse, CGATGTAGCGGTCGAAGC
HPRT Forward, TGACACTGGCAAAACAATGCA 56 94
Reverse, GGTCCTTTTCACCAGCAAGCT
MYH11 Forward, AGAAGCCAGGGAGAAGGAAACCAA 68 131
Reverse, TGGAGCTGACCAGGTCTTCCATTT
SM22␣ Forward, GGTTAGGCCAAGGCTCTACTGT 56 184
Reverse, GGGCCACACTGCACTATGAT
SMemb Forward, TCTTGGGATGAATGTGATG 56 172
Reverse, ATTGATGCGATGAACGAG
Abbreviation: HPRT, hypoxanthine phosphoribosyltransferase 1.
3026 Jia et al GPR30 Promotes Prostate Stromal Cell Activation Endocrinology, August 2016, 157(8):3023–3035

Table 2. Antibody Table


Manufacturer,
Catalog Number,
Peptide/ and/or Name of Species Raised
Protein Antigen Sequence Name of Individual Providing (Monoclonal or Dilution
Target (if Known) Antibody the Antibody Polyclonal) Used
␣-SMA ␣-smooth muscle Boster, BM0002 Mouse monoclonal 1:100 (IHC)
actin 1:300 (WB)
␤-Actin ␤-Actin Boster, BM0627 Mouse monoclonal 1:400 (WB)
Calponin Calponin 1 Santa Cruz Biotechnology, Mouse monoclonal 1:100 (IHC)
sc-58707 1:400 (WB)

Downloaded from https://academic.oup.com/endo/article/157/8/3023/2422352 by biomedicas user on 06 February 2024


CD44 CD44 Abcam, ab51037 Rabbit monoclonal 1:100 (IHC/IF)
1:2000 (WB)
CD44 CD44 Cell Signaling, 3570 Mouse monoclonal 1:100 (IF)
ER␣ Amino acids 1–300 ER␣ Abcam, ab108398 Rabbit monoclonal 1:100 (IHC/IF)
1:1000 (WB)
FAP Fibroblast activation Abcam, ab53066 Rabbit polyclonal 1:100 (IHC/IF)
protein, ␣ 1:500 (WB)
FAP FAP Santa Cruz Biotechnology, Mouse monoclonal 1:100 (IF)
sc-65398
GPR30 Amino acids 307–375 GPR30 Abcam, ab154069 Rabbit polyclonal 1:100 (IHC/IF)
1:1000 (WB)
MYH11 MYH11 Santa Cruz Biotechnology, Mouse monoclonal 1:100 (IHC)
sc-6956 1:500 (WB)
SM22␣ N-terminus amino acids Transgelin Santa Cruz Biotechnology, Rabbit polyclonal 1:100 (IHC)
16 –90 sc-50446 1:300 (WB)
SMemb Nonmuscle myosin Yamasa, 7602 Mouse monoclonal 1:1000 (IHC/IF)
heavy chain 1:2000 (WB)
Abbreviation: WB, Western blot.

thology and the Department of Urology, the Second Affiliated ture, and the DNA of cell nuclei was counterstained with 4⬘,6⬘-
Hospital of Tianjin Medical University (Tianjin, China). Use of diamino-2-phenylindole (DAPI; 1:100 dilution; Roche;
the tissue samples in this study was approved by the institutional 236276). The sections were then analyzed with a confocal laser-
review board. Hematoxylin and eosin (HE) staining and immu- scanning microscope. The mean fluorescence intensity (MFI) in
nohistochemical (IHC) staining were carried out as previously the stromal compartment was quantified with Photoshop soft-
described (29). Briefly, 5-␮m sections were deparaffinized in xy- ware. At least nine randomly selected microscopic fields were
lene and rehydrated in a graded series of alcohol. One section was analyzed, and the mean values were calculated.
HE stained for histological examination, and the other sections
were processed for immunohistochemistry using the avidin-bi- cAMP measurement
otin-peroxidase complex method. The poach method with ci- WPMY-1 cells were transfected with plasmids or siRNA as
trate buffer (pH 6.0) was used for antigen retrieval. Sections were described above. Forty-eight hours after transfection, cells were
incubated with the primary antibodies in 10% bovine serum in rinsed once with PBS and lysed. As positive control, WPMY-1
PBS overnight at 4°C, followed by the respective biotinylated cells were treated with DMSO or 1 ␮M FSK for 10 minutes. The
secondary antibodies and horseradish-peroxidase streptavidin. lysates were subsequently collected and subjected to a cAMP
Primary antibody dilutions used are given in Table 2. The stained luminescence assay according to the manufacturer’s instruction
slides were mounted and visualized in the bright field with an (cAMP-Glo assay, V1501; Promega).
Olympus CX41 microscope. Pictures were obtained using a mag-
nification of ⫻200. Positively stained cells were counted in at Statistical analysis
least nine randomly selected microscopic fields and the mean Data are expressed as the mean ⫾ SD. Significance was as-
values were calculated. sessed using a Student’s paired t test. P ⬍ .05 was considered as
significant.
Immunofluorescence (IF) double-staining assay
Before blocking of paraffin sections with 10% normal bovine Results
serum in PBS, the poach method with citrate buffer (pH 6.0) was
performed for antigen retrieval. Then sections were incubated Increasing number of nonmuscle myosin heavy
with two types of primary antibodies at 4°C overnight using
chain B (SMemb)-positive stem-like cells
antibody dilutions specified in Table 2. Sections were subse-
quently washed and incubated with Alexa Flour 594 anti-mouse accompanies a decrease of SMCs in PCa stroma
IgG (1:500 dilution; Invitrogen) and Alexa Flour 488 anti-rabbit To better understand the phenotypic characteristic of
IgG (1:1000 dilution; Invitrogen) for 1 hour at room tempera- activated stromal cells in human PCa, PCa tissues were
doi: 10.1210/en.2016-1035 press.endocrine.org/journal/endo 3027

whereas no positive staining was de-


tected in NP stroma. Moreover, the
positive rate of CD44, a marker of
prostate-resident mesenchymal stem
cells (31), was 26.5% ⫾ 7.5% in PCa
stroma in contrast to only 1.8% ⫾
0.5% in the NP stroma. Whereas
92.7% ⫾ 5.2% of PCa stromal cells
stained positive for ␣-smooth muscle
actin (␣-SMA), the positive cells ex-

Downloaded from https://academic.oup.com/endo/article/157/8/3023/2422352 by biomedicas user on 06 February 2024


hibited lower immunostaining inten-
sity compared with NP stroma. The
positive rates of smooth muscle (SM)
differentiation markers SM22␣, cal-
ponin, and MYH11 in PCa stroma
(30.5% ⫾ 4.2%, 25.7% ⫾ 6.4%,
and 21.3% ⫾ 7.5%, respectively)
were much lower than in the NP
stroma (93% ⫾ 5.7%, 85.5% ⫾
6.3%, and 93.7% ⫾ 4%, respec-
tively), whereas the SMemb, a
marker indicating the cell phenotype
switch, increased from 0.3% ⫾
0.2% in the NP stroma to 25.2% ⫾
7.1% in the PCa stroma (Figure 1, A
and B).
In addition, basal expression lev-
els of these markers were measured
in primary human prostate NAFs
and CAFs. Consistent with tissue
IHC, a 1.66-, 4.17-, and 2.15-fold
increase of FAP, CD44, and SMemb
mRNA expressions, respectively,
were observed in CAFs compared
with NAFs by real-time RT-PCR.
These changes were accompanied by
a 2.56-, 2.28-, 4-, and 11.1-fold de-
Figure 1. Increased SMemb-positive stem-like cells and decreased SMCs in PCa stroma. A, IHC
analysis of SM differentiation markers and FAP, CD44, and SMemb expression in human NP and crease in ␣-SMA, SM22␣, calponin,
PCa tissue. Bar, 100 ␮m. B, Mean (SE) percentage of stromal area per high-power field (n ⫽ 9) and MYH11 mRNA expression, re-
stained positive for FAP, CD44, Smemb, and SM differentiation markers in NP (n ⫽ 6) and PCa spectively (Figure 1C, upper panel).
(n ⫽ 7) tissue. *, P ⬍ .05, PCa vs NP. C, Real-time RT-PCR and Western blot analysis of FAP,
CD44, Smemb, and SM differentiation markers expression at mRNA (upper panel) and protein
Comparable changes of these mark-
level (lower panel) in primary NAFs and CAFs. ImageJ software (National Institutes of Health) was ers were also observed at the protein
used for densitometric quantification of proteins. Results are presented as mean ⫾ SD (n ⫽ 3). level via Western blotting (Figure
*, P ⬍ .05, CAFs vs NAFs. D, Real-time RT-PCR analysis of FAP, CD44, Smemb, and SM
1C, lower panel). These differences
differentiation markers expression in hTERT-immortalized NAFs and CAFs. Results are presented
as mean ⫾ SD (n ⫽ 3). *, P ⬍ .05, CAFs vs NAFs. E, Serial sections from each group were in gene expression were also detected
analyzed by HE staining and double-IF staining of SMemb with FAP or SMemb with CD44 in PCa in the hTERT-immortalized NAFs
tissue. Cell nuclei were counterstained with DAPI (blue). Bar, 100 ␮m. and CAFs (Figure 1D).
To address the coexpression of
compared with normal prostate (NP) tissues by IHC. The FAP, CD44, and SMemb, double-IF staining was per-
well-established activated fibroblast marker fibroblast ac- formed in PCa tissue sections. Of interest, all SMemb-
tivation protein (FAP) (6, 30) was specifically detected in positive cells strongly coexpressed FAP and thus constitute
PCa stroma with a positive rate of 65.9% ⫾ 7.5%, an SMemb⫹FAP⫹ subtype within the FAP-positive cells.
3028 Jia et al GPR30 Promotes Prostate Stromal Cell Activation Endocrinology, August 2016, 157(8):3023–3035

Nearly all SMemb-positive cells also coexpressed CD44 (Figure performed in tissue sections. As shown in Figure 2, A and
1E). Taken together, the IHC and IF results suggest that the B, all cells with strong FAP-positive staining and some cells
appearance of prostate CAFs coincides with a substantial re- with low FAP-positive staining coexpressed GPR30; how-
duction in numbers of differentiated SMCs and a significant ever, no FAP⫹ER␣⫹ cells were detected in PCa stroma. In
increase of SMemb/CD44/FAP-positive stem-like cells. NP stromal cells, GPR30 was not only expressed at lower
levels compared with PCa stromal cells, but it was also
Prostate CAFs show high GPR30 but low ER␣ found to be primarily localized to the nucleus (Figure 2B,
expression arrow). By contrast, GPR30 was present in the cell mem-
To investigate which ER(s) may be involved in stromal brane and the cytoplasm of PCa stromal cells (Figure 2B,
cell activation, double-IF staining for FAP and ERs was arrowhead). Quantification revealed GPR30 up-regula-

Downloaded from https://academic.oup.com/endo/article/157/8/3023/2422352 by biomedicas user on 06 February 2024

Figure 2. Prostate CAFs show high GPR30 but low ER␣ expression. A, Serial sections from each group were analyzed by HE staining and double-
IF staining of FAP with ER␣ in NP and PCa tissue (upper panel) and mean of florescent intensity of the images (MFI) of stromal ER␣ (lower panel)
(n ⫽ 6). Cell nuclei were counterstained with DAPI (blue). Bar, 100 ␮m. *, P ⬍ .05, PCa vs NP. B, Serial sections from each group were analyzed by
HE staining and double-IF staining of FAP with GPR30 in NP and PCa tissue (upper panel) and MFI of stromal GPR30 (lower panel). Bar, 100 ␮m.
*, P ⬍ .05, PCa vs NP. C and D, Real-time RT-PCR and Western blot analysis of ER␣ and GPR30 expression at mRNA (C) and protein level (D) in
primary NAFs and CAFs. Results are presented as mean ⫾ SD (n ⫽ 3). *, P ⬍ .0,5 CAFs vs NAFs. E, Real-time RT-PCR analysis of ER␣ and GPR30
expression in hTERT-immortalized NAFs and CAFs. Results are presented as mean ⫾ SD (n ⫽ 3). *, P ⬍ .05, CAFs vs NAFs. F, Serial sections from
each group were analyzed by HE staining and double-IF staining of CD44 with GPR30 and SMemb with GPR30 in PCa tissue. Bar, 100 ␮m.
doi: 10.1210/en.2016-1035 press.endocrine.org/journal/endo 3029

CAF cells and obtained congruent


results (Figure 2E). Furthermore,
double-IF staining revealed that
in addition to FAP, GPR30 was
strongly coexpressed with CD44 and
SMemb (Figure 2F).
Taken together, our results sug-
gest that the acquisition of an acti-
vated fibroblasts phenotype is posi-
tively related to the expression of

Downloaded from https://academic.oup.com/endo/article/157/8/3023/2422352 by biomedicas user on 06 February 2024


GPR30 but negatively to the expres-
sion of ER␣ in the PCa stroma.

Overexpression of GPR30 or
knockdown of ER␣ in stromal
cells promote PCa cell
proliferation and migration in a
paracrine manner
The inverse expression of GPR30
and ER␣ in prostate CAFs prompted
us to investigate their roles in PCa
progression via a stromal to epithe-
lial stimulation. WPMY-1 cells were
transfected with expression plasmid
or siRNA for GPR30 and ER␣, re-
spectively (Figure 3, A and C), and
the CM was subsequently collected
to study the effects on PCa cells. We
used two prostate epithelial cell
lines, the less invasive PCa cell line
LNCaP and the highly malignant
Figure 3. High expression of GPR30 or low expression of ER␣ in stromal cells promote PCa cell
proliferation in a paracrine manner. A, C, E, and G, Prostate stromal cells were transfected with PCa cell line PC3 as models. As
expression plasmid (A) or siRNA (C, E, and G) for GPR30 and ER␣. Overexpression and shown in Figure 3, B and D, the CM
knockdown were verified by Western blot analysis after 72 hours. Notice that Western blot collected from WPMY-1 cells over-
images were obtained using different sensitivity settings for overexpression and knockdown
experiments in WPMY-1 cells, lower sensitivity for overexpression (A), and higher sensitivity for expressing GPR30 promoted LN-
knockdown experiments (C). B, D, F, and H, PCa cell lines LNCaP and PC3 were treated with CM CaP and PC3 cell proliferation,
from transfected stromal cells. Cells were counted after 24, 48, 72, and 96 hours, respectively. whereas the silencing of GPR30 ab-
The proliferation curves of LNCaP and PC3 cells are depicted (n ⫽ 3).
rogated the promotion effect. In
contrast, the CM collected from
tion by 3.4-fold and ER␣ down-regulation by 5-fold, re- WPMY-1 cells overexpressing ER␣
spectively, in PCa stroma (Figure 2, A and B, lower panel). suppressed LNCaP and PC3 cell proliferation, whereas
The up-regulation of GPR30 and down-regulation of ER␣ the silencing of ER␣ enhanced the CM ability of the
in the stroma of PCa tissues were confirmed by IHC stain- WPMY-1 cells to promote PCa cell proliferation. The
ing (Supplemental Figure 2). same results were obtained using hTERT-immortalized
In vitro, using real-time RT-PCR, we confirmed that prostate NAF and CAF cells. The CM from immortalized
GPR30 mRNA expression is increased by 5.1-fold in CAF cells with GPR30 knockdown showed a diminished
CAFs compared with NAFs, which is accompanied with a ability to promote PCa cell proliferation (Figure 3, E and
2.5-fold decrease in ER␣ mRNA (Figure 2C). Western F), whereas the CM collected from immortalized NAF
blotting revealed a 2.9-fold increase of GPR30 and 4-fold cells with ER␣ knockdown promoted LNCaP and PC3 cell
decrease of ER␣ at protein levels in CAFs compared with proliferation (Figure 3, G and H).
NAFs (Figure 2D). We confirmed the expression of On the other hand, we investigated the effects of the
GPR30 and ER␣ mRNA in the immortalized NAF and CMs obtained above in a Transwell migration assay (BD
3030 Jia et al GPR30 Promotes Prostate Stromal Cell Activation Endocrinology, August 2016, 157(8):3023–3035

Biosciences). Results showed in-


creased numbers of migrated LNCaP
and PC3 cells with the CM from
WPMY-1 GPR30 cells, whereas the
numbers of migrated LNCaP and
PC3 cells were decreased with the
CM from WPMY-1 ER␣ cells (Fig-
ure 4A). Conversely, the CM of
WPMY-1 siGPR30 significantly re-
duced LNCaP and PC3 cell migra-

Downloaded from https://academic.oup.com/endo/article/157/8/3023/2422352 by biomedicas user on 06 February 2024


tion, whereas the CM of WPMY-1
siER␣ increased LNCaP and PC3
cell migration (Figure 4B). Similarly,
the CM from immortalized CAF
cells with GPR30 knockdown
showed a diminished ability to pro-
mote PCa cell migration (Figure 4C),
whereas the silencing of ER␣ en-
hanced the CM ability of the immor-
talized NAF cells to promote LNCaP
and PC3 cell migration (Figure 4D).
Additionally, these findings were
confirmed by wound-healing assays
(Figure 4, E–H). Taken together, our
data suggest that the expression of
GPR30 in the tumor stroma may
play a promoting role, whereas ER␣
expression may have a suppressing
role upon PCa progression.

High expression of GRP30 and/


or low expression of ER␣ is
positively related to the CAF-
like phenotype of prostate
stromal cells
To elucidate the mechanism by
which stromal GPR30 and ER␣ were
able to exert their effects on PCa pro-
gression, we further investigated
their functions on stromal cell phe-
notypic switching. We scrutinized
Figure 4. CM from stromal cells with high expression of GPR30 or low expression of ER␣ the expression of FAP, CD44,
stimulate PCa cell migration. A–D, The Transwell assay was performed to evaluate the effect of Smemb, and the differentiation
CM from transfected stromal cells on migration of LNCaP and PC3 cells. CMs were placed into
markers at both the mRNA and pro-
the lower chamber of the Transwell plates (BD Biosciences). LNCaP and PC3 cells were put
into the top chamber and allowed to migrate for 24 hours. The Transwell inserts were fixed tein levels in WPMY-1 cells, which
and stained before light microscopy visualization and cell number counting. Results are were transfected with expression
presented as mean ⫾ SD (n ⫽ 5). *, P ⬍ .05 vs controls. Bar, 200 ␮m. E–H, Wound-healing plasmids or siRNA for GPR30 and
assay of LNCaP and PC3 cells incubated for 0 or 24 hours with CM from transfected stromal
cells. ImageJ software (National Institutes of Health) was used for the quantification of ER␣. As shown in Figure 5, A and B,
wound closure. Results are presented as mean ⫾ SD (n ⫽ 3). *, P ⬍ .05 vs controls. Bar, FAP, CD44, and SMemb were sig-
200 ␮m. nificantly up-regulated in WPMY-1
cells upon GPR30 overexpression,
together with the substantial down-
doi: 10.1210/en.2016-1035 press.endocrine.org/journal/endo 3031

regulation of ␣-SMA, SM22␣, cal-


ponin, and MYH11. In contrast, the
expression levels of FAP, CD44, and
SMemb were dramatically reduced
in WPMY-1 cells and the immortal-
ized CAFs upon GPR30 knock-
down, whereas the expression levels
of differentiation markers were in-
creased (Figure 5, C–F).
On the other hand, in WPMY-1

Downloaded from https://academic.oup.com/endo/article/157/8/3023/2422352 by biomedicas user on 06 February 2024


cells with ER␣ overexpression, FAP,
CD44, and SMemb were down-reg-
ulated, but ␣-SMA, SM22␣, cal-
ponin, and MYH11 were up-regu-
lated (Figure 5, G and H), and
knocking down ER␣ resulted in a re-
markable opposite regulation in
both WPMY-1 cells and the immor-
talized NAFs (Figure 5, I–L). All re-
sults suggest that high GPR30 ex-
pression and/or low ER␣ expression
facilitate the induction of a CAF-like
phenotype in prostate stromal cells.

GPR30 represses ER␣ expression


in prostate stromal cells via
inhibition of the cAMP/PKA
signaling pathway
Because GPR30 and ER␣ show
opposite roles in stromal cell activa-
tion, we further investigated whether
GPR30 and ER␣ could regulate each
other’s expression. Interestingly, the
overexpression of GPR30 resulted in
a significant down-regulation of
ER␣ at both the mRNA and protein
levels, whereas the knockdown of
GPR30 resulted in a dramatic up-
regulation of ER␣ in WPMY-1 cells
(Figure 6, A and B). Conversely, the
overexpression or knockdown of
ER␣ induced no significant regula-
Figure 5. High expression of GPR30 and/or low expression of ER␣ is related to the CAF-like
phenotype in prostate stromal cells. A–D, Real-time RT-PCR and Western blot analysis of gene
tion of GPR30 expression (Figure 6,
expression in WPMY-1 cells transfected with GPR30 expression plasmid (A and B) or siRNA (C C and D). The interconnected regu-
and D) at the mRNA level and protein level. E and F, Real-time RT-PCR and Western blot analysis lation was also confirmed in the im-
of gene expression in immortalized CAFs transfected with siGPR30 at the mRNA (E) and protein
mortalized NAFs and CAFs (Figure
level (F). G–J, Real-time RT-PCR and Western blot analysis of gene expression in WPMY-1 cells
transfected with ER␣ expression plasmid (G and H) or siRNA (I and J) at the mRNA and protein 6, E and F).
level. K and L, Real-time RT-PCR and Western blot analysis of gene expression in immortalized It was reported that FSK stimu-
NAFs transfected with siER␣ at mRNA (K) and protein level (L). Results are presented as mean ⫾ lates ER␣ expression by activating
SD (n ⫽ 3). *, P ⬍ .05 vs controls.
the PKA pathway (32), whereas
other reports indicated that GPR30
suppresses PKA activity by inhibit-
3032 Jia et al GPR30 Promotes Prostate Stromal Cell Activation Endocrinology, August 2016, 157(8):3023–3035

ing cAMP production (33, 34). This


led us to examine whether GPR30
represses ER␣ expression though in-
hibiting PKA activity. Thus, we
quantified cAMP concentrations af-
ter transfection of WPMY-1 with
GPR30 expression plasmid or spe-
cific siRNA. As shown in Figure 6G,
cAMP levels were reduced 3.1-fold
in WPMY-1 cells upon GPR30 over-

Downloaded from https://academic.oup.com/endo/article/157/8/3023/2422352 by biomedicas user on 06 February 2024


expression, whereas knockdown in-
creased cAMP concentration by 2.3-
fold. FSK, a cAMP activator used as
a positive control, rapidly stimulated
cAMP production and, as expected,
increased ER␣ protein expression in
WPMY-1 cells (Figure 6H). We then
treated stromal cells with the selec-
tive PKA inhibitor H89 after trans-
fection of siGPR30. As shown in
Figure 6I, H89 blocked the up-regu-
lation of ER␣ protein expression
by GPR30 knockdown in both
WPMY-1 cells and immortalized
CAFs. Thus, these data provide the
evidence that GPR30 negatively reg-
ulates ER␣ expression through in-
hibiting the cAMP/PKA signaling
pathway in prostate stromal cells.
Overall, our data reveal a mecha-
nism summarized in Figure 7:
GPR30 inhibits ER␣ expression and
ER␣ downstream signaling path-
ways. A high GPR30/ER␣ ratio sub-
sequently induces a CAF-like pheno-
type switch of prostate stromal cells.

Discussion
In this study, we found an increased
expression of FAP accompanied by
Figure 6. GPR30 represses ER␣ expression in prostate stromal cell via inhibition of the cAMP/PKA
an enhanced expressions of CD44
signaling pathway. A and B, Real-time RT-PCR and Western blot analysis of ER␣ expression in WPMY1-
GPR30 or WPMY1-siGPR30 at mRNA (A) and protein level (B). C and D, Real-time RT-PCR and Western and SMemb and a substantial down-
blot analysis of GPR30 expression in WPMY1-ER␣ or WPMY1-siER␣ at the mRNA (C) and protein level (D). regulation of SM differentiation
E, Real-time RT-PCR and Western blot analysis of ER␣ expression in immortalized CAFs transfected with
siGPR30 at mRNA (left panel) and protein level (right panel). F, Real-time RT-PCR and Western blot analysis markers in stromal cells of prostate
of GPR30 expression in immortalized NAFs transfected with siER␣ at mRNA (left panel) and protein level cancer tissues and cultured stromal
(right panel). G, WPMY-1 cells were transfected with GPR30 expression plasmid or siRNA for 48 hours or cells derived therefrom. Moreover,
treated with 1 ␮M FSK for 10 minutes, and then concentrations of cAMP were quantified using the
cAMP-Glo assay (n ⫽ 3). *, P ⬍ .05 vs corresponding controls (CTLs). H, WPMY-1 cells were treated with 1 CD44 and SMemb were largely co-
␮M FSK for 24 hours, and ER␣ protein expression was demonstrated by Western blot. I, Western blot expressed in PCa stromal cells, and
analysis of ER␣ protein expression in WPMY-1 cells (left panel) and immortalized CAFs (right panel)
transfected with siGPR30 and treated with 2 ␮M H89 (upper panel), and quantification by densitometry
double-positive CD44⫹SMemb⫹ cells
(lower panel). Results are presented as mean ⫾ SD (n ⫽ 3). *, P ⬍ .05. strongly coexpress FAP.
doi: 10.1210/en.2016-1035 press.endocrine.org/journal/endo 3033

the cell membrane and the cytoplasm, whereas GPR30


showed predominant nuclear localization in NP stromal
cells. In the vast majority of previous studies, GPR30 ap-
peared to be expressed on the cell surface and the endo-
plasmic reticulum of several cancer or stromal cells (40 –
42). However, in cultured breast CAFs, GPR30 was
reported to translocate into the nucleus through an im-
portin-dependent mechanism, and its nuclear localization
is required to stimulate the up-regulation of c-fos, one of

Downloaded from https://academic.oup.com/endo/article/157/8/3023/2422352 by biomedicas user on 06 February 2024


the GPR30 target genes (22, 43). These data suggest that
the cellular localization of GPR30 may be related to its
physiological function and its tissue specificity.
We further demonstrate that the high expression of
GPR30 in prostate stromal cells up-regulates FAP, CD44,
and SMemb and down-regulates SM differentiation mark-
Figure 7. A schematic illustration of the proposed mechanism. GPR30
lowers cAMP and inhibits PKA activity, which represses ER␣ expression ers. Opposite to GPR30, the high expression of ER␣ in
and attenuates ER␣-mediated stimulation of SM differentiation and stromal cells is accompanied by the low expression of FAP,
inhibition of CAF-like phenotype switching. High GPR30/low ER␣ CD44, and SMemb and the high expression of SM differ-
supports the CAF phenotype, which enhances the release of
proliferation and migration-promoting factors into the supernatant for entiation markers. This suggests that high GPR30 and/or
paracrine stimulation of prostate tumor cells. low ER␣ levels facilitate the phenotype switch of stromal
cells toward CAFs. A role of ER␣ in prostate stromal cells
phenotype modulation was reported previously (24).
SMemb, an embryonic-type member of the nonmuscle
However, to our knowledge, our report is the first to link
myosin heavy chain isoform family was previously re-
GPR30 to prostate CAF induction, identifying it as a cru-
ported to be abundantly expressed by activated mesen-
cial modifier of the stromal cell phenotype.
chymal cells in cardiac myxomas and atherosclerosis (35,
Another important finding of our study is the repres-
36), and its expression was coupled to the cell phenotype
sion of ER␣ expression by GPR30, whereas ER␣ demon-
switch and proliferation (37). Coexpression of SMemb
strated no effect on GPR30 expression in prostate stromal
and the stem cell marker CD44 in PCa stroma indicates
cells. Contrary to our finding, Ariazi et al (44) previously
that most stromal stem-like cells have been abnormally
reported the E2-mediated down-regulation of GPR30
reawakened/reactivated and undergone a phenotypic
mRNA via ER in breast cancer cells. Similarly, the group
change to gain a higher proliferative capacity. It is well
accepted that stem cells have a high plasticity (38), and the of Gao et al (45) found that the activation of GPR30 by
existence of CD44⫹SMemb⫹FAP⫹ cells indicates that the G-1 inhibited ER␣ (Ser118) phosphorylation signals in the
reactivated stromal stem-like cells differentiate into FAP- mouse uterus.
positive CAFs during PCa development. Yet we also de- The CM from the stromal cells with high GPR30 or low
tected FAP⫹CD44⫺ cells within the PCa stroma. Accord- ER␣ expression have promoting effects on the growth and
ing to a previous report CD44-positive cells are able to migration of PCa cells. These findings are consistent with
generate CD44-negative cells by undergoing asymmetric a large body of literature demonstrating that activated
division and to develop into fully differentiated myofibro- stromal cells secrete an abundance of paracrine-acting
blasts with reduced CD44 expression upon TGF-␤ stim- growth factors and cytokines that modulate the prolifer-
ulation (31, 39). By this mechanism the FAP⫹CD44⫺ cells ation and motility/invasion of cancer cells. For example,
may be derived from the FAP⫹CD44⫹SMemb⫹ cells. The hepatocyte growth factor secreted by prostate stromal
proliferation of stem-like cells and their transit-amplifying cells can drive proliferation of adjacent epithelial cells
progeny contribute to the generation of reactive stroma. In (46); its production was found induced by estrogens in
view of the several hypotheses about the origin of CAFs, mammary fibroblasts (47). Similarly, Slavin et al (21) re-
our study supports stromal stem-like cells as a source of ported the down-regulation of MMP3 by ER␣ in prostate
prostate CAFs. CAFs, and Yeh et al (48) reported the abrogation of IL-6
In prostate CAFs we found a significantly increased secretion by ER␣ in prostate CAFs. Both MMP3 and IL-6
expression level of GPR30. Moreover, the FAP-positive promote the migration of tumor cells (4, 48, 49) and are
stromal cells, especially the FAP⫹CD44⫹SMemb⫹ sub- also candidate mediators for the observed effects on tumor
type, coexpressed GPR30, which was mainly localized to cell migration.
3034 Jia et al GPR30 Promotes Prostate Stromal Cell Activation Endocrinology, August 2016, 157(8):3023–3035

As we discussed above, GPR30 is up-regulated in pros- Acknowledgments


tate CAFs, and the overexpression of GPR30 in WPMY-1
Address all correspondence and requests for reprints to: Ju
cells induced a CAF-like phenotype to promote PCa cell
Zhang, PhD, or Jiandang Shi, PhD, Department of Biochemistry
proliferation and migration in a paracrine manner. In ad- and Molecular Biology, College of Life Sciences, Bioactive Ma-
dition, a role for GPR30 in prostate epithelium has been terials Key Lab of Ministry of Education, Nankai University,
reported previously. Chan et al (50) reported the activa- Tianjin 300071, China; E-mail: zhangju@nankai.edu.cn (J.Z.);
tion of GPR30 by the nonestrogenic ligand G-1 inhibited or shijd@nankai.edu.cn (J.S.).
the proliferation of PCa cells. Additionally, G-1 effectively This work was supported by the National Natural Science
inhibited the growth of castration-resistant tumors and Foundation of China Grant 81370859 and The 111 Project

Downloaded from https://academic.oup.com/endo/article/157/8/3023/2422352 by biomedicas user on 06 February 2024


Grant B08011.
elicited neutrophil infiltration-associated necrosis in
Disclosure Summary: The authors have nothing to disclose.
xenograft models (51). This seems to disclose an opposing
role of stromal and epithelial GPR30 in PCa progression,
which is similar to the prostatic androgen receptor (AR)
References
that epithelial AR functions as a suppressor and stromal
AR functions as a promoter for PCa cell invasion (52). 1. Clark AK, Taubenberger AV, Taylor RA, et al. A bioengineered
microenvironment to quantitatively measure the tumorigenic prop-
Moreover, G-1 inhibits PCa cell proliferation through the
erties of cancer-associated fibroblasts in human prostate cancer.
sustained activation of ERK1/2 and a c-jun/c-fos-depen- Biomaterials. 2013;34:4777– 4785.
dent up-regulation of p21 (50), whereas in our study 2. Cunha GR, Hayward SW, Wang YZ, Ricke WA. Role of the stromal
microenvironment in carcinogenesis of the prostate. Int J Cancer.
GPR30 suppresses ER␣ expression and was critically
2003;107:1–10.
identified through inhibiting the cAMP/PKA signaling 3. Barron DA, Rowley DR. The reactive stroma microenvironment
pathway in prostate stromal cell lines, although GPR30 and prostate cancer progression. Endocr Relat Cancer. 2012;19:
R187–R204.
overexpression or knockdown had no effect on ERK1/2 4. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006;
phosphorylation in WPMY-1 cells (data not shown). 6:392– 401.
Therefore, the specific downstream signaling of epithelial 5. Ayala G, Tuxhorn JA, Wheeler TM, et al. Reactive stroma as a
predictor of biochemical-free recurrence in prostate cancer. Clin
and stromal GPR30 could be taken into account for the Cancer Res. 2003;9:4792– 4801.
development of PCa therapies targeting GPR30. 6. Tuxhorn JA, Ayala GE, Smith MJ, Smith VC, Dang TD, Rowley DR.
Although our result of the down-regulation of SM dif- Reactive stroma in human prostate cancer: induction of myofibro-
blast phenotype and extracellular matrix remodeling. Clin Cancer
ferentiation makers in prostate CAFs is in agreement with Res. 2002;8:2912–2923.
previous reports (5, 6), no mechanism has been demon- 7. Josson S, Matsuoka Y, Chung LW, Zhau HE, Wang R. Tumor-
strated before. Here we propose two possible mechanisms stroma co-evolution in prostate cancer progression and metastasis.
Semin Cell Dev Biol. 2010;21:26 –32.
according to our results: 1) with PCa development, the 8. Tripathi M, Billet S, Bhowmick NA. Understanding the role of stro-
stromal stem-like cells stimulated via GPR30 up-regula- mal fibroblasts in cancer progression. Cell Adh Migr. 2012;6:231–
235.
tion preferentially propagate over the preexisting SMCs,
9. Micke P, Ostman A. Tumour-stroma interaction: cancer-associated
resulting in the decrease of SM markers in PCa stroma; or fibroblasts as novel targets in anti-cancer therapy? Lung Cancer.
2) GPR30 up-regulation directly down-regulates SM dif- 2004;45(suppl 2):S163–S175.
10. Williams G. Aromatase up-regulation, insulin and raised intracel-
ferentiation marker expression in stromal cells by sup-
lular oestrogens in men, induce adiposity, metabolic syndrome and
pressing ER␣. Therefore, GPR30 and its downstream sig- prostate disease, via aberrant ER-alpha and GPER signalling. Mol
naling pathway may be a crucial event involved in prostate Cell Endocrinol. 2012;351:269 –278.
11. Madar S, Brosh R, Buganim Y, et al. Modulated expression of
CAF formation.
WFDC1 during carcinogenesis and cellular senescence. Carcinogen-
In conclusion, we provide evidence that stromal stem- esis. 2009;30:20 –27.
like cells are a possible source of prostate CAFs, and 12. Karnoub AE, Dash AB, Vo AP, et al. Mesenchymal stem cells within
tumour stroma promote breast cancer metastasis. Nature. 2007;
GPR30 plays a central role in promoting stromal cell ac- 449:557–563.
tivation by inhibiting ER␣ signaling. Further studies will 13. Mishra PJ, Humeniuk R, Medina DJ, et al. Carcinoma-associated
be required to identify the mechanism(s) underlying stro- fibroblast-like differentiation of human mesenchymal stem cells.
Cancer Res. 2008;68:4331– 4339.
mal GPR30 up-regulation and cellular translocalization 14. Zeisberg EM, Potenta S, Xie L, Zeisberg M, Kalluri R. Discovery of
during PCa development. The crucial role of GPR30 in the endothelial to mesenchymal transition as a source for carcinoma-
induction and stimulation of a reactive tumor stroma sug- associated fibroblasts. Cancer Res. 2007;67:10123–10128.
15. Montie JE, Pienta KJ. Review of the role of androgenic hormones in
gests that treatment for PCa by targeting stromal GPR30 the epidemiology of benign prostatic hyperplasia and prostate can-
should be considered in the future. cer. Urology. 1994;43:892– 899.
doi: 10.1210/en.2016-1035 press.endocrine.org/journal/endo 3035

16. Cunha GR, Hayward SW, Wang YZ. Role of stroma in carcino- tradiol-mediated ERK phosphorylation and apoptosis in vascular
genesis of the prostate. Differentiation. 2002;70:473– 485. smooth muscle cells requires GPR 30. Am J Physiol Cell Physiol.
17. Ho S-M, Leung Y-K, Chung I. Estrogens and antiestrogens as eti- 2009;297:C1178 –C1187.
ological factors and therapeutics for prostate cancer. Ann NY Acad 35. Rabkin E, Aikawa M, Stone JR, Fukumoto Y, Libby P, Schoen FJ.
Sci. 2006;1089:177–193. Activated interstitial myofibroblasts express catabolic enzymes and
18. Prossnitz ER, Barton M. Signaling, physiological functions and clin- mediate matrix remodeling in myxomatous heart valves. Circula-
ical relevance of the G protein-coupled estrogen receptor GPER. tion. 2001;104:2525–2532.
Prostaqlandins Other Lipid Mediat. 2009;89:89 –97. 36. Suzuki J, Takayama K, Mitsui F, et al. In situ interleukin-6 tran-
19. Yu L, Shi J, Cheng S, et al. Estrogen promotes prostate cancer cell scription in embryonic nonmuscle myosin heavy chain expressing
migration via paracrine release of ENO1 from stromal cells. Mol immature mesenchyme cells of cardiac myxoma. Cardiovasc Pathol.
Endocrinol. 2012;26:1521–1530. 2000;9:33–37.
20. Yu L, Wang CY, Shi J, et al. Estrogens promote invasion of prostate 37. Shao R, Shi J, Liu H, et al. Epithelial-to-mesenchymal transition and
estrogen receptor ␣ mediated epithelial dedifferentiation mark the

Downloaded from https://academic.oup.com/endo/article/157/8/3023/2422352 by biomedicas user on 06 February 2024


cancer cells in a paracrine manner through up-regulation of matrix
metalloproteinase 2 in prostatic stromal cells. Endocrinology. 2011; development of benign prostatic hyperplasia. Prostate. 2014;74:
152:773–781. 970 –982.
21. Slavin S, Yeh CR, Da J, et al. Estrogen receptor ␣ in cancer-associ- 38. Verfaillie CM. Adult stem cells: assessing the case for pluripotency.
ated fibroblasts suppresses prostate cancer invasion via modulation Trends Cell Biol. 2002;12:502–508.
of thrombospondin 2 and matrix metalloproteinase 3. Carcinogen- 39. Patrawala L, Calhoun T, Schneider-Broussard R, et al. Highly pu-
esis. 2014;35:1301–1309. rified CD44⫹ prostate cancer cells from xenograft human tumors
22. Pupo M, Vivacqua A, Perrotta I, et al. The nuclear localization signal are enriched in tumorigenic and metastatic progenitor cells. Onco-
is required for nuclear GPER translocation and function in breast gene. 2006;25:1696 –1708.
cancer-associated fibroblasts (CAFs). Mol Cell Endocrinol. 2013; 40. Funakoshi T, Yanai A, Shinoda K, Kawano MM, Mizukami Y. G
376:23–32. protein-coupled receptor 30 is an estrogen receptor in the plasma
23. De Francesco EM, Pellegrino M, Santolla MF, et al. GPER mediates membrane. Biochem Biophys Res Commun. 2006;346:904 –910.
activation of HIF1/VEGF signaling by estrogens. Cancer Res. 2014; 41. Thomas P, Pang Y, Filardo EJ, Dong J. Identity of an estrogen mem-
74:4053– 4064. brane receptor coupled to a G protein in human breast cancer cells.
Endocrinology. 2005;146:624 – 632.
24. Zhang Z, Wang L, Mei M, et al. Both nongenomic and genomic
42. Revankar CM, Mitchell HD, Field AS, et al. Synthetic estrogen de-
effects are involved in estradiol’s enhancing the phenotype of
rivatives demonstrate the functionality of intracellular GPR30. ACS
smooth muscle cells in cultured prostate stromal cells. Prostate.
Chem Biol. 2007;2:536 –544.
2010;70:317–332.
43. Madeo A, Maggiolini M. Nuclear alternate estrogen receptor
25. Li F, Yu X, Szynkarski CK, et al. Activation of GPER induces dif-
GPR30 mediates 17␤-estradiol-induced gene expression and migra-
ferentiation and inhibition of coronary artery smooth muscle cell
tion in breast cancer-associated fibroblasts. Cancer Res. 2010;70:
proliferation. PloS One. 2013;8:e64771.
6036 – 6046.
26. Cronauer MV, Eder IE, Hittmair A, et al. A reliable system for the
44. Ariazi EA, Brailoiu E, Yerrum S, et al. The G protein-coupled re-
culture of human prostatic cells. In Vitro Cell Dev Biol Anim. 1997;
ceptor GPR30 inhibits proliferation of estrogen receptor-positive
33:742–744.
breast cancer cells. Cancer Res. 2010;70:1184 –1194.
27. Corvin S, Boesch S, Maneschg C, Radmayr C, Bartsch G, Klocker
45. Gao F, Ma X, Ostmann AB, Das SK. GPR30 activation opposes
H. Effect of heat exposure on viability and contractility of cultured
estrogen-dependent uterine growth via inhibition of stromal
prostatic stromal cells. Eur Urol. 2000;37:499 –504. ERK1/2 and estrogen receptor ␣ (ER␣) phosphorylation signals.
28. Webber MM, Trakul N, Thraves PS, et al. A human prostatic stro- Endocrinology. 2011;152:1434 –1447.
mal myofibroblast cell line WPMY-1: a model for stromal-epithelial 46. Lim A, Shin K, Zhao C, Kawano S, Beachy PA. Spatially restricted
interactions in prostatic neoplasia. Carcinogenesis. 1999;20:1185– Hedgehog signalling regulates HGF-induced branching of the adult
1192. prostate. Nat Cell Biol. 2014;16:1135–1145.
29. Zhou Y, Xiao XQ, Chen LF, et al. Proliferation and phenotypic 47. Zhang HZ, Bennett JM, Smith KT, Sunil N, Haslam SZ. Estrogen
changes of stromal cells in response to varying estrogen/androgen mediates mammary epithelial cell proliferation in serum-free culture
levels in castrated rats. Asian J Androl. 2009;11:451– 459. indirectly via mammary stroma-derived hepatocyte growth factor.
30. Brennen WN, Rosen DM, Wang H, Isaacs JT, Denmeade SR. Tar- Endocrinology. 2002;143:3427–3434.
geting carcinoma-associated fibroblasts within the tumor stroma 48. Yeh CR, Slavin S, Da J, et al. Estrogen receptor ␣ in cancer associated
with a fibroblast activation protein-activated prodrug. J Natl Can- fibroblasts suppresses prostate cancer invasion via reducing CCL5,
cer Inst. 2012;104:1320 –1334. IL6 and macrophage infiltration in the tumor microenvironment.
31. Kim W, Barron DA, San Martin R, et al. RUNX1 is essential for Mol Cancer. 2016;15:7.
mesenchymal stem cell proliferation and myofibroblast differenti- 49. DiNatale BC, Schroeder JC, Perdew GH. Ah receptor antagonism
ation. Proc Natl Acad Sci USA. 2014;111:16389 –16394. inhibits constitutive and cytokine inducible IL6 production in head
32. Tsai H-W, Katzenellenbogen JA, Katzenellenbogen BS, Shupnik and neck tumor cell lines. Mol Carcinog. 2011;50:173–183.
MA. Protein kinase A activation of estrogen receptor ␣ transcription 50. Chan QKY, Lam HM, Ng CF, et al. Activation of GPR30 inhibits
does not require proteasome activity and protects the receptor from the growth of prostate cancer cells through sustained activation of
ligand-mediated degradation. Endocrinology. 2004;145:2730 – Erk1/2, c-jun/c-fos-dependent upregulation of p21, and induction of
2738. G2 cell-cycle arrest. Cell Death Differ. 2010;17:1511–1523.
33. Broselid S, Berg KA, Chavera TA, et al. G protein-coupled receptor 51. Lam HM, Ouyang B, Chen J, et al. Targeting GPR30 with G-1: a
30 (GPR30) forms a plasma membrane complex with membrane- new therapeutic target for castration-resistant prostate cancer. En-
associated guanylate kinases (MAGUKs) and protein kinase A-an- docr Relat Cancer. 2014;21:903–914.
choring protein 5 (AKAP5) that constitutively inhibits cAMP pro- 52. Niu Y, Altuwaijri S, Lai KP, et al. Androgen receptor is a tumor
duction. J Biol Chem. 2014;289:22117–22127. suppressor and proliferator in prostate cancer. Proc Natl Acad Sci
34. Ding Q, Gros R, Limbird LE, Chorazyczewski J, Feldman RD. Es- USA. 2008;105:12182–12187.

You might also like