You are on page 1of 13

0013-7227/06/$15.

00/0 Endocrinology 147(9):4179 – 4191


Printed in U.S.A. Copyright © 2006 by The Endocrine Society
doi: 10.1210/en.2006-0168

Pigment Epithelium-Derived Factor Is Estrogen


Sensitive and Inhibits the Growth of Human Ovarian
Cancer and Ovarian Surface Epithelial Cells
Lydia W. T. Cheung, Simon C. L. Au, Annie N. Y. Cheung, Hextan Y. S. Ngan, Joyce Tombran-Tink,
Nelly Auersperg, and Alice S. T. Wong
Departments of Zoology (L.W.T.C., A.S.T.W.), Pathology (A.N.Y.C.), and Obstetrics and Gynecology (H.Y.S.N.), University of
Hong Kong, Hong Kong; Department of Physiology (S.C.L.A.), Chinese University of Hong Kong, Hong Kong; Division of
Pharmaceutical Sciences (J.T.-T.), University of Missouri, Kansas City, Missouri 64110; Department of Ophthalmology
(J.T.-T.), Yale University School of Medicine, New Haven, Connecticut 06520; and Department of Obstetrics and Gynecology
(N.A.), University of British Columbia, Vancouver, British Columbia, Canada V6H 3V5

Epithelial ovarian carcinoma is the most lethal gynecological tor of PEDF in human OSE. Treatment of the cultured cells
cancer. However, little is known about the molecular mech- with 17␤-estradiol (E2) inhibited the expression of PEDF pro-
anisms underlying the disease development and progression. tein and mRNA in a dose- and time-dependent manner, which
In this study, we found that the expression of pigment epi- could be reversed by the specific estrogen receptor antago-
thelium-derived factor (PEDF) was greatly reduced in ovar- nist, ICI 182,780, indicating that the regulation was estrogen
ian tumors and in ovarian cancer cell lines when compared receptor-mediated. We further showed that this down-regu-
with their normal precursor, ovarian surface epithelium lation of PEDF gene transcription was a direct, primary effect
(OSE). In addition, we showed that exogenous PEDF inhibited of E2. E2 promoted OSE and ovarian cancer cell growth,
the growth of cultured human OSE as well as ovarian cancer whereas simultaneous treatment with E2 and PEDF abro-
cell lines, whereas targeted inhibition of endogenous PEDF gated the estrogenic growth stimulation of these cells. This
using small interfering RNA or neutralizing PEDF antibody study is the first to demonstrate a role of PEDF in OSE biology
promoted the growth of these cells, confirming that the and ovarian cancer and suggests that the loss of PEDF may e of
growth-inhibitory effect was PEDF specific. We also report for relevance in carcinogenesis. (Endocrinology 147: 4179 – 4191,
the first time that estrogen is an important upstream regula- 2006)

O VARIAN CANCER IS the most lethal gynecological


cancer among women in Western countries, and the
vast majority of human ovarian carcinomas are derived from
nal). PEDF action appears to be cell specific. To neurons,
PEDF is neurotrophic, promoting cell survival and differen-
tiation (6 – 8). However, when endothelial cells are exposed
the ovarian surface epithelium (OSE) (1). The OSE is a simple to PEDF, their cell migration and proliferation are inhibited
squamous to cuboidal mesothelial cells that overlies the (9), and they undergo apoptosis (10, 11). Although the spe-
ovary. During each reproductive cycle, OSE on the preovu- cific functions of PEDF in cancer are not known, there is some
latory follicle undergoes apoptosis at the time of ovulation evidence that PEDF could have inhibitory effects on tumor
and then proliferates rapidly to repair the ruptured follicle and growth. First, the levels of PEDF are highest in normal tissue
reconstitutes an intact mesothelium (2). Repeated ovulation is (prostate, liver, and melanocyte), and they decrease during
believed to contribute to OSE neoplastic transformation (3), carcinogenesis (12–15). Second, PEDF has been reported to
indicating that the process of healing damaged OSE may con- have a direct antitumor effect on melanoma (13), osteosar-
tribute to the disease. Therefore, there is great interest in un- coma (16), endometrial (17), and prostate (12, 14) cancer cells
derstanding the cell survival and death signals in these cells. by inhibiting angiogenesis, cell growth, and migration. Fi-
Pigment epithelium-derived factor (PEDF) is a secreted nally, in PEDF-knockout mice, vascular density is markedly
50-kDa glycoprotein first described as an ocular neurotro- increased, and epithelial cell hyperplasia is evident in the
phic protein synthesized by fetal retinal pigment epithelial prostate and exocrine pancreas (12).
cells (4, 5). Subsequent studies demonstrate that it is widely PEDF appears to play an important role in determining the
expressed in most normal tissues (neuronal and nonneuro- tissue growth of many organs, and this may include the
ovary, where a high basal level of PEDF mRNA expression
First Published Online June 15, 2006
Abbreviations: ActD, Actinomycin D; CHX, cycloheximide; E2, 17␤- is found (18). Interestingly, allelic loss in the 17p13.3 region,
estradiol; ER, estrogen receptor; FBS, fetal bovine serum; ICI, ICI 182,780; where the PEDF gene is located, is frequently encountered in
MTT, 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide; early-stage ovarian carcinoma (19), implicating PEDF in this
OSE, ovarian surface epithelium; PEDF, pigment epithelium-derived fac- region as a potential tumor suppressor gene regulating the
tor; rPEDF, recombinant PEDF; siRNA, small interfering RNA; TUNEL,
terminal deoxynucleotidyl transferase-mediated nick-end labeling. behavior of ovarian cancers. As yet, however, no study has
Endocrinology is published monthly by The Endocrine Society (http://
examined the regulation of PEDF expression in normal ova-
www.endo-society.org), the foremost professional society serving the ries or ovarian tumors.
endocrine community. The mechanism underlying the regulation of PEDF is

4179

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 October 2015. at 04:17 For personal use only. No other uses without permission. . All rights reserved.
4180 Endocrinology, September 2006, 147(9):4179 – 4191 Cheung et al. • Down-Regulation and Antiproliferative Role of PEDF

largely unknown. Several lines of evidence prompted us to Treatments


investigate that one of the mechanisms regulating PEDF To study the regulation of PEDF expression by E2, 5 ⫻ 105 cells were
expression in OSE or ovarian cancer cells might involve plated onto six-well dishes. After 48 h preincubation in phenol red-free
estrogen. 1) Estrogen is the major female hormone with mi- medium supplemented with 2.5% charcoal/dextran-treated FBS (for
togenic activities on a variety of tissues including the ovary. removal of endogenous steroids in the serum), the cells were treated
with 0.01–100 nm E2 (Sigma) in fresh medium for 24 h. To test the
Normal OSE and most ovarian carcinomas have specific specificity of E2 effect, some cells were exposed to a blocker of classical
receptors for estrogen (20 –22). A role of estrogen in ovarian ER, ICI 182,780 (ICI; Tocris Cookson Ltd., Bristol, UK), 30 min before and
carcinogenesis has been suggested; however, the epidemio- during treatment with E2. To study the stability of PEDF mRNA, cells
logical data are inconsistent. Some past studies have linked were treated with 100 nm E2 for 3 h, and cells without pretreatment
the use of postmenopausal estrogen replacement therapy to served as controls. Actinomycin D (ActD; 4 ␮g/ml) (Calbiochem, San
Diego, CA) was then added to the cultures, and total RNA was prepared
an increased risk, whereas other reports detected an un- at the times indicated for up to 8 h. To inhibit protein synthesis, cells were
changed or reduced risk of developing the cancer. Estrogen incubated with 4 ␮g/ml cycloheximide (CHX) (Sigma) for 1 h followed
taken as oral contraceptives during premenopausal years by the addition of 100 nm E2 for 24 h. Control cultures were treated with
seems to offer protection (23–25). Although further investi- the vehicle (i.e. ethanol) alone. To examine the effect of E2 and recom-
binant PEDF (rPEDF) (Upstate Biotechnology Inc., Lake Placid, NY) on
gations are required to resolve how exogenous estrogens cell growth, cells were plated for 3-(4,5-dimethyl-thiazol-2-yl)-2,5-di-
affect cancer risk, numerous studies have demonstrated the phenyltetrazolium bromide (MTT) and apoptosis assays. Twenty-four
mitogenic action of estrogen in ovarian cancer. For instance, hours later, the cultures were treated with E2 in the presence or absence
experimental ovarian tumor could be induced by estrogens of rPEDF for 5 d, with fresh hormone added every other day.
(26, 27), and estrogen stimulated the growth of several es-
trogen receptor (ER)-positive ovarian carcinoma cell lines in Immunohistochemistry
vitro (21, 28, 29). However, little is known about the estrogen- Formalin-fixed, paraffin-embedded tissues that include three normal
regulated genes in ovarian tissue. 2) It is of interest to note that ovarian tissues (ages, 23– 47 yr; mean age, 31.3 yr), 13 benign tumors
a putative estrogen-response element has been identified in the (ages, 20 – 67 yr; mean age, 36.8 yr), 12 borderline tumors (ages, 24 –57
5⬘-flanking region of the PEDF gene, further implying that yr; mean age, 35.9 yr), and 24 ovarian carcinomas (ages, 30 – 69 yr; mean
age, 49.6 yr) (10 serous, six endometrioid, four mucinous, and four clear
PEDF may be linked to the regulation of hormone-dependent cell) were obtained with Internal Review Board approval from the ar-
growth in the ovary and that in the ovarian cancer (18). chives of the Department of Pathology of Queen Mary Hospital, the
In this study, we aimed to elucidate the role of PEDF in the University of Hong Kong. Patients were identified anonymous reference
tumorigenesis of ovarian cancer and in particular to inves- numbers. Five-micrometer sections were deparaffinized and rehydrated
in a graded series of ethanol following standard protocol. Sections were
tigate the possible regulation of PEDF by 17␤-estradiol (E2) treated with 3% H2O2 for 10 min to eliminate endogenous peroxidase
in ovarian epithelial cells. Our findings indicate that PEDF activity and then incubated with an anti-PEDF antibody (1:200) (Bio-
plays an important role in regulating the normal OSE cell products, Middletown, MD) for 30 min at room temperature before
function, and its decrease or loss in ovarian tumors could exposure to biotinylated secondary antibodies for 10 min. Antigen-
contribute to tumor development and progression. In addi- antibody complexes were visualized using the substrate-chromogen
mixture (Zymed Laboratories Inc., San Francisco, CA) and counter-
tion, we demonstrate that estrogen-mediated down-regula- stained with hematoxylin. Omission or substitution of the primary
tion of PEDF is a novel way of reducing PEDF levels in OSE antibody with preimmune serum was used as a negative control. Im-
and mediates the effect of E2 on proliferation of these cells. munoreactivity was assessed by the intensity and percentage of positive
staining. Staining intensity was scored as 0 (negative), 1 (faint), 2 (mod-
erate), and 3 (strong). A case was considered to be negative if there was
Materials and Methods no staining or weak staining involving less than 10% cells.
Cell culture and tissue samples
Silencing of PEDF by small interfering RNA (siRNA)
Two immortalized nontumorigenic human OSE cell lines, IOSE-29
and IOSE-397 (30), and the human ovarian epithelial carcinoma cell lines The Smart-pool siRNA for silencing PEDF (catalog no. M-010153-00)
Caov-3 and SKOV-3 were cultured at 37 C in Medium 199:105 (Sigma, was purchased from Dharmacon (Lafayette, CO). Transfection of the
St. Louis, MO) supplemented with 5% fetal bovine serum (FBS) (Hyclone PEDF-specific siRNA (20 nm) was performed using SilentFect (Bio-Rad,
Laboratories Ltd., Logan, UT), 100 U/ml penicillin, and 100 ␮g/ml Carlsbad, CA) per the manufacturer’s instructions. As a nonspecific
streptomycin (Invitrogen, San Diego, CA) in a humidified atmosphere siRNA control, scrambled siRNA (Dharmacon) was used. To determine
of 5% CO2 in air. All cells were passaged using 0.06% trypsin/0.01% the efficiency of siRNA transfection, RT-PCR and Western blot analyses
EDTA (Invitrogen). All experiments were performed using both OSE were performed to detect PEDF expression level at 5 d posttransfection.
lines and repeated two to three times with each experiment yielding To examine the role of PEDF in cell proliferation and viability, trans-
essentially similar results. The results from a representative cell line fected cells were plated for MTT and apoptosis assays 24 h after siRNA
(IOSE-397) are shown in Figs. 3 and 5–7. transfection.
Samples of primary ovarian carcinomas obtained after surgery were
collected from patients at Queen Mary Hospital, Hong Kong. The his- Semiquantitative RT-PCR
tology of all samples was verified by surgical pathologists and found to
contain more than 70% tumor cells. There were 11 serous carcinomas, Total RNA was isolated from cultured cells using the TRIzol reagent
two endometrioid carcinomas, and one mixed endometrioid and serous (Invitrogen) according to the manufacturer’s procedure. First strand
carcinoma (ages, 31– 62 yr; mean age, 44.9 yr). Normal human OSE cells cDNA synthesis was performed from 2.5 ␮g total RNA using Super-
(ages, 34 – 47 yr; mean age, 40.5 yr) were derived from surface scrapings Script Reverse Transcriptase (Invitrogen). cDNA was amplified in a
of normal ovaries from women with nonmalignant gynecological dis- 15-␮l PCR mixture containing 1 mm dNTPs, 1⫻ PCR buffer, 2.5 mm
eases. OSE-2 and OSE-3 were cultured in Medium 199:105 with genta- MgCl2, and 1 U DNA Taq polymerase (Promega, Madison, WI) with 5
micin (50 ␮g/ml) and 15% FBS (1). RNA was extracted from cells har- pmol each primer for human PEDF (sense, 5⬘-CATTCACCGGGCTCTC-
vested in passage 2. The use of these tissues was approved by the TAC-3⬘; antisense: 5⬘-GGCAGCTGGGCAATCTTGCA-3⬘). The condi-
Institutional Ethical Review Board for Research on the use of human tion in the logarithmic phase of PCR amplification was as follows: 5 min
subjects. Informed consent was obtained from all patients. initial denaturation at 94 C, 1 min denaturation at 94 C, 35 sec annealing

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 October 2015. at 04:17 For personal use only. No other uses without permission. . All rights reserved.
Cheung et al. • Down-Regulation and Antiproliferative Role of PEDF Endocrinology, September 2006, 147(9):4179 – 4191 4181

at 67 C, and 1.5 min extension at 72 C for 30 cycles. The number of Hoechst staining
amplification cycles during which PCR product formation was limited
by template concentration was determined in pilot experiments. PCR To confirm the findings of TUNEL assay, apoptotic cells were also
products were analyzed by agarose gel electrophoresis. ␤-Actin was detected by Hoechst staining. Briefly, cells were trypsinized and fixed
used as the internal control (sense, 5⬘-TCACCGAGGCCCCTCTGAAC- with Carnoy’s fixative (methanol/acetic acid, 1:3) for 10 min at room
CCTA-3⬘; antisense, 5⬘-GGCAGTAATCTCCTTCTGCATCCT-3⬘). The temperature. After two PBS washes, DNA-specific fluorochrome
reproducibility of the quantitative measurements was evaluated by Hoechst 33342 (Sigma) was added at a final concentration of 5 ␮g/ml,
three independent cDNA syntheses and PCR amplification from each and the suspension was incubated at room temperature for 30 min in the
preparation of RNA. Densitometric analysis was performed using Scion dark. Cell suspensions were mounted on slide glasses and subjected to
Image software (Scion Corp., Frederick, MD), and the relative PEDF immunofluorescence microscopic examination. Apoptotic cells were iden-
mRNA expression levels were determined as the ratio of the signal tified by chromatin condensation and nuclei fragmentation. The percent-
intensity of PEDF to that of ␤-actin. ages of apoptotic cells were calculated from the ratio of apoptotic cells to
total cells counted. At minimum, 500 cells were counted in five different
fields, and assays were performed in duplicate at least three times.
Real-time PCR
Real-time PCR was performed using the iCycler iQ Real-Time de- Promoter constructs and luciferase assay
tection system and the IQ SYBR Green Supermix (Bio-Rad). PCR primers
for PEDF were as described above. ␤-Actin was analyzed in the same run The PEDF promoter region between positions ⫺864 and ⫹63 was
as internal control. Fluorescent measurements were recorded during cloned into promoterless luciferase reporter vector pGL3-Basic as pre-
each annealing step. The PCR quality and specificity were verified by viously described (32). Cells were transiently transfected with 1 ␮g of the
melting curve analysis and gel electrophoresis. For quantitative com- construct using Lipofectamine 2000 (Invitrogen) as directed by the man-
parison, the relative abundance of the PEDF mRNA was determined by ufacturer. After 6 h, the cells were incubated with or without E2 (100 nm)
dividing the threshold of each sample by the threshold of the internal for an additional 24 h before being harvested for luciferase activity
control ␤-actin. These experiments were carried out in duplicate and measurement. The pSV-␤-galactosidase plasmid was cotransfected as
independently repeated three times. internal control. Luciferase units were calculated as luciferase activity/
␤-galactosidase activity and are presented as the mean ⫾ sd of three
individual experiments with triplication. The fold change was calculated
Western blot analysis by comparison with the promoterless luciferase vector (pGL3-Basic).
Cellular extracts were prepared using RIPA [1% Triton X-100, 50 mm
Tris-HCl (pH 7.4), 0.1% SDS, 150 mm NaCl, and 5 mm EDTA, supple-
Statistical analysis
mented with protease inhibitors containing 1 mm phenylmethylsulfonyl
fluoride, 1 ␮g/ml aprotinin, 1 ␮g/ml leupeptin, and 1 ␮g/ml pepstatin Every experiment was repeated at least twice in either duplicate or
A]. Conditioned media were collected and clarified by centrifugation. triplicate with different cell preparations to ensure consistency of the find-
Total protein content in each sample was determined using the Bradford ings. Statistical analysis was carried out using Fisher’s exact test and
assay (Bio-Rad). Forty micrograms extracted proteins from each cell ANOVA followed by Tukey’s post hoc test (GraphPad Software, San Diego,
culture or 100 ␮g total proteins from the conditioned media was sep- CA) where applicable. P ⬍ 0.05 was considered statistically significant.
arated by 7.5% SDS-PAGE and then transferred to nitrocellulose mem-
brane. Membranes were blocked using 5% nonfat dry milk in PBS
containing 0.05% Tween 20 and incubated overnight at 4 C with rabbit Results
polyclonal human PEDF antibody (1:1000) (Bioproducts) or rabbit poly- Loss of PEDF in human ovarian cancer
clonal ␤-actin antibody (1:1000) (Sigma) as a loading control. For con-
ditioned media, parallel gels were stained with Coomassie blue to con- To understand the significance of PEDF in ovarian carci-
firm equal loading. The immunocomplex was detected with horseradish nogenesis, we first evaluated the expression of the PEDF
peroxidase-conjugated antirabbit IgG (Bio-Rad) and visualized using an
enhanced chemiluminescence detection system (Amersham Biosciences, protein in normal (n ⫽ 3), benign (n ⫽ 13), borderline (n ⫽
Little Chalfont, UK). 12), and malignant (n ⫽ 24) ovarian tissues by immunohis-
tochemistry. Our analyses revealed strong PEDF immuno-
MTT assay staining in normal OSE, cortical stroma, and endothelium
(Fig. 1A, a; and Table 1). In benign serous and mucinous
Cell viability was assessed colorimetrically by the mitochondria-de-
pendent reduction of MTT (Sigma) to formazan (31). Cells were seeded
cystadenomas, a comparable strong epithelial expression
at 2000 per well in 96-well plates. After a 5-d incubation period, 10 ␮l was detected (Fig. 1A, b; and Table 1). Of these, six showed
MTT was added to each well, and plates were incubated at 37 C for 4 h. strong expression of PEDF (46.1%), and five showed mod-
Finally, culture medium was aspirated, and 100 ␮l DMSO was added to erate expression (38.5%). In contrast, tumors of borderline
extract the dye. Conversion of MTT to formazan by metabolically viable malignancy exhibited no or only weak expression level of
cells was monitored by the absorbance of each well at 570 nm with 630
nm as the reference wavelength. Relative cell viability was expressed as PEDF (Fig. 1A, c), and loss of PEDF was confirmed in 22 of
the fold change over control cultures. Assays were performed in trip- 24 tumors (91.7%) (Fig. 1A, d–f; and Table 1). There was no
licates, and data points represent the mean values ⫾ sd from three significant correlation between the PEDF expression and the
independent experiments. clinical data of the tumor histological type (P ⫽ 0.74). Omis-
sion or substitution of the primary antibody with preimmune
Terminal deoxynucleotidyl transferase-mediated nick-end serum did not produce any staining (data not shown). We
labeling (TUNEL) staining also performed real-time PCR to verify our immunohisto-
TUNEL assay was performed using an in situ cell death detection kit chemistry results with ovarian cancer tissue specimens (OC-
(Roche Biochemical, Indianapolis, IN) per the manufacturer’s instruc- 1–14) and normal ovarian epithelium scrapings (OSE-1) or
tions. This system end-labels the fragmented DNA of apoptotic cells. culture of OSE scrapings (OSE-2 and -3). As shown in Fig. 1B,
Omission of the enzyme in the TUNEL reaction was used as a negative PEDF mRNA expression was significantly higher in normal
control, and cells treated with DNAse I were used as a positive control.
The number of TUNEL-positive cells was counted in five different fields OSE (n ⫽ 3) compared with ovarian cancer (n ⫽ 14). No sig-
under a light microscope at ⫻40 magnification, and representative fields nificant difference was seen in uncultured and cultured OSE
were photographed. brushings, in which all cases had relatively high levels of PEDF

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 October 2015. at 04:17 For personal use only. No other uses without permission. . All rights reserved.
4182 Endocrinology, September 2006, 147(9):4179 – 4191 Cheung et al. • Down-Regulation and Antiproliferative Role of PEDF

FIG. 1. Immunohistochemical staining


of PEDF in human ovarian tissues. Rep-
resentative tissue sections are shown.
A, Paraffin-embedded tissue sections
of: a, human normal ovary (surface ep-
ithelium indicated by arrows); b, benign
cystadenoma; c, borderline tumor; and
d to f, ovarian adenocarcinomas were
immunostained for PEDF. Sections
were counterstained with hematoxylin
to reveal the nuclei. All sections are
shown at ⫻400 magnification. B, PEDF
mRNA expression was assessed in
three normal OSE (OSE-1–3) and 14
human ovarian cancer cells (OC-1–14)
by real-time PCR with specific primers,
as described in Materials and Methods.
␤-Actin was used as internal control. In
all cases, fold change in PEDF expres-
sion was compared with the ovarian
cancer cell line Caov-3, which was given
an arbitrary value of 1.

expression. Together, these results suggest that PEDF protein with Coomassie blue (Fig. 2C). Consistently, OSE cells se-
diminishes with ovarian neoplastic progression. creted the most PEDF, and ovarian cancer cells secreted
Corroborating the observations in vivo, PEDF expression much less to no detectable PEDF. The presence of a specific
was further confirmed in established ovarian cell lines; the 50-kDa band indicated that PEDF was appropriately syn-
immortalized normal OSE cell lines were found to express thesized and processed.
PEDF at high levels by real-time PCR (Fig. 2A) and Western
blot analyses (Fig. 2B). In contrast, the levels were minimal
Suppression of PEDF leads to increased cell proliferation
to absent in ovarian cancer cell lines Caov-3 and SKOV-3,
and viability
suggesting that the decreased PEDF protein expression may
result from decreased mRNA synthesis. Media conditioned To study the physiological functions of PEDF, we per-
by these cells were tested for PEDF expression by Western formed the MTT cell viability and proliferation assay. As
blot. Equal loading was confirmed by parallel gels stained shown, rPEDF induced a significant decrease in cell growth

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 October 2015. at 04:17 For personal use only. No other uses without permission. . All rights reserved.
Cheung et al. • Down-Regulation and Antiproliferative Role of PEDF Endocrinology, September 2006, 147(9):4179 – 4191 4183

TABLE 1. Summary of immunoreactivity of PEDF in the ovary


sections

Staining intensity scorea


n
0 (%) 1 (%) 2 (%) 3 (%)
Normal ovary 3 3 (100)
Benign tumors 13 2 (15.4) 5 (38.5) 6 (46.1)
Borderline tumors 12 6 (50) 5 (41.7) 1 (8.3)
Ovarian carcinoma
Serous 10 9 (90) 1 (10)
Endometrioid 6 5 (83.3) 1 (16.7)
Mucinous 4 4 (100)
Clear cell 4 4 (100)
Total 24 22 (91.7) 2 (8.3)
a
0, No staining; 1, weak staining; 2, moderate staining; 3, strong
staining.

when compared with untreated controls (Fig. 3A) (P ⬍ 0.05).


The growth-inhibitory effect of rPEDF was dose dependent
because the effect (decreased by 42.2, 61.2, and 78.9% re-
spectively) increased with concentration of treatment (1, 10,
and 100 nm) (Fig. 3A). The antiproliferative effect of rPEDF
was blocked by the addition of antibodies specific to PEDF
(P ⬍ 0.05), confirming the effect of PEDF was specific (Fig.
3A). The antibody by itself had no effect on cell number (Fig.
3A). The ability of rPEDF to decrease cell number could result
from either decreased cell cycle progression or increased
apoptosis. Our analyses did not detect a significant change
in cell cycle progression as assessed by propidium iodide
staining (data not shown); however, exposure to rPEDF re-
sulted in a concomitant increase of apoptosis with 55.7% of
the cells being TUNEL positive (at 100 nm rPEDF) when
compared with 16.8% in untreated controls (Fig. 3B). The
apoptotic activity was greatly reduced by the addition of
PEDF antibodies. The ability of rPEDF to induce apoptosis
was examined also by counting Hoeschst-stained cells with
fragmented DNA. Similar to the TUNEL staining experi-
ments, a significant increase in rPEDF-induced apoptosis
was observed (Fig. 3B).
RNA interference assay was conducted to further verify
the role of endogenous PEDF in the regulation of cell pro-
liferation and viability. siRNA oligos that specifically target
human PEDF were transiently transfected into OSE cells. As
revealed by real-time PCR and Western blot analyses (Fig.
3C), transfection of PEDF siRNA efficiently repressed PEDF
expression by over 80% at both mRNA and protein levels,
whereas nonspecific siRNA had no effect. Importantly, in-
hibition of PEDF expression by RNA interference resulted in
a significant increase (98.6% increase) in cell proliferation
(Fig. 3D). In addition, PEDF siRNA suppressed apoptosis FIG. 2. PEDF expression in ovarian cells. Expression of PEDF in
(Fig. 3E). No inhibition was observed for nonspecific siRNA immortalized normal OSE and ovarian cancer cell lines was detected
(Fig. 3E). by: A, real-time PCR using primers specific for PEDF and ␤-actin; B,
To examine whether PEDF might also target tumor epi- cell lysates (40 ␮g); and C, conditioned media (100 ␮g) were analyzed
thelial cells, we treated Caov-3 and SKOV-3 ovarian cancer by Western blot for the presence of PEDF proteins. Immunoblotting
for ␤-actin and staining of proteins by Coomassie blue were included
cells with rPEDF proteins in vitro. As shown in Fig. 4A, as loading controls for B and C, respectively. The signal intensity was
rPEDF at 100 nm substantially decreased cell proliferation in determined by densitometry and expressed as the ratio of PEDF
both cancer cell lines (P ⬍ 0.05) when compared with results relative to loading control for each sample (lower panels of B and C).
obtained with untreated controls, and this response was Experiments were repeated three times, and data are shown as
mean ⫾ SD. **, Statistically significant difference between mean
blocked by treatment with an antibody against PEDF (Fig. PEDF expression levels in OSE cells and those observed in ovarian
4A). We also observed a 3.5-fold increase in the rate of ap- cancer cells with P ⬍ 0.005. The fold change in PEDF expression was
optosis in rPEDF-treated cells, with two apoptosis assays compared with the ovarian cancer cell line Caov-3, which was given
confirming the same finding (P ⬍ 0.005) (Fig. 4B). Consistent an arbitrary value of 1.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 October 2015. at 04:17 For personal use only. No other uses without permission. . All rights reserved.
4184 Endocrinology, September 2006, 147(9):4179 – 4191 Cheung et al. • Down-Regulation and Antiproliferative Role of PEDF

FIG. 3. Effect of PEDF on cell proliferation and apoptosis of OSE cells. A, OSE cells were treated with increasing concentrations of rPEDF,
rPEDF ⫹ anti-PEDF antibody (anti-PEDF), or anti-PEDF alone for 5 d. Cell growth was assessed by MTT assay as described in Materials and
Methods. The absorbance of wells not exposed to treatments was arbitrarily set as 1, and cell growth after treatment was expressed as the fold
changes compared with the control. B, Samples were stained for TUNEL using immunofluorescent labels according to the manufacturer’s
protocol and Hoechst stains, then photographed (left) and counted to determine the percentage of apoptotic cells (right). C, OSE cells were
transfected with PEDF siRNA or a nonspecific (NS) siRNA for 5 d. siRNA-mediated depletion of PEDF was analyzed by: a, real-time PCR; and
b, Western blot analysis. D, In parallel experiments, cells were collected for MTT assay. E, Apoptosis was detected by TUNEL and Hoechst
staining. The bar graphs summarize the percentage of apoptotic cells counted in five fields from three experiments. Experiments were repeated
three times, and data are shown as mean ⫾ SD. *, P ⬍ 0.05; or **, P ⬍ 0.005 compared with untreated controls or as indicated.

with these results, siRNA-mediated down-regulation of served for nonspecific siRNA (Figs. 4, C–E). These findings
PEDF increased proliferation and decreased apoptosis of indicate a functional role for PEDF in OSE and cancer cell
Caov-3 and SKOV-3 cells (Fig. 4, C–E). No effects were ob- growth and survival.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 October 2015. at 04:17 For personal use only. No other uses without permission. . All rights reserved.
Cheung et al. • Down-Regulation and Antiproliferative Role of PEDF Endocrinology, September 2006, 147(9):4179 – 4191 4185

FIG. 4. rPEDF induces apoptotic cell death in ovarian cancer cells. Caov-3 and SKOV-3 cells were cultured and treated with 100 nM rPEDF
and/or antihuman PEDF (anti-PEDF) for 5 d. A, Cell proliferation was assessed by MTT assay as described and expressed as relative fold changes
compared with control. B, In parallel experiments, samples were harvested for TUNEL assay and Hoechst staining, and the bar graphs
summarize the percentage of apoptotic cells counted in five fields from three experiments. C, Cells were transfected with PEDF siRNA or
nonspecific (NS) siRNA. Real-time PCR analysis and Western blotting were performed 5 d after transient gene transfer. In parallel experiments,
cells were collected for MTT assay (D) and TUNEL assay and Hoechst staining (E). Experiments were repeated three times, and data are shown
as mean ⫾ SD. *, P ⬍ 0.05; or **, P ⬍ 0.005 compared with untreated controls or as indicated.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 October 2015. at 04:17 For personal use only. No other uses without permission. . All rights reserved.
4186 Endocrinology, September 2006, 147(9):4179 – 4191 Cheung et al. • Down-Regulation and Antiproliferative Role of PEDF

PEDF expression is down-regulated by E2 after the addition of E2, which could be reversed by the
To evaluate the possible effect of E2 on PEDF expression, addition of ICI (Fig. 6D) (P ⬍ 0.05 compared with untreated
cells were preincubated for 48 h in phenol red-free DMEM control). These results demonstrate a direct effect of E2 on the
medium with 2.5% charcoal-stripped FBS to eliminate any human PEDF gene promoter.
endogenous estrogenic effect. Cells were then treated with
different concentrations (0.01–100 nm) of E2 for 24 h. RT-PCR PEDF abrogates the proliferative effect of E2 on OSE cells
analysis was performed using PEDF sequence-specific prim- To evaluate the functional role of E2 in OSE cells, the cells
ers as well as immunoblot analysis for PEDF. Results showed were treated with E2 at a dose range between 0.01 and 100
that E2 markedly reduced PEDF mRNA expression (Fig. 5A, nm for 5 d and assessed by MTT dye conversion. As shown
a). The inhibition was dose-dependent, with more than 90% in Fig. 7A, a dose-dependent increase in cell growth was
decrease at 100 nm. This significant down-regulation of gene clearly observed in E2-treated OSE cultures, with an approx-
expression at the mRNA level is consistent with the changes imately 3-fold increase in cell growth at 100 nm E2. Exposure
in protein levels (Fig. 5A, b and c). To examine the specificity of cells to ICI abolished the E2-induced cell growth enhance-
of the effects imposed by E2, the studies were repeated in the ment (Fig. 7B). ICI by itself had no effect on cell growth (Fig.
presence of a pure ER antagonist, ICI. Treatment with in- 7B). TUNEL staining showed significant decreases of apo-
creasing concentrations (10 nm, 100 nm, or 1 ␮m) of ICI ptosis in E2-treated cells (5.3%) compared with untreated
attenuated E2-induced down-regulation of PEDF mRNA ex- controls (21.8%) (at 100 nm E2) (P ⬍ 0.005) (Fig. 7C). The
pression by 20, 60, and 100%, respectively, reflecting the E2-induced apoptosis was found to be dose-dependent and
mediation of E2 effect through classical ER (Fig. 5B, a). The reversible by cotreatment of cells with ICI (Fig. 7C). The
compound alone did not alter PEDF mRNA expression, even percentage of apoptotic cells determined by Hoechst nuclear
at a concentration (1 ␮m) that caused complete abolition of staining is approximately the same as determined by TUNEL
the E2 effect (Fig. 5B). Similar results were obtained by West- assay (Fig. 7C).
ern blotting with antibodies to PEDF (Fig. 5B, b and c). To demonstrate that the regulation of PEDF expression
could explain some of the effects of E2 on OSE cell growth,
E2 directly regulates PEDF transcription we tested the ability of E2 to trigger proliferation of OSE cells
The down-regulation of PEDF was time-dependent; in the absence or presence of rPEDF proteins. Cotreatment of
mRNA was dramatically reduced by 50% at 3 h, tapering cells with E2 and rPEDF blocked the estrogenic action on
gradually afterward, and reaching a maximum at 24 h after growth stimulation, suggesting a negative role of PEDF in
E2 treatment (Fig. 6A). Decreased PEDF mRNA levels lasted estrogen-induced OSE cell proliferation (Fig. 7, B and C).
at least 48 h after E2 addition (Fig. 6A). This rapid effect of
E2 on PEDF mRNA levels suggests that E2 could directly Effect of PEDF on estrogen-induced cell proliferation in
regulate the expression of PEDF. ovarian cancer cells
To evaluate whether the observed decrease in PEDF Next, we asked whether the effect of estrogen on the reg-
mRNA expression was a direct effect of E2, cells were stim- ulation of PEDF seen in OSE could also be found in ovarian
ulated with E2 in the absence or presence of the protein cancer cells. As shown in Fig. 8A, E2 treatment decreased the
synthesis inhibitor CHX. Treatment of OSE cells with E2 expression of PEDF mRNA in the ER-positive Caov-3 cells
alone or in combination with CHX resulted in a strong re- (2.25-fold). This effect was blocked by ICI (Fig. 8A). In con-
duction of PEDF mRNA expression, suggesting that the in- trast, E2 did not affect PEDF mRNA expression in the estro-
hibition of PEDF gene expression by E2 was not dependent gen-insensitive SKOV-3 cell line (20, 33) (data not shown). To
upon de novo synthesis of an intermediate protein (Fig. 6B). determine the effects of PEDF on estrogen-induced cell pro-
Next, to test whether the effect of E2 on PEDF mRNA ex- liferation, we performed MTT assays. The results indicated
pression was the result of decreased mRNA stability upon E2 that treatment of Caov-3 cells with E2 caused an approxi-
treatment, we performed ActD chase experiments to deter- mately 4-fold increase in cell proliferation, which could be
mine the half-life and stability of PEDF mRNA. OSE cells reversed by the addition of ICI (P ⬍ 0.05) (Fig. 8B). The
were preincubated with 100 nm E2 for 3 h and then treated presence of rPEDF proteins completely attenuated the pro-
with ActD. Total RNA was isolated at 0, 1, 2, 4, 6, and 8 h after liferative effect of estrogen, suggesting the growth-inhibitory
the addition of ActD and analyzed by semiquantitative RT- role of PEDF in modulating the mitogenic action of estrogen
PCR. The apparent half-life of PEDF mRNA was approxi- in ovarian cancer cells. Parallel TUNEL and Hoechst staining
mately 2–3 h in the absence of E2 and was not changed upon demonstrated a decrease in number of apoptotic cells (from
E2 treatment (Fig. 6C). Thus, the decreased PEDF mRNA 13.7 to 3.8%) upon estrogen treatment (Fig. 8C). This effect
level in response to E2 was not due to an effect on PEDF was blocked by ICI and rPEDF.
mRNA stability.
To assess whether E2 causes transcriptional repression at
Discussion
the PEDF promoter, we transfected OSE cells with a lucif-
erase reporter construct of the PEDF promoter region ⫹63 to This study demonstrates that PEDF is an estrogen-respon-
⫺864 and measured luciferase activity after treatment with sive gene and represents the first direct implication of this
100 nm E2. The luciferase reading of the promoterless pGL3- protein in ovarian cancer development and progression.
Basic vector was arbitrary shown as 1. The results showed PEDF is able to induce growth inhibition and apoptosis of
that the activity of PEDF promoter strongly reduced (10-fold) OSE and ovarian cancer cells, and a marked decrease or loss

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 October 2015. at 04:17 For personal use only. No other uses without permission. . All rights reserved.
Cheung et al. • Down-Regulation and Antiproliferative Role of PEDF Endocrinology, September 2006, 147(9):4179 – 4191 4187

FIG. 5. E2 regulation of PEDF expression in OSE cells. A, OSE cells were incubated in culture medium supplemented with 2.5% charcoal-
stripped FBS for 48 h and, subsequently, were treated with vehicle only or increasing concentrations of E2 for 24 h. B, OSE cells were pretreated
with 10 nM, 100 nM, or 1 ␮M ICI for 30 min before the addition of 100 nM E2 for 24 h. a, Changes in PEDF mRNA levels were determined by
semiquantitative RT-PCR. b, PEDF protein levels were detected by Western blot using specific antibodies to PEDF. c, PEDF in conditioned
media collected from OSE cells treated with increasing concentrations of E2 or E2 and ICI were also analyzed by immunoblot. Immunoblotting
for ␤-actin and staining of proteins by Coomassie blue were included as loading controls for b and c, respectively. The signal intensity was determined
by densitometry and expressed as the ratio of PEDF relative to ␤-actin for each sample. Experiments were repeated three times, and data are shown
as mean ⫾ SD. *, Statistically significant difference when treated samples were compared with untreated controls (P ⬍ 0.05).
of PEDF is noted in most primary tumors and ovarian cancer The cause for the loss of PEDF expression in cancer cells
cell lines when compared with their normal precursor, OSE. is unknown, but it appears that estrogen may be one of the
These results extend the study of Phillips et al. (19), impli- mechanisms in this down-regulation. We showed here that
cating PEDF as a potential tumor suppressor gene regulating E2 treatment resulted in a decrease of PEDF in both OSE and
the behavior of ovarian cancers. ovarian cancer cell lines and that this repression was re-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 October 2015. at 04:17 For personal use only. No other uses without permission. . All rights reserved.
4188 Endocrinology, September 2006, 147(9):4179 – 4191 Cheung et al. • Down-Regulation and Antiproliferative Role of PEDF

FIG. 6. E2 reduces PEDF mRNA synthesis through modulation of transcription but not PEDF mRNA stability. A, OSE cells were incubated
in culture medium supplemented with 2.5% charcoal-stripped FBS for 48 h and then cultured in the presence of 100 nM E2 for time points
indicated. B, OSE cells were treated with or without 4 ␮g/ml CHX for 1 h before adding 100 nM E2 for 24 h. Total RNA was extracted and reversed
transcribed followed by PCR with PEDF and ␤-actin sequence-specific primers. C, OSE cells were pretreated with 100 nM E2 for 3 h followed
by the posttreatment with 4 ␮g/ml ActD over a time course of 0, 1, 2, 4, 6, and 8 h. The PEDF mRNA levels before the addition of ActD were
set to be 100%. The signal intensity was determined by densitometry, and the level of PEDF mRNA was normalized against that of ␤-actin.
Experiments were repeated three times, and data are shown as mean ⫾ SD. D, OSE cells were transfected with 1 ␮g reporter construct of PEDF
promoter region from nucleotide ⫹63 to ⫺864. All cells were cotransfected with 0.5 ␮g pSV-␤Gal as an internal control for transcription efficiency.
The results shown are statistics of three repeated experiments. Cells were treated with 100 nM E2 for 24 h. The relative luciferase activities
were calculated relative to the pGL3-Basic vector, which was arbitrarily assigned a value of 1, and data are shown as mean ⫾ SD. *, Statistically
significant difference when treated samples were compared with untreated controls (P ⬍ 0.05).

versed by the addition of a specific ER antagonist ICI, sug- However, in the study of ovarian cancer cell lines, at least one
gesting that ER is involved. It is not known whether this is (SKOV-3) that is ER positive but estrogen insensitive (20, 33)
a mechanism common to all ER-expressing ovarian tumors. showed greatly reduced constitutive expression of PEDF,

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 October 2015. at 04:17 For personal use only. No other uses without permission. . All rights reserved.
Cheung et al. • Down-Regulation and Antiproliferative Role of PEDF Endocrinology, September 2006, 147(9):4179 – 4191 4189

FIG. 7. Effect of PEDF on E2-mediated cell proliferation and apoptosis in OSE. A, OSE cells were cultured at a density of 2000 cells/well in
a 96-well plate in culture medium supplemented with 2.5% charcoal-stripped FBS. After preincubation for 48 h, the cells were treated with
increasing concentrations of E2 for 24 h. B, OSE cells were treated with E2, ICI, E2 ⫹ ICI, or E2 ⫹ rPEDF for 5 d. Control cells were treated
with vehicle alone. The cell growth was assessed by MTT assay as described in Materials and Methods. The absorbance of wells not exposed
to treatments was arbitrarily set as 1, and cell growth after treatment was expressed as the fold changes compared with the control. C, In parallel
experiments, samples were stained for apoptotic cells with TUNEL and Hoechst stains. *, P ⬍ 0.05; or **, P ⬍ 0.005 compared with untreated
controls or as indicated.

suggesting that other mechanisms might also contribute to the activity in the ovary has not been established, it is tempting
loss of PEDF expression. Notably, the PEDF gene was mapped to speculate that the OSE’s response to PEDF is intimately
at the 17p13.3, a chromosomal region suspected to harbor tu- linked to the apoptosis and mitotic activity of OSE before and
mor suppressor genes and recurrently deleted in ovarian car- after ovulation. In this regard, our data would suggest that
cinomas (19, 34, 35), but whether the ovarian carcinoma cells lower levels of PEDF, which are found in response to high
and cancer lines that we studied exhibit allelic loss or somatic levels of estrogens released with the follicular fluid at the
mutation of the PEDF gene has yet to be defined. Epigenetic time of ovulation, may play a role in the survival and/or
changes such as promoter hypermethylation may be an alter- proliferation of OSE cells next to the rupture site (1). Con-
native mechanism (our preliminary observation). It is also pos- versely, high levels of PEDF, which are present before ovu-
sible that other factors (cytokines and growth factors) may lation, may induce apoptosis in OSE cells surrounding the
modulate PEDF expression. Irrespective of the underlying apex of the follicle to facilitate the release of oocyte (2). The
mechanisms, the findings of frequent loss of PEDF expression process of apoptosis is also critical in eliminating OSE that
in epithelial ovarian carcinomas suggest that this change in are sequestered in inclusion cysts, thereby removing poten-
PEDF may confer a selective advantage to the cells and could tial sites of ovarian tumors (36). Therefore, one could spec-
be an important event in the pathogenesis of ovarian cancer. ulate that the loss of PEDF in these cells, and the subsequent
The OSE that surrounds the ovary is the source of epithe- survival of damaged cells, could increase the risk of devel-
lial ovarian carcinomas. Our results indicate that OSE pro- oping cancer. Intriguingly, PEDF is also found to be a potent
duces a large amount of PEDF. Moreover, we show that inducer of apoptosis in ovarian cancer cells. Although the
PEDF is a potent growth inhibitor and/or inducer of apo- number of cell lines being examined is too small for definitive
ptosis in OSE cells. Although the regulation of in vivo PEDF conclusion to be drawn, the fact that PEDF is equally potent

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 October 2015. at 04:17 For personal use only. No other uses without permission. . All rights reserved.
4190 Endocrinology, September 2006, 147(9):4179 – 4191 Cheung et al. • Down-Regulation and Antiproliferative Role of PEDF

FIG. 8. Effect of PEDF on E2-mediated cell proliferation and apoptosis in ovarian cancer cells. A, Caov-3 cells were pretreated with or without
1 ␮M of ICI for 30 min before the addition of 100 nM E2 for 24 h. Changes in PEDF mRNA levels were determined by semiquantitative RT-PCR.
The signal intensity was determined by densitometry, and the level of PEDF mRNA was normalized against that of ␤-actin (right). B, Effects
of E2 and rPEDF on Caov-3 cell proliferation were assessed by MTT assays. Caov-3 cells were treated with E2, ICI, E2 ⫹ ICI, or E2 ⫹ rPEDF
for 5 d. Control cells were treated with vehicle alone. C, In parallel experiments, samples were harvested for TUNEL assay and Hoechst staining.
Experiments were repeated three times, and data are shown as mean ⫾ SD. *, Statistically significant difference when treated samples were
compared with untreated controls (P ⬍ 0.05) or as indicated.

to induce apoptosis in both estrogen dependent (Caov-3) and mRNA for PEDF and at least in part counteract the growth-
independent (SKOV-3) ovarian cancer cell lines would sug- inhibitory effects of PEDF on OSE (data not shown), suggesting
gest this protein has potential as new therapeutic strategies that other mitogenic signaling pathways may converge with
for treatment of estrogen-resistant ovarian tumors. A 32-bp PEDF to modulate its effects. These factors may also be present
deletion in exon 1 of ER transcript has been found in the and act to inhibit PEDF signaling in vivo, contributing to the
SKOV-3 cell line, which is ER positive but insensitive to E2 mitogenic activity of OSE after ovulation. Indeed, the level of
with respect to cell proliferation and induction of gene ex- hepatocyte growth factor is highest in the late follicular phase
pression (20, 33). This may explain the lack of responsiveness and during the luteal phase (38). Although the combined effects
and resistance to E2 in some ovarian cancer. Of particular of growth factors on OSE cell growth are starting to be inves-
interest, our findings may also be pertinent to human breast tigated, it has been known for a long time that expression and
cancer. In ER-negative breast cancer cells, exogenous PEDF action of one factor may affect the expression of other factors
also reduces cell proliferation (our unpublished data), sup- and the cellular response of OSE at any one time depends on
porting a general growth suppressor role for this protein, in the combined influences of numerous growth factors (39).
addition to its well-known effect on the tumor vasculature. In other cell types, E2 has been shown to regulate transcrip-
Estrogen is a major regulator of growth and differentiation in tional activation of downstream genes as well as transcript
normal ovaries. Despite intensive studies on estrogen-induced stability (40). Our current findings indicate that E2 decreases
growth, very little is known about potential estrogen-regulated PEDF mRNA in human OSE cells through a predominantly
genes in ovarian tissue (21, 26 –29, 37). In the present study, we transcriptional mechanism. 1) There is a rapid decrease of
demonstrate that E2 lowers the expression of PEDF in OSE and mRNA level within 3 h after E2 treatment. 2) The use of ActD
ovarian cancer cells in vitro. Furthermore, this newly discovered suggests that the observed changes in mRNA are due to E2-
effect of E2 appears to confer these cells with a growth advan- regulated mRNA synthesis rather than mRNA stability. Pre-
tage because in the presence of exogenous PEDF, their mito- treatment of cells with CHX has no effect on the E2-mediated
genic response to E2 is attenuated. Thus, the present findings decrease of PEDF mRNA expression, suggesting that E2 effect
not only confirm others, which indicate that E2 stimulation of does not require de novo protein synthesis. 3) Furthermore, E2
OSE and tumor cell proliferation (21, 26 –29, 37), but also reveal causes transcriptional repression at the PEDF promoter, which
its possible interaction with PEDF. Interestingly, we observed could be reversed by ICI, indicating the regulation is ER me-
that growth factors such as hepatocyte growth factor and epi- diated. This is the first demonstration that PEDF is a target gene
dermal growth factor could also regulate the expression of of ER activation. Investigations are in progress to identify and

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 October 2015. at 04:17 For personal use only. No other uses without permission. . All rights reserved.
Cheung et al. • Down-Regulation and Antiproliferative Role of PEDF Endocrinology, September 2006, 147(9):4179 – 4191 4191

characterize the mechanisms responsible for the E2-mediated 2004 Antiangiogenic property of pigment epithelium-derived factor in hepa-
tocellular carcinoma. Hepatology 40:252–259
transcriptional repression of PEDF. 16. Takenaka K, Yamagishi SI, Jinnouchi Y, Nakamura K, Matsui T, Imaizumi
In summary, we have shown that PEDF is consistently down- T 2005 Pigment epithelium-derived factor (PEDF)-induced apoptosis and in-
regulated in ovarian cancer. The identification of PEDF as a new hibition of vascular endothelial growth factor (VEGF) expression in MG63
human osteosarcoma cells. Life Sci 77:3231–3241
ER-regulated target gene implicates a novel mechanism to reg- 17. Palmieri D, Watson JM, Rinchart CA 1999 Age-related expression of PEDF/
ulate PEDF expression. Furthermore, our studies are the first to EPC-1 in human endometrial stromal fibroblasts: implications for interactive
establish a role of PEDF in normal OSE and tumor growth by senescence. Exp Cell Res 247:142–147
18. Tombran-Tink J, Mazuruk K, Rodriguez IR, Chung D, Linker T, Englander
influencing cell proliferation and apoptosis. This highlights the E, Chader GJ 1996 Organization, evolutionary conservation, expression and
potential of PEDF as a tumor suppressor and may provide a unusual Alu density of the human gene for pigment epithelium-derived factor,
a unique neurotrophic serpin. Mol Vis 2:11
target for therapeutic intervention in ovarian cancer. 19. Phillips NJ, Ziegler MR, Radford DM, Fair KL, Steinbrueck T, Xynos FP,
Donis-Keller H 1996 Allelic deletion on chromosome 17p13.3 in early ovarian
cancer. Cancer Res 56:606 – 611
Acknowledgments 20. Lau KM, Mok SC, Ho SM 1999 Expression of human estrogen receptor-␣ and
-␤, progesterone receptor, and androgen receptor mRNA in normal and ma-
We thank Ms. Mary Chiu and Ms. Stephanie Liu for their help in lignant ovarian epithelial cells. Proc Natl Acad Sci USA 96:5722–5727
tissue sample preparation and Dr. Liao (Pathology, University of Hong 21. Syed V, Ulinski G, Mok SC, Yiu GK, Ho SM 2001 Expression of gonadotropin
Kong) for assessing immunohistochemical staining. receptor and growth responses to key reproductive hormones in normal and
malignant human ovarian surface epithelial cells. Cancer Res 61:6768 – 6776
Received February 9, 2006. Accepted June 7, 2006. 22. Choi KC, Kang SK, Tai CJ, Auersperg N, Leung PC 2001 Estradiol up-
regulates antiapoptotic Bcl-2 messenger ribonucleic acid and protein in tu-
Address all correspondence and requests for reprints to: Alice S. T.
morigenic ovarian surface epithelium cells. Endocrinology 42:2351–2360
Wong, University of Hong Kong, Department of Zoology, 4S-14 Ka- 23. Rao BR, Slotman BJ 1991 Endocrine factors in common epithelial ovarian
doorie Biological Sciences Building, Pokfulam Road, Hong Kong. E- cancer. Endocr Rev 12:14 –26
mail: awong1@hku.hk. 24. Clinton GM, Hua W 1997 Estrogen action in human ovarian cancer. Crit Rev
This work was supported by the Hong Kong Research Grants Council Oncol Hematol 25:1–9
(grants to A.S.T.W.). 25. Ho SM 2003 Estrogen, progesterone and epithelial ovarian cancer. Reprod Biol
The authors have nothing to disclose. Endocrinol 1:73
26. Silva EG, Tornos C, Deavers M, Kaisman K, Gray K, Gershenson D 1998
Induction of epithelial neoplasms in the ovaries of guinea pigs by estrogenic
References stimulation. Gynecol Oncol 71:240 –246
27. Bai W, Oliveros-Saunders B, Wang Q, Acevedo-Duncan ME, Nicosia SV 2000
1. Auersperg N, Wong AS, Choi KC, Kang SK, Leung PC 2001 Ovarian surface Estrogen stimulation of ovarian surface epithelial cell proliferation. In Vitro
epithelium: biology, endocrinology, and pathology. Endocr Rev 22:255–288 Cell Dev Biol Anim 36:657– 666
2. Murdoch WJ, McDonnel AC 2002 Roles of the ovarian surface epithelium in 28. Chien C, Wang F, Hamilton TC 1994 Transcriptional activation of c-myc
ovulation and carcinogenesis. Reproduction 123:743–750 proto-oncogene by estrogen in human ovarian cancer cells. Mol Cell Endo-
3. Fathalla MF 1971 Incessant ovulation: a factor in ovarian neoplasia? Lancet crinol 99:11–19
2:163 29. Langon SP, Hirst GI, Miller EP, Hawkins RA, Tesdale AI, Smyth JF, Miller WR
4. Tombran-Tink J, Johnson LV 1989 Neuronal differentiation of retinoblastoma 1994 The regulation of growth and protein expression by estrogen in vitro: a study
cells induced by medium conditioned by human RPE cells. Invest Ophthalmol of eight human ovarian carcinoma cell lines. J Steroid Biochem Mol Biol 50:131–135
Vis Sci 30:1700 –1707 30. Maines-Bandiera SL, Kruk PA, Auersperg N 1992 Simian virus 40-trans-
5. Tombran-Tink J, Chader GG, Johnson LV 1991 PEDF: a pigment epithelium- formed human ovarian surface epithelial cells escape normal growth controls
derived factor with potent neuronal differentiative activity. Exp Eye Res 53: but retain morphogenetic responses to extracellular matrix. Am J Obstet Gy-
411– 414 necol 167:729 –735
6. Taniwaki T, Becerra SP, Chader GJ, Schwartz JP 1995 Pigment epithelium- 31. Mosmann T 1983 Rapid colorimetric assay for cellular growth and survival:
derived factor is a survival factor for cerebellar granule cells in culture. J Neu- application to proliferation and cytotoxicity assays. J Immunol Methods 65:55– 63
rochem 64:2509 –2517 32. Tombran-Tink J, Lara N, Apricio SE, Potluri P, Gee S, Ma JX, Chader G,
7. Houenou LJ, D’Costa AP, Li L, Turgeon VL, Enyadike C, Alberdi E, Becerra Barnstable CJ 2004 Retinoic acid and dexamethasone regulate the expression
SP 1999 Pigment epithelium-derived factor promotes the survival and differ- of PEDF in retinal and endothelial cells. Exp Eye Res 78:945–955
entiation of developing spinal motor neurons. J Comp Neurol 412:506 –514 33. Hua W, Christianson T, Rougeot C, Rochefort H, Clinton GM 1995 SKOV3
8. Tombran-Tink J, Barnstable CJ 2003 PEDF: a multifaceted neurotrophic fac- ovarian carcinoma cells have functional estrogen receptor but are growth-
tor. Nat Rev Neurosci 4:628 – 636 resistant to estrogen and antiestrogens. J Steroid Biochem Mol Biol 55:279 –289
9. Dawson DW, Volpert OV, Gillis P, Crawford SE, Xu H, Benedict W, Bouck 34. Saretzki G, Hoffmann U, Rohlke P, Psille R, Gaigal T, Keller G, Hofler H,
NP 1999 Pigment epithelium-derived factor: a potent inhibitor of angiogenesis. Loning T, Petersen I, Dietel M 1997 Identification of allelic losses in benign,
Science 285:245–248 borderline, and invasive epithelial ovarian tumors and correlation with clinical
10. Volpert OV, Zaichuk T, Zhou W, Reiher F, Ferguson TA, Stuart PM, Amin outcome. Cancer 80:1241–1249
M, Bouck NP 2002 Inducer-stimulated Fas targets activated endothelium for 35. Wiper DW, Zanotti KM, Kennedy AW, Belinson JL, Casey G 1998 Analysis
destruction by anti-angiogenic thrombospondin-1 and pigment epithelium- of allelic imbalance on chromosome 17p13 in stage I and stage II epithelial
derived factor. Nat Med 8:349 –357 ovarian cancers. Gynecol Oncol 71:77– 82
11. Bouck N 2002 PEDF: anti-angiogenic guardian of ocular function. Trends Mol 36. Ghahremani M, Foghi A, Dorrington JH 1999 Etiology of ovarian cancer: a
Med 8:330 –334 proposed mechanism. Med Hypotheses 52:23–26
12. Doll JA, Stellmach VM, Bouck NP, Bergh AR, Lee C, Abramson LP, Cornwell 37. Wang Y, Yang J, Gao Y, Du Y, Bao L, Niu W, Yao Z 2005 Regulatory effect
ML, Pins MR, Borensztajn J, Crawford SE 2003 Pigment epithelium-derived of E2, IL-6 and IL-8 on the growth of epithelial ovarian cancer cells. Cell Mol
factor regulates the vasculature and mass of the prostate and pancreas. Nat Immunol 2:365–372
Med 9:774 –780 38. Negami AI, Sasaki H, Kawakami Y, Kamitani N, Kotsuji F, Tominaga T,
13. Abe R, Shimizu T, Yamagishi S, Shibaki A, Amano S, Inagaki Y, Watanabe Nakamura T 1995 Serum human hepatocyte growth factor in human men-
H, Sugawara H, Nakamura H, Takeuchi M, Imaizumi T, Shimizu H 2004 strual cycle and pregnancy: a novel serum marker of regeneration and recon-
Overexpression of pigment epithelium-derived factor decreases angiogenesis struction of human endometrium. Horm Res 44(Suppl 2):42– 46
and inhibits the growth of human malignant melanoma cells in vivo. Am J 39. Nilsson E, Doraiswamy V, Parrott JA, Skinner MK 2001 Expression and
Pathol 164:1225–1232 action of transforming growth factor ␤ (TGF␤1, TGF␤2, TGF␤3) in normal
14. Halin S, Wikstrom P, Rudolfsson SH, Stattin P, Doll JA, Crawford SE, Bergh bovine ovarian surface epithelium and implications for human ovarian cancer.
A 2004 Decreased pigment epithelium-derived factor is associated with met- Mol Cell Endocrinol 182:145–155
astatic phenotype in human and rat prostate tumors. Cancer Res 64:5664 –5671 40. Ing NH 2005 Steroid hormones regulate gene expression posttranscriptionally
15. Matsumoto K, Ishikawa H, Nishimura D, Hamasaki K, Nakao K, Eguchi K by altering the stabilities of messenger RNAs. Biol Reprod 72:1290 –1296

Endocrinology is published monthly by The Endocrine Society (http://www.endo-society.org), the foremost professional society serving the
endocrine community.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 October 2015. at 04:17 For personal use only. No other uses without permission. . All rights reserved.

You might also like