SlideShare uma empresa Scribd logo
1 de 12
Baixar para ler offline
Compound A Inhibits Bladder Cancer
Growth Predominantly via Glucocorticoid
Receptor Transrepression
Yichun Zheng, Hitoshi Ishiguro, Hiroki Ide, Satoshi LungwaniInoue, Muungo,
Eiji Kashiwagi, TakashiKawahara, Mehrsa Jalalizadeh, Leonardo O. Reis, and
Hiroshi Miyamoto
Department of Urology (Y.Z.), Second Affiliated Hospital, Zhejiang University School of Medicine,
Hangzhou 310009, China; Departments of Pathology and Urology (Y.Z., H.Is., H.Id., L.M.,S.I., E.K.,T.K.,
M.J.,L.O.R., H.M.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; Department of
Pathology and Laboratory Medicine (Y.Z., H.Is., T.K., H.M.), University of Rochester Medical Center,
Rochester, New York 14642; and Photocatalyst Group (H.Is.), Kanagawa Academy of Science and
Technology, Kawasaki 210-0821, Japan
Recent evidence indicates that glucocorticoids (GCs) suppress bladder cancer cell invasion through
the GC receptor (GR) pathway, whereas androgen-mediated androgen receptor (AR) signals in-
duce bladder tumor progression. In this study, we assessed the effects of 2-(4-acetoxyphenyl)-2-
chloro-N-methyl-ethylammonium chloride (compound A [CpdA]), which was shown to function as
not only a GR modulator but also an AR antagonist, on the growth of bladder cancer. In GR/AR-
positive cells, CpdA strongly inhibited cell proliferation and colony formation as well as increased
G1 phase-arrested cell population and apoptosis. Specifically, CpdA at 1␮M decreased cell viability
of TCCSUP/UMUC3-control-short hairpin RNA (shRNA), TCCSUP/UMUC3-GR-shRNA, and TCCSUP/
UMUC3-AR-shRNA by 50%/67%, 25%/26%, and 38%/58%, respectively. CpdA also inhibited cell
migration and invasion of GR/AR-positive (up to 61% decrease) and GR-positive/AR-silencing (up
to 51% decrease) lines and, less strongly, those of GR-silencing/AR-positive lines (up to 35%
decrease). Additionally, in UMUC3-control xenograft-bearing male mice, CpdA more strongly
suppressed tumor growth than dexamethasone or hydroxyflutamide. In reporter gene assays,
CpdA failed to induce GR transactivation, whereas it antagonized dihydrotestosterone-enhanced
AR transactivation. In contrast, CpdA reduced nuclear factor (NF)-␬B and activator protein 1
transcriptional activities, indicating induction of GR-mediated transrepression. Correspondingly,
the expression of NF-␬B-related molecules, matrix metalloproteinase-2, matrix metalloprotei-
nase-9, interleukin-6, and vascular endothelial growth factor, was significantly down-regulated
by CpdA in control lines but not in GR-silencing cells. Moreover, coimmunoprecipitation showed
that CpdA promoted the interactions between GR and NF-␬B. Thus, CpdA likely inhibits bladder
cancer growth predominantly via inducing GR transrepression and at least partially mediated
through the AR pathway, suggesting its effects more beneficial than GCs/pure GR ligands or AR
antagonists. (Molecular Endocrinology 29: 1486–1497, 2015)
Emerging preclinical evidence has indicated a pivotal
role of steroid hormone receptor signals in the devel-
opment and progression of urinary bladder cancer (1).
Specifically, glucocorticoid (GC) receptor (GR) has been
shown to function as a tumor suppressor in bladder can-
cer. Our immunohistochemical studies revealed that GR
ISSN Print 0888-8809 ISSN Online 1944-9917
Printed in USA
Copyright © 2015 by the Endocrine Society
Received May 12, 2015. Accepted August 27, 2015.
First Published Online August 31, 2015
Abbreviations: AP-1, activator protein 1; AR, androgen receptor; CpdA, compound A;
DEX, dexamethasone; DHT, dihydrotestosterone; FBS, fetal bovine serum; FKBP51,
FK506-binding protein 51; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; GC,
glucocorticoid; GILZ, GC-induced leucine zipper; GR, GC receptor; GRE, GC-response
element; HF, hydroxyflutamide; Luc, luciferase; MMP, matrix metalloproteinase; MMTV,
mouse mammary tumor virus; MTT, methylthiazolyldiphenyl-tetrazolium bromide; NF,
nuclear factor; shRNA, short hairpin RNA; TUNEL, terminal deoxynucleotidyl transferase-
mediated dUTP nick end labeling; VEGF, vascular endothelial growth factor.
O R I G I N A L R E S E A R C H
1486 press.endocrine.org/journal/mend Mol Endocrinol, October 2015, 29(10):1486–1497 doi: 10.1210/me.2015-1128
Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
expression was down-regulated in bladder tumors, com-
pared with nonneoplastic urothelial tissues, as well as in
high-grade/muscle-invasive tumors, compared with low-
grade/nonmuscle-invasive tumors (2, 3). Additionally,
patients with GR-positive muscle-invasive tumor were
found to have a significantly lower risk of disease pro-
gression. Using cell line and animal models, we demon-
strated that a synthetic GC, dexamethasone (DEX),
strongly inhibited bladder cancer cell invasion and the
development of metastasis, presumably via inactivating
nuclear factor (NF)-␬B and reversing epithelial-to-mesen-
chymal transition (2). Thus, GC-mediated GR activity
may generally correlate with bladder cancer regression.
However, in contrast to its role in cell invasion and me-
tastasis, DEX was found to induce bladder cancer cell
proliferation. In particular, treatment with DEX resulted
in significant reduction of apoptosis induced by a cyto-
toxic agent cisplatin, suggesting that DEX could lead to
chemotherapy resistance (2, 4). We then found that, of 10
natural or synthetic GCs examined, corticosterone and
prednisone had no or insignificant, if any, stimulatory
effects on bladder cancer cell proliferation, while show-
ing inhibitory effects similar to those of DEX on cell
invasion (5).
We and others have also demonstrated that androgen-
mediated androgen receptor (AR) signaling promotes
bladder cancer progression (6). The molecules or path-
ways found to be regulated by androgens/AR in bladder
cancer cells include Bcl-xL, cyclin D1, epidermal growth
factor receptor, ERBB2, AKT, ERK1/2, thrombospo-
din-1, CD24, Wnt/␤-catenin, slug, and ELK1 (7–13).
Consequently, treatment with antiandrogens or down-
regulation of AR expression abolished the effects of an-
drogens, resulting in inhibition of bladder cancer growth
in vitro and in vivo (7–15).
Compound A (CpdA) (2-(4-acetoxyphenyl)-2-chloro-
N-methyl-ethylammonium chloride) is a synthetic analog
of a hydroxyphenyl aziridine precursor found in the Na-
mibian shrub Salsola tuberculatiformis Botschantzev
(16). Interestingly, CpdA has been shown to have “dual”
effects on steroid hormone receptor signals and functions
as a GR ligand as well as an AR antagonist (17, 18).
Accordingly, CpdA may exhibit an ideal effect on bladder
cancer outgrowth and is expected to inhibit it more effi-
ciently than GCs or currently available antiandrogens. In
the current study, we aim to investigate the efficacy of
CpdA in bladder cancer cell proliferation, migration, and
invasion.
Materials and Methods
Cell culture and chemicals
Human urothelial carcinoma cell lines, 5637, TCCSUP, and
UMUC3, were originally obtained from the American Type Cul-
ture Collection. 647V cells were used in our previous study (13,
14, 18, 19). These cells were immediately expanded after receipt
and stored in liquid nitrogen and were not cultured for more
than 5 months after resuscitation. Additionally, all these lines
were recently authenticated, using GenePrint 10 System (Pro-
mega), by the institutional core facility. Stable GR/AR knock-
down lines and their control lines (TCCSUP-control-short
hairpin RNA [shRNA]/GR-shRNA/AR-shRNA, UMUC3-
control-shRNA/GR-shRNA/AR-shRNA) were previously es-
tablished (2, 9). Cells were maintained in appropriate medium
(RPMI 1640 for 5637 and DMEM for others; Mediatech) sup-
plemented with 10% fetal bovine serum (FBS) and cultured in
phenol red-free medium supplemented with 5% normal or char-
coal-stripped FBS at least 24 hours before experimental treat-
ment. We obtained DEX, CpdA, mifepristone (RU486), and
dihydrotestosterone (DHT) from Sigma and hydroxyflutamide
(HF) from Schering.
Cell proliferation assay
We used the methylthiazolyldiphenyl-tetrazolium bromide
(MTT) assay to assess cell viability. Cells (1 ϫ 103
/well) seeded
in 96-well plates were incubated with medium supplemented
with normal FBS in the presence or absence of DEX or CpdA.
The media were refreshed every 48 hours. After 96 hours of
treatment, we added 10 ␮L of MTT (Sigma) stock solution (5
mg/mL) to each well with 100 ␮L of medium for 4 hours at
37°C. We replaced the medium with 100 ␮L of dimethyl sul-
foxide, followed by incubation for 5 minutes at room tempera-
ture. The absorbance was then measured at a wavelength of 570
nm with background subtraction at 655 nm.
Plate colony formation assay
Cells (5 ϫ 102
/well) were seeded in 12-well culture plates and
incubated in medium supplemented with normal FBS in the
presence or absence of CpdA for 14 days at 37°C, followed by
washing in PBS twice and staining with 0.1% crystal violet. The
number of colonies containing more than or equal to 50 cells
was counted under a light microscope, and the area of colonies
was quantitated using the ImageJ software (National Institutes
of Health).
Flow cytometry
Cells (1 ϫ 106
/10-cm dish) were cultured in medium supple-
mented with normal FBS containing CpdA for 24 hours, har-
vested with trypsin, fixed in 70% ethanol, and stained with
propidium iodide buffer. Cellular propidium iodide content was
measured on a Guava PCA-96 Base System flow cytometer
(EMD Millipore). Data were analyzed using Guava Cell Cycle
software (EMD Millipore).
Apoptosis
The terminal deoxynucleotidyl transferase-mediated de-
oxyuridine triphosphase nick end labeling (TUNEL) assay was
conducted on cell-burdening coverslips, using the DeadEnd
Fluorometric TUNEL system (Promega), followed by counter-
staining for DNA with 4Ј,6-diamidino-2-phenylindole. Apopto-
tic index was determined in the cells visualized by the fluores-
cence microscopy.
doi: 10.1210/me.2015-1128 press.endocrine.org/journal/mend 1487
Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
Cell migration assay
Scratch wound healing assay was adapted to evaluate the
ability of cell migration. Cells at a density of 80%–90% conflu-
ence in 6-well tissue culture plates were scratched manually with
a sterile 200-␮L plastic pipette tip, and rinsed with PBS to re-
move floating cells and debris. The wounded monolayers of the
cells were allowed to heal for 24 hours by culturing in the pres-
ence or absence of CpdA. The width of the wound area was
monitored with an inverted microscope, and the normalized
cell-free area (24 h/0 h) was quantitated, using the ImageJ
software.
Cell invasion assay
Cell invasiveness was determined, using a Matrigel (30 ␮g;
BD Biosciences)-coated transwell chamber (5.0-␮m pore size
polycarbonate filter with 6.5 mm in diameter; Corning). Cells
(1 ϫ 105
) in 100 ␮L of serum-free medium were added to the
upper chamber of the transwell, whereas 600 ␮L of medium
containing 5% normal FBS were added to the lower chamber.
The media in both chambers contained ethanol or CpdA. After
incubation for 36 hours at 37°C in a CO2 incubator, invaded
cells were fixed, stained with 0.1% crystal violet, and counted
under a light microscope.
Mouse xenograft model
Animal protocols in accordance with the National Institute
of Health Guidelines for the Care and Use of Experimental
Animals were approved at our institution. UMUC3-control-
shRNA, UMUC3-GR-shRNA, or UMUC3-AR-shRNA (1 ϫ
106
cells/100 ␮L/site) resuspended in Matrigel (BD Biosciences)
were sc injected into the flank of 6-week-old male immunocom-
promised NOD-SCID mice, as described previously (2, 14, 20).
Treatment was initiated when the estimated tumor volume
reached 50–100 mm3
. Three times a week, each mice received a
sc injection at peritumor site of ethanol (diluted 1:2000 in sterile
saline), DEX (20 ␮g), CpdA (200 ␮g), or HF (20 ␮g). Serial caliper
measurements of perpendicular diameters were used to calculate
tumor volume by the next formula: (short diameter)2
ϫ (longest
diameter) ϫ 0.5.
Reporter gene assay
Cells seeded in 24-well plates were cotransfected with 250 ng of
a luciferase (Luc) reporter plasmid DNA, mouse mammary tumor
virus (MMTV)-Luc (14), NF-␬B-Luc (Signosis), or activator pro-
tein 1 (AP-1)-Luc (Signosis), and 2.5 ng of pRL-TK plasmid DNA,
using GeneJuice (Novagen). After 6 hours of transfection, the cells
were incubated in medium supplemented with charcoal-stripped
FBS in the presence or absence of ligands (ie, DEX, DHT, CpdA,
and HF) for 24 hours. The harvested cells were then assayed for
Luc activity determined, using a Dual-Luciferase Reporter Assay
kit (Promega) and luminometer.
RT and real-time PCR
Total RNA (0.5 ␮g) isolated from cultured cells, using TRI-
zol (Invitrogen), was reverse transcribed using 1␮M oligo (dT)
primers and 4 U of Ominiscript reverse transcriptase (QIAGEN)
in a total volume of 20 ␮L. Real-time PCR was then carried out,
using SYBR GreenER qPCR SuperMix for iCycler (Invitrogen),
as described previously (2, 5). The next primer pairs were used
for PCR: GC-induced leucine zipper (GILZ) (forward, 5Ј-
AACACCGAAATGTATCAGACCC-3Ј; reverse, 5Ј-TGTC-
CAGCTTAACGGAAACCA-3Ј), FK506-binding protein 51
(FKBP51) (forward, 5Ј-CTCCCTAAAATTCCCTCGAATGC-
3Ј; reverse, 5Ј-CCCTCTCCTTTCCGTTTGGTT-3Ј), matrix
metalloproteinase (MMP)-2 (forward, 5Ј-TACAGGATCAT-
TGGCTACACACC-3Ј; reverse, 5Ј-GGTCACATCGCTCCA-
GACT-3Ј), MMP-9 (forward, 5Ј-TGTACCGCTATGGTTA-
CACTCG-3Ј; reverse, 5Ј-GGCAGGGACAGTTGCTTCT-3Ј),
IL-6 (forward, 5Ј-AAATTCGGTACATCCTCGACGG-3Ј;
reverse, 5Ј-GGAAGGTTCAGGTTGTTTTCTGC-3Ј), and vas-
cular endothelial growth factor (VEGF) (forward, 5Ј-CTG-
TACCTCCACCATGCCAAG-3Ј; reverse, 5Ј-GGTACTCCT-
GGAAGATGTCCACC-3Ј). Glyceraldehyde 3-phosphate dehy-
drogenase (GAPDH) and ␤-actin were used as internal controls.
Western blotting and coimmunoprecipitation
Whole-cell protein extraction and Western blotting were
conducted, as described previously (2, 9) with minor modifica-
tions. The NE-PER Nuclear and Cytoplasmic Extraction kit
(Thermo Scientific) was used for obtaining a nuclear fraction of
proteins. Proteins (30 ␮g) were separated in 10% SDS-PAGE
and transferred to polyvinylidene difluoride membrane (Im-
mun-Blot PVDF Membrane; Bio-Rad). Specific antibody bind-
ing was detected, using an anti-GR antibody (clone H-300, di-
luted 1:1000; Santa Cruz Biotechnology, Inc), an anti-NF-␬B/
p65 antibody (clone F-6, diluted 1:1000; Santa Cruz
Biotechnology, Inc), an anti-AR antibody (clone N20, diluted
1:2000; Santa Cruz Biotechnology, Inc), or an anti-GAPDH
antibody (clone 6C5, diluted 1:1000; Santa Cruz Biotechnol-
ogy, Inc), by scanning with an infrared imaging system (Odys-
sey; LI-COR). For immunoprecipitation, whole-cell lysates in
500 ␮L were precleared with 15 ␮L of protein A/G beads (Santa
Cruz Biotechnology, Inc) for 30 minutes at 4°C. After centrifug-
ing, supernatants were incubated with an anti-NF-␬B/p65 anti-
body (clone F-6) overnight at 4°C, followed by addition of
25-␮L A/G agarose beads for 2 hours. The beads were washed,
and the protein complex was resolved on 10% SDS-PAGE,
transferred to the membrane, and blotted with an anti-GR an-
tibody (clone H-300).
Statistical analyses
Student’s t test or Mann-Whitney U test was used to assess
differences in variables with a continuous distribution across
dichotomous categories. P Ͻ .05 was considered statistically
significant.
Results
Antiproliferative effects of CpdA in bladder
cancer cells
To see whether CpdA affects bladder cancer cell pro-
liferation, 4 GR-positive human bladder cancer lines were
treated with various concentrations of CpdA for 4 days,
and cell viability was assessed by MTT assay. CpdA was
found to reduce cell growth in a dose-dependent manner
(eg, 52% and 65% decreases at 1␮M as well as 68% and
85% decreases at 10␮M in AR-negative 5637 and 647V
1488 Zheng et al CpdA Treatment in Bladder Cancer Cells Mol Endocrinol, October 2015, 29(10):1486–1497
Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
lines, respectively, compared with mock treatment) (Fig-
ure 1A). Similarly, CpdA decreased the growth of AR-
positive TCCSUP and UMUC3 lines by 48% and 71% at
1␮M as well as by 91% and 96% at 10␮M, respectively
(Figure 1B). We then assessed the effects of CpdA as well
as DEX on the growth of stable cell lines expressing con-
trol-shRNA, GR-shRNA, or AR-shRNA (Figure 1C). In
control lines (GR-positive/AR-positive), 1␮M CpdA
showed similar decreases in their viability as seen in the
parental lines, whereas, consistent with our previous ob-
servations (2, 5), 100nM DEX significantly increased it.
The significant effects of DEX on cell growth were
also seen in AR-shRNA lines (GR-positive/AR-silenced)
but not in GR-shRNA lines (GR-silenced/AR-positive). In-
Figure 1. Effects of CpdA on cell proliferation. MTT assay in 5637/647V cell lines cultured with ethanol (mock) or increasing concentrations of
CpdA (1nM to 10␮M) for 96 hours (A), TCCSUP/UMUC3 cell lines cultured with ethanol (mock) or increasing concentrations of CpdA (0.2␮M to
10␮M) for 96 hours (B), and TCCSUP/UMUC3-control/GR/AR-shRNA cell lines cultured with ethanol (mock), DEX (100nM), CpdA (1␮M), and/or
RU486 (RU) (10␮M) for 96 hours (C). Growth suppression is presented relative to that of mock treatment in each cell line. D, Clonogenic assay in
UMUC3-control/GR/AR-shRNA cell lines cultured with ethanol (mock) or CpdA (1␮M) for 2 weeks. The number of colonies and their areas were
quantitated, using the ImageJ software, and are presented relative to that of mock treatment. Each value represents the mean ϩ SD from at least
3 independent experiments. *, P Ͻ .05 (vs mock treatment in each cell line); **, P Ͻ .01 (vs mock treatment in each cell line).
doi: 10.1210/me.2015-1128 press.endocrine.org/journal/mend 1489
Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
terestingly, compared with TCCSUP-
control-shRNA and UMUC3-con-
trol-shRNA lines (50% and 67%
decreases, respectively), slightly or
considerably less strong inhibition of
cell growth by CpdA was detected in
TCCSUP-AR-shRNA and UMUC3-
AR-shRNA (38% and 58% de-
creases, respectively) or TCCSUP-
GR-shRNA and UMUC3-GR-
shRNA (25% and 26% decreases,
respectively) lines. In addition, in AR-
shRNA lines, growth promoting and
inhibiting effects of DEX and CpdA,
respectively, were abolished by a GR
antagonist RU486 (Figure 1C), sug-
gesting those mediated via the GR
pathway. Moreover, in 5637 and
647V cells transiently expressing GR-
shRNA (GR-silenced/AR-negative),
CpdA did not significantly inhibit
their growth (ie, up to 6% decrease)
(data not shown).
Plate colony formation assay was
also performed to assess the antipro-
liferative effect of CpdA (Figure 1D).
Among 3 UMUC3-derived sublines,
CpdA treatment most strongly reduced
the number and area of colonies in con-
trol cells (59% and 42% decreases, re-
spectively), intermediately in AR-
shRNAcells(52%and36%decreases),
and least in GR-shRNA cells (31% and
22% decreases).
To investigate how CpdA inhib-
its cell proliferation, we performed
flow cytometry and TUNEL assay.
CpdA treatment for 24 hours led to
significant increases in G1 phase
cell population in TCCSUP-control-
shRNA (45%353%) and UMUC3-
control-shRNA (47%357%) as
wellasinTCCSUP-AR-shRNA(45%3
52%) and UMUC3-AR-shRNA
(46%356%) but not in TCCSUP-GR-
shRNA (44%346%) and UMUC3-
GR-shRNA (43%347%) (Figure 2A).
The effects of CpdA on apoptosis were
then assessed in the parental UMUC3/
TCCSUP (Figure 2B) and stable
UMUC3-control/GR/AR-shRNA (Fig-
ure 2C) cell lines. CpdA treatment for
Figure 2. Effects of CpdA on cell cycle and apoptosis. A, Flow cytometry in TCCSUP/UMUC3-
control/GR/AR-shRNA cell lines cultured with ethanol (mock) or CpdA (1␮M) for 24 hours.
Representative analyses and the percentages of cells in G1 phase are shown. TUNEL assay in
TCCSUP/UMUC3 cell lines cultured with ethanol (mock) or increasing concentrations of CpdA
(0.1␮M to 1␮M) for 48 hours (B) and UMUC3-control/GR/AR-shRNA cell lines cultured with
ethanol (mock), CpdA (1␮M), and/or DEX (100nM) for 48 hours (C). The percentages of TUNEL-
positive cells were counted under a fluorescence microscopy. Each value represents the mean ϩ
SD from at least 3 independent experiments. *, P Ͻ .05 (vs mock treatment in parental or each
control-shRNA cell line); **, P Ͻ .01 (vs mock treatment in parental or each control-shRNA cell
line).
1490 Zheng et al CpdA Treatment in Bladder Cancer Cells Mol Endocrinol, October 2015, 29(10):1486–1497
Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
48hoursincreasedapoptoticindicesinadose-dependentman-
ner (up to 12.5% and 11.8% in TCCSUP and UMUC3,
respectively). In accordance with our previous findings
(2), in UMUC3-control-shRNA cells, DEX consider-
ably reduced apoptosis from 2.1% to 1.2% (P ϭ .045).
In contrast, CpdA significantly increased apoptosis in
UMUC3-control-shRNA (2.1%312.2%), UMUC3-
GR-shRNA (2.0%35.2%), and UMUC3-AR-shRNA
(2.0%38.8%) cells. Although DEX was shown to in-
hibit apoptotic cell death of UMUC3 induced by cis-
platin (2, 5), it failed to considerably block that in-
duced by CpdA (12.2% vs 11.5%).
Suppressive effects of CpdA on bladder cancer cell
migration and invasion
A scratch wound healing assay and a transwell inva-
sion assay were performed to assess the effects of CpdA
on cell migration and invasion, re-
spectively, in bladder cancer lines. In
the wound healing assay (Figure
3A), CpdA more significantly inhib-
ited wound closure in TCCSUP-
control-shRNA (47% decrease) or
UMUC3-control-shRNA (47% de-
crease) as well as TCCSUP-AR-
shRNA (43% decrease) or UMUC3-
AR-shRNA (29% decrease), compared
with TCCSUP-GR-shRNA (16%
decrease) or UMUC3-GR-shRNA
(11% decrease). Moreover, GR
knockdown resulted in a significant
increase (23% vs control line) in cell
migration of TCCSUP, suggesting
an inhibitory role of GR in cell mi-
gration, although the increase (5%)
in UMUC3 cells was not significant.
Similarly, in the transwell assay (Fig-
ure 3B), CpdA treatment demon-
strated significant decreases in the in-
vasive properties of control-shRNA
(61% decrease), GR-shRNA (35%
decrease), and AR-shRNA (51% de-
crease) cells. In addition, in mock-
treated cells, GR (9% increase)
or AR (4% decrease) knockdown
did not significantly change their
invasion.
Antitumor activity of CpdA in
mouse xenograft models for
bladder cancer
We used mouse xenograft models
to investigate whether CpdA inhib-
its bladder tumor growth in vivo, in comparison with
treatment with DEX or a clinically used AR antagonist
HF. UMUC3-control-shRNA (Figure 4A), UMUC3-GR-
shRNA (Figure 4B), or UMUC3-AR-shRNA (Figure 4C)
cells were implanted sc into the flank of SCID male mice,
and, after 2–4 weeks, we commenced injections of DEX,
CpdA, or HF. Consistent with our previous findings (2),
DEX failed to significantly inhibit the growth of control-
shRNA (4%–15% decreases), GR-shRNA (up to 25%
increase or up to 10% decrease), or AR-shRNA (up to 3%
increase or up to 18% decrease) xenografts (P Ͼ .10). HF
similarly inhibited the growth of control-shRNA (up to
39% decrease) and GR-shRNA (up to 45% decrease)
xenografts, but not that of AR-shRNA xenografts (up to
16% decrease). In accordance with our in vitro data,
CpdA reduced the size of control-shRNA xenografts
Figure 3. Effects of CpdA on cell migration and invasion. Wound healing assay in TCCSUP/
UMUC3-control/GR/AR-shRNA cell lines (A). The cells grown to confluence were gently
scratched, and the wound area was measured after a 24-hour culture with ethanol (mock) or
CpdA (1␮M). The migration determined by the rate of cells filling the wound area is presented
relative to that of mock treatment in each control-shRNA cell line. Transwell invasion assay in
UMUC3-control/GR/AR-shRNA cell lines (B). The cells were cultured in the Matrigel-coated
transwell chamber for 36 hours in the presence of ethanol (mock) or CpdA (1␮M). The number
of invaded cells present in the lower chamber was counted under a light microscope (ϫ100
objective in 5 random fields). Cell invasion is presented relative to that of mock treatment in
control-shRNA cell line. Each value represents the mean ϩ SD from 3 independent experiments.
*, P Ͻ .05 (mock vs CpdA treatment in each cell line); **, P Ͻ .01 (mock vs CpdA treatment in
each cell line); #, P Ͻ .05 (vs mock treatment in control-shRNA cell line).
doi: 10.1210/me.2015-1128 press.endocrine.org/journal/mend 1491
Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
much more strongly (up to 65% decrease) and that of
GR-shRNA xenografts similarly (up to 50% decrease),
compared with HF. In addition, CpdA inhibited the
growth of AR-shRNA xenografts (up to 45% decrease).
Nonsignificant effects of CpdA on GR
transactivation
As is the case with other steroid hormone receptors,
GR, upon binding to GCs, is known to induce GC-re-
sponse element (GRE)-mediated gene transcription
(“transactivation”) (21–23). Therefore, we next studied
the effects of CpdA on GR transactivation in bladder
cancer cells. GR-mediated transcriptional activity was de-
termined in the cell extracts with transfection of a Luc
reporter plasmid (MMTV-Luc) and treatment with CpdA
or DEX. Similar to our previous findings (2), 100nM
DEX significantly increased Luc activity in UMUC3-con-
trol-shRNA (16.4-fold) and UMUC3-AR-shRNA (15.7-
fold) but not in UMUC3-GR-shRNA (1.9-fold), com-
pared with respective mock treatments (Figure 5A). In
contrast, CpdA even at 10␮M showed nonsignificant ef-
fects (up to 1.6-fold increase) on GR transactivation in
these 3 sublines.
Figure 5. Effects of CpdA on GR transactivation. A, UMUC3-control/
GR/AR-shRNA cell lines were cotransfected with MMTV-Luc and pRL-
TK and subsequently cultured with ethanol (mock), DEX (100nM), or
CpdA (1␮M or 10␮M) for 24 hours. Luc activity is presented relative to
that with mock treatment in each cell line. TCCSUP/UMUC3 cell lines
cultured with ethanol (mock), DEX (100nM), or CpdA (1␮M) for 24
hours were subjected to RNA extraction and subsequent real-time RT-
PCR for GILZ (B) and FKBP51 (C). Expression of each specific gene was
normalized to that of GAPDH. Transcription amount is presented
relative to that of mock treatment in each cell line. Each value
represents the mean ϩ SD from at least 3 independent experiments. *,
P Ͻ .05 (vs mock treatment in each cell line); **, P Ͻ .01 (vs mock
treatment in each cell line).
Figure 4. Effects of CpdA on tumor growth in mouse xenograft
models for bladder cancer. CpdA, DEX, HF, or vehicle control was
injected sc at peritumor sites in UMUC3-control-shRNA (A), UMUC3-
GR-shRNA (B), or UMUC3-AR-shRNA (C) bearing NOD-SICD male mice
(n ϭ 5 per each condition). Tumor size was monitored every other
days. Each value represents the mean of estimated tumor volume.
*, P Ͻ .05 (mock vs CpdA); #, P Ͻ .05 (DEX vs CpdA); ϩ, P Ͻ .05 (HF
vs CpdA).
1492 Zheng et al CpdA Treatment in Bladder Cancer Cells Mol Endocrinol, October 2015, 29(10):1486–1497
Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
We further assessed the effects of CpdA on the expres-
sion of 2 canonical targets of GR transcription, GILZ (23,
24) and FKBP51 (25, 26), using a quantitative RT-PCR.
As expected, DEX significantly augmented the levels of
GILZ (Figure 5B) and FKBP51 (Figure 5C) expression in
GR-positive bladder cancer lines, whereas CpdA failed to
induce them.
Induction of GR transrepression by CpdA
GC-activated GR monomers are also known to inter-
act with other transcription factors, such NF-␬B and
AP-1, and subsequently regulate their transcriptional ac-
tivities (“transrepression”) (22, 23, 27). Indeed, in pros-
tate cancer cells, CpdA was shown to induce GR transre-
pression (28). We therefore investigated the ability of
CpdA to affect GR-mediated transrepression in bladder
cancer cells. NF-␬B and AP-1 transcriptional activities
were first measured by Luc assay. DEX reduced NF-␬B
transcriptional activity by 22%, compared with mock
treatment, and 1␮M–10␮M CpdA reduced it by 27%–
37% (Figure 6A). Similarly, DEX (15% decrease) and
CpdA (26%–36% decreases) inhibited AP-1 transcrip-
tion (Figure 6B).
To confirm the suppressive effects of CpdA on NF-␬B
activity, we determined the expression levels of NF-␬B
regulated genes that are also known to contribute to tu-
mor invasion and metastasis, including MMP-2, MMP-9,
IL-6, and VEGF, in bladder cancer cells treated with
CpdA. In accordance with the results of the Luc and trans-
well assays, CpdA considerably reduced their levels in
TCCSUP cells (Figure 6C) as well as in UMUC3-control-
shRNA and UMUC3-AR-shRNA cells but not in
UMUC3-GR-shRNA cells (Figure 6D).
Coimmunoprecipitation assay and Western blot anal-
ysis were then performed to determine whether CpdA
could have an influence on the interaction between GR
and NF-␬B as well as their expression. Coimmunoprecipi-
tation showed that both CpdA and DEX induced the in-
teraction between GR and NF-␬B (Figure 7A). There were
no significant changes in GR or NF-␬B expression in
mock- vs CpdA-treated TCCSUP/UMUC3, whereas as
we showed previously (2), DEX decreased the levels of
GR but not NF-␬B (Figure 7B).
Figure 6. Effects of CpdA on GR transrepression. UMUC3 cells were cotransfected with NF-␬B-Luc (A)/AP-1-Luc (B) and pRL-TK and subsequently
cultured with ethanol (mock), DEX (100nM), or CpdA (1␮M or 10␮M) for 24 hours. Luc activity is presented relative to that with mock treatment.
TCCSUP (C) and UMUC3-control/GR/AR-shRNA (D) cell lines cultured with ethanol (mock) or CpdA (1␮M) for 24 hours were subjected to RNA
extraction and subsequent real-time RT-PCR for MMP-2, MMP-9, IL-6, and VEGF. Expression of each specific gene was normalized to that of
GAPDH and ␤-actin. Transcription amount is presented relative to that of mock treatment in TCCSUP or UMUC3-control-shRNA. Each value
represents the mean ϩ SD from at least 3 independent experiments. *, P Ͻ .05 (vs mock treatment in each cell line); **, P Ͻ .01 (vs mock
treatment in each cell line).
Figure 7. CpdA-induced interaction between NF-␬B and GR. A, Cell
lysates from UMUC3/TCCSUP cultured with ethanol (mock), 1␮M
CpdA, or 100nM DEX for 24 hours were immunoprecipitated with an
anti-NF-␬B antibody or IgG and then immunoblotted for GR. B,
TCCSUP/UMUC3 cell lines cultured with ethanol (mock), 1␮M CpdA,
or 10nM DEX for 24 hours were analyzed on Western blotting, using
an antibody to GR (95 kDa) or NF-␬B (65 kDa). GAPDH (37 kDa) served
as an internal control.
doi: 10.1210/me.2015-1128 press.endocrine.org/journal/mend 1493
Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
Antiandrogenic effects of CpdA
Using a reporter gene assay, antiandrogenic activity of
CpdA was assessed in bladder cancer cells. AR-mediated
transcriptional activity was determined in the cell extracts
with transfection of MMTV-Luc, as used for GR trans-
activation, and treatment with CpdA and/or a potent an-
drogen DHT. In UMUC3-GR-shRNA cells, DHT in-
creased Luc activity, and CpdA as well as an antiandrogen
HF, showing nonsignificant agonist activity, could block
DHT-induced AR transcription (Figure 8A). Subcellular
localization of AR was also examined in UMUC3 cells
treated with CpdA and/or DHT by Western blotting.
CpdA failed to prevent DHT-induced AR nuclear trans-
location (Figure 8B). However, CpdA by itself did not
induce nuclear translocation of endogenous AR in blad-
der cancer cells.
Discussion
Several GCs, such as DEX, prednisone, and hydrocorti-
sone, have clinically been used as cytotoxic agents, pre-
dominantly for lymphomas and castration-resistant pros-
tate cancer (29). GCs are also known to reduce acute
toxicity of other cytotoxic drugs, particularly hypereme-
sis during systemic chemotherapy, as well as to protect
normal tissue against their long-term effects (30). As a
result, GCs are often prescribed as comedication, without
expecting their antitumor activities, in patients with solid
tumor, including bladder cancer. We have recently dem-
onstrated that DEX strongly suppresses GR-positive
bladder cancer cell invasion and metastasis in vitro and in
vivo (2). Nonetheless, DEX was found to promote blad-
der cancer cell proliferation despite its induction of cell-
cycle arrest at G1 phase and inhibited antiproliferative
effects of cisplatin via prevention of apoptosis (2, 4). Sim-
ilar findings have been reported in other types of malig-
nancies, suggesting that DEX can
reduce the sensitivity of chemother-
apeutic agents (31–33). More re-
cently, we have found that cortico-
sterone and prednisone suppress
bladder cancer cell invasion without
promoting cell proliferation or re-
ducing cisplatin cytotoxicity (5).
Thus, in conjunction with the results
in our immunohistochemical studies
(2, 3), GC-mediated GR activation
is likely to associate with bladder
cancer regression. However, no GR
ligands have been shown to signifi-
cantly reduce the viability of bladder
cancer cells. In the current study, we
demonstrate that a dual GR/AR modulator CpdA
strongly inhibits bladder cancer cell proliferation, via in-
creasing both G1 phase population and apoptosis, as well
as its migration and invasion.
The action of GCs is often complex and is generally
dependent on a balance between transactivation and tran-
srepression of GR (23, 34). Therapeutic effects of GCs are
thought to be due to transrepression, whereas adverse
effects associated with GC therapy are often induced by
transactivation. However, none of natural or synthetic
GCs have been shown to produce only the beneficial
changes via transrepression without the negative effects
resulting from transactivation (34). Recent advances in
drug design and compound screening have enabled the
identification of “dissociated” GR ligands that selectively
modulate GR functions presumably via altering GR struc-
ture which is favorable for transrepression over transac-
tivation (23, 35). CpdA, with a GR binding affinity sim-
ilar to that of DEX (17), is such a dissociated compound
isolated from natural sources. Remarkably, the literature
data indicated that CpdA was unable to induce GR trans-
activation. For instance, no significant increase in the ex-
pression of a GR-dependent target FKBP51 was seen in
leukemia cells (26). We confirmed this in bladder cancer
cells by showing that CpdA did not enhance GRE reporter
activity as well as the expression of GILZ and FKBP51
genes. Previous investigation of molecular mechanisms
underlying the effect of CpdA further revealed that the
lack of transactivation by CpdA on GRE-driven promot-
ers correlated with its inability to provoke ligand-medi-
ated GR dimerization as well as GR phosphorylation at a
specific residue (eg, Ser211
) (18). Based on these findings,
it is anticipated that treatment with CpdA in vivo is asso-
ciated with fewer GC-induced side effects.
Again, CpdA is known to preferentially induce GR-
mediated transrepression the major mechanism of which
Figure 8. Effects of CpdA on AR. A, UMUC3-GR-shRNA cells were cotransfected with MMTV-
Luc and pRL-TK and subsequently cultured with ethanol (mock), DHT (1nM), CpdA (1␮M), and/or
HF (5␮M) for 24 hours. Luc activity is presented relative to that with mock treatment. Each value
represents the mean ϩ SD from at least 3 independent experiments. *, P Ͻ .05 (vs mock
treatment); #, P Ͻ .05 (vs DHT treatment). B, Nuclear protein fractions from UMUC3 cells
cultured with ethanol (mock), 1nM DHT, and/or 1␮M CpdA for 24 hours were analyzed on
Western blotting, using an antibody to AR (110 kDa). Histone 3 (15 kDa) served as an internal
control.
1494 Zheng et al CpdA Treatment in Bladder Cancer Cells Mol Endocrinol, October 2015, 29(10):1486–1497
Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
is believed to be the ability of the receptor to inhibit the
activity of other transcription factors, including NF-␬B
and AP-1. In prostate cancer cells, not only CpdA (28) but
also DEX (36) reduced their growth mainly via inhibiting
NF-␬B activation. We also previously demonstrated the
data suggesting that inactivation of NF-␬B and inhibition
of the production of NF-␬B-dependent cytokines, such as
IL-6, might be a central mechanism involved in DEX/GR-
mediated suppression of bladder cancer cell invasion (2).
Similarly, in the present study, we showed that CpdA
inhibited transcriptional activities of NF-␬B and AP-1 as
well as the expression of NF-␬B-regulated genes, includ-
ing MMP-2, MMP-9, IL-6, and VEGF, in GR-positive
cells. CpdA could also induce the interaction between GR
and NF-␬B. It is thus likely that CpdA treatment leads to
a shift of GR functions toward transrepression in bladder
cancer cells. However, the reduction of MMP-2/MMP-9/
IL-6/VEGF gene expression by CpdA might simply rep-
resent its inhibitory effect on cell invasion rather than GR
transrepression, although nonsignificant and significant
inhibitions of these genes’ expression (Figure 6D) and cell
invasion (Figure 3B), respectively, were seen in CpdA-
treated AR-positive/GR knockdown UMUC3. Androgen
deprivation has been shown to inhibit the expression of
cell invasion-related genes, such as MMP-9 and VEGF, in
bladder cancer cells (8, 14). Indeed, the expression levels
of these 4 genes were slightly lower in mock-treated
UMUC3-AR-shRNA than in mock-treated UMUC3-con-
trol-shRNA (12%–25% decreases) (Figure 6D). None-
theless, in the current transwell assay (Figure 3B), there
was no significant effect of AR knockdown on bladder
cancer cell invasion. Meanwhile, it was noteworthy that
the suppressive effects of CpdA on bladder cancer cell
invasion (eg, 61% decrease in UMUC3-control-shRNA)
were found to be even stronger than those of DEX, cor-
ticosterone, or prednisone in the transwell assays per-
formed under the same conditions (eg, 34%–50% de-
creases) (2, 5).
Cytotoxic and proapoptotic activities of CpdA have
been related to its effects on GR functions, such as its
expression and nuclear translocation, in addition to
transactivation and transrepression. Underlying mecha-
nisms for GC-induced apoptosis may also vary depending
on cell type. GC treatment has been shown to promote
apoptosis in several types of malignancies, including leu-
kemia, osteosarcoma, lung small cell carcinoma, and
prostate cancer (36, 37), whereas it inhibits apoptosis in
other types, such as breast cancer (38) and fibrosarcoma
(39). DEX treatment is also associated with inhibition of
apoptosis of bladder cancer cells in our previous (2, 5)
and current studies. It has been suggested that the levels of
GR expression and GR-mediated transactivation play an
important role in determining the promotion of apoptosis
vs survival in a cell-specific manner (37, 38). In our study,
DEX is found to induce both transactivation and transre-
pression of GR and reduces its expression in bladder can-
cer cells, whereas CpdA induces only GR transrepression
and does not modify GR expression. These differences
may have contributed to the distinct effects of DEX vs
CpdA as GR ligands on bladder cancer cell proliferation,
whereas both compounds similarly induce G1 arrest. Ad-
ditionally, down-regulation of GR expression by DEX
has been implicated in a limitation of GCs with long-term
use. In this respect, CpdA inducing no significant changes
in GR expression may also be superior to DEX.
CpdA has been found to antagonize androgen actions
via mechanisms similar to those for classical antiandro-
gens, such as flutamide (16, 28). Although CpdA inhibits
androgen-enhanced AR transactivation, it possesses par-
tial agonist activity and induces nuclear translocation of
AR in prostate cancer cells (28). Indeed, clinically used
antiandrogens, including flutamide and bicalutamide,
failed to prevent androgen-induced AR nuclear translo-
cation (40, 41). We demonstrated in bladder cancer cells
that CpdA restored DHT-induced AR transcription yet
did not affect DHT-mediated AR nuclear translocation.
However, in contrast to the observations in prostate can-
cer (28), CpdA appeared to have only nonsignificant ag-
onist activity in bladder cancer cells without inducing AR
nuclear translocation. Thus, CpdA was confirmed to act
as an AR antagonist in bladder cancer cells. More impor-
tantly, the inhibitory effects of CpdA on bladder cancer
growth were invariably stronger in control cells than in
AR-silencing cells stably expressing AR-shRNA and were
still seen in GR-silencing cells stably expressing GR-
shRNA. Because stable expression of GR-shRNA in blad-
der cancer cells does not completely knock down endog-
enous GR (2), there is a possibility that residual GR in the
cells may have contributed to the latter effect. Nonethe-
less, AR-mediated pathway is still thought to involve the
inhibition of bladder cancer cell proliferation, migration,
and invasion by CpdA, which further supports the role of
AR signals in bladder cancer progression.
Using mouse xenograft models for bladder cancer, we
showed in vivo evidence suggesting that CpdA inhibited
the progression of bladder cancer via both GR and AR
pathways. CpdA was also found to more significantly
suppress the growth of GR-positive and/or AR-positive
xenografts than DEX or HF. Our previous immunohisto-
chemical studies in bladder tissue microarrays revealed
that most (eg, 87%) of bladder cancers expressed the GR
(3), whereas AR positivity in tumors correlated with dis-
ease progression in patients with muscle-invasive bladder
cancer (13, 42). Therefore, CpdA has the potential to
doi: 10.1210/me.2015-1128 press.endocrine.org/journal/mend 1495
Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
efficiently regress tumor growth, especially in male pa-
tients with GR-positive/AR-positive bladder cancer
whose androgen levels are not low, and its effects may be
more beneficial than GCs/pure GR ligands or AR antag-
onists. However, the current study has compared the ef-
ficacy of CpdA with that of only one compound each (ie,
DEX and HF) in tumor growth. Moreover, we injected
each drug 3 times a week. Although no data appears to be
available about the half-life of each compound when in-
jected peritumorally in mice, potentially different half-
lives that are probably less than 48 hours may have af-
fected the results from our xenograft study.
In conclusion, CpdA was found to inhibit bladder can-
cer growth predominantly via inducing GR-mediated
transrepression as well as at least partially via inactivating
androgen-enhanced AR signals. The current data also
support previous observations indicating that bladder
cancer cells possess functional GR and AR. Further study
to extensively compare the antitumor activities of this
unique compound vs various GR ligands and AR antag-
onists in bladder cancer is required.
Acknowledgments
Address all correspondence and requests for reprints to: Hiroshi
Miyamoto, MD, PhD, The James Buchanan Brady Urological
Institute at the Johns Hopkins Hospital, 600 North Wolfe
Street, Marburg 148, Baltimore, MD 21287. E-mail:
hmiyamo1@jhmi.edu.
This work was supported part by the National Natural Sci-
ence Foundation of China Grant NFSC 81202022 and the Basic
Research Program of the Department of Education of Zhejiang
Province Grant Y201225369 (to Y.Z.).
Disclosure Summary: The authors have nothing to disclose.
References
1. Miyamoto H, Zheng Y, Izumi K. Nuclear hormone receptor signals
as new therapeutic targets for urothelial carcinoma. Curr Cancer
Drug Tar. 2012;12:14–22.
2. Zheng Y, Izumi K, Li Y, Ishiguro H, Miyamoto H. Contrary regu-
lation of bladder cancer cell proliferation and invasion by dexam-
ethasone-mediated glucocorticoid receptor signals. Mol Cancer
Ther. 2012;11:2621–2632.
3. Ishiguro H, Kawahara T, Zheng Y, Netto GJ, Miyamoto H. Re-
duced glucocorticoid receptor expression predicts bladder tumor
recurrence and progression. Am J Clin Pathol. 2014;142:157–164.
4. Zhang C, Wenger T, Mattern J, et al. Clinical and mechanistic
aspects of glucocorticoid-induced chemotherapy resistance in the
majority of solid tumors. Cancer Biol Ther. 2007;6:278–287.
5. Ishiguro H, Kawahara T, Zheng Y, Kashiwagi E, Li Y, Miyamoto
H. Differential regulation of bladder cancer growth by various glu-
cocorticoids: corticosterone and prednisone inhibit cell invasion
without promoting cell proliferation or reducing cisplatin cytotox-
icity. Cancer Chemother Pharmacol. 2014;74:249–255.
6. Li Y, Izumi K, Miyamoto H. The role of the androgen receptor in
the development and progression of bladder cancer. Jpn J Clin On-
col. 2012;42:569–577.
7. Johnson AM, O’Connell MJ, Miyamoto H, et al. Androgenic de-
pendence of exophytic tumor growth in a transgenic mouse model
of bladder cancer: a role for thrombospondin-1. BMC Urol. 2008;
8:7.
8. Wu JT, Han BM, Yu SQ, Wang HP, Xia SJ. Androgen receptor is a
potential therapeutic target for bladder cancer. Urology. 2010;75:
820–827.
9. Zheng Y, Izumi K, Yao JL, Miyamoto H. Dihydrotestosterone up-
regulates the expression of epidermal growth factor receptor and
ERBB2 in androgen receptor-positive bladder cancer cells. Endocr
Relat Cancer. 2011;18:451–464.
10. Overdevest JB, Knubel KH, Duex JE, et al. CD24 expression is
important in male urothelial tumorigenesis and metastasis in mice
and is androgen regulated. Proc Natl Acad Sci USA. 2012;109:
E3588–E3596.
11. Li Y, Zheng Y, Izumi K, et al. Androgen activates ␤-catenin signal-
ing in bladder cancer cells. Endocr Relat Cancer. 2013;20:293–
304.
12. Jing Y, Cui D, Guo W, et al. Activated androgen receptor promotes
bladder cancer metastasis via Slug mediated epithelial-mesenchy-
mal transition. Cancer Lett. 2014;348:135–145.
13. Kawahara T, Shareef HK, Aljarah AK, et al. ELK1 is up-regulated
by androgen in bladder cancer cells and promotes tumor progres-
sion. Oncotarget. Available at: http://www.impactjournals.com/
oncotarget/index.php?journalϭoncotarget&pageϭarticle&opϭ
view&path%5B%5Dϭ5007.
14. Miyamoto H, Yang Z, Chen YT, et al. Promotion of bladder cancer
development and progression by androgen receptor signals. J Natl
Cancer Inst. 2007;99:558–568.
15. Izumi K, Zheng Y, Li Y, Zaengle J, Miyamoto H. Epidermal growth
factor induces bladder cancer cell proliferation through activation
of the androgen receptor. Int J Oncol. 2012;41:1587–1592.
16. Louw A, Swart P. Salsola tuberculatiformis Botschantzev and an
aziridine precursor analog mediate the in vivo increase in free cor-
ticosterone and decrease in corticosteroid-binding globulin in fe-
male Wistar rats. Endocrinology. 1999;140:2044–2053.
17. Tanner TM, Verrijdt G, Rombauts W, Louw A, Hapgood JP,
Claessens F. Anti-androgenic properties of compound A, an analog
of a non-steroidal plant compound. Mol Cell Endocrinol. 2003;
201:155–164.
18. De Bosscher K, Vanden Berghe W, Beck IM, et al. A fully dissoci-
ated compound of plant origin for inflammatory gene repression.
Proc Natl Acad Sci USA. 2005;102:15827–15832.
19. Li Y, Ishiguro H, Kawahara T, Miyamoto Y, Izumi K, Miyamoto
H. GATA3 in the urinary bladder: suppression of neoplastictumori-
genesis and down-regulation by androgens. Am J Cancer Res. 2014;
4:461–473.
20. Kawahara T, Kashiwagi E, Ide H, et al. Cyclosporine A and tacroli-
mus inhibit bladder cancer growth through down-regulation of
NFATc1. Oncotarget. 2015;6:1582–1593.
21. Evans RM. The steroid and thyroid hormone receptor superfamily.
Science. 1988;240:889–895.
22. Zhou J, Cidlowski JA. The human glucocorticoid receptor: one
gene, multiple proteins and diverse responses. Steroids. 2005;70:
407–417.
23. Ratman D, Vanden Berghe W, Dejager L, et al. How glucocorticoid
receptors modulate the activity of other transcription factors: a
scope beyond tethering. Mol Cell Endocrinol. 2013;380:41–54.
24. Ayroldi E, Riccardi C. Glucocorticoid-induced leucine zipper
(GILZ): a new important mediator of glucocorticoid action. FASEB
J. 2009;23:3649–3658.
25. Kester HA, van der Leede BM, van der Saag PT, van der Burg B.
Novel progesterone target genes identified by an improved differ-
ential display technique suggest that progestin-induced growth in-
1496 Zheng et al CpdA Treatment in Bladder Cancer Cells Mol Endocrinol, October 2015, 29(10):1486–1497
Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
hibition of breast cancer cells coincides with enhancement of dif-
ferentiation. J Biol Chem. 1997;272:16637–16643.
26. Lesovaya EA, Yemelyanov AY, Kirsanov KI, Yakubovskaya MG,
Budunova IV. Antitumor effect of non-steroid glucocorticoid re-
ceptor ligand CpdA on leukemia cell lines CEM and K562. Bio-
chemistry. 2011;76:1242–1252.
27. De Bosscher K, Vanden Berghe W, Haegeman G. The interplay
between the glucocorticoid receptor and nuclear factor-␬B or acti-
vator protein-1: molecular mechanisms for gene repression. Endocr
Rev. 2003;24:488–522.
28. Yemelyanov A, Czwornog J, Gera L, Joshi S, Chatterto RT Jr,
Budunova I. Novel steroid receptor phyto-modulator compound A
inhibits growth and survival of prostate cancer cells. Cancer Res.
2008;68:4763–4773.
29. Ishiguro H, Kawahara T, Li Y, Miyamoto H. Anti-tumor activities
of dexamethasone. In: Sauvage A, Levy M, eds. Dexamethasone:
Therapeutic Uses, Mechanism of Action and Potential Side Effects.
New York, NY: Nova Science Publishers; 2013:117–135.
30. Basch E, Prestrud AA, Hesketh PJ, et al. Antiemetics: American
Society of Clinical Oncology clinical practice guideline update.
J Clin Oncol. 2011;29:4189–4198.
31. Qian YH, Xiao Q, Chen H, Xu J. Dexamethasone inhibits camp-
tothecin-induced apoptosis in C6-glioma via activation of Stat5/
Bcl-xL pathway. Biochim Biophys Acta. 2009;1793:764–771.
32. Chen YX, Wang Y, Fu CC, et al. Dexamethasone enhances cell
resistance to chemotherapy by increasing adhesion to extracellular
matrix in human ovarian cancer cells. Endocr Relat Cancer. 2010;
17:39–50.
33. Ge H, Ni S, Wang X, et al. Dexamethasone reduces sensitivity to
cisplatin by blunting p53-dependent cellular senescence in non-
small cell lung cancer. PLoS One. 2012;7:e51821.
34. Patel R, Williams-Dautovich J, Cummins CL. Minireview: new mo-
lecular mediators of glucocorticoid receptor activity in metabolic
tissues. Mol Endocrinol. 2014;28:999–1011.
35. Schäcke H, Berger M, Rehwinkel H, Asadullah K. Selective gluco-
corticoid receptor agonists (SEGRAs): novel ligands with an im-
proved therapeutic index. Mol Cell Endocrinol. 2007;275:109–
117.
36. Nishimura K, Nonomura N, Satoh E, et al. Potential mechanism for
the effects of dexamethasone on growth of androgen-independent
prostate cancer. J Natl Cancer Inst. 2001;93:1739–1746.
37. Schlossmacher G, Stevens A, White A. Glucocorticoid receptor-
mediated apoptosis: mechanisms of resistance in cancer cells. J En-
docrinol. 2011;211:17–25.
38. Aziz MH, Shen H, Maki CG. Glucocorticoid receptor activation
inhibits p53-induced apoptosis of MCF10Amyc cells via induction
of protein kinase C⑀. J Biol Chem. 2012;287:29825–29836.
39. Gascoyne DM, Kypta RM, Vivanco Md. Glucocorticoids inhibit
apoptosis during fibrosarcoma development by transcriptionally
activating Bcl-xL. J Biol Chem. 2003;278:18022–18029.
40. Masiello D, Cheng S, Bubley GJ, Lu ML, Balk SP. Bicalutamide
functions as an androgen receptor antagonist by assembly of a
transcriptionally inactive receptor. J Biol Chem. 2002;277:26321–
26326.
41. Kawahara T, Miyamoto H. Androgen receptor antagonists in the
treatment of prostate cancer. Clin Immunol Endocr Metab Drugs.
2014;1:11–19.
42. Miyamoto H, Yao JL, Chaux A, et al. Expression of androgen and
oestrogen receptors and its prognostic significance in urothelial
neoplasm of the urinary bladder. BJU Int. 2012;109:1716–1726.
doi: 10.1210/me.2015-1128 press.endocrine.org/journal/mend 1497
Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020

Mais conteúdo relacionado

Mais procurados

Functional p53 is required for rapid restoration of daunorubicin-induced lesi...
Functional p53 is required for rapid restoration of daunorubicin-induced lesi...Functional p53 is required for rapid restoration of daunorubicin-induced lesi...
Functional p53 is required for rapid restoration of daunorubicin-induced lesi...Enrique Moreno Gonzalez
 
Mi r 449b inhibits the migration and invasion of colorectal cancer cells thro...
Mi r 449b inhibits the migration and invasion of colorectal cancer cells thro...Mi r 449b inhibits the migration and invasion of colorectal cancer cells thro...
Mi r 449b inhibits the migration and invasion of colorectal cancer cells thro...Clinical Surgery Research Communications
 
Ultrasound reverses adriamycin resistance in non-small cell lung cancer via p...
Ultrasound reverses adriamycin resistance in non-small cell lung cancer via p...Ultrasound reverses adriamycin resistance in non-small cell lung cancer via p...
Ultrasound reverses adriamycin resistance in non-small cell lung cancer via p...Clinical Surgery Research Communications
 
Lnc rna h19 induces retinal müller cell apoptosis via mir-29b foxo4 axis in d...
Lnc rna h19 induces retinal müller cell apoptosis via mir-29b foxo4 axis in d...Lnc rna h19 induces retinal müller cell apoptosis via mir-29b foxo4 axis in d...
Lnc rna h19 induces retinal müller cell apoptosis via mir-29b foxo4 axis in d...Clinical Surgery Research Communications
 
Roles of circular rn as and their interactions with micro rnas in human disor...
Roles of circular rn as and their interactions with micro rnas in human disor...Roles of circular rn as and their interactions with micro rnas in human disor...
Roles of circular rn as and their interactions with micro rnas in human disor...Clinical Surgery Research Communications
 
Abnormal expression of Pygopus 2 correlates with a malignant phenotype in hum...
Abnormal expression of Pygopus 2 correlates with a malignant phenotype in hum...Abnormal expression of Pygopus 2 correlates with a malignant phenotype in hum...
Abnormal expression of Pygopus 2 correlates with a malignant phenotype in hum...Enrique Moreno Gonzalez
 
全人關懷獎的簡報
全人關懷獎的簡報全人關懷獎的簡報
全人關懷獎的簡報bgbgbg
 
Connexin-43 can delay early recurrence and metastasis in patients with hepati...
Connexin-43 can delay early recurrence and metastasis in patients with hepati...Connexin-43 can delay early recurrence and metastasis in patients with hepati...
Connexin-43 can delay early recurrence and metastasis in patients with hepati...Enrique Moreno Gonzalez
 
CXCR7 is induced by hypoxia and mediates glioma cell migration towards SDF-1a...
CXCR7 is induced by hypoxia and mediates glioma cell migration towards SDF-1a...CXCR7 is induced by hypoxia and mediates glioma cell migration towards SDF-1a...
CXCR7 is induced by hypoxia and mediates glioma cell migration towards SDF-1a...Enrique Moreno Gonzalez
 
Prognostic and clinicopathological significance of programmed death ligand 1 ...
Prognostic and clinicopathological significance of programmed death ligand 1 ...Prognostic and clinicopathological significance of programmed death ligand 1 ...
Prognostic and clinicopathological significance of programmed death ligand 1 ...Clinical Surgery Research Communications
 
Circ ldlrad3 regulates cell proliferation, migration and invasion of pancreat...
Circ ldlrad3 regulates cell proliferation, migration and invasion of pancreat...Circ ldlrad3 regulates cell proliferation, migration and invasion of pancreat...
Circ ldlrad3 regulates cell proliferation, migration and invasion of pancreat...Clinical Surgery Research Communications
 
ACUTE MYELOID LEUKEMIA
ACUTE MYELOID LEUKEMIAACUTE MYELOID LEUKEMIA
ACUTE MYELOID LEUKEMIAflasco_org
 
Casc15 promotes lens epithelial cell apoptosis in age related cataracts by re...
Casc15 promotes lens epithelial cell apoptosis in age related cataracts by re...Casc15 promotes lens epithelial cell apoptosis in age related cataracts by re...
Casc15 promotes lens epithelial cell apoptosis in age related cataracts by re...Clinical Surgery Research Communications
 
Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...
Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...
Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...Clinical Surgery Research Communications
 

Mais procurados (20)

Functional p53 is required for rapid restoration of daunorubicin-induced lesi...
Functional p53 is required for rapid restoration of daunorubicin-induced lesi...Functional p53 is required for rapid restoration of daunorubicin-induced lesi...
Functional p53 is required for rapid restoration of daunorubicin-induced lesi...
 
Mi r 449b inhibits the migration and invasion of colorectal cancer cells thro...
Mi r 449b inhibits the migration and invasion of colorectal cancer cells thro...Mi r 449b inhibits the migration and invasion of colorectal cancer cells thro...
Mi r 449b inhibits the migration and invasion of colorectal cancer cells thro...
 
Ultrasound reverses adriamycin resistance in non-small cell lung cancer via p...
Ultrasound reverses adriamycin resistance in non-small cell lung cancer via p...Ultrasound reverses adriamycin resistance in non-small cell lung cancer via p...
Ultrasound reverses adriamycin resistance in non-small cell lung cancer via p...
 
Lnc rna h19 induces retinal müller cell apoptosis via mir-29b foxo4 axis in d...
Lnc rna h19 induces retinal müller cell apoptosis via mir-29b foxo4 axis in d...Lnc rna h19 induces retinal müller cell apoptosis via mir-29b foxo4 axis in d...
Lnc rna h19 induces retinal müller cell apoptosis via mir-29b foxo4 axis in d...
 
Roles of circular rn as and their interactions with micro rnas in human disor...
Roles of circular rn as and their interactions with micro rnas in human disor...Roles of circular rn as and their interactions with micro rnas in human disor...
Roles of circular rn as and their interactions with micro rnas in human disor...
 
Abnormal expression of Pygopus 2 correlates with a malignant phenotype in hum...
Abnormal expression of Pygopus 2 correlates with a malignant phenotype in hum...Abnormal expression of Pygopus 2 correlates with a malignant phenotype in hum...
Abnormal expression of Pygopus 2 correlates with a malignant phenotype in hum...
 
全人關懷獎的簡報
全人關懷獎的簡報全人關懷獎的簡報
全人關懷獎的簡報
 
Connexin-43 can delay early recurrence and metastasis in patients with hepati...
Connexin-43 can delay early recurrence and metastasis in patients with hepati...Connexin-43 can delay early recurrence and metastasis in patients with hepati...
Connexin-43 can delay early recurrence and metastasis in patients with hepati...
 
CXCR7 is induced by hypoxia and mediates glioma cell migration towards SDF-1a...
CXCR7 is induced by hypoxia and mediates glioma cell migration towards SDF-1a...CXCR7 is induced by hypoxia and mediates glioma cell migration towards SDF-1a...
CXCR7 is induced by hypoxia and mediates glioma cell migration towards SDF-1a...
 
Prognostic and clinicopathological significance of programmed death ligand 1 ...
Prognostic and clinicopathological significance of programmed death ligand 1 ...Prognostic and clinicopathological significance of programmed death ligand 1 ...
Prognostic and clinicopathological significance of programmed death ligand 1 ...
 
Circ ldlrad3 regulates cell proliferation, migration and invasion of pancreat...
Circ ldlrad3 regulates cell proliferation, migration and invasion of pancreat...Circ ldlrad3 regulates cell proliferation, migration and invasion of pancreat...
Circ ldlrad3 regulates cell proliferation, migration and invasion of pancreat...
 
Suppress lung cancer progression via up regulation of linc rna-p21
Suppress lung cancer progression via up regulation of linc rna-p21Suppress lung cancer progression via up regulation of linc rna-p21
Suppress lung cancer progression via up regulation of linc rna-p21
 
ACUTE MYELOID LEUKEMIA
ACUTE MYELOID LEUKEMIAACUTE MYELOID LEUKEMIA
ACUTE MYELOID LEUKEMIA
 
Prolonged Simvastatin Treatment Provided a Decrease in Apoptotic, Inflammator...
Prolonged Simvastatin Treatment Provided a Decrease in Apoptotic, Inflammator...Prolonged Simvastatin Treatment Provided a Decrease in Apoptotic, Inflammator...
Prolonged Simvastatin Treatment Provided a Decrease in Apoptotic, Inflammator...
 
Casc15 promotes lens epithelial cell apoptosis in age related cataracts by re...
Casc15 promotes lens epithelial cell apoptosis in age related cataracts by re...Casc15 promotes lens epithelial cell apoptosis in age related cataracts by re...
Casc15 promotes lens epithelial cell apoptosis in age related cataracts by re...
 
Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...
Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...
Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...
 
Nrgastro.2012.208
Nrgastro.2012.208Nrgastro.2012.208
Nrgastro.2012.208
 
2014 Zorniak JNS
2014 Zorniak JNS2014 Zorniak JNS
2014 Zorniak JNS
 
1476-4598-2-38
1476-4598-2-381476-4598-2-38
1476-4598-2-38
 
Effect of miR-21 on Oral Squamous Cell Carcinoma Cell Proliferation and Apopt...
Effect of miR-21 on Oral Squamous Cell Carcinoma Cell Proliferation and Apopt...Effect of miR-21 on Oral Squamous Cell Carcinoma Cell Proliferation and Apopt...
Effect of miR-21 on Oral Squamous Cell Carcinoma Cell Proliferation and Apopt...
 

Semelhante a Mend1486

Cancer Res-2014-Chakraborty-3489-500
Cancer Res-2014-Chakraborty-3489-500Cancer Res-2014-Chakraborty-3489-500
Cancer Res-2014-Chakraborty-3489-500Rachel Stupay
 
Cancer Discovery-2014-Horwitz-730-43
Cancer Discovery-2014-Horwitz-730-43Cancer Discovery-2014-Horwitz-730-43
Cancer Discovery-2014-Horwitz-730-43Elad Horwitz
 
Cancer Immunol Res-2015-Manuel-2326-6066.CIR-14-0214 (3)
Cancer Immunol Res-2015-Manuel-2326-6066.CIR-14-0214 (3)Cancer Immunol Res-2015-Manuel-2326-6066.CIR-14-0214 (3)
Cancer Immunol Res-2015-Manuel-2326-6066.CIR-14-0214 (3)Melanie Lampa
 
Lnc rna pcat29 suppresses cell proliferation, invasion, and migration in rena...
Lnc rna pcat29 suppresses cell proliferation, invasion, and migration in rena...Lnc rna pcat29 suppresses cell proliferation, invasion, and migration in rena...
Lnc rna pcat29 suppresses cell proliferation, invasion, and migration in rena...Clinical Surgery Research Communications
 
Clin Cancer Res-1999-Chen-3583-93
Clin Cancer Res-1999-Chen-3583-93Clin Cancer Res-1999-Chen-3583-93
Clin Cancer Res-1999-Chen-3583-93PreveenRamamoorthy
 
Epigenetic regulation and role of metastasis suppressor genes in pancreatic d...
Epigenetic regulation and role of metastasis suppressor genes in pancreatic d...Epigenetic regulation and role of metastasis suppressor genes in pancreatic d...
Epigenetic regulation and role of metastasis suppressor genes in pancreatic d...Enrique Moreno Gonzalez
 
Overview of New Targets For Anti-tumor Drugs.pdf
Overview of New Targets For Anti-tumor Drugs.pdfOverview of New Targets For Anti-tumor Drugs.pdf
Overview of New Targets For Anti-tumor Drugs.pdfDoriaFang
 
Gastric Cancer and the Role of Hedgehog- Interacting Protein One as a Prognos...
Gastric Cancer and the Role of Hedgehog- Interacting Protein One as a Prognos...Gastric Cancer and the Role of Hedgehog- Interacting Protein One as a Prognos...
Gastric Cancer and the Role of Hedgehog- Interacting Protein One as a Prognos...CrimsonpublishersCancer
 
Molecular Biology of Esophageal and Gastric Cancer
Molecular Biology of Esophageal and Gastric CancerMolecular Biology of Esophageal and Gastric Cancer
Molecular Biology of Esophageal and Gastric CancerSiddharth Sreemahadevan
 
Low expression of N-myc downstream-regulated gene 2 in oesophageal squamous c...
Low expression of N-myc downstream-regulated gene 2 in oesophageal squamous c...Low expression of N-myc downstream-regulated gene 2 in oesophageal squamous c...
Low expression of N-myc downstream-regulated gene 2 in oesophageal squamous c...Enrique Moreno Gonzalez
 
Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...
Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...
Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...JohnJulie1
 

Semelhante a Mend1486 (17)

440
440440
440
 
Dihydrotanshinone I Induces Apoptosis Through Reactive Oxygen Species–Mediate...
Dihydrotanshinone I Induces Apoptosis Through Reactive Oxygen Species–Mediate...Dihydrotanshinone I Induces Apoptosis Through Reactive Oxygen Species–Mediate...
Dihydrotanshinone I Induces Apoptosis Through Reactive Oxygen Species–Mediate...
 
Cancer Res-2014-Chakraborty-3489-500
Cancer Res-2014-Chakraborty-3489-500Cancer Res-2014-Chakraborty-3489-500
Cancer Res-2014-Chakraborty-3489-500
 
Cancer Discovery-2014-Horwitz-730-43
Cancer Discovery-2014-Horwitz-730-43Cancer Discovery-2014-Horwitz-730-43
Cancer Discovery-2014-Horwitz-730-43
 
Cancer Immunol Res-2015-Manuel-2326-6066.CIR-14-0214 (3)
Cancer Immunol Res-2015-Manuel-2326-6066.CIR-14-0214 (3)Cancer Immunol Res-2015-Manuel-2326-6066.CIR-14-0214 (3)
Cancer Immunol Res-2015-Manuel-2326-6066.CIR-14-0214 (3)
 
Lnc rna pcat29 suppresses cell proliferation, invasion, and migration in rena...
Lnc rna pcat29 suppresses cell proliferation, invasion, and migration in rena...Lnc rna pcat29 suppresses cell proliferation, invasion, and migration in rena...
Lnc rna pcat29 suppresses cell proliferation, invasion, and migration in rena...
 
Mechanism of Jianpi Huayu Decoction Enhancing Dormancy of Liver Cancer by Inh...
Mechanism of Jianpi Huayu Decoction Enhancing Dormancy of Liver Cancer by Inh...Mechanism of Jianpi Huayu Decoction Enhancing Dormancy of Liver Cancer by Inh...
Mechanism of Jianpi Huayu Decoction Enhancing Dormancy of Liver Cancer by Inh...
 
B0211117
B0211117B0211117
B0211117
 
Clin Cancer Res-1999-Chen-3583-93
Clin Cancer Res-1999-Chen-3583-93Clin Cancer Res-1999-Chen-3583-93
Clin Cancer Res-1999-Chen-3583-93
 
Src kinases_Shhyam Moorthy.PDF
Src kinases_Shhyam Moorthy.PDFSrc kinases_Shhyam Moorthy.PDF
Src kinases_Shhyam Moorthy.PDF
 
Epigenetic regulation and role of metastasis suppressor genes in pancreatic d...
Epigenetic regulation and role of metastasis suppressor genes in pancreatic d...Epigenetic regulation and role of metastasis suppressor genes in pancreatic d...
Epigenetic regulation and role of metastasis suppressor genes in pancreatic d...
 
Overview of New Targets For Anti-tumor Drugs.pdf
Overview of New Targets For Anti-tumor Drugs.pdfOverview of New Targets For Anti-tumor Drugs.pdf
Overview of New Targets For Anti-tumor Drugs.pdf
 
Gastric Cancer and the Role of Hedgehog- Interacting Protein One as a Prognos...
Gastric Cancer and the Role of Hedgehog- Interacting Protein One as a Prognos...Gastric Cancer and the Role of Hedgehog- Interacting Protein One as a Prognos...
Gastric Cancer and the Role of Hedgehog- Interacting Protein One as a Prognos...
 
Molecular Biology of Esophageal and Gastric Cancer
Molecular Biology of Esophageal and Gastric CancerMolecular Biology of Esophageal and Gastric Cancer
Molecular Biology of Esophageal and Gastric Cancer
 
Low expression of N-myc downstream-regulated gene 2 in oesophageal squamous c...
Low expression of N-myc downstream-regulated gene 2 in oesophageal squamous c...Low expression of N-myc downstream-regulated gene 2 in oesophageal squamous c...
Low expression of N-myc downstream-regulated gene 2 in oesophageal squamous c...
 
Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...
Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...
Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...
 
Nrgastro.2013.52
Nrgastro.2013.52Nrgastro.2013.52
Nrgastro.2013.52
 

Mais de University of Zambia, School of Pharmacy, Lusaka, Zambia

Mais de University of Zambia, School of Pharmacy, Lusaka, Zambia (20)

7 biotechnology and human disease
7 biotechnology and human disease7 biotechnology and human disease
7 biotechnology and human disease
 
6 radiopharmaceutical systems
6 radiopharmaceutical systems6 radiopharmaceutical systems
6 radiopharmaceutical systems
 
4 preformulation
4 preformulation4 preformulation
4 preformulation
 
2 colloidal system
2 colloidal system2 colloidal system
2 colloidal system
 
1 general polymer science
1 general polymer science1 general polymer science
1 general polymer science
 
15 sedimentation
15 sedimentation15 sedimentation
15 sedimentation
 
15 lyophilization
15 lyophilization15 lyophilization
15 lyophilization
 
15 heat transfer
15 heat transfer15 heat transfer
15 heat transfer
 
15 extraction
15 extraction15 extraction
15 extraction
 
15 evaporation transpiration sublimation
15 evaporation transpiration sublimation15 evaporation transpiration sublimation
15 evaporation transpiration sublimation
 
15 distillation
15 distillation15 distillation
15 distillation
 
15 crystallization
15 crystallization15 crystallization
15 crystallization
 
15 coagulation and flocculation
15 coagulation and flocculation15 coagulation and flocculation
15 coagulation and flocculation
 
15 mixing
15 mixing15 mixing
15 mixing
 
15 filtration
15 filtration15 filtration
15 filtration
 
15 drying
15 drying15 drying
15 drying
 
15 communition
15 communition15 communition
15 communition
 
15 adsorption
15 adsorption15 adsorption
15 adsorption
 
14 rheology
14 rheology14 rheology
14 rheology
 
13 polymer science
13 polymer science13 polymer science
13 polymer science
 

Último

Call Girls Service In Goa 💋 9316020077💋 Goa Call Girls By Russian Call Girl...
Call Girls Service In Goa  💋 9316020077💋 Goa Call Girls  By Russian Call Girl...Call Girls Service In Goa  💋 9316020077💋 Goa Call Girls  By Russian Call Girl...
Call Girls Service In Goa 💋 9316020077💋 Goa Call Girls By Russian Call Girl...russian goa call girl and escorts service
 
Punjab❤️Call girls in Mohali ☎️7435815124☎️ Call Girl service in Mohali☎️ Moh...
Punjab❤️Call girls in Mohali ☎️7435815124☎️ Call Girl service in Mohali☎️ Moh...Punjab❤️Call girls in Mohali ☎️7435815124☎️ Call Girl service in Mohali☎️ Moh...
Punjab❤️Call girls in Mohali ☎️7435815124☎️ Call Girl service in Mohali☎️ Moh...Sheetaleventcompany
 
Local Housewife and effective ☎️ 8250192130 🍉🍓 Sexy Girls VIP Call Girls Chan...
Local Housewife and effective ☎️ 8250192130 🍉🍓 Sexy Girls VIP Call Girls Chan...Local Housewife and effective ☎️ 8250192130 🍉🍓 Sexy Girls VIP Call Girls Chan...
Local Housewife and effective ☎️ 8250192130 🍉🍓 Sexy Girls VIP Call Girls Chan...Russian Call Girls Amritsar
 
Call Girls Hyderabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Hyderabad Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Hyderabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Hyderabad Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Udaipur Call Girls 📲 9999965857 Call Girl in Udaipur
Udaipur Call Girls 📲 9999965857 Call Girl in UdaipurUdaipur Call Girls 📲 9999965857 Call Girl in Udaipur
Udaipur Call Girls 📲 9999965857 Call Girl in Udaipurseemahedar019
 
Dehradun Call Girls Service 08854095900 Real Russian Girls Looking Models
Dehradun Call Girls Service 08854095900 Real Russian Girls Looking ModelsDehradun Call Girls Service 08854095900 Real Russian Girls Looking Models
Dehradun Call Girls Service 08854095900 Real Russian Girls Looking Modelsindiancallgirl4rent
 
VIP Call Girl Sector 10 Noida Call Me: 9711199171
VIP Call Girl Sector 10 Noida Call Me: 9711199171VIP Call Girl Sector 10 Noida Call Me: 9711199171
VIP Call Girl Sector 10 Noida Call Me: 9711199171Call Girls Service Gurgaon
 
pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...
pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...
pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...Call Girls Noida
 
Call Girls Thane Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Thane Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Thane Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Thane Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Nanded Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Nanded Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real MeetNanded Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Nanded Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real MeetCall Girls Service
 
Vip sexy Call Girls Service In Sector 137,9999965857 Young Female Escorts Ser...
Vip sexy Call Girls Service In Sector 137,9999965857 Young Female Escorts Ser...Vip sexy Call Girls Service In Sector 137,9999965857 Young Female Escorts Ser...
Vip sexy Call Girls Service In Sector 137,9999965857 Young Female Escorts Ser...Call Girls Noida
 
❤️♀️@ Jaipur Call Girl Agency ❤️♀️@ Manjeet Russian Call Girls Service in Jai...
❤️♀️@ Jaipur Call Girl Agency ❤️♀️@ Manjeet Russian Call Girls Service in Jai...❤️♀️@ Jaipur Call Girl Agency ❤️♀️@ Manjeet Russian Call Girls Service in Jai...
❤️♀️@ Jaipur Call Girl Agency ❤️♀️@ Manjeet Russian Call Girls Service in Jai...Gfnyt.com
 
Krishnagiri call girls Tamil aunty 7877702510
Krishnagiri call girls Tamil aunty 7877702510Krishnagiri call girls Tamil aunty 7877702510
Krishnagiri call girls Tamil aunty 7877702510Vipesco
 
Call Girls Service Faridabad 📲 9999965857 ヅ10k NiGhT Call Girls In Faridabad
Call Girls Service Faridabad 📲 9999965857 ヅ10k NiGhT Call Girls In FaridabadCall Girls Service Faridabad 📲 9999965857 ヅ10k NiGhT Call Girls In Faridabad
Call Girls Service Faridabad 📲 9999965857 ヅ10k NiGhT Call Girls In Faridabadgragmanisha42
 
VIP Call Girls Sector 67 Gurgaon Just Call Me 9711199012
VIP Call Girls Sector 67 Gurgaon Just Call Me 9711199012VIP Call Girls Sector 67 Gurgaon Just Call Me 9711199012
VIP Call Girls Sector 67 Gurgaon Just Call Me 9711199012Call Girls Service Gurgaon
 
Call Girl Raipur 📲 9999965857 ヅ10k NiGhT Call Girls In Raipur
Call Girl Raipur 📲 9999965857 ヅ10k NiGhT Call Girls In RaipurCall Girl Raipur 📲 9999965857 ヅ10k NiGhT Call Girls In Raipur
Call Girl Raipur 📲 9999965857 ヅ10k NiGhT Call Girls In Raipurgragmanisha42
 
Call Girls Chandigarh 👙 7001035870 👙 Genuine WhatsApp Number for Real Meet
Call Girls Chandigarh 👙 7001035870 👙 Genuine WhatsApp Number for Real MeetCall Girls Chandigarh 👙 7001035870 👙 Genuine WhatsApp Number for Real Meet
Call Girls Chandigarh 👙 7001035870 👙 Genuine WhatsApp Number for Real Meetpriyashah722354
 
Ozhukarai Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Ozhukarai Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real MeetOzhukarai Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Ozhukarai Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real MeetCall Girls Service
 
(Sonam Bajaj) Call Girl in Jaipur- 09257276172 Escorts Service 50% Off with C...
(Sonam Bajaj) Call Girl in Jaipur- 09257276172 Escorts Service 50% Off with C...(Sonam Bajaj) Call Girl in Jaipur- 09257276172 Escorts Service 50% Off with C...
(Sonam Bajaj) Call Girl in Jaipur- 09257276172 Escorts Service 50% Off with C...indiancallgirl4rent
 
Hubli Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Hubli Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real MeetHubli Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Hubli Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real MeetCall Girls Service
 

Último (20)

Call Girls Service In Goa 💋 9316020077💋 Goa Call Girls By Russian Call Girl...
Call Girls Service In Goa  💋 9316020077💋 Goa Call Girls  By Russian Call Girl...Call Girls Service In Goa  💋 9316020077💋 Goa Call Girls  By Russian Call Girl...
Call Girls Service In Goa 💋 9316020077💋 Goa Call Girls By Russian Call Girl...
 
Punjab❤️Call girls in Mohali ☎️7435815124☎️ Call Girl service in Mohali☎️ Moh...
Punjab❤️Call girls in Mohali ☎️7435815124☎️ Call Girl service in Mohali☎️ Moh...Punjab❤️Call girls in Mohali ☎️7435815124☎️ Call Girl service in Mohali☎️ Moh...
Punjab❤️Call girls in Mohali ☎️7435815124☎️ Call Girl service in Mohali☎️ Moh...
 
Local Housewife and effective ☎️ 8250192130 🍉🍓 Sexy Girls VIP Call Girls Chan...
Local Housewife and effective ☎️ 8250192130 🍉🍓 Sexy Girls VIP Call Girls Chan...Local Housewife and effective ☎️ 8250192130 🍉🍓 Sexy Girls VIP Call Girls Chan...
Local Housewife and effective ☎️ 8250192130 🍉🍓 Sexy Girls VIP Call Girls Chan...
 
Call Girls Hyderabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Hyderabad Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Hyderabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Hyderabad Just Call 9907093804 Top Class Call Girl Service Available
 
Udaipur Call Girls 📲 9999965857 Call Girl in Udaipur
Udaipur Call Girls 📲 9999965857 Call Girl in UdaipurUdaipur Call Girls 📲 9999965857 Call Girl in Udaipur
Udaipur Call Girls 📲 9999965857 Call Girl in Udaipur
 
Dehradun Call Girls Service 08854095900 Real Russian Girls Looking Models
Dehradun Call Girls Service 08854095900 Real Russian Girls Looking ModelsDehradun Call Girls Service 08854095900 Real Russian Girls Looking Models
Dehradun Call Girls Service 08854095900 Real Russian Girls Looking Models
 
VIP Call Girl Sector 10 Noida Call Me: 9711199171
VIP Call Girl Sector 10 Noida Call Me: 9711199171VIP Call Girl Sector 10 Noida Call Me: 9711199171
VIP Call Girl Sector 10 Noida Call Me: 9711199171
 
pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...
pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...
pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...
 
Call Girls Thane Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Thane Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Thane Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Thane Just Call 9907093804 Top Class Call Girl Service Available
 
Nanded Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Nanded Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real MeetNanded Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Nanded Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
 
Vip sexy Call Girls Service In Sector 137,9999965857 Young Female Escorts Ser...
Vip sexy Call Girls Service In Sector 137,9999965857 Young Female Escorts Ser...Vip sexy Call Girls Service In Sector 137,9999965857 Young Female Escorts Ser...
Vip sexy Call Girls Service In Sector 137,9999965857 Young Female Escorts Ser...
 
❤️♀️@ Jaipur Call Girl Agency ❤️♀️@ Manjeet Russian Call Girls Service in Jai...
❤️♀️@ Jaipur Call Girl Agency ❤️♀️@ Manjeet Russian Call Girls Service in Jai...❤️♀️@ Jaipur Call Girl Agency ❤️♀️@ Manjeet Russian Call Girls Service in Jai...
❤️♀️@ Jaipur Call Girl Agency ❤️♀️@ Manjeet Russian Call Girls Service in Jai...
 
Krishnagiri call girls Tamil aunty 7877702510
Krishnagiri call girls Tamil aunty 7877702510Krishnagiri call girls Tamil aunty 7877702510
Krishnagiri call girls Tamil aunty 7877702510
 
Call Girls Service Faridabad 📲 9999965857 ヅ10k NiGhT Call Girls In Faridabad
Call Girls Service Faridabad 📲 9999965857 ヅ10k NiGhT Call Girls In FaridabadCall Girls Service Faridabad 📲 9999965857 ヅ10k NiGhT Call Girls In Faridabad
Call Girls Service Faridabad 📲 9999965857 ヅ10k NiGhT Call Girls In Faridabad
 
VIP Call Girls Sector 67 Gurgaon Just Call Me 9711199012
VIP Call Girls Sector 67 Gurgaon Just Call Me 9711199012VIP Call Girls Sector 67 Gurgaon Just Call Me 9711199012
VIP Call Girls Sector 67 Gurgaon Just Call Me 9711199012
 
Call Girl Raipur 📲 9999965857 ヅ10k NiGhT Call Girls In Raipur
Call Girl Raipur 📲 9999965857 ヅ10k NiGhT Call Girls In RaipurCall Girl Raipur 📲 9999965857 ヅ10k NiGhT Call Girls In Raipur
Call Girl Raipur 📲 9999965857 ヅ10k NiGhT Call Girls In Raipur
 
Call Girls Chandigarh 👙 7001035870 👙 Genuine WhatsApp Number for Real Meet
Call Girls Chandigarh 👙 7001035870 👙 Genuine WhatsApp Number for Real MeetCall Girls Chandigarh 👙 7001035870 👙 Genuine WhatsApp Number for Real Meet
Call Girls Chandigarh 👙 7001035870 👙 Genuine WhatsApp Number for Real Meet
 
Ozhukarai Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Ozhukarai Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real MeetOzhukarai Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Ozhukarai Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
 
(Sonam Bajaj) Call Girl in Jaipur- 09257276172 Escorts Service 50% Off with C...
(Sonam Bajaj) Call Girl in Jaipur- 09257276172 Escorts Service 50% Off with C...(Sonam Bajaj) Call Girl in Jaipur- 09257276172 Escorts Service 50% Off with C...
(Sonam Bajaj) Call Girl in Jaipur- 09257276172 Escorts Service 50% Off with C...
 
Hubli Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Hubli Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real MeetHubli Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Hubli Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
 

Mend1486

  • 1. Compound A Inhibits Bladder Cancer Growth Predominantly via Glucocorticoid Receptor Transrepression Yichun Zheng, Hitoshi Ishiguro, Hiroki Ide, Satoshi LungwaniInoue, Muungo, Eiji Kashiwagi, TakashiKawahara, Mehrsa Jalalizadeh, Leonardo O. Reis, and Hiroshi Miyamoto Department of Urology (Y.Z.), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; Departments of Pathology and Urology (Y.Z., H.Is., H.Id., L.M.,S.I., E.K.,T.K., M.J.,L.O.R., H.M.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; Department of Pathology and Laboratory Medicine (Y.Z., H.Is., T.K., H.M.), University of Rochester Medical Center, Rochester, New York 14642; and Photocatalyst Group (H.Is.), Kanagawa Academy of Science and Technology, Kawasaki 210-0821, Japan Recent evidence indicates that glucocorticoids (GCs) suppress bladder cancer cell invasion through the GC receptor (GR) pathway, whereas androgen-mediated androgen receptor (AR) signals in- duce bladder tumor progression. In this study, we assessed the effects of 2-(4-acetoxyphenyl)-2- chloro-N-methyl-ethylammonium chloride (compound A [CpdA]), which was shown to function as not only a GR modulator but also an AR antagonist, on the growth of bladder cancer. In GR/AR- positive cells, CpdA strongly inhibited cell proliferation and colony formation as well as increased G1 phase-arrested cell population and apoptosis. Specifically, CpdA at 1␮M decreased cell viability of TCCSUP/UMUC3-control-short hairpin RNA (shRNA), TCCSUP/UMUC3-GR-shRNA, and TCCSUP/ UMUC3-AR-shRNA by 50%/67%, 25%/26%, and 38%/58%, respectively. CpdA also inhibited cell migration and invasion of GR/AR-positive (up to 61% decrease) and GR-positive/AR-silencing (up to 51% decrease) lines and, less strongly, those of GR-silencing/AR-positive lines (up to 35% decrease). Additionally, in UMUC3-control xenograft-bearing male mice, CpdA more strongly suppressed tumor growth than dexamethasone or hydroxyflutamide. In reporter gene assays, CpdA failed to induce GR transactivation, whereas it antagonized dihydrotestosterone-enhanced AR transactivation. In contrast, CpdA reduced nuclear factor (NF)-␬B and activator protein 1 transcriptional activities, indicating induction of GR-mediated transrepression. Correspondingly, the expression of NF-␬B-related molecules, matrix metalloproteinase-2, matrix metalloprotei- nase-9, interleukin-6, and vascular endothelial growth factor, was significantly down-regulated by CpdA in control lines but not in GR-silencing cells. Moreover, coimmunoprecipitation showed that CpdA promoted the interactions between GR and NF-␬B. Thus, CpdA likely inhibits bladder cancer growth predominantly via inducing GR transrepression and at least partially mediated through the AR pathway, suggesting its effects more beneficial than GCs/pure GR ligands or AR antagonists. (Molecular Endocrinology 29: 1486–1497, 2015) Emerging preclinical evidence has indicated a pivotal role of steroid hormone receptor signals in the devel- opment and progression of urinary bladder cancer (1). Specifically, glucocorticoid (GC) receptor (GR) has been shown to function as a tumor suppressor in bladder can- cer. Our immunohistochemical studies revealed that GR ISSN Print 0888-8809 ISSN Online 1944-9917 Printed in USA Copyright © 2015 by the Endocrine Society Received May 12, 2015. Accepted August 27, 2015. First Published Online August 31, 2015 Abbreviations: AP-1, activator protein 1; AR, androgen receptor; CpdA, compound A; DEX, dexamethasone; DHT, dihydrotestosterone; FBS, fetal bovine serum; FKBP51, FK506-binding protein 51; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; GC, glucocorticoid; GILZ, GC-induced leucine zipper; GR, GC receptor; GRE, GC-response element; HF, hydroxyflutamide; Luc, luciferase; MMP, matrix metalloproteinase; MMTV, mouse mammary tumor virus; MTT, methylthiazolyldiphenyl-tetrazolium bromide; NF, nuclear factor; shRNA, short hairpin RNA; TUNEL, terminal deoxynucleotidyl transferase- mediated dUTP nick end labeling; VEGF, vascular endothelial growth factor. O R I G I N A L R E S E A R C H 1486 press.endocrine.org/journal/mend Mol Endocrinol, October 2015, 29(10):1486–1497 doi: 10.1210/me.2015-1128 Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
  • 2. expression was down-regulated in bladder tumors, com- pared with nonneoplastic urothelial tissues, as well as in high-grade/muscle-invasive tumors, compared with low- grade/nonmuscle-invasive tumors (2, 3). Additionally, patients with GR-positive muscle-invasive tumor were found to have a significantly lower risk of disease pro- gression. Using cell line and animal models, we demon- strated that a synthetic GC, dexamethasone (DEX), strongly inhibited bladder cancer cell invasion and the development of metastasis, presumably via inactivating nuclear factor (NF)-␬B and reversing epithelial-to-mesen- chymal transition (2). Thus, GC-mediated GR activity may generally correlate with bladder cancer regression. However, in contrast to its role in cell invasion and me- tastasis, DEX was found to induce bladder cancer cell proliferation. In particular, treatment with DEX resulted in significant reduction of apoptosis induced by a cyto- toxic agent cisplatin, suggesting that DEX could lead to chemotherapy resistance (2, 4). We then found that, of 10 natural or synthetic GCs examined, corticosterone and prednisone had no or insignificant, if any, stimulatory effects on bladder cancer cell proliferation, while show- ing inhibitory effects similar to those of DEX on cell invasion (5). We and others have also demonstrated that androgen- mediated androgen receptor (AR) signaling promotes bladder cancer progression (6). The molecules or path- ways found to be regulated by androgens/AR in bladder cancer cells include Bcl-xL, cyclin D1, epidermal growth factor receptor, ERBB2, AKT, ERK1/2, thrombospo- din-1, CD24, Wnt/␤-catenin, slug, and ELK1 (7–13). Consequently, treatment with antiandrogens or down- regulation of AR expression abolished the effects of an- drogens, resulting in inhibition of bladder cancer growth in vitro and in vivo (7–15). Compound A (CpdA) (2-(4-acetoxyphenyl)-2-chloro- N-methyl-ethylammonium chloride) is a synthetic analog of a hydroxyphenyl aziridine precursor found in the Na- mibian shrub Salsola tuberculatiformis Botschantzev (16). Interestingly, CpdA has been shown to have “dual” effects on steroid hormone receptor signals and functions as a GR ligand as well as an AR antagonist (17, 18). Accordingly, CpdA may exhibit an ideal effect on bladder cancer outgrowth and is expected to inhibit it more effi- ciently than GCs or currently available antiandrogens. In the current study, we aim to investigate the efficacy of CpdA in bladder cancer cell proliferation, migration, and invasion. Materials and Methods Cell culture and chemicals Human urothelial carcinoma cell lines, 5637, TCCSUP, and UMUC3, were originally obtained from the American Type Cul- ture Collection. 647V cells were used in our previous study (13, 14, 18, 19). These cells were immediately expanded after receipt and stored in liquid nitrogen and were not cultured for more than 5 months after resuscitation. Additionally, all these lines were recently authenticated, using GenePrint 10 System (Pro- mega), by the institutional core facility. Stable GR/AR knock- down lines and their control lines (TCCSUP-control-short hairpin RNA [shRNA]/GR-shRNA/AR-shRNA, UMUC3- control-shRNA/GR-shRNA/AR-shRNA) were previously es- tablished (2, 9). Cells were maintained in appropriate medium (RPMI 1640 for 5637 and DMEM for others; Mediatech) sup- plemented with 10% fetal bovine serum (FBS) and cultured in phenol red-free medium supplemented with 5% normal or char- coal-stripped FBS at least 24 hours before experimental treat- ment. We obtained DEX, CpdA, mifepristone (RU486), and dihydrotestosterone (DHT) from Sigma and hydroxyflutamide (HF) from Schering. Cell proliferation assay We used the methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay to assess cell viability. Cells (1 ϫ 103 /well) seeded in 96-well plates were incubated with medium supplemented with normal FBS in the presence or absence of DEX or CpdA. The media were refreshed every 48 hours. After 96 hours of treatment, we added 10 ␮L of MTT (Sigma) stock solution (5 mg/mL) to each well with 100 ␮L of medium for 4 hours at 37°C. We replaced the medium with 100 ␮L of dimethyl sul- foxide, followed by incubation for 5 minutes at room tempera- ture. The absorbance was then measured at a wavelength of 570 nm with background subtraction at 655 nm. Plate colony formation assay Cells (5 ϫ 102 /well) were seeded in 12-well culture plates and incubated in medium supplemented with normal FBS in the presence or absence of CpdA for 14 days at 37°C, followed by washing in PBS twice and staining with 0.1% crystal violet. The number of colonies containing more than or equal to 50 cells was counted under a light microscope, and the area of colonies was quantitated using the ImageJ software (National Institutes of Health). Flow cytometry Cells (1 ϫ 106 /10-cm dish) were cultured in medium supple- mented with normal FBS containing CpdA for 24 hours, har- vested with trypsin, fixed in 70% ethanol, and stained with propidium iodide buffer. Cellular propidium iodide content was measured on a Guava PCA-96 Base System flow cytometer (EMD Millipore). Data were analyzed using Guava Cell Cycle software (EMD Millipore). Apoptosis The terminal deoxynucleotidyl transferase-mediated de- oxyuridine triphosphase nick end labeling (TUNEL) assay was conducted on cell-burdening coverslips, using the DeadEnd Fluorometric TUNEL system (Promega), followed by counter- staining for DNA with 4Ј,6-diamidino-2-phenylindole. Apopto- tic index was determined in the cells visualized by the fluores- cence microscopy. doi: 10.1210/me.2015-1128 press.endocrine.org/journal/mend 1487 Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
  • 3. Cell migration assay Scratch wound healing assay was adapted to evaluate the ability of cell migration. Cells at a density of 80%–90% conflu- ence in 6-well tissue culture plates were scratched manually with a sterile 200-␮L plastic pipette tip, and rinsed with PBS to re- move floating cells and debris. The wounded monolayers of the cells were allowed to heal for 24 hours by culturing in the pres- ence or absence of CpdA. The width of the wound area was monitored with an inverted microscope, and the normalized cell-free area (24 h/0 h) was quantitated, using the ImageJ software. Cell invasion assay Cell invasiveness was determined, using a Matrigel (30 ␮g; BD Biosciences)-coated transwell chamber (5.0-␮m pore size polycarbonate filter with 6.5 mm in diameter; Corning). Cells (1 ϫ 105 ) in 100 ␮L of serum-free medium were added to the upper chamber of the transwell, whereas 600 ␮L of medium containing 5% normal FBS were added to the lower chamber. The media in both chambers contained ethanol or CpdA. After incubation for 36 hours at 37°C in a CO2 incubator, invaded cells were fixed, stained with 0.1% crystal violet, and counted under a light microscope. Mouse xenograft model Animal protocols in accordance with the National Institute of Health Guidelines for the Care and Use of Experimental Animals were approved at our institution. UMUC3-control- shRNA, UMUC3-GR-shRNA, or UMUC3-AR-shRNA (1 ϫ 106 cells/100 ␮L/site) resuspended in Matrigel (BD Biosciences) were sc injected into the flank of 6-week-old male immunocom- promised NOD-SCID mice, as described previously (2, 14, 20). Treatment was initiated when the estimated tumor volume reached 50–100 mm3 . Three times a week, each mice received a sc injection at peritumor site of ethanol (diluted 1:2000 in sterile saline), DEX (20 ␮g), CpdA (200 ␮g), or HF (20 ␮g). Serial caliper measurements of perpendicular diameters were used to calculate tumor volume by the next formula: (short diameter)2 ϫ (longest diameter) ϫ 0.5. Reporter gene assay Cells seeded in 24-well plates were cotransfected with 250 ng of a luciferase (Luc) reporter plasmid DNA, mouse mammary tumor virus (MMTV)-Luc (14), NF-␬B-Luc (Signosis), or activator pro- tein 1 (AP-1)-Luc (Signosis), and 2.5 ng of pRL-TK plasmid DNA, using GeneJuice (Novagen). After 6 hours of transfection, the cells were incubated in medium supplemented with charcoal-stripped FBS in the presence or absence of ligands (ie, DEX, DHT, CpdA, and HF) for 24 hours. The harvested cells were then assayed for Luc activity determined, using a Dual-Luciferase Reporter Assay kit (Promega) and luminometer. RT and real-time PCR Total RNA (0.5 ␮g) isolated from cultured cells, using TRI- zol (Invitrogen), was reverse transcribed using 1␮M oligo (dT) primers and 4 U of Ominiscript reverse transcriptase (QIAGEN) in a total volume of 20 ␮L. Real-time PCR was then carried out, using SYBR GreenER qPCR SuperMix for iCycler (Invitrogen), as described previously (2, 5). The next primer pairs were used for PCR: GC-induced leucine zipper (GILZ) (forward, 5Ј- AACACCGAAATGTATCAGACCC-3Ј; reverse, 5Ј-TGTC- CAGCTTAACGGAAACCA-3Ј), FK506-binding protein 51 (FKBP51) (forward, 5Ј-CTCCCTAAAATTCCCTCGAATGC- 3Ј; reverse, 5Ј-CCCTCTCCTTTCCGTTTGGTT-3Ј), matrix metalloproteinase (MMP)-2 (forward, 5Ј-TACAGGATCAT- TGGCTACACACC-3Ј; reverse, 5Ј-GGTCACATCGCTCCA- GACT-3Ј), MMP-9 (forward, 5Ј-TGTACCGCTATGGTTA- CACTCG-3Ј; reverse, 5Ј-GGCAGGGACAGTTGCTTCT-3Ј), IL-6 (forward, 5Ј-AAATTCGGTACATCCTCGACGG-3Ј; reverse, 5Ј-GGAAGGTTCAGGTTGTTTTCTGC-3Ј), and vas- cular endothelial growth factor (VEGF) (forward, 5Ј-CTG- TACCTCCACCATGCCAAG-3Ј; reverse, 5Ј-GGTACTCCT- GGAAGATGTCCACC-3Ј). Glyceraldehyde 3-phosphate dehy- drogenase (GAPDH) and ␤-actin were used as internal controls. Western blotting and coimmunoprecipitation Whole-cell protein extraction and Western blotting were conducted, as described previously (2, 9) with minor modifica- tions. The NE-PER Nuclear and Cytoplasmic Extraction kit (Thermo Scientific) was used for obtaining a nuclear fraction of proteins. Proteins (30 ␮g) were separated in 10% SDS-PAGE and transferred to polyvinylidene difluoride membrane (Im- mun-Blot PVDF Membrane; Bio-Rad). Specific antibody bind- ing was detected, using an anti-GR antibody (clone H-300, di- luted 1:1000; Santa Cruz Biotechnology, Inc), an anti-NF-␬B/ p65 antibody (clone F-6, diluted 1:1000; Santa Cruz Biotechnology, Inc), an anti-AR antibody (clone N20, diluted 1:2000; Santa Cruz Biotechnology, Inc), or an anti-GAPDH antibody (clone 6C5, diluted 1:1000; Santa Cruz Biotechnol- ogy, Inc), by scanning with an infrared imaging system (Odys- sey; LI-COR). For immunoprecipitation, whole-cell lysates in 500 ␮L were precleared with 15 ␮L of protein A/G beads (Santa Cruz Biotechnology, Inc) for 30 minutes at 4°C. After centrifug- ing, supernatants were incubated with an anti-NF-␬B/p65 anti- body (clone F-6) overnight at 4°C, followed by addition of 25-␮L A/G agarose beads for 2 hours. The beads were washed, and the protein complex was resolved on 10% SDS-PAGE, transferred to the membrane, and blotted with an anti-GR an- tibody (clone H-300). Statistical analyses Student’s t test or Mann-Whitney U test was used to assess differences in variables with a continuous distribution across dichotomous categories. P Ͻ .05 was considered statistically significant. Results Antiproliferative effects of CpdA in bladder cancer cells To see whether CpdA affects bladder cancer cell pro- liferation, 4 GR-positive human bladder cancer lines were treated with various concentrations of CpdA for 4 days, and cell viability was assessed by MTT assay. CpdA was found to reduce cell growth in a dose-dependent manner (eg, 52% and 65% decreases at 1␮M as well as 68% and 85% decreases at 10␮M in AR-negative 5637 and 647V 1488 Zheng et al CpdA Treatment in Bladder Cancer Cells Mol Endocrinol, October 2015, 29(10):1486–1497 Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
  • 4. lines, respectively, compared with mock treatment) (Fig- ure 1A). Similarly, CpdA decreased the growth of AR- positive TCCSUP and UMUC3 lines by 48% and 71% at 1␮M as well as by 91% and 96% at 10␮M, respectively (Figure 1B). We then assessed the effects of CpdA as well as DEX on the growth of stable cell lines expressing con- trol-shRNA, GR-shRNA, or AR-shRNA (Figure 1C). In control lines (GR-positive/AR-positive), 1␮M CpdA showed similar decreases in their viability as seen in the parental lines, whereas, consistent with our previous ob- servations (2, 5), 100nM DEX significantly increased it. The significant effects of DEX on cell growth were also seen in AR-shRNA lines (GR-positive/AR-silenced) but not in GR-shRNA lines (GR-silenced/AR-positive). In- Figure 1. Effects of CpdA on cell proliferation. MTT assay in 5637/647V cell lines cultured with ethanol (mock) or increasing concentrations of CpdA (1nM to 10␮M) for 96 hours (A), TCCSUP/UMUC3 cell lines cultured with ethanol (mock) or increasing concentrations of CpdA (0.2␮M to 10␮M) for 96 hours (B), and TCCSUP/UMUC3-control/GR/AR-shRNA cell lines cultured with ethanol (mock), DEX (100nM), CpdA (1␮M), and/or RU486 (RU) (10␮M) for 96 hours (C). Growth suppression is presented relative to that of mock treatment in each cell line. D, Clonogenic assay in UMUC3-control/GR/AR-shRNA cell lines cultured with ethanol (mock) or CpdA (1␮M) for 2 weeks. The number of colonies and their areas were quantitated, using the ImageJ software, and are presented relative to that of mock treatment. Each value represents the mean ϩ SD from at least 3 independent experiments. *, P Ͻ .05 (vs mock treatment in each cell line); **, P Ͻ .01 (vs mock treatment in each cell line). doi: 10.1210/me.2015-1128 press.endocrine.org/journal/mend 1489 Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
  • 5. terestingly, compared with TCCSUP- control-shRNA and UMUC3-con- trol-shRNA lines (50% and 67% decreases, respectively), slightly or considerably less strong inhibition of cell growth by CpdA was detected in TCCSUP-AR-shRNA and UMUC3- AR-shRNA (38% and 58% de- creases, respectively) or TCCSUP- GR-shRNA and UMUC3-GR- shRNA (25% and 26% decreases, respectively) lines. In addition, in AR- shRNA lines, growth promoting and inhibiting effects of DEX and CpdA, respectively, were abolished by a GR antagonist RU486 (Figure 1C), sug- gesting those mediated via the GR pathway. Moreover, in 5637 and 647V cells transiently expressing GR- shRNA (GR-silenced/AR-negative), CpdA did not significantly inhibit their growth (ie, up to 6% decrease) (data not shown). Plate colony formation assay was also performed to assess the antipro- liferative effect of CpdA (Figure 1D). Among 3 UMUC3-derived sublines, CpdA treatment most strongly reduced the number and area of colonies in con- trol cells (59% and 42% decreases, re- spectively), intermediately in AR- shRNAcells(52%and36%decreases), and least in GR-shRNA cells (31% and 22% decreases). To investigate how CpdA inhib- its cell proliferation, we performed flow cytometry and TUNEL assay. CpdA treatment for 24 hours led to significant increases in G1 phase cell population in TCCSUP-control- shRNA (45%353%) and UMUC3- control-shRNA (47%357%) as wellasinTCCSUP-AR-shRNA(45%3 52%) and UMUC3-AR-shRNA (46%356%) but not in TCCSUP-GR- shRNA (44%346%) and UMUC3- GR-shRNA (43%347%) (Figure 2A). The effects of CpdA on apoptosis were then assessed in the parental UMUC3/ TCCSUP (Figure 2B) and stable UMUC3-control/GR/AR-shRNA (Fig- ure 2C) cell lines. CpdA treatment for Figure 2. Effects of CpdA on cell cycle and apoptosis. A, Flow cytometry in TCCSUP/UMUC3- control/GR/AR-shRNA cell lines cultured with ethanol (mock) or CpdA (1␮M) for 24 hours. Representative analyses and the percentages of cells in G1 phase are shown. TUNEL assay in TCCSUP/UMUC3 cell lines cultured with ethanol (mock) or increasing concentrations of CpdA (0.1␮M to 1␮M) for 48 hours (B) and UMUC3-control/GR/AR-shRNA cell lines cultured with ethanol (mock), CpdA (1␮M), and/or DEX (100nM) for 48 hours (C). The percentages of TUNEL- positive cells were counted under a fluorescence microscopy. Each value represents the mean ϩ SD from at least 3 independent experiments. *, P Ͻ .05 (vs mock treatment in parental or each control-shRNA cell line); **, P Ͻ .01 (vs mock treatment in parental or each control-shRNA cell line). 1490 Zheng et al CpdA Treatment in Bladder Cancer Cells Mol Endocrinol, October 2015, 29(10):1486–1497 Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
  • 6. 48hoursincreasedapoptoticindicesinadose-dependentman- ner (up to 12.5% and 11.8% in TCCSUP and UMUC3, respectively). In accordance with our previous findings (2), in UMUC3-control-shRNA cells, DEX consider- ably reduced apoptosis from 2.1% to 1.2% (P ϭ .045). In contrast, CpdA significantly increased apoptosis in UMUC3-control-shRNA (2.1%312.2%), UMUC3- GR-shRNA (2.0%35.2%), and UMUC3-AR-shRNA (2.0%38.8%) cells. Although DEX was shown to in- hibit apoptotic cell death of UMUC3 induced by cis- platin (2, 5), it failed to considerably block that in- duced by CpdA (12.2% vs 11.5%). Suppressive effects of CpdA on bladder cancer cell migration and invasion A scratch wound healing assay and a transwell inva- sion assay were performed to assess the effects of CpdA on cell migration and invasion, re- spectively, in bladder cancer lines. In the wound healing assay (Figure 3A), CpdA more significantly inhib- ited wound closure in TCCSUP- control-shRNA (47% decrease) or UMUC3-control-shRNA (47% de- crease) as well as TCCSUP-AR- shRNA (43% decrease) or UMUC3- AR-shRNA (29% decrease), compared with TCCSUP-GR-shRNA (16% decrease) or UMUC3-GR-shRNA (11% decrease). Moreover, GR knockdown resulted in a significant increase (23% vs control line) in cell migration of TCCSUP, suggesting an inhibitory role of GR in cell mi- gration, although the increase (5%) in UMUC3 cells was not significant. Similarly, in the transwell assay (Fig- ure 3B), CpdA treatment demon- strated significant decreases in the in- vasive properties of control-shRNA (61% decrease), GR-shRNA (35% decrease), and AR-shRNA (51% de- crease) cells. In addition, in mock- treated cells, GR (9% increase) or AR (4% decrease) knockdown did not significantly change their invasion. Antitumor activity of CpdA in mouse xenograft models for bladder cancer We used mouse xenograft models to investigate whether CpdA inhib- its bladder tumor growth in vivo, in comparison with treatment with DEX or a clinically used AR antagonist HF. UMUC3-control-shRNA (Figure 4A), UMUC3-GR- shRNA (Figure 4B), or UMUC3-AR-shRNA (Figure 4C) cells were implanted sc into the flank of SCID male mice, and, after 2–4 weeks, we commenced injections of DEX, CpdA, or HF. Consistent with our previous findings (2), DEX failed to significantly inhibit the growth of control- shRNA (4%–15% decreases), GR-shRNA (up to 25% increase or up to 10% decrease), or AR-shRNA (up to 3% increase or up to 18% decrease) xenografts (P Ͼ .10). HF similarly inhibited the growth of control-shRNA (up to 39% decrease) and GR-shRNA (up to 45% decrease) xenografts, but not that of AR-shRNA xenografts (up to 16% decrease). In accordance with our in vitro data, CpdA reduced the size of control-shRNA xenografts Figure 3. Effects of CpdA on cell migration and invasion. Wound healing assay in TCCSUP/ UMUC3-control/GR/AR-shRNA cell lines (A). The cells grown to confluence were gently scratched, and the wound area was measured after a 24-hour culture with ethanol (mock) or CpdA (1␮M). The migration determined by the rate of cells filling the wound area is presented relative to that of mock treatment in each control-shRNA cell line. Transwell invasion assay in UMUC3-control/GR/AR-shRNA cell lines (B). The cells were cultured in the Matrigel-coated transwell chamber for 36 hours in the presence of ethanol (mock) or CpdA (1␮M). The number of invaded cells present in the lower chamber was counted under a light microscope (ϫ100 objective in 5 random fields). Cell invasion is presented relative to that of mock treatment in control-shRNA cell line. Each value represents the mean ϩ SD from 3 independent experiments. *, P Ͻ .05 (mock vs CpdA treatment in each cell line); **, P Ͻ .01 (mock vs CpdA treatment in each cell line); #, P Ͻ .05 (vs mock treatment in control-shRNA cell line). doi: 10.1210/me.2015-1128 press.endocrine.org/journal/mend 1491 Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
  • 7. much more strongly (up to 65% decrease) and that of GR-shRNA xenografts similarly (up to 50% decrease), compared with HF. In addition, CpdA inhibited the growth of AR-shRNA xenografts (up to 45% decrease). Nonsignificant effects of CpdA on GR transactivation As is the case with other steroid hormone receptors, GR, upon binding to GCs, is known to induce GC-re- sponse element (GRE)-mediated gene transcription (“transactivation”) (21–23). Therefore, we next studied the effects of CpdA on GR transactivation in bladder cancer cells. GR-mediated transcriptional activity was de- termined in the cell extracts with transfection of a Luc reporter plasmid (MMTV-Luc) and treatment with CpdA or DEX. Similar to our previous findings (2), 100nM DEX significantly increased Luc activity in UMUC3-con- trol-shRNA (16.4-fold) and UMUC3-AR-shRNA (15.7- fold) but not in UMUC3-GR-shRNA (1.9-fold), com- pared with respective mock treatments (Figure 5A). In contrast, CpdA even at 10␮M showed nonsignificant ef- fects (up to 1.6-fold increase) on GR transactivation in these 3 sublines. Figure 5. Effects of CpdA on GR transactivation. A, UMUC3-control/ GR/AR-shRNA cell lines were cotransfected with MMTV-Luc and pRL- TK and subsequently cultured with ethanol (mock), DEX (100nM), or CpdA (1␮M or 10␮M) for 24 hours. Luc activity is presented relative to that with mock treatment in each cell line. TCCSUP/UMUC3 cell lines cultured with ethanol (mock), DEX (100nM), or CpdA (1␮M) for 24 hours were subjected to RNA extraction and subsequent real-time RT- PCR for GILZ (B) and FKBP51 (C). Expression of each specific gene was normalized to that of GAPDH. Transcription amount is presented relative to that of mock treatment in each cell line. Each value represents the mean ϩ SD from at least 3 independent experiments. *, P Ͻ .05 (vs mock treatment in each cell line); **, P Ͻ .01 (vs mock treatment in each cell line). Figure 4. Effects of CpdA on tumor growth in mouse xenograft models for bladder cancer. CpdA, DEX, HF, or vehicle control was injected sc at peritumor sites in UMUC3-control-shRNA (A), UMUC3- GR-shRNA (B), or UMUC3-AR-shRNA (C) bearing NOD-SICD male mice (n ϭ 5 per each condition). Tumor size was monitored every other days. Each value represents the mean of estimated tumor volume. *, P Ͻ .05 (mock vs CpdA); #, P Ͻ .05 (DEX vs CpdA); ϩ, P Ͻ .05 (HF vs CpdA). 1492 Zheng et al CpdA Treatment in Bladder Cancer Cells Mol Endocrinol, October 2015, 29(10):1486–1497 Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
  • 8. We further assessed the effects of CpdA on the expres- sion of 2 canonical targets of GR transcription, GILZ (23, 24) and FKBP51 (25, 26), using a quantitative RT-PCR. As expected, DEX significantly augmented the levels of GILZ (Figure 5B) and FKBP51 (Figure 5C) expression in GR-positive bladder cancer lines, whereas CpdA failed to induce them. Induction of GR transrepression by CpdA GC-activated GR monomers are also known to inter- act with other transcription factors, such NF-␬B and AP-1, and subsequently regulate their transcriptional ac- tivities (“transrepression”) (22, 23, 27). Indeed, in pros- tate cancer cells, CpdA was shown to induce GR transre- pression (28). We therefore investigated the ability of CpdA to affect GR-mediated transrepression in bladder cancer cells. NF-␬B and AP-1 transcriptional activities were first measured by Luc assay. DEX reduced NF-␬B transcriptional activity by 22%, compared with mock treatment, and 1␮M–10␮M CpdA reduced it by 27%– 37% (Figure 6A). Similarly, DEX (15% decrease) and CpdA (26%–36% decreases) inhibited AP-1 transcrip- tion (Figure 6B). To confirm the suppressive effects of CpdA on NF-␬B activity, we determined the expression levels of NF-␬B regulated genes that are also known to contribute to tu- mor invasion and metastasis, including MMP-2, MMP-9, IL-6, and VEGF, in bladder cancer cells treated with CpdA. In accordance with the results of the Luc and trans- well assays, CpdA considerably reduced their levels in TCCSUP cells (Figure 6C) as well as in UMUC3-control- shRNA and UMUC3-AR-shRNA cells but not in UMUC3-GR-shRNA cells (Figure 6D). Coimmunoprecipitation assay and Western blot anal- ysis were then performed to determine whether CpdA could have an influence on the interaction between GR and NF-␬B as well as their expression. Coimmunoprecipi- tation showed that both CpdA and DEX induced the in- teraction between GR and NF-␬B (Figure 7A). There were no significant changes in GR or NF-␬B expression in mock- vs CpdA-treated TCCSUP/UMUC3, whereas as we showed previously (2), DEX decreased the levels of GR but not NF-␬B (Figure 7B). Figure 6. Effects of CpdA on GR transrepression. UMUC3 cells were cotransfected with NF-␬B-Luc (A)/AP-1-Luc (B) and pRL-TK and subsequently cultured with ethanol (mock), DEX (100nM), or CpdA (1␮M or 10␮M) for 24 hours. Luc activity is presented relative to that with mock treatment. TCCSUP (C) and UMUC3-control/GR/AR-shRNA (D) cell lines cultured with ethanol (mock) or CpdA (1␮M) for 24 hours were subjected to RNA extraction and subsequent real-time RT-PCR for MMP-2, MMP-9, IL-6, and VEGF. Expression of each specific gene was normalized to that of GAPDH and ␤-actin. Transcription amount is presented relative to that of mock treatment in TCCSUP or UMUC3-control-shRNA. Each value represents the mean ϩ SD from at least 3 independent experiments. *, P Ͻ .05 (vs mock treatment in each cell line); **, P Ͻ .01 (vs mock treatment in each cell line). Figure 7. CpdA-induced interaction between NF-␬B and GR. A, Cell lysates from UMUC3/TCCSUP cultured with ethanol (mock), 1␮M CpdA, or 100nM DEX for 24 hours were immunoprecipitated with an anti-NF-␬B antibody or IgG and then immunoblotted for GR. B, TCCSUP/UMUC3 cell lines cultured with ethanol (mock), 1␮M CpdA, or 10nM DEX for 24 hours were analyzed on Western blotting, using an antibody to GR (95 kDa) or NF-␬B (65 kDa). GAPDH (37 kDa) served as an internal control. doi: 10.1210/me.2015-1128 press.endocrine.org/journal/mend 1493 Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
  • 9. Antiandrogenic effects of CpdA Using a reporter gene assay, antiandrogenic activity of CpdA was assessed in bladder cancer cells. AR-mediated transcriptional activity was determined in the cell extracts with transfection of MMTV-Luc, as used for GR trans- activation, and treatment with CpdA and/or a potent an- drogen DHT. In UMUC3-GR-shRNA cells, DHT in- creased Luc activity, and CpdA as well as an antiandrogen HF, showing nonsignificant agonist activity, could block DHT-induced AR transcription (Figure 8A). Subcellular localization of AR was also examined in UMUC3 cells treated with CpdA and/or DHT by Western blotting. CpdA failed to prevent DHT-induced AR nuclear trans- location (Figure 8B). However, CpdA by itself did not induce nuclear translocation of endogenous AR in blad- der cancer cells. Discussion Several GCs, such as DEX, prednisone, and hydrocorti- sone, have clinically been used as cytotoxic agents, pre- dominantly for lymphomas and castration-resistant pros- tate cancer (29). GCs are also known to reduce acute toxicity of other cytotoxic drugs, particularly hypereme- sis during systemic chemotherapy, as well as to protect normal tissue against their long-term effects (30). As a result, GCs are often prescribed as comedication, without expecting their antitumor activities, in patients with solid tumor, including bladder cancer. We have recently dem- onstrated that DEX strongly suppresses GR-positive bladder cancer cell invasion and metastasis in vitro and in vivo (2). Nonetheless, DEX was found to promote blad- der cancer cell proliferation despite its induction of cell- cycle arrest at G1 phase and inhibited antiproliferative effects of cisplatin via prevention of apoptosis (2, 4). Sim- ilar findings have been reported in other types of malig- nancies, suggesting that DEX can reduce the sensitivity of chemother- apeutic agents (31–33). More re- cently, we have found that cortico- sterone and prednisone suppress bladder cancer cell invasion without promoting cell proliferation or re- ducing cisplatin cytotoxicity (5). Thus, in conjunction with the results in our immunohistochemical studies (2, 3), GC-mediated GR activation is likely to associate with bladder cancer regression. However, no GR ligands have been shown to signifi- cantly reduce the viability of bladder cancer cells. In the current study, we demonstrate that a dual GR/AR modulator CpdA strongly inhibits bladder cancer cell proliferation, via in- creasing both G1 phase population and apoptosis, as well as its migration and invasion. The action of GCs is often complex and is generally dependent on a balance between transactivation and tran- srepression of GR (23, 34). Therapeutic effects of GCs are thought to be due to transrepression, whereas adverse effects associated with GC therapy are often induced by transactivation. However, none of natural or synthetic GCs have been shown to produce only the beneficial changes via transrepression without the negative effects resulting from transactivation (34). Recent advances in drug design and compound screening have enabled the identification of “dissociated” GR ligands that selectively modulate GR functions presumably via altering GR struc- ture which is favorable for transrepression over transac- tivation (23, 35). CpdA, with a GR binding affinity sim- ilar to that of DEX (17), is such a dissociated compound isolated from natural sources. Remarkably, the literature data indicated that CpdA was unable to induce GR trans- activation. For instance, no significant increase in the ex- pression of a GR-dependent target FKBP51 was seen in leukemia cells (26). We confirmed this in bladder cancer cells by showing that CpdA did not enhance GRE reporter activity as well as the expression of GILZ and FKBP51 genes. Previous investigation of molecular mechanisms underlying the effect of CpdA further revealed that the lack of transactivation by CpdA on GRE-driven promot- ers correlated with its inability to provoke ligand-medi- ated GR dimerization as well as GR phosphorylation at a specific residue (eg, Ser211 ) (18). Based on these findings, it is anticipated that treatment with CpdA in vivo is asso- ciated with fewer GC-induced side effects. Again, CpdA is known to preferentially induce GR- mediated transrepression the major mechanism of which Figure 8. Effects of CpdA on AR. A, UMUC3-GR-shRNA cells were cotransfected with MMTV- Luc and pRL-TK and subsequently cultured with ethanol (mock), DHT (1nM), CpdA (1␮M), and/or HF (5␮M) for 24 hours. Luc activity is presented relative to that with mock treatment. Each value represents the mean ϩ SD from at least 3 independent experiments. *, P Ͻ .05 (vs mock treatment); #, P Ͻ .05 (vs DHT treatment). B, Nuclear protein fractions from UMUC3 cells cultured with ethanol (mock), 1nM DHT, and/or 1␮M CpdA for 24 hours were analyzed on Western blotting, using an antibody to AR (110 kDa). Histone 3 (15 kDa) served as an internal control. 1494 Zheng et al CpdA Treatment in Bladder Cancer Cells Mol Endocrinol, October 2015, 29(10):1486–1497 Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
  • 10. is believed to be the ability of the receptor to inhibit the activity of other transcription factors, including NF-␬B and AP-1. In prostate cancer cells, not only CpdA (28) but also DEX (36) reduced their growth mainly via inhibiting NF-␬B activation. We also previously demonstrated the data suggesting that inactivation of NF-␬B and inhibition of the production of NF-␬B-dependent cytokines, such as IL-6, might be a central mechanism involved in DEX/GR- mediated suppression of bladder cancer cell invasion (2). Similarly, in the present study, we showed that CpdA inhibited transcriptional activities of NF-␬B and AP-1 as well as the expression of NF-␬B-regulated genes, includ- ing MMP-2, MMP-9, IL-6, and VEGF, in GR-positive cells. CpdA could also induce the interaction between GR and NF-␬B. It is thus likely that CpdA treatment leads to a shift of GR functions toward transrepression in bladder cancer cells. However, the reduction of MMP-2/MMP-9/ IL-6/VEGF gene expression by CpdA might simply rep- resent its inhibitory effect on cell invasion rather than GR transrepression, although nonsignificant and significant inhibitions of these genes’ expression (Figure 6D) and cell invasion (Figure 3B), respectively, were seen in CpdA- treated AR-positive/GR knockdown UMUC3. Androgen deprivation has been shown to inhibit the expression of cell invasion-related genes, such as MMP-9 and VEGF, in bladder cancer cells (8, 14). Indeed, the expression levels of these 4 genes were slightly lower in mock-treated UMUC3-AR-shRNA than in mock-treated UMUC3-con- trol-shRNA (12%–25% decreases) (Figure 6D). None- theless, in the current transwell assay (Figure 3B), there was no significant effect of AR knockdown on bladder cancer cell invasion. Meanwhile, it was noteworthy that the suppressive effects of CpdA on bladder cancer cell invasion (eg, 61% decrease in UMUC3-control-shRNA) were found to be even stronger than those of DEX, cor- ticosterone, or prednisone in the transwell assays per- formed under the same conditions (eg, 34%–50% de- creases) (2, 5). Cytotoxic and proapoptotic activities of CpdA have been related to its effects on GR functions, such as its expression and nuclear translocation, in addition to transactivation and transrepression. Underlying mecha- nisms for GC-induced apoptosis may also vary depending on cell type. GC treatment has been shown to promote apoptosis in several types of malignancies, including leu- kemia, osteosarcoma, lung small cell carcinoma, and prostate cancer (36, 37), whereas it inhibits apoptosis in other types, such as breast cancer (38) and fibrosarcoma (39). DEX treatment is also associated with inhibition of apoptosis of bladder cancer cells in our previous (2, 5) and current studies. It has been suggested that the levels of GR expression and GR-mediated transactivation play an important role in determining the promotion of apoptosis vs survival in a cell-specific manner (37, 38). In our study, DEX is found to induce both transactivation and transre- pression of GR and reduces its expression in bladder can- cer cells, whereas CpdA induces only GR transrepression and does not modify GR expression. These differences may have contributed to the distinct effects of DEX vs CpdA as GR ligands on bladder cancer cell proliferation, whereas both compounds similarly induce G1 arrest. Ad- ditionally, down-regulation of GR expression by DEX has been implicated in a limitation of GCs with long-term use. In this respect, CpdA inducing no significant changes in GR expression may also be superior to DEX. CpdA has been found to antagonize androgen actions via mechanisms similar to those for classical antiandro- gens, such as flutamide (16, 28). Although CpdA inhibits androgen-enhanced AR transactivation, it possesses par- tial agonist activity and induces nuclear translocation of AR in prostate cancer cells (28). Indeed, clinically used antiandrogens, including flutamide and bicalutamide, failed to prevent androgen-induced AR nuclear translo- cation (40, 41). We demonstrated in bladder cancer cells that CpdA restored DHT-induced AR transcription yet did not affect DHT-mediated AR nuclear translocation. However, in contrast to the observations in prostate can- cer (28), CpdA appeared to have only nonsignificant ag- onist activity in bladder cancer cells without inducing AR nuclear translocation. Thus, CpdA was confirmed to act as an AR antagonist in bladder cancer cells. More impor- tantly, the inhibitory effects of CpdA on bladder cancer growth were invariably stronger in control cells than in AR-silencing cells stably expressing AR-shRNA and were still seen in GR-silencing cells stably expressing GR- shRNA. Because stable expression of GR-shRNA in blad- der cancer cells does not completely knock down endog- enous GR (2), there is a possibility that residual GR in the cells may have contributed to the latter effect. Nonethe- less, AR-mediated pathway is still thought to involve the inhibition of bladder cancer cell proliferation, migration, and invasion by CpdA, which further supports the role of AR signals in bladder cancer progression. Using mouse xenograft models for bladder cancer, we showed in vivo evidence suggesting that CpdA inhibited the progression of bladder cancer via both GR and AR pathways. CpdA was also found to more significantly suppress the growth of GR-positive and/or AR-positive xenografts than DEX or HF. Our previous immunohisto- chemical studies in bladder tissue microarrays revealed that most (eg, 87%) of bladder cancers expressed the GR (3), whereas AR positivity in tumors correlated with dis- ease progression in patients with muscle-invasive bladder cancer (13, 42). Therefore, CpdA has the potential to doi: 10.1210/me.2015-1128 press.endocrine.org/journal/mend 1495 Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
  • 11. efficiently regress tumor growth, especially in male pa- tients with GR-positive/AR-positive bladder cancer whose androgen levels are not low, and its effects may be more beneficial than GCs/pure GR ligands or AR antag- onists. However, the current study has compared the ef- ficacy of CpdA with that of only one compound each (ie, DEX and HF) in tumor growth. Moreover, we injected each drug 3 times a week. Although no data appears to be available about the half-life of each compound when in- jected peritumorally in mice, potentially different half- lives that are probably less than 48 hours may have af- fected the results from our xenograft study. In conclusion, CpdA was found to inhibit bladder can- cer growth predominantly via inducing GR-mediated transrepression as well as at least partially via inactivating androgen-enhanced AR signals. The current data also support previous observations indicating that bladder cancer cells possess functional GR and AR. Further study to extensively compare the antitumor activities of this unique compound vs various GR ligands and AR antag- onists in bladder cancer is required. Acknowledgments Address all correspondence and requests for reprints to: Hiroshi Miyamoto, MD, PhD, The James Buchanan Brady Urological Institute at the Johns Hopkins Hospital, 600 North Wolfe Street, Marburg 148, Baltimore, MD 21287. E-mail: hmiyamo1@jhmi.edu. This work was supported part by the National Natural Sci- ence Foundation of China Grant NFSC 81202022 and the Basic Research Program of the Department of Education of Zhejiang Province Grant Y201225369 (to Y.Z.). Disclosure Summary: The authors have nothing to disclose. References 1. Miyamoto H, Zheng Y, Izumi K. Nuclear hormone receptor signals as new therapeutic targets for urothelial carcinoma. Curr Cancer Drug Tar. 2012;12:14–22. 2. Zheng Y, Izumi K, Li Y, Ishiguro H, Miyamoto H. Contrary regu- lation of bladder cancer cell proliferation and invasion by dexam- ethasone-mediated glucocorticoid receptor signals. Mol Cancer Ther. 2012;11:2621–2632. 3. Ishiguro H, Kawahara T, Zheng Y, Netto GJ, Miyamoto H. Re- duced glucocorticoid receptor expression predicts bladder tumor recurrence and progression. Am J Clin Pathol. 2014;142:157–164. 4. Zhang C, Wenger T, Mattern J, et al. Clinical and mechanistic aspects of glucocorticoid-induced chemotherapy resistance in the majority of solid tumors. Cancer Biol Ther. 2007;6:278–287. 5. Ishiguro H, Kawahara T, Zheng Y, Kashiwagi E, Li Y, Miyamoto H. Differential regulation of bladder cancer growth by various glu- cocorticoids: corticosterone and prednisone inhibit cell invasion without promoting cell proliferation or reducing cisplatin cytotox- icity. Cancer Chemother Pharmacol. 2014;74:249–255. 6. Li Y, Izumi K, Miyamoto H. The role of the androgen receptor in the development and progression of bladder cancer. Jpn J Clin On- col. 2012;42:569–577. 7. Johnson AM, O’Connell MJ, Miyamoto H, et al. Androgenic de- pendence of exophytic tumor growth in a transgenic mouse model of bladder cancer: a role for thrombospondin-1. BMC Urol. 2008; 8:7. 8. Wu JT, Han BM, Yu SQ, Wang HP, Xia SJ. Androgen receptor is a potential therapeutic target for bladder cancer. Urology. 2010;75: 820–827. 9. Zheng Y, Izumi K, Yao JL, Miyamoto H. Dihydrotestosterone up- regulates the expression of epidermal growth factor receptor and ERBB2 in androgen receptor-positive bladder cancer cells. Endocr Relat Cancer. 2011;18:451–464. 10. Overdevest JB, Knubel KH, Duex JE, et al. CD24 expression is important in male urothelial tumorigenesis and metastasis in mice and is androgen regulated. Proc Natl Acad Sci USA. 2012;109: E3588–E3596. 11. Li Y, Zheng Y, Izumi K, et al. Androgen activates ␤-catenin signal- ing in bladder cancer cells. Endocr Relat Cancer. 2013;20:293– 304. 12. Jing Y, Cui D, Guo W, et al. Activated androgen receptor promotes bladder cancer metastasis via Slug mediated epithelial-mesenchy- mal transition. Cancer Lett. 2014;348:135–145. 13. Kawahara T, Shareef HK, Aljarah AK, et al. ELK1 is up-regulated by androgen in bladder cancer cells and promotes tumor progres- sion. Oncotarget. Available at: http://www.impactjournals.com/ oncotarget/index.php?journalϭoncotarget&pageϭarticle&opϭ view&path%5B%5Dϭ5007. 14. Miyamoto H, Yang Z, Chen YT, et al. Promotion of bladder cancer development and progression by androgen receptor signals. J Natl Cancer Inst. 2007;99:558–568. 15. Izumi K, Zheng Y, Li Y, Zaengle J, Miyamoto H. Epidermal growth factor induces bladder cancer cell proliferation through activation of the androgen receptor. Int J Oncol. 2012;41:1587–1592. 16. Louw A, Swart P. Salsola tuberculatiformis Botschantzev and an aziridine precursor analog mediate the in vivo increase in free cor- ticosterone and decrease in corticosteroid-binding globulin in fe- male Wistar rats. Endocrinology. 1999;140:2044–2053. 17. Tanner TM, Verrijdt G, Rombauts W, Louw A, Hapgood JP, Claessens F. Anti-androgenic properties of compound A, an analog of a non-steroidal plant compound. Mol Cell Endocrinol. 2003; 201:155–164. 18. De Bosscher K, Vanden Berghe W, Beck IM, et al. A fully dissoci- ated compound of plant origin for inflammatory gene repression. Proc Natl Acad Sci USA. 2005;102:15827–15832. 19. Li Y, Ishiguro H, Kawahara T, Miyamoto Y, Izumi K, Miyamoto H. GATA3 in the urinary bladder: suppression of neoplastictumori- genesis and down-regulation by androgens. Am J Cancer Res. 2014; 4:461–473. 20. Kawahara T, Kashiwagi E, Ide H, et al. Cyclosporine A and tacroli- mus inhibit bladder cancer growth through down-regulation of NFATc1. Oncotarget. 2015;6:1582–1593. 21. Evans RM. The steroid and thyroid hormone receptor superfamily. Science. 1988;240:889–895. 22. Zhou J, Cidlowski JA. The human glucocorticoid receptor: one gene, multiple proteins and diverse responses. Steroids. 2005;70: 407–417. 23. Ratman D, Vanden Berghe W, Dejager L, et al. How glucocorticoid receptors modulate the activity of other transcription factors: a scope beyond tethering. Mol Cell Endocrinol. 2013;380:41–54. 24. Ayroldi E, Riccardi C. Glucocorticoid-induced leucine zipper (GILZ): a new important mediator of glucocorticoid action. FASEB J. 2009;23:3649–3658. 25. Kester HA, van der Leede BM, van der Saag PT, van der Burg B. Novel progesterone target genes identified by an improved differ- ential display technique suggest that progestin-induced growth in- 1496 Zheng et al CpdA Treatment in Bladder Cancer Cells Mol Endocrinol, October 2015, 29(10):1486–1497 Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020
  • 12. hibition of breast cancer cells coincides with enhancement of dif- ferentiation. J Biol Chem. 1997;272:16637–16643. 26. Lesovaya EA, Yemelyanov AY, Kirsanov KI, Yakubovskaya MG, Budunova IV. Antitumor effect of non-steroid glucocorticoid re- ceptor ligand CpdA on leukemia cell lines CEM and K562. Bio- chemistry. 2011;76:1242–1252. 27. De Bosscher K, Vanden Berghe W, Haegeman G. The interplay between the glucocorticoid receptor and nuclear factor-␬B or acti- vator protein-1: molecular mechanisms for gene repression. Endocr Rev. 2003;24:488–522. 28. Yemelyanov A, Czwornog J, Gera L, Joshi S, Chatterto RT Jr, Budunova I. Novel steroid receptor phyto-modulator compound A inhibits growth and survival of prostate cancer cells. Cancer Res. 2008;68:4763–4773. 29. Ishiguro H, Kawahara T, Li Y, Miyamoto H. Anti-tumor activities of dexamethasone. In: Sauvage A, Levy M, eds. Dexamethasone: Therapeutic Uses, Mechanism of Action and Potential Side Effects. New York, NY: Nova Science Publishers; 2013:117–135. 30. Basch E, Prestrud AA, Hesketh PJ, et al. Antiemetics: American Society of Clinical Oncology clinical practice guideline update. J Clin Oncol. 2011;29:4189–4198. 31. Qian YH, Xiao Q, Chen H, Xu J. Dexamethasone inhibits camp- tothecin-induced apoptosis in C6-glioma via activation of Stat5/ Bcl-xL pathway. Biochim Biophys Acta. 2009;1793:764–771. 32. Chen YX, Wang Y, Fu CC, et al. Dexamethasone enhances cell resistance to chemotherapy by increasing adhesion to extracellular matrix in human ovarian cancer cells. Endocr Relat Cancer. 2010; 17:39–50. 33. Ge H, Ni S, Wang X, et al. Dexamethasone reduces sensitivity to cisplatin by blunting p53-dependent cellular senescence in non- small cell lung cancer. PLoS One. 2012;7:e51821. 34. Patel R, Williams-Dautovich J, Cummins CL. Minireview: new mo- lecular mediators of glucocorticoid receptor activity in metabolic tissues. Mol Endocrinol. 2014;28:999–1011. 35. Schäcke H, Berger M, Rehwinkel H, Asadullah K. Selective gluco- corticoid receptor agonists (SEGRAs): novel ligands with an im- proved therapeutic index. Mol Cell Endocrinol. 2007;275:109– 117. 36. Nishimura K, Nonomura N, Satoh E, et al. Potential mechanism for the effects of dexamethasone on growth of androgen-independent prostate cancer. J Natl Cancer Inst. 2001;93:1739–1746. 37. Schlossmacher G, Stevens A, White A. Glucocorticoid receptor- mediated apoptosis: mechanisms of resistance in cancer cells. J En- docrinol. 2011;211:17–25. 38. Aziz MH, Shen H, Maki CG. Glucocorticoid receptor activation inhibits p53-induced apoptosis of MCF10Amyc cells via induction of protein kinase C⑀. J Biol Chem. 2012;287:29825–29836. 39. Gascoyne DM, Kypta RM, Vivanco Md. Glucocorticoids inhibit apoptosis during fibrosarcoma development by transcriptionally activating Bcl-xL. J Biol Chem. 2003;278:18022–18029. 40. Masiello D, Cheng S, Bubley GJ, Lu ML, Balk SP. Bicalutamide functions as an androgen receptor antagonist by assembly of a transcriptionally inactive receptor. J Biol Chem. 2002;277:26321– 26326. 41. Kawahara T, Miyamoto H. Androgen receptor antagonists in the treatment of prostate cancer. Clin Immunol Endocr Metab Drugs. 2014;1:11–19. 42. Miyamoto H, Yao JL, Chaux A, et al. Expression of androgen and oestrogen receptors and its prognostic significance in urothelial neoplasm of the urinary bladder. BJU Int. 2012;109:1716–1726. doi: 10.1210/me.2015-1128 press.endocrine.org/journal/mend 1497 Downloadedfromhttps://academic.oup.com/mend/article-abstract/29/10/1486/2556433bygueston03March2020