ArticlePDF Available

Mutual antagonism of estrogen receptors alpha and beta and their preferred interactions with steroid receptor coactivators in human osteoblastic cell lines

Authors:
  • Robert Bosch Center for Tumor Diseases

Abstract and Figures

Estrogen is a major sex steroid that affects the growth, maintenance, and homeostasis of the skeleton. Two isoforms of the estrogen receptor (ERalpha and ERbeta) mediate the transcriptional effects of estrogen. Although both isoforms of ER are present and functional in some human osteoblast (OB) cell lines, there is minimal information on the differential regulation of transcription by ERalpha and ERbeta homo- or heterodimers. This report demonstrates that ERalpha and ERbeta coexpression decreases the transcriptional capacity (relative to each ER isoform alone) on an estrogen response element-dependent reporter gene in OBs but not in other non-osteoblastic cell lines. These data suggest that ERalpha and ERbeta coexpression can differentially influence the degree of transcriptional activation in certain cell types. Interestingly, the overexpression of the steroid hormone receptor coactivator-1 (SRC1) resulted in preferential transcriptional enhancement by ERbeta as well as coexpressed ERalpha and ERbeta, whereas SRC2 overexpression appeared to preferentially enhance ERalpha transactivation. SRC3 overexpression failed to enhance estrogen-dependent transcription of any ER combination in OBs. Similar overexpression experiments in COS7 cells exhibited preferential enhancement of ERalpha function with all SRCs, including SRC3. Our data also demonstrated that SRC3 mRNA is reduced in osteoblastic cells, suggesting that SRC3 may have only a minor role in these cells. These data suggest that the transactivation capacity of various ER isoforms is both SRC species and cell type dependent.
Content may be subject to copyright.
Mutual antagonism of estrogen receptors and and their
preferred interactions with steroid receptor coactivators in
human osteoblastic cell lines
D G Monroe, S A Johnsen, M Subramaniam, B J Getz, S Khosla
1
,
B L Riggs
1
and T C Spelsberg
Department of Molecular Biology and Biochemistry, Mayo Clinic and Mayo Foundation, 200 1st Avenue SW, Rochester, Minnesota 55905, USA
1
Endocrine Research Unit, Mayo Clinic and Mayo Foundation, Rochester, Minnesota 55905, USA
(Requests for offprints should be addressed to D G Monroe; Email: Monroe.David@mayo.edu)
Abstract
Estrogen is a major sex steroid that aects the growth,
maintenance, and homeostasis of the skeleton. Two iso-
forms of the estrogen receptor (ERand ER) mediate
the transcriptional eects of estrogen. Although both
isoforms of ER are present and functional in some human
osteoblast (OB) cell lines, there is minimal information on
the dierential regulation of transcription by ERand
ERhomo- or heterodimers. This report demonstrates
that ERand ERcoexpression decreases the transcrip-
tional capacity (relative to each ER isoform alone) on an
estrogen response element-dependent reporter gene in
OBs but not in other non-osteoblastic cell lines. These
data suggest that ERand ERcoexpression can dier-
entially influence the degree of transcriptional activation in
certain cell types. Interestingly, the overexpression of the
steroid hormone receptor coactivator-1 (SRC1) resulted in
preferential transcriptional enhancement by ERas well
as coexpressed ERand ER, whereas SRC2 over-
expression appeared to preferentially enhance ER
transactivation. SRC3 overexpression failed to enhance
estrogen-dependent transcription of any ER combination
in OBs. Similar overexpression experiments in COS7 cells
exhibited preferential enhancement of ERfunction with
all SRCs, including SRC3. Our data also demonstrated
that SRC3 mRNA is reduced in osteoblastic cells, sug-
gesting that SRC3 may have only a minor role in these
cells. These data suggest that the transactivation capacity of
various ER isoforms is both SRC species and cell type
dependent.
Journal of Endocrinology (2003) 176, 349–357
Introduction
Estrogen is involved in mediating important physiological
processes in numerous target tissues including breast,
uterus, brain, and bone (reviewed in Rickard et al. 2000).
The eects of estrogen were initially considered to
be mediated by a single estrogen receptor (ER)
(Mangelsdorf et al. 1995); however, a second estrogen
receptor (ER) that increases the potential diversity of
estrogen responses was discovered (Kuiper et al. 1996,
Mosselman et al. 1996, Tremblay et al. 1997). The two
ER isoforms function by binding 17-estradiol (E2) with
high anity. The ‘activated’ receptor isoforms subse-
quently dimerize with either themselves (e.g. forming
homodimers) or with the other ER isoform (e.g. forming
heterodimers) (Cowley et al. 1997). The receptor recog-
nizes specific cis-acting DNA elements (estrogen response
elements or EREs) located within the regulatory regions of
target genes and activates transcription through recruit-
ment of numerous coactivators and components of the
basal transcriptional machinery. ERand ERshare
significant homology within the DNA-binding domain
and ligand-binding domains (Rickard et al. 2000), sug-
gesting that both ER isoforms bind similar ligands and
EREs (Yi et al. 2002). However, significant dierences in
tissue specificity and responses to certain antiestrogens
(Paech et al. 1997) demonstrate that the ER isoforms are
unique and may perform dierent functions in dierent
cell types.
It is well accepted that estrogen plays a pivotal role in
bone cell metabolism and overall skeletal homeostasis,
which involves the regulatory actions of the bone-forming
osteoblasts (OBs). The development of specific ER
knockouts in mice has proven useful in understanding
estrogen action on the skeleton. Homozygous deletion of
the ERgene in mice (ERKO) results in decreased
longitudinal bone growth, decreased cortical bone density,
as well as decreased bone formation (Korach et al. 1996,
Lindberg et al. 2002), suggesting that ERis of critical
importance in promoting overall bone growth. In contrast,
349
Journal of Endocrinology (2003) 176, 349–357
0022–0795/03/0176–349 2003 Society for Endocrinology Printed in Great Britain
Online version via http://www.endocrinology.org
homozygous deletion of the ERgene in female mice
(BERKO) results in increased longitudinal bone growth
and increased bone mineral density (Windahl et al.
1999, Lindberg et al. 2002, Sims et al. 2002). Taken
together, these data suggest that ERmay exert a negative
eect on ER-mediated bone growth in mice. Although
this concept of ERantagonism has been suggested in
other cell systems (Hall & McDonnell 1999, Pettersson
et al. 2000), the physiological consequences of ER
coexpression with ERin bone cells, such as OBs, are
unknown. The adoption of similar principles of ER
function between the animal and human skeleton is
complicated by the observation that, in contrast to current
mouse models, a natural mutation of ERin man, which
inactivates the receptor, results in continued longitudinal
bone growth (Smith et al. 1994). This suggests that
estrogen has dierent eects on ER-mediated bone
growth in humans and mice. Thus, the understanding of
ER isoform actions and interactions in a human OB
system is critical to developing models for estrogen
action on the skeleton and developing new selective ER
modulators for the clinical treatment of osteoporosis.
Nuclear hormone receptor coactivators are involved in
enhancing the ligand-dependent transcriptional signal of
numerous nuclear hormone receptors, including ER. The
p160 family of coactivators includes steroid hormone
receptor coactivator (SRC)-1 (also called p160/
ERAP160) (Onate et al. 1995), SRC2 (also called GRIP1
and TIF2) (Voegel et al. 1996), and SRC3 (also called
TRAM1, ACTR, AIB1, RAC3, and p/CIP) (Li et al.
1997). Gene deletion experiments in mice demonstrate
that while homozygous deletion of SRC1 results in partial
hormone resistance (Xu et al. 1998, Weiss et al. 1999), the
phenotype of SRC1 null mice otherwise appears largely
normal. Interestingly, the SRC3 null mice demonstrate
dwarfism and a more severe reproductive phenotype (Xu
et al. 2000). However, the ultrastructure, densitometry,
and skeletal/bone growth patterns have not been exam-
ined in any SRC knockout model, nor have the coactiva-
tion properties of the SRCs been studied in a human OB
system.
This paper addresses several important and novel aspects
of ER function in OBs: the interactions between and
transcriptional regulation by coexpressed ERand ER,
the eects of SRC overexpression on ER-dependent
transcription, and the comparison of these eects between
osteoblastic and non-osteoblastic cells. As a first step to
understanding these aspects of ER function in human
OBs, it is important to first characterize these responses.
Materials and Methods
DNA constructs and reagents
Full-length cDNAs for ERand ERcontaining an
N-terminal FLAG epitope were constructed in the
pcDNA4/TO expression vector (Invitrogen, Carlsbad,
CA, USA) using PCR. Briefly, oligonucleotide primers
were designed to amplify the full-length ER(amino
acids 1–595) that contained BamHI and XhoI restriction
sites (5and 3respectively) for subcloning into pcDNA4/
TO. Full-length ER(amino acids 1–530) was con-
structed using primers containing HindIII and XbaI
restrictions sites (5and 3respectively) and subsequent
subcloning into pcDNA4/TO. A construct containing a
canonical ERE upstream of luciferase (ERE-TK-LUC)
was used as a reporter in transfection experiments.
pRL-TK (expressing Renilla luciferase) was used as a
transfection eciency control (Promega, Madison, WI,
USA). The ER antagonist ICI 182,780 was generously
provided by Zeneca Pharmaceuticals (Macclesfield,
Cheshire, UK) and E2 was purchased from Sigma (St
Louis, MO, USA).
Cell culture and transfection
Human fetal osteoblasts (hFOB) (Harris et al. 1995)
were maintained in phenol-red free Dulbecco’s modi-
fied Eagles’s medium (DMEM)/F12 media contain-
ing 10% (v/v) fetal bovine serum (FBS) supplemented
with 300 mg/l G418 and 1antibiotic/antimycotic
(Invitrogen). MG63, COS7, Hs578T, U2OS and SaOS2
cells were maintained in the same media lacking the G418
selection. All cells were transfected at a density of 50% in
six-well dishes in serum-free media with the indicated
amount of ER expression plasmid, 1·0 µg ERE-TK-LUC,
and 1·0 µg pRL-TK using lipofectamine PLUS reagent
(Invitrogen). Three hours after transfection, the hFOB
cells were washed and treated with ICI 182,780 (10 nM)
in DMEM/F12 media containing 10% (v/v) charcoal-
stripped FBS for 24 h to eliminate basal estrogen-
independent activation of the reporter construct. All other
cell lines were maintained in similar media lacking ICI
182,780 during this period. Subsequently, the cells were
treated either with ethanol vehicle or 10 nM E2 in
DMEM/F12 media containing 10% (v/v) charcoal-
stripped FBS for an additional 24 h. It is important
to mention that following removal of the ICI 182,780
from the hFOB cell media, no subsequent estrogen-
independent activation of the reporter construct was
observed in the absence of E2 during the 24-h treatment
period (data not shown). The cells were harvested and
30 µl cell extract was assayed using the Dual Luciferase
Reporter System (Promega) with a Turner Designs
20/20 luminometer (Sunnyvale, CA, USA). Promoter
activity was quantified as a ratio of firefly luciferase
to Renilla luciferase (expressed as relative light units;
RLU). Experiments were repeated three times and a
representative experiment is shown.
Western blot analysis
hFOB cells were transfected with 1·0 µg ERor ER
expression plasmid using lipofectamine PLUS reagent.
D G MONROE and others · ER isoforms and interactions with SRCs350
www.endocrinology.orgJournal of Endocrinology (2003) 176, 349–357
Total protein extracts were prepared in RIPA buer (1%
(v/v) NP40, 0·5% (v/v) sodium deoxycholate, 0·1%
(v/v), SDS, and 1protease inhibitor cocktail (Roche
Diagnostics Corporation, Indianapolis, IN, USA) in
1phosphate-buered saline) and 75 µg of the protein
extract was subjected to Western blot analysis and detected
using an -FLAG-M2 antibody (Sigma) followed by an
-mouse IgG secondary antibody (Sigma). The proteins
were visualized using an enhanced chemiluminescence kit
(Amersham Pharmacia, Piscataway, NJ, USA).
Real-time PCR analysis
Total cellular RNA from triplicate plates of COS7,
MCF7, hFOB, MG63, U2OS, and SaOS2 cells were
harvested using Trizol Reagent (Invitrogen). Four micro-
grams of total RNA were heat denatured at 68 Cfor
15 min in a reverse transcription reaction buer (11st
strand buer (50 mM Tris–HCl, 75 mM KCl, 3 mM
MgCl
2
), 50 mM dithiothreitol, 1 µM dNTPs, and 500 ng
oligo-dT primer). Following heat denaturation, 1 unit
MMLV-RT (Invitrogen) was added and the mixture
incubated at 37 C for 45 min followed by a 68 C
incubation for an additional 15 min. The resultant cDNA
products were diluted to 50 µl with PCR grade water and
2 µl was used in a real-time PCR reaction (1PCR
buer (20 mM Tris–HCl, 50 mM KCl), 3 mM MgCl
2
,
300 nM of both 5and 3primer, 1Sybr Green (Mol-
ecular Probes, Eugene, OR, USA), and 1 unit Taq
Polymerase (Promega)). The primer sequences used for
real time PCR are as follows: SRC1 5primer (5-TGC
CTC CGG GTA TCA GTC ACC AG), SRC1 3primer
(5-AGG CGT GGG CTG GTT CTG GAC AG), SRC2
5primer (5-GTG GTA TGC CAG CAA CTA TGA
GC), SRC2 3primer (5-TGG ATC AGG TTG CTG
ACT TAT TCC G), SRC3 5primer (5-ACA ACC
AGA TCC AGC CTT TGG TC), SRC3 3primer
(5-TGG ATG CAG CCT GCG GGT GTT GC),
-actin 5primer (5-TCA CCC ACA CTG TGC CCA
TCT ACG A), and -actin 3primer (5-CAG CGG
AAC CGC TCA TTG CCA ATG G). The reactions
were amplified using the I-Cycler (BioRad, Hercules,
CA, USA) using the following thermal protocol: 1 cycle at
94 C (1 min), 35 cycles at 94 C (30 s), 65 C (30 s), and
72 C (30 s). The C
T
measurement is defined at the
fractional cycle number at which the amount of amplified
target reaches a fixed threshold above background Sybr
Green fluorescence. The amount of target in the cDNA
sample relative to -actin was calculated by the formula:
2
C
T
, where C
T
is the dierence between the
average C
T
value for the target (SRC1, SRC2, or SRC3)
and -actin.
Results
Development of FLAG-tagged ER expression constructs and
determination of transfection conditions
For this study, N-terminal FLAG epitope-tagged ERand
ERexpression constructs were developed (see Materials
and Methods and Fig. 1A). Figure 1B demonstrates that
transfection of equimolar amounts of ERand ER
expression construct in hFOB cells produces proteins of
66 kDa and 54 kDa respectively, which are expressed at
approximately equal levels. To demonstrate that these
receptors are functional, either the ER–FLAG or ER
FLAG construct was transiently co-transfected with an
ERE–reporter construct into hFOB. Figure 1C demon-
strates that these receptors are functional, i.e. displays
E2-inducible transcription of an ERE-dependent reporter
gene when the cells were treated with E2. Under these
conditions, each isoform exhibits a comparable activity. In
order to determine the optimal amount of expression
construct to achieve the maximal transcriptional response,
a titration of ER–FLAG and ER–FLAG expression
constructs was performed in hFOB cells. Interestingly,
Fig. 2 demonstrates that cells containing the lowest
Figure 1 Development of FLAG epitope-tagged (TAG) ERand
ERexpression constructs. (A) Expression constructs containing an
N-terminal FLAG epitope were constructed using PCR and
subcloned into the pcDNA4/TO expression vector. (B) One
microgram of each ER–FLAG construct was transfected into hFOB
cells. Following 24 h to allow for expression, the cells were
harvested and 75 g total cellular extract was separated by PAGE
and transferred to a nitrocellulose membrane. The membrane was
probed with an anti-FLAG-M2 antibody and visualized using
enhanced chemiluminescence. (C) Ten nanograms of each
ER–FLAG construct was cotransfected with 1 g ERE-TK-LUC and
1gRenilla luciferase construct into hFOB cells. Following a 24-h
E2 treatment (10 nM E2), the cells were harvested and assayed for
luciferase activity. The bars represent fold induction by E2S.D.
and the asterisks represent significance at the P<0·001 level
(ANOVA) compared with the vehicle (EtOH control).
ER isoforms and interactions with SRCs ·D G MONROE and others 351
www.endocrinology.org Journal of Endocrinology (2003) 176, 349–357
amounts of the transfected ER constructs (10 ng) display
the greatest fold of E2-inducible transcription (approxi-
mately fourfold) for both ER isoforms. Other experiments,
using 5 ng ER, demonstrated similar levels of induction as
the 10 ng dose (data not shown). Therefore, 10 ng total
ER expression construct(s) was used throughout this study.
ERand ERcoexpression exhibits lowered transactivation
capacity in OBs but not in non-OBs
Gene deletion experiments in mice suggest that ERand
ERhave opposing actions on bone growth (Korach et al.
1996, Windahl et al. 1999). Therefore, since ERand
ERare coexpressed in OBs (Arts et al. 1997, Rickard
et al. 2000), we tested the hypothesis that coexpression of
ERand ERhas an antagonistic eect on ERE-
dependent (E2-inducible) reporter activity. Figure 3A
demonstrates that expression of ERor ERalone (10 ng
each) results in equivalent E2 induction of ERE-
dependent transcription in hFOB cells. Interestingly,
coexpression of equimolar amounts of ERand ER
(10 ng total ER) results in a statistically significant 31%
decrease in E2-inducible transcription in hFOB cells (Fig.
3A). Similar results are shown in MG63 osteosarcomas,
which also lack detectable endogenous ER expression
(Lambertini et al. 2002), where a statistically significant
47% decrease in E2-inducible transcription was observed
(Fig. 3B). This demonstrates that various OB cell lines,
coexpressing both ERand ERin equimolar ratios,
exhibit diminished E2-inducible responses. Since it is well
established in the literature that coexpression of equimolar
amounts of ERand ERexpression construct results in
primarily heterodimer formation (Cowley et al. 1997,
Tremblay et al. 1999), these studies reflect the transcrip-
tional eects of the ER/heterodimer and ERand
ERhomodimers on ERE-dependent promoter activity.
Furthermore, these data suggest that the ER heterodimer
has a unique transcriptional function since its E2-inducible
activity is significantly lower than either ER isoform alone.
To examine the cell type specificity of this ER isoform
antagonism, similar experiments were conducted in the
monkey kidney cell line, COS7, and the human breast
carcinoma, Hs578T, both of which lack detectable
endogenous ER expression (Gopalakrishna et al. 1999,
Kahlert et al. 2000). Figure 4A demonstrates that the
coexpression of ERand ERin COS7 cells results in no
dierence in E2-inducible transcription compared with
Figure 2 The effect of cotransfecting different ER amounts into
hFOB cells on the ERE reporter gene activity under E2 stimulation.
Increasing amounts of ERor ERexpression construct (10, 20,
40, 60, 80 ng) were cotransfected with 1 g ERE-TK-LUC and 1 g
Renilla luciferase into hFOB cells. Following a 24-h E2 treatment
(10 nM), the cells were harvested and assayed for luciferase
activity. The bars represent fold induction by E2S.D. and the
asterisks represent significance at the P<0·01 level (ANOVA)
compared with the 10 ng data point for each ERand ER
titration.
Figure 3 ERand ERcoexpression results in transcriptional
antagonism on ERE-reporter activity in hFOB cells and MG63
osteosarcomas. (A) ER(10 ng), ER(10 ng), or ER(5 ng) and
ER(5 ng) together (ER/) were cotransfected with 1 g
ERE-TK-LUC and 1 gRenilla luciferase into hFOB cells. Following
a 24-h E2 treatment (10 nM E2), the cells were harvested and
assayed for luciferase activity. (B) This panel represents the
identical experimental procedure as in (A) except that the
transfected cells were MG63 osteosarcoma cells. Each
experimental condition was performed in triplicate and a
representative experiment is shown. Luciferase values represent
the meansS.D. and the asterisks represent significance at the
P<0·001 level (ANOVA).
Figure 4 ERand ERcoexpression does not result in
transcriptional antagonism on ERE-reporter activity in COS7 and
Hs578T cells. (A) A total of 10 ng ER,ER,orERand ER
together (ER/) were cotransfected with 1 g ERE-TK-LUC and
1gRenilla luciferase into COS7 cells. Following a 24-h E2
treatment (10 nM E2), the cells were harvested and assayed for
luciferase activity. Luciferase values represent the meansS.D. (B)
This panel represents the identical experimental procedure as in
(A) except that the transfected cells were Hs578T breast
carcinoma cells. Each experimental condition was performed in
triplicate and a representative experiment is shown.
D G MONROE and others · ER isoforms and interactions with SRCs352
www.endocrinology.orgJournal of Endocrinology (2003) 176, 349–357
either isoform alone. In contrast, in Hs578T cells, ER
had weaker transcriptional activation capability compared
with ERand coexpression of ERand ERresulted
in an activity intermediate to either ER isoform alone
(Fig. 4B). Importantly, coexpression of ERand ERin
Hs578T cells does not exhibit a similar type of transcrip-
tional antagonism as seen in OBs, where coexpressed ER
and ERresulted in lowered E2-inducible activity than
either ER isoform alone (Fig. 3). Notably, the observation
that ERis a weaker activator than ERin Hs578T cells
has been described in other non-OB cell types (i.e.
Hep2 G cells) (Hall & McDonnell 1999), demonstrating
fundamental dierences between the ERand ER
homodimer activities when expressed in breast carcinoma
and OB cells.
Enhancement of ER signaling by SRCs is both SRC and cell
type dependent
Transcriptional responses elicited by the ER are mediated
by a group of nuclear transcription factors termed ‘steroid
receptor coactivators’ or SRCs (McKenna et al. 1999).
The specific modulation of SRC expression levels or their
coactivation properties with ER may explain the transcrip-
tional dierences of ER function in dierent cell types
(Figs 3 and 4). Although SRC-dependent coactivation of
both ER isoforms has been demonstrated in some cell
types, the eects of SRC coexpression on ER function in
OBs have not been explored. Therefore, in order to
understand the contribution of SRC1, SRC2, and SRC3
to E2 responses in hFOBs, titration of these SRC mol-
ecules with either ER isoform alone, or coexpressed ER
and ER, was performed. Titration of the SRC1 expres-
sion construct ranging from 0 to 80 ng resulted in only a
slight enhancement of ER-dependent transcription
on the ERE-luciferase construct (Fig. 5A). However,
overexpression of SRC1 resulted in a dose-dependent
enhancement of ER-dependent transcription (Fig. 5A).
The results support a preferential enhancement of ER
function. The transcriptional enhancement of coexpressed
ERand ER, reported in the literature to create pri-
marily heterodimers (Cowley et al. 1997, Tremblay et al.
1999), appeared similar to that of ERat higher SRC1
concentrations. Titration of SRC2 resulted in a dose-
dependent enhancement in cells containing ER,ER,or
Figure 5 Overexpression of SRC1, SRC2, or SRC3 results in an
SRC-specific and ER isoform-specific enhancement of
E2-dependent transcription in hFOB cells. Ten nanograms of ER,
ER,orERand ER(ER/) was cotransfected with 1 g
ERE-TK-LUC, 1 gRenilla luciferase, and increasing concentrations
(0, 20, 40, 60, 80 ng) of (A) SRC1, (B), SRC2, or (C) SRC3
expression constructs into hFOB cells. Following a 24-h E2
treatment (10 nM), the cells were harvested and assayed for
luciferase activity. Luciferase values represent the meansS.D.The
asterisks in (A) denote statistical significance between ERand
ERor ER/at the P<0·001 level (ANOVA). The asterisks in (B)
denote statistical significance among all ER combinations at the
P<0·01 level (ANOVA). Since estrogen-independent effects of SRC
overexpression were not observed, only the data points
representing estrogen treatment are shown. Each experimental
condition was performed in triplicate and a representative
experiment is shown.
ER isoforms and interactions with SRCs ·D G MONROE and others 353
www.endocrinology.org Journal of Endocrinology (2003) 176, 349–357
coexpressed ERand ER; however, SRC2 consistently
enhanced ER-dependent transcription to a greater
extent than ER(Fig. 5B). The coexpression of ERand
ERresults in an intermediate transcriptional activity of
either ER isoform alone. Surprisingly, overexpression of
SRC3 had no statistically significant eect on transcrip-
tional enhancement with any ER isoform combination
tested (Fig. 5C).
To examine the cell type specificity of these SRC-
dependent responses, similar studies were conducted in
COS7 cells. Figure 6A–C demonstrates that ERfunction
is enhanced to a greater extent than that of ERby all the
SRCs tested, and that the transcriptional enhancement
of coexpressed ERand ERappears intermediate to
either ER isoform alone in the presence of all the SRCs.
Thus, the preferential enhancement of SRC-dependent
coactivation, as observed in Fig. 5 with hFOB cells,
was not observed in COS7 cells. This demonstrates an
apparent cell type preference of SRC function with the
various ER dimers.
SRC3 mRNA is reduced in osteoblastic cell lines when
compared with non-osteoblastic cell lines
A plausible explanation for the lack of transcriptional
enhancement of SRC3 with any ER combination in the
hFOB cells is that sucient SRC3 is already present in
these cells and that additional (e.g. transfected) SRC3 will
not increase the ER-mediated transcriptional response.
Therefore, we examined the relative levels of endogen-
ous SRC1, SRC2, and SRC3 mRNAs in hFOB and
COS7 cells using real-time PCR. Figure 7 demonstrates
that no statistically significant dierence was observed in
SRC1 and SRC2 mRNA levels between the hFOB and
COS7 cells. However, a 20-fold decrease in SRC3
mRNA was observed in hFOB cells when compared with
COS7 cells, strengthening the notion that sucient SRC3
is not present in hFOB cells to increase ER transactivation.
To further explore this phenomenon, SRC3 mRNA
levels were examined in another non-OB cell line, MCF7,
and three additional human osteoblastic cell lines (MG63,
U2OS, and SaOS2). Figure 8 demonstrates that all the
osteoblastic cell lines tested have significantly reduced
SRC3 mRNA levels when compared with either COS7
or MCF7 cells.
Figure 6 Overexpression of SRC1, SRC2, or SRC3 results in
preferential enhancement of E2-dependent transcription by ERin
COS7 cells. Ten nanograms of ER,ER,orERand ER(ER/)
were cotransfected with 1 g ERE-TK-LUC, 1 gRenilla luciferase,
and increasing concentrations (0, 20, 40, 60, 80 ng) of (A) SRC1,
(B), SRC2, or (C) SRC3 expression constructs into COS7 cells.
Following a 24-h E2 treatment (10 nM), the cells were harvested
and assayed for luciferase activity. Luciferase values represent the
meansS.D. All asterisks denote statistical significance among all
ER combinations at the P<0·001 level (ANOVA). Since
estrogen-independent effects of SRC overexpression were not
observed, only the data points representing estrogen treatment are
shown. Each experimental condition was performed in triplicate
and a representative experiment is shown.
D G MONROE and others · ER isoforms and interactions with SRCs354
www.endocrinology.orgJournal of Endocrinology (2003) 176, 349–357
Discussion
This report addresses two important and largely unex-
plored aspects of ER function in OBs; the function of
ERand ERcoexpression and the eects of SRC
overexpression on E2-dependent transcription. We have
demonstrated that dual expression of ERand ERin
either hFOB or MG63 osteosarcoma results in an
estrogen-dependent transcriptional response that is 31%
and 47% lower respectively than either ER isoform acting
alone. Similar experiments in two non-OB cell lines,
COS7 and Hs578T, failed to exhibit the same transcrip-
tional antagonism when ERand ERwere coexpressed,
suggesting that transcriptional antagonism elicited by ER
and ERcoexpression is a cell type-specific phenomenon.
Earlier studies demonstrated that the ratio of ER:ER
changes significantly throughout OB dierentiation (Arts
et al. 1997). Our data demonstrating lowered activation
potential when ERand ERare coexpressed suggest that
altering ERand ERratios (which presumably shifts the
balance of ER homo- and heterodimers) during OB
dierentiation may potentially have important implications
in the overall magnitude of estrogen responses. These data
suggest that factors other than ER/heterodimerization
itself, presumably cell type-specific factors, are responsible
for determining the degree of E2-dependent transcription
at a classical ERE in various cell types. We are currently
investigating how dual expression of both ER isoforms
would aect dierent classes of E2-responsive elements
(i.e. YERE/SP1 and AP1 elements) in this human OB
system.
A potential mechanism explaining the dierences in ER
activity in divergent cell types (OB versus non-OB) may
involve the dierential expression or recruitment of tran-
scriptional accessory factors, such as the SRCs. Therefore,
we overexpressed SRC1, SRC2, and SRC3 with various
ER isoform combinations to test this hypothesis. We have
demonstrated that in hFOB cells, the overexpression of
SRC1 preferentially enhances ER-induced transcription
whereas SRC2 enhances ER-induced transcription to a
greater extent than either ERor coexpressed ERand
ER. This finding may have fundamentally important
implications on the activation of estrogen-responsive genes
that are dependent on a classical ERE for activation. The
activation of certain genes would be theoretically depen-
dent on the particular ER dimer bound at the promoter,
the concentration of each coactivator in the OB nucleus,
and possibly the promoter element involved. Our hypoth-
esis is that the modulation of ER dimers (e.g. homo- or
heterodimers), as occurs in in vivo osteoblastic dierentia-
tion (Arts et al. 1997), acts to ‘select’ a particular set of
accessory factors (e.g. SRCs) for transcriptional activation.
Interestingly, recent biochemical purification of SRC1
and SRC3 from HeLa nuclear extracts revealed that these
coactivators reside in unique complexes containing unique
enzymatic activities (Wu et al. 2002). Therefore, specific
ER dimers may determine the anity for a particular
coactivator complex and elicit specific coactivation func-
tions based on which coactivator complex is recruited.
Further delineation of the specific SRC complexes
involved in the regulation of estrogen-responsive genes
and of how the ER dimers dierentially interact with
these coactivator complexes would significantly aid in
testing this hypothesis.
Our data also suggest that the patterns of SRC-
dependent coactivation of transcription with a particular
ER dimer is a tissue-specific phenomenon, as SRC1,
SRC2, or SRC3 overexpression enhanced ER-induced
transcription to higher levels than either ERor coex-
pressed ERand ERin COS7 cells. This agrees with a
previous observation of SRC1 enhancement of ER-
induced transcription in COS1 cells (Sheppard et al.
2001). More specifically, our data extend this observation
to demonstrate that SRC2 and SRC3 overexpression also
preferentially enhances ER-induced transcription over
that of either ERor coexpressed ERand ER. Taken
Figure 7 Comparison of SRC1, SRC2, and SRC3 mRNA levels
demonstrates reduced SRC3 mRNA in hFOB cells. mRNA from
COS7 and hFOB cells were subjected to RT-PCR using real-time
PCR technology as described in the Materials and Methods
section. The data are plotted relative to -actin and normalized to
the COS7 data (arbitrarily set at 100). Values represent the
meansS.D. (n=3) and the asterisk denotes statistical significance
at the P<0·001 level (ANOVA).
Figure 8 SRC3 mRNA levels are reduced in various osteoblastic
cells compared with non-osteoblastic cells. mRNA from COS7,
MCF7, hFOB, MG63, U2OS, and SaOS2 cells were subjected to
RT-PCR using real-time PCR technology as described in the
Materials and Methods section. The data are plotted relative to
-actin and normalized to the COS7 data (arbitrarily set at 100).
Values represent the meansS.D. (n=3) and the asterisks denote
statistical significance at the P<0·001 level (ANOVA).
ER isoforms and interactions with SRCs ·D G MONROE and others 355
www.endocrinology.org Journal of Endocrinology (2003) 176, 349–357
together with the observation of SRC overexpression in
hFOBs, these data suggest that fundamental dierences
exist in the ability of SRCs to enhance ER isoform
function that is critically dependent on the cell type. This
also demonstrates that the lack of ER-induced transcrip-
tional enhancement in hFOB cells with SRC3 is a
biologically relevant phenomenon, and not due to techni-
cal issues with the SRC3 construct, since SRC3 enhanced
ER-dependent transcription in the COS7 cell line.
A plausible mechanism to explain the dierences in the
responses of various ER dimers to a particular SRC within
a specific cell type may lie in the physical interaction
interface between the ER and SRC molecules. Previous
studies demonstrated the stoichiometry for ER–SRC
interactions are one SRC molecule per ER dimer
(Kalkhoven et al. 1998, Tremblay et al. 1999). Therefore,
the SRC interaction interface is unique depending on
whether ERhomodimers, ERhomodimers, or ER/
heterodimers are present at the ERE. Our data suggest
that these unique interaction surfaces may be critically
important in determining the strength of coactivation
depending on which SRC molecule is present. Recent
reports have demonstrated that variations in ERE
sequences influence the magnitude of SRC coactivation of
ER function (Hall et al. 2002) which, in turn, is also
aected by various estrogenic compounds (Bramlett et al.
2001, Hall et al. 2002). However, the novelty of our data
lies in the demonstration that SRCs can aect estrogen
signaling at the same ERE with the same ligand (e.g. E2)
in dierent ways dependent on the ER dimer and cell
type.
An extreme example of this cell-specific coactivation is
illustrated with the overexpression of SRC3 having no
eect on ER-induced transcription in OBs compared with
the statistically significant eects of SRC3 overexpression
in COS7 cells. Thus, it appears that osteoblastic cells are
somehow impaired in their capability to utilize SRC3 in
estrogenic responses, possibly due to factors specific to
cell types other than OBs. Our data also indicate that
comparatively low levels of endogenous SRC3 exist in
multiple osteoblastic cell lines. Collectively, these obser-
vations suggest that SRC3 has little function in mediating
estrogenic responses in osteoblastic cells and that these
responses are most likely dependent on SRC1 and/or
SRC2 coactivation of the ER.
In summary, the present study serves to address two
fundamental aspects of ER signaling that occurs in OBs.
First, the notion that ERand ERcoexpression results in
transcriptional antagonism in OBs, as suggested by the
bone phenotypes of the ERKO (Couse & Korach 1999)
and BERKO (Windahl et al. 1999) gene disruption
mouse models (ERand ERrespectively). Secondly, to
characterize the transcriptional responses of the ER iso-
forms (homo- and heterodimers) to the nuclear receptor
coactivators, SRC1, SRC2, and SRC3. The responses of
ER homo- and heterodimers to SRC overexpression
illuminate an alternative form of transcriptional regulation
that is dependent on the ER dimerization status, the
specific SRC present at the promoter, and the cell type.
Acknowledgements
We thank Dr Bert O’Malley for providing the SRC1
expression construct (Onate et al. 1995), Dr Pierre
Chambon for the TIF2 (SRC2) expression construct
(Voegel et al. 1996), and Dr Paul Meltzer for the AIB-1
(SRC3) expression construct (Anzick et al. 1997). This
work was supported by an NIH grant (PO1-AG04875),
NIH training grant CA09441, and the Mayo Foundation.
References
Anzick SL, Kononen J, Walker RL, Azorsa DO, Tanner MM, Guan
XY, Sauter G, Kallioniemi OP, Trent JM & Meltzer PS 1997
AIB1, a steroid receptor coactivator amplified in breast and ovarian
cancer. Science 277 965–968.
Arts J, Kuiper GGJM, Janssen JMMF, Gustafsson J-A, Lowik CWGM,
Pols HAP & Van Leeuwen PTM 1997 Dierential expression of
estrogen receptors and mRNA during dierentiation of human
osteoblasts SV-HFO cells. Endocrinology 138 5067–5070.
Bramlett KS, Wu Y & Burris TP 2001 Ligands specific coactivator
nuclear receptor (NR) box anity for estrogen receptor subtypes.
Molecular Endocrinology 15 909–922.
Couse JF & Korach KS 1999 Estrogen receptor null mice: what have
we learned and where will they lead us? Endocrine Reviews 20
358–417.
Cowley SM, Hoare S, MosselmanS&Parker MG 1997 Estrogen
receptors alpha and beta form heterodimers on DNA. Journal of
Biological Chemistry 272 19858–19862.
Gopalakrishna R, Gundimeda U, Fontana JA & Clarke R 1999
Dierential distribution of protein phosphatase 2A in human breast
carcinoma cell lines and its relation to estrogen receptor status.
Cancer Letters 136 143–151.
Hall JM & McDonnell DP 1999 The estrogen receptor isoform
(ER) of the human estrogen receptor modulates ER
transcriptional activity and is a key regulator of cellular response to
estrogens and antiestrogens. Endocrinology 140 5566–5578.
Hall JM, McDonnell DP & Korach KS 2002 Allosteric regulation of
estrogen receptor structure, function, and coactivator recruitment by
dierent estrogen response elements. Molecular Endocrinology 16
469–486.
Harris SA, Enger RJ, Riggs BL & Spelsberg TC 1995 Development
and characterization of a conditionally immortalized human fetal
osteoblastic cell line. Journal of Bone and Mineral Research 10
178–186.
Kahlert S, Nuedling S, van Eickels M, Vetter H, MeyerR&Grohe
C 2000 Estrogen receptor alpha rapidly activates the IGF-1 receptor
pathway. Journal of Biological Chemistry 275 18447–18453.
Kalkhoven E, Valentine JE, Heery DM & Parker MG 1998 Isoforms
of steroid receptor co-activator 1 dier in their ability to potentiate
transcription by the oestrogen receptor. EMBO Journal 17 232–243.
Korach KS, Couse JF, Curtis SW, Washburn TF, Lindzey J, Kimbro
KS, Eddy EM, Migliaccio S, Snedeker SM, Lubahn DB,
Schomberg DW & Smith EP 1996 Estrogen receptor gene
disruption: molecular characterization and experimental and clinical
phenotypes. Recent Progress in Hormone Research 51 159–186.
Kuiper GGJM, Enmark E, Pelto-Huikko M, NilssonS&Gustafsson
J-Å 1996 Cloning of a novel estrogen receptor expressed in rat
prostate and ovary. PNAS 93 5925–5930.
D G MONROE and others · ER isoforms and interactions with SRCs356
www.endocrinology.orgJournal of Endocrinology (2003) 176, 349–357
Lambertini E, Penolazzi L, Aguiari G, del Senno L, Pezzetti F,
SollazzoV&PivaR2002 Osteoblastic dierentiation induced by
transcription factor decoy against estrogen receptor alpha gene.
Biochemical and Biophysical Research Communications 292 761–770.
Li H, Gomes PJ & Chen JD 1997 RAC3, a steroid/nuclear
receptor-associated coactivator that is related to SRC-1 and TIF2.
PNAS 94 8479–8484.
Lindberg MK, Moverare S, Skrtic S, Alatalo S, Halleen J, Mohan S,
Gustafsson J-Å & Ohlsson C 2002 Two dierent pathways for the
maintenance of trabecular bone in adult male mice. Journal of Bone
and Mineral Research 17 555–562.
Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G,
Umesono K, Blumberg B, Kaster P, Mark M, Chambon P &
Evans RM 1995 The nuclear receptor superfamily: the second
decade. Cell 83 835–839.
McKenna NJ, Lanz RB & O’Malley BW 1999 Nuclear receptor
coregulators: cellular and molecular biology. Endocrine Reviews 20
321–344.
Mosselman S, PolmanJ&Dijkema R 1996 ER: identification and
characterization of a novel human estrogen receptor. FEBS Letters
392 49–53.
Onate SA, Tsai SY, Tsai MJ & O’Malley BW 1995 Sequence and
characterization of a coactivator for the steroid hormone receptor
superfamily. Science 270 1354–1357.
Paech K, Webb P, Kuiper GGJM, Nilsson S, Gustafsson J-Å, Kushner
PJ & Scanlan TS 1997 Dierential ligand activation of estrogen
receptors ERand ERat AP1 sites. Science 277 1508–1510.
Pettersson K, Delaunay F & Gustafsson J-Å 2000 Estrogen receptor
acts as a dominant regulator of estrogen signaling. Oncogene 19
4970–4978.
Rickard D, Harris SA, Turner R, Khosla S & Spelsberg TC 2000
Estrogen and progestins. In Principles of Bone Biology, edn 2,
pp 655–676. Eds P Bilezikian, LG Raisz & GA Rodan. San Diego:
Academic Press.
Sheppard HM, Harries JC, Hussain S, BevanC&Heery DM 2001
Analysis of the steroid receptor coactivator 1 (SRC1)–CREB
binding protein interaction interface and its importance for the
function of SRC1. Molecular and Cellular Biology 21 39–50.
Sims NA, Dupont S, Krust A, Clement-Lacroix P, Minet D,
Resche-Rigon M, Gaillard-KellyM&BaronR2002 Deletion of
estrogen receptors reveals a regulatory role for estrogen receptor-b
in bone remodeling in females but not in males. Bone 30 18–25.
Smith EP, Boyd J, Frank GR, Takahashi H, Cohen RM, Specker B,
Williams TC, Lubahn DB & Korach KS 1994 Estrogen resistance
caused by a mutation in the estrogen receptor gene in a man.
New England Journal of Medicine 331 1056–1061.
Tremblay GB, Tremblay A, Copeland NG, Gilbert DJ, Jenkins NA,
LabrieF&GiguereV1997Cloning, chromosomal location and
functional analysis of the murine estrogen receptor .Molecular
Endocrinology 11 353–365.
Tremblay GB, Tremblay A, LabrieF&GiguereV1999Dominant
activity of activation function 1 (AF1) and dierential stoichiometric
requirements for AF-1 and -2 in the estrogen receptor
heterodimeric complex. Molecular and Cellular Biology 19
1919–1927.
Voegel JJ, Heine MJ, Zechel C, ChambonP&Gronemeyer H 1996
TIF2, a 160 kDa transcriptional mediator for the ligand-dependent
activation function AF-2 of nuclear receptors. EMBO Journal 15
3667–3675.
Weiss RE, Xu J, Ning G, Pohlenz J, O’Malley BW & RefetoS
1999 Mice deficient in the steroid receptor co-activator 1 (SRC1)
are resistant to thyroid hormone. EMBO Journal 18 1900–1904.
Windahl S, Vidal O, Andersson G, Gustafsson J-Å & Ohlsson C 1999
Increased cortical bone mineral content but unchanged trabecular
bone mineral density in adult female estrogen receptor
/mice. Journal of Clinical Investigation 104 895–901.
Wu R-C, Qin J, Hashimoto Y, Wong J, Xu J, Tsai SY, Tsai MJ &
O’Malley BW 2002 Regulation of SRC-3 (pCIP/ACTR/AIB-1/
RAC-3/TRAM-1) coactivator activity by IB kinase.
Molecular and Cellular Biology 22 3549–3561.
Xu J, Qiu Y, DeMayo FJ, Tsai SY, Tsai MJ & O’Malley BW 1998
Partial hormone resistance in mice with disruption of the steroid
receptor coactivator-1 (SRC-1) gene. Science 279 1922–1925.
Xu J, Liao L, Ning G, Yoshida-Komiya H, DengC&OMalley BW
2000 The steroid receptor coactivator SRC3 (pCIP/RAC3/AIB1/
ACTR/TRAM-1) is required for normal growth. PNAS 97
6379–6384.
Yi P, Driscoll MD, Huang J, Bhagat S, Hilf R, Bambara RA &
Muyan M 2002 The eects of estrogen-responsive element- and
ligand-induced structural changes on the recruitment of cofactors
and transcriptional responses by ERand ER.Molecular
Endocrinology 16 674–693.
Received in final form 5 November 2002
Accepted 7 November 2002
ER isoforms and interactions with SRCs ·D G MONROE and others 357
www.endocrinology.org Journal of Endocrinology (2003) 176, 349–357
... This is consistent with reports in the literature, from our laboratory and others, that ERβ acts as a natural antagonist to ERα through heterodimerization. (27)(28)(29) To further address this hypothesis, we used siRNAs to suppress ERβ in MC3T3-E1 osteoblastic cells, which express both ERα and ERβ (data not shown), and examined ERα-specific activation of an estrogen response element (ERE)-luciferase construct using the ERα-selective agonist PPT. (30) The ERβ siRNA resulted in a 52% decrease in ERβ transcripts compared with the negative control siRNA (Fig. 5A). ...
... This clearly demonstrates that suppression of ERβ in osteoblastic cells enhances ERα-specific transcriptional activation and is entirely consistent with the known literature. (27)(28)(29) ...
... (27) Similarly, we have also shown that coexpression of both ERα and ERβ in hFOB and MG63 osteoblastic cells attenuates estrogen-dependent activity relative to each receptor alone. (28) We have extended these findings in this report by demonstrating that suppression of ERβ in MC3T3-E1 osteoblastic cells enhances ERα-specific transcriptional activity using the ERα-selective agonist PPT. Taken together, these data suggest that the effects of osteoblast-lineage-specific deletion of ERβ may be the result of enhanced ERα action in these cells. ...
Article
Full-text available
While the role of ERα in regulating bone metabolism has been extensively studied, ERβ has been largely dismissed as a relevant modulator of bone mass. Previous studies examining ERβ utilized a germline knock-out mouse expressing transcript variants of ERβ and displaying systemic hormonal changes that confounded interpretation of the skeletal phenotype. Thus, we used a conditional ERβ mouse model to achieve deletion of ERβ specifically in early osteoprogenitor cells using the Prx1-Cre driver. We observed marked increases in the trabecular bone volume fraction (of 58% [P < 0.003] and 93% [P < 0.0003] in 6- and 12-week old female ERβ(Prx1-CKO) mice, respectively) but no changes in cortical bone. Serum estradiol and IGF-I levels were unaltered in ERβ(Prx1-CKO) mice. Bone formation and resorption indices by histomorphometry and serum assays were unchanged in these mice, suggesting that alterations in bone turnover may have occurred early in development. However, the ratio of colony-forming unit-osteoblasts (CFU-OBs) to CFU-fibroblasts (CFU-Fs) was increased in bone marrow cultures from ERβ(Prx1-CKO) compared to control mice, indicating increased differentiation of osteoblast precursor cells into osteoblasts in ERβ(Prx1-CKO) mice. Detailed quantitative polymerase chain reaction analyses of 128 genes in 16 pre-specified pathways revealed significant downregulation of 11 pathways in ERβ(Prx1-CKO) mice. Thus, deletion of ERβ specifically in osteoblast lineage cells, in the absence of all splice variants, increases trabecular bone mass and modulates multiple pathways related to bone metabolism. These findings suggest that pharmacological inhibition of ERβ in bone may provide a novel approach to treat osteoporosis. This article is protected by copyright. All rights reserved.
... As expected, 4OHT failed to recruit SRC-1, -2 or -3 to the PRL array with either receptor. Thus, the PRL-HeLa array model utilizes high throughput microscopy to visually detect differences in coregulator recruitment between ERα and ERβ, and between agonists (E2) and SERM (4OHT) ligands, which is similar to previous work by others using conventional in vitro chromatin immunoprecipitation (ChIP) assays (Monroe et al., 2003, Wong et al., 2001. ...
Article
Environmental exposures to chemically heterogeneous endocrine-disrupting chemicals (EDCs) mimic or interfere with hormone actions and negatively affect human health. Despite public interest and the prevalence of EDCs in the environment, methods to mechanistically classify these diverse chemicals in a high throughput (HT) manner have not been actively explored. Here, we describe the use of multiparametric, HT microscopy-based platforms to examine how a prototypical EDC, bisphenol A (BPA), and 18 poorly studied BPA analogs (BPXs), affect estrogen receptor (ER). We show that short exposure to BPA and most BPXs induces ERα and/or ERβ loading to DNA changing target gene transcription. Many BPXs exhibit higher affinity for ERβ and act as ERβ antagonists, while they act largely as agonists or mixed agonists and antagonists on ERα. Finally, despite binding to ERs, some BPXs exhibit lower levels of activity. Our comprehensive view of BPXs activities allows their classification and the evaluation of potential harmful effects. The strategy described here used on a large-scale basis likely offers a faster, more cost-effective way to identify safer BPA alternatives.
... These differences in transcriptional abilities between ERα and ERβ may be attributed to differential recruitment of coactivators, or differences in the ability of the same coactivator to facilitate transcription of the ER subtypes (129). While some studies using recombinant SRC-1 (129) are consistent with our findings that SRC-1 from brain interacts more with ERα than with ERβ, other studies suggest that SRC-1 associates equally with each ER subtype (130,131). While these later findings are consistent with our results using SRC-1 from hippocampus, we observed that SRC-1 from hypothalamus interacted more with ERα than with ERβ. ...
Article
Steroid hormones have profound effects on homeostasis, development , reproduction and behaviour. Many of the biological effects of steroid hormones are mediated through their respective receptors, which are members of the steroid ⁄ nuclear receptor superfamily of transcriptional activators (1, 2). Receptors for oestrogens (ER) and progestins (PR) can function in a classical, genomic mechanism by acting as ligand-dependent nuclear transcription factors. Nuclear receptor coregulators, consisting of coactivators and corepressors, are critical for modulating the transcriptional activity of ER and PR, as well as other nuclear receptors. Although ER and PR can also function in the brain independent of ligand and at the membrane to rapidly activate cytoplasmic signalling pathways (3–6), these receptors elicit many changes in behaviour and physiology by acting through classical, genomic mechanisms. This review focuses on the function of these important nuclear receptor coactivators in geno-mic mechanisms of ER and PR action in the brain and in behaviour.
... For RNA extraction, four hearts from each group were frozen immediately in liquid nitrogen after the end of the postconditioning procedure (10 min after reperfusion) to visualize the early molecular changes in this protection. RT-PCR was done in triplicate as described previously [22]. The following primer sequences were used: rat BNP sense primer, 5′-ACA ATC CAC GAT GCA GAA GCT -3′ and antisense primer, 5′-GGG CCT TGG TCC TTT GAG A -3′, ANP sense primer 5′-ATG GGC TCC TTC TCC ATC AC -3′ and antisense primer 5′-TCT ACC GGC ATC TTC TCC TC-3′, and TNF-α sense primer 5′-CCA GGA GAA AGT CAG CCT CCT -3′ and antisense primer 5′-TCA TAC CAG GGC TTG AGC TCA -3′. ...
Article
Full-text available
The role of pacing postconditioning (PPC) in the heart protection against ischemia-reperfusion injury is not completely understood. The aim of this study was to investigated if 17-β-estradiol (estrogen, E2), endogenous atrial natriuretic peptide (ANP), endogenous brain natriuretic peptide (BNP), and tumor necrosis factor-alpha (TNF-α) are involved in PPC-mediated protection. Langendorff perfused female Wistar rat hearts were used for this study. Hearts challenged with regional ischemia for 30 min subjected to no further treatment served as a control. The PPC protocol was 3 cycles of 30 s pacing alternated between the right atrium and left ventricle (LV). Protection was assessed by recovery of LV contractility and coronary vascular-hemodynamics. Ischemia induced a significant (P < 0.05) deterioration in the heart function compared with baseline data. PPC alone or in combination with short-term E2 treatment (E2 infusion at the beginning of reperfusion) significantly (P < 0.05) improved the heart functions. Short-term E2 treatment post-ischemically afforded protection similar to that of PPC. However, long-term E2 substitution for 6 weeks completely attenuated the protective effects of PPC. Although no changes were noted in endogenous ANP levels, PPC significantly increased BNP expression level and decreased TNF-α in the cardiomyocyte lysate and coronary effluent compared to ischemia and controls. Our data suggested a protective role for short-term E2 treatment similar to that of PPC mediated by a pathway recruiting BNP and downregulating TNF-α. Our study further suggested a bad influence for long-term E2 substitution on the heart as it completely abrogated the protective effects of PPC.
... For example, at high concentrations (i.e., 50 µg/mL or above), polysaccharides of T. wilfordii and LPS acted in a pattern of mutual antagonism, in which PS appears to act as an LPS antagonist as well as exhibit LPSlike effects on its own (Golenbock et al., 1991). Plausible mechanisms underlying this unique antagonism could be (1) chemical or physical interaction, in which two agonists form a complex in the medium and therefore deactivate each other, or (2) transcriptional interaction of receptors, in which the coexpression of a group of receptors for similar agonists results in a decrease in transcriptional capacity (e.g., the relationship between α and β estrogen receptors in certain types of cells) (Monroe et al., 2003), but this requires experimental verification. ...
Article
The organic extracts of Tripterygium wilfordii. Hook F. (Celastraceae) have been used in the treatment of inflammatory and autoimmune diseases for many years in China, although they also possess significant toxicity. These extracts are anti-inflammatory and immunosuppressive in nature, suppressing lipopolyscacharide (LPS)-stimulated upregulation of NF kappa B gene expression. In the current study, an ethyl acetate (EA) extract and a crude hot water soluble polysaccharide (PS) fraction from T. wilfordii. were compared. Both fractions were effective in reducing LPS-stimulated NO accumulation in cultured RAW 267.4 macrophages in a dose-dependent manner (IC50 values of 1.7 and 32.1 μ g/mL for EA and PS, respectively). The LC50 of EA (19.8–22.3 μ g/mL) was significantly greater than that of PS, which exhibited no apparent cytotoxicity toward either macrophages or EAhy 926 endothelial cells (up to 10 mg/mL). We also showed that PS was able to stimulate NO production by untreated macrophages in a dose-dependent manner. Thus, depending on the experimental condition of cultured macrophages, a crude PS fraction of T. wilfordii. may exhibit immunostimulatory or anti-inflammatory effects. This unique property, together with its relatively low level of toxicity, supports further investigation of its role in diseases in which inflammation and immunomodulation are critical.
Article
Full-text available
Aging is associated with visceral adiposity, metabolic disorders, and chronic low-grade inflammation. 17α-estradiol (17α-E2), a naturally occurring enantiomer of 17β-estradiol (17β-E2), extends life span in male mice through unresolved mechanisms. We tested whether 17α-E2 could alleviate age-related metabolic dysfunction and inflammation. 17α-E2 reduced body mass, visceral adiposity, and ectopic lipid deposition without decreasing lean mass. These declines were associated with reductions in energy intake due to the activation of hypothalamic anorexigenic pathways and direct effects of 17α-E2 on nutrient-sensing pathways in visceral adipose tissue. 17α-E2 did not alter energy expenditure or excretion. Fasting glucose, insulin, and glycosylated hemoglobin were also reduced by 17α-E2, and hyperinsulinemic-euglycemic clamps revealed improvements in peripheral glucose disposal and hepatic glucose production. Inflammatory mediators in visceral adipose tissue and the circulation were reduced by 17α-E2. 17α-E2 increased AMPKα and reduced mTOR complex 1 activity in visceral adipose tissue but not in liver or quadriceps muscle, which is in contrast to the generalized systemic effects of caloric restriction. These beneficial phenotypic changes occurred in the absence of feminization or cardiac dysfunction, two commonly observed deleterious effects of exogenous estrogen administration. Thus, 17α-E2 holds potential as a novel therapeutic for alleviating age-related metabolic dysfunction through tissue-specific effects.
Article
Full-text available
The cyclopentenone 15-deoxy-#12,14-prostaglandin J2 (15d- PGJ2) inhibits proliferation of cancer cells, including breast cancers, by peroxisome proliferator-activated receptor-; (PPAR;)-dependent and PPAR;-independent mechanisms. However, little is known about its effect on the transcriptional activity of estrogen receptor-A (ERA) that plays vital roles in the growth of breast cancers. Here, we show that 15d-PGJ2 inhibits both 17B-estradiol (E2)-dependent and E2-indepen- dent ERA transcriptional activity by PPAR;-independent mechanism. In addition, 15d-PGJ2 directly modifies ERA protein via its reactive cyclopentenone moiety, evidenced by incorporation of biotinylated 15d-PGJ2 into ERA, both in vitro and in vivo. Nanoflow reverse-phase liquid chroma- tography tandem mass spectrometry analysis identifies two cysteines (Cys227 and Cys240) within the COOH-terminal zinc finger of ERA DNA-binding domain (DBD) as targets for covalent modification by 15d-PGJ2. Gel mobility shift and chromatin immunoprecipitation assays show that 15d-PGJ2 inhibits DNA binding of ERA and subsequent repression of ERA target gene expression, such as pS2 and c-Myc. Therefore, our results suggest that 15d-PGJ2 can block ERA function by covalent modification of cysteine residues within the vulnerable COOH-terminal zinc finger of ERA DBD, resulting in fundamental inhibition of both hormone- dependent and hormone-independent ERA transcriptional activity. (Cancer Res 2007;67(6):2595-602)
Article
Estradiol17β (E2) and the phytoestrogens genistein (G), and daidzein (D) increase creatine kinase (CK) specific activity in primary cell cultures of human female to a greater extent in cells from pre-menopausal than post-menopausal women. Pretreatment with the non-calcemic analog of Vitamin D, JK 1624 F2-2 (JKF), upregulated this estrogenic response at all ages. In contrast, biochainin A (BA) and quercertin (Qu) increased CK with no age dependence or modulation by JKF pretreatment. Both ERα and ERβ present in the cells were upregulated by pretreatment with JKF, as measured by Western blot analysis. Real time PCR showed no significant change in ERα mRNA but a marked decrease in ERβ mRNA in both age groups after JKF treatment. Cells from both age groups had surface binding sites for E2, shown by assays using cell impermeable Europium labeled ovalbumin-E2 conjugate (Eu-Ov-E2). Binding of to intracellular E2 receptors (ERs) was similar in both age groups with differences in phytoestrogenic competition. JKF pretreatment increased nuclear but decreased membranal binding in both age groups. These results provide evidence for membranal, in addition to nuclear estrogen receptors which are differentially modulated by a Vitamin D analog.
Article
Full-text available
Nuclear receptors (NRs) act as ligand‐inducible transcription factors which regulate the expression of target genes upon binding to cognate response elements. The ligand‐dependent activity of the NR activation function AF‐2 is believed to be mediated to the transcription machinery through transcriptional mediators/intermediary factors (TIFs). We report here the cloning of the 160 kDa human nuclear protein TIF2, which exhibits all properties expected for a mediator of AF‐2: (i) it interacts in vivo with NRs in an agonist‐dependent manner; (ii) it binds directly to the ligand‐binding domains (LBDs) of NRs in an agonist‐ and AF‐2‐integrity‐dependent manner in vitro; (iii) it harbours an autonomous transcriptional activation function; (iv) it relieves nuclear receptor autosquelching; and (v) it enhances the activity of some nuclear receptor AF‐2s when overexpressed in mammalian cells. TIF2 exhibits partial sequence homology with the recently isolated steroid receptor coactivator SRC‐1, indicating the existence of a novel gene family of nuclear receptor transcriptional mediators.
Article
Full-text available
Estrogen signaling is mediated by ER and -. ERs are converted from an inactive form to a tran- scriptionally active state through conformational changes induced by ligand and estrogen-responsive element (ERE) sequences. We show here that ER and ER bind to an ERE independently from ER li- gands. We found that although the binding affinity of ER for an ERE is 2-fold lower than that of ER, both ERs use the same nucleotides for DNA contacts. We show that both EREs and ligands are independent modulators of ER conformation. Specifically, the ERE primarily determines the receptor-DNA affinity, whereas the structure of the ER ligand dictates the affinity of ER for particular cofactors. We found that the ligand-dependent cofactor transcriptional inter- mediary factor-2, through a distinct surface, also in- teracts with ER preferentially and independently of ligand. The extent of interaction, however, is depen- dent upon the ER-ERE affinity. In transfected cells, ER is more transcriptionally active than ER. The ERE sequence, however, determines the potency of gene induction when either ER subtype binds to an agonist. Antagonists prevent ERs from inducing tran- scription independently from ERE sequences. Thus, ERE- and ligand-induced structural changes are in- dependent determinants for the recruitment of cofactors and transcriptional responses. The ability of ER to differentially recruit a cofactor could con- tribute to ER subtype-specific gene responses. (Molecular Endocrinology 16: 674-693, 2002)
Article
Full-text available
Estrogen receptor β (ERβ) is a novel steroid receptor that is expressed in rat prostate and ovary. We have cloned the mouse homolog of ERβ and mapped the gene, designated Estrb, to the central region of chromosome 12. The cDNA encodes a protein of 485 amino acids that shares, respectively, 97% and 60% identity with the DNA- and ligand-binding domains of mouse (m) ERα. Mouse ERβ binds to an inverted repeat spaced by three nucleotides in a gel mobility shift assay and transactivates promoters containing synthetic or natural estrogen response elements in an estradiol (E2)-dependent manner. Scatchard analysis indicates that mERβ has slightly lower affinity for E2 [dissociation constant (Kd) = 0.5 nm] when compared with mERα (Kd = 0.2 nm). Antiestrogens, including 4-hydroxytamoxifen (OHT), ICI 182,780, and a novel compound, EM-800, inhibit E2-dependent transactivation efficiently. However, while OHT displays partial agonistic activity with ERα on a basal promoter linked to estrogen response elements in Cos-1 cells, this effect is not observed with mERβ. Cotransfection of mERβ and H-RasV12 causes enhanced activation in the presence of E2. Mutagenesis of a serine residue (position 60), located within a mitogen-activated protein kinase consensus phosphorylation site abolishes the stimulatory effect of Ras, suggesting that the activity of mERβ is also regulated by the mitogen-activated protein kinase pathway. Surprisingly, the coactivator SRC-1 up-regulates mERβ transactivation both in the absence and presence of E2, and in vitro interaction between SRC-1 and the ERβ ligand-binding domain is enhanced by E2. Moreover, the ligand-independent stimulatory effect of SRC-1 on ERβ transcriptional activity is abolished by ICI 182,780, but not by OHT. Our results demonstrate that while ERβ shares many of the functional characteristics of ERα, the molecular mechanisms regulating the transcriptional activity of mERβ may be distinct from those of ERα.
Article
Steroids, thyroid hormones, vitamin D3, and retinoids are lipophilic small molecules that regulate diverse biological effects such as cell differentiation, development, and homeostasis. The actions of these hormones are mediated by steroid/nuclear receptors which function as ligand-dependent transcriptional regulators. Transcriptional activation by ligand-bound receptors is a complex process requiring dissociation and recruitment of several additional cofactors. We report here the cloning and characterization of receptor-associated coactivator 3 (RAC3), a human transcriptional coactivator for steroid/nuclear receptors. RAC3 interacts with several liganded receptors through a mechanism which requires their respective ligand-dependent activation domains. RAC3 can activate transcription when tethered to a heterologous DNA-binding domain. Overexpression of RAC3 enhances the ligand-dependent transcriptional activation by the receptors in mammalian cells. Sequence analysis reveals that RAC3 is related to steroid receptor coactivator 1 (SRC-1) and transcriptional intermediate factor 2 (TIF2), two of the most potent coactivators for steroid/nuclear receptors. Thus, RAC3 is a member of a growing coactivator network that should be useful as a tool for understanding hormone action and as a target for developing new therapeutic agents that can block hormone-dependent neoplasia.
Article
We have cloned a novel member of the nuclear receptor superfamily. The cDNA of clone 29 was isolated from a rat prostate cDNA library and it encodes a protein of 485 amino acid residues with a calculated molecular weight of 54.2 kDa. Clone 29 protein is unique in that it is highly homologous to the rat estrogen receptor (ER) protein, particularly in the DNA-binding domain (95%) and in the C-terminal ligand-binding domain (55%). Expression of clone 29 in rat tissues was investigated by in situ hybridization and prominent expression was found in prostate and ovary. In the prostate clone 29 is expressed in the epithelial cells of the secretory alveoli, whereas in the ovary the granulosa cells in primary, secondary, and mature follicles showed expression of clone 29. Saturation ligand-binding analysis of in vitro synthesized clone 29 protein revealed a single binding component for 17beta -estradiol (E2) with high affinity (Kd = 0.6 nM). In ligand-competition experiments the binding affinity decreased in the order E2 > diethylstilbestrol > estriol > estrone > 5alpha -androstane-3beta ,17beta -diol >> testosterone = progesterone = corticosterone = 5alpha -androstane-3alpha ,17beta -diol. In cotransfection experiments of Chinese hamster ovary cells with a clone 29 expression vector and an estrogen-regulated reporter gene, maximal stimulation (about 3-fold) of reporter gene activity was found during incubation with 10 nM of E2. Neither progesterone, testosterone, dexamethasone, thyroid hormone, all-trans-retinoic acid, nor 5alpha -androstane-3alpha ,17beta -diol could stimulate reporter gene activity, whereas estrone and 5alpha -androstane-3beta ,17beta -diol did. We conclude that clone 29 cDNA encodes a novel rat ER, which we suggest be named rat ERbeta to distinguish it from the previously cloned ER (ERalpha ) from rat uterus.
Article
Estrogens have been shown to be essential for maintaining a sufficiently high bone mineral density and ERα expression has been demonstrated in bone cells. Recently, a novel estrogen receptor, estrogen receptor β (ERβ) has been identified. Here we demonstrate that also ERβ is expressed in human osteoblasts, and that ERα and ERβ are differentially expressed during human osteoblast differentiation. ERβ mRNA expression increased gradually during osteoblast culture, resulting in an average increase of 9.9 ± 5.3 fold (mean ± S.D., n = 3) at day 21 (mineralization phase) as compared to day 6 (proliferation phase). In contrast, ERα mRNA expression levels increased only slightly until day 10 (2.3 ± 1.7 fold) and then remained constant. The observed differential regulation of ERα and β is suggestive for an additional functional role of ERβ to ERα in bone metabolism.
Article
The human estrogen receptor a (ERa) and the recently identified ERb share a high degree of amino acid homology; however, there are significant differences in regions of these receptors that would be expected to influence transcriptional activity. Consequently, we com- pared the mechanism(s) by which these receptors regulate target gene transcription, and evaluated the cellular consequences of coexpres- sion of both ER subtypes. Previously, it has been determined that ERa contains two distinct activation domains, ERa-AF-1 and ERa-AF-2, whose transcriptional activity is influenced by cell and promoter con- text. We determined that ERb, like ERa, contains a functional AF-2, however, the ERb-AF-2 domain functions independently within the receptor. Of additional significance was the finding that ERb does not contain a strong AF-1 within its amino-terminus but, rather, contains a repressor domain that when removed, increases the overall tran- scriptional activity of the receptor. The importance of these findings was revealed when it was determined that ERb functions as a trans- dominant inhibitor of ERa transcriptional activity at subsaturating hormone levels and that ERb decreases overall cellular sensitivity to estradiol. Additionally, the partial agonist activity of tamoxifen man- ifest through ERa in some contexts was completely abolished upon coexpression of ERb. In probing the mechanisms underlying ERb- mediated repression of ERa transcriptional activity we have deter- mined that 1) ERa and ERb can form heterodimers within target cells; and 2) ERb interacts with target gene promoters in a ligand-inde- pendent manner. Cumulatively, these data indicate that one role of ERb is to modulate ERa transcriptional activity, and thus the relative expression level of the two isoforms will be a key determinant of cellular responses to agonists and antagonists. (Endocrinology 140: 5566 -5578, 1999)
Article
We report the establishment of a human fetal osteoblast cell line derived from biopsies obtained from a spontaneous miscarriage. Primary cultures isolated from fetal tissue were transfected with a gene coding for a temperature-sensitive mutant (tsA58) of SV40 large T antigen along with a gene coding for neomycin (G418) resistance. Individual neomycin resistant colonies were screened for alkaline phosphatase (AP)-specific staining. The clone with the highest AP level, hFOB 1.19, was examined further for other osteoblast phenotypic markers. Incubation of hFOB cells at the permissive temperature (33.5 degrees C) resulted in rapid cell division, whereas little or no cell division occurred at the restrictive temperature (39.5 degrees C). Both AP activity and osteocalcin (OC) secretion increased in a dose-dependent manner following dihydroxyvitamin D3 (1,25-D3) treatment when cultured at either temperature. However, AP and 1,25-D3-induced OC levels were elevated in confluent hFOB cells cultured at 39.5 degrees C compared with 33.5 degrees C. Treatment of hFOB cells with 1-34 parathyroid hormone (PTH) resulted in an increase in cAMP levels. Upon reaching confluence, hFOB cultures went through programmed differentiation and formed mineralized nodules as observed by von Kossa staining. Further, immunostaining of postconfluent, differentiated hFOB cells showed that high levels of osteopontin, osteonectin, bone sialoprotein, and type I collagen were expressed. Therefore, the clonal cell line hFOB 1.19 provides a homogeneous, rapidly proliferating model system to study certain stages of human osteoblast differentiation.
Article
Mutations in the estrogen-receptor gene have been thought to be lethal. A 28-year-old man whose estrogen resistance was caused by a disruptive mutation in the estrogen-receptor gene underwent studies of pituitary-gonadal function and bone density and received transdermal estrogen for six months. Estrogen-receptor DNA, extracted from lymphocytes, was evaluated by analysis of single-strand-conformation polymorphisms and by direct sequencing. The patient was tall (204 cm [80.3 in.]) and had incomplete epiphyseal closure, with a history of continued linear growth into adulthood despite otherwise normal pubertal development. He was normally masculinized and had bilateral axillary acanthosis nigricans. Serum estradiol and estrone concentrations were elevated, and serum testosterone concentrations were normal. Serum follicle-stimulating hormone and luteinizing hormone concentrations were increased. Glucose tolerance was impaired, and hyperinsulinemia was present. The bone mineral density of the lumbar spine was 0.745 g per square centimeter, 3.1 SD below the mean for age-matched normal women; there was biochemical evidence of increased bone turnover. The patient had no detectable response to estrogen administration, despite a 10-fold increase in the serum free estradiol concentration. Conformation analysis of his estrogen-receptor gene revealed a variant banding pattern in exon 2. Direct sequencing of exon 2 revealed a cytosine-to-thymine transition at codon 157 of both alleles, resulting in a premature stop codon. The patient's parents were heterozygous carriers of this mutation, and pedigree analysis revealed consanguinity. Disruption of the estrogen receptor in humans need not be lethal. Estrogen is important for bone maturation and mineralization in men as well as women.