SlideShare a Scribd company logo
1 of 11
Download to read offline
Steroidogenic Factor-1 Influences Protein-
Deoxyribonucleic Acid Interactions within the Cyclic
Adenosine 3؅,5؅-Monophosphate-Responsive Regions of
the Murine Steroidogenic Acute Regulatory
Protein Gene*
CLAVIA R. WOOTON-KEE AND BARBARA J. CLARK
Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine,
Louisville, Kentucky 40292
ABSTRACT
De novo synthesis of the steroidogenic acute regulatory protein
(StAR) in response to trophic hormonal stimulation of steroidogenic
cells is required for the delivery of cholesterol from the mitochondrial
outer membrane to the mitochondrial inner membrane and the cy-
tochrome P450 side-chain cleavage enzyme. StAR expression is tran-
scriptionally regulated by cAMP-mediated mechanisms, and we have
identified a 45-bp region within the mouse promoter that is important
for cAMP responsiveness of the gene. This region, located between
Ϫ105 and Ϫ60 of the start site of transcription, contains a SF-1-
binding site, a highly conserved C/EBP␤ ϪAP-1-nuclear receptor half-
site sequences (CAN region), and a GATA-4-binding site. The SF-1
element and CAN region are required for full basal activity, whereas
the GATA-4 element may account for 20% of the cAMP response in
MA-10 mouse Leydig tumor cells. A cAMP-dependent protein-DNA
complex was observed with the CAN region and mutation of a non-
consensus AP-1 site within this region greatly diminished promoter
strength. Complex protein-DNA interactions within the cAMP re-
sponse region (Ϫ105/Ϫ60) were shown to require the SF-1 element
(Ϫ95), suggesting that SF-1 is required for protein-DNA interaction
at the CAN (Ϫ79) region and maximal activity of the promoter. (En-
docrinology 141: 1345–1355, 2000)
STEROID HORMONE biosynthesis in the adrenal and
gonads is stimulated by trophic hormones via a cAMP-
dependent second messenger system. Within minutes of hor-
monal stimulation, cholesterol is mobilized to the outer mi-
tochondrial membrane and transferred to the mitochondrial
inner membrane where it is converted to pregnenolone by
the cytochrome P450 side-chain cleavage enzyme (P450scc)
(1–4). This acute response is the rate-limiting step in steroi-
dogenesis and is dependent upon the de novo synthesis of the
steroidogenic acute regulatory protein (StAR) (5). Without a
functional StAR protein, cholesterol accumulates in the outer
mitochondrial membrane and steroid production ceases.
Thus, StAR functions in the transfer of cholesterol to the inner
mitochondrial membrane (5). This function of StAR is ele-
gantly demonstrated by characterization of StAR knockout
mice that lack steroid production (6). These data confirmed
that the StAR mutations identified in patients with congen-
ital lipoid adrenal hyperplasia are the underlying cause for
the disorder (7, 8).
Hormonal treatment of steroidogenic cells controls StAR
gene expression. Our previous studies have shown that treat-
ment of MA-10 mouse Leydig tumor cells with trophic hor-
mones or the cAMP analog, (Bu)2cAMP, induces StAR mes-
sage within 30 min, and maximal steady state levels are
reached within 4 h (9). This induction of StAR gene tran-
scription does not require de novo protein synthesis, suggest-
ing that posttranslational mechanisms are involved in the
acute induction of StAR gene in response to increases in
intracellular levels of cAMP (10).
Several factors that participate in StAR gene activation
have been reported. One factor is the orphan nuclear receptor
steroidogenic factor-1 (SF-1). SF-1 has been shown to have a
critical role in the regulation of many of the steroid hydrox-
ylase genes, and gene knockout studies in mice have shown
that SF-1 is critical for development of the gonad and adrenal
glands (11–14). Regulation of SF-1 may occur through direct
phosphorylation by a protein kinase A (PKA)-dependent
pathway. This proposal is supported by studies in which
SF-1 trans-activation of the steroid hydroxylase genes was
lost in PKA-deficient cell lines (15, 16). Furthermore, poten-
tial PKA phosphorylation sites are present in SF-1, and in
vitro studies have directly demonstrated a PKA-dependent
phosphorylation of SF-1 (17–19). In addition, activation of the
mitogen-activated protein kinase pathway has been shown
to phosphorylate and active SF-1, supporting a role for phos-
phorylation of SF-1 for its function (20). Our initial analysis
of two SF-1 sites in the mouse promoter, located at Ϫ45 and
Ϫ135 from the transcription start site, indicated that these
elements contributed to basal promoter activity but were not
essential for the cAMP-dependent induction (10). The in-
volvement of a SF-1 site in StAR gene activation was more
recently confirmed with the rat promoter (21). Mutation of
the SF-1 elements at Ϫ135 and Ϫ95, either individually or in
combination, decreased SF-1-dependent reporter gene ex-
Received August 9, 1999.
Address all correspondence and requests for reprints to: Dr. Barbara
J. Clark, Department of Biochemistry and Molecular Biology, University
of Louisville School of Medicine, Louisville, Kentucky 40292. E-mail:
bjclark@louisville.edu.
* This work was supported by NIH Grant DK-51656.
0013-7227/00/$03.00/0 Vol. 141, No. 4
Endocrinology Printed in U.S.A.
Copyright © 2000 by The Endocrine Society
1345
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
pression in a nonsteroidogenic cell line cotransfected for SF-1
expression (21). In these studies the (Bu)2cAMP-mediated
increase in promoter activity was also depressed, suggesting
that SF-1 may mediate part of the cAMP response. However,
it is not known what effect the Ϫ95 SF-1 mutation would
have in a steroidogenic cell line, where factors other than SF-1
may be required for basal and cAMP-dependent StAR tran-
scription. On the other hand, the SF-1 element located at
Ϫ105/Ϫ95 in the human promoter was shown to be critical
for basal and maximal cAMP responsiveness in human gran-
ulosa-luteal cells and in H295R adrenocortical cells (22, 23).
Thus, it appears that the role of SF-1 in StAR expression may
be promoter and/or tissue specific.
C/EBP␤ has also been shown to modulate StAR basal
promoter activity. C/EBP␤ is a basic leucine zipper tran-
scription factor that is expressed in liver, intestine, lung, and
adipose tissues (24). More recently, C/EBP␤ has been iden-
tified as the only isoform of C/EBP present in unstimulated
primary Leydig cell cultures and MA-10 Leydig cells (25).
Two putative CCAAT sites located at Ϫ113 and Ϫ87 in the
murine StAR promoter were identified, and C/EBP␤ in
MA-10 nuclear extracts was shown to bind to the Ϫ113 el-
ement (26). A weak protein-DNA interaction was observed
with the Ϫ87 region of the promoter, but the factors were not
identified. Mutation of the Ϫ87, but not the Ϫ113, site abol-
ished SF-1-dependent trans-activation of StAR-luciferase re-
porter gene expression in transfected COS-1 cells (26). How-
ever, transient transfection of MA-10 cells with a StAR-
luciferase reporter gene construct containing mutations at
either the Ϫ87 or Ϫ113 C/EBP␤-binding site did not affect
the cAMP-dependent response, but greatly reduced the basal
activity of the promoter. A protein-protein interaction be-
tween C/EBP␤ and SF-1 was further demonstrated; thus,
these studies suggested a possible C/EBP␤-SF-1 interaction
that is required for basal promoter activity.
Although SF-1 and C/EBP␤ participate in basal expression
of StAR, the mechanism for cAMP-dependent induction has
not been elucidated. Previously, we narrowed the cAMP-
responsive region of murine StAR to Ϫ254 bp of the start site
of transcription (10). Sequence analysis of this region iden-
tified potential binding sites for transcription factors that
have been shown to mediate cAMP-dependent responses in
other genes. Two elements that are part of the focus in the
present study are a C/EBP␤/nonconsensus activating pro-
tein-1/nuclear receptor half-site located between Ϫ87/Ϫ79
(CAN region) and a putative binding site for members of the
GATA family of zinc finger transcription factors located be-
tween Ϫ68/Ϫ40. Activating protein-1 is composed of either
Fos-Jun or Jun-Jun dimers that bind to the consensus se-
quence TGA(G/C)TCA and regulates genes either constitu-
tively or by cAMP- or Ca2ϩ
-mediated mechanisms (27–29).
The GATA family of transcription factors regulates gene
expression, differentiation, and cell proliferation by binding
to the consensus DNA sequence (A/T)GATA(A/G) (30).
More recently, three members of the GATA transcription
factor family were shown to be expressed in the developing
gonads: GATA-1, GATA-4, and GATA-6 (31–33). GATA-1
message has been isolated in MA-10 cells, and GATA-1 was
shown to up-regulate the basal promoter activity of the rat
inhibin ␣-subunit gene (34). GATA-4 and GATA-6 have also
been identified in testicular tissue, and transient expression
of GATA-4 was shown to trans-activate an inhibin ␣-
promoter/reporter construct in mouse Leydig and granulosa
tumor cell lines (35). Thus, a new role is emerging for GATA
transcription factors in Leydig cell function.
To clarify the role of SF-1 in StAR expression in MA-10
mouse Leydig tumor cells, we extended our previous studies
and demonstrated that SF-1 binds to a third element in mouse
promoter at Ϫ95. We show that this SF-1 site, denoted SF1–3,
is required for full basal activity of the StAR promoter. Ad-
ditionally, SF-1 functions in part to stabilize protein-DNA
interactions at the C/EBP␤/nonconsensus activating pro-
tein-1 (AP-1)/nuclear receptor half-site (CAN) region located
at Ϫ79. We also demonstrate that GATA-4 binds to the StAR
promoter at Ϫ68 bp and contributes to 20% of the cAMP-
dependent induction. We conclude that the SF1–3, CAN, and
GATA-4 DNA-binding sites serve critical roles in maintain-
ing full basal promoter activity, which, in turn, is necessary
for maximal cAMP induction.
Materials and Methods
Materials
N6
,2Ј-O-(Bu)2cAMP [(Bu)2cAMP)] was purchased from Sigma (St.
Louis, MO). Restriction enzymes, Klenow enzyme, T4 DNA ligase, Lu-
ciferase Assay System, and pGL2-luciferase reporter vectors were pur-
chased from Promega Corp. (Madison, WI). [␥-32
P]ATP was obtained
from NEN Life Science Products (Wilmington, DE). Custom oligonu-
cleotides were purchased from Genosys (The Woodlands, TX) and Gen-
emed Synthesis, Inc. (San Francisco, CA). Glutathione-S-transferase
(GST)-SF-1 plasmid was donated by Dr. Keith Parker, University of
Texas Southwestern Medical Center (Dallas, TX). Antibodies were pur-
chased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA) and Up-
state Biotechnology, Inc. (Lake Placid, NY). X-ray film was obtained
from Eastman Kodak Co. (Rochester, NY) and NEN Life Science Prod-
ucts (Boston, MA).
Cell culture
The MA-10 mouse Leydig tumor cell line was a gift from Dr. M. Ascoli
(Department of Pharmacology, University of Iowa of Medicine, Iowa
City, IA). The cells were grown in Waymouth’s MB/752 medium con-
taining 15% horse serum and 40 ␮g gentamicin sulfate/ml.
Transient transfections and reporter assays
MA-10 cells were plated at 300,000 cells/well in a 24-well plate 24 h
before transfection. Two micrograms of StAR-luciferase reporter gene
plasmid and 2 ␮g pCMV-␤-galactosidase (CMV, cytomegalovirus) ex-
pression vector plasmid were cotransfected into cells using 15 ␮g/ml
Lipofectamine (Life Technologies, Inc., Grand Island, NY) and Way-
mouth’s medium without antibiotics and serum as described previously
(10). Twenty-four hours posttransfection, the cells were treated with 1
mm (Bu)2cAMP for 16–19 h. The Promega Corp. Luciferase Assay Sys-
tem was used to prepare cell lysates and measure luciferase activity
(Lumat LB 9507 luminometer, Wallac, Inc., Gaithersburg, MD). ␤-
Galactosidase activity was assayed using ␤-d-galactopyranosidase
(Roche Molecular Biochemicals, Indianapolis, IN) as the substrate and
measuring the absorbance at 595 nm. Relative light unit (RLU) values
were normalized to ␤-galactosidase activity for each sample. Each treat-
ment was performed in triplicate, and the mean Ϯ sem were determined.
The data were expressed as fold induction relative to the Ϫ254/ϩ35 luc
control, which was set at 100. The mean Ϯ sem for the fold induction in
response to (Bu)2cAMP for all independent transfection experiments
was calculated, and a pooled Student’s t test was performed on the data.
P Ͻ 0.05 was considered statistically significant.
1346 SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS Endo • 2000
Vol 141 • No 4
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
StAR-luciferase constructs
PCR was used to generate the 5Ј-deletion constructs for the StAR
promoter. Linearized Ϫ254/ϩ35 StAR promoter-pGL2-luciferase (Ϫ254
StAR promoter construct) was used as the template. To amplify the
region Ϫ150 to ϩ35 (Ϫ150/ϩ35 luc), MluI Ϫ150 and GL2-primers were
used in the PCR reaction, and the product was digested with MluI and
HindIII and cloned into the MluI and HindIII sites of pGL2-basic. The
regions Ϫ105 to ϩ35 (Ϫ105/ϩ35 luc) and Ϫ68 to ϩ35 region (Ϫ68/ϩ35
luc) were amplified using SF1–3 top primer and GL2 primers, and
Ϫ68/Ϫ44 top primer and GL2 primers, respectively, and the PCR prod-
uct for each clone was Klenow treated, digested with HindIII, and cloned
into the SmaI-HindIII sites of pGL2-basic. The site-specific mutants gen-
erated in the Ϫ254/ϩ35 luc plasmid were constructed by generating two
overlapping PCR products that share an engineered restriction site to
alter the core sequences of interest. To produce the 5Ј-PCR product, the
GL1 primer was used to amplify the top strand, and the bottom strand
was amplified with the site-specific primer. The 3Ј-PCR product was
produced with the site-specific mutant primer (top strand) and GL2
(bottom strand). The PCR products were digested with the appropriate
restriction enzymes and cloned into the SmaI-KpnI restriction sites of
pGL2-basic. The sequences of oligonucleotides used in the reactions are
listed under Oligonucleotides used in EMSA and PCR. The SF1–3mut/
GATA-4mut-luc, GATAmut/AP-1mut-luc, and SF1–3mut/AP-1mut-
luc constructs were created in a similar manner; the mutant SF1–3 site
was engineered into GATAmut-luc using the SF1–3mut-luc primer to
generate SF1–3mut/GATAmut-luc, the AP-1mut site was engineered
into GATAmut-luc using the AP-1mut-luc primer to generate
GATAmut/AP-1mut-luc, and the AP-1mut site was engineered into
SF1–3mut-luc using the AP-1mut-luc construct to generate SF1–3mut/
AP-1mut-luc. One exception for SF1–3mut/AP-1mut-luc is the 5Ј-PCR
product was digested with MluI and BglII, the 3Ј-PCR product was
digested with BglII and XbaI, and the two products were cloned into the
MluI-NheI sites of pGL2 basic. All sequences were confirmed by double
stranded sequencing using the Sequenase II kit (United States Biochem-
ical Corp., Cleveland, OH).
Electrophoretic mobility shift assay (EMSA)
Double stranded oligonucleotides were generated by mixing equal
molar concentrations of top and bottom strand primers in a high salt
annealing buffer [10 mm Tris (pH 7.5), 50 mm NaCl, and 1 mm EDTA]
and heating for 2 min at 95 C followed by slow equilibration to room
temperature. The oligonucleotides were end labeled with T4 DNA ki-
nase and [␥-32
P]ATP (NEN Life Science Products). To generate a probe
for the Ϫ105/Ϫ44 region of StAR, linearized Ϫ254 StAR promoter con-
struct was used as the DNA template in a PCR reaction with SF1–3 (top)
and Ϫ68/Ϫ44 (bottom). The Ϫ105/Ϫ44 fragment containing site-
specific mutations or double mutations was generated as described
above using the corresponding mutated reporter gene constructs as the
template and appropriate oligonucleotides. Each PCR fragment was gel
purified and radiolabeled as described above. Restriction digestion anal-
ysis was used to confirm the PCR-generated probe contained the desired
mutations. MA-10 nuclear extracts (2.5–15.0 ␮g) or 0.25 ␮g GST-SF-1
were incubated with 50 fmol radiolabeled oligonucleotide for 30 min on
ice. The binding buffer contained the following: 50 mm Tris-Cl (pH 8.0),
100 mm KCl, 12.5 mm MgCl2, 1 mm EDTA, 1 mm dithiothreitol, 20%
glycerol, and 2.5 ␮g poly(dI:dC). Cold competition assays were per-
formed with a 20- to 100-fold molar excess of oligonucleotides. Antibody
supershift assays were performed with 2.5 or 15 ␮g MA-10 nuclear
extract proteins or 0.25 ␮g GST-SF-1 plus 4 ␮g monoclonal GATA-1,
polyclonal GATA-4, polyclonal C/EBP␤, or polyclonal SF-1 antibodies.
For the supershift assays, all components of the binding reaction, ex-
cluding the radiolabeled probe, were preincubated on ice for 30 min.
Radiolabeled probe was then added, and the incubation was continued
for 30 min. Binding reactions were resolved on a 4% nondenaturing
polyacrylamide gel. The dried gels were exposed to x-ray film, and the
radioactive bands were visualized by autoradiography.
The following oligonucleotides were used in the EMSA binding re-
actions. Only the top strand is listed, and changed base pairs in the
mutated probes are underlined.
Sequences of oligonucleotides used for PCR and
EMSA probes
The following sequences were used: SF1–3, 5Ј-CATTCCATCCTT-
GACCCTCTGCA-3Ј; SF1–3mut, 5Ј-CATTCCATCCTCGAGCCTCT-
GCA-3Ј; Ϫ68/Ϫ44, 5Ј-TTTTTTATCTCAAGTGATGATGCAC-3Ј; Ϫ68/
Ϫ44 GATAmut, 5Ј-TTTTTCCGGACAAGTGATGATGCAC-3Ј; Ϫ68/
Ϫ44 SREBPmut (SREBP, sterol regulatory binding protein), 5Ј-TTTTT-
TATCTCTCGAGATGATGCAC-3Ј; Ϫ87/Ϫ64, 5Ј-TGCACAATGACT-
GATGACTTTTT-3Ј; Ϫ87/Ϫ64 AP-1mut, 5Ј-TGCACAATAGATCTT-
GACTTTTT-3Ј; Ϫ87/Ϫ64 1/2 mut, 5Ј-TGCACAATGACTGA-
GATCTTTTT-3Ј; GL1*, 5Ј-TGTATCTTATGGTACTGTAACTG-3Ј; GL2*,
5Ј-CTTTATGTTTTTGGCGTCTTCCA-3Ј; and Ϫ150 Mlu, 5Ј-ACA-
CACGCGTAGTCTGCTCCCTCCCACCTTGGCCA-3Ј (* indicates the
primers used for pGL2-basic vector).
Nuclear extract preparation
Confluent MA-10 mouse Leydig tumor cell cultures were treated with
fresh Waymouth’s medium in the absence (control) or presence of
(Bu)2cAMP for 4 h, and nuclear extracts were prepared following pre-
viously described protocols (36). The extracts were flash-frozen in liquid
nitrogen and stored at Ϫ80 C until use.
Results
5Ј-Deletion analysis localizes the cAMP-responsive region
between Ϫ105 and Ϫ68 of the mStAR promoter
To more closely map the cAMP-responsive elements
within the Ϫ254 bp region of the murine StAR gene, a series
of StAR promoter deletion constructs was transiently trans-
fected into MA-10 cells and tested for cAMP responsiveness
(Fig. 1). (Bu)2cAMP treatment of MA-10 cells resulted in an
8.7-fold increase in luciferase reporter gene activity for the
Ϫ254 StAR promoter construct. Deletion to Ϫ150 caused both
basal and (Bu)2cAMP-stimulated promoter activity to in-
crease 4- and 6-fold, respectively, which represents a 70%
increase in the cAMP-dependent fold induction compared
with the Ϫ254 StAR construct. Further deletion to Ϫ105
caused a 46% decrease in basal activity compared with Ϫ254
StAR, but the fold induction in response to the cAMP stim-
ulus was not diminished. However, a 5-fold decrease in the
cAMP-dependent induction was observed with the Ϫ68
StAR promoter construct. These results suggest that multiple
elements may be required for the cAMP response of the
mouse StAR promoter: a possible negative regulatory region
located between the Ϫ150/Ϫ105 and a positive regulatory
region located between the Ϫ105/Ϫ68 region of the pro-
moter. The Ϫ254/Ϫ65 region of the promoter was cloned
upstream of a heterologous promoter (Ϫ254/Ϫ66StAR-
pGL3luc), but the single copy of this region was not sufficient
to confer cAMP-dependent responsiveness (data not shown).
Therefore, elements 5Ј and 3Ј of Ϫ68 appear to required for
the induction of StAR gene by cAMP.
SF-1 and GATA-4 bind to elements within Ϫ105/Ϫ44
region of the mouse StAR promoter
As the 5Ј-deletion analysis indicated that the cAMP-
responsive region was within Ϫ105 bp of the transcriptional
start site, we were interested in testing for possible cAMP-
dependent protein-DNA interactions within this region. We
first compared the sequence of murine StAR promoter (Ϫ105
to ϩ35) to the transcription factor database and identified
several potential transcription factor-binding sites that in-
SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS 1347
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
cluded SF-1, C/EBP, AP-1, and GATA-1. Shown in Fig. 2A
is the sequence of the Ϫ150 region of murine StAR, with the
location of these elements indicated. An alignment with the
human and rat sequences is also shown to indicate the high
degree of sequence conservation within this region of the
StAR promoter. We previously characterized the SF-1–1
(Ϫ135), SF-1–2 (Ϫ45), and the C/EBP␤ (Ϫ113) elements (10,
26), whereas the SF-1 site located at Ϫ95 bp has been shown
to bind SF-1 in the human and rat StAR promoters (21, 37).
The region between Ϫ87/Ϫ64 is not as well conserved and
unique to the mouse sequence are potential C/EBP␤, non-
consensus AP-1, and nuclear-receptor half-site elements that
we refer to as the CAN region. Another highly conserved
region, located between Ϫ68/Ϫ44, is a putative binding site
for GATA-1 and SREBP. This region is adjacent to the pro-
posed cAMP-responsive region (Ϫ105/Ϫ68) identified by
5Ј-deletion analysis and was deleted in the nonresponsive
3Ј-deletion construct (Ϫ254/Ϫ66StAR-pGL3luc), indicating
that this region also may contribute to the cAMP response.
Based on the location of these elements, overlapping DNA
probes were generated to encompass the SF-1 (Ϫ105/Ϫ83
SF1–3), the C/EBP␤-AP-1-nuclear receptor half-site se-
quences (Ϫ87/Ϫ64 CAN), and the GATA/SREBP element
(Ϫ68/Ϫ44 GATA) and used in EMSAs. Mutations that were
introduced into the core binding sequences of the oligonu-
cleotides are shown in Fig. 2B. The SF1–3 DNA probe (Ϫ105/
Ϫ83) formed a specific protein-DNA complex that was ob-
served using nuclear proteins isolated from either untreated
(lane 1) or (Bu)2cAMP-treated (lane 5) MA-10 cells (Fig. 3).
The specificity of complex formation was shown by compe-
tition with unlabeled SF1–3 probe (lanes 2 and 6), and the
complex was not formed when an oligomer that contained
two point mutations in the CATCCTTG core sequence (SF1–
FIG. 1. 5Ј-Deletion analysis of the mouse StAR promoter. MA-10 cells
were cotransfected with the indicated StAR-promoter-luciferase re-
porter gene constructs (denoted by the position of the base pairs
5-prime to the transcriptional start site) and the pCMV-␤-galactosi-
dase expression vector. The transfected cells were treated in triplicate
with fresh Waymouth’s medium in either the absence (basal) or pres-
ence of 1 mM (Bu)2cAMP for 16–19 h, and the luciferase activity was
determined and normalized to ␤-galactosidase expression (RLU/b-
gal). For each experiment, the data were expressed as a percentage
of the Ϫ254 StAR-promoter construct (Ϫ254/ϩ35 StAR promoter-
pGL2luciferase), which was set at 100%, and are graphed as the
mean Ϯ SEM values for the relative luciferase activity from three to
five experiments. The fold induction values represent the ratio of
(Bu)2cAMP/basal activity for each construct. Shown are the mean Ϯ
SEM values for three to five experiments. The inset graph shows the
RLU/␤-gal activities of the Ϫ254, Ϫ105, and Ϫ68 StAR-promoter
constructs. pGL2-basic represents vector control, and NT represents
nontransfected MA-10 cells. *, Statistically significant (P Ͻ 0.05)
increase (Ϫ150/ϩ35) or decrease (Ϫ68/ϩ35 luc) relative to the fold
induction of the Ϫ254 StAR promoter-luciferase construct in response
to (Bu)2cAMP.
FIG. 2. A, Comparison of mouse (M),
human (H), and rat (R) StAR promoter
sequences within Ϫ150 bp before the
transcriptional start site. The location
of the DNA elements for SF-1, C/EBP␤-,
GATA-4-, and putative SREBP-binding
sites are indicated above the core se-
quences by the respective name of the
highlighted factors. The putative bind-
ing sites for the CAN region (Ϫ87/Ϫ64)
include C/EBP, AP-1, and an imperfect
nuclear receptor half-site (1/2). B,
Shown are the region of the promoter
and the name given to the introduced
mutation in the Ϫ254/ϩ35 StAR-
promoter constructs and the EMSA
probes. The point mutations are indi-
cated in bold.
1348 SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS Endo • 2000
Vol 141 • No 4
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
3mut) was used in the binding reaction (lanes 4 and 8). A
protein-DNA complex of similar mobility was observed
when the probe was incubated with purified GST-SF-1 (lane
9), and addition of an antibody specific for SF-1 greatly
diminished or abolished DNA-protein interaction (lanes 3
and 7). Thus, these data verify that SF-1 also binds to this
region of the mouse promoter. As the mobility and intensity
of the SF-1 complex are the same with nuclear proteins from
either control or (Bu)2cAMP-stimulated cells, these data in-
dicate that SF-1 binding is independent of the cAMP stimulus
in MA-10 cells. This element is referred to as SF1–3 to indicate
the third SF-1-binding site within 150 bp of the mouse pro-
moter (Fig. 2A).
Two protein-DNA complexes were observed with the
CAN (Ϫ87/Ϫ64) DNA probe (Fig. 4, A and B). Complex A
is a doublet that is present in binding reactions with nuclear
proteins from both control and (Bu)2cAMP-treated MA-10
cells, whereas complex B is a slower migrating band that is
present only with nuclear proteins from stimulated cells (Fig.
4A). As this region contains a CAAT box, and C/EBP␤ has
been proposed or shown to interact with this region (26, 38),
we used a C/EBP␤ antibody to test for the presence of
C/EBP␤ in complex A or B. As shown in Fig. 4B, addition of
the antibody did not affect protein-DNA interactions with
the CAN probe and nuclear proteins from stimulated cells
(lane 2). We verified that a protein-DNA complex is formed
with a consensus C/EBP oligonucleotide (lane 3) and that
this complex is supershifted by antibody (lane 4), confirming
that C/EBP␤ is present in the nuclear extract (25, 26). Cold
competition with the consensus C/EBP oligonucleotide also
failed to indicate any involvement of C/EBP␤ with this el-
ement (lane 5). The specificity of complex formation was
demonstrated by cold competition with the CAN probe (lane
8) and a mutation in the putative nuclear receptor half-site
(1/2mut) was also effective as a competitor probe (lane 6).
Mutation in the putative AP-1 site (AP-1mut), on the other
hand, did not compete with the CAN probe for protein
binding (lane 7). We next tested for Fos/Jun binding to this
FIG. 3. EMSA analysis reveals SF-1 binding to the Ϫ105/Ϫ83 region
of mouse StAR. EMSA was used to examine binding of MA-10 nuclear
extract to the radiolabeled SF1–3 probe, which represents the Ϫ105/
Ϫ83 region of the StAR promoter. Binding reactions containing 50
fmol radiolabeled probe and 2.5 ␮g nuclear extracts prepared from
unstimulated (lanes 1–4) or (Bu)2cAMP-treated (lanes 5–8) MA-10
cells or 0.25 ␮g purified GST-SF-1 protein (lane 9) were preincubated
on ice for 30 min. For cold competition or antibody interference anal-
ysis, a 100-fold molar excess of cold probe (indicated as ؉ in lanes 2
and 6) or 4 ␮g SF-1 polyclonal antibody (indicated as Ab in lanes 3 and
7) were preincubated with nuclear extract for 30 min; then the ra-
diolabeled probe was added to all reactions, and the incubation was
continued for 30 min on ice. The DNA-protein complexes were re-
solved on a nondenaturing 4% polyacrylamide gel, the gel was dried,
and the complexes were visualized by autoradiography. Mutations
introduced into SF1–3 (SF1–3 mut) are listed in Fig. 2B. Lane 10
shows incubation of SF-1 polyclonal antibody and radiolabeled probe
without nuclear extract. Free probe is not shown on the gel.
FIG. 4. A cAMP-dependent factor(s) binds to the Ϫ87/Ϫ64 CAN re-
gion of mouse StAR. EMSA was used to examine binding of MA-10
nuclear extract to radiolabeled CAN probe, which represents the
Ϫ87/Ϫ64 region of the StAR promoter. A, Concentration-dependent
protein-DNA complex formation. Binding reactions were preincu-
bated for 30 min on ice with nuclear extracts prepared from unstimu-
lated (control) or (Bu)2cAMP-treated [(Bu)2] MA-10 cells. Shown is a
titration of 5, 10, and 15 ␮g control or (Bu)2cAMP nuclear extracts,
respectively. B, Effects of cold competition and C/EBP␤ antibody on
protein-DNA interactions. Either a 50-fold molar excess of the indi-
cated cold probe or C/EBP␤ polyclonal antibody (4 ␮g) antibody was
included in a preincubation with nuclear extracts for 30 min; then
radiolabeled probe was added, and the incubation was continued for
30 min on ice. The protein-DNA complexes were resolved on a non-
denaturing 4% polyacrylamide gel, the gel was dried, and the com-
plexes were visualized by autoradiography. The sequences of the
oligomer probes for the CAN region containing mutations in the po-
tential AP-1 (AP-1 mut) and imperfect nuclear receptor half-sites (1/2
mut) are listed in Fig. 2B. con. C/EBP, Commercially purchased con-
sensus C/EBP oligomer. The arrows indicate the positions of com-
plexes A and B and free probe. SS, The C/EBP␤ antibody supershift.
SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS 1349
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
region, but addition of antibodies that recognize all family
members did not affect complex formation (data not shown).
A prominent protein-DNA complex was apparent with
the GATA oligonucleotide (Ϫ68/Ϫ44 GATA) using nuclear
proteins from either untreated or (Bu)2cAMP-stimulated
MA-10 cells (Fig. 5). Complex formation was abolished by
cold competition with the GATA oligonucleotide (lane 2) or
the SREBPmut oligonucleotide that contains a mutation in
the putative SREBP site (lane 4). Conversely, using the SREB-
Pmut as a probe did not affect complex formation (lane 3),
indicating that the SREBP element is not involved in protein-
DNA interactions at this region. On the other hand, no com-
plex was formed with a probe that had a mutation within the
GATA DNA-binding site (lane 5), and the mutant GATA
probe (GATA mut) did not compete for protein binding (lane
6). A complex of similar mobility was observed using a probe
with the consensus sequence for GATA binding (lanes 7 and
8) and cold competition experiments using this consensus
GATA probe resulted in the lack of complex formation (lane
10). Although this sequence was identified as a potential
GATA-1-binding site, GATA-1 antibodies did not affect com-
plex formation (compare lanes 1 and 9). However, binding to
the consensus GATA probe appeared to be diminished by
GATA-1 antibodies (compare lanes 7 and 8). Therefore, we
tested for other members of the GATA family, and GATA-4
antibodies resulted in a supershift of the complex (lanes
11–13). The supershifted complex was apparent using nu-
clear extracts from either control (lane 11) or (Bu)2cAMP-
treated (lanes 12 and 13) MA-10 cells with the StAR GATA
probe (lanes 11 and 12) or the consensus GATA probe (lane
13). A less prominent band appears with the Ϫ68/Ϫ44 GATA
probe; however, the significance of this interaction is not
known. At present, these results indicate that GATA-4 in
MA-10 cells binds to the mouse StAR promoter.
The Ϫ87/Ϫ68 region of the StAR promoter is required for
maximal basal promoter activity of StAR
To test the functional significance of the specific protein-
DNA interactions characterized above on StAR promoter
activity, the mutations that abolished protein binding were
individually introduced into the Ϫ254 StAR promoter con-
struct, and luciferase activity was measured in transiently
transfected MA-10 cells (Fig. 6A). Mutation of the SF1–3 site
resulted in a 43% reduction in basal luciferase activity com-
pared with the Ϫ254 StAR promoter construct (WT), but the
fold induction in response to (Bu)2cAMP stimulation was not
significantly different. Mutation of the GATA-4-binding site
also reduced basal promoter strength (ϳ36%); however, the
cAMP response was also decreased from 8.7-fold (WT) to
6.9-fold (GATA). Lastly, mutation of the putative AP-1 site
(AP-1mut) or the nuclear receptor half-site (1/2mut) resulted
in decreased basal activity by 63% or 57%, respectively, but,
again, fold activation in response to (Bu)2cAMP was un-
changed compared with that with the Ϫ254 StAR promoter
construct (WT). Although the fold induction by (Bu)2cAMP-
treatment was not diminished with the SF1–3 and CAN
region mutations, overall promoter strength was reduced.
This decrease in response appeared to closely parallel the
severity of the mutation on basal promoter activity. These
data indicate that the SF1–3, CAN, and GATA sites all con-
tribute to basal promoter strength. However, neither the
SF1–3 nor the CAN region is individually required for the
cAMP-dependent response of the StAR promoter. On the
other hand, GATA-4 may account for 20–30% of the cAMP
response of the murine StAR promoter.
Mutation of multiple elements affects basal activity of the
StAR promoter
As mutations in the SF-1- and GATA-4-binding site indi-
vidually affected StAR basal and cAMP-dependent promoter
activity, respectively, as well as the close proximity of the two
binding sites, we introduced mutations that abolished both
SF-1 and GATA-4 binding into Ϫ254 StAR promoter con-
struct and tested the effect on promoter activity in transiently
transfected MA-10 cells (Fig. 6B). The overall basal activity
FIG. 5. GATA-4 in MA-10 nuclear extracts binds the highly con-
served region (Ϫ68/Ϫ44) of the mouse StAR promoter. EMSA was
used to examine binding of MA-10 nuclear extract to radiolabeled
GATA probe, which represents the Ϫ68/Ϫ44 region of the StAR pro-
moter. Binding reactions were incubated for 30 min on ice with 5 ␮g
nuclear extracts prepared from unstimulated (lanes 1–11) or
(Bu)2cAMP-treated (lanes 12 and 13) MA-10 cells. For cold competi-
tion or antibody interference analysis, a 20-fold molar excess of the
indicated cold probe, denoted by cc (lanes 2, 4, 6, and 10), or 4 ␮g
GATA-1 monoclonal antibody (lanes 8 and 9) or GATA-4 polyclonal
antibody (lanes 11–13) were included in a preincubation with nuclear
extracts; then the radiolabeled probe was added, and the incubation
was continued for 30 min on ice. Lanes 14 and 15 represent probe
alone or probe and antibody minus nuclear extract, respectively. The
protein-DNA complexes were resolved on a nondenaturing 4% poly-
acrylamide gel, the gel was dried, and the complexes were visualized
by autoradiography. The radiolabeled probes are denoted as GATA for
the Ϫ68/Ϫ44 region, GATA mut for the Ϫ68/Ϫ44 region containing
mutations in the GATA core sequence, and SREBP mut for the Ϫ68/
Ϫ44 region containing mutations in the putative SREBP element. The
sequences for these probes are all shown in Fig. 2B. The con. GATA
probe represents the 20-mer consensus oligonucleotide for GATA that
was used as a positive control for GATA-binding proteins. The arrows
indicate the supershift with GATA-4 Ab (SS) and the free probe; the
open arrowhead represents the specific complex.
1350 SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS Endo • 2000
Vol 141 • No 4
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
of StAR promoter was dramatically decreased (88%) com-
pared with that of the Ϫ254 StAR promoter construct (WT),
indicating that mutation of both SF1–3 and GATA had an
additive effect on promoter strength. Despite minimal basal
activity, the promoter caused a 4.8-fold increase in the cAMP-
dependent response. However, this represents only 45% of
the cAMP response for the Ϫ254 StAR promoter construct.
Thus, the double mutation has a greater effect than the single
GATA mutation on the cAMP response. Therefore, we ex-
amined the possibility that either the SF1–3 or GATA-4 site
acts functionally with the AP-1 site to promote the cAMP
response (Fig. 6B). Double mutations in the SF-1 and AP-1
elements (SF1–3/AP-1) or the GATA-4 and AP-1 (GATA/
AP-1) elements did not result in any greater effect on StAR
promoter activity compared with an additive effect of the
single mutations alone. The basal activities of SF1–3mut/
AP-1mut and GATAmut/AP-1mut were 16% (AP-1 plus
SF-1) and 23% (SF-1 plus GATA) that of the Ϫ254 StAR
promoter construct, respectively, whereas the (Bu)2cAMP-
dependent increases were 90% (SF1–3 effect) and 64%
(GATA-4 effect) that of the Ϫ254 StAR. Thus, it appears that
mutation of the AP-1 element has the single greatest effect on
StAR promoter activity, due mainly to decreased basal ex-
pression, whereas the SF1–3 and GATA-4 double mutations
had the most severe effect on promoter function. One pos-
sibility is that SF1–3 and GATA-4 function to stabilize a
common factor(s) in the CAN (Ϫ87/Ϫ64) region. To test this
proposal, we examined the effects of abolishing one DNA-
binding site on protein-DNA interactions at other sites
within the Ϫ105/Ϫ44 bp of StAR promoter.
Protein-DNA interactions within the cAMP-responsive
regions of the StAR promoter
Four major protein-DNA complexes (I–IV) were detected
by EMSA analysis using a DNA probe that spans the Ϫ105
to Ϫ44 region (Ϫ105/Ϫ44) of the StAR promoter (Fig. 7A).
Complex formation appeared to be independent of cAMP
treatment in that similar complexes were formed with nu-
clear extracts from both control and (Bu)2cAMP-treated
MA-10 cells. Antibodies to SF-1, GATA-4, and C/EBP␤ were
used to help identify the presence of these proteins in the four
complexes (Fig. 7B). The SF-1 antibody abolished complex I
and diminished complex III (lane 2), whereas GATA-4 an-
tibodies caused the appearance of a supershifted complex
concomitant with diminished complex II (lane 3). C/EBP␤
antibodies had no effect on protein-DNA interactions within
this region of the StAR promoter (lane 4). Cold competition
experiments with the oligonucleotides corresponding to the
SF1–3 (Ϫ105/Ϫ44), CAN (Ϫ87/Ϫ64), and GATA (Ϫ68/Ϫ44)
regions of the promoter were performed to verify the con-
tribution of each site to complex formation. First, cold com-
petition with unlabeled Ϫ105/Ϫ44 probe greatly diminished
complex formation (lane 5). The SF1–3 probe eliminated com-
plex I and diminished complexes III and IV without affecting
complex II formation (lane 6), whereas the GATA probe
competed for complex II formation without affecting com-
plex I, III, or IV (lane 9). The CAN probe eliminated complex
IV (lane 7), whereas the consensus C/EBP␤ oligonucleotide
(lane 8) had no effect on the protein-DNA interactions with
the StAR promoter.
To further assess complex composition, a series of com-
plementary experiments was performed in which the same
sequence mutations used in the functional studies were in-
troduced into the Ϫ105/Ϫ44 probe, and the effects on pro-
tein-DNA interactions were determined (Fig. 7C). Complex
I was abolished, and complexes III and IV were diminished
by mutation of the SF1–3 site (SF1–3mut, lane 2), which is
consistent with the results of the SF-1 antibody and cold
competition experiments. Complex II was not affected by the
SF1–3 mutation, and addition of the GATA-4 antibody
caused a supershift of the remaining complex II (lane 3).
Conversely, mutation of the GATA-4 site eliminated com-
plex II formation (GATAmut, lane 4), and the remaining
complexes (I, III, and IV) were greatly diminished by SF-1
antibodies (lane 5). The CAN probe was an effective com-
FIG. 6. Mutational analysis reveals regulatory regions of mouse
StAR. MA-10 cells were cotransfected with the indicated StAR pro-
moter-luciferase reporter gene constructs and pCMV-␤-galactosidase
as detailed in Materials and Methods. The transfected cells were
treated in triplicate with fresh Waymouth’s medium in either the
absence (basal) or presence of 1 mM (Bu)2cAMP for 16–19 h, and
luciferase activity was determined and normalized to ␤-galactosidase
expression (RLU/b-gal). For each experiment, the data were ex-
pressed as a percentage of the Ϫ254 StAR promoter basal activity,
which was set at 100%. Graphed are the mean Ϯ SEM values from
three to six experiments for the relative luciferase activity of StAR
promoter containing mutations in either a single element (A) or two
elements (B). The fold induction values represent the ratio of
(Bu)2cAMP/basal activity for each construct. Shown are the mean Ϯ
SEM values for three to five experiments. *, Statistically significant
(P Ͻ 0.05) difference relative to the fold induction of the Ϫ254 StAR
promoter construct in response to (Bu)2cAMP. WT, Wild-type Ϫ254/
ϩ35 StAR promoter-luciferase construct; pGL2-basic, empty vector;
NT, nontransfected.
SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS 1351
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
petitor for complexes III and IV that remained with the
GATAmut probe (lane 6) probe, but did not further reduce
complex formation with the AP-1mut probe (lane 7), sug-
gesting that factors binding to the CAN region contribute to
formation of these complexes. Indeed, mutation of the AP-
1-like site eliminated complex IV and surprisingly also re-
duced all other protein-DNA interactions (lane 10). The re-
maining complexes I and III were eliminated by the addition
of SF-1 antibodies (lane 9), whereas GATA-4 antibodies re-
sulted in a supershift of complex II (lanes 8).
FIG. 7. The SF1–3- and GATA-4-binding sites are essential for the formation of multiple complexes within the Ϫ105/Ϫ44 region of StAR. EMSA
was used to examine the binding of MA-10 nuclear extract to the radiolabeled Ϫ105/Ϫ44 region of the StAR promoter. A, Concentration-
dependent protein-DNA complex formation. Fifty femtomoles of radiolabeled probe (Ϫ105/Ϫ44) were incubated with 5, 10, or 15 ␮g nuclear
extracts from untreated (control; lanes 1–3) or (Bu)2cAMP-treated [(Bu)2; lanes 4–6] MA-10 cells. The arrows indicate the positions of four
protein-DNA complexes, denoted I–IV. Free probe is not shown on this gel. B, Cold competition or antibody interference analysis. SF-1, GATA-4,
or C/EBP␤ antibody (4 ␮g) or the indicated cold competitor probes were incubated with 10 ␮g nuclear extracts [(Bu)2cAMP-treated]; then the
radiolabeled Ϫ105/Ϫ44 probe was added, and the incubation was continued for 30 min on ice. The protein-DNA complexes were resolved on
a nondenaturing 4% polyacrylamide gel, the gel was dried, and the complexes were visualized by autoradiography. The cold competitors were
Ϫ105/Ϫ44, SF1–3 (Ϫ105/Ϫ83), CAN (Ϫ87/Ϫ64 region), GATA (Ϫ68/Ϫ44 region), and consensus C/EBP. Arrows indicate complexes I–IV, and
the open arrowhead indicates the GATA-4 supershifted complex (SS). C and D, Effects of mutation of an individual element (C) or two elements
(D) on protein-DNA interactions. The sequences for the mutations that were introduced into the Ϫ105/44 probe are given in Table 2B. The
radiolabeled probes used in the binding reaction are indicated: Ϫ105/Ϫ44, SF1–3 mut, GATA mut, and AP-1 mut, or their combination (D).
Experiments in the presence of antibodies or cold competitor CAN probe were performed as described in B.
1352 SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS Endo • 2000
Vol 141 • No 4
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
As StAR basal promoter activity was greatly reduced by
mutations within two of the three SF1–3, CAN, or GATA
elements (Fig. 6B), we also tested the effects of these muta-
tions on protein-DNA interactions (Fig. 7D). A double mu-
tation in SF-1 and GATA (SF1–3/GATA mut) resulted in the
loss of all complex formation (Fig. 7D, lane 2), whereas com-
plex II was still present with the probe containing a double
mutation in SF1–3 and AP-1 (SF1–3/AP-1 mut, lane 3). Dou-
ble mutations of GATA and AP-1 caused a marked decrease
in protein-DNA interactions for complexes III, and IV and
eliminated complex II (lane 4). These data are consistent with
an additive effect of the single mutations on protein-DNA
interactions, with the exception that complex I did not appear
affected by the GATAmut/AP-1mut probe. This was not an
expected result based on the decrease in complex I with the
AP-1 mut probe (Fig. 7C, compare lanes 10 and 11). These
results indicate that complexes III and IV are dependent
upon SF-1 and possibly, to a lesser extent, GATA-4.
In sum, the individual mutations in the SF1–3 or AP-1-like
regions and the SF-1 antibodies do not affect GATA-4 bind-
ing; therefore, we conclude that GATA-4 binding within the
Ϫ105/Ϫ44 region is independent of other factors. Further,
the GATA-4 mutation does not prevent SF-1-dependent com-
plex formation (I, III, and IV), indicating that SF-1 binding is
not dependent on GATA-4 binding. CAN factors appear to
contribute to complex IV formation, as mutation of the AP-
1-like element or cold competition with the CAN region
eliminated this complex. However, protein binding within
the CAN region in the context of the longer probe appears
to be dependent on SF-1 binding to SF1–3 and may be in-
fluenced by GATA-4 binding. Conversely, an intact AP-1
element appears to be required for efficient SF-1 binding,
indicating that protein binding to SF1–3 and CAN regions
may be interdependent. Together, these data clearly dem-
onstrate that complex I is due to SF-1 binding, and complex
II is due to GATA-4 binding. The data are consistent with
complexes III and IV being dependent upon SF-1, with com-
plex III most likely containing SF-1, whereas CAN factor
binding contributes to complex IV.
Discussion
These studies have identified two regulatory regions of the
StAR promoter: a negative region located between Ϫ254/
Ϫ150, and a cAMP-responsive region located between
Ϫ105/Ϫ60. Putative negative regulatory regions were pre-
viously identified for the rat and porcine promoters as well
(21, 39). Our promoter deletion analysis has eliminated SF-1
(SF-1–1) and C/EBP␤ binding at Ϫ135 and Ϫ113 as essential
for the cAMP response of the StAR gene and has pointed to
SF1–3 (Ϫ95), CAN-binding protein(s) (Ϫ87/Ϫ68), and
GATA-4 (Ϫ68) as potential factors.
Identification of the Ϫ105 to Ϫ95 region as a SF-1-binding
site was an expected result, as this element was previously
shown to bind SF-1 in the human and rat StAR promoters (21,
37). However, these are the first studies to show a distinction
between regulations of the human and mouse StAR pro-
moter by cAMP in steroidogenic cell lines. Mutation of the
SF-1 site (Ϫ95) in the human promoter demonstrated 90%
and 80% decreases in basal and forskolin-stimulated pro-
moter activity in human granulosa-lutein cells (37). Our re-
cent studies in the human H295R adrenocortical cells also
have shown that the SF-1 element at Ϫ95 is required for
cAMP and angiotensin II induction of human StAR promot-
er-luciferase reporter gene expression (23). Transient trans-
fection of COS-1 cells with the rat StAR promoter confirmed
a SF-1-dependent activation of the StAR promoter that re-
quired the SF-1 site at Ϫ95 and that mutation of this site
resulted in a decrease in the cAMP response that was due to
lowered basal promoter activity (21). Our results demon-
strate that the mouse and rat promoters are functionally more
similar to each other than to the human StAR promoter. This
apparent promoter-specific difference may be reflective of a
divergence within the StAR promoter sequences between the
human and the mouse. The human sequence has an addi-
tional five bases downstream of the SF-1 site and does not
contain the CAAT box, the the AP-1-like site, or the nuclear
receptor half-site that have been shown to be important for
the murine promoter in MA-10 cells (26, 38). Thus, it is
apparent that SF-1 is important in StAR gene regulation but
is not sufficient for the cAMP-dependent response in MA-10
cells. Possibly, the sequence divergence reflects greater in-
teractions of SF-1 with other factors binding in this region of
the mouse StAR promoter.
We have confirmed the functional importance of the AP-
1-like element within the Ϫ87/Ϫ68 (CAN) region of mouse
StAR promoter for basal transcriptional activity (26, 38). In
addition, this study provides the first evidence that
(Bu)2cAMP treatment of MA-10 cells results in the formation
of a unique protein-DNA complex at this region of the StAR
promoter (complex B). Identification of the CAN factor(s)
should provide an important link to elucidating the mech-
anism of action for cAMP-dependent regulation of StAR.
Presently, our data suggest that multiple elements may con-
tribute to activation of the StAR promoter. Double mutations
in either the SF1–3 and AP-1 or GATA and AP-1 elements
have a greater effect on basal promoter activity that repre-
sents the combined effects of the single mutations. The
GATAmut/AP-1 mut also resulted in decreased cAMP re-
sponse that is attributed to the GATA element. On the other
hand, the effect of the SF1–3 and GATA double mutation on
basal promoter activity was greater than the combined ef-
fects of the single mutations. As mutations that disrupted
the AP-1-like site (AP-1mut, SF1–3mut/AP-1mut, and
GATAmut/AP-1mut) have similar effects on basal activity as
the SF1–3mut/GATAmut, these data suggest that all three
elements work together to promote full basal activity of the
StAR promoter that, in turn, is required for a maximal cAMP
response. What is not clear at the time is why the cAMP-
dependent response, although reduced, remains intact de-
spite apparent elimination of protein-DNA binding interac-
tions within the Ϫ105/Ϫ44 region of StAR promoter with the
SF1–3mut/GATAmut. As we did not observe a cAMP-
dependent complex with the Ϫ105/Ϫ44 probe, the cAMP-
dependent CAN-binding factor(s) may not have been de-
tected in this analysis. Indeed, the cAMP-dependent complex
was observed only with increased concentrations of nuclear
extract, suggesting that this factor(s) may be expressed in
lesser abundance. Thus, it is possible that the AP-1 region is
still functional in the SF1–3mut/GATAmut double mutation.
SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS 1353
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
Previously, we demonstrated that a protein(s) in MA-10
nuclear extracts binds to the CAAT box located in the Ϫ93/
Ϫ71 region, but with lower affinity compared with the
C/EBP␤ site located at Ϫ113 (26). Antibody supershift EMSA
experiments showed that C/EBP␤ binds to the Ϫ113 site, but
binding to the proximal site was not tested. We now have
directly tested C/EBP␤ binding to the CAN region and verify
that C/EBP␤ does not bind to this region of the StAR pro-
moter. It is possible that our previous studies did not detect
the factor(s) we now observe because the probe used pre-
viously (Ϫ93/Ϫ71) did not contain the entire CAN region
(Ϫ87/Ϫ64) binding sites, which may have affected protein
binding to this region. Indeed, complexes A and B appear to
be dependent upon an intact AP-1-like element and the flank-
ing TGATGA sequence. Functionally, mutation of the puta-
tive AP-1 site resulted in approximately a 63% decrease in the
basal promoter activity of StAR without affecting the cAMP
response, a result very similar to the effects seen by mutating
the CAAT box at Ϫ87 (26). Therefore, mutation of the CAAT
box most likely affects a factor(s) binding to the AP-1-like
element in the CAN region or vice versa.
Our results in MA-10 cells are distinct from the recent
report of murine StAR promoter activity in rat ovarian cells,
which demonstrated that C/EBP␤ is a prominent component
in a DNA-protein complex formed with the Ϫ87/Ϫ64 region
(38). Interestingly, the binding site recognized by C/EBP␤ in
ovarian cell extracts is not the CAAT box core, but is the
AP-1-like element. One possibility for this apparent tissue-
specific difference in C/EBP␤ binding is that different factors
bind to the CAN region. This tissue-specific difference may
reflect the cycloheximide sensitivity of the cAMP response
for StAR. Previously, StAR gene expression was reported to
be mediated by a cycloheximide-sensitive mechanism in hu-
man granulosa-lutein cells (40). C/EBP␤ expression has been
shown to be increased by FSH in ovarian granulosa cells and
by (Bu)2cAMP in MA-10 cells (25, 38). Subsequent to sub-
mission of this paper, it was reported that C/EBP␤ is induced
in human granulosa cells by 8-bromo-cAMP (41). The data
supported C/EBP␤ in StAR basal promoter activity as well
as the proposal that the cycloheximide-sensitive, cAMP-
dependent response may be linked to the induction of
C/EBP␤. Thus, it is probable that a SF-1/C/EBP␤ or a
GATA-4/C/EBP␤ interaction mediates the observed cyclo-
heximide-sensitive response in rat ovarian granulosa cells.
Consistent with this proposal, C/EBP␤ and SF-1 were shown
to interact directly in a GST pull-down assay (26). However,
StAR gene expression in MA-10 cells and Y1 mouse adre-
nocortical cells is not dependent upon de novo protein syn-
thesis (10). Therefore, another factor(s) that is insensitive to
cycloheximide may be used in MA-10 Leydig cells and Y1
cells to bind to the CAN region and promote maximal basal
activation of StAR. A difference in tissue-specific transcrip-
tion factor expression may thus account for differences in the
acute regulation of StAR in granulosa vs. Leydig cell cultures.
We identified GATA-4 as the factor that binds to the highly
conserved Ϫ68/Ϫ44 region of the mouse StAR promoter.
Functionally, GATA-4 contributes to 20% of the cAMP re-
sponse. Independently, GATA-4 was recently reported to
bind to the same region and was shown to be required for
maximal cAMP response in rat granulosa cells (38). Mutation
of both the GATA-4 and AP-1-like elements decreased StAR
basal promoter activity 97% and dramatically decreased the
FSH-dependent increase in reporter gene expression in lu-
teal-granulosa cells. In contrast, we demonstrate that a sim-
ilar double mutation (GATAmut/AP1mut) had no greater
effect on StAR promoter activity in MA-10 cells than the
combined effects of the individual mutations within these
elements. Again, this discrepancy between our results and
those reported for rat granulosa cells indicates that the mu-
rine StAR gene may be regulated in a tissue-specific manner.
In MA-10 cells, we propose the GATA-4 transcription factor
has a role in stabilizing the factor(s) bound to the putative
AP-1 site and that GATA-4 and SF-1 work together to sta-
bilize this common factor(s). However, these putative CAN-
binding proteins and SF-1 can function in the absence of
GATA-4.
In sum, SF-1 binding is important for the basal activity of
the StAR gene, most likely by affecting the interactions
among the other proteins within the CAN region of the
promoter. GATA-4 binding is independent of SF-1 binding
and has an apparently minimal effect on SF-1- and CAN-
protein binding, but may influence factor binding. Interac-
tion between SF-1 and GATA-4 has been shown for regula-
tion of the Mu¨llerian-inhibiting substance gene; therefore,
one model for SF-1 and GATA-4 is that they interact and
function to stabilize binding of the CAN-binding factor(s)
(42). Alternatively, it is possible that there is a redundancy
of function for SF-1, CAN-binding protein, and GATA-4.
This proposal would be consistent with our mutational anal-
ysis that shows that elimination of one binding factor does
not compromise the cAMP response of the StAR promoter.
Thus, each of the DNA-binding factors may work together to
stabilize the unknown cAMP-mediated transcription factors
and/or coactivators, but each factor alone may be sufficient
to mediate part of the response.
Acknowledgments
We thank Dr. Keith Parker, University of Texas Southwestern Med-
ical Center (Dallas, TX), for providing the GST-SF-1 expression plasmid.
We also thank Drs. Carolyn M. Klinge and Keith C. Falkner, Department
of Biochemistry and Molecular Biology, University of Louisville School
of Medicine (Louisville, KY), for their critical review of this manuscript.
Finally, we thank Ms. Rebecca Combs for her excellent technical
assistance.
References
1. Stone D, Hechter O 1954 Studies on ACTH action in perfused bovine adrenals:
site of action of ACTH in corticosteroidogenesis. Arch Biochem Biophys
51:457–469
2. Karaboyas G, Koritz S 1965 Identity of the site of action of cAMP and ACTH
in corticosteroidogenesis in rat adrenal and beef adrenal cortex slices. Bio-
chemistry 4:462–468
3. Simpson ER, Jefcoate CR, Brownie AC, Boyd GS 1972 The effect of ether
anaesthesia stress on cholesterol-side-chain cleavage and cytochrome P450 in
rat-adrenal mitochondria. Eur J Biochem 28:442–450
4. Privalle CT, Crivello JF, Jefcoate CR 1983 Regulation of intramitochondrial
cholesterol transfer to side-chain cleavage cytochrome P-450 in rat adrenal
gland. Proc Natl Acad Sci USA 80:702–706
5. Stocco DM, Clark BJ 1996 Regulation of the acute production of steroids in
steroidogenic cells. Endocr Rev 17:221–244
6. Caron KM, Soo SC, Wetsel WC, Stocco DM, Clark BJ, Parker KL 1997
Targeted disruption of the mouse gene encoding steroidogenic acute regula-
tory protein provides insights into congenital lipoid adrenal hyperplasia. Proc
Natl Acad Sci USA 94:11540–11545
7. Lin D, Sugawara T, Strauss III JF, Clark BJ, Stocco DM, Saenger P, Rogol A,
1354 SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS Endo • 2000
Vol 141 • No 4
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
Miller WL 1995 Role of steroidogenic acute regulatory protein in adrenal and
gonadal steroidogenesis. Science 267:1828–1831
8. Miller WL 1997 Congenital lipoid adrenal hyperplasia: the human gene knock-
out for the steroidogenic acute regulatory protein. J Mol Endocrinol 19:227–240
9. Clark BJ, Combs R, Hales KH, Hales DB, Stocco DM 1997 Inhibition of
transcription affects synthesis of steroidogenic acute regulatory protein and
steroidogenesis in MA-10 mouse Leydig tumor cells. Endocrinology
138:4893–4901
10. Caron KM, Ikeda Y, Soo SC, Stocco DM, Parker KL, Clark BJ 1997 Charac-
terization of the promoter region of the mouse gene encoding the steroidogenic
acute regulatory protein. Mol Endocrinol 11:138–147
11. Lala DS, Rice DA, Parker KL 1992 Steroidogenic factor I, a key regulator of
steroidogenic enzyme expression, is the mouse homolog of fushi tarazu-factor
I. Mol Endocrinol 6:1249–1258
12. Honda S, Morohashi K, Nomura M, Takeya H, Kitajima M, Omura T 1993
Ad4BP regulating steroidogenic P-450 gene is a member of steroid hormone
receptor superfamily. J Biol Chem 268:7494–7502
13. Morohashi K, Zanger UM, Honda S, Hara M, Waterman MR, Omura T 1993
Activation of CYP11A and CYP11B gene promoters by the steroidogenic cell-
specific transcription factor, Ad4BP. Mol Endocrinol 7:1196–1204
14. Luo X, Ikeda Y, Parker KL 1994 A cell-specific nuclear receptor is essential for
adrenal and gonadal development and sexual differentiation. Cell 77:481–490
15. Wong M, Rice DA, Parker KL, Schimmer BP 1989 The roles of cAMP and
cAMP-dependent protein kinase in the expression of cholesterol side chain
cleavage and steroid 11␤-hydroxylase genes in mouse adrenocortical tumor
cells. J Biol Chem 264:12867–12871
16. Parissenti AM, Parker KL, Schimmer BP 1993 Identification of promoter
elements in the mouse 21-hydroxylase (Cyp21) gene that require a functional
cyclic adenosine 3Ј,5Ј-monophosphate- dependent protein kinase. Mol Endo-
crinol 7:283–290
17. Zhang P, Mellon SH 1996 The orphan nuclear receptor steroidogenic factor-1
regulates the cyclic adenosine 3Ј,5Ј-monophosphate-mediated transcriptional
activation of rat cytochrome P450c17 (17␣-hydroxylase/c17–20 lyase). Mol
Endocrinol 10:147–158
18. Carlone DL, Richards JS 1997 Evidence that functional interactions of CREB
and SF-1 mediate hormone regulated expression of the aromatase gene in
granulosa cells and constitutive expression in R2C cells. J Steroid Biochem Mol
Biol 61:223–231
19. Jacob AL, Lund J 1998 Mutations in the activation function-2 core domain of
steroidogenic factor-1 dominantly suppresses PKA-dependent transactivation
of the bovine CYP17 gene. J Biol Chem 273:13391–13394
20. Hammer GD, Krylova I, Zhang Y, Darimont BD, Simpson K, Weigel NL,
Ingraham HA 1999 Phosphorylation of the nuclear receptor SF-1 modulates
cofactor recruitment: integration of hormone signaling in reproduction and
stress. Mol Cell 3:521–526
21. Sandhoff TW, Hales DB, Hales KH, McLean MP 1998 Transcriptional reg-
ulation of the rat steroidogenic acute regulatory protein gene by steroidogenic
factor 1. Endocrinology 139:4820–4831
22. Sugawara T, Kiriakidou M, McAllister JM, Kallen CB, Strauss III JF 1997
Multiple steroidogenic factor 1 binding elements in the human steroidogenic
acute regulatory protein gene 5Ј-flanking region are required for maximal
promoter activity and cyclic AMP responsiveness. Biochemistry 36:7249–7255
23. Clark BJ, Combs R 1999 Angiotensin II and cAMP induce human steroido-
genic acute regulatory protein transcription through a common steroidogenic
factor-1 element. Endocrinology 140:4390–4397
24. Lekstrom-Himes J, Xanthopoulos KG 1998 Biological role of the CCAAT/
enhancer-binding protein family of transcription factors. J Biol Chem
273:28545–28548
25. Nalbant D, Williams SC, Stocco DM, Khan SA 1998 Luteinizing hormone-
dependent gene regulation in Leydig cells may be mediated by CCAAT/
enhancer-binding protein-␤. Endocrinology 139:272–279
26. Reinhart AJ, Williams SC, Clark BJ, Stocco DM 1999 SF-1 (steroidogenic
factor-1) and C/EBP␤ (CCAAT/enhancer binding protein-␤) cooperate to
regulate the murine StAR (steroidogenic acute regulatory) promoter. Mol
Endocrinol 13:729–741
27. Mukai K, Mitani F, Agake R, Ishimura Y 1998 Adrenocorticotropic hormone
stimulates CYP11B1 gene transcription through a mechanism involving AP-1
factors. Eur J Biochem 256:190–200
28. Sun Y, Duckworth ML 1999 Identification of a placental-specific enhancer in
the rat placental lactogen II gene that contains binding sites for members of the
Ets and AP-1 (activator protein 1) families of transcription factors. Mol En-
docrinol 13:385–399
29. Duval DL, Ellsworth BS, Clay CM 1999 Is gonadotrope expression of the
gonadotropin releasing hormone receptor gene mediated by autocrine/para-
crine stimulation of an activin response element? Endocrinology
140:1949–1952
30. Orkin SH 1992 GATA-binding transcription factors in hematopoietic cells.
Blood 80:575–581
31. Yomogida K, Ohtani H, Harigae H, Ito E, Nishimune Y, Engel JD, Yamamoto
M 1994 Developmental stage- and spermatogenic cycle-specific expression of
transcription factor GATA-1 in mouse Sertoli cells. Development
120:1759–1766
32. Morrisey EE, Ip HS, Lu MM, Parmacek MS 1996 GATA-6: a zinc finger
transcription factor that is expressed in multiple cell lineages derived from
lateral mesoderm. Dev Biol 177:309–322
33. Heikinheimo M, Ermolaeva M, Bielinska M, Rahman NA, Narita N,
Huhtaniemi IT, Tapanainen JS, Wilson DB 1997 Expression and hormonal
regulation of transcription factors GATA-4 and GATA-6 in the mouse ovary.
Endocrinology 138:3505–3514
34. Feng ZM, Wu AZ, Chen CL 1998 Testicular GATA-1 factor up-regulates the
promoter activity of rat inhibin ␣-subunit gene in MA-10 Leydig tumor cells.
Mol Endocrinol 12:378–390
35. Ketola I, Rahman N, Toppari J, Bielinska M, Porter-Tinge SB, Tapanainen
JS, Huhtaniemi IT, Wilson DB, Heikinheimo M 1999 Expression and regu-
lation of transcription factors GATA-4 and GATA-6 in developing mouse
testis. Endocrinology 140:1470–1480
36. Dignam JD, Lebovitz RM, Roeder RG 1983 Accurate transcription initiation
by RNA polymerase II in a soluble extract from isolated mammalian nuclei.
Nucleic Acids Res 11:1475–1489
37. Sugawara T, Holt JA, Kiriakidou M, Strauss III JF 1996 Steroidogenic factor
1-dependent promoter activity of the human steroidogenic acute regulatory
protein (StAR) gene. Biochemistry 35:9052–9059
38. Silverman E, Eimerl S, Orly J 1999 CCAAT enhancer-binding protein ␤ and
GATA-4 binding regions within the promoter of the steroidogenic acute reg-
ulatory protein (StAR) gene are required for transcription in rat ovarian cells.
J Biol Chem 274:17987–17996
39. LaVoie HA, Garmey JC, Veldhuis JD 1999 Mechanisms of insulin-like growth
factor I augmentation of follicle-stimulating hormone-induced porcine steroi-
dogenic acute regulatory protein gene promoter activity in granulosa cells.
Endocrinology 140:146–153
40. Kiriakidou M, McAllister JM, Sugawara T, Strauss III JF 1996 Expression of
steroidogenic acute regulatory protein (StAR) in the human ovary. J Clin
Endocrinol Metab 81:4122–4128
41. Christenson LK, Johnson PF, McAllister JM, Strauss III JF 1999 CCAAT/
enhancer-binding proteins regulate expression of the human steroidogenic
acute regulatory protein (StAR) gene. J Biol Chem 274:26591–26598
42. Tremblay JJ, Viger RS 1999 Transcription factor GATA-4 enhances Mullerian
inhibiting substance gene transcription through a direct interaction with the
nuclear receptor SF-1. Mol Endocrinol 13:1388–1401
SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS 1355
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.

More Related Content

What's hot

Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...
Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...
Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...Dougan McGrath
 
Signal transduction proteins and pathways in oncogenesis
Signal transduction proteins and pathways in oncogenesisSignal transduction proteins and pathways in oncogenesis
Signal transduction proteins and pathways in oncogenesisShashidhara TS
 
Phosphoinositide 3 kinase
Phosphoinositide 3 kinasePhosphoinositide 3 kinase
Phosphoinositide 3 kinaseShijina Nair
 
RECEPTOR SERINE THREONINE KINASE
RECEPTOR SERINE THREONINE KINASERECEPTOR SERINE THREONINE KINASE
RECEPTOR SERINE THREONINE KINASEBidhan Sarkar
 
Fat and Beyond: The Diverse Biology of PPAR gamma
Fat and Beyond: The Diverse Biology of PPAR gammaFat and Beyond: The Diverse Biology of PPAR gamma
Fat and Beyond: The Diverse Biology of PPAR gammaRamesh Pothuraju
 
Signaling Pathways
Signaling PathwaysSignaling Pathways
Signaling PathwaysEuplectes
 
PPAR alpha poster
PPAR alpha posterPPAR alpha poster
PPAR alpha posterEric Cobb
 
4) Signaling, Ola Elgaddar
4) Signaling, Ola Elgaddar4) Signaling, Ola Elgaddar
4) Signaling, Ola ElgaddarOla Elgaddar
 
Signalling mechanism in cell growth
Signalling mechanism in cell growthSignalling mechanism in cell growth
Signalling mechanism in cell growthbijitadutta123
 
Unit 2 & 4 GPCRs WHOLE UNITS (GENERAL PHARMACOLOGY)
Unit  2 & 4 GPCRs WHOLE UNITS (GENERAL PHARMACOLOGY)Unit  2 & 4 GPCRs WHOLE UNITS (GENERAL PHARMACOLOGY)
Unit 2 & 4 GPCRs WHOLE UNITS (GENERAL PHARMACOLOGY)Priyansha Singh
 
sugar sensing and signaling in plants
sugar sensing and signaling in plantssugar sensing and signaling in plants
sugar sensing and signaling in plantsAnjali Dahiya
 
Stat3 protein in psoriasis by yousry
Stat3 protein in psoriasis  by yousryStat3 protein in psoriasis  by yousry
Stat3 protein in psoriasis by yousryM.YOUSRY Abdel-Mawla
 
Cancer signal-transduction
Cancer signal-transductionCancer signal-transduction
Cancer signal-transductionDeepika Tripathi
 

What's hot (19)

CDD 2016
CDD 2016CDD 2016
CDD 2016
 
Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...
Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...
Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...
 
Signal transduction proteins and pathways in oncogenesis
Signal transduction proteins and pathways in oncogenesisSignal transduction proteins and pathways in oncogenesis
Signal transduction proteins and pathways in oncogenesis
 
Phosphoinositide 3 kinase
Phosphoinositide 3 kinasePhosphoinositide 3 kinase
Phosphoinositide 3 kinase
 
RECEPTOR SERINE THREONINE KINASE
RECEPTOR SERINE THREONINE KINASERECEPTOR SERINE THREONINE KINASE
RECEPTOR SERINE THREONINE KINASE
 
PI3 kinase pathway
PI3 kinase pathwayPI3 kinase pathway
PI3 kinase pathway
 
Fat and Beyond: The Diverse Biology of PPAR gamma
Fat and Beyond: The Diverse Biology of PPAR gammaFat and Beyond: The Diverse Biology of PPAR gamma
Fat and Beyond: The Diverse Biology of PPAR gamma
 
Signaling Pathways
Signaling PathwaysSignaling Pathways
Signaling Pathways
 
PPAR alpha poster
PPAR alpha posterPPAR alpha poster
PPAR alpha poster
 
Signal Transduction
Signal TransductionSignal Transduction
Signal Transduction
 
4) Signaling, Ola Elgaddar
4) Signaling, Ola Elgaddar4) Signaling, Ola Elgaddar
4) Signaling, Ola Elgaddar
 
Signalling mechanism in cell growth
Signalling mechanism in cell growthSignalling mechanism in cell growth
Signalling mechanism in cell growth
 
Unit 2 & 4 GPCRs WHOLE UNITS (GENERAL PHARMACOLOGY)
Unit  2 & 4 GPCRs WHOLE UNITS (GENERAL PHARMACOLOGY)Unit  2 & 4 GPCRs WHOLE UNITS (GENERAL PHARMACOLOGY)
Unit 2 & 4 GPCRs WHOLE UNITS (GENERAL PHARMACOLOGY)
 
Sukhjinder singh
Sukhjinder singhSukhjinder singh
Sukhjinder singh
 
sugar sensing and signaling in plants
sugar sensing and signaling in plantssugar sensing and signaling in plants
sugar sensing and signaling in plants
 
Surp09 Signaling
Surp09 SignalingSurp09 Signaling
Surp09 Signaling
 
Stat3 protein in psoriasis by yousry
Stat3 protein in psoriasis  by yousryStat3 protein in psoriasis  by yousry
Stat3 protein in psoriasis by yousry
 
Tgf β signalling from cell
Tgf β signalling from cellTgf β signalling from cell
Tgf β signalling from cell
 
Cancer signal-transduction
Cancer signal-transductionCancer signal-transduction
Cancer signal-transduction
 

Similar to StAR

Pathways of intracellular signal transduction
Pathways of intracellular signal transductionPathways of intracellular signal transduction
Pathways of intracellular signal transductionMohamedEramHosen
 
Teloomerase Research Paper
Teloomerase Research PaperTeloomerase Research Paper
Teloomerase Research PaperDawn Robertson
 
Altering_Phenylalanine_67_Serine
Altering_Phenylalanine_67_SerineAltering_Phenylalanine_67_Serine
Altering_Phenylalanine_67_SerineAmber Anger
 
Na f activates map ks and induces apoptosis in odontoblast-like
Na f activates map ks and induces apoptosis in odontoblast-likeNa f activates map ks and induces apoptosis in odontoblast-like
Na f activates map ks and induces apoptosis in odontoblast-likeGanesh Murthi
 
Epidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinaseEpidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinaseGedion Yilma
 
Enzyme linked receptors
Enzyme linked receptorsEnzyme linked receptors
Enzyme linked receptorsFarazaJaved
 
Investigation of Intracellular Signal Transduction in Multiple Endocrine Neop...
Investigation of Intracellular Signal Transduction in Multiple Endocrine Neop...Investigation of Intracellular Signal Transduction in Multiple Endocrine Neop...
Investigation of Intracellular Signal Transduction in Multiple Endocrine Neop...KayvonneFerguson
 
The B30. 2 domain of pyrin, the familial mediterranean fever protein, interac...
The B30. 2 domain of pyrin, the familial mediterranean fever protein, interac...The B30. 2 domain of pyrin, the familial mediterranean fever protein, interac...
The B30. 2 domain of pyrin, the familial mediterranean fever protein, interac...José Luis Moreno Garvayo
 
Translational Control of Autism Spectrum Disorders in Eif4ebp2 knockout Mouse...
Translational Control of Autism Spectrum Disorders in Eif4ebp2 knockout Mouse...Translational Control of Autism Spectrum Disorders in Eif4ebp2 knockout Mouse...
Translational Control of Autism Spectrum Disorders in Eif4ebp2 knockout Mouse...Rey Christian Pacis
 
1-s2.0-S0167488911000772-main
1-s2.0-S0167488911000772-main1-s2.0-S0167488911000772-main
1-s2.0-S0167488911000772-mainAndreas D. Song
 
Rho Kinase Promotes Alloimmune Responses by Regulating the Proliferation and ...
Rho Kinase Promotes Alloimmune Responses by Regulating the Proliferation and ...Rho Kinase Promotes Alloimmune Responses by Regulating the Proliferation and ...
Rho Kinase Promotes Alloimmune Responses by Regulating the Proliferation and ...Federal University of Bahia
 

Similar to StAR (20)

Pathways of intracellular signal transduction
Pathways of intracellular signal transductionPathways of intracellular signal transduction
Pathways of intracellular signal transduction
 
Teloomerase Research Paper
Teloomerase Research PaperTeloomerase Research Paper
Teloomerase Research Paper
 
Altering_Phenylalanine_67_Serine
Altering_Phenylalanine_67_SerineAltering_Phenylalanine_67_Serine
Altering_Phenylalanine_67_Serine
 
Na f activates map ks and induces apoptosis in odontoblast-like
Na f activates map ks and induces apoptosis in odontoblast-likeNa f activates map ks and induces apoptosis in odontoblast-like
Na f activates map ks and induces apoptosis in odontoblast-like
 
journal.pone.0046482.PDF
journal.pone.0046482.PDFjournal.pone.0046482.PDF
journal.pone.0046482.PDF
 
Vaillencourt
VaillencourtVaillencourt
Vaillencourt
 
srep43028
srep43028srep43028
srep43028
 
Epidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinaseEpidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinase
 
J. Biol. Chem.-2015-Maganti-9812-22
J. Biol. Chem.-2015-Maganti-9812-22J. Biol. Chem.-2015-Maganti-9812-22
J. Biol. Chem.-2015-Maganti-9812-22
 
Enzyme linked receptors
Enzyme linked receptorsEnzyme linked receptors
Enzyme linked receptors
 
Investigation of Intracellular Signal Transduction in Multiple Endocrine Neop...
Investigation of Intracellular Signal Transduction in Multiple Endocrine Neop...Investigation of Intracellular Signal Transduction in Multiple Endocrine Neop...
Investigation of Intracellular Signal Transduction in Multiple Endocrine Neop...
 
fujioka2004
fujioka2004fujioka2004
fujioka2004
 
fujioka2004
fujioka2004fujioka2004
fujioka2004
 
The B30. 2 domain of pyrin, the familial mediterranean fever protein, interac...
The B30. 2 domain of pyrin, the familial mediterranean fever protein, interac...The B30. 2 domain of pyrin, the familial mediterranean fever protein, interac...
The B30. 2 domain of pyrin, the familial mediterranean fever protein, interac...
 
Translational Control of Autism Spectrum Disorders in Eif4ebp2 knockout Mouse...
Translational Control of Autism Spectrum Disorders in Eif4ebp2 knockout Mouse...Translational Control of Autism Spectrum Disorders in Eif4ebp2 knockout Mouse...
Translational Control of Autism Spectrum Disorders in Eif4ebp2 knockout Mouse...
 
GW8510 manuscript
GW8510 manuscriptGW8510 manuscript
GW8510 manuscript
 
1-s2.0-S0167488911000772-main
1-s2.0-S0167488911000772-main1-s2.0-S0167488911000772-main
1-s2.0-S0167488911000772-main
 
signal transduction
signal transductionsignal transduction
signal transduction
 
Hippo signal pathway
Hippo signal pathwayHippo signal pathway
Hippo signal pathway
 
Rho Kinase Promotes Alloimmune Responses by Regulating the Proliferation and ...
Rho Kinase Promotes Alloimmune Responses by Regulating the Proliferation and ...Rho Kinase Promotes Alloimmune Responses by Regulating the Proliferation and ...
Rho Kinase Promotes Alloimmune Responses by Regulating the Proliferation and ...
 

StAR

  • 1. Steroidogenic Factor-1 Influences Protein- Deoxyribonucleic Acid Interactions within the Cyclic Adenosine 3؅,5؅-Monophosphate-Responsive Regions of the Murine Steroidogenic Acute Regulatory Protein Gene* CLAVIA R. WOOTON-KEE AND BARBARA J. CLARK Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, Kentucky 40292 ABSTRACT De novo synthesis of the steroidogenic acute regulatory protein (StAR) in response to trophic hormonal stimulation of steroidogenic cells is required for the delivery of cholesterol from the mitochondrial outer membrane to the mitochondrial inner membrane and the cy- tochrome P450 side-chain cleavage enzyme. StAR expression is tran- scriptionally regulated by cAMP-mediated mechanisms, and we have identified a 45-bp region within the mouse promoter that is important for cAMP responsiveness of the gene. This region, located between Ϫ105 and Ϫ60 of the start site of transcription, contains a SF-1- binding site, a highly conserved C/EBP␤ ϪAP-1-nuclear receptor half- site sequences (CAN region), and a GATA-4-binding site. The SF-1 element and CAN region are required for full basal activity, whereas the GATA-4 element may account for 20% of the cAMP response in MA-10 mouse Leydig tumor cells. A cAMP-dependent protein-DNA complex was observed with the CAN region and mutation of a non- consensus AP-1 site within this region greatly diminished promoter strength. Complex protein-DNA interactions within the cAMP re- sponse region (Ϫ105/Ϫ60) were shown to require the SF-1 element (Ϫ95), suggesting that SF-1 is required for protein-DNA interaction at the CAN (Ϫ79) region and maximal activity of the promoter. (En- docrinology 141: 1345–1355, 2000) STEROID HORMONE biosynthesis in the adrenal and gonads is stimulated by trophic hormones via a cAMP- dependent second messenger system. Within minutes of hor- monal stimulation, cholesterol is mobilized to the outer mi- tochondrial membrane and transferred to the mitochondrial inner membrane where it is converted to pregnenolone by the cytochrome P450 side-chain cleavage enzyme (P450scc) (1–4). This acute response is the rate-limiting step in steroi- dogenesis and is dependent upon the de novo synthesis of the steroidogenic acute regulatory protein (StAR) (5). Without a functional StAR protein, cholesterol accumulates in the outer mitochondrial membrane and steroid production ceases. Thus, StAR functions in the transfer of cholesterol to the inner mitochondrial membrane (5). This function of StAR is ele- gantly demonstrated by characterization of StAR knockout mice that lack steroid production (6). These data confirmed that the StAR mutations identified in patients with congen- ital lipoid adrenal hyperplasia are the underlying cause for the disorder (7, 8). Hormonal treatment of steroidogenic cells controls StAR gene expression. Our previous studies have shown that treat- ment of MA-10 mouse Leydig tumor cells with trophic hor- mones or the cAMP analog, (Bu)2cAMP, induces StAR mes- sage within 30 min, and maximal steady state levels are reached within 4 h (9). This induction of StAR gene tran- scription does not require de novo protein synthesis, suggest- ing that posttranslational mechanisms are involved in the acute induction of StAR gene in response to increases in intracellular levels of cAMP (10). Several factors that participate in StAR gene activation have been reported. One factor is the orphan nuclear receptor steroidogenic factor-1 (SF-1). SF-1 has been shown to have a critical role in the regulation of many of the steroid hydrox- ylase genes, and gene knockout studies in mice have shown that SF-1 is critical for development of the gonad and adrenal glands (11–14). Regulation of SF-1 may occur through direct phosphorylation by a protein kinase A (PKA)-dependent pathway. This proposal is supported by studies in which SF-1 trans-activation of the steroid hydroxylase genes was lost in PKA-deficient cell lines (15, 16). Furthermore, poten- tial PKA phosphorylation sites are present in SF-1, and in vitro studies have directly demonstrated a PKA-dependent phosphorylation of SF-1 (17–19). In addition, activation of the mitogen-activated protein kinase pathway has been shown to phosphorylate and active SF-1, supporting a role for phos- phorylation of SF-1 for its function (20). Our initial analysis of two SF-1 sites in the mouse promoter, located at Ϫ45 and Ϫ135 from the transcription start site, indicated that these elements contributed to basal promoter activity but were not essential for the cAMP-dependent induction (10). The in- volvement of a SF-1 site in StAR gene activation was more recently confirmed with the rat promoter (21). Mutation of the SF-1 elements at Ϫ135 and Ϫ95, either individually or in combination, decreased SF-1-dependent reporter gene ex- Received August 9, 1999. Address all correspondence and requests for reprints to: Dr. Barbara J. Clark, Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, Kentucky 40292. E-mail: bjclark@louisville.edu. * This work was supported by NIH Grant DK-51656. 0013-7227/00/$03.00/0 Vol. 141, No. 4 Endocrinology Printed in U.S.A. Copyright © 2000 by The Endocrine Society 1345 The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
  • 2. pression in a nonsteroidogenic cell line cotransfected for SF-1 expression (21). In these studies the (Bu)2cAMP-mediated increase in promoter activity was also depressed, suggesting that SF-1 may mediate part of the cAMP response. However, it is not known what effect the Ϫ95 SF-1 mutation would have in a steroidogenic cell line, where factors other than SF-1 may be required for basal and cAMP-dependent StAR tran- scription. On the other hand, the SF-1 element located at Ϫ105/Ϫ95 in the human promoter was shown to be critical for basal and maximal cAMP responsiveness in human gran- ulosa-luteal cells and in H295R adrenocortical cells (22, 23). Thus, it appears that the role of SF-1 in StAR expression may be promoter and/or tissue specific. C/EBP␤ has also been shown to modulate StAR basal promoter activity. C/EBP␤ is a basic leucine zipper tran- scription factor that is expressed in liver, intestine, lung, and adipose tissues (24). More recently, C/EBP␤ has been iden- tified as the only isoform of C/EBP present in unstimulated primary Leydig cell cultures and MA-10 Leydig cells (25). Two putative CCAAT sites located at Ϫ113 and Ϫ87 in the murine StAR promoter were identified, and C/EBP␤ in MA-10 nuclear extracts was shown to bind to the Ϫ113 el- ement (26). A weak protein-DNA interaction was observed with the Ϫ87 region of the promoter, but the factors were not identified. Mutation of the Ϫ87, but not the Ϫ113, site abol- ished SF-1-dependent trans-activation of StAR-luciferase re- porter gene expression in transfected COS-1 cells (26). How- ever, transient transfection of MA-10 cells with a StAR- luciferase reporter gene construct containing mutations at either the Ϫ87 or Ϫ113 C/EBP␤-binding site did not affect the cAMP-dependent response, but greatly reduced the basal activity of the promoter. A protein-protein interaction be- tween C/EBP␤ and SF-1 was further demonstrated; thus, these studies suggested a possible C/EBP␤-SF-1 interaction that is required for basal promoter activity. Although SF-1 and C/EBP␤ participate in basal expression of StAR, the mechanism for cAMP-dependent induction has not been elucidated. Previously, we narrowed the cAMP- responsive region of murine StAR to Ϫ254 bp of the start site of transcription (10). Sequence analysis of this region iden- tified potential binding sites for transcription factors that have been shown to mediate cAMP-dependent responses in other genes. Two elements that are part of the focus in the present study are a C/EBP␤/nonconsensus activating pro- tein-1/nuclear receptor half-site located between Ϫ87/Ϫ79 (CAN region) and a putative binding site for members of the GATA family of zinc finger transcription factors located be- tween Ϫ68/Ϫ40. Activating protein-1 is composed of either Fos-Jun or Jun-Jun dimers that bind to the consensus se- quence TGA(G/C)TCA and regulates genes either constitu- tively or by cAMP- or Ca2ϩ -mediated mechanisms (27–29). The GATA family of transcription factors regulates gene expression, differentiation, and cell proliferation by binding to the consensus DNA sequence (A/T)GATA(A/G) (30). More recently, three members of the GATA transcription factor family were shown to be expressed in the developing gonads: GATA-1, GATA-4, and GATA-6 (31–33). GATA-1 message has been isolated in MA-10 cells, and GATA-1 was shown to up-regulate the basal promoter activity of the rat inhibin ␣-subunit gene (34). GATA-4 and GATA-6 have also been identified in testicular tissue, and transient expression of GATA-4 was shown to trans-activate an inhibin ␣- promoter/reporter construct in mouse Leydig and granulosa tumor cell lines (35). Thus, a new role is emerging for GATA transcription factors in Leydig cell function. To clarify the role of SF-1 in StAR expression in MA-10 mouse Leydig tumor cells, we extended our previous studies and demonstrated that SF-1 binds to a third element in mouse promoter at Ϫ95. We show that this SF-1 site, denoted SF1–3, is required for full basal activity of the StAR promoter. Ad- ditionally, SF-1 functions in part to stabilize protein-DNA interactions at the C/EBP␤/nonconsensus activating pro- tein-1 (AP-1)/nuclear receptor half-site (CAN) region located at Ϫ79. We also demonstrate that GATA-4 binds to the StAR promoter at Ϫ68 bp and contributes to 20% of the cAMP- dependent induction. We conclude that the SF1–3, CAN, and GATA-4 DNA-binding sites serve critical roles in maintain- ing full basal promoter activity, which, in turn, is necessary for maximal cAMP induction. Materials and Methods Materials N6 ,2Ј-O-(Bu)2cAMP [(Bu)2cAMP)] was purchased from Sigma (St. Louis, MO). Restriction enzymes, Klenow enzyme, T4 DNA ligase, Lu- ciferase Assay System, and pGL2-luciferase reporter vectors were pur- chased from Promega Corp. (Madison, WI). [␥-32 P]ATP was obtained from NEN Life Science Products (Wilmington, DE). Custom oligonu- cleotides were purchased from Genosys (The Woodlands, TX) and Gen- emed Synthesis, Inc. (San Francisco, CA). Glutathione-S-transferase (GST)-SF-1 plasmid was donated by Dr. Keith Parker, University of Texas Southwestern Medical Center (Dallas, TX). Antibodies were pur- chased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA) and Up- state Biotechnology, Inc. (Lake Placid, NY). X-ray film was obtained from Eastman Kodak Co. (Rochester, NY) and NEN Life Science Prod- ucts (Boston, MA). Cell culture The MA-10 mouse Leydig tumor cell line was a gift from Dr. M. Ascoli (Department of Pharmacology, University of Iowa of Medicine, Iowa City, IA). The cells were grown in Waymouth’s MB/752 medium con- taining 15% horse serum and 40 ␮g gentamicin sulfate/ml. Transient transfections and reporter assays MA-10 cells were plated at 300,000 cells/well in a 24-well plate 24 h before transfection. Two micrograms of StAR-luciferase reporter gene plasmid and 2 ␮g pCMV-␤-galactosidase (CMV, cytomegalovirus) ex- pression vector plasmid were cotransfected into cells using 15 ␮g/ml Lipofectamine (Life Technologies, Inc., Grand Island, NY) and Way- mouth’s medium without antibiotics and serum as described previously (10). Twenty-four hours posttransfection, the cells were treated with 1 mm (Bu)2cAMP for 16–19 h. The Promega Corp. Luciferase Assay Sys- tem was used to prepare cell lysates and measure luciferase activity (Lumat LB 9507 luminometer, Wallac, Inc., Gaithersburg, MD). ␤- Galactosidase activity was assayed using ␤-d-galactopyranosidase (Roche Molecular Biochemicals, Indianapolis, IN) as the substrate and measuring the absorbance at 595 nm. Relative light unit (RLU) values were normalized to ␤-galactosidase activity for each sample. Each treat- ment was performed in triplicate, and the mean Ϯ sem were determined. The data were expressed as fold induction relative to the Ϫ254/ϩ35 luc control, which was set at 100. The mean Ϯ sem for the fold induction in response to (Bu)2cAMP for all independent transfection experiments was calculated, and a pooled Student’s t test was performed on the data. P Ͻ 0.05 was considered statistically significant. 1346 SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS Endo • 2000 Vol 141 • No 4 The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
  • 3. StAR-luciferase constructs PCR was used to generate the 5Ј-deletion constructs for the StAR promoter. Linearized Ϫ254/ϩ35 StAR promoter-pGL2-luciferase (Ϫ254 StAR promoter construct) was used as the template. To amplify the region Ϫ150 to ϩ35 (Ϫ150/ϩ35 luc), MluI Ϫ150 and GL2-primers were used in the PCR reaction, and the product was digested with MluI and HindIII and cloned into the MluI and HindIII sites of pGL2-basic. The regions Ϫ105 to ϩ35 (Ϫ105/ϩ35 luc) and Ϫ68 to ϩ35 region (Ϫ68/ϩ35 luc) were amplified using SF1–3 top primer and GL2 primers, and Ϫ68/Ϫ44 top primer and GL2 primers, respectively, and the PCR prod- uct for each clone was Klenow treated, digested with HindIII, and cloned into the SmaI-HindIII sites of pGL2-basic. The site-specific mutants gen- erated in the Ϫ254/ϩ35 luc plasmid were constructed by generating two overlapping PCR products that share an engineered restriction site to alter the core sequences of interest. To produce the 5Ј-PCR product, the GL1 primer was used to amplify the top strand, and the bottom strand was amplified with the site-specific primer. The 3Ј-PCR product was produced with the site-specific mutant primer (top strand) and GL2 (bottom strand). The PCR products were digested with the appropriate restriction enzymes and cloned into the SmaI-KpnI restriction sites of pGL2-basic. The sequences of oligonucleotides used in the reactions are listed under Oligonucleotides used in EMSA and PCR. The SF1–3mut/ GATA-4mut-luc, GATAmut/AP-1mut-luc, and SF1–3mut/AP-1mut- luc constructs were created in a similar manner; the mutant SF1–3 site was engineered into GATAmut-luc using the SF1–3mut-luc primer to generate SF1–3mut/GATAmut-luc, the AP-1mut site was engineered into GATAmut-luc using the AP-1mut-luc primer to generate GATAmut/AP-1mut-luc, and the AP-1mut site was engineered into SF1–3mut-luc using the AP-1mut-luc construct to generate SF1–3mut/ AP-1mut-luc. One exception for SF1–3mut/AP-1mut-luc is the 5Ј-PCR product was digested with MluI and BglII, the 3Ј-PCR product was digested with BglII and XbaI, and the two products were cloned into the MluI-NheI sites of pGL2 basic. All sequences were confirmed by double stranded sequencing using the Sequenase II kit (United States Biochem- ical Corp., Cleveland, OH). Electrophoretic mobility shift assay (EMSA) Double stranded oligonucleotides were generated by mixing equal molar concentrations of top and bottom strand primers in a high salt annealing buffer [10 mm Tris (pH 7.5), 50 mm NaCl, and 1 mm EDTA] and heating for 2 min at 95 C followed by slow equilibration to room temperature. The oligonucleotides were end labeled with T4 DNA ki- nase and [␥-32 P]ATP (NEN Life Science Products). To generate a probe for the Ϫ105/Ϫ44 region of StAR, linearized Ϫ254 StAR promoter con- struct was used as the DNA template in a PCR reaction with SF1–3 (top) and Ϫ68/Ϫ44 (bottom). The Ϫ105/Ϫ44 fragment containing site- specific mutations or double mutations was generated as described above using the corresponding mutated reporter gene constructs as the template and appropriate oligonucleotides. Each PCR fragment was gel purified and radiolabeled as described above. Restriction digestion anal- ysis was used to confirm the PCR-generated probe contained the desired mutations. MA-10 nuclear extracts (2.5–15.0 ␮g) or 0.25 ␮g GST-SF-1 were incubated with 50 fmol radiolabeled oligonucleotide for 30 min on ice. The binding buffer contained the following: 50 mm Tris-Cl (pH 8.0), 100 mm KCl, 12.5 mm MgCl2, 1 mm EDTA, 1 mm dithiothreitol, 20% glycerol, and 2.5 ␮g poly(dI:dC). Cold competition assays were per- formed with a 20- to 100-fold molar excess of oligonucleotides. Antibody supershift assays were performed with 2.5 or 15 ␮g MA-10 nuclear extract proteins or 0.25 ␮g GST-SF-1 plus 4 ␮g monoclonal GATA-1, polyclonal GATA-4, polyclonal C/EBP␤, or polyclonal SF-1 antibodies. For the supershift assays, all components of the binding reaction, ex- cluding the radiolabeled probe, were preincubated on ice for 30 min. Radiolabeled probe was then added, and the incubation was continued for 30 min. Binding reactions were resolved on a 4% nondenaturing polyacrylamide gel. The dried gels were exposed to x-ray film, and the radioactive bands were visualized by autoradiography. The following oligonucleotides were used in the EMSA binding re- actions. Only the top strand is listed, and changed base pairs in the mutated probes are underlined. Sequences of oligonucleotides used for PCR and EMSA probes The following sequences were used: SF1–3, 5Ј-CATTCCATCCTT- GACCCTCTGCA-3Ј; SF1–3mut, 5Ј-CATTCCATCCTCGAGCCTCT- GCA-3Ј; Ϫ68/Ϫ44, 5Ј-TTTTTTATCTCAAGTGATGATGCAC-3Ј; Ϫ68/ Ϫ44 GATAmut, 5Ј-TTTTTCCGGACAAGTGATGATGCAC-3Ј; Ϫ68/ Ϫ44 SREBPmut (SREBP, sterol regulatory binding protein), 5Ј-TTTTT- TATCTCTCGAGATGATGCAC-3Ј; Ϫ87/Ϫ64, 5Ј-TGCACAATGACT- GATGACTTTTT-3Ј; Ϫ87/Ϫ64 AP-1mut, 5Ј-TGCACAATAGATCTT- GACTTTTT-3Ј; Ϫ87/Ϫ64 1/2 mut, 5Ј-TGCACAATGACTGA- GATCTTTTT-3Ј; GL1*, 5Ј-TGTATCTTATGGTACTGTAACTG-3Ј; GL2*, 5Ј-CTTTATGTTTTTGGCGTCTTCCA-3Ј; and Ϫ150 Mlu, 5Ј-ACA- CACGCGTAGTCTGCTCCCTCCCACCTTGGCCA-3Ј (* indicates the primers used for pGL2-basic vector). Nuclear extract preparation Confluent MA-10 mouse Leydig tumor cell cultures were treated with fresh Waymouth’s medium in the absence (control) or presence of (Bu)2cAMP for 4 h, and nuclear extracts were prepared following pre- viously described protocols (36). The extracts were flash-frozen in liquid nitrogen and stored at Ϫ80 C until use. Results 5Ј-Deletion analysis localizes the cAMP-responsive region between Ϫ105 and Ϫ68 of the mStAR promoter To more closely map the cAMP-responsive elements within the Ϫ254 bp region of the murine StAR gene, a series of StAR promoter deletion constructs was transiently trans- fected into MA-10 cells and tested for cAMP responsiveness (Fig. 1). (Bu)2cAMP treatment of MA-10 cells resulted in an 8.7-fold increase in luciferase reporter gene activity for the Ϫ254 StAR promoter construct. Deletion to Ϫ150 caused both basal and (Bu)2cAMP-stimulated promoter activity to in- crease 4- and 6-fold, respectively, which represents a 70% increase in the cAMP-dependent fold induction compared with the Ϫ254 StAR construct. Further deletion to Ϫ105 caused a 46% decrease in basal activity compared with Ϫ254 StAR, but the fold induction in response to the cAMP stim- ulus was not diminished. However, a 5-fold decrease in the cAMP-dependent induction was observed with the Ϫ68 StAR promoter construct. These results suggest that multiple elements may be required for the cAMP response of the mouse StAR promoter: a possible negative regulatory region located between the Ϫ150/Ϫ105 and a positive regulatory region located between the Ϫ105/Ϫ68 region of the pro- moter. The Ϫ254/Ϫ65 region of the promoter was cloned upstream of a heterologous promoter (Ϫ254/Ϫ66StAR- pGL3luc), but the single copy of this region was not sufficient to confer cAMP-dependent responsiveness (data not shown). Therefore, elements 5Ј and 3Ј of Ϫ68 appear to required for the induction of StAR gene by cAMP. SF-1 and GATA-4 bind to elements within Ϫ105/Ϫ44 region of the mouse StAR promoter As the 5Ј-deletion analysis indicated that the cAMP- responsive region was within Ϫ105 bp of the transcriptional start site, we were interested in testing for possible cAMP- dependent protein-DNA interactions within this region. We first compared the sequence of murine StAR promoter (Ϫ105 to ϩ35) to the transcription factor database and identified several potential transcription factor-binding sites that in- SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS 1347 The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
  • 4. cluded SF-1, C/EBP, AP-1, and GATA-1. Shown in Fig. 2A is the sequence of the Ϫ150 region of murine StAR, with the location of these elements indicated. An alignment with the human and rat sequences is also shown to indicate the high degree of sequence conservation within this region of the StAR promoter. We previously characterized the SF-1–1 (Ϫ135), SF-1–2 (Ϫ45), and the C/EBP␤ (Ϫ113) elements (10, 26), whereas the SF-1 site located at Ϫ95 bp has been shown to bind SF-1 in the human and rat StAR promoters (21, 37). The region between Ϫ87/Ϫ64 is not as well conserved and unique to the mouse sequence are potential C/EBP␤, non- consensus AP-1, and nuclear-receptor half-site elements that we refer to as the CAN region. Another highly conserved region, located between Ϫ68/Ϫ44, is a putative binding site for GATA-1 and SREBP. This region is adjacent to the pro- posed cAMP-responsive region (Ϫ105/Ϫ68) identified by 5Ј-deletion analysis and was deleted in the nonresponsive 3Ј-deletion construct (Ϫ254/Ϫ66StAR-pGL3luc), indicating that this region also may contribute to the cAMP response. Based on the location of these elements, overlapping DNA probes were generated to encompass the SF-1 (Ϫ105/Ϫ83 SF1–3), the C/EBP␤-AP-1-nuclear receptor half-site se- quences (Ϫ87/Ϫ64 CAN), and the GATA/SREBP element (Ϫ68/Ϫ44 GATA) and used in EMSAs. Mutations that were introduced into the core binding sequences of the oligonu- cleotides are shown in Fig. 2B. The SF1–3 DNA probe (Ϫ105/ Ϫ83) formed a specific protein-DNA complex that was ob- served using nuclear proteins isolated from either untreated (lane 1) or (Bu)2cAMP-treated (lane 5) MA-10 cells (Fig. 3). The specificity of complex formation was shown by compe- tition with unlabeled SF1–3 probe (lanes 2 and 6), and the complex was not formed when an oligomer that contained two point mutations in the CATCCTTG core sequence (SF1– FIG. 1. 5Ј-Deletion analysis of the mouse StAR promoter. MA-10 cells were cotransfected with the indicated StAR-promoter-luciferase re- porter gene constructs (denoted by the position of the base pairs 5-prime to the transcriptional start site) and the pCMV-␤-galactosi- dase expression vector. The transfected cells were treated in triplicate with fresh Waymouth’s medium in either the absence (basal) or pres- ence of 1 mM (Bu)2cAMP for 16–19 h, and the luciferase activity was determined and normalized to ␤-galactosidase expression (RLU/b- gal). For each experiment, the data were expressed as a percentage of the Ϫ254 StAR-promoter construct (Ϫ254/ϩ35 StAR promoter- pGL2luciferase), which was set at 100%, and are graphed as the mean Ϯ SEM values for the relative luciferase activity from three to five experiments. The fold induction values represent the ratio of (Bu)2cAMP/basal activity for each construct. Shown are the mean Ϯ SEM values for three to five experiments. The inset graph shows the RLU/␤-gal activities of the Ϫ254, Ϫ105, and Ϫ68 StAR-promoter constructs. pGL2-basic represents vector control, and NT represents nontransfected MA-10 cells. *, Statistically significant (P Ͻ 0.05) increase (Ϫ150/ϩ35) or decrease (Ϫ68/ϩ35 luc) relative to the fold induction of the Ϫ254 StAR promoter-luciferase construct in response to (Bu)2cAMP. FIG. 2. A, Comparison of mouse (M), human (H), and rat (R) StAR promoter sequences within Ϫ150 bp before the transcriptional start site. The location of the DNA elements for SF-1, C/EBP␤-, GATA-4-, and putative SREBP-binding sites are indicated above the core se- quences by the respective name of the highlighted factors. The putative bind- ing sites for the CAN region (Ϫ87/Ϫ64) include C/EBP, AP-1, and an imperfect nuclear receptor half-site (1/2). B, Shown are the region of the promoter and the name given to the introduced mutation in the Ϫ254/ϩ35 StAR- promoter constructs and the EMSA probes. The point mutations are indi- cated in bold. 1348 SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS Endo • 2000 Vol 141 • No 4 The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
  • 5. 3mut) was used in the binding reaction (lanes 4 and 8). A protein-DNA complex of similar mobility was observed when the probe was incubated with purified GST-SF-1 (lane 9), and addition of an antibody specific for SF-1 greatly diminished or abolished DNA-protein interaction (lanes 3 and 7). Thus, these data verify that SF-1 also binds to this region of the mouse promoter. As the mobility and intensity of the SF-1 complex are the same with nuclear proteins from either control or (Bu)2cAMP-stimulated cells, these data in- dicate that SF-1 binding is independent of the cAMP stimulus in MA-10 cells. This element is referred to as SF1–3 to indicate the third SF-1-binding site within 150 bp of the mouse pro- moter (Fig. 2A). Two protein-DNA complexes were observed with the CAN (Ϫ87/Ϫ64) DNA probe (Fig. 4, A and B). Complex A is a doublet that is present in binding reactions with nuclear proteins from both control and (Bu)2cAMP-treated MA-10 cells, whereas complex B is a slower migrating band that is present only with nuclear proteins from stimulated cells (Fig. 4A). As this region contains a CAAT box, and C/EBP␤ has been proposed or shown to interact with this region (26, 38), we used a C/EBP␤ antibody to test for the presence of C/EBP␤ in complex A or B. As shown in Fig. 4B, addition of the antibody did not affect protein-DNA interactions with the CAN probe and nuclear proteins from stimulated cells (lane 2). We verified that a protein-DNA complex is formed with a consensus C/EBP oligonucleotide (lane 3) and that this complex is supershifted by antibody (lane 4), confirming that C/EBP␤ is present in the nuclear extract (25, 26). Cold competition with the consensus C/EBP oligonucleotide also failed to indicate any involvement of C/EBP␤ with this el- ement (lane 5). The specificity of complex formation was demonstrated by cold competition with the CAN probe (lane 8) and a mutation in the putative nuclear receptor half-site (1/2mut) was also effective as a competitor probe (lane 6). Mutation in the putative AP-1 site (AP-1mut), on the other hand, did not compete with the CAN probe for protein binding (lane 7). We next tested for Fos/Jun binding to this FIG. 3. EMSA analysis reveals SF-1 binding to the Ϫ105/Ϫ83 region of mouse StAR. EMSA was used to examine binding of MA-10 nuclear extract to the radiolabeled SF1–3 probe, which represents the Ϫ105/ Ϫ83 region of the StAR promoter. Binding reactions containing 50 fmol radiolabeled probe and 2.5 ␮g nuclear extracts prepared from unstimulated (lanes 1–4) or (Bu)2cAMP-treated (lanes 5–8) MA-10 cells or 0.25 ␮g purified GST-SF-1 protein (lane 9) were preincubated on ice for 30 min. For cold competition or antibody interference anal- ysis, a 100-fold molar excess of cold probe (indicated as ؉ in lanes 2 and 6) or 4 ␮g SF-1 polyclonal antibody (indicated as Ab in lanes 3 and 7) were preincubated with nuclear extract for 30 min; then the ra- diolabeled probe was added to all reactions, and the incubation was continued for 30 min on ice. The DNA-protein complexes were re- solved on a nondenaturing 4% polyacrylamide gel, the gel was dried, and the complexes were visualized by autoradiography. Mutations introduced into SF1–3 (SF1–3 mut) are listed in Fig. 2B. Lane 10 shows incubation of SF-1 polyclonal antibody and radiolabeled probe without nuclear extract. Free probe is not shown on the gel. FIG. 4. A cAMP-dependent factor(s) binds to the Ϫ87/Ϫ64 CAN re- gion of mouse StAR. EMSA was used to examine binding of MA-10 nuclear extract to radiolabeled CAN probe, which represents the Ϫ87/Ϫ64 region of the StAR promoter. A, Concentration-dependent protein-DNA complex formation. Binding reactions were preincu- bated for 30 min on ice with nuclear extracts prepared from unstimu- lated (control) or (Bu)2cAMP-treated [(Bu)2] MA-10 cells. Shown is a titration of 5, 10, and 15 ␮g control or (Bu)2cAMP nuclear extracts, respectively. B, Effects of cold competition and C/EBP␤ antibody on protein-DNA interactions. Either a 50-fold molar excess of the indi- cated cold probe or C/EBP␤ polyclonal antibody (4 ␮g) antibody was included in a preincubation with nuclear extracts for 30 min; then radiolabeled probe was added, and the incubation was continued for 30 min on ice. The protein-DNA complexes were resolved on a non- denaturing 4% polyacrylamide gel, the gel was dried, and the com- plexes were visualized by autoradiography. The sequences of the oligomer probes for the CAN region containing mutations in the po- tential AP-1 (AP-1 mut) and imperfect nuclear receptor half-sites (1/2 mut) are listed in Fig. 2B. con. C/EBP, Commercially purchased con- sensus C/EBP oligomer. The arrows indicate the positions of com- plexes A and B and free probe. SS, The C/EBP␤ antibody supershift. SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS 1349 The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
  • 6. region, but addition of antibodies that recognize all family members did not affect complex formation (data not shown). A prominent protein-DNA complex was apparent with the GATA oligonucleotide (Ϫ68/Ϫ44 GATA) using nuclear proteins from either untreated or (Bu)2cAMP-stimulated MA-10 cells (Fig. 5). Complex formation was abolished by cold competition with the GATA oligonucleotide (lane 2) or the SREBPmut oligonucleotide that contains a mutation in the putative SREBP site (lane 4). Conversely, using the SREB- Pmut as a probe did not affect complex formation (lane 3), indicating that the SREBP element is not involved in protein- DNA interactions at this region. On the other hand, no com- plex was formed with a probe that had a mutation within the GATA DNA-binding site (lane 5), and the mutant GATA probe (GATA mut) did not compete for protein binding (lane 6). A complex of similar mobility was observed using a probe with the consensus sequence for GATA binding (lanes 7 and 8) and cold competition experiments using this consensus GATA probe resulted in the lack of complex formation (lane 10). Although this sequence was identified as a potential GATA-1-binding site, GATA-1 antibodies did not affect com- plex formation (compare lanes 1 and 9). However, binding to the consensus GATA probe appeared to be diminished by GATA-1 antibodies (compare lanes 7 and 8). Therefore, we tested for other members of the GATA family, and GATA-4 antibodies resulted in a supershift of the complex (lanes 11–13). The supershifted complex was apparent using nu- clear extracts from either control (lane 11) or (Bu)2cAMP- treated (lanes 12 and 13) MA-10 cells with the StAR GATA probe (lanes 11 and 12) or the consensus GATA probe (lane 13). A less prominent band appears with the Ϫ68/Ϫ44 GATA probe; however, the significance of this interaction is not known. At present, these results indicate that GATA-4 in MA-10 cells binds to the mouse StAR promoter. The Ϫ87/Ϫ68 region of the StAR promoter is required for maximal basal promoter activity of StAR To test the functional significance of the specific protein- DNA interactions characterized above on StAR promoter activity, the mutations that abolished protein binding were individually introduced into the Ϫ254 StAR promoter con- struct, and luciferase activity was measured in transiently transfected MA-10 cells (Fig. 6A). Mutation of the SF1–3 site resulted in a 43% reduction in basal luciferase activity com- pared with the Ϫ254 StAR promoter construct (WT), but the fold induction in response to (Bu)2cAMP stimulation was not significantly different. Mutation of the GATA-4-binding site also reduced basal promoter strength (ϳ36%); however, the cAMP response was also decreased from 8.7-fold (WT) to 6.9-fold (GATA). Lastly, mutation of the putative AP-1 site (AP-1mut) or the nuclear receptor half-site (1/2mut) resulted in decreased basal activity by 63% or 57%, respectively, but, again, fold activation in response to (Bu)2cAMP was un- changed compared with that with the Ϫ254 StAR promoter construct (WT). Although the fold induction by (Bu)2cAMP- treatment was not diminished with the SF1–3 and CAN region mutations, overall promoter strength was reduced. This decrease in response appeared to closely parallel the severity of the mutation on basal promoter activity. These data indicate that the SF1–3, CAN, and GATA sites all con- tribute to basal promoter strength. However, neither the SF1–3 nor the CAN region is individually required for the cAMP-dependent response of the StAR promoter. On the other hand, GATA-4 may account for 20–30% of the cAMP response of the murine StAR promoter. Mutation of multiple elements affects basal activity of the StAR promoter As mutations in the SF-1- and GATA-4-binding site indi- vidually affected StAR basal and cAMP-dependent promoter activity, respectively, as well as the close proximity of the two binding sites, we introduced mutations that abolished both SF-1 and GATA-4 binding into Ϫ254 StAR promoter con- struct and tested the effect on promoter activity in transiently transfected MA-10 cells (Fig. 6B). The overall basal activity FIG. 5. GATA-4 in MA-10 nuclear extracts binds the highly con- served region (Ϫ68/Ϫ44) of the mouse StAR promoter. EMSA was used to examine binding of MA-10 nuclear extract to radiolabeled GATA probe, which represents the Ϫ68/Ϫ44 region of the StAR pro- moter. Binding reactions were incubated for 30 min on ice with 5 ␮g nuclear extracts prepared from unstimulated (lanes 1–11) or (Bu)2cAMP-treated (lanes 12 and 13) MA-10 cells. For cold competi- tion or antibody interference analysis, a 20-fold molar excess of the indicated cold probe, denoted by cc (lanes 2, 4, 6, and 10), or 4 ␮g GATA-1 monoclonal antibody (lanes 8 and 9) or GATA-4 polyclonal antibody (lanes 11–13) were included in a preincubation with nuclear extracts; then the radiolabeled probe was added, and the incubation was continued for 30 min on ice. Lanes 14 and 15 represent probe alone or probe and antibody minus nuclear extract, respectively. The protein-DNA complexes were resolved on a nondenaturing 4% poly- acrylamide gel, the gel was dried, and the complexes were visualized by autoradiography. The radiolabeled probes are denoted as GATA for the Ϫ68/Ϫ44 region, GATA mut for the Ϫ68/Ϫ44 region containing mutations in the GATA core sequence, and SREBP mut for the Ϫ68/ Ϫ44 region containing mutations in the putative SREBP element. The sequences for these probes are all shown in Fig. 2B. The con. GATA probe represents the 20-mer consensus oligonucleotide for GATA that was used as a positive control for GATA-binding proteins. The arrows indicate the supershift with GATA-4 Ab (SS) and the free probe; the open arrowhead represents the specific complex. 1350 SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS Endo • 2000 Vol 141 • No 4 The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
  • 7. of StAR promoter was dramatically decreased (88%) com- pared with that of the Ϫ254 StAR promoter construct (WT), indicating that mutation of both SF1–3 and GATA had an additive effect on promoter strength. Despite minimal basal activity, the promoter caused a 4.8-fold increase in the cAMP- dependent response. However, this represents only 45% of the cAMP response for the Ϫ254 StAR promoter construct. Thus, the double mutation has a greater effect than the single GATA mutation on the cAMP response. Therefore, we ex- amined the possibility that either the SF1–3 or GATA-4 site acts functionally with the AP-1 site to promote the cAMP response (Fig. 6B). Double mutations in the SF-1 and AP-1 elements (SF1–3/AP-1) or the GATA-4 and AP-1 (GATA/ AP-1) elements did not result in any greater effect on StAR promoter activity compared with an additive effect of the single mutations alone. The basal activities of SF1–3mut/ AP-1mut and GATAmut/AP-1mut were 16% (AP-1 plus SF-1) and 23% (SF-1 plus GATA) that of the Ϫ254 StAR promoter construct, respectively, whereas the (Bu)2cAMP- dependent increases were 90% (SF1–3 effect) and 64% (GATA-4 effect) that of the Ϫ254 StAR. Thus, it appears that mutation of the AP-1 element has the single greatest effect on StAR promoter activity, due mainly to decreased basal ex- pression, whereas the SF1–3 and GATA-4 double mutations had the most severe effect on promoter function. One pos- sibility is that SF1–3 and GATA-4 function to stabilize a common factor(s) in the CAN (Ϫ87/Ϫ64) region. To test this proposal, we examined the effects of abolishing one DNA- binding site on protein-DNA interactions at other sites within the Ϫ105/Ϫ44 bp of StAR promoter. Protein-DNA interactions within the cAMP-responsive regions of the StAR promoter Four major protein-DNA complexes (I–IV) were detected by EMSA analysis using a DNA probe that spans the Ϫ105 to Ϫ44 region (Ϫ105/Ϫ44) of the StAR promoter (Fig. 7A). Complex formation appeared to be independent of cAMP treatment in that similar complexes were formed with nu- clear extracts from both control and (Bu)2cAMP-treated MA-10 cells. Antibodies to SF-1, GATA-4, and C/EBP␤ were used to help identify the presence of these proteins in the four complexes (Fig. 7B). The SF-1 antibody abolished complex I and diminished complex III (lane 2), whereas GATA-4 an- tibodies caused the appearance of a supershifted complex concomitant with diminished complex II (lane 3). C/EBP␤ antibodies had no effect on protein-DNA interactions within this region of the StAR promoter (lane 4). Cold competition experiments with the oligonucleotides corresponding to the SF1–3 (Ϫ105/Ϫ44), CAN (Ϫ87/Ϫ64), and GATA (Ϫ68/Ϫ44) regions of the promoter were performed to verify the con- tribution of each site to complex formation. First, cold com- petition with unlabeled Ϫ105/Ϫ44 probe greatly diminished complex formation (lane 5). The SF1–3 probe eliminated com- plex I and diminished complexes III and IV without affecting complex II formation (lane 6), whereas the GATA probe competed for complex II formation without affecting com- plex I, III, or IV (lane 9). The CAN probe eliminated complex IV (lane 7), whereas the consensus C/EBP␤ oligonucleotide (lane 8) had no effect on the protein-DNA interactions with the StAR promoter. To further assess complex composition, a series of com- plementary experiments was performed in which the same sequence mutations used in the functional studies were in- troduced into the Ϫ105/Ϫ44 probe, and the effects on pro- tein-DNA interactions were determined (Fig. 7C). Complex I was abolished, and complexes III and IV were diminished by mutation of the SF1–3 site (SF1–3mut, lane 2), which is consistent with the results of the SF-1 antibody and cold competition experiments. Complex II was not affected by the SF1–3 mutation, and addition of the GATA-4 antibody caused a supershift of the remaining complex II (lane 3). Conversely, mutation of the GATA-4 site eliminated com- plex II formation (GATAmut, lane 4), and the remaining complexes (I, III, and IV) were greatly diminished by SF-1 antibodies (lane 5). The CAN probe was an effective com- FIG. 6. Mutational analysis reveals regulatory regions of mouse StAR. MA-10 cells were cotransfected with the indicated StAR pro- moter-luciferase reporter gene constructs and pCMV-␤-galactosidase as detailed in Materials and Methods. The transfected cells were treated in triplicate with fresh Waymouth’s medium in either the absence (basal) or presence of 1 mM (Bu)2cAMP for 16–19 h, and luciferase activity was determined and normalized to ␤-galactosidase expression (RLU/b-gal). For each experiment, the data were ex- pressed as a percentage of the Ϫ254 StAR promoter basal activity, which was set at 100%. Graphed are the mean Ϯ SEM values from three to six experiments for the relative luciferase activity of StAR promoter containing mutations in either a single element (A) or two elements (B). The fold induction values represent the ratio of (Bu)2cAMP/basal activity for each construct. Shown are the mean Ϯ SEM values for three to five experiments. *, Statistically significant (P Ͻ 0.05) difference relative to the fold induction of the Ϫ254 StAR promoter construct in response to (Bu)2cAMP. WT, Wild-type Ϫ254/ ϩ35 StAR promoter-luciferase construct; pGL2-basic, empty vector; NT, nontransfected. SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS 1351 The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
  • 8. petitor for complexes III and IV that remained with the GATAmut probe (lane 6) probe, but did not further reduce complex formation with the AP-1mut probe (lane 7), sug- gesting that factors binding to the CAN region contribute to formation of these complexes. Indeed, mutation of the AP- 1-like site eliminated complex IV and surprisingly also re- duced all other protein-DNA interactions (lane 10). The re- maining complexes I and III were eliminated by the addition of SF-1 antibodies (lane 9), whereas GATA-4 antibodies re- sulted in a supershift of complex II (lanes 8). FIG. 7. The SF1–3- and GATA-4-binding sites are essential for the formation of multiple complexes within the Ϫ105/Ϫ44 region of StAR. EMSA was used to examine the binding of MA-10 nuclear extract to the radiolabeled Ϫ105/Ϫ44 region of the StAR promoter. A, Concentration- dependent protein-DNA complex formation. Fifty femtomoles of radiolabeled probe (Ϫ105/Ϫ44) were incubated with 5, 10, or 15 ␮g nuclear extracts from untreated (control; lanes 1–3) or (Bu)2cAMP-treated [(Bu)2; lanes 4–6] MA-10 cells. The arrows indicate the positions of four protein-DNA complexes, denoted I–IV. Free probe is not shown on this gel. B, Cold competition or antibody interference analysis. SF-1, GATA-4, or C/EBP␤ antibody (4 ␮g) or the indicated cold competitor probes were incubated with 10 ␮g nuclear extracts [(Bu)2cAMP-treated]; then the radiolabeled Ϫ105/Ϫ44 probe was added, and the incubation was continued for 30 min on ice. The protein-DNA complexes were resolved on a nondenaturing 4% polyacrylamide gel, the gel was dried, and the complexes were visualized by autoradiography. The cold competitors were Ϫ105/Ϫ44, SF1–3 (Ϫ105/Ϫ83), CAN (Ϫ87/Ϫ64 region), GATA (Ϫ68/Ϫ44 region), and consensus C/EBP. Arrows indicate complexes I–IV, and the open arrowhead indicates the GATA-4 supershifted complex (SS). C and D, Effects of mutation of an individual element (C) or two elements (D) on protein-DNA interactions. The sequences for the mutations that were introduced into the Ϫ105/44 probe are given in Table 2B. The radiolabeled probes used in the binding reaction are indicated: Ϫ105/Ϫ44, SF1–3 mut, GATA mut, and AP-1 mut, or their combination (D). Experiments in the presence of antibodies or cold competitor CAN probe were performed as described in B. 1352 SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS Endo • 2000 Vol 141 • No 4 The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
  • 9. As StAR basal promoter activity was greatly reduced by mutations within two of the three SF1–3, CAN, or GATA elements (Fig. 6B), we also tested the effects of these muta- tions on protein-DNA interactions (Fig. 7D). A double mu- tation in SF-1 and GATA (SF1–3/GATA mut) resulted in the loss of all complex formation (Fig. 7D, lane 2), whereas com- plex II was still present with the probe containing a double mutation in SF1–3 and AP-1 (SF1–3/AP-1 mut, lane 3). Dou- ble mutations of GATA and AP-1 caused a marked decrease in protein-DNA interactions for complexes III, and IV and eliminated complex II (lane 4). These data are consistent with an additive effect of the single mutations on protein-DNA interactions, with the exception that complex I did not appear affected by the GATAmut/AP-1mut probe. This was not an expected result based on the decrease in complex I with the AP-1 mut probe (Fig. 7C, compare lanes 10 and 11). These results indicate that complexes III and IV are dependent upon SF-1 and possibly, to a lesser extent, GATA-4. In sum, the individual mutations in the SF1–3 or AP-1-like regions and the SF-1 antibodies do not affect GATA-4 bind- ing; therefore, we conclude that GATA-4 binding within the Ϫ105/Ϫ44 region is independent of other factors. Further, the GATA-4 mutation does not prevent SF-1-dependent com- plex formation (I, III, and IV), indicating that SF-1 binding is not dependent on GATA-4 binding. CAN factors appear to contribute to complex IV formation, as mutation of the AP- 1-like element or cold competition with the CAN region eliminated this complex. However, protein binding within the CAN region in the context of the longer probe appears to be dependent on SF-1 binding to SF1–3 and may be in- fluenced by GATA-4 binding. Conversely, an intact AP-1 element appears to be required for efficient SF-1 binding, indicating that protein binding to SF1–3 and CAN regions may be interdependent. Together, these data clearly dem- onstrate that complex I is due to SF-1 binding, and complex II is due to GATA-4 binding. The data are consistent with complexes III and IV being dependent upon SF-1, with com- plex III most likely containing SF-1, whereas CAN factor binding contributes to complex IV. Discussion These studies have identified two regulatory regions of the StAR promoter: a negative region located between Ϫ254/ Ϫ150, and a cAMP-responsive region located between Ϫ105/Ϫ60. Putative negative regulatory regions were pre- viously identified for the rat and porcine promoters as well (21, 39). Our promoter deletion analysis has eliminated SF-1 (SF-1–1) and C/EBP␤ binding at Ϫ135 and Ϫ113 as essential for the cAMP response of the StAR gene and has pointed to SF1–3 (Ϫ95), CAN-binding protein(s) (Ϫ87/Ϫ68), and GATA-4 (Ϫ68) as potential factors. Identification of the Ϫ105 to Ϫ95 region as a SF-1-binding site was an expected result, as this element was previously shown to bind SF-1 in the human and rat StAR promoters (21, 37). However, these are the first studies to show a distinction between regulations of the human and mouse StAR pro- moter by cAMP in steroidogenic cell lines. Mutation of the SF-1 site (Ϫ95) in the human promoter demonstrated 90% and 80% decreases in basal and forskolin-stimulated pro- moter activity in human granulosa-lutein cells (37). Our re- cent studies in the human H295R adrenocortical cells also have shown that the SF-1 element at Ϫ95 is required for cAMP and angiotensin II induction of human StAR promot- er-luciferase reporter gene expression (23). Transient trans- fection of COS-1 cells with the rat StAR promoter confirmed a SF-1-dependent activation of the StAR promoter that re- quired the SF-1 site at Ϫ95 and that mutation of this site resulted in a decrease in the cAMP response that was due to lowered basal promoter activity (21). Our results demon- strate that the mouse and rat promoters are functionally more similar to each other than to the human StAR promoter. This apparent promoter-specific difference may be reflective of a divergence within the StAR promoter sequences between the human and the mouse. The human sequence has an addi- tional five bases downstream of the SF-1 site and does not contain the CAAT box, the the AP-1-like site, or the nuclear receptor half-site that have been shown to be important for the murine promoter in MA-10 cells (26, 38). Thus, it is apparent that SF-1 is important in StAR gene regulation but is not sufficient for the cAMP-dependent response in MA-10 cells. Possibly, the sequence divergence reflects greater in- teractions of SF-1 with other factors binding in this region of the mouse StAR promoter. We have confirmed the functional importance of the AP- 1-like element within the Ϫ87/Ϫ68 (CAN) region of mouse StAR promoter for basal transcriptional activity (26, 38). In addition, this study provides the first evidence that (Bu)2cAMP treatment of MA-10 cells results in the formation of a unique protein-DNA complex at this region of the StAR promoter (complex B). Identification of the CAN factor(s) should provide an important link to elucidating the mech- anism of action for cAMP-dependent regulation of StAR. Presently, our data suggest that multiple elements may con- tribute to activation of the StAR promoter. Double mutations in either the SF1–3 and AP-1 or GATA and AP-1 elements have a greater effect on basal promoter activity that repre- sents the combined effects of the single mutations. The GATAmut/AP-1 mut also resulted in decreased cAMP re- sponse that is attributed to the GATA element. On the other hand, the effect of the SF1–3 and GATA double mutation on basal promoter activity was greater than the combined ef- fects of the single mutations. As mutations that disrupted the AP-1-like site (AP-1mut, SF1–3mut/AP-1mut, and GATAmut/AP-1mut) have similar effects on basal activity as the SF1–3mut/GATAmut, these data suggest that all three elements work together to promote full basal activity of the StAR promoter that, in turn, is required for a maximal cAMP response. What is not clear at the time is why the cAMP- dependent response, although reduced, remains intact de- spite apparent elimination of protein-DNA binding interac- tions within the Ϫ105/Ϫ44 region of StAR promoter with the SF1–3mut/GATAmut. As we did not observe a cAMP- dependent complex with the Ϫ105/Ϫ44 probe, the cAMP- dependent CAN-binding factor(s) may not have been de- tected in this analysis. Indeed, the cAMP-dependent complex was observed only with increased concentrations of nuclear extract, suggesting that this factor(s) may be expressed in lesser abundance. Thus, it is possible that the AP-1 region is still functional in the SF1–3mut/GATAmut double mutation. SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS 1353 The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
  • 10. Previously, we demonstrated that a protein(s) in MA-10 nuclear extracts binds to the CAAT box located in the Ϫ93/ Ϫ71 region, but with lower affinity compared with the C/EBP␤ site located at Ϫ113 (26). Antibody supershift EMSA experiments showed that C/EBP␤ binds to the Ϫ113 site, but binding to the proximal site was not tested. We now have directly tested C/EBP␤ binding to the CAN region and verify that C/EBP␤ does not bind to this region of the StAR pro- moter. It is possible that our previous studies did not detect the factor(s) we now observe because the probe used pre- viously (Ϫ93/Ϫ71) did not contain the entire CAN region (Ϫ87/Ϫ64) binding sites, which may have affected protein binding to this region. Indeed, complexes A and B appear to be dependent upon an intact AP-1-like element and the flank- ing TGATGA sequence. Functionally, mutation of the puta- tive AP-1 site resulted in approximately a 63% decrease in the basal promoter activity of StAR without affecting the cAMP response, a result very similar to the effects seen by mutating the CAAT box at Ϫ87 (26). Therefore, mutation of the CAAT box most likely affects a factor(s) binding to the AP-1-like element in the CAN region or vice versa. Our results in MA-10 cells are distinct from the recent report of murine StAR promoter activity in rat ovarian cells, which demonstrated that C/EBP␤ is a prominent component in a DNA-protein complex formed with the Ϫ87/Ϫ64 region (38). Interestingly, the binding site recognized by C/EBP␤ in ovarian cell extracts is not the CAAT box core, but is the AP-1-like element. One possibility for this apparent tissue- specific difference in C/EBP␤ binding is that different factors bind to the CAN region. This tissue-specific difference may reflect the cycloheximide sensitivity of the cAMP response for StAR. Previously, StAR gene expression was reported to be mediated by a cycloheximide-sensitive mechanism in hu- man granulosa-lutein cells (40). C/EBP␤ expression has been shown to be increased by FSH in ovarian granulosa cells and by (Bu)2cAMP in MA-10 cells (25, 38). Subsequent to sub- mission of this paper, it was reported that C/EBP␤ is induced in human granulosa cells by 8-bromo-cAMP (41). The data supported C/EBP␤ in StAR basal promoter activity as well as the proposal that the cycloheximide-sensitive, cAMP- dependent response may be linked to the induction of C/EBP␤. Thus, it is probable that a SF-1/C/EBP␤ or a GATA-4/C/EBP␤ interaction mediates the observed cyclo- heximide-sensitive response in rat ovarian granulosa cells. Consistent with this proposal, C/EBP␤ and SF-1 were shown to interact directly in a GST pull-down assay (26). However, StAR gene expression in MA-10 cells and Y1 mouse adre- nocortical cells is not dependent upon de novo protein syn- thesis (10). Therefore, another factor(s) that is insensitive to cycloheximide may be used in MA-10 Leydig cells and Y1 cells to bind to the CAN region and promote maximal basal activation of StAR. A difference in tissue-specific transcrip- tion factor expression may thus account for differences in the acute regulation of StAR in granulosa vs. Leydig cell cultures. We identified GATA-4 as the factor that binds to the highly conserved Ϫ68/Ϫ44 region of the mouse StAR promoter. Functionally, GATA-4 contributes to 20% of the cAMP re- sponse. Independently, GATA-4 was recently reported to bind to the same region and was shown to be required for maximal cAMP response in rat granulosa cells (38). Mutation of both the GATA-4 and AP-1-like elements decreased StAR basal promoter activity 97% and dramatically decreased the FSH-dependent increase in reporter gene expression in lu- teal-granulosa cells. In contrast, we demonstrate that a sim- ilar double mutation (GATAmut/AP1mut) had no greater effect on StAR promoter activity in MA-10 cells than the combined effects of the individual mutations within these elements. Again, this discrepancy between our results and those reported for rat granulosa cells indicates that the mu- rine StAR gene may be regulated in a tissue-specific manner. In MA-10 cells, we propose the GATA-4 transcription factor has a role in stabilizing the factor(s) bound to the putative AP-1 site and that GATA-4 and SF-1 work together to sta- bilize this common factor(s). However, these putative CAN- binding proteins and SF-1 can function in the absence of GATA-4. In sum, SF-1 binding is important for the basal activity of the StAR gene, most likely by affecting the interactions among the other proteins within the CAN region of the promoter. GATA-4 binding is independent of SF-1 binding and has an apparently minimal effect on SF-1- and CAN- protein binding, but may influence factor binding. Interac- tion between SF-1 and GATA-4 has been shown for regula- tion of the Mu¨llerian-inhibiting substance gene; therefore, one model for SF-1 and GATA-4 is that they interact and function to stabilize binding of the CAN-binding factor(s) (42). Alternatively, it is possible that there is a redundancy of function for SF-1, CAN-binding protein, and GATA-4. This proposal would be consistent with our mutational anal- ysis that shows that elimination of one binding factor does not compromise the cAMP response of the StAR promoter. Thus, each of the DNA-binding factors may work together to stabilize the unknown cAMP-mediated transcription factors and/or coactivators, but each factor alone may be sufficient to mediate part of the response. Acknowledgments We thank Dr. Keith Parker, University of Texas Southwestern Med- ical Center (Dallas, TX), for providing the GST-SF-1 expression plasmid. We also thank Drs. Carolyn M. Klinge and Keith C. Falkner, Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine (Louisville, KY), for their critical review of this manuscript. Finally, we thank Ms. Rebecca Combs for her excellent technical assistance. References 1. Stone D, Hechter O 1954 Studies on ACTH action in perfused bovine adrenals: site of action of ACTH in corticosteroidogenesis. Arch Biochem Biophys 51:457–469 2. Karaboyas G, Koritz S 1965 Identity of the site of action of cAMP and ACTH in corticosteroidogenesis in rat adrenal and beef adrenal cortex slices. Bio- chemistry 4:462–468 3. Simpson ER, Jefcoate CR, Brownie AC, Boyd GS 1972 The effect of ether anaesthesia stress on cholesterol-side-chain cleavage and cytochrome P450 in rat-adrenal mitochondria. Eur J Biochem 28:442–450 4. Privalle CT, Crivello JF, Jefcoate CR 1983 Regulation of intramitochondrial cholesterol transfer to side-chain cleavage cytochrome P-450 in rat adrenal gland. Proc Natl Acad Sci USA 80:702–706 5. Stocco DM, Clark BJ 1996 Regulation of the acute production of steroids in steroidogenic cells. Endocr Rev 17:221–244 6. Caron KM, Soo SC, Wetsel WC, Stocco DM, Clark BJ, Parker KL 1997 Targeted disruption of the mouse gene encoding steroidogenic acute regula- tory protein provides insights into congenital lipoid adrenal hyperplasia. Proc Natl Acad Sci USA 94:11540–11545 7. Lin D, Sugawara T, Strauss III JF, Clark BJ, Stocco DM, Saenger P, Rogol A, 1354 SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS Endo • 2000 Vol 141 • No 4 The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.
  • 11. Miller WL 1995 Role of steroidogenic acute regulatory protein in adrenal and gonadal steroidogenesis. Science 267:1828–1831 8. Miller WL 1997 Congenital lipoid adrenal hyperplasia: the human gene knock- out for the steroidogenic acute regulatory protein. J Mol Endocrinol 19:227–240 9. Clark BJ, Combs R, Hales KH, Hales DB, Stocco DM 1997 Inhibition of transcription affects synthesis of steroidogenic acute regulatory protein and steroidogenesis in MA-10 mouse Leydig tumor cells. Endocrinology 138:4893–4901 10. Caron KM, Ikeda Y, Soo SC, Stocco DM, Parker KL, Clark BJ 1997 Charac- terization of the promoter region of the mouse gene encoding the steroidogenic acute regulatory protein. Mol Endocrinol 11:138–147 11. Lala DS, Rice DA, Parker KL 1992 Steroidogenic factor I, a key regulator of steroidogenic enzyme expression, is the mouse homolog of fushi tarazu-factor I. Mol Endocrinol 6:1249–1258 12. Honda S, Morohashi K, Nomura M, Takeya H, Kitajima M, Omura T 1993 Ad4BP regulating steroidogenic P-450 gene is a member of steroid hormone receptor superfamily. J Biol Chem 268:7494–7502 13. Morohashi K, Zanger UM, Honda S, Hara M, Waterman MR, Omura T 1993 Activation of CYP11A and CYP11B gene promoters by the steroidogenic cell- specific transcription factor, Ad4BP. Mol Endocrinol 7:1196–1204 14. Luo X, Ikeda Y, Parker KL 1994 A cell-specific nuclear receptor is essential for adrenal and gonadal development and sexual differentiation. Cell 77:481–490 15. Wong M, Rice DA, Parker KL, Schimmer BP 1989 The roles of cAMP and cAMP-dependent protein kinase in the expression of cholesterol side chain cleavage and steroid 11␤-hydroxylase genes in mouse adrenocortical tumor cells. J Biol Chem 264:12867–12871 16. Parissenti AM, Parker KL, Schimmer BP 1993 Identification of promoter elements in the mouse 21-hydroxylase (Cyp21) gene that require a functional cyclic adenosine 3Ј,5Ј-monophosphate- dependent protein kinase. Mol Endo- crinol 7:283–290 17. Zhang P, Mellon SH 1996 The orphan nuclear receptor steroidogenic factor-1 regulates the cyclic adenosine 3Ј,5Ј-monophosphate-mediated transcriptional activation of rat cytochrome P450c17 (17␣-hydroxylase/c17–20 lyase). Mol Endocrinol 10:147–158 18. Carlone DL, Richards JS 1997 Evidence that functional interactions of CREB and SF-1 mediate hormone regulated expression of the aromatase gene in granulosa cells and constitutive expression in R2C cells. J Steroid Biochem Mol Biol 61:223–231 19. Jacob AL, Lund J 1998 Mutations in the activation function-2 core domain of steroidogenic factor-1 dominantly suppresses PKA-dependent transactivation of the bovine CYP17 gene. J Biol Chem 273:13391–13394 20. Hammer GD, Krylova I, Zhang Y, Darimont BD, Simpson K, Weigel NL, Ingraham HA 1999 Phosphorylation of the nuclear receptor SF-1 modulates cofactor recruitment: integration of hormone signaling in reproduction and stress. Mol Cell 3:521–526 21. Sandhoff TW, Hales DB, Hales KH, McLean MP 1998 Transcriptional reg- ulation of the rat steroidogenic acute regulatory protein gene by steroidogenic factor 1. Endocrinology 139:4820–4831 22. Sugawara T, Kiriakidou M, McAllister JM, Kallen CB, Strauss III JF 1997 Multiple steroidogenic factor 1 binding elements in the human steroidogenic acute regulatory protein gene 5Ј-flanking region are required for maximal promoter activity and cyclic AMP responsiveness. Biochemistry 36:7249–7255 23. Clark BJ, Combs R 1999 Angiotensin II and cAMP induce human steroido- genic acute regulatory protein transcription through a common steroidogenic factor-1 element. Endocrinology 140:4390–4397 24. Lekstrom-Himes J, Xanthopoulos KG 1998 Biological role of the CCAAT/ enhancer-binding protein family of transcription factors. J Biol Chem 273:28545–28548 25. Nalbant D, Williams SC, Stocco DM, Khan SA 1998 Luteinizing hormone- dependent gene regulation in Leydig cells may be mediated by CCAAT/ enhancer-binding protein-␤. Endocrinology 139:272–279 26. Reinhart AJ, Williams SC, Clark BJ, Stocco DM 1999 SF-1 (steroidogenic factor-1) and C/EBP␤ (CCAAT/enhancer binding protein-␤) cooperate to regulate the murine StAR (steroidogenic acute regulatory) promoter. Mol Endocrinol 13:729–741 27. Mukai K, Mitani F, Agake R, Ishimura Y 1998 Adrenocorticotropic hormone stimulates CYP11B1 gene transcription through a mechanism involving AP-1 factors. Eur J Biochem 256:190–200 28. Sun Y, Duckworth ML 1999 Identification of a placental-specific enhancer in the rat placental lactogen II gene that contains binding sites for members of the Ets and AP-1 (activator protein 1) families of transcription factors. Mol En- docrinol 13:385–399 29. Duval DL, Ellsworth BS, Clay CM 1999 Is gonadotrope expression of the gonadotropin releasing hormone receptor gene mediated by autocrine/para- crine stimulation of an activin response element? Endocrinology 140:1949–1952 30. Orkin SH 1992 GATA-binding transcription factors in hematopoietic cells. Blood 80:575–581 31. Yomogida K, Ohtani H, Harigae H, Ito E, Nishimune Y, Engel JD, Yamamoto M 1994 Developmental stage- and spermatogenic cycle-specific expression of transcription factor GATA-1 in mouse Sertoli cells. Development 120:1759–1766 32. Morrisey EE, Ip HS, Lu MM, Parmacek MS 1996 GATA-6: a zinc finger transcription factor that is expressed in multiple cell lineages derived from lateral mesoderm. Dev Biol 177:309–322 33. Heikinheimo M, Ermolaeva M, Bielinska M, Rahman NA, Narita N, Huhtaniemi IT, Tapanainen JS, Wilson DB 1997 Expression and hormonal regulation of transcription factors GATA-4 and GATA-6 in the mouse ovary. Endocrinology 138:3505–3514 34. Feng ZM, Wu AZ, Chen CL 1998 Testicular GATA-1 factor up-regulates the promoter activity of rat inhibin ␣-subunit gene in MA-10 Leydig tumor cells. Mol Endocrinol 12:378–390 35. Ketola I, Rahman N, Toppari J, Bielinska M, Porter-Tinge SB, Tapanainen JS, Huhtaniemi IT, Wilson DB, Heikinheimo M 1999 Expression and regu- lation of transcription factors GATA-4 and GATA-6 in developing mouse testis. Endocrinology 140:1470–1480 36. Dignam JD, Lebovitz RM, Roeder RG 1983 Accurate transcription initiation by RNA polymerase II in a soluble extract from isolated mammalian nuclei. Nucleic Acids Res 11:1475–1489 37. Sugawara T, Holt JA, Kiriakidou M, Strauss III JF 1996 Steroidogenic factor 1-dependent promoter activity of the human steroidogenic acute regulatory protein (StAR) gene. Biochemistry 35:9052–9059 38. Silverman E, Eimerl S, Orly J 1999 CCAAT enhancer-binding protein ␤ and GATA-4 binding regions within the promoter of the steroidogenic acute reg- ulatory protein (StAR) gene are required for transcription in rat ovarian cells. J Biol Chem 274:17987–17996 39. LaVoie HA, Garmey JC, Veldhuis JD 1999 Mechanisms of insulin-like growth factor I augmentation of follicle-stimulating hormone-induced porcine steroi- dogenic acute regulatory protein gene promoter activity in granulosa cells. Endocrinology 140:146–153 40. Kiriakidou M, McAllister JM, Sugawara T, Strauss III JF 1996 Expression of steroidogenic acute regulatory protein (StAR) in the human ovary. J Clin Endocrinol Metab 81:4122–4128 41. Christenson LK, Johnson PF, McAllister JM, Strauss III JF 1999 CCAAT/ enhancer-binding proteins regulate expression of the human steroidogenic acute regulatory protein (StAR) gene. J Biol Chem 274:26591–26598 42. Tremblay JJ, Viger RS 1999 Transcription factor GATA-4 enhances Mullerian inhibiting substance gene transcription through a direct interaction with the nuclear receptor SF-1. Mol Endocrinol 13:1388–1401 SF-1 INFLUENCE ON PROTEIN-DNA INTERACTIONS 1355 The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 24 April 2015. at 10:09 For personal use only. No other uses without permission. . All rights reserved.