Download Coordinated Regulation of AIB1 Transcriptional Activity by

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Cell encapsulation wikipedia , lookup

Hedgehog signaling pathway wikipedia , lookup

Cytokinesis wikipedia , lookup

Cellular differentiation wikipedia , lookup

Cell nucleus wikipedia , lookup

Protein moonlighting wikipedia , lookup

G protein–coupled receptor wikipedia , lookup

Nuclear magnetic resonance spectroscopy of proteins wikipedia , lookup

Histone acetylation and deacetylation wikipedia , lookup

Amitosis wikipedia , lookup

Paracrine signalling wikipedia , lookup

Signal transduction wikipedia , lookup

List of types of proteins wikipedia , lookup

Phosphorylation wikipedia , lookup

Protein phosphorylation wikipedia , lookup

Transcript
THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 281, NO. 31, pp. 21848 –21856, August 4, 2006
© 2006 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.
Coordinated Regulation of AIB1 Transcriptional Activity
by Sumoylation and Phosphorylation*
Received for publication, April 19, 2006, and in revised form, June 5, 2006 Published, JBC Papers in Press, June 7, 2006, DOI 10.1074/jbc.M603772200
Huijian Wu, Luyang Sun, Ying Zhang, Yupeng Chen, Bin Shi, Ruifang Li, Yan Wang, Jing Liang, Dongwei Fan,
Ge Wu, Dan Wang, Shaosi Li, and Yongfeng Shang1
From the Department of Biochemistry and Molecular Biology, Peking University Health Science Center, 38 Xue Yuan Road,
Beijing 100083, China
Nuclear receptors play critical roles in animal development
and homeostasis through their bimodal function as repressors
or activators of gene transcription (1). The repression and activation activity of nuclear receptors are determined by their
association with corepressors and coactivators, respectively.
Among the coactivators, the steroid receptor coactivator
(SRC)2 family is both necessary and sufficient for nuclear receptor-mediated gene activation as we previously demonstrated
(2). The SRC family of coactivators includes three distinct but
related p160 members: SRC-1, GRIP1 (or SRC-2), and AIB1
(also named RAC3, ACTR, SRC-3, or p/CIP in mice) (3– 6).
They share 40% overall sequence similarity, and they all contain, in the N terminus, a basic helix-loop-helix domain that is
* This work was supported by the National Natural Science Foundation of
China (Grants 30225043, 30393110, and 30470912 to Y. S.) and by the
Ministry of Science and Technology of China (863 Program Grant
2002BA711A01 and 973 Program Grant 2005CB522404 to Y. S.). The costs
of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked “advertisement” in
accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
1
To whom correspondence should be addressed. Tel.: 86-10-8280-5118; Fax:
86-10-8280-1355; E-mail: [email protected].
2
The abbreviations used are: SRC, steroid receptor coactivator; MAPK, mitogenactivated protein kinase; SUMO, small ubiquitin-like modifier; CREB, cAMPresponse element-binding protein; E1, ubiquitin-activating enzyme; E2,
ubiquitin carrier protein; E3, ubiquitin-protein isopeptide ligase; DBD,
DNA-binding domain; ERK, extracellular signal-regulated kinase; HA, hemagglutinin; E2, 17␤-estradiol; wt, wild type; aa, amino acid(s).
21848 JOURNAL OF BIOLOGICAL CHEMISTRY
known for protein dimerization-DNA interaction as well as a
Per-Arnt-Sim domain, which is known to be involved in protein-protein interaction. Three LXXLL motifs, also known as
nuclear receptor boxes (for nuclear receptor binding), are centrally located in all three proteins (7–9). In the C terminus, the
SRC proteins contain two activation domains, AD1 and AD2,
which are associated with histone acetyltransferases (CREB-binding protein/p300 and pCAF) and coactivator-associated arginine
methyltransferase 1, respectively, and function to modify the configuration of the chromatin structure (10, 11).
Despite the structural similarities among the members of the
SRC family of coactivators, evidence has been accumulated to
suggest different physiological functions for SRC-1, GRIP1, and
AIB1. Genetic studies with gene ablation showed that, although
both male and female SRC1-null mice are viable and fertile, they
exhibit partial resistance to several hormones, including estrogen, progestin, androgen, and thyroid hormones (12, 13). Elimination of GRIP1 impairs fertility in both male and female mice
(14), and GRIP-null mice are protected against obesity and display enhanced adaptive thermogenesis, whereas SRC-1 null
mice are prone to obesity due to reduced energy expenditure
(15). It has been documented that, unlike SRC-1 and GRIP1,
AIB1 is more promiscuous; it can enhance the transcriptional
activity of a number of different activators (16, 17). Indeed,
AIB1-null mice are not only abnormal in reproductive function
but also small in animal size (18, 19). At the molecular level, we
recently demonstrated SRC-1, GRIP1, and AIB1 are differentially implicated in steroid receptor-mediated gene transcription (20 –22).
The differential activity for SRC-1, GRIP1, and AIB1 is probably determined primarily by their intrinsic protein structures.
Ultimately, the function of a protein is also regulated by additional mechanisms particularly post-translational modifications. It has been reported that the activity of the SRC proteins is regulated by phosphorylation. For example, SRC-1
has been shown to be phosphorylated by mitogen-activated
protein kinase (MAPK) at Thr-1179 and Ser-1185. Mutations of these sites attenuated SRC-1 ability to coactivate
progesterone receptor-dependent transcription (23); MAPK
also phosphorylates GRIP1 but at Ser-736, the integrity of
which is required for a full GRIP1 transcriptional activation
function (24). Interestingly, these phosphorylation sites are
not conserved in AIB1. AIB1 was shown to be phosphorylated by several different kinases, including MAPK, I␬B
kinase, and receptor tyrosine kinase HER2/neu (16, 25, 26),
with Ser-505, Ser-543, Ser-860, and Ser-867 being identified
VOLUME 281 • NUMBER 31 • AUGUST 4, 2006
Downloaded from www.jbc.org at Harvard Libraries on July 28, 2006
AIB1, a member of the steroid receptor coactivator (SRC)
family that participates in gene transcriptional activation by
nuclear receptors and other transcription factors, is required for
animal growth and reproductive development and implicated in
breast carcinogenesis. The mechanisms underlying the AIB1
pleiotropic functions are not fully understood and neither is the
regulation of its activity. Here, we showed that AIB1 was a
sumoylated protein and the sumoylation attenuated the transactivation activity of AIB1, which is in contrast to the sumoylation of its paralogs, GRIP1 and SRC-1. The transactivation activity of AIB1 is enhanced by its phosphorylation by several
kinases, including mitogen-activated protein kinase. We demonstrated in this report that estrogen treatment led to an
increased phosphorylation and decreased sumoylation of AIB1
and that the sumoylation coordinated with phosphorylation in
regulating the transcriptional activity of AIB1, providing a
mechanism for post-translational modifications in regulating
the transcriptional output of AIB1.
Regulation of AIB1 by Sumoylation and Phosphorylation
EXPERIMENTAL PROCEDURES
Plasmids and Antibodies—AIB1 mutants (K723R, K786R,
K1194R, K723R/K786R/K1194R, and S505A/S543A/S860A/
S867A) were generated using site-directed mutagenesis
according to the manufacturer’s instructions (Stratagene, La
Jolla, CA). Full-length AIB1 and the mutants were PCR-amplified and subcloned into pcDNA3.1-Gal4-DNA binding domain
(DBD) to generate Gal4-DBD-AIB1 fusion constructs.
pcDNA3-HA-SUMO-1 and pcDNA3-Ubc9-SV5 and dominant negative mutant DN-Ubc9 (C93S) were from Dr. Ronald
Hay (University of St. Andrews, St. Andrews, UK). FLAGSENP1, FLAG-SENP1 mutant (R630L/K631M), FLAG-SENP2,
His-SENP3 were provided by Dr. Edward Yeh (The University
of Texas, M.D. Anderson Cancer Center). Polyclonal anti-extracellular signal-regulated kinase (ERK) 1/2 and anti-phosphor-ERK1/2, polyclonal antibodies against Gal4-DBD, mouse
monoclonal anti-HA antibody, and anti-SUMO-1 were
obtained from Santa Cruz Biotechnology (Santa Cruz, CA).
Mouse monoclonal anti-FLAG was from Sigma. Rabbit polyAUGUST 4, 2006 • VOLUME 281 • NUMBER 31
clonal anti-phosphoserine was from Zymed Laboratories
(South San Francisco, CA).
Cell Culture and Reporter Assays—MCF-7 human breast
cancer cells and COS-7 African green monkey kidney cells were
maintained in Dulbecco’s modified Eagle’s medium containing
10% fetal bovine serum. Transient transfections were performed with Lipofectamine (Invitrogen) according to the company’s specification. For luciferase assays, MCF-7 or COS-7
cells were seeded onto 24-well plates for 18 h and transfected
with appropriate plasmid constructs. Five hours after transfection, cells were switched to phenol red-free medium containing
10% charcoal-dextran-treated fetal bovine serum for 48 h followed by treatment with or without 10 nM 17␤-estradiol (E2) or
5 ␮M ICI-182,780 for 16 h. The cells then were harvested, and
luciferase and Renilla activities were measured using a dual
luciferase kit (Promega, Madison, WI).
Immunoprecipitation and Immunoblotting—MCF-7 or
COS-7 cells were collected in ice-cold phosphate-buffered
saline, and cell extracts were prepared in lysis buffer (50 mM
Tris-HCl, pH 8.0, 150 mM NaCl, 0.1% SDS, 1% Nonidet P-40,
and 0.5% sodium deoxycholate) with 1:100 diluted protease
inhibitor mixture (Roche Applied Science, Indianapolis, IN).
The cytosolic and nuclear protein extracts were prepared as
previously described (43). The cell extracts was pre-cleared
using protein-A-Sepharose at 4 °C for 2 h. Following centrifugation at 14,000 ⫻ g for 10 min, the supernatant was incubated
with appropriate antibodies for 16 h at 4 °C. Immune complexes were then captured using protein A-Sepharose. Following centrifugation at 6,000 ⫻ g for 10 min, the protein-ASepharose was washed four times in washing buffer (20 mM
Tris-HCl, pH 7.5, 500 or 800 mM NaCl, 0.5% Nonidet P-40, and
0.05% sodium deoxycholate). Bound proteins were released
from the protein-A-Sepharose by boiling for 5 min in 2 ⫻ SDS
loading buffer, resolved on 6 – 8% SDS-PAGE, and transferred
onto Pure Nitrocellulose Blotting Membrane (Pall Corp.). The
membrane was then probed using a secondary antibody and
visualized using the enhanced chemiluminescence detection
reagents (Amersham Biosciences) according to the manufacturer’s instructions.
RESULTS
AIB1 Is Modified by SUMO-1—Sumoylation has recently
been identified as an important mechanism that regulates protein functions in essential biological processes, such as gene
transcription, subnuclear structure organization, and cell cycle
progression. Within the SRC family of coactivators, both
GRIP1 (41) and SRC-1 (38) have been reported to be sumoylated. To understand the basis for diverse activities of AIB1 and
the mechanism of the regulation of AIB1 activity, we wanted to
know if AIB1 is also a sumoylated protein in vivo. For this purpose, we performed immunoprecipitation analysis of cellular
extracts from breast cancer cells MCF-7 using anti-AIB1 antibodies, followed by immunoblotting with anti-SUMO-1. As
shown in Fig. 1A, immunoprecipitated AIB1 could also be
detected with an anti-SUMO-1 antibody, indicating that AIB1
is a sumoylated protein. Interestingly, AIB1 sumoylation
quickly disappeared upon treatment of MCF-7 cells with 17␤estradiol (E2); 5 min after E2 treatment, sumoylated AIB1 was
JOURNAL OF BIOLOGICAL CHEMISTRY
21849
Downloaded from www.jbc.org at Harvard Libraries on July 28, 2006
as potential MAPK sites (16). Sequence alignments for the
SRC coactivators revealed that, whereas Ser-505 and Ser-543
of AIB1 are conserved in SRC-1 and GRIP1, Ser-860 and
Ser-867 are unique for AIB1. Thus, different phosphorylation profiles may contribute, in part, to the differential activity of AIB1, GRIP1, and SRC-1.
In addition to phosphorylation, recent studies have identified
sumoylation as another important post-translational modification that regulates the biological functions of a protein. In
sumoylation, SUMO (small ubiquitin-like modifier) is covalently attached to a lysine residue in a ␺KXE sequence (where ␺
is a large hydrophobic residue and X represents any amino acid)
on the substrate protein, especially nuclear proteins, via a pathway similar to ubiquitination in which a battery of activating
(E1), conjugating (E2), and ligating (E3) enzymes participate
(27). Similar to phosphorylation, sumoylation is a reversible
process, and desumoylation is the primary function of four
SUMO-specific proteases: SENP1 is a nuclear protease that
appears to deconjugate a large number of sumoylated proteins
(28 –31); SENP2 is a nuclear envelope-associated protease that
has an activity similar to that of SENP1 when overexpressed
(32, 33); SENP3 localizes in the nucleolus (28, 34), and SENP6
was found primarily in the cytoplasm (34, 35). SUMO modification correlates with either an enhanced or attenuated activity
of target proteins, for example, as has been shown for nuclear
receptors such as androgen receptor (36), glucocorticoid receptor (37), progesterone receptor (38, 39), and estrogen receptor
(40) and their cofactors, including SRC-1 (38), GRIP1 (41), and
p300 (42).
In this report, we demonstrated that SUMO-1 is conjugated
to AIB1 protein, and in contrast to GRIP1 and SRC-1, SUMO
modification attenuates, rather than enhances, the transactivation activity of AIB1. In addition, sumoylation of AIB1 coincided with a decreased level of AIB1 phosphorylation by
MAPK, whereas AIB1 phosphorylation by MAPK was concomitant with a loss of the SUMO moiety. Therefore, phosphorylation and sumoylation appear to coordinately regulate the transcriptional activity of AIB1.
Regulation of AIB1 by Sumoylation and Phosphorylation
no longer detectable, although the total AIB1 level was
unchanged (Fig. 1A, lower panel).
Most SUMO conjugates are nuclear proteins, and it is likely
that most of the major functions of SUMO take place in the
21850 JOURNAL OF BIOLOGICAL CHEMISTRY
VOLUME 281 • NUMBER 31 • AUGUST 4, 2006
Downloaded from www.jbc.org at Harvard Libraries on July 28, 2006
FIGURE 1. AIB1 is modified by SUMO-1 in the nucleus. A, MCF-7 cells were
treated with E2 at the indicated periods of time. Cellular protein extracts were
prepared and immunoprecipitated (IP) with anti-AIB1 antibodies followed by
Western blotting (WB) analyses with anti-SUMO-1 or anti-AIB1 antibodies. B, the
cytosolic and nuclear protein extracts from MCF-7 cells treated or untreated with
E2 for 15 min were first immunoprecipitated with anti-AIB1 antibodies followed
by Western blotting with the anti-SUMO-1 antibody. Fibrillarin and ␤-actin were
measured by Western blotting to monitor the efficiency in preparation of nuclear
and cytosolic protein extracts, respectively.
nucleus. In addition, AIB1 functions as a cofactor that coactivates estrogen receptor-mediated transcription in the nucleus
under the treatment of E2. In light of the observation that
sumoylated AIB1 was only detected in the absence of E2 stimulation, we wished to know whether the sumoylation of AIB1
occurred in cytoplasm or in the nucleus. For this purpose, we
prepared cytosolic and nuclear protein extracts separately from
MCF-7 cells cultured in the absence of E2. Immunoprecipitation was performed with these two cellular protein fractions
using the anti-AIB1 antibodies, and immunoprecipitates were
then probed with the anti-SUMO-1 by Western blotting. As
shown in Fig. 1B, sumoylated AIB1 was only detected in nuclear
fraction of the cellular protein extracts, indicating that sumoylated AIB1 localizes in the nucleus. The efficiency for preparation of nuclear extracts and cytosolic extracts was monitored by
measuring a nuclear protein marker, fibrillarin, and a cytosolic
protein marker, ␤-actin (Fig. 1B).
Identification of Sumoylation Sites in AIB1 Sequence—
SUMO attachment to target proteins occurs at specific lysine
residues, which are in most cases embedded in a consensus
sequence ␺KXE. In searching the amino acid stretch for the
consensus SUMO modification motifs, we found three potential SUMO-1 attachment sites, lysines 723, 786, and 1194, in the
AIB1 sequence. The lysines 723 and 786 locate in the first
nuclear receptor interaction domain. The Lys-1194 locates in
the Q-rich region downstream to the CREB-binding protein/
p300 interaction domain. Interestingly, the Lys-723 and Lys786 sites are conserved residues that are sumoylated in the two
other members of the SRC family, SRC-1 (Lys-732 and Lys-774)
and GRIP1 (Lys-731 and Lys-788) (38, 41). To examine whether
these sites indeed serve as SUMO-1 acceptors in AIB1 as well as
to test the effect of AIB1 sumoylation on its transactivation
activity, we first generated AIB1 mutants in which these lysine
residues were individually (K723R, K786R, or K1194R) or collectively (K723R/K786R/K1194R) changed to arginine residues. Wild-type (wt) and the mutated AIB1 were then fused to
Gal4-DNA binding domain (Gal4-DBD). The Gal4-DBD AIB1
fusions were then expressed in COS-7 cells with or without
cotransfection of a HA-tagged SUMO-1 plasmid. The cell
lysates were immunoprecipitated with an anti-Gal4-DBD antibody followed by immunoprobing with an anti-HA antibody.
As shown in Fig. 2A, HA tag could be detected in all three single
lysine-mutated AIB1, K723R, K786R, or K1194R, whereas in
AIB1 carrying K723R/K786R/K1194R, HA tag was no longer
detected, suggesting that lysines 723, 786, and 1194 could all be
the acceptors for SUMO conjugation in AIB1. The level of
Gal4-DBD expression was similar in all experimental conditions (Fig. 2A, lower panel).
To further define the sumoylation site(s) in AIB1, we next
constructed the following Gal4-DBD-fused AIB1 fragments:
N-terminal fragments (aa 1–740) containing Lys-723 or K723R,
N-terminal fragments (aa 1–792) containing K723R and Lys786 or K723R and K786R, and C-terminal fragments (aa 793–
1424) containing Lys-1194 or K1194R (Fig. 2B). These constructs were transfected into COS-7 cells together with the
HA-tagged SUMO-1 plasmid. Forty-eight hours after transfection, cellular extracts were immunoprecipitated with the antibody against Gal4-DBD, and the immunoprecipitates were
Regulation of AIB1 by Sumoylation and Phosphorylation
FIGURE 2. Identification of sumoylation sites in AIB1. A, COS-7 cells were
transfected with a SUMO-1 expression plasmid (HA-SUMO-1) along with Gal4DBD-fused wild-type or mutated AIB1 carrying K723R, K786R, K1194R, or
K723R/K786R/K1194R (3K/3R). Cells were harvested at 48 h after the transfection, and the cellular protein extracts were immunoprecipitated (IP) with antiGal4-DBD antibodies and subsequently immunoblotted (WB) with an anti-HA
antibody. B, Gal4-DBD-fused constructs with AIB1 fragments: N-terminal (aa
1–740) containing Lys-723 or K723R, N-terminal (aa 1–792) containing K723R
and Lys-786 or K723R and K786R, and C-terminal (aa 793–1424) containing
Lys-1194 or K1194R; SRC-1 fragments: N-terminal (aa 1–779) and C-terminal
(aa 780 –1441); and GRIP1 fragments: N-terminal (aa 1–789) and C-terminal
(aa 790 –1462). These constructs were transfected into COS-7 cells together
with the HA-tagged SUMO-1 plasmid. Forty-eight hours after transfection,
cellular extracts were immunoprecipitated with the antibody against Gal4DBD, and the immunoprecipitates were immunoblotted using the antibody
against HA.
immunoblotted using the antibody against HA. As shown in
Fig. 2B, SUMO conjugation was detected in the N-terminal
fragments containing Lys-723 or containing K723R and Lys786 but not in the N-terminal fragments containing K723R or
containing K723R and K786R. In the C-terminal fragments,
AUGUST 4, 2006 • VOLUME 281 • NUMBER 31
SUMO conjugation was detected in the AIB1 fragment containing Lys-1194 but not that containing K1194R. In addition,
corresponding C-terminal fragments of SRC-1 (aa 780 –1441)
or GRIP1 (aa 790 –1462) did not show any SUMO conjugations.
These experiments clearly showed that Lys-723, Lys-786, and
Lys-1194 are all sumoylation sites in AIB1 and an additional
sumoylation site exists in AIB1 compared with SRC-1 and
GRIP1.
Sumoylation of AIB1 Attenuates Its Transcriptional Activity—To examine the effect of AIB1 sumoylation on the
transactivation activity of AIB1, we next transfected COS-7
cells with a Gal4 promoter-driven luciferase (Gal4-luc) construct together with the Gal4-DBD-AIB1 (wt or mutants), and
the reporter activity was measured. As shown in Fig. 3, whereas
AIB1 carrying K1194R had a similar effect as wt AIB1 on Gal4luc activity, Gal4-luc activity was significantly increased in cells
that were transfected with AIB1 carrying either K723R, K786R,
or K723R/K786R/K1194R mutations (Fig. 3, upper panel). The
maximal Gal4-luc activity was observed in cells that were transfected with AIB1 carrying K723R/K786R/K1194R mutations.
In addition, the enhancement effect of these transfections on
the Gal4-luc activity was dose-dependent. Similar results were
obtained in COS-7 cells transfected with a luciferase expression
JOURNAL OF BIOLOGICAL CHEMISTRY
21851
Downloaded from www.jbc.org at Harvard Libraries on July 28, 2006
FIGURE 3. Sumoylation attenuated AIB1 transcriptional activity. A, COS-7
cells grown in the presence of E2 were transfected with increased amounts of
wild-type (WT) or mutated AIB1 fused to Gal4-DBD together with a Gal4luciferase reporter. Forty-eight hours after transfection, cells were harvested
and luciferase activity was determined. Each bar represents mean ⫾ S.D. from
three independent experiments. B, COS-7 cells grown in the presence of E2
were transfected with wild-type (WT) or mutated AIB1 together with an estrogen receptor-responsive element-luciferase reporter. Forty-eight hours after
transfection, cells were harvested and luciferase activity was determined.
Each bar represents mean ⫾ S.D. from three independent experiments.
Regulation of AIB1 by Sumoylation and Phosphorylation
21852 JOURNAL OF BIOLOGICAL CHEMISTRY
VOLUME 281 • NUMBER 31 • AUGUST 4, 2006
Downloaded from www.jbc.org at Harvard Libraries on July 28, 2006
port the notion that sumoylation of
AIB1 attenuated its transactivation
activity and to investigate which desumoylating enzyme might be
involved in regulating AIB1 activity,
we next cotransfected de-sumoylating enzyme SENP1, SENP2, or
SENP3 with Gal4-DBD-fused wt
AIB1 and AIB1 mutant 3K/3R. As
shown in Fig. 4, while cotransfection
of SENP2 or SENP3 had minimal
effects on Gal4-luc activity in COS-7
cells, cotransfection of SENP1
resulted in dramatic increases in the
Gal4-luc activity in cells transfected
with wt AIB1 (Fig. 4A). In AIB1
mutant 3K/3R-transfected cells, although the base level of the Gal4-luc
was higher than that of wt AIB1transfected cells, transfection of
SENP1 did not increase the Gal4-luc
activity. In addition, the effect of
SENP1 on Gal4-luc in wt AIB1-transfected cells was dose-dependent (Fig.
4B). Moreover, cotransfection of an
SENP1 mutant, which carries mutated residues (R630L/K631M) that
are crucial to its catalytic activity, did
not significantly alter Gal4-DBDAIB1-mediated transcription. Collectively, these data indicated that
SENP1 functions to de-sumoylate
AIB1. The expression of the Gal4DBD-fused wt AIB1 and SENP1 was
confirmed by Western blotting (Fig.
4C). Moreover, cotransfection of a
mutant of the E2 SUMO-conjugating enzyme Ubc9, Ubc9/C93S,
which had a dominant negative
effect on its wild type (29), enhanced
Gal4-DNA-AIB1-mediated transcription (data not shown). All these
data strongly supported the notion
FIGURE 4. SENP1 is responsible for de-conjugating the SUMO moiety from AIB1 protein. A, SENP1, but not that sumoylation of AIB1 attenuates
SENP2 or SENP3, markedly enhanced AIB1 transactivation activity. COS-7 cells were transfected with wild-type its transcriptional activity, and
AIB1 or 3K/3R mutant that were fused to Gal4-DBD together with a Gal4-luc reporter construct and constructs SENP1 is responsible for deconjufor the expression of SENP1, SENP1 mutant (mu), SENP2, or SENP3. Forty-eight hours after transfection, cells
were collected and the luciferase activity was measured. Each bar represents mean ⫾ S.D. from three inde- gating the SUMO mark from AIB1
pendent experiments. B, dose-dependent response of SENP1 action. Similar experiments stated above were protein.
performed with transfections of increased amounts of SENP1 or SENP1 mutant. C, Western blotting (WB)
AIB1 Modifications by Sumoylaanalysis of the expression of SENP1 and Gal4-DBD-fused AIB1.
tion and Phosphorylation—It has
construct driven by estrogen receptor-responsive element (Fig. been documented that AIB1 is regulated by phosphorylation,
3, lower panel). These data suggest that the sumoylation of and the transcriptional activity of AIB1 is enhanced by MAPK
activation (16, 25). MAPK can specifically phosphorylate AIB1
AIB1 attenuated its transactivation potential.
Sumoylation is a reversible chemical modification, and on Ser-505, Ser-543, Ser-860, and Ser-867 (16). To investigate
deconjugation of SUMO is believed to be a function of one of the mechanism of sumoylation and phosphorylation in relation
four SUMO-specific proteases: SENP1, SENP2, SENP3, or to AIB1 transcriptional activity, we first confirmed MAPK activaSENP6. Of these, SENP1, SENP2, and SENP3 function in the tion and AIB1 phosphorylation in our system. MCF-7 cells were
nucleus, whereas SENP6 is a cytoplasmic enzyme. To further sup- treated with E2 for different periods of time, and cellular pro-
Regulation of AIB1 by Sumoylation and Phosphorylation
tein extracts were prepared for Western blotting analysis of
p42/p44 MAPK activation and AIB1 phosphorylation. As
shown in Fig. 5A, E2 treatment resulted in increased levels of
phosphorylated ERK1/2 in MCF-7 cells; the effect was more
evident after 15 min of E2 treatment. Reprobing the immunoblots with antibodies against ERK1/2 showed that the
total level of ERK1/2 was not changed (Fig. 5A, lower panel).
In the meantime, immunoprecipitation with anti-AIB1 antibodies followed by immunoblotting with an anti-phosphorserine antibody showed E2-induced phosphorylation of AIB1
(Fig. 5B).
To examine the relationship between phosphorylation and
sumoylation of AIB1, MCF-7 cells were treated with E2, a pure
E2 antagonist ICI-182,780, or an ERK1/2-specific inhibitor
U0126, and the cellular proteins were extracted and immunoprecipitated with AIB1 antibodies followed by Western blotting
analysis of the immunoprecipitates with anti-SUMO-1 or antiphosphor-serine. As shown in Fig. 5C, E2 treatment was associated with an increased AIB1 phosphorylation and decreased
AUGUST 4, 2006 • VOLUME 281 • NUMBER 31
DISCUSSION
AIB1 is highly expressed in the mammary gland, uterus, testis, pituitary gland, and muscle (44) and is strongly amplified/
overexpressed in 64% of primary breast cancers as well as in
some of primary ovarian tumors (3, 45); thus, AIB1 has important physiological and pathological roles. At the molecular
level, AIB1, together with the other two members of the SRC
family of coactivators, GRIP1 and SRC-1, coactivates gene tranJOURNAL OF BIOLOGICAL CHEMISTRY
21853
Downloaded from www.jbc.org at Harvard Libraries on July 28, 2006
FIGURE 5. Coordinated modifications of AIB1 by SUMO and MAPK pathways. A, activation of MAPK by E2. MCF-7 cells were treated with E2 for the
time indicated. Cellular proteins were extracted, and Western blot analyses
were performed with antibodies against total or phosphorylated form of ERK.
B, MCF-7 cells were treated with E2 for the time indicated. Cellular proteins
were extracted and immunoprecipitated (IP) with anti-AIB1 antibodies followed by Western blotting (WB) analysis with an anti-phosphor-serine antibody. C, dynamic interplays between phosphorylation and sumoylation of
AIB1. Cellular protein extracts were prepared from MCF-7 cells that were
treated with or without E2 for 15 min in the presence of specific ERK inhibitor
U0126 or anti-estrogen ICI-182,780 (ICI). Protein extracts were then immunoprecipitated (IP) with anti-AIB1 antibodies followed by Western blotting analyses with anti-SUMO-1, anti-phosphor-serine, or anti-AIB1 antibodies.
AIB1 sumoylation, whereas both ICI-182,780 and U0126
induced increased AIB1 sumoylation but decreased AIB1 phosphorylation, indicating that the sumoylation and phosphorylation of AIB1 may occur in a coordinated fashion.
Coordinated Regulation of AIB1 Activity by Sumoylation and
Phosphorylation—To explore a functional correlation between
the coordinated sumoylation and phosphorylation of AIB1, we
next examined the effects of SUMO and MAPK pathways on
the transactivation potential of AIB1. The SUMO target-mutated AIB1 (K723R, K786R, K1194R, and K723R/K786R/
K1194R) as well as phosphorylation target-mutated AIB1
(S505A/S543A/S860A/S867A) were used in experiments to
analyze the effect of these AIB1 mutants on the coactivation of
the Gal4-luc activity under various experimental conditions,
including treatment with E2, ICI-182,780, or U0126 or cotransfection with SENP1 in COS-7 cells. As shown in Fig. 6, treatment with ICI-182,780 or U0126, which was associated with an
increased sumoylation and a decreased phosphorylation of
AIB1 as demonstrated above, resulted in decreased Gal4-luc
activities. Meanwhile, treatment with E2 or cotransfection of
SENP1 led to an increased Gal4-luc activity. The highest Gal4luc activity was detected with AIB1 mutant carrying all sumoylation site mutations (K723R/K786R/K1194R), whereas the
lowest Gal4-luc activity was detected with AIB1 mutant carrying all phosphorylation site mutations (S505A/S543A/S860A/
S867A). All these data suggest a coordinated pattern of sumoylation and phosphorylation in regulating AIB1 transcriptional
activity.
To further support the coordinated pattern of sumoylation
and phosphorylation of AIB1, MCF-7 cells were transfected
with the Gal4-DBD-fused wt AIB1 or the AIB1 mutant carrying
all sumoylation site mutations (K723R/K786R/K1194R) under
no treatment or treatment with E2, and coimmunoprecipitation experiments were performed with the antibody against
Gal4-DBD and then immunoblotted with anti-phosphor-serine. The results of the experiments indicated that phosphorylation was detected in sumoylation site-mutated AIB1 even
under no treatment of E2 (Fig. 6D, left panel). Similar experiments were performed with Gal4-DBD-fused wt AIB1 or the
AIB1 mutant carrying all phosphorylation site mutations
(S505A/S543A/S860A/S867A). The results of these experiments revealed the retention of sumoylation in phosphorylation site-mutated AIB1 even after the treatment of E2 (Fig. 6D,
right panel). These data strongly support a coordinated pattern
between phosphorylation and sumoylation in regulating AIB1
activity. Moreover, these data may also help to explain the
higher base levels of Gal4-luc activity in 3K/3R transfections
and the overall lower activation of the Gal4-luc activity in
4S/4A transfections described in Figs. 4A, 6A, 6B, and 6C.
Regulation of AIB1 by Sumoylation and Phosphorylation
scription mediated by nuclear receptors as well as by other
unrelated transcription factors, such as NF-␬B, AP-1, signal
transducers and activators of transcriptions, ETS, p53, and E2F
(46, 47). Although closely related, AIB1, GRIP1, and SRC-1
21854 JOURNAL OF BIOLOGICAL CHEMISTRY
VOLUME 281 • NUMBER 31 • AUGUST 4, 2006
Downloaded from www.jbc.org at Harvard Libraries on July 28, 2006
FIGURE 6. SUMO and MAPK pathways coordinated AIB1 transcriptional
activities. COS-7 cells were transfected with Gal4-luc and Gal4-DBD-AIB1
(wt or mutant) (3K/3R: K723R/K786R/K1194R; 4S/4A: S505A/S543A/S860A/
S867A) in the absence or presence of SENP1 plasmids. Twenty hours after
transfections, cells were treated with E2 (A), ICI-182,780 (ICI) (B), or U0126
(C) for an additional 12 h, and the luciferase activity was measured. Each
bar represents mean ⫾ S.D. from three independent experiments. D,
MCF-7 cells were transfected with the Gal4-DBD-fused wt AIB1 or the AIB1
mutant carrying all sumoylation site mutations (K723R/K786R/K1194R or
3K/3R) or the AIB1 mutant carrying all phosphorylation site mutations
(S505A/S543A/S860A/S867A) under no treatment or treatment with E2,
and coimmunoprecipitation experiments were performed with the antibody against Gal4-DBD and then immunoblotted with anti-phosphor-serine or with anti-SUMO-1.
exhibit differential functions that are manifested in animal phenotypes as well as in transcriptional outputs. Recently, we demonstrated that SRC coactivator proteins function in heterodimers in which AIB1 is the common dimerization partner
for GRIP1 and SRC-1 (20).
The basic mechanisms underlying ligand-dependent transcription initiation by nuclear receptors indicate a sequential
recruitment of an assortment of coactivators. Increasing numbers of coactivators have been identified in recent years, and
both biochemical and genetic studies have demonstrated that
these coactivators are differentially used by transcription factors, including nuclear receptors, in a cell/tissue type- and promoter-specific manner. However, the molecular basis underlying this specificity remains largely unknown. In the past years,
both nuclear receptors and cofactors have been shown to be
targeted for post-translational modifications by diverse cellular
signaling pathways. It is proposed that post-translational modifications of these cofactor proteins provide the basis for a combinatorial code required for specific gene activation by nuclear
receptors and coactivators and that this code also enables
coactivators to efficiently stimulate the activity of other classes
of transcription factors (46).
Phosphorylation is a fundamental mechanism of posttranslational modifications implicated in signal transduction. Phosphorylation of AIB1 has been shown to involve
different kinases, such as MAPK, I␬B kinase, and receptor
tyrosine kinase HER2/neu (16, 19, 24 –26, 48). It has been
shown that different signals differentially phosphorylate
AIB1 at different combinations of sites and that these phosphorylated profiles of AIB1 encode for distinct physiological
functions (16). In addition, studies in recent years have identified another important post-translational modification,
sumoylation. SUMO has been shown to covalently modify a
large number of proteins with important roles in many cellular processes, including gene expression, chromatin structure, signal transduction, and maintenance of the genome.
Relevant to this report, two members of the SRC family of
coactivators, GRIP1 and SRC-1, have been shown to be modified by SUMO (38, 41). In addition, sumoylated GRIP1 and
SRC-1 are associated with their enhanced transactivation
activity. In this report, we showed that the AIB1 is also a
sumoylated protein. But surprisingly, sumoylation attenuated AIB1 transactivation activity.
Currently, there are no clear molecular explanations for the
mechanism by which SUMO addition regulates transcription
factor activity. Clearly, modification with SUMO alters the surface of the target protein and might cause either general conformational changes or specific changes at critical interfaces,
thereby influencing the ability of the protein to interact with its
partners. One possibility is that sumoylation promotes or
inhibits protein-protein interactions and thereby regulates the
assembly of transcriptional complexes. Alternatively, sumoylation might block other lysine-targeted modifications such as
acetylation, methylation, or ubiquitination. Other possibilities
include subnuclear redistribution of sumoylated transcription
regulators. Sumoylation of transcription regulators could serve
to organize specific protein complexes into nuclear matrix sites
within nuclear bodies, thus regulating their activities either
Regulation of AIB1 by Sumoylation and Phosphorylation
REFERENCES
1. Glass, C. K., and Rosenfeld, M. G. (2000) Genes Dev. 14, 121–141
2. Shang, Y., Hu, X., DiRenzo, J., Lazar, M. A., and Brown, M. (2000) Cell 103,
843– 852
3. Anzick, S. L., Kononen, J., Walker, R. L., Azorsa, D. O., Tanner, M. M.,
Guan, X. Y., Sauter, G., Kallioniemi, O. P., Trent, J. M., and Meltzer, P. S.
(1997) Science 277, 965–968
4. Hong, H., Kohli, K., Trivedi, A., Johnson, D. L., and Stallcup, M. R. (1996)
Proc. Natl. Acad. Sci. U. S. A. 93, 4948 – 4952
5. Li, H., Gomes, P. J., and Chen, J. D. (1997) Proc. Natl. Acad. Sci. U. S. A. 94,
8479 – 8484
6. Onate, S. A., Tsai, S. Y., Tsai, M. J., and O’Malley, B. W. (1995) Science 270,
1354 –1357
7. Heery, D. M., Kalkhoven, E., Hoare, S., and Parker, M. G. (1997) Nature
387, 733–736
AUGUST 4, 2006 • VOLUME 281 • NUMBER 31
8. Darimont, B. D., Wagner, R. L., Apriletti, J. W., Stallcup, M. R., Kushner,
P. J., Baxter, J. D., Fletterick, R. J., and Yamamoto, K. R. (1998) Genes Dev.
12, 3343–3356
9. McInerney, E. M., Rose, D. W., Flynn, S. E., Westin, S., Mullen, T. M.,
Krones, A., Inostroza, J., Torchia, J., Nolte, R. T., Assa-Munt, N., Milburn, M. V., Glass, C. K., and Rosenfeld, M. G. (1998) Genes Dev. 12,
3357–3368
10. Sheppard, H. M., Harries, J. C., Hussain, S., Bevan, C., and Heery, D. M.
(2001) Mol. Cell. Biol. 21, 39 –50
11. Stallcup, M. R. (2001) Oncogene 20, 3014 –3020
12. Xu, J., Qiu, Y., DeMayo, F. J., Tsai, S. Y., Tsai, M. J., and O’Malley, B. W.
(1998) Science 279, 1922–1925
13. Weiss, R. E., Xu, J., Ning, G., Pohlenz, J., O’Malley, B. W., and Refetoff, S.
(1999) EMBO J. 18, 1900 –1904
14. Gehin, M., Mark, M., Dennefeld, C., Dierich, A., Gronemeyer, H., and
Chambon, P. (2002) Mol. Cell. Biol. 22, 5923–5937
15. Picard, F., Gehin, M., Annicotte, J. S., Rocchi, S., Champy, M. F., O’Malley,
B. W., Chambon, P., and Auwerx, J. (2002) Cell 111, 931–941
16. Wu, R. C., Qin, J., Yi, P., Wong, J., Tsai, S. Y., Tsai, M. J., and O’Malley,
B. W. (2004) Mol. Cell 15, 937–949
17. Torres-Arzayus, M. I., Font de Mora, J., Yuan, J., Vazquez, F., Bronson, R.,
Rue, M., Sellers, W. R., and Brown, M. (2004) Cancer Cell 6, 263–274
18. Xu, J., Liao, L., Ning, G., Yoshida-Komiya, H., Deng, C., and O’Malley,
B. W. (2000) Proc. Natl. Acad. Sci. U. S. A. 97, 6379 – 6384
19. Wang, Z., Rose, D. W., Hermanson, O., Liu, F., Herman, T., Wu, W.,
Szeto, D., Gleiberman, A., Krones, A., Pratt, K., Rosenfeld, R., Glass,
C. K., and Rosenfeld, M. G. (2000) Proc. Natl. Acad. Sci. U. S. A. 97,
13549 –13554
20. Zhang, H., Yi, X., Sun, X., Yin, N., Shi, B., Wu, H., Wang, D., Wu, G., and
Shang, Y. (2004) Genes Dev. 18, 1753–1765
21. Shang, Y., and Brown, M. (2002) Science 295, 2465–2468
22. Wu, H., Chen, Y., Liang, J., Shi, B., Wu, G., Zhang, Y., Wang, D., Li, R., Yi,
X., Zhang, H., Sun, L., and Shang, Y. (2005) Nature 438, 981–987
23. Rowan, B. G., Weigel, N. L., and O’Malley, B. W. (2000) J. Biol. Chem. 275,
4475– 4483
24. Lopez, G. N., Turck, C. W., Schaufele, F., Stallcup, M. R., and Kushner, P. J.
(2001) J. Biol. Chem. 276, 22177–22182
25. Font de Mora, J., and Brown, M. (2000) Mol. Cell. Biol. 20, 5041–5047
26. Shou, J., Massarweh, S., Osborne, C. K., Wakeling, A. E., Ali, S., Weiss, H.,
and Schiff, R. (2004) J. Natl. Cancer Inst. 96, 926 –935
27. Gill, G. (2004) Genes Dev. 18, 2046 –2059
28. Nishida, T., Tanaka, H., and Yasuda, H. (2000) Eur. J. Biochem. 267,
6423– 6427
29. Cheng, J., Wang, D., Wang, Z., and Yeh, E. T. (2004) Mol. Cell. Biol. 24,
6021– 6028
30. Gong, L., Millas, S., Maul, G. G., and Yeh, E. T. (2000) J. Biol. Chem. 275,
3355–3359
31. Bailey, D., and O’Hare, P. (2002) J. Gen. Virol. 83, 2951–2964
32. Hang, J., and Dasso, M. (2002) J. Biol. Chem. 277, 19961–19966
33. Zhang, H., Saitoh, H., and Matunis, M. J. (2002) Mol. Cell. Biol. 22,
6498 – 6508
34. Yeh, E. T., Gong, L., and Kamitani, T. (2000) Gene (Amst.) 248, 1–14
35. Kim, K. I., Baek, S. H., Jeon, Y. J., Nishimori, S., Suzuki, T., Uchida, S.,
Shimbara, N., Saitoh, H., Tanaka, K., and Chung, C. H. (2000) J. Biol.
Chem. 275, 14102–14106
36. Poukka, H., Karvonen, U., Janne, O. A., and Palvimo, J. J. (2000) Proc. Natl.
Acad. Sci. U. S. A. 97, 14145–14150
37. Tian, S., Poukka, H., Palvimo, J. J., and Janne, O. A. (2002) Biochem. J. 367,
907–911
38. Chauchereau, A., Amazit, L., Quesne, M., Guiochon-Mantel, A., and
Milgrom, E. (2003) J. Biol. Chem. 278, 12335–12343
39. Abdel-Hafiz, H., Takimoto, G. S., Tung, L., and Horwitz, K. B. (2002)
J. Biol. Chem. 277, 33950 –33956
40. Sentis, S., Le Romancer, M., Bianchin, C., Rostan, M. C., and Corbo, L.
(2005) Mol. Endocrinol. 19, 2671–2684
41. Kotaja, N., Karvonen, U., Janne, O. A., and Palvimo, J. J. (2002) J. Biol.
Chem. 277, 30283–30288
42. Girdwood, D., Bumpass, D., Vaughan, O. A., Thain, A., Anderson, L. A.,
JOURNAL OF BIOLOGICAL CHEMISTRY
21855
Downloaded from www.jbc.org at Harvard Libraries on July 28, 2006
negatively or positively. In any event, how sumoylation affects
the transcription activity of its target protein remains an
intriguing question for further investigation. Our observation
that sumoylation renders AIB1 functionally opposite to its close
cousins GRIP1 and SRC-1 is even more intriguing; after all,
these proteins are so much alike. We reported a sumoylation
site in AIB1 (Lys-1194) outside of the nuclear receptor box,
which has not been reported for GRIP1 and SRC-1. Interestingly, AIB1 was also shown to harbor unique MAPK phosphorylation sites that are not conserved in SRC-1 and GRIP1;
among MAPK-targeted Ser-505, Ser-543, Ser-860, and Ser-867
of AIB1, Ser-505 and Ser-543 are conserved in SRC-1 and
GRIP1, and Ser-860 and Ser-867 are unique for AIB1 (16).
Whether or not these unique modifications in AIB1 contribute
in anyway to specific AIB1 activities will be a subject of future
investigations. Nevertheless, it is clear that functional specificity of a protein is partly determined by the specificity of the
regulation/modification of the protein, and, as mentioned
above, AIB1, GRIP1, and SRC-1 exhibit differential activities
that may be consequences of differential post-translational
modifications.
Dynamic interplays by different post-translational modifications are important for functional dynamics of proteins. The
antagonistic relationship between phosphorylation and sumoylation has also been reported for transcription factor EIk-1 (49).
Our results indicated that a dynamic phosphorylation and
sumoylation may play important roles in regulating AIB1 activity. The overall transactivation potential of AIB1 could be
enhanced by activation of the MAPK pathway accompanying a
loss of SUMO mark in AIB1. On the other hand, the transcription activation potential of AIB1 is dampened upon its sumoylation, which is concomitant with its dephosphorylation. Thus,
it is logical to assume that a balance between phosphorylation
and sumoylation is critical in the final readout of the AIB1 activity. In addition, it is not clear how E2 treatment leads to desumoylation of AIB1. Clearly, the fast effect of E2 treatment on
the sumoylation status of AIB1 suggests a non-genomic action
for estrogen. It is conceivable that, similar to the activation of
MAPK pathway, estrogen treatment emanates stimuli that activate signaling pathways responsible for SUMO de-conjugation.
Future studies will focus on the delineation of the molecular
details that influence the switch between phosphorylation and
sumoylation.
Regulation of AIB1 by Sumoylation and Phosphorylation
Snowden, A. W., Garcia-Wilson, E., Perkins, N. D., and Hay, R. T. (2003)
Mol. Cell 11, 1043–1054
43. Tsai, E., Jain, J., Pesavento, P., Rao, A., and Goldfeld, A. (1996) Mol. Cell.
Biol. 16, 459 – 467
44. Suen, C. S., Berrodin, T. J., Mastroeni, R., Cheskis, B. J., Lyttle, C. R., and
Frail, D. E. (1998) J. Biol. Chem. 273, 27645–27653
45. Bautista, S., Valles, H., Walker, R. L., Anzick, S., Zeillinger, R., Meltzer, P.,
and Theillet, C. (1998) Clin. Cancer Res. 4, 2925–2929
46. Wu, R. C., Smith, C. L., and O’Malley, B. W. (2005) Endocr. Rev. 26,
393–399
47. Yin, N., Wang, D., Zhang, H., Yi, X., Sun, X., Shi, B., Wu, H., Wu, G., Wang,
X., and Shang, Y. (2004) Cancer Res. 64, 5870 –5875
48. Wu, R. C., Qin, J., Hashimoto, Y., Wong, J., Xu, J., Tsai, S. Y., Tsai, M. J., and
O’Malley, B. W. (2002) Mol. Cell. Biol. 22, 3549 –3561
49. Yang, S. H., Jaffray, E., Hay, R. T., and Sharrocks, A. D. (2003) Mol. Cell 12,
63–74
Downloaded from www.jbc.org at Harvard Libraries on July 28, 2006
21856 JOURNAL OF BIOLOGICAL CHEMISTRY
VOLUME 281 • NUMBER 31 • AUGUST 4, 2006