You are on page 1of 12

Estrogen Signaling:

A Subtle Balance
ER ER
Between ER and ER
he biological actions of estrogens are mediated by estrogen binding to one of two specific

T estrogen receptors (ERs), ER and ER, which belong to the nuclear receptor superfamily, a

family of ligand-regulated transcription factors. ER and ER are products of different genes and

exhibit tissue- and cell-type specific expression. The characterization of mice lacking ER, or ER,

or both has revealed that both receptor subtypes have overlapping but also unique roles in

estrogen-dependent action in vivo. Additionally, ER and ER have different transcriptional

activities in certain ligand, cell-type, and promoter contexts. Both receptors, however, are

coexpressed in a number of tissues and form functional heterodimers. The biological roles of

ER/ heterodimers in the presence of each respective homodimer are unknown. When

coexpressed, ER exhibits an inhibitory action on ER-mediated gene expression and in many

instances opposes the actions of ER. A number of ER and ER isoforms have also been

described, many of which alter estrogen-mediated gene expression. Uncovering the molecular

mechanisms regulating the expression of both ERs, and how ER and ER directly or indirectly

affect each other’s function are paramount to understanding the cellular and biological events of

estrogen-mediated gene regulation in normal and diseased tissues.

Jason Matthews and Jan-Åke Gustafsson


Department of Biosciences at Novum, Karolinska Institutet,
S-141 57 Huddinge Sweden
August 2003
Volume 3, Issue 5 281
Review

INTRODUCTION specificity, is identical between the two receptors. Thus, it is not


surprising that both receptors bind estrogen responsive elements
Estrogens regulate many physiological processes, including normal (EREs) with similar specificity and affinity, although differential
cell growth, development, and tissue-specific gene regulation in subtype affinities and responses to a subset of natural EREs have
the reproductive tract and in the central nervous and skeletal been described (8). The ligand-binding domains (LBDs) are also
systems (1, 2). Estrogens also influence the pathological processes conserved and both receptors exhibit similar affinities for the
of hormone-dependent diseases, such as breast, endometrial, and endogenous estrogen, 17-estradiol (E2). However, ER and ER
ovarian cancers, as well as osteoporosis (2). The biological actions exhibit different affinities for some natural compounds and novel
of estrogens are mediated by binding to one of two specific subtype-specific ligands have been reported (9, 10).
estrogen receptors (ERs), ER or ER, which belong to the Characterization of mice lacking either ER, or ER, or both
nuclear receptor (NR) superfamily, a family of ligand-regulated (ERKO, ERKO, and ERKO, respectively) has demonstrated
transcription factors (1). Ligand-binding induces conformation that each subtype has similar but also unique roles in estrogen
changes in the receptor leading to dimerization, protein–DNA action in vivo. Both ERs are widely distributed throughout the
interaction, recruitment of coregulator proteins, and other body, displaying distinct but overlapping expression patterns in a
transcription factors, and ultimately the formation of the variety of tissues (1, 2). ER is expressed primarily in the uterus,
preinitiation complex (3). ERs regulate gene expression by binding liver, kidney, and heart, whereas ER is expressed primarily in the
to their cognate response element or through protein–protein ovary, prostate, lung, gastrointestinal tract, bladder, and
interactions with other transcription factors (4). The discovery in hematopoietic and central nervous systems. ER and  are,
rat prostate of ER (5) and its subsequent cloning in other species however, coexpressed in a number of tissues including the
(6) has caused a paradigm shift in our understanding of estrogen mammary gland, epididymis, thyroid, adrenal, bone, and certain
action. regions of the brain. Although both ER subtypes may be expressed
ER and ER contain the evolutionarily conserved structural in the same tissue, they might not be expressed in the same cell
and functional domains typical of NR family members, including type. Nonetheless, ER and ER proteins have been
domains involved in DNA-binding, dimerization, ligand binding, simultaneously detected in many cell types including neurons and
and transcriptional activation (7) (Figure 1). ER and ER share a thymocytes (11, 12), and these as well as other cell types that
high degree of sequence identity within their DNA-binding coexpress both ER subtypes are targets for heterodimerization and
domains (DBDs). The amino acid sequence of the P-box, a motif potential interplay between the two receptors.
within the DBD critical for receptor–DNA recognition and The transactivating functions of ER and ER are mediated
by two separate but not mutually exclusive transcription activation
DNA binding Ligand binding functions (AFs) that allow the receptors to stimulate the
AF-1 transcription of estrogen-regulated genes: an N-terminal ligand-
dimerization dimerization AF-2
independent activation function (AF-1), and a C-terminal ligand-
hER A/B C D E F
dependent activation function (AF-2) located within the LBD (7).
The AFs contribute to estrogen-mediated transcription and
hER-46
mediate cell- and promoter-specificity. A comparison of the AF-1
domains of the two ERs has revealed that this domain is very
hER1long
active in ER on a variety of estrogen responsive promoters, but
under identical conditions, the activity of AF-1 in ER is minimal
hER1short
(13). Furthermore, these two receptors exhibit distinctive
hER2 responses to the synthetic antiestrogens tamoxifen and raloxifene.
For example, these ligands are partial ER agonists for ER but act
hER5 as pure ER antagonists for ER (13). The differences between the
respective N-terminal regions of the ERs have been suggested as a
hER4 possible explanation for their diversity of responsiveness to several
ligands. The AF-2 mediated transcriptional activities of ERs are
hER5 dependent on interactions with and recruitment of cofactor
Figure 1. Schematic representation of human estrogen proteins to estrogen responsive promoters. Cofactors can be
receptor  and  and their respective isoforms. Note that classified as coactivators, which promote NR activity, or
both hER2 and hER∆5 can have either the hER1 long or the corepressors, which attenuate NR activity.
short N-termini. In addition, there is no evidence of full-length Crystallographic studies have revealed that the LBDs of both
hER4 or hER5. The different fill patterns of the 3' end of hER2,
4, and 5 represent the divergent C-terminal regions of these
ER and ER share a similar overall architecture and have
isoforms. demonstrated that the AF-2 interaction surfaces are composed of

282
Biological Actions of ER and ER

amino acids in helices 3, 4, 5, and 12, and that binding of ligands regulating the expression of both ERs, and the complex interplay
alters the position of helix 12. When the ER LBD is complexed between the two receptors is paramount in understanding the
with agonists such as E2, helix 12 is positioned over the ligand- cellular and biological events of estrogen-mediated gene
binding pocket and forms an interaction surface for the regulation. This review focuses on the molecular mechanisms that
recruitment of coactivators. In contrast, when either the ER- or regulate expression of the ER and ER genes and how ER and
ER-LBDs are complexed with antagonists, helix 12 is displaced ER directly or indirectly affect the expression of the other
from its agonist position and occupies the hydrophobic groove subtype.
formed by helices 3, 4, and 5, causing helix 12 to disrupt the
coactivator interaction surface (14, 15). Functional and structural ER AND ER ISOFORMS
studies have shown that coactivators interact with the AF-2 region
via short leucine-rich motifs (i.e., LxxLL) termed “NR boxes” to ER and ER are products of distinct genes on different
transduce ligand signals to the basal transcriptional machinery. chromosomes. ER is located at chromosomal locus 6q25.1 (25),
These motifs represent the primary docking sites to the AF-2 whereas ER is found at position 14q22–24 (26). Several ER and
domain and exist in many cofactors (16). A large subset of these ER splicing variants have been described, but whether all
proteins include the steroid receptor coactivator (SRC) family transcripts are expressed as functional proteins and have biological
(SRC1, SRC2/TIF2/GRIP1, and SRC3/AIB1/RAC3) (17) and functions remains unclear.
components of the mammalian mediator complex [thyroid The generation of human ER mRNA transcripts is a
receptor associated proteins (TRAP), vitamin-D receptor complex process that involves at least seven different promoters
interacting proteins (DRIP), activator-recruited cofactor (ARC)] (3, and exhibits cell line-dependent promoter usage [reviewed in
18), which possess chromatin-remodeling ability and tether (27)]. Most ER variants differ only in the 5' UTR and result in the
activated receptors to the basal transcriptional machinery (3). expression of the full-length 66-kDa form of ER (27). Flouriot
Additional cofactors, such as the histone acetyltransferase (HAT), and colleagues have, however, identified a shorter 46-kDa isoform
CREB-binding protein (CBP), the highly related p300, and the of ER generated from an internal ATG start codon (28) (Figure
histone methyltrasferases CARM-1 and PRMT1 are tethered to the 1). The shorter 46-kDa isoform of hER lacks exon 1 and
ERs through interactions with the SRC family of coactivators (3, consequently the N-terminal AF-1 region (28). This isoform is
18). The recruitment of cofactors with histone modifying and present in human osteoblasts and in the breast cancer cell line
chromatin remodeling activities by ERs overcomes the repressive MCF-7, and heterodimerizes with wild-type ER, thereby
features of chromatin leading to active transcription through the suppressing its AF-1–dependent transcriptional activity. This
general transcriptional machinery (19). inhibition was observed in HepG2 cells, where ER activity is
In the absence of ligand, some NRs associate with the NR primarily mediated through AF-1, but not in HeLa cells in which
corepressors, namely, silencing mediator of retinoic acid and estrogen responses are predominantly mediated through AF-2
thyroid hormone receptors (SMRT) and nuclear receptor (28). Although AF-1 specific repression mediated by the 46-kDa
corepressor (NCoR) (18). Both SMRT and NCoR recruit SIN3 and ER isoform might be necessary for cell proliferation, much of the
histone deacetylases (HDACs) to form a large corepressor complex identification and characterization has been performed in cell-lines
that contains histone deacetylase activity, implicating histone and this isoform has not yet been identified in human tissue
deacetylation in transcriptional repression (20). samples. Therefore, its precise role in modulating the pleiotrophic
It has been suggested that corepressors and deacetylases are effects of estrogen remains to be determined.
recruited to ER target genes where they antagonize coactivator and The first characterization of the ER promoter identified a
HAT complexes. Indeed the histone deacetylase activity recruited region referred to as exon 0N (29), and subsequent analysis
by corepressor complexes such as NCoR–SIN3–HDAC2 is identified a second upstream promoter region referred to as exon
required for the transcriptional repression of tamoxifen-bound 0K (30). The Human Genome Project has aided investigators in
ER, and the loss of corepressors might be one mechanism of identifying at least five additional exons located between the 0K
tamoxifen resistance (20). and 0N regions, suggesting that ER is under sophisticated
Although ER and ER share similar mechanisms of action, regulatory control similar to that reported for ER (31). In
several differences in the transcriptional abilities of each receptor addition, several alternative splicing variants of ER, some with
as well as distinct phenotypes between gene-null animals have extended N-termini and others with truncations and/or insertions
been identified, suggesting that these receptors may regulate in the C-terminal LBD have been reported (26) (Figure 1). The
distinct cellular pathways. When ERs are coexpressed, ER expression levels of many of these isoforms are higher in human
exhibits an inhibitory action on ER-mediated gene expression (4, breast tissues than that of wild-type ER, and there are data
21, 22). A number of isoforms of ER and ER have also been supporting the protein expression of several ER isoforms (32, 33).
described, many of which also alter estrogen-mediated gene The original human ER clone encoded a protein of 485 amino
expression (23, 24). Uncovering the molecular mechanisms acids (6); however, cloning of an additional N-terminal sequence

August 2003
Volume 3, Issue 5 283
Review

has extended the N-terminus resulting in a 530 amino acid protein progression of this type of neoplasia. Transcriptional enhancer
(34). These proteins have been designated ER1 long (530 aa) and elements located in the 5' UTR of the ER promoter have been
ER1 short (485 aa), and the ER1 long form is currently identified and shown to contribute to the differential expression of
regarded as the full-length wild-type ER. Although some ER in breast cancers. Two coregulator proteins, activator protein
transfection studies suggest that both forms are functionally 2- (AP2-) and estrogen receptor promoter B associated factor
equivalent (35), the possibly distinct roles of the variable N- (ERBF-1), are partly responsible for the enhancement of ER
termini have yet to be fully explored. Wilkinson and colleagues expression (44, 45); however, AP2- is not expressed in all breast
have described the identification of an putative upstream in-frame tumor cells and ERBF-1 specifically enhances the expression of
initiation codon in hER, yielding a predicted protein of 548 ER from a distal promoter region that may not be utilized in all
amino acids; unlike ER1, the ER 548 amino-acid isoform cell types (44).
exhibits tamoxifen and raloxifene partial-agonist responses on an The expression level of ER is also intricately regulated
ERE driven reporter gene (36). However, a recent study failed to through DNA methylation and chromatin condensation of
detect hER 548 in samples from African, Caucasian, and Asian promoter regions via hypermethylation of CpG islands in the ER
populations (37), therefore questioning the physiological relevance promoter. Hypermethylation of the ER promoter is associated
and importance of this isoform. with a marked decrease in ER mRNA expression among ER-
ER variants lacking exons 5 and/or 6 have been identified in expressing cancer cell lines, and the inhibition of DNA-
human cancers and various cell lines (38). Loss of exon 5 results methyltransferases reactivates ER expression in these cell lines
in a codon frameshift and produces a truncated receptor that lacks (46). Increased CpG methylation of the ER promoters is reported
the majority of the LBD. Coexpression of this isoform with either to be significantly more extensive in BRCA1-linked human tumors,
wild-type ER1 or ER inhibits E2-dependent ERE-driven gene suggesting that inhibition of this process may be of particular
activity; thus, this isoform may function as an ER inhibitor in vivo. benefit to patients that inherit a mutation in BRCA1 (47).
Another ER isoform, ER2 (also called ERcx) (24), is identical The molecular mechanisms that govern the regulation of ER
to the ER1 long form except that 26 unique amino-acid residues, expression are relatively unclear (29). To date, mRNA transcripts
termed exon 9, replace the amino acids of exon 8. ER2 does not that include the 0K versus the 0N exon are expressed in differing
bind ligand, nor does it activate the transcription of an estrogen- amounts in different cell lines (and types) though whether the five
sensitive reporter gene. Furthermore, ER2 shows preferential (thus far described) putative promoter regions exhibit a similar
heterodimerization with ER rather than with ER, inhibiting ER cell-type-specific usage and whether these transcripts encode
DNA binding and having a dominant-negative effect on ligand- receptors that differ from wild-type receptor remains to be
dependent ER reporter gene activity (24). Moreover, hER2 determined. Several response elements have been identified in the
inhibits the transcriptional activity mediated by raloxifene-ER 5' UTR of the ER promoter, but no specific transcription factor
complexes on the raloxifene responsive tumor growth factor–beta has been identified that might contribute to the differential
3 (TGF-3) promoter, whereas no transcriptional inhibition was expression of ER protein. As in the case of ER, the ER
observed with ER1 on raloxifene–ER complexes (39). These promoter regions are also subject to DNA methylation. Of
data suggest that the ER2 and ER1 isoforms may differentially particular note is the ER 0N promoter, which is methylated in a
modulate estrogen action. Several additional ER isoforms have number of prostate cancer cell lines and carcinogenic tissue
been reported, although full-length sequences have not been samples (48). This may have important clinical implications
determined (40) (Figure 1). Some isoforms exhibit species-specific because ER is the dominantly expressed ER in the prostatic
expression; for example, an ER variant in rat termed ER 503 epithelium and ERKO mice display age-related prostatic
containing an eighteen amino-acid insertion in the LBD has not hyperplasia (49). Ongoing studies from our group indicate that the
been detected in humans. ER 503 and ER1 heterodimerize and 0K and 0N promoters are differentially methylated in a wide range
bind to consensus EREs but the affinity of many ligands for ER of human breast cancer cells, supporting the findings that DNA
503 is 1000-fold lower than for ER1 (41). methylation is an important regulatory mechanism of ER
expression levels.
REGULATION OF ER AND ER EXPRESSION LEVELS
THE EXPRESSION ER AND ER IS AUTOREGULATED
The relative expression levels of ER and ER are significantly
altered during the development of breast cancer (42,43); however, Several groups have demonstrated that exposure to estrogen can
the molecular details surrounding the receptors’ regulation remain modulate the expression of ER and ER (50–52), but little is
poorly understood. known about the biological importance and the molecular
The mechanisms that regulate ER mRNA levels have been mechanisms involved in this autoregulation. In the case of ER,
best studied in the context of primary breast carcinoma and breast exposure to E2 results in a ligand-dependent increase in the
cancer cell lines, because ER participates in the initiation and activities of two distinct promoter regions, although it is not

284
Biological Actions of ER and ER

known whether all of the A


Direct DNA binding of ER homo- and heterodimers
distinct ER promoters are
responsive to estrogen (51,
AF-2 AF-2 AF-2 AF-2 AF-2 AF-2
52). It has been hypothesized

1
AF

AF

AF
-

-
AF

AF

AF
-1

-1

-1
that autoregulation may
contribute to ER ERE ERE ERE
overexpression in some breast ER homodimers ER homodimers ER/ER heterodimers
tumors (52). Similarly, little is
known about the potential
ER-dependent regulation or B
autoregulation of the ER Tethering to other transcription factors
promoters. Our group and
AF-2
others have observed a time-

AF
AF-2 AF-2 AF-2

-1

-1
AF
AF

AF
dependent estrogen-induced

-1
-1
increase in ER mRNA in the AF-2 AF-2
AF

AF
?
-1

-1
human breast cancer cell-line
T47D [(38) (unpublished
results)]. Subsequent
chromatin
ER ER ER/ER coexpression
immunoprecipitation (ChIP)
Figure 2. Alternative estrogen-dependent transcriptional regulation by ER–ER homo-
assays have revealed the time-
and heterodimers. The different modes of direct DNA binding of ER subtypes either as homo- or
dependent recruitment of heterodimers to estrogen response elements (EREs) (A). Representation of the transcriptional regulation by
ER to an ERE half-site in the ER subtypes through tethering to other transcription factors, such as activating protein 1 (AP-1) and
ER 0N promoter stimulating protein 1 (Sp1) (B). The relative estrogen-dependent transcriptional activities are represented by
(unpublished results). the magnitude of the arrow. ER and ER are represented in blue and white, respectively.
Interestingly, an ERE half-site
has also been identified in the 0K exon though it is unclear modes including direct DNA binding (as homodimers or as
whether ER or ER regulates this promoter. In contrast, analyses heterodimers) or through tethering to other transcription factors
of neuronal tissue from ERKO and from wild-type mice show such as activating protein-1 (AP-1) and stimulating protein 1
that estrogen treatment tends to decrease the number of ER- (Sp1) (Figure 2). In many instances ER and ER exhibit
immunoreactive cells in wild-type mice, whereas such cells were opposing actions in the regulation of several promoters and
increased ERKO mice (50). Moreover, long-term exposure of specific response elements. Tethered to AP-1, ER exhibits E2-
primary endothelial cells to E2 increases the expression of ER dependent activation of transcription at AP-1 sites, whereas E2-
but decreases that of ER (53). Taken together, these studies bound ER has no effect (4). Raloxifene binding to ER causes an
highlight the complex interplay between ER and ER in the increase in reporter gene expression, whereas binding to ER
regulation and autoregulation of their respective promoters and results in minimal activation examined under identical conditions
suggest that this regulation exhibits cell-type and tissue variation. (4). The ER–AP-1 complex mediates the estrogen-dependent
activation of the progesterone receptor promoter (55).
ER OPPOSES ER-MEDIATED TRANSCRIPTION ER and ER oppose each other’s function in the regulation
of the cyclin D1 promoter (21). Unlike ER, E2-bound ER
ER appears to act as a dominant regulator of estrogen signaling, represses cyclin D1 expression and blocks ER-E2–mediated
and when coexpressed with ER, ER causes a concentration- induction when both receptors are present. The increased
dependent reduction in ER-mediated transcriptional activation expression of the cyclin D1 gene participates in estrogen-provoked
(21, 54). These antagonistic effects of ER on ER might arise proliferation in vivo, and the cyclin D1 protein is overexpressed in
from differences in their respective transactivation regions (54). over 50% of mammary carcinomas. Several groups have shown
ER activation requires the combination of the two AFs for that ER and ER form functional heterodimers in vitro and in
synergistic transcriptional activation, but the individual regions vivo and that if both isoforms are expressed, the heterodimers
exhibit independent activity in a cell-type– and promoter- predominate (56). The role that heterodimers play in estrogen
dependent manner. Both subtypes contain the potent C-terminal signaling, specifically in mediating the antagonistic effects of ER
AF-2 (7), but unlike ER, ER contains a weaker N-terminal AF- on ER transcriptional activity, is unknown.
1, which might possess repressive activity (35). ERs also modulate target-gene expression through interaction
ERs can regulate gene expression through several different with the transcription factor Sp1 (57). Both ER and ER

August 2003
Volume 3, Issue 5 285
Review

specifically interact with Sp1, and agonists as well as antagonists 3/AIB1/RAC3 enhance ER-mediated transcription in a cell-type
activate the ER–Sp1 complex; however, little, if any, increase in specific manner (63, 64). The affinities of ER and ER for
transcriptional activation is observed for Sp1-associated ER different coactivators and various NR boxes present within
complexes (57). This differential responsiveness for each subtype coactivators also display ligand-dependent variability (65). For
has been mapped to the AF-1 domains––in domain swapping example, in the presence of E2, SRC-1 or SRC-2/TIF2/GRIP1 can
experiments, complete reversal of phenotypes was observed (57). be recruited to both ER subtypes with similar affinity, whereas in
Furthermore, coexpression of ER with increasing concentrations the presence of genistein, both SRC coactivators are more strongly
of ER results in repression of ER–Sp1 activity, supporting a recruited to ER than ER (65).
functional antagonism between ER and ER. The contexts of response elements and cell-type specificity
It may be argued that the ER-dependent antagonism of affect the actions of regulatory proteins, such as coactivators.
ER-mediated responses is restricted to a limited number of genes Although the mechanisms are not understood, it is clear that
and does not represent a general mechanism in ER signaling. within a regulatory protein complex, a given factor may activate
Nonetheless, global antagonism by ER on ER transcriptional transcription, repress it, or have no regulatory activity. Different
activity is supported by a number of recent studies describing response elements induce alterations in the efficiency of
ER-directed repression of several ER-mediated effects including cofactor–AF-2 interactions thereby inducing differential binding of
fat reduction and cellular proliferation in the uterus and prostate SRC coactivators to ER or ER in the presence of diverse ERE
(58). In addition, we have applied high density microarray analysis elements (66). These differences may serve as a basis for promoter-
of bone and liver tissue isolated from ERKO, ERKO, or specific activities between the two ER subtypes. In addition, the
ERKO animals, and the data support the repressive effects of activity of SRC-2/TIF2/GRIP1 can alter between that of a
ER on ER-mediated transcriptional activity (22). In the absence coactivator and a corepressor depending on the response element
of ER, ER partially mediates the effect of estrogen on gene (67). On the collagenase-3 gene, SRC-2/TIF2/GRIP1, but not SRC-
transcription. Collectively, data from cell-based assays, gene 1 or SRC-3/AIB1/RAC3, enhances glucocorticoid
expression studies, and analysis of ER null animals strongly receptor–mediated transcriptional repression (67). These data
support the contention that ER attenuates ER activity. suggest that differential recruitment and/or variability in the
expression levels of coactivators can play a significant role in
RECRUITMENT OF SUBTYPE-SELECTIVE COFACTORS mediating transcriptional differences between ER and ER.

There has been a strong focus on the cloning of a large number of SUBTYPE-SPECIFIC TARGET GENES
NR coactivators and corepressors and the characterization their
respective enzymatic activities (18). These proteins participate in ER and ER exhibit similar affinity for different EREs (8), which
the determination of target-gene expression and act to bridge is not surprising given the high degree of sequence identity
activated receptors to the basal transcriptional machinery. The between their DBDs. Thus, to date, subtype-selective EREs have
precise roles of each type of coactivator, as well as the functional not been described, although there are examples of subtype-
interactions among them, during NR-dependent transcription have selective interactions at other response elements, such as SFREs
not been well defined. Altered expression of ER cofactors has been [steroidogenic factor 1 (SF-1) response elements]. Laudet and
reported during human breast tumorigenesis (59), and selective colleagues have explored the potential interplay among ER, ER,
cofactor recruitment by ER and ER may serve as a mechanism and members of the estrogen receptor related receptors (ERRs)
for differential transcriptional activities between the two receptors. (68). They have shown that in addition to binding EREs, ER and
A number of proteins have been isolated that interact in a ERRs also bind to SFRE whereas ER does not. Despite the ability
ligand-dependent manner via their NR-box sequences with the of ER and ER to form heterodimers, ER is unable to drive ER
AF-2 region of ERs. The thyroid receptor–associated protein 220 to the SFRE of an artificial reporter or to the SFRE-containing
(TRAP220), a component of the mediator complex, which bridges osteopontin promoter (68). This suggests that ER subtype-specific
ERs to RNA polymerase II, interacts with both ER and ER but regulation might preferentially take place with response elements
exhibits selective affinity for ER (60). This subtype selective other than those with an ERE background, or perhaps, through
affinity is influenced by the respective C-termini, the F domains tethering to additional transcription factors, as is observed at AP-
(61). The SRC family members are important mediators of AF- 1– or Sp1-responsive elements (4, 57). However, different EREs
2–dependent responses and have been shown to interact in vivo cause distinct conformational changes in receptors that are
with both ERs (17). SRC-3/AIB1/RAC3 is overexpressed in a independent of ligand-induced effects, and these changes in turn
variety of hormonally regulated cancers including breast, uterine, influence interactions with cofactors (66). Because the ERE
and prostate cancers (62), and overexpression of cofactors is sequence directly influences the amount of ER bound to DNA and
suspected to contribute to the enhancement of estrogen-dependent indirectly influences the affinity of ER for coactivators, the context
tumorigenesis (59). Both SRC-2/TIF2/GRIP1 and SRC- of the ERE may also contribute to subtype-specific gene

286
Biological Actions of ER and ER

ER ER
+ E2 + E2

CBP S CBP
SRC
R C
AF-2 AF-2 AF-2 AF-2

-1
AF
AF

-1

AF
AF

-1
-1

Mediator CBP SRC Mediator


CBP S
RC
AF-2 AF-2 AF-2 AF-2

-1
AF
-1

pol II
AF

pol II

AF
AF

-1
-1

Figure 3. The influence of chromatin on the transcriptional activities of ER and ER. Both ER subtypes exhibit
comparable activity on naked DNA, but differential activity on chromatin templates. ER strongly activates transcription on chromatin
templates, whereas chromatin structure is very restrictive for transcription mediated by ER. These differences may be due to the inability of
ER to recruit a stable preinitiation complex to chromatin templates, demonstrating that chromatin represents an important regulatory aspect
of ER-mediated gene expression.

regulation. Moreover, in light of the recent findings on the in the proper activation and repression of transcription has been
influence of chromatin on ER activity [see below, (69)], it is demonstrated experimentally in a number of in vitro and cell-
conceivable that although both subtypes are able to interact with based assays (70). The basic N-termini of histones are subject to a
an ERE in vitro, this interaction may not result in the formation of number of posttranslational modifications, including lysine
an active complex on chromatin templates in vivo. acetylation, lysine and arginine methylation, serine
The identification of subtype-specific target genes will be phosphorylation, and ubiquitinylation (71). These modifications
important for understanding the biological functions of each form the basis for the “histone code hypothesis,” which predicts
subtype but it will also present investigators with a significant that modifications of a histone tail influence the regulatory features
challenge. We and others have approached this problem by of target loci (72).
applying high-density microarray analysis to tissues isolated from Recently, the field of transcriptional regulation has witnessed
ER-null mice (as well as to cell lines that express both receptors) a rapid emergence of the ChIP assay, which is becoming a routine
followed by extensive bioinformatics to identify not only new technique for studying the temporal regulation of chromatin
regulatory pathways and new target genes, but also possible templates and has helped to uncover new mechanisms of
subtype-specific target genes. This strategy is central in clarifying transcriptional regulation. ChIP assays offer the powerful
the complexities of estrogen receptor signaling networks. advantage of studying transcription factor binding to endogenous
promoters in vivo under different physiological conditions. Two
INFLUENCE OF CHROMATIN ON ER TRANSCRIPTIONAL independent laboratories have investigated the dynamics of ligand-
ACTIVITY dependent cofactor recruitment by ER to estrogen responsive
promoters (73, 74). Their data demonstrate that ER reaches
In eukaryotes, DNA is associated with histones and organized into maximal occupancy of the cathepsin D and pS2 promoters after
nucleosomes, which together form chromatin. The packaging of forty-five to sixty minutes of treatment, although significant
genomic DNA into chromatin restricts access of the transcriptional promoter occupancy is also apparent minutes after exposure to E2
machinery, thereby causing a repression in transcription. Although [(74) and our unpublished data]. This may not be surprising
chromatin was once overlooked and considered a passive player in because significant levels of ER are bound to promoters in the
transcriptional regulation, the significant role that chromatin has absence of ligand (73, 74). Following the initial peak of binding

August 2003
Volume 3, Issue 5 287
Review

occupancy, ER leaves the promoters and returns in a second peak ER (78). Interestingly, both p68/p72 and TATA-binding protein
after 120 to 150 minutes of treatment (73, 74), suggesting that (TBP) show selectivity for the N-terminal A/B region of ER (78,
ER cycles on and off the promoter during active rounds of 79). Screening for novel subtype specific AF-1 interaction partners,
transcription. It has been demonstrated that recruitment of E3 and further biochemical and cell-based assays will be necessary to
ligases, enzymes that covalently add one or more ubiquitins to uncover the molecular mechanisms behind the differential
target proteins, and subunits of the 19S proteasome are necessary activities of ER and ER on chromatin templates.
in the cycling of ER to promoters (75). ChIP data also support
the sequential formation of complexes of different coregulators PLASMA MEMBRANE ESTROGEN RECEPTORS
during the onset of transcription; however, there are
inconsistencies concerning the simultaneous recruitment of Increasing evidence suggests that distinct pools of ERs that localize
specific cofactors, namely the mediator and p300/CBP–SRC to the plasma membrane play important roles in estrogen-
complexes (73, 74). Additionally, the kinetic profiles were not dependent responses. Several groups have demonstrated the E2-
sufficient to address the extent of cycling that occurs during induced rapid generation of a number of signaling cascades
promoter activation or the mechanisms and participants involved including cyclic AMP, inositol-1,4,5-trisphosphate (IP3),
in repression of these “activated promoters.” Moreover, little is phospholipase C, and members of the MAP kinase family
known about temporal recruitment of ER to estrogen responsive [reviewed in (80)]. Many of these effects are thought to arise
promoters and also about the potential role ER may exert on through the activation of signaling cascades by distinct pools of
dynamics of ER-mediated gene expression. ER has been ERs, which locate to the caveolae of the plasma membrane.
conclusively demonstrated to have an antagonistic effect on ER Transient transfection of both ER subtypes from single cDNAs
activity, and ChIP assays offer a powerful means of investigating results in a small but significant pool of receptors localizing to the
the effects of ER on ER and cofactor recruitment during the plasma membrane, suggesting that the plasma membrane pool of
activation and shut down of transcription on endogenous ERs arises from the same gene product as that localized to the
promoters. nuclear compartment (81). The precise mechanism and structural
Kraus and colleagues have recently investigated the potential features of ERs that result in membrane localization have not been
impact of chromatin on ER and ER-mediated transcriptional determined. Recently, however, Ser522 of ER has been shown to be
activities using a well-established in vitro chromatin assembly necessary for the efficient translocation and function of ER at the
assay (76). They have elegantly shown that complexes of the plasma membrane; mutation of this residue to alanine significantly
mediator, CBP/p300, and SRC are required for maximal reduces ER translocation to the plasma membrane (82).
transcriptional enhancement of ER-dependent chromatin Interestingly, Levin and colleagues have also reported the
templates and that the mediator has distinct functions during differential regulation of c-Jun kinase by the different membrane
transcriptional activation as compared to the p300/CBP–SRC pools of E2 bound ER and ER; whereas ER activated c-Jun N-
complex (77). The p300/CBP-SRC complex was primarily involved terminal kinase (JNK), ER inhibited its activation (81). The E2-
in the stable formation of the preinitiation complex in the initial dependent inhibition of JNK activation has been suggested as a
round of transcription, whereas the mediator was not only mechanism for breast cancer survival since the inhibition of JNK
involved in the first round of complex formation but also in prevents the inactivating phosphorylation of Bcl2 and the
subsequent rounds of reinitiation (77). Perhaps more intriguing subsequent activation of the caspases resulting in cell death (83).
was the finding that ER and ER exhibited similar abilities to These data demonstrate that opposing actions of ER subtypes not
activate transcription on naked DNA, but that ER is much more only occur at the transcriptional level but also in the plasma
efficient at activating transcription on chromatin templates (69) membrane pool of ERs, which could have significant
(Figure 3). The difference is attributable to the N-terminal AF-1 consequences for estrogen-dependent cell biology.
region of ER because the inability of ER to function on
chromatin templates was overcome by swapping its AF-1 region CONCLUSIONS AND FUTURE PROSPECTS
with that of ER. The studies of Kraus and colleagues suggest that
the AF-1 regions of the ERs harbor distinct features affecting the Despite being discovered almost ten years after ER, ER appears
recruitment of cofactors, which function on genomic templates to be the dominant player in a number of ER-dependent signaling
and not naked DNA. In agreement with other reports, pathways. A number of cell-based and animal models have
coexpression of ER and ER results in repression of ER- conclusively demonstrated that ER exhibits an antagonistic effect
mediated transcription (69). on ER-mediated transcriptional activity. As has been observed in
Relatively few proteins appear to bind primarily to the N- a number of other NR signaling paradigms, chromatin structure is
terminal AF-1 region of ERs. For example, the RNA-binding an important regulatory factor of ER activity. Uncovering the
protein p68/72, which is found in a complex containing SRC mechanisms behind the reduced activity of ER on chromatin
coactivators and the RNA coactivator SRA, binds selectively to templates and the role the AF-1 domain plays in mediating this

288
Biological Actions of ER and ER

effect will be important in interpreting the functional roles of each selective ligands and identification of subtype-specific signaling
ER subtype. Researchers are only beginning to decipher the pathways will be essential in planning novel therapeutic strategies
intricate interplay among ER, ER, and their respective isoforms, for the treatment estrogen-dependent pathologies.
and although a large body of data suggests that the functional
differences between the ER subtypes reside within the N-termini, Acknowledgments
continued efforts will undoubtedly uncover new modes of action
in ER signaling networks. The improved design of ER- and ER- This work was supported by grants from the Swedish Cancer

References elements. Nucleic Acids Res. 29, This is a key reference that
2905–2919 (2001). This paper is a describes how the structure of
1. Pettersson, K. and Gustafsson, J.-Å. very good account of the nuclear receptors, specifically ER
Role of estrogen receptor  in estrogen interactions of ER and ER with LBD, is altered upon
action. Annu. Rev. Physiol. 63, 165–192 numerous natural estrogen response agonist/antagonist binding.
(2001). elements. 15. Pike, A.C., Brzozowski, A.M.,
2. Couse, J.F. and Korach, K.S. Estrogen 9. Kuiper, G.G., Lemmen, J.G., Carlsson, Hubbard, R.E., Bonn, T., Thorsell,
receptor null mice: What have we B., Corton, J.C., Safe, S.H., van der A.G., Engstrom, O., Ljunggren, J.,
learned and where will they lead us? Saag, P.T., van der Burg, B., and Gustafsson, J.-Å., and Carlquist, M.
Endocr. Rev. 20, 358–417 (1999). Gustafsson, J.-Å. Interaction of Structure of the ligand-binding domain
3. Rachez, C. and Freedman, L.P. estrogenic chemicals and of oestrogen receptor  in the presence
Mediator complexes and transcription. phytoestrogens with estrogen receptor of a partial agonist and a full
Curr. Opin. Cell Biol. 13, 274–280 . Endocrinology 139, 4252–4263 antagonist. EMBO J. 18, 4608–4618
(2001). (1998). (1999).
4. Paech, K., Webb, P., Kuiper, G.G., 10. Sun, J., Meyers, M.J., Fink, B.E., 16. Leers, J., Treuter, E., and Gustafsson,
Nilsson, S., Gustafsson, J.-Å., Kushner, Rajendran, R., Katzenellenbogen, J.A., J.-Å. Mechanistic principles in NR box-
P.J., and Scanlan, T.S. Differential and Katzenellenbogen, B.S. Novel dependent interaction between nuclear
ligand activation of estrogen receptors ligands that function as selective hormone receptors and the coactivator
ER and ER at AP1 sites. Science estrogens or antiestrogens for estrogen TIF2. Mol. Cell. Biol. 18, 6001–6013
277, 1508–1510 (1997). An receptor- or estrogen receptor-. (1998).
extremely important paper. The first Endocrinology 140, 800–804 (1999). 17. Leo, C. and Chen, J.D. The SRC family
demonstration that ER and ER 11. Greco, B., Allegretto, E.A., Tetel, M.J., of nuclear receptor coactivators. Gene
can exert opposing effects in the and Blaustein, J.D. Coexpression of 245, 1–11 (2000).
modulation of gene expression. ER with ER and progestin receptor 18. Glass, C.K. and Rosenfeld, M.G. The
5. Kuiper, G.G., Enmark, E., Pelto- proteins in the female rat forebrain: coregulator exchange in transcriptional
Huikko, M., Nilsson, S., and Effects of estradiol treatment. functions of nuclear receptors. Genes
Gustafsson, J.-Å. Cloning of a novel Endocrinology 142, 5172–5181 (2001). Dev. 14, 121–141 (2000).
receptor expressed in rat prostate and 12. Mor, G., Munoz, A., Redlinger, R. Jr., 19. Kraus, W.L., and Wong, J. Nuclear
ovary. Proc. Natl. Acad. Sci. U.S.A. 93, Silva, I., Song, J., Lim, C., and Kohen, receptor-dependent transcription with
5925–5930 (1996). This paper F. The role of the Fas/Fas ligand system chromatin. Is it all about enzymes? Eur.
describes the initial cloning of ER, in estrogen-induced thymic alteration. J. Biochem. 269, 2275–2283 (2002).
which caused a paradigm shift in Am. J. Reprod. Immunol. 46, 298–307 20. Lavinsky, R.M., Jepsen, K., Heinzel, T.
our understanding on estrogen (2001). et al. Diverse signaling pathways
action. 13. Barkhem, T., Carlsson, B., Nilsson, Y., modulate nuclear receptor recruitment
6. Mosselman, S., Polman, J., and Enmark, E., Gustafsson, J.-Å., and of N-CoR and SMRT complexes. Proc.
Dijkema, R. ER: Identification and Nilsson, S. Differential response of Natl. Acad. Sci. U.S.A. 95, 2920–2925
characterization of a novel human estrogen receptor  and estrogen (1998).
estrogen receptor. FEBS Lett. 392, receptor  to partial estrogen 21. Liu, M.M., Albanese, C., Anderson,
49–53 (1996). agonists/antagonists. Mol. Pharmacol. C.M., Hilty, K., Webb, P., Uht, R.M.,
7. Nilsson, S., Makela, S., Treuter, E. et 54, 105–112 (1998). Price, R.H., Jr., Pestell, R.G., and
al. Mechanisms of estrogen action. 14. Brzozowski, A.M., Pike, A.C., Dauter, Kushner, P.J. Opposing action of
Physiol. Rev. 81, 1535–1565 (2001). Z. et al. Molecular basis of agonism estrogen receptors  and  on cyclin
8. Klinge, C.M. Estrogen receptor and antagonism in the oestrogen D1 gene expression. J. Biol. Chem. 277,
interaction with estrogen response receptor. Nature 389, 753–758 (1997). 24353–24360 (2002).

August 2003
Volume 3, Issue 5 289
Review

22. Lindberg, M.K., Moverare, S., Skrtic, Sonntag-Buck, V., and Gannon, F. heterodimerization with ER  in vivo
S., Gao, H., Dahlman-Wright, K., Identification of a new isoform of the and in vitro. Biochem. Biophys. Res.
Gustafsson, J.-Å., and Ohlsson, C. human estrogen receptor- (hER-) Commun. 243, 122–126 (1998).
Estrogen receptor (ER)- reduces ER- that is encoded by distinct transcripts 36. Hall, J.M. and McDonnell, D.P. The
regulated gene transcription, and that is able to repress hER- estrogen receptor -isoform (ER) of
supporting a “Ying Yang” relationship activation function 1. EMBO J. 19, the human estrogen receptor
between ER and ER in mice. Mol. 4688–4700 (2000). This paper modulates ER transcriptional activity
Endocrinol. 17, 203–208 (2003). In describes the identification and and is a key regulator of the cellular
this study, microarray analysis of characterization of the ER-46 kDa response to estrogens and
tissue isolated from ER null mice isoform and demonstrates that this antiestrogens, Endocrinology 140,
demonstrated a global repression of isoform inhibits the ligand- 5566–5578 (1999).
ER-mediated gene expression by independent activities mediated by 37. Wilkinson, H.A., Dahllund, J., Liu, H.,
ER. Thus, supporting the notion wild-type ER. Yudkovitz, J., Cai, S.J., Nilsson, S.,
that ER antagonizes the activities 29. Li, L.C., Yeh, C.C., Nojima, D., and Schaeffer, J.M., and Mitra, S.W.
of ER. Dahiya, R. Cloning and Identification and characterization of a
23. Inoue, S., Ogawa, S., Horie, K., characterization of human estrogen functionally distinct form of human
Hoshino, S., Goto, W., Hosoi, T., receptor  promoter. Biochem. Biophys. estrogen receptor . Endocrinology 143,
Tsutsumi, O., Muramatsu, M., and Res. Commun. 275, 682–689 (2000). 1558–1561 (2002).
Ouchi, Y. An estrogen receptor  30. Hirata, S., Shoda, T., Kato, J., and 38. Vladusic, E.A., Hornby, A.E., Guerra-
isoform that lacks exon 5 has Hoshi, K. The multiple untranslated Vladusic, F.K., and Lupu, R.
dominant negative activity on both first exons system of the human Expression of estrogen receptor 
ER and ER. Biochem. Biophys. Res. estrogen receptor  (ER ) gene. J. messenger RNA variant in breast
Commun. 279, 814–819 (2000). Steroid Biochem. Mol. Biol. 78, 33–40 cancer. Cancer Res. 58, 210–214
24. Ogawa, S., Inoue, S., Watanabe, T., (2001). (1998).
Orimo, A., Hosoi, T., Ouchi, Y., and 31. Sand, P., Luckhaus, C., Schlurmann, 39. Peng, B., Lu, B., Leygue, E., and
Muramatsu, M. Molecular cloning and K., Gotz, M., and Deckert, J. Murphy, L.C. Putative functional
characterization of human estrogen Untangling the human estrogen characteristics of human estrogen
receptor cx: A potential inhibitor of receptor gene structure. J. Neural receptor- isoforms. J. Mol. Endocrinol.
estrogen action in human. Nucleic Acids Transm. 109, 567–583 (2002). 30, 13–29 (2003).
Res. 26, 3505–3512 (1998). 32. Fuqua, S.A., Schiff, R., Parra, I. Et al. 40. Moore, J. T., McKee, D. D., Slentz-
25. Menasce, L.P., White, G.R., Harrison, Expression of wild-type estrogen Kesler, K. et al. Cloning and
C.J., and Boyle, J.M. Localization of receptor  and variant isoforms in characterization of human estrogen
the estrogen receptor locus (ESR) to human breast cancer. Cancer Res. 59, receptor  isoforms. Biochem. Biophys.
chromosome 6q25.1 by FISH and a 5425–5428 (1999). Res. Commun. 247, 75–78 (1998).
simple post-FISH banding technique. 33. Saji, S., Omoto, Y., Shimizu, C. et al. 41. Petersen, D.N., Tkalcevic, G.T., Koza-
Genomics 17, 263–265 (1993). Expression of estrogen receptor ERcx Taylor, P.H., Turi, T.G., and Brown,
26. Enmark, E., Pelto-Huikko, M., protein in ER-positive breast cancer: T.A. Identification of estrogen receptor
Grandien, K., Lagercrantz, S., Specific correlation with progesterone 2, a functional variant of estrogen
Lagercrantz, J., Fried, G., receptor. Cancer Res. 62, 4849–4853 receptor  expressed in normal rat
Nordenskjold, M., and Gustafsson, J.- (2002). tissues. Endocrinology 139, 1082–1092
Å. Human estrogen receptor -gene 34. Xu, L., Pan-Hammarstrom, Q., Forsti, (1998).
structure, chromosomal localization, A., Hemminki, K., Hammarstrom, L., 42. Leygue, E., Dotzlaw, H., Watson, P.H.,
and expression pattern. J. Clin. Labuda, D., Gustafsson, J.-Å., and and Murphy, L.C. Altered estrogen
Endocrinol. Metab. 82, 4258–4265 Dahlman-Wright, K. Human estrogen receptor  and  messenger RNA
(1997). receptor  548 is not a common expression during human breast
27. Reid, G., Denger, S., Kos, M., and variant in three distinct populations. tumorigenesis. Cancer Res. 58,
Gannon, F. Human estrogen receptor- Endocrinology 144, 3541–3546 (2003). 3197–3201 (1998).
: Regulation by synthesis, 35. Ogawa, S., Inoue, S., Watanabe, T., 43. Roger, P., Sahla, M.E., Makela, S.,
modification and degradation. Cell. Hiroi, H., Orimo, A., Hosoi, T., Ouchi, Gustafsson, J.-Å., Baldet, P., and
Mol. Life Sci. 59, 821–831 (2002). Y., and Muramatsu, M. The complete Rochefort, H. Decreased expression of
28. Flouriot, G., Brand, H., Denger, S., primary structure of human estrogen estrogen receptor  protein in
Metivier, R., Kos, M., Reid, G., receptor  (hER ) and its proliferative preinvasive mammary

290
Biological Actions of ER and ER

tumors. Cancer Res. 61, 2537–2541 M., and Gannon, F. Tissue-specific Troup, S., Adeyinka, A., and Watson, P.
(2001). expression of human ER and ER in H. Altered expression of estrogen
44. Tanimoto, K., Eguchi, H., Yoshida, T., the male. Mol. Cell. Endocrinol. 178, receptor coregulators during human
Hajiro-Nakanishi, K., and Hayashi, S. 155–160 (2001). breast tumorigenesis. Cancer Res. 60,
Regulation of estrogen receptor  gene 52. Castles, C.G., Oesterreich, S., Hansen, 6266–6271 (2000).
mediated by promoter B responsible R., and Fuqua, S.A. Auto-regulation of 60. Kang, Y.K., Guermah, M., Yuan, C.X.,
for its enhanced expressionin human the estrogen receptor promoter. J. and Roeder, R.G. The TRAP/Mediator
breast cancer. Nucleic Acids Res. 27, Steroid Biochem. Mol. Biol. 62, 155–163 coactivator complex interacts directly
903–909 (1999). (1997). with estrogen receptors  and 
45. Turner, B.C., Zhang, J., Gumbs, A.A. et 53. Ihionkhan, C.E., Chambliss, K.L., through the TRAP220 subunit and
al. Expression of AP-2 transcription Gibson, L.L., Hahner, L.D., directly enhances estrogen receptor
factors in human breast cancer Mendelsohn, M.E., and Shaul, P.W. function in vitro. Proc. Natl. Acad. Sci.
correlates with the regulation of Estrogen causes dynamic alterations in U.S.A. 99, 2642–2647 (2002).
multiple growth factor signalling endothelial estrogen receptor 61. Warnmark, A., Almlof, T., Leers, J.,
pathways. Cancer Res. 58, 5466–5472 expression. Circ. Res. 91, 814–820 Gustafsson, J.-Å., and Treuter, E.
(1998). (2002). Differential recruitment of the
46. Ottaviano, Y.L., Issa, J.P., Parl, F.F., 54. Pettersson, K., Delaunay, F., and mammalian mediator subunit
Smith, H.S., Baylin, S.B., and Gustafsson, J.-Å. Estrogen receptor  TRAP220 by estrogen receptors ER
Davidson, N.E. Methylation of the acts as a dominant regulator of and . J. Biol. Chem. 276,
estrogen receptor gene CpG island estrogen signaling. Oncogene 19, 23397–23404 (2001).
marks loss of estrogen receptor 4970–4978 (2000). 62. Anzick, S.L., Kononen, J., Walker, R.L.
expression in human breast cancer 55. Petz, L.N., Ziegler, Y.S., Loven, M.A., et al. AIB1, a steroid receptor
cells. Cancer Res. 54, 2552–2555 and Nardulli, A.M. Estrogen receptor  coactivator amplified in breast and
(1994). and activating protein-1 mediate ovarian cancer. Science 277, 965–968
47. Archey, W.B., McEachern, K.A., estrogen responsiveness of the (1997).
Robson, M., Offit, K., Vaziri, S. A., progesterone receptor gene in MCF-7 63. Suen, C. S., Berrodin, T.J., Mastroeni,
Casey, G., Borg, A., and Arrick, B.A. breast cancer cells. Endocrinology 143, R., Cheskis, B.J., Lyttle, C.R., and Frail,
Increased CpG methylation of the 4583–4591 (2002). D.E. A transcriptional coactivator,
estrogen receptor gene in BRCA1- 56. Pettersson, K., Grandien, K., Kuiper, steroid receptor coactivator-3,
linked estrogen receptor-negative G.G., and Gustafsson, J.-Å. Mouse selectively augments steroid receptor
breast cancers. Oncogene 21, estrogen receptor  forms estrogen transcriptional activity. J. Biol. Chem.
7034–7041 (2002). response element-binding 273, 27645–27653 (1998).
48. Sasaki, M., Tanaka, Y., Perinchery, G., heterodimers with estrogen receptor . 64. Monroe, D.G., Johnsen, S.A.,
Dharia, A., Kotcherguina, I., Fujimoto, Mol. Endocrinol. 11, 1486–1496 Subramaniam, M., Getz, B.J., Khosla,
S., and Dahiya, R. Methylation and (1997). S., Riggs, B.L., and Spelsberg, T.C.
inactivation of estrogen, progesterone, 57. Saville, B., Wormke, M., Wang, F., Mutual antagonism of estrogen
and androgen receptors in prostate Nguyen, T., Enmark, E., Kuiper, G., receptors  and  and their preferred
cancer. J. Natl. Cancer Inst. 94, Gustafsson, J.-Å., and Safe, S. Ligand-, interactions with steroid receptor
384–390 (2002). cell-, and estrogen receptor subtype coactivators in human osteoblastic cell
49. Weihua, Z., Warner, M., and (/)-dependent activation at GC-rich lines. J. Endocrinol. 176, 349–357
Gustafsson, J.-Å. Estrogen receptor  (Sp1) promoter elements. J. Biol. Chem. (2003).
in the prostate. Mol. Cell. Endocrinol. 275, 5379–5387 (2000). This is an 65. Routledge, E.J., White, R., Parker,
193, 1–5 (2002). important paper demonstrating the M.G., and Sumpter, J.P. Differential
50. Nomura, M., Korach, K.S., Pfaff, D.W., differences in responsiveness of ER effects of xenoestrogens on coactivator
and Ogawa, S. Estrogen receptor  and ER at Sp1 response elements. recruitment by estrogen receptor (ER)
(ER) protein levels in neurons 58. Weihua, Z., Makela, S., Andersson,  and ER. J. Biol. Chem. 275,
depend on estrogen receptor alpha L.C. et al. A role for estrogen receptor 35986–35993 (2000).
(ER) gene expression and on its  in the regulation of growth of the 66. Yi, P., Driscoll, M.D., Huang, J.,
ligand in a brain region-specific ventral prostate. Proc. Natl. Acad. Sci. Bhagat, S., Hilf, R., Bambara, R.A., and
manner. Brain Res. Mol. Brain Res. 110, U.S.A. 98, 6330–6335 (2001). Muyan, M. The effects of estrogen-
7–14 (2003). 59. Murphy, L.C., Simon, S.L., Parkes, A., responsive element- and ligand-
51. Denger, S., Reid, G., Brand, H., Kos, Leygue, E., Dotzlaw, H., Snell, L., induced structural changes on the

August 2003
Volume 3, Issue 5 291
Review

recruitment of cofactors and immunopreciptation assays to nuclear estrogen receptors (ERs)


transcriptional responses by ER  and demonstrate the cycling and originate from a single transcript:
ER . Mol. Endocrinol. 16, 674–693 temporal recruitment of ER and Studies of ER and ER expressed in
(2002). cofactors during the transcriptional Chinese hamster ovary cells. Mol.
67. Rogatsky, I., Luecke, H.F., Leitman, activation of the estrogen responsive Endocrinol. 13, 307–319 (1999).
D.C., and Yamamoto, K.R. Alternate cathepsin D promoter. 82. Razandi, M., Alton, G., Pedram, A.,
surfaces of transcriptional coregulator 74. Burakov, D., Crofts, L.A., Chang, C.P., Ghonshani, S., Webb, P., and Levin,
GRIP1 function in different and Freedman, L.P. Reciprocal E.R. Identification of a structural
glucocorticoid receptor activation and recruitment of DRIP/mediator and determinant necessary for the
repression contexts. Proc. Natl. Acad. p160 coactivator complexes in vivo by localization and function of estrogen
Sci. U.S.A. 99, 16701–16706 (2002). estrogen receptor. J. Biol. Chem. 277, receptor  at the plasma membrane.
68. Vanacker, J.M., Pettersson, K., 14359–14362 (2002). Mol. Cell. Biol. 23, 1633–1646 (2003).
Gustafsson, J.-Å., and Laudet, V. 75. Lonard, D.M., Nawaz, Z., Smith, C.L., 83. Razandi, M., Pedram, A., and Levin,
Transcriptional targets shared by and O’Malley, B.W. The 26S E.R. Plasma membrane estrogen
estrogen receptor- related receptors proteasome is required for estrogen receptors signal to anti-apoptosis in
(ERRs) and estrogen receptor (ER) , receptor- and coactivator turnover breast cancer. Mol. Endocrinol. 14,
but not by ER. EMBO J. 18, and for efficient estrogen receptor- 1434–1437 (2000).
4270–4279 (1999). transactivation. Mol. Cell 5, 939–948
69. Cheung, E., Schwabish, M.A., and (2000). Jason Matthews,
Kraus, W.L. Chromatin exposes 76. Kraus, W.L. and Kadonaga, J.T. p300 PhD, received his
intrinsic differences in the and estrogen receptor cooperatively undergraduate
transcriptional activities of estrogen activate transcription via differential degree from the
receptors  and . EMBO J. 22, enhancement of initiation and University of
600–611 (2003). A key reference that reinitiation, Genes Dev. 12, 331–342 Western Ontario in
describes for the first time the (1998). Canada before
influence of chromatin structure on 77. Acevedo, M.L. and Kraus, W.L. pursuing his
the transcriptional activities of ER Mediator and p300/CBP-steroid doctoral studies in biochemistry and
and ER. This study shows that the receptor coactivator complexes have environmental toxicology at Michigan State
AF-1 domain of ER, but not that of distinct roles, but function University in East Lansing. He is currently a
ER, contains a transferable synergistically, during estrogen postdoctoral fellow in Prof. Gustafsson’s
activation potential, which permits receptor -dependent transcription laboratory at the Karolinska Institute,
the receptor to efficiently activate with chromatin templates. Mol. Cell. Department of Biosciences in Huddinge,
transcription on chromatin Biol. 23, 335–348 (2003). Sweden.
templates. 78. Endoh, H., Maruyama, K., Masuhiro, Jan-Åke
70. Wolffe, A.P. Transcriptional regulation Y. et al. Purification and identification Gustafsson, MD,
in the context of chromatin structure. of p68 RNA helicase acting as a PhD, is Professor of
Essays Biochem. 37, 45–57 (2001). transcriptional coactivator specific for Medical Nutrition
71. Berger, S.L. An embarrassment of the activation function 1 of human and Director of the
niches: The many covalent estrogen receptor . Mol. Cell. Biol. 19, Center for
modifications of histones in 5363–5372 (1999). Biotechnology at
transcriptional regulation. Oncogene 79. Warnmark, A., Wikstrom, A., Wright, Novum, the South
20, 3007–3013 (2001). A.P., Gustafsson, J.-Å., and Hard, T. Campus of the Karolinska Institute; his
72. Strahl, B.D. and Allis, C.D. The The N-terminal regions of estrogen main interest is nuclear receptor signaling.
language of covalent histone receptor  and  are unstructured in He is foreign associate of the US National
modifications. Nature 403, 41–45 vitro and show different TBP binding Academy of Sciences. Address
(2000). properties. J. Biol. Chem. 276, correspondence to J-ÅG. E-mail jan-
73. Shang, Y., Hu, X., DiRenzo, J., Lazar, 45939–45944 (2001). ake.gustafsson@mednut.ki.se;
M.A., and Brown, M. Cofactor 80. Levin, E.R. Cellular functions of fax: + 46-8-779 87 95
dynamics and sufficiency in estrogen plasma membrane estrogen receptors.
receptor-regulated transcription. Cell Steroids 67, 471–475 (2002).
103, 843–852 (2000). An important 81. Razandi, M., Pedram, A., Greene, G.L.,
study that used chromatin and Levin, E.R. Cell membrane and

292

You might also like