Sunteți pe pagina 1din 8

Reviews in Endocrine & Metabolic Disorders 2002;3:193200

# 2002 Kluwer Academic Publishers. Manufactured in The Netherlands.

Estrogen Receptors: Structure, Mechanisms and Function


Sylvia Curtis Hewitt and Kenneth S Korach
Receptor Biology, LRDT, National Institute of Environmental Health
Sciences, NIH, PO Box 12233, Research Triangle Park, NC 27709

Key Words. estrogen receptor, reproductive tract, transgenic mice, for which no ligands have been identied [1]. A second
estrogen action ER gene was cloned from prostate tissue in 1996 [2], and
thus, there are two ER molecules: the original ER ``a'',
and the recently discovered ER ``b''. By comparing the
Introduction ER sequences it is apparent that both share a general
domain structure common to ligand modulated nuclear
The biological effects of estrogen (E) in mammalian target transcription factors [3]. The current understanding of
tissues are important for numerous physiological pro- ER mechanisms of action can be summarized with
cesses. E is known to induce responses in the reproductive reference to the overall structure of the ER molecules
tract, mammary tissue and pituitary but also affects non- (Fig. 1). The functions of some regions of the ER
reproductive processes such as bone formation and molecules have been dened using deletion and mutation
cardiovascular health. Since the initial identication and as well as structural analysis. The best characterized
isolation of the estrogen receptor (ER) molecule decades functions are a zinc-nger containing domain (C domain,
ago, the mechanism of estrogen (E) action in cells has been Fig. 1) that binds with high afnity and specicity to
intensely studied. At the core, our understanding of the EREs in target genes, and a ligand-binding domain
basic mechanism has remained the same, however, the (LBD, domain E), which binds E as well as other
details continue to be dened and many new aspects estrogenic ligands. The ERE is a 13 base pair inverted
remain to be discovered. The dogma of high afnity E repeat sequence (GGTCAnnnTGACC), and in vitro DNA
binding and modulation of transcription via high afnity binding studies have indicated that the ER binds as a
estrogen responsive elements (EREs) in target genes still dimer [4], with one ER molecule contacting each 5 base
remains the basis of E action in target tissues. However, pair inverted repeat [5]. Although the DNA binding is a
this model has become much more complex with the dimerization stimulus, sequences in the LBD are also
understanding that many other factors cooperate and involved in dimerization [4], and crystallized truncated
interact to modulate transcription. In addition there are ER containing only ligand binding domain is clearly
``twists'' in the ``plot'', in alternatives to E activation of shown to be a dimer in the presence of ligand [6].
ER as well as ER's ability to modulate transcription of In addition, regions in the amino terminus (AF-1) and
genes that lack EREs. Advances in technology and within the LBD (AF-2) are involved in ligand-indepen-
methods available for molecular, structural, biochemical, dent (AF-1) and ligand-dependent (AF-2) transcriptional
genetic and physiological analysis of ER function have activation, as deletion of or introduction of mutations in
allowed for the denition of many of the details of ER these regions result in a diminished ability to induce E
function in in vitro model systems and the expansion of responsive genes [7,8]. The mechanism by which
these models into genetically modied mouse models. transcription is mediated by ER is thought to be through
This chapter will review the current understanding of the interaction of AFs with ``transcriptional machinery'',
mechanism of ER function and then describe insights which is a general term referring to the complex of
gained from use of transgenic mouse models. molecules that assembles and ultimately results in
synthesis of mRNA [9]. Much is now known about
RNA polymerase II and the enzymes and factors that
Estrogen Receptors: Structure and allow transcription. Transcriptional co-regulators are
Mechanism of Action molecules that mediate the interaction between ERs
and the transcriptional machinery. Many co-regulators
ERs are members of a family of nuclear transcription
factors including receptors for sex steroids, thyroid Address correspondence to: Kenneth S. Korach.
hormone, retinoids as well as many ``orphan'' receptors, E-mail: korach@niehs.nih.gov

193
194 Hewitt and Korach

Fig. 1. Comparison of domain structures of ERa and ERb. The


estrogen receptors are members of the nuclear receptor superfamily, Fig. 2. Mechanisms of ER mediated transcription. ER-mediated
and share a domain structure, which is depicted schematically. The transcription is initiated following E binding or ligand-independent
ERs have 6 domains, AF, and the number of amino acids in these activation resulting from growth factor receptor pathway signaling
domains as well as the functions associated with these domains are and cross-talk with ER. Once activated, ER dimers recruit
indicated for each form of ER. AF-1 and AF-2 refer to regions that coactivators and can mediate transcription of genes via direct
mediate the transcriptional activation functions of the ERs. The binding to EREs in target genes (``classical'' mechanism).
degree of homology between ERa and ERb in the C and E domains Alternatively, ER can recruit coactivators to an AP-1 complex or
are indicated below these domains. remove repressors from the AP-1 complex (AP-1 mediated
mechanism). Finally, ER can interact with promoters containing Sp1
binding sequences and ERE-half sites (Sp1Nx ERE(1/2) mediated).
have been isolated that interact with ER in a ligand
dependent manner [911]. The most well-characterized
co-regulators interact with the AF-2 of the ER, although (Fig. 2). First, ER can regulate expression of genes that
recently molecules that interact with the N-terminus have lack EREs. One such mechanism that has been studied in
been described. Notably, the co-activator, p68, which some detail is the ER regulation of genes containing AP-
interacts with the AF-1 region of ERa, has RNA helicase 1 elements that bind the fos/jun dimer, which recruits
activity and associates with SRA, a RNA molecule with ERa and co-activators [16]. In addition, ER may also
co-activator activity [1214]. Tsai's review in this sequester repressors away from the fos/jun transcription
volume describes in greater detail the roles and apparatus, resulting in activation through AP-1 [16]. This
mechanisms of co-activators in steroid hormone action. alternative activity of ER adds complexity to the actions
The general mechanism of ER activation is depicted and roles of estrogen. The insulin-like growth factor
in Fig. 2. Upon binding to ligand, the conformation of the (IGF-1) gene is induced by E via an AP-1 element.
LBD is altered, allowing interaction with co-activators if Studies using aERKO mice indicated the IGF-1 pathway
the ligand is an agonist, but preventing this interaction in was not induced by E or various selective estrogen
the case of an antagonist [6,9]. Interestingly, crystal responsive modulators (SERMs), ligands that bind ER
structures of the liganded-ERa LBD indicate that the but exhibit selective tissue dependent activity [17]. This
binding of antagonist results in ER helix 12 shifting to a observation indicates that this ER/AP-1 pathway func-
conformation that interferes with binding of co-activa- tions in vivo and also illustrates the requirement for ERa
tors by allowing an LXXLL-like sequence in helix 12 to for this ERE-independent regulation.
bind the co-activator binding pocket of the LBD [15]. Estrogen regulation is also seen in genes such as c-
Conversely, the binding of agonist repositions helix 12, FOS and TGFa, which lack a full ERE sequence. This
exposing the co-activator binding site of the LBD. ER regulation is mediated by an interaction between ERa
dimers bind ERE sequences in target genes, and if and SP1 proteins, which bind ERE-half sites and GC rich
agonist and co-activator are associated, induce transcrip- sequences, respectively, in the regulatory regions of these
tion by interacting with and activating necessary genes [18].
components of the transcriptional apparatus. Another alternative mechanism of ER action involves
ligand-independent ER activation, in which growth
factor receptor signaling results in ER target gene
Alternative Mechanisms of ER Activity induction. Both in vivo and in vitro studies have indicated
that ER mediated responses can be induced or increased
In addition to the classical mechanism of ER function, by EGF and IGF-1 [1924]. Thus, cross-talk and signal
several alternative mechanisms have been elucidated amplication between membrane-bound growth factor
Estrogen Receptors 195

Table 1. The Phenotypes reported in mice with disrupted ERa, ERb, or both ERa and ERb

Tissue a b ab

Mammary Immature-ductal rudiment Normal structure and lactation Immature-ductal rudiment


Fertility Both sexes are infertile Fertile males Both sexes are infertile
Subfertile females: Infrequent
pregnancies, small litter sizes
Pituitary Elevated LH production, low prolactin Normal Elevated LH production
Ovary E and T elevated Appears Normal, inefcient ovulation Progressive degeneration of
Follicles don't mature, hemorrhagic cystic in superovulation trial, ``trapped germ cells, transdifferentiation
follicles begin developing at puberty as a follicle'' phenotype at superovutation of granualosa cells into
result of chronic elevated LH. Sertoli cells
LH and FSH receptors expressed
Reduced ovulations in superovulation trial,
``trapped follicle'' phenotype after
superovulation
Uterus: E resposiveness Immature. Insensitive to E-no mitosis or Normal responses to E Insensitive to E-like aERKO
induction of E responsive genes
Uterus: Progesterone PR present, P responsive genes induced, nd nd
responsiveness decidualization is E independent
Uterus: Implantation No implantation Normal; pups carried to term nd
Testes Progressive uid retention and dilation of Normal Progressive uid retention
seminiferous tubules, eventual loss and dilation of seminiferous
of sperm tubules, eventual loss of sperm
Mating behaviors Disrupted mating behavior Normal mating behavior Disrupted mating behavior
Cardiovascular responses E protection retained in vascular injury study E protection retained in vascular E protection lost in vascular
injury study injury study
Bone Both sexes shorter. Female:smaller diameter Increased density in females, Shorter
male: lower density no effect in males

receptor pathways and nuclear receptors has been the bERKO males are fertile and the bERKO females are
proposed (reviewed in Yee and Lee [25] and Cenni and subfertile [29,30].
Picard [26]). In support of this hypothesis, the estrogen
receptor a knockout (aERKO) mouse, which lacks ERa,
has been used to show that this receptor is essential for Role of ER in Female Fertility
EGF-induced DNA synthesis in uterine epithelium [27].
Further complicating these alternative modes of ER The most signicant phenotype in the aERKO and
activity, EGF and IGF-1 can stimulate a reporter gene bERKO females is complete infertility in the aERKO and
driven by GC rich promoter element from the cathepsin decreased fertility of the bERKO as a result of failure of
D gene [18]. several components necessary for successful reproduc-
tion. Normally, gonadotropin production by the pituitary
is regulated by a negative feedback loop in which rising
Animal Models for the Study of ER E levels down-regulate transcription of the LHb gene,
and thus, withdrawal of E by ovariectomy causes an
The structure and mechanisms of ER-mediated regula- increase in serum LH. The aERKO female also has
tion of various target genes have been extensively elevated LH transcript in the pituitary as well as elevated
characterized using isolated components or in vitro cell serum LH [31]. In contrast, LH levels are normal in the
models. The ability to produce transgenic mice with bERKO (unpublished), indicating that the negative
disruption of the ER genes has allowed study of the feedback is mediated by ERa rather than ERb.
physiological processes that require ERs in live animals Immunohistochemical analysis of the expression of
and provided insight into differing physiological roles of ERa and ERb protein in a normal ovary indicate that,
ERa and ERb. Table 1 summarizes many of the although both ERs are present, their distribution differs,
phenotypes reported in mice with disrupted ERa with ERb predominantly in the granulosa cells of the
(aERKO), ERb (bERKO) or both ERa and ERb follicles and ERa localized in the thecal and interstitial
(abERKO) (reviewed in Couse and Korach [28]). The regions of the ovary (Fig. 3). Therefore, one might expect
most signicant effects, as might be expected, are seen in the a or b ERKO mouse ovaries to exhibit different
reproductive systems of these animals. Both male and phenotypes. The aERKO ovaries develop hemorrhagic
female aERKO and abERKO mice are infertile, whereas cysts [29], and lack mature follicles and corpora lutea,
196 Hewitt and Korach

ance (Fig. 4), yet a similar result was obtained in a


superovulation trial in the bERKO (6 oocytes/trial
bERKO vs. 33.7 oocytes/trial WT), indicating a role
for ERb in ovulation as well. Analysis of the structures in
the ovary following the superovulation trial indicated
that although fewer ovulations occurred in the aERKO
and bERKO animals, several pre-ovulatory follicles were
present (Fig. 5). This is similar to the ``trapped follicle''
phenotype that has been reported in several other mouse
models including RIP140 (an estrogen receptor inter-
acting co-represser) knockout [35], progesterone
receptor knockout [36,37] and the Cox2 (cycloxygenase
2) knockout [38]. Further studies, however, have not yet
revealed why these follicles fail to rupture in the aERKO
and bERKO. It is clear that one cause of the infertility in
the aERKO female is dysregulation of LH. The de-
creased response of the follicles in both the aERKO and
bERKO animals even when gonadotropins are provided
exogenously may underlie their ovulatory defects.

Role of ER in the Uterus

The rodent uterus is essential for the implantation and


support of developing embryos during pregnancy. In
addition, the initiation and maintenance of the pregnancy
is dependant on ovarian hormones. The pre-ovulatory
peak of E is important in proliferation of the uterine
epithelium in preparation for implantation, while rising
progesterone (P) levels following ovulation are impor-
tant for implantation of the embryo and the formation of
decidual tissue in the stroma. The uterus of the aERKO is
Fig. 3. Localization of ERa and ERb expression in the ovary.
hypoplastic, similar in appearance to be a pre-pubertal
Immunohistochemical staining for ERa or ERb in a normal ovary
shows the differential expression pattern of these ERs. uterus (Fig. 4), and does not respond to E in terms of
weight increase, epithelial proliferation, or induction of
indicating the absence of ovulation (Fig. 4). The ovaries E responsive genes [29,32]. Furthermore, the aERKO
also produce elevated levels of E in the serum [29,32]. does not support the implantation of donor embryos
This is similar to the ovaries observed in LH over- (SCH, unpublished). All of these responses are normal in
expressing transgenic mice [33], indicating that perhaps the bERKO, and the bERKO can carry a pregnancy to
the chronically elevated LH level in the aERKO leads to term [30]. Thus, the dysfunction of the ovary is not the
the ovarian phenotype. In support of this hypothesis, only component that causes the aERKO infertility, while
treatment of aERKO females with GnRH antagonist the inefcient ovarian function in the bERKO appears to
decreases the serum LH level and also prevents the account for the subfertility. Interestingly, deciduomas
ovaries from developing hemorrhagic cysts [34]. can be induced experimentally in the aERKO uteri,
Attempts to superovulate the aERKO using exogenous indicating that this aspect of uterine function does not
gonadotropins prior to the development of hemorrhagic require ERa [39]. Overall, it is apparent that ERa is the
cysts resulted in ovulations, but with a signicantly lower ER subtype required for uterine reproductive function.
efciency than in WT mice (14.5 oocytes/trial aERKO Although the phenotypes in the aERKO uterus and
vs. 40 oocytes/trial WT) [34], although LH and FSH ovary are severe enough to account for the infertility, it is
receptors are expressed in the aERKO ovaries. Oocytes important to note that lack of ERa also leads to severe
from both WT and aERKO animals were fertilized in disruption in mating behaviors, while the bERKO
vitro with equal efciency, indicating that ERa has a role exhibits normal mating behaviors [40,41]. This indicates
in ovulation but is not required for proper oocyte that ERa, but not ERb, has a requisite role in regulation
development. The bERKO ovary has a normal appear- of female mating behavior.
Estrogen Receptors 197

Fig. 4. Ovarian histology, reproductive tracts and mammary gland whole mounts from WT, aERKO, bERKO and abERKO tissues. Arrows indicate
rudimentary ductal tree in mammay glands.

Role of ER in Mammary Glands


At birth the mouse mammary gland consists of a
rudimental ductal tree, which, as the female mouse
matures, elongates in response to E and branches in
response to progesterone (P) to eventually ll the stroma.
In the aERKO, the ductal rudiment fails to elongate [42],
while in the bERKO the gland develops normally (Fig. 4).
Moreover, the bERKO mother successfully nurses her
young, indicating normal lactation function as well. These
observations indicate ERa is required for normal
mammary gland maturation and development. Interest-
ingly, the rudimentary ductal tree in the aERKO can be
stimulated with P to develop lobular alveolar structures,
indicating that the ducts retain their ability to respond to P.

Role of ER in Male Fertility

The infertility of the aERKO males indicated for the rst


Fig. 5. aERKO and bERKO ovaries following superovulation.
time that E has an essential role in male reproduction.
``Trapped'' follicles are indicated by arrows. Note the corpus lutea Evaluation of the testis histopathology indicated dilation
(CL) in the bERKO section. of the seminiferous tubules and lack of sperm in mature
198 Hewitt and Korach

animals [43]. When germ cells from aERKO males were follicles, containing an oocyte, a normal complement of
transplanted into testis of germ-cell depleted WT recipi- granulosa cells, and surrounding theca. However, there is
ents, normal offspring were produced by the recipients also a marked presence of abnormal follicles that, when
[44,45]. This experimental observation combined with the viewed in a 2-dimensional section, appear similar to
ability of ERa-disrupted sperm from ERa heterozygous seminiferous tubules. These follicles most often lack an
(ERa + ) males to produce offspring indicate that ERa oocyte and granulosa cells, but rather possess Sertoli-like
is not needed for sperm function, but is required in the cells, located along the basement membrane and
male reproductive tract to allow maturation of sperm. exhibiting the characteristic cytosolic extensions and
Although the lack of sperm alone results in male tripartite nucleoli. Further analysis indicates the presence
infertility, it should be noted that the aERKO and of what may be intermediate structures, i.e., follicles
abERKO males also exhibit decient mating behaviors containing a degenerating oocyte, surrounded by both
[46,47], while the bERKO mating behaviors are normal granulosa cells and Sertoli-like cells. Because this
[41], indicating the importance of ERa in normal male phenotype emerges progressively with age, it is currently
mating behaviors. thought that these tubular structures represent ``ghosts''
of follicles in which the germ cell has died and a
population of remaining granulosa cells is ``trans-
Phenotypes in the abERKO differentiated'' into Sertoli-like cells.

The phenotypes exhibited by the abERKO are similar to


those of the aERKO (Table 1) [48], emphasizing the Phenotypes in Non-Reproductive Tissues
importance of ERa in both male and female reproduction.
One exception is the phenotype observed in the ovary Epidemiological data showing increased risks for osteo-
(Figs. 4 and 6) [48]. The abERKO ovary exhibits normal porosis and cardiovascular disease in postmenopausal

Fig. 6. Details of follicles from abERKO ovary. All three representative follicles are from the same ovary. The top panel shows a normal
developing follicle, with higher power detail to right showing granulosa cells. The bottom panel shows a seminiferous tubule-like structure with
higher power detail to right. Note the Sertoli-like cells along the basement membrane. The middle panel shows an intermediate follicle with
characteristics of both.
Estrogen Receptors 199

women indicates a possible role for E in bone and cardio- 4. Glass CK. Differential recognition of target genes by nuclear
receptor monomers, dimers, and heterodimers. Endocr Rev
vascular tissue. Therefore the phenotypes of the ERKO
1994;15:391407.
mice in these tissues have been studied. Analysis of bone 5. Klinge CM. Estrogen receptor interaction with estrogen response
tissues has shown aERKO femurs are shorter [49] as are elements. Nucleic Acids Res 2001;29:29052919.
male aERKO and abERKO but not bERKO femurs [50]. 6. Pike ACW, Brzozowski AM, Hubbard RE. A structural biologist's
The shorter femurs are associated with lower serum IGF- view of the oestrogen receptor. J Steroid Biochem Mol Biol
2000;74:261268.
1 levels. aERKO femurs are also of smaller diameter
7. Metzger D, Ali S, Bornert JM, Chambon P. Characterization of the
in the female and lower density in the male [28]. An amino-terminal transcriptional activation function of the human
increase in bone mineral content and density was seen in estrogen receptor in animal and yeast cells. J Biol Chem
bERKO females, with no change in bERKO males 1995;270:95359542.
[51,52], indicating that ERb may actually exert a 8. Parker MG. Structure and function of estrogen receptors. Vitam
Horm 1995;51:267287.
negative effect on bone density in the female mouse.
9. Edwards DP. The role of coactivators and corepressors in the
Cardiovascular tissues from ERKO mice were biology and mechanism of action of steroid hormone receptors.
analyzed by studying the protective effectiveness of J Mammary Gland Biol Neoplasia 2000;5:307324.
estradiol in an aortic injury model. The protective effect 10. Parker MG. Transcriptional activation by oestrogen receptors.
of E was retained in the bERKO tissues (53). Initial Biochem Soc Symp 1998;63:4550.
11. McKenna NJ, Lanz RB, O'Malley BW. Nuclear receptor
studies indicated the protective effect was also present coregulators: Cellular and molecular biology. Endocr Rev
aERKO tissue (54). However, studies utilizing an ERa 1999;20:321344.
decient mouse derived in another laboratory 12. Lanz RB, McKenna NJ, Onate SA, Albrecht U, Wong J, Tsai SY,
(ERaKOSt) indicated a lack of estrogen protection. In Tsai MJ, O'Malley BW. A steroid receptor coactivator, sra,
addition, the abERKO tissues lacked estrogen protection functions as an rna and is present in an src-1 complex. Cell
1999;97:1727.
as well (55). The initial nding of retained estrogen 13. Endoh H, Maruyama K, Masuhiro Y, Kobayashi Y, Goto M, Tai H,
protection in the original aERKO model is most likely Yanagisawa J, Metzger D, Hashimoto S, Kato S. Purication and
due to the expression of an ERa splice variant resulting identication of p68 rna helicase acting as a transcriptional
in residual truncated ERa (32) combined with the super- coactivator specic for the activation function 1 of human estrogen
physiological dose of estradiol required in the study (55). receptor alpha. Mol Cell Biol 1999;19:53635372.
14. Watanabe M, Yanagisawa J, Kitagawa H, Takeyama K, Ogawa S,
Overall, the studies indicate that ERa mediates the Arao Y, Suzawa M, Kobayashi Y, Yano T, Yoshikawa H, Masuhiro
estrogen protective response observed in this model. Y, Kato S. A subfamily of rna-binding dead-box proteins acts as an
estrogen receptor alpha coactivator through the n-terminal
activation domain (af-1) with an rna coactivator, sra. EMBO J
2001;20:13411352.
Summary 15. Chen JD. Steroid/nuclear Receptor Coactivators. 2000:391448.
16. Kushner PJ, Agard DA, Greene GL, Scanlan TS, Shiau AK, Uht
This brief overview has described the well-established RM, Webb P. Estrogen receptor pathways to ap-1. J Steroid
Biochem Mol Biol 2000;74:311317.
mechanisms and physiological functions of ER as well as
17. Klotz DM, Hewitt SC, Korach KS, Diaugustine RP. Activation of a
the frontiers of study that continue to illustrate new uterine insulin-like growth factor i signaling pathway by clinical
details of the mechanisms of action and roles of ER as an and environmental estrogens: Requirement of estrogen receptor-
important transcriptional modulator. Future investigation alpha. Endocrinology 2000;141:34303439.
promises continuing advances that are eagerly awaited 18. Safe S. Transcriptional Activation of Genes by 17 Beta-Estradiol
Through Estrogen Receptor-sp1 Interactions. 2001:231252.
and will have many important applications in diagnostic
19. Nelson KG, Takahashi T, Bossert NL, Walmer DK, McLachlan JA.
and therapeutic aspects of reproduction and cancers of Epidermal growth factor replaces estrogen in the stimulation of
reproductive tissues. female genital-tract growth and differentiation. Proc Natl Acad Sci
USA 1991;88:2125.
20. Ignar Trowbridge DM, Nelson KG, Bidwell MC, Curtis SW,
Washburn TF, McLachlan JA, Korach KS. Coupling of dual
References signaling pathways: Epidermal growth factor action involves the
estrogen receptor. Proc Natl Acad Sci USA 1992;89:46584662.
1. Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G, 21. Ignar Trowbridge DM, Teng CT, Ross KA, Parker MG, Korach KS,
Umesono K, Blumberg B, Kastner P, Mark M, Chambon P. The McLachlan JA. Peptide growth factors elicit estrogen receptor-
nuclear receptor superfamily: The second decade. Cell dependent transcriptional activation of an estrogen-responsive
1995;83:835839. element. Mol Endocrinol 1993;7:992998.
2. Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA. 22. Ignar-Trowbridge DM, Pimentel M, Teng CT, Korach KS,
Cloning of a novel receptor expressed in rat prostate and ovary. McLachlan JA. Cross talk between peptide growth factor and
Proc Natl Acad Sci USA 1996;93:59255930. estrogen receptor signaling systems. Environ Health Perspect
3. Tsai MJ, O'Malley BW. Molecular mechanisms of action of 1995;103 (Suppl 7):3538.
steroid/thyroid receptor superfamily members. Ann Rev Biochem 23. Ignar-Trowbridge DM, Pimentel M, Parker MG, McLachlan JA,
1994;63:451486. Korach KS. Peptide growth factor cross-talk with the estrogen
200 Hewitt and Korach

receptor requires the a/b domain and occurs independently of related behaviors in female mice. Endocrinology 1998;139:5070
protein kinase c or estradiol. Endocrinology 1996;137:17351744. 5081.
24. Stoica A, Saceda M, Fakhro A, Joyner M, Martin MB. Role of 41. Ogawa S, Chan J, Chester AE, Gustafsson JA, Korach KS, Pfaff
insulin-like growth factor-i in regulating estrogen receptor-alpha DW. Survival of reproductive behaviors in estrogen receptor beta
gene expression. J Cell Biochem 2000;76:605614. gene-decient (beta erko) male and female mice. Proc Natl Acad
25. Yee D, Lee AV. Crosstalk between the insulin-like growth factors Sci USA 1999;96:1288712892.
and estrogens in breast cancer. J Mammary Gland Biol Neoplasia 42. Bocchinfuso WP, Lindzey JK, Hewitt SC, Clark JA, Myers PH,
2000;5:107115. Cooper R, Korach KS. Induction of mammary gland development
26. Cenni B, Picard D. Ligand-independent activation of steroid in estrogen receptor-alpha knockout mice. Endocrinology
receptors: New roles for old players. Trends Endocrinol Metab 2000;141:29822994.
1999;10:4146. 43. Eddy EM, Washburn TF, Bunch DO, Goulding EH, Gladen BC,
27. Curtis SW, Washburn T, Sewall C, DiAugustine R, Lindzey J, Lubahn DB, Korach KS. Targeted disruption of the estrogen
Couse JF, Korach KS. Physiological coupling of growth factor and receptor gene in male mice causes alteration of spermatogenesis
steroid receptor signaling pathways: Estrogen receptor knockout and infertility. Endocrinology 1996;137:47964805.
mice lack estrogen-like response to epidermal growth factor. Proc 44. Mahato D, Goulding EH, Korach KS, Eddy EM. Spermatogenic
Natl Acad Sci USA 1996;93:1262612630. cells do not require estrogen receptor alpha for development or
28. Couse JF, Korach KS. Estrogen receptor null mice: What have we function. Endocrinology 2000;141:12731276.
learned and where will they lead us? Endocr Rev 1999;20:358417. 45. Mahato D, Goulding EH, Korach KS, Eddy EM. Estrogen receptor-
29. Lubahn DB, Moyer JS, Golding TS, Couse JF, Korach KS, Smithies alpha is required by the supporting somatic cells for spermatogen-
O. Alteration of reproductive function but not prenatal sexual esis. Mol Cell Endocrinol 2001;178:5763.
development after insertional disruption of the mouse estrogen 46. Ogawa S, Lubahn DB, Korach KS, Pfaff DW. Behavioral effects of
receptor gene. Proc Natl Acad Sci USA 1993;90:1116211166. estrogen receptor gene disruption in male mice. Proc Natl Acad Sci
30. Krege JH, Hodgin JB, Couse JF, Enmark E, Warner M, Mahler JF, USA 1997;94:14761481.
Sar M, Korach KS, Gustafsson JA, Smithies O. Generation and 47. Ogawa S, Chester AE, Hewitt SC, Walker VR, Gustafsson JA,
reproductive phenotypes of mice lacking estrogen receptor beta. Smithies O, Korach KS, Pfaff DW. Abolition of male sexual
Proc Natl Acad Sci USA 1998;95:1567715682. behaviors in mice lacking estrogen receptors alpha and beta
31. Scully KM, Gleiberman AS, Lindzey J, Lubahn DB, Korach KS, (alpha beta erko). Proc Natl Acad Sci USA 2000;97:1473714741.
Rosenfeld MG. Role of estrogen receptor-alpha in the anterior 48. Couse JF, Hewitt SC, Bunch DO, Sar M, Walker VR, Davis BJ,
pituitary gland. Mol Endocrinol 1997;11:674681. Korach KS. Postnatal sex reversal of the ovaries in mice lacking
32. Couse JF, Curtis SW, Washburn TF, Lindzey J, Golding TS, Lubahn estrogen receptors alpha and beta. Science 1999;286:23282331.
DB, Smithies O, Korach KS. Analysis of transcription and estrogen 49. Vidal O, Lindberg M, Savendahl L, Lubahn DB, Ritzen EM,
insensitivity in the female mouse after targeted disruption of the Gustafsson JA, Ohlsson C. Disproportional body growth in female
estrogen receptor gene. Mol Endocrinol 1995;9:14411454. estrogen receptor-alpha-inactivated mice. Biochem Biophys Res
33. Risma KA, Clay CM, Nett TM, Wagner T, Yun J, Nilson JH. Commun 1999;265:569571.
Targeted overexpression of luteinizing hormone in transgenic mice 50. Vidal O, Lindberg MK, Hollberg K, Baylink DJ, Andersson G,
leads to infertility, polycystic ovaries, and ovarian tumors. Proc Lubahn DB, Mohan S, Gustafsson JA, Ohlsson C. Estrogen
Natl Acad Sci USA 1995;92:13221326. receptor specicity in the regulation of skeletal growth and
34. Couse JF, Bunch DO, Lindzey J, Schomberg DW, Korach KS. maturation in male mice. Proc Natl Acad Sci USA
Prevention of the polycystic ovarian phenotype and characteriza- 2000;97:54745479.
tion of ovulatory capacity in the estrogen receptor-alpha knockout 51. Windahl SH, Vidal O, Andersson G, Gustafsson JA, Ohlsson C.
mouse. Endocrinology 1999;140:58555865. Increased cortical bone mineral content but exchanged trabecular
35. White R, Leonardsson G, Rosewell I, Jacobs MA, Milligan S, bone mineral density in female er beta( / ) mice. J Clin Invest
Parker M. The nuclear receptor co-repressor nrip1 (rip140) is 1999;104:895901.
essential for female fertility. Nat Med 2000;6:13681374. 52. Windahl SH, Hollberg K, Vidal O, Gustafsson JA, Ohlsson C,
36. Lydon JP, DeMayo FJ, Funk CR, Mani SK, Hughes AR, Andersson G. Female estrogen receptor beta / mice are
Montgomery CA, Shyamala G, Conneely OM, O'Malley BW. partially protected against age-related trabecular bone loss.
Mice lacking progesterone receptor exhibit pleiotropic reproduc- J Bone Miner Res 2001;16:13881398.
tive abnormalities. Genes Dev 1995;9:22662278. 53. Karas RH, Hodgin JB, Kwoun M, Krege JH, Aronovitz M, Mackey
37. Lydon JP, DeMayo FJ, Conneely OM, O'Malley BW. Reproductive W, Gustafsson JA, Korach KS, Smithies O, Mendelsohn ME.
phenotypes of the progesterone receptor null mutant mouse. Estrogen inhibits the vascular injury response in estrogen receptor
J Steroid Biochem Mol Biol 1996;56:6777. beta-decient female mice. Proc Natl Acad Sci USA
38. Lim H, Paria BC, Das SK, Dinchuk JE, Langenbach R, Trzaskos 1999;96:1513315136.
JM, Dey SK. Multiple reproductive failures in cyclooxygenase 2- 54. Iafrati MD, Karas RH, Aronovitz M, Kim S, Sullivan TR, Jr.,
decient mice. Cell 1997;91:197208. Lubahn DB, O'Donnell TF, Jr., Korach KS, Mendelsohn ME.
39. Curtis SW, Clark J, Myers P, Korach KS. Disruption of estrogen Estrogen inhibits the vascular injury response in estrogen receptor
signaling does not prevent progesterone action in the estrogen alpha-decient mice. Nat Med 1997;3:545548.
receptor or knockout mouse uterus. Proc Natl Acad Sci USA 55. Paras G, Krust A, Karas RH, Dupont S, Aronovitz M, Chambon P,
1999;96:36463651. Mendelsohn ME. Estrogen receptor-alpha mediates the pro-
40. Ogawa S, Eng V, Taylor J, Lubahn DB, Korach KS, Pfaff DW. tective effects of estrogen against vascular injury. Circ Res
Roles of estrogen receptor-alpha gene expression in reproduction- 2002;90(10):10871092.

S-ar putea să vă placă și