Download Print

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

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

Document related concepts

Growth hormone therapy wikipedia , lookup

Hormone replacement therapy (menopause) wikipedia , lookup

Hypothalamus wikipedia , lookup

Hormone replacement therapy (male-to-female) wikipedia , lookup

Hyperthyroidism wikipedia , lookup

Graves' disease wikipedia , lookup

Transcript
Physiol Rev
82: 923–944, 2002; 10.1152/physrev.00014.2002.
Estrogen and Thyroid Hormone Receptor Interactions:
Physiological Flexibility by Molecular Specificity
NANDINI VASUDEVAN, SONOKO OGAWA, AND DONALD PFAFF
Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, New York
923
924
924
924
926
930
930
931
933
933
935
936
936
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
I. Introduction
II. Hormonal Induction of Genes in the Central Nervous System and in Cell Lines
A. Estrogen induction of genes in the brain
B. Isoforms from genes for ER and TR: distinct and overlapping functions
C. Molecular interactions between the ER and TR isoforms: cell culture studies
III. Physiological Data and Their Implications
A. Lordosis behavior
B. Differences in isoforms from nuclear receptor genes: use of knock-out models
C. Patterns of behavior
D. Physiological implications of thyroid hormone modulation of estrogen action
IV. Role of Promoter and Cell Specificity in Distinct Transcriptional Results
V. Questions Unanswered
A. Gene duplication and splice variants
B. Rapid versus slow effects of estrogen: two separate opportunities for thyroid
hormone modulation?
C. Thyroid hormone elevation: does it signal cold temperatures?
VI. Summary
937
938
938
Vasudevan, Nandini, Sonoko Ogawa, and Donald Pfaff. Estrogen and Thyroid Hormone Receptor Interactions:
Physiological Flexibility by Molecular Specificity. Physiol Rev 82: 923–944, 2002; 10.1152/physrev.00014.2002.—The
influence of thyroid hormone on estrogen actions has been demonstrated both in vivo and in vitro. In transient
transfection assays, the effects of liganded thyroid hormone receptors (TR) on transcriptional facilitation by
estrogens bound to estrogen receptors (ER) display specificity according to the following: 1) ER isoform, 2) TR
isoform, 3) the promoter through which transcriptional facilitation occurs, and 4) cell type. Some of these molecular
phenomena may be related to thyroid hormone signaling of seasonal limitations upon reproduction. The various
combinations of these molecular interactions provide multiple and flexible opportunities for relations between two
major hormonal systems important for neuroendocrine feedbacks and reproductive behaviors.
I. INTRODUCTION
Cross-talk between members of the nuclear receptor
superfamily theoretically can multiply the possible modes
of gene regulation, leading to a greater and more flexible
array of transcriptional responses to environmental
changes. In the central nervous system (CNS), such gene
regulation conceivably can help to coordinate behavioral
responses of the organism to climatic and social stimuli
(217). Such cross-talk can also underlie metastatic processes. The activation of the estrogen-dependent growth
responses by a nonestrogen such as the growth factor,
insulin growth factor (IGF), may promote the growth of
various cell types. Cross-talk between IGF and estrogens,
for example, can lead to cell proliferation in breast carwww.prv.org
cinoma (41 and references therein) and hence is of considerable interest as a target for adjunct therapy.
Estrogen (ER) and thyroid hormone receptors (TR)
are members of the nuclear receptor superfamily that
bind the low-molecular-weight ligands, estrogens and thyroid hormones, respectively. They transduce these signals
into gene regulation events. These receptors have a modular protein structure with high homology in the central
DNA binding domain. They are ligand-activated transcription factors that influence transcription from target genes
(43, 105). Nuclear receptors bind enhancer elements on
DNA called hormone response elements to regulate transcription from genes (43, 105).
How do nuclear receptors regulate gene transcription? Gene activation events require the recruitment of
0031-9333/02 $15.00 Copyright © 2002 the American Physiological Society
923
924
VASUDEVAN, OGAWA, AND PFAFF
II. HORMONAL INDUCTION OF GENES
IN THE CENTRAL NERVOUS SYSTEM
AND IN CELL LINES
A. Estrogen Induction of Genes in the Brain
Estrogens are necessary for the induction of the primary female-typical sexual behavior lordosis (91) in several species. Estrogens regulate the expression of several
genes in the brain, some of which are responsible for the
facilitation of lordosis (reviewed in Ref. 145). For example, the nonapeptide oxytocin (OT) and its receptor, the
oxytocin receptor (OTR), are expressed in the uterus
Physiol Rev • VOL
during pregnancy (96) and are thought to be important for
gestation, parturition, and lactation. Mice that do not have
OT (OT knock-out mice) cannot lactate and therefore
cannot nurse pups (123). The expression of OT and OTR
in the CNS is believed to be important for the facilitation
of affiliative (24, 44) and sexual behaviors (7, 8, 22) that
are, in turn, required for optimal reproduction.
Estrogen administration to ovariectomized female
rats increases OTR mRNA in the ventromedial hypothalamus (VMH), a brain region critical for lordosis (153).
Estrogens also upregulate OTR mRNA in the medial
amygdala, hippocampus, and anterior pituitary but do not
change the concentration of OTR mRNA in the caudate
putamen or arcuate nucleus of the rat (153). Concomitantly, estrogen treatment increases oxytocin binding in
the bed nucleus of the stria terminalis, lateral ventral
septum, amygdala, and VMH (20, 59, 188). In situ hybridization studies show that OTR mRNA is highest in the rat
VMH at proestrous, showing a correlation with the estrogen surge that occurs during this part of the estrous cycle
(12). When antisense oligonucleotides are infused into the
VMH of estrogen-primed female rats, the rats show significantly higher rejection behavior and lower lordosis,
thus demonstrating the need for an intact OT-OTR system
for reproductive success (107).
In a similar manner, estrogen treatment also upregulates preproenkephalin (PPE) mRNA in the rodent VMH
(97, 168, 169). The importance of the expression of this
gene is underscored by the ability of antisense oligonucleotides against PPE in the hypothalamus to reduce lordosis behavior (121).
B. Isoforms From Genes for ER and TR:
Distinct and Overlapping Functions
1. Two ER genes: ␣ and ␤
Among vertebrates, the ER exists in two isoforms, ␣
and ␤, which are products of different genes (186). The
newly discovered ER␤ isoform was cloned from rat prostate and the ovary. The isoforms exhibit considerable
homology in the DNA binding domain and COOH-terminal
AF2 domain but high divergence in the NH2-terminal
transactivation AF-1 domain (65). Both isoforms can bind
several ligands with similar affinities (92); they also bind
the consensus the estrogen response element (ERE) with
similar affinities (31). The dissociation of ER␣ and ER␤
from such a consensus ERE is similarly affected in the
presence and absence of ligand at elevated temperature
(136). They also bind many EREs (72) derived from estrogen-regulated genes that have deviations from the consensus ERE sequence. Both receptors contain a functionally conserved AF-2 domain, which can be stimulated by
binding the steroid receptor coactivator (SRC1) (31, 186).
Despite limited homology in the NH2-terminal AF-1 do-
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
specific coactivators by ligand-bound nuclear receptors
(33, 116, 122). This leads to the remodeling of chromatin,
since many coactivators possess histone acetyltransferase (HAT) activity (27, 178). The remodeling and
“opening” up of chromatin leads to gene activation (27).
Unlike the ER, the TR can regulate transcription even in
the absence of ligand (213). Two well-characterized corepressors termed nuclear corepressor (NCoR) and SMRT
exist for the TRs and the retinoic acid receptors (RAR)␣
(71, 93). In the absence of ligands, TR and RARs recruit
corepressors, which have histone deacetylase activity and
antagonize HAT coactivators. This, in turn, represses gene
transcription (68).
Estrogens are critical in the control of reproduction
in both male and female mammals (88). The deletion of
the ER␣ isoform causes infertility in both male and female
mice (104, 128). The nonreproductive functions of estrogens include maintenance of bone mass (64 and references therein) and cardioprotective effects (170). The
central role of estrogens in mammalian reproduction is
reflected in the neuroendocrine control of gonadotrophin
production from the anterior pituitary and feedback regulation of gonadotropin releasing hormone (GnRH) in the
hypothalamus (88, 89). Recently, neuroprotective effects
as well as effects on mood and cognition have also been
described (99, 147, 204).
Unlike estrogens, which have a more focused role,
the thyroid hormones triiodothyronine (T3) and thyroxine
(T4) exhibit a large range of actions (135, 208). In the adult
homeothermic animal, they exert control over lipogenesis, lipolysis, and thermogenesis. They are critical for
growth, development, and differentiation (134, 172). In
humans, neonatal hypothyroidism results in cretinism, a
disorder characterized by mental retardation and skeletal
defects (35). The effects of thyroid hormone on the brain
are especially well illustrated by thyroid hormone control of
myelin basic protein and the consequent myelination of
axons in the brain (45). This article attempts to review the
relevant information on estrogen and thyroid hormone interactions with a perspective on neuroendocrine functions.
PATTERNS OF ER AND TR ACTIONS IN CNS
2. Differences between ER␣ and ER␤
At AP-1 sites, classical estrogens such as diethylstilbestrol and 17␤-estradiol activated transcription when
bound to ER␣ but were antiestrogens when bound to ER␤
(137). On the human RAR␣-1 promoter, ER␣ activates
transcription in response to estrogens through nonclassical ERE and not by direct DNA-receptor binding. However, in response to estrogens, ER␤ does not activate this
promoter; it activates it in response to tamoxifen, raloxifene, and ICI-164,384 (220). Therefore, the ER isoforms
show considerable promoter site specificity. In vivo and
in vitro, heterodimerization between ER␣ and ER␤ has
been shown (129, 143), and tissues that coexpress both
isoforms are thought, therefore, to respond differently to
various ER ligands compared with tissues that express
predominantly one isoform (65, 66). Therefore, there is
likely a considerable contribution of ER␤ to the pharmacology of estrogens and antiestrogens.
3. TRs: two genes and four isoforms
The four TR isoforms are protooncogene products
derived by differential splicing of two different genes:
Physiol Rev • VOL
TR␣1 and TR␣2 are from the TR␣ gene, while TR␤1 and
TR␤2 are from a separate TR␤ gene. In chicken, a shorter
TR␣1 transcript lacking the NH2-terminal A/B domain is
also present. Also, two TR␤1 transcripts, which possess
very short A/B domains, are also present in the chicken.
Xenopus laevis has several transcripts with homology to
TR␤1 but none similar to TR␤2 (98). Again, although there
is considerable homology in the central DNA binding
domain among the isoforms, significant dissimilarity exists in the NH2-terminal A/B domain. Not all TR isoforms
can bind ligand; the TR␣2 isoforms lacks the ability to
bind ligand due to a loss of 40 amino acids in the COOHterminal hormone-binding domain. The role of TR␣2 in
physiology is unclear; ex vivo studies in cell culture implicate it as a dominant negative inhibitor of TR action.
However, the potency of dominant negative action is
lower than the unliganded TR isoforms, possibly due to
deficient interactions with corepressors (182).
4. Transcriptional properties of TR isoforms
The transcriptional properties of different TR isoforms have been poorly studied, but there do exist some
differences. The differences in the TR isoforms could
allow for differential interactions with other proteins,
thereby regulating transcription. For example, unliganded
TR␤2 can bind SRC-1, unlike TR␣1 and TR␤1 (125). The
TR␤2 is a more potent mediator of ligand-independent
activation than TR␣1 or TR␤1 of T3 target genes such as
the thyroid stimulating hormone (TSH) subunit gene and
the TRH gene (70, 94, 171). This ability is independent of
NCoR and may be due to differential binding of coactivators. The TR␤2 isoform also is unable to mediate ligandindependent repression on the growth hormone promoter, unlike the TR␤1 and TR␣1 isoforms, due to lack of
NCoR binding ability (70). Zhu et al. (215) have noted an
increased ability of TR␤1 to transactivate from a F2 thyroid hormone response element (TRE) compared with
TR␣1 (215). Differential interaction with other proteins,
including other nuclear receptors, may therefore play a
role in thyroid hormone physiology.
Since the consensus DNA sequences bound by ER
and TR share a common half site, it is possible that
competition between the two receptors may lead to antagonism of the other’s effect. Indeed, this was first demonstrated for the vitellogenin ERE by Glass et al. (60); the
thyroid hormone receptor could decrease ER␣-mediated
transactivation.
Steroid hormone receptors have been shown to decrease ligand-dependent TR transactivation from a TRE
(210). Estrogens were also shown to suppress the T3
effect on the ␣-glycoprotein hormone subunit promoter
(207). In both pituitary-derived GH3 cells and JEG-3 choriocarcinoma cells, T3 mediates suppression of the ␣-glycoprotein hormone subunit promoter. 17␤-Estradiol sup-
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
main, both ER␣ and ER␤ contain a mitogen-activated
protein kinase (MAPK) phosphorylation site that results
in enhanced transcription (186). Hall and McDonnell (66)
show that despite similar binding affinities for several
ligands, activation of transcription from simple target promoters containing EREs by ER␤ is dependent on pure
agonists. On the other hand, ER␣ can activate transcription when bound to agonists and partial agonists. If a
chimeric ER␤ receptor containing the A/B domain of the
ER␣ is tested for transcriptional activation, antiestrogens
such as tamoxifen, which showed no transcriptional activation with ER␤, could now show some degree of transactivation (110). Jones et al. (76) investigated the ability
of ER␣ and ER␤ to activate transcription from a number
of different promoters that are estrogen responsive but
lack classical EREs in human breast, bone, and uterine
cell lines. These included a collagenase promoter containing an AP-1 element important in estrogen induction, a
nonconsensus ERE containing complement C3 promoter,
and a transforming growth factor (TGF)-␣ promoter containing both ERE and Sp1 elements. All antiestrogens
studied were agonistic on the collagenase reporter in the
uterine cell lines when ER␤ was transfected, but tamoxifen alone was agonistic when ER␣ was transfected (76).
Also, the ability of ER␤ to repress transactivation of
NF␬B in osteoblasts occurs only in the presence of 17␤estradiol, whereas ER␣ can repress NF␬B transactivation
in the absence or presence of ligand (152). This suggests
important mechanistic differences, possibly arising from
differences in amino acid sequence in the AF-1 domain
(66, 195).
925
926
VASUDEVAN, OGAWA, AND PFAFF
pressed this inhibition. In vitro synthesized ER␣ could
bind to the TRE present in this promoter, thereby suggesting competition between these two nuclear receptor
systems as a distinct possibility (207). However, in both
these studies, pure TR and ER isoforms were not used.
Hence, the investigation of possible differential interactions between distinct ER and TR isoforms is of interest.
C. Molecular Interactions Between the ER and TR
Isoforms: Cell Culture Studies
1. Modulation of ER␣ transcriptional activity by the
ligand-binding TR isoforms
The interaction of ER␣, the classical ER isoform, and
the ligand-binding TR isoforms has been observed on a
consensus vitellogenin ERE linked to a minimal thymidine kinase promoter in kidney fibroblast, CV-1, cells
(190, 218). CV-1 cells were chosen since they have low
endogenous ER and TR isoforms (215). On cotransfection
of ER␣ and TR␣1 expression vectors, the T3-liganded
TR␣1 isoform could interfere with the ER␣ induction of
this simple promoter (Fig. 1A) (190, 218). However, the
TR␤1 or TR␤2 isoforms did not have any effect on the
ER␣ induction of this promoter (190, 218) (Fig. 1, B and
C). This demonstrates that on a simple consensus ERE,
there is considerable difference in the modulation of transcriptional activity of ER␣ by the ligand-binding TR isoforms.
2. Interactions of ER␤ with the ligand binding TR
isoforms on the consensus ERE
ER␤ could also induce this promoter in CV-1 cells,
albeit at a lower level than the ER␣ isoform (Fig. 2). When
the ligand binding TR isoforms were cotransfected with
ER␤ in CV-1 cells, the TR isoforms showed differential
effects on ER␤-mediated induction from the consensus
Physiol Rev • VOL
3. ER␣ versus ER␤: TR␣1 modulation of induction
of the PPE promoter in CV-1 cells
Similar to the inhibitory effect by the TR␣1 on ER␣mediated transcription from the consensus ERE, the
TR␣1 inhibited ER␣ induction from the PPE promoter
(Fig. 3A). In contrast, the TR␣1 isoform stimulated ER␤mediated transcription (Fig. 3B), demonstrating differences in the interaction of a given TR isoform with an ER
isoform. In the CV-1 cell line, the TR␤ isoforms had no
effect on ER␣ or ER␤-mediated induction of the PPE
promoter (192). Again, similar to the consensus ERE, the
ability of the TR␣1 isoform to inhibit or stimulate ERmediated transcription of the PPE depends on the ER
isoform present in the cell (192).
4. TR isoform modulation of ER␣-mediated induction
of the OTR promoter
To test the interactions between the TR and ER
isoforms in a cell line with neuronal properties and to
compare these interactions with those occurring in a
nonneuronal cell line, the OTR promoter containing the
distal ERE was investigated in both CV-1 and SK-N-BE2C
cell lines (189). Again, the TR␣1 isoform was capable of
inhibiting the ER␣-mediated induction from the OTR promoter in both CV-1 and SK-N-BE2C cell lines. However, a
complex pattern of interactions emerged when the TR␤
isoforms were expressed in conjunction with the ER␣
isoform. Although the TR␤2 isoforms were inhibitory to
ER␣-mediated induction of this OTR promoter in either
cell line, the TR␤1 effect on ER␣ induction depends on
the cell line. In the CV-1 cell line, this isoform stimulated
the ER␣ induction while inhibiting it in the neuronal cell
line (189). A neuronal specific cofactor, NIX1, which can
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
Two different promoters were initially used to examine the interactions between the ER and the TR. The
consensus ERE derived from the vitellogenin gene promoters has long been used as a model system to explain
molecular mechanisms by which estrogen regulates
genes. In CV-1 kidney fibroblast cells, the consensus ERE
linked to a CAT reporter has been shown previously to be
transcriptionally upregulated by estrogen-liganded ER␣
(218). Transiently transfected TR␣1 was able to inhibit
this ER␣-mediated induction, but TR␤1 and TR␤2 had no
effect (218). Contrasting to another promoter, when three
tandem copies of the estrogen response EREs from the
progesterone receptor promoter are used in the CV-1 cell
line, no TR isoform could inhibit the ER␣-mediated induction (173). This suggested that the interactions between
TR and ER isoform were different on different promoters.
ERE. In contrast to the inhibitory effect of the TR␣1
isoform on the consensus ERE, the TR␣1 isoform stimulated ER␤-mediated transcription (Fig. 2A). The TR␤1
isoform also stimulated ER␤-mediated transcription (Fig.
2B), while the TR␤2 isoform inhibited ER␤-mediated transcription (Fig. 2C). However, neither TR␤1 nor TR␤2 had
any effect on ER␣-mediated transcription. This shows
that a single TR isoform can lead to differential transcriptional outcomes depending on the ER isoform present in
the cell.
Are the interactions between the various TR and ER
isoforms different on a physiologically relevant promoter? To address this question, the estrogen-responsive,
behaviorally relevant PPE and OTR promoters cloned
upstream of reporter genes were transfected into CV-1
cell lines and neuronal (SK-N-BE2C) cell lines. The PPE
promoter has two EREs located within 450 bp of the
transcription start site (77), whereas the OTR promoter
has a distal ERE located ⬃4 kb from the transcription
start site (11).
PATTERNS OF ER AND TR ACTIONS IN CNS
927
bind liganded TR␤1 and downregulate transcription,
could be responsible for this phenomenon (63). The expression of the cofactor appears to be confined to dentate
gyrus, the amygdala, as well as thalamic and hypothalamic regions and may contribute to the differences in
transcriptional activation observed with the TR␤1 isoform in these cell lines. Therefore, cell-specific effects are
also important in the interactions between nuclear receptor isoforms. These data underscore the need to test
physiological promoters in different cell lines.
Table 1 summarizes the transcriptional pattern obtained using different promoters and various combinations of ER and TR isoforms. The different outcomes
show that the interaction between the ER and TR isoforms demonstrates considerable promoter specificity. It
is easy to visualize that different levels of TR and ER
Physiol Rev • VOL
isoforms in cells may, therefore, allow for flexible regulation of EREs depending on stimuli.
5. Mechanisms of TR␣1-mediated inhibition
A) COMPETITIVE DNA BINDING. What are the possible
mechanisms by which the TR isoforms interact with the
ER isoforms? Since the consensus DNA sequences, the
hormone response element (HRE), bound by both ER and
TR, are very similar, TR can interfere with ER-mediated
transcription by competing with the ER for binding to the
ERE on DNA (60). A TR␣1 mutant called the TR P-box
mutant has a mutation in the DNA binding region; this
disallows binding by this isoform to DNA (209). If DNA
binding were important in the inhibitory interaction between TR␣1 and the ER␣ isoform, then such a mutant
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
FIG. 1. Effect of the ligand binding thyroid hormone receptor (TR) isoforms on estrogen receptor (ER)␣-mediated
induction of the consensus estrogen response element (ERE) in CV-1 cells. CV-1, a kidney fibroblast cell line, was grown
in DMEM supplemented with 10% fetal bovine serum. The reporter plasmid is the ERE-tk-CAT and has a single consensus
vitellogenin ERE upstream of a minimal thymidine kinase promoter linked to chloramphenicol acetyltransferase (CAT)
(218). The cells seeded in 6-well plates (Falcon) were transfected at 60% confluency. The expression plasmids used were
the pSG-hER␣ coding for the ER␣ protein and the pCDNAI-rTR␣1, -␤1, and -␤2 coding for the TR isoforms (218). The
pSV-␤-galactosidase (pSV␤gal) plasmid coding for the enzyme ␤-galactosidase was used as a normalization control for
transfection efficiency and lysate preparation. The reporter plasmid (200 ng), the expression plasmids for the nuclear
receptors (80 ng), and the pSV␤gal (80 ng) plasmids were cotransfected into CV-1 cells using the Effectene reagent
(Qiagen) according to the manufacturer’s instructions. Twenty-four hours after transfections, phenol red-free media
supplemented with hormone-free sera were added to the cells. Either 17␤ -estradiol (E) (10⫺7 M) or triiodothyronine (T)
(10⫺6 M) or both (E⫹T) were added to wells. A set of wells received the vehicle, ethanol, alone. Forty-eight hours after
hormone treatment, cells were lysed using Reporter lysis buffer (Promega) according to the manufacturer’s instructions,
and CAT and ␤-gal assays were performed on every sample. The CAT activity was normalized to the ␤-gal activity for
every sample. Results (fold over vehicle control) represent means ⫾ SE (n ⫽ 5/treatment group). Statistical comparisons
between treatment groups were done using ANOVA followed by Student-Newman-Keuls post hoc tests. A (TR␣1):
*P ⬍ 0.001 compared with the vehicle-treated group. #P ⬍ 0.001 compared with the estrogen-treated group. B (TR␤1):
*P ⬍ 0.05 compared with the vehicle-treated group. C (TR␤2): *P ⬍ 0.001 compared with the vehicle-treated group.
[Modified from Vasudevan et al. (190).]
928
VASUDEVAN, OGAWA, AND PFAFF
should not be able to inhibit the ER␣ induction. With the
consensus ERE in CV-1 cells, inhibition by the TR␣1
isoform was lost when the TR P-box mutant is used (190,
218), implicating DNA binding and hence competition for
the ERE as important in this inhibition. In addition, the
TR␤1 and TR␣1 isoforms can bind to the consensus ERE,
thus making inhibition by competitive DNA binding possible (218). However, differences arise when “physiologi-
cal” promoters such as the OTR and PPE promoters are
used. Despite lack of DNA binding ability, the TR P-box
mutant could nonetheless inhibit the ER␣-mediated induction of both PPE (Fig. 4A) and OTR promoters, suggesting that DNA competition may not be a universal
mechanism in inhibition. Also, to check if the levels of ER
isoforms expressed in the CV-1 cell lines significantly
differ, binding of [3H]estradiol to extracts of cells trans-
FIG. 3. TR␣1 has opposing effects on ER␣-mediated (A) and ER␤-mediated (B) induction from the rat preproenkephalin (PPE) promoter in CV-1 cells. The protocol for transfection and the expression and normalization plasmids
used have been described in the legends to Figures 1 and 2 and in previous studies (137, 218). The rat PPE gene promoter
(⫺437 to ⫹53 bp) containing two putative EREs was inserted into pUTKAT1 (77, 218). The CAT activity was normalized
to the ␤-gal activity for every sample. Results (fold over vehicle control) represent means ⫾ SE. Statistical comparisons
between treatment groups were done using ANOVA followed by Student-Newman-Keuls post hoc tests. A (n ⫽ 8/
treatment group): *P ⬍ 0.001 compared with the vehicle-treated group. #P ⬍ 0.01 compared with the estrogen-treated
group but ⬎0.05 compared with vehicle-treated group. B (n ⫽ 5/treatment group): *P ⬍ 0.05 compared with the
vehicle-treated group. #P ⬍ 0.01 compared with estrogen-treated group. [Modified from Vasudevan et al. (192).]
Physiol Rev • VOL
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
FIG. 2. Effect of the ligand binding TR
isoforms on the ER␤-mediated induction
from the consensus ERE in CV-1 cells.
The protocol for transfection as well as
the reporter and expression plasmids for
the TR isoforms has been detailed in the
legend to Figure 1. The ER␤ expression
plasmid used was the pCMV-ER␤ (137).
The CAT activity was normalized to the
␤-gal activity for every sample. Results
(fold over vehicle control) represent
means ⫾ SE (n ⫽ 8 or 9/treatment group).
Statistical comparisons between treatment groups were done using ANOVA followed by Student-Newman-Keuls post
hoc tests. A (TR␣1): *P ⬍ 0.01 compared
with the vehicle-treated group. #P ⬍ 0.001
compared with the vehicle-treated group.
⵩ P ⬍ 0.001 compared with the estrogentreated group. B (TR␤1): *P ⬍ 0.001 compared with the vehicle treated group.
#P ⬍ 0.001 compared with the estrogentreated group. C (TR␤2): *P ⬍ 0.01 compared with the vehicle-treated group.
#P ⬍ 0.001 compared with the vehicletreated group. ⵩ P ⬍ 0.01 compared with
the estrogen-treated group. [Modified
from Vasudevan et al. (190).]
929
PATTERNS OF ER AND TR ACTIONS IN CNS
TABLE 1. ER␣ compared with ER␤ in the specificities of interactions with different TR isoforms: promoter and
cell type dependence
T4/ TR Interactions
ER Driving E-Facilitated
Transcription
I
II
III
ER␣
ER␤
ER␣
ER␤
ER␣
ER␤
ER␣
ER␤
cf
of
cf
cf
TR␣1
TR␤1
TR␤2
2
1
2
No E induction
2
No E induction
2
1
⬃
⬃
1
No E induction
2
No E induction
⬃
1
⬃
⬃
2
No E induction
2
No E induction
⬃
2
Promoter
PPE
PPE
OT receptor
Cell Type
CV1
OT receptor
cf
SK-N-BE2C
ERE consensus
CV1
fected with either ER␣ or ER␤ was done. There was no
difference in the levels of ER␣ or ER␤; this also correlates
well with the similar level of transcriptional activation
promoted by either isoform in response to 17␤-estradiol.
The nuclear corepressor NCoR mediates basal repression
by unliganded TR isoforms (30). However, because the
inhibition detailed in Table 1 is ligand dependent, this
must represent a novel, NCoR-independent mechanism.
B) RESCUE OF TR␣1 INHIBITION BY THE EXPRESSION OF A
COACTIVATOR. The p160 group of coactivators, of which
FIG. 4. Mechanisms of TR-mediated interference of ER-driven transcription. A: a TR␣1 mutant unable to bind DNA,
the TR P-box mutant, can still cause inhibition of the ER␣-mediated induction from the rat PPE promoter in CV-1 cells.
B: SRC-1 overexpression rescues TR␣1 inhibition of ER␣-mediated induction from the rat OTR in CV-1 cells. A: CV-1 cells
were cotransfected with PPE reporter plasmids and the expression plasmids for ER␣ and the TR P-box mutant (209) as
detailed in the legend to Figure 1. [Modified from Vasudevan et al. (192).] B: SRC-1, a general steroid receptor
coactivator, was overexpressed in CV-1 along with the TR␣1 and ER␣ isoforms as detailed in the legend to Figure 1. The
samples that were transfected with SRC-1 expression vector (4th, 5th, and 6th bar from left) were treated with
17␤-estradiol (E) (10⫺7 M) or both 17␤-estradiol and triiodothyronine (E ⫹ T). Corresponding hormone treatment was
given to a set of samples that received the empty control SRC-1 expression plasmid (first 3 bars from left). [Modified from
Vasudevan et al. (189).] Forty-eight hours after hormone treatment, cells were lysed, and each sample was assayed for
both ␤-gal and CAT activity. The reporter gene activity was normalized to the ␤-gal activity for every sample. Results
(fold over vehicle control) represent means ⫾ SE. Statistical comparisons between treatment groups were done using
ANOVA followed by Student-Newman-Keuls post hoc tests. A (n ⫽ 8/treatment group): *P ⬍ 0.01 compared with
vehicle-treated group. #P ⬍ 0.01 compared with estrogen treatment. B (n ⫽ 5/treatment group): *P ⬍ 0.05 compared with
the vehicle-treated group. #P ⬍ 0.05 compared with the vehicle-treated ⫺SRC-1 group. ƒP ⬍ 0.001 compared with the
estrogen-treated ⫺SRC-1 group.
Physiol Rev • VOL
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
Summary of the interaction between the ligand-binding thyroid hormone receptor (TR) isoforms and the estrogen receptor (ER)␣ and ER␤
isoforms in CV-1 (kidney fibroblast) and SK-N-BE2C (neuroblastoma) cells. All effects of thyroid hormone-liganded TR isoforms are tested only if
there is estrogen (E) induction via either ER␣ or ER␤ of the promoter. The promoters used were as follows: ppe-cat, physiological rat
preproenkephalin (PPE) gene promoter (⫺437 to ⫹53) linked to the chloramphenicol acetyltransferase reporter gene (77); otr-luc, physiological rat
oxytocin (OT) receptor gene promoter linked to the luciferase reporter gene (11); eretkcat, a single copy of the consensus (ERE) derived from the
vitellogenin gene linked to the chloramphenicol acetyltransferase reporter gene (218). 1, T4 through TR isoform increases E-induced transcription;
2, T4 through TR isoform decreases E-induced transcription; ⬃, T4 through TR isoform has no effect on E-induced transcription. I, data from
Vasudevan et al. (192); II, data from Vasudevan et al. (189); III, data from Vasudevan et al. (190).
930
VASUDEVAN, OGAWA, AND PFAFF
6. PPE and OTR, “downstream genes,” providing
routes from estrogens/ERs to behavior
The PPE gene plays a role in analgesic responses
which can help the female to put up with somatosensory
stimuli during mating which otherwise would be treated
as noxious (17). Therefore, PPE gene induction represents a causal route which allows us to link a hormone’s
genomic effects with a specific behavior, lordosis. In the
rat VMH, PPE mRNA in the afternoon of proestrous was
significantly higher than diestrous (56). In the female ewe,
PPE mRNA increased in the VMH both during lactation
and with estrogen treatment (21). On a single dose of
Physiol Rev • VOL
17-estradiol-3-benzoate given to female ovariectomized
mice, PPE mRNA was upregulated in the VMH, medial
amygdala (MeAmyg), and arcuate nucleus (ARC) at 24
and 48 h (154). However, PPE is not regulated in the
caudate putamen or in the cortical amygdala by estrogens
(154). A single dose of estradiol benzoate to ovariectomized female rats resulted in a biphasic increase in PPE
mRNA producing cells both in the ARC and VMH with a
peak at 48 h (151). This biphasic response of PPE consists
of a primary peak at 1 h and a second peak between 24
and 48 h postinjection. The rapid first peak was stress
induced and could be blocked by adrenalectomy or constant low levels of corticosterone. A peak of plasma corticosterone also coincided with this peak. In the medial
amygdala, the antiestrogen tamoxifen blocked the second
peak of PPE mRNA expression. These data indicate that
both steroids and noxious mild somatosensory stimuli
interact to give increases in PPE expression (177). This is
consistent with a role for PPE in female reproductive
behavior. Acute, mild stress in the form of male approach
behavior may activate limbic and hypothalamic circuits
known to be important for the full display of reproductive
behavior (177).
The OTR gene is also critical for reproductive success. Infusion of oxytocin into the VMH increases sexual
behavior and maternal behavior. How does it do so? It is
thought that most laboratory tests for sexual behavior and
maternal behavior involve unfamiliar, novel and potentially threatening surroundings for rodents. Exposure to
such apparatuses or to an unfamiliar animal could trigger
inhibitory stress responses. In Swiss Webster mice pretreated with estrogens, peripherally administered OT increased entries into open arms in the elevated plus maze
(108). In mice given intracerebroventricular injections of
OT, entries into open arms were increased compared with
mice given arginine vasopressin (108). Therefore, estrogen upregulation of OT could be a vital component in
anxiolytic actions, decreasing stress and facilitating social
interactions (109).
Therefore, both PPE and OTR are downstream genes
with proven behavioral roles and are upregulated by a
primary reproductive effector, estrogen. These downstream gene products are expressed in behaviorally relevant neurons that possess ERs. Indeed, they can be visualized as systems that link a small hormonal signal with an
identifiable behavior.
III. PHYSIOLOGICAL DATA
AND THEIR IMPLICATIONS
A. Lordosis Behavior
Neuronal and genetic mechanisms for lordosis behavior have been worked out in such detail (reviewed in
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
SRC-1 is a member, has been shown to bind both the ER
ligand binding domain and the TR (103). Therefore, SRC-1
appeared to be an attractive candidate that could be
tested for its ability to restore transcriptional activation
by ER␣ on the rat OTR and PPE promoters in the presence of the inhibitory TR␣1 isoform. To explore the idea
that coactivators are sequestered by the TR isoforms, we
overexpressed a general steroid coactivator, the SRC-1,
along with TR␣1 and ER␣. On both PPE and OTR (Fig.
4B), promoters as well as the minimal promoter containing the consensus ERE, SRC-1 overexpression could rescue TR␣1 inhibition, suggesting that squelching of common coactivators is an important mechanism of inhibition
by the TR isoforms. Recently, Auger et al. (10) have
shown that reduction in brain SRC-1 levels by antisense
oligonucleotide injection reduces the ability of the ER to
defeminize the brain during postnatal sexual differentiation in Sprague-Dawley rats. Inhibition of T3-dependent
transcriptional activation by other nuclear receptors such
as the glucocorticoid and estrogen receptor has been
reported to be due to titration of essential coactivators
(209). The ligand-bound TR␣ and TR␤ proteins could
interfere with progesterone receptor (PR)-mediated
transactivation from a progesterone responsive reporter
in the CV-1 cell line. Deletion of the DNA binding region
did not affect the inhibitory properties; however, deletion
of the six amino acids in the ligand binding domain
needed for binding coactivators abolished the interference (214). Therefore, squelching of proteins has important consequences for gene regulation. For example, the
coactivator proteins RIP140 and TIF2 compete for a common binding site on the glucocorticoid receptor (GR),
allowing TIF2 to relieve the inhibitory effect of RIP140 on
GR action (180). Although it is not clear if ER-containing
neurons also coexpress SRC-1, the widespread distribution of SRC-1 in the brain makes this likely (10). Also,
many ER containing VMH neurons coexpress SRC-1, thus
making such alleviation of inhibition possible in vivo (10).
Therefore, the ability of the TR␣1 isoform to bind DNA as
well to squelch coactivators such as SRC-1 provides a
rationale for the inhibition observed with this isoform.
PATTERNS OF ER AND TR ACTIONS IN CNS
B. Differences in Isoforms From Nuclear Receptor
Genes: Use of Knock-out Models
1. TR gene knock-out mice
Differences in the physiological roles of the different
isoforms have been explored using knock-out mice models. Despite similar ligand binding characteristics, the
differential distribution of the TR isoforms as well as data
obtained from knockout mice suggest a unique role for
the various TR isoforms. The TR␣1 and the TR␤ isoforms
have both common and specific roles in vivo. Predicated
on the high concentration of TR␤2 in the anterior pituitary
(69), the TR␤2 isoform plays a major role in the negativePhysiol Rev • VOL
feedback regulation of TSH by thyroid hormone. Lack of
TR␤2, therefore, causes hyperthyroidism in mice (2). On
the other hand, lack of the TR␣1 isoform results in a mild
hypothyroidism in mice (75). The distribution of the TR␤2
isoform in the developing retina of the mouse is also
indicative of an important developmental role for this
isoform. In rodents, cones contain different opsins sensitive to different wavelengths. The TR␤2 isoform is responsible for a commitment to M-cone (M, middle or green
wavelengths) identity. Deletion of this isoform results in a
lack of M cones and a concomitant increase in S-opsin (S,
short wavelength) immunoreactive cones (119). The inability for one isoform to substitute for another is also
exemplified by T3-controlled type 1 deiodinase expression
(6). Although both TR␣1 and TR␤ are present in both liver
and kidney, expression of the deiodinase was highly dependent on TR␤ in the liver and completely dependent on
TR␤ in the kidney (6). Another example, the TR␤1 isoform, is implicated in hearing loss, whereas the TR␣
knock-out mice remain unaffected (2).
The TR␣ and TR␤ isoforms also play distinct roles in
the facilitation of lordosis in female mice. Deletion of the
TR␣1 isoform resulted in decreased lordosis behavior in
female mice, whereas loss of the TR␤ isoforms resulted in
increased lordosis (36). OT immunoreactivity in the paraventricular nucleus (PVN) was elevated in TR␤ knock-out
female mice treated with estradiol compared with wildtype mice given the same treatment, implicating OT increase in the PVN as important in increased lordosis (36).
Both behavioral and molecular data on the cross-talk
between the ER␣ and TR isoforms on the PPE and OTR
promoters point to opposing effects of the TR␣ and TR␤
isoforms.
2. ER␣ knock-out mice versus ER␤ knock-out mice:
behavioral phenotypes
Reproductive and affiliative behaviors are differentially affected according to which of the ERs is deleted in
mice. ER␣ knock-out (ERKO) female mice show virtually
no lordosis (128, 132), whereas ER␤ knock-out mice
(BERKO) not only show normal lordosis behavior but
express this behavior during a larger portion of the estrous cycle than wild-type littermate controls (126).
ERKO females have striking deficits in maternal behavior.
Dramatically, aggressive behaviors in young adult BERKO
males are heightened (124), whereas they are markedly
suppressed in ERKO males (131). Note that there appears
to be genotype/age interactions in aggressive behavior by
BERKO mice, in that the relatively inexperienced young
BERKO mice are more aggressive in resident-intruder
tests. Finally, the increase in locomotor activity in both
genetic females and genetic males, following estrogen
administration, depends absolutely on the patency of the
ER␣ gene but not the ER␤ gene (127).
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
Ref. 145) that the behavior virtually stands as an expression system for ER transcriptional activation. In turn,
interactions between liganded TRs and ER function can
be charted thereby.
The neuroanatomy of estrogens liganded to ER␣ or
ER␤ has been charted in considerable detail (144, 146,
175). VMH neurons binding estrogens sit on top the lordosis behavior neuronal circuit (148). Hormone implant
experiments establish that it is the estrogenic sensitivity
of these neurons that accounts for hormonal facilitation
of lordosis. New RNA and protein synthesis are required
for the behavioral facilitation. Specific, hypothalamically
expressed genes have the following properties: they are
turned on by estrogens, and their products facilitate lordosis (150). Therefore, in the manner of a logical syllogism, their induction comprises part of the mechanisms
by which estrogenic hormones turn on the behavior.
These particular genes in no way exhaust the possibilities of hormone-stimulated messages, which are behaviorally relevant. New DNA-microarray experiments
have revealed hitherto unimagined genes that are hormone sensitive (113) which indicate linked glial/neuronal
and possible leptomeningeal/neuronal cooperation in
neuroendocrine function.
Normal gene expression for ER␣ is required for normal lordosis behavior (128, 132). In dramatic contrast,
active gene expression for ER␤ actually suppresses lordosis; ER␤ knock-out female mice show the behavior
during a larger portion of their estrous cycles than wildtype female littermate controls (126).
Therefore, estrogen-dependent lordosis behavior is
well suited to look for thyroid/estrogenic interactions.
Thyroid hormone administration, in fact, reduces lordosis
behavior performance both in female rats (38) and in
female mice (114). The molecular mechanisms involved
are not necessarily simple and require further investigation; that is, against all predictions, the contribution of the
TR␣ gene to the regulation of female reproductive behavior is diametrically opposite to that of the TR␤ gene (36).
931
932
VASUDEVAN, OGAWA, AND PFAFF
3. Cross-talk between the ER␣ and ER␤ isoforms
It has been suggested that the inability of ERKO mice
to induce OTR in response to estradiol benzoate treatment (212) is a factor in their failure to promote social
interactions. Antagonistic effects of two ER isoforms expressed in the same cell have also been reported. On a
consensus ERE promoter in HeLa cells, Hall and McDonnell (66) have noted that coexpression of ER␤ along with
ER␣ reduces the transactivation seen with ER␣. We were
interested in investigating if there is a similar effect on a
physiological promoter in both the cell lines. However,
although ER␤ did not affect the transactivation observed
with the ER␣ isoform on the OTR promoter in response to
17␤-estradiol in the CV-1 cell line (Fig. 5), it decreased the
transcriptional activation observed on this promoter in
the SK-N-BE2C cell line (Fig. 5). Again, cell-specific effects such as the expression of tissue-specific cofactors
may play a role in this phenomenon. The inability of
estrogen-liganded ER␤ to activate transcription from a
physiological OTR promoter may also help explain a result obtained from ERKO mice. The OTR gene promoter is
upregulated by estrogens in several brain regions. Unlike
in the rat, estrogens do not induce OTR expression in the
mouse hippocampus but induce it in the cortex (212).
However, the ERKO mouse, which does not have ER␣ but
has ER␤, lacks estrogen induction of OTR in many brain
regions, such as the cortex, as monitored by OT binding
(212). This may be explained by the differential distribution of ER isoforms as well as the inability of the ER␤ to
induce the OTR. In the mouse hippocampus, ER␣ expresPhysiol Rev • VOL
sion is sparse, although ER␤ expression is intense (174).
It has been suggested that ER␤ distribution in the rat
brain results in overall “global” functions for ER␤ in estrogen action, augmenting cognitive processes and neuronal regeneration (176).
However, the cortex of the mouse expresses both
ER␣ and ER␤ isoforms. Although the ER␣ isoform consistently supported 17␤-estradiol induction of the rat OTR
promoter in both cell lines tested, the ER␤ isoform failed
to induce the OTR gene promoter under identical conditions. This would predict that brain regions rich in ER␤
but poor in ER␣ such as the hippocampus would fail to
support a significant induction of OTR gene expression in
wild-type mice. When the predominant ER isoform
present in brain areas is the noninducing ER␤ isoform,
this is precisely the case. The inability of ER␤ to induce
transcription in response to 17␤-estradiol has also been
reported in transiently transfected NG108 –15 neuroblastoma glioma cells using a neuropeptide Y-Y1 receptor
gene promoter (117). This receptor promoter has two
half-site ERE, and estrogen-mediated transcription is
strictly dependent on the presence of transfected ER␣.
Coexpression of both ER isoforms abolished the ER␣mediated transactivation, suggesting an antagonistic effect of ER␤ on this physiological promoter (117).
4. Mechanisms for ER␣ cross-talk with ER␤
Do relative amounts of ER␣ and ER␤ contribute to
this result? As monitored by [3H]estradiol binding, ER␣
and ER␤ appeared to be expressed equivalently to each
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
FIG. 5. Effect of the coexpression of ER␣ and ER␤ on oxytocin receptor (OTR) gene transcription in CV-1 and
SK-N-BE2C cells. Both ER␣ and ER␤ expression plasmids were cotransfected into CV-1 and SK-N-BE2C cells at equal
concentrations along with the OTR-luciferase construct. A corresponding set of samples received the ER␣ or the ER␤
expression construct alone with the OTR-luciferase reporter construct. After treatment of each set with 10⫺7 M
17␤-estradiol or ethanol for 48 h (n ⫽ 6/treatment group), cells are lysed and assayed for ␤-gal and luciferase activity.
The results are analyzed using ANOVA followed by Student-Newman-Keuls post hoc test to compare between treatment
groups. CV-1 cells: *P ⬍ 0.001 compared with vehicle treatment of the ER␣ group. #P ⬍ 0.01 compared with vehicle
treatment of the ER␣ and ER␤ group. SK-N-BE2C cells: ‚P ⬍ 0.01 when compared with vehicle treatment of the same
group. ƒP ⬍ 0.01 when compared with estrogen treatment of the ER␣ group. [Modified from Vasudevan et al. (189).]
933
PATTERNS OF ER AND TR ACTIONS IN CNS
ucts. On the right side of Table 2 are summarized some of
the data gathered from genetic females and genetic male
mice on a number of behavioral and histochemical assays.
The variety of relations between ER␣ and ER␤ are reminiscent of the differences seen between ER␣ and ER␤ in
their molecular interactions, reviewed above, with a given
TR isoform in the cell culture model systems. For both the
molecular and the behavioral studies, the specificities of
interactions among ER gene products and TR gene product isoforms additionally provide us with internal
controls.
In the early 1940s, Beadle and Tatum (13), in their
famous genetic and biochemical studies on the fungus
Neurospora crassa, found that the loss of an enzyme led
to a specific biochemical defect. The underlying cause of
lack of enzyme activity was discovered to be a loss of a
gene, thus leading to the well-known “one gene-one enzyme” hypothesis (13, 158, 183). The data presented above
show us the combinatorial possibilities of nuclear receptor gene product interactions. These force us to redirect
our thinking into a new, more organismal framework
whereby patterns of gene expressions and interactions in
the central nervous system underlie patterns of behavior.
C. Patterns of Behavior
D. Physiological Implications of Thyroid Hormone
Modulation of Estrogen Action
The above data suggest that isoforms deriving from
closely related genes for nuclear receptors play unique
roles that are clearly not equivalent in whole animal studies. The ERKO mice do not exhibit the same behavioral
phenotype as the BERKO mice in several behavior tests,
especially those designed to elicit socially motivated responses. Similarly, the ␣TRKO females treated with estrogens do not display the same levels of sexual receptivity as the ␤TRKO females (36). Hence, the ␣KO are not
equivalent to the ␤KO mice in either ER or TR knock-out
models. Table 2 highlights theoretical scenarios for functional relations between ERKO and BERKO gene prod-
TABLE
1. Neuroendocrine data
The involvement of thyroid hormone in the neuroendocrine control of reproduction has been documented
especially well in starlings and sheep. In these species
inhabiting temperate latitudes, seasonal reproduction ensures the birth of young in conditions that maximize
survival. Therefore, the termination of the breeding season in sheep and the initiation of anestrous occur during
the long-day period (spring and summer). An endogenous
rhythm, which is entrained by such changes in day length,
2. Behavioral phenotypes shown by ␣ERKO and ␤ERKO mice
Necessary?
ER␣ and ER␤
Sufficient?
Neither ER␣
nor ER␤
Neither ER␣
nor ER␤
Either ER␣ or
ER␤
␣, ␤ each for its
own
␣,␤ each for its
own
ER␣ Absent
ER␤
Optimal ER␣/
ER␤ balance
ER␣ Absent
ER␤
Optimal ER␣/
ER␤ balance
Requirement
ER␣ and ER␤
must synergize
ER␣ and ER␤ can
substitute for
each other
ER␣ vs. ER␤
contributions
not related
ER␤ can reduce
ER␣ effect
ER␣ vs. ER␤
always opposed
Assays in the Male
Assays in the Female
None
None
E induction of PR (ICC) ER reduction of ER
(ICC)
E induction of PR ER
reduction of ER (ICC)
Maternal behavior (␣), suppression of
aggression (␣), food intake reduction (␣),
anxiety reduction (␣)
Lordosis behavior
Mounting behavior (␣), anxiety
response (␣)
None
Intromissions and ejaculation
Aggression
None
ER␣ knock out (␣ERKO) and ER␤ knock out (␤ERKO) male (124, 130, 131, 133) and female (126 –128, 132) mice were tested on a variety of
behavioral assays. The criteria for the requirement for either ER␣ or ER␤ are detailed in the table.
Physiol Rev • VOL
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
other in both CV-1 cells (190) and SK-N-BE2C (189) cell
lines. Although not statistically significant, ER␤ binding to
[3H]estradiol appeared to be slightly lower and may have
contributed to the noninducibility of the rat OTR promoter by ER␤. In contrast, in CV-1 kidney fibroblast cells
(Fig. 3B), ER␤ was capable of mediating the 17␤-estradiol
induction of the natural PPE gene promoter fragment.
Also, ER␤ is capable of promoting neurite elongation in
SK-N-BE2C human neuroblastoma cells in response to
added 17␤-estradiol, thus proving that both cell lines are
responsive to estrogens via both ER␣ and ER␤ (140).
Therefore, the nonresponsiveness of the OTR gene promoter on transfection of the ER␤ isoform in both the cell
lines tested is promoter and ER isoform specific. The
lower transcriptional efficiency of ER␤ has been noted
using the consensus ERE construct, ERE-tk-luc, in COS-1
and HepG2 cells. With the use of Gal4 DNA binding fusion
proteins fused to the AF-1 domains of either ER␣ or ER␤,
it was determined that the AF-1 activity of ER␤ was
negligible compared with ER␣ (32). On promoters and in
cell lines which require both AF-1 and AF-2 activity, ER␤
appears to be a poorer transcriptional activator than
ER␣ (32).
934
VASUDEVAN, OGAWA, AND PFAFF
2. Mechanisms of thyroid hormone-induced
anestrous in sheep
The mechanisms of thyroid hormone-induced
anestrous in sheep have centered mainly on the gonadal
hypothalamic-pituitary axis. There is no effect of THX in
female ewes on the ability of a rise in estrogens to elicit
the LH surge or in the ability of progesterone to suppress
LH secretion (199). However, there is intensified estrogen-mediated negative feedback in control ewes compared with THX ewes (199). High-frequency pulses of
Physiol Rev • VOL
both GnRH and LH are observed in THX ewes that did not
make the transition to anestrous (198). Central infusion of
T4 to THX and ovarectomized ewes given Silastic implants
of estradiol benzoate restored anestrous to these ewes.
This demonstrated that thyroxine acts centrally in the
brain in ewes to promote changes in GnRH and LH that
signal anestrous (194). Also, TR␣ has been colocalized in
46% of GnRH neurons in sheep (74). In rats, hyperthyroid
rats had 25% less LH on proestrous, showing depression
of the LH surges. Also, the amount of estrogens required
to initiate a LH surge was greater in hyperthyroid animals
(53). Hyperthyroid animals could, however, respond to
GnRH, suggesting that the pituitary was not a site of
action for thyroid hormone. The hypothalamus is more
plausible, since stimulation of the arcuate nuclei-median
eminence area (ARC-ME) resulted in hyperthyroid rats
secreting less LH than control rats (53). In rats devoid of
the thyroid gland, the synthesis and metabolism of LH
was not affected, but the secretion of LH was higher (52).
Propylthiouracil (PTU), a goitrogen, given transiently to
neonatal rats dramatically increases sperm production
and testis size in the adult rat. However, it leads to a
significant drop in GnRH-stimulated LH production. Gonadal feedback is enhanced in PTU-treated males resulting in chronically reduced circulating levels of LH and
follicle stimulating hormone (86).
3. Thyroid hormone effects on reproductive behavior
in rodents
Thyroid hormone elevation has also been shown to
have an adverse effect of reproduction in rodents (38,
114). Concomitant administration of T4 to ovariectomized
rats (38) and mice (114) treated with estrogens has been
shown to reduce lordosis, compared with ovariectomized
rodents that received estrogens alone. TR knock-out female ovariectomized and estrogen-treated mice deleted
for TR␤ isoforms showed higher lordosis than the
␤TRWT, suggesting that TR␤ may exert an inhibitory
influence on ER-controlled reproduction (36).
Because reproductive behavior is controlled by estrogens via the ER, it is possible that a reduction in ER
target genes such as OT, OTR, and PPE could be responsible for TR-mediated inhibition (149, 217). Indeed, injections of thyroid hormone to estrogen-treated female rodents lead to a decrease of OT mRNA in the PVN (39). Ex
vivo studies indicate that thyroid hormone upregulates
the human OT promoter fivefold through the composite
element containing an imperfect ERE located at ⫺148/
⫺172 bp upstream of the transcriptional start site (4).
TR␣1 protein can bind to this composite element and
interfere with the transcriptional induction by estrogens
(4). Thyroid hormone elevation also reduces the expression of another estrogen-induced gene in the VMH, the
PPE gene, which facilitates lordosis behavior (37).
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
controls the timing of seasonal reproduction (78, 166,
167). In starlings, thyroidectomy of starlings prevented
the start of photorefractoriness and allowed for continuation of the breeding season (201). Thyroxine rise during
long days in European starlings is permissive for the
neuroendocrine shift to the nonbreeding season, which is
primarily dictated by day length (15). In the thyroidectomized male American tree sparrow, administration of T4
given intracerebroventricularly could restore all components of seasonality. Therefore, T4 was capable of acting
centrally to program already photostimulated male American male sparrows (203).
However, in mammals, both retinal photoreceptors
and the pineal gland are required for reproductive responses to photoperiod (120). Long days induced a drop
in lutenizing hormone (LH) in thyroid-intact ewes, though
thyroidectomy blocked this effect (112). However, thyroidectomy had no effect on the circadian pattern of
circulating melatonin or prolactin and the change that
occurs with the photoperiod (34). Thyroid hormones may
play a permissive role to photoperiod; they need to be
present at the end of the breeding season for anestrous to
commence. This critical period of neuroendocrine responses to thyroid hormone is late in the breeding season.
The minimal effective duration of exposure to circulating
levels of thyroid hormone was 60 –90 days beginning in
late December such that anestrous could develop in
spring (184). Do thyroid hormones also play a role in the
maintenance of anestrous once it develops? In ewes thyroidectomized (THX) just as they entered anestrous, the
timing of the LH rise late in anestrous, indicative of the
next breeding season, was the same as non-THX controls.
Therefore, although thyroid hormones play a role in initiating anestrous, they do not have any role in the maintenance of the anestrous and the timing of the subsequent
breeding season (185) in sheep. However, in the male
American tree sparrow, T4, T3, and reverse T3 given intracerebroventricularly could allow for thyroid hormonedependent photoperiodic testicular growth. The order of
potency was T4 ⬎ T3 ⬎ reverse T3 (203). These data
demonstrate that thyroid hormone may play slightly different roles in the maintenance of nonreproductive conditions in mammals and birds.
PATTERNS OF ER AND TR ACTIONS IN CNS
4. Modulation of estrogen action by thyroid hormone
in other species
5. Estrogen and thyroid hormone influence each
other’s nonreproductive functions
One of the most prominent effects of estrogens is to
promote mitosis in the uterine luminal epithelium,
stroma, and myometrium. The ability of hypothyroid rats
to increase the mitotic index in these uterine regions is
reduced compared with the euthyroid controls (87). This
diminished uterine response is not due to a shift in the
dose-response curve of the estrogen; rather, it is possible
that thyroid hormones have a direct effect on the uterus
such that it lowers its responsiveness to estrogens (57). In
pregnancy, despite lower levels of free T4 and free T3,
there is no rise in serum TSH (50). A similar absence of
TSH rise is seen in postmenopausal women receiving
estrogen replacement therapy (1). Although estrogen
treatment did not augment the serum concentrations of
TSH in euthyroid or untreated hypothyroid rats, it increased the suppressive T3 effect on serum TSH in hypothyroid animals. An increase in the number of pituitary
nuclear receptors for T3 was seen after estrogen treatment in rats, suggesting that the augmentation of the T3
effect may be due to increase in thyroid hormone receptors (51). In bone tissue, where estrogens promote bone
mineral density, hyperthyroidism has been shown to increase bone turnover and decrease bone density (49). In
the pituitary, there was a significant increase in weight
and total cellular RNA when ovariectomized rats were
given estrogens. This increase was inhibited by concomitant administration of T3 (216). Estrogen produces an
anorectic effect in rats, presumably by acting on the
hypothalamus. T4 given daily subcutaneously could antagonize the estrogen-mediated anorectic effect. It has been
proposed that this antagonism could be related to thyroxPhysiol Rev • VOL
ine’s reductive effect on blood glucose level and subsequent decrease in satiety (211).
In the gonadectomized rat, there is a subset of estrogen-driven physiological responses that require thyroid
hormonal interplay. These include the estrogen suppression of somatic growth and the effects of estrogens on
serum triglycerides. It also includes estrogenic suppression of LH secretion (negative feedback control) (48).
Surprisingly, this subset of T3-dependent estrogen responses also can be promoted by tamoxifen acting as an
estrogen agonist. Because T3 regulates growth and energy
metabolism, this may provide an interactive mechanism
for relating metabolic state to reproductive biology (48).
Thyroid hormone may also affect the clearance rate
of estrogen, although there are conflicting studies on this
theme. Hypothyroidism lowers the clearance rate of estrogen in women (102). In another study in women, hyperthyroidism increased the clearance rate of estrogen
(164). This is also predicted by data in the male Japanese
quail (165). Also, in hyperthyroid rabbits, the clearance
rate of estrogen is increased (187). In female Japanese
quail, thyroidectomy actually increased the clearance rate
of estrogen, contrary to the data in males (141). However,
in male cytomegalous monkeys, there was no change in
the clearance rate of estrogen on thyroid hormone treatment (18).
IV. ROLE OF PROMOTER AND CELL
SPECIFICITY IN DISTINCT
TRANSCRIPTIONAL RESULTS
How do differences in sequences bound by the ER
and TR isoforms explain their transcriptional differences
in the context of cell lines and promoters? Different hormone response elements are allosteric mediators of
receptor conformation. Therefore, hormone response
elements not only position receptors close to basal
transcription complexes but also serve to direct the mode
of regulation of the target gene (100). Studies with the
consensus vitellogenin A2 ERE, or the imperfect pS2,
vitellogenin B1 or oxytocin (OT) ERE show that the A2
was the most potent activator of transcription followed by
the OT ERE (205). DNase footprinting revealed that
MCF-7 proteins protected the OT and A2 EREs to a
greater extent than the pS2 or B1 EREs. Although the
receptor-interacting domains of the glucocorticoid receptor interacting protein 1 (GRIP) and SRC-1 bound effectively to ER␣, TIF2 was bound less by B1-bound ER␣ than
A2-bound ER␣, suggesting that allosteric modulation of
ER␣ conformation by different EREs influences coactivator recruitment (205).
Different isoform conformations within the cell
could also have an effect in the recruitment of coactivators. The TR␤2 isoform, for example, can bind p160 class
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
In other species, thyroid hormone has also been
shown to modulate estrogen action. In the fish tilapia,
Oreochromis neoloticus, three distinct populations of
GnRH exist: the terminal nerve neurons in the forebrain,
the preoptic neurons, and the midbrain neurons. In castrated male tilapia, terminal nerve neurons express
GnRH, which is lowered by exogenous T4 treatment (138).
Interestingly, in the tilapia, the ontogeny of terminal nerve
neuron GnRH is concomitant with a decrease in T4 levels.
Sexually mature tilapia have low levels of thyroid hormones but high levels of terminal nerve GnRH (197). In
oviparous species such as the clawed toad, Xenopus laevis, metamorphosis is dependent on thyroid hormone
while vitellogenesis is strongly dependent on estrogens
(155). T3 could enhance ER␣ production and autoinduction and thereby enhance the estrogenic activation of
vitellogenin genes (156).
935
936
VASUDEVAN, OGAWA, AND PFAFF
Physiol Rev • VOL
of functions is indeed unique to a certain isoform and
cannot be substituted by the other isoforms. The experiments suggest that a variety of transcriptional outcomes
are possible depending on the combination of isoforms
present in the cell. These represent different flexible outputs, presumably to ensure homeostasis, in response to
the same external stimulus, i.e., presence of ligand. We
suggest that the isoforms have evolved to allow for different cell responses to the same signal, possibly by differential cross-talk between isoforms, to achieve neuroendocrine integration.
V. QUESTIONS UNANSWERED
A. Gene Duplication and Splice Variants
Gene duplication events and splice variations comprise likely origins for a number of hormone receptor
isoforms. Those isoforms, which are gene duplication
products, provide fascinating opportunities to compare
their physiological roles. Some current examples from
molecular endocrinology and neuronal biophysics suggest that diametrically opposite functions represent an
interesting possibility.
Gustafsson and colleagues’ (122) have made the case
that in peripheral organs ER␤ can oppose the actions of
ER␣. Neurobiological results both with behavioral end
points (126, 131–133) and during transfection studies
(189, 190, 192) support this point of view. With respect to
the CNS, both transcriptional end points in cell lines (189,
192) and behavioral results (36) offer the possibility of
markedly different TR␣ and TR␤ actions.
Among nuclear hormone receptors, PRs show the
wide diversity of actions of PR splice variants; that is,
under circumstances where PR␤ can act as a transcriptional activator, the smaller splice variant PR␣ can have
the opposite transcriptional effect (58, 193, 200). The
same can be illustrated for neuropeptides. OT and vasopressin are extremely similar nonapeptides, which almost
certainly represent gene duplication products. OT is well
known to promote affiliative behaviors (23, 142). In contrast, vasopressin promotes aggressive and territory-defensive behaviors (5, 46, 47). The electrophysiological
actions of neurotransmitter receptors make the same
case. For the transmitter norepinephrine (␣1 vs. ␣2 receptors), dopamine (D1 vs. D2 receptors), serotonin (5-HT1
vs. 5-HT2 receptors) and acetylcholine (M1 vs M2) receptors, markedly different and even opposing actions of
likely gene duplication products are the rule rather than
the exception. All of these examples constitute new opportunities for the fine-grained analysis of gene domains
governing specific neuroendocrine functions.
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
of coactivators in the absence of the hormone and, therefore, mediate ligand-independent activation of target
genes. This is mediated by contacts in the unique NH2
terminus of TR␤2 and an internal interaction domain of
SRC-1 and GRIP-1 coactivators. These contacts are different from the LXXLL motifs that mediate hormone-dependent coactivator contacts and hence hormone-dependent
transcriptional activation (206). The NH2-terminal region
in the human PR (hPR) also modulates differential coactivator and repressor binding. The hPR exists as two
different isoforms: hPRA, which is a strong ligand-dependent repressor of transcription, and the hPRB, which is a
transcriptional activator in most cell and promoter contexts. An inhibitory domain (ID) present in the hPRA and
B is active only in the hPRA isoform, facilitating the
interaction between this isoform and the transrepressor
SMRT. This and the inability of the hPRA isoform to
engage coactivators could provide an explanation of the
differential activities of the isoforms (58). Also, the specificity of NCoR interactions with TR␤1 could be because
of the ability of this TR isoform to interact with a novel
NCoR interacting domain, called N3 which is specific for
TR and interacts very poorly with other nuclear receptors,
such as the RAR␣ (29). Possible differences in isoform
conformation may allow for tissue-specific expression.
For example, although a ERE oligonucleotide derived
from the rat 3-hydroxy-3-methylglutaryl CoA (HMG CoA)
reductase promoter confers estrogen responsiveness to a
heterologous promoter in several cell lines, the “natural”
HMG CoA promoter containing the ERE shows estrogen
induction in MCF-7 breast carcinoma cells but not in
hepatic cell lines (40).
Within the mammalian CNS, the physiological results
of hormones liganding to specific nuclear receptor gene
products, as well as the consequences of their interactions, may often depend on the details of neuronal or glial
localization; that is, the neuroanatomical pattern of expression of ER␣ is much different from that for ER␤
(175), and the TR␣ and TR␤ gene products likewise are
distributed differently both inside and outside the CNS
(19, 90, 101, 111). Most importantly, differential distributions that depended on promoter sequences active during
certain developmental stages may well underlie functional differences between nuclear receptor isoforms. In
turn, what about the requirement that for direct TR/ER
interactions they must be expressed in the same neuron?
Kia and colleagues (83– 85), using a sensitive double in
situ hybridization methodology, showed that a high percentage of forebrain neurons expressing ER also expressed TRs.
Why are isoforms of the nuclear receptors such as ER
and TR maintained in organisms? The differential colocalization of the isoforms is a clue that they have distinct
nonoverlapping functions. The phenotypes of the isoformspecific knock-outs also appear to indicate that a subset
PATTERNS OF ER AND TR ACTIONS IN CNS
B. Rapid Versus Slow Effects of Estrogen:
Two Separate Opportunities For Thyroid
Hormone Modulation?
Physiol Rev • VOL
greater transcription than either pulse alone (Fig. 6). Reversal of the order of addition, 17␤-estradiol in the first
pulse and E-BSA in the second pulse, did not have any
effect on transcription, showing that primary membrane
effects of estrogens are important to the nuclear effects of
estrogens. Addition of inhibitors to the signal transduction cascade in the first 2-h pulse along with E-BSA could
inhibit this synergism, demonstrating that activation of
protein kinases A and C plays a role in facilitation of
transcription by the nuclear 17␤-estradiol-bound ER. In
contrast, addition of these inhibitors in the second 2-h
pulse along with 17␤-estradiol did not reduce synergism.
This synergistic transcription could be blocked by concomitant addition of the nuclear ER antagonist ICI in
either the 20-min first or 2-h second pulse, demonstrating
⫺9
FIG. 6. E-BSA (10
M), given for only 20 min in the first pulse, can
potentiate transcription mediated by 17␤-estradiol (10⫺9 M) in the second pulse. SK-N-BE2C, a human neuroblastoma cell line, was cotransfected with ERE-luciferase, pSG-hER␣, and pSV␤gal plasmids, 48 h after
plating, using the Effectene reagent (Qiagen), according to the manufacturer’s protocol. The day after the transfection, 17␤-estradiol was
added using a two-pulse paradigm. Briefly, 17␤-estradiol (10⫺9 M) was
added to the media twice in a first pulse of 20 min and a second pulse of
2 h with these two pulses separated by an interval of 4 h. At the end of
the second 2-h pulse of 17␤-estradiol, the cells were washed free of
hormones, fresh media were added, and incubation continued for a
further 16 h. Cell lysates were then prepared using the Reporter lysis
buffer as per the manufacturer’s instructions (Promega), and the luciferase activity normalized to the ␤-galactosidase (␤-gal) activity for every
sample. The results (fold over control) are expressed as fold induction
over that achieved by the vehicle ethanol alone. Control condition:
17␤-estradiol (10⫺9 M) in the first pulse was followed by E-BSA (10⫺9 M)
in the second pulse (4th bar from left). Test of “potentiation” hypothesis:
E-BSA given in the first pulse was followed by 17␤-estradiol in the
second pulse (far right bar). Results (n ⫽ 4/treatment group) are represented as means ⫾ SE. Statistical analysis was done using one-way
ANOVA using Student-Newman-Keuls post hoc test to compare between
treatment groups. *P ⬍ 0.05 compared with all other treatment groups.
#P ⬍ 0.05 compared with the vehicle group. The control condition was
also greater to groups where E-BSA in the first pulse followed vehicle in
the second pulse and where 17␤-estradiol in the first pulse was followed
by vehicle in the second pulse. The activity in the control group equaled
that of 17␤-estradiol given in both pulses (data not shown). [Modified
from Vasudevan et al. (191).]
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
Although the classical mode of estrogen action is via
the nuclear ER and leads to the modulation of transcription rate of estrogen-target genes, nonnuclear ER-mediated effects exist and are certainly prominent in neurophysiological literature (14, 80 – 82). These nonnuclear ER
effects are typically rapid and are mediated by a putative
membrane receptor (mER) and involve several signal
transduction pathways (79 and references therein). Membrane-limited estradiol conjugates are used to investigate
the membrane effects of estrogens. The MAPK has been
implicated as a downstream effector molecule in estrogen’s rapid action at the membrane in a neuroblastoma
cell line, the SK-N-SH cell line (196). In a cell line with
endogenous ER␣ (the breast carcinoma MCF-7 line), nongenomic mechanisms have been shown to activate MAPK
(73). Protein kinase C activation in growth plate chrondocytes is seen on administration of both 17␤-estradiol
and E-BSA (181). The rapid rise in intracellular free Ca2⫹
has been thought to be mediated by mERs (9, 115, 179).
The identity of the putative mER has been explored in
several studies. In Chinese hamster ovary (CHO) cells,
ER␣ expressed in a transient transfection assay gave rise
to a single transcript and functional protein expression in
both nuclear and membrane fractions (159). Microscopy
on fetal rat hippocampal neurons reacted with affinitypurified anti-ER␣ revealed abundant staining at the neurites. Also, incubation with antisense oligonucleotides directed against ER␣ reduced the immunostaining (28).
Although work on the nuclear ER and membrane ER
has been ongoing for several years, they have usually
been presented as alternate modes of estrogen action.
However, this laboratory has recently shown that these
two modes of estrogen action can in fact synergize. Therefore, it is possible that an early, preliminary administration of estrogen leads to rapid activation of signal transduction cascades at the membrane, and this facilitates
transcription by a later administration of 17␤-estradiol (191).
The study utilized a two-pulse hormonal schedule in
an ex vivo neuroblastoma cell culture model system. Such
a pulse regimen was shown to be previously effective in
investigating estrogen’s effects on cell division (67). The
first pulse (of either 20-min or 2-h duration) utilized EBSA, a membrane-limited estrogen conjugate designed to
limit estrogen actions to the membrane and the second
2-h pulse utilized 17␤-estradiol to stimulate transcription.
Transcription from a transiently transfected reporter
gene, consisting of three tandem consensus EREs linked
to luciferase, was then measured. A 20-min first pulse
using E-BSA and a second 2-h pulse utilizing 17␤-estradiol, separated by a hormone free interval of 4 h, showed
937
938
VASUDEVAN, OGAWA, AND PFAFF
that in this model system the mER is similar to the nuclear
ER (191).
Could thyroid hormone modulation of ER gene transcription affect either or both of these modes of estrogen
action? Discrete targets of modulation impart specificity
to thyroid hormone action. Also, the mechanisms of stimulatory cross-talk between the ER and TR remain currently unknown and would be an avenue of future research.
C. Thyroid Hormone Elevation: Does It Signal
Cold Temperatures?
Physiol Rev • VOL
VI. SUMMARY
Transcriptional influences of enhancer complexes on
gene promoters can depend on complex interplay among
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
Impetus to the examination of TR and ER cross-talk
was provided by a series of studies investigating the nature of the mammalian response to cold temperatures and
its dependency on thyroid hormone. Cold temperatures
are also correlated with seasonal shifts in photoperiod
(short day length), which in turn signal anestrous in several mammalian species. Cold exposure increases the
blood levels of thyroid hormones and TSH. Despite elevated inhibitory levels of thyroid hormones, it also causes
an increase of TRH mRNA in the PVN, demonstrating the
ability of cold to override inhibition by T3 (219). Such cold
stress also transiently elevated corticotropin releasing
hormone (CRH) levels, cortisol, and ACTH levels in rats
(55). Acute stressors such as uncontrollable foot shock in
rats also elevated brain T3 levels in both male and female
rats (54). The role of brown adipose tissue (BAT) in
adaptive nonshivering thermogenesis in mammals has
been well studied. Intracellular conversion of T4 to T3 is
required for the optimal thermogenic function of BAT
(16). In thyroidectomized rats, the acute thermogenic response of BAT to cold exposure (measured by increase in
mitochondrial GDP binding) was decreased (139). The
5⬘-deiodinase present in BAT catalyzes the formation of
T3 from T4 and is elevated by cold temperatures (161).
Such activation of the deiodinase saturates the nuclear TR
in the BAT, and it plays a central role in thyroid hormonedependent thermogenic response (25). Uncoupling proteins, named UCP1, -2, and -3 (based on sequence homology), are inner mitochondrial membrane proteins that
generate heat by uncoupling oxidative phosphorylation
from the respiratory chain. UCP2 is ubiquitously expressed while UCP3 is predominant in skeletal muscle
(163). Such thermogenesis may be adaptive nonshivering
thermogenesis that occurs in newborns and small mammals (rodents) in response to cold or may be due to
metabolic substrate excess. Hence, the UCPs play a central role in energy balance and cold acclimitization (162).
At cold temperatures, thyroid hormone-stimulated production of uncoupling protein (UCP1) is the major mechanism by which T3 produces the thermogenic effect in
BAT. It transcriptionally upregulates the rat UCP gene via
two TREs located at around ⫺2,300 bp situated ⬃27 bp
apart in the rat promoter. These two TRE bind both TR
homodimers and the TR-RXR heterodimer and synergize
to give an increase in transcription (157). Increases in
stability of the UCP transcript have also been shown to be
dependent on thyroid hormone (160). Hypothyroidism
(16) and inhibitors to the 5⬘-deiodinase (161) result in
both lower basal levels of UCP1 and in lower response on
stimulation with cold than euthyroid controls. UCP1 levels are both positively correlated with decreased temperatures and with overfeeding and negatively correlated
with starvation and genetic obesity. UCP1-deleted mice
do not show adrenergic adaptive nonshivering thermogenesis in response to cold or thermogenesis in response
to fatty acid supply (diet-induced thermogenesis) (118),
arguing for a central role for UCP1 in thermogenesis. The
roles of UCP2 and UCP3 are more unclear. The human
UCP3 promoter has a TRE (3), which may explain the
upregulation of this protein by thyroid hormone (61).
However, the UCP3-deleted mice had no phenotypic abnormality and had normal body temperature responses to
thyroid hormone and cold exposure (62). UCP2 mRNA is
increased by T3 injections in the BAT, white adipose
tissue (106), and the heart, suggesting that the thyroid
hormone control on basal metabolic rate may be mediated by UCP2 (95). The TR␤-selective ligand GC-1 could
restore the UCP1 level in hypothyroid mice but could not
restore core body temperature, indicating that TR␣1 may
be important in maintaining core body temperature. In
accordance, mice with a TR␣1 ablation have reduced core
body temperature compared with wild type (202). Therefore, thyroid hormone upregulation of all three UCP
mRNAs demonstrates a major role for isoform-specific TR
in thermogenesis.
Ovariectomized Syrian hamsters exposed to cold
showed decreased lordosis behavior and ER immunoreactivity in the VMH when treated with estradiol benzoate
after cold exposure (42). To test the hypothesis that cold
exposure may decrease lordosis in rodents, mice were
exposed to cold and then tested both for locomotor activity using running wheels and lordosis behavior. The
main effect of cold temperature exposure was to reduce
female approaches to the stud male, perhaps as a consequence of markedly decreased locomotor activity (26). If
it could be shown that access of T3 to TRs in nerve cells
of the female mouse forebrain would be increased by this
cold exposure, then it would be possible that TRs which
mediate thermogenesis in response to cold also “transduce” this environmental signal to neuroendocrine systems mediating sexual behavior.
PATTERNS OF ER AND TR ACTIONS IN CNS
We thank Drs. Y. S. Zhu, Chris Krebs, and Kami Kia for
helpful discussions during preparation of this review.
This work was supported by National Institute of Child
Health and Human Development Grant HD-05751.
Current address of N. Vasudevan: 208 Mueller Labs, Pennsylvania State University, University Park, PA 16802.
Address for reprint requests and other correspondence: D.
Pfaff, Box 275, The Rockefeller University, 1230 York Ave., New
York, NY 10021 (E-mail: [email protected]).
REFERENCES
1. ABDALLA HI, HART DM, AND BEASTALL GH. Reduced serum free
thyroxine concentration in postmenopausal women receiving oestrogen treatment. Br Med J 288: 754 –755, 1984.
2. ABEL DE, BOERS ME, MOURA CP, MOURA E, KAULBACH H, ZAKARIA M,
LOWELL B, RADOVICK S, LIBERMAN CM, AND WONDISFORD F. Divergent
roles for thyroid hormone receptor beta isoforms in the endocrine
axis and auditory system. J Clin Invest 104: 291–300, 1999.
3. ACIN A, RODRIGUEZ M, RIQUE H, CANET E, BOUTIN JA, AND GALIZZI JP.
Cloning and characterization of the 5⬘ flanking region of the human
uncoupling protein 3 (UCP3) gene. Biochem Biophys Res Commun
258: 278 –283, 1999.
4. ADAN RA, COX JJ, VAN KATS JP, AND BURBACH JP. Thyroid hormone
regulates the oxytocin gene. J Biol Chem 267: 3771–3777, 1992.
5. ALBERS HE, HENNESSEY AC, AND WHITMAN DC. Vasopressin and the
regulation of hamster social behavior. Ann NY Acad Sci 652: 227–
242, 1992.
6. AMMA LL, BARROS-CAMPOS A, WANG Z, VENNSTROM B, AND FORREST D.
Distinct tissue-specific roles for thyroid hormone receptors beta
and alpha in regulation of type 1 deiodinase expression. Mol Endocrinol 15: 467– 475, 2001.
7. ARLETTI R, BENELLI A, AND BERTOLINI A. Oxytocin involvement in
male and female sexual behavior. Ann NY Acad Sci 652: 180 –193,
1992.
8. ARLETTI R AND BERTOLINI A. Oxytocin stimulates lordosis behavior in
female rats. Neuropeptides 6: 247–253, 1985.
9. AUDY MC, VACHER P, AND DUFY B. 17␤-Estradiol stimulates a rapid
Ca2⫹ influx in LNCaP human prostate cancer cells. Eur J Endocrinol 135: 367–373, 1996.
10. AUGER AP, TETEL MJ, AND MCCARTHY MM. Steroid receptor coactivator (SRC-1) mediates the development of sex-specific brain morphology and behavior. Proc Natl Acad Sci USA 97: 7551–7555, 2000.
Physiol Rev • VOL
11. BALE TL AND DORSA DM. Cloning, novel promoter sequence, and
estrogen regulation of a rat oxytocin receptor gene. Endocrinology
138: 1151–1158, 1997.
12. BALE TL, DORSA DM, AND JOHNSTON CA. Oxytocin receptor mRNA
expression in the ventromedial hypothalamus during the estrous
cycle. J Neurosci 15: 5058 –5064, 1995.
13. BEADLE GW AND TATUM EL. Genetic control of biochemical reactions in neurospora. Proc Natl Acad Sci USA 27: 499 –506, 1941.
14. BECKER JB AND RUDICK CN. Rapid effects of estrogen or progesterone on the amphetamine-induced increase in striatal dopamine are
enhanced by estrogen priming: a microdialysis study. Pharmacol
Biochem Behav 64: 53–57, 1999.
15. BENTLEY GE, GOLDSMITH AR, DAWSON A, GLENNIE LM, TALBOT RT, AND
SHARP PJ. Photorefractoriness in European starlings (Sturnus vulgaris) is not dependent upon the long-day-induced rise in plasma
thyroxine. Gen Comp Endocrinol 107: 428 – 438, 1997.
16. BIANCO AC AND SILVA JE. Intracellular conversion of thyroxine to
triiodothyronine is required for the optimal thermogenic function
of brown adipose tissue. J Clin Invest 79: 295–300, 1987.
17. BODNAR R, COMMONS K, AND PFAFF D. Central Neural States Relating
Sex and Pain. Baltimore, MD: Johns Hopkins Univ. Press, 2000.
18. BOURGET C, FEMINO A, FRANZ C, HASTINGS S, AND LONGCOPE C. The
effects of L-thyroxine and dexamethasone on steroid dynamics in
male cynomologous monkeys. J Steroid Biochem 28: 575–579,
1987.
19. BRADLEY D, TOWLE H, AND YOUNG W. Spatial and temporal expression of a- and b-thyroid hormone receptor mRNAs, including the
b2-subtype, in the developing mammalian nervous system. J Neurosci 12: 2288 –2302, 1992.
20. BRETON C AND ZINGG HH. Expression and region-specific regulation
of the oxytocin receptor gene in rat brain. Endocrinology 138:
1857–1862, 1997.
21. BROAD KD, KENDRICK KM, SIRINATHSINGHJI DJ, AND KEVERNE EB.
Changes in preopiomelanocortin and preproenkephalin mRNA levels in the ovine brain during pregnancy, parturition and lactation
and in response to oestrogen and progesterone. J Neuroendocrinol
5: 711–719, 1993.
22. CALDWELL JD, PRANGE AF, AND PEDERSEN CA. Oxytocin facilitates the
sexual receptivity of estrogen-treated female rats. Neuropeptides 7:
175–189, 1986.
23. CARTER C. Oxytocin and sexual behavior. Neurosci Biobehav Rev
16: 131–144, 1992.
24. CARTER CS AND DEVRIES AC. Oxytocin and vasopressin can influence social behaviors and the HPA axis. In: Stress and Neuroendocrine Systems, edited by Yamashita H. Heidelberg, Germany:
Springer-Verlag, 1999.
25. CARVALHO SD, KIMURA ET, BIANCO AC, AND SILVA JE. Central role of
brown adipose tissue thyroxine 5⬘ deiodinase on thyroid hormonedependent thermogenic response to cold. Endocrinology 128:
2149 –2159, 1991.
26. CHAN J, OGAWA S, AND PFAFF DW. Reproduction-related behaviors of
Swiss-Webster female mice living in a cold environment. Proc Natl
Acad Sci USA 98: 700 –704, 2001.
27. CHEN H, LIN RJ, SCHILTZ RL, CHAKRAVARTI D, NASH A, NAGY L, PRIVALSKY ML, NAKATANI Y, AND EVANS RM. Nuclear receptor coactivator
ACTR is a novel histone acetyltransferase and forms a multimeric
activation complex with P/CAF and CBP/p300. Cell 90: 569 –580,
1997.
28. CLARKE CH, NORFLEET AM, CLARKE MS, WATSON CS, CUNNINGHAM KA,
AND THOMAS ML. Perimembrane localization of the estrogen receptor alpha protein in neuronal processes of cultured hippocampal
neurons. Neuroendocrinology 71: 34 – 42, 2000.
29. COHEN RN, BRZOSTEK S, KIM B, CHOREV M, WONDISFORD FE, AND
HOLLENBERG AN. The specificity of interactions between nuclear
hormone receptors and corepressors is mediated by distinct amino
acid sequences within the interacting domains. Mol Endocrinol 15:
1049 –1061, 2001.
30. COHEN RN, WONDISFORD FE, AND HOLLENBERG AN. Two separate
NCoR (nuclear receptor corepressor) interaction domains mediate
corepressor action on thyroid hormone response elements. Mol
Endocrinol 12: 1567–1581, 1998.
31. COWLEY S, HOARE S, MOSSELMAN S, AND PARKER MG. ERalpha and
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
transcription factors. Here we reviewed evidence that two
classes of transcription factors, TRs and Ers, can interact
in several different modes, which depend on TR isoform,
ER isoform, gene promoter, and cell type.
The in vivo data utilizing gene knock-out mice also
point to different roles for different isoforms for both TRs
and ERs. This is evident from the behavioral data where
the deletion of one isoform does not lead to the same
phenotype as deletion of the other isoforms. For ER␣
compared with ER␤, enough behavioral assays have been
completed to show that different patterns of ER gene
expression are required for different patterns of neuronal
function and behavior. We note that nonredundancy of
isoforms could explain their continued maintenance in
the mammalian genome. In turn, different molecular patterns of TR-ER interactions may underlie alterations in
behavior. The genes for nuclear receptors represent scientifically accessible causal routes for effects of small,
circulating molecules on behaviors of considerable biological import.
939
940
32.
33.
34.
35.
36.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
ERbeta form heterodimers on DNA. J Biol Chem 272: 19858 –19862,
1997.
COWLEY SM AND PARKER MG. A comparison of the transcriptional
activation by ER alpha and ER beta. J Steroid Biochem Mol Biol 69:
165–175, 1999.
CURTIS-HEWITT S, COUSE JF, AND KORACH KS. Estrogen receptor
transcription and transactivation: estrogen receptor knockout
mice: what their phenotypes reveal about mechanisms of estrogen
action. Breast Cancer Res 2: 345–352, 2000.
DAHL GE, EVANS NP, MOENTER SM, AND KARSCH FJ. The thyroid gland
is required for reproductive neuroendocrine responses to photoperiod in the ewe. Endocrinology 135: 10 –15, 1994.
DELANGE FM AND ERMANS AM. Endemic goiter and cretinism. In: The
Thyroid: Physiology and Treatment of Disease, edited by Hershman JM and Bray GA. New York: Pergamon, 1979, p. 417– 451.
DELLOVADE TL, CHAN J, VENNSTROM B, FORREST D, AND PFAFF DW. The
two thyroid hormone receptor genes have opposite effects on
estrogen stimulated sex behaviors. Nature Neurosci 3: 472– 475,
2000.
DELLOVADE TL, KIA HK, ZHU YS, AND PFAFF DW. Thyroid hormone
coadministration inhibits the estrogen-stimulated elevation of preproenkephalin mRNA in female rat hypothalamic neurons. Neuroendocrinology 70: 168 –174, 1999.
DELLOVADE TL, ZHU YS, KREY L, AND PFAFF DW. Thyroid hormone
and estrogen interact to regulate behavior. Proc Natl Acad Sci USA
93: 12581–12586, 1996.
DELLOVADE TL, ZHU YS, AND PFAFF DW. Thyroid hormones and
estrogen affect oxytocin gene expression in hypothalamic neurons.
J Neuroendocrinol 11: 1–10, 1999.
DI CROCE L, VICENT GP, PECCI A, BRUSCALUPI G, TRENTALANCE A, AND
BEATO M. The promoter of the rat 3-hydroxy-3-methylglutaryl coenzyme A reductase gene contains a tissue specific estrogen responsive region. Mol Endocrinol 13: 1225–1236, 1999.
DUPONT J AND LE ROITH D. Insulin-like growth factor 1 and oestradiol promote cell proliferation of MCF-7 breast cancer cells: new
insights into their synergistic effects. Mol Pathol 54: 149 –154, 2001.
EARLY AH, WADE GN, AND LEMPICKI RL. Effects of cold exposure on
estrous behavior and neural estrogen receptor in Syrian hamsters.
Physiol Behav 65: 763–768, 1999.
EVANS RM. The steroid and thyroid hormone receptor superfamily.
Science 240: 889 – 895, 1988.
FAHRBACH SE, MORRELL JI, AND PFAFF DW. Possible role for endogenous oxytocin in estrogen-facilitated maternal behavior in rats.
Neuroendocrinology 40: 526 –532, 1985.
FARSETTI A, ROBBINS J, AND NIKODEM V. Molecular basis of thyroid
hormone regulation of myelin basic protein gene expression in
rodent brain. J Biol Chem 266: 23226 –23232, 1992.
FERRIS C. Role of vasopressin in aggressive and dominant/subordinate behaviors. Ann NY Acad Sci 652: 212–226, 1992.
FERRIS C AND DELVILLE Y. Vasopressin and serotonin interactions in
the control of agonistic behavior. Psychoneuroendocrinology 19:
593– 601, 1994.
FITTS JM, KLEIN RM, AND POWERS AC. Estrogen and tamoxifen
interplay with T3 in male rats: pharmacologically distinct classes of
estrogen responses affecting growth, bone, and lipid metabolism,
and their relation to serum GH and IGF-I. Endocrinology 142:
4223– 4235, 2001.
FRANKLYN J, BETTERIDGE J, HOLDER R, DAYKIN J, LILLEY J, AND SHEPPARD M. Bone mineral density in thyroxine treated females with or
without a previous history of thyrotoxicosis. Clin Endocrinol 41:
425– 432, 1994.
FRANKLYN JA, SHEPPARD MC, AND RAMSDEN DB. Serum free thyroxine
and free triiodothyronine concentrations in pregnancy. Br Med J
287: 394, 1983.
FRANKLYN JA, WOOD DF, BALFOUR NJ, RAMSDEN DB, DOCHERTY K, AND
SHEPPARD MC. Modulation by estrogen of thyroid hormone effects
of thyrotrophin gene expression. J Endocrinol 115: 53–59, 1987.
FREEMAN ME, LAROCHELLE FT, AND MOORE RB. Thyroid hormone
regulation of the pulsatile discharges of lutenizing hormone in
ovariectomized rats. Endocrinology 97: 738 –743, 1975.
FREEMAN ME, LAROCHELLE FT, AND MOORE RB. Effect of thyroid
status on spontaneous and induced surges of lutenizing hormone.
Endocrinology 99: 713–719, 1976.
Physiol Rev • VOL
54. FRIEDMAN Y, BACCHUS R, RAYMOND R, JOFFE RT, AND NOBREGA JN.
Acute stress increases thyroid hormone levels in rat brain. Biol
Psychiatry 45: 234 –237, 1999.
55. FUKUHARA K, KVETNANSKY R, CIZZA G, PACAK K, OHARA H, GOLDSTEIN
DS, AND KOPIN IJ. Interrelations between sympathoadrenal system
and hypothalamo-pituitary adrenocortical/thyroid systems in rats
exposed to cold stress. J Neuroendocrinol 8: 533–541, 1996.
56. FUNABASHI T, BROOKS PJ, KLEOPOULOS SP, GRANDISON L, MOBBS CV,
AND PFAFF DW. Changes in preproenkephalin messenger RNA level
in the rat ventromedial hypothalamus during the estrous cycle.
Brain Res 1: 129 –134, 1995.
57. GARDNER RM, KIRKLAND JL, IRELAND JS, AND STANCEL GM. Regulation
of the uterine response to estrogen by thyroid hormone. Endocrinology 103: 1164 –1172, 1978.
58. GIANGRANDE PH, KIMBREL EA, EDWARDS DP, AND MCDONNELL DP. The
opposing transcriptional activities of the two isoforms of the human progesterone receptor are due to differential cofactor binding.
Mol Cell Biol 20: 3102–3115, 2000.
59. GIMPL G AND FAHRENHOLZ F. The oxytocin receptor system: structure, function, and regulation. Physiol Rev 81: 629 – 683, 2001.
60. GLASS CK, HOLLOWAY J, DEVARY OV, AND ROSENFELD MG. The thyroid
hormone receptor binds with opposite transcriptional effects to a
common sequence motif in thyroid hormone and estrogen response elements. Cell 54: 313–323, 1988.
61. GONG DW, HE Y, KARAS M, AND REITMAN M. Uncoupling protein-3 is
a mediator of thermogenesis regulated by thyroid hormone, beta3adrenergic agonists and leptin. J Biol Chem 272: 24129 –24132,
1997.
62. GONG DW, MONEMDJOU S, GAVRILOVA O, LEON LR, MARCUS-SAMUELS B,
CHOU CJ, EVERETT C, KOZAK LP, LI C, DENG C, HARPER ME, AND
REITMAN ML. Lack of obesity and normal response to fasting and
thyroid hormone in mice lacking uncoupling protein 3. J Biol Chem
275: 16251–16257, 2000.
63. GREINER EF, KIRFEL J, GRESCHIK H, HUANG D, BECKER P, KAPFHAMMER
JP, AND SCHULE R. Differential ligand-dependent protein-protein
interactions between nuclear receptors and a neuronal-specific
cofactor. Proc Natl Acad Sci USA 97: 7160 –7165, 2000.
64. GRUMBACH MM. Estrogen, bone, growth and sex: a sea change in
conventional wisdom. J Pediatr Endocrinol Metab 13: 1439 –1455,
2000.
65. GUSTAFSSON JA. Estrogen receptor beta: a new dimension in estrogen mechanism of action. J Endocrinol 163: 379 –383, 1999.
66. HALL JM AND MCDONNELL DP. The estrogen receptor beta-isoform
(ER␤) of the human estrogen receptor modulates ER␣ transcriptional activity and is a key regulator of the cellular response to
estrogens and antiestrogens. Endocrinology 140: 5566 –5578, 1999.
67. HARRIS J AND GORSKI J. Evidence for a discontinuous requirement
for estrogen in stimulation of deoxyribonucleic acid synthesis in
the immature rat uterus. Endocrinology 103: 240 –245, 1978.
68. HEINZEL T, LAVINSKY RM, MULLEN TM, SODERSTROM M, LAHERTY CD,
TORCHIA J, YANG WM, BRARD G, NGO SD, DAVIE JR, SETO E, EISENMAN
RN, ROSE DW, GLASS CK, AND ROSENFELD MG. A complex containing
N-CoR, mSin3 and histone deacetylase mediates transcriptional
repression. Nature 387: 43– 48, 1997.
69. HODIN RA, LAZAR MA, WINTMAN BI, DARLING DS, KOENIG RJ, LARSEN
PR, MOORE DD, AND CHIN WW. Identification of a thyroid hormone
receptor that is pituitary-specific. Science 244: 76 –79, 1989.
70. HOLLENBERG AN, MONDEN T, MADURA JP, LEE K, AND WONDISFORD FE.
Function of nuclear co-repressor protein on thyroid hormone response elements is regulated by the receptor A/B domain. J Biol
Chem 271: 28516 –28520, 1996.
71. HORLEIN A, NAAR A, HEINZEL T, TORCHIA J, GLOSS B, KUROKAWA R,
RYAN A, KAMEI Y, SODERSTROM M, GLASS C, AND ROSENFELD M. Ligandindependent repression by the thyroid hormone receptor mediated
by a nuclear receptor co-repressor. Nature 377: 397– 404, 1995.
72. HYDER SM, CHIAPPETTA C, AND STANCEL GM. Interaction of human
estrogen receptors alpha and beta with the same naturally occurring estrogen response elements. Biochem Pharmacol 57: 597– 601,
1999.
73. IMPROTA-BREARS T, WHORTON AR, CODAZZI F, YORK JD, MEYER T, AND
MCDONNELL DP. Estrogen-induced activation of mitogen-activated
protein kinase requires mobilization of intracellular calcium. Proc
Natl Acad Sci USA 96: 4686 – 4691, 1999.
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
37.
VASUDEVAN, OGAWA, AND PFAFF
PATTERNS OF ER AND TR ACTIONS IN CNS
Physiol Rev • VOL
95.
96.
97.
98.
99.
100.
101.
102.
103.
104.
105.
106.
107.
108.
109.
110.
111.
112.
113.
114.
of a novel amino-terminal domain important for ligand-independent activation. J Biol Chem 272: 24927–24933, 1997.
LANNI A, DEFELICE M, LOMBARDI A, MORENO M, FLEURY C, RICQUIER D,
AND GOGLIA F. Induction of UCP2 mRNA by thyroid hormones in rat
heart. FEBS Lett 418: 171–174, 1997.
LARCHER A, NECULCEA J, BRETON C, ARSLAN A, ROZEN F, RUSSO C, AND
ZINGG H. Oxytocin receptor gene expression in the rat uterus during
pregnancy and the estrous cycle and in response to gonadal steroid
treatment. Endocrinology 136: 5350 –5356, 1995.
LAUBER AH, ROMANO GJ, MOBBS CV, HOWELLS RD, AND PFAFF DW.
Estradiol induction of proenkephalin messenger RNA in hypothalamus: dose-response and relation to reproductive behavior in the
female rat. Mol Brain Res 8: 47–54, 1990.
LAZAR M. Thyroid hormone receptors: multiple forms, multiple
possiblilites. Endocr Rev 14: 184 –193, 1993.
LEE SJ AND MCEWEN BS. Neurotrophic and neuroprotective actions
of estrogens and their therapeutic implications. Annu Rev Pharmacol Toxicol 41: 569 –591, 2001.
LEFSTIN JA AND YAMAMOTO KR. Allosteric effects of DNA on transcriptional regulators. Nature 392: 885– 888, 1998.
LEONARD JL, FARWELL AP, YEN PM, CHIN WW, AND STULA M. Differential expression of thyroid hormone receptor isoforms in neurons
and astroglial cells. Endocrinology 135: 548 –555, 1994.
LONGCOPE C, ABEND S, BRAVERMAN LE, AND EMERSON CH. Androstenedione and estrone dynamics in hypothyroid women. J Clin
Endocrinol Metab 70: 903–907, 1990.
LOPEZ GN, WEBB P, SHINSAKO JH, BAXTER JD, GREENE GL, AND KUSHNER PJ. Titration by estrogen receptor activation function-2 of
targets that are downstream from coactivators. Mol Endocrinol 13:
897–909, 1999.
LUBAHN DB, MOYER JS, GOLDING TS, COUSE JF, KORACH KS, AND
SMITHIES O. Alteration of reproductive function but not prenatal
sexual development after insertional disruption of the mouse estrogen receptor gene. Proc Natl Acad Sci USA 90: 11162–11166,
1993.
MANGELSDORF DJ, THUMMEL C, BEATO M, HERRLICH P, SCHULTZ G,
UMESENO K, BLUMBER B, KASTNER P, MARK M, CHAMBON P, AND EVANS
RM. Overview: the nuclear receptor superfamily: the second decade. Cell 83: 835– 840, 1995.
MASAKI T, YOSHIMATSU H, KAKUMA T, HIDAKA S, KUROKAWA M, AND
SAKATA T. Enhanced expression of uncoupling protein 2 gene in rat
white adipose tissue and skeletal muscle following chronic treatment with thyroid hormone. FEBS Lett 418: 323–326, 1997.
MCCARTHY MM, KLEOPOULOS SP, MOBBS CV, AND PFAFF DW. Infusion
of antisense oligodeoxynucleotides to the oxytocin receptor in the
ventromedial hypothalamus reduces estrogen induced sexual receptivity and oxytocin receptor binding in the female rat. Neuroendocrinology 59: 432– 440, 1994.
MCCARTHY MM, MCDONALD CH, BROOKS PJ, AND GOLDMAN D. An
anxiolytic action of oxytocin is enhanced by estrogen in the mouse.
Physiol Behav 60: 1209 –1215, 1996.
MCCARTHY MM, SCHWARTZ-GIBLIN S, AND WANG SM. Does estrogen
facilitate social behavior by reducing anxiety? Ann NY Acad Sci
807: 541–542, 1997.
MCINERNEY EM, WEIS KE, SUN J, MOSSELMAN S, AND KATZENELLENBOGEN BS. Transcription activation by the human estrogen receptor
subtype beta studied with ER beta and ER alpha receptor chimeras.
Endocrinology 139: 4513– 4522, 1998.
MILNE M, KANG MI, CARDONA G, QUAIL JM, BRAVERMAN LE, CHIN WW,
AND BARAN DT. Expression of multiple thyroid hormone receptor
isoforms in rat femoral and vertebral bone and in bone marrow
osteogenic cultures. J Cell Biochem 74: 684 – 693, 1999.
MOENTER SM, WOODFILL CJI, AND KARSCH FJ. Role of the thyroid
gland in seasonal reproduction: thyroidectomy blocks seasonal
suppression of reproductive neuroendocrine activity in ewes. Endocrinology 128: 1337–1344, 1991.
MONG J, KREBS CJ, AND PFAFF DW. Perspective: micoarrays and
differential display PCR-tools for studying transcript levels of
genes in neuroendocrine systems. Endocrinology 143: 2002–2006,
2002.
MORGAN MA, DELLOVADE TL, AND PFAFF DW. Effect of thyroid hormone administration on estrogen-induced sex behavior in female
mice. Horm Behav 37: 15–22, 2000.
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
74. JANSEN HT, LUBBERS LS, MACCHIA E, DEGROOT LJ, AND LEHMAN MN.
Thyroid hormone receptor (alpha) distribution in hamster and
sheep brain: colocalisation in gonadotropin-releasing hormone and
other identified neurons. Endocrinology 138: 5039 –5047, 1997.
75. JOHANSSON C, GOTHE S, FORREST D, VENNSTROM B, AND THOREN P.
Cardiovascular phenotype and temperature control in mice lacking
thyroid hormone receptor-beta or both alpha1 and beta. Am J
Physiol Heart Circ Physiol 276: H2006 –H2012, 1999.
76. JONES PS, PARROTT E, AND WHITE INH. Activation of transcription by
estrogen receptor alpha and beta is cell type and promoter dependent. J Biol Chem 274: 32008 –32014, 1999.
77. JOSHI J AND SABOL SL. Proenkephalin gene expression in C6 rat
glioma cells: potentiation of cyclic adenosine 3⬘,5⬘-monophosphate-dependent transcription by glucocorticoids. Mol Endocrinol
5: 1069 –1080, 1991.
78. KARSCH FJ, ROBINSON JE, WOODFILL CJI, AND BROWN MB. Circannual
cycles of luteinizing hormone and prolactin secrection in ewes
during prolonged exposure to a fixed photoperiod: evidence for an
endogeneous reproductive rhythm. Biol Reprod 41: 1034 –1046,
1989.
79. KELLY MJ AND LEVIN ER. Rapid actions of plasma membrane estrogen receptors. Trends Endocrinol Metab 12: 152–156, 2001.
80. KELLY MJ, MOSS RL, AND DUDLEY CA. Differential sensitivity of
preoptic-septal neurons to microelectrophoresed estrogen during
the estrous cycle. Brain Res 114: 152–157, 1976.
81. KELLY MJ, MOSS RL, AND DUDLEY CA. The effects of microelectrophoretically applied estrogen, cortisol and acetylcholine on medial
preoptic-septal unit activity throughout the estrous cycle of the
female rat. Exp Brain Res 30: 53– 64, 1977.
82. KELLY MJ, MOSS RL, DUDLEY CA, AND FAWCETT CP. The specificity of
the response of preoptic-septal area neurons to estrogen: 17␣
estradiol versus 17␤ estradiol and the response of extrahypothalamic neurons. Exp Brain Res 30: 43–52, 1977.
83. KIA HK, CHIN WW, KOIBUCHI N, AND PFAFF DW. Co-expression of
mRNAs for estrogen receptors (ER) a and b with thyroid hormone
receptors (TR) b1 mRNA in individual neurons in mouse forebrain.
Neuroendocrinology. In press.
84. KIA HK, KREBS CJ, KOIBUCHI N, CHIN WW, AND PFAFF DW. Coexpression of estrogen and thyroid hormone receptors in individual hypothalamic neurons. J Comp Neurol 437: 286 –295, 2001.
85. KIA HK, YUEN G, KREBS CJ, AND PFAFF DW. Colocalization of estrogen receptor alpha and NMDA-2D receptor mRNAs in amygdala
and hypothalamic nuclei of the mouse forebrain. Mol Brain Res
104: 47–54, 2002.
86. KIRBY JD, ARAMBEPOLA N, HEISKANEN P, KIRBY YK, RHOADS ML, NITTA
H, JETTON AE, IWAMOTO G, JACKSON GL, TUREK FW, AND COOKE PS.
Neonatal hypothyroidism permanently alters follicle stimulating
hormone and luteinizing hormone production in the male rat. Endocrinology 138: 2713–2721, 1997.
87. KIRKLAND J, GARDNER R, MUKKU V, AKHTAR M, AND STANCEL G. Hormonal control of uterine growth: the effect of hypothyroidism on
estrogen-stimulated cell division. Endocrinology 108: 2346 –2351,
1981.
88. KNOBIL E AND NEILL J. The Physiology of Reproduction (2nd ed.).
New York: Raven, 1994.
89. KNOBIL E AND NEILL J. Encyclopedia of Reproduction. San Diego,
CA: Academic, 1999.
90. KOIBUCHI N AND CHIN WW. Thyroid hormone action and brain development. Trends Endocrinol Metab 11: 123–128, 2000.
91. KOW L AND DWP. Mapping of neural and signal transduction pathways for lordosis in the search for estrogen actions on the central
nervous system. Behav Brain Res 92: 169 –180, 1998.
92. KUIPER GG, CARLSSON B, GRANDIEN K, ENMARK E, HAGGBLAD J, NILSSON
S, AND GUSTAFSSON JA. Comparison of the ligand binding specificity
and transcript tissue distribution of estrogen receptors alpha and
beta. Endocrinology 138: 863– 870, 1997.
93. KUROKAWA R, SODERSTROM M, HORLEIN A, HALACHMI S, BROWN M,
ROSENFELD MG, AND GLASS CK. Polarity-specific activities of retinoic
acid receptors determined by a co-repressor. Nature 377: 451– 454,
1995.
94. LANGLOIS MF, ZANGER K, MONDEN T, SAFER JD, HOLLENBERG AN, AND
WONDISFORD FE. A unique role of the beta-2 thyroid hormone receptor isoform in negative regulation by thyroid hormone. Mapping
941
942
VASUDEVAN, OGAWA, AND PFAFF
Physiol Rev • VOL
136.
137.
138.
139.
140.
141.
142.
143.
144.
145.
146.
147.
148.
149.
150.
151.
152.
153.
154.
of thyroid hormone actions in vivo. In: Molecular Endocrinology:
Basic Concepts and Clinical Correlations, edited by Weintraub B.
New York: Raven, 1995, p. 249 –268.
PACE P, TAYLOR J, SUNTHARALINGAM S, COOMBES CR, AND ALI S. Human
estrogen receptor beta binds DNA in a manner similar to and
dimerises with estrogen receptor alpha. J Biol Chem 272: 25832–
25838, 1997.
PAECH K, WEBB P, KUIPER GG, NILSSON S, GUSTAFSSON J, KUSHNER PJ,
AND SCANLAN TS. Differential ligand activation of estrogen receptors
ERalpha and ERbeta at AP1 sites. Science 277: 1508 –1510, 1997.
PARHAR IS, SOGA T, AND SAKUMA Y. Thyroid hormone and estrogen
regulate brain region specific messenger ribonucleic acids encoding three gonadotropin releasing hormone genes in sexually immature male fish, Oreochromis niloticus. Endocrinology 141: 1618 –
1626, 2000.
PARK IR, MOUNT DB, AND HIMMS-HAGEN J. Role of T3 in thermogenic
and trophic response of brown adipose tissue to cold. Am J Physiol
Endocrinol Metab 257: E81–E87, 1989.
PATRONE C, POLLIO G, VEGETO E, ENMARK E, DE CURTIS I, GUSTAFSSON
JA, AND MAGGI A. Estradiol induces differential neuronal phenotypes by activating estrogen receptor alpha or beta. Endocrinology
141: 1839 –1845, 2000.
PECZELY P. The role of thyroid and adrenal cortical hormones in the
modulation of the gonadal function in birds. Acta Biol Hung 36:
45–70, 1985.
PETERSEN CA, CALDWELL JD, PETERSON G, WALKER CH, AND MASON
GA. Oxytocin activation of maternal behavior in the rat. Ann NY
Acad Sci 652: 58 – 69, 1992.
PETTERSSON K, GRANDIEN K, KUIPER GG, AND GUSTAFSSON JA. Mouse
estrogen receptor beta forms estrogen response element-binding
heterodimers with estrogen receptor alpha. Mol Endocrinol 11:
1486 –1496, 1997.
PFAFF D. Autoradiographic localization of radioactivity in rat brain
after injection of tritiated sex hormones. Science 161: 1355–1356,
1968.
PFAFF D. Lordosis. In: Encyclopedia of Reproduction, edited by
Knobil E and Neill J. San Diego, CA: Academic, 1999, vol. 2, p.
1074 –1075.
PFAFF D AND KEINER M. Atlas of estradiol-concentrating cells in the
central nervous system of the female rat. J Comp Neurol 151:
121–158, 1973.
PFAFF D, VASUDEVAN N, KIA H, ZHU YS, CHAN J, GAREY J, MORGAN M,
AND OGAWA S. Estrogens, brain and behavior: studies in fundamental neurobiology and observations related to women’s health. J
Steroid Biochem 74: 365–373, 2000.
PFAFF DW. Estrogens and Brain Function: Neural Analysis of a
Hormone Controlled Mammalian Reproductive Behavior. New
York: Springer-Verlag, 1980.
PFAFF DW, KOW LM, ZHU YS, SCOTT REM, WU-PENG SX, AND DELLOVADE T. Hypothalamic cellular and molecular mechanisms helping
to satisfy axiomatic requirements for reproduction. J Neuroendocrinol 8: 325–336, 1996.
PFAFF DW, OGAWA S, KIA HK, VASUDEVAN N, KREBS C, FROHLICH J, AND
KOW LM. Genetic mechanisms in neural and hormonal controls
over female reproductive behaviors. In: Hormones, Brain and
Behavior, edited by Pfaff DW, Arnold A, Etgen A, Fahrbach S, and
Rubin R. San Diego, CA: Academic, 2002, vol. 3. In press.
PRIEST CA, ECKERSELL CB, AND MICEVYCH PE. Estrogen regulates
preproenkephalin-A mRNA levels in the rat ventromedial nucleus:
temporal and cellular aspects. Mol Brain Res 28: 251–262, 1995.
QUAEDACKERS ME, VAN DEN BRINK CE, WISSINK S, SCHREURS RHMM,
GUSTAFSSON JA, VAN DER SAAG PT, AND VAN DER BURG B. 4-Hydroxytamoxifen trans-represses nuclear factor-kappaB activity in
human osteoblastic U2-OS cells through estrogen receptor (ER)
alpha, and not through ERbeta. Endocrinology 142: 1156 –1166,
2001.
QUINONES-JENAB V, JENAB S, OGAWA S, ADAN RA, BURBACH JP, AND
PFAFF DW. Effects of estrogen on oxytocin receptor messenger
ribonucleic acid expression in the uterus, pituitary, and forebrain
of the female rat. Neuroendocrinology 65: 9 –17, 1997.
QUINONES-JENAB V, OGAWA S, JENAB S, AND PFAFF D. Estrogen regulation of preproenkephalin messenger RNA in the forebrain of
female mice. J Chem Neuroanat 12: 29 –36, 1996.
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
115. MORLEY P, WHITFILED JF, VANDERHYDEN BC, TSANG BK, AND SCHWARTZ
JL. A new, nongenomic estrogen action: the rapid release of intracellular calcium. Endocrinology 131: 1305–1312, 1992.
116. MURAMATSU M AND INOUE S. Estrogen receptors: how do they control
reproductive and non-reproductive functions? Biochem Biophys
Res Commun 270: 1–10, 2000.
117. MUSSO R, MAGGI A, AND EVA C. 17-␤ Estradiol stimulates mouse
neuropeptide Y-Y1 receptor gene transcription by binding to estrogen receptor alpha in neuroblastoma cells. Neuroendocrinology 72:
360 –367, 2000.
118. NEDERGAARD J, GOLOZOUBOVA V, MATTHIAS A, ASADI A, JACOBSSON A,
AND CANNON B. UCP1: the only protein able to mediate adaptive
non-shivering thermogenesis and metabolic inefficiency. Biochim
Biophys Acta 1504: 82–106, 2001.
119. NG L, HURLEY JB, DIERKS B, SRINIVAS M, SALTO C, VENNSTROM B, REH
TA, AND FORREST D. A thyroid hormone receptor that is required for
the development of green cone photoreceptors. Nature Genet 27:
94 –98, 2001.
120. NICHOLLS TJ, GOLDSMITH AR, AND DAWSON A. Photorefractoriness in
birds and comparison with mammals. Physiol Rev 68: 133–176,
1988.
121. NICOT A, OGAWA S, YB, CARR K, AND PFAFF DW. Effects of an
intrahypothalamic injection of antisense oligonucleotides for preproenkephalin mRNA in female rats: evidence for opioid involvement in lordosis reflex. Brain Res 777: 60 – 68, 1997.
122. NILSSON S, MAKELA S, TREUTER E, TUJAGUE M, THOMSEN J, ANDERSSON
G, ENMARK E, PETTERSSON K, WARNER M, AND GUSTAFSSON J. Mechanisms of estrogen action. Physiol Rev 81: 1535–1565, 2001.
123. NISHIMORI K, YOUNG LJ, GUO Q, WANG Z, INSEL TR, AND MATZUK MM.
Oxytocin is required for nursing but is not essential for parturition
or reproductive behavior. Proc Natl Acad Sci USA 93: 11699 –11704,
1996.
124. NOMURA M, DURBACK L, CHAN J, GUSTAFSSON JA, SMITHIES O, KORACH
KS, PFAFF DW, AND OGAWA S. Genotype/age interactions on aggressive behavior in gonadally intact estrogen receptor b knockout
(bERKO) male mice. Horm Behav 41: 288 –296, 2002.
125. OBERSTE-BERGHAUS C, ZANGER K, KH, COHEN RN, HOLLENBERG AN,
AND WONDISFORD FE. Thyroid hormone-independent interaction between the thyroid hormone receptor beta2 amino terminus and
coactivators. J Biol Chem 275: 1787–1792, 2000.
126. OGAWA S, CHAN J, CHESTER AE, GUSTAFSSON JA, KORACH KS, AND
PFAFF DW. Survival of reproductive behaviors in estrogen receptor
beta gene-deficient (betaERKO) male and female mice. Proc Natl
Acad Sci USA 96: 12887–12892, 1999.
127. OGAWA S, CHAN J, KORACH KS, AND PFAFF DW. Estrogen facilitates
running wheel activity via estrogen receptor-a. Soc Neurosci Abstr
27: 534.14, 2001.
128. OGAWA S, ENG V, TAYLOR J, LUBAHN DB, KORACH KS, AND PFAFF DW.
Roles of estrogen receptor-alpha gene expression in reproductionrelated behaviors in female mice. Endocrinology 139: 5070 –5081,
1998.
129. OGAWA S, INOUE S, WATANABE T, HIROI H, ORIMO A, HOSOI T, OUCHI Y,
AND MURAMATSU M. The complete primary structure of human estrogen receptor beta (hER beta) and its heterodimerization with
ER alpha in vivo and in vitro. Biochem Biophys Res Commun 243:
122–126, 1998.
130. OGAWA S, KORACH KS, GUSTAFSSON J, AND PFAFF DW. Survival of
reproductive behaviors in estrogen receptor b gene deficient
(bERKO) male mice. Soc Neurosci Abstr 245.15, 1999.
131. OGAWA S, LUBAHN DB, KORACH KS, AND PFAFF DW. Behavioral effects
of estrogen receptor gene disruption in male mice. Proc Natl Acad
Sci USA 94: 1476 –1481, 1997.
132. OGAWA S, TAYLOR J, LUBAHN DB, KORACH KS, AND PFAFF DW. Reversal
of sex roles in genetic female mice by disruption of estrogen
receptor gene. Neuroendocrinology 64: 467– 470, 1996.
133. OGAWA S, WASHBURN T, TAYLOR J, LUBAHN D, KORACH K, AND PFAFF D.
Modifications of testosterone-dependent behaviors by estrogen receptor-alpha gene disruption in male mice. Endocrinology 139:
5058 –5069, 1998.
134. OPPENHEIMER JH AND SCHWARTZ HL. Molecular basis of thyroid hormone-dependent brain development. Endocr Rev 18: 462– 475,
1997.
135. OPPENHEIMER JH, SCHWARTZ HL, AND STRAIT KA. An integrated view
PATTERNS OF ER AND TR ACTIONS IN CNS
Physiol Rev • VOL
175.
176.
177.
178.
179.
180.
181.
182.
183.
184.
185.
186.
187.
188.
189.
190.
191.
192.
193.
194.
of the estrogen receptor-alpha knockout mouse. Endocrinology
138: 5649 –5652, 1997.
SHUGHRUE PJ, LANE MV, AND MERCHENTHALER I. Comparative distribution of estrogen receptor-alpha and -beta mRNA in the rat central
nervous system. J Comp Neurol 388: 507–525, 1997.
SHUGHRUE PJ AND MERCHENTHALER I. Estrogen is more than just a
“sex hormone”: novel sites for estrogen action in the hippocampus
and cerebral cortex. Front Neuroendocrinol 1: 95–101, 2000.
SINCHAK K, ECKERSELL C, QUEZADA V, NORELL A, AND MICEVYCH P.
Preproenkephalin mRNA levels are regulated by acute stress and
estrogen stimulation. Physiol Behav 69: 425– 432, 2000.
SPENCER TE, JENSTER G, BURCIN MM, ALLIS CD, ZHOU J, MIZZEN CA,
MCKENNA NJ, ONATE SA, TSAI SY, TSAI MJ, AND O’MALLEY BW. Steroid
receptor coactivator-1 is a histone acetyltransferase. Nature 389:
194 –198, 1997.
STEFANO GB, PREVOT V, BEAUVILLAIN JC, FIMIANI C, WELTERS I, CADET
P, BRETON C, PESTEL J, SALZET M, AND BILFINGER TV. Estradiol
coupling to human monocyte nitric oxide release is dependent on
intracellular calcium transients: evidence for an estrogen surface
receptor. J Immunol 163: 3758 –3763, 1999.
SUBRAMANIAM N, TREUTER E, AND OKRET S. Receptor interacting
protein RIP140 inhibits both positive and negative gene regulation
by glucocorticoids. J Biol Chem 274: 18121–18127, 1999.
SYLVIA VL, WALTON J, LOPEZ D, DEAN DD, BOYAN BD, AND SCHWARTZ Z.
17Beta estradiol-BSA conjugates and 17beta estradiol regulate
growth plate chondrocytes by common membrane associated
mechanisms involving PKC dependent and independent signal
transduction. J Cell Biochem 81: 413– 429, 2001.
TAGAMI T, KOPP P, JOHNSON W, ARSEVEN OK, AND JAMESON LJ. The
thyroid hormone receptor variant alpha2 is a weak antagonist
because it is deficient in interactions with nuclear receptor corepressors. Endocrinology 139: 2535–2544, 1998.
TATUM EL AND BEADLE GW. Genetic control of biochemical reactions in Neurospora: an “aminobenzoicless” mutant. Proc Natl
Acad Sci USA 28: 234 –243, 1942.
THRUN LA, DAHL GE, EVANS NP, AND KARSCH FJ. A critical period for
thyroid hormone action on seasonal changes in reproductive neuroendocrine function in the ewe. Endocrinology 138: 3402–3409,
1997.
THRUN LA, DAHL GE, EVANS NP, AND KARSCH FJ. Effect of thyroidectomy on maintenance of seasonal reproductive suppression in
the ewe. Biol Reprod 56: 1035–1040, 1997.
TREMBLAY GB, TREMBLAY A, COPELAND NG, GILBERT DJ, JENKINS NA,
LABRIE F, AND GIGUERE V. Cloning, chromosomal localization, and
functional analysis of the murine estrogen receptor beta. Mol Endocrinol 11: 353–365, 1997.
TREMBLAY RR, BRAITHWAITE S, HO-KIM MA, AND DUBE JY. Effect of
thyroid state on estradiol-17beta metabolism in the rabbit. Steroids
29: 649 – 656, 1977.
TRIBOLLET E, AUDIGIER S, DUBOIS-DAUPHIN M, AND DREIFUSS JJ. Gonadal steroids regulate oxytocin receptors but not vasopressin
receptors in the brain of male and female rats. An autoradiographical study. Brain Res 511: 129 –140, 1990.
VASUDEVAN N, DAVIDKOVA G, ZHU YS, KOIBUCHI N, CHIN WW, AND
PFAFF DW. Differential interaction of estrogen receptor and thyroid
hormone receptor isoforms on the rat oxytocin receptor promoter
leads to differences in transcriptional regulation. Neuroendocrinology 74: 309 –324, 2001.
VASUDEVAN N, KOIBUCHI N, CHIN WW, AND PFAFF DW. Differential
crosstalk between estrogen receptor (ER) alpha and ER beta and
the thyroid hormone receptor isoforms results in flexible regulation of the consensus ERE. Mol Brain Res 95: 9 –17, 2001.
VASUDEVAN N, KOW LM, AND PFAFF DW. Early membrane estrogenic
effects required for full expression of slower genomic actions in a
nerve cell line. Proc Natl Acad Sci USA 98: 12267–12271, 2001.
VASUDEVAN N, ZHU YS, DANIEL S, KOIBUCHI N, CHIN WW, AND PFAFF
DW. Crosstalk between estrogen receptors and thyroid hormone
receptor isoforms results in differential regulation of the preproenkephalin gene. J Neuroendocrinology 13: 229 –790, 2001.
VEGETO E, WAGNER BL, IMHOF MO, AND MCDONNELL DP. The molecular pharmacology of ovarian steroid receptors. Vitam Horm 52:
99 –128, 1996.
VIGUIE C, BATTAGLIA DF, KRASA HB, THRUN LA, AND KARSCH FJ.
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
155. RABELO EM, BAKER BS, AND TATA JR. Interplay between thyroid
hormone and estrogen in modulating expression of their receptor
and vitellogenin genes during Xenopus metamorphosis. Mech Dev
45: 49 –57, 1994.
156. RABELO EM AND TATA JR. Thyroid hormone potentiates estrogen
activation of vitellogenin genes and autoinduction of estrogen receptor in adult Xenopus hepatocytes. Mol Cell Endocrinol 96:
37– 44, 1993.
157. RABELO R, REYES C, SCHIFMAN A, AND SILVA JE. Interactions among
receptors, thyroid hormone response elements and ligands in the
regulation of the rat uncoupling protein gene expression by thyroid
hormone. Endocrinology 137: 3478 –3487, 1996.
158. RAJU TN. The Nobel chronicles 1958: George Wells Beadle (1903–
89), Edward Lawrie Tatum (1909 –75) and Joshua Lederberg (b
1925). Lancet 353: 2082, 1999.
159. RAZANDI M, PEDRAM A, GREENE GL, AND LEVIN ER. Cell membrane
and nuclear estrogen receptor (ERs) originate from a single transcript: studies of ERalpha and ERbeta expressed in Chinese hamster ovary cells. Mol Endocrinol 13: 307–319, 1999.
160. REHNMARK S, BIANCO AC, KIEFFER JD, AND SILVA JE. Transcriptional
and posttranscriptional mechanisms in uncoupling protein mRNA
response to cold. Am J Physiol Endocrinol Metab 262: E58 –E67,
1992.
161. REITER RJ, KLAUS S, EBBINGHAUS C, HELDMAIER G, REDLIN U, RICQUIER
D, VAUGHAN MK, AND STEINLECHNER S. Inhibition of 5⬘-deiodination
of thyroxine suppresses the cold-induced increase in brown adipose tissue messenger ribonucleic acid for mitochondrial uncoupling protein without influencing lipoprotein lipase activity. Endocrinology 126: 2550 –2554, 1990.
162. RIA LE AND GONZALEZ-BARROSO MM. Physiological regulation of the
transport activity in the uncoupling proteins UCP1 and UCP2.
Biochim Biophys Acta 1504: 70 – 81, 2001.
163. RICQUIER D AND BOUILLAUD F. Mitochondrial uncoupling proteins:
from mitochondria to the regulation of energy balance. J Physiol
529: 3–10, 2000.
164. RIDGWAY EC, LONGCOPE C, AND MALOOF F. Metabolic clearance and
blood production rates of estradiol in hyperthyroidism. J Clin
Endocrinol Metab 41: 491– 497, 1975.
165. ROBINSON GA AND GIBBINS AM. Increased clearance rate of plasma
thyroxine accompanies estradiol-induced vitellogenesis in male
Japanese quail. Poult Sci 64: 2397–2399, 1985.
166. ROBINSON JE AND KARSCH FJ. Refractoriness to inductive day lengths
terminates the breeding season of the Suffolk ewe. Biol Reprod 3:
656 – 663, 1984.
167. ROBINSON JE AND KARSCH FJ. Timing of the breeding season of the
ewe: what is the role of daylength? Reprod Nutr Dev 28: 365–374,
1988.
168. ROMANO GJ, HARLAN RE, SHIVERS BD, HOWELLS RD, AND PFAFF DW.
Estrogen increases proenkephalin messenger ribonucleic acid levels in the ventromedial hypothalamus of the rat. Mol Endocrinol 2:
1320 –1328, 1988.
169. ROMANO GJ, MOBBS CV, HOWELLS RD, AND PFAFF DW. Estrogen
regulation of proenkephalin gene expression in the ventromedial
hypothalamus of the rat: temporal qualities and synergism with
progesterone. Mol Brain Res 5: 51–58, 1989.
170. ROSANO GM AND PANINA G. Oestrogens and the heart. Therapie 54:
381–385, 1999.
171. SAFER JD, LANGLOIS MF, COHEN R, MONDEN T, JOHN-HOPE D, JM,
HOLLENBERG AN, AND WONDISFORD FE. Isoform variable action
among thyroid hormone receptor mutants provides insight into
pituitary resistance to thyroid hormone. Mol Endocrinol 11: 16 –26,
1997.
172. SCHWARTZ HL. Effect of thyroid hormone in growth and development. In: Molecular Basis of Thyroid Hormone Action, edited by
Oppenheimer JH and Samuels HH. New York: Academic, 1983, p.
413– 444.
173. SCOTT RE, WU-PENG XS, YEN PM, CHIN WW, AND PFAFF DW. Interactions of estrogen- and thyroid hormone receptors on a progesterone receptor estrogen response element (ERE) sequence: a
comparison with the vitellogenin A2 consensus ERE. Mol Endocrinol 11: 1581–1592, 1997.
174. SHUGHRUE P, SCRIMO P, LANE M, ASKEW R, AND MERCHENTHALER I. The
distribution of estrogen receptor-beta mRNA in forebrain regions
943
944
195.
196.
197.
198.
200.
201.
202.
203.
204.
205.
206.
Thyroid hormones act primarily within the brain to promote the
seasonal inhibition of luteinizing hormone secretion in the ewe.
Endocrinology 140: 1111–1117, 1999.
WATANABE T, INOUE S, OGAWA S, ISHII Y, HIROI H, IKEDA K, ORIMO A,
AND MURAMATSU M. Agonistic effect of tamoxifen is dependent on
cell type, ERE-promoter context, and estrogen receptor subtype:
functional difference between estrogen receptors alpha and beta.
Biochem Biophys Res Commun 236: 140 –145, 1997.
WATTERS JJ, CAMPBELL JS, CUNNINGHAM MJ, KREBS EG, AND DORSA
DM. Rapid membrane effects of steroids in neuroblastoma cells:
effects of estrogen on mitogen activated protein kinase signaling
cascade and c-fos immediate early gene transcription. Endocrinology 138: 4030 – 4033, 1997.
WEBER GM, OKIMOTO DK, RICHMAN NH, AND GRAU EG. Patterns of
thyroxine and triiodothyronine in serum and follicle-bound oocytes
of the tilapia, Oreochromis mossambicus, during oogenesis. Gen
Comp Endocrinol 85: 392– 404, 1992.
WEBSTER JR, MOENTER SJ, BARRELL GK, LEHMAN MN, AND KARSCH JF.
Role of the thyroid gland in seasonal reproduction. III. Thyroidectomy blocks seasonal suppression of gonadotropin-releasing hormone secretion in sheep. Endocrinology 129: 1635–1643, 1991.
WEBSTER JR, MOENTER SM, WOODFILL CJI, AND KARSCH JF. Role of the
thyroid gland in seasonal reproduction. II. Thyroxine allows a
season-specific suppression of gonadotropin secretion in sheep.
Endocrinology 129: 176 –183, 1991.
WEN DX, XU YF, MAIS DE, GOLDMAN ME, AND MCDONNELL DP. The A
and B isoforms of the human progesterone receptor operate
through distinct signaling pathways within target cells. Mol Cell
Biol 8356 – 8364, 1994.
WIESELTHIER AS AND VAN TIENHOVEN A. The effect of thyroidectomy
on testicular size and on the protorefractory period in the starling
(Sturnus vulgaris L). J Exp Zool 179: 331–338, 1972.
WIKSTROM L, JOHANSSON C, SALTO C, BARLOW C, CAMPOS BARROS A,
BAAS F, FORREST D, THOREN P, AND VENNSTROM B. Abnormal heart
rate and body temperature in mice lacking thyroid hormone receptor ␣1. EMBO J 17: 455– 461, 1998.
WILSON FW AND REINERT BD. Thyroid hormone acts centrally to
programme photostimulated male american tree sparrows (Spizella arborea) for vernal and autumnal components of seasonality.
J Neuroendocrinol 12: 87–95, 2000.
WISE PM, DUBAL DB, WILSON ME, RAU SW, AND LIU Y. Estrogens:
trophic and protective factors in the adult brain. Front Neuroendocrinol 22: 33– 66, 2001.
WOOD JR, LIKHITE VS, LOVEN MA, AND NARDULLI AM. Allosteric modulation of estrogen receptor conformation by different estrogen
response elements. Mol Endocrinol 15: 1114 –1126, 2001.
YANG Z AND PRIVALSKY ML. Isoform-specific transcriptional regulation by thyroid hormone receptors: hormone independent activa-
Physiol Rev • VOL
207.
208.
209.
210.
211.
212.
213.
214.
215.
216.
217.
218.
219.
220.
tion operates through a steroid receptor mode of coactivator interaction. Mol Endocrinol 5: 1170 –1185, 2001.
YARWOOD NJ, GURR JA, SHEPPARD MC, AND FRANKLYN JA. Estradiol
modulates thyroid hormone regulation of the human glycoprotein
hormone alpha subunit gene. J Biol Chem 268: 21984 –21989, 1993.
YEN PM. Physiological and molecular basis of thyroid hormone
action. Physiol Rev 81: 1097–1142, 2001.
YEN PM, IKEDA M, WILCOX EC, BRUBAKER JH, SPANJAARD RA, SUGAWARA A, AND CHIN WW. Half-site arrangement of hybrid glucocorticoid and thyroid hormone response elements specifies thyroid
hormone receptor complex binding to DNA and transcriptional
activity. J Biol Chem 269: 12704 –12709, 1994.
YEN PM, WILCOX EC, AND CHIN WW. Steroid hormone receptors
selectively affect transcriptional activation but not basal repression
by thyroid hormone receptors. Endocrinology 136: 440 – 445, 1995.
YOUNG JK. Thyroxine treatment reduces the anorectic effect of
estradiol in rats. Behav Neurosci 100: 284 –287, 1986.
YOUNG LJ, WANG Z, DONALDSON R, AND RISSMAN EF. Estrogen receptor alpha is essential for induction of oxytocin receptor by estrogen. Neuroreport 9: 933–936, 1998.
ZHANG J AND LAZAR MA. The mechanism of action of thyroid hormones. Annu Rev Physiol 62: 439 – 466, 2000.
ZHANG X, JEYAKUMAR M, AND BAGCHI MK. Ligand-dependent crosstalk between steroid and thyroid receptors. Evidence for common
transcriptional coactivator(s). J Biol Chem 271: 14825–14833, 1996.
ZHU XG, MCPHIE P, LIN KH, AND CHENG SY. The differential hormone
dependent transcriptional activation of thyroid hormone receptor
isoforms is mediated by interplay of their domains. J Biol Chem
272: 9048 –9054, 1997.
ZHU YS, DELLOVADE TL, AND PFAFF DW. Gender-specific induction of
RNA synthesis in rat pituitary by estrogen and its interaction with
thyroid hormones. J Neuroendocrinol 9: 395– 403, 1997.
ZHU YS, DELLOVADE TL, AND PFAFF DW. Interactions between hormonal and environmental signals on hypothalamic neurons: molecular mechanisms signaling environmental events. Trends Endocrinol Metab 8: 111–115, 1997.
ZHU YS, YEN PM, CHIN WW, AND PFAFF DW. Estrogen and thyroid
hormone interaction on regulation of gene expression. Proc Natl
Acad Sci USA 93: 12587–12592, 1996.
ZOELLER RT, KABEER N, AND ALBERS HE. Cold exposure elevates
cellular levels of messenger ribonucleic acid encoding thyrotropinreleasing hormone in paraventricular nucleus despite elevated levels of thyroid hormone. Endocrinology 127: 2955–2962, 1990.
ZOU A, MARSCHKE KB, ARNOLD KE, BERGER EM, FITZGERALD P, MAIS
DE, AND ALLEGRETTO EA. Estrogen receptor beta activates the human retinoic acid receptor alpha-1 promoter in response to tamoxifen and other estrogen receptor antagonists, but not in response to
estrogen. Mol Endocrinol 13: 418 – 430, 1999.
82 • OCTOBER 2002 •
www.prv.org
Downloaded from http://physrev.physiology.org/ by 10.220.33.4 on July 4, 2017
199.
VASUDEVAN, OGAWA, AND PFAFF