Anda di halaman 1dari 11

Ligands That Act through Nuclear Receptors,  52

Nuclear Receptor Signaling Mechanisms,  54


Receptor Regulation of Gene Transcription,  57

CHAPTER
CHAPTER 4 
Mechanism of Action of Hormones
That Act on Nuclear Receptors
MITCHELL A. LAZAR

Hormones can be divided into two groups on the basis of Numerous other signaling molecules share with thyroid
where they function in a target cell. The first group includes and steroid hormones the ability to function in the nucleus
hormones that do not enter cells; instead, they signal by to convey intercellular and environmental signals. Not all
means of second messengers generated by interactions of these molecules are produced in glandular tissues.
with receptors at the cell surface. All polypeptide hormones Although some signaling molecules, such as classic endo­
(e.g., growth hormone), monoamines (e.g., serotonin), and crine hormones, arrive at target tissues through the blood­
prostaglandins (e.g., prostaglandin E2), use cell surface stream, others have paracrine functions (i.e., acting on
receptors (see Chapter 5). The second group, the focus of adjacent cells) or autocrine functions (i.e., acting on the
this chapter, includes hormones that can enter cells. These cell of origin).
hormones bind to intracellular receptors that function in In addition to the classic steroid and thyroid hormones,
the nucleus of the target cell to regulate gene expression. lipophilic signaling molecules that use nuclear receptors
Classic hormones that use intracellular receptors include include derivatives of vitamins A and D, endogenous
thyroid and steroid hormones. metabolites such as oxysterols and bile acids, and non-
Hormones serve as a major form of communication natural chemicals encountered in the environment (i.e.,
between different organs and tissues. They allow special­ xenobiotics). These molecules are referred to as nuclear
ized cells in complex organisms to respond in a coor­ receptor ligands. The nuclear receptors for all of these signal­
dinated manner to changes in the internal and external ing molecules are structurally related and are collectively
environments. Classic endocrine hormones are secreted by referred to as the nuclear receptor superfamily.1-3 The
endocrine glands and are distributed throughout the body study of these receptors is a rapidly evolving field, and
through the bloodstream. These hormones were discovered more detailed information can be obtained by visiting
by purifying the biologically active substances from clearly the Nuclear Receptor Signaling Atlas web site (http://
definable glands. www.nursa.org [accessed September 2010]).
51
52   Mechanism of Action of Hormones That Act on Nuclear Receptors

TABLE 4-1 
LIGANDS THAT ACT THROUGH
Nuclear Receptor Ligands and Their Receptors
NUCLEAR RECEPTORS
Ligand Receptor
General Features of Nuclear Classic Hormones
Receptor Ligands Thyroid hormone Thyroid hormone receptor (TR), subtypes
Unlike polypeptide hormones that function through cell α, β
surface receptors, no ligands for nuclear receptors are Estrogen Estrogen receptor (ER), subtypes α, β
directly encoded in the genome. All nuclear receptor Testosterone Androgen receptor (AR)
ligands are small (molecular mass <1000 d) and lipophilic, Progesterone Progesterone receptor (PR)
enabling them to enter cells by passive diffusion, although Aldosterone Mineralocorticoid receptor (MR)
in some cases, a membrane transport protein is involved. Cortisol Glucocorticoid receptor (GR)
For example, several active and specific thyroid hormone Vitamins
transporters have been identified, including monocarbox­
ylate transporter 8 (MCT8), MCT10, and organic anion 1,25-(OH)2-Vitamin D3 Vitamin D receptor (VDR)
transporting polypeptide 1C1 (OATP1C1).4,5 The lipophi­ All-trans-retinoic acid Retinoic acid receptor, subtypes α, β, γ
licity of nuclear receptor ligands allows them to be absorbed 9-cis-Retinoic acid Retinoid X receptor (RXR), subtypes α, β, γ
from the gastrointestinal tract, facilitating their use in Metabolic Intermediates and Products
replacement or pharmacologic therapies for various disease
Fatty acids Peroxisome proliferator-activated receptor
states.
(PPAR), subtypes α, δ, γ
Another common feature of naturally occurring nuclear
Oxysterols Liver X receptor (LXR), subtypes α, β
receptor ligands is that all are derived from dietary, envi­
Bile acids Bile acid receptor (BAR)
ronmental, or metabolic precursors. In this sense, the func­
Heme Rev-Erb subtypes α, β
tion of these ligands and their receptors is to translate cues
from the external and internal environments into changes Xenobiotics
in gene expression. Their critical role in maintaining ?? Pregnane X receptor (PXR), constitutive
homeostasis in multicellular organisms is highlighted by androstane receptor (CAR)
the fact that nuclear receptors are found in all vertebrates
and insects but not in single-cell organisms such as yeast.6
Because nuclear receptor ligands are lipophilic, most are
readily absorbed from the gastrointestinal tract. This makes Precursors of vitamin D are synthesized and stored
nuclear receptors excellent targets for pharmaceutical in skin and activated by ultraviolet light; vitamin D can
interventions. In addition to natural ligands, many drugs also be derived from dietary sources. Vitamin D is then
in clinical use target nuclear receptors, ranging from those converted in the liver to 25(OH)D (25-hydroxyvitamin D,
used to treat specific hormone deficiencies to those used to calcidiol) and in the kidney to 1,25(OH)2 D3 (1,25-
treat common multigenic conditions such as inflamma­ dihydroxyvitamin D3, calcitriol), the most potent natural
tion, cancer, and type 2 diabetes. ligand of the vitamin D receptor (VDR). The 1-hydroxylation
of calcidiol is tightly regulated, and calcitriol acts as a cir­
culating hormone.
Subclasses of Nuclear Receptor Ligands Vitamin A is stored in the liver and is activated by
One classification of nuclear receptor ligands is outlined in metabolism to all-trans-retinoic acid, which is a high-
Table 4-1 and is described in the following paragraphs. affinity ligand for retinoic acid receptors (RARs).9 Retinoic
acid is likely to function as a signaling molecule in para­
Classic Hormones crine as well as endocrine pathways. Retinoic acid is also
The classic hormones that use nuclear receptors for signal­ converted to its 9-cis-isomer, which is a ligand for another
ing are thyroid hormone and steroid hormones. Steroid nuclear receptor, the retinoid X receptor (RXR).10 These
hormones include cortisol, aldosterone, estradiol, proges­ retinoids and their receptors are essential for normal devel­
terone, and testosterone. In some cases (e.g., thyroid opment of multiple organs and tissues, and they have
hormone receptor α and β genes [THRA and THRB], estro­ pharmaceutical utility for conditions ranging from skin
gen receptor α and β genes [ESR1 and ESR2]), there are diseases to leukemia.10
multiple receptor genes that encode multiple receptors.
Multiple receptors for the same hormone can also be Metabolic Intermediates and Products
derived from a single gene by alternative promoter usage Certain nuclear receptors respond to naturally occurring
or by alternative splicing (e.g., THRB1 and THRB2). endogenous metabolic products. The peroxisome
Some receptors can mediate the signals of multiple hor­ proliferator-activated receptors (PPARs) constitute the
mones. For example, the mineralocorticoid receptor, also best defined subfamily of metabolite-sensing nuclear
known as the aldosterone receptor or nuclear receptor 3C2 receptors.11 All three PPAR subtypes are activated by
(NR3C2), has equal affinity for cortisol7 and probably func­ polyunsaturated fatty acids. No single fatty acid has par­
tions as a glucocorticoid receptor in some tissues, such as the ticularly high affinity for any PPAR, and it is possible that
brain; likewise, the androgen receptor binds and responds these receptors function as integrators of the concentration
to both testosterone and dihydrotestosterone (DHT).8 of a number of fatty acids.
PPARα is expressed primarily in liver; the natural ligand
Vitamins with highest affinity for PPARα is an eicosanoid, 8(S)-
Vitamins are essential constituents of a healthful diet. Two hydroxyeicosatetraenoic acid,12,13 although there is evi­
fat-soluble vitamins, A and D, are precursors of important dence that the natural ligand may be a fatty acid derived
signaling molecules that function as ligands for nuclear from lipolysis of circulating triglyceride-rich lipoproteins.14
receptors. The fibrate class of lipid-lowering pharmaceuticals are
Mechanism of Action of Hormones That Act on Nuclear Receptors   53

potent ligands for PPARα, and the very name of this recep­ to respond to metabolites and environmental compounds
tor is derived from its ability to induce the proliferation of were originally discovered as orphans. Therefore, future
peroxisomes in the liver.15 Indeed, stimulation of fatty acid research will likely find that additional orphan receptors
oxidation is an important physiologic role of PPARα. function as receptors for physiologic, pharmacologic, or
The other PPARs (δ and γ) are structurally related but are environmental ligands. For example, the nuclear receptor
not activated by peroxisome proliferators. PPARδ, also NR1D1 (formerly called Rev-Erbα), which is a regulator of
known as PPARβ, is ubiquitous, and its ligands—other than circadian rhythms,24 has been shown to be a receptor for
fatty acids—are not well characterized. Activation of PPARδ molecular heme,25,26 which is involved in the coordination
appears to increase oxidative metabolism in fat and skeletal of circadian rhythms and metabolism.
muscle.16 PPARγ is expressed primarily in adipocytes and is
necessary for differentiation along the adipocyte lineage.17
PPARγ is also expressed in other cell types, including
Variant Receptors
colonocytes, macrophages, and vascular endothelial The carboxyl-terminus of the nuclear receptors is respon­
cells, where it may play physiologic and pathologic roles. sible for hormone binding. In a few nuclear receptors,
The natural ligand for PPARγ is not known, although pros­ including THRA and the glucocorticoid receptor, alterna­
taglandin J derivatives have been shown to bind and acti­ tive splicing produces variant receptors with unique
vate the receptor at concentrations in the micromolar C-termini that do not bind ligand.27,28 These variant recep­
range.13,18,19 PPARγ appears to be the target of thiazolidine­ tors are normally expressed, but their biologic relevance is
dione (TZD) antidiabetic drugs that improve insulin sen­ uncertain. They may modulate the action of the classic
sitivity.17,20 These pharmaceutical agents bind to PPARγ receptor to which they are related by inhibiting its
with nano­molar affinities, and non-TZD PPARγ ligands are function.
also insulin sensitizers, further implicating PPARγ in this Another type of normally occurring variant nuclear
physio­logic role. receptors lacks a classic DNA-binding domain (discussed
Another metabolite-responsive nuclear receptor, the later). These include NROB1 (formerly called DAX1), which
liver X receptor (LXR), is activated by oxysterol intermedi­ is mutated in human disease,29 and PTPN6 (formerly called
ates in cholesterol biosynthesis. Mice lacking LXRα have a SHP1).30 Their ligands have not been identified, and it is
dramatically impaired ability to metabolize cholesterol.21 A likely that NROB1 and PTPN6 bind to and repress the
receptor known as farnesyl X receptor (FXR) binds and is actions of other receptors.
activated by bile acids, and it likely plays a role in regula­ Rare, naturally occurring mutations of hormone recep­
tion of bile synthesis and circulation in normal conditions tors can cause hormone resistance in affected patients.31
and in disease states.22 Inheritance of the hormone resistance phenotype is domi­
nant if the mutant receptor inhibits the action of the
Xenobiotics normal receptor, as occurs with resistance to thyroid
Other nuclear receptors appear to function as integrators hormone or PPARγ ligands.31 Inheritance is recessive if the
of exogenous environmental signals, including natural mutation results in a complete loss of receptor function, as
endobiotics (e.g., medicinal agents and toxins found in with the syndrome of hereditary calcitriol-resistant rickets.32
plants) and xenobiotics (i.e., compounds that are not natu­ Inheritance can also be X-linked, as with the mutated
rally occurring). In these cases, the role of the activated androgen receptor in androgen insensitivity syndromes,
nuclear receptor is to induce cytochrome P450 enzymes including testicular feminization.33
that facilitate detoxification of potentially dangerous com­
pounds in the liver. Receptors in this class include sterol
and xenobiotic receptor (SXR), also known as pregnane
Regulation of Ligand Levels
X receptor (PXR), and constitutive androstane receptor Ligand levels can be regulated in several ways (Table 4-2).
(CAR).22 A dietary precursor may not be available in required
Unlike other nuclear receptors that have high affinity amounts (e.g., in hypothyroidism due to iodine deficiency).
for very specific ligands, xenobiotic receptors have low Pituitary hormones (e.g., thyroid-stimulating hormone)
affinity for a large number of ligands, reflecting their func­ regulate the synthesis and secretion of classic thyroid and
tion in defense against a varied and challenging environ­ steroid hormones. If the glands that synthesize these hor­
ment. Although these xenobiotic compounds are not mones fail, hormone deficiency can occur.
hormones in the classic sense, the function of these nuclear Many of the nuclear receptor ligands are enzymatically
receptors is consistent with the general theme of helping converted from inactive prohormones to biologically active
the organism to cope with environmental challenges. hormones; examples include the 5′ deiodination of thyrox­
ine (T4) to triiodothyronine (T3) (see Chapter 11). In other
cases, one hormone is a precursor for another, for example,
Orphan Receptors in aromatization of testosterone to estradiol. Biotransfor­
The nuclear receptor superfamily is one of the largest fami­ mation may occur in a specific tissue that is not the main
lies of transcription factors. The hormones and vitamins target of the hormone, as with renal 1-hydroxylation of
just described account for the functions of only a fraction
of the total number of nuclear receptors. The remaining
TABLE 4-2 
receptors have been designated as orphan receptors because
their putative ligands are not known.23 Mechanisms Regulating Ligand Levels
From analyses of mice and humans with mutations in Precursor availability
various orphan receptors, it is clear that many of them are Synthesis
required for life or for development of specific organs, Secretion
ranging from brain nuclei to endocrine glands. Some Activation (prohormone → active hormone)
orphan receptors appear to be active in the absence of any Deactivation (active hormone) → inactive hormone)
ligand (i.e., constitutively active) and may not respond to Elimination (hepatic, renal clearance)
a natural ligand. Nevertheless, all of the receptors known
54   Mechanism of Action of Hormones That Act on Nuclear Receptors

vitamin D (see Chapter 28), or it may occur primarily in Prohormone


target tissues (e.g., 5α-reduction of testosterone to DHT; see (ligand precursor)
Chapter 19). Deficiency or pharmacologic inhibition of an
Hormone
enzyme can also reduce hormone levels.34 (ligand)
Hormones can be inactivated by standard hepatic or
renal clearance mechanisms or by more specific enzymatic
processes. In the latter case, reduction in enzyme activity
due to gene mutations or pharmacologic agents can result
Phenotypic change
in symptoms of hormone excess, such as renal deactivation
(hormone effect)
of cortisol by 11β-hydroxysteroid dehydrogenase (11β-
HSD). Because cortisol can activate the mineralocorticoid
receptor, insufficient 11β-HSD activity due to licorice inges­
tion, gene mutation, or extremely high cortisol levels
causes syndromes of apparent mineralocorticoid excess.35 Transcription mRNA Protein

HRE
NUCLEAR RECEPTOR SIGNALING Hormone-responsive gene
MECHANISMS
Nucleus
Nuclear receptors are multifunctional proteins that trans­
duce the signals of their cognate ligands. General features
of nuclear receptor signaling are illustrated in Figure 4-1.
For hormone action in the nucleus, the ligand and the
nuclear receptor must physically get into the nucleus. The
nuclear receptor also must bind its ligand with high affin­
ity. Because a major function of the receptor is to selec­ Figure 4-1  Mechanism of signal transduction by hormones and other
tively regulate target gene transcription, it must recognize ligands that act through nuclear receptors. HRE, hormone response element;
and bind to promoter elements in appropriate target genes. mRNA, messenger ribonucleic acid.
One discriminatory mechanism is dimerization of a recep­
tor with a second copy of itself or with another nuclear
receptor. The DNA-bound receptor must work in the
context of chromatin to signal the basal transcription
Domain Structure of Nuclear Receptors
machinery to increase or decrease transcription of the Nuclear receptors are proteins with molecular masses
target gene. In the regulation of signaling by nuclear recep­ between 50,000 and 100,000 d. They share a common
tors, some basic mechanisms are used by many or all series of domains, referred to as domains A through F (Fig.
members of the nuclear receptor superfamily, whereas 4-2). This linear depiction is useful for describing and com­
other mechanisms impart the specificity that is crucial to paring receptors, but it does not capture the roles of protein
the vastly different biologic effects of the many hormones folding and tertiary structure in mediating various receptor
and ligands that use these related receptors. functions. Investigations have revealed the structures of

A/B C D E F

Activation function-1 (AF-1)

DNA-binding (DBD)

P-box (half-site specificity)

Dimerization

Nuclear localization
signal (NLS)

C-terminal extension (flanking


DNA binding specificity)

Ligand binding domain (LBD)

Ligand-dependent activation

Activation function-2 (AF-2)


helix 12

Repression
Figure 4-2  Domain structures of nuclear receptors.
Mechanism of Action of Hormones That Act on Nuclear Receptors   55

individual domains and the first solved structure of a full- Hormone Binding
length nuclear receptor.
High-affinity binding of a lipophilic ligand is a shared
characteristic of many nuclear receptors. This defining
Nuclear Localization function of the receptor is mediated by the C-terminal
The nuclear receptors, like all cellular proteins, are synthe­ ligand-binding domain (LBD), domains D and E in Figure
sized on ribosomes that reside outside the nucleus. Import 4-2. This region of the receptor has many other functions,
of the nuclear receptors into the nucleus requires the including induction of dimerization and transcriptional
nuclear localization signal (NLS), which is located near the regulation (see later discussions).
border of the C and D domains (see Fig. 4-2). As a result of The structure of the LBD has been solved for a number
their NLSs, most of the nuclear receptors reside in the of receptors. All share a similar overall structure consisting
nucleus, with or without their ligands. A major exception of 12 α-helical segments in a highly folded tertiary struc­
is the glucocorticoid receptor; in the absence of hormone, ture (Fig. 4-3A). The ligand binds within a hydrophobic
it is tethered in the cytoplasm to a complex of chaperone pocket composed of amino acids in helices 3, 4, and 5 (H3,
molecules, including heat shock proteins (HSPs). Hormone H4, and H5, respectively). The major structural change
binding to the glucocorticoid receptor induces a conforma­ induced by ligand binding is an internal folding of the
tional change that results in dissociation of the chaperone most C-terminal helix (H12), which forms a cap on the
complex, allowing the hormone-activated glucocorticoid ligand-binding pocket (see Fig. 4-3B). Although the overall
receptor to translocate to the nucleus by means of its NLS. mechanism of ligand binding is similar for all receptors,

Helix 4-5
(H4-5) H4-5

Helix 3
(H3) H3
Helix 11
(H11) H11 H12
Activating ligand
(agonist)

Helix 12
(H12)

Corepressor Coactivator
(CoR) (CoA)

A B

CoRNR box NR box


region of CoR region of CoA
H4-5
H4-5

Activating ligand
(agonist)
H3

H3
H11
H11 H12

H12
Dissociation
of CoR

C D
Figure 4-3 Structural basis of nuclear receptor ligand binding and cofactor recruitment. A and B, Apo-receptor (no ligand bound). B and D, Ligand-bound
receptor. C and D, Structures showing the positional binding of a corepressor (C) or coactivator (D). NR, nuclear receptor.
56   Mechanism of Action of Hormones That Act on Nuclear Receptors

the molecular details are essential for determining ligand Dimer


specificity.36,37Ligand binding is the most critical determi­ interface
nant of receptor specificity.

Target Gene Recognition by Receptors


Another crucial specificity factor for nuclear receptors is
their ability to recognize and bind to the subset of genes
that is to be regulated by their cognate ligand. Target genes
contain specific DNA sequences that are called hormone P-box
response elements (HREs). Binding to the HRE is mediated
A G AA C A N TCTTGA
by the central C domain of the nuclear receptors (see Fig.
4-2). This region is typically composed of 66 to 68 amino TCTTGT A G AA C T
acids, including two subdomains that are called zinc fingers
because the structure of each subdomain is maintained by A Steroid hormone receptor homodimer
four cysteine residues coordinated with a zinc atom.
The first of these zinc-ordered modules contains basic
amino acids that contact DNA; as with the LBD, the overall
structure of the DNA-binding domain (DBD) is similar for Dimer
interface
all members of the nuclear receptor superfamily. The spe­ RXR NR
cificity of DNA binding is determined by multiple
factors (Table 4-3). All steroid hormone receptors, except
for the estrogen receptor, bind to the double-stranded DNA
sequence AGAACA (Fig. 4-4).
By convention, the double-stranded sequence is
described by the sequence of one of its complementary
strands, with the bases ordered from the 5′ to the 3′ end.
Other nuclear receptors recognize the sequence AGGTCA.
The primary determinant of this specificity is a group of
amino acid residues in the P box of the DBD (see Fig. 4-4). P-box
These hexamer DNA sequences are referred to as half-sites. N
AGGTCA AGGTCA
The only difference between these hexameric half-sites
is the central two base pairs (underlined in Fig. 4-4). For TCCAGT TCCAGT
some nuclear receptors, the C-terminal extension of the
DBD contributes specificity for extended half-sites contain­ B Nuclear receptor (NR)-RXR heterodimer
ing additional, highly specific DNA sequences 5′ to the
Figure 4-4 Structural basis for nuclear receptor (NR) DNA-binding speci-
hexamer. Another source of specificity for target genes is ficity is shown in the ribbon diagrams of receptor DNA-binding domains
the spacing and orientation of these half-sites, which in (DBDs). A, Steroid hormone receptor binding as a homodimer to the
most cases are bound by receptor dimers. inverted repeat (arrows) of the AGAACA half-site. The central base pairs are
underlined. B, RXR-NR heterodimer binding to the direct repeat of AGGTCA.
The position of the P box, the region of the DBD that makes direct contact
Receptor Dimerization with DNA, is shown. N, number of base pairs between the two half-sites;
The nuclear receptor DBD has affinity for the hexameric RXR, retinoid X receptor.
half-site or for extended half-sites; however, many HREs
are composed of repeats of the half-site sequence, and most
nuclear receptors bind these HREs as dimers. Steroid recep­ with three base pairs in between (IR3) (see Fig. 4-4A). The
tors, including estrogen receptors, function primarily as major dimerization domain in steroid receptors is within
homodimers, which preferentially bind to two half-sites the C domain, although the LBD contributes. Ligand
oriented toward each other (i.e., inverted repeats [IRs]) binding facilitates dimerization and DNA binding of steroid
hormone receptors. Most other receptors, including THR,
RAR, PPAR, LXR, and VDR, bind to DNA as heterodimers
with RXR (see Fig. 4-4B).
TABLE 4-3 
Heterodimerization with RXR is mediated by two dis­
Determinants of Target Gene Specificity tinct interactions, one involving LBDs and the other
of Nuclear Receptors involving DBDs. The receptor LBD mediates the strongest
interaction, which occurs even in the absence of DNA.
Specificity Region of Receptor
These receptor heterodimers bind to two half-sites arranged
1.  Binding to DNA DBD (C domain) as direct repeats (DRs) with variable numbers of base pairs
2.  Binding to specific hexamer P box in C domain (AGGTCA vs in between.
AGAACA) The spacing of the half-sites is a major determinant
3.  Binding to sequences 5′ to C-terminal extension of DBD of target gene specificity, and it results from the second
hexamer receptor-receptor interaction, which involves the DBDs
4.  Binding to hexamer repeats Dimerization domain (C domain for and is highly sensitive to the spacing of the half-sites. For
steroid receptors; D, E, and F for example, VDR/RXR heterodimers bind preferentially to
others)
DRs separated by three bases (DR3 sites), TR/RXR binds
5.  Recognition of hexamer spacing Heterodimerization with RXR
DR4, and RAR/RXR binds DR5 with highest affinity.38
(nonsteroid receptors, C domain)
Studies of isolated DBD binding to DNA have shown
DBD, DNA-binding domain; RXR, retinoid-X receptor. that these spacing requirements are related to the fact that
Mechanism of Action of Hormones That Act on Nuclear Receptors   57

the RXR binds to the upstream half-site (i.e., farthest from TABLE 4-5 
the start of transcription). As a result of the periodicity
of the DNA helix, each base pair separating the half-sites Nuclear Receptor Coregulators
leads to a rotation of about 36 degrees of one half-site rela­ Coactivators
tive to the other. Subtle differences in the structure of the
Chromatin remodeling
receptor DBDs make the interactions more or less favorable
Swi/Snf complex
at different degrees of rotation.39 Solution of the crystal
Histone acetyltransferase
structure of a nuclear receptor heterodimer (specifically,
p160 family (SRC-1, GRIP-1, pCIP)
the PPARγ-RXRα heterodimer) bound to DNA demon­
p300/CBP
strated that the heterodimer forms a nonsymmetric
pCAF (p300/CBP-associated factor)
complex, allowing the LBD of PPARγ to cooperate with
Activation
both DBDs to enhance response element binding.40
TRAP/DRIP
The discovery of nuclear receptor binding sites has been
largely empiric, based on the finding of binding sites in Corepressors
small numbers of known target genes. Unbiased analysis NCoR (nuclear receptor corepressor)
of thousands of nuclear receptor binding locations in living SMRT (silencing mediator for retinoid and thyroid hormone receptors)
cells has confirmed the canonical binding sequences for
several nuclear receptors, including those of the estrogen CBP, calcium-binding protein; DRIP, vitamin D receptor–interacting proteins;
receptor,41,42 the androgen receptor,43,44 the glucocorticoid TRAP, thyroid hormone receptor–associated proteins.
receptor,45 and PPAR/RXR heterodimers.46,47

The ligand-bound nuclear receptor communicates stim­


ulatory signals to GTFs on the gene to which it is bound.
RECEPTOR REGULATION OF Ligands specifically recruit a set of proteins, collectively
GENE TRANSCRIPTION called coregulators, to the nuclear receptor LBD.49 Positively
acting coregulators, called coactivators, specifically recog­
Nuclear receptors mediate a variety of effects on gene tran­ nize the ligand-bound conformation of the LBD and bind
scription. The most common modes of regulation are to the nuclear receptor on DNA only when an activating
ligand-dependent gene activation, ligand-independent (agonist) hormone or ligand is bound.50 A number of coac­
repression of transcription, and ligand-dependent negative tivator proteins that bind to liganded nuclear receptors
regulation of transcription (Table 4-4). have been described (Table 4-5).50
The most important determinant of coactivator binding
is the position of H12, which changes dramatically when
Ligand-Dependent Activation activating ligands bind receptors (see Fig. 4-3D). Along
Ligand-dependent activation is the best understood func­ with H3, H4, and H5, H12 forms a hydrophobic cleft that
tion of nuclear receptors and their ligands. The ligand- is bound by short polypeptide regions of the coactivator
bound receptor increases transcription of a target gene to molecules.51-53 These polypeptides, called NR boxes, have
which it is bound. The DBD brings the receptor domains characteristic sequences of LxxLL, in which L is leucine and
that mediate transcriptional activation to a specific gene. xx can be any two amino acids.54
Transcriptional activation itself is mediated primarily by Coactivators increase the rate of gene transcription. This
the LBD, which can function in the same way even when is accomplished by enzymatic functions, including DNA
it is transferred to a DNA-binding protein that is not related unwinding in response to histone acetyltransferase (HAT)
to nuclear receptors. The activation function (AF) of the activity.55 HAT activity is critically important for activation
LBD is referred to as AF-2 (see Fig. 4-2). because chromosomal DNA is tightly wrapped around
Gene transcription is mediated by a large complex of nucleosomal units composed of core histone proteins;
factors that ultimately regulate the activity of ribonucleic acetylation of lysine tails on histones opens up this chro­
acid (RNA) polymerase, the enzyme that uses the chromo­ matin structure.
somal DNA template to direct the synthesis of messenger The best understood class of coactivator proteins is the
RNA. Most mammalian genes are transcribed by RNA poly­ p160 family, whose name is based on their protein size
merase II using a large set of cofactor proteins that include (approximately 160 kd). The family contains at least three
basal transcription factors and associated factors collec­ such molecules, and each has many names (see Table 4-5).56
tively referred to as general transcription factors (GTFs). These factors possess HAT activity and recruit other coacti­
Details about GTFs are of fundamental importance and are vators, such as calcium-binding protein (CBP) and p300,
available elsewhere.48 which are also HATs. A third HAT, p300/CBP-associated
factor (PCAF, also designated TAF5L or TAF6L), is also
TABLE 4-4  recruited by liganded receptors. These HATs, along with a
complex of SMARC molecules (SWI/SNF-related, matrix-
Regulation of Gene Transcription associated, actin-dependent regulators of chromatin) that
by Nuclear Receptors direct adenosine triphosphate (ATP)-dependent DNA
Mode of Regulation Examples unwinding, create a chromatin structure that favors tran­
scription (Fig. 4-5).57
1.  Ligand-dependent gene activation DNA binding and recruitment of Recruitment of multiple HATs may reflect different spec­
coactivators ificities for core histones and, potentially for some nonhis­
2.  Ligand-independent gene DNA binding and recruitment of tone proteins. Some HATs also interact directly with GTFs
repression corepressors and further enhance their activities. The thyroid hormone
3.  Ligand-dependent negative DNA binding and recruitment of receptor–associated protein (TRAP) complex and the
regulation of gene expression corepressors or recruitment of
coactivators of DNA
vitamin D receptor–interacting protein (DRIP) complex
link nuclear receptors to GTFs.58,59 The HATs and TRAP
58   Mechanism of Action of Hormones That Act on Nuclear Receptors

HDAC3
Other HDACs
HAT Coactivators
Ligand p160
Corepressor CBP/p300 TRAP/DRIP
(N-CoR, SMRT) PCAF Coactivators
CoRNR CoRNR NR box NR box NR box NR box
LBD LBD
HDAC HAT
DBD DBD
HRE HRE HRE GTFs

Repression Activation
Figure 4-5 Coactivators and corepressors in transcriptional regulation by nuclear receptors. CBP, calcium-binding protein; CoRNR, coreceptor nuclear
receptor box; DBD, DNA-binding domain; DRIP, vitamin D receptor–interacting protein; GTFs, general transcription factors; HAT, histone acetyltransferase;
HDAC, histone deacetylase; HRE, hormone response element; LBD, ligand-binding domain; N-CoR, nuclear receptor corepressor; NR, nuclear receptor;
PCAF, CBP/p300-associated factor; SMRT, silencing mediator of retinoid and thyroid receptors; TRAP, thyroid hormone receptor–associated protein.

factors are recruited to the liganded, target gene–bound determines repression and activation by nuclear receptors
receptor in an ordered manner that also involves cycling (see Fig. 4-5).64
on and off of receptor gene targets.60 Although the mecha­ The transcriptional functions of NCoR and SMRT are the
nism is not understood, the cycling may allow multiple opposite of those of the coactivators. The corepressors
coactivators using similar NR boxes to contribute to themselves do not possess enzyme activity but do recruit
gene regulation without binding to a single receptor histone deacetylases (HDACs) to the target gene, thereby
simultaneously. reversing the effects of histone acetylation described earlier
and leading to a compact, repressed state of chromatin.
Repression of Gene Expression Although the mammalian genome contains multiple
HDACs, several of which may play a role in nuclear recep­
by Unliganded Receptor tor function, the main one involved in repression is
Although DNA binding is ligand dependent for steroid HDAC3, whose enzyme activity depends on interaction
hormone receptors, other nuclear receptors are bound to with NCoR or SMRT.65 The ability of NCoR to bind and
DNA even in the absence of their cognate ligand. The activate HDAC3 is required for normal metabolic and cir­
unliganded, DNA-bound receptor is not passively waiting cadian physiology.66 The corepressors interact directly with
for hormone; instead, it actively represses transcription of GTFs to inhibit their transcriptional activities, and they
the target gene. This repression turns off the target gene also exist in large, multiprotein complexes whose range of
and amplifies the magnitude of the subsequent activation functions is not fully understood.
by hormone or ligand. For instance, if the level of gene
transcription in the repressed state is 10% of the basal level Ligand-Dependent Negative Regulation
in the absence of a receptor, hormone activation to 10-fold
above that basal level represents a 100-fold difference of
of Gene Expression: Transrepression
transcription rate between hormone-deficient (repressed) The ligand-dependent switch between repressed and acti­
genes and hormone-activated genes (Fig. 4-6).61 vated receptor conformations explains how hormones acti­
In many ways, the molecular mechanism of repression vate gene expression. However, many important gene
is the mirror image of ligand-dependent activation. The targets of hormones are turned off in the presence of the
unliganded nuclear receptor recruits negatively acting ligand. This is referred to as ligand-dependent negative regula-
coregulators, called corepressors, to the target gene (see tion of transcription, or transrepression, to distinguish it from
Fig. 4-3C). The two major corepressors are large (approxi­ the repression of basal transcription by unliganded
mately 270 kd) proteins: nuclear receptor corepressor receptors.
(NCoR) and silencing mediator for retinoid and thyroid The mechanism of negative regulation is less well under­
receptors (SMRT).62 NCoR and SMRT specifically recognize stood than ligand-dependent activation, and there may be
the unliganded conformation of nuclear receptors and use several mechanisms.67 One mechanism involves nuclear
an amphipathic helical sequence similar to the NR box receptor binding to DNA-binding sites that reverse the
of coactivators to bind to a hydrophobic pocket in the paradigm of ligand-dependent activation (i.e., negative
receptor. response elements). For example, when the unliganded
For corepressors, the peptide responsible for receptor thyroid receptor binds to the negative response element of
binding is called the CoRNR box, and it contains the the genes for the β-subunit of thyroid-stimulating hormone
sequence (I or L)xx(I or V)I, in which I is isoleucine, L is or thyroid-stimulating hormone–releasing hormone, tran­
leucine, V is valine, and xx represents any two amino scription is activated.68 The role and recruitment of coregu­
acids.63 The receptor uses helices 3 to 5 to form the hydro­ lators in this process require further investigation. In other
phobic pocket, as in coactivator binding, but H12 does not cases, negative regulation may result from ligand binding
promote and even hinders corepressor binding. This nega­ to nuclear receptors that bind to other transcription factors
tive function of H12 highlights the role of the ligand- without binding DNA. This interaction leads to removal
dependent change in the position of H12 as the switch that of coactivators such as p300 and CBP from the other
Mechanism of Action of Hormones That Act on Nuclear Receptors   59

Coactivator

HRE
Activated state
10x

Dissociation
of
Corepressor
Relative level of gene transcription

Recruitment
HRE
of
Coactivator
x 100-fold
Basal state

Corepressor

HRE
0.1x
Repressed state

Figure 4-6  Repression and activation functions augment the dynamic range of transcriptional regulation by nuclear receptors. The magnitudes of activation
and repression were arbitrarily set at 10-fold for this theoretical example. In cells, these magnitudes vary as a function of coactivator and corepressor con-
centration and in a target gene–specific manner. HRE, hormone response element.

transcription factors that positively regulate the gene. In such as cAMP-response element–binding protein (CREB).
this model, inhibition of the activity of the positively Such signals can also lead to phosphorylation of nuclear
acting factors results in the observed negative regulation. receptors. Multiple signal-dependent kinases can phos­
phorylate nuclear receptors, leading to conformational
changes that regulate function.70 Phosphorylation can lead
Roles of Other Nuclear Receptor Domains to changes in DNA binding, ligand binding, or coactivator
The amino-terminal A/B domain of the nuclear receptors is binding; these consequences depend on the specific kinase,
the most variable region among all members of the super­ receptor, and domain of the receptor that is phosphory­
family in terms of length and amino acid sequence. Sub­ lated. The properties of coactivators and corepressors are
types of the same receptor often have completely different also regulated by phosphorylation.71
A/B domains. The function of this domain is the least well
defined of any. It is not required for unliganded repression
or for ligand-dependent activation. In many receptors, the
Receptor Antagonists
A/B domain contains positive transcriptional activity, often Certain ligands function as receptor antagonists by com­
referred to as activation function 1 (AF-1) (see Fig. 4-2). It is peting with agonists for the ligand-binding site. In the case
ligand independent but probably interacts with coactivators of steroid hormone receptors, the position of H12 in the
and may influence the magnitude of activation by agonists antagonist-bound receptor is not identical to that in the
or partial agonists. This activation function is tissue specific unliganded receptor or in the agonist-bound receptor. H12,
and tends to be more important for steroid hormone recep­ which has a sequence that resembles the NR box, binds to
tors, whose A/B domains are notably longer than those of the coactivator-binding pocket of the receptor and thereby
other members of the superfamily.69 The F domain of the prevents coactivator binding.72 This antagonist-bound con­
nuclear receptors is hypervariable in length and sequence, formation of the receptor also favors corepressor binding
and its function is not known. to steroid hormone receptors.

Cross-Talk with Other Signaling Pathways Tissue Selectivity of Ligands Interacting


Hormones and cytokines that signal through cell surface
with Nuclear Receptors
receptors also regulate gene transcription, often by activat­ Many endogenous hormones that act through nuclear
ing protein kinases that phosphorylate transcription factors receptors do so in a tissue-specific manner. The most
60   Mechanism of Action of Hormones That Act on Nuclear Receptors

12. Forman BM, Chen J, Evans RM. Hypolipidemic drugs, polyunsaturated


TABLE 4-6 
fatty acids, and eicosanoids are ligands for peroxisome proliferator-
Factors Modulating Receptor Activity activated receptors alpha and delta. Proc Natl Acad Sci U S A. 1997;94:4312-
4317.
in Different Tissues 13. Yu K, Bayonea W, Kallen CB, et al. Differential activation of peroxisome
proliferator-activated receptors by eicosanoids. J Biol Chem. 1995;
Concentration of receptor
270:23975-23983.
  Cell-specificity 14. Ziouzenkova O, Perrey S, Asatryan L, et al. Lipolysis of triglyceride-rich
  Variation within a given cell type lipoproteins generates PPAR ligands: evidence for an antiinflammatory
Post-translational modification of receptor (e.g., phosphorylation) role for lipoprotein lipase. Proc Natl Acad Sci U S A. 2003;100:2730-
2735.
Regulation of intracellular ligand levels (see Table 4-2)
15. Issemann I, Green S. Activation of a member of the steroid hormone
Pioneer factors determining receptor binding sites (e.g., FOX proteins) receptor superfamily by peroxisome proliferators. Nature. 1990;347:
Function of ligand 645-650.
  Agonist 16. Barish GD, Narkar VA, Evans RM. PPAR delta: a dagger in the heart of
the metabolic syndrome. J Clin Invest. 2006;116:590-597.
  Partial agonist
17. Rangwala SM, Lazar MA. Peroxisome proliferator-activated receptor
  Antagonist gamma in diabetes and metabolism. Trends Pharmacol Sci. 2004;25:
Concentration and types of coregulators 331-336.
  Coactivators 18. Forman BM, Tontonoz P, Chen J, et al. 15-Deoxy, delta 12, 14-
prostaglandin J2 is a ligand for the adipocyte determination factor
  Corepressors
PPARg. Cell. 1995;83:803-812.
19. Kliewer SA, Lenhard JM, Willson TM, et al. A prostaglandin J2 metabo­
lite binds peroxisome proliferator-activated receptor gamma and pro­
motes adipocyte differentiation. Cell. 1995;83:813-819.
obvious mechanism is differential expression of the recep­ 20. Lehmann JM, Moore LB, Smith-Oliver TA, et al. An antidiabetic thia­
tors, both in space (e.g., cell-type specificity)73 and in time zolidinedione is a high affinity ligand for the nuclear peroxisome
proliferator-activated receptor g (PPARg). J Biol Chem. 1995;270:12953-
(e.g., circadian variation).74 Ligand levels may be regulated 12956.
intracellularly (see earlier discussion and Table 4-2), and 21. Tontonoz P, Mangelsdorf DJ. Liver X receptor signaling pathways in
post-translational modification (e.g., phosphorylation) cardiovascular disease. Mol Endocrinol. 2003;17:985-993.
22. Moore DD, Kato S, Xie W, et al. International Union of Pharmacology:
influences cell-specific receptor function. Although nuclear LXII. The NR1H and NR1I receptors: constitutive androstane receptor,
receptors bind at thousands of sites on genomic DNA, the pregnene X receptor, farnesoid X receptor alpha, farnesoid X receptor
specific binding sites are regulated in a cell type–specific beta, liver X receptor alpha, liver X receptor beta, and vitamin D recep­
manner. For example, the estrogen receptor binds to over­ tor. Pharmacol Rev. 2006;58:742-759.
lapping but clearly different sets of genomic sites in the 23. Benoit G, Cooney A, Giguere V, et al. International Union of Pharmacol­
ogy: LXVI. Orphan nuclear receptors. Pharmacol Rev. 2006;58:798-836.
uterus and in the breast, probably because of the differen­ 24. Preitner N, Damiola F, Lopez-Molina L, et al. The orphan nuclear recep­
tial actions of so-called pioneer transcription factors, tor REV-ERBalpha controls circadian transcription within the positive
including forkhead box (FOX) proteins.75 limb of the mammalian circadian oscillator. Cell. 2002;110:251-260.
25. Raghuram S, Stayrook KR, Huang P, et al. Identification of heme as the
Some ligands function as antagonists in certain tissues ligand for the orphan nuclear receptors REV-ERBalpha and REV-ERBbeta.
but as full or partial agonists in others. These selective Nat Struct Mol Biol. 2007;14:1207-1213.
receptor modulators include compounds such as tamoxi­ 26. Yin L, Wu N, Curtin JC, et al. Rev-erbalpha, a heme sensor that
fen, a selective estrogen receptor modulator (SERM). SERMs coordinates metabolic and circadian pathways. Science. 2007;318:
are estrogen receptor antagonists with respect to the func­ 1786-1789.
27. Lu NZ, Cidlowski JA. The origin and functions of multiple human
tions of AF-2, including coregulator binding, and they glucocorticoid receptor isoforms. Ann N Y Acad Sci. 2004;1024:
require the AF-1 for their agonist activity.76 This agonism, 102-123.
like AF-1 activity, tends to be tissue specific and therefore 28. Zhang J, Lazar MA. The mechanism of action of thyroid hormone recep­
has great therapeutic utility.77 Table 4-6 summarizes factors tors. Annu Rev Physiol. 2000;62:439-466.
29. Achermann JC, Jameson JL. Fertility and infertility: genetic contribu­
contributing to the tissue specificity of receptor activity. tions from the hypothalamic-pituitary-gonadal axis. Mol Endocrinol.
1999;13:812-818.
REFERENCES 30. Seol W, Choi HS, Moore DD. An orphan nuclear hormone receptor that
lacks a DNA binding domain and heterodimerizes with other receptors.
1. Chambon P. The nuclear receptor superfamily: a personal retrospect on Science. 1996;272:1336-1339.
the first two decades. Mol Endocrinol. 2005;19:1418-1428. 31. Gurnell M, Chatterjee VK. Nuclear receptors in disease: thyroid receptor
2. Evans RM. The nuclear receptor superfamily: a rosetta stone for physiol­ beta, peroxisome-proliferator-activated receptor gamma and orphan
ogy. Mol Endocrinol. 2005;19:1429-1438. receptors. Essays Biochem. 2004;40:169-189.
3. O’Malley BW. A life-long search for the molecular pathways of steroid 32. Niu DM, Hwang B, Tiu CM, et al. Contributions of bone maturation
hormone action. Mol Endocrinol. 2005;19:1402-1411. measurements to the differential diagnosis of neonatal transient hypo­
4. Visser WE, Friesema EC, Jansen J, et al. Thyroid hormone transport in thyroidism versus dyshormonogenetic congenital hypothyroidism.
and out of cells. Trends Endocrinol Metab. 2008;19:50-56. Acta Paediatr. 2004;93:1301-1306.
5. Heuer H, Visser TJ. Minireview: pathophysiological importance of 33. McPhaul MJ. Molecular defects of the androgen receptor. Recent Prog
thyroid hormone transporters. Endocrinology. 2009;150:1078-1083. Horm Res. 2002;57:181-194.
6. Escriva H, Delaunay F, Laudet V. Ligand binding and nuclear receptor 34. Palermo M, Quinkler M, Stewart PM. Apparent mineralocorticoid excess
evolution. Bioessays. 2000;22:717-727. syndrome: an overview. Arq Bras Endocrinol Metabol. 2004;48:687-696.
7. Arriza JL, Weinberger C, Cerelli G, et al. Cloning of human mineralo­ 35. Stewart PM, Krozowski ZS. 11 Beta-hydroxysteroid dehydrogenase.
corticoid receptor complementary cDNA: structure and functional Vitam Horm. 1999;57:249-324.
kinship with the glucocorticoid receptor. Science. 1987;237:268-275. 36. Moras D, Gronemeyer H. The nuclear receptor ligand-binding domain:
8. Roy AK, Tyagi RK, Song CS, et al. Androgen receptor: structural domains structure and function. Curr Opin Cell Biol. 1998;10:384-391.
and functional dynamics after ligand-receptor interaction. Ann N Y Acad 37. Weatherman RV, Fletterick RJ, Scanlan TS. Nuclear receptor ligands and
Sci. 2001;949:44-57. ligand-binding domains. Annu Rev Biochem. 1999;68:559-581.
9. Germain P, Chambon P, Eichele G, et al. International Union of Phar­ 38. Umesono K, Murakami KK, Thompson CC, et al. Direct repeats as selec­
macology: LX. Retinoic acid receptors. Pharmacol Rev. 2006;58: tive response elements for the thyroid hormone, retinoic acid, and
712-725. vitamin D3 receptors. Cell. 1991;65:1255-1266.
10. Germain P, Chambon P, Eichele G, et al. International Union of Phar­ 39. Rastinejad F, Perlmann T, Evans RM, et al. Structural determinants of
macology: LXIII. Retinoid X receptors. Pharmacol Rev. 2006;58: nuclear receptor assembly on DNA direct repeats. Nature. 1995;375:
760-772. 203-211.
11. Michalik L, Auwerx J, Berger JP, et al. International Union of Pharmacol­ 40. Chandra V, Huang P, Hamuro Y, et al. Structure of the intact PPAR-
ogy: LXI. Peroxisome proliferator-activated receptors. Pharmacol Rev. gamma-RXR-nuclear receptor complex on DNA. Nature. 2008;456:
2006;58:726-741. 350-356.
Mechanism of Action of Hormones That Act on Nuclear Receptors   61

41. Carroll JS, Meyer CA, Song J, et al. Genome-wide analysis of estrogen 59. Rachez C, Suidan Z, Ward J, et al. A novel protein complex that interacts
receptor binding sites. Nat Genet. 2006;38:1289-1297. with the vitamin D3 receptor in a ligand-dependent manner and
42. Lin CY, Vega VB, Thomsen JS, et al. Whole-genome cartography of enhances VDR transactivation in a cell-free system. Genes Dev.
estrogen receptor alpha binding sites. PLoS Genet. 2007;3:e87. 1998;12:1787-1800.
43. Bolton EC, So AY, Chaivorapol C, et al. Cell- and gene-specific regula­ 60. Shang Y, Hu X, DiRenzo J, et al. Cofactor dynamics and sufficiency
tion of primary target genes by the androgen receptor. Genes Dev. in estrogen receptor-regulated transcription. Cell. 2000;103:843-
2007;21:2005-2017. 852.
44. Wang Q, Li W, Liu XS, et al. A hierarchical network of transcription 61. Hu X, Lazar MA. Transcriptional repression by nuclear hormone recep­
factors governs androgen receptor-dependent prostate cancer growth. tors. Trends Endocrinol Metab. 2000;11:6-10.
Mol Cell. 2007;27:380-392. 62. Privalsky ML. The role of corepressors in transcriptional regulation by
45. Wang JC, Derynck MK, Nonaka DF, et al. Chromatin immunoprecipita­ nuclear hormone receptors. Annu Rev Physiol. 2004;66:315-360.
tion (ChIP) scanning identifies primary glucocorticoid receptor target 63. Hu X, Lazar MA. The CoRNR motif contols the recruitment of corepres­
genes. Proc Natl Acad Sci U S A. 2004;101:15603-15608. sors to nuclear hormone receptors. Nature. 1999;402:93-96.
46. Lefterova MI, Zhang Y, Steger DJ, et al. PPARgamma and C/EBP factors 64. Glass CK, Rosenfeld MG. The coregulator exchange in transcriptional
orchestrate adipocyte biology via adjacent binding on a genome-wide functions of nuclear receptors. Genes Dev. 2000;14:121-141.
scale. Genes Dev. 2008;22:2941-2952. 65. Ishizuka T, Lazar MA. The nuclear receptor corepressor deacetylase acti­
47. Nielsen R, Pedersen TA, Hagenbeek D, et al. Genome-wide profiling of vating domain is essential for repression by thyroid hormone receptor.
PPARgamma:RXR and RNA polymerase II occupancy reveals temporal Mol Endocrinol. 2005;19:1443-1451.
activation of distinct metabolic pathways and changes in RXR dimer 66. Alenghat T, Meyers K, Mullican SE, et al. Nuclear receptor corepressor
composition during adipogenesis. Genes Dev. 2008;22:2953-2967. and histone deacetylase 3 govern circadian metabolic physiology.
48. Roeder RG. Role of general and gene-specific cofactors in the regulation Nature. 2008;456:997-1000.
of eukaryotic transcription. Cold Spring Harb Symp Quant Biol. 67. Lazar MA. Thyroid hormone action: a binding contract. J Clin Invest.
1998;63:201-218. 2003;112:497-499.
49. Halachmi S, Marden E, Martin G, et al. Estrogen receptor-associated 68. Chiamolera MI, Wondisford FE. Minireview: thyrotropin-releasing
proteins: possible mediators of hormone-induced transcription. Science. hormone and the thyroid hormone feedback mechanism. Endocrinology.
1994;264:1455-1458. 2009;150:1091-1096.
50. Smith CL, O’Malley BW. Coregulator function: a key to understanding 69. Kumar R, Thompson EB. Transactivation functions of the N-terminal
tissue specificity of selective receptor modulators. Endocr Rev. domains of nuclear hormone receptors: protein folding and coactivator
2004;25:45-71. interactions. Mol Endocrinol. 2003;17:1-10.
51. Feng W, Ribieiro RC, Wagner RL, et al. Hormone-dependent coactivator 70. Shao D, Lazar MA. Modulating nuclear receptor function: may the phos
binding to a hydrophobic cleft on nuclear receptors. Science. be with you. J Clin Invest. 1999;103:1617-1618.
1998;280:1747-1749. 71. Han SJ, Lonard DM, O’Malley BW. Multi-modulation of nuclear recep­
52. Nolte RT, Wisely GB, Westin S, et al. Ligand binding and co-activator tor coactivators through posttranslational modifications. Trends Endo-
assembly of the peroxisome prolierator-activated receptor-gamma. crinol Metab. 2009;20:8-15.
Nature. 1998;395:137-143. 72. Gronemeyer H, Gustafsson JA, Laudet V. Principles for modulation
53. Shiau AK, Barstad D, Loria PM, et al. The structural basis of estrogen of the nuclear receptor superfamily. Nat Rev Drug Discov. 2004;3:
receptor/coactivator recognition and the antagonism of this interaction 950-964.
by tamoxifen. Cell. 1998;95:927-937. 73. Bookout AL, Jeong Y, Downes M, et al. Anatomical profiling of nuclear
54. Heery DM, Kalkhoven E, Hoare S, et al. A signature motif in transcrip­ receptor expression reveals a hierarchical transcriptional network. Cell.
tional co-activators mediates binding to nuclear receptors. Nature. 2006;126:789-799.
1997;387:733-736. 74. Yang X, Downes M, Yu RT, et al. Nuclear receptor expression links the
55. Fischle W, Wang Y, Allis CD. Histone and chromatin cross-talk. Curr circadian clock to metabolism. Cell. 2006;126:801-810.
Opin Cell Biol. 2003;15:172-183. 75. Lupien M, Eeckhoute J, Meyer CA, et al. FoxA1 translates epigenetic
56. Glass CK, Rose DW, Rosenfeld MG. Nuclear receptor coactivators. Curr signatures into enhancer-driven lineage-specific transcription. Cell.
Opin Cell Biol. 1997;9:222-232. 2008;132:958-970.
57. Aoyagi S, Trotter KW, Archer TK. ATP-dependent chromatin remodeling 76. Osborne CK, Zaho H, Fuqua SA. Selective estrogen receptor modulators:
complexes and their role in nuclear receptor-dependent transcription structure, function, and clinical use. J Clin Oncol. 2000;18:3172-
in vivo. Vitam Horm. 2005;70:281-307. 3186.
58. Ito M, Yuan CX, Malik S, et al. Identity between TRAP and SMCC com­ 77. Shang Y, Brown M. Molecular determinants for the tissue specificity of
plexes indicates novel pathways for the function of nuclear receptors SERMs. Science. 2002;295:2465-2468.
and diverse mammalian activators. Mol Cell. 1999;3:361-370.

Anda mungkin juga menyukai