SlideShare a Scribd company logo
1 of 25
Download to read offline
1
SOAL DAN PEMBAHASAN
FARMAKOLOGI MOLEKULAR
PPAR DAN RESEPTOR ESTROGEN
*daftar pustaka dan jurnal terlampir di akhir
1. Jelaskan mekanisme obat golongan tiazolidindion berikatan dengan PPARγ secara
molekular !
Jawab :
Tahapannya :
a) Obat golongan tiazolindidion sebagai ligan melalui membran plasma masuk ke
dalam sitoplasma
b) Ligan berikatan dengan PPARγ yang sebelumnya dalam keadaan inaktif distabilkan
oleh co-repressor
c) Co-repressor lepas dari PPARγ
d) Co-activator berikatan dengan PPARγ-ligan
e) PPARγ membentuk kompleks heterodimer dengan RXR (Retinoid X Receptor)
untuk regulasi transkripsi
f) Transkripsi gen di dalam nucleus dengan disand oleh PPRE (Peroxisome
Proliferator Receptor Response Element)
g) Translasi
h) Menghasilkan protein yang akan mempengaruhi fungsi pada berbagai macam
jaringan :
Protein Kondisi Protein Jaringan
GLUT 4 ↑ Otot
↑ ambilan glukosa dan oksidasi asam lemak
Sel Adiposa
↑ ambilan glukosa, adipogenesis, diferensiasi, dan
simpanan trigliserida
GLUT 2 ↑ Liver
↑ oksidasi asam lemak, simpanan lipid
↓ glukoneogenesis
NF-kB ↓ Makrofag
2
↑ polarisasi makrofag
↓ inflamasi
PI3K ↑ Jantung dan pembuluh darah
↓ inflamasi
VEGF ↑ Jantung dan pembuluh darah
↓ inflamasi
CD36 ↑ Liver
↑ oksidasi asam lemak dan simpanan lemak
↓ glukoneogenesis
Makrofag
↑ polarisasi makrofag
↓ inflamasi
Sel Adiposa
↑ ambilan glukosa, adipogenesis, diferensiasi, dan
simpanan trigliserida
2. Jelaskan aktivitas genomik reseptor estrogen !
Jawab :
Tahapannya :
3
a) PA (Palmitoylation) menyebabkan ER (Estrogen Receptor) terlokalisasi di
membran plasma sel
b) E2 (17 β-estradiol) berdifusi melalui membran plasma sel
c) Masuk ke sitoplasma
d) E2 berikatan dengan ER
e) Terjadi relokasisasi
f) Perlibatan domain N-terminal dari interaksi intermolecular dan intramolecular
g) Ikatan DNA domain (DBD, c region) menyebabkan ER dapat dimerisasi dan
berikatan dengan sekuens DNA yang disandi oleh ERE (Estrogen Receptor
Element) melalui struktur 2 “zinc finger”
h) Aktivitas trans aktivasi
i) Aktivasi transkripsi melalui perlibatan koaktivator dan komponen dari mesin
transkripsi basal
j) D-region mendorong dimerisasi reseptor dan ikatan reseptor dengan chaperone Hsp
(Heat shock protein)
k) Domain ligan berikatan (LBD, E/F region, C terminal) meliputi domain E2
berikatan
l) Sinergis dengan N-terminal domain dalam transkripsi gen
m) AF 1 dan AF 2 sebagai activation function dari ER berikatan dengan koaktivator
n) ER α-E2 induksi aktivasi transkripsi sedangkan ER β-E2 menghambat transkripsi
o) Respons estrogenik
p) ER meningkatkan ikatan dengan Sp-1 (faktor transkripsi specificity protein 1)
q) Ko-aktivasi
r) Aktivasi Erα-E2 dari AP 1 responsive elements membutuhkan domain AF 1 dan
AF 2 dari reseptor agar dapat berikatan dengan meningkatkan aktivitas komponen-
komponen p160 (seperti SRC-1 dan SRC-2) oleh Fos/Jun
s) Transkripsi gen
t) Fungsi sel
4
Daftar Pustaka :
1. Choi, Sung Hee et al. Re-highlighting the Action of PPARγ in Treating Metabolic
Diseases. F1000 Research. 2019.
2. Marino, Maria et al. Estrogen Signaling Multiple Pathways to Impact Gene
Trascription. Current Genomics. 2006.
Current Genomics, 2006, 7, 497-508 497
1389-2029/06 $50.00+.00 ©2006 Bentham Science Publishers Ltd.
Estrogen Signaling Multiple Pathways to Impact Gene Transcription
Maria Marino1,*, Paola Galluzzo1
and Paolo Ascenzi1,2,3
1
Department of Biology and 2
Interdepartmental Laboratory for Electron Microscopy, University Roma Tre, Viale Gug-
lielmo Marconi, 446 I-00146 Roma, Italy; 3
National Institute for Infectious Diseases I.R.C.C.S. “Lazzaro Spallanzani”,
Via Portuense 292, I-00149 Roma, Italy
Abstract: Steroid hormones exert profound effects on cell growth, development, differentiation, and homeostasis. Their
effects are mediated through specific intracellular steroid receptors that act via multiple mechanisms. Among others, the
action mechanism starting upon 17-estradiol (E2) binds to its receptors (ER) is considered a paradigmatic example of
how steroid hormones function. Ligand-activated ER dimerizes and translocates in the nucleus where it recognizes spe-
cific hormone response elements located in or near promoter DNA regions of target genes. Behind the classical genomic
mechanism shared with other steroid hormones, E2 also modulates gene expression by a second indirect mechanism that
involves the interaction of ER with other transcription factors which, in turn, bind their cognate DNA elements. In this
case, ER modulates the activities of transcription factors such as the activator protein (AP)-1, nuclear factor-B (NF-B)
and stimulating protein-1 (Sp-1), by stabilizing DNA-protein complexes and/or recruiting co-activators. In addition, E2
binding to ER may also exert rapid actions that start with the activation of a variety of signal transduction pathways (e.g.
ERK/MAPK, p38/MAPK, PI3K/AKT, PLC/PKC). The debate about the contribution of different ER-mediated signaling
pathways to coordinate the expression of specific sets of genes is still open. This review will focus on the recent knowl-
edge about the mechanism by which ERs regulate the expression of target genes and the emerging field of integration of
membrane and nuclear receptor signaling, giving examples of the ways by which the genomic and non-genomic actions of
ERs on target genes converge.
Received on: August 8, 2006 - Revised on: October 21, 2006 - Accepted on: October 30, 2006
Key Words: Estrogen, estrogen receptors, genomic and non-genomic action mechanism, gene transcription.
1. INTRODUCTION
The principle estrogenic hormone, 17-estradiol (E2),
synthesized by testosterone aromatization in the ovary and in
other tissues, plays a central role in the control of sexual be-
havior and reproductive functions. At present it is well rec-
ognized that the impact of E2 in human physiology is wider
than previously thought impact including the differentiation
of several tissues and organs, the modulation of inflamma-
tion, and brain and cardiovascular functions as well [see 1-
3].
E2 regulates human physiology via diffusion through the
plasma membrane of target cells and signaling through intra-
cellular hormone-specific estrogen receptors (ERs). Two
distinct types of signaling can be mediated, often referred to
as genomic and non-genomic or non-genotropic pathways. In
the genomic pathway, estrogens bind to ERs in the nucleus,
inducing a conformational change in the receptors that cause
dissociation from chaperones, dimerization, and activation of
the receptor transcriptional domain [4-6].
The canonical model for ER-mediated regulation of gene
expression involves the direct binding of dimeric ER to
DNA sequences known as estrogen response elements (EREs),
*Address correspondence to this author at the Department of Biology, Uni-
versity “Roma Tre”, Viale Guglielmo Marconi 446, I-00146 Roma, Italy;
Tel: +39-06-55176345; Fax: +39-06-55176321;
E-mail: m.marino@uniroma3.it
which are specific, inverted palindromic sequences [7]. In
addition, ER can indirectly associate with promoters through
protein-protein interactions with other DNA-binding tran-
scription factors [8-10]. In either case, interaction of ERs
with E2 leads to transcriptional activation of the associated
genes via recruitment of coactivators and components of the
basal transcriptional machinery [11-14]. In addition to the
nuclear ERs, plasma membrane-associated ERs mediate the
non-genomic signaling pathway [see 15-19], which can lead
both to cytoplasmic alterations and to regulation of gene
expression [16, 20, 1].
Regulation of transcription by nuclear ER is more com-
plicated than the classical paradigm would predict [5, 18].
The two nuclear ERs, ER and ER, exhibit distinct tran-
scriptional properties and can form both homodimers and
heterodimers [22-24]. Recent studies point to the fact that
signaling pathways modulate both ERs and some co-regula-
tory molecules activities [13, 25].
To understand the connection between physiological and
molecular functions of ERs, the field requires an in-depth
understanding of the spectrum of genes regulated in each
tissue and cell type. This review will focus on the current
state of knowledge about the mechanism by which ERs regu-
late the expression of target genes and the emerging field of
integration of membrane and nuclear receptor signaling, giv-
ing examples of the ways by which the genomic and non-
genomic actions of ERs on target genes converge.
498 Current Genomics, 2006, Vol. 7, No. 8 Marino et al.
2. THE STRUCTURE OF ESTROGEN RECEPTORS
Human ER and ER are encoded by different genes
located on different chromosomes (locus 6q25.1 and locus
14q23-24.1, respectively) [26-29]. ER and ER, like all the
members of the nuclear receptor super-family, are modular
proteins sharing common regions, named A/B, C, D, and
E/F, as well as a high sequence homology (Fig. (1A)). These
regions participate in the formation of independent but inter-
acting functional domains. The N-terminal domain (A/B re-
gion) is involved in both inter-molecular and intra-molecular
interactions as well as in the activation of gene transcription.
The DNA binding domain (DBD, C region) allows ER to
dimerize and to bind to the specific ERE sequence on DNA
through its two “zinc finger” structures (Fig. (1B)). The hinge
domain (D region) has a role in receptor dimerization and in
binding to chaperone heat-shock proteins (Hsp). The ligand
binding domain (LBD, E/F region, C-terminal) comprises
the E2-binding domain and works, synergistically with the
N-terminal domain in the regulation of gene transcription [5,
30-32].
ERs contain two regions called activation functions
(AFs) important for ligand-dependent transcriptional activity
(Fig. (1A)) [5, 30-32]. AF-1 and AF-2 regions of ERs, inter-
acting with a number of transcription co-activators, can acti-
vate transcription independently but in most cases, they syn-
ergize with one another in a promoter- and cell-context spe-
cific manner [33].
AF-1 could be activated even in a ligand-independent
manner, depending on the phosphorylation status of ER. In
particular, the Ser118 residue in the AF-1 region of ER, as
well as residues Ser106 and Ser124 in the AF-1 region of
ER, are the phosphorylation sites essential for the ligand-
independent activation of ERs through the Ras-mitogen acti-
vated protein kinase (MAPK) signaling cascade [see 34, 35].
Recent progress in studies on genomic and cDNA se-
quences has accelerated the identification of gene splice
variants in the NR super-family. Numerous mRNA splice
variants exist for both ERs and the best-characterized splice
variants are ER46 and ERcx, which are frequently co-
expressed with their wild-type counterparts. The exact func-
tion and potential role of these and other ERs splice variants
in physiology and human disease remain to be elucidated
[see 36].
Fig. (1). Domain organization of human ER and ER (A). ERs consist of the N- terminal region involved in transactivation (A/B domains,
AF-1), the DNA binding domain (DBD, C domain), the hinge region involved in dimerization (D domain), the C-terminal region containing
ligand binding domain (LBD, E/F domain, AF-2) and transactivation function-2 (AF-2). The percentage indicates the homology between
ER and ER. (B) Binding mode of ERE to dimeric ER (PDB ID:1HCQ) [163]. Spheres indicate the zinc atoms. For details, see text.
Estrogen Signaling and Gene Transcription Current Genomics, 2006, Vol. 7, No. 8 499
3. ESTROGEN RECEPTOR GENOMIC ACTIVITY
3.1. Direct Association to DNA
The pioneering work by O’Malley and colleagues dem-
onstrated that ERs function as ligand-activated transcription
factors [37]. The trans-activation activity of ERs initiate
through the ligand-bound receptor to its cognate, cis-acting
enhancers, ERE [38]. The consensus palindromic element
ERE was initially described based on the estrogen-respon-
sive sequence in the Xenopus laevis vitellogenin A2 pro-
moter: 5’-GGTCACAGTGACC-3’ [39-41]. This “perfect”
ERE sequence was shown to function in an orientation- and
distance-independent manner, both of which are properties
of an enhancer [7, 42]. When ER directly interacts with the
promoter/enhancer, binding to a full ERE is apparently the
dominant mode of interaction. The human full EREs have a
3-bp spacer between the two half-sites, the exceptions being
response elements in the human transforming growth factor
(TGF)- promoter, with a 4-bp spacer, and in the promoter
of the rat luteinizing hormone  gene, with a 5-bp spacer
[42]. Controversy still exists concerning ER DNA binding
via ERE half sites, although a number of examples exist [43-
46].
Since the identification of a canonical ERE, several com-
putational approaches have been undertaken to identify tar-
get genes based on the presence of EREs within promoter
proximal regions [47, 48]. For instance, for the 38 estrogen-
responsive genes reviewed by Klinge [7], most of the func-
tional EREs located within the promoters or 3’-untranslated
regions are not the traditional consensus sequence. Thus,
many target genes contain response elements that bear little
similarity to consensus EREs. In one of the most comprehen-
sive studies, Bourdeau and coworkers screened for all EREs
in the human and mouse genomes and identified in excess of
70,000 EREs within the human genome, over 17,000 of
which were within 15 kb of mRNA start sites [48]. Elimina-
tion of EREs that were not conserved between the human
and mouse genomes reduced the number of gene proximal
EREs to 660. A number of these sites were validated as
genuine ER interaction sites, supporting the use of computa-
tional models to predict putative ER target genes to some
degree [49].
The sequence of the response element affects the affinity
that a given receptor has for binding DNA. ER binds with
the highest affinity the canonical ERE sequence found within
the vitellogenin A2 gene, and less well the imperfect EREs
found within the vitellogenin B1 (5’-AGTCACTGTGACC-
3’) [39], pS2 (GGTCACGGTGGCC-3’) [50], and oxytocin
(5’-GGTCAAGGTCACC-3’) [51] genes. This explains, at
least in part, how the sequence of the response element can
be one important determinant of the extent to which ERs can
activate gene expression [52-55].
The conformation of transcription factors can be altered
through binding to DNA [see 56]. The specific ERE se-
quences could exert distinct, allosteric effects on the con-
formation of ER and ER [52, 57, 58]. Just as ligand-
induced changes in ER conformation influence ER interac-
tions with co-activators, consensus and imperfect EREs also
influence the ability of ERs to bind co-activators. Note that
the steroid receptor coactivator-2 (SRC-2) interacts better
with ER bound to EREs from the vitellogenin A2 than from
the vitellogenin B1 gene [54].
3.2. Indirect Association to DNA
The ER signaling mechanisms discussed until now pro-
vide an explanation for the regulation of genes in which a
functional ERE-like sequence can be documented within the
promoter. Another category of gene promoters, lacking any
ERE-like sequences, requires a second DNA-binding tran-
scription factor to mediate ER association with the DNA
[42]. This mechanism is generally referred to as “transcrip-
tional cross-talk” [59-60]. Roughly 35% of the categorized
human primary E2-responsive genes are transcripted via ER-
indirect DNA association [42].
Stimulating protein-1 (Sp-1) is the predominant mediator
of ER-DNA indirect binding [42] and increasing numbers of
genes are found to be induced by E2 via this mechanism
including the low-density lipoprotein (LDL) receptor [8],
endothelial nitric oxide sinthase (eNOS) [61], c-fos [62],
cyclin D1 [63], and the retinoic acid receptor-1 genes [64,
65]. In response to estrogenic stimulation, ER enhances the
binding of Sp-1 to its site, containing GC-rich promoter se-
quences [46] and contributes to co-activator recruitment. The
DNA-binding domain of ER is dispensable for such activa-
tion [42, 66, 67].
Another example is the interaction between ER and the
c-rel subunit of the nuclear factor-B (NF-B) complex.
This interaction prevents NF-B from binding to and stimu-
lating expression from the interleukin-6 (IL-6) promoter
[68]. In this way, E2 inhibits expression of the cytokine IL-6
[68-70].
Other intermediary factors through which ER can associ-
ate with promoters/enhancers include: activating transcrip-
tion factor (ATF)-2/c-jun or ATF-2/cAMP response element
binding protein (CREB) for the cyclin D1 gene, ATF-
1/CREB for the Bcl-2 gene, and nuclear transcription factor-
Y for the mouse E2F1 gene [42].
ERs utilize protein-protein interactions also to enhance
transcription of genes that contain activator protein-1 (AP-1)
sites [71] related, but not identical, to those for the
ATF/CREB transcription factors. The AP-1 complex, com-
posed of Jun protein dimers and of Jun/Fos heterodimers,
plays an important role in cell proliferation. Notably, ER
activation of IGF-1 and collagenase expression is mediated
through the interaction of receptor with Fos and Jun at AP-1
binding sites [42]. Collagenase, insulin-like growth factor
(IGF)-1 receptor, ovalbumin, and cyclin D1 are examples of
genes activated by the ER-E2 complex via AP-1 [72, 73].
ER and ER have been shown to signal in opposite
ways at AP-1 sites. ER activates transcription in the pres-
ence of E2, whereas ER-E2 inhibits AP-1-dependent tran-
scription [74, 60]. Studies show that ER-E2 activation of
AP-1-responsive elements requires both AF-1 and AF-2 do-
mains of the receptor, which bind and enhance the activity of
the p160 components (e.g. SRC-1 and SRC-2) of the co-
activator complex recruited to the site by Fos/Jun. Interest-
ingly, human ER, which lacks a functional AF-1, is unable
to activate transcription of AP-1-regulated genes when
bound with ER agonists, indicating the possibility of distinct
500 Current Genomics, 2006, Vol. 7, No. 8 Marino et al.
physiological actions of the two ERs via the regulation of
unique subsets of genes [4]. Similar to AP-1, E2 binding to
ER induces transcriptional activation when associated with
Sp-1 in GC-rich regions. However, E2 interaction with ER
does not result in the formation of a transcriptionally active
complex at a promoter containing Sp-1 elements. As an ex-
ample ER and ER, in the presence of E2, oppose each
other’s function in the regulation of the cyclin D1 promoter
[75]. There is considerable evidence that cyclin D1, impor-
tant for progression of cells through the G1 phase of the cell
cycle, is a well-defined target for ER-E2 action in mam-
mary carcinoma cells [76-78], although no detectable “per-
fect” or ERE-like sequence in the cyclin D1 gene promoter
has been reported [79]. Deletion of AP-1 and Sp-1 respon-
sive element motifs in the cyclin D1 gene promoter resulted
in attenuation of promoter responsiveness to E2 [72, 80].
Unlike ER, E2-bound ER represses cyclin D1 expression
[81] and blocks ER-E2-mediated induction when both re-
ceptor isoforms are present [22]. Consequently, these differ-
ences in transcriptional activity between the ER and ER
may account for the major differences in their tissue specific
biologic actions. This complexity is further enhanced by the
presence of different ER splicing forms, by the ability of
ERs to form homodimers and heterodimers, and by their
capacity to interact with different co-regulators [82].
3.3. Transcriptional Co-Factors
Both in the direct and indirect action modes, the ligand-
activated ERs are not the transcription controllers. In fact,
ERs need to interact with co-regulatory proteins (co-
activators or co-repressors) to form a platform upon which
additional proteins are assembled [12, 13]. Cofactors interact
with ERs through their Leu rich motif (i.e., Leu-Xxx-Xxx-
Leu-Leu, where Xxx is any amino acid). Several classes of
ER cofactors have been identified. The first identified and
well-characterized co-activator family consists of three re-
lated members SRC-1, which is the founding member of the
family, SRC-2, and SRC-3 [see 83]. A large co-activator
complex, referred to as thyroid hormone receptor associated
protein/vitamin D receptor-integrating protein (TRAP/DRIP)
complex, could connect ERs directly to the basal transcrip-
tion machinery via its intrinsic chromatin remodeling func-
tions. In addition, histone acetyl transferase (e.g., CBP/p300),
histone methyl transferase (e.g., CARM1 and PRMT1) and
the nucleosome remodeling complexes (e.g., SWI/SNF) are
necessary to release the chromatin-dependent inhibition of
gene transcription [13].
Although there are far fewer nuclear receptor co-repre-
ssors, these (macro)molecules serve important roles in nega-
tively regulating ER-dependent gene expression. Two AF-2
interacting proteins, receptor-interacting protein-140 and
short hetero-dimer partner, exhibit negative co-regulatory
functions because they can antagonize SRC-1 co-activators
in vivo and compete for AF-2 binding in vitro [84-86]. On
the other hand, ERs could also associate with specific tran-
scriptional repressors such as the nuclear receptor corepres-
sor and specific histone deacetylase complexes [13].
The relative expression of co-activators and co-repre-
ssors, within a cell, influences the ability of ER ligands (e.g.,
E2 and selective ER modulators (SERMs)) to regulate gene
expression [2, 13, 87].
Because of the homology in their AF-2 domains (see Fig.
(1A)), ER and ER should be similar in co-activator re-
cruitment, but certain differences have been reported. For
E2-bound receptors, ER, but not ER, binds well to the
receptor-interacting component of the mammalian mediator
complex, TRAP220. There are differences between the rela-
tive affinities of ER and ER for members of the p160 co-
activator family [13, 88]. More pronounced differences are
observed in the case of SERM-bound ERs [see 2, 13]. For
ERE-dependent gene expression, the SERM 4-hydroxy-
tamoxifen is a partial agonist of ER, but is generally unable
to stimulate ER transcriptional activity [89-91]. Con-
versely, when assessing ER activity on AP-1 containing re-
porter genes, 4-hydroxytamoxifen will stimulate ER and
ER transcriptional activity in a cell-dependent fashion [74].
A mechanism for shuttling off transcription involves the
covalent post-translational modification of ERs and co-
activators (e.g., lysine acetylation and arginine methylation),
which can inhibit the binding of co-activators to nuclear re-
ceptors or other transcriptional activators by altering critical
protein-protein interaction surface [see 13, 92]. Thus, the
acetylation of SRC-3 by p300 has been shown to cause a
disruption of receptor-co-activator complexes, leading to a
decrease in receptor-mediated gene activation [see 92]. Us-
ing a variety of biochemical and cell-based assays, Krauss
and co-workers have shown ER, but not ER, is a target for
acetylation by p300 and have identified acetylation as modu-
lator of the ligand-dependent gene regulatory activity of ER
[93].
A number of cellular signaling pathways also influence
the ER-dependent gene expression modulating ER confor-
mational changes or co-regulators recruitment [5, 13]. It has
been recognized only recently that both co-activators and co-
repressors are also substrates for kinases, their phosphoryla-
tion affects their ability to interact with steroid receptors [94,
95].
4. ESTROGEN RECEPTOR NON-GENOMIC ACTIV-
ITY
The “genomic action” of steroid hormones occurs after a
time-lag of at least 2 hours after E2 stimulation and explains
some of hormone functions in physiological and pathological
situations [see 96, 97]. This picture was challenged when a
physiological dose of E2 was reported to increase the uterine
cAMP level in ovariectomized rats within 15 seconds [98],
an effect too rapid to be accounted for genomic action(s).
This event was not abrogated by transcriptional inhibitors
and was termed “rapid or non-genomic”. Actually the term
“non-genomic” is not adequate when referring to rapid
changes that may also initiate new gene transcription [see 96,
99].
Various signaling pathways are activated upon E2 bind-
ing to ERs. These rapid events may be classified into four
main signaling cascade: phospholipase C (PLC)/protein ki-
nase C (PKCs) [100-106], Ras/Raf/MAPK [72, 107-113],
phosphatidyl inositol 3 kinase (PI3K)/AKT [15, 16, 80, 81,
97, 114-118], and cAMP/ protein kinase A (PKA) [104, 119-
123].
These pathways present numerous interactions with sev-
eral other pathways. The ER-E2 complex interacts with the
Estrogen Signaling and Gene Transcription Current Genomics, 2006, Vol. 7, No. 8 501
IGF-1 receptor, leading to IGF-1 receptor activation and
hence to MAPK signaling pathway activation [124]. In addi-
tion, the ER-E2 complex activates the EGF receptor by a
mechanism that involves activation of guanine nucleotide
exchange proteins (G-proteins), Src, and matrix metallopro-
teinases, leading to an increase in extracellular regulated
kinases (ERK) and PI3K/AKT activities [109, 125-129]. In
endothelial cells the Src/PI3K/AKT pathway mediates rapid
E2-dependent activation of eNOS and the release of nitric
oxide. AKT and PKC could also modulate the MAPK path-
way through Raf phosphorylation [97, 116, 130, 131].
It is important to note that activation of signaling path-
ways by E2 is cell type-specific. Indeed, the effect of E2 on
PKC activity has been observed in the preoptic area of fe-
male rat brain slices, but not in the hypothalamus or cortex
[132]. The activation of G-protein/Src/PI3K/MAPK pathway
by E2 was evident in late, but not early, differentiated rat
pre-adipocytes [109]. The differential requirement of Src/
PI3K or intracellular calcium for MAPK activation is also
observed in diverse cell types [15, 109, 129]. Different PKC
isoforms are rapidly activated by E2 in HepG2 and MCF7
cells [102]. As a whole, these studies indicate that the rapid
actions of E2 depend on a number of conditions such as the
set of signal transduction molecules and downstream targets
present in the target cell, thus the responses are likely to be
diverse.
All these results point to the concept that ER is the pri-
mary endogenous mediator of rapid E2 actions. Less infor-
mation is available on the role played by the ER-E2 com-
plex to activate rapid non-genomic mechanisms. A subpopu-
lation of ER transfected into Chinese Hamster ovary cells is
capable of activating inositol tris-phosphate production,
ERK and JNK phosphorylation [133]. Geraldes and cowork-
ers reported that E2 reduces ERK activity through ER
stimulation in porcine smooth muscle cells [134]. We have
recently reported the ability of the ER-E2 complex to acti-
vate the p38 member of MAPK family, but not ERK or
AKT, in human colon cancer cells [81, 135]. Although the
scarce information does not allow a complete discussion on
the contribution of ER in E2-induced rapid signals, these
data indicate that also ER could originate cell-specific sig-
nal transduction cascade.
The rapidity by which E2 induces rapid signals as well as
the localization of signaling complex raises the requirement
of a plasma membrane ER. Debate continues over whether
structural changes target nuclear ERs in separate pools local-
izing them to the membrane [61, 97, 99, 117, 136], or
whether membrane ER represents a novel receptor [137-
142]. Besides these data, much evidence favors the idea that
the membrane-localized ER is the same protein as the nu-
clear-localized receptor [72, 80, 133, 143, 144]. Even if the
definitive proof that membrane and nuclear ER are the same
protein requires isolation and “sequencing” of the two recep-
tor pools, ER and ER must be considered a population of
protein(s) which localization in the cell is able to dynami-
cally change, shuttling from membrane to cytosol and to the
nucleus, depending on ligand binding [87, 97, 135,145].
Current evidence indicates that a small population of
ER and ER localize at the plasma membrane exists within
caveolar rafts. It is at the plasma membrane that E2-liganded
ER associates with the scaffolding protein caveolin-1 and a
variety of signal transduction cascade activation occurs [e.g.,
PLC, PKC, ERK, PI3K, and nitric oxide synthase (NOS)].
ERs do not contain a trans-membrane domain [15, 18], thus
the ability of ER and ER to associate with the plasma
membrane could be due to its association with membrane
proteins and/or by post-translational addition of lipids to
ER [16, 146].
Fatty acids and isoprenoids are two of the most common
lipid moieties found on post-translational modified proteins
bound to membranes. No consensus sequences for N-acyla-
tion (i.e., miristoylation) or S-prenylation have been found in
ER and ER [147]. On the contrary, S-acylation (i.e.,
palmitoylation) does not require any consensus sequence, but
just reactive Cys residues [148].
Cys residues present in the ER and ER LBD could
undergo S-acylation. In particular, the amino acid sequence
encompassing the Cys447 residue of ER and Cys399 of
ER is highly homologous to that surrounding the S-
palmitoylated Cys132 residue of human caveolin-1 [147].
Based on this observation we demonstrated that ER un-
dergo S-palmitoylation which represents the major determi-
nant for its residence at the plasma membrane and in its as-
sociation with caveolin-1 [146, 147]. It is noteworthy that
ER is also a palmitoylable protein [Marino M., unpublished
results].
Because ER has no intrinsic kinase domains the local-
ization of ERs at the plasma membrane facilitate the associa-
tion between ER and signaling proteins allowing the activa-
tion of rapid events. Src, Shc, proline-, glutamic acid-,
leucine- rich protein /modulator of non-genomic activity of
estrogen receptor (PELP1/MNAR), the p85 subunit of
PI3K, receptor tyrosine kinases (i.e., EGF and IGF-1 recep-
tors), as well as G-protein isoforms (i.e., Gs and Gq) have
all been reported to serve as components of large complexes
of interacting proteins. Through the mediation of these
molecules, E2 activates the MAPK and PI3K/AKT pathways
[16, 136, 149-151].
Although the list of signaling and adapter proteins inter-
acting with ER is growing, protein-protein complex forma-
tion occurs only 5 to 15 min after E2 stimulation [152].
Thus, the conformational changes of the ER LBD domain,
which follows E2 entry into the cell, seems to be important
in allowing the ER-E2 complex to detach from the mem-
brane and allocate with growth factor receptors or adapter
proteins to activate downstream signals.
4.1. Cell Functions Regulated by Non-Genomic Signals
The rapid activities of ERs are widely accepted and dis-
agreement on the involvement of nuclear receptors is quite
settled. However, other controversies in this field are still
present and related to whether or not all of these rapid effects
are of physiological relevance [153]. The main difficulties
are linked to the experimental models used. In fact, the study
of signaling pathways can be done mainly on isolated, often
immortalized, cells and it is very complicated to obtain simi-
lar information on a whole organism in which the use of sig-
naling inhibitors could have many side effects other than to
inhibit just one kinase.
502 Current Genomics, 2006, Vol. 7, No. 8 Marino et al.
Nevertheless, the physiological significance of rapid
membrane-starting pathways has been clarified at least for
some E2 targets. In the nervous system, E2 affects neural
functions (e.g., cognition, behavior, stress responses, and
reproduction) in part by inducing such rapid responses [96].
In the skeleton, ER, present in caveolae of bone-forming
osteoblasts, transmits survival signals through activation of
the Src/Shc/ERK pathway and prolongs the life span of os-
teoblasts [21]. At the same time, E2 delivers a pro-apoptotic
signal to bone-resorbing osteoclasts, shortening their life
span [21]. Although these studies have been done mainly in
cell-culture systems, their results suggest that ER rapid sig-
naling actions have also a role in vivo. In the liver, rapid E2-
induced signals (i.e., PLC/PKC) are deeply linked to the ex-
pression of the LDL receptor and to a decreased level of se-
rum LDL-cholesterol [103]. Finally, vascular protection by
E2 in ischemia/reperfusion injury in vivo requires E2-
induced activation of endothelial NOS, as mediated by the
PI3K/AKT pathway [117, 130].
The mechanism(s) by which E2 exerts proliferative ef-
fects is assumed to be exclusively mediated by rapid mem-
brane-starting actions [72, 80, 101, 102, 114, 115]. E2 treat-
ment of mammary-derived MCF-7 cells triggers the associa-
tion of ER with Src and p85 leading to DNA synthesis
[115]. In HepG2 cells multiple and parallel membrane-
starting pathways are rapidly activated by the ER-E2 com-
plex [72, 80, 101] and the blockade of PLC/PKC, ERK, and
PI3K/AKT pathways completely prevents the E2-induced
DNA synthesis [72, 80]. ERK/MAPK and PI3K/AKT path-
ways, rapidly activated by the ER-E2 complex, also have a
critical role in E2 action as a survival agent. In fact, these
pathways enhance the expression of the anti-apoptotic pro-
tein Bcl-2, block the activation of the p38/MAPK, reduce the
pro-apoptotic caspase-3 activation, and promote G1-to-S
phase transition via the enhancement of the cyclin D1 ex-
pression [72, 80, 81].
What is the contribution of ER to E2-induced cell pro-
liferation? ER appears to act as a dominant regulator in E2
signaling, and when co-expressed with ER it causes a con-
centration-dependent reduction of ER-mediated transcrip-
tional activation [22] and the repression of ER-mediated
effects including cell proliferation. Consistent with this no-
tion, E2 increases cell proliferation and causes tumor forma-
tion in MCF-7 cells expressing only ER [22]. On the other
hand, ER inhibits the E2-induced proliferation of trans-
fected MCF-7 cells and prevents tumor formation in a mouse
xenograft model in response to E2 [154]. This effect is
linked to the ER repressive effect on ER-induced gene
transcription by binding to other transcription factors (e.g.,
AP-1, Sp-1) [22]. Recently, ER has been reported to rapidly
induce a persistent membrane-initiated activation of p38/
MAPK without any interference on survival proliferative
pathways, thus impairing the activation of cell cycle compo-
nents (i.e., cyclin D1 expression) [81].
5. INTEGRATION OF NUCLEAR AND EXTRA-
NUCLEAR ACTION OF E2
Even though the membrane ERs and associated non-
genomic actions is an area of active research, the nuclear
effects of membrane ERs has not received much attention
[see 16, 19]. In human vascular smooth muscle cells tran-
siently transfected with ER an E2-dependent and an E2-
independent translocation of ER from the membrane to the
nucleus was observed. The latter was blocked by MAPK
inhibitors [155]. The ability of membrane ER and/or the
growth factor receptor tyrosine kinases to signal via multiple
kinases to the nucleus undoubtedly impacts all aspects of
cellular function.
E2-induced ERK activation up regulates AP-1 mediated
genes (e.g., c-fos) [156]. This results in part from serum re-
sponse factor/elk-1 stimulation by E2, and in part by re-
cruitment of nuclear ER and co-activators to AP-1 sites on
gene promoters [16]. Other targets include several members
of the signal transducer and activators of transcription
(STAT) family such as STAT1, STAT3 and STAT5. In en-
dothelial cells, activation of both STAT3 and STAT5 by E2
was mediated through signaling pathways involving MAPK,
PI3K and Src and it functions to regulate -casein expression
[15].
Similarly, PI3K activation by E2-induced signaling from
the membrane ER rapidly up regulates hundreds of genes in
a target cell [157]. Microarray analysis of gene expression in
vascular endothelial cells showed that about 250 genes were
up-regulated 40 min after treatment. This effect could be
prevented by the PI3K inhibitor, LY294,002 [157]. Interest-
ingly, the transcriptional activity of the ER-E2 complex is
inhibited by a pre-treatment with the ERK inhibitors
PD98,059 and U0126 [20, 114]. This suggests that stimula-
tion of some gene expression (i.e., cyclin D1 and prolactin)
by E2 occurs through ERK and PI3K activation.
CREB is the most studied of the several transcription
factors rapidly activated by E2. In a hippocampal cell line
[158], adipocyte cells [109], and colonic carcinoma cells
[159], CREB transcriptional activity can be induced by E2 or
E2-BSA through MAPK pathway, independently from the
PKA pathway. Such activation of CREB induces expression
of several genes (e.g., c-fos, uncoupling protein-2). In con-
trast, in neuroblastoma cells activation of CREB by ER-
mediated rapid signals is dependent on the cAMP/PKA
pathway, leading to neurotensin gene expression [160].
In addition, ERs are possible ER-mediated rapid signal
targets. Indeed, it has been long known that E2 treatment can
increase the phosphorylation state of ERs, via ERK and
PI3K, the mutation of important phosphorylation sites re-
duces their transcription activity [19]. The rapid E2-evoked
phosphorylation of ER contributes to the stimulation of ER
dimerization and its nuclear translocation. As an example,
the phosphorylation of ER on Ser305 enhances cyclin D1
transcription in breast cancer. E2 also induces phosphoryla-
tion of ER in Ser118, Ser167, and Tyr537 residues through
the non-genomic activation of the MAPK signaling pathway
[19]. Furthermore, the Ser167 residue of ER also can be
phosphorylated in response to rapid E2-mediated PI3K/AKT
activation, whereas E2-induced p38/MAPK phosphorylation
of Thr311 promotes ER nuclear localization and interaction
with specific receptor coactivators [19].
Besides these functions, the complexity of the mecha-
nism of ER action suggests a more finely tuned control ex-
erted by E2-induced rapid signals on cellular molecular
Estrogen Signaling and Gene Transcription Current Genomics, 2006, Vol. 7, No. 8 503
events. In particular, the extra-nuclear signals induced by E2
occur before the appearance of nuclear effects and the cell
context in which the genomic events occur will be different
depending on which signal pathway is activated. Thus, the
integration between these molecular events is required to
obtain the complete cellular response.
The complex relationship between membrane and nu-
clear effects induced by E2 also involves membrane-
initiating phosphorylation of co-activators recruiting these
proteins to the nuclear transcriptosome [13, 16]. This aug-
ments the recruitment of co-activator proteins, such as SRC-
1 by ER [13]. One can envisage a carefully controlled modu-
lation of nuclear ER-induced transcription, depending upon
which signaling pathway(s) are activated by E2 in a given
cell context. It is likely that discrete signaling pathways
regulate the access of co-repressors to target gene promoters,
although this mechanism is not well studied. As a corollary
to this, phosphorylation of co-activators at discrete motifs
could be inhibitory as well.
The possible convergence of ER genomic and non-
genomic activities at multiple response elements provides an
extremely fine degree of control for the regulation of tran-
scription by ERs. It has been estimated that more than 500
kinases are encoded within the human genome. The ability
of ER-E2 membrane starting signals and/or growth factor
receptor to signal through multiple cascades to the nucleus,
undoubtedly has an impact on all aspects of cellular function,
contributing to E2-induced cell proliferation and survival, all
essential features of cell physiology as well as of tumor biol-
ogy [16].
Examples of such fine-tuned ER multiple control action
are cyclin D1 and vascular endothelial growth factor (VEGF)
genes. Cyclin D1, a well-defined target for E2 in mammary
gland, is important for the progression of cells throughout
the G1 phase of the cell cycle. The cyclin D1 promoter is
complex and contains binding sites for several transcription
factors, but no ERE-like sequences have been identified [79].
It has been suggested that activation of the cyclin D1 gene
transcription by E2 results from different ER activities: di-
rect ER/Sp-1 or ER/AP-1 interaction [161] as well as ER-
dependent non-genomic mechanisms [72, 80]. The cyclin D1
promoter also contains binding sites for STAT5 and NF-B,
and these could be targets for ERs through both genomic and
non-genomic actions [15]. The VEGF gene is another exam-
ple of cross-talk between ERs non-genomic and genomic
action. In fact, VEGF gene promoter contains both an ERE-
like variant and GC-rich sequences that bind ER and ER-Sp-
1 complex [42]. Both must be occupied for the E2 maximal
activation [15].
As a whole, these data strongly suggest that E2-induced
rapid signaling reaches to the nucleus through these and
other, undiscovered, pathways and synergize each other to
provide plasticity for cell response to sex steroids (see Fig.
(2)).
6. CONCLUSION
The regulation of gene expression by E2 is a multi-
factorial process, involving both genomic and non-genomic
actions that converge at certain response elements located in
the promoters of target genes. The final gene responses,
however, could depend on a number of conditions such as
the combination of transcription factors bound to a specific
gene promoter, the cellular localization of ERs, the levels of
various co-regulator proteins and signal transduction compo-
nents, as well as the nature of extra-cellular stimuli. These
variables are highly specific for cell types. Thus, E2 could
use different signaling pathways depending both on the cel-
lular type and on the physiological status of the cell. In this
way E2 evokes distinct gene responses in different types of
target cells [15, 16, 97, 162].
The possibility that E2 could act on ER pools localized in
different cell compartments (i.e., membrane versus cytoso-
lic) gives rise to questioning the ability of these different ER
pools to send parallel or synergic signals to the nucleus. For
example, it has been observed that a naturally occurring
variant of the metastatic tumor antigen 1 sequesters ER in
the cytoplasm of breast cancer cells. The result of this cyto-
solic retention is the reduction of E2-mediated transcription
and the enhancement of E2-initiated ERK activation [136].
These data suggest that the same ER molecule is involved in
genomic and in rapid signal transduction cascade. More data
are needed to confirm this hypothesis and the use of dynamic
imaging in the near future will help to clarify this issue.
Based upon findings highlighted in this review, one may
envisage a dynamic integrated model of action for ERs in-
side the cell. In this model, ERs would shuttle from cell
membrane to the cytoplasm and to the nucleus, in a dynamic
equilibrium between different cell compartments. Each could
play a different role in a multi step process of target gene
activation by ER and co-activators from their upstream non-
genomic to their downstream genomic responses would lead
to activation of transcription (Fig. (2)).
The cell context specific environment (e.g., differentia-
tion, ER level, and ER co-expression) has an impact on the
integration of rapid signaling by E2 from the membrane and
on subsequent nuclear transcription. This leads to different
signal cascades, different gene expression in response to the
same hormone, and different cell biological outcome.
The field is moving quickly. The challenges in the near
future are to continue identifying the discrete actions of each
ER intracellular pool, in order to clarify the role of ER, and
to identify the potential cross-talk between ERs and other
nuclear receptors. As we gain a deeper understanding of the
complex controls exerted by ER and start identifying the
critical players, it is likely that some of these putative mole-
cules might emerge target candidates for therapeutic devel-
opment in the treatment of hormone-responsive diseases,
such as for different types of cancer.
ACKNOWLEDGEMENTS
Some experimental concepts described in the current
paper are based on work conducted in the laboratories of the
authors. These experimental studies were supported by
grants from the Ministry of Education, University, and Re-
search of Italy (PRIN-COFIN 2004 to M.M.). The Authors
wish to thank past and present members of their laboratory
who contributed with data and discussions to the ideas pre-
sented here.
504 Current Genomics, 2006, Vol. 7, No. 8 Marino et al.
ABBREVIATIONS
AF = Activation function
AP-1 = Activator protein-1
CREB = cAMP responsive element binding protein
DBD = DNA-binding domain
EGF = Epidermal growth factor
ER = Estrogen receptor
ERE = Estrogen responsive element
ERK = Extracellular regulated kinase
E2 = 17-estradiol
G-proteins = Guanine nucleotide exchange proteins
Hsp = Heath shock protein
IGF-1 = Insulin-like growth factor-1
IL-6 = Interleukin-6
LDL = Low dendity lipoprotein
LBD = Ligand-binding domain
MAPK = Mitogen-activated protein kinase
MNAR = Modulator of non-genomic activity of estro-
gen receptor (also named Pro-, Glu-, and
Leu-rich protein-1 PELP1)
NF-B = Nuclear factor B
PI3K = Phosphatidyl inositol 3-kinase
NOS = Nitric oxide synthase
PKA = Protein kinase A
PKC = Protein kinase C
PLC = Phospholipase C
SERMs = Selective estrogen modulators
Sp-1 = Stimulating protein-1
SRC = Steroid receptor co-activator family
STAT = Signal transducers and activators of
transcription
Fig. (2). Schematic model illustrating the relationship between rapid, intermediate, and long term actions of E2 on target cells. Palmitoyla-
tion (PA) allows the estrogen receptor (ER) localization at the plasma membrane. 17-estradiol (E2) binding induces ER re-
localization, association to signaling proteins, and triggers the activation of signaling cascades. The kinase activations phosphory-
late ER, modulate transcriptional coactivators recruitment, and enhance AP-1 and Sp-1 activation. After dimerization ERs directly
interact with ERE on DNA. ERs-DNA indirect association occurs through protein-protein interactions with the Sp-1 and AP-1 tran-
scription factors. AP-1, activating protein-1; MNAR, modulator of non-genomic activity of ER; PA, palmitic acid; Sp-1, stimulating
factor-1. For details, see text.
Estrogen Signaling and Gene Transcription Current Genomics, 2006, Vol. 7, No. 8 505
TGF- = Transforming growth factor-
TRAP = Thyroid hormone receptor associated protein
(also named vitamin D receptor-integrating
protein DRIP)
VEGF = Vascular endothelial growth factor
REFERENCES
[1] Gruber, C.J., Tschugguel, W., Schneeberger, C., Huber, J.C. Pro-
duction and actions of estrogens. New Engl. J. Med. 2002, 346:
340-352.
[2] Pearce, S.T., Jordan, V.C. The biological role of estrogen receptors
 and  in cancer. Crit. Rev. Oncol. Hematol. 2004, 50: 3-22.
[3] Deroo, B.J., Korach, K.S. Estrogen receptors and human disease. J.
Clin. Invest. 2006, 116: 561-570.
[4] Hall, J.M., Couse, J.F., Korach, KS. The multifaceted mechanisms
of estradiol and estrogen receptor signaling. J. Biol. Chem. 2001,
276: 36869-36872.
[5] Nilsson, S., Makela, S., Treuter, E., Tujague, M., Thomsen, J.,
Andersson, G., Enmark, E., Pettersson, K., Warner, M.,
Gustafsson, J.-Å. Mechanisms of estrogen action. Physiol. Rev.
2001, 81: 1535-1565.
[6] Hall J.M., McDonnell, D.P. Coregulators in nuclear estrogen recep-
tor action: from concept to therapeutic targeting. Mol. Interv. 2005,
5: 343-357.
[7] Klinge, C.M. Estrogen receptor interaction with estrogen response
elements. Nucleic Acids Res. 2001, 29: 2905-2919.
[8] Li, C., Briggs, M.R., Ahlborn T.E., Kraemer, F.B., Liu, J. Re-
quirement of Sp1 and estrogen receptor-interaction in 17-
estradiol-mediated transcriptional activation of the low density
lipoprotein receptor gene expression. Endocrinology 2001, 142:
1546–1553.
[9] Safe, S. Transcriptional activation of genes by 17-estradiol
through estrogen receptor-Sp1 interactions. Vitam. Horm. 2001, 62:
231-252.
[10] Stossi, F., Likhite, V.S., Katzenellenbogen, J.A., Katzenellenbogen,
B.S. Estrogen-occupied estrogen receptor represses cyclin G2 gene
expression and recruits a repressor complex at the cyclin G2 pro-
moter. J. Biol. Chem. 2006, 281: 16272-16278.
[11] Glass, C.K., Rosenfeld, M.G. The coregulator exchange in tran-
scriptional functions of nuclear receptors. Genes Dev. 2000, 14:
121-141.
[12] McKenna, N.J., O’Malley, B.W. Combinatorial control of gene
expression by nuclear receptors and coregulators. Cell 2002, 108:
465-474.
[13] Smith, C.L., O’Malley, B.W. Coregulator function: a key to under-
standing tissue specificity of selective receptor modulators. Endocr.
Rev. 2004, 25: 45-71.
[14] Kato, S., Sato, T., Watanabe, T., Takemasa, S.; Masuhiro, Y.,
Ohtake, F., Matsumoto, T. Function of nuclear sex hormone recep-
tors in gene regulation. Cancer Chemother. Pharmacol. 2005, 56
(Suppl.1): 4-9.
[15] Björnström, L., Sjöberg, M. Mechanisms of estrogen receptor
signaling: convergence of genomic and nongenomic actions on tar-
get genes. Mol. Endocrinol. 2005, 19: 833-842.
[16] Levin, E.R. Integration of the extra-nuclear and nuclear actions of
estrogen. Mol. Endocrinol. 2005, 19: 1951-1959.
[17] Marino, M., Ascenzi, P., Acconcia, F. S-palmitoylation modulates
estrogen receptor alpha localization and functions. Steroids 2006,
71: 298-303.
[18] Ascenzi, P., Bocedi, A., Marino, M. Structure-function relationship
of estrogen receptor  and : impact on human health. Mol. Aspects
Med. 2006, 27: 299-402.
[19] Zhang, D., Trudeau, V.L. Integration of membrane and nuclear
estrogen receptor signaling. Comp. Biochem. Physiol. A Mol. In-
tegr. Physiol. 2006, 144: 306-315.
[20] Acconcia, F., Marino, M. Synergism between genomic and non-
genomic estrogen action mechanisms. IUBMB Life 2003, 55: 145-
150.
[21] Kousteni, S., Han, L., Chen, J.R., Almeida, M., Plotkin, L.I., Bel-
lido, T., Manolagas, S.C. Kinase-mediated regulation of common
transcription factors accounts for the bone-protective effects of sex
steroids. J. Clin. Invest. 2003, 111: 1651-1664.
[22] Matthews, J., Gustafsson, J.-Å. Estrogen signalling: A subtle bal-
ance between ER and ER. Mol. Interventions 2003, 3: 281-292.
[23] Li, X., Huang, J., Yi, P., Bambara, R.A., Hilf, R., Muyan, M. Sin-
gle-chain estrogen receptors (ERs) reveal that the ER/ heterodi-
mer emulates functions of the ER dimer in genomic estrogen sig-
naling pathways. Mol. Cell. Biol. 2004, 24: 7681-7694.
[24] Monroe, D.G., Secreto, F.J., Subramaniam, M., Getz, B.J., Khosla,
S., Spelsberg, T.C. Estrogen receptor  and  heterodimers exert
unique effects on estrogen-and tamoxifen-dependent gene expres-
sion in human U2OS osteosarcoma cells. Mol. Endocrinol. 2005,
19: 1555-1568.
[25] Mendelsohn, M.E. Genomic and nongenomic effects of estrogen in
the vasculature. Am. J. Cardiol. 2002, 90: 3F-6F.
[26] Gosden, J.R., Middleton, P.G., Rout, D. Localization of the human
oestrogen receptor gene to chromosome 6q24-q27 by in situ hy-
bridization. Cytogenet. Cell Genet. 1986, 43: 218-220.
[27] Enmark, E., Pelto-Huikko, M., Grandien, K., Lagercrantz, S., La-
gercrantz, J., Fried, G., Nordenskjold, M., Gustafsson, J.-Å. Human
estrogen receptor -gene structure, chromosomal localization, and
expression pattern. J. Clin. Endocrinol. Metab. 1997, 82: 4258-
4265.
[28] Luisi, S., Galleri, L., Marini, F., Ambrosiani, G., Brandi, M.L.,
Petraglia, F. Estrogen receptor gene polymorphisms are associated
with recurrence of endometriosis. Fertil. Steril. 2006, 85: 764-766.
[29] Zhou, W., Liu, Z., Wu, J., Liu, J.H., Hyder, S.M., Antoniou, E.,
Lubahn, D.B. Identification and characterization of two novel
splicing isoforms of human estrogen-related receptor . J. Clin.
Endocrinol. Metab. 2006, 91: 569-579.
[30] Mosselman, S., Polman, J., Dijkema, R. ER: identification and
characterization of a novel human estrogen receptor. FEBS Lett.
1996, 392: 49-53.
[31] Claessens, F., Gewirth, D.T. DNA recognition by nuclear recep-
tors. In: McEwan, I.J. (Ed.), Essay in Biochemistry: The Nuclear
Receptor Superfamily Portland Press, London, 2004, 59-72.
[32] Kumar, R., Johnson, B.H., Thompson, E.B. Overview of the struc-
tural basis for transcription regulation by nuclear hormone recep-
tors. In: McEwan, I.J. (Ed.), Essay in Biochemistry: The Nuclear
Receptor Superfamily Portland Press, London, 2004, pp. 27-39.
[33] McEwan, I.J. Sex, drugs and gene expression: signalling by mem-
bers of the nuclear receptor superfamily. In: McEwan, I.J. (Ed.),
Essays in Biochemistry: the Nuclear Receptor Superfamily. Port-
land Press, London, 2004, pp. 1-10.
[34] Ortì, E., Bodwell, J.E., Munck, A. Phosphorylation of steroid hor-
mone receptors. Endocr. Rev. 1992, 13: 105-128.
[35] Lannigan, D.A. Estrogen receptor phosphorylation. Steroids 2003,
68: 1-9.
[36] Herynk, M.H., Fuqua, S.A. Estrogen receptor mutations in human
disease. Endocr. Rev. 2004, 25: 869-898.
[37] O’Malley, B.W. A life-long search for the molecular pathways of
steroid hormone action. Mol. Endocrinol. 2005, 19: 1402-1411.
[38] Beato, M. Gene regulation by steroid hormones. Cell 1989, 56:
335-344.
[39] Walker, P., Germond, J.-E., Brown-Luedi, M., Givel, F., Wahli, W.
Sequence homologies in the region preceding the transcription ini-
tiation site of the liver estrogen-responsive vitellogenin and apo-
VLDLII genes. Nucleic Acids Res. 1984, 12: 8611-8626.
[40] Klein-Hitpass, L., Schorpp, M., Wagner, U., Ryffel, G.U. An es-
trogen-responsive element derived from the 5’ flanking region of
the Xenopus vitellogenin A2 gene functions in transfected human
cells. Cell 1986, 46: 1053-1061.
[41] Ponglikitmongkol, M., Green, S., Chambon, P. Genomic organiza-
tion of the human oestrogen receptor gene. EMBO J. 1988, 7:
3385-3388.
[42] O’Lone, R., Frith, M.C., Karlsson, E.K., Hansen, U. Genomic
targets of nuclear estrogen receptors. Mol. Endocrinol. 2004, 18:
1859-1875.
[43] Kato, S., Tora, L., Yamauchi, J., Masushige, S., Bellard, M.,
Chambon, P. A far upstream estrogen response element of the
ovalbumin gene contains several half palindromic 5’-TGACC-3’
motifs acting synergistically. Cell 1992, 68: 731-742.
[44] Klinge, C.M., Bodenner, D.L., Desai, D., Niles, R.M., Traish, A.M.
Binding of type II nuclear receptors and estrogen receptor to full
and half-site estrogen response elements in vitro. Nucleic Acids
Res. 1997, 25: 1903-1912.
506 Current Genomics, 2006, Vol. 7, No. 8 Marino et al.
[45] Porter, W., Wang, F., Wang, W.; Duan, R., Safe, S. Role of estro-
gen receptor/Sp1 complexes in estrogen-induced heat shock protein
27 gene expression. Mol. Endocrinol. 1996, 10: 1371-1378.
[46] Porter, W., Saville, B., Hoivik, D., Safe, S. Functional synergy
between the transcription factor Sp1 and the estrogen receptor.
Mol. Endocrinol. 1997, 11: 1569-1580.
[47] Bajic, V.B., Tan, S.L., Chong, A., Tang, S., Strom, A., Gustafsson,
J.-Å., Lin, C.Y., Liu, E.T. Dragon ERE Finder version 2: A tool for
accurate detection and analysis of estrogen response elements in
vertebrate genomes. Nucleic Acids Res. 2003, 31: 3605-3607.
[48] Bourdeau, V., Deschenes, J., Métivier, R., Nagai, Y., Nguyen, D.,
Bretschneider, N., Gannon, F., White, J.H., Mader, S. Genome-
wide identification of high-affinity estrogen response elements in
human and mouse. Mol. Endocrinol. 2004, 18: 1411-1427.
[49] Carroll, J.S., Brown, M. Estrogen receptor target gene: an evolving
concept in molecular endocrinology. Mol. Endocrinol. 2006, 20:
1707-1714.
[50] Nunez, A.M., Jakowlev, S., Briand, J.P., Gaire, M., Krust, A., Rio,
M.C., Chambon, P. Characterization of the estrogen-induced pS2
protein secreted by the human breast cancer cell line MCF-7. En-
docrinology 1987, 121: 1759-1765.
[51] Sausville, E., Carney, D., Battey, J. The human vasopressin gene is
linked to the oxytocin gene and is selectively expressed in a cul-
tured lung cancer cell line. J. Biol. Chem. 1985, 260: 10236-10241.
[52] Loven, M.A., Wood, J.R., Nardulli, A.M. Interaction of estrogen
receptors  and  with estrogen response elements. Mol. Cell. En-
docrinol. 2001, 181: 151-163.
[53] Loven, M.A., Likhite, V.S., Choi, I., Nardulli, A.M. Estrogen re-
sponse elements alter coactivator recruitment through allosteric
modulation of estrogen receptor  conformation. J. Biol. Chem.
2001, 276: 45282-45288.
[54] Wood, J.R., Likhite, V.S., Loven, M.A., Nardulli, A.M. Allosteric
modulation of estrogen receptor conformation by different estrogen
response elements. Mol. Endocrinol. 2001, 15: 1114-1126.
[55] Yi, P., Driscoll, M.D., Huang, J., Bhagat, S., Hill, R., Bambara,
R.A., Muyan, M. The effects of estrogen-responsive element- and
ligand-induced structural changes on the recruitment of cofactors
and transcriptional responses by ER and ER. Mol. Endocrinol.
2002, 16: 674-693.
[56] Lefstin, J.A., Yamamoto, K.R. Allosteric effects of DNA on
transcriptional regulators. Nature 1998, 392: 885-888.
[57] Hall, J.M., McDonnell, D.P., Korach, K.S. Allosteric regulation of
estrogen receptor structure, function, and coactivator recruitment
by different estrogen response elements. Mol. Endocrinol. 2002,
16: 469-486.
[58] Ikeda, M., Wilcox, E.C., Chin, W.W. Different DNA elements can
modulate the conformation of thyroid hormone receptor heterodi-
mer and its transcriptional activity. J. Biol. Chem. 1996, 271:
23096-23104.
[59] Gottlicher, M., Heck, S., Herrlich, P. Transcriptional cross-talk, the
second mode of steroid hormone receptor action. J. Mol. Med.
1998, 76: 480-489.
[60] Aranda, A., Pascual A. Nuclear hormone receptors and gene ex-
pression. Physiol Rev. 2001, 81: 1269-1304.
[61] Chambliss, K.L., Shaul, P.W. Rapid activation of endothelial NO
synthase by estrogen: evidence for a steroid receptor fast-action
complex (SRFC) in caveolae. Steroids 2002, 67: 413-419.
[62] Duan, R., Porter, W., Safe, S. Estrogen-induced c-fos protoonco-
gene expression in MCF-7 human breast cancer cells: role of estro-
gen receptor Sp1 complex formation. Endocrinology 1998, 139:
1981-1990.
[63] Castro-Rivera, E., Samudio, I., Safe, S. Estrogen regulation of
cyclin D1 gene expression in ZR-75 breast cancer cells involves
multiple enhancer elements. J. Biol. Chem. 2001, 276: 30853-
30861.
[64] Sun, G., Porter, W., Safe, S. Estrogen-induced retinoic acid recep-
tor 1 gene expression: role of estrogen receptor-Sp1 complex.
Mol. Endocrinol. 1998, 12: 882-890.
[65] Zhang, J., Hu, X., Lazar M.A. A novel role for helix 12 of retinoid
X receptor in regulating repression. Mol. Cell. Biol. 1999, 19:
6448-6457.
[66] Batistuzzo de Medeiros, S.R., Krey, G., Hihi, A.K., Wahli, W.
Functional interaction between the estrogen receptor and the tran-
scription activator Sp1 regulate the estrogen-dependent transcrip-
tional activity of the vitellogenin A1 promoter. J. Biol. Chem.
1997, 272: 18250-18260.
[67] Qin, C., Singh, P., Safe, S. Transcriptional activation of insulin-like
growth factor-binding protein-4 by 17-estradiol in MCF-7 cells:
role of estrogen receptor-SP1 complexes. Endocrinology 1999,
140: 2501-2508.
[68] Galien, R., Garcia, T. Estrogen receptor impairs interleukin-6
expression by preventing protein binding on the NF-B site.
Nucleic Acids Res. 1997, 25: 2424-2429.
[69] Ray, A., Prefontaine, K.E., Ray, P. Down-modulation of interleu-
kin-6 gene expression by estradiol 17 in the absence of high affin-
ity DNA binding by the estrogen receptor. J. Biol. Chem. 1994,
269: 12940-12946.
[70] Kalaitzidis, D., Gilmore, T.D. Transcription factor cross-talk: the
estrogen receptor and NF-B. Trends Endocrinol. Metab. 2005, 16:
46-52.
[71] Gaub, M.P., Bellard, M., Scheuer, I., Chambon, P., Sassone-Corsi,
P. Activation of the ovalbumin gene by the estrogen receptor in-
volves the fos-jun complex. Cell 1990, 3: 1267-1276.
[72] Marino, M., Acconcia, F., Bresciani, F., Weisz, A., Trentalance A.
Distinct nongenomic signal transduction pathways controlled by
17-estradiol regulate DNA synthesis and cyclin D1 gene transcrip-
tion in HepG2 cells. Mol. Biol. Cell 2002, 13: 3720-3729.
[73] Fujimoto, N., Honda, H., Kitamura, S. Effects of environmental
estrogenic chemicals on AP-1 mediated transcription with estrogen
receptors  and . J. Steroid Biochem. Mol. Biol. 2004, 88: 53-59.
[74] Paech, K., Webb, P., Kuiper, G.G., Nilsson, S., Gustafsson, J.,
Kushner, P.J., Scanlan, T.S. Differential ligand activation of estro-
gen receptors ER and ER at AP-1 sites. Science 1997, 277:
1508-1510.
[75] Liu, M.M., Albanese, C., Anderson, C.M., Hilty, K., Webb, P.,
Uht, R.M., Price, R.H.Jr., Pestell, R.G., Kushner, P.J. Opposing ac-
tion of estrogen receptors  and  on cyclin D1 gene expression. J.
Biol. Chem. 2002, 277: 24353-24360.
[76] Altucci, L., Addeo, R., Cicatiello, L., Dauvois, S., Parker, M.G.,
Truss, M., Beato, M., Sica, V., Bresciani, F., Weisz, A. 17-
Estradiol induces cyclin D1 gene transcription, p36D1-p34cdk4
complex activation and p105Rb phosphorylation during mitogenic
stimulation of G1-arrested human breast cancer cells. Oncogene
1996, 12: 2315-2324.
[77] Foster, J.S., Wimalasena, J. Estrogen regulates activity of cyclin-
dependent kinases and retinoblastoma protein phosphorylation in
breast cancer cells. Mol. Endocrinol. 1996, 10: 488-498.
[78] Prall, O.W., Sarcevic, B., Musgrove, E.A., Watts, C.K., Sutherland,
R.L. Estrogen-induced activation of Cdk4 and Cdk2 during G1-S
phase progression is accompanied by increased cyclin D1 expres-
sion and decreased cyclin-dependent kinase inhibitor association
with cyclin E-Cdk2. J. Biol. Chem. 1997, 272: 10882-10894.
[79] Herbert, B., Truss, M., Beato, M., Müller, R. Inducibile regulatory
elements in the human cyclin D1 promoter. Oncogene 1994, 9:
1295-1304.
[80] Marino, M., Acconcia, F., Trentalance, A. Biphasic estradiol in-
duced AKT-phosphorylation is modulated by PTEN via MAP
kinase in HepG2 cells. Mol. Biol. Cell 2003, 14: 2583-2591.
[81] Acconcia, F., Totta, P., Ogawa, S., Cardillo, I., Inoue, S., Leone, S.,
Trentalance, A., Muramatsu, M., Marino, M. Survival versus apop-
totic 17-estradiol effect: role of ER and ER activated non-
genomic signalling. J. Cell. Physiol. 2005, 203: 193-201.
[82] Bardin, A., Boulle, N., Lazennec, G., Vignon, F., Pujol, P. Loss of
ER expression as a common step in estrogen-dependent tumor
progression. Endocr. Relat. Cancer 2004, 11: 537-551.
[83] McKenna, N.J., Lanz, R.B., O’Malley, B.W. Nuclear receptor
coregulators: cellular and molecular biology. Endocr. Rev. 1999,
20: 321-344.
[84] Cavailles, V., Dauvois, S., L’Horset, F., Lopez, G., Hoare, S.,
Kushner, P.J., Parker, M.G. Nuclear factor RIP140 modulates tran-
scriptional activation by the estrogen receptor. EMBO J. 1995, 14:
3741-3751.
[85] Treuter, E., Johansson, L., Thomsen, J.S., Wärnmark, A., Leers, J.,
Pelto-Huikko, M., Sjöberg, M., Wright, A.P., Spyrou, G.,
Gustafsson, J.-Å. Competition between thyroid hormone receptor-
associated protein (TRAP) 220 and transcriptional intermediary
factor (TIF) 2 for binding to nuclear receptors. Implications for the
recruitment of TRAP and p160 coactivator complexes. J. Biol.
Chem. 1999, 274: 6667-6677.
[86] Johansson, L., Bavner, A., Thomsen, J.S., Farnegardh, M.,
Gustafsson, J.-Å., Treuter, E. The orphan nuclear receptor SHP
utilizes conserved LXXLL-related motifs for interactions with
Estrogen Signaling and Gene Transcription Current Genomics, 2006, Vol. 7, No. 8 507
ligand-activated estrogen receptors. Mol. Cell. Biol. 2000, 20:
1124-1133.
[87] Leclercq, G., Lacroix, M., Laios, I., Laurent, G. Estrogen receptor
: impact of ligands on intracellular shuttling and turnover rate in
breast cancer cells. Curr. Cancer Drug Targets 2006, 6: 39-64.
[88] Wärnmark, A., Almlöf, T., Leers, J., Gustafsson, J.-Å., Treuter, E.
Differential recruitment of the mammalian mediator subunit
TRAP220 by estrogen receptors ER and ER. J. Biol. Chem.
2001, 276: 23397-23404.
[89] Barkhem, T., Carlsson, B., Nilsson, Y., Enmark, E., Gustafsson, J.-
Å., Nilsson, S. Differential response of estrogen receptor  and es-
trogen receptor  to partial estrogen agonists/antagonists. Mol.
Pharmacol. 1998, 54: 105-112.
[90] McInerney, E.M., Weis, K.E., Sun, J., Mosselman, S., Katzenellen-
bogen, B.S. Transcription activation by the human estrogen recep-
tor subtype  (ER ) studied with ER and ER receptor chimeras.
Endocrinology 1998, 139: 4513-4522.
[91] Hall, J.M., McDonnell, D.P. The estrogen receptor -isoform
(ER) of the human estrogen receptor modulates ER transcrip-
tional activity and is a key regulator of the cellular response to es-
trogens and antiestrogens. Endocrinology 1999, 140: 5566-5578.
[92] Acevedo, M.L., Kraus, W.L. Transcriptional activation by nuclear
receptors. In: McEwan, I.J. (Ed.), Essays in Biochemistry: the Nu-
clear Receptor Superfamily. Portland Press, London, 2004, pp. 73-
88.
[93] Kim, M.Y., Woo, E.M.; Chong, Y.T., Homenko, D.R., Kraus, W.L.
Acetylation of estrogen receptor alpha by p300 at lysines 266 and
268 enhances the DNA binding and transactivation activities of the
receptor.. Mol. Endocrinol. 2006, 20: 1479-1493.
[94] Rowan, B.G., Weigel, N.L., O’Malley, B.W. Phosphorylation of
steroid receptor coactivator-1. Identification of the phosphorylation
sites and phosphorylation through the mitogen-activated protein
kinase pathway. J. Biol. Chem. 2000, 275: 4475-4483.
[95] Ko, L., Cardona, G.R., Iwasaki, T., Bramlett, K.S., Burris, T.P.,
Chin, W.W. Ser-884 adjacent to the LXXLL motif of coactivator
TRBP defines selectivity for ERs and TRs. Mol. Endocrinol. 2002,
16: 128-140.
[96] Farach-Carson, M.C., Davis, P.J. Steroid hormone interactions with
target cells: cross talk between membrane and nuclear pathways. J.
Pharmacol. Exper. Therap. 2003, 30, 839-845.
[97] Marino, M., Acconcia, F., Ascenzi, P. Estrogen receptor signalling:
Bases for drug actions. Curr. Drug Targets - Immune, Endocrine 
Metabolic Disorders 2005, 5: 305-314.
[98] Szego, C.M., Davis, J.S. Adenosine 3’,5’-monophosphate in rat
uterus: acute elevation by estrogen. Proc. Natl. Acad. Sci. U.S.A.
1967, 58: 1711-1718.
[99] Kampa, M., Castanas, E. Membrane steroid receptor signaling in
normal and neoplastic cells. Mol. Cell. Endocrinol. 2006, 246: 76-
82.
[100] Morley, P., Whitfield, J.F., Vanderhyden, B.C., Tsang, B.K.,
Schwartz, J.L. A new, nongenomic estrogen action: the rapid re-
lease of intracellular calcium. Endocrinology 1992, 131: 1305-
1312.
[101] Marino, M., Pallottini, V., Trentalance, A. Estrogens cause rapid
activation of IP3-PKC- signal transduction pathway in HEPG2
cells. Biochem. Biophys. Res. Commun. 1998, 245: 254-258.
[102] Marino, M., Ficca, R., Ascenzi, P., Trentalance, A. Nitric oxide
inhibits selectively the 17-estradiol-induced gene expression
without affecting nongenomic events in HeLa cells. Biochem. Bio-
phys. Res. Commun. 2001, 286: 529-533.
[103] Marino, M., Distefano, E., Trentalance, A., Smith, C. L. Estradiol
induced IP3 mediate the estrogen receptor activity expressed in
human cells. Mol. Cell. Endocrinol. 2001, 182: 19-26.
[104] Picotto, G., Vazquez, G., Boland, R. 17-oestradiol increases intra-
cellular Ca2+
concentration in rat enterocytes. Potential role of
phospholipase C-dependent store-operated Ca2+
influx. Biochem. J.
1999, 339: 71-77.
[105] Perret, S., Dockery, P., Harvey, B.J. 17-oestradiol stimulates
capacitative Ca2+
entry in human endometrial cells. Mol. Cell. En-
docrinol. 2001, 176: 77-84.
[106] Incerpi, S., D’Arezzo, S., Marino, M., Musanti, R., Pallottini, V.,
Pascolini, A., Trentalance, A. Short-term activation by low 17-
estradiol concentrations of the Na+
/H+
exchanger in rat aortic
smooth muscle cells: physiopathological implications. Endocrinol-
ogy 2003, 144: 4315-4324.
[107] Watters, J.J., Campbell, J.S., Cunningham, M.J., Krebs, E.G.,
Dorsa, D.M. Rapid membrane effects of steroids in neuroblastoma
cells: effects of estrogen on mitogen activated protein kinase sig-
nalling cascade and c-fos immediate early gene transcription. En-
docrinology 1997, 138: 4030-4033.
[108] Russell, K.S., Haynes, M.P., Sinha, D., Clerisme, E., Bender, J.R.
Human vascular endothelial cells contain membrane binding sites
for estradiol, which mediate rapid intracellular signaling. Proc.
Natl. Acad. Sci. U. S. A. 2000, 97, 5930–5935.
[109] Dos Santos, E.G., Dieudonne, M.N., Pecquery, R., Le Moal, V.,
Giudicelli, Y., Lacasa, D. Rapid nongenomic E2 effects on p42/p44
MAPK, activator protein-1, and cAMP response element binding
protein in rat white adipocytes. Endocrinology 2002, 143: 930-940.
[110] Migliaccio, A., Castoria, G., Di Domenico, M., de Falco, A., Bilan-
cio, A., Auricchio, F. Src is an initial target of sex steroid hormone
action. Ann. N.Y. Acad. Sci. 2002, 963: 185-190.
[111] Tanaka, Y., Gavrielides, M.V., Mitsuuchi, Y., Fujii, T., Kazanietz,
M.G. Protein kinase C promotes apoptosis in LNCaP prostate can-
cer cells through activation of p38 MAPK and inhibition of the Akt
survival pathway. J. Biol. Chem. 2003, 278: 33753-33762.
[112] Klinge, C.M., Blankenship, K.A., Risinger, K.E., Bhatnagar, S.,
Noisin, E.L., Sumanasekera, W.K., Zhao, L., Brey, D.M., Keynton,
R.S. Resveratrol and estradiol rapidly activate MAPK signaling
through estrogen receptors  and  in endothelial cells. J. Biol.
Chem. 2005, 280, 7460-7468.
[113] Woo, C.H., Lim, J.H., Kim, J.H. VCAM-1 upregulation via PKC-
p38 kinase-linked cascade mediates the TNF--induced leukocyte
adhesion and emigration in the lung airway epithelium. Am. J.
Physiol. Lung Cell. Mol. Physiol. 2005, 288: L307-L316.
[114] Castoria, G., Barone, M.V., Di Domenico, M., Bilancio, A.,
Ametrano, D., Migliaccio, A., Auricchio, F. Non-trascriptional ac-
tion of oestradiol and progestin triggers DNA synthesis. EMBO J.
1999, 18: 2500-2510.
[115] Castoria, G., Migliaccio, A., Bilancio, A., Di Domenico, M., de
Falco, A., Lombardi, M., Fiorentino, R., Varricchio, L., Barone,
M.V., Auricchio, F. PI3-kinase in concert with Src promotes the S-
phase entry of oestradiol-stimulated MCF-7 cells. EMBO J. 2001,
20: 6050-6059.
[116] Chambliss, K.L., Simon, L., Yuhanna, I.S., Mineo, C., Shaul, P.W.
Dissecting the basis of nongenomic activation of eNOS by estra-
diol: role of ER domains with known nuclear functions. Mol. En-
docrinol. 2005, 19: 277-289.
[117] Simoncini, T., Hafezi-Moghadam, A., Brazil, D.P., Ley, K., Chin,
W.W., Liao, J.K. Interaction of oestrogen receptor with the regula-
tory subunit of phosphatidylinositol-3-OH kinase. Nature 2000,
407: 538-541.
[118] Alexaki, V.I., Charalampopoulos, I., Kampa, M., Nifli, A.P., Hat-
zoglou, A., Gravanis, A., Castanas, E. Activation of membrane
estrogen receptors induce pro-survival kinases. J. Steroid Biochem.
Mol. Biol. 2006, 98: 97-110.
[119] Farhat, M.Y., Abi-Younes, S., Dingaan, B., Vargas, R., Ramwell,
P.W. Estradiol increases cyclic adenosine monophosphate in rat
pulmonary vascular smooth muscle cells by a nongenomic mecha-
nism. J. Pharmacol. Exp. Ther. 1996, 276: 652-657.
[120] Gu, Q., Moss, R.L. 17-estradiol potentiates kainate-induced cur-
rents via activation of the cAMP cascade. J. Neurosci. 1996, 16:
3620-3629.
[121] Picotto, G.; Massheimer, V., Boland, R. Acute stimulation of intes-
tinal cell calcium influx induced by 17-estradiol via the cAMP
messenger system. Mol. Cell. Endocrinol. 1996, 119: 129-134.
[122] Chen, Z.J., Yu, L., Chang, C.H. Stimulation of membrane-bound
guanylate cyclase activity by 17- estradiol. Biochem. Biophys.
Res. Commun. 1998, 252: 639-642.
[123] Malyala, A., Kelly, M.J., Ronnekleiv, O.K. Estrogen modulation of
hypothalamic neurons, activation of multiple signaling pathways
and gene expression changes. Steroids 2005, 70. 397-406.
[124] Kahlert, S., Nuedling, S., van Eickels, M., Vetter, H., Meyer, R.,
Grohe, C. Estrogen receptor  rapidly activates the IGF-1 receptor
pathway. J. Biol. Chem. 2000, 275: 18447-18453.
[125] Improta-Brears, T., Whorton, A.R., Codazzi, F., York, J.D., Meyer,
T., McDonnell, D.P. Estrogen-induced activation of mitogen-
activated protein kinase requires mobilization of intracellular cal-
cium. Proc. Natl. Acad. Sci. U. S. A. 1999, 96: 4686-4691.
[126] Driggers, P.H., Segars, J.H. Estrogen action and cytoplasmic sig-
naling pathways: Part II. The role of growth factors and phosphory-
508 Current Genomics, 2006, Vol. 7, No. 8 Marino et al.
lation in estrogen signaling. Trends Endocrinol. Metab. 2002, 13:
422-427.
[127] Razandi, M., Alton, G., Pedram, A., Ghonshani, S., Webb, P.,
Levin, E.R. Identification of a structural determinant necessary for
the localization and function of estrogen receptor  at the plasma
membrane. Mol. Cell. Biol. 2003, 23: 1633-1646.
[128] Zhang, Z., Kumar, R., Santen, R.J., Song, R.X.D. The role of
adapter protein Shc in estrogen non-genomic action. Steroids 2004,
69: 523-529.
[129] Kupzig, S., Walker, S.A., Cullen, P.J. The frequencies of calcium
oscillations are optimized for efficient calcium-mediated activation
of Ras and the ERK/MAPK cascade. Proc. Natl. Acad. Sci. U. S. A.
2005, 102: 7577-7582.
[130] Chambliss, K.L., Yuhanna, I.S., Anderson, R.G., Mendelsohn,
M.E., Shaul, P.W. ER has nongenomic action in caveolae. Mol.
Endocrinol. 2002, 16: 938-946.
[131] Kim, K.H., Bender, J.R. Rapid, estrogen receptor-mediated signal-
ing: why is the endothelium so special? Sci. STKE 2005, 14: pe28.
[132] Ansonoff, M.A., Etgen, A.M. Estradiol elevates protein kinase C
catalytic activity in the preoptic area of female rats. Endocrinology
1998, 139: 3050-3056.
[133] Razandi, M., Pedram, A., Greene, G.L., Levin, E.R. Cell membrane
and nuclear estrogen receptors (ERs) originate from a single tran-
script: studies of ER and ER expressed in Chinese hamster ovary
cells. Mol. Endocrinol. 1999, 13: 307-319.
[134] Geraldes, P., Sirois, M.G., Tanguay, J.F. Specific contribution of
estrogen receptors on mitogen-activated protein kinase pathways
and vascular cell activation. Circ. Res. 2003, 93: 399-405.
[135] Marino, M., Galluzzo, P., Leone, S., Acconcia, F., Ascenzi, P.
Nitric oxide impairs the 17-estradiol-induced apoptosis in human
colon adenocarcinoma cells. Endocr. Relat. Cancer 2006, 13: 559-
569.
[136] Acconcia, F., Kumar, R. Signaling regulation of genomic and non-
genomic functions of estrogen receptors. Cancer Lett. 2005, 238: 1-
14.
[137] Ahola, T.M., Manninen, T., Alkio, N., Ylikomi, T. G protein-
coupled receptor 30 is critical for a progestin-induced growth inhi-
bition in MCF-7 breast cancer cells. Endocrinology 2002, 143:
3376-3384.
[138] Filardo, E.J., Quinn, J.A., Frackelton, A.R. Jr., Bland, K.I. Estrogen
action via the G protein-coupled receptor, GPR30: stimulation of
adenylyl cyclase and cAMP-mediated attenuation of the epidermal
growth factor receptor-to-MAPK signaling axis. Mol. Endocrinol.
2002, 16: 70-84.
[139] Ropero, A.B., Soria, B., Nadal, A. A nonclassical estrogen mem-
brane receptor triggers rapid differential actions in the endocrine
pancreas. Mol. Endocrinol. 2002, 16: 497-505.
[140] Toran-Allerand, C.D., Guan, X., MacLusky, N.J., Horvath, T.L.,
Diano, S., Singh, M., Connolly, E.S.Jr., Nethrapalli, I.S., Tinnikov,
A.A. ER-X: a novel, plasma membrane-associated, putative estro-
gen receptor that is regulated during development and after
ischemic brain injury. J. Neurosci. 2002, 22: 8391-8401.
[141] Thomas, P., Pang, Y., Filardo, E.J., Dong, J. Identity of an estrogen
membrane receptor coupled to a G protein in human breast cancer
cells. Endocrinology 2005, 146: 624-632.
[142] Vivacqua, A., Bonofiglio, D., Recchia, A.G., Musti, A.M., Picard,
D., Andò, S., Maggiolini, M. The G protein-coupled receptor
GPR30 mediates the proliferative effects induced by 17-estradiol
and hydroxytamoxifen in endometrial cancer cells. Mol. Endocri-
nol. 2006, 20: 631-646.
[143] Pappas, T.C., Gametchu, B., Watson, C.S. Membrane estrogen
receptors identified by multiple antibody labeling and impeded-
ligand binding. FASEB J. 1995, 9: 404-410.
[144] Norfleet, A.M., Thomas, M.L., Gametchu, B., Watson, C.S. Estro-
gen receptor- detected on the plasma membrane of aldehyde-fixed
GH3/B6/F10 rat pituitary tumor cells by enzyme-linked immuno-
cytochemistry. Endocrinology 1999, 140: 3805-3814.
[145] Dan, P., Cheung, J.C., Scriven, D.R., Moore, E.D. Epitope depend-
ent localization of estrogen receptor-, but not -, in en face arte-
rial endothelium. Am. J. Physiol. 2003, 284: H1295-H1306.
[146] Acconcia, F., Ascenzi, P., Bocedi, A., Spisni, E., Tomasi, V., Tren-
talance, A., Visca, P., Marino, M. Palmitoylation-dependent estro-
gen receptor  membrane localization: regulation by 17-estradiol.
Mol. Biol. Cell 2005, 16: 231-237.
[147] Acconcia, F., Bocedi, A., Ascenzi, P., Marino, M. Does palmitoyla-
tion target estrogen receptors to plasma membrane caveolae?
IUBMB Life 2003, 55: 33-35.
[148] Bijlmakers, M.J., Marsh, M. The on-off story of protein palmitoy-
lation. Trends Cell Biol. 2003, 13: 32-42.
[149] Kennedy, A.M., Shogren, K.L., Zhang, M., Turner, R.T., Spels-
berg, T.C., Maran, A. 17-estradiol-dependent activation of signal
transducer and activator of transcription-1 in human fetal os-
teoblasts is dependent on Src kinase activity. Endocrinology 2005,
146, 201-207.
[150] Song, R.X.D., Zhang, Z., Santen, R.J. Estrogen rapid action via
protein complex formation involving ER and Src. Trends Endo-
crinol. Metab. 2005, 16: 347-353.
[151] Greger, J.G., Guo, Y., Henderson, R., Ross, J.F., Cheskis, B.J.
Characterization of MNAR expression. Steroids 2006, 71: 317-322.
[152] Song, R.X.D., Barnes, C.J., Zhang, Z., Bao, Y., Kumar, R., Santen,
R.J. The role of Shc and insulin-like growth factor 1 receptor in
mediating the translocation of estrogen receptor  to the plasma
membrane. Proc. Natl. Acad. Sci. U.S.A. 2004, 101: 2076-2081.
[153] Warner, M., Gustafsson, J.-Å. Nongenomic effects of estrogen:
why all the uncertainty? Steroids 2006, 71: 91-95.
[154] Paruthiyil, S., Parmar, H., Kerekatte, V., Cunha, G.R., Firestone,
G.L., Leitman, D.C. Estrogen receptor  inhibits human breast can-
cer cell proliferation and tumor formation by causing a G2 cell cy-
cle arrest. Cancer Res. 2004, 64: 423-428.
[155] Lu, Q., Ebling, H., Mittler, J., Baur, W.E., Karas, R.H. MAP kinase
mediates growth factor-induced nuclear translocation of estrogen
receptor . FEBS Lett. 2002, 516: 1-8.
[156] Yamakawa, K., Arita, J. Cross-talk between the estrogen receptor-,
protein kinase A-, and mitogen-activated protein kinase-mediated
signaling pathways in the regulation of lactotroph proliferation in
primary culture. J. Steroid Biochem. Mol. Biol. 2004, 88: 123-130.
[157] Pedram, A., Razandi, M., Aitkenhead, M., Hughes, C.C., Levin,
E.R. Integration of the non-genomic and genomic actions of estro-
gen. Membrane-initiated signaling by steroid to transcription and
cell biology. J. Biol. Chem. 2002, 277: 50768-50775.
[158] Wade, C.B., Dorsa, D.M. Estrogen activation of cyclic adenosine
5- monophosphate response element-mediated transcription re-
quires the extracellularly regulated kinase/mitogen-activated pro-
tein kinase pathway. Endocrinology 2003, 144: 832-838.
[159] Hennessy, B.A., Harvey, B.J., Healy, V. 17-Estradiol rapidly
stimulates c-fos expression via the MAPK pathway in T84 cells.
Mol. Cell. Endocrinol. 2005, 229: 39-47.
[160] Watters, J.J., Dorsa, D.M. Transcriptional effects of estrogen on
neuronal neurotensin gene expression involve cAMP/protein kinase
A-dependent signaling mechanisms. J. Neurosci. 1998, 18: 6672-
6680.
[161] Foster, J.S., Henley, D.C., Ahamed, S. Estrogens and cell-cycle
regulation in breast cancer. Trends Endocrinol. Metabol. 2001, 12:
320-327.
[162] Marino, M., Ascenzi, P. Estrogen receptor-: Plasma membrane
localization and functions. Immun. Endoc.  Metab. Agents in
Med. Chem. 2006, 6: 281-286.
[163] Schwabe, J.W.R., Chapman, L., Finch, J.T., Rhodes, D. The crystal
structure of the estrogen receptor DNA-binding domain bound to
DNA: how receptors discriminate between their response elements.
Cell 1993, 75: 567-578.
 
Open Peer Review
F1000 Faculty Reviews are written by members of
the prestigious  . They are
F1000 Faculty
commissioned and are peer reviewed before
publication to ensure that the final, published version
is comprehensive and accessible. The reviewers
who approved the final version are listed with their
names and affiliations.
Any comments on the article can be found at the
end of the article.
REVIEW
Re-highlighting the action of PPARγ in treating metabolic
 
diseases [version 1; peer review: 2 approved]
Sung Hee Choi ,   
Sung Soo Chung , Kyong Soo Park 1,4
Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul
National University, Seoul, South Korea
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a member of the
nuclear receptor family and plays an important role in adipocyte
differentiation, glucose homeostasis, and insulin sensitivity.
Thiazolidinediones (TZDs), synthetic ligands of PPARγ, have been used for
the treatment of diabetes mellitus for two decades. TZDs were expected to
be amazing drugs not only for type 2 diabetes but also for metabolic
syndrome and atherosclerotic vascular disease because they can reduce
both insulin resistance and inflammation in experimental studies. However,
serious unwanted effects pushed TZDs back to an optional second-tier
drug for type 2 diabetes. Nevertheless, PPARγ is still one of the most
important targets for the treatment of insulin resistance and diabetes
mellitus, and novel strategies to modulate PPARγ activity to enhance its
beneficial effects and reduce unwanted adverse effects are anticipated.
Recent studies showed that post-translational modification (PTM) of
PPARγ regulates PPARγ activity or stability and may be a novel way to
optimize PPARγ activity with reduced adverse effects. In this review, we will
focus on recent advances in PTM of PPARγ and the mechanisms
regulating PPARγ function as well as in the development of PPARγ
modulators or agonists.
Keywords
PPARgamma, post-translational modification, metabolic disease
1,2 1,3 1,4
1
2
3
4
   
Reviewer Status
  Invited Reviewers
 
version 1
published
24 Jul 2018
 
1 2
, SBP Medical Discovery Institute,
Laszlo Nagy
Florida, USA
University of Debrecen, Debrecen, Hungary
, University of Debrecen, Debrecen,
Attila Pap
Hungary
1
, Skolkovo Innovative
Alexander Orekhov
Center, Moscow, Russian Federation
2
 24 Jul 2018,  (F1000 Faculty Rev):1127 (
First published: 7
)
https://doi.org/10.12688/f1000research.14136.1
 24 Jul 2018,  (F1000 Faculty Rev):1127 (
Latest published: 7
)
https://doi.org/10.12688/f1000research.14136.1
v1
Page 1 of 9
F1000Research 2018, 7(F1000 Faculty Rev):1127 Last updated: 17 JUL 2019
 
 Kyong Soo Park ( )
Corresponding author: kspark@snu.ac.kr
  : Conceptualization, Data Curation, Resources, Writing – Original Draft Preparation, Writing – Review  Editing;  :
Author roles: Choi SH Chung SS
Conceptualization, Data Curation, Resources, Supervision, Writing – Original Draft Preparation;  : Conceptualization, Data Curation,
Park KS
Resources, Supervision, Writing – Review  Editing
 No competing interests were disclosed.
Competing interests:
 This work was supported by National Research Foundation Grant by Ministry of Science and ICT, Republic of Korea
Grant information:
(NRF-2016R1A2B3010373).
The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
 © 2018 Choi SH  . This is an open access article distributed under the terms of the  , which
Copyright: et al Creative Commons Attribution Licence
permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
 Choi SH, Chung SS and Park KS. 
How to cite this article: Re-highlighting the action of PPARγ in treating metabolic diseases [version 1;
 F1000Research 2018,  (F1000 Faculty Rev):1127 ( )
peer review: 2 approved] 7 https://doi.org/10.12688/f1000research.14136.1
 24 Jul 2018,  (F1000 Faculty Rev):1127 ( ) 
First published: 7 https://doi.org/10.12688/f1000research.14136.1
Page 2 of 9
F1000Research 2018, 7(F1000 Faculty Rev):1127 Last updated: 17 JUL 2019
Introduction
Insulin resistance is the key pathophysiologic abnormality of
many metabolic diseases such as type 2 diabetes mellitus, obesity,
dyslipidemia, and cardiovascular diseases1
. Therefore, reducing
insulin resistance is the most important strategy for improving
metabolic deterioration. Thiazolidinediones (TZDs), peroxisome
proliferator-activated receptor γ (PPARγ) agonists, have shown
many beneficial effects not only by enhancing insulin sensitiv-
ity but also by demonstrating anti-inflammatory and antioxidant
properties, whose actions are related to anti-atherosclerosis2,3
.
Thus, TZDs were considered a magic bullet for the treatment
of type 2 diabetes and atherosclerosis. Indeed, TZDs dem-
onstrated a preventive role for recurrent ischemic stroke in
several clinical trials4
and for restenosis after percutaneous
coronary intervention (PCI)5–7
. However, TZDs increased the risk
of peripheral edema, bone loss, and congestive heart failure8–10
.
A meta-analysis of clinical trials showed that rosiglitazone sig-
nificantly increased the risk of myocardial infarction11
. Although
later studies revealed that rosiglitazone did not increase the risk
of heart attack and the US Food and Drug Administration (FDA)
removed the warning labels from rosiglitazone-containing
drugs regarding the issue of increasing heart attack in 2013,
rosiglitazone’s cardiovascular safety issue alongside the
above-mentioned adverse effects still lead to many physicians
hesitating to prescribe TZDs in their clinical practice. Never-
theless, PPARγ is still one of the most important targets for the
treatment of insulin resistance and type 2 diabetes, and novel
strategies to modulate PPARγ activity to enhance its beneficial
effects and reduce unwanted adverse effects are strongly
anticipated. Recent studies showed that post-translational modi-
fication (PTM) of PPARγ regulates PPARγ activity or stability
and may be a novel way to optimize PPARγ activity with
reduced adverse effects. In addition, selective PPARγ modulators
(sPPARγMs), dual or pan PPAR agonists, have been developed and
tested for their metabolic effects in animal studies and in some
clinical trials.
PPARγ, a therapeutic target for insulin resistance
(Figure 1)
PPARγ is a master regulator of adipocyte differentiation. It is
also involved in glucose homeostasis and insulin sensitivity.
The expression of PPARγ is most abundant in adipose tissue12
.
Evidence has shown that the primary target of TZDs is adipose
tissue, where it increases the expression of Glut4 and CAP13
, and
an animal model lacking PPARγ in adipose tissue had a signifi-
cantly lower response to TZDs14,15
. TZDs inhibit the expression
of TNF-α, IL-6, and resistin in adipose tissue, which promote
insulin resistance and chronic inflammation16,17
, while TZDs
increased the production of adiponectin and fibroblast growth
factor 21 (FGF21), which enhance fatty acid oxidation and
insulin sensitivity18,19
. TZDs increase lipogenesis by aP2, LPL,
CD36, fatty acid transport protein, PEPCK, and the glyc-
erol transporter aquaporin 72
and make adipose tissue store
more lipid, while TZDs remove lipid accumulation in other
tissues such as muscle and liver20
.
From these studies, it seems that improvement of insulin sen-
sitivity in liver and muscle might be secondary to the effects
of TZDs in adipose tissue. However, there is also evidence
showing that TZDs have an insulin-sensitizing effect on other
peripheral organs. It has been demonstrated that ablation of
liver PPARγ in mice reduced hepatic steatosis but worsened
hyperlipidemia, triglyceride clearance, and muscle insulin
resistance21
. The expression of PPARγ in skeletal muscle is
relatively low compared to adipose tissue, and the physiological
significance of PPARγ in skeletal muscle has been shown to work
indirectly in previous studies22
. However, selective activation of
PPARγ in skeletal muscle showed significant protection from
high-fat diet-induced insulin resistance and associated changes
in muscle phenotype, such as decreasing the quantity of lipid
in myocytes and increasing the number of oxidative muscle
fiber types23
. It suggests that the activation of PPARγ can
act directly on muscle tissue to improve insulin sensitivity.
Macrophage PPARγ is also implicated in anti-inflammation and
lipid metabolism24
, and mice lacking macrophage PPARγ are
more prone to whole-body insulin resistance25,26
.
PPARγ agonists and their effects on the vascular
system: friend or foe?
PPARγ is expressed in the endothelium and vascular smooth
muscle in the blood vessel wall27,28
. Despite controversial car-
diovascular effects of TZDs in humans, most experimental
studies showed beneficial effects on vascular systems. TZDs
inhibit the proliferation and migration of vascular smooth
muscle cells (VSMCs), with potential favorable effects on
atherosclerosis29,30
. Smooth muscle-specific dominant-negative
PPARγ transgenic mice showed a loss of nitric oxide responsive-
ness and high contractility31
, which resulted in systolic hyper-
tension. In humans, dominant-negative mutations of PPARγ are
associated with early hypertension and insulin resistance32
.
Activation of PPARγ inhibits CCAAT/enhancer-binding
protein-δ (C/EBPδ), which is a well-known mediator of the
proinflammatory response in vascular cells33
.
TZDs also reduce activation and inflammation in endothelial
cells by suppressing the expression of inflammation-associated
genes34–37
. On the other hand, TZDs induce vascular endothe-
lial growth factor (VEGF) in endothelial cells and increase
endothelial cell proliferation and migration by the Akt-dependent
pathway38–40
. In recent data, rosiglitazone significantly increased
endothelial cell migration and vascular leakage in an animal
study with increased VEGF expression and suppressed tight
junction proteins, which caused instability of the endothelial
membrane41
. This result could be related to vascular permeabil-
ity, peripheral edema, and congestive heart failure associated with
the use of TZDs, contrary to their beneficial effect on vascular
cells. We still need more concrete evidence to understand the role
of TZDs in the whole vascular system under various conditions.
Regulation of PPARγ by PTMs to reduce the side
effects of TZDs
The PTM of PPARγ involves several pathways, including
phosphorylation, SUMOylation, ubiquitination, β-O-linked
N-acetylglucosamine modification (O-GlcNAcylation), and
acetylation. These PTMs are known to regulate both PPARγ
expression and its transcriptional activity42
and have been recently
suggested as a good modality for reducing the side effects of
PPARγ activation by TZDs43
(Figure 2).
Page 3 of 9
F1000Research 2018, 7(F1000 Faculty Rev):1127 Last updated: 17 JUL 2019
Phosphorylation
Phosphorylation at serine 112 (S112) in the N-terminal AF-1
domain was first identified, and various studies revealed that net
results of PPARγ phosphorylation may inhibit or stimulate its
transcriptional activity depending on the cellular contexts and
kinases involved44–48
. Phosphorylation at S273 in the ligand-
binding domain is mediated by cyclin-dependent kinase
5 (Cdk5), which is activated by pro-inflammatory stimuli and
free fatty acids49
. S273 phosphorylation affects the expression of
insulin-sensitizing adipokines such as adiponectin and adipsin
but not those affecting adipogenesis. PPARγ partial agonist
MRL24 specifically blocks the phosphorylation of PPARγ at
S273 and has higher anti-diabetic activity and fewer side effects
than does rosiglitazone49
. SR1664 and similar non-agonist PPARγ
ligands were also developed for blocking cdk5-mediated
phosphorylation and showed improved insulin sensitivity in
high-fat diet-fed mice without causing side effects such as
fluid retention and weight gain43,50
. More recently, it has been
reported that phosphorylation at S273 is also facilitated by
MEK/ERK, and inhibition of MEK and ERK improves insu-
lin resistance, suggesting that MEK and ERK inhibitors can
be therapeutic targets for diabetes through the modulation of
PPARγ function51
.
SUMOylation
Small ubiquitin-like modifier (SUMO) modification is a revers-
ible process and may affect protein stability, transcriptional
activity, and protein–protein interaction. PPARγ is known
Figure 1. Effect of PPARγ activation on various tissues. ACSL1, acyl-CoA synthetase long chain family member 1; CD36, cluster of
differentiation 36; CPT1b, carnitine palmitoyltransferase 1B; FA, fatty acid; FFA, free fatty acid; FGF, fibroblast growth factor; GK, glucokinase;
GLUT, glucose transporter; GlyK, glycerol kinase; IRS2, insulin receptor substrate 1; LPL, lipoprotein lipase; NF-κB, nuclear factor kappa-light-
chain-enhancer of activated B cells; MCP-1, monocyte chemoattractant protein 1; MMP-9, matrix metalloproteinase 9; NO, nitric oxide; PAI-1,
plasminogen activator inhibitor type 1; PCK2, peroxisome proliferator-activated receptor gamma 2 binding site; PEPCK, phosphoenolpyruvate
carboxykinase; PI3K, phosphoinositide 3-kinase; PPARγ, peroxisome proliferator-activated receptor γ; PPRE, peroxisome proliferator-activated
receptor response element; RXR, retinoid X receptor; STAT6, signal transducer and activator of transcription 6; TG, triglyceride; TNF-α, tumor
necrosis factor α; TZD, thiazolidinedione; VCAM-1, vascular cell adhesion molecule-1; VEGF, vascular endothelial growth factor.
Page 4 of 9
F1000Research 2018, 7(F1000 Faculty Rev):1127 Last updated: 17 JUL 2019
Figure 2. Regulation of PPARγ by post translational modification. Ac, acetyl; ACSL1, acyl-CoA synthetase long chain family
member 1; AF1, activation function 1; AF2, activation function 2; CD36, cluster of differentiation 36; Cdk, cyclin-dependent kinase; CPT1b,
carnitine palmitoyltransferase 1B; DBD, DNA-binding domain; FABP3, fatty-acid-binding protein 3, muscle and heart; FAO, fatty acid
oxidation; FGF, fibroblast growth factor; GlcNAC, N-acetylglucosamine; LBD, ligand-binding domain; Lys, lysine; MAPK, mitogen-activated
protein kinase; P, phosphate; PPARγ, peroxisome proliferator-activated receptor γ; PRDM16, PR domain containing 16; SENP2, small
ubiquitin-like modifier-specific protease 2; Ser, serine; SIRT1, sirtuin 1; SUMO, small ubiquitin-like modifier; Thr, threonine; Ub, ubiquitin;
WAT, white adipose tissue.
as a target of SUMOylation. Lysine 107 (K107) of PPARγ2
is the major SUMOylation site, and deSUMOylation of this
site increases the transcriptional activity of PPARγ52
. The
K107R mutant form of PPARγ stimulates adipogenesis and sup-
presses neointimal formation after balloon injury more effec-
tively than does the PPARγ wild-type form53,54
. SUMOylation
at K107 of PPARγ may be linked to S112 phosphorylation53
.
PPARγ SUMOylation at K107 is markedly increased in
FGF21-knockout mice, suggesting that FGF21 regulates PPARγ
SUMOylation by an unknown mechanism19
. SUMOylation of
PPARγ at K395 (K365 of PPARγ1) is stimulated by PPARγ
agonists, and this modification inhibits the transcription of inflam-
matory response genes, such as iNOS, through recruiting transcrip-
tional repressors to the NFkB complex in macrophages55
.
SUMO-specific protease 2 (SENP2) is the major deSUMOylation
enzyme of PPARγ56
. Overexpression of SENP2 in C2C12 cells
effectively induces PPARγ target genes such as Fabp3 and Cd36
but not Adrp; thus, SENP2 can induce the expression of PPARγ
target genes in a selective manner56
. SENP2 deSUMOylates
PPARγ and PPARδ and activates genes involved in fatty acid
oxidation such as Cpt1b and Acsl1, which results in an increase
of fatty acid oxidation in muscle. Interestingly, palmitate
increases SENP2 expression via the TLR4-MyD88-NFkB
pathway. These results suggest that SENP2 is an important
regulator of fatty acid metabolism in skeletal muscle57
.
Ubiquitination
Ubiquitination is the covalent attachment of ubiquitin, a 76-
amino-acid peptide, to lysine residues in the substrate protein.
PPARγ has a short half-life and is degraded by the polyubiquitin-
proteasome pathway58
. Inhibition of proteasome activity by
proteasome inhibitors increases PPARγ stability, suggesting
ubiquitin modification of PPARγ is an important determinant of
PPARγ activity58
. Several ubiquitin ligases, such as FBOX9 and
Cul4B, and an ubiquitin-specific protease (HAUSP) targeting
PPARγ have been identified, and an increase in PPARγ stability
generally promotes PPARγ activity and adipogenesis59–61
. Inter-
estingly, PPARγ agonists, TZDs, stimulate the ubiquitination of
PPARγ, which can be mediated by an ubiquitin ligase, Siah158,62
.
Therefore, PPARγ ubiquitination may be differently regulated
by several ubiquitin E3 ligases or proteases upon various
conditions.
O-GlcNAcylation
O-GlcNAcylation is the post-translational cycling of a single
β-O-linked N-acetylglucosamine (O-GlcNAc) on the hydroxyl
groups of serine or threonine residues of target proteins. A major
Page 5 of 9
F1000Research 2018, 7(F1000 Faculty Rev):1127 Last updated: 17 JUL 2019
Soal dan Pembahasan Farmakologi Molekular - PPAR dan Reseptor Estrogen
Soal dan Pembahasan Farmakologi Molekular - PPAR dan Reseptor Estrogen
Soal dan Pembahasan Farmakologi Molekular - PPAR dan Reseptor Estrogen
Soal dan Pembahasan Farmakologi Molekular - PPAR dan Reseptor Estrogen

More Related Content

Similar to Soal dan Pembahasan Farmakologi Molekular - PPAR dan Reseptor Estrogen

Control of gene expression in plants
Control of gene expression in plantsControl of gene expression in plants
Control of gene expression in plantsAbhilash Panju
 
Eukaryotic and prokaryotic_gene_structure
Eukaryotic and prokaryotic_gene_structureEukaryotic and prokaryotic_gene_structure
Eukaryotic and prokaryotic_gene_structureDaniel Murillo
 
Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)
Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)
Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)Daire Murphy
 
Horrix et al. - 2010
Horrix et al. - 2010Horrix et al. - 2010
Horrix et al. - 2010Cristina Voss
 
Role of-mek1-in-tlr4-mediated-signaling
Role of-mek1-in-tlr4-mediated-signalingRole of-mek1-in-tlr4-mediated-signaling
Role of-mek1-in-tlr4-mediated-signalingscience journals
 
Lecture notes GENE REGULATION IN EUKARYOTES.pdf
Lecture notes GENE REGULATION IN EUKARYOTES.pdfLecture notes GENE REGULATION IN EUKARYOTES.pdf
Lecture notes GENE REGULATION IN EUKARYOTES.pdfKristu Jayanti College
 
Epigenetic regulation in plants
Epigenetic regulation in plantsEpigenetic regulation in plants
Epigenetic regulation in plantsRyza Priatama
 
CHEM1110 Chemistry For The Life Sciences I.docx
CHEM1110 Chemistry For The Life Sciences I.docxCHEM1110 Chemistry For The Life Sciences I.docx
CHEM1110 Chemistry For The Life Sciences I.docxwrite31
 
CELLULAR REPROGRAMMING: Current Technology, Perspectives and Generation of iP...
CELLULAR REPROGRAMMING: Current Technology, Perspectives and Generation of iP...CELLULAR REPROGRAMMING: Current Technology, Perspectives and Generation of iP...
CELLULAR REPROGRAMMING: Current Technology, Perspectives and Generation of iP...Munna Yadav
 
Receptors with intrinsic protein kinase activity
Receptors with intrinsic protein kinase activityReceptors with intrinsic protein kinase activity
Receptors with intrinsic protein kinase activityDr.M.Prasad Naidu
 
RAGE-Mediated Cell Signaling.pdf
RAGE-Mediated Cell Signaling.pdfRAGE-Mediated Cell Signaling.pdf
RAGE-Mediated Cell Signaling.pdfNazmunNahar479158
 
Genetic fine structure
Genetic fine structureGenetic fine structure
Genetic fine structureSujan Karki
 
Mod 4 regulation of gene expression -notes SH.pdf
Mod 4 regulation of gene expression -notes SH.pdfMod 4 regulation of gene expression -notes SH.pdf
Mod 4 regulation of gene expression -notes SH.pdfMSCW Mysore
 
M Sc Molecular Biology Final- project SV.pptx
M Sc Molecular Biology Final-  project SV.pptxM Sc Molecular Biology Final-  project SV.pptx
M Sc Molecular Biology Final- project SV.pptxOmekhan1
 
Eicosanoids: lipid mediators of inflammation in transplantation
Eicosanoids: lipid mediators of inflammation in transplantationEicosanoids: lipid mediators of inflammation in transplantation
Eicosanoids: lipid mediators of inflammation in transplantationFederal University of Bahia
 

Similar to Soal dan Pembahasan Farmakologi Molekular - PPAR dan Reseptor Estrogen (20)

fujioka2004
fujioka2004fujioka2004
fujioka2004
 
Control of gene expression in plants
Control of gene expression in plantsControl of gene expression in plants
Control of gene expression in plants
 
Eukaryotic and prokaryotic_gene_structure
Eukaryotic and prokaryotic_gene_structureEukaryotic and prokaryotic_gene_structure
Eukaryotic and prokaryotic_gene_structure
 
Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)
Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)
Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)
 
Horrix et al. - 2010
Horrix et al. - 2010Horrix et al. - 2010
Horrix et al. - 2010
 
Role of-mek1-in-tlr4-mediated-signaling
Role of-mek1-in-tlr4-mediated-signalingRole of-mek1-in-tlr4-mediated-signaling
Role of-mek1-in-tlr4-mediated-signaling
 
Lecture notes GENE REGULATION IN EUKARYOTES.pdf
Lecture notes GENE REGULATION IN EUKARYOTES.pdfLecture notes GENE REGULATION IN EUKARYOTES.pdf
Lecture notes GENE REGULATION IN EUKARYOTES.pdf
 
Peroxisomes in dermatology
Peroxisomes in dermatologyPeroxisomes in dermatology
Peroxisomes in dermatology
 
Ch17 217 228
Ch17 217 228Ch17 217 228
Ch17 217 228
 
Epigenetic regulation in plants
Epigenetic regulation in plantsEpigenetic regulation in plants
Epigenetic regulation in plants
 
CHEM1110 Chemistry For The Life Sciences I.docx
CHEM1110 Chemistry For The Life Sciences I.docxCHEM1110 Chemistry For The Life Sciences I.docx
CHEM1110 Chemistry For The Life Sciences I.docx
 
CELLULAR REPROGRAMMING: Current Technology, Perspectives and Generation of iP...
CELLULAR REPROGRAMMING: Current Technology, Perspectives and Generation of iP...CELLULAR REPROGRAMMING: Current Technology, Perspectives and Generation of iP...
CELLULAR REPROGRAMMING: Current Technology, Perspectives and Generation of iP...
 
ERAP zero.pptx
ERAP zero.pptxERAP zero.pptx
ERAP zero.pptx
 
Receptors with intrinsic protein kinase activity
Receptors with intrinsic protein kinase activityReceptors with intrinsic protein kinase activity
Receptors with intrinsic protein kinase activity
 
RAGE-Mediated Cell Signaling.pdf
RAGE-Mediated Cell Signaling.pdfRAGE-Mediated Cell Signaling.pdf
RAGE-Mediated Cell Signaling.pdf
 
Genetic fine structure
Genetic fine structureGenetic fine structure
Genetic fine structure
 
Mod 4 regulation of gene expression -notes SH.pdf
Mod 4 regulation of gene expression -notes SH.pdfMod 4 regulation of gene expression -notes SH.pdf
Mod 4 regulation of gene expression -notes SH.pdf
 
M Sc Molecular Biology Final- project SV.pptx
M Sc Molecular Biology Final-  project SV.pptxM Sc Molecular Biology Final-  project SV.pptx
M Sc Molecular Biology Final- project SV.pptx
 
Genetic regulation 06 08-13
Genetic regulation    06 08-13Genetic regulation    06 08-13
Genetic regulation 06 08-13
 
Eicosanoids: lipid mediators of inflammation in transplantation
Eicosanoids: lipid mediators of inflammation in transplantationEicosanoids: lipid mediators of inflammation in transplantation
Eicosanoids: lipid mediators of inflammation in transplantation
 

More from Nesha Mutiara

Pemantauan Terapi Obat Pasien Diabetes, Hipertensi, dan Hipoalbuminemia
Pemantauan Terapi Obat Pasien Diabetes, Hipertensi, dan HipoalbuminemiaPemantauan Terapi Obat Pasien Diabetes, Hipertensi, dan Hipoalbuminemia
Pemantauan Terapi Obat Pasien Diabetes, Hipertensi, dan HipoalbuminemiaNesha Mutiara
 
Farmakoterapi Infeksi : Studi Kasus Tatalaksana Malaria
Farmakoterapi Infeksi : Studi Kasus Tatalaksana MalariaFarmakoterapi Infeksi : Studi Kasus Tatalaksana Malaria
Farmakoterapi Infeksi : Studi Kasus Tatalaksana MalariaNesha Mutiara
 
Farmakoterapi Infeksi : Studi Kasus Tatalaksana TBC Kategori 1
Farmakoterapi Infeksi : Studi Kasus Tatalaksana TBC Kategori 1Farmakoterapi Infeksi : Studi Kasus Tatalaksana TBC Kategori 1
Farmakoterapi Infeksi : Studi Kasus Tatalaksana TBC Kategori 1Nesha Mutiara
 
Farmakoterapi Infeksi : Studi Kasus Tatalaksana TBC MDR
Farmakoterapi Infeksi : Studi Kasus Tatalaksana TBC MDRFarmakoterapi Infeksi : Studi Kasus Tatalaksana TBC MDR
Farmakoterapi Infeksi : Studi Kasus Tatalaksana TBC MDRNesha Mutiara
 
Farmasi Klinik - Medication Error di Bidang Onkologi
Farmasi Klinik - Medication Error di Bidang OnkologiFarmasi Klinik - Medication Error di Bidang Onkologi
Farmasi Klinik - Medication Error di Bidang OnkologiNesha Mutiara
 
Farmakoterapi Renal : Studi Kasus CKD (Chronic Kidney Disease)
Farmakoterapi Renal : Studi Kasus CKD (Chronic Kidney Disease)Farmakoterapi Renal : Studi Kasus CKD (Chronic Kidney Disease)
Farmakoterapi Renal : Studi Kasus CKD (Chronic Kidney Disease)Nesha Mutiara
 
Farmakoterapi Syaraf : Studi Kasus Stroke
Farmakoterapi Syaraf : Studi Kasus StrokeFarmakoterapi Syaraf : Studi Kasus Stroke
Farmakoterapi Syaraf : Studi Kasus StrokeNesha Mutiara
 
Pharmaceutical Care : Evaluasi Literatur Uji Klinik
Pharmaceutical Care : Evaluasi Literatur Uji KlinikPharmaceutical Care : Evaluasi Literatur Uji Klinik
Pharmaceutical Care : Evaluasi Literatur Uji KlinikNesha Mutiara
 
Farmasi Klinik : Studi Kasus Diabetes Ketoasidosis
Farmasi Klinik : Studi Kasus Diabetes KetoasidosisFarmasi Klinik : Studi Kasus Diabetes Ketoasidosis
Farmasi Klinik : Studi Kasus Diabetes KetoasidosisNesha Mutiara
 
Bioanalisis - Penentuan Bioekivalensi Obat Sulfametoksazol
Bioanalisis - Penentuan Bioekivalensi Obat SulfametoksazolBioanalisis - Penentuan Bioekivalensi Obat Sulfametoksazol
Bioanalisis - Penentuan Bioekivalensi Obat SulfametoksazolNesha Mutiara
 
Bioanalisis - Uji Sensitivitas Metode Spektrofotometri Visible Berdasarkan Pe...
Bioanalisis - Uji Sensitivitas Metode Spektrofotometri Visible Berdasarkan Pe...Bioanalisis - Uji Sensitivitas Metode Spektrofotometri Visible Berdasarkan Pe...
Bioanalisis - Uji Sensitivitas Metode Spektrofotometri Visible Berdasarkan Pe...Nesha Mutiara
 
Farmasi Rumah Sakit - Evaluasi Penggunaan Antibiotika dengan Metode Gyssens
Farmasi Rumah Sakit - Evaluasi Penggunaan Antibiotika dengan Metode GyssensFarmasi Rumah Sakit - Evaluasi Penggunaan Antibiotika dengan Metode Gyssens
Farmasi Rumah Sakit - Evaluasi Penggunaan Antibiotika dengan Metode GyssensNesha Mutiara
 
Kapita Selekta Apoteker - Cara Penyimpanan yang Baik pada Produk Vaksin Coron...
Kapita Selekta Apoteker - Cara Penyimpanan yang Baik pada Produk Vaksin Coron...Kapita Selekta Apoteker - Cara Penyimpanan yang Baik pada Produk Vaksin Coron...
Kapita Selekta Apoteker - Cara Penyimpanan yang Baik pada Produk Vaksin Coron...Nesha Mutiara
 
Kapita Selekta Apoteker - Implementasi Home Care Telepharmacy dalam Monitorin...
Kapita Selekta Apoteker - Implementasi Home Care Telepharmacy dalam Monitorin...Kapita Selekta Apoteker - Implementasi Home Care Telepharmacy dalam Monitorin...
Kapita Selekta Apoteker - Implementasi Home Care Telepharmacy dalam Monitorin...Nesha Mutiara
 
Kapita Selekta Apoteker - Kompetensi Apoteker Sebagai Pendukung Kemajuan Indu...
Kapita Selekta Apoteker - Kompetensi Apoteker Sebagai Pendukung Kemajuan Indu...Kapita Selekta Apoteker - Kompetensi Apoteker Sebagai Pendukung Kemajuan Indu...
Kapita Selekta Apoteker - Kompetensi Apoteker Sebagai Pendukung Kemajuan Indu...Nesha Mutiara
 
Farmasi Rumah Sakit - Sistem Distribusi Obat di Rumah Sakit
Farmasi Rumah Sakit - Sistem Distribusi Obat di Rumah SakitFarmasi Rumah Sakit - Sistem Distribusi Obat di Rumah Sakit
Farmasi Rumah Sakit - Sistem Distribusi Obat di Rumah SakitNesha Mutiara
 
Rangkuman Obat Off Label
Rangkuman Obat Off LabelRangkuman Obat Off Label
Rangkuman Obat Off LabelNesha Mutiara
 
Laporan Praktikum Bioanalisis - Uji Ekivalensi In Vitro
Laporan Praktikum Bioanalisis - Uji Ekivalensi In VitroLaporan Praktikum Bioanalisis - Uji Ekivalensi In Vitro
Laporan Praktikum Bioanalisis - Uji Ekivalensi In VitroNesha Mutiara
 
Farmasi Rumah Sakit - Tingkat Kelulusan Akreditasi dan Manajemen Organisasi RS
Farmasi Rumah Sakit - Tingkat Kelulusan Akreditasi dan Manajemen Organisasi RS Farmasi Rumah Sakit - Tingkat Kelulusan Akreditasi dan Manajemen Organisasi RS
Farmasi Rumah Sakit - Tingkat Kelulusan Akreditasi dan Manajemen Organisasi RS Nesha Mutiara
 
Formulasi Sediaan Steril Vial Anestesi Lokal (Lidokain HCl)
Formulasi Sediaan Steril Vial Anestesi Lokal (Lidokain HCl)Formulasi Sediaan Steril Vial Anestesi Lokal (Lidokain HCl)
Formulasi Sediaan Steril Vial Anestesi Lokal (Lidokain HCl)Nesha Mutiara
 

More from Nesha Mutiara (20)

Pemantauan Terapi Obat Pasien Diabetes, Hipertensi, dan Hipoalbuminemia
Pemantauan Terapi Obat Pasien Diabetes, Hipertensi, dan HipoalbuminemiaPemantauan Terapi Obat Pasien Diabetes, Hipertensi, dan Hipoalbuminemia
Pemantauan Terapi Obat Pasien Diabetes, Hipertensi, dan Hipoalbuminemia
 
Farmakoterapi Infeksi : Studi Kasus Tatalaksana Malaria
Farmakoterapi Infeksi : Studi Kasus Tatalaksana MalariaFarmakoterapi Infeksi : Studi Kasus Tatalaksana Malaria
Farmakoterapi Infeksi : Studi Kasus Tatalaksana Malaria
 
Farmakoterapi Infeksi : Studi Kasus Tatalaksana TBC Kategori 1
Farmakoterapi Infeksi : Studi Kasus Tatalaksana TBC Kategori 1Farmakoterapi Infeksi : Studi Kasus Tatalaksana TBC Kategori 1
Farmakoterapi Infeksi : Studi Kasus Tatalaksana TBC Kategori 1
 
Farmakoterapi Infeksi : Studi Kasus Tatalaksana TBC MDR
Farmakoterapi Infeksi : Studi Kasus Tatalaksana TBC MDRFarmakoterapi Infeksi : Studi Kasus Tatalaksana TBC MDR
Farmakoterapi Infeksi : Studi Kasus Tatalaksana TBC MDR
 
Farmasi Klinik - Medication Error di Bidang Onkologi
Farmasi Klinik - Medication Error di Bidang OnkologiFarmasi Klinik - Medication Error di Bidang Onkologi
Farmasi Klinik - Medication Error di Bidang Onkologi
 
Farmakoterapi Renal : Studi Kasus CKD (Chronic Kidney Disease)
Farmakoterapi Renal : Studi Kasus CKD (Chronic Kidney Disease)Farmakoterapi Renal : Studi Kasus CKD (Chronic Kidney Disease)
Farmakoterapi Renal : Studi Kasus CKD (Chronic Kidney Disease)
 
Farmakoterapi Syaraf : Studi Kasus Stroke
Farmakoterapi Syaraf : Studi Kasus StrokeFarmakoterapi Syaraf : Studi Kasus Stroke
Farmakoterapi Syaraf : Studi Kasus Stroke
 
Pharmaceutical Care : Evaluasi Literatur Uji Klinik
Pharmaceutical Care : Evaluasi Literatur Uji KlinikPharmaceutical Care : Evaluasi Literatur Uji Klinik
Pharmaceutical Care : Evaluasi Literatur Uji Klinik
 
Farmasi Klinik : Studi Kasus Diabetes Ketoasidosis
Farmasi Klinik : Studi Kasus Diabetes KetoasidosisFarmasi Klinik : Studi Kasus Diabetes Ketoasidosis
Farmasi Klinik : Studi Kasus Diabetes Ketoasidosis
 
Bioanalisis - Penentuan Bioekivalensi Obat Sulfametoksazol
Bioanalisis - Penentuan Bioekivalensi Obat SulfametoksazolBioanalisis - Penentuan Bioekivalensi Obat Sulfametoksazol
Bioanalisis - Penentuan Bioekivalensi Obat Sulfametoksazol
 
Bioanalisis - Uji Sensitivitas Metode Spektrofotometri Visible Berdasarkan Pe...
Bioanalisis - Uji Sensitivitas Metode Spektrofotometri Visible Berdasarkan Pe...Bioanalisis - Uji Sensitivitas Metode Spektrofotometri Visible Berdasarkan Pe...
Bioanalisis - Uji Sensitivitas Metode Spektrofotometri Visible Berdasarkan Pe...
 
Farmasi Rumah Sakit - Evaluasi Penggunaan Antibiotika dengan Metode Gyssens
Farmasi Rumah Sakit - Evaluasi Penggunaan Antibiotika dengan Metode GyssensFarmasi Rumah Sakit - Evaluasi Penggunaan Antibiotika dengan Metode Gyssens
Farmasi Rumah Sakit - Evaluasi Penggunaan Antibiotika dengan Metode Gyssens
 
Kapita Selekta Apoteker - Cara Penyimpanan yang Baik pada Produk Vaksin Coron...
Kapita Selekta Apoteker - Cara Penyimpanan yang Baik pada Produk Vaksin Coron...Kapita Selekta Apoteker - Cara Penyimpanan yang Baik pada Produk Vaksin Coron...
Kapita Selekta Apoteker - Cara Penyimpanan yang Baik pada Produk Vaksin Coron...
 
Kapita Selekta Apoteker - Implementasi Home Care Telepharmacy dalam Monitorin...
Kapita Selekta Apoteker - Implementasi Home Care Telepharmacy dalam Monitorin...Kapita Selekta Apoteker - Implementasi Home Care Telepharmacy dalam Monitorin...
Kapita Selekta Apoteker - Implementasi Home Care Telepharmacy dalam Monitorin...
 
Kapita Selekta Apoteker - Kompetensi Apoteker Sebagai Pendukung Kemajuan Indu...
Kapita Selekta Apoteker - Kompetensi Apoteker Sebagai Pendukung Kemajuan Indu...Kapita Selekta Apoteker - Kompetensi Apoteker Sebagai Pendukung Kemajuan Indu...
Kapita Selekta Apoteker - Kompetensi Apoteker Sebagai Pendukung Kemajuan Indu...
 
Farmasi Rumah Sakit - Sistem Distribusi Obat di Rumah Sakit
Farmasi Rumah Sakit - Sistem Distribusi Obat di Rumah SakitFarmasi Rumah Sakit - Sistem Distribusi Obat di Rumah Sakit
Farmasi Rumah Sakit - Sistem Distribusi Obat di Rumah Sakit
 
Rangkuman Obat Off Label
Rangkuman Obat Off LabelRangkuman Obat Off Label
Rangkuman Obat Off Label
 
Laporan Praktikum Bioanalisis - Uji Ekivalensi In Vitro
Laporan Praktikum Bioanalisis - Uji Ekivalensi In VitroLaporan Praktikum Bioanalisis - Uji Ekivalensi In Vitro
Laporan Praktikum Bioanalisis - Uji Ekivalensi In Vitro
 
Farmasi Rumah Sakit - Tingkat Kelulusan Akreditasi dan Manajemen Organisasi RS
Farmasi Rumah Sakit - Tingkat Kelulusan Akreditasi dan Manajemen Organisasi RS Farmasi Rumah Sakit - Tingkat Kelulusan Akreditasi dan Manajemen Organisasi RS
Farmasi Rumah Sakit - Tingkat Kelulusan Akreditasi dan Manajemen Organisasi RS
 
Formulasi Sediaan Steril Vial Anestesi Lokal (Lidokain HCl)
Formulasi Sediaan Steril Vial Anestesi Lokal (Lidokain HCl)Formulasi Sediaan Steril Vial Anestesi Lokal (Lidokain HCl)
Formulasi Sediaan Steril Vial Anestesi Lokal (Lidokain HCl)
 

Recently uploaded

Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111Sapana Sha
 
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxSOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxiammrhaywood
 
Employee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxEmployee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxNirmalaLoungPoorunde1
 
Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)eniolaolutunde
 
Pharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdfPharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdfMahmoud M. Sallam
 
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdfBASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdfSoniaTolstoy
 
Computed Fields and api Depends in the Odoo 17
Computed Fields and api Depends in the Odoo 17Computed Fields and api Depends in the Odoo 17
Computed Fields and api Depends in the Odoo 17Celine George
 
Mastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory InspectionMastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory InspectionSafetyChain Software
 
ENGLISH5 QUARTER4 MODULE1 WEEK1-3 How Visual and Multimedia Elements.pptx
ENGLISH5 QUARTER4 MODULE1 WEEK1-3 How Visual and Multimedia Elements.pptxENGLISH5 QUARTER4 MODULE1 WEEK1-3 How Visual and Multimedia Elements.pptx
ENGLISH5 QUARTER4 MODULE1 WEEK1-3 How Visual and Multimedia Elements.pptxAnaBeatriceAblay2
 
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...Krashi Coaching
 
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...Marc Dusseiller Dusjagr
 
Introduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxIntroduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxpboyjonauth
 
Alper Gobel In Media Res Media Component
Alper Gobel In Media Res Media ComponentAlper Gobel In Media Res Media Component
Alper Gobel In Media Res Media ComponentInMediaRes1
 
भारत-रोम व्यापार.pptx, Indo-Roman Trade,
भारत-रोम व्यापार.pptx, Indo-Roman Trade,भारत-रोम व्यापार.pptx, Indo-Roman Trade,
भारत-रोम व्यापार.pptx, Indo-Roman Trade,Virag Sontakke
 
Hybridoma Technology ( Production , Purification , and Application )
Hybridoma Technology  ( Production , Purification , and Application  ) Hybridoma Technology  ( Production , Purification , and Application  )
Hybridoma Technology ( Production , Purification , and Application ) Sakshi Ghasle
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13Steve Thomason
 
Blooming Together_ Growing a Community Garden Worksheet.docx
Blooming Together_ Growing a Community Garden Worksheet.docxBlooming Together_ Growing a Community Garden Worksheet.docx
Blooming Together_ Growing a Community Garden Worksheet.docxUnboundStockton
 
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdfssuser54595a
 
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTiammrhaywood
 

Recently uploaded (20)

Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111
 
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxSOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
 
Employee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxEmployee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptx
 
Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)
 
Pharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdfPharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdf
 
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdfBASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
 
Computed Fields and api Depends in the Odoo 17
Computed Fields and api Depends in the Odoo 17Computed Fields and api Depends in the Odoo 17
Computed Fields and api Depends in the Odoo 17
 
Mastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory InspectionMastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory Inspection
 
ENGLISH5 QUARTER4 MODULE1 WEEK1-3 How Visual and Multimedia Elements.pptx
ENGLISH5 QUARTER4 MODULE1 WEEK1-3 How Visual and Multimedia Elements.pptxENGLISH5 QUARTER4 MODULE1 WEEK1-3 How Visual and Multimedia Elements.pptx
ENGLISH5 QUARTER4 MODULE1 WEEK1-3 How Visual and Multimedia Elements.pptx
 
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
 
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
 
Introduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxIntroduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptx
 
Alper Gobel In Media Res Media Component
Alper Gobel In Media Res Media ComponentAlper Gobel In Media Res Media Component
Alper Gobel In Media Res Media Component
 
भारत-रोम व्यापार.pptx, Indo-Roman Trade,
भारत-रोम व्यापार.pptx, Indo-Roman Trade,भारत-रोम व्यापार.pptx, Indo-Roman Trade,
भारत-रोम व्यापार.pptx, Indo-Roman Trade,
 
Hybridoma Technology ( Production , Purification , and Application )
Hybridoma Technology  ( Production , Purification , and Application  ) Hybridoma Technology  ( Production , Purification , and Application  )
Hybridoma Technology ( Production , Purification , and Application )
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13
 
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
 
Blooming Together_ Growing a Community Garden Worksheet.docx
Blooming Together_ Growing a Community Garden Worksheet.docxBlooming Together_ Growing a Community Garden Worksheet.docx
Blooming Together_ Growing a Community Garden Worksheet.docx
 
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
 
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
 

Soal dan Pembahasan Farmakologi Molekular - PPAR dan Reseptor Estrogen

  • 1. 1 SOAL DAN PEMBAHASAN FARMAKOLOGI MOLEKULAR PPAR DAN RESEPTOR ESTROGEN *daftar pustaka dan jurnal terlampir di akhir 1. Jelaskan mekanisme obat golongan tiazolidindion berikatan dengan PPARγ secara molekular ! Jawab : Tahapannya : a) Obat golongan tiazolindidion sebagai ligan melalui membran plasma masuk ke dalam sitoplasma b) Ligan berikatan dengan PPARγ yang sebelumnya dalam keadaan inaktif distabilkan oleh co-repressor c) Co-repressor lepas dari PPARγ d) Co-activator berikatan dengan PPARγ-ligan e) PPARγ membentuk kompleks heterodimer dengan RXR (Retinoid X Receptor) untuk regulasi transkripsi f) Transkripsi gen di dalam nucleus dengan disand oleh PPRE (Peroxisome Proliferator Receptor Response Element) g) Translasi h) Menghasilkan protein yang akan mempengaruhi fungsi pada berbagai macam jaringan : Protein Kondisi Protein Jaringan GLUT 4 ↑ Otot ↑ ambilan glukosa dan oksidasi asam lemak Sel Adiposa ↑ ambilan glukosa, adipogenesis, diferensiasi, dan simpanan trigliserida GLUT 2 ↑ Liver ↑ oksidasi asam lemak, simpanan lipid ↓ glukoneogenesis NF-kB ↓ Makrofag
  • 2. 2 ↑ polarisasi makrofag ↓ inflamasi PI3K ↑ Jantung dan pembuluh darah ↓ inflamasi VEGF ↑ Jantung dan pembuluh darah ↓ inflamasi CD36 ↑ Liver ↑ oksidasi asam lemak dan simpanan lemak ↓ glukoneogenesis Makrofag ↑ polarisasi makrofag ↓ inflamasi Sel Adiposa ↑ ambilan glukosa, adipogenesis, diferensiasi, dan simpanan trigliserida 2. Jelaskan aktivitas genomik reseptor estrogen ! Jawab : Tahapannya :
  • 3. 3 a) PA (Palmitoylation) menyebabkan ER (Estrogen Receptor) terlokalisasi di membran plasma sel b) E2 (17 β-estradiol) berdifusi melalui membran plasma sel c) Masuk ke sitoplasma d) E2 berikatan dengan ER e) Terjadi relokasisasi f) Perlibatan domain N-terminal dari interaksi intermolecular dan intramolecular g) Ikatan DNA domain (DBD, c region) menyebabkan ER dapat dimerisasi dan berikatan dengan sekuens DNA yang disandi oleh ERE (Estrogen Receptor Element) melalui struktur 2 “zinc finger” h) Aktivitas trans aktivasi i) Aktivasi transkripsi melalui perlibatan koaktivator dan komponen dari mesin transkripsi basal j) D-region mendorong dimerisasi reseptor dan ikatan reseptor dengan chaperone Hsp (Heat shock protein) k) Domain ligan berikatan (LBD, E/F region, C terminal) meliputi domain E2 berikatan l) Sinergis dengan N-terminal domain dalam transkripsi gen m) AF 1 dan AF 2 sebagai activation function dari ER berikatan dengan koaktivator n) ER α-E2 induksi aktivasi transkripsi sedangkan ER β-E2 menghambat transkripsi o) Respons estrogenik p) ER meningkatkan ikatan dengan Sp-1 (faktor transkripsi specificity protein 1) q) Ko-aktivasi r) Aktivasi Erα-E2 dari AP 1 responsive elements membutuhkan domain AF 1 dan AF 2 dari reseptor agar dapat berikatan dengan meningkatkan aktivitas komponen- komponen p160 (seperti SRC-1 dan SRC-2) oleh Fos/Jun s) Transkripsi gen t) Fungsi sel
  • 4. 4 Daftar Pustaka : 1. Choi, Sung Hee et al. Re-highlighting the Action of PPARγ in Treating Metabolic Diseases. F1000 Research. 2019. 2. Marino, Maria et al. Estrogen Signaling Multiple Pathways to Impact Gene Trascription. Current Genomics. 2006.
  • 5. Current Genomics, 2006, 7, 497-508 497 1389-2029/06 $50.00+.00 ©2006 Bentham Science Publishers Ltd. Estrogen Signaling Multiple Pathways to Impact Gene Transcription Maria Marino1,*, Paola Galluzzo1 and Paolo Ascenzi1,2,3 1 Department of Biology and 2 Interdepartmental Laboratory for Electron Microscopy, University Roma Tre, Viale Gug- lielmo Marconi, 446 I-00146 Roma, Italy; 3 National Institute for Infectious Diseases I.R.C.C.S. “Lazzaro Spallanzani”, Via Portuense 292, I-00149 Roma, Italy Abstract: Steroid hormones exert profound effects on cell growth, development, differentiation, and homeostasis. Their effects are mediated through specific intracellular steroid receptors that act via multiple mechanisms. Among others, the action mechanism starting upon 17-estradiol (E2) binds to its receptors (ER) is considered a paradigmatic example of how steroid hormones function. Ligand-activated ER dimerizes and translocates in the nucleus where it recognizes spe- cific hormone response elements located in or near promoter DNA regions of target genes. Behind the classical genomic mechanism shared with other steroid hormones, E2 also modulates gene expression by a second indirect mechanism that involves the interaction of ER with other transcription factors which, in turn, bind their cognate DNA elements. In this case, ER modulates the activities of transcription factors such as the activator protein (AP)-1, nuclear factor-B (NF-B) and stimulating protein-1 (Sp-1), by stabilizing DNA-protein complexes and/or recruiting co-activators. In addition, E2 binding to ER may also exert rapid actions that start with the activation of a variety of signal transduction pathways (e.g. ERK/MAPK, p38/MAPK, PI3K/AKT, PLC/PKC). The debate about the contribution of different ER-mediated signaling pathways to coordinate the expression of specific sets of genes is still open. This review will focus on the recent knowl- edge about the mechanism by which ERs regulate the expression of target genes and the emerging field of integration of membrane and nuclear receptor signaling, giving examples of the ways by which the genomic and non-genomic actions of ERs on target genes converge. Received on: August 8, 2006 - Revised on: October 21, 2006 - Accepted on: October 30, 2006 Key Words: Estrogen, estrogen receptors, genomic and non-genomic action mechanism, gene transcription. 1. INTRODUCTION The principle estrogenic hormone, 17-estradiol (E2), synthesized by testosterone aromatization in the ovary and in other tissues, plays a central role in the control of sexual be- havior and reproductive functions. At present it is well rec- ognized that the impact of E2 in human physiology is wider than previously thought impact including the differentiation of several tissues and organs, the modulation of inflamma- tion, and brain and cardiovascular functions as well [see 1- 3]. E2 regulates human physiology via diffusion through the plasma membrane of target cells and signaling through intra- cellular hormone-specific estrogen receptors (ERs). Two distinct types of signaling can be mediated, often referred to as genomic and non-genomic or non-genotropic pathways. In the genomic pathway, estrogens bind to ERs in the nucleus, inducing a conformational change in the receptors that cause dissociation from chaperones, dimerization, and activation of the receptor transcriptional domain [4-6]. The canonical model for ER-mediated regulation of gene expression involves the direct binding of dimeric ER to DNA sequences known as estrogen response elements (EREs), *Address correspondence to this author at the Department of Biology, Uni- versity “Roma Tre”, Viale Guglielmo Marconi 446, I-00146 Roma, Italy; Tel: +39-06-55176345; Fax: +39-06-55176321; E-mail: m.marino@uniroma3.it which are specific, inverted palindromic sequences [7]. In addition, ER can indirectly associate with promoters through protein-protein interactions with other DNA-binding tran- scription factors [8-10]. In either case, interaction of ERs with E2 leads to transcriptional activation of the associated genes via recruitment of coactivators and components of the basal transcriptional machinery [11-14]. In addition to the nuclear ERs, plasma membrane-associated ERs mediate the non-genomic signaling pathway [see 15-19], which can lead both to cytoplasmic alterations and to regulation of gene expression [16, 20, 1]. Regulation of transcription by nuclear ER is more com- plicated than the classical paradigm would predict [5, 18]. The two nuclear ERs, ER and ER, exhibit distinct tran- scriptional properties and can form both homodimers and heterodimers [22-24]. Recent studies point to the fact that signaling pathways modulate both ERs and some co-regula- tory molecules activities [13, 25]. To understand the connection between physiological and molecular functions of ERs, the field requires an in-depth understanding of the spectrum of genes regulated in each tissue and cell type. This review will focus on the current state of knowledge about the mechanism by which ERs regu- late the expression of target genes and the emerging field of integration of membrane and nuclear receptor signaling, giv- ing examples of the ways by which the genomic and non- genomic actions of ERs on target genes converge.
  • 6. 498 Current Genomics, 2006, Vol. 7, No. 8 Marino et al. 2. THE STRUCTURE OF ESTROGEN RECEPTORS Human ER and ER are encoded by different genes located on different chromosomes (locus 6q25.1 and locus 14q23-24.1, respectively) [26-29]. ER and ER, like all the members of the nuclear receptor super-family, are modular proteins sharing common regions, named A/B, C, D, and E/F, as well as a high sequence homology (Fig. (1A)). These regions participate in the formation of independent but inter- acting functional domains. The N-terminal domain (A/B re- gion) is involved in both inter-molecular and intra-molecular interactions as well as in the activation of gene transcription. The DNA binding domain (DBD, C region) allows ER to dimerize and to bind to the specific ERE sequence on DNA through its two “zinc finger” structures (Fig. (1B)). The hinge domain (D region) has a role in receptor dimerization and in binding to chaperone heat-shock proteins (Hsp). The ligand binding domain (LBD, E/F region, C-terminal) comprises the E2-binding domain and works, synergistically with the N-terminal domain in the regulation of gene transcription [5, 30-32]. ERs contain two regions called activation functions (AFs) important for ligand-dependent transcriptional activity (Fig. (1A)) [5, 30-32]. AF-1 and AF-2 regions of ERs, inter- acting with a number of transcription co-activators, can acti- vate transcription independently but in most cases, they syn- ergize with one another in a promoter- and cell-context spe- cific manner [33]. AF-1 could be activated even in a ligand-independent manner, depending on the phosphorylation status of ER. In particular, the Ser118 residue in the AF-1 region of ER, as well as residues Ser106 and Ser124 in the AF-1 region of ER, are the phosphorylation sites essential for the ligand- independent activation of ERs through the Ras-mitogen acti- vated protein kinase (MAPK) signaling cascade [see 34, 35]. Recent progress in studies on genomic and cDNA se- quences has accelerated the identification of gene splice variants in the NR super-family. Numerous mRNA splice variants exist for both ERs and the best-characterized splice variants are ER46 and ERcx, which are frequently co- expressed with their wild-type counterparts. The exact func- tion and potential role of these and other ERs splice variants in physiology and human disease remain to be elucidated [see 36]. Fig. (1). Domain organization of human ER and ER (A). ERs consist of the N- terminal region involved in transactivation (A/B domains, AF-1), the DNA binding domain (DBD, C domain), the hinge region involved in dimerization (D domain), the C-terminal region containing ligand binding domain (LBD, E/F domain, AF-2) and transactivation function-2 (AF-2). The percentage indicates the homology between ER and ER. (B) Binding mode of ERE to dimeric ER (PDB ID:1HCQ) [163]. Spheres indicate the zinc atoms. For details, see text.
  • 7. Estrogen Signaling and Gene Transcription Current Genomics, 2006, Vol. 7, No. 8 499 3. ESTROGEN RECEPTOR GENOMIC ACTIVITY 3.1. Direct Association to DNA The pioneering work by O’Malley and colleagues dem- onstrated that ERs function as ligand-activated transcription factors [37]. The trans-activation activity of ERs initiate through the ligand-bound receptor to its cognate, cis-acting enhancers, ERE [38]. The consensus palindromic element ERE was initially described based on the estrogen-respon- sive sequence in the Xenopus laevis vitellogenin A2 pro- moter: 5’-GGTCACAGTGACC-3’ [39-41]. This “perfect” ERE sequence was shown to function in an orientation- and distance-independent manner, both of which are properties of an enhancer [7, 42]. When ER directly interacts with the promoter/enhancer, binding to a full ERE is apparently the dominant mode of interaction. The human full EREs have a 3-bp spacer between the two half-sites, the exceptions being response elements in the human transforming growth factor (TGF)- promoter, with a 4-bp spacer, and in the promoter of the rat luteinizing hormone gene, with a 5-bp spacer [42]. Controversy still exists concerning ER DNA binding via ERE half sites, although a number of examples exist [43- 46]. Since the identification of a canonical ERE, several com- putational approaches have been undertaken to identify tar- get genes based on the presence of EREs within promoter proximal regions [47, 48]. For instance, for the 38 estrogen- responsive genes reviewed by Klinge [7], most of the func- tional EREs located within the promoters or 3’-untranslated regions are not the traditional consensus sequence. Thus, many target genes contain response elements that bear little similarity to consensus EREs. In one of the most comprehen- sive studies, Bourdeau and coworkers screened for all EREs in the human and mouse genomes and identified in excess of 70,000 EREs within the human genome, over 17,000 of which were within 15 kb of mRNA start sites [48]. Elimina- tion of EREs that were not conserved between the human and mouse genomes reduced the number of gene proximal EREs to 660. A number of these sites were validated as genuine ER interaction sites, supporting the use of computa- tional models to predict putative ER target genes to some degree [49]. The sequence of the response element affects the affinity that a given receptor has for binding DNA. ER binds with the highest affinity the canonical ERE sequence found within the vitellogenin A2 gene, and less well the imperfect EREs found within the vitellogenin B1 (5’-AGTCACTGTGACC- 3’) [39], pS2 (GGTCACGGTGGCC-3’) [50], and oxytocin (5’-GGTCAAGGTCACC-3’) [51] genes. This explains, at least in part, how the sequence of the response element can be one important determinant of the extent to which ERs can activate gene expression [52-55]. The conformation of transcription factors can be altered through binding to DNA [see 56]. The specific ERE se- quences could exert distinct, allosteric effects on the con- formation of ER and ER [52, 57, 58]. Just as ligand- induced changes in ER conformation influence ER interac- tions with co-activators, consensus and imperfect EREs also influence the ability of ERs to bind co-activators. Note that the steroid receptor coactivator-2 (SRC-2) interacts better with ER bound to EREs from the vitellogenin A2 than from the vitellogenin B1 gene [54]. 3.2. Indirect Association to DNA The ER signaling mechanisms discussed until now pro- vide an explanation for the regulation of genes in which a functional ERE-like sequence can be documented within the promoter. Another category of gene promoters, lacking any ERE-like sequences, requires a second DNA-binding tran- scription factor to mediate ER association with the DNA [42]. This mechanism is generally referred to as “transcrip- tional cross-talk” [59-60]. Roughly 35% of the categorized human primary E2-responsive genes are transcripted via ER- indirect DNA association [42]. Stimulating protein-1 (Sp-1) is the predominant mediator of ER-DNA indirect binding [42] and increasing numbers of genes are found to be induced by E2 via this mechanism including the low-density lipoprotein (LDL) receptor [8], endothelial nitric oxide sinthase (eNOS) [61], c-fos [62], cyclin D1 [63], and the retinoic acid receptor-1 genes [64, 65]. In response to estrogenic stimulation, ER enhances the binding of Sp-1 to its site, containing GC-rich promoter se- quences [46] and contributes to co-activator recruitment. The DNA-binding domain of ER is dispensable for such activa- tion [42, 66, 67]. Another example is the interaction between ER and the c-rel subunit of the nuclear factor-B (NF-B) complex. This interaction prevents NF-B from binding to and stimu- lating expression from the interleukin-6 (IL-6) promoter [68]. In this way, E2 inhibits expression of the cytokine IL-6 [68-70]. Other intermediary factors through which ER can associ- ate with promoters/enhancers include: activating transcrip- tion factor (ATF)-2/c-jun or ATF-2/cAMP response element binding protein (CREB) for the cyclin D1 gene, ATF- 1/CREB for the Bcl-2 gene, and nuclear transcription factor- Y for the mouse E2F1 gene [42]. ERs utilize protein-protein interactions also to enhance transcription of genes that contain activator protein-1 (AP-1) sites [71] related, but not identical, to those for the ATF/CREB transcription factors. The AP-1 complex, com- posed of Jun protein dimers and of Jun/Fos heterodimers, plays an important role in cell proliferation. Notably, ER activation of IGF-1 and collagenase expression is mediated through the interaction of receptor with Fos and Jun at AP-1 binding sites [42]. Collagenase, insulin-like growth factor (IGF)-1 receptor, ovalbumin, and cyclin D1 are examples of genes activated by the ER-E2 complex via AP-1 [72, 73]. ER and ER have been shown to signal in opposite ways at AP-1 sites. ER activates transcription in the pres- ence of E2, whereas ER-E2 inhibits AP-1-dependent tran- scription [74, 60]. Studies show that ER-E2 activation of AP-1-responsive elements requires both AF-1 and AF-2 do- mains of the receptor, which bind and enhance the activity of the p160 components (e.g. SRC-1 and SRC-2) of the co- activator complex recruited to the site by Fos/Jun. Interest- ingly, human ER, which lacks a functional AF-1, is unable to activate transcription of AP-1-regulated genes when bound with ER agonists, indicating the possibility of distinct
  • 8. 500 Current Genomics, 2006, Vol. 7, No. 8 Marino et al. physiological actions of the two ERs via the regulation of unique subsets of genes [4]. Similar to AP-1, E2 binding to ER induces transcriptional activation when associated with Sp-1 in GC-rich regions. However, E2 interaction with ER does not result in the formation of a transcriptionally active complex at a promoter containing Sp-1 elements. As an ex- ample ER and ER, in the presence of E2, oppose each other’s function in the regulation of the cyclin D1 promoter [75]. There is considerable evidence that cyclin D1, impor- tant for progression of cells through the G1 phase of the cell cycle, is a well-defined target for ER-E2 action in mam- mary carcinoma cells [76-78], although no detectable “per- fect” or ERE-like sequence in the cyclin D1 gene promoter has been reported [79]. Deletion of AP-1 and Sp-1 respon- sive element motifs in the cyclin D1 gene promoter resulted in attenuation of promoter responsiveness to E2 [72, 80]. Unlike ER, E2-bound ER represses cyclin D1 expression [81] and blocks ER-E2-mediated induction when both re- ceptor isoforms are present [22]. Consequently, these differ- ences in transcriptional activity between the ER and ER may account for the major differences in their tissue specific biologic actions. This complexity is further enhanced by the presence of different ER splicing forms, by the ability of ERs to form homodimers and heterodimers, and by their capacity to interact with different co-regulators [82]. 3.3. Transcriptional Co-Factors Both in the direct and indirect action modes, the ligand- activated ERs are not the transcription controllers. In fact, ERs need to interact with co-regulatory proteins (co- activators or co-repressors) to form a platform upon which additional proteins are assembled [12, 13]. Cofactors interact with ERs through their Leu rich motif (i.e., Leu-Xxx-Xxx- Leu-Leu, where Xxx is any amino acid). Several classes of ER cofactors have been identified. The first identified and well-characterized co-activator family consists of three re- lated members SRC-1, which is the founding member of the family, SRC-2, and SRC-3 [see 83]. A large co-activator complex, referred to as thyroid hormone receptor associated protein/vitamin D receptor-integrating protein (TRAP/DRIP) complex, could connect ERs directly to the basal transcrip- tion machinery via its intrinsic chromatin remodeling func- tions. In addition, histone acetyl transferase (e.g., CBP/p300), histone methyl transferase (e.g., CARM1 and PRMT1) and the nucleosome remodeling complexes (e.g., SWI/SNF) are necessary to release the chromatin-dependent inhibition of gene transcription [13]. Although there are far fewer nuclear receptor co-repre- ssors, these (macro)molecules serve important roles in nega- tively regulating ER-dependent gene expression. Two AF-2 interacting proteins, receptor-interacting protein-140 and short hetero-dimer partner, exhibit negative co-regulatory functions because they can antagonize SRC-1 co-activators in vivo and compete for AF-2 binding in vitro [84-86]. On the other hand, ERs could also associate with specific tran- scriptional repressors such as the nuclear receptor corepres- sor and specific histone deacetylase complexes [13]. The relative expression of co-activators and co-repre- ssors, within a cell, influences the ability of ER ligands (e.g., E2 and selective ER modulators (SERMs)) to regulate gene expression [2, 13, 87]. Because of the homology in their AF-2 domains (see Fig. (1A)), ER and ER should be similar in co-activator re- cruitment, but certain differences have been reported. For E2-bound receptors, ER, but not ER, binds well to the receptor-interacting component of the mammalian mediator complex, TRAP220. There are differences between the rela- tive affinities of ER and ER for members of the p160 co- activator family [13, 88]. More pronounced differences are observed in the case of SERM-bound ERs [see 2, 13]. For ERE-dependent gene expression, the SERM 4-hydroxy- tamoxifen is a partial agonist of ER, but is generally unable to stimulate ER transcriptional activity [89-91]. Con- versely, when assessing ER activity on AP-1 containing re- porter genes, 4-hydroxytamoxifen will stimulate ER and ER transcriptional activity in a cell-dependent fashion [74]. A mechanism for shuttling off transcription involves the covalent post-translational modification of ERs and co- activators (e.g., lysine acetylation and arginine methylation), which can inhibit the binding of co-activators to nuclear re- ceptors or other transcriptional activators by altering critical protein-protein interaction surface [see 13, 92]. Thus, the acetylation of SRC-3 by p300 has been shown to cause a disruption of receptor-co-activator complexes, leading to a decrease in receptor-mediated gene activation [see 92]. Us- ing a variety of biochemical and cell-based assays, Krauss and co-workers have shown ER, but not ER, is a target for acetylation by p300 and have identified acetylation as modu- lator of the ligand-dependent gene regulatory activity of ER [93]. A number of cellular signaling pathways also influence the ER-dependent gene expression modulating ER confor- mational changes or co-regulators recruitment [5, 13]. It has been recognized only recently that both co-activators and co- repressors are also substrates for kinases, their phosphoryla- tion affects their ability to interact with steroid receptors [94, 95]. 4. ESTROGEN RECEPTOR NON-GENOMIC ACTIV- ITY The “genomic action” of steroid hormones occurs after a time-lag of at least 2 hours after E2 stimulation and explains some of hormone functions in physiological and pathological situations [see 96, 97]. This picture was challenged when a physiological dose of E2 was reported to increase the uterine cAMP level in ovariectomized rats within 15 seconds [98], an effect too rapid to be accounted for genomic action(s). This event was not abrogated by transcriptional inhibitors and was termed “rapid or non-genomic”. Actually the term “non-genomic” is not adequate when referring to rapid changes that may also initiate new gene transcription [see 96, 99]. Various signaling pathways are activated upon E2 bind- ing to ERs. These rapid events may be classified into four main signaling cascade: phospholipase C (PLC)/protein ki- nase C (PKCs) [100-106], Ras/Raf/MAPK [72, 107-113], phosphatidyl inositol 3 kinase (PI3K)/AKT [15, 16, 80, 81, 97, 114-118], and cAMP/ protein kinase A (PKA) [104, 119- 123]. These pathways present numerous interactions with sev- eral other pathways. The ER-E2 complex interacts with the
  • 9. Estrogen Signaling and Gene Transcription Current Genomics, 2006, Vol. 7, No. 8 501 IGF-1 receptor, leading to IGF-1 receptor activation and hence to MAPK signaling pathway activation [124]. In addi- tion, the ER-E2 complex activates the EGF receptor by a mechanism that involves activation of guanine nucleotide exchange proteins (G-proteins), Src, and matrix metallopro- teinases, leading to an increase in extracellular regulated kinases (ERK) and PI3K/AKT activities [109, 125-129]. In endothelial cells the Src/PI3K/AKT pathway mediates rapid E2-dependent activation of eNOS and the release of nitric oxide. AKT and PKC could also modulate the MAPK path- way through Raf phosphorylation [97, 116, 130, 131]. It is important to note that activation of signaling path- ways by E2 is cell type-specific. Indeed, the effect of E2 on PKC activity has been observed in the preoptic area of fe- male rat brain slices, but not in the hypothalamus or cortex [132]. The activation of G-protein/Src/PI3K/MAPK pathway by E2 was evident in late, but not early, differentiated rat pre-adipocytes [109]. The differential requirement of Src/ PI3K or intracellular calcium for MAPK activation is also observed in diverse cell types [15, 109, 129]. Different PKC isoforms are rapidly activated by E2 in HepG2 and MCF7 cells [102]. As a whole, these studies indicate that the rapid actions of E2 depend on a number of conditions such as the set of signal transduction molecules and downstream targets present in the target cell, thus the responses are likely to be diverse. All these results point to the concept that ER is the pri- mary endogenous mediator of rapid E2 actions. Less infor- mation is available on the role played by the ER-E2 com- plex to activate rapid non-genomic mechanisms. A subpopu- lation of ER transfected into Chinese Hamster ovary cells is capable of activating inositol tris-phosphate production, ERK and JNK phosphorylation [133]. Geraldes and cowork- ers reported that E2 reduces ERK activity through ER stimulation in porcine smooth muscle cells [134]. We have recently reported the ability of the ER-E2 complex to acti- vate the p38 member of MAPK family, but not ERK or AKT, in human colon cancer cells [81, 135]. Although the scarce information does not allow a complete discussion on the contribution of ER in E2-induced rapid signals, these data indicate that also ER could originate cell-specific sig- nal transduction cascade. The rapidity by which E2 induces rapid signals as well as the localization of signaling complex raises the requirement of a plasma membrane ER. Debate continues over whether structural changes target nuclear ERs in separate pools local- izing them to the membrane [61, 97, 99, 117, 136], or whether membrane ER represents a novel receptor [137- 142]. Besides these data, much evidence favors the idea that the membrane-localized ER is the same protein as the nu- clear-localized receptor [72, 80, 133, 143, 144]. Even if the definitive proof that membrane and nuclear ER are the same protein requires isolation and “sequencing” of the two recep- tor pools, ER and ER must be considered a population of protein(s) which localization in the cell is able to dynami- cally change, shuttling from membrane to cytosol and to the nucleus, depending on ligand binding [87, 97, 135,145]. Current evidence indicates that a small population of ER and ER localize at the plasma membrane exists within caveolar rafts. It is at the plasma membrane that E2-liganded ER associates with the scaffolding protein caveolin-1 and a variety of signal transduction cascade activation occurs [e.g., PLC, PKC, ERK, PI3K, and nitric oxide synthase (NOS)]. ERs do not contain a trans-membrane domain [15, 18], thus the ability of ER and ER to associate with the plasma membrane could be due to its association with membrane proteins and/or by post-translational addition of lipids to ER [16, 146]. Fatty acids and isoprenoids are two of the most common lipid moieties found on post-translational modified proteins bound to membranes. No consensus sequences for N-acyla- tion (i.e., miristoylation) or S-prenylation have been found in ER and ER [147]. On the contrary, S-acylation (i.e., palmitoylation) does not require any consensus sequence, but just reactive Cys residues [148]. Cys residues present in the ER and ER LBD could undergo S-acylation. In particular, the amino acid sequence encompassing the Cys447 residue of ER and Cys399 of ER is highly homologous to that surrounding the S- palmitoylated Cys132 residue of human caveolin-1 [147]. Based on this observation we demonstrated that ER un- dergo S-palmitoylation which represents the major determi- nant for its residence at the plasma membrane and in its as- sociation with caveolin-1 [146, 147]. It is noteworthy that ER is also a palmitoylable protein [Marino M., unpublished results]. Because ER has no intrinsic kinase domains the local- ization of ERs at the plasma membrane facilitate the associa- tion between ER and signaling proteins allowing the activa- tion of rapid events. Src, Shc, proline-, glutamic acid-, leucine- rich protein /modulator of non-genomic activity of estrogen receptor (PELP1/MNAR), the p85 subunit of PI3K, receptor tyrosine kinases (i.e., EGF and IGF-1 recep- tors), as well as G-protein isoforms (i.e., Gs and Gq) have all been reported to serve as components of large complexes of interacting proteins. Through the mediation of these molecules, E2 activates the MAPK and PI3K/AKT pathways [16, 136, 149-151]. Although the list of signaling and adapter proteins inter- acting with ER is growing, protein-protein complex forma- tion occurs only 5 to 15 min after E2 stimulation [152]. Thus, the conformational changes of the ER LBD domain, which follows E2 entry into the cell, seems to be important in allowing the ER-E2 complex to detach from the mem- brane and allocate with growth factor receptors or adapter proteins to activate downstream signals. 4.1. Cell Functions Regulated by Non-Genomic Signals The rapid activities of ERs are widely accepted and dis- agreement on the involvement of nuclear receptors is quite settled. However, other controversies in this field are still present and related to whether or not all of these rapid effects are of physiological relevance [153]. The main difficulties are linked to the experimental models used. In fact, the study of signaling pathways can be done mainly on isolated, often immortalized, cells and it is very complicated to obtain simi- lar information on a whole organism in which the use of sig- naling inhibitors could have many side effects other than to inhibit just one kinase.
  • 10. 502 Current Genomics, 2006, Vol. 7, No. 8 Marino et al. Nevertheless, the physiological significance of rapid membrane-starting pathways has been clarified at least for some E2 targets. In the nervous system, E2 affects neural functions (e.g., cognition, behavior, stress responses, and reproduction) in part by inducing such rapid responses [96]. In the skeleton, ER, present in caveolae of bone-forming osteoblasts, transmits survival signals through activation of the Src/Shc/ERK pathway and prolongs the life span of os- teoblasts [21]. At the same time, E2 delivers a pro-apoptotic signal to bone-resorbing osteoclasts, shortening their life span [21]. Although these studies have been done mainly in cell-culture systems, their results suggest that ER rapid sig- naling actions have also a role in vivo. In the liver, rapid E2- induced signals (i.e., PLC/PKC) are deeply linked to the ex- pression of the LDL receptor and to a decreased level of se- rum LDL-cholesterol [103]. Finally, vascular protection by E2 in ischemia/reperfusion injury in vivo requires E2- induced activation of endothelial NOS, as mediated by the PI3K/AKT pathway [117, 130]. The mechanism(s) by which E2 exerts proliferative ef- fects is assumed to be exclusively mediated by rapid mem- brane-starting actions [72, 80, 101, 102, 114, 115]. E2 treat- ment of mammary-derived MCF-7 cells triggers the associa- tion of ER with Src and p85 leading to DNA synthesis [115]. In HepG2 cells multiple and parallel membrane- starting pathways are rapidly activated by the ER-E2 com- plex [72, 80, 101] and the blockade of PLC/PKC, ERK, and PI3K/AKT pathways completely prevents the E2-induced DNA synthesis [72, 80]. ERK/MAPK and PI3K/AKT path- ways, rapidly activated by the ER-E2 complex, also have a critical role in E2 action as a survival agent. In fact, these pathways enhance the expression of the anti-apoptotic pro- tein Bcl-2, block the activation of the p38/MAPK, reduce the pro-apoptotic caspase-3 activation, and promote G1-to-S phase transition via the enhancement of the cyclin D1 ex- pression [72, 80, 81]. What is the contribution of ER to E2-induced cell pro- liferation? ER appears to act as a dominant regulator in E2 signaling, and when co-expressed with ER it causes a con- centration-dependent reduction of ER-mediated transcrip- tional activation [22] and the repression of ER-mediated effects including cell proliferation. Consistent with this no- tion, E2 increases cell proliferation and causes tumor forma- tion in MCF-7 cells expressing only ER [22]. On the other hand, ER inhibits the E2-induced proliferation of trans- fected MCF-7 cells and prevents tumor formation in a mouse xenograft model in response to E2 [154]. This effect is linked to the ER repressive effect on ER-induced gene transcription by binding to other transcription factors (e.g., AP-1, Sp-1) [22]. Recently, ER has been reported to rapidly induce a persistent membrane-initiated activation of p38/ MAPK without any interference on survival proliferative pathways, thus impairing the activation of cell cycle compo- nents (i.e., cyclin D1 expression) [81]. 5. INTEGRATION OF NUCLEAR AND EXTRA- NUCLEAR ACTION OF E2 Even though the membrane ERs and associated non- genomic actions is an area of active research, the nuclear effects of membrane ERs has not received much attention [see 16, 19]. In human vascular smooth muscle cells tran- siently transfected with ER an E2-dependent and an E2- independent translocation of ER from the membrane to the nucleus was observed. The latter was blocked by MAPK inhibitors [155]. The ability of membrane ER and/or the growth factor receptor tyrosine kinases to signal via multiple kinases to the nucleus undoubtedly impacts all aspects of cellular function. E2-induced ERK activation up regulates AP-1 mediated genes (e.g., c-fos) [156]. This results in part from serum re- sponse factor/elk-1 stimulation by E2, and in part by re- cruitment of nuclear ER and co-activators to AP-1 sites on gene promoters [16]. Other targets include several members of the signal transducer and activators of transcription (STAT) family such as STAT1, STAT3 and STAT5. In en- dothelial cells, activation of both STAT3 and STAT5 by E2 was mediated through signaling pathways involving MAPK, PI3K and Src and it functions to regulate -casein expression [15]. Similarly, PI3K activation by E2-induced signaling from the membrane ER rapidly up regulates hundreds of genes in a target cell [157]. Microarray analysis of gene expression in vascular endothelial cells showed that about 250 genes were up-regulated 40 min after treatment. This effect could be prevented by the PI3K inhibitor, LY294,002 [157]. Interest- ingly, the transcriptional activity of the ER-E2 complex is inhibited by a pre-treatment with the ERK inhibitors PD98,059 and U0126 [20, 114]. This suggests that stimula- tion of some gene expression (i.e., cyclin D1 and prolactin) by E2 occurs through ERK and PI3K activation. CREB is the most studied of the several transcription factors rapidly activated by E2. In a hippocampal cell line [158], adipocyte cells [109], and colonic carcinoma cells [159], CREB transcriptional activity can be induced by E2 or E2-BSA through MAPK pathway, independently from the PKA pathway. Such activation of CREB induces expression of several genes (e.g., c-fos, uncoupling protein-2). In con- trast, in neuroblastoma cells activation of CREB by ER- mediated rapid signals is dependent on the cAMP/PKA pathway, leading to neurotensin gene expression [160]. In addition, ERs are possible ER-mediated rapid signal targets. Indeed, it has been long known that E2 treatment can increase the phosphorylation state of ERs, via ERK and PI3K, the mutation of important phosphorylation sites re- duces their transcription activity [19]. The rapid E2-evoked phosphorylation of ER contributes to the stimulation of ER dimerization and its nuclear translocation. As an example, the phosphorylation of ER on Ser305 enhances cyclin D1 transcription in breast cancer. E2 also induces phosphoryla- tion of ER in Ser118, Ser167, and Tyr537 residues through the non-genomic activation of the MAPK signaling pathway [19]. Furthermore, the Ser167 residue of ER also can be phosphorylated in response to rapid E2-mediated PI3K/AKT activation, whereas E2-induced p38/MAPK phosphorylation of Thr311 promotes ER nuclear localization and interaction with specific receptor coactivators [19]. Besides these functions, the complexity of the mecha- nism of ER action suggests a more finely tuned control ex- erted by E2-induced rapid signals on cellular molecular
  • 11. Estrogen Signaling and Gene Transcription Current Genomics, 2006, Vol. 7, No. 8 503 events. In particular, the extra-nuclear signals induced by E2 occur before the appearance of nuclear effects and the cell context in which the genomic events occur will be different depending on which signal pathway is activated. Thus, the integration between these molecular events is required to obtain the complete cellular response. The complex relationship between membrane and nu- clear effects induced by E2 also involves membrane- initiating phosphorylation of co-activators recruiting these proteins to the nuclear transcriptosome [13, 16]. This aug- ments the recruitment of co-activator proteins, such as SRC- 1 by ER [13]. One can envisage a carefully controlled modu- lation of nuclear ER-induced transcription, depending upon which signaling pathway(s) are activated by E2 in a given cell context. It is likely that discrete signaling pathways regulate the access of co-repressors to target gene promoters, although this mechanism is not well studied. As a corollary to this, phosphorylation of co-activators at discrete motifs could be inhibitory as well. The possible convergence of ER genomic and non- genomic activities at multiple response elements provides an extremely fine degree of control for the regulation of tran- scription by ERs. It has been estimated that more than 500 kinases are encoded within the human genome. The ability of ER-E2 membrane starting signals and/or growth factor receptor to signal through multiple cascades to the nucleus, undoubtedly has an impact on all aspects of cellular function, contributing to E2-induced cell proliferation and survival, all essential features of cell physiology as well as of tumor biol- ogy [16]. Examples of such fine-tuned ER multiple control action are cyclin D1 and vascular endothelial growth factor (VEGF) genes. Cyclin D1, a well-defined target for E2 in mammary gland, is important for the progression of cells throughout the G1 phase of the cell cycle. The cyclin D1 promoter is complex and contains binding sites for several transcription factors, but no ERE-like sequences have been identified [79]. It has been suggested that activation of the cyclin D1 gene transcription by E2 results from different ER activities: di- rect ER/Sp-1 or ER/AP-1 interaction [161] as well as ER- dependent non-genomic mechanisms [72, 80]. The cyclin D1 promoter also contains binding sites for STAT5 and NF-B, and these could be targets for ERs through both genomic and non-genomic actions [15]. The VEGF gene is another exam- ple of cross-talk between ERs non-genomic and genomic action. In fact, VEGF gene promoter contains both an ERE- like variant and GC-rich sequences that bind ER and ER-Sp- 1 complex [42]. Both must be occupied for the E2 maximal activation [15]. As a whole, these data strongly suggest that E2-induced rapid signaling reaches to the nucleus through these and other, undiscovered, pathways and synergize each other to provide plasticity for cell response to sex steroids (see Fig. (2)). 6. CONCLUSION The regulation of gene expression by E2 is a multi- factorial process, involving both genomic and non-genomic actions that converge at certain response elements located in the promoters of target genes. The final gene responses, however, could depend on a number of conditions such as the combination of transcription factors bound to a specific gene promoter, the cellular localization of ERs, the levels of various co-regulator proteins and signal transduction compo- nents, as well as the nature of extra-cellular stimuli. These variables are highly specific for cell types. Thus, E2 could use different signaling pathways depending both on the cel- lular type and on the physiological status of the cell. In this way E2 evokes distinct gene responses in different types of target cells [15, 16, 97, 162]. The possibility that E2 could act on ER pools localized in different cell compartments (i.e., membrane versus cytoso- lic) gives rise to questioning the ability of these different ER pools to send parallel or synergic signals to the nucleus. For example, it has been observed that a naturally occurring variant of the metastatic tumor antigen 1 sequesters ER in the cytoplasm of breast cancer cells. The result of this cyto- solic retention is the reduction of E2-mediated transcription and the enhancement of E2-initiated ERK activation [136]. These data suggest that the same ER molecule is involved in genomic and in rapid signal transduction cascade. More data are needed to confirm this hypothesis and the use of dynamic imaging in the near future will help to clarify this issue. Based upon findings highlighted in this review, one may envisage a dynamic integrated model of action for ERs in- side the cell. In this model, ERs would shuttle from cell membrane to the cytoplasm and to the nucleus, in a dynamic equilibrium between different cell compartments. Each could play a different role in a multi step process of target gene activation by ER and co-activators from their upstream non- genomic to their downstream genomic responses would lead to activation of transcription (Fig. (2)). The cell context specific environment (e.g., differentia- tion, ER level, and ER co-expression) has an impact on the integration of rapid signaling by E2 from the membrane and on subsequent nuclear transcription. This leads to different signal cascades, different gene expression in response to the same hormone, and different cell biological outcome. The field is moving quickly. The challenges in the near future are to continue identifying the discrete actions of each ER intracellular pool, in order to clarify the role of ER, and to identify the potential cross-talk between ERs and other nuclear receptors. As we gain a deeper understanding of the complex controls exerted by ER and start identifying the critical players, it is likely that some of these putative mole- cules might emerge target candidates for therapeutic devel- opment in the treatment of hormone-responsive diseases, such as for different types of cancer. ACKNOWLEDGEMENTS Some experimental concepts described in the current paper are based on work conducted in the laboratories of the authors. These experimental studies were supported by grants from the Ministry of Education, University, and Re- search of Italy (PRIN-COFIN 2004 to M.M.). The Authors wish to thank past and present members of their laboratory who contributed with data and discussions to the ideas pre- sented here.
  • 12. 504 Current Genomics, 2006, Vol. 7, No. 8 Marino et al. ABBREVIATIONS AF = Activation function AP-1 = Activator protein-1 CREB = cAMP responsive element binding protein DBD = DNA-binding domain EGF = Epidermal growth factor ER = Estrogen receptor ERE = Estrogen responsive element ERK = Extracellular regulated kinase E2 = 17-estradiol G-proteins = Guanine nucleotide exchange proteins Hsp = Heath shock protein IGF-1 = Insulin-like growth factor-1 IL-6 = Interleukin-6 LDL = Low dendity lipoprotein LBD = Ligand-binding domain MAPK = Mitogen-activated protein kinase MNAR = Modulator of non-genomic activity of estro- gen receptor (also named Pro-, Glu-, and Leu-rich protein-1 PELP1) NF-B = Nuclear factor B PI3K = Phosphatidyl inositol 3-kinase NOS = Nitric oxide synthase PKA = Protein kinase A PKC = Protein kinase C PLC = Phospholipase C SERMs = Selective estrogen modulators Sp-1 = Stimulating protein-1 SRC = Steroid receptor co-activator family STAT = Signal transducers and activators of transcription Fig. (2). Schematic model illustrating the relationship between rapid, intermediate, and long term actions of E2 on target cells. Palmitoyla- tion (PA) allows the estrogen receptor (ER) localization at the plasma membrane. 17-estradiol (E2) binding induces ER re- localization, association to signaling proteins, and triggers the activation of signaling cascades. The kinase activations phosphory- late ER, modulate transcriptional coactivators recruitment, and enhance AP-1 and Sp-1 activation. After dimerization ERs directly interact with ERE on DNA. ERs-DNA indirect association occurs through protein-protein interactions with the Sp-1 and AP-1 tran- scription factors. AP-1, activating protein-1; MNAR, modulator of non-genomic activity of ER; PA, palmitic acid; Sp-1, stimulating factor-1. For details, see text.
  • 13. Estrogen Signaling and Gene Transcription Current Genomics, 2006, Vol. 7, No. 8 505 TGF- = Transforming growth factor- TRAP = Thyroid hormone receptor associated protein (also named vitamin D receptor-integrating protein DRIP) VEGF = Vascular endothelial growth factor REFERENCES [1] Gruber, C.J., Tschugguel, W., Schneeberger, C., Huber, J.C. Pro- duction and actions of estrogens. New Engl. J. Med. 2002, 346: 340-352. [2] Pearce, S.T., Jordan, V.C. The biological role of estrogen receptors and in cancer. Crit. Rev. Oncol. Hematol. 2004, 50: 3-22. [3] Deroo, B.J., Korach, K.S. Estrogen receptors and human disease. J. Clin. Invest. 2006, 116: 561-570. [4] Hall, J.M., Couse, J.F., Korach, KS. The multifaceted mechanisms of estradiol and estrogen receptor signaling. J. Biol. Chem. 2001, 276: 36869-36872. [5] Nilsson, S., Makela, S., Treuter, E., Tujague, M., Thomsen, J., Andersson, G., Enmark, E., Pettersson, K., Warner, M., Gustafsson, J.-Å. Mechanisms of estrogen action. Physiol. Rev. 2001, 81: 1535-1565. [6] Hall J.M., McDonnell, D.P. Coregulators in nuclear estrogen recep- tor action: from concept to therapeutic targeting. Mol. Interv. 2005, 5: 343-357. [7] Klinge, C.M. Estrogen receptor interaction with estrogen response elements. Nucleic Acids Res. 2001, 29: 2905-2919. [8] Li, C., Briggs, M.R., Ahlborn T.E., Kraemer, F.B., Liu, J. Re- quirement of Sp1 and estrogen receptor-interaction in 17- estradiol-mediated transcriptional activation of the low density lipoprotein receptor gene expression. Endocrinology 2001, 142: 1546–1553. [9] Safe, S. Transcriptional activation of genes by 17-estradiol through estrogen receptor-Sp1 interactions. Vitam. Horm. 2001, 62: 231-252. [10] Stossi, F., Likhite, V.S., Katzenellenbogen, J.A., Katzenellenbogen, B.S. Estrogen-occupied estrogen receptor represses cyclin G2 gene expression and recruits a repressor complex at the cyclin G2 pro- moter. J. Biol. Chem. 2006, 281: 16272-16278. [11] Glass, C.K., Rosenfeld, M.G. The coregulator exchange in tran- scriptional functions of nuclear receptors. Genes Dev. 2000, 14: 121-141. [12] McKenna, N.J., O’Malley, B.W. Combinatorial control of gene expression by nuclear receptors and coregulators. Cell 2002, 108: 465-474. [13] Smith, C.L., O’Malley, B.W. Coregulator function: a key to under- standing tissue specificity of selective receptor modulators. Endocr. Rev. 2004, 25: 45-71. [14] Kato, S., Sato, T., Watanabe, T., Takemasa, S.; Masuhiro, Y., Ohtake, F., Matsumoto, T. Function of nuclear sex hormone recep- tors in gene regulation. Cancer Chemother. Pharmacol. 2005, 56 (Suppl.1): 4-9. [15] Björnström, L., Sjöberg, M. Mechanisms of estrogen receptor signaling: convergence of genomic and nongenomic actions on tar- get genes. Mol. Endocrinol. 2005, 19: 833-842. [16] Levin, E.R. Integration of the extra-nuclear and nuclear actions of estrogen. Mol. Endocrinol. 2005, 19: 1951-1959. [17] Marino, M., Ascenzi, P., Acconcia, F. S-palmitoylation modulates estrogen receptor alpha localization and functions. Steroids 2006, 71: 298-303. [18] Ascenzi, P., Bocedi, A., Marino, M. Structure-function relationship of estrogen receptor and : impact on human health. Mol. Aspects Med. 2006, 27: 299-402. [19] Zhang, D., Trudeau, V.L. Integration of membrane and nuclear estrogen receptor signaling. Comp. Biochem. Physiol. A Mol. In- tegr. Physiol. 2006, 144: 306-315. [20] Acconcia, F., Marino, M. Synergism between genomic and non- genomic estrogen action mechanisms. IUBMB Life 2003, 55: 145- 150. [21] Kousteni, S., Han, L., Chen, J.R., Almeida, M., Plotkin, L.I., Bel- lido, T., Manolagas, S.C. Kinase-mediated regulation of common transcription factors accounts for the bone-protective effects of sex steroids. J. Clin. Invest. 2003, 111: 1651-1664. [22] Matthews, J., Gustafsson, J.-Å. Estrogen signalling: A subtle bal- ance between ER and ER. Mol. Interventions 2003, 3: 281-292. [23] Li, X., Huang, J., Yi, P., Bambara, R.A., Hilf, R., Muyan, M. Sin- gle-chain estrogen receptors (ERs) reveal that the ER/ heterodi- mer emulates functions of the ER dimer in genomic estrogen sig- naling pathways. Mol. Cell. Biol. 2004, 24: 7681-7694. [24] Monroe, D.G., Secreto, F.J., Subramaniam, M., Getz, B.J., Khosla, S., Spelsberg, T.C. Estrogen receptor and heterodimers exert unique effects on estrogen-and tamoxifen-dependent gene expres- sion in human U2OS osteosarcoma cells. Mol. Endocrinol. 2005, 19: 1555-1568. [25] Mendelsohn, M.E. Genomic and nongenomic effects of estrogen in the vasculature. Am. J. Cardiol. 2002, 90: 3F-6F. [26] Gosden, J.R., Middleton, P.G., Rout, D. Localization of the human oestrogen receptor gene to chromosome 6q24-q27 by in situ hy- bridization. Cytogenet. Cell Genet. 1986, 43: 218-220. [27] Enmark, E., Pelto-Huikko, M., Grandien, K., Lagercrantz, S., La- gercrantz, J., Fried, G., Nordenskjold, M., Gustafsson, J.-Å. Human estrogen receptor -gene structure, chromosomal localization, and expression pattern. J. Clin. Endocrinol. Metab. 1997, 82: 4258- 4265. [28] Luisi, S., Galleri, L., Marini, F., Ambrosiani, G., Brandi, M.L., Petraglia, F. Estrogen receptor gene polymorphisms are associated with recurrence of endometriosis. Fertil. Steril. 2006, 85: 764-766. [29] Zhou, W., Liu, Z., Wu, J., Liu, J.H., Hyder, S.M., Antoniou, E., Lubahn, D.B. Identification and characterization of two novel splicing isoforms of human estrogen-related receptor . J. Clin. Endocrinol. Metab. 2006, 91: 569-579. [30] Mosselman, S., Polman, J., Dijkema, R. ER: identification and characterization of a novel human estrogen receptor. FEBS Lett. 1996, 392: 49-53. [31] Claessens, F., Gewirth, D.T. DNA recognition by nuclear recep- tors. In: McEwan, I.J. (Ed.), Essay in Biochemistry: The Nuclear Receptor Superfamily Portland Press, London, 2004, 59-72. [32] Kumar, R., Johnson, B.H., Thompson, E.B. Overview of the struc- tural basis for transcription regulation by nuclear hormone recep- tors. In: McEwan, I.J. (Ed.), Essay in Biochemistry: The Nuclear Receptor Superfamily Portland Press, London, 2004, pp. 27-39. [33] McEwan, I.J. Sex, drugs and gene expression: signalling by mem- bers of the nuclear receptor superfamily. In: McEwan, I.J. (Ed.), Essays in Biochemistry: the Nuclear Receptor Superfamily. Port- land Press, London, 2004, pp. 1-10. [34] Ortì, E., Bodwell, J.E., Munck, A. Phosphorylation of steroid hor- mone receptors. Endocr. Rev. 1992, 13: 105-128. [35] Lannigan, D.A. Estrogen receptor phosphorylation. Steroids 2003, 68: 1-9. [36] Herynk, M.H., Fuqua, S.A. Estrogen receptor mutations in human disease. Endocr. Rev. 2004, 25: 869-898. [37] O’Malley, B.W. A life-long search for the molecular pathways of steroid hormone action. Mol. Endocrinol. 2005, 19: 1402-1411. [38] Beato, M. Gene regulation by steroid hormones. Cell 1989, 56: 335-344. [39] Walker, P., Germond, J.-E., Brown-Luedi, M., Givel, F., Wahli, W. Sequence homologies in the region preceding the transcription ini- tiation site of the liver estrogen-responsive vitellogenin and apo- VLDLII genes. Nucleic Acids Res. 1984, 12: 8611-8626. [40] Klein-Hitpass, L., Schorpp, M., Wagner, U., Ryffel, G.U. An es- trogen-responsive element derived from the 5’ flanking region of the Xenopus vitellogenin A2 gene functions in transfected human cells. Cell 1986, 46: 1053-1061. [41] Ponglikitmongkol, M., Green, S., Chambon, P. Genomic organiza- tion of the human oestrogen receptor gene. EMBO J. 1988, 7: 3385-3388. [42] O’Lone, R., Frith, M.C., Karlsson, E.K., Hansen, U. Genomic targets of nuclear estrogen receptors. Mol. Endocrinol. 2004, 18: 1859-1875. [43] Kato, S., Tora, L., Yamauchi, J., Masushige, S., Bellard, M., Chambon, P. A far upstream estrogen response element of the ovalbumin gene contains several half palindromic 5’-TGACC-3’ motifs acting synergistically. Cell 1992, 68: 731-742. [44] Klinge, C.M., Bodenner, D.L., Desai, D., Niles, R.M., Traish, A.M. Binding of type II nuclear receptors and estrogen receptor to full and half-site estrogen response elements in vitro. Nucleic Acids Res. 1997, 25: 1903-1912.
  • 14. 506 Current Genomics, 2006, Vol. 7, No. 8 Marino et al. [45] Porter, W., Wang, F., Wang, W.; Duan, R., Safe, S. Role of estro- gen receptor/Sp1 complexes in estrogen-induced heat shock protein 27 gene expression. Mol. Endocrinol. 1996, 10: 1371-1378. [46] Porter, W., Saville, B., Hoivik, D., Safe, S. Functional synergy between the transcription factor Sp1 and the estrogen receptor. Mol. Endocrinol. 1997, 11: 1569-1580. [47] Bajic, V.B., Tan, S.L., Chong, A., Tang, S., Strom, A., Gustafsson, J.-Å., Lin, C.Y., Liu, E.T. Dragon ERE Finder version 2: A tool for accurate detection and analysis of estrogen response elements in vertebrate genomes. Nucleic Acids Res. 2003, 31: 3605-3607. [48] Bourdeau, V., Deschenes, J., Métivier, R., Nagai, Y., Nguyen, D., Bretschneider, N., Gannon, F., White, J.H., Mader, S. Genome- wide identification of high-affinity estrogen response elements in human and mouse. Mol. Endocrinol. 2004, 18: 1411-1427. [49] Carroll, J.S., Brown, M. Estrogen receptor target gene: an evolving concept in molecular endocrinology. Mol. Endocrinol. 2006, 20: 1707-1714. [50] Nunez, A.M., Jakowlev, S., Briand, J.P., Gaire, M., Krust, A., Rio, M.C., Chambon, P. Characterization of the estrogen-induced pS2 protein secreted by the human breast cancer cell line MCF-7. En- docrinology 1987, 121: 1759-1765. [51] Sausville, E., Carney, D., Battey, J. The human vasopressin gene is linked to the oxytocin gene and is selectively expressed in a cul- tured lung cancer cell line. J. Biol. Chem. 1985, 260: 10236-10241. [52] Loven, M.A., Wood, J.R., Nardulli, A.M. Interaction of estrogen receptors and with estrogen response elements. Mol. Cell. En- docrinol. 2001, 181: 151-163. [53] Loven, M.A., Likhite, V.S., Choi, I., Nardulli, A.M. Estrogen re- sponse elements alter coactivator recruitment through allosteric modulation of estrogen receptor conformation. J. Biol. Chem. 2001, 276: 45282-45288. [54] Wood, J.R., Likhite, V.S., Loven, M.A., Nardulli, A.M. Allosteric modulation of estrogen receptor conformation by different estrogen response elements. Mol. Endocrinol. 2001, 15: 1114-1126. [55] Yi, P., Driscoll, M.D., Huang, J., Bhagat, S., Hill, R., Bambara, R.A., Muyan, M. The effects of estrogen-responsive element- and ligand-induced structural changes on the recruitment of cofactors and transcriptional responses by ER and ER. Mol. Endocrinol. 2002, 16: 674-693. [56] Lefstin, J.A., Yamamoto, K.R. Allosteric effects of DNA on transcriptional regulators. Nature 1998, 392: 885-888. [57] Hall, J.M., McDonnell, D.P., Korach, K.S. Allosteric regulation of estrogen receptor structure, function, and coactivator recruitment by different estrogen response elements. Mol. Endocrinol. 2002, 16: 469-486. [58] Ikeda, M., Wilcox, E.C., Chin, W.W. Different DNA elements can modulate the conformation of thyroid hormone receptor heterodi- mer and its transcriptional activity. J. Biol. Chem. 1996, 271: 23096-23104. [59] Gottlicher, M., Heck, S., Herrlich, P. Transcriptional cross-talk, the second mode of steroid hormone receptor action. J. Mol. Med. 1998, 76: 480-489. [60] Aranda, A., Pascual A. Nuclear hormone receptors and gene ex- pression. Physiol Rev. 2001, 81: 1269-1304. [61] Chambliss, K.L., Shaul, P.W. Rapid activation of endothelial NO synthase by estrogen: evidence for a steroid receptor fast-action complex (SRFC) in caveolae. Steroids 2002, 67: 413-419. [62] Duan, R., Porter, W., Safe, S. Estrogen-induced c-fos protoonco- gene expression in MCF-7 human breast cancer cells: role of estro- gen receptor Sp1 complex formation. Endocrinology 1998, 139: 1981-1990. [63] Castro-Rivera, E., Samudio, I., Safe, S. Estrogen regulation of cyclin D1 gene expression in ZR-75 breast cancer cells involves multiple enhancer elements. J. Biol. Chem. 2001, 276: 30853- 30861. [64] Sun, G., Porter, W., Safe, S. Estrogen-induced retinoic acid recep- tor 1 gene expression: role of estrogen receptor-Sp1 complex. Mol. Endocrinol. 1998, 12: 882-890. [65] Zhang, J., Hu, X., Lazar M.A. A novel role for helix 12 of retinoid X receptor in regulating repression. Mol. Cell. Biol. 1999, 19: 6448-6457. [66] Batistuzzo de Medeiros, S.R., Krey, G., Hihi, A.K., Wahli, W. Functional interaction between the estrogen receptor and the tran- scription activator Sp1 regulate the estrogen-dependent transcrip- tional activity of the vitellogenin A1 promoter. J. Biol. Chem. 1997, 272: 18250-18260. [67] Qin, C., Singh, P., Safe, S. Transcriptional activation of insulin-like growth factor-binding protein-4 by 17-estradiol in MCF-7 cells: role of estrogen receptor-SP1 complexes. Endocrinology 1999, 140: 2501-2508. [68] Galien, R., Garcia, T. Estrogen receptor impairs interleukin-6 expression by preventing protein binding on the NF-B site. Nucleic Acids Res. 1997, 25: 2424-2429. [69] Ray, A., Prefontaine, K.E., Ray, P. Down-modulation of interleu- kin-6 gene expression by estradiol 17 in the absence of high affin- ity DNA binding by the estrogen receptor. J. Biol. Chem. 1994, 269: 12940-12946. [70] Kalaitzidis, D., Gilmore, T.D. Transcription factor cross-talk: the estrogen receptor and NF-B. Trends Endocrinol. Metab. 2005, 16: 46-52. [71] Gaub, M.P., Bellard, M., Scheuer, I., Chambon, P., Sassone-Corsi, P. Activation of the ovalbumin gene by the estrogen receptor in- volves the fos-jun complex. Cell 1990, 3: 1267-1276. [72] Marino, M., Acconcia, F., Bresciani, F., Weisz, A., Trentalance A. Distinct nongenomic signal transduction pathways controlled by 17-estradiol regulate DNA synthesis and cyclin D1 gene transcrip- tion in HepG2 cells. Mol. Biol. Cell 2002, 13: 3720-3729. [73] Fujimoto, N., Honda, H., Kitamura, S. Effects of environmental estrogenic chemicals on AP-1 mediated transcription with estrogen receptors and . J. Steroid Biochem. Mol. Biol. 2004, 88: 53-59. [74] Paech, K., Webb, P., Kuiper, G.G., Nilsson, S., Gustafsson, J., Kushner, P.J., Scanlan, T.S. Differential ligand activation of estro- gen receptors ER and ER at AP-1 sites. Science 1997, 277: 1508-1510. [75] Liu, M.M., Albanese, C., Anderson, C.M., Hilty, K., Webb, P., Uht, R.M., Price, R.H.Jr., Pestell, R.G., Kushner, P.J. Opposing ac- tion of estrogen receptors and on cyclin D1 gene expression. J. Biol. Chem. 2002, 277: 24353-24360. [76] Altucci, L., Addeo, R., Cicatiello, L., Dauvois, S., Parker, M.G., Truss, M., Beato, M., Sica, V., Bresciani, F., Weisz, A. 17- Estradiol induces cyclin D1 gene transcription, p36D1-p34cdk4 complex activation and p105Rb phosphorylation during mitogenic stimulation of G1-arrested human breast cancer cells. Oncogene 1996, 12: 2315-2324. [77] Foster, J.S., Wimalasena, J. Estrogen regulates activity of cyclin- dependent kinases and retinoblastoma protein phosphorylation in breast cancer cells. Mol. Endocrinol. 1996, 10: 488-498. [78] Prall, O.W., Sarcevic, B., Musgrove, E.A., Watts, C.K., Sutherland, R.L. Estrogen-induced activation of Cdk4 and Cdk2 during G1-S phase progression is accompanied by increased cyclin D1 expres- sion and decreased cyclin-dependent kinase inhibitor association with cyclin E-Cdk2. J. Biol. Chem. 1997, 272: 10882-10894. [79] Herbert, B., Truss, M., Beato, M., Müller, R. Inducibile regulatory elements in the human cyclin D1 promoter. Oncogene 1994, 9: 1295-1304. [80] Marino, M., Acconcia, F., Trentalance, A. Biphasic estradiol in- duced AKT-phosphorylation is modulated by PTEN via MAP kinase in HepG2 cells. Mol. Biol. Cell 2003, 14: 2583-2591. [81] Acconcia, F., Totta, P., Ogawa, S., Cardillo, I., Inoue, S., Leone, S., Trentalance, A., Muramatsu, M., Marino, M. Survival versus apop- totic 17-estradiol effect: role of ER and ER activated non- genomic signalling. J. Cell. Physiol. 2005, 203: 193-201. [82] Bardin, A., Boulle, N., Lazennec, G., Vignon, F., Pujol, P. Loss of ER expression as a common step in estrogen-dependent tumor progression. Endocr. Relat. Cancer 2004, 11: 537-551. [83] McKenna, N.J., Lanz, R.B., O’Malley, B.W. Nuclear receptor coregulators: cellular and molecular biology. Endocr. Rev. 1999, 20: 321-344. [84] Cavailles, V., Dauvois, S., L’Horset, F., Lopez, G., Hoare, S., Kushner, P.J., Parker, M.G. Nuclear factor RIP140 modulates tran- scriptional activation by the estrogen receptor. EMBO J. 1995, 14: 3741-3751. [85] Treuter, E., Johansson, L., Thomsen, J.S., Wärnmark, A., Leers, J., Pelto-Huikko, M., Sjöberg, M., Wright, A.P., Spyrou, G., Gustafsson, J.-Å. Competition between thyroid hormone receptor- associated protein (TRAP) 220 and transcriptional intermediary factor (TIF) 2 for binding to nuclear receptors. Implications for the recruitment of TRAP and p160 coactivator complexes. J. Biol. Chem. 1999, 274: 6667-6677. [86] Johansson, L., Bavner, A., Thomsen, J.S., Farnegardh, M., Gustafsson, J.-Å., Treuter, E. The orphan nuclear receptor SHP utilizes conserved LXXLL-related motifs for interactions with
  • 15. Estrogen Signaling and Gene Transcription Current Genomics, 2006, Vol. 7, No. 8 507 ligand-activated estrogen receptors. Mol. Cell. Biol. 2000, 20: 1124-1133. [87] Leclercq, G., Lacroix, M., Laios, I., Laurent, G. Estrogen receptor : impact of ligands on intracellular shuttling and turnover rate in breast cancer cells. Curr. Cancer Drug Targets 2006, 6: 39-64. [88] Wärnmark, A., Almlöf, T., Leers, J., Gustafsson, J.-Å., Treuter, E. Differential recruitment of the mammalian mediator subunit TRAP220 by estrogen receptors ER and ER. J. Biol. Chem. 2001, 276: 23397-23404. [89] Barkhem, T., Carlsson, B., Nilsson, Y., Enmark, E., Gustafsson, J.- Å., Nilsson, S. Differential response of estrogen receptor and es- trogen receptor to partial estrogen agonists/antagonists. Mol. Pharmacol. 1998, 54: 105-112. [90] McInerney, E.M., Weis, K.E., Sun, J., Mosselman, S., Katzenellen- bogen, B.S. Transcription activation by the human estrogen recep- tor subtype (ER ) studied with ER and ER receptor chimeras. Endocrinology 1998, 139: 4513-4522. [91] Hall, J.M., McDonnell, D.P. The estrogen receptor -isoform (ER) of the human estrogen receptor modulates ER transcrip- tional activity and is a key regulator of the cellular response to es- trogens and antiestrogens. Endocrinology 1999, 140: 5566-5578. [92] Acevedo, M.L., Kraus, W.L. Transcriptional activation by nuclear receptors. In: McEwan, I.J. (Ed.), Essays in Biochemistry: the Nu- clear Receptor Superfamily. Portland Press, London, 2004, pp. 73- 88. [93] Kim, M.Y., Woo, E.M.; Chong, Y.T., Homenko, D.R., Kraus, W.L. Acetylation of estrogen receptor alpha by p300 at lysines 266 and 268 enhances the DNA binding and transactivation activities of the receptor.. Mol. Endocrinol. 2006, 20: 1479-1493. [94] Rowan, B.G., Weigel, N.L., O’Malley, B.W. Phosphorylation of steroid receptor coactivator-1. Identification of the phosphorylation sites and phosphorylation through the mitogen-activated protein kinase pathway. J. Biol. Chem. 2000, 275: 4475-4483. [95] Ko, L., Cardona, G.R., Iwasaki, T., Bramlett, K.S., Burris, T.P., Chin, W.W. Ser-884 adjacent to the LXXLL motif of coactivator TRBP defines selectivity for ERs and TRs. Mol. Endocrinol. 2002, 16: 128-140. [96] Farach-Carson, M.C., Davis, P.J. Steroid hormone interactions with target cells: cross talk between membrane and nuclear pathways. J. Pharmacol. Exper. Therap. 2003, 30, 839-845. [97] Marino, M., Acconcia, F., Ascenzi, P. Estrogen receptor signalling: Bases for drug actions. Curr. Drug Targets - Immune, Endocrine Metabolic Disorders 2005, 5: 305-314. [98] Szego, C.M., Davis, J.S. Adenosine 3’,5’-monophosphate in rat uterus: acute elevation by estrogen. Proc. Natl. Acad. Sci. U.S.A. 1967, 58: 1711-1718. [99] Kampa, M., Castanas, E. Membrane steroid receptor signaling in normal and neoplastic cells. Mol. Cell. Endocrinol. 2006, 246: 76- 82. [100] Morley, P., Whitfield, J.F., Vanderhyden, B.C., Tsang, B.K., Schwartz, J.L. A new, nongenomic estrogen action: the rapid re- lease of intracellular calcium. Endocrinology 1992, 131: 1305- 1312. [101] Marino, M., Pallottini, V., Trentalance, A. Estrogens cause rapid activation of IP3-PKC- signal transduction pathway in HEPG2 cells. Biochem. Biophys. Res. Commun. 1998, 245: 254-258. [102] Marino, M., Ficca, R., Ascenzi, P., Trentalance, A. Nitric oxide inhibits selectively the 17-estradiol-induced gene expression without affecting nongenomic events in HeLa cells. Biochem. Bio- phys. Res. Commun. 2001, 286: 529-533. [103] Marino, M., Distefano, E., Trentalance, A., Smith, C. L. Estradiol induced IP3 mediate the estrogen receptor activity expressed in human cells. Mol. Cell. Endocrinol. 2001, 182: 19-26. [104] Picotto, G., Vazquez, G., Boland, R. 17-oestradiol increases intra- cellular Ca2+ concentration in rat enterocytes. Potential role of phospholipase C-dependent store-operated Ca2+ influx. Biochem. J. 1999, 339: 71-77. [105] Perret, S., Dockery, P., Harvey, B.J. 17-oestradiol stimulates capacitative Ca2+ entry in human endometrial cells. Mol. Cell. En- docrinol. 2001, 176: 77-84. [106] Incerpi, S., D’Arezzo, S., Marino, M., Musanti, R., Pallottini, V., Pascolini, A., Trentalance, A. Short-term activation by low 17- estradiol concentrations of the Na+ /H+ exchanger in rat aortic smooth muscle cells: physiopathological implications. Endocrinol- ogy 2003, 144: 4315-4324. [107] Watters, J.J., Campbell, J.S., Cunningham, M.J., Krebs, E.G., Dorsa, D.M. Rapid membrane effects of steroids in neuroblastoma cells: effects of estrogen on mitogen activated protein kinase sig- nalling cascade and c-fos immediate early gene transcription. En- docrinology 1997, 138: 4030-4033. [108] Russell, K.S., Haynes, M.P., Sinha, D., Clerisme, E., Bender, J.R. Human vascular endothelial cells contain membrane binding sites for estradiol, which mediate rapid intracellular signaling. Proc. Natl. Acad. Sci. U. S. A. 2000, 97, 5930–5935. [109] Dos Santos, E.G., Dieudonne, M.N., Pecquery, R., Le Moal, V., Giudicelli, Y., Lacasa, D. Rapid nongenomic E2 effects on p42/p44 MAPK, activator protein-1, and cAMP response element binding protein in rat white adipocytes. Endocrinology 2002, 143: 930-940. [110] Migliaccio, A., Castoria, G., Di Domenico, M., de Falco, A., Bilan- cio, A., Auricchio, F. Src is an initial target of sex steroid hormone action. Ann. N.Y. Acad. Sci. 2002, 963: 185-190. [111] Tanaka, Y., Gavrielides, M.V., Mitsuuchi, Y., Fujii, T., Kazanietz, M.G. Protein kinase C promotes apoptosis in LNCaP prostate can- cer cells through activation of p38 MAPK and inhibition of the Akt survival pathway. J. Biol. Chem. 2003, 278: 33753-33762. [112] Klinge, C.M., Blankenship, K.A., Risinger, K.E., Bhatnagar, S., Noisin, E.L., Sumanasekera, W.K., Zhao, L., Brey, D.M., Keynton, R.S. Resveratrol and estradiol rapidly activate MAPK signaling through estrogen receptors and in endothelial cells. J. Biol. Chem. 2005, 280, 7460-7468. [113] Woo, C.H., Lim, J.H., Kim, J.H. VCAM-1 upregulation via PKC- p38 kinase-linked cascade mediates the TNF--induced leukocyte adhesion and emigration in the lung airway epithelium. Am. J. Physiol. Lung Cell. Mol. Physiol. 2005, 288: L307-L316. [114] Castoria, G., Barone, M.V., Di Domenico, M., Bilancio, A., Ametrano, D., Migliaccio, A., Auricchio, F. Non-trascriptional ac- tion of oestradiol and progestin triggers DNA synthesis. EMBO J. 1999, 18: 2500-2510. [115] Castoria, G., Migliaccio, A., Bilancio, A., Di Domenico, M., de Falco, A., Lombardi, M., Fiorentino, R., Varricchio, L., Barone, M.V., Auricchio, F. PI3-kinase in concert with Src promotes the S- phase entry of oestradiol-stimulated MCF-7 cells. EMBO J. 2001, 20: 6050-6059. [116] Chambliss, K.L., Simon, L., Yuhanna, I.S., Mineo, C., Shaul, P.W. Dissecting the basis of nongenomic activation of eNOS by estra- diol: role of ER domains with known nuclear functions. Mol. En- docrinol. 2005, 19: 277-289. [117] Simoncini, T., Hafezi-Moghadam, A., Brazil, D.P., Ley, K., Chin, W.W., Liao, J.K. Interaction of oestrogen receptor with the regula- tory subunit of phosphatidylinositol-3-OH kinase. Nature 2000, 407: 538-541. [118] Alexaki, V.I., Charalampopoulos, I., Kampa, M., Nifli, A.P., Hat- zoglou, A., Gravanis, A., Castanas, E. Activation of membrane estrogen receptors induce pro-survival kinases. J. Steroid Biochem. Mol. Biol. 2006, 98: 97-110. [119] Farhat, M.Y., Abi-Younes, S., Dingaan, B., Vargas, R., Ramwell, P.W. Estradiol increases cyclic adenosine monophosphate in rat pulmonary vascular smooth muscle cells by a nongenomic mecha- nism. J. Pharmacol. Exp. Ther. 1996, 276: 652-657. [120] Gu, Q., Moss, R.L. 17-estradiol potentiates kainate-induced cur- rents via activation of the cAMP cascade. J. Neurosci. 1996, 16: 3620-3629. [121] Picotto, G.; Massheimer, V., Boland, R. Acute stimulation of intes- tinal cell calcium influx induced by 17-estradiol via the cAMP messenger system. Mol. Cell. Endocrinol. 1996, 119: 129-134. [122] Chen, Z.J., Yu, L., Chang, C.H. Stimulation of membrane-bound guanylate cyclase activity by 17- estradiol. Biochem. Biophys. Res. Commun. 1998, 252: 639-642. [123] Malyala, A., Kelly, M.J., Ronnekleiv, O.K. Estrogen modulation of hypothalamic neurons, activation of multiple signaling pathways and gene expression changes. Steroids 2005, 70. 397-406. [124] Kahlert, S., Nuedling, S., van Eickels, M., Vetter, H., Meyer, R., Grohe, C. Estrogen receptor rapidly activates the IGF-1 receptor pathway. J. Biol. Chem. 2000, 275: 18447-18453. [125] Improta-Brears, T., Whorton, A.R., Codazzi, F., York, J.D., Meyer, T., McDonnell, D.P. Estrogen-induced activation of mitogen- activated protein kinase requires mobilization of intracellular cal- cium. Proc. Natl. Acad. Sci. U. S. A. 1999, 96: 4686-4691. [126] Driggers, P.H., Segars, J.H. Estrogen action and cytoplasmic sig- naling pathways: Part II. The role of growth factors and phosphory-
  • 16. 508 Current Genomics, 2006, Vol. 7, No. 8 Marino et al. lation in estrogen signaling. Trends Endocrinol. Metab. 2002, 13: 422-427. [127] Razandi, M., Alton, G., Pedram, A., Ghonshani, S., Webb, P., Levin, E.R. Identification of a structural determinant necessary for the localization and function of estrogen receptor at the plasma membrane. Mol. Cell. Biol. 2003, 23: 1633-1646. [128] Zhang, Z., Kumar, R., Santen, R.J., Song, R.X.D. The role of adapter protein Shc in estrogen non-genomic action. Steroids 2004, 69: 523-529. [129] Kupzig, S., Walker, S.A., Cullen, P.J. The frequencies of calcium oscillations are optimized for efficient calcium-mediated activation of Ras and the ERK/MAPK cascade. Proc. Natl. Acad. Sci. U. S. A. 2005, 102: 7577-7582. [130] Chambliss, K.L., Yuhanna, I.S., Anderson, R.G., Mendelsohn, M.E., Shaul, P.W. ER has nongenomic action in caveolae. Mol. Endocrinol. 2002, 16: 938-946. [131] Kim, K.H., Bender, J.R. Rapid, estrogen receptor-mediated signal- ing: why is the endothelium so special? Sci. STKE 2005, 14: pe28. [132] Ansonoff, M.A., Etgen, A.M. Estradiol elevates protein kinase C catalytic activity in the preoptic area of female rats. Endocrinology 1998, 139: 3050-3056. [133] Razandi, M., Pedram, A., Greene, G.L., Levin, E.R. Cell membrane and nuclear estrogen receptors (ERs) originate from a single tran- script: studies of ER and ER expressed in Chinese hamster ovary cells. Mol. Endocrinol. 1999, 13: 307-319. [134] Geraldes, P., Sirois, M.G., Tanguay, J.F. Specific contribution of estrogen receptors on mitogen-activated protein kinase pathways and vascular cell activation. Circ. Res. 2003, 93: 399-405. [135] Marino, M., Galluzzo, P., Leone, S., Acconcia, F., Ascenzi, P. Nitric oxide impairs the 17-estradiol-induced apoptosis in human colon adenocarcinoma cells. Endocr. Relat. Cancer 2006, 13: 559- 569. [136] Acconcia, F., Kumar, R. Signaling regulation of genomic and non- genomic functions of estrogen receptors. Cancer Lett. 2005, 238: 1- 14. [137] Ahola, T.M., Manninen, T., Alkio, N., Ylikomi, T. G protein- coupled receptor 30 is critical for a progestin-induced growth inhi- bition in MCF-7 breast cancer cells. Endocrinology 2002, 143: 3376-3384. [138] Filardo, E.J., Quinn, J.A., Frackelton, A.R. Jr., Bland, K.I. Estrogen action via the G protein-coupled receptor, GPR30: stimulation of adenylyl cyclase and cAMP-mediated attenuation of the epidermal growth factor receptor-to-MAPK signaling axis. Mol. Endocrinol. 2002, 16: 70-84. [139] Ropero, A.B., Soria, B., Nadal, A. A nonclassical estrogen mem- brane receptor triggers rapid differential actions in the endocrine pancreas. Mol. Endocrinol. 2002, 16: 497-505. [140] Toran-Allerand, C.D., Guan, X., MacLusky, N.J., Horvath, T.L., Diano, S., Singh, M., Connolly, E.S.Jr., Nethrapalli, I.S., Tinnikov, A.A. ER-X: a novel, plasma membrane-associated, putative estro- gen receptor that is regulated during development and after ischemic brain injury. J. Neurosci. 2002, 22: 8391-8401. [141] Thomas, P., Pang, Y., Filardo, E.J., Dong, J. Identity of an estrogen membrane receptor coupled to a G protein in human breast cancer cells. Endocrinology 2005, 146: 624-632. [142] Vivacqua, A., Bonofiglio, D., Recchia, A.G., Musti, A.M., Picard, D., Andò, S., Maggiolini, M. The G protein-coupled receptor GPR30 mediates the proliferative effects induced by 17-estradiol and hydroxytamoxifen in endometrial cancer cells. Mol. Endocri- nol. 2006, 20: 631-646. [143] Pappas, T.C., Gametchu, B., Watson, C.S. Membrane estrogen receptors identified by multiple antibody labeling and impeded- ligand binding. FASEB J. 1995, 9: 404-410. [144] Norfleet, A.M., Thomas, M.L., Gametchu, B., Watson, C.S. Estro- gen receptor- detected on the plasma membrane of aldehyde-fixed GH3/B6/F10 rat pituitary tumor cells by enzyme-linked immuno- cytochemistry. Endocrinology 1999, 140: 3805-3814. [145] Dan, P., Cheung, J.C., Scriven, D.R., Moore, E.D. Epitope depend- ent localization of estrogen receptor-, but not -, in en face arte- rial endothelium. Am. J. Physiol. 2003, 284: H1295-H1306. [146] Acconcia, F., Ascenzi, P., Bocedi, A., Spisni, E., Tomasi, V., Tren- talance, A., Visca, P., Marino, M. Palmitoylation-dependent estro- gen receptor membrane localization: regulation by 17-estradiol. Mol. Biol. Cell 2005, 16: 231-237. [147] Acconcia, F., Bocedi, A., Ascenzi, P., Marino, M. Does palmitoyla- tion target estrogen receptors to plasma membrane caveolae? IUBMB Life 2003, 55: 33-35. [148] Bijlmakers, M.J., Marsh, M. The on-off story of protein palmitoy- lation. Trends Cell Biol. 2003, 13: 32-42. [149] Kennedy, A.M., Shogren, K.L., Zhang, M., Turner, R.T., Spels- berg, T.C., Maran, A. 17-estradiol-dependent activation of signal transducer and activator of transcription-1 in human fetal os- teoblasts is dependent on Src kinase activity. Endocrinology 2005, 146, 201-207. [150] Song, R.X.D., Zhang, Z., Santen, R.J. Estrogen rapid action via protein complex formation involving ER and Src. Trends Endo- crinol. Metab. 2005, 16: 347-353. [151] Greger, J.G., Guo, Y., Henderson, R., Ross, J.F., Cheskis, B.J. Characterization of MNAR expression. Steroids 2006, 71: 317-322. [152] Song, R.X.D., Barnes, C.J., Zhang, Z., Bao, Y., Kumar, R., Santen, R.J. The role of Shc and insulin-like growth factor 1 receptor in mediating the translocation of estrogen receptor to the plasma membrane. Proc. Natl. Acad. Sci. U.S.A. 2004, 101: 2076-2081. [153] Warner, M., Gustafsson, J.-Å. Nongenomic effects of estrogen: why all the uncertainty? Steroids 2006, 71: 91-95. [154] Paruthiyil, S., Parmar, H., Kerekatte, V., Cunha, G.R., Firestone, G.L., Leitman, D.C. Estrogen receptor inhibits human breast can- cer cell proliferation and tumor formation by causing a G2 cell cy- cle arrest. Cancer Res. 2004, 64: 423-428. [155] Lu, Q., Ebling, H., Mittler, J., Baur, W.E., Karas, R.H. MAP kinase mediates growth factor-induced nuclear translocation of estrogen receptor . FEBS Lett. 2002, 516: 1-8. [156] Yamakawa, K., Arita, J. Cross-talk between the estrogen receptor-, protein kinase A-, and mitogen-activated protein kinase-mediated signaling pathways in the regulation of lactotroph proliferation in primary culture. J. Steroid Biochem. Mol. Biol. 2004, 88: 123-130. [157] Pedram, A., Razandi, M., Aitkenhead, M., Hughes, C.C., Levin, E.R. Integration of the non-genomic and genomic actions of estro- gen. Membrane-initiated signaling by steroid to transcription and cell biology. J. Biol. Chem. 2002, 277: 50768-50775. [158] Wade, C.B., Dorsa, D.M. Estrogen activation of cyclic adenosine 5- monophosphate response element-mediated transcription re- quires the extracellularly regulated kinase/mitogen-activated pro- tein kinase pathway. Endocrinology 2003, 144: 832-838. [159] Hennessy, B.A., Harvey, B.J., Healy, V. 17-Estradiol rapidly stimulates c-fos expression via the MAPK pathway in T84 cells. Mol. Cell. Endocrinol. 2005, 229: 39-47. [160] Watters, J.J., Dorsa, D.M. Transcriptional effects of estrogen on neuronal neurotensin gene expression involve cAMP/protein kinase A-dependent signaling mechanisms. J. Neurosci. 1998, 18: 6672- 6680. [161] Foster, J.S., Henley, D.C., Ahamed, S. Estrogens and cell-cycle regulation in breast cancer. Trends Endocrinol. Metabol. 2001, 12: 320-327. [162] Marino, M., Ascenzi, P. Estrogen receptor-: Plasma membrane localization and functions. Immun. Endoc. Metab. Agents in Med. Chem. 2006, 6: 281-286. [163] Schwabe, J.W.R., Chapman, L., Finch, J.T., Rhodes, D. The crystal structure of the estrogen receptor DNA-binding domain bound to DNA: how receptors discriminate between their response elements. Cell 1993, 75: 567-578.
  • 17.   Open Peer Review F1000 Faculty Reviews are written by members of the prestigious  . They are F1000 Faculty commissioned and are peer reviewed before publication to ensure that the final, published version is comprehensive and accessible. The reviewers who approved the final version are listed with their names and affiliations. Any comments on the article can be found at the end of the article. REVIEW Re-highlighting the action of PPARγ in treating metabolic   diseases [version 1; peer review: 2 approved] Sung Hee Choi ,    Sung Soo Chung , Kyong Soo Park 1,4 Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea Abstract Peroxisome proliferator-activated receptor γ (PPARγ) is a member of the nuclear receptor family and plays an important role in adipocyte differentiation, glucose homeostasis, and insulin sensitivity. Thiazolidinediones (TZDs), synthetic ligands of PPARγ, have been used for the treatment of diabetes mellitus for two decades. TZDs were expected to be amazing drugs not only for type 2 diabetes but also for metabolic syndrome and atherosclerotic vascular disease because they can reduce both insulin resistance and inflammation in experimental studies. However, serious unwanted effects pushed TZDs back to an optional second-tier drug for type 2 diabetes. Nevertheless, PPARγ is still one of the most important targets for the treatment of insulin resistance and diabetes mellitus, and novel strategies to modulate PPARγ activity to enhance its beneficial effects and reduce unwanted adverse effects are anticipated. Recent studies showed that post-translational modification (PTM) of PPARγ regulates PPARγ activity or stability and may be a novel way to optimize PPARγ activity with reduced adverse effects. In this review, we will focus on recent advances in PTM of PPARγ and the mechanisms regulating PPARγ function as well as in the development of PPARγ modulators or agonists. Keywords PPARgamma, post-translational modification, metabolic disease 1,2 1,3 1,4 1 2 3 4     Reviewer Status   Invited Reviewers   version 1 published 24 Jul 2018   1 2 , SBP Medical Discovery Institute, Laszlo Nagy Florida, USA University of Debrecen, Debrecen, Hungary , University of Debrecen, Debrecen, Attila Pap Hungary 1 , Skolkovo Innovative Alexander Orekhov Center, Moscow, Russian Federation 2  24 Jul 2018,  (F1000 Faculty Rev):1127 ( First published: 7 ) https://doi.org/10.12688/f1000research.14136.1  24 Jul 2018,  (F1000 Faculty Rev):1127 ( Latest published: 7 ) https://doi.org/10.12688/f1000research.14136.1 v1 Page 1 of 9 F1000Research 2018, 7(F1000 Faculty Rev):1127 Last updated: 17 JUL 2019
  • 18.    Kyong Soo Park ( ) Corresponding author: kspark@snu.ac.kr   : Conceptualization, Data Curation, Resources, Writing – Original Draft Preparation, Writing – Review  Editing;  : Author roles: Choi SH Chung SS Conceptualization, Data Curation, Resources, Supervision, Writing – Original Draft Preparation;  : Conceptualization, Data Curation, Park KS Resources, Supervision, Writing – Review  Editing  No competing interests were disclosed. Competing interests:  This work was supported by National Research Foundation Grant by Ministry of Science and ICT, Republic of Korea Grant information: (NRF-2016R1A2B3010373). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.  © 2018 Choi SH  . This is an open access article distributed under the terms of the  , which Copyright: et al Creative Commons Attribution Licence permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.  Choi SH, Chung SS and Park KS.  How to cite this article: Re-highlighting the action of PPARγ in treating metabolic diseases [version 1;  F1000Research 2018,  (F1000 Faculty Rev):1127 ( ) peer review: 2 approved] 7 https://doi.org/10.12688/f1000research.14136.1  24 Jul 2018,  (F1000 Faculty Rev):1127 ( )  First published: 7 https://doi.org/10.12688/f1000research.14136.1 Page 2 of 9 F1000Research 2018, 7(F1000 Faculty Rev):1127 Last updated: 17 JUL 2019
  • 19. Introduction Insulin resistance is the key pathophysiologic abnormality of many metabolic diseases such as type 2 diabetes mellitus, obesity, dyslipidemia, and cardiovascular diseases1 . Therefore, reducing insulin resistance is the most important strategy for improving metabolic deterioration. Thiazolidinediones (TZDs), peroxisome proliferator-activated receptor γ (PPARγ) agonists, have shown many beneficial effects not only by enhancing insulin sensitiv- ity but also by demonstrating anti-inflammatory and antioxidant properties, whose actions are related to anti-atherosclerosis2,3 . Thus, TZDs were considered a magic bullet for the treatment of type 2 diabetes and atherosclerosis. Indeed, TZDs dem- onstrated a preventive role for recurrent ischemic stroke in several clinical trials4 and for restenosis after percutaneous coronary intervention (PCI)5–7 . However, TZDs increased the risk of peripheral edema, bone loss, and congestive heart failure8–10 . A meta-analysis of clinical trials showed that rosiglitazone sig- nificantly increased the risk of myocardial infarction11 . Although later studies revealed that rosiglitazone did not increase the risk of heart attack and the US Food and Drug Administration (FDA) removed the warning labels from rosiglitazone-containing drugs regarding the issue of increasing heart attack in 2013, rosiglitazone’s cardiovascular safety issue alongside the above-mentioned adverse effects still lead to many physicians hesitating to prescribe TZDs in their clinical practice. Never- theless, PPARγ is still one of the most important targets for the treatment of insulin resistance and type 2 diabetes, and novel strategies to modulate PPARγ activity to enhance its beneficial effects and reduce unwanted adverse effects are strongly anticipated. Recent studies showed that post-translational modi- fication (PTM) of PPARγ regulates PPARγ activity or stability and may be a novel way to optimize PPARγ activity with reduced adverse effects. In addition, selective PPARγ modulators (sPPARγMs), dual or pan PPAR agonists, have been developed and tested for their metabolic effects in animal studies and in some clinical trials. PPARγ, a therapeutic target for insulin resistance (Figure 1) PPARγ is a master regulator of adipocyte differentiation. It is also involved in glucose homeostasis and insulin sensitivity. The expression of PPARγ is most abundant in adipose tissue12 . Evidence has shown that the primary target of TZDs is adipose tissue, where it increases the expression of Glut4 and CAP13 , and an animal model lacking PPARγ in adipose tissue had a signifi- cantly lower response to TZDs14,15 . TZDs inhibit the expression of TNF-α, IL-6, and resistin in adipose tissue, which promote insulin resistance and chronic inflammation16,17 , while TZDs increased the production of adiponectin and fibroblast growth factor 21 (FGF21), which enhance fatty acid oxidation and insulin sensitivity18,19 . TZDs increase lipogenesis by aP2, LPL, CD36, fatty acid transport protein, PEPCK, and the glyc- erol transporter aquaporin 72 and make adipose tissue store more lipid, while TZDs remove lipid accumulation in other tissues such as muscle and liver20 . From these studies, it seems that improvement of insulin sen- sitivity in liver and muscle might be secondary to the effects of TZDs in adipose tissue. However, there is also evidence showing that TZDs have an insulin-sensitizing effect on other peripheral organs. It has been demonstrated that ablation of liver PPARγ in mice reduced hepatic steatosis but worsened hyperlipidemia, triglyceride clearance, and muscle insulin resistance21 . The expression of PPARγ in skeletal muscle is relatively low compared to adipose tissue, and the physiological significance of PPARγ in skeletal muscle has been shown to work indirectly in previous studies22 . However, selective activation of PPARγ in skeletal muscle showed significant protection from high-fat diet-induced insulin resistance and associated changes in muscle phenotype, such as decreasing the quantity of lipid in myocytes and increasing the number of oxidative muscle fiber types23 . It suggests that the activation of PPARγ can act directly on muscle tissue to improve insulin sensitivity. Macrophage PPARγ is also implicated in anti-inflammation and lipid metabolism24 , and mice lacking macrophage PPARγ are more prone to whole-body insulin resistance25,26 . PPARγ agonists and their effects on the vascular system: friend or foe? PPARγ is expressed in the endothelium and vascular smooth muscle in the blood vessel wall27,28 . Despite controversial car- diovascular effects of TZDs in humans, most experimental studies showed beneficial effects on vascular systems. TZDs inhibit the proliferation and migration of vascular smooth muscle cells (VSMCs), with potential favorable effects on atherosclerosis29,30 . Smooth muscle-specific dominant-negative PPARγ transgenic mice showed a loss of nitric oxide responsive- ness and high contractility31 , which resulted in systolic hyper- tension. In humans, dominant-negative mutations of PPARγ are associated with early hypertension and insulin resistance32 . Activation of PPARγ inhibits CCAAT/enhancer-binding protein-δ (C/EBPδ), which is a well-known mediator of the proinflammatory response in vascular cells33 . TZDs also reduce activation and inflammation in endothelial cells by suppressing the expression of inflammation-associated genes34–37 . On the other hand, TZDs induce vascular endothe- lial growth factor (VEGF) in endothelial cells and increase endothelial cell proliferation and migration by the Akt-dependent pathway38–40 . In recent data, rosiglitazone significantly increased endothelial cell migration and vascular leakage in an animal study with increased VEGF expression and suppressed tight junction proteins, which caused instability of the endothelial membrane41 . This result could be related to vascular permeabil- ity, peripheral edema, and congestive heart failure associated with the use of TZDs, contrary to their beneficial effect on vascular cells. We still need more concrete evidence to understand the role of TZDs in the whole vascular system under various conditions. Regulation of PPARγ by PTMs to reduce the side effects of TZDs The PTM of PPARγ involves several pathways, including phosphorylation, SUMOylation, ubiquitination, β-O-linked N-acetylglucosamine modification (O-GlcNAcylation), and acetylation. These PTMs are known to regulate both PPARγ expression and its transcriptional activity42 and have been recently suggested as a good modality for reducing the side effects of PPARγ activation by TZDs43 (Figure 2). Page 3 of 9 F1000Research 2018, 7(F1000 Faculty Rev):1127 Last updated: 17 JUL 2019
  • 20. Phosphorylation Phosphorylation at serine 112 (S112) in the N-terminal AF-1 domain was first identified, and various studies revealed that net results of PPARγ phosphorylation may inhibit or stimulate its transcriptional activity depending on the cellular contexts and kinases involved44–48 . Phosphorylation at S273 in the ligand- binding domain is mediated by cyclin-dependent kinase 5 (Cdk5), which is activated by pro-inflammatory stimuli and free fatty acids49 . S273 phosphorylation affects the expression of insulin-sensitizing adipokines such as adiponectin and adipsin but not those affecting adipogenesis. PPARγ partial agonist MRL24 specifically blocks the phosphorylation of PPARγ at S273 and has higher anti-diabetic activity and fewer side effects than does rosiglitazone49 . SR1664 and similar non-agonist PPARγ ligands were also developed for blocking cdk5-mediated phosphorylation and showed improved insulin sensitivity in high-fat diet-fed mice without causing side effects such as fluid retention and weight gain43,50 . More recently, it has been reported that phosphorylation at S273 is also facilitated by MEK/ERK, and inhibition of MEK and ERK improves insu- lin resistance, suggesting that MEK and ERK inhibitors can be therapeutic targets for diabetes through the modulation of PPARγ function51 . SUMOylation Small ubiquitin-like modifier (SUMO) modification is a revers- ible process and may affect protein stability, transcriptional activity, and protein–protein interaction. PPARγ is known Figure 1. Effect of PPARγ activation on various tissues. ACSL1, acyl-CoA synthetase long chain family member 1; CD36, cluster of differentiation 36; CPT1b, carnitine palmitoyltransferase 1B; FA, fatty acid; FFA, free fatty acid; FGF, fibroblast growth factor; GK, glucokinase; GLUT, glucose transporter; GlyK, glycerol kinase; IRS2, insulin receptor substrate 1; LPL, lipoprotein lipase; NF-κB, nuclear factor kappa-light- chain-enhancer of activated B cells; MCP-1, monocyte chemoattractant protein 1; MMP-9, matrix metalloproteinase 9; NO, nitric oxide; PAI-1, plasminogen activator inhibitor type 1; PCK2, peroxisome proliferator-activated receptor gamma 2 binding site; PEPCK, phosphoenolpyruvate carboxykinase; PI3K, phosphoinositide 3-kinase; PPARγ, peroxisome proliferator-activated receptor γ; PPRE, peroxisome proliferator-activated receptor response element; RXR, retinoid X receptor; STAT6, signal transducer and activator of transcription 6; TG, triglyceride; TNF-α, tumor necrosis factor α; TZD, thiazolidinedione; VCAM-1, vascular cell adhesion molecule-1; VEGF, vascular endothelial growth factor. Page 4 of 9 F1000Research 2018, 7(F1000 Faculty Rev):1127 Last updated: 17 JUL 2019
  • 21. Figure 2. Regulation of PPARγ by post translational modification. Ac, acetyl; ACSL1, acyl-CoA synthetase long chain family member 1; AF1, activation function 1; AF2, activation function 2; CD36, cluster of differentiation 36; Cdk, cyclin-dependent kinase; CPT1b, carnitine palmitoyltransferase 1B; DBD, DNA-binding domain; FABP3, fatty-acid-binding protein 3, muscle and heart; FAO, fatty acid oxidation; FGF, fibroblast growth factor; GlcNAC, N-acetylglucosamine; LBD, ligand-binding domain; Lys, lysine; MAPK, mitogen-activated protein kinase; P, phosphate; PPARγ, peroxisome proliferator-activated receptor γ; PRDM16, PR domain containing 16; SENP2, small ubiquitin-like modifier-specific protease 2; Ser, serine; SIRT1, sirtuin 1; SUMO, small ubiquitin-like modifier; Thr, threonine; Ub, ubiquitin; WAT, white adipose tissue. as a target of SUMOylation. Lysine 107 (K107) of PPARγ2 is the major SUMOylation site, and deSUMOylation of this site increases the transcriptional activity of PPARγ52 . The K107R mutant form of PPARγ stimulates adipogenesis and sup- presses neointimal formation after balloon injury more effec- tively than does the PPARγ wild-type form53,54 . SUMOylation at K107 of PPARγ may be linked to S112 phosphorylation53 . PPARγ SUMOylation at K107 is markedly increased in FGF21-knockout mice, suggesting that FGF21 regulates PPARγ SUMOylation by an unknown mechanism19 . SUMOylation of PPARγ at K395 (K365 of PPARγ1) is stimulated by PPARγ agonists, and this modification inhibits the transcription of inflam- matory response genes, such as iNOS, through recruiting transcrip- tional repressors to the NFkB complex in macrophages55 . SUMO-specific protease 2 (SENP2) is the major deSUMOylation enzyme of PPARγ56 . Overexpression of SENP2 in C2C12 cells effectively induces PPARγ target genes such as Fabp3 and Cd36 but not Adrp; thus, SENP2 can induce the expression of PPARγ target genes in a selective manner56 . SENP2 deSUMOylates PPARγ and PPARδ and activates genes involved in fatty acid oxidation such as Cpt1b and Acsl1, which results in an increase of fatty acid oxidation in muscle. Interestingly, palmitate increases SENP2 expression via the TLR4-MyD88-NFkB pathway. These results suggest that SENP2 is an important regulator of fatty acid metabolism in skeletal muscle57 . Ubiquitination Ubiquitination is the covalent attachment of ubiquitin, a 76- amino-acid peptide, to lysine residues in the substrate protein. PPARγ has a short half-life and is degraded by the polyubiquitin- proteasome pathway58 . Inhibition of proteasome activity by proteasome inhibitors increases PPARγ stability, suggesting ubiquitin modification of PPARγ is an important determinant of PPARγ activity58 . Several ubiquitin ligases, such as FBOX9 and Cul4B, and an ubiquitin-specific protease (HAUSP) targeting PPARγ have been identified, and an increase in PPARγ stability generally promotes PPARγ activity and adipogenesis59–61 . Inter- estingly, PPARγ agonists, TZDs, stimulate the ubiquitination of PPARγ, which can be mediated by an ubiquitin ligase, Siah158,62 . Therefore, PPARγ ubiquitination may be differently regulated by several ubiquitin E3 ligases or proteases upon various conditions. O-GlcNAcylation O-GlcNAcylation is the post-translational cycling of a single β-O-linked N-acetylglucosamine (O-GlcNAc) on the hydroxyl groups of serine or threonine residues of target proteins. A major Page 5 of 9 F1000Research 2018, 7(F1000 Faculty Rev):1127 Last updated: 17 JUL 2019