Estrogen and Signaling in The Cells of Immune System: Advances in Neuroimmune Biology January 2012

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/279852548

Estrogen and Signaling in the Cells of Immune System

Article  in  Advances in Neuroimmune Biology · January 2012


DOI: 10.3233/NIB-2012-012039

CITATIONS READS

24 3,813

3 authors:

Deena Khan Cat Cowan


Virginia Polytechnic Institute and State University Virginia Polytechnic Institute and State University
17 PUBLICATIONS   893 CITATIONS    21 PUBLICATIONS   460 CITATIONS   

SEE PROFILE SEE PROFILE

Ansar Ahmed
Virginia Polytechnic Institute and State University
144 PUBLICATIONS   7,858 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Neutrophils in systemic lupus erythematous View project

All content following this page was uploaded by Ansar Ahmed on 07 July 2015.

The user has requested enhancement of the downloaded file.


Advances in Neuroimmune Biology 3 (2012) 73–93 73
DOI 10.3233/NIB-2012-012039
IOS Press

Estrogen and Signaling in the Cells


of Immune System

PY
Deena Khan, Catharine Cowan and S. Ansar Ahmed∗
Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine,
Virginia Tech, Blacksburg, VA, USA
CO
Abstract. Hypothalamus-pituitary-gonadal axis regulation of estrogen, which acts on reproductive tissues, is well established.
However, it is also evident that estrogens physiologically act on not only reproductive tissues but also on a broad range of tissues
such as immune system. It is well documented that estrogen regulates all facets of the immunoregulation thereby affecting
the outcome of autoimmune and inflammatory immune responses. Given the broader role of estrogen in immunobiology, it is
OR

important to understand how estrogens act on the cells of the immune system. Estrogens act in estrogen receptor dependent and/or
independent manner to affect the regulation of cytokines and chemokines. This review focuses on sources and biosynthesis of
estrogens, differential expression of estrogen receptors on the cells of immune system, key signaling pathways and its effect on the
induction of key pro- and anti-inflammatory cytokines. Since estrogen has contrasting effects in female-predominant autoimmune
diseases such as multiple sclerosis (MS) and systemic lupus erythematosus (SLE), we briefly overview the mechanistic role
of estrogen on these chronic diseases. Recent evidence suggests that estrogens also alter microRNAs, which regulate a broad
TH

range of transcription of genes. The review also addresses this and other newly discovered mechanisms of estrogen-induced
immunomodulation in health and disease.

Keywords: Estrogen, receptor, immune, autoimmune, cytokine


AU

SOURCES AND BIOSYNTHESIS OF only women. However, studies in males with either
ESTROGENS defective estrogen synthesis or signaling have clearly
demonstrated that estrogen is also physiologically
There has been a paradigm shift in the under- important in males [2–4]. Estrogens occur in three
standing of biological effects of estrogen in recent major natural forms: estrone (E1), 17␤-estradiol (E2),
years. Estrogens should no longer be merely con- and estriol (E3). In non-pregnant female individuals,
sidered as reproductive hormones since estrogens act 17␤-estradiol is the predominant form of estrogen in
not only on reproductive tissues, but also physio- the reproductive stages from puberty to menopause.
logically act on many non-reproductive tissues. This In postmenopausal women, estrone is more preva-
include: cells of the immune, central nervous, car- lent; whereas during pregnancy, estriol is the primary
diovascular, and skeletal systems, as well as cells estrogen. 17␤-estradiol is generally believed to be the
from liver, skin and kidneys [1]. Thus, a wide-range most potent estrogen synthesized in the human body.
of tissues are natural targets for estrogen action. Although, estrone and estriol were considered to be
Interestingly, it was originally believed that estrogen inactive in the past, it has now been shown that these
exclusively affects females, hence of importance to compounds have tissue specific roles and are high affin-
ity ligands for estrogen receptors [5–7].
∗ Correspondence to: Dr. Ansar Ahmed, Head; Department of Other sources of exposure to estrogens include:
Biomedical Sciences and Pathobiology, Center of Molecular
Medicine and Infectious Diseases, 1410 Prices Ford Road, Blacks-
(i) intake of synthetic pharmaceuticals (oral con-
burg. VA 24060-0442 USA. Tel.: +1 540 231 4652; E-mail: traceptives and hormone replacement); (ii) unin-
ansrahmd@vt.edu. tentional exposure to environmental contaminants

ISSN 1878-948X/12/$27.50 © 2012 – IOS Press and the authors. All rights reserved
74 D. Khan et al. / Estrogen and Signaling in the Cells

(xenoestrogens; e.g., pesticides and herbicides) and immunoglobulin class, Th1/Th2 balance and IL-4
certain commercial products (plastics and cosmetics); levels in allergies [16]. Another natural source of estro-
(iii) consumption of plant-derived estrogens (phy- gens come from fungi and is called mycoestrogens.
toestrogens), and (iv) exposure to fungus-derived A common mycoestrogen found in animal feed is zear-
estrogens (mycoestrogens). These structurally diverse alenone produced by Fusarium fungi and is recently
compounds act on estrogen receptors to induce estro- been shown to modulate immune response in pigs [17].
genic activity. It is beyond the focus of this review to discuss the bio-
17␣ ethinyl estradiol is a common synthetic estrogen logical effects of xenoestrogens, phytoestrogens and

PY
which is prescribed either as oral contraceptive to pre- mycoestrogens. Therefore, this review is restricted to
menopausal women or as estrogen replacement therapy key findings of natural endogenous estrogens (17␤-
to postmenopausal women [8]. Some of pharmaceuti- estradiol) on the cells of the immune system.
cal estrogens mimic estrogen action in some tissues Estrogens are primarily synthesized by both gonadal
but behave as antagonist in certain other tissues and and extragonadal tissues. The synthesis and release of
CO
are referred to as Selective Estrogen Receptor Mod- estrogens are centrally regulated by the hypothalamus-
ulators (SERMs) [9]. Therefore, these SERMs have pituitary-gonadal axis. Hypothalamus in brain secretes
been advantageously used clinically to achieve desired gonadotrophin releasing hormone (GnRH), which then
effects in certain tissues. acts on pituitary to release luteinizing hormone (LH)
Humans and animals are unintentionally exposed and Follicular stimulating hormone (FSH). These two
to xenoestrogens. Although these compounds struc- hormones act on gonads to release estrogen in a cyclic
turally differ with estrogen, yet they are estrogenic manner during menstrual cycle. Estrogen in turn con-
OR

and have significant effect on the ecology and human trols the release of GnRH from hypothalamus in a
health. Some of the xenoestrogens include: synthetic negative feedback loop. This Hypothalamus-pituitary-
substances and environmental contaminants found in gonadal axis regulates reproduction by controlling
detergents and surfactants (polycyclic aromatic hydro- uterine and ovarian cycles and for proper female devel-
carbons; octylphenols), plastics (phthalates, bisphe- opment [18]. In premenopausal female, estrogens are
TH

nol A), pesticides (methoxychlor, dichlorodiphenyl- produced primarily in the follicles of ovaries, cor-
trichloroethane or DDT, hexachlorobenzene, and pus luteum and in the placenta of pregnant women.
dieldrin), and industrial chemicals (polychlorinated On the other hand, in postmenopausal women and
biphenyls or PCBs, 2,3,7,8-tetrachlorodibenzo-p- in men, estrogens are synthesized by extragonadal
dioxin or TCDD) [10]. These environmental contam- tissues. These include: cells in the liver and brain, mes-
AU

inants are of importance since they interfere with enchymal cells of adipose and breast tissue, osteoblasts
physiological estrogen signaling and thus are com- and chondrocytes of bone, adrenal glands, vascular
monly termed as endocrine disruptors. In addition, they endothelium, and aortic smooth muscle cells [19].
are also known to alter the synthesis of estrogen and The estrogen synthesized in these extragonadal sites
inhibit the inactivation of estrogen by sulphation thus act locally in a paracrine or intracrine fashion and
resulting in hormonal imbalance [11]. Even though only occasionally escapes into circulation [20]. The
these compounds are less potent than natural estrogens, estrogen levels also fluctuate in female mice between
they tend to accumulate in body fat and are capable to 20–35 pg/ml in diestrus; 70–200 pg/ml during estrus,
altering the endocrine function to affect reproductive and 5,000–10,000 pg/ml during pregnancy [21–23]. In
capacity, immune and other biological functions [12]. women, estrogen levels vary through different physio-
Studies in various animals have revealed that endocrine logical stages such as menstrual cycle and pregnancy,
disruptors alter the cytokines, immunoglobulin and T and also by age. During menstrual cycle, estrogen lev-
helper cell (Th) profile (reviewed in detail in [13, 14]). els are highest during ovulation induction 1000 pg/ml;
Some plant products also possess estrogenic activ- peak (late follicular) around 200–500 pg/ml [24]. In
ity and are termed phytoestrogens [15]. Leguminous humans, estrogen levels markedly increase to around
plants such as soybeans, whole grain cereals, and 16,000–30,000 pg/ml during pregnancy [24, 25]. The
some seeds have high amount of phytoestrogens. levels of estrogen decrease significantly in post-
Soybeans have high levels of isoflavones (e.g., genis- menopausal women to 5–20 pg/ml [19, 24]. Estrogen
tein), which are estrogenic and have anti-inflammatory replacement therapy of post-menopausal women clin-
effects that are associated with alteration of chemotac- ically restores estrogen levels to around 100 pg/ml by
tic factors, adhesion molecules, and reactive oxygen giving hormone replacement therapy [24]. In men the
radicals. Phytoestrognes are also known to modulate estrogen level is around 30–35 pg/ml [19, 26].
D. Khan et al. / Estrogen and Signaling in the Cells 75

Cholesterol (C27) acts as a precursor of many receptor (NR) family of transcription factors encoded
steroids found in human/animal body. In theca interna by chromosomes 6 and 14, respectively. ERs are
cells of ovary, after hydroxylation and side chain ligand-activated transcription factor that regulate a
cleavage of cholesterol by delta-5 and delta-4 path- broad range of estrogen-responsive genes. Nearly six
way leads to the generation of a key intermediary decades ago, an estrogen binding protein was identified
androstenedione. Androstenedione then crosses basal by Elwood Jensen, now known as estrogen recep-
membrane into granulosa cells where it is converted tor alpha ER␣ [30]. Interestingly, ER␤ was identified
to testosterone, which in turn undergoes conversion to much later (1996) [31]. Generation of ER␣ knockout

PY
estradiol by an enzyme called cytochrome P450 aro- and ER␤ knockout mice have been invaluable in clas-
matase (P450AROM ). Alternatively, androstenedione is sifying physiological role of these receptors in various
aromatized to estrone, which is subsequently converted tissues [32]. ERs are evolutionarily conserved and have
to estradiol by 17␤-hydroxysteroid dehydrogenase. In structural and functional five distinct domains known:
men, a small amount of estradiol is produced by pre- the DNA binding domain (DBD; domain C), the lig-
CO
cursor hormone especially from testosterone in testes. and binding domain (LBD; E/F domain), the hinge
In post menopausal women and in men, aromatase domain (D), and two transcriptional activation function
converts androstenedione to estrogen in extragonadal domains AF-1 (in A/B domain) and AF-2 (in F domain)
tissue [19]. (Fig. 1). The NH2-terminal A/B domain is the only
In the plasma, estrogen and other sex hormone are variable domain in both sequence and length. The A/B
bound to steroid hormone binding globulin (SHBG). domain has sites for post-transcriptional modifications
Effectively, SHBG regulates the bioavailability of free by kinases of growth factor pathways, which stimulate
OR

estrogen levels in the plasma [27]. SHBG plays an AF-1 constitutively in a hormone independent manner.
important role in estrogen signal transduction since The DNA binding domain or C domain is central and
any change in the levels of SHBG will alter the lev- most conserved domain and is involved in recognition
els of free estrogen. Free estrogen is now capable and binding to the DNA. The hinge domain is essential
of acting on target cells to induce biological activ- for nuclear localization and for its post-translational
TH

ity. Recent studies have shown that SHBG binds to its modifications by acetylation, lipophilic moieties, and
specific receptor (RSHBG ) on surface of various cells ubiquitination. The C-terminal is essential for lig-
[28]. This SHBG- RSHBG complex can be activated and binding, ER homo or hetero dimerization, and
by sex hormone to induce cell signaling, thereby pro- interaction with co-regulatory proteins through ligand
viding additional molecular mechanism by which sex dependent AF-2 region. The AF domains are required
AU

hormone can affect cellular functions [29]. for transcriptional activation and aid in the recruit-
ment of co-regulatory proteins for gene expression.
ESTROGEN RECEPTORS IN THE CELLS Although both ERs have similar affinity for estrogen
OF THE IMMUNE SYSTEM and bind to the same DNA response element, they
have distinct, non-redundant roles and in some tis-
Estrogen signaling is an intricate balance between sues entirely opposing roles. Estrogen receptors exert
two distinct receptors ER␣ (NR3A1) and ER␤ their effects either through genomic or non-genomic
(NR3A2) and their splice variants from nuclear signaling pathways depending on the tissue.

Fig. 1. Structural description of domain structure and percent sequence homology of human ER␣ and ER␤. Full length human ER␣ comprise of
595 amino acids and has 2 shorter isoforms, while ER␤ has 530 amino acid and 4 other splice variants. ERs usually bind to Estrogen Response
Element (ERE), which is a 5 base pair palindrome (GGTCA) with a 3 base pair spacer (nnn).
76 D. Khan et al. / Estrogen and Signaling in the Cells

Different splice variants or isoform of both ER␣ and expressions of ERs in cells of the immune system have
ER␤ have been identified in different cell lines [33, 34]. an impact on the net effect of estrogen on immune
In human, full length ER␣ is 595 amino acids long [35] responses. To date, ERs have been found in nearly all
and has 2 shorter isoforms (hER␣-46 and hER␣-36), cells of the immune system including cells of the innate
which lack AF-1 portion of NH2 terminal [33, 34]. and acquired immunity. This suggests that the immune
ER␤ is smaller than ER␣ and is 530 amino acid long system is a natural target for estrogen. Hematopoi-
[36] and has 4 other isoforms either because of dele- etic progenitors have differentially expressed ERs
tion of 5th exon or due to different COOH-terminal depending on the developmental stage, with reduced

PY
regions [37–40]. Even though ER␤ is smaller than expression during fetus and increased expression after
ER␣, they have similarities in DNA binding domain birth [50]. ER␣ is found in most of the cells of immune
and ligand binding domain. Similarly, mouse and rats system, while ER␤ appears to be somewhat restricted.
ERs have sequence homology and different isoforms ER␣ is preferentially expressed in thymus stromal
with altered ligand binding and other functionalities cells, thymocytes, hematopoietic cells, and bone mar-
CO
(reviewed in detail in [41]). The binding site of estrogen row [51–54], and murine splenic DC and peritoneal
to its receptor is generous in size, therefore, a wide vari- macrophages [55]. ER␤ is expressed in thymus and
ety of compounds with diverse structures are capable spleen of human mid-gestational fetus [49], and lym-
of binding to the estrogen receptors [42]. phocytes in human lymph nodes, rat thymocyte and
The distribution of ERs in various cells and tis- stromal cells [54], and in murine bone marrow and
sues are also critical in determining the overall affect thymus [56–58]. Splenic B cells express both ER␣
of ligand-dependent signaling. The distribution of and ER␤ [59]. ER␣ levels are higher than ER␤ in
OR

ER␣ and ER␤ is overlapping in some tissues and CD4+ ; low levels of both ER␣ and ER␤ are found in
highly specific in some. Analysis and knowledge of CD8+ cells, whereas B cells have more ER␤ than ER␣
tissue distribution of ERs is critical for designing [60, 61]. In human blood T lymphocytes, B lympho-
potential ER-targeted therapies. Different molecular cytes and NK cells ER␣46 isoform is the predominant
mechanisms play important roles in modulating the ER [62]. Overall, the presence of ERs in immune cells
TH

ER-mediated response. This includes different ligand indicates that the immune system is a target for estro-
affinity, signaling events, transactivation, interaction gen action. Different tissues have varying levels of ER␣
with specific co-factor interaction, homo- or hetero- and ER␤ which in turn affects the overall outcome of
dimerization, splice variant ER isoforms. Numerous immune response.
reports have been published regarding differential ER-mediated signaling in the cells of the immune
AU

expression of ER␣ and ER␤, which is determined by a system induces a variety of cytokines including
variety of techniques including polymerase chain reac- IFN␥, IL-17, IL-6 among others. Interestingly, activa-
tion, Western blotting or intracellular flow cytometry. tion of CD4+ T cells with proinflammatory cytokines
ER␣ is expressed highly in epididymis, bone, breast, increases ER expression. For example, IFN␥ and IFN␣
uterus, testis (leydig cells), ovary (theca cells), kid- treatment can upregulate ER␣ expression in mouse
ney, adrenal, prostate gland, bladder, liver, and thymus, splenocytes or cell lines by activating STAT1 pathway
white adipose tissue and various regions of the brains. [63]. In myasthenia gravis patients, there is upregu-
ER␤ is expressed predominantly in colon, epithelium lation of ER␣ in thymocytes indicating that there is
of prostate gland, testis, granulosa cells of ovary, bone modulation of ER expression depending on lympho-
marrow, salivary gland, vascular endothelium, lung, cyte subset, localization and disease condition [61].
bladder and brain [43–45]. ER␤ is predominant in ovar- The above studies demonstrate cross talk between
ian granulosa cells of follicles, while ER␣ is mostly cytokines and ERs expression.
present in theca and interstitial cells [31, 46, 47]. The To define the specific role of ER␣ and ER␤ in
expression of ERs is also dependent on the differenti- immune modulation, mice deficient in either ER␣ or
ation and developmental stage of the tissues such as, ER␤ and selective agonist or antagonist have been
e.g., in uterus and pituitary gland; ER␤ is expressed in used. ER␣ is important in thymus and spleen devel-
immature whereas ER␣ predominates in fully mature opment since, ER␣ knockout mice have hypoplasia
tissue [48, 49]. of the thymuses and spleen [58, 64], increase in num-
Since estrogen is known to modulate immune ber of immature CD4+ CD8+ thymocyte and decreased
system, extensive studies have been performed to CD4+ CD8− cells and systemic autoimmune diseases
define the role of estrogen-mediated signaling in with immune-complex mediated glomerulonephritis,
physiological and pathological conditions. Differential proteinuria, infiltration of B cells in kidney, damage
D. Khan et al. / Estrogen and Signaling in the Cells 77

of tubular cells and presence of serum anti-DNA anti- ER␤ either inhibits the action of ER␣ regulated gene
bodies [58, 65]. Majority of the studies indicate that or reduces ER␣ protein level. For instance, ER␤ pro-
ER␣ plays a prominent role in immunmodulation when tects against ER␣-induced hyper proliferation thereby
compared with ER␤ [66]. ER␤ is required in prolif- counteracting the biological activity of ER␣. ER␤ also
eration of progenitor cells in the bone marrow. Old alters the recruitment of c-Fos and c-Jun to AP-1
mice deficient in ER␤ have been shown to develop regulated promoters antagonizing ER␣-mediated reg-
myeloproliferative disease similar to chronic myeloid ulation of AP-1 dependent transcription. In addition,
leukemia with increased number of B cells in bone ER␤ also alters the expression of c-Fos and decreases

PY
marrow and spleen; increase in number of Gr-1hi/Mac- ER␣ expression by proteolytic degradation [69]. The
1hi-positive granulocytes and B cells in blood and relative expression of ERs in a cell produces a distinct
some of the mice even develop severe lymphoprolif- response to ER selective or partial agonist/antagonists.
erative phenotype [67]. ER␤ has been shown to be One possible reason for diversity of responsiveness
required for estrogen-mediated thymic cortex atrophy could be due to differences in activation of AF-1
CO
and shift of thymocyte phenotype in female mice [58]. domain in ER␣ when compared with ER␤ [70].
ER␤ deficient mice have hyperplasia of bone marrow
[67]. On the other hand, in ER␣ and ER␤ knockout Estrogen-receptor mediated cell signaling
mice significant reduction of B lymphopoiesis was
seen upon estrogen treatment. This reduction in lym- Numerous studies have confirmed that multi-
phopoiesis is mediated either through ER␣ or both ple mechanistically distinct molecular pathways are
receptors. Furthermore, estrogen treatment of ER␤ involved in the signaling through ERs. These path-
OR

knockout mice and wild type littermates resulted in sig- ways are broadly classified as ligand dependent or
nificant increase in immunoglobulin secreting B cells. ligand independent (Fig. 2). Binding of either endoge-
This indicates that although both ERs are required for nous hormone or synthetic compounds with estrogenic
complete downregulation of B lymphopoiesis, only properties to the ligand-binding domain of ERs in
ER␣ is required to up-regulate immunoglobulin pro- the cytosol triggers the ligand dependent pathway.
TH

duction in bone marrow and spleen [68]. Following this initial step, the ER subtypes undergo
It has been shown that ER␣ and ER␤ have oppos- different biological processes such as conformational
ing role in transcription of estrogen-responsive genes. changes, receptor dimerization (either homodimers -
AU

LIGAND DEPENDENT SIGNALING LIGAND INDEPENDENT SIGNALING

EGF
Growth Factor Receptor activation
ESTROGEN
Membrane-anchored ER

EGFR
ERα/β Activation of MAPK and
GFR downstream kinases
other pathways
phophorylate ERs, leading to
dimerization
Ligand binding
Receptor Dimerization C ERα/β

“Non-genomic” signaling
B D
A

“Classical” signaling “Tethered” signaling

Fig. 2. Pictorial illustration of key estrogen signaling pathways. Estrogen signals via membrane or intracellular ER␣/␤ in either ligand dependent
or ligand independent pathway. A) In the ligand dependent pathway, estrogen binds to its receptor, which then dimerizes and translocates to
nucleus and binds to estrogen response element (ERE) and regulates estrogen responsive genes in “classical” pathway. B) In “tethered” pathway,
activated ERs interact with other signaling molecules and transcription factors and bind to non-ERE sites and regulate gene expression. C)
In another ligand dependent pathway, upon ligand receptor interaction, there is rapid physiological “non-genomic” signaling by activation
of MAPK or other cytoplasmic signaling pathways. D) In the ligand independent pathway, ERs are activated downstream of other activated
signaling cascades such as growth factor receptor (GFR) and modulate gene expression in absence of estrogen in both ERE dependent genomic
fashion or by interacting with other transcription factors.
78 D. Khan et al. / Estrogen and Signaling in the Cells

ER␣: ER␣ or ER␤: ER␤ or heterodimers ER␣: ER␤), regulated by NF-␬B (IL-1 alpha, IL-1 beta, IL-10,
and then translocate to nuclei and bind to specific and IFN-gamma) [83]. Interestingly, we have recently
estrogen-responsive element (ERE) in the regulatory demonstrated that estrogen promotes truncation of
regions of estrogen responsive genes. This is referred p65 by serine protease [84]. While estrogen inhib-
to as the “classical” signaling pathway. The final out- ited full-length p65 translocation, shorter form of p65
come is dependent on the interaction with effectors was found in nuclei of estrogen-treated cells, when
such as coregulatory proteins, chromatin remodelers, detected in Western blotting by using antibody spe-
co-activators, co-repressors and other active signal- cific for N-terminal p65. Inhibition of serine protease

PY
ing pathways in the cells [9, 71–73]. Different ligand activity restored the full-sized length of p65 in nuclear
binding induces unique ER conformational changes extracts of cells from estrogen-treated mice compara-
that recruit specific co-regulatory proteins, which inter- ble to placebo-treated mice [84]. In lymphoid cells,
act with AF-1 and AF-2 at N or C-terminal ends NF-␬B regulates TNF secretion, and estrogen has also
of ERs, respectively [74, 75]. Although, the general been demonstrated to inhibit TNF secretion in T cells,
CO
belief is that ERs bind to the consensus ERE con- macrophages, and dendritic cells [85]. However an ER-
sisting of a 5-base pair palindrome with a 3-base pair mediated mechanism for this inhibition has not been
spacer: GGTCAnnnTGACC, some reports have sug- demonstrated.
gested deviation in the consensus sequence of ERE Third ligand dependent pathway is also known as
[76] or binding to non–estrogen response elements by “non-genomic pathway” termed due to its rapid phys-
tethering to other transcription factors [73]. iological effect within seconds to minutes without
The process whereby ERs interact with other tran- involving gene regulation. In this process, there is
OR

scription factors is referred as transcription factor cross ligand interaction with ERs localized to the plasma
talk or the “tethered” pathway. It is noteworthy that ERs membrane via palmitoylation on cysteine447 [86].
can affect ER responsive genes even in the absence Recently, a genetically and structurally unrelated mem-
of binding to ERE. This is accomplished by binding brane receptor, GPR30 (an orphan G-protein coupled
of ERs to different transcription factors and thus not receptor 30) has been demonstrated to bind to estro-
TH

interacting with ERE directly. There are studies that gen and mediate downstream non-genomic signaling.
demonstrate ligand activated ERs interact with var- However, it has been shown that estrogen binding to
ious transcription factor complexes such as Fos/Jun GRP30 induces expression of the ER␣ variant ER␣36,
AP-1 (activating protein-1) [77], SP-1 (stimulating which then mediates downstream non-genomic signal-
protein-1; GC-rich motifs), CREB, Runx1, NF-␬B, ing; demonstrating that the GPR30 pathway still relies
AU

STAT5 and p53 [78–81]. While most of these asso- on ER expression and function [87]. The role of GPR30
ciations have been demonstrated in epithelial cancer in immune cells remains unclear, as knockout mouse
cells, many of these transcription factors play well- models demonstrate thymic atrophy; however GPR30
characterized roles in immune cell responses as well. expression could not be detected in thymocytes [88].
For example, estrogen is known to inhibit NF-␬B- This implies that GRP30 may have indirect effects on
driven transcription by preventing p65 translocation the thymus.
to the nucleus [82]. It has been demonstrated that in In non-genomic pathway, after activation of ERs,
vitro estrogen (1 nm) treatment of Raw 264.7 cells cytoplasmic signaling pathways are stimulated, includ-
decreases LPS induced DNA binding and transcrip- ing protein kinase C (PKC), mitogen-activated protein
tional activity of p65 by blocking nuclear translocation kinases (MAPK), phosphoinositide-3 kinase (PI3K),
and also activates phosphatidylinositol 3-kinase with- as well as increased release of intracellular Ca and
out activating or modifying Ikappa-Balpha degradation activation of calcium-calmodulin-dependent kinases.
or MAPK.This activity is mediated through ER␣ and Activation of non-genomic pathway likely depends on
not ER␤ [82]. expression levels of the different ERs, splice variants
Studies from our laboratory have added new and post-translational modifications [73].
thoughts to estrogen-mediated NF-␬B signaling path- Some studies on immune cells have demonstrated
way that promotes inflammation. In vivo estrogen the existence of the non-genomic actions of estro-
treatment of mice, inhibited nuclear localization of gen and ERs interactions. Membrane bound ER␣ has
NF-␬B p65, c-Rel, and RelB, but not p50. However, been detected in peripheral blood mononuclear cells
estrogen treatment increased the activity of NF-␬B as [89]. Estrogen has been shown to rapidly induce ERK
measured by luciferase reporter assay increased bind- (MAPK) phosphorylation in glial cells, indicating at
ing of NF-␬B to DNA, and the levels of cytokines least this non-genomic pathway is conserved between
D. Khan et al. / Estrogen and Signaling in the Cells 79

epithelial and immune cells, and implying others likely diestrus levels) may stimulate secretion while high
are as well [90, 91]. In another example, estrogen doses (approximately pregnancy levels) are inhibitory
decreases the activity of the small GTPase Rac1 in [85]. The converse can also be true. These variability
monocytes, without altering Rac1 expression. Rac1 likely results from the multiple signaling pathways and
regulates actin polymerization in the cytoplasm and cross-talk that can occur upon ligand binding to ERs,
modulates monocyte adhesion [92]. However it is as well as variability in expression of ER subtypes and
unclear whether this effect is due to direct ER inter- splice variants among immune cells. Following is a
action or via downstream proteins. brief review of selected key pro- and anti- inflamma-

PY
The fourth ER signaling pathway, is known as the tory cytokines secreted from cellular players of innate
ligand-independent pathway. In this pathway, ERs and adaptive immune system that are responsive to
are phosphorylated by activated signaling cascades estrogen treatment, a subject reviewed in detail earlier
[93]. Ligand-bound growth factor receptors activate [85, 89, 96].
downstream kinases, which then phosphorylate ERs
CO
and stimulate both direct ERE-dependent genomic
actions and transcription factor associations. For exam- Cytokines from innate immune cells
ple, HER2 signaling activates the MAP-kinase ERK, The cells of innate immune cells predominantly
which can phosphorylate ER on serine 118, and lead macrophages and dendritic cells (DCs) produce
to increased ER-AF-1 activity [94]. The role of this cytokines which influence the direction of adaptive
pathway in immune cells is perhaps the least charac- immune response. As mentioned earlier, the influence
terized. Whether activation of kinases such as ERK by of estrogen on these cytokines depends on number of
OR

immunologic stimuli leads to similar ER phosphoryla- factors including but not limited to, dose of estrogen,
tion events in immune cells, although likely, needs to method (in vitro or in vivo) and duration of treatment,
be determined. species and cell type studied and the kind of activa-
The signaling mechanism via ERs is dependent tion/stimuli used [85). For example, on exposure to
on number of variables including but not limited estrogen, PMA- activated human monoblastic U937
TH

to ER type, expression and its post-translational cells have increased TNF␣ production [97], while
modification; type and concentration of ligand avail- LPS-activated bone marrow derived macrophages have
ability; tissue type and coregulators present in the decreased TNF␣ production [98]. 17␤-estradiol at
cell. Post-translational modifications of ERs mod- 0.01-0.001 ng/ml increases TNF␣ secretion from LPS-
ulate their signaling ability. While phosphorylation activated rat peritoneal macrophages while both low
AU

triggers downstream events, glycosylation is involved (<0.1 pg/ml) and high dose (>0.1 ng/ml) doses of estra-
in directing ERs to their final intracellular local- diol decreased TNF␣ secretion from the LPS-activated
ization. Acetylation has been shown to enhance rat peritoneal macrophages [99]. In vivo estrogen or
ER-DNA binding activity, hormone sensitivity and estriol treatment also increases TNF expression from
transcriptional activity. Ligand mediated sumoylation peritoneal macrophages, Kupffer cells and in sera
facilitates ERalpha-dependent transcription, nitrosyla- of animals challenged with LPS [100, 101] Treat-
tion impairs their genomic action, DNA binding ability ment of CD11c+ murine spleen DCs with estrogen,
and ubiquitination tags ERs to undergo degradation. upregulated intracellular IL-6 and IL-10, but did not
In addition, myristoylation and palmitoylation affect affect TNF␣ and IL-12 expression [102]. Another
interaction of ERs with membrane proteins, trafficking, recent report has shown that in vivo estrogen or ER␣
as well as signal transduction [95]. select agonist exposure of wildtype and ER␤ knock-
out mice downregulated TNF␣ and IL-6 secretion
Estrogen-mediated regulation of pro-inflammatory from splenocytes and macrophages following trauma-
cytokines hemorrhage [103]. In ovariectomized mice, there is
decreased TNF␣ expression in brain following intrac-
Estrogen treatment is known to markedly regulate erebral injection of LPS. However, addition of estrogen
cytokine synthesis from a number of different cell upregulated TNF␣ levels [104]. These findings suggest
types. Estrogen-regulated cytokine secretion is vari- that both ER␣ and ER␤ are essential for mediating
able, depending on the cell type, ER expression profile, immunoprotective effect by decreasing the production
concentration of ligand, type of ligand and length of of proinflammatory cytokines [103] and the affect of
exposure and in vitro versus in vivo treatment [85, estradiol on cytokine induction is dependent on the
89]. In many cases, low doses of estrogen (roughly dose and route of treatment.
80 D. Khan et al. / Estrogen and Signaling in the Cells

Similarly, IL-1␣ and IL-1␤, are proinflammatory Another important pro-inflammatory cytokine
cytokines that are secreted mainly from macrophage secreted by antigen presenting cells is IL-6, which
or monocyte, and have been shown to be critical in together with TGF␤ is involved in priming naı̈ve CD4+
not only fighting off infections but also is a key player T cells to commit to IL-17 secreting Th17 cell type.
in diverse autoinflammatory diseases [105]. IL-1␤ sig- Increase in estrogen-mediated IL-6 has been shown in
nal through IL-1R of Toll like receptor (TLR) family human peripheral monocytes with and without LPS
activates NF-␬B translocation and expression of mul- treatment [116]. Various reports have demonstrated
tiple pro-inflammatory genes. There are reports that that IL-6 is variably inhibited by estrogen treatment

PY
have shown that estrogen upregulate IL-1␤ induction, in whole blood, PBMCs and whole bone marrow,
possibly by direct binding to IL-1␤ promoter. In vitro but increased from macrophages [115, 117]. Even
constructs demonstrated ER binding directly to IL-1␤ in macrophages, it is chronic in vivo estrogen expo-
promoter in murine macrophages at moderate phys- sure that leads to stimulation, while short-term in
iological estrogen concentrations [106]. Likewise, in vitro exposure causes suppression [112]. However,
CO
peritoneal macrophages from male and female rats, others have shown that short-term estrogen exposure
moderate to low estrogen stimulated secretion of IL-1␤ of immature DCs has been demonstrated to increase
[107]. Exposure of estrogen to LPS-stimulated HL-60 IL-6, IL-8, and MCP-1 secretion. In addition, estro-
promyelocytic leukemia cells also upregulated IL-1␤ gen provides signal migration of mature DCs towards
induction [108]. lymph-node derived CCL19/MIP3beta in migration
Increased IL-1␣ mRNA has been demonstrated in assay, indicating that estrogen regulates DC-mediated
17␤-estradiol-treated rheumatoid fibroblast-like syn- T and B cell responses [118].
OR

ovial cells and from primary synovial cells from


RA patients in ER␣ dependent manner. It has been Cytokines from cells of the adaptive immune
demonstrated that estrogen–mediated dissociation of system
corepressor HDAC2 from ER␣, results in physical The key effector cells of adaptive immunity com-
interaction of ER with Sp1 transcription factor and prise of (i) IFN␥-secreting Th1 cells, which are critical
TH

activation of Sp1 through the GC-rich region within the for immunity against intracellular pathogens; (ii) IL-
IL-1␣ gene promoter [109]. Administration of estrogen 4-secreting Th2 cells essential for immunity against
in ovariectomized rats with acute endoluminal arterial helminth parasite and key player in allergic response;
injury decreases IL-1␣ induction when compared with (iii) IL-17-secreting cells which regulate neutrophilic
untreated ovariectomized rats [110]. inflammation; and (iv) suppressor T regulatory cells
AU

Activation of estrogen-treated splenic lymphoid (Treg), which downregulate immune responses by


cells with LPS increased both IL-1␣ and IL-1␤ via secreting TGF␤ and IL-10.
NF-␬B mediated signaling [83]. Increased IL-1␣, IFN␥ is a prototypic cytokine released by Th1
IL-1␤ and IL-6 and increased astroglial responses cells, but a number of other cell types such as CD8
have been observed in male rats in vivo treated T cells, NK cells, NKT cells, are also know to
with 17␤-estradiol following spinal cord injury [111]. secrete IFN␥. Numerous reports have indicated that
Conversely, short term in vitro estrogen exposure there is gender bias in IFN␥ production. There is
downregulates IL-1 secretion from macrophage by increased IFN␥ secretion in virus or Listeria mex-
activating Akt pathway while chronic administration icana or mycobacteria-stimulated spleen or lymph
enhances IL-1 levels from LPS-activated peritoneal node cultures from female BALB/c, C57BL/6 [119],
macrophages [112]. However, LPS-activated PBMCs NZBxNZW- F1 [120], DBA/2 mice [121] when com-
from postmenopausal women given estrogen for 6 pared to their male counterparts etc. Elevated levels
months had decreased IL-1␤ but not IL-1Ra [113]. of IFN␥ have been demonstrated in Concanavalin A
LPS activation of microglia and astrocytes exposed or antiCD3 antibody activated splenic lymphoid cell
to estrogen or ER agonist decreases IL-1␤ and TNF␣ cultures and purified T cells from in vivo estrogen
induction [114]. Interestingly, IL-6, TNF␣, IL-1Ra, IL- treated mice [122]. A putative ERE has been identi-
1␤, and ratio of IL-1␤ /IL-1Ra were decreased in whole fied in the promoter 5 -prime flanking region of IFN␥
blood cell cultures exposed to varying dose of estrogen gene [123]. In presence of IL-27, estrogen induces
(10−12 – 10−6 mol/l) [115]. These studies demonstrate T-bet, which primes CD4+ cells to differentiate into
that the duration and type of estrogen exposure plays a Th1 cell type and IFN␥-mediated downstream proin-
crucial role in modulating IL-1␣ and IL-1␤ levels and flammatory events [124]. Studies have also demon-
therefore influence the overall immune response. strated that 17␤-estradiol, estrone and estriol treatment
D. Khan et al. / Estrogen and Signaling in the Cells 81

stimulates secretion of IFN␥ from neuroantigen spe- exaggerated or dysregulated IL-17 production results
cific CD4+ T cells from patients with MS [125, 126]. has been observed in various chronic and autoimmune
There is increase in IFN␥+ -secreting cells in lymph diseases such as MS and SLE. A recent report from
nodes of estrogen-treated female ER␣+/+ mice and our laboratory has shown that in vivo estrogen treat-
not in ER␣ deficient mice demonstrating that ER␣, but ment primes splenic lymphocytes to secrete copious
not ER␤, is critical for the enhanced E2-driven Th1 cell amounts of IL-17 when activated with IL-6+TGF␤ and
responsiveness [66]. In vitro and in vivo exposure to antiCD3 antibodies [144].
estrogen increased IFN␥ synthesis from alpha-GalCer Cytokines from Tregs cells-T-regulatory cells (Tregs)

PY
activated iNKT via ER␣ receptor [127]. On the con- exist as natural regulatory cells in the thymus and
trary, estrogen has been shown to inhibit secretion in induced Tregs cells in the peripheral lymphoid organs,
T cells from postmenopausal women and in dendritic which are typically CD4+ CD25+FoxP3+ [145]. These
cells [89, 128, 129]. Physiological (preovulatory) lev- cells exert profound effects to dampen the immune
els of estrogen increase IFN␥ secretion from PHA and responses especially after the antigens are cleared.
CO
LPS-stimulated PBMCs, whereas exposure to preg- Tregs can downregulate all types of T cells includ-
nancy levels of estrogen decreases IFN␥ production ing pro-inflammatory Th-1 and Th-17 cells. Tregs
[130]. In the third trimester of pregnancy, there is use a variety of mechanisms to downregulate these
decreased IFN␥ expression in RA patient compared cells including through secretion of “suppressive”
to healthy women. cytokines, IL-10 and TGF␤. Disturbances in Treg cells
Although estrogen has been shown to increase the can promote autoimmunity and are discussed in the
secretion of Th-1 defining cytokine, IFN␥, there are following section.
OR

other reports, which have demonstrated that estro- Estrogen has been shown to have opposing effects
gen primes CD4 T cells to entirely opposite pathway in autoimmune diseases by exaggerating the patho-
of IL-4 and IL-10-secreting Th2 cell subset. In vivo genesis in systemic lupus erythematosus (SLE)
ovalbumin treatment of ovarietomized rats enhances and attenuating diseases like multiple sclerosis.
estrogen mediated IL-4 secretion from bone mar- Anti-inflammatory action of estrogen is mediated
TH

row cells when compared to control rats [131]. High by decreasing the production of pro-inflammatory
physiological levels of estrogen in T cells increase cytokines or by increasing the secretion of anti-
IL-4 secretion and GATA-3 expression, a transcrip- inflammatory cytokine such as IL-10 and TGF␤
tion factor that can bind the IL-4 promoter [132]. secreted by Treg cells. These cytokines are known
GATA-3 over-expression is associated with ER over- to dampen Th1, Th2 and Th17-mediated immune
AU

expression in hormonally responsive breast cancer, response by blocking of IFN␥, IL-4 and IL-17, respec-
however estrogen treatment in that model did not tively. During pregnancy, when there are high levels
increase GATA-3 expression [133]. Exposure to high of estrogens, there is increase in anti-inflammatory IL-
levels of estrogen either during pregnancy or given 10 levels, decreased maturation of DCs and IL-12p70
experimentally, shifts the cytokine balance to Th2 type [146]. TGF␤ is conventionally regarded as an anti-
[130]. Increased IL-4 and IL-10 accompanied with inflammatory cytokine, transcribed by SP-1, a known
decreased IFN␥ have been reported in PHA-stimulated estrogen-binding partner via the tethered pathway
PBMCs from pregnant women when compared to [73, 147]. TGF␤ secretion is stimulated by estro-
cells from non-pregnant controls suggesting the role gen treatment in astrocytes and fibroblasts [148, 149].
of higher estrogen levels in skewing the response from Estrogen at high physiological levels stimulated pro-
Th1 to Th2 [134, 135]. duction of IL-10 [125] in T cells from human patients.
Th17 cells, Tc17 cells, NKT and ␥␦ T cells secrete IL-10 transcription can be induced by CREB [150],
various proinflammatory cytokine but the most potent a transcription factor known to associate with ER
and important of them all is IL-17 [136]. IL-6 along in breast cancer models [81, 151], and likewise a
TGF␤ is required for commitment of naı̈ve CD4+ putative ERE has been identified in the IL-10 pro-
T cells to Th17 cells. IL-17 aids in recruitment of neu- moter region [151]. It has been demonstrated that
trophil to the site of tissue damage and has been shown Treg suppresses osteoclast differentiation from human
to either protect and enhance immunity against differ- embryonic bone marrow cells by upregulating TGF␤
ent pathogens and vaccine or enhance their clearance and IL-10 induction. Addition of estrogen at concentra-
e.g., Mycoplasma pulmonis, Shigella flexneri, Listeria tions between 10−7 and 10−9 mol/l further suppressed
monocytogenes, Francisella tularensis, Yersinia pestis, osteoclast differentiation by increasing IL-10 and
Helicobacter pylori, influenza A [137–143]. However, TGF␤ expression in Tregs cells. This indicates the
82 D. Khan et al. / Estrogen and Signaling in the Cells

stimulatory effect of estrogen on Tregs [152]. Estro- Estrogen treatment has been shown to decrease
gen treatment also increases FoxP3 expression in Tregs clinical and histological signs of EAE in mice. It is
[153]. Additionally, programmed death -1 (PD-1), a thought that estrogen accomplishes this by inhibit-
negative co-stimulatory molecule that is expressed ing the autoantigen specific proinflammatory cytokine
intracellularly by Tregs is also upregulated by estrogen production, and inhibiting inflammation and demyeli-
treatment. In ER knockout mice, there is suppressed nation. For example, estrogen has been shown to inhibit
PD-1 expression and Tregs activity [154]. It has been production of inflammatory molecules (such as IFN␥,
demonstrated that the protective effects of estrogen on tumor necrosis factor-alpha (TNF␣), iNOS, and MCP-

PY
EAE, by increasing FoxP3 expression and suppressive 1) in microglia cells from EAE mice [157]. Estrogen
activity of Tregs are mediated through ER␣ but not treatment is neuroprotective as it helps in preserv-
ER␤ [155]. Furthermore, physiological levels of estro- ing the integrity of neurons and axon with decreased
gen in vitro directly converted activated CD4+ CD25– activation of microglial and monocytes in the central
T cells into CD4+ CD25+ Treg cells which correlated nervous system [159]. The anti-inflammatory and neu-
CO
with expression of ER␣ in these cells [156]. From roprotective effects of estrogen on EAE are mediated
published reports, it is evident that estrogen plays an through ER␣ but not ER␤ [157, 159, 160]. Immuno-
important role in regulating the number and activity of modulatory effects of estrogen are also demonstrated
Treg cells in various autoimmune diseases such as MS in a study where splenic dendritic cells (DC) exposed to
and RA. In the following section we will compare the estrogen, in vitro, had therapeutic effects on acute EAE
opposing roles of estrogen on two female predominant with inhibition of CD4+ T cells expansion, increased
autoimmune diseases. proportions of Tregs and CD4+ CD8− suppressor
OR

T cells [129], increased T cell apoptosis [161], and


increased Th2 cytokines such as IL-10, IL-4 accompa-
nied with decreased Th1 cytokines such as TNF␣ and
CONTRASTING INFLUENCE OF IFN␥ [162]. Interestingly, estrogen profoundly altered
ESTROGEN ON MULTIPLE SCLEROSIS expression pattern of 315 genes in spinal cord tissue
TH

AND SYSTEMIC LUPUS of mice protected from EAE, of which 302 genes were
ERYTHEMATOSUS down-regulated and only 13 genes up-regulated [163].
In pregnant mice with EAE, there is reduced CNS
Clearly estrogen can mediate both inflammatory and pathology and less TNF␣ and IL-17 production along
anti-inflammatory effects, depending on the cellular with reduced T cells activation when compared to non-
AU

context, specific ligand as well as receptor reper- pregnant controls [164]. In addition, myeloid-derived
toire. Likewise varying effects of estrogen on immune cells especially DCs have shown to mediate protec-
diseases has been observed. Estrogens can both pro- tive effects of estrogen. In vivo treatment of mice
mote autoimmune disease, as in the case of systemic with pregnancy specific estrogen, estriol, generated
lupus erythematosus (SLE), and reduce or even pro- tolerogenic DCs [165]. These DCs had upregulated
tect against autoimmune disease, as in the case of activation markers (CD80 and CD86) and inhibitory
multiple sclerosis (MS). Experimental autoimmune costimulatory markers (PD-L1, PD-L2, B7-H3, and
encephalitis (EAE) is a Th-1 cell mediated autoim- B7-H4). Furthermore, DCs from estriol treated mice
mune disease and serves as a model for MS, an had increased IL-10 and TGF␤ but decreased proin-
inflammatory demyelinating disease of the central ner- flammatory IL-12, IL-23 and IL-6 mRNA expression.
vous system. Transfer of DCs from estriol treated mice protected
MS and EAE are characterized by activation of the recipient mice from active induction of EAE
auto-reactive CD4+ T cells that target myelin anti- [165]. With use of conditional ER␣ deficient mice
gens such as myelin oligodendrocyte glycoprotein and bone marrow chimeras, it has been demonstrated
(MOG) [157, 158]. This results in infiltration of that ER␣ in hematopoietic cells but not in endothe-
CD4+ T cells (predominantly Th-1) and other pro- lial cells is essential for estrogen-mediated inhibition
inflammatory cells into the central nervous system of Th1 and Th17 cell differentiation and protection
(CNS) and de-myelination of axons by macrophages, from EAE [166]. ER␣ signaling in T cells is indispens-
ultimately leading to axonal death and decreased able and sufficient for estrogen-mediated protection
CNS function [158]. Clinically, MS often presents as against Th17-cells driven CNS inflammation in
a relapsing-remitting disease, with variable rates of EAE mice [166] and CD4+ T cells homing in the CNS
relapse between individuals. [167].
D. Khan et al. / Estrogen and Signaling in the Cells 83

The anti-inflammatory role of estrogen was impli- tions, serositis and renal disease. Female patients have
cated when it was observed that rates of relapse in more psychiatric symptoms and headaches whereas
women were significantly reduced during pregnancy males have more seizures and peripheral neuropathy
when estrogen levels are high, however rates rose [179].
substantially post-partum when estrogen drops precipi- Estrogen can promote SLE progression via a num-
tously [168, 169]. A small clinical trial of women with ber of mechanisms including induction of cytokine
MS given E3 at pregnancy levels showed a decrease and autoantibodies and autoantigens [180, 181]. First,
in the number and size of brain lesions measured by by induction of Th-2 type cytokines including IL-4,

PY
MRI [170]. Experiments in mouse EAE models have IL-6 and IL-10, as discussed in the review of cytokines,
demonstrated E2 and to a greater extent E3 treatment estrogen promotes skewing of T cell populations
is protective against development of EAE and reduces towards Th-2, the canonical B cell “helper” which
the severity of disease after onset [157]. There is a then activates auto-reactive B cells. This imbalance in
decrease in inflammation upon estrogen treatment by cytokine is accompanied with flares of SLE and throm-
CO
a number of mechanisms, including reduced secretion bosis and has been confirmed in pregnant females,
of TNF␣ by autoreactive T cells and macrophages, where there is high amount of estrogen in the circu-
reduced recruitment of inflammatory cells to the CNS, lation, and also in females given exogenous estrogen
as well as induction of CD4+ CD25+ Tregs. It also [182]. However, from randomized trials and large
down-regulates cellular adhesion molecules such as observational studies it is now demonstrated that SLE
VCAM and ICAM in endothelial cells and modulates patients on HRT have only marginal increase in the
the GTPase Rac1 in macrophages, inhibiting adhesion risk of mild/moderate flare and thrombosis and no risk
OR

and migration [171]. The anti-inflammatory actions of of major flare [183, 184]. Estrogen also stimulated
estrogens thus inhibit autoimmunity in the context of secretion of IL-10 in monocytes along with increased
a Th-1 T cell-mediated disease such as MS. anti-double-stranded DNA antibody and immunoglob-
In the case of SLE, estrogen is associated with ulin G production by peripheral blood mononuclear
increased severity and increased flares of the disease, cells from patients with SLE [185, 186].
TH

both in humans and in animal models [85, 89]. The Estrogen has been shown to regulate B cell matura-
female: male ratio of 9 : 1 to 20 : 1 has been reported tion and selection, in a mouse model of lupus [187].
for SLE susceptibility [172, 173]. Lupus disease man- It has been demonstrated that high levels of estro-
ifests as multiple symptoms that vary by patient. gen in serum promotes maturation of a pathogenic
Lupus involves nephritis, skin rash, arrhythmias and naive autoreactive B cell population and decreases the
AU

neuronal effects such as numbness, tingling, and psy- maturation of a potentially protective autoreactive B
chiatric changes [174]. Dysregulation of the immune cell repertoire [188]. In addition, estrogen treatment
system at multiple levels, including loss of tolerance, increased IgG and IgM secretion from PBMCs as
altered T cell signaling, and T cell, B cell, and mono- well as serum levels of IgG and IgA in rodents and
cyte hyperactivity, lead to activation of autoreactive B humans [186, 189–191]. Furthermore, in mouse model
cells and secretion of pathogenic antibodies includ- for induced lupus, exposure to estrogen increases the
ing anti-ds DNA and anti-phoshpholipid antibodies titer of antiDNA antibodies and also leads to systemic
[175–178]. The accumulation of immune complexes inflammation with increased B cell-activating factor
which lodge in small capillaries, triggering further and IFN levels and induction of an IFN signature
localized inflammatory cascades, have been shown to [192]. Estrogen exposure increases calcineurin expres-
cause SLE nephritis, skin rash and are likely respon- sion from T cells from SLE patients when compared
sible for many other symptoms. There is a shift in with healthy females [193]. Not only this, estrogen also
T cell populations from Th-1 to Th-2, associated with upregulates the expression of endogenous autoanti-
an increase in serum concentrations of Th-2 associ- gens such as human endogenous retroviruses [194],
ated cytokines IL-4, IL-6 and IL-10 and reduction in and reactivity to exogenous antigens [195]. HERV is
the Th-1 cytokine IL-2 and IFN␥ [89]. IFN␥ still plays found to be increased in SLE wherein HERV act as
a role in glomerulonephritis implying different effects autoantigens by molecularly mimicking RNP antigens
of estrogen in various tissue pathology. Gender differ- in the patient’s body [196].
ences in SLE predilection and pathogenesis are well In mice model for SLE, it has been reported that there
documented. Clinically, females have more frequent is differential expression of ERs in MRL/MP-lpr/lpr
relapses and Raynaud’s phenomenon, arthritis and and NZB/W mice when compared with BALB/c mice
leucopenia, while males have more skin manifesta- [197]. One reason for hyper-responsiveness of estrogen
84 D. Khan et al. / Estrogen and Signaling in the Cells

in SLE patients could be due to altered quantitative or C57BL/6 aged matched controls [210]. Further-
and/or qualitative expression of ERs. For example, more, the mRNA level of IRF5 in splenic cells was
in SLE patients, there is high expression of ER␣ on found to be decreased in ER␣−/− mice when compared
CD4+ T cells while there is decreased ER␤ on PBMCs, with female ER␣+/+ mice and treatment of splenic
which inversely correlates with the SLE disease activ- cells with 17␤–estradiol upregulated IRF5 mRNA lev-
ity index (SLEDAI) or prednisolone dose [198]. In els. Impressively, IRF5 mRNA was high in nuclei
addition, estrogen treatment modulated expression of of splenic B cells from female mice when compared
ERs in immune cells of autoimmune-prone SNF(1) to male mice [210]. Interestingly, IFN␥ and IFN␣

PY
and non-autoimmune DBF(1) mice. Estrogen exposure can upregulate ER␣, which demonstrates the feedback
increased ER␣ expressing CD4+ and CD8+ T cells and interactions of cytokines and ERs. Overall, it is clear
percent ER␣+ DCs and macrophages in SNF(1) mice that estrogen effects vary based on the autoimmune
but not in DBF(1) [199]. Studies have shown that ER␣− disease setting.
mediated signaling is important in estrogen-induced
CO
development of lupus phenotype. Estrogen treatment
of wildtype mice had accelerated lupus development, MIRNA AND ESTROGEN
increased autoantibody and IL-5, IL-6, IL-10, IFN␥
and TNF␣ production with increased kidney damage From recent reports it is now evident that estrogens
when compared with estrogen-treated ER␣ defi- play an important in immune response by epige-
cient mice [199]. In lupus-prone (NZB × NZW)F(1) netic microRNA regulation [211, 212]. MicroRNAs
mice, ER␣ deficiency attenuated glomerulonephritis, (miRNA) are endogenous non-coding RNAs of an
OR

decreased anti-histone/DNA antibodies and increased average of 21-22 nt in length [213]. The role of miRNA
survival [200]. Furthermore, with ER selective ago- in post-transcriptional gene regulation has recently
nists, importance of ER␣ in lupus development has been defined. MiRNA bind to the 3 untranslated region
been demonstrated [201]. Together, these studies (UTR) of target mRNA by partial homology result-
indicate that ER␣ is essential in estrogen-mediated ing in either degradation of mRNA or inhibition of
TH

exaggeration of lupus disease. translation. miRNAs also participate in the regula-


Polymorphism in ER␣ gene expression has also tion of autoimmunity: deficiency in Dicer or Drosha
been reported in SLE patients [202, 203]. Furthermore, leads to autoimmunity in mice [214]. Aberrant miRNA
out of 13 genes identified as significantly altered during expression has been demonstrated in several autoim-
menstrual cycle in females but not in males, six were mune diseases including multiple sclerosis (MS), and
AU

statistically different in SLE patients when compared SLE [215]. MicroRNAs have recently been implicated
with normal controls [204]. Tumor necrosis factor in the pathogenesis of MS by multiple studies, how-
receptor superfamily member (TNFRSF14) also called ever there is great variability between these studies
Herpes virus entry mediator (HVEM) is one such gene, and the precise disease-associated miRNAs [216–221].
which is quantitatively altered in both females and SLE This variation is likely due to differences in tissue
patients. It is a ligand for B and T lymphocyte atten- and patient populations studied as well as techniques
uator (BTLA). Interaction of TNFRSF14 with BTLA employed to identify miRNAs. The possible contribu-
downregulates lymphocyte activation and homeostasis tion of hormonal regulation of these disease-associated
[205]. In menstrual cycle, estrogen increases expres- miRNAs has not been addressed directly.
sion of TNFRSF14 mRNA in PBMCs, however in SLE Of the MS-associated miRNAs identified, only a few
patients TNFRSF14 mRNA is lowered which results have been shown to be regulated by estrogen [212].
in partial activity of BTLA thus leading to net immune These include miR-145, miR-200b, and miR-486.
enhancement [205, 206]. Keller et al. demonstrated upregulation of miR-145
In addition, SLE patients have other risk alle- in whole blood from relapsing-remitting MS patients
les susceptibility loci such as interferon regulatory compared to healthy controls [219]. While the tar-
factor (IRF5) [207]. It is suggested that immune com- get of miR-145 in immune cells is unknown, it has
plex of nuclear antigens activate intracellular TLRs been show to be critically involved with multiple stem
such as TLR7 and TLR9-mediated IRF5 pathway, cell differentiation pathways, including Sox9 and Oct4
which upregulates IFN␣ secretion in SLE patients and [222, 223]. In vivo estrogen treatment downregulated
MRL/lpr mice, [208, 209]. Increased levels of IRF5 expression of miR-145 in murine splenic lymphocytes
have been reported in female NZB and NZB/W F(1) [212]. Both miR-200b and miR-486 were found to be
mice when compared to male NZB and NZB/W F(1) upregulated in CD4+ cells from relapsing-remitting
D. Khan et al. / Estrogen and Signaling in the Cells 85

MS suffers compared to healthy controls. Estrogen Microarray analysis data revealed regulation of
downregulated miR-200b, which has a broad range 25 miRNA by estrogen in splenic lymphocytes.
of target mRNAs in different cell types, including While miR-223, miR18a and miR-708 were increased,
the transcription factors ZEB1/2, RND3 and Ets-1 miR-146a, miR-125a, miR-125b, miR-143, miR-145,
[224–226]. It is possible that estrogen downregulation let-7e, miR-126 were found to be decreased in
of miR-145 and miR-200b could be one of the rea- freshly-isolated splenic lymphocytes from estrogen-
sons for estrogen-regulated alleviation of MS since treated mice when compared to placebo controls.
both of them are found to be upregulated in MS Increased activity of miR-146a significantly lowered

PY
patients. Unlike the previous miRNAs, estrogen treat- LPS-induced IFN␥ and iNOS expression, however
ment upregulated expression of miR-486 by nearly 7 decreased activity of miR-223 decreased LPS-induced
fold, in splenic lymphoid cells [212]. The antiapoptotic IFN␥ in cells from estrogen-treated mice suggest-
glycoprotein OLFM4 is a putative target of miR-486 ing important role of miRNA in estrogen-mediated
in gastric cancers, however its expression and role in immune regulation [212]. Since some of the miRNA
CO
lymphocytes remains to be determined [227]. Together, dysregulated in SLE and modulated by estrogen are
these miRNAs can be used as potential targets to treat common such as miR-146a, miR-125a etc., it is plau-
MS. sible that estrogen-mediated exaggeration of SLE may
In pediatric SLE patients, miR-181a is significantly be due to dysregulated miRNA expression. Recent
downregulated [228]. It is noteworthy that, miR-181a studies add new dimension to our current understand-
is critical modulator of B and T cell differentiation, ing of immune regulation by estrogen.
maturation and function. In another report, 11 miRNA
OR

were significantly altered in CD4+ T cells of SLE


patients of which miR-126 was increased which tar- CONCLUSION
geted DNA methyltransferase 1 (Dnmt1). The decrease
in Dnmt1 resulted in demethylation and increased Sex hormone regulation of the immune system
expression of CD11a and CD70 resulting in hyperac- is now very well recognized. Estrogens act on the
TH

tivity of T and B cells [229]. MiR-146a and miR-125a immune system by estrogen receptor or non-receptor
are decreased in human SLE patients [230, 231]. dependent mechanisms. While it is clear that estrogen
Decrease in expression of miR-146a contributes to regulates the immune system, the outcome of estrogen-
alteration in type 1 IFN signaling with increased IRF- induced immune response is variable. It is therefore
5, STAT-1 and IFN scores [232]. Decreased miR-125a difficult to generalize the effect of estrogen since
AU

in SLE patient results in upregulated miR-125a tar- the overall outcome i.e., either immuno-suppression
get RANTES via KLF13 expression [231]. miR-21 is or immune enhancement depends on multiple fac-
upregulated in SLE patients and positively correlates tors such as type of tissue; health of host; presence
with disease activity. miR-21 has been shown to target or absence of estrogens; dose and type of estrogen;
a protein translation inhibitor, PDCD4, which results type of receptor expression; qualitative or quantita-
in aberrant T cell activity [233]. tive expression of receptor; and signaling pathways,
In our recent study, common set of dysregulated presence of adaptor molecules, coactivators and core-
miRNAs, in splenic lymphocytes, purified T and pressors. It is now recognized that estrogen exists in
B cells, were identified in three genetically dif- multiple forms natural, synthetic, plant-derived or fun-
ferent murine lupus models, MRL-lpr, B6-lpr and gal form. It is possible that there may be a subset
NZB/W(F1 ) [234]. By miRNA microarray assays and of individuals who could be very sensitive to estro-
Real-time RT-PCR analysis, miR-182-96-183 cluster, gen. It is noteworthy that a majority of autoimmune
miR-31, and miR-155 were found to be decreased in diseases are female predominant [235]. The precise
splenic lymphocytes of all three strains with active dis- reasons for female gender predisposition of autoim-
ease when compared with their age-matched controls. mune disease remain unclear. The evident gender
However, miR-146a, miR-101a, and miR-17-92 were bias has been inconclusively associated with different
markedly upregulated only in splenic T, and not in B hypothesis such as X-chromosomal inactivation, fetal
cells from MRL-lpr mice. There was difference in level microchimerism, X chromosomal abnormalities, epi-
of expression across different strains e.g., miR-127 genetic miRNA and histone deacetylation. It is well
and miR-379 were greatly upregulated in splenocytes established that sex hormones play a critical role in
from lpr mice, but were only moderately increased in the disease pathogenesis since the fluctuations in the
diseased NZB/W mice [234]. levels of estrogen with age and estrus cycle modulate
86 D. Khan et al. / Estrogen and Signaling in the Cells

the immune parameters and disease severity. Although [12] Lathers CM. Endocrine disruptors: A new scientific role for
studies are being conducted to define the role of estro- clinical pharmacologists? Impact on human health, wildlife,
and the environment. J Clin Pharmacol. 2002; 42(1): 7-23.
gen in these chronic lifelong debilitating diseases, their Epub 2002/01/26.
precise affects needs to be answered. The complex- [13] Ahmed SA. The immune system as a potential target
ity of genetic factors, age, gender and other signaling for environmental estrogens (endocrine disrupters): a new
pathways together with environmental exposure to new emerging field. Toxicology. 2000; 150(1-3): 191-206.
[14] Milla S, Depiereux S, Kestemont P. The effects of estrogenic
chemicals makes it hard to conclusively predict the and androgenic endocrine disruptors on the immune system
outcome of sex hormone in clinical conditions. It is of fish: a review. Ecotoxicology. 2011; 20(2): 305-319. Epub

PY
unwise to generalize the estrogen-mediated immune 2011/01/07.
cell signaling and thus each disease state must be [15] Jordan VC, Mittal S, Gosden B, Koch R, Lieberman ME.
Structure-activity relationships of estrogens. Environ Health
examined independently. Therefore, future mechanis- Perspect. 1985; 61: 97-110. Epub 1985/09/01.
tic studies are warranted to understand the specific role [16] Chalubinski M, Kowalski ML. Endocrine disrupters–
of estrogen in immune regulation in a particular clinical potential modulators of the immune system and aller-
CO
gic response. Allergy. 2006; 61(11): 1326-1335. Epub
condition and disease.
2006/09/28.
[17] Marin DE, Taranu I, Burlacu R, Manda G, Motiu M, Neagoe
I, et al. Effects of zearalenone and its derivatives on porcine
REFERENCES immune response. Toxicol In Vitro. 2011. Epub 2011/07/19.
[18] Cunningham JG, Klein BG. Texbook of Veterinary Physi-
[1] Gustafsson JA. What pharmacologists can learn from recent ology. 4th ed: Saunders Elsevier; 2007.
advances in estrogen signalling. Trends Pharmacol Sci. [19] Simpson ER. Sources of estrogen and their importance.
OR

2003; 24(9): 479-485. Epub 2003/09/12. J Steroid Biochem Mol Biol. 2003; 86(3-5): 225-230.
[2] Carani C, Qin K, Simoni M, Faustini-Fustini M, Serpente S, [20] Labrie F, Belanger A, Luu-The V, Labrie C, Simard J, Cusan
Boyd J, et al. Effect of testosterone and estradiol in a man L, et al. DHEA and the intracrine formation of androgens
with aromatase deficiency. The New England Journal of and estrogens in peripheral target tissues: its role during
Medicine. 1997; 337(2): 91-95. Epub 1997/07/10. aging Steroids. 1998; 63(5-6): 322-328.
[3] Smith EP, Boyd J, Frank GR, Takahashi H, Cohen RM, [21] Bebo BF Jr, Fyfe-Johnson A, Adlard K, Beam AG,
Specker B, et al. Estrogen resistance caused by a mutation Vandenbark AA, Offner H. Low-dose estrogen therapy
TH

in the estrogen-receptor gene in a man. The New Eng- ameliorates experimental autoimmune encephalomyelitis in
land Journal of Medicine. 1994; 331(16): 1056-1061. Epub two different inbred mouse strains. Journal of immunology.
1994/10/20. 2001; 166(3): 2080-2089.
[4] Herrmann BL, Saller B, Janssen OE, Gocke P, Bockisch A, [22] Couse JF, Korach KS. Estrogen receptor null mice: What
Sperling H, et al. Impact of estrogen replacement therapy have we learned and where will they lead us? Endocr Rev.
in a male with congenital aromatase deficiency caused by a 1999; 20(3): 358-417. Epub 1999/06/16.
AU

novel mutation in the CYP19 gene. J Clin Endocrinol Metab. [23] Walmer DK, Wrona MA, Hughes CL, Nelson KG. Lacto-
2002; 87(12): 5476-5484. Epub 2002/12/06. ferrin expression in the mouse reproductive tract during the
[5] Gruber DM, Huber JC. Conjugated estrogens–the natural natural estrous cycle: correlation with circulating estradiol
SERMs. Gynecological endocrinology: the Official Journal and progesterone. Endocrinology. 1992; 131(3): 1458-1466.
of the International Society of Gynecological Endocrinol- Epub 1992/09/01.
ogy. 1999; 13(Suppl 6): 9-12. Epub 2000/06/22. [24] Askanase AD, Buyon JP. Reproductive health in SLE. Best
[6] Kuiper GG, Carlsson B, Grandien K, Enmark E, Haggblad J, practice & research Clinical rheumatology. 2002; 16(2):
Nilsson S, et al. Comparison of the ligand binding specificity 265-280. Epub 2002/06/04.
and transcript tissue distribution of estrogen receptors alpha [25] Levitz M, Young BK. Estrogens in pregnancy. Vitamins and
and beta. Endocrinology. 1997; 138(3): 863-870. Hormones. 1977; 35: 109-147. Epub 1977/01/01.
[7] Cutolo M, Villaggio B, Seriolo B, Montagna P, Capellino [26] Dogru MT, Basar MM, Yuvanc E, Simsek V, Sahin O. The
S, Straub RH, et al. Synovial fluid estrogens in rheuma- relationship between serum sex steroid levels and heart rate
toid arthritis. Autoimmunity Reviews. 2004; 3(3): 193-198. variability parameters in males and the effect of age. Turk
Epub 2004/04/28. Kardiyoloji Dernegi arsivi: Turk Kardiyoloji Derneginin
[8] Miller L, Hughes JP. Continuous combination oral yayin organidir. 2010; 38(7): 459-465. Epub 2011/01/06.
contraceptive pills to eliminate withdrawal bleeding: [27] Rosenbaum W, Christy NP, Kelly WG. Electrophoretic evi-
A randomized trial. Obstet Gynecol. 2003; 101(4): 653-661. dence for the presence of an estrogen-binding beta-globulin
Epub 2003/04/10. in human plasma. J Clin Endocrinol Metab. 1966; 26(12):
[9] Katzenellenbogen BS, Katzenellenbogen JA. Biomedicine. 1399-1403. Epub 1966/12/01.
Defining the “S” in SERMs. Science. 2002; 295(5564): [28] Rosner W, Hryb DJ, Kahn SM, Nakhla AM, Romas NA.
2380-2381. Epub 2002/03/30. Interactions of sex hormone-binding globulin with target
[10] Bolger R, Wiese TE, Ervin K, Nestich S, Checovich W. cells. Mol Cell Endocrinol. 2010; 316(1): 79-85. Epub
Rapid screening of environmental chemicals for estrogen 2009/08/25.
receptor binding capacity. Environ Health Perspect. 1998; [29] Rosner M, Fuchs C, Siegel N, Valli A, Hengstschlager M.
106(9): 551-557. Epub 1998/08/29. Functional interaction of mammalian target of rapamycin
[11] Waring RH, Harris RM. Endocrine disrupters: a human complexes in regulating mammalian cell size and cell
risk? Mol Cell Endocrinol. 2005; 244(1-2): 2-9. Epub cycle. Hum Mol Genet. 2009; 18(17): 3298-3310. Epub
2005/11/08. 2009/06/10.
D. Khan et al. / Estrogen and Signaling in the Cells 87

[30] Jensen EV. On the mechanism of estrogen action. Perspect [45] Couse JF, Lindzey J, Grandien K, Gustafsson JA, Korach
Biol Med. 1962; 6: 47-59. Epub 1962/01/01. KS. Tissue distribution and quantitative analysis of estro-
[31] Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafs- gen receptor-alpha (ERalpha) and estrogen receptor-beta
son JA. Cloning of a novel receptor expressed in rat prostate (ERbeta) messenger ribonucleic acid in the wild-type and
and ovary. Proceedings of the National Academy of Sciences ERalpha-knockout mouse. Endocrinology. 1997; 138(11):
of the United States of America. 1996; 93(12): 5925-5930. 4613-4621. Epub 1997/11/05.
Epub 1996/06/11. [46] Hiroi H, Inoue S, Watanabe T, Goto W, Orimo A, Momoeda
[32] Krege JH, Hodgin JB, Couse JF, Enmark E, Warner M, M, et al. Differential immunolocalization of estrogen recep-
Mahler JF, et al. Generation and reproductive phenotypes tor alpha and beta in rat ovary and uterus. J Mol Endocrinol.
of mice lacking estrogen receptor beta. Proceedings of 1999; 22(1): 37-44. Epub 1999/01/30.

PY
the National Academy of Sciences of the United States [47] Sar M, Welsch F. Differential expression of estrogen
of America. 1998; 95(26): 15677-15682. Epub 1998/ receptor-beta and estrogen receptor-alpha in the rat ovary.
12/23. Endocrinology. 1999; 140(2): 963-971. Epub 1999/02/02.
[33] Flouriot G, Brand H, Denger S, Metivier R, Kos M, Reid [48] Nishihara E, Nagayama Y, Inoue S, Hiroi H, Muramatsu M,
G, et al. Identification of a new isoform of the human estro- Yamashita S, et al. Ontogenetic changes in the expression
gen receptor-alpha (hER-alpha) that is encoded by distinct of estrogen receptor alpha and beta in rat pituitary gland
CO
transcripts and that is able to repress hER-alpha activa- detected by immunohistochemistry. Endocrinology. 2000;
tion function 1. EMBO J. 2000; 19(17): 4688-4700. Epub 141(2): 615-20. Epub 2000/01/29.
2000/09/06. [49] Brandenberger AW, Tee MK, Lee JY, Chao V, Jaffe RB. Tis-
[34] Wang Z, Zhang X, Shen P, Loggie BW, Chang Y, Deuel sue distribution of estrogen receptors alpha (ER-alpha) and
TF. Identification, cloning, and expression of human estro- beta (ER-beta) mRNA in the midgestational human fetus.
gen receptor-alpha36, a novel variant of human estrogen J Clin Endocrinol Metab. 1997; 82(10): 3509-3512. Epub
receptor-alpha66. Biochem Biophys Res Commun. 2005; 1997/11/05.
336(4): 1023-1027. Epub 2005/09/17. [50] Carreras E, Turner S, Paharkova-Vatchkova V, Mao A,
OR

[35] Green S, Walter P, Kumar V, Krust A, Bornert JM, Argos P, Dascher C, Kovats S. Estradiol acts directly on bone mar-
et al. Human oestrogen receptor cDNA: Sequence, expres- row myeloid progenitors to differentially regulate GM-CSF
sion and homology to v-erb-A. Nature. 1986; 320(6058): or Flt3 ligand-mediated dendritic cell differentiation. Jour-
134-139. Epub 1986/03/13. nal of Immunology. 2008; 180(2): 727-738. Epub 2008/
[36] Leygue E, Dotzlaw H, Lu B, Glor C, Watson PH, Mur- 01/08.
phy LC. Estrogen receptor beta: Mine is longer than yours? [51] Seiki K, Sakabe K. Sex hormones and the thymus in rela-
J Clin Endocrinol Metab. 1998; 83(10): 3754-3755. Epub tion to thymocyte proliferation and maturation. Arch Histol
TH

1998/10/13. Cytol. 1997; 60(1): 29-38.


[37] Inoue S, Ogawa S, Horie K, Hoshino S, Goto W, Hosoi T, [52] Smithson G, Medina K, Ponting I, Kincade PW. Estrogen
et al. An estrogen receptor beta isoform that lacks exon 5 suppresses stromal cell-dependent lymphopoiesis in culture.
has dominant negative activity on both ERalpha and ERbeta. Journal of Immunology. 1995; 155(7): 3409-3417.
Biochem Biophys Res Commun. 2000; 279(3): 814-819. [53] Kawashima I, Seiki K, Sakabe K, Ihara S, Akatsuka A, Kat-
Epub 2001/02/13. sumata Y. Localization of estrogen receptors and estrogen
AU

[38] Moore JT, McKee DD, Slentz-Kesler K, Moore LB, Jones receptor-mRNA in female mouse thymus. Thymus. 1992;
SA, Horne EL, et al. Cloning and characterization of human 20(2): 115-121. Epub 1992/09/01.
estrogen receptor beta isoforms. Biochem Biophys Res [54] Mor G, Munoz A, Redlinger R Jr., Silva I, Song J, Lim C,
Commun. 1998; 247(1): 75-78. Epub 1998/06/24. et al. The role of the Fas/Fas ligand system in estrogen-
[39] Ogawa S, Inoue S, Watanabe T, Orimo A, Hosoi T, Ouchi induced thymic alteration. Am J Reprod Immunol. 2001;
Y, et al. Molecular cloning and characterization of human 46(4): 298-307.
estrogen receptor betacx: a potential inhibitor ofestrogen [55] Lambert KC, Curran EM, Judy BM, Lubahn DB, Estes
action in human. Nucleic Acids Res. 1998; 26(15): 3505- DM. Estrogen receptor-alpha deficiency promotes increased
3512. Epub 1998/07/22. TNF-alpha secretion and bacterial killing by murine
[40] Poola I, Abraham J, Baldwin K, Saunders A, Bhatnagar R. macrophages in response to microbial stimuli in vitro. Jour-
Estrogen receptors beta4 and beta5 are full length function- nal of Leukocyte Biology. 2004; 75(6): 1166-1172. Epub
ally distinct ERbeta isoforms: cloning from human ovary 2004/03/17.
and functional characterization. Endocrine. 2005; 27(3): [56] Vidal O, Kindblom LG, Ohlsson C. Expression and local-
227-238. Epub 2005/10/19. ization of estrogen receptor-beta in murine and human
[41] Heldring N, Pike A, Andersson S, Matthews J, Cheng G, bone. J Bone Miner Res. 1999; 14(6): 923-929. Epub
Hartman J, et al. Estrogen receptors: How do they signal and 1999/06/03.
what are their targets. Physiol Rev. 2007; 87(3): 905-931. [57] Smithson G, Couse JF, Lubahn DB, Korach KS, Kincade
Epub 2007/07/07. PW. The role of estrogen receptors and androgen recep-
[42] Brzozowski AM, Pike AC, Dauter Z, Hubbard RE, Bonn T, tors in sex steroid regulation of B lymphopoiesis. Journal of
Engstrom O, et al. Molecular basis of agonism and antag- Immunology. 1998; 161(1): 27-34.
onism in the oestrogen receptor. Nature. 1997; 389(6652): [58] Erlandsson MC, Ohlsson C, Gustafsson JA, Carlsten H.
753-758. Epub 1997/10/24 21:29. Role of oestrogen receptors alpha and beta in immune organ
[43] Mosselman S, Polman J, Dijkema R. ER beta: identification development and in oestrogen-mediated effects on thymus.
and characterization of a novel human estrogen receptor. Immunology. 2001; 103(1): 17-25.
FEBS letters. 1996; 392(1): 49-53. [59] Grimaldi CM, Cleary J, Dagtas AS, Moussai D, Diamond
[44] Shughrue PJ, Komm B, Merchenthaler I. The distribution B. Estrogen alters thresholds for B cell apoptosis and activa-
of estrogen receptor-beta mRNA in the rat hypothalamus. tion. The Journal of Clinical Investigation. 2002; 109(12):
Steroids. 1996; 61(12): 678-681. Epub 1996/12/01. 1625-1633.
88 D. Khan et al. / Estrogen and Signaling in the Cells

[60] Phiel KL, Henderson RA, Adelman SJ, Elloso MM. Differ- subtypes. Mol Endocrinol. 2001; 15(6): 909-922. Epub
ential estrogen receptor gene expression in human peripheral 2001/05/29.
blood mononuclear cell populations. Immunol Lett. 2005; [75] McInerney EM, Tsai MJ, O’Malley BW, Katzenellen-
97(1): 107-113. Epub 2005/01/01. bogen BS. Analysis of estrogen receptor transcriptional
[61] Nancy P, Berrih-Aknin S. Differential estrogen receptor enhancement by a nuclear hormone receptor coactivator.
expression in autoimmune myasthenia gravis. Endocrinol- Proceedings of the National Academy of Sciences of the
ogy. 2005; 146(5): 2345-2353. Epub 2005/01/22. United States of America. 1996; 93(19): 10069-10073.
[62] Pierdominici M, Maselli A, Colasanti T, Giammarioli AM, [76] O’Lone R, Frith MC, Karlsson EK, Hansen U. Genomic
Delunardo F, Vacirca D, et al. Estrogen receptor profiles in targets of nuclear estrogen receptors. Mol Endocrinol. 2004;
human peripheral blood lymphocytes. Immunology Letters. 18(8): 1859-1875. Epub 2004/03/20.

PY
2010; 132(1-2): 79-85. Epub 2010/06/15. [77] Kushner PJ, Agard DA, Greene GL, Scanlan TS, Shiau AK,
[63] Panchanathan R, Shen H, Zhang X, Ho SM, Choubey D. Uht RM, et al. Estrogen receptor pathways to AP-1. J Steroid
Mutually positive regulatory feedback loop between inter- Biochem Mol Biol. 2000; 74(5): 311-317.
ferons and estrogen receptor-alpha in mice: implications for [78] Biswas DK, Singh S, Shi Q, Pardee AB, Iglehart JD. Cross-
sex bias in autoimmunity. PloS One. 2010; 5(5): e10868. roads of estrogen receptor and NF-kappaB signaling. Sci
Epub 2010/06/09. STKE. 2005; 2005(288): pe27. Epub 2005/06/16.
CO
[64] Staples JE, Gasiewicz TA, Fiore NC, Lubahn DB, Korach [79] Fox EM, Andrade J, Shupnik MA. Novel actions of estro-
KS, Silverstone AE. Estrogen receptor alpha is necessary gen to promote proliferation: integration of cytoplasmic and
in thymic development and estradiol-induced thymic alter- nuclear pathways. Steroids. 2009; 74(7): 622-627. Epub
ations. Journal of Immunology. 1999; 163(8): 4168-74. 2008/11/11.
[65] Shim GJ, Kis LL, Warner M, Gustafsson JA. Autoimmune [80] Saville B, Wormke M, Wang F, Nguyen T, Enmark E,
glomerulonephritis with spontaneous formation of splenic Kuiper G, et al. Ligand-, cell-, and estrogen receptor sub-
germinal centers in mice lacking the estrogen receptor alpha type (alpha/beta)-dependent activation at GC-rich (Sp1)
gene. Proceedings of the National Academy of Sciences promoter elements. J Biol Chem. 2000; 275(8): 5379-5387.
OR

of the United States of America. 2004; 101(6): 1720-1724. Epub 2000/02/22.


Epub 2004/01/28. [81] Bjornstrom L, Sjoberg M. Mechanisms of estrogen receptor
[66] Maret A, Coudert JD, Garidou L, Foucras G, Gourdy P, Krust signaling: convergence of genomic and nongenomic actions
A, et al. Estradiol enhances primary antigen-specific CD4 on target genes. Mol Endocrinol. 2005; 19(4): 833-842.
T cell responses and Th1 development in vivo. Essential [82] Ghisletti S, Meda C, Maggi A, Vegeto E. 17beta-estradiol
role of estrogen receptor alpha expression in hematopoi- inhibits inflammatory gene expression by controlling NF-
etic cells. European Journal of Immunology. 2003; 33(2): kappaB intracellular localization. Mol Cell Biol. 2005;
TH

512-521. 25(8): 2957-2968. Epub 2005/03/31.


[67] Shim GJ, Wang L, Andersson S, Nagy N, Kis LL, Zhang [83] Dai R, Phillips RA, Ahmed SA. Despite inhibition of
Q, et al. Disruption of the estrogen receptor beta gene in nuclear localization of NF-kappa B p65, c-Rel, and RelB,
mice causes myeloproliferative disease resembling chronic 17-beta estradiol up-regulates NF-kappa B signaling in
myeloid leukemia with lymphoid blast crisis. Proceedings mouse splenocytes: the potential role of Bcl-3. Journal of
of the National Academy of Sciences of the United States Immunology. 2007; 179(3): 1776-1783. Epub 2007/07/21.
AU

of America. 2003; 100(11): 6694-6699. [84] Dai R, Phillips RA, Karpuzoglu E, Khan D, Ahmed SA.
[68] Erlandsson MC, Jonsson CA, Islander U, Ohlsson C, Estrogen regulates transcription factors STAT-1 and NF-
Carlsten H. Oestrogen receptor specificity in oestradiol- kappaB to promote inducible nitric oxide synthase and
mediated effects on B lymphopoiesis and immunoglobulin inflammatory responses. Journal of Immunology. 2009;
production in male mice. Immunology. 2003; 108(3): 346- 183(11): 6998-7005. Epub 2009/11/06.
351. [85] Straub RH. The complex role of estrogens in inflammation.
[69] Matthews J, Wihlen B, Tujague M, Wan J, Strom A, Endocrine reviews. 2007; 28(5): 521-574. Epub 2007/07/21.
Gustafsson JA. Estrogen receptor (ER) beta modulates [86] Hammes SR, Levin ER. Extranuclear steroid receptors:
ERalpha-mediated transcriptional activation by altering the nature and actions. Endocrine Reviews. 2007; 28(7): 726-
recruitment of c-Fos and c-Jun to estrogen-responsive 741. Epub 2007/10/06.
promoters. Mol Endocrinol. 2006; 20(3): 534-543. Epub [87] Kang L, Zhang X, Xie Y, Tu Y, Wang D, Liu Z, et al. Involve-
2005/11/19. ment of estrogen receptor variant ER-alpha36, not GPR30,
[70] Barkhem T, Carlsson B, Nilsson Y, Enmark E, Gustafs- in nongenomic estrogen signaling. Mol Endocrinol. 2010;
son J, Nilsson S. Differential response of estrogen receptor 24(4): 709-21. Epub 2010/03/04.
alpha and estrogen receptor beta to partial estrogen ago- [88] Isensee J, Meoli L, Zazzu V, Nabzdyk C, Witt H, Soewarto
nists/antagonists. Mol Pharmacol. 1998; 54(1): 105-112. D, et al. Expression pattern of G protein-coupled receptor 30
Epub 1998/07/11. in LacZ reporter mice. Endocrinology. 2009; 150(4): 1722-
[71] Nilsson S, Makela S, Treuter E, Tujague M, Thomsen J, 1730. Epub 2008/12/20.
Andersson G, et al. Mechanisms of estrogen action. Physiol [89] Kassi E, Moutsatsou P. Estrogen receptor signaling and its
Rev. 2001; 81(4): 1535-1565. relationship to cytokines in systemic lupus erythematosus. J
[72] Levin ER. Plasma membrane estrogen receptors. Biomed Biotechnol. 2010; 2010: 317452. Epub 2010/07/10.
Trends Endocrinol Metab. 2009; 20(10): 477-482. Epub [90] Bruce-Keller AJ, Barger SW, Moss NI, Pham JT, Keller JN,
2009/09/29. Nath A. Pro-inflammatory and pro-oxidant properties of the
[73] Nilsson S, Gustafsson JA. Estrogen receptors: therapies tar- HIV protein Tat in a microglial cell line: attenuation by 17
geted to receptor subtypes. Clin Pharmacol Ther. 2011; beta-estradiol. J Neurochem. 2001; 78(6): 1315-1324. Epub
89(1): 44-55. Epub 2010/12/03. 2001/10/02.
[74] Bramlett KS, Wu Y, Burris TP. Ligands specify coactiva- [91] Bruce-Keller AJ, Keeling JL, Keller JN, Huang FF, Camon-
tor nuclear receptor (NR) box affinity for estrogen receptor dola S, Mattson MP. Antiinflammatory effects of estrogen on
D. Khan et al. / Estrogen and Signaling in the Cells 89

microglial activation. Endocrinology. 2000; 141(10): 3646- [108] Mori H, Sawairi M, Itoh N, Hanabayashi T, Tamaya T.
3656. Epub 2000/10/03. Effects of sex steroids on cell differentiation and interleukin-
[92] Friedrich EB, Clever YP, Wassmann S, Hess C, Nickenig 1 beta production in the human promyelocytic leukemia cell
G. 17Beta-estradiol inhibits monocyte adhesion via down- line HL-60. J Reprod Med. 1992; 37(10): 871-878.
regulation of Rac1 GTPase. J Mol Cell Cardiol. 2006; 40(1): [109] Itoh Y, Hayashi H, Miyazawa K, Kojima S, Akahoshi T,
87-95. Epub 2005/12/08. Onozaki K. 17beta-estradiol induces IL-1alpha gene expres-
[93] Kato S, Endoh H, Masuhiro Y, Kitamoto T, Uchiyama S, sion in rheumatoid fibroblast-like synovial cells through
Sasaki H, et al. Activation of the estrogen receptor through estrogen receptor alpha (ERalpha) and augmentation of
phosphorylation by mitogen-activated protein kinase. Sci- transcriptional activity of Sp1 by dissociating histone
ence. 1995; 270(5241): 1491-1494. Epub 1995/12/01. deacetylase 2 from ERalpha. Journal of Immunology. 2007;

PY
[94] Gee JM, Robertson JF, Gutteridge E, Ellis IO, Pin- 178(5): 3059-366. Epub 2007/02/22.
der SE, Rubini M, et al. Epidermal growth factor [110] Miller AP, Feng W, Xing D, Weathington NM, Blalock
receptor/HER2/insulin-like growth factor receptor sig- JE, Chen YF, et al. Estrogen modulates inflammatory
nalling and oestrogen receptor activity in clinical breast mediator expression and neutrophil chemotaxis in injured
cancer. Endocr Relat Cancer. 2005; 12 (Suppl 1): S99-S111. arteries. Circulation. 2004; 110(12): 1664-1669. Epub 2004/
Epub 2005/08/23. 09/09.
CO
[95] Ascenzi P, Bocedi A, Marino M. Structure-function rela- [111] Ritz MF, Hausmann ON. Effect of 17beta-estradiol on func-
tionship of estrogen receptor alpha and beta: impact on tional outcome, release of cytokines, astrocyte reactivity and
human health. Mol Aspects Med. 2006; 27(4): 299-402. inflammatory spreading after spinal cord injury in male rats.
Epub 2006/08/18. Brain Res. 2008; 1203: 177-188. Epub 2008/03/05.
[96] Bouman A, Heineman MJ, Faas MM. Sex hormones and the [112] Calippe B, Douin-Echinard V, Laffargue M, Laurell H,
immune response in humans. Hum Reprod Update. 2005; Rana-Poussine V, Pipy B, et al. Chronic estradiol admin-
11(4): 411-23. Epub 2005/04/09. istration in vivo promotes the proinflammatory response of
[97] Carruba G, D’Agostino P, Miele M, Calabro M, Barbera macrophages to TLR4 activation: involvement of the phos-
OR

C, Bella GD, et al. Estrogen regulates cytokine produc- phatidylinositol 3-kinase pathway. Journal of Immunology.
tion and apoptosis in PMA-differentiated, macrophage-like 2008; 180(12): 7980-7988.
U937 cells. J Cell Biochem. 2003; 90(1): 187-196. [113] Rogers A, Clowes JA, Pereda CA, Eastell R. Different
[98] Zhang X, Wang L, Zhang H, Guo D, Qiao Z, Qiao J. Estrogen effects of raloxifene and estrogen on interleukin-1beta and
inhibits lipopolysaccharide-induced tumor necrosis factor- interleukin-1 receptor antagonist production using in vitro
alpha release from murine macrophages. Methods Find Exp and ex vivo studies. Bone. 2007; 40(1): 105-110. Epub
Clin Pharmacol. 2001; 23(4): 169-173. 2006/08/29.
TH

[99] Chao TC, Van Alten PJ, Greager JA, Walter RJ. Steroid sex [114] Lewis DK, Johnson AB, Stohlgren S, Harms A, Sohrabji
hormones regulate the release of tumor necrosis factor by F. Effects of estrogen receptor agonists on regulation of the
macrophages. Cell Immunol. 1995; 160(1): 43-49. inflammatory response in astrocytes from young adult and
[100] Zuckerman SH, Ahmari SE, Bryan-Poole N, Evans GF, middle-aged female rats. J Neuroimmunol. 2008; 195(1-2):
Short L, Glasebrook AL. Estriol: a potent regulator of TNF 47-59. Epub 2008/03/11.
and IL-6 expression in a murine model of endotoxemia. [115] Rogers A, Eastell R. The effect of 17beta-estradiol on pro-
AU

Inflammation. 1996; 20(6): 581-597. Epub 1996/12/01. duction of cytokines in cultures of peripheral blood. Bone.
[101] Ikejima K, Enomoto N, Iimuro Y, Ikejima A, Fang D, Xu J, 2001; 29(1): 30-4.
et al. Estrogen increases sensitivity of hepatic Kupffer cells [116] Asai K, Hiki N, Mimura Y, Ogawa T, Unou K, Kaminishi M.
to endotoxin. Am J Physiol. 1998; 274(4 Pt 1): G669-G676. Gender differences in cytokine secretion by human periph-
Epub 1998/05/12. eral blood mononuclear cells: role of estrogen in modulating
[102] Yang L, Hu Y, Hou Y. Effects of 17beta-estradiol on the mat- LPS-induced cytokine secretion in an ex vivo septic model.
uration, nuclear factor kappa B p65 and functions of murine Shock. 2001; 16(5): 340-343. Epub 2001/11/09.
spleen CD11c-positive dendritic cells. Mol Immunol. 2006; [117] Rachon D, Mysliwska J, Suchecka-Rachon K, Wieck-
43(4): 357-366. iewicz J, Mysliwski A. Effects of oestrogen deprivation on
[103] Hildebrand F, Hubbard WJ, Choudhry MA, Thobe BM, interleukin-6 production by peripheral blood mononuclear
Pape HC, Chaudry IH. Are the protective effects of 17beta- cells of postmenopausal women. The Journal of Endocrinol-
estradiol on splenic macrophages and splenocytes after ogy. 2002; 172(2): 387-395. Epub 2002/02/09.
trauma-hemorrhage mediated via estrogen-receptor (ER)- [118] Bengtsson AK, Ryan EJ, Giordano D, Magaletti DM,
alpha or ER-beta? Journal of Leukocyte Biology. 2006; Clark EA. 17beta-estradiol (E2) modulates cytokine and
79(6): 1173-1180. Epub 2006/03/15. chemokine expression in human monocyte-derived den-
[104] Soucy G, Boivin G, Labrie F, Rivest S. Estradiol is dritic cells. Blood. 2004; 104(5): 1404-1410. Epub
required for a proper immune response to bacterial and 2004/05/15.
viral pathogens in the female brain. Journal of Immunology. [119] Huygen K, Palfliet K. Strain variation in interferon gamma
2005; 174(10): 6391-6398. production of BCG-sensitized mice challenged with PPD
[105] Dinarello CA. Interleukin-1 in the pathogenesis and treat- II. Importance of one major autosomal locus and additional
ment of inflammatory diseases. Blood. 2011; 117(14): sexual influences. Cell Immunol. 1984; 85(1): 75-81.
3720-3732. Epub 2011/02/10. [120] McMurray RW, Hoffman RW, Nelson W, Walker SE.
[106] Ruh MF, Bi Y, D’Alonzo R, Bellone CJ. Effect of estrogens Cytokine mRNA expression in the B/W mouse model of
on IL-1beta promoter activity. J Steroid Biochem Mol Biol. systemic lupus erythematosus–analyses of strain, gender,
1998; 66(4): 203-210. Epub 1998/09/23. and age effects. Clin Immunol Immunopathol. 1997; 84(3):
[107] Hu SK, Mitcho YL, Rath NC. Effect of estradiol on inter- 260-268.
leukin 1 synthesis by macrophages. Int J Immunopharmacol. [121] Satoskar A, Alexander J. Sex-determined susceptibil-
1988; 10(3): 247-52. ity and differential IFN-gamma and TNF-alpha mRNA
90 D. Khan et al. / Estrogen and Signaling in the Cells

expression in DBA/2 mice infected with Leishmania mexi- [135] Matthiesen L, Khademi M, Ekerfelt C, Berg G, Sharma
cana. Immunology. 1995; 84(1): 1-4. S, Olsson T, et al. In-situ detection of both inflammatory
[122] Karpuzoglu-Sahin E, Hissong BD, Ansar Ahmed S. and anti-inflammatory cytokines in resting peripheral blood
Interferon-gamma levels are upregulated by 17-beta- mononuclear cells during pregnancy. J Reprod Immunol.
estradiol and diethylstilbestrol. J Reprod Immunol. 2001; 2003; 58(1): 49-59. Epub 2003/03/01.
52(1-2): 113-127. [136] Blaschitz C, Raffatellu M. Th17 cytokines and the gut
[123] Fox HS, Bond BL, Parslow TG. Estrogen regulates the IFN- mucosal barrier. J Clin Immunol. 2011; 30(2): 196-203.
gamma promoter. Journal of Immunology. 1991; 146(12): Epub 2010/02/04.
4362-4367. [137] Kabir S. The role of interleukin-17 in the Helicobacter pylori
[124] Karpuzoglu E, Phillips RA, Gogal RM Jr., Ansar Ahmed S. induced infection and immunity. Helicobacter. 2011; 16(1):

PY
IFN-gamma-inducing transcription factor, T-bet is upregu- 1-8. Epub 2011/01/19.
lated by estrogen in murine splenocytes: role of IL-27 but [138] DeLyria ES, Redline RW, Blanchard TG. Vaccination of
not IL-12. Mol Immunol. 2007; 44(7): 1808-1814. Epub mice against H pylori induces a strong Th-17 response and
2006/10/19. immunity that is neutrophil dependent. Gastroenterology.
[125] Gilmore W, Weiner LP, Correale J. Effect of estradiol on 2009; 136(1): 247-256. Epub 2008/10/25.
cytokine secretion by proteolipid protein-specific T cell [139] McKinstry KK, Strutt TM, Buck A, Curtis JD, Dib-
CO
clones isolated from multiple sclerosis patients and nor- ble JP, Huston G, et al. IL-10 deficiency unleashes an
mal control subjects. Journal of Immunology. 1997; 158(1): influenza-specific Th17 response and enhances survival
446-451. against high-dose challenge. Journal of immunology. 2009;
[126] Correale J, Arias M, Gilmore W. Steroid hormone regu- 182(12): 7353-7363. Epub 2009/06/06.
lation of cytokine secretion by proteolipid protein-specific [140] Sieve AN, Meeks KD, Bodhankar S, Lee S, Kolls JK,
CD4+ T cell clones isolated from multiple sclerosis patients Simecka JW, et al. A novel IL-17-dependent mechanism
and normal control subjects. Journal of Immunology. 1998; of cross protection: respiratory infection with mycoplasma
161(7): 3365-3374. protects against a secondary listeria infection. European
OR

[127] Gourdy P, Araujo LM, Zhu R, Garmy-Susini B, Diem S, Journal of Immunology. 2009; 39(2): 426-438. Epub
Laurell H, et al. Relevance of sexual dimorphism to regula- 2009/01/31.
tory T cells: estradiol promotes IFN-gamma production by [141] Lin Y, Ritchea S, Logar A, Slight S, Messmer M, Rangel-
invariant natural killer T cells. Blood. 2005; 105(6): 2415- Moreno J, et al. Interleukin-17 is required for T helper 1 cell
2420. immunity and host resistance to the intracellular pathogen
[128] Liu Y, Guo YL, Zhou SJ, Liu F, Du FJ, Zheng XJ, et Francisella tularensis. Immunity. 2009; 31(5): 799-810.
al. CREB is a positive transcriptional regulator of gamma Epub 2009/10/27.
TH

interferon in latent but not active tuberculosis infections. [142] Sellge G, Magalhaes JG, Konradt C, Fritz JH, Salgado-
Clin Vaccine Immunol. 2010; 17(9): 1377-1380. Epub 2010/ Pabon W, Eberl G, et al. Th17 cells are the dominant T
08/06. cell subtype primed by Shigella flexneri mediating pro-
[129] Pettersson A, Ciumas C, Chirsky V, Link H, Huang YM, tective immunity. Journal of Immunology. 2010; 184(4):
Xiao BG. Dendritic cells exposed to estrogen in vitro 2076-2085. Epub 2010/01/22.
exhibit therapeutic effects in ongoing experimental aller- [143] Lin JS, Kummer LW, Szaba FM, Smiley ST. IL-17 con-
AU

gic encephalomyelitis. J Neuroimmunol. 2004; 156(1-2): tributes to cell-mediated defense against pulmonary Yersinia
58-65. Epub 2004/10/07. pestis infection. Journal of Immunology. 2011; 186(3):
[130] Matalka KZ. The effect of estradiol, but not progesterone, 1675-1684. Epub 2010/12/22.
on the production of cytokines in stimulated whole blood, [144] Khan D, Dai R, Karpuzoglu E, Ahmed SA. Estrogen
is concentration-dependent. Neuro Endocrinol Lett. 2003; increases, whereas IL-27 and IFN-gamma decrease, spleno-
24(3-4): 185-191. Epub 2003/10/03. cyte IL-17 production in WT mice. European journal of
[131] de Oliveira AP, Domingos HV, Cavriani G, Damazo AS, Immunology. 2010; 40(9): 2549-2556. Epub 2010/07/14.
Dos Santos Franco AL, Oliani SM, et al. Cellular recruit- [145] LeRoith T, Ansar Ahmed S. Regulatory T Cells and Viral
ment and cytokine generation in a rat model of allergic lung Disease. In: Khatami M, editor. Inflammatory Diseases/
inflammation are differentially modulated by progesterone Book 1. ISBN 978-953-307-762-8. Chapter 2. Rijeka, Croa-
and estradiol. Am J Physiol Cell Physiol. 2007; 293(3): tia: INTech-Open Access; 2011. pp. 121-144.
C1120-C1128. Epub 2007/07/20. [146] Bachy V, Williams DJ, Ibrahim MA. Altered dendritic cell
[132] Lambert KC, Curran EM, Judy BM, Milligan GN, Lubahn function in normal pregnancy. J Reprod Immunol. 2008;
DB, Estes DM. Estrogen receptor alpha (ERalpha) defi- 78(1): 11-21. Epub 2007/11/17.
ciency in macrophages results in increased stimulation of [147] Sysa P, Potter JJ, Liu X, Mezey E. Transforming growth
CD4+T cells while 17beta-estradiol acts through ERalpha to factor-beta1 up-regulation of human alpha(1)(I) collagen is
increase IL-4 and GATA-3 expression in CD4+T cells inde- mediated by Sp1 and Smad2 transacting factors. DNA Cell
pendent of antigen presentation. Journal of Immunology. Biol. 2009; 28(9): 425-434. Epub 2009/06/30.
2005; 175(9): 5716-5723. Epub 2005/10/21. [148] Dhandapani KM, Wade FM, Mahesh VB, Brann DW.
[133] Hoch RV, Thompson DA, Baker RJ, Weigel RJ. GATA- Astrocyte-derived transforming growth factor-{beta} medi-
3 is expressed in association with estrogen receptor in ates the neuroprotective effects of 17{beta}-estradiol:
breast cancer. Int J Cancer. 1999; 84(2): 122-128. Epub involvement of nonclassical genomic signaling pathways.
1999/03/30. Endocrinology. 2005; 146(6): 2749-2759. Epub 2005/
[134] Marzi M, Vigano A, Trabattoni D, Villa ML, Salvaggio 03/05.
A, Clerici E, et al. Characterization of type 1 and type 2 [149] Mercier I, Colombo F, Mader S, Calderone A. Ovarian
cytokine production profile in physiologic and pathologic hormones induce TGF-beta(3) and fibronectin mRNAs but
human pregnancy. Clin Exp Immunol. 1996; 106(1): 127- exhibit a disparate action on cardiac fibroblast proliferation.
133. Epub 1996/10/01. Cardiovasc Res. 2002; 53(3): 728-739. Epub 2002/02/28.
D. Khan et al. / Estrogen and Signaling in the Cells 91

[150] Alvarez Y, Municio C, Alonso S, Sanchez Crespo M, pregnancy-specific serum factors. J Neuroimmunol. 2011;
Fernandez N. The induction of IL-10 by zymosan in den- 230(1-2): 105-113. Epub 2010/10/19.
dritic cells depends on CREB activation by the coactivators [165] Papenfuss TL, Powell ND, McClain MA, Bedarf A, Singh
CREB-binding protein and TORC2 and autocrine PGE2. A, Gienapp IE, et al. Estriol generates tolerogenic dendritic
Journal of Immunology. 2009; 183(2): 1471-1479. Epub cells in vivo that protect against autoimmunity. J Immunol.
2009/07/01. 2011; 186(6): 3346-3355. Epub 2011/02/15.
[151] Kube D, Platzer C, von Knethen A, Straub H, Bohlen H, [166] Lelu K, Laffont S, Delpy L, Paulet PE, Perinat T, Tschanz
Hafner M, et al. Isolation of the human interleukin 10 pro- SA, et al. Estrogen receptor alpha signaling in T lympho-
moter. Characterization of the promoter activity in Burkitt’s cytes is required for estradiol-mediated inhibition of Th1
lymphoma cell lines. Cytokine. 1995; 7(1): 1-7. Epub and Th17 cell differentiation and protection against exper-

PY
1995/01/01. imental autoimmune encephalomyelitis. J Immunol. 2011;
[152] Luo CY, Wang L, Sun C, Li DJ. Estrogen enhances the func- 187(5): 2386-2393. Epub 2011/08/04.
tions of CD4(+)CD25(+)Foxp3(+) regulatory T cells that [167] Lelu K, Delpy L, Robert V, Foulon E, Laffont S, Pelletier
suppress osteoclast differentiation and bone resorption in L, et al. Endogenous estrogens, through estrogen recep-
vitro. Cellular & molecular immunology. 2011; 8(1): 50-58. tor alpha, constrain autoimmune inflammation in female
Epub 2011/01/05. mice by limiting CD4+ T-cell homing into the CNS. Eur
CO
[153] Polanczyk MJ, Hopke C, Huan J, Vandenbark AA, Offner J Immunol. 2010; 40(12): 3489-3498. Epub 2010/11/26.
H. Enhanced FoxP3 expression and Treg cell function in [168] Confavreux C, Hutchinson M, Hours MM, Cortinovis-
pregnant and estrogen-treated mice. J Neuroimmunol. 2005; Tourniaire P, Moreau T. Rate of pregnancy-related relapse in
170(1-2): 85-92. Epub 2005/10/29. multiple sclerosis. Pregnancy in Multiple Sclerosis Group.
[154] Polanczyk MJ, Hopke C, Vandenbark AA, Offner H. Treg The New England Journal of Medicine. 1998; 339(5): 285-
suppressive activity involves estrogen-dependent expression 291. Epub 1998/07/31.
of programmed death-1 (PD-1). International Immunology. [169] Vukusic S, Hutchinson M, Hours M, Moreau T, Cortinovis-
2007; 19(3): 337-343. Epub 2007/02/03. Tourniaire P, Adeleine P, et al. Pregnancy and multiple
OR

[155] Offner H, Vandenbark AA. Congruent effects of estrogen sclerosis (the PRIMS study): clinical predictors of post-
and T-cell receptor peptide therapy on regulatory T cells in partum relapse. Brain. 2004; 127(Pt 6): 1353-1360. Epub
EAE and MS. International Reviews of Immunology. 2005; 2004/05/08.
24(5-6): 447-477. Epub 2005/12/02. [170] Sicotte NL, Liva SM, Klutch R, Pfeiffer P, Bouvier S, Odesa
[156] Tai P, Wang J, Jin H, Song X, Yan J, Kang Y, et al. Induc- S, et al. Treatment of multiple sclerosis with the pregnancy
tion of regulatory T cells by physiological level estrogen. hormone estriol. Ann Neurol. 2002; 52(4): 421-428. Epub
Journal of cellular physiology. 2008; 214(2): 456-464. Epub 2002/09/27.
TH

2007/07/27. [171] Caulin-Glaser T, Watson CA, Pardi R, Bender JR. Effects


[157] Offner H, Polanczyk M. A potential role for estrogen in of 17beta-estradiol on cytokine-induced endothelial cell
experimental autoimmune encephalomyelitis and multiple adhesion molecule expression. The Journal of clinical inves-
sclerosis. Annals of the New York Academy of Sciences. tigation. 1996; 98(1): 36-42. Epub 1996/07/01.
2006; 1089: 343-372. [172] Lockshin MD. Sex ratio and rheumatic disease: excerpts
[158] Gold SM, Voskuhl RR. Estrogen and testosterone therapies from an Institute of Medicine report. Lupus. 2002; 11(10):
AU

in multiple sclerosis. Prog Brain Res. 2009; 175: 239-251. 662-666. Epub 2002/11/05.
Epub 2009/08/08. [173] Beeson PB. Age and sex associations of 40 autoimmune
[159] Polanczyk M, Zamora A, Subramanian S, Matejuk A, Hess diseases. Am J Med. 1994; 96(5): 457-462.
DL, Blankenhorn EP, et al. The protective effect of 17beta- [174] Joseph FG, Scolding NJ. Neurolupus. Pract Neurol. 2010;
estradiol on experimental autoimmune encephalomyelitis 10(1): 4-15. Epub 2010/02/05.
is mediated through estrogen receptor-alpha. Am J Pathol. [175] Boumpas DT, Fessler BJ, Austin HA 3rd, Balow JE,
2003; 163(4): 1599-1605. Klippel JH, Lockshin MD. Systemic lupus erythematosus:
[160] Morales LB, Loo KK, Liu HB, Peterson C, Tiwari-Woodruff emerging concepts. Part 2: Dermatologic and joint dis-
S, Voskuhl RR. Treatment with an estrogen receptor ease, the antiphospholipid antibody syndrome, pregnancy
alpha ligand is neuroprotective in experimental autoimmune and hormonal therapy, morbidity and mortality, and patho-
encephalomyelitis. J Neurosci. 2006; 26(25): 6823-6833. genesis. Ann Intern Med. 1995; 123(1): 42-53. Epub 1995/
[161] Xiao BG, Liu X, Link H. Antigen-specific T cell functions 07/01.
are suppressed over the estrogen-dendritic cell-indoleamine [176] Lourenco EV, La Cava A. Cytokines in systemic lupus
2,3-dioxygenase axis. Steroids. 2004; 69(10): 653-659. erythematosus. Curr Mol Med. 2009; 9(3): 242-254. Epub
Epub 2004/10/07. 2009/04/10.
[162] Liu HY, Buenafe AC, Matejuk A, Ito A, Zamora A, [177] Tenbrock K, Juang YT, Kyttaris VC, Tsokos GC. Altered
Dwyer J, et al. Estrogen inhibition of EAE involves effects signal transduction in SLE T cells. Rheumatology (Oxford).
on dendritic cell function. J Neurosci Res. 2002; 70(2): 2007; 46(10): 1525-1530. Epub 2007/06/26.
238-248. [178] Zhang J, Jacobi AM, Wang T, Berlin R, Volpe BT, Diamond
[163] Matejuk A, Dwyer J, Hopke C, Vandenbark AA, Offner B. Polyreactive autoantibodies in systemic lupus erythe-
H. 17Beta-estradiol treatment profoundly down-regulates matosus have pathogenic potential. J Autoimmun. 2009;
gene expression in spinal cord tissue in mice protected 33(3-4): 270-274. Epub 2009/04/29.
from experimental autoimmune encephalomyelitis. Arch [179] Yacoub Wasef SZ. Gender differences in systemic lupus
Immunol Ther Exp (Warsz). 2003; 51(3): 185-193. Epub erythematosus. Gend Med. 2004; 1(1): 12-17. Epub
2003/08/05. 2005/08/24.
[164] Gatson NN, Williams JL, Powell ND, McClain MA, Hennon [180] Talal N. Natural history of murine lupus. Modulation by sex
TR, Robbins PD, et al. Induction of pregnancy during estab- hormones. Arthritis Rheum. 1978; 21(5 Suppl): S58-S63.
lished EAE halts progression of CNS autoimmune injury via Epub 1978/06/01.
92 D. Khan et al. / Estrogen and Signaling in the Cells

[181] Sekigawa I, Fujishiro M, Yamaguchi A, Kawasaki M, Inui [197] Greenstein B, Roa R, Dhaher Y, Nunn E, Greenstein A,
A, Nozawa K, et al. A new hypothesis of the possible Khamashta M, et al. Estrogen and progesterone receptors
mechanisms of gender differences in systemic lupus ery- in murine models of systemic lupus erythematosus. Int
thematosus. Clinical and experimental rheumatology. 2010; Immunopharmacol. 2001; 1(6): 1025-1035.
28(3): 419-423. Epub 2010/05/13. [198] Inui A, Ogasawara H, Naito T, Sekigawa I, Takasaki Y,
[182] Walker SE. Estrogen and autoimmune disease. Clin Rev Hayashida Y, et al. Estrogen receptor expression by periph-
Allergy Immunol. 2011; 40(1): 60-65. Epub 2010/02/26. eral blood mononuclear cells of patients with systemic lupus
[183] Holroyd CR, Edwards CJ. The effects of hormone replace- erythematosus. Clin Rheumatol. 2007; 26(10): 1675-1678.
ment therapy on autoimmune disease: rheumatoid arthritis Epub 2007/09/18.
and systemic lupus erythematosus. Climacteric. 2009; [199] Feng F, Nyland J, Banyai M, Tatum A, Silverstone AE,

PY
12(5): 378-386. Epub 2009/07/11. Gavalchin J. The induction of the lupus phenotype by estro-
[184] Gompel A, Piette JC. Is there a place for postmenopausal gen is via an estrogen receptor-alpha-dependent pathway.
hormone therapy use in women with lupus? Panminerva Clin Immunol. 2010; 134(2): 226-236. Epub 2009/11/21.
Med. 2008; 50(3): 247-254. Epub 2008/10/18. [200] Bynote KK, Hackenberg JM, Korach KS, Lubahn DB, Lane
[185] Kanda N, Tamaki K. Estrogen enhances immunoglobulin PH, Gould KA. Estrogen receptor-alpha deficiency attenu-
production by human PBMCs. J Allergy Clin Immunol. ates autoimmune disease in (NZB × NZW)F1 mice. Genes
CO
1999; 103(2 Pt 1): 282-288. and Immunity. 2008; 9(2): 137-152. Epub 2008/01/18.
[186] Kanda N, Tsuchida T, Tamaki K. Estrogen enhancement [201] Li J, McMurray RW. Effects of estrogen receptor subtype-
of anti-double-stranded DNA antibody and immunoglob- selective agonists on autoimmune disease in lupus-prone
ulin G production in peripheral blood mononuclear cells NZB/NZW F1 mouse model. Clin Immunol. 2007; 123(2):
from patients with systemic lupus erythematosus. Arthritis 219-226. Epub 2007/03/06.
Rheum. 1999; 42(2): 328-337. [202] Lee YJ, Shin KS, Kang SW, Lee CK, Yoo B, Cha HS,
[187] Cohen-Solal JF, Jeganathan V, Hill L, Kawabata D, et al. Association of the oestrogen receptor alpha gene
Rodriguez-Pinto D, Grimaldi C, et al. Hormonal regulation polymorphisms with disease onset in systemic lupus erythe-
OR

of B-cell function and systemic lupus erythematosus. Lupus. matosus. Ann Rheum Dis. 2004; 63(10): 1244-1249. Epub
2008; 17(6): 528-532. Epub 2008/06/10. 2004/09/14.
[188] Grimaldi CM, Jeganathan V, Diamond B. Hormonal reg- [203] Johansson M, Arlestig L, Moller B, Smedby T, Rantapaa-
ulation of B cell development: 17 beta-estradiol impairs Dahlqvist S. Oestrogen receptor {alpha} gene polymor-
negative selection of high-affinity DNA-reactive B cells phisms in systemic lupus erythematosus. Ann Rheum Dis.
at more than one developmental checkpoint. Journal of 2005; 64(11): 1611-1617. Epub 2005/04/09.
Immunology. 2006; 176(5): 2703-2710. Epub 2006/02/24. [204] Kawasaki M, Sekigawa I, Nozawa K, Kaneko H, Takasaki Y,
TH

[189] Ahmed SA, Aufdemorte TB, Chen JR, Montoya AI, Olive D, Takamori K, et al. Changes in the gene expression of periph-
Talal N. Estrogen induces the development of autoantibodies eral blood mononuclear cells during the menstrual cycle of
and promotes salivary gland lymphoid infiltrates in normal females is associated with a gender bias in the incidence of
mice. J Autoimmun. 1989; 2(4): 543-552. Epub 1989/08/01. systemic lupus erythematosus. Clinical and Experimental
[190] Franklin RD, Kutteh WH. Characterization of immunoglob- Rheumatology. 2009; 27(2): 260-266. Epub 2009/05/29.
ulins and cytokines in human cervical mucus: influence of [205] Sedy JR, Gavrieli M, Potter KG, Hurchla MA, Lindsley
AU

exogenous and endogenous hormones. Journal of Reproduc- RC, Hildner K, et al. B and T lymphocyte attenuator regu-
tive Immunology. 1999; 42(2): 93-106. Epub 1999/04/30. lates T cell activation through interaction with herpesvirus
[191] Verthelyi D, Ahmed SA. 17 beta-estradiol, but not entry mediator. Nat Immunol. 2005; 6(1): 90-98. Epub
5 alpha-dihydrotestosterone, augments antibodies to 2004/11/30.
double-stranded deoxyribonucleic acid in nonautoimmune [206] Cai G, Freeman GJ. The CD160, BTLA, LIGHT/HVEM
C57BL/6J mice. Endocrinology. 1994; 135(6): 2615-2622. pathway: a bidirectional switch regulating T-cell activation.
Epub 1994/12/01. Immunol Rev. 2009; 229(1): 244-258. Epub 2009/05/12.
[192] Venkatesh J, Yoshifuji H, Kawabata D, Chinnasamy P, [207] Koga M, Kawasaki A, Ito I, Furuya T, Ohashi J, Kyo-
Stanevsky A, Grimaldi CM, et al. Antigen is required for goku C, et al. Cumulative association of eight susceptibility
maturation and activation of pathogenic anti-DNA antibod- genes with systemic lupus erythematosus in a Japanese
ies and systemic inflammation. Journal of Immunology. female population. J Hum Genet. 2011; 56(7): 503-507.
2011; 186(9): 5304-5312. Epub 2011/03/30. Epub 2011/05/13.
[193] Rider V, Abdou NI. Gender differences in autoimmunity: [208] Salloum R, Niewold TB. Interferon regulatory factors in
molecular basis for estrogen effects in systemic lupus human lupus pathogenesis. Translational Research: The
erythematosus. Int Immunopharmacol. 2001; 1(6): 1009- Journal of Laboratory and Clinical Medicine. 2011; 157(6):
1024. 326-331. Epub 2011/05/18.
[194] Durali D, de Goer de Herve MG, Giron-Michel J, Azzarone [209] Tada Y, Kondo S, Aoki S, Koarada S, Inoue H, Suematsu R,
B, Delfraissy JF, Taoufik Y. In human B cells, IL-12 triggers et al. Interferon regulatory factor 5 is critical for the devel-
a cascade of molecular events similar to Th1 commitment. opment of lupus in MRL/lpr mice. Arthritis Rheum. 2011;
Blood. 2003; 102(12): 4084-4089. 63(3): 738-748. Epub 2011/02/10.
[195] Sekigawa I, Naito T, Hira K, Mitsuishi K, Ogasawara H, [210] Shen H, Panchanathan R, Rajavelu P, Duan X, Gould KA,
Hashimoto H, et al. Possible mechanisms of gender bias in Choubey D. Gender-dependent expression of murine Irf5
SLE: a new hypothesis involving a comparison of SLE with gene: implications for sex bias in autoimmunity. Journal
atopy. Lupus. 2004; 13(4): 217-222. Epub 2004/06/05. of Molecular Cell Biology. 2010; 2(5): 284-290. Epub
[196] Perl A, Nagy G, Koncz A, Gergely P, Fernandez D, Doherty 2010/08/31.
E, et al. Molecular mimicry and immunomodulation by [211] Dai R, Ahmed SA. MicroRNA, a new paradigm
the HRES-1 endogenous retrovirus in SLE. Autoimmunity. for understanding immunoregulation, inflammation, and
2008; 41(4): 287-297. Epub 2008/04/25. autoimmune diseases. Translational Research: The Journal
D. Khan et al. / Estrogen and Signaling in the Cells 93

of Laboratory and Clinical Medicine. 2011; 157(4): 163- during pregnancy and labor. Proceedings of the National
179. Epub 2011/03/23. Academy of Sciences of the United States of America. 2010;
[212] Dai R, Phillips RA, Zhang Y, Khan D, Crasta O, Ahmed SA. 107(48): 20828-20833. Epub 2010/11/17.
Suppression of LPS-induced Interferon-gamma and nitric [225] Xia H, Ng SS, Jiang S, Cheung WK, Sze J, Bian XW,
oxide in splenic lymphocytes by select estrogen-regulated et al. miR-200a-mediated downregulation of ZEB2 and
microRNAs: a novel mechanism of immune modulation. CTNNB1 differentially inhibits nasopharyngeal carcinoma
Blood. 2008; 112(12): 4591-4597. Epub 2008/09/16. cell growth, migration and invasion. Biochem Biophys Res
[213] Bartel DP. MicroRNAs: target recognition and regulatory Commun. 2010; 391(1): 535-541. Epub 2009/11/26.
functions. Cell. 2009; 136(2): 215-233. Epub 2009/01/27. [226] Chan YC, Khanna S, Roy S, Sen CK. miR-200b targets Ets-
[214] Zhou X, Jeker LT, Fife BT, Zhu S, Anderson MS, McManus 1 and is down-regulated by hypoxia to induce angiogenic

PY
MT, et al. Selective miRNA disruption in T reg cells leads response of endothelial cells. J Biol Chem. 2010; 286(3):
to uncontrolled autoimmunity. J Exp Med. 2008; 205(9): 2047-2056. Epub 2010/11/18.
1983-1991. Epub 2008/08/30. [227] Oh HK, Tan AL, Das K, Ooi CH, Deng NT, Tan IB, et al.
[215] Pauley KM, Cha S, Chan EK. MicroRNA in autoimmu- Genomic loss of miR-486 regulates tumor progression and
nity and autoimmune diseases. J Autoimmun. 2009; 32(3-4): the OLFM4 antiapoptotic factor in gastric cancer. Clin Can-
189-194. Epub 2009/03/24. cer Res. 2011; 17(9): 2657-2667. Epub 2011/03/19.
CO
[216] Cox MB, Cairns MJ, Gandhi KS, Carroll AP, Moscovis S, [228] Lashine YA, Seoudi AM, Salah S, Abdelaziz AI. Expression
Stewart GJ, et al. MicroRNAs miR-17 and miR-20a inhibit signature of microRNA-181-a reveals its crucial role in the
T cell activation genes and are under-expressed in MS whole pathogenesis of paediatric systemic lupus erythematosus.
blood. PloS One. 2010; 5(8): e12132. Epub 2010/08/17. Clinical and experimental rheumatology. 2011; 29(2): 351-
[217] Lindberg RL, Hoffmann F, Mehling M, Kuhle J, Kappos 357. Epub 2011/03/10.
L. Altered expression of miR-17-5p in CD4+ lymphocytes [229] Zhao S, Wang Y, Liang Y, Zhao M, Long H, Ding S, et al.
of relapsing-remitting multiple sclerosis patients. Euro- MicroRNA-126 regulates DNA methylation in CD4+ T cells
pean Journal of Immunology. 2010; 40(3): 888-898. Epub and contributes to systemic lupus erythematosus by target-
OR

2010/02/12. ing DNA methyltransferase 1. Arthritis Rheum. 2011; 63(5):


[218] Du C, Liu C, Kang J, Zhao G, Ye Z, Huang S, et al. 1376-1386. Epub 2011/05/04.
MicroRNA miR-326 regulates TH-17 differentiation and [230] Hai-yan W, Yang L, Mei-hong C, Hui Z. Expression of
is associated with the pathogenesis of multiple sclero- MicroRNA-146a in peripheral blood mononuclear cells in
sis. Nature Immunology. 2009; 10(12): 1252-1259. Epub patients with systemic lupus Erythematosus. Zhongguo Yi
2009/10/20. Xue Ke Xue Yuan Xue Bao. 2011; 33(2): 185-188. Epub
[219] Keller A, Leidinger P, Lange J, Borries A, Schroers H, 2011/05/03.
TH

Scheffler M, et al. Multiple sclerosis: microRNA expression [231] Zhao X, Tang Y, Qu B, Cui H, Wang S, Wang L,
profiles accurately differentiate patients with relapsing- et al. MicroRNA-125a contributes to elevated inflammatory
remitting disease from healthy controls. PloS One. 2009; chemokine RANTES levels via targeting KLF13 in sys-
4(10): e7440. Epub 2009/10/14. temic lupus erythematosus. Arthritis Rheum. 2010; 62(11):
[220] Otaegui D, Baranzini SE, Armananzas R, Calvo B, Munoz- 3425-3435. Epub 2010/07/01.
Culla M, Khankhanian P, et al. Differential micro RNA [232] Tang Y, Luo X, Cui H, Ni X, Yuan M, Guo Y, et al.
AU

expression in PBMC from multiple sclerosis patients. PloS MicroRNA-146A contributes to abnormal activation of the
One. 2009; 4(7): e6309. Epub 2009/07/21. type I interferon pathway in human lupus by targeting the
[221] Junker A, Krumbholz M, Eisele S, Mohan H, Augstein F, key signaling proteins. Arthritis Rheum. 2009; 60(4): 1065-
Bittner R, et al. MicroRNA profiling of multiple sclerosis 1075. Epub 2009/04/01.
lesions identifies modulators of the regulatory protein CD47. [233] Stagakis E, Bertsias G, Verginis P, Nakou M, Hatziapos-
Brain. 2009; 132(Pt 12): 3342-3352. Epub 2009/12/03. tolou M, Kritikos H, et al. Identification of novel microRNA
[222] Yang B, Guo H, Zhang Y, Chen L, Ying D, Dong S. signatures linked to human lupus disease activity and patho-
MicroRNA-145 regulates chondrogenic differentiation of genesis: miR-21 regulates aberrant T cell responses through
mesenchymal stem cells by targeting Sox9. PloS One. 2011; regulation of PDCD4 expression. Ann Rheum Dis. 2011;
6(7): e21679. Epub 2011/07/30. 70(8): 1496-506. Epub 2011/05/24.
[223] Bussolati B, Moggio A, Collino F, Aghemo G, D’Armento [234] Dai R, Zhang Y, Khan D, Heid B, Caudell D, Crasta O,
G, Grange C, et al. Hypoxia modulates the undifferentiated et al. Identification of a common lupus disease-associated
phenotype of human renal inner medullary CD133+ progen- microRNA expression pattern in three different murine
itors through Oct4/miR-145 balance. Am J Physiol Renal models of lupus. PloS One. 2010; 5(12): e14302. Epub
Physiol. 2011. Epub 2011/09/09. 2010/12/21.
[224] Renthal NE, Chen CC, Williams KC, Gerard RD, Prange- [235] Ansar Ahmed S, Penhale WJ, Talal N. Sex hormones,
Kiel J, Mendelson CR. miR-200 family and targets, ZEB1 immune responses, and autoimmune diseases. Mechanisms
and ZEB2, modulate uterine quiescence and contractility of sex hormone action. Am J Pathol. 1985; 121(3): 531-551.

View publication stats

You might also like