0% found this document useful (0 votes)
14 views14 pages

Fonc 10 587386

The document discusses the role of Estrogen Receptor Beta (ERβ) as a potential tumor suppressor in various cancers, particularly breast cancer. High expression levels of the full-length ERβ1 isoform are associated with improved survival outcomes, while lower levels are found in aggressive cancers. The review emphasizes the need for selective ERβ agonists to exploit its therapeutic potential while minimizing side effects compared to traditional chemotherapy.

Uploaded by

Yuling Xie
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
14 views14 pages

Fonc 10 587386

The document discusses the role of Estrogen Receptor Beta (ERβ) as a potential tumor suppressor in various cancers, particularly breast cancer. High expression levels of the full-length ERβ1 isoform are associated with improved survival outcomes, while lower levels are found in aggressive cancers. The review emphasizes the need for selective ERβ agonists to exploit its therapeutic potential while minimizing side effects compared to traditional chemotherapy.

Uploaded by

Yuling Xie
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 14

REVIEW

published: 23 October 2020


doi: 10.3389/fonc.2020.587386

Estrogen Receptor Beta (ERβ): A


Ligand Activated Tumor Suppressor
Rahul Mal 1 , Alexa Magner 1 , Joel David 1 , Jharna Datta 1 , Meghna Vallabhaneni 1 ,
Mahmoud Kassem 1,2 , Jasmine Manouchehri 1 , Natalie Willingham 1 , Daniel Stover 1,2 ,
Jeffery Vandeusen 1,2 , Sagar Sardesai 1,2 , Nicole Williams 1,2 , Robert Wesolowski 1,2 ,
Maryam Lustberg 1,2 , Ramesh K. Ganju 1 , Bhuvaneswari Ramaswamy 1,2 and
Mathew A. Cherian 1,2*
1
Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States, 2 Stefanie Spielman
Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States

Estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ) belong to a superfamily
of nuclear receptors called steroid hormone receptors, which, upon binding ligand,
dimerize and translocate to the nucleus where they activate or repress the transcription
of a large number of genes, thus modulating critical physiologic processes. ERβ has
multiple isoforms that show differing association with prognosis. Expression levels of the
full length ERβ1 isoform are often lower in aggressive cancers as compared to normal
Edited by: tissue. High ERβ1 expression is associated with improved overall survival in women with
Shilpa S. Dhar, breast cancer. The promise of ERβ activation, as a potential targeted therapy, is based
University of Texas MD Anderson
Cancer Center, United States on concurrent activation of multiple tumor suppressor pathways with few side effects
Reviewed by: compared to chemotherapy. Thus, ERβ is a nuclear receptor with broad-spectrum tumor
John Hawse, suppressor activity, which could serve as a potential treatment target in a variety of human
Mayo Clinic, United States
Abdul Malik Tyagi,
cancers including breast cancer. Further development of highly selective agonists that
Emory University, United States lack ERα agonist activity, will be necessary to fully harness the potential of ERβ.
*Correspondence:
Keywords: ESR1, ESR2, ERα, ERβ, breast cancer
Mathew A. Cherian
[email protected]

INTRODUCTION
Specialty section:
This article was submitted to
In 2020, 9–10 million cancer deaths are projected to occur worldwide (WHO). Despite tremendous
Molecular and Cellular Oncology,
a section of the journal
progress in unbiased, high throughput analysis of genomic alterations in cancer, the identification
Frontiers in Oncology of key drivers that can be successfully targeted for therapy remains a challenge. The continued
dissection of signaling pathways in breast cancer, using well-designed pre-clinical experiments is
Received: 27 July 2020
Accepted: 15 September 2020
essential for the development of effective new therapies.
Published: 23 October 2020 Estrogens play a key role in cell growth and differentiation of primary and secondary
reproductive organs (1). The actions of estrogens, including those of 17β estradiol, which accounts
Citation:
Mal R, Magner A, David J, Datta J, for the majority of biological estrogenic activity in human plasma, are mediated by estrogen
Vallabhaneni M, Kassem M, receptors. These include the transcriptionally active estrogen receptors, including ERα and ERβ,
Manouchehri J, Willingham N, and membrane localized estrogen receptors, namely the G protein coupled estrogen receptor
Stover D, Vandeusen J, Sardesai S, (GPR30) and palmitoylated forms of ERα and ERβ that localize to the plasma membrane. Estradiol
Williams N, Wesolowski R, mediates long-term effects on cellular function through the transcriptional effects of ERα and
Lustberg M, Ganju RK,
ERβ, while the membrane localized estrogen receptors mediate rapid signaling effects. Both ERα
Ramaswamy B and Cherian MA
(2020) Estrogen Receptor Beta (ERβ):
and ERβ belong to the superfamily of nuclear receptors called steroid hormone receptors, which
A Ligand Activated Tumor Suppressor. upon binding ligand dimerize, dissociate from the molecular chaperone HSP90, bind to DNA,
Front. Oncol. 10:587386. either directly or through interaction with other transcription factors, and activate or repress the
doi: 10.3389/fonc.2020.587386 transcription of a large number of genes, thus modulating critical physiologic processes (2–6).

Frontiers in Oncology | www.frontiersin.org 1 October 2020 | Volume 10 | Article 587386


Mal et al. ERβ: A Ligand Activated Tumor Suppressor

NATURAL ERβ LIGANDS TABLE 1 | Structure of major the estrogen receptor ligands and their binding
affinity for ERα and Erβ.
As with ERα, estrogenic compounds including estradiol, estrone,
Compound Structure Binding affinity (Ki, nM)
and estriol activate ERβ. Relative to ERα, ERβ binds estriol
and ring B unsaturated estrogens with higher affinity, while the ERα ERβ
reverse is true of 17β-estradiol and estrone (7–10). On the other
Estradiol 0.115 0.15
hand, the dihydrotestosterone metabolites 5-androstenediol and
(0.04–0.24) (0.10–2.08)
3β androstanediol are relatively selective (3-fold) for ERβ
over ERα (11). The structures of these natural ligands and
their binding affinities to ERα and ERβ have been shown in
Table 1. These testosterone derived ligands of ERβ may be Estrone 0.445 1.75
(0.3–1.01) (0.35–9.24)
unique in their ability to recruit the transcriptional repressor
CtBP (C-terminal Binding Protein) to promoters that include
activator protein 1 (AP-1) binding sites, thus suppressing
AP-1 mediated transcriptional up-regulation in microglia and Estriol 0.45 0.7
(0.35–1.4) (0.63–0.7)
astrocytes and downregulating inflammatory responses in the
CNS (12). It also follows that a reduction in ADIOL levels
may promote an inflammatory response. Furthermore, these
effects are induced by Androstanediol (ADIOL) but not by 17β- 5-androsten-3β, 3.6 0.9
Estradiol, suggesting that certain downstream effects may be 17β-diol
ligand specific. This suggests that, similar to the case with ERα,
ligands are best referred to as modulators rather than pure
Genistein 2.6–126 0.3–12.8
agonists or antagonists given that each ligand may induce a
different output from the receptor depending on which binding
partners are recruited.
Daidzein 2 85.3

ERβ: GENETIC LOCUS, DOMAIN


STRUCTURE, AND ISOFORMS LY500307 2.68 0.19

ESR2 (Estrogen Receptor 2), the gene that expresses ERβ, maps
to 14q22-24, a distinct locus from that of ESR1 (Estrogen
Receptor 1), the gene that expresses ERα. ESR2 spans 254 kb
and contains eight encoding exons (13). The tertiary structure
of ERα and ERβ includes an N terminal activation function-1
All structures were obtained from PubChem: https://pubchem.ncbi.nlm.nih.gov/.
(AF1) or A/B domain that mediates weak ligand independent
transcriptional activity; a C domain or DNA binding domain
that binds to a cognate DNA binding element called an estrogen
response element with high affinity, and also incorporates a weak
dimerization capacity; a flexible D domain or hinge domain Binding of anti-estrogens results in the formation of perinuclear
that includes a nuclear localization signal motif as well as a clusters of the complex of estrogen receptor and ligand, while
weak dimerization interface; the E domain or ligand binding that of agonist results in persistent nuclear localization (6).
domain that includes a ligand binding pocket and AF2 function Binding of ligands also results in association of a fraction of
as well as a dimerization interface and activates transcription receptors with the small molecular chaperone HSP27, receptor
in response to ligand binding; and a C-terminal F domain that acylation, augmented interaction with caveolin and membrane
negatively regulates ligand dependent dimerization (Figures 1, localization (14).
2). Binding of ligand to the E/AF2/ligand-binding domain results Domains of ERα and ERβ, protein domain function, and
in homo and heterodimerization with ERα and with other sequence identity are depicted in Figures 1, 2. There are multiple
ERβ isoforms and dissociation from the chaperones HSP70 and splice isoforms of ERβ (Figure 1). However, only the 530
HSP90 as well as cytoplasmic relocalization of HSP90 (2–6). amino acid full-length isoform possesses a complete ligand-
binding pocket and is sensitive to activation by estradiol and
Abbreviations: ADIOL, Androstanediol; AF1, Activation Function-1; CCND1, other ligands. The different isoforms of ERβ have differing
cell division protein cyclin D1; CtBP, C-terminal Binding Protein; EGFR, associations with prognosis. For example, ERβ1 tissue expression
epidermal growth factor receptor; ERβ, Estrogen Receptor β; ERα, Estrogen levels are considered to be lower in aggressive cancers (15–19).
Receptor α; EREs, estrogen response elements; E6AP, E6 Associated Protein; Interestingly, high ERβ1 expression is associated with a higher
MAPK, Mitogen Activated Protein Kinases; GSK, Glycogen Synthase Kinase;
HER2, Human Epidermal Growth Factor 2; hMSH2, Human MutS homologue-
overall survival in women with breast cancer (20, 21). In contrast,
2; HSP90, heat shock protein-90; KO, knockout; PARP, poly (ADP-ribose) ERβ2 over-expression, in certain contexts, is associated with poor
polymerase-1; SRC-3, steroid receptor co-activator 3. outcomes (22).

Frontiers in Oncology | www.frontiersin.org 2 October 2020 | Volume 10 | Article 587386


Mal et al. ERβ: A Ligand Activated Tumor Suppressor

FIGURE 1 | Exon map of ESR2 isoforms.

FIGURE 2 | Protein domain similarity of ERα and ERβ.

POST-TRANSLATIONAL MODIFICATIONS be necessary for transcriptional activity (25, 26). Similarly,


OF ERβ RECEPTOR phosphorylation of serine-60 and serine-75 residues increases
the degradation of ERβ. Additionally, phosphorylation of
Post-translational modifications of estrogen receptors serine-75 and serine-87 facilitates recruitment of the E3
contribute to genomic and rapid non-genomic signaling, ubiquitin ligase E6AP to the unliganded receptor, induces the
protein stability, dimerization, chromatin binding, and transition of ERβ from inactive clusters to a more mobile state
interaction with other regulators (23). The majority in the nuclear compartment, and increases its proteasomal
of these posttranslational modifications localize to the degradation (27).
AF1 domain. Interestingly, phosphorylation of ERβ at serines 75, 87, and
105, induced by ERK1/2 and p38 mitogen activated protein
Phosphorylation kinases (MAPK), augment the ligand dependent transcriptional
Multiple amino acid residues of ERβ have been identified activity of ERβ. Also of note, ERβ activation induces p38
as sites of phosphorylation. Glycogen Synthase Kinase MAPK activity that, in turn, induces ERβ expression and activity
(GSK) induced Serine-6 phosphorylation primes ERβ K4 suggesting a positive feedback loop (28–30). A phosphomimetic
for increased sumoylation and decreased ubiquitination, these mutant of S105 displayed enhanced transcriptional activity and
modifications being mutually exclusive and competitive. (24). cell migration and invasion.
Phosphorylation of tyrosine 36 is necessary for the tumor A distinct phosphorylation site, Serine-106, is involved
suppressor activity of the receptor in U87 glioblastoma cells, in recruitment of SRC-1 (also known as nuclear receptor
MCF7 breast cancer cells, and SKOV3 ovarian cancer cells. coactivator-1), stimulation of estrogen receptor beta
Interestingly, phosphorylation of Y36 of ERβ up-regulates its degradation, and activation of the transcriptional activity
ubiquitination and turnover, suggesting that turnover may of ERβ. Phosphorylation of an additional serine residue, S124

Frontiers in Oncology | www.frontiersin.org 3 October 2020 | Volume 10 | Article 587386


Mal et al. ERβ: A Ligand Activated Tumor Suppressor

also plays a role in the recruitment of SRC-1. Furthermore, a number of different proteins, suggesting that this modification
phosphorylation of Tyrosine-488 also activates transcriptional may regulate ERβ stability (38).
activity of ERβ, and is homologous to the Y537 residue of ERα, a
hotspot for activating mutations in breast cancer (31, 32).
A unique characteristic of ERβ is a phosphorylation site at MECHANISM OF TUMOR SUPPRESSOR
serine 236 in the hinge region of the receptor that is mediated FUNCTION: KEY DIFFERENCES OF ERα
via Erb-B2/B3 and Akt activation, leading to the recruitment of AND ERβ
the E3 ubiquitin ligase Mdm2 to ERβ, its polyubiquitination, and
26S proteasome mediated degradation. Phosphorylation of this Given that ERβ and ERα are both expressed in many
negatively charged hinge region by Akt may also decrease the tissues, one key question is whether they act synergistically or
transcriptional activity of ERβ (33, 34). antagonistically. The classic pathway of estrogen action includes
ligand binding to the ER ligand-binding pocket causing a
Ubiquitination and Sumoylation conformational change that induces dissociation of the ER-
The interaction of ERβ with the ubiquitin ligase E6AP enhances heat shock protein-90 (HSP90) chaperone complex receptor
the transcriptional activity of the receptor, however, the specific dimerization and binding to DNA (2). Ligand bound ER can also
target lysine for ubiquitination is yet to be described (27). interact with other trans-acting factors, both co-transcriptional
Ligand dependent degradation by MDM2 E3 ligase has also been activators, and repressors, that modulate transcription. One
described but, similarly, the exact lysine residue has not yet been difference between ERβ and ERα is the decreased transactivation
identified (27, 33). ERβ is also post-translationally modified at potential of the ERβ AF1 domain and reduced cooperative
lysine 4 by small ubiquitin like modifiers. Glycogen Synthase interaction with its AF2 domain (39). Of note, unlike ERα, ERβ
Kinase (GSK)-induced serine-6 phosphorylation primes ERβ binds to the co-repressor nuclear corepressor (NcoR) in the
K4 for increased sumoylation and decreased ubiquitination, unliganded state resulting in a lower basal level of transcriptional
these modifications being mutually exclusive and competitive, upregulation when expressed in the absence of ligand (40).
causing increased and decreased protein stability, and decreased Binding of ER to estrogen response elements (ERE) within
and increased transcriptional activity respectively (24, 27). ERβ promoters and enhancers induces transcription. These cis-
sumoylation is also up-regulated downstream of Ras-MAPK regulatory sites interact with the estrogen receptors in several
signaling (24). The E3 ubiquitin ligase CHIP ubiquitinates ERβ, different modes (41, 42). First, the receptor itself can bind to
resulting in basal turnover and inhibition of CHIP with small its cognate DNA binding element, the ERE, directly. However,
molecules such as Diptoindonesin G (Dip G) causes stabilization the EREs exist in multiple conformations: consensus EREs
of ERβ (35). (palindromic ERE repeats), non-consensus EREs, single binding
site EREs, multiple binding site EREs, and composite ERE
sites. In comparison to ERα, ERβ has reduced binding to
Palmitoylation non-consensus EREs, which represent the majority of estrogen
Palmitoylation of ERβ at cysteine-399 by palmitoyl transferases
responsive elements (e.g., C-fos, c-jun, pS2, cathepsin D, and
is necessary for its plasma membrane localization (36).
choline acetyltransferase cis regulatory sequences). Estrogen
Experiments suggest that palmitoylation of ERβ may be
receptors can also interact with DNA indirectly through tethering
obligatory for its membrane localization and binding with
to other DNA-bound transcription factors like AP-1 (42, 43).
caveolin-1. Binding to caveolin-1 is necessary for the activation
In fact, genome-wide mapping of ERβ binding sites reveals
of p38 MAPK mediated pro-apoptotic effects of ERβ (36). Thus,
extensive overlap with AP-1 binding sites (44). I a study by
palmitoylation may be a key post-translation modification for
Hall and McDonnell, it was found that in transient transfection
ERβ mediated pro-apoptotic effects. Interaction with the small
assays ERβ antagonizes the effect of ERα on certain E2 responsive
heat shock protein HSP27 is also necessary for membrane
E2-AP-1 composite promoters. This suggests that selective ERβ
localization (14).
activation could decrease proliferation induced by ERα (19).
Studies by Kim et al. (45) demonstrated the ligand and cell
O-Linked N-Acetyl context dependent ERα/SP1 and ERβ/SP1 action using an
N-Acetyl-D-Glucosamine Glycosylation estradiol responsive promoter containing three tandem GC
O-Linked N-acetyl glucosamine glycosylation and O-linked rich Sp1 binding elements. Both ERα and ERβ proteins were
phosphorylation of serine-16 of ERβ has been demonstrated found to interact with SP1 via their C-termini. ERα mediated
to contribute to the stability of the protein: The wild-type transcriptional activation of the GC rich promoter element was
estrogen receptor beta turns over rapidly with estrogen treatment observed in 3 out of 4 cell lines tested, but ERβ failed to
(7–8 h half-life) as compared to the S16 mutant with an activate transcription from the same promoter. Exchange of AF1
increased half-life (15–16 h). However, O-glycosylation at the domains of ER subtypes gave chimeric ERα/β and ERβ/α proteins
same residue results in stabilization of the protein, suggesting that that resembled wild type ER in terms of physical associations
phosphorylation and O-glycosylation have opposite effects on with the SP1 protein. Transcriptional activation studies with
protein stability (37). Another study showed that Serine-60 can chimeric ERα/β and ERβ/α showed that only ERα/β chimeras
also be modified through O-linked GlcNAcylation; this residue is that included the ERα AF1 domain activated transcription from
located in the PEST region, which mediates rapid degradation of the GC rich promoter element. This suggests that only the AF1

Frontiers in Oncology | www.frontiersin.org 4 October 2020 | Volume 10 | Article 587386


Mal et al. ERβ: A Ligand Activated Tumor Suppressor

domain of ERα is capable of productive interaction with SP1. not ERβ. The selective recruitment of SRC-3 to cis-regulatory
In fact, a specific region of the AF1 domain, including amino elements by ERα but not ERβ in the presence of tamoxifen could
acid residues 79–117 of ERα, was identified as being necessary explain some of the previously studied differences between the
for transcriptional activation from an SP1 binding element. In two estrogen receptors in relation to cancer outcomes (53).
contrast, ERβ represses transcription of ERα by binding Sp1 and Estradiol increases the proliferation of and causes tumor
recruiting a corepressor complex containing NCoR to the ERα formation of MCF-7 breast cancer cell line xenografts in an
gene (ESR1) promoter (46). ERα dependent manner (54). In contrast, the over-expression of
The transactivation properties of ERα and ERβ were also ERβ in MCF7 cells reduces proliferation in vitro and prevents
examined with different ligands in the context of an estrogen tumor formation in mice in the presence of supplemental
response half element in tandem with a AP-1 binding element estradiol. Furthermore, ERβ was shown to repress c-myc and
(47). Upon treatment with estradiol, ERα was found to upregulate cyclin D1 expression, and to increase the expression of p21
transcription from this composite promoter. In contrast, ERβ was and p27Kip1, leading to G2 cell cycle arrest in this model.
found to down-regulate transcription from the same promoter in In addition, ERβ regulates proliferation and migration through
the presence of estradiol, suggesting opposing effects for the two modulation of mitofusin 2 expression in MCF7 cells (54).
receptors at ERE-AP-1 composite response elements. Moreover, estrogen up-regulates the expression of LRP16 mRNA
The cell division protein cyclin D1 (CCND1), one target of through the activation of ERα, but not ERβ, which promotes
AP-1 and SP1 mediated transcription, is upregulated by ERα MCF-7 cell proliferation (55). Thus, ERβ and ERα have shown
and induces estrogen-mediated proliferation (48). The CCND1 opposing effects on proliferation and the expression of various
promoter contains a cAMP response element and an AP- oncogenes and tumor suppressors in breast cancer cell lines in
1 binding site, both of which play partially redundant roles the presence of estradiol.
in ERα mediated transcriptional up-regulation of cyclin D1. Malignant pleural mesotheliomas with high levels of AKT1
Surprisingly, activation of this promoter by ERβ was shown expression are associated with a worse prognosis, according
only to occur with antiestrogens. Estradiol, which up-regulates to a study done by Pinton et al. (56). An inhibitory feedback
cyclin D1 transcription in cells that over-express ERα, inhibits loop of ERβ and AKT was described, suggesting an ERβ
its transcription in cells that over-express ERβ. Additionally, it based therapeutic strategy for this malignancy. Interestingly,
was found that the presence of ERβ inhibits transcriptional up- patients with high levels of AKT1 expression in tumors had
regulation of cyclin D1 by ERα in the presence of estradiol, a worse prognosis. These experiments demonstrated the role
suggesting that ERβ may exert dominant negative effects on of the AKT1/SIRT1/FOXM1 axis in the repression of ERβ
ERα mediated cyclin D1 transcription. Opposing actions and expression. Conversely, ERβ agonists increase the acetylation
dominance of ERβ over ERα with respect to activation of cyclin and inactivation of AKT1 in breast cancer (57). Other groups
D1 gene expression may explain why ERβ is a negative regulator have demonstrated that the growth inhibitory effects of ERβ are
of the proliferative effects of estrogen. repressed by ERK1/2 activation and PI3K-Akt activation, and
It is important to note that ERβ knock down in ER rescued, in this context, by small molecule inhibitors of ERK and
positive breast cancer cell lines induces an invasive phenotype, PI3K (58).
increases anchorage independent proliferation, and elevates An additional opposing relationship between ERβ and PI3K-
EGFR signaling. This contrasts with the effects of ERα on Akt activation occurs through the E3 ubiquitin ligase MDM2:
EGFR signaling, which are collaborative in nature, and could heregulin-β, the ligand for HER2/HER3 dimers, activates Akt
explain how ERβ induction decreases anchorage independent and triggers the recruitment of MDM2 to ERβ, leading to
proliferation and invasiveness (49, 50). ERβ polyubiquitination and increased turnover (33). The ERβ
Human MutS homolog-2 (hMSH2) is a tumor suppressor transcriptional co-activator and histone acetyl transferase CBP
mutated in hereditary non-polyposis colorectal cancer. Both ERα stabilizes the interaction of ERβ with MDM2. Moreover, PI3K-
and ERβ interact with mismatch repair proteins, although the Akt activation abrogates the potential for CBP to activate
nature of the interactions diverge: ERα and hMSH2 interact ERβ mediated transcription (33, 34). Conversely, ERβ increases
in a ligand-dependent fashion, while ERβ and hMSH2 interact active PTEN and reduces Akt phosphorylation when activated
in a ligand-independent manner (51). In transient transfection by estradiol (59). Thus, there are multiple lines of evidence
assays, hMSH2 potentiated the transactivation function of ligand suggesting an antagonistic relationship between ERβ and Akt
bound ERα but not that of ERβ. This suggests that hMSH2 may signaling and the potential for synergy with PI3K-Akt inhibitors.
have a role as a coactivator of ERα dependent gene expression but These findings contrast with the effects of PI3K-Akt activation
not that of ERβ. on signaling by ERα (60, 61). Interestingly, however, in certain
Another key difference is their interactions with steroid contexts ERβ activation was shown to be associated with
receptor co-activator 3 (SRC-3)/AIB-1, an oncogenic co-activator upregulation of serum and glucocorticoid dependent kinase (62)
in both endocrine and non-endocrine cancers (52). SRC-3 and PIM1(63) expression, both of which have some overlap with
expression was positively associated with ERα expression, while Akt in terms of function.
being inversely associated with that of ERβ (53). In the same ERβ activation has been shown to modulate another target
study, HER2 positive primary tumor cell lines treated with of the Akt pathway: the FOXO family of transcription factors.
tamoxifen induced SRC-3 recruitment to an estrogen response ERβ ligands 3β androstanediol, diaryl propiononitrile, and 8-
element and enhanced the interaction of SRC-3 with ERα but vinylestra 1, 3, 5 triene-3,17-diol induce apoptosis via the

Frontiers in Oncology | www.frontiersin.org 5 October 2020 | Volume 10 | Article 587386


Mal et al. ERβ: A Ligand Activated Tumor Suppressor

p53 independent up-regulation of PUMA by FOXO3 (64). these actions and causes anti-proliferative, anti-invasive and anti-
This transcriptional upregulation of FOXO3a by ERβ agonists migratory effects as well as profound metabolic effects (80).
is additional evidence for the potential for synergy with Studies with breast cancer cells have also demonstrated
inhibitors of the PI3K-Akt pathway, activation of which results key differences in the interactions of estrogen receptors with
in phosphorylation of FOXOs by Akt leading to 14–3–3 TP53: ERα recruits SUV39H1/H2 to induce histone H3 lysine
binding, cytoplasmic retention, and degradation of FOXO 9 trimethylation to silence TP53-activated transcription, an
transcription factors (65). Potential for therapeutic synergy effect that is opposed by ERβ, which induces activating
between ERβ and FOXO transcription factors is further heterochromatin conformation by inducing histone H3 lysine
suggested by studies in prostate cancer models, which reveal 4 trimethylation and RNA polymerase II recruitment to ERα-
transcriptional upregulation of FOXO1 by the un-liganded repressed TP53-activated genes (81).
ERβ receptor (66, 67). These studies are unique in that they ERβ also binds to the aryl hydrocarbon nuclear translocator
suggest that the un-liganded receptor is a tumor suppressor, (ARNT) with increased affinity compared to ERα, suggesting that
and the estradiol bound receptor behaves like an oncogene in ARNT is a selective co-activator of ERβ (82). This association of
prostate cancer. ERβ with ARNT may lead to unique effects on hypoxia inducible
Other experiments suggest that non-genomic signaling factor-1α mediated upregulation of angiogenic factors via
by ERβ may be necessary for its tumor suppressor effects downregulation of aryl hydrocarbon nuclear translocator levels,
and this effect may involve interaction with and activation leading to the inhibition of hypoxic signaling and angiogenesis
of p38 MAPK at the plasma membrane, effects that are (82–84). Loss of ERβ in prostate cancer induces transcriptional
distinct from those of ERα that predominantly activates ERK down-regulation of prolyl hydroxylase 2, resulting in decreased
and PI3K-Akt through rapid non-genomic signaling (36, 68, HIF1-α hydroxylation, and thereby decreased ubiquitination by
69). Thus, a key distinction between non-genomic effects the von Hippel-Lindau tumor suppressor and increased HIF1α
of ERβ and ERα is the lack of activation of PI3K by protein expression (85).
ERβ (70). An additional difference between ERβ and ERα lies in the
Certain plant-derived flavonoids with xeno-estrogenic capacity for ERβ to interact with HSP27 (86). Another unique
activity, such as naringenin, may synergize with estradiol protein-protein interaction of ERβ is that with MNK2 isoform
in activating p38 MAPK, inducing caspase 3 activation, b, which is of interest given the role of MNK kinases 1 and 2
PARP cleavage, and apoptosis of human cancer cell lines that in stimulating protein translation when concentration of eIF4A
predominantly express ERβ (71). These same ligands selectively components such as eIF4E may be rate limiting (87). This
inhibit the non-genomic activation of ERK1/2 and Akt by ERα, interaction is unique to ERβ and for the b isoform of MNK2,
without impeding its genomic action on an estrogen response but not ERα or MNK1. On the other hand, estradiol induced
element driven promoter (72–74). Similar effects with respect to activation of AMPK has been shown to be mediated by selective
rapid non-genomic activation of p38 MAPK have been described interaction of the alpha catalytic subunit of AMPK with ERα but
for the flavonoid quercetin, although this alkaloid induced p38 not ERβ (88). Any functional effects on protein translation or cell
MAPK activation both through ERα and ERβ (22, 75). An viability remain to be determined.
artificial xenoestrogen, Bisphenol A, on the other hand, has One gene that was shown to be selectively upregulated by
the opposite effect, activating ERα dependent rapid membrane ERβ and not by ERα, is TFGβ- inducible early gene-1/ Kruppel
proximal signaling to ERK1/2 and Akt while inhibiting ERβ Like Factor 10 (KLF10) (89). Consistent with the low degree of
induced p38 MAPK activation, which may explain some of the sequence conservation in the AF1 domains of ERα and ERβ, this
endocrine disrupting effects that have been attributed to this effect is mediated by the ERβ AF1 domain. The protein product
compound (74, 76, 77). It is also important to note that ERβ of this gene induces apoptosis in pancreatic cancer cell lines (90).
activation induces p38 MAPK activity and p38 MAPK activity This decrease in cell viability may be due to induction of oxidative
induces ERβ expression (36), and activity (29), suggesting a stress (91).
positive feedback loop. Another notable difference between ERα and ERβ are their
Likewise, several other natural-product estrogen receptor effects on the unfolded protein response (UPR). The ERβ
ligands have estrogenic effects and may have chemo-preventive specific agonist ERB-041 has been shown to attenuate UPR
effects in breast cancer (78). For example, the isoflavone associated IRE1α and XBP1 splicing induced by tunicamycin,
compounds genistein and daidzein that are present in soy- a classic chemical activator of UPR, and, conversely, UPR
products have weak estrogen-like effects (Table 1) (79). Studies downregulates ERβ expression (92). In addition, ERβ1 was
have shown that both these compounds at low concentrations shown to sensitize breast cancer cells to endoplasmic reticulum
increase the proliferation of ERα+ MCF7 breast cancer cells (ER) stress by attenuating XBP-1 splicing and inducing the
while at higher doses are inhibitory. In contrast, in ERα-ve cells degradation of IRE1α by upregulating its E3 ligase synoviolin
(MDA-MB-231) this biphasic effect is not observed. Rather, both 1 (93). In contrast, spliced and unspliced XBP-1 induce ligand
of these phytoestrogens demonstrate an anti-proliferative effect. independent ERα transcriptional activity (94). Additionally,
This data indicates that both genistein and daidzein exert their XBP-1 is an ERα upregulated gene in ERα expressing cell lines
proliferative effect (at low doses) through ERα while ERβ, which (95). Key differences between ERα and ERβ are summarized in
is expressed at low levels in both ER+ and ER– cells, opposes Table 2.

Frontiers in Oncology | www.frontiersin.org 6 October 2020 | Volume 10 | Article 587386


Mal et al. ERβ: A Ligand Activated Tumor Suppressor

TABLE 2 | Key differences between the action of ERα and ERβ on several genes Secreto et al. (63) did similar experiments using Tet-
or proteins. inducible ERα and ERβ over-expression in the HS578T triple
Effect on ERα ERβ
negative breast cancer cell line. In this study, treatment
genes/proteins of doxycycline induced ERβ expressing cells with estradiol
but not tamoxifen resulted in decreased proliferation and
ERE-AP-1 Upregulates transcription Downregulates also modulation of expression of a number of genes. These
transcription included the downregulation of cyclin E2, connective tissue
ER(α/β)/SP-1 Transcriptional activation No transcriptional growth factor, Jagged1, and the upregulation of complement
activation
component 3, nuclear receptor interacting protein 1, CXCL14,
CCND1 transcription Positive regulator Negative regulator
and surprisingly, Pim1.
EGFR signaling Increases signaling Decreases signaling
Another study in U2OS osteosarcoma cells showed a different
hMSH2 Increased transactivation No effect
sub-set of ERβ regulated genes, including upregulation of
function
autotaxin and cystatin D (100). Other studies of ERβ induced
SRC-3/AIB-1 Enhanced interaction in No effect with
presence of tamoxifen tamoxifen
gene expression in U2OS cells reveal quite marked differences
TP53 Silences TP53-activated Induces ERα-silenced-
in transcriptional effects of different ligands, suggesting that
gene transcription TP53-activated gene each different agonist may have distinct effects (101). These
transcription ligand dependent differences in ERβ induced transcription were
Serum and Upregulates SGK3 Upregulates SGK1 further emphasized in a study that examined gene expression
glucocorticoid in ERα and ERβ overexpressing U2OS cells in response to
dependent kinase
estradiol, tamoxifen, and raloxifene (102). In this study, there
Unfolded protein Up-regulates XBP-1 Induces degradation of
was only a small overlap between ERα and ERβ regulated genes
response expression IRE1 and
downregulates XBP-1
(17%). Additionally only 27% of genes were regulated by both
splicing tamoxifen and raloxifene in ERα and ERβ expressing U2OS
Non-genomic effect Activates PI3-Akt and ERK Activates p38 MAPK cells, further emphasizing the potential for unique transcriptional
signaling—enhances signaling—enhances effects of individual ligands. Among the genes that were markedly
tumorigenicity pro-apoptotic/tumor upregulated by ERβ was the IL-10 family member Mda-7/IL-24.
suppressor function
This cytokine signals via the IL20 receptor to induce bystander
cell cytotoxicity in cancer cell lines but not untransformed cells
(103). The findings in regard to upregulation of Mda-7/IL20 by
ERβ activation was confirmed in another study of U2OS cells that
HIGH-THROUGHPUT ANALYSIS OF ERβ overexpress ERβ (104).
ACTION
Genome wide studies of ERβ induced transcriptional effects have CELL NON-AUTONOMOUS EFFECTS OF
shown clear differences from those of ERα. Using derivative ERβ
sub-lines of the ERα+ breast cancer cell line T47D, stably
transfected with a Tet-off ERβ expression construct, Williams In addition to its role in directly suppressing the viability of
et al. showed that of the 1434 transcripts whose expression was cancer cells, ERβ affects cancer cells in vivo through cell non -
altered by ERα, ERβ expression negatively impacted ERα induced autonomous mechanisms. Zhao et al. reported the effects of a
modulation of 998 (96). Surprisingly, they noted that expression highly selective agonist, LY500307 in a TNBC xenograft model
of a DNA binding domain-deleted form of ERβ resulted in the in mice. They observed suppression of lung metastasis in this
same effect on ERα induced gene expression for many of these model and demonstrated increased IL-1β secretion by cancer
genes. Leucine-rich repeat-containing 15 and Apolipoprotein D cells upon exposure to LY500307, which resulted in increased
mRNAs were markedly up regulated in estradiol treated ERβ neutrophil recruitment into tumors and regression (105). These
expressing cells. ERβ markedly downregulated other genes such results suggest that ERβ activation may recruit immune cells
as the cytokine IL-20 and the anti-apoptotic gene Bcl2. into the tumor microenvironment. Notably, estrogen induced
A similar study by Lim et al. (97) again in T47D cells, ERβ activation has been shown to cause NLRP3 inflammosome
identified a number of genes whose upregulation by estradiol activation—a key upstream pathway for IL-1β processing and
was inhibited upon ERβ overexpression including XBP1, PCNA, secretion—in models of endometrial cancer, although a pro-
RAC1, CDK1, Cdc6, DNA replication helicase 2-like, and cell neoplastic role was suggested in this model (106).
division cycle 28 protein kinase regulatory subunit 2. Studies Interestingly ERβ was shown to play a pro-inflammatory
by Chang et al. (98) in MCF7 cells showed that ERβ induces role in the estrogen driven non-malignant gynecologic disorder,
the expression of S100P while attenuating estradiol induced endometriosis. ERβ is expressed at many fold high levels in
uregulation of FOXM1, CDC25A, E2F1, and survivin/BIRC5. endometriotic tissue as compared to normal endometrium (107).
Yet another study in MCF7 cells showed that both ERβ1 and Over-expression of ERβ enhances the growth of endometriotic
ERβ2/ERβcx downregulated the expression of the proliferation lesions in mice and interacts with components of the NLRP3
related genes cathepsin D and IGFBP4 (99). inflammosome such as NALP3, caspase-1, and caspase 9

Frontiers in Oncology | www.frontiersin.org 7 October 2020 | Volume 10 | Article 587386


Mal et al. ERβ: A Ligand Activated Tumor Suppressor

and induces the processing of pro-IL-1β to its active form REGULATION OF CO-TRANSCRIPTIONAL
(108). In addition, several other inflammatory mediators AND POST-TRANSCRIPTIONAL RNA
such as macrophage inflammatory protein 1α, macrophage
PROCESSING BY ERβ
inflammatory protein 2, IL-16, monocyte chemoattractant
protein 5, B lymphocyte chemoattractant, and triggering receptor In addition to its effects on rapid non-genomic signaling and
expressed on myeloid cells (TREM1) were upregulated upon ERβ transcription, ERβ induces exon skipping of specific genes in
over-expression in endometrial cells. However, other cytokines response to estradiol (38). In breast cancer cells that were
were down-regulated such as TNF-α, monokine induced by responsive to hormone therapy, there was a two-fold increase
interferon gamma, macrophage colony stimulating factor (M- in splicing events in cells that expressed ERβ. ERβ also induced
CSF), interferon gamma inducible protein 10/CXCL10, and the promoter-switching of 61 genes and differential splicing of
keratinocyte chemoattractant/CXCL1. 28 genes and the transcription and splicing of transcriptional
Conversely, several studies do suggest an anti-inflammatory regulators such as NCOR2, which mediates gene repression by
role for ERβ in other contexts. For example, ERβ activation the tamoxifen-bound ERα (38).
by selective agonists was shown to suppress TNFα induced
gene expression by recruiting the co-activator SRC-2 forming
a repressive complex (109). ERβ activation suppresses NF- ERβ EXPRESSION AND ROLE IN VARIOUS
κB activation and secretion of multiple cytokines in prostate CANCER TYPES
cancer cell lines (110). In addition, an ERβ selective agonist,
LY3201 inhibited NF-κB activation and neuroinflammation in ERβ is unique in that it functions as a tumor suppressor in diverse
murine models of demyelinating disorders, as did 5-androsten- biologic contexts. ERβ has been implicated in various cancer
3β, 17β-diol /ADIOL, another ERβ agonist (12, 111). Thus, the types, including breast, prostate, lung, glioblastoma, thyroid, and
ramifications of ERβ activation on immune activation should ovarian cancer (15–19).
be analyzed separately in each disease context, keeping the Concerning breast cancer, ERβ expression by IHC is
possibility of ligand specific effects in mind, to determine the net detectable in 20–30% of invasive breast cancers (127). In regard
effects on immune activation. to association with patient outcomes in breast cancer, ERβ
expression is an independent prognostic marker in ERα+-
progesterone receptor positive breast cancer (128). Mann et al.
ERβ KNOCKOUT MICE (129) found that ERβ status is a predictor of survival in women
with breast cancer when treated with adjuvant hormonal therapy.
Krege et al. (112) developed and characterized the first ERβ KO They also found that expression of ERβ in more than 10% of
mouse model in the late nineties. These mice develop normally the cancer cells was associated with improved survival. This
and are morphologically and histologically indistinguishable association has been confirmed in a meta-analysis showing
from littermates. ERβ−/− female mice are sub-fertile, with improved disease-free survival in patients with tumors positive
smaller litters than WT mice, which is corrected by ovulation for ERβ1 and ERβ2 isoforms (130).
inducers suggesting defects in ovulation. ERβ−/− male mice Although therapeutic efforts so far have focused more on
reproduce normally but older mice develop prostatic and triple negative breast cancer (TNBC), ERβ expression in tissue
bladder hyperplasia. This phenotype contrasts with that of microarrays of breast cancer was significantly associated with
ERα KO mice, which are infertile and display a complete expression of ERα and progesterone receptor and, when analyzed
absence of breast development in females and reproductive by molecular classification, higher in luminal A (72%) and
tract, gonadal and behavioral deficits in males and females luminal B (68%) breast cancers as compared to HER2+ and
(113–117). Further investigation of the ERβ KO phenotype basal like sub-types (55 and 60%) (131). In this study, ERβ
revealed the development of age related myeloproliferative expression was inversely associated with HER2 and EGFR
disease resembling chronic myeloid leukemia with lymphoid expression. These data suggests potential for therapeutic utility
blast crisis (118) and prostatic hyperplasia and prostatic in ERα+ breast cancer. In ER+ve breast cancer ERα drives
intraepithelial neoplasia in the ventral prostate (119), consistent proliferation, while ERβ has anti-proliferative effects (11).
with a tumor suppressor role. Moreover, in mammary specific Various mechanisms have been suggested. The attenuation of AP-
Cre- p53F/F ERβF/F mice, loss of ERβ accelerated the onset 1 and Sp1 mediated estradiol induced transcriptional activation
of mammary tumors, establishing ERβ as a tumor suppressor is a potential, although not clearly established, mechanism for
in the mammary epithelium (120). Additionally, deficits were inhibition (132) and the AF1 domain of ERβ is necessary for
noted in mammary epithelial organization and differentiation this effect (133, 134). In regard to TNBC, Reese et al. suggested
to secretory epithelium (121), colonic epithelial differentiation, that ERβ induced the up-regulation of secreted proteins known as
and apoptosis (122), pulmonary development (123, 124), and cystatins that down-regulate canonical TGFβ signaling, is critical
neuronal migration in the developing cortex (118) of ERβ KO for the suppression of the metastatic phenotype (135).
mice. ERβ KO female mice are partially protected from age- Some studies, however, suggest a pro-invasive effect of the
related trabecular bone loss (125), and have increased radial weak and modestly selective ERβ ligand, di-aryl propionitrile,
cortical bone growth (126), suggesting that maintenance of bone in triple negative SUM149 inflammatory breast cancer cells,
density is primarily a function of ERα. an effect that MEK inhibition reversed (136). Another study

Frontiers in Oncology | www.frontiersin.org 8 October 2020 | Volume 10 | Article 587386


Mal et al. ERβ: A Ligand Activated Tumor Suppressor

suggested ERβ1 expression in TNBC is upregulated by insulin patients with ovarian tumors that expressed cytoplasmic ERβ
like growth factor 2 and was associated with a worse overall had prolonged survival compared to patients whose tumors
survival and that ERβ activation by diaryl propionitrile is did not express the receptor (151). Interestingly, another study
associated with increased proliferation in TNBC and increased concluded that nuclear expression of ERα or ERβ was not
secretion of VEGF, amphiregulin, and WNT 10b/12 (137). associated with clinical outcome (152). Pujol et al. further
Hence, we still do not fully understand the mechanism of described expression of ERα and ERβ in ovarian cancer, normal
action of ERβ in breast cancer. Highly specific and selective ovarian tissue, metastatic tissue, and benign tissue (153). Normal
agonists are required to fully characterize the role of ERβ in breast ovarian tissue expressed ERβ at higher levels than in ovarian
cancer, and, thus, determine its potential as a therapeutic strategy. cancer cell lines. Seventy-eight percent of benign and borderline
The development of highly specific inhibitors is especially tumors expressed ERβ mRNA, whereas 29% expressed ERα.
germane to the application of this strategy for breast cancers In another study of ovarian cancer, metastatic cancer tissues
given the concurrent expression of ERα in the majority of expressed only ERα mRNA and protein, suggesting that ERβ
breast cancers. expression is down regulated at metastatic sites consistent with
In the human male prostate, ERβ is expressed in stromal its role as a tumor suppressor (154). Lastly, Chan et al. (155)
and luminal epithelial cells (138). Studies using ER-knockout showed that ERβ1 expression is lower in high grade cancers and
mice have shown that mice lacking the ERβ gene develop that higher cytoplasmic expression of ERα and ERβ1 is associated
prostate cancer at an increased rate with the addition of the with better disease-free and overall survival. They also noted that
pertinent hormones (139). ERβ may play a role in prostate higher nuclear ERβ5 and lower cytoplasmic ERβ5 was associated
differentiation and proliferation, and may modulate both the with clear cell histology and a worse prognosis.
initial phases of prostate cancer as well as androgen-independent An overwhelming amount of literature suggests that there
tumor growth (1). is an inverse relationship between the expression of ERβ
Concerning the role of ERβ in lung cancer, the available data and the presence of colorectal polyps. The effects of ERβ in
suggest both pro-tumorigenic as well as tumor suppressor roles colorectal cancer may include reduced metastasis (via inhibiting
depending on the experimental context. In healthy lung tissue, PROX1), enhanced apoptosis (via p53), an anti-inflammatory
both pneumocytes and bronchial epithelial cells express ERβ. It response (via NF-kB), and reduced proliferation (by inhibiting
is also required for the maintenance of extracellular matrix in the c-myc) (156).
lung (140, 141). ERβ stimulates the proliferation of non-small cell Zeng et al. (157) showed that the ERβ selective agonist DPN
lung carcinoma cells in certain experimental contexts (142, 143). suppressed proliferation of papillary thyroid carcinoma (PTC)
Other reports suggest that cytoplasmic expression of ERβ is cell lines in contradistinction to the effects of the ERα specific
associated with therapy resistance to EGFR inhibitors (144). In agonist PPT, which increased proliferation. Similarly, Dong et al.
addition, other studies have shown that nuclear ERβ is associated (158) examined the effects of estradiol in the BCPAP PTC
with a better prognosis, while cytoplasmic ERβ localization is cell line and discovered that ERβ, in contrast to the effects of
associated with a poorer prognosis (145–147). When exclusively ERα, suppresses motility of these cells on exposure to estradiol.
cytoplasmic, ERβ expression has been associated with increased Reduced expression of ERβ1 in ERα negative PTC was associated
growth of non-small cell lung cancer via extra nuclear non- with greater invasiveness and metastasis (159). Expression of
genomic activation of cAMP, Akt, and MAPK signaling (148). the ERβ2 isoform in PTC was also associated with reduced
Thus, in non-small cell lung cancer the effects of ERβ are complex lymph node metastasis in pre-menopausal patients with PTC
and possibly differ from the classic tumor suppressor role it plays (160). However, another study by Dong et al. observed that
in other malignancies. both nuclear, and combined nuclear and cytoplasmic staining for
One study suggests that glioblastoma cells predominantly ERβ1, by immuno-histochemistry was lower in PTC compared
express the ERβ1 and ERβ5 isoforms, and the ERβ2 and ERβ4 to the lesions of a benign nodular thyroid goiter (BNTG) but
isoforms to a lesser degree (149). ERβ knockout human GBM cell that nuclear and combined nuclear and cytoplasmic staining for
lines have increased invasive properties and re-introduction of ERβ2 was actually higher in PTC than benign lesions (159, 161).
ERβ1 reverses these effects. In contrast, the over-expression of Similarly, in the medullary thyroid carcinoma cell line TT, the
ERβ5 actually increased anchorage-independent growth. In vivo, expression of ERβ suppressed growth, again in contradistinction
the expression of ERβ1 in U251-ERβ-KO GBM xenografts was to the effects of ERα over-expression (162).
associated with longer survival and was the only isoform with a Renal cell carcinoma (RCC) cell lines express ERβ, which
tumor-suppressive effect. mediates estradiol-induced decreases in viability (163).
Estrogen receptor beta may have therapeutic relevance in Moreover, ERβ protein expression was lower in RCC than
Hodgkin lymphoma. One article suggests that the ERβ agonist in normal kidney when examined by immuno-histochemistry.
2,3-bis (4-hydroxyphenyl)-propionitrile (DPN) could reduce These findings suggest a tumor suppressor role for ERβ in RCC.
Hodgkin lymphoma cell growth up to 60%. The mechanism of
reduced cell growth was suggested to be via the overexpression of
damage-regulated autophagy modulator 2 (DRAM2) and protein CONCLUSIONS
1 light chain 3 (150).
Expression of estrogen receptor beta also seems to have an The promise of ERβ activation lies in the possibility of a targeted
effect on patients with ovarian cancer. One study showed that therapy that concurrently activates multiple tumor suppressor

Frontiers in Oncology | www.frontiersin.org 9 October 2020 | Volume 10 | Article 587386


Mal et al. ERβ: A Ligand Activated Tumor Suppressor

pathways while causing relatively few side effects. Thus, ERβ is a action of ERβ agonist is required for the design of potential
nuclear receptor with broad-spectrum tumor suppressor activity synergistic combinations with other therapeutic drug classes for
that could serve as a potential treatment target in a variety of breast cancer.
human cancers. Further refinement of selective ERβ modulators
with the development of highly selective ERβ agonists that AUTHOR CONTRIBUTIONS
lack ERα agonist activity will be necessary to fully harness the
potential of this promising therapeutic avenue. Gene expression RM, AM, JDav, JDat, MV, MK, JM, NWillin, and MC conducted a
signature and non-genomic signaling pathway activation should review of the literature and prepared the body of the manuscript.
be defined for each compound given the potential for unique DS, JV, SS, NWillia, RW, ML, RG, and BR critically reviewed
ligand dependent effects on the recruitment of binding partners the publication. MC drafted the manuscript. All the authors
(101). In addition, further investigation into the mechanism of endorsed the final form of the manuscript.

REFERENCES 15. Bialesova L, Xu L, Gustafsson J-Å, Haldosen L-A, Zhao C, Dahlman-


Wright K. Estrogen receptor β2 induces proliferation and invasiveness of
1. Signoretti S, Loda M. Estrogen receptor β in prostate cancer: triple negative breast cancer cells: association with regulation of PHD3 and
brake pedal or accelerator? Am J Pathol. (2001) 159:13. HIF-1α. Oncotarget. (2017) 8:76622. doi: 10.18632/oncotarget.20635
doi: 10.1016/S0002-9440(10)61666-5 16. Dey P, Velazquez-Villegas LA, Faria M, Turner A, Jonsson P, Webb P,
2. Pratt WB, Toft DO. Steroid receptor interactions with heat shock et al. Estrogen receptor β2 induces hypoxia signature of gene expression
protein and immunophilin chaperones. Endocr Rev. (1997) 18:306–60. by stabilizing HIF-1α in prostate cancer. PLoS One. (2015) 10:e0128239.
doi: 10.1210/edrv.18.3.0303 doi: 10.1371/journal.pone.0128239
3. Joab I, Radanyi C, Renoir M, Buchou T, Catelli MG, Binart N, et al. 17. Faria M, Karami S, Granados-Principal S, Dey P, Verma A, Choi DS, et al.
Common non-hormone binding component in non-transformed chick The ERβ4 variant induces transformation of the normal breast mammary
oviduct receptors of four steroid hormones. Nature. (1984) 308:850–3. epithelial cell line MCF-10A; the ERβ variants ERβ2 and ERβ5 increase
doi: 10.1038/308850a0 aggressiveness of TNBC by regulation of hypoxic signaling. Oncotarget.
4. Catelli MG, Binart N, Jung-Testas I, Renoir JM, Baulieu EE, Feramisco (2018) 9:12201. doi: 10.18632/oncotarget.24134
JR, et al. The common 90-kd protein component of non-transformed 18. Moore JT, McKee DD, Slentz-Kesler K, Moore LB, Jones SA,
’8S’ steroid receptors is a heat-shock protein. EMBO J. (1985) 4:3131–5. Horne EL, et al. Cloning and characterization of human estrogen
doi: 10.1002/j.1460-2075.1985.tb04055.x receptor β isoforms. Biochem Biophys Res Commun. (1998) 247:75–8.
5. Smith DF, Toft DO. Steroid receptors and their associated proteins. Mol doi: 10.1006/bbrc.1998.8738
Endocrinol. (1993) 7:4–11. doi: 10.1210/mend.7.1.8446107 19. Hall JM, McDonnell DP. The estrogen receptor β-isoform (ERβ) of the
6. Devin-Leclerc J, Meng X, Delahaye F, Leclerc P, Baulieu E-E, Catelli human estrogen receptor modulates ERα transcriptional activity and is
M-G. Interaction and dissociation by ligands of estrogen receptor and a key regulator of the cellular response to estrogens and antiestrogens.
Hsp90: the antiestrogen RU 58668 induces a protein synthesis-dependent Endocrinology. (1999) 140:5566–78. doi: 10.1210/endo.140.12.7179
clustering of the receptor in the cytoplasm. Mol Endocrinol. (1998) 12:842– 20. Reese JM, Suman VJ, Subramaniam M, Wu X, Negron V, Gingery A, et al.
54. doi: 10.1210/mend.12.6.0121 ERβ1: characterization, prognosis, and evaluation of treatment strategies
7. Perkins MS, Louw-du Toit R, Africander D. A comparative characterization in ERα-positive and-negative breast cancer. BMC Cancer. (2014) 14:749.
of estrogens used in hormone therapy via estrogen receptor (ER)- doi: 10.1186/1471-2407-14-749
alpha and -beta. J Steroid Biochem Mol Biol. (2017) 174:27–39. 21. Wang J, Zhang C, Chen K, Tang H, Tang J, Song C, et al. ERβ1 inversely
doi: 10.1016/j.jsbmb.2017.07.022 correlates with PTEN/PI3K/AKT pathway and predicts a favorable prognosis
8. Bhavnani BR, Tam SP, Lu X. Structure activity relationships and differential in triple-negative breast cancer. Breast Cancer Res Treat. (2015) 152:255–69.
interactions and functional activity of various equine estrogens mediated via doi: 10.1007/s10549-015-3467-3
estrogen receptors (ERs) ERα and ERβ. Endocrinology. (2008) 149:4857–70. 22. Galluzzo P, Martini C, Bulzomi P, Leone S, Bolli A, Pallottini V, et al.
doi: 10.1210/en.2008-0304 Quercetin-induced apoptotic cascade in cancer cells: antioxidant versus
9. Barkhem T, Carlsson B, Nilsson Y, Enmark E, Gustafsson J, Nilsson S. estrogen receptor α-dependent mechanisms. Mol Nutr Food Res. (2009)
Differential response of estrogen receptor alpha and estrogen receptor beta 53:699–708. doi: 10.1002/mnfr.200800239
to partial estrogen agonists/antagonists. Mol Pharmacol. (1998) 54:105–12. 23. Le Romancer M, Poulard C, Cohen P, Sentis S, Renoir J-M, Corbo L.
doi: 10.1124/mol.54.1.105 Cracking the estrogen receptor’s posttranslational code in breast tumors.
10. Katzenellenbogen BS. Biology and receptor interactions of estriol and estriol Endocr Rev. (2011) 32:597–622. doi: 10.1210/er.2010-0016
derivatives in vitro and in vivo. J Steroid Biochem. (1984) 20:1033–7. 24. Picard N, Caron V, Bilodeau S, Sanchez M, Mascle X, Aubry M, et al.
doi: 10.1016/0022-4731(84)90015-3 Identification of estrogen receptor beta as a SUMO-1 target reveals a novel
11. Paterni I, Granchi C, Katzenellenbogen JA, Minutolo F. Estrogen receptors phosphorylated sumoylation motif and regulation by glycogen synthase
alpha (ERα) and beta (ERβ): subtype-selective ligands and clinical potential. kinase 3β. Mol Cell Biol. (2012) 32:2709–21. doi: 10.1128/MCB.06624-11
Steroids. (2014) 90:13–29. doi: 10.1016/j.steroids.2014.06.012 25. Yuan B, Cheng L, Gupta K, Chiang H-C, Gupta HB, Sareddy GR,
12. Saijo K, Collier JG, Li AC, Katzenellenbogen JA, Glass CK. An et al. Tyrosine phosphorylation regulates ERβ ubiquitination, protein
ADIOL-ERβ-CtBP transrepression pathway negatively regulates microglia- turnover, and inhibition of breast cancer. Oncotarget. (2016) 7:42585.
mediated inflammation. Cell. (2011) 145:584–595. doi: 10.1016/j.cell.2011. doi: 10.18632/oncotarget.10018
03.050 26. Yuan B, Cheng L, Chiang H-C, Xu X, Han Y, Su H, et al. A phosphotyrosine
13. Enmark E, Pelto-Huikko M, Grandien K, Lagercrantz S, Lagercrantz J, switch determines the antitumor activity of ERβ. J Clin Invest. (2014)
Fried G, et al. Human estrogen receptor β-gene structure, chromosomal 124:3378–90. doi: 10.1172/JCI74085
localization, and expression pattern. J Clin Endocrinol Metab. (1997) 27. Picard N, Charbonneau C, Sanchez M, Licznar A, Busson M, Lazennec
82:4258–65. doi: 10.1210/jcem.82.12.4470 G, et al. Phosphorylation of activation function-1 regulates proteasome-
14. Razandi M, Pedram A, Levin ER. Heat shock protein 27 is required for sex dependent nuclear mobility and E6-associated protein ubiquitin ligase
steroid receptor trafficking to and functioning at the plasma membrane. Mol recruitment to the estrogen receptor β. Mol Endocrinol. (2008) 22:317–30.
Cell Biol. (2010) 30:3249–61. doi: 10.1128/MCB.01354-09 doi: 10.1210/me.2007-0281

Frontiers in Oncology | www.frontiersin.org 10 October 2020 | Volume 10 | Article 587386


Mal et al. ERβ: A Ligand Activated Tumor Suppressor

28. Lam H-M, Babu CS, Wang J, Yuan Y, Lam Y-W, Ho S-M, et al. 47. Paech K, Webb P, Kuiper GG, Nilsson S, Gustafsson J, Kushner PJ, et al.
Phosphorylation of human estrogen receptor-beta at serine 105 inhibits Differential ligand activation of estrogen receptors ERα and ERβ at AP1 sites.
breast cancer cell migration and invasion. Mol Cell Endocrinol. (2012) Science. (1997) 277:1508–10. doi: 10.1126/science.277.5331.1508
358:27–35. doi: 10.1016/j.mce.2012.02.012 48. Liu MM, Albanese C, Anderson CM, Hilty K, Webb P, Uht RM, et al.
29. Driggers PH, Segars JH, Rubino DM. The proto-oncoprotein Brx activates Opposing action of estrogen receptors alpha and beta on cyclin D1 gene
estrogen receptor β by a p38 mitogen-activated protein kinase pathway. J expression. J Biol Chem. (2002) 277:24353–60. doi: 10.1074/jbc.M201829200
Biol Chem. (2001) 276:46792–7. doi: 10.1074/jbc.M106927200 49. Pinton G, Thomas W, Bellini P, Manente AG, Favoni RE, Harvey BJ,
30. Caiazza F, Galluzzo P, Lorenzetti S, Marino M. 17β-estradiol induces ERβ up- et al. Estrogen receptor beta exerts tumor repressive functions in human
regulation via p38/MAPK activation in colon cancer cells. Biochem Biophys malignant pleural mesothelioma via EGFR inactivation and affects response
Res Commun. (2007) 359:102–7. doi: 10.1016/j.bbrc.2007.05.059 to gefitinib. PLoS One. (2010) 5:e14110. doi: 10.1371/journal.pone.0014110
31. Tharun IM, Nieto L, Haase C, Scheepstra M, Balk M, Möcklinghoff S, et al. 50. Ignar-Trowbridge DM, Nelson KG, Bidwell MC, Curtis SW, Washburn TF,
Subtype-specific modulation of estrogen receptor-coactivator interaction by McLachlan JA, et al. Coupling of dual signaling pathways: epidermal growth
phosphorylation. ACS Chem Biol. (2015) 10:475-484. doi: 10.1021/cb5007097 factor action involves the estrogen receptor. Proc Natl Acad Sci U S A. (1992)
32. Nguyen HD, Phan TT, Carraz M, Brunsveld L. Estrogen receptor α/β- 89:4658–62. doi: 10.1073/pnas.89.10.4658
cofactor motif interactions; interplay of tyrosine 537/488 phosphorylation 51. Wada-Hiraike O, Yano T, Nei T, Matsumoto Y, Nagasaka K, Takizawa S, et al.
and LXXLL motifs. Mol Biosyst. (2012) 8:3134–41. doi: 10.1039/c2mb25257k The DNA mismatch repair gene hMSH2 is a potent coactivator of oestrogen
33. Sanchez M, Picard N, Sauve K, Tremblay A. Coordinate regulation receptor alpha. Br J Cancer. (2005) 92:2286–91. doi: 10.1038/sj.bjc.6602614
of estrogen receptor β degradation by Mdm2 and CREB-binding 52. Tien JC, Xu J. Steroid receptor coactivator-3 as a potential molecular
protein in response to growth signals. Oncogene. (2013) 32:117. target for cancer therapy. Expert Opin Ther Targets. (2012) 16:1085–96.
doi: 10.1038/onc.2012.19 doi: 10.1517/14728222.2012.718330
34. Sanchez M, Sauvé K, Picard N, Tremblay A. The hormonal response of 53. McIlroy M, Fleming FJ, Buggy Y, Hill AD, Young LS. Tamoxifen-induced ER-
estrogen receptor β is decreased by the phosphatidylinositol 3-kinase/Akt alpha-SRC-3 interaction in HER2 positive human breast cancer; a possible
pathway via a phosphorylation-dependent release of CREB-binding protein. mechanism for ER isoform specific recurrence. Endocr Related Cancer.
J Biol Chem. (2007) 282:4830–40. doi: 10.1074/jbc.M607908200 (2006) 13:1135–45. doi: 10.1677/erc.1.01222
35. Zhao Z, Wang L, James T, Jung Y, Kim I, Tan R, et al. Reciprocal regulation of 54. Paruthiyil S, Parmar H, Kerekatte V, Cunha GR, Firestone GL, Leitman
ERα and ERβ stability and activity by Diptoindonesin G. Chem Biol. (2015) DC. Estrogen receptor beta inhibits human breast cancer cell proliferation
22:1608–21. doi: 10.1016/j.chembiol.2015.10.011 and tumor formation by causing a G2 cell cycle arrest. Cancer Res. (2004)
36. Galluzzo P, Caiazza F, Moreno S, Marino M. Role of ERβ palmitoylation 64:423–8. doi: 10.1158/0008-5472.CAN-03-2446
in the inhibition of human colon cancer cell proliferation. Endocr Related 55. Ma L, Liu Y, Geng C, Qi X, Jiang J. Estrogen receptor beta inhibits estradiol-
Cancer. (2007) 14:153–67. doi: 10.1677/ERC-06-0020 induced proliferation and migration of MCF-7 cells through regulation of
37. Cheng X, Hart GW. AlternativeO-glycosylation/O-phosphorylation of mitofusin 2. Int J Oncol. (2013) 42:1993–2000. doi: 10.3892/ijo.2013.1903
serine-16 in murine estrogen receptor β post-translational regulation of 56. Pinton G, Zonca S, Manente AG, Cavaletto M, Borroni E,
turnover and transactivation activity. J Biol Chem. (2001) 276:10570–5. Daga A, et al. SIRT1 at the crossroads of AKT1 and ERβ in
doi: 10.1074/jbc.M010411200 malignant pleural mesothelioma cells. Oncotarget. (2016) 7:14366–79.
38. Dago DN, Scafoglio C, Rinaldi A, Memoli D, Giurato G, Nassa doi: 10.18632/oncotarget.7321
G, et al. Estrogen receptor beta impacts hormone-induced alternative 57. Manente AG, Pinton G, Zonca S, Cilli M, Rinaldi M, Daga A, et al.
mRNA splicing in breast cancer cells. BMC Genomics. (2015) 16:367. Intracellular lactate-mediated induction of estrogen receptor beta (ERβ) in
doi: 10.1186/s12864-015-1541-1 biphasic malignant pleural mesothelioma cells. Oncotarget. (2015) 6:25121–
39. Zwart W, de Leeuw R, Rondaij M, Neefjes J, Mancini MA, Michalides R. 34. doi: 10.18632/oncotarget.4486
The hinge region of the human estrogen receptor determines functional 58. Cotrim CZ, Fabris V, Doria ML, Lindberg K, Gustafsson JA, Amado F,
synergy between AF-1 and AF-2 in the quantitative response to estradiol et al. Estrogen receptor beta growth-inhibitory effects are repressed through
and tamoxifen. J Cell Sci. (2010) 123(Pt 8):1253–61. doi: 10.1242/jcs. activation of MAPK and PI3K signalling in mammary epithelial and breast
061135 cancer cells. Oncogene. (2013) 32:2390–402. doi: 10.1038/onc.2012.261
40. Webb P, Valentine C, Nguyen P, Price RH Jr, Marimuthu A, West BL, et al. 59. Hartz AM, Madole EK, Miller DS, Bauer B. Estrogen receptor beta
ERβ Binds N-CoR in the presence of estrogens via an LXXLL-like motif in signaling through phosphatase and tensin homolog/phosphoinositide 3-
the N-CoR C-terminus. Nucl Recep. (2003) 1:4. doi: 10.1186/1478-1336-1-4 kinase/Akt/glycogen synthase kinase 3 down-regulates blood-brain barrier
41. Deroo BJ, Buensuceso AV. Minireview: estrogen receptor-β: mechanistic breast cancer resistance protein. J Pharmacol Exp Ther. (2010) 334:467–76.
insights from recent studies. Mol Endocrinol. (2010) 24:1703–14. doi: 10.1124/jpet.110.168930
doi: 10.1210/me.2009-0288 60. Stoica GE, Franke TF, Moroni M, Mueller S, Morgan E, Iann MC, et al.
42. Katzenellenbogen BS, Katzenellenbogen JA. Estrogen receptor transcription Effect of estradiol on estrogen receptor-α gene expression and activity can
and transactivation Estrogen receptor alpha and estrogen receptor beta: be modulated by the ErbB2/PI 3-K/Akt pathway. Oncogene. (2003) 22:7998.
regulation by selective estrogen receptor modulators and importance in doi: 10.1038/sj.onc.1206769
breast cancer. Breast Cancer Res. (2000) 2:335. doi: 10.1186/bcr78 61. Campbell RA, Bhat-Nakshatri P, Patel NM, Constantinidou D, Ali S,
43. Deroo BJ, Korach KS. Estrogen receptors and human disease. J Clin Invest. Nakshatri H. Phosphatidylinositol 3-Kinase/AKT-mediated activation of
(2006) 116:561–70. doi: 10.1172/JCI27987 estrogen receptor α A new model for anti-estrogen resistance. J Biol Chem.
44. Zhao C, Gao H, Liu Y, Papoutsi Z, Jaffrey S, Gustafsson JA, et al. Genome- (2001) 276:9817–24. doi: 10.1074/jbc.M010840200
wide mapping of estrogen receptor-beta-binding regions reveals extensive 62. Monsivais D, Dyson MT, Yin P, Navarro A, Coon JST, Pavone ME, et al.
cross-talk with transcription factor activator protein-1. Cancer Res. (2010) Estrogen receptor β regulates endometriotic cell survival through serum and
70:5174–183. doi: 10.1158/0008-5472.CAN-09-4407 glucocorticoid-regulated kinase activation. Fertil Steril. (2016) 105:1266–73.
45. Kim K, Thu N, Saville B, Safe S. Domains of estrogen receptor alpha doi: 10.1016/j.fertnstert.2016.01.012
(ERα) required for ERα/Sp1-mediated activation of GC-rich promoters by 63. Secreto FJ, Monroe DG, Dutta S, Ingle JN, Spelsberg TC. Estrogen receptor
estrogens and antiestrogens in breast cancer cells. Mol Endocrinol. (2003) alpha/beta isoforms, but not betacx, modulate unique patterns of gene
17:804–17. doi: 10.1210/me.2002-0406 expression and cell proliferation in Hs578T cells. J Cell Biochem. (2007)
46. Bartella V, Rizza P, Barone I, Zito D, Giordano F, Giordano C, et al. Estrogen 101:1125–47. doi: 10.1002/jcb.21205
receptor beta binds Sp1 and recruits a corepressor complex to the estrogen 64. Dey P, Ström A, Gustafsson J-Å. Estrogen receptor β upregulates FOXO3a
receptor alpha gene promoter. Breast Cancer Res Treat. (2012) 134:569–81. and causes induction of apoptosis through PUMA in prostate cancer.
doi: 10.1007/s10549-012-2090-9 Oncogene. (2014) 33:4213. doi: 10.1038/onc.2013.384

Frontiers in Oncology | www.frontiersin.org 11 October 2020 | Volume 10 | Article 587386


Mal et al. ERβ: A Ligand Activated Tumor Suppressor

65. Tzivion G, Dobson M, Ramakrishnan G. FoxO transcription factors; 84. Park C, Lee Y. Overexpression of ERβ is sufficient to inhibit hypoxia-
Regulation by AKT and 14-3-3 proteins. Biochim Biophys Acta. (2011) inducible factor-1 transactivation. Biochem Biophys Res Commun. (2014)
1813:1938–45. doi: 10.1016/j.bbamcr.2011.06.002 450:261–6. doi: 10.1016/j.bbrc.2014.05.107
66. Nakajima Y, Osakabe A, Waku T, Suzuki T, Akaogi K, Fujimura T, et al. 85. Mak P, Chang C, Pursell B, Mercurio AM. Estrogen receptor β sustains
Estrogen exhibits a biphasic effect on prostate tumor growth through epithelial differentiation by regulating prolyl hydroxylase 2 transcription.
the estrogen receptor β-KLF5 pathway. Mol Cell Biol. (2016) 36:144–56. Proc Natl Acad Sci U S A. (2013) 110:4708–13. doi: 10.1073/pnas.1221654110
doi: 10.1128/MCB.00625-15 86. Miller H, Poon S, Hibbert B, Rayner K, Chen YX, O’Brien ER. Modulation
67. Nakajima Y, Akaogi K, Suzuki T, Osakabe A, Yamaguchi C, Sunahara N, of estrogen signaling by the novel interaction of heat shock protein 27,
et al. Estrogen regulates tumor growth through a nonclassical pathway that a biomarker for atherosclerosis, and estrogen receptor beta: mechanistic
includes the transcription factors ERβ and KLF5. Sci Signal. (2011) 4:ra22. insight into the vascular effects of estrogens. Arteriosc Thromb Vasc Biol.
doi: 10.1126/scisignal.2001551 (2005) 25:e10–4. doi: 10.1161/01.ATV.0000156536.89752.8e
68. Marino M, Ascenzi P. Membrane association of estrogen receptor α and β 87. Slentz-Kesler K, Moore JT, Lombard M, Zhang J, Hollingsworth R,
influences 17β-estradiol-mediated cancer cell proliferation. Steroids. (2008) Weiner MP. Identification of the human Mnk2 gene (MKNK2) through
73:853–8. doi: 10.1016/j.steroids.2007.12.003 protein interaction with estrogen receptor beta. Genomics. (2000) 69:63–71.
69. Acconcia F, Totta P, Ogawa S, Cardillo I, Inoue S, Leone S, et al. Survival doi: 10.1006/geno.2000.6299
versus apoptotic 17β-estradiol effect. Role of ERα and ERβ activated non- 88. Lipovka Y, Chen H, Vagner J, Price TJ, Tsao TS, Konhilas JP. Oestrogen
genomic signaling. J Cell Physiol. (2005) 203:193–201. doi: 10.1002/jcp.20219 receptors interact with the α-catalytic subunit of AMP-activated protein
70. Lee Y-R, Park J, Yu H-N, Kim J-S, Youn HJ, Jung SH. Up-regulation kinase. Biosci Rep. (2015) 35:e00264. doi: 10.1042/BSR20150074
of PI3K/Akt signaling by 17β-estradiol through activation of estrogen 89. Hawse JR, Subramaniam M, Monroe DG, Hemmingsen AH, Ingle JN,
receptor-α, but not estrogen receptor-β, and stimulates cell growth in Khosla S, et al. Estrogen receptor beta isoform-specific induction of
breast cancer cells. Biochem Biophys Res Commun. (2005) 336:1221–6. transforming growth factor beta-inducible early gene-1 in human osteoblast
doi: 10.1016/j.bbrc.2005.08.256 cells: an essential role for the activation function 1 domain. Mol Endocrinol.
71. Totta P, Acconcia F, Leone S, Cardillo I, Marino M. Mechanisms of (2008) 22:1579–95. doi: 10.1210/me.2007-0253
naringenin-induced apoptotic cascade in cancer cells: involvement of 90. Chalaux E, López-Rovira T, Rosa JL, Pons G, Boxer LM, Bartrons R,
estrogen receptor a and ß signalling. IUBMB Life. (2004) 56:491–9. et al. A zinc-finger transcription factor induced by TGF-β promotes
doi: 10.1080/15216540400010792 apoptotic cell death in epithelial Mv1Lu cells. FEBS Lett. (1999) 457:478–82.
72. Bulzomi P, Bolli A, Galluzzo P, Leone S, Acconcia F, Marino M. Naringenin doi: 10.1016/S0014-5793(99)01051-0
and 17β-estradiol coadministration prevents hormone-induced human 91. Ribeiro A, Bronk SF, Roberts PJ, Urrutia R, Gores GJ. The transforming
cancer cell growth. IUBMB Life. (2010) 62:51–60. doi: 10.1002/iub.279 growth factor beta(1)-inducible transcription factor TIEG1, mediates
73. Virgili F, Acconcia F, Ambra R, Rinna A, Totta P, Marino M. Nutritional apoptosis through oxidative stress. Hepatology. (1999) 30:1490–7.
flavonoids modulate estrogen receptor α signaling. IUBMB Life. (2004) doi: 10.1002/hep.510300620
56:145–51. doi: 10.1080/15216540410001685083 92. Crider A, Nelson T, Davis T, Fagan K, Vaibhav K, Luo M, et al.
74. Marino M, Pellegrini M, La Rosa P, Acconcia F. Susceptibility of estrogen Estrogen Receptor β Agonist Attenuates endoplasmic reticulum stress-
receptor rapid responses to xenoestrogens: physiological outcomes. Steroids. induced changes in social behavior and brain connectivity in mice. Mol
(2012) 77:910–7. doi: 10.1016/j.steroids.2012.02.019 Neurobiol. (2018) 55:7606–18. doi: 10.1007/s12035-018-0929-8
75. Bulzomi P, Galluzzo P, Bolli A, Leone S, Acconcia F, Marino M. The pro- 93. Rajapaksa G, Nikolos F, Bado I, Clarke R, Gustafsson JÅ, Thomas C. ERβ
apoptotic effect of quercetin in cancer cell lines requires ERβ-dependent decreases breast cancer cell survival by regulating the IRE1/XBP-1 pathway.
signals. J Cell Physiol. (2012) 227:1891–8. doi: 10.1002/jcp.22917 Oncogene. (2015) 34:4130–41. doi: 10.1038/onc.2014.343
76. Bolli A, Galluzzo P, Ascenzi P, Del Pozzo G, Manco I, Vietri MT, et al. Laccase 94. Ding L, Yan J, Zhu J, Zhong H, Lu Q, Wang Z, et al. Ligand-independent
treatment impairs bisphenol A-induced cancer cell proliferation affecting activation of estrogen receptor alpha by XBP-1. Nucleic Acids Res. (2003)
estrogen receptor α-dependent rapid signals. IUBMB Life. (2008) 60:843–52. 31:5266–74. doi: 10.1093/nar/gkg731
doi: 10.1002/iub.130 95. Sengupta S, Sharma CG, Jordan VC. Estrogen regulation of X-box
77. Bolli A, Bulzomi P, Galluzzo P, Acconcia F, Marino M. Bisphenol A impairs binding protein-1 and its role in estrogen induced growth of breast and
estradiol-induced protective effects against DLD-1 colon cancer cell growth. endometrial cancer cells. Horm Mol Biol Clin Invest. (2010) 2:235–43.
IUBMB Life. (2010) 62:684–7. doi: 10.1002/iub.370 doi: 10.1515/hmbci.2010.025
78. Pan MH, Chiou YS, Chen LH, Ho CT. Breast cancer chemoprevention by 96. Williams C, Edvardsson K, Lewandowski SA, Ström A, Gustafsson JA. A
dietary natural phenolic compounds: specific epigenetic related molecular genome-wide study of the repressive effects of estrogen receptor beta on
targets. Mol Nutr Food Res. (2015) 59:21–35. doi: 10.1002/mnfr.201400515 estrogen receptor alpha signaling in breast cancer cells. Oncogene. (2008)
79. Chi F, Wu R, Zeng YC, Xing R, Liu Y, Xu ZG. Post-diagnosis soy food intake 27:1019–32. doi: 10.1038/sj.onc.1210712
and breast cancer survival: a meta-analysis of cohort studies. Asian Pac J 97. Lin C-Y, Ström A, Li Kong S, Kietz S, Thomsen JS, Tee JBS, et al. Inhibitory
Cancer Prev. (2013) 14:2407–12. doi: 10.7314/APJCP.2013.14.4.2407 effects of estrogen receptor beta on specific hormone-responsive gene
80. Uifălean A, Schneider S, Gierok P, Ionescu C, Iuga CA, Lalk M. The expression and association with disease outcome in primary breast cancer.
impact of soy isoflavones on MCF-7 and MDA-MB-231 breast cancer Breast Cancer Res. (2007) 9:R25. doi: 10.1186/bcr1667
cells using a global metabolomic approach. Int J Mol Sci. (2016) 17:1443. 98. Chang EC, Frasor J, Komm B, Katzenellenbogen BS. Impact of estrogen
doi: 10.3390/ijms17091443 receptor β on gene networks regulated by estrogen receptor α in breast cancer
81. Lu W, Katzenellenbogen BS. Estrogen Receptor-β modulation of the cells. Endocrinology. (2006) 147:4831–42. doi: 10.1210/en.2006-0563
ERα-p53 loop regulating gene expression, proliferation, and apoptosis in 99. Omoto Y, Eguchi H, Yamamoto-Yamaguchi Y, Hayashi S-i. Estrogen receptor
breast cancer. Horm Cancer. (2017) 8:230–42. doi: 10.1007/s12672-017- (ER) β1 and ERβcx/β2 inhibit ERα function differently in breast cancer cell
0298-1 line MCF7. Oncogene. (2003) 22:5011–20. doi: 10.1038/sj.onc.1206787
82. Ruegg J, Swedenborg E, Wahlstrom D, Escande A, Balaguer P, Pettersson 100. Stossi F, Barnett DH, Frasor J, Komm B, Lyttle CR, Katzenellenbogen
K, et al. The transcription factor aryl hydrocarbon receptor nuclear BS. Transcriptional profiling of estrogen-regulated gene expression via
translocator functions as an estrogen receptor beta-selective coactivator, and estrogen receptor (ER) α or ERβ in human osteosarcoma cells: distinct and
its recruitment to alternative pathways mediates antiestrogenic effects of common target genes for these receptors. Endocrinology. (2004) 145:3473–
dioxin. Mol Endocrinol. (2008) 22:304–16. doi: 10.1210/me.2007-0128 86. doi: 10.1210/en.2003-1682
83. Lim W, Park Y, Cho J, Park C, Park J, Park Y-K, et al. Estrogen receptor 101. Paruthiyil S, Cvoro A, Zhao X, Wu Z, Sui Y, Staub RE, et al.
beta inhibits transcriptional activity of hypoxia inducible factor-1 through Drug and cell type-specific regulation of genes with different classes
the downregulation of arylhydrocarbon receptor nuclear translocator. Breast of estrogen receptor beta-selective agonists. PLoS One. (2009) 4:e6271.
Cancer Res. (2011) 13:R32. doi: 10.1186/bcr2854 doi: 10.1371/journal.pone.0006271

Frontiers in Oncology | www.frontiersin.org 12 October 2020 | Volume 10 | Article 587386


Mal et al. ERβ: A Ligand Activated Tumor Suppressor

102. Tee MK, Rogatsky I, Tzagarakis-Foster C, Cvoro A, An J, Christy RJ, et al. to induce breast tumorigenesis. Breast Cancer Res. (2017) 19:79.
Estradiol and selective estrogen receptor modulators differentially regulate doi: 10.1186/s13058-017-0872-z
target genes with estrogen receptors alpha and beta. Mol Biol Cell. (2004) 121. Förster C, Mäkela S, Wärri A, Kietz S, Becker D, Hultenby K, et al.
15:1262–72. doi: 10.1091/mbc.e03-06-0360 Involvement of estrogen receptor β in terminal differentiation of mammary
103. Chada S, Mhashilkar AM, Ramesh R, Mumm JB, Sutton RB, Bocangel D, gland epithelium. Proc Natl Acad Sci U S A. (2002) 99:15578–83.
et al. Bystander activity of Ad-mda7: human MDA-7 protein kills melanoma doi: 10.1073/pnas.192561299
cells via an IL-20 receptor-dependent but STAT3-independent mechanism. 122. Wada-Hiraike O, Imamov O, Hiraike H, Hultenby K, Schwend T, Omoto Y,
Mol Therapy. (2004) 10:1085–95. doi: 10.1016/j.ymthe.2004.08.020 et al. Role of estrogen receptor beta in colonic epithelium. Proc Natl Acad Sci
104. Monroe DG, Getz BJ, Johnsen SA, Riggs BL, Khosla S, Spelsberg U S A. (2006) 103:2959–64. doi: 10.1073/pnas.0511271103
TC. Estrogen receptor isoform-specific regulation of endogenous gene 123. Patrone C, Cassel TN, Pettersson K, Piao YS, Cheng G, Ciana P,
expression in human osteoblastic cell lines expressing either ERα or ERβ. et al. Regulation of postnatal lung development and homeostasis
J Cell Biochem. (2003) 90:315–26. doi: 10.1002/jcb.10633 by estrogen receptor beta. Mol Cell Biol. (2003) 23:8542–52.
105. Zhao L, Huang S, Mei S, Yang Z, Xu L, Zhou N, et al. Pharmacological doi: 10.1128/MCB.23.23.8542-8552.2003
activation of estrogen receptor beta augments innate immunity to suppress 124. Morani A, Barros RP, Imamov O, Hultenby K, Arner A, Warner M,
cancer metastasis. Proc Natl Acad Sci U S A. (2018) 115:E3673–E81. et al. Lung dysfunction causes systemic hypoxia in estrogen receptor beta
doi: 10.1073/pnas.1803291115 knockout (ERβ−/− ) mice. Proc Natl Acad Sci U S A. (2006) 103:7165–9.
106. Liu SG, Wu XX, Hua T, Xin XY, Feng DL, Chi SQ, et al. NLRP3 doi: 10.1073/pnas.0602194103
inflammasome activation by estrogen promotes the progression of 125. Windahl SH, Hollberg K, Vidal O, Gustafsson JA, Ohlsson C, Andersson
human endometrial cancer. Oncotargets Therapy. (2019) 12:6927–36. G. Female estrogen receptor β−/− mice are partially protected against
doi: 10.2147/OTT.S218240 age-related trabecular bone loss. J Bone Miner Res. (2001) 16:1388–98.
107. Dyson MT, Bulun SE. Cutting SRC-1 down to size in endometriosis. Nat doi: 10.1359/jbmr.2001.16.8.1388
Med. (2012) 18:1016–8. doi: 10.1038/nm.2855 126. Windahl SH, Vidal O, Andersson G, Gustafsson JA, Ohlsson C. Increased
108. Han SJ, Jung SY, Wu SP, Hawkins SM, Park MJ, Kyo S, et al. cortical bone mineral content but unchanged trabecular bone mineral
Estrogen receptor β modulates apoptosis complexes and the inflammasome density in female ERβ−/− mice. J Clin Invest. (1999) 104:895–901.
to drive the pathogenesis of endometriosis. Cell. (2015) 163:960–74. doi: 10.1172/JCI6730
doi: 10.1016/j.cell.2015.10.034 127. Hawse JR, Carter JM, Aspros KGM, Bruinsma ES, Koepplin JW, Negron V,
109. Cvoro A, Tatomer D, Tee M-K, Zogovic T, Harris HA, Leitman DC. Selective et al. Optimized immunohistochemical detection of estrogen receptor beta
estrogen receptor-β agonists repress transcription of proinflammatory genes. using two validated monoclonal antibodies confirms its expression in normal
J Immunol. (2008) 180:630–6. doi: 10.4049/jimmunol.180.1.630 and malignant breast tissues. Breast Cancer Res Treat. (2020) 179:241–9.
110. Xiao L, Luo Y, Tai R, Zhang N. Estrogen receptor β suppresses inflammation doi: 10.1007/s10549-019-05441-3
and the progression of prostate cancer. Mol Med Rep. (2019) 19:3555–63. 128. Maehle BO, Collett K, Tretli S, Akslen LA, Grotmol T. Estrogen
doi: 10.3892/mmr.2019.10014 receptor beta–an independent prognostic marker in estrogen receptor alpha
111. Wu W-f, Tan X-j, Dai Y-b, Krishnan V, Warner M, Gustafsson J-Å. and progesterone receptor-positive breast cancer? Acta Pathol Microbiol
Targeting estrogen receptor β in microglia and T cells to treat experimental Immunol Scand. (2009) 117:644–50. doi: 10.1111/j.1600-0463.2009.02510.x
autoimmune encephalomyelitis. Proc Natl Acad Sci U S A. (2013) 110:3543– 129. Mann S, Laucirica R, Carlson N, Younes PS, Ali N, Younes A, et al. Estrogen
8. doi: 10.1073/pnas.1300313110 receptor beta expression in invasive breast cancer. Human Pathol. (2001)
112. Krege JH, Hodgin JB, Couse JF, Enmark E, Warner M, Mahler 32:113–8. doi: 10.1053/hupa.2001.21506
JF, et al. Generation and reproductive phenotypes of mice lacking 130. Tan W, Li Q, Chen K, Su F, Song E, Gong C. Estrogen receptor beta as a
estrogen receptor β. Proc Natl Acad Sci U S A. (1998) 95:15677–82. prognostic factor in breast cancer patients: a systematic review and meta-
doi: 10.1073/pnas.95.26.15677 analysis. Oncotarget. (2016) 7:10373–85. doi: 10.18632/oncotarget.7219
113. Ogawa S, Chester AE, Hewitt SC, Walker VR, Gustafsson J-Å, Smithies 131. Marotti JD, Collins LC, Hu R, Tamimi RM. Estrogen receptor-beta
O, et al. Abolition of male sexual behaviors in mice lacking estrogen expression in invasive breast cancer in relation to molecular phenotype:
receptors α and β (αβERKO). Proc Natl Acad Sci U S A. (2000) 97:14737–41. results from the Nurses’ Health Study. Modern Pathol. (2010) 23:197–204.
doi: 10.1073/pnas.250473597 doi: 10.1038/modpathol.2009.158
114. Ogawa S, Taylor JA, Lubahn DB, Korach KS, Pfaff DW. Reversal of sex 132. Maruyama S, Fujimoto N, Asano K, Ito A. Suppression by estrogen
roles in genetic female mice by disruption of estrogen receptor gene. receptor beta of AP-1 mediated transactivation through estrogen
Neuroendocrinology. (1996) 64:467. doi: 10.1159/000127154 receptor alpha. J Steroid Biochem Mol Biol. (2001) 78:177–84.
115. Ogawa S, Eng V, Taylor J, Lubahn DB, Korach KS, Pfaff DW. Roles of estrogen doi: 10.1016/S0960-0760(01)00083-8
receptor-α gene expression in reproduction-related behaviors in female mice. 133. Gougelet A, Mueller SO, Korach KS, Renoir JM. Oestrogen receptors
Endocrinology. (1998) 139:5070–81. doi: 10.1210/endo.139.12.6357 pathways to oestrogen responsive elements: the transactivation function-1
116. Ogawa S, Washburn TF, Taylor J, Lubahn DB, Korach KS, Pfaff DW. acts as the keystone of oestrogen receptor (ER)β-mediated transcriptional
Modifications of testosterone-dependent behaviors by estrogen receptor- repression of ERα. J Steroid Biochem Mol Biol. (2007) 104:110–22.
α gene disruption in male mice. Endocrinology. (1998) 139:5058–69. doi: 10.1016/j.jsbmb.2007.03.002
doi: 10.1210/endo.139.12.6358 134. Saville B, Wormke M, Wang F, Nguyen T, Enmark E, Kuiper G, et al. Ligand-
117. Rissman EF, Wersinger SR, Taylor JA, Lubahn DB. Estrogen receptor , cell-, and estrogen receptor subtype (alpha/beta)-dependent activation
function as revealed by knockout studies: neuroendocrine and behavioral at GC-rich (Sp1) promoter elements. J Biol Chem. (2000) 275:5379–87.
aspects. Horm Behav. (1997) 31:232–43. doi: 10.1006/hbeh.1997.1390 doi: 10.1074/jbc.275.8.5379
118. Shim GJ, Wang L, Andersson S, Nagy N, Kis LL, Zhang Q, et al. Disruption 135. Reese JM, Bruinsma ES, Nelson AW, Chernukhin I, Carroll JS, Li Y, et al. ERβ
of the estrogen receptor beta gene in mice causes myeloproliferative -mediated induction of cystatins results in suppression of TGFbeta signaling
disease resembling chronic myeloid leukemia with lymphoid blast crisis. and inhibition of triple-negative breast cancer metastasis. Proc Natl Acad Sci
Proc Natl Acad Sci U S A. (2003) 100:6694–9. doi: 10.1073/pnas.07318 U S A. (2018) 115:E9580–E9. doi: 10.1073/pnas.1807751115
30100 136. Ohshiro K, Schwartz AM, Levine PH, Kumar R. Alternate estrogen receptors
119. Imamov O, Morani A, Shim GJ, Omoto Y, Thulin-Andersson C, Warner M, promote invasion of inflammatory breast cancer cells via non-genomic
et al. Estrogen receptor beta regulates epithelial cellular differentiation in signaling. PLoS One. (2012) 7:e30725. doi: 10.1371/journal.pone.0030725
the mouse ventral prostate. Proc Natl Acad Sci U S A. (2004) 101:9375–80. 137. Hamilton N, Márquez-Garbán D, Mah V, Fernando G, Elshimali Y, Garbán
doi: 10.1073/pnas.0403041101 H, et al. Biologic roles of estrogen receptor-β and insulin-like growth factor-
120. Bado I, Nikolos F, Rajapaksa G, Wu W, Castaneda J, Krishnamurthy 2 in triple-negative breast cancer. BioMed Res Int. (2015) 2015:925703.
S, et al. Somatic loss of estrogen receptor beta and p53 synergize doi: 10.1155/2015/925703

Frontiers in Oncology | www.frontiersin.org 13 October 2020 | Volume 10 | Article 587386


Mal et al. ERβ: A Ligand Activated Tumor Suppressor

138. Christoforou P, Christopoulos PF, Koutsilieris M. The role of potential marker of ovarian carcinogenesis. Cancer Res. (1998) 58:53
estrogen receptor β in prostate cancer. Mol Med. (2014) 20:427–34. 67–73.
doi: 10.2119/molmed.2014.00105 154. Rutherford T, Brown WD, Sapi E, Aschkenazi S, Munoz A, Mor G.
139. Kuiper G, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson J-A. Cloning of Absence of estrogen receptor-beta expression in metastatic ovarian cancer.
a novel receptor expressed in rat prostate and ovary. Proc Natl Acad Sci U S Obstet Gynecol. (2000) 96:417–21. doi: 10.1097/00006250-200009000-
A. (1996) 93:5925–30. doi: 10.1073/pnas.93.12.5925 00018
140. Carey MA, Card JW, Voltz JW, Germolec DR, Korach KS, Zeldin DC. The 155. Chan KKL, Siu MKY, Jiang YX, Wang JJ, Wang Y, Leung THY, et al.
impact of sex and sex hormones on lung physiology and disease: lessons Differential expression of estrogen receptor subtypes and variants in ovarian
from animal studies. Am J Physiol Lung Cell Mol Physiol. (2007) 293:L272–8. cancer: effects on cell invasion, proliferation and prognosis. BMC Cancer.
doi: 10.1152/ajplung.00174.2007 (2017) 17:606. doi: 10.1186/s12885-017-3601-1
141. Brandenberger AW, Tee MK, Lee JY, Chao V, Jaffe RB. Tissue distribution 156. Williams C, DiLeo A, Niv Y, Gustafsson JA. Estrogen receptor beta as
of estrogen receptors alpha (ER-α) and beta (ER-β) mRNA in the target for colorectal cancer prevention. Cancer Lett. (2016) 372:48–56.
midgestational human fetus. J Clin Endocrinol Metab. (1997) 82:3509–12. doi: 10.1016/j.canlet.2015.12.009
doi: 10.1210/jc.82.10.3509 157. Zeng Q, Chen GG, Vlantis AC, van Hasselt CA. Oestrogen
142. Hershberger PA, Stabile LP, Kanterewicz B, Rothstein ME, Gubish CT, Land mediates the growth of human thyroid carcinoma cells via an
S, et al. Estrogen receptor beta (ERβ) subtype-specific ligands increase oestrogen receptor-ERK pathway. Cell Prolif. (2007) 40:921–35.
transcription, p44/p42 mitogen activated protein kinase (MAPK) activation doi: 10.1111/j.1365-2184.2007.00471.x
and growth in human non-small cell lung cancer cells. J Steroid Biochem Mol 158. Dong W, Zhang H, Li J, Guan H, He L, Wang Z, et al. Estrogen
Biol. (2009) 116:102–9. doi: 10.1016/j.jsbmb.2009.05.004 induces metastatic potential of papillary thyroid cancer cells through
143. Hiramitsu S, Ishikawa T, Lee W-R, Khan T, Crumbley C, Khwaja N, estrogen receptor alpha and beta. Int J Endocrinol. (2013) 2013:941568.
et al. Estrogen receptor beta-mediated modulation of lung cancer cell doi: 10.1155/2013/941568
proliferation by 27-hydroxycholesterol. Front Endocrinol. (2018) 9:470. 159. Dong WW, Li J, Li J, Zhang P, Wang ZH, Sun W, et al. Reduced expression of
doi: 10.3389/fendo.2018.00470 oestrogen receptor-beta is associated with tumour invasion and metastasis in
144. Ding X, Li L, Tang C, Meng C, Xu W, Wei X, et al. Cytoplasmic oestrogen receptor-alpha-negative human papillary thyroid carcinoma. Int J
expression of estrogen receptor β may predict poor outcome of EGFR-TKI Exp Pathol. (2018) 99:15–21. doi: 10.1111/iep.12266
therapy in metastatic lung adenocarcinoma. Oncol Lett. (2018) 16:2382–90. 160. Dong W, Li J, Zhang H, Huang Y, He L, Wang Z, et al. Altered expression
doi: 10.3892/ol.2018.8936 of estrogen receptor beta2 is associated with different biological markers and
145. Baik CS, Eaton KD. Estrogen signaling in lung cancer: an opportunity for clinicopathological factors in papillary thyroid cancer. Int J Clin Exp Pathol.
novel therapy. Cancers. (2012) 4:969–88. doi: 10.3390/cancers4040969 (2015) 8:7149–56.
146. Kawai H. Estrogen receptors as the novel therapeutic biomarker 161. Dong W, Li J, Huang Y, Zhang H, Shan Z, Teng W. Differential expression
in non-small cell lung cancer. World J Clin Oncol. (2014) 5:1020. patterns of estrogen receptor (ER)-beta splice variants between papillary
doi: 10.5306/wjco.v5.i5.1020 thyroid cancer and nodular thyroid goiter. Med Sci Monit. (2012) 18:Br351–
147. Hsu L-H, Chu N-M, Kao S-H. Estrogen, estrogen receptor and lung cancer. 5. doi: 10.12659/MSM.883344
Int J Mol Sci. (2017) 18:1713. doi: 10.3390/ijms18081713 162. Cho MA, Lee MK, Nam KH, Chung WY, Park CS, Lee JH, et al. Expression
148. Zhang G, Liu X, Farkas AM, Parwani AV, Lathrop KL, Lenzner D, et al. and role of estrogen receptor alpha and beta in medullary thyroid carcinoma:
Estrogen receptor β functions through nongenomic mechanisms in lung different roles in cancer growth and apoptosis. J Endocrinol. (2007) 195:255–
cancer cells. Mol Endocrinol. (2009) 23:146–56. doi: 10.1210/me.2008-0431 63. doi: 10.1677/JOE-06-0193
149. Liu J, Sareddy GR, Zhou M, Viswanadhapalli S, Li X, Lai Z, et al. Differential 163. Yu CP, Ho JY, Huang YT, Cha TL, Sun GH, Yu DS, et al. Estrogen
effects of estrogen receptor beta isoforms on glioblastoma progression. inhibits renal cell carcinoma cell progression through estrogen receptor-
Cancer Res. (2018) 78:3176–89. doi: 10.1158/0008-5472.CAN-17-3470 beta activation. PLoS One. (2013) 8:e56667. doi: 10.1371/journal.pone.00
150. Pierdominici M, Maselli A, Locatelli SL, Ciarlo L, Careddu G, 56667
Patrizio M, et al. Estrogen receptor beta ligation inhibits Hodgkin
lymphoma growth by inducing autophagy. Oncotarget. (2017) 8:8522–35. Conflict of Interest: The authors declare that the research was conducted in the
doi: 10.18632/oncotarget.14338 absence of any commercial or financial relationships that could be construed as a
151. Schuler-Toprak S, Weber F, Skrzypczak M, Ortmann O, Treeck O. potential conflict of interest.
Estrogen receptor beta is associated with expression of cancer associated
genes and survival in ovarian cancer. BMC Cancer. (2018) 18:981. Copyright © 2020 Mal, Magner, David, Datta, Vallabhaneni, Kassem, Manouchehri,
doi: 10.1186/s12885-018-4898-0 Willingham, Stover, Vandeusen, Sardesai, Williams, Wesolowski, Lustberg, Ganju,
152. De Stefano I, Zannoni GF, Prisco MG, Fagotti A, Tortorella L, Vizzielli G, Ramaswamy and Cherian. This is an open-access article distributed under the terms
et al. Cytoplasmic expression of estrogen receptor beta (ERβ) predicts poor of the Creative Commons Attribution License (CC BY). The use, distribution or
clinical outcome in advanced serous ovarian cancer. Gynecol Oncol. (2011) reproduction in other forums is permitted, provided the original author(s) and the
122:573–9. doi: 10.1016/j.ygyno.2011.05.025 copyright owner(s) are credited and that the original publication in this journal
153. Pujol P, Rey JM, Nirde P, Roger P, Gastaldi M, Laffargue F, et al. Differential is cited, in accordance with accepted academic practice. No use, distribution or
expression of estrogen receptor-alpha and -beta messenger RNAs as a reproduction is permitted which does not comply with these terms.

Frontiers in Oncology | www.frontiersin.org 14 October 2020 | Volume 10 | Article 587386

You might also like