You are on page 1of 17

17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022].

See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MINIREVIEW

Gonadotropin-releasing hormone: GnRH receptor signaling


in extrapituitary tissues
Lydia W. T. Cheung and Alice S. T. Wong
School of Biological Sciences, University of Hong Kong, China

Keywords Gonadotropin-releasing hormone (GnRH) has historically been known as


cross-talk; extrapituitary; GnRH; GnRH a pituitary hormone; however, in the past few years, interest has been
receptor; MAPK; metastasis; pituitary;
raised in locally produced, extrapituitary GnRH. GnRH receptor
receptor tyrosine kinase; signaling; tumor
(GnRHR) was found to be expressed in normal human reproductive tissues
Correspondence (e.g. breast, endometrium, ovary, and prostate) and tumors derived from
A. S. T. Wong, School of Biological these tissues. Numerous studies have provided evidence for a role of GnRH
Sciences, University of Hong Kong, 4S-14 in cell proliferation. More recently, we and others have reported a novel
Kadoorie Biological Sciences Building, role for GnRH in other aspects of tumor progression, such as metastasis
Pokfulam Road, Hong Kong, China and angiogenesis. The multiple actions of GnRH could be linked to the
Fax: +852 2559 9114
divergence of signaling pathways that are activated by GnRHR. Recent
Tel: +852 2299 0865
E-mail: awong1@hku.hk
observations also demonstrate cross-talk between GnRHR and growth fac-
tor receptors. Intriguingly, the classical Gaq–11-phospholipase C signal
(Received 14 April 2008, revised 28 May transduction pathway, known to function in pituitary gonadotropes, is not
2008, accepted 11 June 2008) involved in GnRH actions at nonpituitary targets. Herein, we review the
key findings on the role of GnRH in the control of tumor growth, progres-
doi:10.1111/j.1742-4658.2008.06677.x sion, and dissemination. The emerging role of GnRHR in actin cytoskele-
ton remodeling (small Rho GTPases), expression and ⁄ or activity of
adhesion molecules (integrins), proteolytic enzymes (matrix metalloprotein-
ases) and angiogenic factors is explored. The signal transduction mecha-
nisms of GnRHR in mediating these activities is described. Finally, we
discuss how a common GnRHR may mediate different, even opposite,
responses to GnRH in the same tissue ⁄ cell type and whether an additional
receptor(s) for GnRH exists.

gonadotropins, luteinizing hormone and follicle-stimu-


Introduction
lating hormone, which in turn stimulate the gonads
The hypothalamic gonadotropin-releasing hormone for steroid production. Subsequently, a second iso-
(GnRH) is a decapeptide that plays a critical role in form of GnRH (His5, Trp7, Tyr8) (GnRH-II) has
the regulation of reproduction. GnRH-I (pGlu-His- been isolated from chicken brain. It is also highly
Trp-Ser-Tyr-Gly-Leu-Arg-Pro-Gly-NH2) is the first conserved among vertebrates, including mammals [1].
GnRH isoform discovered in mammalian brain. Its However, in contrast to GnRH-I, GnRH-II is
major role is to stimulate pituitary secretion of expressed at significantly higher levels outside the

Abbreviations
EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; ERK, extracellular signal-related kinase; FAK, focal adhesion
kinase; FGF, fibroblast growth factor; GnRH, gonadotropin-releasing hormone; GnRHR, gonadotropin-releasing hormone receptor;
JNK, Jun N-terminal kinase; MAPK, mitogen-activated protein kinase; MMP, matrix metalloproteinase; NF-jB, nuclear factor kappa B; PI3K,
phosphatidylinositol 3-kinase; PKC, protein kinase C; Pyk2, proline-rich tyrosine kinase 2; RTK, receptor tyrosine kinase; uPA, urokinase-type
plasminogen activator; VEGF, vascular endothelial growth factor.

FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS 5479
17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
GnRH receptor signaling L. W. T. Cheung and A. S. T. Wong

brain and is particularly abundant in the kidney, their tissue of origin, ovarian surface epithelium [9,10].
bone marrow, and prostate [2]. This leads to the Interestingly, levels of GnRHR seem to be associated
speculation that GnRH-II may have distinct physio- with cancer grading and have been reported to be
logical functions from those of GnRH-I. In line with elevated in advanced stage (stages III and IV) as
this is the observation that although GnRH-II can compared to early stage (stages I and II) ovarian
stimulate gonadotropin secretion, its efficiency is carcinomas [11]. Our recent findings that GnRH can
much lower than that of GnRH-I (only about 2% of promote the motility and invasiveness of ovarian can-
that of GnRH-I) [3]. This suggests that the primary cer cells further corroborate the view that GnRH may
role of GnRH-II is not in the regulation of gonado- play a crucial role in tumor progression ⁄ metastasis
tropin secretion. Instead, this peptide has been shown [12,13], and these findings will be discussed in a later
to act as a neuromodulator [4]. The exact actions of section.
GnRH-II in peripheral tissues are not entirely under- Using [125I][d-Trp6]GnRH, specific receptor binding
stood, but this is certainly an important topic for has been detected in membranes from 24 of 31 (77%)
investigation which may offer an opportunity to eluci- endometrial carcinomas and from three of 13 (23.1%)
date the undisclosed complexity of GnRH. nonmalignant human endometrial specimens [14].
In this minireview, we will focus on recent progress GnRHR mRNA has been clearly detected in surgical
in understanding the roles of GnRH-I and GnRH-II endometrial carcinoma specimens and endometrial
in extrapituitary tissues, in particular its emerging carcinoma cell lines [15,16]. As with normal myome-
role in tumor growth, invasion, and metastasis. We trium, most benign neoplasms studied thus far,
will also describe the molecular mechanisms underlying including uterine leiomyoma, also possess GnRHR
these effects, focusing on the roles of proteolysis, [17].
adhesion, and signaling, as well as our still-emerging Early studies showed that the human placenta con-
understanding of receptor cross-talk with other tains specific binding sites for GnRH that interact with
pathways. Finally, we will discuss two important GnRH agonists and antagonists [18]. Later on,
outstanding questions in the field regarding what might GnRHR was localized to the cytotrophoblast and
distinguish the different responses to the same ligand syncytiotrophoblast cell layers [19,20]. Temporal
(GnRH) and whether an additional receptor(s) for expression of GnRHR in the placental cells at different
GnRH exists in humans. weeks of gestation has been observed, in parallel with
the time-course of chorionic gonadotropin secretion
during pregnancy [21], suggesting that the expression
Localization of GnRH receptor (GnRHR)
of the receptor is a function of pregnancy stage.
in peripheral reproductive tissues
The presence of GnRHR has been demonstrated in
The initial interest in extrapituitary GnRHR stemmed numerous human breast cancer cell lines and tumor
primarily from observations in the 1980s that GnRH biopsy specimens [22–24]. GnRHR was immunolocal-
analogs can inhibit the growth of nonpituitary tumor ized in the cytoplasm in 37 of 58 (64%) invasive ductal
cell lines [5]. Soon after this, a functional type I carcinoma cases [23]. The expression of GnRHR in
GnRHR was demonstrated in a variety of normal normal human breast tissue is still controversial, but
human reproductive tissues (e.g. breast, endometrium, the sample size may have been too small to allow any
ovary, and prostate) and tumors derived from these definite conclusion [22,25].
tissues. GnRHR is also present in prostate cancer cells, as
In the ovary, GnRHR mRNAs are expressed in shown by radioligand-binding studies, PCR, and
granulosa-luteal cells, and increased expression of western blotting analysis [26,27]. GnRHR immunore-
GnRHR correlates with follicular growth and develop- activity is localized to the luminal and basal epithelial
ment [6]. GnRHR binding has been demonstrated in cells in benign and malignant prostate tissues. In this
luteinized granulosa cells, late follicles and developing study, the relative GnRHR mRNA levels showed a wide
corpora lutea, but not in primordial, early antral and range of individual differences that were unrelated to
preovulatory follicles [7,8]. This stage-specific expres- the histological grades of the 16 cases [27]. There does,
sion of GnRHR in the human granulosa and luteal however, appear to be significantly higher expression of
cells suggests a role for GnRH in the regulation of GnRHR in hormone-refractory prostate carcinoma
ovarian physiology, particularly ovulation, follicular than in other types of prostate tumor (n = 80) [28].
atresia and luteolysis. The presence of GnRHR protein Although these extrapituitary GnRHRs share the
and mRNA has also been demonstrated in human same cDNA nucleotide sequence and encode tran-
ovarian tumor specimens, ovarian cancer cell lines and scripts and proteins of the same size as the pituitary

5480 FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS
17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
L. W. T. Cheung and A. S. T. Wong GnRH receptor signaling

GnRHR [20,26,29], they also differ in several ways. GnRH-II has antiproliferative effects on ovarian cancer
First, cell surface receptor expression in extrapituitary cells [41–43]. Although it has been suggested that this
sites is low as compared to that of the pituitary effect of GnRH-II is mediated through the type I
[15,27]. This may underlie the greater effect of the GnRHR [43], there are other findings implicating a
GnRHR ligands on the gonadotropes. Second, there type I GnRHR-independent action [41,42].
are at least two classes of GnRHR: one has high affin- It is interesting to note that although both GnRH-I
ity [with nanomolar dissociation constants (Kd)] for agonists and antagonists exert antiproliferative effects,
GnRH, and one has low affinity (with micromolar Kd the effects of GnRH-I antagonists are stronger than
values) for GnRH. The high-affinity GnRH-binding those of the agonists [44]. This difference has also been
sites are commonly regarded as being the same as the seen in an in vivo model, which demonstrates a signifi-
GnRHR of the pituitary gland. Whereas in most of cant inhibition of tumor growth by GnRH-I antago-
the reported cases, both the low-affinity and high-affin- nists but not GnRH-I agonists [45]. The advantage of
ity GnRHR have been found in extrapituitary tissues GnRH antagonists over the agonistic peptides is prob-
[30–33], in some cases, only low-affinity GnRHR could ably due to the fact that they inhibit the secretion of
be detected [10,18,34], and in others, e.g. in endome- gonadotropins and reduce sex steroid levels immedi-
trial cancers and nonmalignant endometrial specimens, ately after application, thus achieving rapid therapeutic
only the high-affinity GnRHR has been demonstrated effects, whereas repeated exposure to agonistic agents
[14]. The exact role of each of these receptors and the is required to induce functional desensitization of the
implications of differential levels of expression remain gonadotropes [46].
to be elucidated. Treatment of human endometrial cancer cells (cell
line Ishikawa) with the GnRH-I antagonist SB-75
results in growth inhibition, mainly due to the Fas ⁄ Fas
Functions of GnRH-I and GnRH-II in
ligand-mediated apoptotic pathway, whereas GnRH-I
cancers
agonists have no effect on the same cell line [15,47,48].
Another endometrial carcinoma cell line, HEC-1A, also
Tumor growth
exhibits differential responses to different GnRH agon-
Over the last two decades, both GnRH agonists and ists and antagonists [15,30,36,48]. GnRH-II has been
antagonists have been widely used as therapeutics in shown to have antiproliferative effects on endometrial
treating sex steroid-dependent tumors. The majority carcinoma cells [41]. The effects of GnRH-I are
of these GnRH analogs, when given continuously, abrogated after type I GnRHR knockout [36], whereas
inhibit gonadotropin synthesis and secretion via those of the GnRH-I antagonist cetrorelix and of
downregulation of the pituitary GnRHRs. This indi- GnRH-II persist [41]. These findings suggest that the
rect mechanism of action has provided the rationale antiproliferative effects of cetrorelix and GnRH-II are
for the use of GnRH analogs in the treatment of hor- not mediated through the type I GnRHR.
mone-dependent tumors for many years. Only since GnRH-I has been demonstrated to have antiprolifer-
the detection of GnRHR in extrapituitary tissues has ative effects on prostate cancer cells [49–51], except in
there been increasing interest in its direct action on one in vivo study [52]. This antiproliferative effect
tumor cells. appears to be independent of the androgen receptor
GnRH-I analogs have direct antiproliferative effects status of the prostate carcinoma cells, as both andro-
on ovarian cancer cells, which is linked to the disrup- gen-sensitive LNCaP cells and androgen-resistant
tion of the cell cycle at G0 ⁄ G1 [31,35,36]. On the other DU-145 cells remain sensitive to GnRH [49,50]. Acti-
hand, several independent in vitro studies failed to vation of GnRHR may mediate these effects via direct
demonstrate significant growth inhibition by GnRH-I induction of apoptosis through caspase activation [53].
agonists, even at fairly high concentrations (micromolar Compatible with a role for GnRH in survival at low
range) [37,38]. In fact, a biphasic impact of GnRH-I doses, an enhancing effect of GnRH was observed
agonists on growth has been reported: whereas GnRH-I when cells were treated with a low concentration
agonists at high dose (1 lm) inhibit cell proliferation (100 pm) of GnRH-I agonist [54]. GnRH-II was shown
in vitro, cells treated with agonists at low dose (10 nm) to have an antiproliferative effect on DU-145 cells and
show significant growth stimulation [39]. Further growth-stimulatory effect on TSU-Pr1 cells, but the
studies demonstrated that nanomolar concentrations of type I GnRHR was not involved [55].
GnRH-I agonists also increase cell survival under The influence of GnRH on the growth of human
multiple stress conditions, including DNA replication- breast cancer cells was first studied with MCF-7 cells
specific cytotoxic agents and UV radiation [40]. [56], and both in vitro and in vivo proliferation of

FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS 5481
17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
GnRH receptor signaling L. W. T. Cheung and A. S. T. Wong

breast cancer cells could be inhibited by both agonistic GnRH-I and GnRH-II on cell invasion supports the
and antagonistic analogs of GnRH [57,58]. However, view that the same receptor, type I GnRHR, is essen-
higher efficiency of GnRH antagonists in growth inhi- tial for the effects of GnRH-I and GnRH-II in ovarian
bition than that of GnRH agonists has been reported cancer cells.
[24,58]. The decrease in uPA activity of cytosol from Dun-
ning R3327H rat prostate tumors after treatment with
GnRH-I analogs suggests that GnRH may be an
Invasion and metastasis
important factor in reducing the invasiveness of pros-
The observation that GnRH controls tumor growth tate cancer [63]. High doses of GnRH-I agonists and
suggests a regulatory role for this peptide in the meta- antagonists have been reported to attenuate the
static behavior of cancer cells. This hypothesis is sup- invading capacity of both androgen-dependent and
ported by studies showing that GnRH-I and GnRH-II androgen-independent prostate cancer cells by modu-
can affect the expression of several extracellular lating E-cadherin-mediated cell–cell contacts and pro-
matrix-degrading enzymes in human extravillous cyto- duction of uPA and its inhibitor (plasminogen
trophoblasts and decidual stromal cells to facilitate activator inhibitor-1) [64–66]. GnRH has also been
implantation [59,60]. However, its potential role in shown to regulate cell motility through its interaction
cancer metastasis has just begun to be revealed. with the small GTPases Rac1, Cdc42, and RhoA,
Metastasis is a complex phenomenon that requires which are involved in the regulation of actin polymer-
several specific steps, such as decreased adhesion, ization [67].
increased motility, and proteolysis. The effects of GnRH Up to now, there has been only one study, by Von
in tumor metastasis are mediated through the regulation Alten et al., investigating the role of GnRH in breast
of adhesion molecules, Rho GTPases, and two families cancer metastasis, using a coculture system with
of metastasis-related proteinases, the matrix metallopro- human osteosarcoma cells to analyze tumor cell
teinases (MMPs) and the urokinase-type plasminogen invasion to bone [68]. The consequences of GnRHR
activator (uPA) system, at several levels: mRNA activation are complex and appear to be cell context
transcription, secretion, and proenzyme activation. dependent: whereas treatment of cells with the
The ability of GnRH to regulate metastasis was first GnRH-I agonist triptorelin, the GnRH-II agonist
reported in melanoma cells [61]. High doses of GnRH-I [d-Lys6]GnRH-II and the GnRH-I antagonist cetrorelix
analog, at micromolar concentrations, significantly decreases the invasion rate in most breast cancer cell
reduces the ability of melanoma cells to invade lines, these agents have no significant effect in the
and migrate [61]. Preliminary data (R. M. Moretti, GnRHR-positive MDA-MB-435 cells [68]. Further
M. Monagnani Marelli, J. C. van Groeninghen, M. investigations are required to elucidate the reason why
Motta & P. Limonta, unpublished results, 2003) indicate the MDA-MB-435 cell line reacts differently.
that this inhibitory action is due to the effects of Organ-specific homing and colonization of cancer
integrins and MMPs [62]. cells are important and interesting features of metasta-
We were the first to report possible metastatic activ- sis. A role for GnRH has also been suggested in the
ity of GnRH-I in tumors of the female reproductive regulation of the immune response and metastasis.
tract [12]. GnRH-I exerts a biphasic effect on cellular GnRH-I and GnRH-II are expressed in human normal
migration and invasion: whereas lower (nanomolar) and cancerous T-cells. GnRH triggers laminin receptor
concentrations of the GnRH-I agonist stimulate cellu- gene expression, adhesion to laminin, in vitro chemo-
lar migration and invasion in a dose-dependent man- taxis, and in vivo homing to specific organs [69].
ner, high (micromolar) concentrations are not as
efficient. This proinvasive effect is mediated through
Angiogenesis
activation of metastasis-related proteinases, in particu-
lar MMP-2 and MMP-9 [12]. Moreover, GnRH-I is Angiogenesis is crucial to a number of physiological
able to transactivate the MMP-2 and MMP-9 promot- and pathological processes, such as reproduction,
ers, which means that GnRH can be considered to be development, and tissue repair, as well as tumor
a new member of MMP-2 and MMP-9 transcriptional growth and metastasis. Vascular endothelial growth
modulators. Like GnRH-I, native GnRH-II and its factor (VEGF) is implicated as the most important
synthetic analog also induce a similar biphasic regula- angiogenesis inducer, because of its potency in a
tion of ovarian cancer invasion [13]. The finding that variety of normal and tumor cells. Other angiogenic
small interfering RNA-mediated downregulation of factors include fibroblast growth factor (FGF), plate-
type I GnRHR completely reversed the effects of both let-derived growth factor and the angiopoietin family.

5482 FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS
17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
L. W. T. Cheung and A. S. T. Wong GnRH receptor signaling

The effect of GnRH on angiogenesis in the ovary, in very interesting to determine whether GnRH also plays
which this neovascularization is necessary for follicular a key role in tumor angiogenesis.
and luteal function, has been demonstrated. A recent
in vivo study using rats revealed that an application of
Intracellular signal transduction
the GnRH-I agonist leuprolide acetate decreases the
protein expression of VEGF and angiopoietin-1 and Upon GnRH binding, GnRHR undergoes a conforma-
their receptors in ovarian follicles, and that this can be tional change and stimulates a unique G-protein. Inter-
reversed by coinjection of the GnRH antagonist antide estingly, the classical Gaq–11-phospholipase C signal
[70]. A similar inhibitory effect on angiogenesis can be transduction pathway, which is known to operate in
observed in marmosets injected with the GnRH-I the pituitary, is not involved in the antitumor activity
antagonist antarelix [71]. However, VEGF mRNA of GnRH analogs. Rather, GnRHRs couple to Gai in
expression is unaffected by the treatment. The clinical these tumors and result in the activation of several
response of uterine shrinkage after GnRH analog downstream signaling cascades [73,74], such as mito-
treatment and a pathological role of FGF-2, VEGF gen-activated protein kinase (MAPK), phosphatidyl-
and platelet-derived growth factor in uterine leiomy- inositol-3-kinase (PI3K), and nuclear factor kappa B
oma growth and vascularization has also been sug- (NF-jB) signaling. The GnRH-induced signaling path-
gested [72]. Considering that angiogenesis is an ways in extrapituitary tissues are shown schematically
important process in many human cancers, it would be in Fig. 1.

Fig. 1. Schematic representation of GnRHR signaling in extrapituitary tissues. Binding of GnRH to GnRHR triggers several intracellular signal-
ing cascades and cross-talk with mitogenic signaling, depending on the cell context. Some of these signaling modules can transduce extra-
cellular signals to the nucleus and thereby regulate genes that are involved in cell growth, metastasis, or survival. Arr, b-arrestin; CREB,
cAMP response element-binding protein; FGFR, fibroblast growth factor receptor; HB-EGF, heparin-binding EGF; IjB, inhibitory factor kap-
pa B; IGFR, IGF receptor; MEK, mitogen-activated protein kinase kinase; MLK3, mixed-lineage kinase 3; PTP, protein tyrosine phosphatase;
Sos, son of sevenless; TNF-a, tumor necrosis factor alpha.

FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS 5483
17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
GnRH receptor signaling L. W. T. Cheung and A. S. T. Wong

implicated in nerve growth factor-mediated neuronal


MAPK
differentiation of PC12 cells, whereas a rapid and
The major MAPK cascades include extracellular sig- transient activation is associated with growth factor-
nal-regulated kinase (ERK), Jun N-terminal kinase mediated proliferation of PC12 cells [80]. Thus, the
(JNK), and p38 MAPK. Many studies have shown duration of kinase activation seems to be a major
that the MAPK pathway is critical for GnRH activi- determinant of signal outcome. We have shown differ-
ties, which provides an important link for the trans- ential regulation of ERK1 ⁄ 2, p38 MAPK, and JNK
mission of signals from the cell surface to the nucleus. by GnRH-I with sustained signaling through the JNK
Activation of MAPK by GnRH involves distinct pathway in ovarian cancer cells [12]. Consistently,
upstream pathways in generating tissue-specific and GnRH-stimulated MMP-2 and MMP-9 expression,
cell-specific signaling (Fig. 1). This can occur at differ- secretion and cell invasion were attenuated by specific
ent levels via different mechanisms: (a) second messen- inhibition of JNK but not of ERK1 ⁄ 2 and p38
gers [protein kinase C (PKC) and cAMP] [26]; MAPK, suggesting that prolonged activation of JNK
(b) receptor-interacting proteins such as Src and may contribute to a more invasive phenotype. Strong
b-zarrestin [53,75]; and (c) upstream kinases such as and sustained activation of MAPK has been reported
MAPK ⁄ ERK kinase and PI3K [53,76]. For example, to be necessary for its cytoplasm-to-nucleus transloca-
GnRH-I induces apoptosis in DU-145 prostate cancer tion, and thereby contributes to the regulation of gene
cells via JNK, which is activated through two indepen- expression [79,81]. It will be interesting to see whether
dent mechanisms [53]. Activation of the pathway is sustained activation of JNK leads to its nuclear trans-
dependent on c-Src with concomitant decrease in Akt location, which is required for GnRH-stimulated cell
activity, and the combination of these two events invasion. The JNK pathway targets multiple transcrip-
relieves the inhibition of the upstream activator of tion factors, including c-Jun, c-Fos, ATF and PEA,
JNK, MLK3 [53] (Fig. 1). Interestingly, although and putative binding sites for these DNA-binding pro-
ERK1 ⁄ 2 is phosphorylated through epidermal growth teins are present in the MMP promoters [82]. Whether
factor receptor (EGFR) under the same conditions, these putative regulatory elements participate in the
this pathway is not involved in the apoptotic effects. GnRH-dependent activation of the MMP-2 and
These findings demonstrate that activation of the two MMP-9 genes remains to be determined.
MAPKs, which lead to distinct physiological out-
comes, is separated already at the upstream levels. In
Cross-talk with mitogenic signaling
the ovarian cancer cell line CaOV-3, prolonged stimu-
lation of ERK1 ⁄ 2 through Shc and son of sevenless is Cross-talk between cell surface receptors, which has
required for GnRH-I-mediated growth inhibition [76]. been recognized as a mechanism capable of generating
Consistent with the fact that sustained activation of signal diversity, is now receiving further interest.
ERK1 ⁄ 2 is often correlated with cell cycle progression, Figure 1 illustrates the cross-talk between GnRHR
GnRH-I-induced growth inhibition is attributed to G1 and receptor tyrosine kinases (RTKs). For instance,
arrest [76]. Moreover, the signaling cascade was shown GnRH causes transactivation of RTKs, such as EGFR
to be initiated by Gbc, supporting the notion that the [75,78,83]. MMP-2 and MMP-9 seem to be essential
post-GnRHR signaling cascade in extrapituitary cells for GnRH-induced EGFR activation by cleavage of
is different from that in pituitary cells. GnRH-induced the heparin-binding epidermal growth factor (EGF)
MAPK activation has also been shown in another precursor [84]. Transactivation of EGFR has been
ovarian cancer cell line, OVCAR-3. Both ERK1 ⁄ 2 and shown to activate ERK1 ⁄ 2, as GnRH-induced
p38 MAPK mediate the antiproliferative effects of ERK1 ⁄ 2 phosphorylation can be abolished in the pres-
GnRH-I and GnRH-II in a PKC-dependent manner ence of the EGFR inhibitor AG1478 [53,78]. However,
[43,77]. GnRH-II induces the activation of activator the biological importance of ERK1 ⁄ 2 activation in
protein-1 transcription factor via p38 MAPK, suggest- response to this cross-talk still remains elusive.
ing a potential role of activator protein-1 in ovarian Negative cross-talk between GnRHR and growth
cancer cell growth [77]. The JNK pathway also drives factor receptors has also been described. For instance,
tumor invasion and migration in ovarian cancer cells the antiproliferative effects of GnRH-I and GnRH-II
[12], but the activation mechanism(s) remains to be agonists are mediated through attenuation of EGFR
elucidated. signaling in many reproductive tumor cells [57,66,85–
Temporal and spatial differences in cellular signaling 87]. In prostate cancer cells, cetrorelix is able to coun-
may have significant phenotypic manifestations [78,79]. ter EGFR-dependent adhesive signaling through
For example, sustained activation of ERK1 ⁄ 2 has been a PKC-dependent mechanism [66]. Activation of

5484 FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS
17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
L. W. T. Cheung and A. S. T. Wong GnRH receptor signaling

GnRHR appears to mediate these effects via activation tor has no effect on the activation of NF-jB [94]. It
of phosphotyrosine phosphatase, thereby reducing has also been shown that GnRH-I suppresses interleu-
EGF-induced EGFR autophosphorylation, resulting in kin-8 expression via attenuation of tumor necrosis fac-
downregulation of mitogenic signal transduction and tor alpha-induced NF-jB signaling in endometriotic
cell proliferation [85,86,88]. A negative regulatory stromal cells (Fig. 1) [95]. These data suggest that
interaction between GnRHR and mitogenic signaling modulation of cytokine signal transduction by GnRH
pathways has also been reported in human prostate may be one of the mechanisms contributing to its
cancer cells via insulin-like growth factor. GnRH-I growth-inhibitory effect.
agonists inhibit expression of the insulin-like growth The non-RTKs focal adhesion kinase (FAK) and
factor receptor, receptor tyrosine phosphorylation, and proline-rich tyrosine kinase 2 (Pyk2) are the predomi-
the subsequent downstream activation of Akt [89–91]. nant mediators of integrin signaling. GnRHR has been
Another example is FGF-2. GnRH analog treatment shown to signal through these molecules, suggesting a
has been shown to block cell proliferation and inva- role for GnRH in cytoskeletal reorganization. In
sion induced by FGF-2 stimulation [65]. human endometrial cancer cells (HEC-1A), b3-integrin-
dependent activation of FAK is associated with the
inhibitory effects of GnRH-I and GnRH-II on growth
PI3K
[96]. Leiomyoma regression induced by GnRH-I agon-
The PI3K signaling pathway and its downstream ists has been suggested to be mediated, at least in part,
target Akt (also named protein kinase B) has been through a mechanism involving suppression of FAK
implicated in promoting cell survival, proliferation, [97]. Maudsley et al. demonstrated a novel signaling
and invasion. In uterine leiomyomas, the GnRH-I ago- cascade of GnRHR that functionally antagonizes the
nist leuprolide acetate causes a significant reduction in actions of testosterone and inhibits prostate tumor
PI3K ⁄ Akt activity and inhibits the expression of the growth [98]. GnRH controls the tyrosine phosphoryla-
antiapoptotic proteins (c-FLIP and PED ⁄ PEA15), tion status of the focal adhesion proteins Pyk2 and
thereby inducing apoptosis [92]. In the SKOV-3 ovar- Hic-5. This alteration of the focal adhesion dynamics
ian cancer cell line, GnRH-I and GnRH-II interfere then results in nuclear translocation of the androgen
with activation of the PI3K ⁄ Akt cascade, and this is is receptor, which renders it transcriptionally inactive
associated with the inhibitory effects of GnRH on cell [98].
invasion [13].
Although PI3K ⁄ Akt and MAPK are two parallel
Mechanisms underlying the diverse
pathways in some cell types, they are two related path-
responses to GnRH action
ways in the mediation of GnRH actions, as inhibition
of PI3K ⁄ Akt can alter the activation of MAPK. For As discussed earlier, GnRH and its agonists have a
instance, in prostate cancer cells, stimulation of dual and biphasic action: whereas low concentrations
PI3K ⁄ Akt releases mixed-lineage kinase 3, which in (0.1–10 nm) of GnRH stimulate cell proliferation,
turn activates the JNK pathway, and this positive reg- migration and invasion in a dose-dependent manner,
ulation is important for the proapoptotic effect of high concentrations (100 nm to 1 lm) inhibit these
GnRH-I (Fig. 1) [53]. PI3K ⁄ Akt is also an upstream functions [12,13,39]. Moreover, the same dose of
kinase of ERK1 ⁄ 2, and EGFR transactivation by GnRH can elicit completely opposite responses in cells
GnRH-I may be required for the activation of this derived from the same tissue. We demonstrated that in
cascade [75,93]. two human ovarian cancer cell lines, OVCAR-3 and
SKOV-3, GnRH-I and GnRH-II induce invasion of
OVCAR-3 cells, but inhibit the invasiveness of SKOV-
Other signaling pathways
3 cells [13]. A similar difference has been found in the
Activation of NF-jB is important for the protection effects of GnRH on cell proliferation and cell migra-
against apoptosis in ovarian tumors induced by the tion in the prostate carcinoma cell lines TSU-Pr1 and
GnRH-I agonist tiptorelin [94]. The effect is probably DU-145 [67]. Whereas GnRH-I and GnRH-II stimu-
mediated by the Gai-coupled GnRHR, and receptor lated cell proliferation, induced actin cytoskeleton
activation causes nuclear translocation of NF-jB [94]. remodeling and promoted migration in TSU-Pr1 cells,
Unlike the other signaling pathways studied, the they were inhibitory in DU-145 cells [67]. The observa-
GnRH-I-induced NF-jB activation appears to be inde- tion that GnRH-I and GnRH-II have no significant
pendent of the cross-talk between GnRHR and growth effect in cell lines with type I GnRHR depletion indi-
factor signaling, as treatment with phosphatase inhibi- cates that the type I GnRHR is indispensable for the

FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS 5485
17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
GnRH receptor signaling L. W. T. Cheung and A. S. T. Wong

Table 1. Potential mechanisms that underlie the diverse responses Third, multiple GnRH-dependent signaling path-
to GnRH. ways may occur via different subunits of a single
Determinants References G-protein [105]. After ligand-induced dissociation,
both the a-subunit and bc-subunits are capable of
Treatment conditions, e.g. duration [99–101] activating various effectors, such as adenylyl cyclase,
and dose
phospholipase C, and ion channels, thereby conferring
Different Ga subtypes [73,74,101,103,104]
Different G-protein subunits [105–109]
on the receptor the potential for dual signaling
GnRHR expression level [6,110–112] [106,107]. The effector pathway to be activated is
Ligand selectivity [114,115] specific to the upstream subunits. For instance,
Presence of GnRHR splice variants [118,119] whereas the a-subunit of Gi inhibits adenylyl cyclase
Intrinsic cellular properties [61,62] activity, the bc-subunits may stimulate the activities of
some adenylyl cyclase subtypes [108,109].
effects of both GnRH-I and GnRH-II [13,67]. Thus, The receptor expression level is also known to be a
one intriguing question is how a common GnRHR determinant for different signal outcomes [6,110,111].
may mediate different, even opposite, responses to In gonadotropes, different cell surface densities of
GnRH in the same cell type ⁄ tissue. The reasons are GnRHR result in the differential regulation of luteiniz-
unknown, but several possibilities (as summarized in ing hormone and follicle-stimulating hormone subunit
Table 1) can be envisaged. First, the treatment condi- gene expression by GnRH-I [112]. We and others have
tion may be one determinant of the outcome. The previously shown that low doses of GnRH upregulate
pulsatility of GnRH release is necessary for the hor- the expression of its receptor, whereas high doses
mone to stimulate pituitary gonadotropes. On the decrease it [12,111,113]. This difference in regulation
other hand, sustained administration of the peptide suggests that high levels of GnRHR expression may
brings about a short initial stimulation that is rapidly enhance the cellular response to GnRH stimulation,
followed by a decrease in gonadotropin synthesis and presumably due to more efficient signal amplification
secretion [99]. In support of this, the signal response is or altered signaling through coupling to different
different at different doses. It has been shown that pul- G-proteins.
satile GnRH stimulates more sustained ERK activity Moreover, ligand selectivity has been proposed to
(more than 8 h), whereas continuous infusion of aT3-1 explain the opposite (stimulatory and inhibitory)
cells with GnRH stimulates short-term (2 h) ERK effects of GnRH. For instance, in positively respond-
activity [100]. There is also evidence that GnRH treat- ing prostate carcinoma cell lines, GnRH-I is more
ment stimulates cAMP production at nanomolar con- effective than GnRH-II. On the other hand, in nega-
centrations, but has an inhibitory effect at micromolar tively responding cell lines, GnRH-II is much more
concentrations [101]. It should be pointed out that the effective than GnRH-I. Given the short plasma half-
nanomolar concentration (0.01–1 nm) corresponds to life of GnRH, efforts have been made to obtain
the physiological circulating level, and the effects GnRH analogs, to resist degradation and to increase
caused by this dose range may represent the physiolog- potency. However, the different GnRH agonists may
ical functions of GnRH [54,102]. selectively stabilize different receptor-active conforma-
Second, GnRH action has been shown to be medi- tions and therefore different ligand-induced selective
ated by coupling to different Ga-proteins, depending signaling pathways [114]. In this regard, it has been
on the time and dose of exposure [101,103]. In general, shown that the highly variable amino acid at posi-
Gaq and Gas are associated with a stimulatory effect tion 8 of GnRH plays a discriminating role in selecting
[103], whereas Gai often mediates the antiproliferative the receptor conformational state [115].
and proapoptotic effects of GnRH [73,74]. Low GnRH The presence of splice variants of the GnRHR tran-
concentrations promote the coupling of GnRHR to script may be another possible reason for the different
Gas [101]. High concentrations of GnRH cause a or opposite responses to GnRH. To date, variant tran-
switch in receptor coupling from Gas to Gai [101]. scripts of GnRHR have been isolated in many species,
Moreover, stimulation of cAMP production by GnRH e.g. chicken [116], mouse [117] and human [118].
is through coupling to Gas, whereas inhibition of Although these splice variants are totally incapable of
cAMP production at high concentrations of GnRH is ligand binding or signal transduction, they have been
through coupling to Gai [101,104]. These findings sug- implicated in the functional regulation of the wild-type
gest that the intracellular milieu in different tissues receptor. Previous studies have reported their inhibi-
results in differential coupling and different phenotypic tory activity on full-length GnRHR function [119].
effects. This inhibition is specific, augmented by increasing

5486 FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS
Table 2. Physiological characteristics and responses to GnRH of common cancer cell lines. fl, decrease; ›, increase; –, no effect; ?, undetermined.
L. W. T. Cheung and A. S. T. Wong

Anchorage- Invasive Effect of Effect of GnRH


Original Tumorigenicity independent capability GnRH on on motility ⁄
Cell line Origin Isolation histology in nude mice growth in vitro growth invasion References

CaOV-3 Ovary Primary Serous Yes High Low fl › [12,76,125,126]


carcinoma
DU-145 Prostate Brain Poorly Yes Low High fl;– fl [50,55,67,120,127,128,132]
differentiated
HEC-1A Endometrium Primary Moderately Yes High High fl;– ? [15,30,36,48,129–131]
carcinoma differentiated
Ishikawa Endometrium Primary Well Yes High Low fl;– ? [15,47,48,129–131]
carcinoma differentiated
LNCaP Prostate Lymph Well Yes Very low Very low fl;– ? [49,120,127,128]
node differentiated

FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS
MCF-7 Breast Pleural Ductal Only achievable Low in Low fl fl [56,68,122–124,135]
effusion with estrogen absence of
added estrogen
MDA-MB-435 Breast Pleural Ductal Yes High High fl – [68,121,133–135]
effusion
OVCAR-3 Ovary Ascites Serous Yes Low Low fl › [12,13,43,136,137]
SKOV-3 Ovary Ascites Serous Yes High High fl fl [13,43,125]
TSU-Pr1 Prostate Lymph Poorly Yes High High ›; – › [54,55,67,120,127]
node differentiated

5487
GnRH receptor signaling

17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
GnRH receptor signaling L. W. T. Cheung and A. S. T. Wong

amounts of the cotransfected splice variant cDNA and tor that is not rapidly desensitized or internalized.
possibly by preventing or diverting the normal process- There is evidence that GnRH-II may act through the
ing of GnRHR or enhancing GnRHR degradation type I GnRHR. In monkey pituitary cultures, in which
[118]. the type II GnRHR is functional, GnRH-II has been
Finally, it is also possible that differences in found to stimulate gonadotropin secretion exclusively
response may be ascribed to the intrinsic properties of through the type I GnRHR [146]. In contrast, other
the cells. The physiological characteristics of the evidence suggests that the neuromodulatory action of
human cancer cell lines mentioned in this minireview GnRH-II on mammalian behavior is not mediated via
are summarized in Table 2. For example, in contrast the type I receptor in musk shrews [147]. Thus, it
to SKOV-3 cells, OVCAR-3 cells have low invasive appears that GnRH-II may selectively interact with
potential. Thus, whereas low doses of GnRH-I and different GnRHRs to mediate its different actions, pre-
GnRH-II can exert a significant invasive effect in sumably due to the structural differences between the
OVCAR-3 cells, they fail to stimulate SKOV-3 maxi- two GnRHR subtypes.
mally [12,13]. Both GnRH-I and GnRH-II only exert Alternatively, it is possible that the human type II
inhibitory effects on SKOV-3 cells at high doses [13]. GnRHR may be encoded by a different gene that has
yet to be identified. Database searches have revealed
the presence of more than two other GnRHR genes in
Novel receptor(s) for GnRH in humans?
the human genome apart from the conventional type I
An important issue that remains unresolved in this receptor gene [148]. These genes are located on sepa-
field is whether one or more other GnRHR subtypes rate chromosomes. Whether functional, full-length
exist in humans. The discovery of GnRH-II has stimu- transcripts can be produced from these receptor-like
lated the search for a cognate type II GnRHR. Molec- genes remains to be determined. Recently, a novel
ular cloning of the type II GnRHR in goldfish, GnRH-II-binding protein, in addition to a conven-
marmoset and monkey has shown that the type II tional GnRHR, has been identified by using photo-
receptor is structurally and functionally distinct from affinity labeling with an azidobenzoyl-conjugated
the type I receptor [138–140]. In humans, a type II GnRH-II in prostate cancer cells [149]. Taken together,
GnRHR has not been found. However, a search of the these observations thus suggest the potential existence
human genome database has revealed a putative of novel receptors for GnRH-I and GnRH-II.
type II GnRHR gene on chromosome 1q21.1
[140,141]. Expression of this type II GnRHR mRNA
Concluding remarks
has been shown in many human tissues, including
endometrium, ovary, placenta, and prostate cancer This overview shows that GnRH modulates a variety
cells [42,55,140–142]. Although these findings suggest of cellular functions in extrapituitary tissues, such as
that the human type II receptor gene is transcription- cell growth, invasion, and angiogenesis. However, the
ally active, the mRNA is disrupted by a frameshift in effects of GnRH are complex and appear to be cell
coding exon 1 and a premature stop codon in exon 2, context dependent. The ability of GnRH to elicit very
suggesting that a conventional seven-transmembrane different, even opposite (positive and negative),
receptor cannot be translated from this gene. The gene responses in extrapituitary tissues may arise from dif-
also overlaps two flanking genes and displays alterna- ferential usage of signal transduction pathways and
tive splicing [143]. Thus, the functionality of these receptor cross-talk. Clearly, further studies are
human type II GnRHR splice variants and their required to unravel this complex signaling network
involvement in transmitting signals from GnRH-II are and the coordinated regulatory roles of different
still in question. factors in specific cellular events during tumorigenesis.
One noteworthy feature of the primate type II High-throughput gene profiling and bioinformatics
GnRHR is that, unlike the type I receptor, it possesses approaches should be helpful to expand this area of
a C-terminal tail, which is responsible for the recep- research. The information may also serve as a basis
tor’s susceptibility to rapid desensitization and inter- for investigators in the field to explore the signaling
nalization [138,144,145]. Finch et al. showed that mechanisms of other G-protein-coupled receptors.
GnRH was able to efficiently inhibit the proliferation Most studies thus far have only been conducted in
of breast cancer cells when engineered with sheep cellular models, but in vivo approaches will be essential
type I GnRHR, but not with Xenopus type II GnRHR for a complete understanding of the specific role of
[145]. This clearly implies that the antiproliferative each GnRH isoform, including the putative GnRH-III
effect of GnRH is mediated most efficiently by a recep- isolated from the human brain [150]. Given the clinical

5488 FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS
17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
L. W. T. Cheung and A. S. T. Wong GnRH receptor signaling

utility of GnRH analogs in hormone-dependent dis- mone binding sites in human epithelial ovarian carci-
eases, better characterization of GnRH actions and its nomata. Eur J Cancer Clin Oncol 25, 215–221.
key molecular effectors is a necessary first step to more 11 Chien CH, Chen CH, Lee CY, Chang TC, Chen RJ &
effective and perhaps new therapeutic strategies for Chow SN (2004) Detection of gonadotropin-releasing
improving clinical outcome. hormone receptor and its mRNA in primary human
epithelial ovarian cancers. Int J Gynecol Cancer 14,
451–458.
Acknowledgements 12 Cheung LW, Leung PC & Wong AS (2006) Gona-
dotropin-releasing hormone promotes ovarian cancer
This work was supported by the Hong Kong Research
cell invasiveness through c-Jun NH2-terminal kinase-
Grant Council grant 778108 to A. S. T. Wong.
mediated activation of matrix metalloproteinase
(MMP)-2 and MMP-9. Cancer Res 66, 10902–
References 10910.
13 Chen CL, Cheung LW, Lau MT, Choi JH, Auersperg
1 Chen A, Yahalom D, Ben-Aroya N, Kaganovsky E,
N, Wang HS, Wong AS & Leung PC (2007) Differen-
Okon E & Koch Y (1998) A second isoform of gona-
tial role of gonadotropin-releasing hormone on human
dotropin-releasing hormone is present in the brain of
ovarian epithelial cancer cell invasion. Endocrine 31,
human and rodents. FEBS Lett 435, 199–203.
311–320.
2 White RB, Eisen JA, Kasten TL & Fernald RD (1998)
14 Srkalovic G, Wittliff JL & Schally AV (1990)
Second gene for gonadotropin-releasing hormone in
Detection and partial characterization of receptors for
humans. Proc Natl Acad Sci USA 95, 305–309.
[D-Trp6]-luteinizing hormone-releasing hormone and
3 Densmore VS & Urbanski HF (2003) Relative effect
epidermal growth factor in human endometrial
of gonadotropin-releasing hormone (GnRH)-I and
carcinoma. Cancer Res 50, 1841–1846.
GnRH-II on gonadotropin release. J Clin Endocrinol
15 Chatzaki E, Bax CM, Eidne KA, Anderson L, Grudz-
Metab 88, 2126–2134.
inskas JG & Gallagher CJ (1996) The expression of
4 Kauffman AS (2004) Emerging functions of gonado-
gonadotropin-releasing hormone and its receptor in
tropin-releasing hormone II in mammalian physiology
endometrial cancer, and its relevance as an autocrine
and behaviour. J Neuroendocrinol 16, 794–806.
growth factor. Cancer Res 56, 2059–2065.
5 Schally AV, Comaru-Schally AM & Redding TW
16 Imai A, Ohno T, Iida K, Fuseya T, Furui T &
(1984) Antitumor effects of analogs of hypothalamic
Tamaya T (1994) Presence of gonadotropin-releasing
hormones in endocrine-dependent cancers. Proc Soc
hormone receptor and its messenger ribonucleic acid in
Exp Biol Med 175, 259–281.
endometrial carcinoma and endometrium. Gynecol
6 Peng C, Fan NC, Ligier M, Vaananen J & Leung PC
Oncol 55, 144–148.
(1994) Expression and regulation of gonadotropin-
17 Marinaccio M, Reshkin S, Pinto V & Paradiso A
releasing hormone (GnRH) and GnRH receptor mes-
(1994) The estimation of LHRH receptors in the tissue
senger ribonucleic acids in human granulosa-luteal
of human leiomyoma, myometrium and endometrium.
cells. Endocrinology 135, 1740–1746.
Minerva Ginecol 46, 519–526.
7 Brus L, Lambalk CB, de Koning J, Helder MN, Jans-
18 Iwashita M, Evans MI & Catt KJ (1986) Characteriza-
sens RM & Schoemaker J (1997) Specific gonadotro-
tion of a gonadotropin-releasing hormone receptor site
phin-releasing hormone analogue binding
in term placenta and chorionic villi. J Clin Endocrinol
predominantly in human luteinized follicular aspirates
Metab 62, 127–133.
and not in human pre-ovulatory follicles. Hum Reprod
19 Casan EM, Raga F & Polan ML (1999) GnRH
12, 769–773.
mRNA and protein expression in human preimplanta-
8 Choi JH, Gilks CB, Auersperg N & Leung PC (2006)
tion embryos. Mol Hum Reprod 5, 234–239.
Immunolocalization of gonadotropin-releasing hor-
20 Boyle TA, Belt-Davis DI & Duello TM (1998) Nucleo-
mone (GnRH)-I, GnRH-II, and type I GnRH receptor
tide sequence analyses predict that human pituitary
during follicular development in the human ovary.
and human placental gonadotropin-releasing hormone
J Clin Endocrinol Metab 91, 4562–4570.
receptors have identical primary structures. Endocrine
9 Choi KC, Auersperg N & Leung PC (2001) Expression
9, 281–287.
and antiproliferative effect of a second form of gona-
21 Lin LS, Roberts VJ & Yen SS (1995) Expression of
dotropin-releasing hormone in normal and neoplastic
human gonadotropin-releasing hormone receptor gene
ovarian surface epithelial cells. J Clin Endocrinol
in the placenta and its functional relationship to
Metab 86, 5075–5078.
human chorionic gonadotropin secretion. J Clin Endo-
10 Emons G, Pahwa GS, Brack C, Sturm R, Oberheuser
crinol Metab 80, 580–585.
F & Knuppen R (1989) Gonadotropin releasing hor-

FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS 5489
17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
GnRH receptor signaling L. W. T. Cheung and A. S. T. Wong

22 Kottler ML, Starzec A, Carre MC, Lagarde JP, Mar- through specific tumour receptors. Br J Cancer 62,
tin A & Counis R (1997) The genes for gonadotropin- 96–99.
releasing hormone and its receptor are expressed in 34 Pahwa GS, Kullander S, Vollmer G, Oberheuser F,
human breast with fibrocystic disease and cancer. Int J Knuppen R & Emons G (1991) Specific low affinity
Cancer 71, 595–599. binding sites for gonadotropin-releasing hormone in
23 Moriya T, Suzuki T, Pilichowska M, Ariga N, Kimura human endometrial carcinomata. Eur J Obstet Gynecol
N, Ouchi N, Nagura H & Sasano H (2001) Immuno- Reprod Biol 41, 135–142.
histochemical expression of gonadotropin releasing 35 Thompson MA, Adelson MD & Kaufman LM (1991)
hormone receptor in human breast carcinoma. Pathol Lupron retards proliferation of ovarian epithelial
Int 51, 333–337. tumor cells cultured in serum-free medium. J Clin
24 Eidne KA, Flanagan CA, Harris NS & Millar RP Endocrinol Metab 72, 1036–1041.
(1987) Gonadotropin-releasing hormone (GnRH)-bind- 36 Volker P, Grundker C, Schmidt O, Schulz KD &
ing sites in human breast cancer cell lines and inhibi- Emons G (2002) Expression of receptors for luteinizing
tory effects of GnRH antagonists. J Clin Endocrinol hormone-releasing hormone in human ovarian and
Metab 64, 425–432. endometrial cancers: frequency, autoregulation, and
25 Eidne KA, Flanagan CA & Millar RP (1985) Gonado- correlation with direct antiproliferative activity of
tropin-releasing hormone binding sites in human breast luteinizing hormone-releasing hormone analogues.
carcinoma. Science 229, 989–991. Am J Obstet Gynecol 186, 171–179.
26 Limonta P, Moretti RM, Marelli MM, Dondi D, Par- 37 Connor JP, Buller RE & Conn PM (1994) Effects of
enti M & Motta M (1999) The luteinizing hormone- GnRH analogs on six ovarian cancer cell lines in cul-
releasing hormone receptor in human prostate cancer ture. Gynecol Oncol 54, 80–86.
cells: messenger ribonucleic acid expression, molecular 38 Slotman BJ, Poels LG & Rao BR (1989) A direct
size, and signal transduction pathway. Endocrinology LHRH-agonist action on cancer cells is unlikely to be
140, 5250–5256. the cause of response to LHRH-agonist treatment.
27 Tieva A, Stattin P, Wikstrom P, Bergh A & Damber JE Anticancer Res 9, 77–80.
(2001) Gonadotropin-releasing hormone receptor 39 Arencibia JM & Schally AV (2000) Luteinizing hor-
expression in the human prostate. Prostate 47, 276–284. mone-releasing hormone as an autocrine growth factor
28 Straub B, Muller M, Krause H, Schrader M, Goessl C, in ES-2 ovarian cancer cell line. Int J Oncol 16, 1009–
Heicappell R & Miller K (2001) Increased incidence of 1013.
luteinizing hormone-releasing hormone receptor gene 40 Gunthert AR, Grundker C, Bottcher B & Emons G
messenger RNA expression in hormone-refractory (2004) Luteinizing hormone-releasing hormone
human prostate cancers. Clin Cancer Res 7, 2340–2343. (LHRH) inhibits apoptosis induced by cytotoxic agent
29 Kakar SS, Grizzle WE & Neill JD (1994) The nucleo- and UV-light but not apoptosis mediated through
tide sequences of human GnRH receptors in breast CD95 in human ovarian and endometrial cancer cells.
and ovarian tumors are identical with that found in Anticancer Res 24, 1727–1732.
pituitary. Mol Cell Endocrinol 106, 145–149. 41 Grundker C, Schlotawa L, Viereck V, Eicke N, Horst
30 Emons G, Schroder B, Ortmann O, Westphalen S, A, Kairies B & Emons G (2004) Antiproliferative
Schulz KD & Schally AV (1993) High affinity binding effects of the GnRH antagonist cetrorelix and of
and direct antiproliferative effects of luteinizing hor- GnRH-II on human endometrial and ovarian cancer
mone-releasing hormone analogs in human endome- cells are not mediated through the GnRH type I recep-
trial cancer cell lines. J Clin Endocrinol Metab 77, tor. Eur J Endocrinol 151, 141–149.
1458–1464. 42 Grundker C, Gunthert AR, Millar RP & Emons G
31 Emons G, Ortmann O, Becker M, Irmer G, Springer (2002) Expression of gonadotropin-releasing hor-
B, Laun R, Holzel F, Schulz KD & Schally AV (1993) mone II (GnRH-II) receptor in human endometrial
High affinity binding and direct antiproliferative and ovarian cancer cells and effects of GnRH-II on
effects of LHRH analogues in human ovarian cancer tumor cell proliferation. J Clin Endocrinol Metab 87,
cell lines. Cancer Res 53, 5439–5446. 1427–1430.
32 Segal-Abramson T, Kitroser H, Levy J, Schally AV & 43 Kim KY, Choi KC, Auersperg N & Leung PC (2006)
Sharoni Y (1992) Direct effects of luteinizing hor- Mechanism of gonadotropin-releasing hormone
mone-releasing hormone agonists and antagonists on (GnRH)-I and -II-induced cell growth inhibition in
MCF-7 mammary cancer cells. Proc Natl Acad Sci ovarian cancer cells: role of the GnRH-I receptor and
USA 89, 2336–2339. protein kinase C pathway. Endocr Relat Cancer 13,
33 Qayum A, Gullick W, Clayton RC, Sikora K & Wax- 211–220.
man J (1990) The effects of gonadotrophin releasing 44 Yano T, Pinski J, Radulovic S & Schally AV (1994)
hormone analogues in prostate cancer are mediated Inhibition of human epithelial ovarian cancer cell

5490 FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS
17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
L. W. T. Cheung and A. S. T. Wong GnRH receptor signaling

growth in vitro by agonistic and antagonistic analogues 55 Enomoto M, Endo D, Kawashima S & Park MK
of luteinizing hormone-releasing hormone. Proc Natl (2004) Human type II GnRH receptor mediates effects
Acad Sci USA 91, 1701–1705. of GnRH on cell proliferation. Zool Sci 21, 763–770.
45 Yano T, Pinski J, Halmos G, Szepeshazi K, Groot K 56 Miller WR, Scott WN, Morris R, Fraser HM &
& Schally AV (1994) Inhibition of growth of OV-1063 Sharpe RM (1985) Growth of human breast cancer
human epithelial ovarian cancer xenografts in nude cells inhibited by a luteinizing hormone-releasing hor-
mice by treatment with luteinizing hormone-releasing mone agonist. Nature 313, 231–233.
hormone antagonist SB-75. Proc Natl Acad Sci USA 57 Gunthert AR, Grundker C, Olota A, Lasche J, Eicke N
91, 7090–7094. & Emons G (2005) Analogs of GnRH-I and GnRH-II
46 Schally AV (1999) Luteinizing hormone-releasing hor- inhibit epidermal growth factor-induced signal trans-
mone analogs: their impact on the control of tumori- duction and resensitize resistant human breast cancer
genesis. Peptides 20, 1247–1262. cells to 4OH-tamoxifen. Eur J Endocrinol 153, 613–625.
47 Imai A, Takagi A, Horibe S, Takagi H & Tamaya T 58 Sharoni Y, Bosin E, Miinster A, Levy J & Schally AV
(1998) Fas and Fas ligand system may mediate anti- (1989) Inhibition of growth of human mammary
proliferative activity of gonadotropin-releasing hor- tumor cells by potent antagonists of luteinizing hor-
mone receptor in endometrial cancer cells. Int J Oncol mone-releasing hormone. Proc Natl Acad Sci USA 86,
13, 97–100. 1648–1651.
48 Kleinman D, Douvdevani A, Schally AV, Levy J & 59 Raga F, Casan EM, Wen Y, Huang HY, Bonilla-
Sharoni Y (1994) Direct growth inhibition of human Musoles F & Polan ML (1999) Independent regulation
endometrial cancer cells by the gonadotropin-releasing of matrix metalloproteinase-9, tissue inhibitor of
hormone antagonist SB-75: role of apoptosis. Am J metalloproteinase-1 (TIMP-1), and TIMP-3 in human
Obstet Gynecol 170, 96–102. endometrial stromal cells by gonadotropin-releasing
49 Limonta P, Dondi D, Moretti RM, Maggi R & Motta hormone: implications in early human implantation.
M (1992) Antiproliferative effects of luteinizing hor- J Clin Endocrinol Metab 84, 636–642.
mone-releasing hormone agonists on the human pros- 60 Chou CS, Zhu H, Shalev E, MacCalman CD & Leung
tatic cancer cell line LNCaP. J Clin Endocrinol Metab PC (2002) The effects of gonadotropin-releasing hor-
75, 207–212. mone (GnRH) I and GnRH II on the urokinase-type
50 Dondi D, Limonta P, Moretti RM, Marelli MM, Ga- plasminogen activator ⁄ plasminogen activator inhibitor
rattini E & Motta M (1994) Antiproliferative effects of system in human extravillous cytotrophoblasts in vitro.
luteinizing hormone-releasing hormone (LHRH) agon- J Clin Endocrinol Metab 87, 5594–5603.
ists on human androgen-independent prostate cancer 61 Moretti RM, Montagnani Marelli M, Van Groening-
cell line DU 145: evidence for an autocrine-inhibitory hen JC & Limonta P (2002) Locally expressed LHRH
LHRH loop. Cancer Res 54, 4091–4095. receptors mediate the oncostatic and antimetastatic
51 Montagnani Marelli M, Moretti RM, Dondi D, activity of LHRH agonists on melanoma cells. J Clin
Limonta P & Motta M (1997) Effects of LHRH Endocrinol Metab 87, 3791–3797.
agonists on the growth of human prostatic tumor cells: 62 Moretti RM, Monagnani Marelli M, van Groeninghen
‘in vitro’ and ‘in vivo’ studies. Arch Ital Urol Androl 69, JC, Motta M & Limonta P (2003) Inhibitory activity
257–263. of luteinizing hormone-releasing hormone on tumor
52 Jungwirth A, Pinski J, Galvan G, Halmos G, growth and progression. Endocr Relat Cancer 10, 161–
Szepeshazi K, Cai RZ, Groot K, Vadillo-Buenfil M & 167.
Schally AV (1997) Inhibition of growth of androgen- 63 Hierowski MT, Wettler O & Schally AV (1984) Plas-
independent DU-145 prostate cancer in vivo by lutein- minogen activator and nuclear androgen receptor in
ising hormone-releasing hormone antagonist Cetrorelix rat prostate tumors after treatment with D-Trp-6-LH-
and bombesin antagonists RC-3940-II and RC-3950- RH. Biomed Pharmacother 38, 297–303.
II. Eur J Cancer 33, 1141–1148. 64 Dondi D, Festuccia C, Piccolella M, Bologna M &
53 Kraus S, Levy G, Hanoch T, Naor Z & Seger R Motta M (2006) GnRH agonists and antagonists
(2004) Gonadotropin-releasing hormone induces decrease the metastatic progression of human prostate
apoptosis of prostate cancer cells: role of c-Jun NH2- cancer cell lines by inhibiting the plasminogen activa-
terminal kinase, protein kinase B, and extracellular tor system. Oncol Rep 15, 393–400.
signal-regulated kinase pathways. Cancer Res 64, 65 Gnanapragasam VJ, Darby S, Khan MM, Lock WG,
5736–5744. Robson CN & Leung HY (2005) Evidence that pros-
54 Enomoto M, Seong JY, Kawashima S & Park MK tate gonadotropin-releasing hormone receptors mediate
(2004) Proliferation of TSU-Pr1, a human prostatic an anti-tumourigenic response to analogue therapy in
carcinoma cell line is stimulated by gonadotropin- hormone refractory prostate cancer. J Pathol 206,
releasing hormone. Life Sci 74, 3141–3152. 205–213.

FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS 5491
17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
GnRH receptor signaling L. W. T. Cheung and A. S. T. Wong

66 Yates C, Wells A & Turner T (2005) Luteinising hor- kinase and apoptosis in ovarian cancer cells. J Clin
mone-releasing hormone analogue reverses the cell Endocrinol Metab 89, 3020–3026.
adhesion profile of EGFR overexpressing DU-145 78 Shah BH, Farshori MP, Jambusaria A & Catt KJ
human prostate carcinoma subline. Br J Cancer 92, (2003) Roles of Src and epidermal growth factor recep-
366–375. tor transactivation in transient and sustained ERK1 ⁄ 2
67 Enomoto M, Utsumi M & Park MK (2006) Gonado- responses to gonadotropin-releasing hormone receptor
tropin-releasing hormone induces actin cytoskeleton activation. J Biol Chem 278, 19118–19126.
remodeling and affects cell migration in a cell-type-spe- 79 Marshall CJ (1995) Specificity of receptor tyrosine
cific manner in TSU-Pr1 and DU145 cells. Endocrinol- kinase signaling: transient versus sustained extracellu-
ogy 147, 530–542. lar signal-regulated kinase activation. Cell 80, 179–185.
68 von Alten J, Fister S, Schulz H, Viereck V, Frosch 80 Kao S, Jaiswal RK, Kolch W & Landreth GE (2001)
KH, Emons G & Grundker C (2006) GnRH analogs Identification of the mechanisms regulating the differ-
reduce invasiveness of human breast cancer cells. ential activation of the mapk cascade by epidermal
Breast Cancer Res Treat 100, 13–21. growth factor and nerve growth factor in PC12 cells.
69 Chen A, Ganor Y, Rahimipour S, Ben-Aroya N, Koch J Biol Chem 276, 18169–18177.
Y & Levite M (2002) The neuropeptides GnRH-II and 81 Pouyssegur J, Volmat V & Lenormand P (2002) Fidel-
GnRH-I are produced by human T cells and trigger ity and spatio-temporal control in MAP kinase
laminin receptor gene expression, adhesion, chemotaxis (ERKs) signalling. Biochem Pharmacol 64, 755–763.
and homing to specific organs. Nat Med 8, 1421–1426. 82 Chakraborti S, Mandal M, Das S, Mandal A & Chak-
70 Parborell F, Irusta G, Rodriguez Celin A & Tesone raborti T (2003) Regulation of matrix metalloprotein-
M (2008) Regulation of ovarian angiogenesis and ases: an overview. Mol Cell Biochem 253, 269–285.
apoptosis by GnRH-I analogs. Mol Reprod Dev 75, 83 Shah BH, Soh JW & Catt KJ (2003) Dependence of
623–631. gonadotropin-releasing hormone-induced neuronal
71 Taylor PD, Hillier SG & Fraser HM (2004) Effects of MAPK signaling on epidermal growth factor receptor
GnRH antagonist treatment on follicular development transactivation. J Biol Chem 278, 2866–2875.
and angiogenesis in the primate ovary. J Endocrinol 84 Roelle S, Grosse R, Aigner A, Krell HW, Czubayko F
183, 1–17. & Gudermann T (2003) Matrix metalloproteinases 2
72 Di Lieto A, De Falco M, Pollio F, Mansueto G, and 9 mediate epidermal growth factor receptor trans-
Salvatore G, Somma P, Ciociola F, De Rosa G & activation by gonadotropin-releasing hormone. J Biol
Staibano S (2005) Clinical response, vascular change, Chem 278, 47307–47318.
and angiogenesis in gonadotropin-releasing hormone 85 Moretti RM, Marelli MM, Dondi D, Poletti A,
analogue-treated women with uterine myomas. J Soc Martini L, Motta M & Limonta P (1996) Luteinizing
Gynecol Investig 12, 123–128. hormone-releasing hormone agonists interfere with the
73 Kraus S, Naor Z & Seger R (2006) Gonadotropin- stimulatory actions of epidermal growth factor in
releasing hormone in apoptosis of prostate cancer cells. human prostatic cancer cell lines, LNCaP and
Cancer Lett 234, 109–123. DU 145. J Clin Endocrinol Metab 81, 3930–3937.
74 Imai A, Horibe S, Takagi A & Tamaya T (1997) Gi 86 Grundker C, Volker P & Emons G (2001) Antiprolif-
protein activation of gonadotropin-releasing hormone- erative signaling of luteinizing hormone-releasing hor-
mediated protein dephosphorylation in human endo- mone in human endometrial and ovarian cancer cells
metrial carcinoma. Am J Obstet Gynecol 176, 371–376. through G protein alpha(I)-mediated activation of
75 Kraus S, Benard O, Naor Z & Seger R (2003) c-Src is phosphotyrosine phosphatase. Endocrinology 142,
activated by the epidermal growth factor receptor in a 2369–2380.
pathway that mediates JNK and ERK activation by 87 Eicke N, Gunthert AR, Emons G & Grundker C
gonadotropin-releasing hormone in COS7 cells. J Biol (2006) GnRH-II agonist [D-Lys6]GnRH-II inhibits the
Chem 278, 32618–32630. EGF-induced mitogenic signal transduction in human
76 Kimura A, Ohmichi M, Kurachi H, Ikegami H, endometrial and ovarian cancer cells. Int J Oncol 29,
Hayakawa J, Tasaka K, Kanda Y, Nishio Y, Jikihara 1223–1229.
H, Matsuura N et al. (1999) Role of mitogen-activated 88 Lamharzi N, Halmos G, Jungwirth A & Schally AV
protein kinase ⁄ extracellular signal-regulated kinase (1998) Decrease in the level and mRNA expression of
cascade in gonadotropin-releasing hormone-induced LH-RH and EGF receptors after treatment with LH-
growth inhibition of a human ovarian cancer cell line. RH antagonist cetrorelix in DU-145 prostate tumor
Cancer Res 59, 5133–5142. xenografts in nude mice. Int J Oncol 13, 429–435.
77 Kim KY, Choi KC, Park SH, Chou CS, Auersperg N 89 Montagnani Marelli M, Moretti RM, Mai S, Procacci
& Leung PC (2004) Type II gonadotropin-releasing P & Limonta P (2007) Gonadotropin-releasing hor-
hormone stimulates p38 mitogen-activated protein mone agonists reduce the migratory and the invasive

5492 FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS
17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
L. W. T. Cheung and A. S. T. Wong GnRH receptor signaling

behavior of androgen-independent prostate cancer cells 99 Kaiser UB, Conn PM & Chin WW (1997) Studies of
by interfering with the activity of IGF-I. Int J Oncol gonadotropin-releasing hormone (GnRH) action using
30, 261–271. GnRH receptor-expressing pituitary cell lines. Endocr
90 Marelli MM, Moretti RM, Dondi D, Motta M & Rev 18, 46–70.
Limonta P (1999) Luteinizing hormone-releasing hor- 100 Haisenleder DJ, Cox ME, Parsons SJ & Marshall
mone agonists interfere with the mitogenic activity of JC (1998) Gonadotropin-releasing hormone pulses
the insulin-like growth factor system in androgen-inde- are required to maintain activation of mitogen-
pendent prostate cancer cells. Endocrinology 140, activated protein kinase: role in stimulation of
329–334. gonadotrope gene expression. Endocrinology 139,
91 Lamharzi N, Schally AV & Koppan M (1998) Lutein- 3104–3111.
izing hormone-releasing hormone (LH-RH) antagonist 101 Krsmanovic LZ, Mores N, Navarro CE, Arora KK &
Cetrorelix inhibits growth of DU-145 human andro- Catt KJ (2003) An agonist-induced switch in G protein
gen-independent prostate carcinoma in nude mice and coupling of the gonadotropin-releasing hormone recep-
suppresses the levels and mRNA expression of IGF-II tor regulates pulsatile neuropeptide secretion. Proc
in tumors. Regul Pept 77, 185–192. Natl Acad Sci USA 100, 2969–2974.
92 Bifulco G, Miele C, Pellicano M, Trencia A, Ferraioli 102 Loumaye E & Catt KJ (1982) Homologous regulation
M, Paturzo F, Tommaselli GA, Beguinot F & Nappi of gonadotropin-releasing hormone receptors in cul-
C (2004) Molecular mechanisms involved in GnRH tured pituitary cells. Science 215, 983–985.
analogue-related apoptosis for uterine leiomyomas. 103 Liu F, Usui I, Evans LG, Austin DA, Mellon PL,
Mol Hum Reprod 10, 43–48. Olefsky JM & Webster NJ (2002) Involvement of both
93 Shah BH, Neithardt A, Chu DB, Shah FB & Catt KJ G(q ⁄ 11) and G(s) proteins in gonadotropin-releasing
(2006) Role of EGF receptor transactivation in phos- hormone receptor-mediated signaling in L beta T2
phoinositide 3-kinase-dependent activation of MAP cells. J Biol Chem 277, 32099–32108.
kinase by GPCRs. J Cell Physiol 206, 47–57. 104 Krsmanovic LZ, Mores N, Navarro CE, Tomic M &
94 Grundker C, Schulz K, Gunthert AR & Emons G Catt KJ (2001) Regulation of Ca2+-sensitive adenylyl
(2000) Luteinizing hormone-releasing hormone induces cyclase in gonadotropin-releasing hormone neurons.
nuclear factor kappaB-activation and inhibits apopto- Mol Endocrinol 15, 429–440.
sis in ovarian cancer cells. J Clin Endocrinol Metab 85, 105 Grosse R, Schmid A, Schoneberg T, Herrlich A, Muhn
3815–3820. P, Schultz G & Gudermann T (2000) Gonadotropin-
95 Sakamoto Y, Harada T, Horie S, Iba Y, Taniguchi releasing hormone receptor initiates multiple signaling
F, Yoshida S, Iwabe T & Terakawa N (2003) pathways by exclusively coupling to G(q ⁄ 11) proteins.
Tumor necrosis factor-alpha-induced interleukin-8 J Biol Chem 275, 9193–9200.
(IL-8) expression in endometriotic stromal cells, 106 Guo CH, Janovick JA, Kuphal D & Conn PM (1995)
probably through nuclear factor-kappa B activation: Transient transfection of GGH3-1¢ cells [GH3 cells
gonadotropin-releasing hormone agonist treatment stably transfected with the gonadotropin-releasing
reduced IL-8 expression. J Clin Endocrinol Metab 88, hormone (GnRH) receptor complementary deoxyribo-
730–735. nucleic acid] with the carboxyl-terminal of beta-adren-
96 Choi JR, Park DW, Choi DS & Min CK (2006) Role ergic receptor kinase 1 blocks prolactin release:
of integrin, FAK (focal adhesion kinase) and ERK evidence for a role of the G protein beta gamma-sub-
(extracellular signal regulated kinase) on the sup- unit complex in GnRH signal transduction. Endocri-
pressed cell proliferation of endometrial cancer cells by nology 136, 3031–3036.
GnRH (gonadotropin-releasing hormone). Korean J 107 Clapham DE & Neer EJ (1993) New roles for G-pro-
Fertil Steril 33, 115–123. tein beta gamma-dimers in transmembrane signalling.
97 Chegini N & Kornberg L (2003) Gonadotropin releas- Nature 365, 403–406.
ing hormone analogue therapy alters signal transduc- 108 Federman AD, Conklin BR, Schrader KA, Reed RR
tion pathways involving mitogen-activated protein and & Bourne HR (1992) Hormonal stimulation of ade-
focal adhesion kinases in leiomyoma. J Soc Gynecol nylyl cyclase through Gi-protein beta gamma subunits.
Investig 10, 21–26. Nature 356, 159–161.
98 Maudsley S, Davidson L, Pawson AJ, Freestone SH, 109 Tang WJ & Gilman AG (1991) Type-specific regula-
Lopez de Maturana R, Thomson AA & Millar RP tion of adenylyl cyclase by G protein beta gamma
(2006) Gonadotropin-releasing hormone functionally subunits. Science 254, 1500–1503.
antagonizes testosterone activation of the human 110 Kaiser UB, Jakubowiak A, Steinberger A & Chin WW
androgen receptor in prostate cells through focal adhe- (1993) Regulation of rat pituitary gonadotropin-releas-
sion complexes involving Hic-5. Neuroendocrinology ing hormone receptor mRNA levels in vivo and
84, 285–300. in vitro. Endocrinology 133, 931–934.

FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS 5493
17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
GnRH receptor signaling L. W. T. Cheung and A. S. T. Wong

111 Kang SK, Cheng KW, Nathwani PS, Choi KC & lines from different laboratories. Breast Cancer Res
Leung PC (2000) Autocrine role of gonadotropin- Treat 9, 111–121.
releasing hormone and its receptor in ovarian cancer 123 Dickson RB, Bates SE, McManaway ME & Lippman
cell growth. Endocrine 13, 297–304. ME (1986) Characterization of estrogen responsive
112 Kaiser UB, Sabbagh E, Katzenellenbogen RA, Conn transforming activity in human breast cancer cell lines.
PM & Chin WW (1995) A mechanism for the differen- Cancer Res 46, 1707–1713.
tial regulation of gonadotropin subunit gene expres- 124 Xie D, Miller CW, O’Kelly J, Nakachi K, Sakashita
sion by gonadotropin-releasing hormone. Proc Natl A, Said JW, Gornbein J & Koeffler HP (2001) Breast
Acad Sci USA 92, 12280–12284. cancer. Cyr61 is overexpressed, estrogen-inducible, and
113 McArdle CA, Gorospe WC, Huckle WR & Conn PM associated with more advanced disease. J Biol Chem
(1987) Homologous down-regulation of gonadotropin- 276, 14187–14194.
releasing hormone receptors and desensitization of 125 Yao JJ, Liu Y, Lacorazza HD, Soslow RA, Scandura
gonadotropes: lack of dependence on protein kinase C. JM, Nimer SD & Hedvat CV (2007) Tumor promoting
Mol Endocrinol 1, 420–429. properties of the ETS protein MEF in ovarian cancer.
114 Lu ZL, Gallagher R, Sellar R, Coetsee M & Millar RP Oncogene 26, 4032–4037.
(2005) Mutations remote from the human gonadotro- 126 Hongo A, Kuramoto H, Nakamura Y, Hasegawa K,
pin-releasing hormone (GnRH) receptor-binding sites Nakamura K, Kodama J & Hiramatsu Y (2003) Anti-
specifically increase binding affinity for GnRH II but tumor effects of a soluble insulin-like growth factor I
not GnRH I: evidence for ligand-selective, receptor- receptor in human ovarian cancer cells: advantage of
active conformations. J Biol Chem 280, 29796–29803. recombinant protein administration in vivo. Cancer Res
115 Lu ZL, Coetsee M, White CD & Millar RP (2007) 63, 7834–7839.
Structural determinants for ligand–receptor conforma- 127 Webber MM, Bello D & Quader S (1997) Immortal-
tional selection in a peptide G protein-coupled recep- ized and tumorigenic adult human prostatic epithelial
tor. J Biol Chem 282, 17921–17929. cell lines: characteristics and applications. Part 2.
116 Shimizu M & Bedecarrats GY (2006) Identification of Tumorigenic cell lines. Prostate 30, 58–64.
a novel pituitary-specific chicken gonadotropin-releas- 128 Sobel RE & Sadar MD (2005) Cell lines used in pros-
ing hormone receptor and its splice variants. Biol tate cancer research: a compendium of old and new
Reprod 75, 800–808. lines – part 1. J Urol 173, 342–359.
117 Zhou W & Sealfon SC (1994) Structure of the mouse 129 Kyo S, Nakamura M, Kiyono T, Maida Y, Kanaya T,
gonadotropin-releasing hormone receptor gene: variant Tanaka M, Yatabe N & Inoue M (2003) Successful
transcripts generated by alternative processing. DNA immortalization of endometrial glandular cells with
Cell Biol 13, 605–614. normal structural and functional characteristics. Am J
118 Kottler ML, Bergametti F, Carre MC, Morice S, Dec- Pathol 163, 2259–2269.
oret E, Lagarde JP, Starzec A & Counis R (1999) Tis- 130 Tanaka R, Saito T, Ashihara K, Nishimura M,
sue-specific pattern of variant transcripts of the human Mizumoto H & Kudo R (2003) Three-dimensional
gonadotropin-releasing hormone receptor gene. Eur J coculture of endometrial cancer cells and fibroblasts in
Endocrinol 140, 561–569. human placenta derived collagen sponges and expres-
119 Grosse R, Schoneberg T, Schultz G & Gudermann T sion matrix metalloproteinases in these cells. Gynecol
(1997) Inhibition of gonadotropin-releasing hormone Oncol 90, 297–304.
receptor signaling by expression of a splice variant of 131 Sillem M, Prifti S, Koumouridis A & Runnebaum B
the human receptor. Mol Endocrinol 11, 1305–1318. (1999) Invasiveness corresponds to differentiation
120 Bahk JY, Hyun JS, Lee H, Kim MO, Cho GJ, Lee rather than to proteinase secretion in endometrial can-
BH & Choi WS (1998) Expression of gonadotropin- cer cell lines. Eur J Gynaecol Oncol 20, 367–370.
releasing hormone (GnRH) and GnRH receptor 132 Stone KR, Mickey DD, Wunderli H, Mickey GH &
mRNA in prostate cancer cells and effect of GnRH on Paulson DF (1978) Isolation of a human prostate
the proliferation of prostate cancer cells. Urol Res 26, carcinoma cell line (DU 145). Int J Cancer 21, 274–
259–264. 281.
121 Chatzistamou I, Schally AV, Varga JL, Groot K, 133 Honjo Y, Nangia-Makker P, Inohara H & Raz A
Busto R, Armatis P & Halmos G (2001) Inhibition (2001) Down-regulation of galectin-3 suppresses
of growth and metastases of MDA-MB-435 human tumorigenicity of human breast carcinoma cells. Clin
estrogen-independent breast cancers by an antagonist Cancer Res 7, 661–668.
of growth hormone-releasing hormone. Anticancer 134 Mukhopadhyay R, Theriault RL & Price JE (1999)
Drugs 12, 761–768. Increased levels of alpha6 integrins are associated with
122 Osborne CK, Hobbs K & Trent JM (1987) Biological the metastatic phenotype of human breast cancer cells.
differences among MCF-7 human breast cancer cell Clin Exp Metastasis 17, 325–332.

5494 FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS
17424658, 2008, 22, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/j.1742-4658.2008.06677.x by CochraneChina, Wiley Online Library on [21/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
L. W. T. Cheung and A. S. T. Wong GnRH receptor signaling

135 Thompson EW, Paik S, Brunner N, Sommers CL, 143 Morgan K, Conklin D, Pawson AJ, Sellar R, Ott TR &
Zugmaier G, Clarke R, Shima TB, Torri J, Donahue Millar RP (2003) A transcriptionally active human
S, Lippman ME et al. (1992) Association of increased type II gonadotropin-releasing hormone receptor gene
basement membrane invasiveness with absence of homolog overlaps two genes in the antisense orientation
estrogen receptor and expression of vimentin in human on chromosome 1q.12. Endocrinology 144, 423–436.
breast cancer cell lines. J Cell Physiol 150, 534–544. 144 Pawson AJ, Faccenda E, Maudsley S, Lu ZL, Naor Z
136 Hamilton TC, Young RC, McKoy WM, Grotzinger & Millar RP (2007) Mammalian type I GnRH recep-
KR, Green JA, Chu EW, Whang-Peng J, Rogan AM, tors undergo slow, constitutive, agonist-independent
Green WR & Ozols RF (1983) Characterization of a internalization. Endocrinology 149, 1415–1422.
human ovarian carcinoma cell line (NIH:OVCAR-3) 145 Finch AR, Green L, Hislop JN, Kelly E & McArdle
with androgen and estrogen receptors. Cancer Res 43, CA (2004) Signaling and antiproliferative effects of
5379–5389. type I and II gonadotropin-releasing hormone recep-
137 Maeda T, Tashiro H, Katabuchi H, Begum M, Ohtake tors in breast cancer cells. J Clin Endocrinol Metab 89,
H, Kiyono T & Okamura H (2005) Establishment of 1823–1832.
an immortalised human ovarian surface epithelial cell 146 Okada Y, Murota-Kawano A, Kakar SS & Winters SJ
line without chromosomal instability. Br J Cancer 93, (2003) Evidence that gonadotropin-releasing hormone
116–123. (GnRH) II stimulates luteinizing hormone and follicle-
138 Millar R, Lowe S, Conklin D, Pawson A, Maudsley S, stimulating hormone secretion from monkey pituitary
Troskie B, Ott T, Millar M, Lincoln G, Sellar R et al. cultures by activating the GnRH I receptor. Biol
(2001) A novel mammalian receptor for the evolution- Reprod 69, 1356–1361.
arily conserved type II GnRH. Proc Natl Acad Sci 147 Kauffman AS, Wills A, Millar RP & Rissman EF
USA 98, 9636–9641. (2005) Evidence that the type-2 gonadotrophin-releas-
139 Illing N, Troskie BE, Nahorniak CS, Hapgood JP, ing hormone (GnRH) receptor mediates the behaviour-
Peter RE & Millar RP (1999) Two gonadotropin- al effects of GnRH-II on feeding and reproduction in
releasing hormone receptor subtypes with distinct musk shrews. J Neuroendocrinol 17, 489–497.
ligand selectivity and differential distribution in brain 148 van Biljon W, Wykes S, Scherer S, Krawetz SA &
and pituitary in the goldfish (Carassius auratus). Proc Hapgood J (2002) Type II gonadotropin-releasing
Natl Acad Sci USA 96, 2526–2531. hormone receptor transcripts in human sperm. Biol
140 Neill JD, Duck LW, Sellers JC & Musgrove LC (2001) Reprod 67, 1741–1749.
A gonadotropin-releasing hormone (GnRH) receptor 149 Maiti K, Oh DY, Moon JS, Acharjee S, Li JH, Bai
specific for GnRH II in primates. Biochem Biophys DG, Park HS, Lee K, Lee YC, Jung NC et al. (2005)
Res Commun 282, 1012–1018. Differential effects of gonadotropin-releasing hormone
141 Neill JD (2002) GnRH and GnRH receptor genes in (GnRH)-I and GnRH-II on prostate cancer cell signal-
the human genome. Endocrinology 143, 737–743. ing and death. J Clin Endocrinol Metab 90, 4287–4298.
142 Eicke N, Gunthert AR, Viereck V, Siebold D, Behe 150 Yahalom D, Chen A, Ben-Aroya N, Rahimipour S,
M, Becker T, Emons G & Grundker C (2005) GnRH- Kaganovsky E, Okon E, Fridkin M & Koch Y (1999)
II receptor-like antigenicity in human placenta and in The gonadotropin-releasing hormone family of neuro-
cancers of the human reproductive organs. Eur J peptides in the brain of human, bovine and rat: identi-
Endocrinol 153, 605–612. fication of a third isoform. FEBS Lett 463, 289–294.

FEBS Journal 275 (2008) 5479–5495 ª 2008 The Authors Journal compilation ª 2008 FEBS 5495

You might also like