You are on page 1of 13

Journal of Neuroendocrinology, 2012, 24, 1463–1475

© 2012 The Authors.


REVIEW ARTICLE Journal of Neuroendocrinology © 2012 British Society for Neuroendocrinology

Gonadotrophin-Releasing Hormone Signalling Downstream of Calmodulin


P. Melamed*†, D. Savulescu*, S. Lim†, A. Wijeweera*, Z. Luo†1, M. Luo†2 and L. Pnueli*
*Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel.
†Department of Biological Sciences, National University of Singapore, Singapore, Singapore.

Journal of Gonadotrophin-releasing hormone (GnRH) regulates reproduction via binding a G-protein


coupled receptor on the surface of the gonadotroph, through which it transmits signals, mostly
Neuroendocrinology via the mitogen-activated protein (MAPK) cascade, to increase synthesis of the gonadotrophin
hormones: luteinising hormone (LH) and follicle-stimulating hormone (FSH). Activation of the
MAPK cascade requires an elevation in cytosolic Ca2+ levels, which is a result of both calcium
influx and mobilisation from intracellular stores. However, Ca2+ also transmits signals via an
MAPK-independent pathway, through binding calmodulin (CaM), which is then able to bind a
number of proteins to impart diverse downstream effects. Although the ability of GnRH to
activate CaM was recognised over 20 years ago, only recently have some of the downstream
effects been elucidated. GnRH was shown to activate the CaM-dependent phosphatase,
calcineurin, which targets gonadotrophin gene expression both directly and indirectly via
transcription factors such as nuclear factor of activated T-cells and Nur77, the Transducer of
Regulated CREB (TORC) co-activators and also the prolyl isomerase, Pin1. Gonadotrophin gene
expression is also regulated by GnRH-induced CaM-dependent kinases (CaMKs); CaMKI is able to
Correspondence to: derepress the histone deacetylase-inhibition of b-subunit gene expression, whereas CaMKII
P. Melamed, Faculty of Biology, appears to be essential for the GnRH-activation of all three subunit genes. Asides from
Technion-Israel Institute of activating gonadotrophin gene expression, GnRH also exerts additional effects on gonadotroph
Technology, Haifa 32000, Israel
function, some of which clearly occur via CaM, including the proliferation of immature gonado-
(e-mail: philippa.melamed@gmail.
com). trophs, which is dependent on calcineurin. In this review, we summarise these pathways, and
1
discuss the additional functions that have been proposed for CaM with respect to modifying
Present address: Stowers Institute for
Medical Research, Kansas City MO,
GnRH-induced signalling pathways via the regulation of the small GTP-binding protein, Gem,
USA. and/or the regulator of G-protein signalling protein 2.
2
Present address: Center for Cell and Key words: GnRH, gonadotrophin, gonadotroph, calmodulin, calcineurin, CaMK
Gene Therapy, Baylor College of
Medicine, Houston TX, USA doi: 10.1111/j.1365-2826.2012.02359.x

these cells in numerous other ways, including cell proliferation and


Gonadotrophin-releasing hormone (GnRH), the gonadotroph
apoptosis, cell shape and mobility, as well as their responsiveness
and calmodulin
to other hormones (1–6).
Reproduction is regulated through a complex interplay of hormones The GnRH arrives at the pituitary in pulses whose amplitude and
and feedback operating along the hypothalamic-pituitary-gonadal frequency vary with ensuing sexual maturity and during the
axis, of which the hypothalamic hormone, gonadotrophin-releasing oestrous cycle; these distinct pulse frequencies direct the preferen-
hormone (GnRH), is the predominant activator. GnRH binds a tial expression of either LHb or FSHb (7–10). Subsequent to their
membrane-bound receptor (GnRHR) on the pituitary gonadotrophs synthesis, the gonadotrophic hormones, LH and FSH, are secreted
to stimulate transcription of the three gonadotrophin genes: the into the circulation from where they regulate gonadal activity. The
common a subunit (aGSU) and the hormone specific b-subunits pivotal role of GnRH in functioning of the gonadotrophs and the
[luteinising hormone (LH)b and follicle-stimulating hormone (FSH)b] regulation of reproduction is seen at puberty, when GnRH delivery
(Fig. 1), as well as stimulating the secretion of LH. As part of its to the gonadotroph is the only endogenous block to the reawaken-
regulation of gonadotroph activity and function, GnRH also affects ing of the pituitary-gonadal axis and reproductive potential (11).
1464 P. Melamed et al.

GnRH
Ca2+
GnRHR
as aq
PKA
Ca2+
Ca2+ PLCb
Calmodulin

PKC
CaMKI CaMKII Calcineurin
P
ERK1/2
Pin1
14-3-3 P P
P TORC NFAT3
HDAC4
Cytosol

Nucleus
HDACsHDAC4 CoR Pin1 SF-1

Gonadotrophin b-subunit gene TORC CDK7


transcription (LHb and FSHb)
NFAT4 SF-1 NFAT3 AP-1 P
SF-1

U
–P TORC U
Gonadotrophin gene
transcription (GSU)
P U P P
Nur77 CREB AP-1 Pitx1 SF-1 Egr-1

Gonadotrophin gene transcription (LHb )


Gonadotrophin gene transcription (FSHb )

Fig. 1. Model of gonadotrophin-releasing hormone (GnRH)-induced pathways downstream of calmodulin towards gonadotrophin gene expression. Upon bind-
ing its G-protein coupled receptor on the cell membrane, GnRH induces calcium influx (as well as mobilisation: not shown), which activates calmodulin (CaM).
Some of the effects downstream of CaM via the CaM kinases and the phosphatase, calcineurin, which lead to gonadotrophin gene expression, are shown. Solid
lines represent effects that are likely direct, and dashed lines represent pathways that may or may not have intermediate elements that are not shown. Blue
lines represent transcriptional, and black lines post-translational effects on the target. The pink double-headed arrows indicate proteins that shuttle between
cellular compartments. CaMK, calmodulin-dependent kinase; CoR, receptor corepressor; ERK, extracellular-signal regulated kinase; HDAC, histone deacetylase;
NFAT, nuclear factor of activated T-cells; PKA, protein kinase A; PKC, protein kinase C; PLC, phospholipase C; TORC, transducer of regulated CREB.

Upon binding to its G-protein coupled receptor on the gonado- suggested that Ca2+ sensing might play a role in the decoding of
troph, GnRH induces the production of inositol triphosphate (IP3) GnRH pulse-frequency (18).
and diacylglycerol for downstream signalling, much of which is via Apart from its role in the activation of PKC and the downstream
protein kinase C (PKC) (12). The elevation of IP3 results in calcium MAPK pathways, Ca2+ is also able to induce distinct cellular out-
mobilisation from the endoplasmic reticulum (ER), with external comes via additional binding protein effectors, most notably cal-
calcium also entering through voltage-gated calcium channels on modulin (CaM), which is expressed ubiquitously. CaM does not
the cell membrane. These GnRH-induced events trigger the major contain its own enzymatic activity but, in different cell contexts,
mitogen-activated protein kinase (MAPK) cascades, culminating in interacts with a variety of proteins, including kinases, phosphatases
activation of extracellular-signal regulated kinase (ERK) 1/2, c-Jun and ion channels to transduce the signalling effects to diverse
NH2-terminal kinase (JNK), p38 MAPK and ERK5 (10,12,13). The ele- pathways (19). This is possible because the CaM N-terminal and
vation in available intracellular Ca2+ is temporary because the Ca2+ C-terminal domains are connected by a linker region which allows
is quickly sequestered by binding proteins and is also pumped out it to assume distinct conformations when bound to different
of the cytosol after inhibition of the ER IP3-sensitive Ca2+ channels targets. In the absence of Ca2+, it adopts a closed conformation
by the high levels of cytosolic Ca2+ (14). The resulting short pulses but, after Ca2+ binding, the conformational change exposes more
of free Ca2+ play crucial roles in cell signalling in many cell types, hydrophobic groups, thus enhancing the ability of CaM to interact
and their roles in activating regulated LH secretion have been docu- with the various effector proteins (19-21).
mented extensively (14,15), while also playing additional roles in Early studies in gonadotrophs demonstrated that GnRH induces
the regulation of other aspects of cell function and gene expression a change in localisation of CaM from the cytoplasm to the cell
(16,17). Diverse studies have indicated the varying involvement of membrane and also indicated that it has a role in GnRH-induced
this Ca2+ in expression of the gonadotrophin genes (12) and it was LH release, as well as in the expression of primary response genes,

© 2012 The Authors. Journal of Neuroendocrinology, 2012, 24, 1463–1475


Journal of Neuroendocrinology © 2012 British Society for Neuroendocrinology
GnRH signalling downstream of calmodulin 1465

c-fos, c-jun and junB, (22–24). More recently, two of the better chaperone proteins that mask the nuclear localisation signal on the
characterised signalling pathways downstream of CaM, the CaM- HDAC and may expose a nuclear export signal before exporting the
dependent kinases (CaMKs) and the phosphatase, calcineurin, have HDAC out of the nucleus (30–32). HDAC4 and HDAC5 are phos-
been shown to play roles in transducing GnRH-activated signals to phorylated after GnRH exposure and this is almost completely abol-
gonadotrophin gene transcription and other aspects of gonadotroph ished by a dominant negative (dn) CaMKI. Moreover, over-
function. In this review, we summarise these pathways, and expression of the dnCaMKI also abolished the ability of GnRH to
examine other evidence for a role of CaM in modulating GnRH- de-repress expression of the FSHb gene, although not the LHb gene
induced ERK signalling, possibly involving the regulator of G-protein (26,33). GnRH causes dissociation of these HDACs from the b-sub-
signalling (RGS) protein, RGS2, or the small GTP-binding protein, unit gene promoters, and their nuclear export is dependent on the
Gem. serines targeted by CaMKI (Fig. 1). Given that knockdown of HDAC4
alone was sufficient to allow FSHb gene expression, it appears that
GnRH-activated CaMKI mediates the de-repression of this gene in
CaMKs and gonadotrophin gene expression
immature gonadotrophs via targeting these HDACs to cause their
The CaMKs are serine/threonine kinases, which include CaMKI, CaM- removal from the gene promoter. Interestingly, however, this effect
KII and CaMKIV, as well as the upstream CaMK kinase (CaMKK). appears to be unique to the FSHb gene because CaMK inhibition
These kinases have comprehensive functions and are involved in failed to block the effect of GnRH on the LHb gene, indicating that
gene expression, cell proliferation, apoptosis and cytoskeletal reor- GnRH likely directs de-repression of the LHb gene downstream of
ganisation, as well as synaptic development and plasticity and CaMK activation (26,33).
behaviour (21,25). CaMKI, CaMKII and CaMKK are widely expressed,
whereas CaMKIV is more tissue-restricted and is not found in the
CaMKII
gonadotrophs (26).
All of the CaMKs contain a Ca2+/CaM -binding region that over- CaMKII differs in structure from the other CaMKs and, unlike other
laps with an autoinhibitory domain. Binding of Ca2+/CaM disrupts CaMKs, it does not act as a monomer but forms part of a complex
the interaction between the autoinhibitory domain and the catalytic of 12 or more subunits with at least six catalytic domains extend-
domain, thus activating the kinase (21). CaMKK is an upstream acti- ing away from the core. The ability of this complex to contain
vator of CaMKI and CaMKIV that shares similar structure with the diverse proteins likely endows its ability to activate multiple down-
other CaMKs; however, after its initial activation, the CaMKKb iso- stream effects (21). Similar to other CaMKs, CaMKII also contains
form can also function independently of Ca2+/CaM because of the an autoinhibitory domain that blocks catalytic activity until Ca2+/
release of its autoinhibitory domain; moreover, it may also have CaM levels are increased. After the initial activation of one subunit
some autonomous activity through its autophosphorylation (21,27). of the holoenzyme by Ca2+/CaM, it can then phosphorylate a
This isoform also acts as a scaffold for a signalling complex includ- neighbouring Ca2+/CaM-bound subunit in the same complex. This
ing CaM and the CaMKK substrate, which was recently shown to phosphorylation disrupts the interaction between the autoinhibitory
include AMP-dependent kinase (AMPK) (28). Interestingly, GnRH domain and the catalytic site, such that the subunit subsequently
was shown to induce a prolonged increase in AMPK activity in the retains activity independent of Ca2+/CaM-binding, at the same time
gonadotrophs (29), although a role for CaMKK in this activation as also increasing the affinity of CaMKII for Ca2+/CaM. Thus, Ca2+/
has yet to be reported. CaM must bind two molecules in the same holoenzyme to attain
initial activation of CaMKII, which reflects the amount of available
Ca2+; however, subsequent CaMKII activity is maintained even after
CaMKI
transiently-elevated Ca2+ levels drop (21,34). Because of the unique
The activity of CaMKI is entirely dependent on activation by Ca2+/ characteristics of CaMKII, which render it highly sensitive to Ca2+
CaM, and CaMKI activity drops as soon as the levels of Ca2+ return pulse frequency, it was proposed that CaMKII might provide a
to basal levels. This dependency is a result of fact that the Ca2+/ mechanism of decoding Ca2+ oscillations that occur in response to
CaM-induced displacement of the autoinhibitory domain is required diverse stimuli (35,36).
to expose the threonine that is targeted by CaMKK. Phosphorylation GnRH activates CaMKII in rat primary pituitary cells and in go-
of this site by CaMKK increases CaMKI activity, probably by enhanc- nadotroph cell lines, and treatment with the CaMK inhibitor, KN-93,
ing the stability of the activation loop that increases the compe- decreased the GnRH induction of promoter activity and/or mRNA
tency of the catalytic site (21). levels of all three gonadotrophin subunit genes (9,26,37,38). More-
CaMKI is expressed in the aT3-1 partially-differentiated immorta- over, the differential expression of these genes was mimicked by
lised gonadotroph cells and is notably phosphorylated within 5 min the administration of Ca2+ pulses at frequencies similar to those
of GnRH treatment; this elevated state is still seen after 60 min of seen after GnRH treatment (18). It was therefore suggested by sev-
continuous GnRH exposure (26). CaMKI has a diverse set of protein eral groups that Ca2+/CaM- CaMKII might play a role in decoding
targets, including transcription factors and the class 2a histone GnRH pulse frequency towards gonadotrophin gene expression. In
deacetylases (HDACs), which shuttle between the nucleus and one model, it was proposed that CaMKII activation might favour
cytoplasm. The CaMK-mediated phosphorylation of these class 2a aGSU and LHb gene expression, which are dominant at higher
HDACs was shown to signal binding of the HDAC by 14-3-3 GnRH pulse frequencies, whereas at lower frequencies that favour

Journal of Neuroendocrinology, 2012, 24, 1463–1475 © 2012 The Authors.


Journal of Neuroendocrinology © 2012 British Society for Neuroendocrinology
1466 P. Melamed et al.

FSHb transcription, the activated CaMKII, which might be less cru-


Calcineurin regulates gonadotrophin gene expression via
cial for FSHb expression, would have time to decay (8). However, a
nuclear factor of activated T-cells (NFAT) and Nur77
latter study showed that CaMKII activation by GnRH at 30- or
240 min-pulses, which are optimal for LHb or FSHb activation, NFAT is the most extensively studied calcineurin target; first identi-
respectively, is not frequency-dependent (9). Moreover, a number of fied in T-cells, it is rapidly induced to activate the IL-2 promoter.
subsequent studies, both experimental and computational, have However, it is widely expressed and plays extensive roles in regulat-
indicated that feedback may be more crucial to the frequency ing gene expression for diverse aspects of cell function. Four NFAT
decoding, and have implicated MAPK phosphatases to play a key isoforms have been identified, NFAT1 (NFATc2), NFAT2 (NFATc1),
role (10,39,40), whereas feedback inhibition of gene-specific tran- NFAT3 (NFATc4) and NFAT4 (NFATc3), all of which are comprised of
scription factors has also been suggested (41,42). an N-terminal transactivation domain, a DNA-binding domain and
The ability of the CaMKII inhibitor to reduce GnRH-induced tran- a NFAT regulatory region. This regulatory region is phosphorylated
scription of all three gonadotrophin b-subunit genes indicates that, on multiple serines by various kinases, including casein kinase 1
even if it does not decode the GnRH pulse frequency, CaMKII does and glycogen synthase kinase 3, which act to repress NFAT activity
mediate some of the effects of GnRH, although its targets in this by keeping it in the cytoplasm. NFAT is activated by calcineurin-
pathway largely remain to be elucidated. Recently, however, GnRH- mediated dephosphorylation of these same residues, allowing the
activation of c-fos was shown to occur via CaMKII phosphorylation NFAT to translocate to the nucleus to activate transcription of its
of serum response factor, which likely plays a role in GnRH induc- target genes. In both the cytoplasm and nucleus, NFAT interacts
tion of FSHb and possibly aGSU transcription (43–46). with a variety of other transcription factors, which include AP-1,
GATA4, Foxp and MEF2, and these interactions play a crucial role in
determining the specificity of the NFAT-binding to its target genes.
Diverse roles for calcineurin in the gonadotroph
Nuclear NFAT is inactivated by phosphorylation, which causes it to
Calcineurin is a Ca2+/CaM-dependent serine/threonine protein phos- return to the cytoplasm (58,59).
phatase comprising two subunits: the catalytic calcineurin A sub- In aT3-1 cells, NFAT 2, 3 and 4 are expressed, with NFAT3 being
unit (CnA), and a Ca2+-binding regulatory subunit, calcineurin B notably translocated into the nucleus, in a calcineurin-dependent
(CnB). The CnA contains three regulatory domains: the CaM-binding manner, after GnRH treatment (50) (Fig. 1). In these cells, NFAT 2
domain, the autoinhibitory domain and a domain that interacts and 4 appear to be constitutively nuclear, although, in studies in
with CnB. In the absence of Ca2+/CaM, the autoinhibitory domain which NFAT2-EFP was transfected into LbT2 cells, the over-
binds in the active site and inhibits phosphatase activity. A rise in expressed protein did oscillate between cellular compartments in
intracellular Ca2+, which binds CnB, facilitates Ca2+/CaM binding by response to GnRH (57). NFAT3 is required for GnRH-induced aGSU
the CaM-binding domain of CnA, resulting in a displacement of the expression in the aT3-1 cells, whereas NFAT4 may play a role in its
autoinhibitory domain and activation of the phosphatase, which is basal expression. In support of a role for NFAT3 in mediating the
subsequently independent of CnB (47,48). effect of GnRH on aGSU gene expression, the calcineurin-respon-
CnA expression levels increase after GnRH treatment in both aT3- sive region of the promoter maps to a sequence ( 420 to
1 cells and the more fully differentiated immortalised gonadotrope 346 bp upstream of the tss) already shown to mediate both the
LbT2 cells (26,49,50). The activity of calcineurin, as well as other GnRH and calcium responses, and this common sequence
phosphatases, in mediating GnRH signals to gonadotrophin genes (tTTCCTGTT) contains a consensus NFAT-binding element (50,60,61).
are clearly tightly regulated by GnRH and likely crucial for maintain- Recently, calcineurin-driven NFAT was also reported to mediate
ing the integrity of the signal (10,40,51–54). It has been proposed GnRH-induced expression of the early response genes, Jun and
that a continual phosphorylation/dephosphorylation cycle of key Aft3. It was shown that NFAT binds the Jun promoter and interacts
substrates is required for GnRH stimulation of gonadotrophin with Jun through AP-1 sites (62). Given that AP-1 sites also regu-
genes to avoid desensitisation, that they act to down-regulate and late aGSU expression, this suggests an additional mechanism of the
constrain the magnitude or duration of GnRH signalling, and also role of NFAT with respect to transducing the effect of GnRH to this
that they play a role in decoding the GnRH pulse frequency gene.
(10,40,51,55,56). However, unlike the MAPK phosphatases, the effects NFAT also regulates FSHb gene transcription, although this effect
of calcineurin in these cells all appear to be stimulatory to gonado- is indirect, mediating GnRH stimulation of the Nur77 transcription
trophin gene expression and are mediated through multiple factor. The expression of the Nur77 is elevated by GnRH in a potent
targets. Inhibition of calcineurin is repressive to transcription and/ and rapid manner, and this is calcineurin-dependent (26,50,52,63)
or promoter activation of all three gonadotrophin subunits (Fig. 1). In T-cells, the transcription of Nur77 occurs via calcineurin-
(26,50,54,57). For the aGSU, calcineurin is both necessary and suffi- activated NFAT, which interacts with MEF2D at the Nur77 gene
cient to induce gene expression, whereas, for FSHb, it is necessary promoter (64). Also in the gonadotroph, NFAT3 is recruited to the
but not sufficient, although it clearly augments the effect of GnRH. Nur77 promoter after GnRH treatment concomitant with the asso-
For the LHb gene, its exact role remains to be elucidated, although it ciation of RNAPII, and NFAT appears to play a central role because
likely includes indirect effects such as those occurring via Pin1, its knockdown reduced the GnRH stimulatory effect on Nur77
which also impacts other aspect of gonadotroph cell function (see expression. In the aT3-1 cells, Nur77 over-expression is sufficient to
below) (Fig. 1). de-repress expression of the FSHb gene, whereas FSHb promoter

© 2012 The Authors. Journal of Neuroendocrinology, 2012, 24, 1463–1475


Journal of Neuroendocrinology © 2012 British Society for Neuroendocrinology
GnRH signalling downstream of calmodulin 1467

activity and transcript levels are reduced after NFAT3 knockdown in interaction with TORC1, presumably favouring the recruitment of
LbT2 cells, suggesting that NFAT3-activated Nur77 is required for CBP (74). Given that this serine is required for the full effect of
GnRH-induction of the FSHb gene (26,50). GnRH but not for that of TORC1, TORC1 may not be required for
Apart from the involvement of calcineurin in the GnRH stimula- the GnRH-stimulated FSHb promoter activity. However, TORC1 does
tion of Nur77 expression, it is likely also involved in the GnRH-acti- appear to play a role in regulating FSHb gene expression because
vated dephosphorylation of Nur77 at S354 (Fig. 1). Mutation of this TORC1 knockout male mice have significantly lower circulating FSH
residue indicates that the dephosphorylation is a means of activat- levels than their wild-type counterparts; although this was not seen
ing Nur77 transcriptional activity (26). This is also the mechanism in female mice, which were tested only at pro-oestrous (75). The
of Nur77 activation by adrenocorticotrophic hormone, involving role of TORC in FSHb gene expression has thus to be fully eluci-
dephosphorylation at the same residue, although, unlike at the dated, although it might provide a different mechanism of activat-
FSHb gene, the phosphorylation prevented DNA binding in the con- ing transcription as part of an alternatively regulated GnRH-
texts tested (65). Although we have yet to determine the function independent pathway, at the same time as still allowing for cross-
of this dephosphorylation in FSHb gene activation, Nur77 is known talk with the GnRH-activated cascade.
to interact with SMRT, which represses expression of the FSHb gene Although calcineurin is known to promote TORC nuclear localisa-
in these cells (26,66,67). It is therefore quite possible that the tion, and GnRH treatment causes dephosphorylation of TORC1 in a
dephosphorylation causes a switch in Nur77 interacting proteins, calcineurin-dependent manner, the levels of nuclear TORC1 were
allowing it to dissociate from SMRT and recruit coactivators. not increased by GnRH treatment. TORC1 appears to be inherently
unstable in these cells and, after GnRH is added, the existing TORC1
is rapidly degraded. Subsequently, the newly synthesised protein
A role for calcineurin-regulated Transducer of Regulated
appears to have a protected N-terminus which is associated with
CREB (TORC)
its enhanced activity (50). Instability of the TORC1 before GnRH
The TORCs are transcriptional coactivators whose activity is regu- treatment is a result of the actions of calpain and the proteasome,
lated by calcineurin-mediated dephosphorylation. Also known as and we have proposed that the degradation of the newly synthes-
CREB-Regulated Transcription Coactivators, this group of proteins ised TORC is regulated by GnRH actions on the calpain-calpastatin
was originally identified as interacting with, and potently increasing system, possibly through ERK-activated phosphorylation. This is
the activity of CREB, however, they are now known to coactivate similar to the regulation of PKCe degradation in rat pituitary GH4C1
additional transcription factors, including c-jun, c-fos, ATF1 and cells by thyrotrophin-releasing hormone (76). However, other expla-
ATF4 (68,69). Unlike the CREB coactivation by CBP, the actions of nations are also quite possible.
TORC are independent of CREB phosphorylation at Ser 133,
although TORC appears to coactivate at only a subset of CREB tar-
Wide-ranging effects of calcineurin via dephosphorylation
gets (70,71). Dephosphorylation of the TORC proteins by calcineurin
of Pin1
allows their nuclear import and thus their transcriptional activity.
Conversely, they are negatively regulated by phosphorylation, for We recently reported a novel target of calcineurin in the gonado-
example by protein kinase A-induced salt-inducible kinase, which trophs: the ubiquitously expressed peptidyl-prolyl isomerase, Pin1
prevents their nuclear localisation (72,73). (49). Pin1 is the only known enzyme that binds a phosphorylated
Although enriched in the brain and lymphocytes, TORC proteins serine or threonine preceding a proline (pSer/Thr-Pro) and isomeris-
are expressed quite ubiquitously at low levels, and we were able to es the cis-trans conformation of the prolyl-peptidyl bond. It con-
detect TORC1 and TORC3 proteins (but not TORC2) in the aT3-1 cells sists of a short N-terminal WW domain that binds only to pSer/
(Melamed, unpublished data). TORC1 over-expression was sufficient Thr-Pro motifs, providing target specificity, and a C-terminal cata-
to increase aGSU promoter activity, whereas its knockdown indicated lytic domain that isomerises the prolyl-peptidyl bond. Pin1 thus
that TORC1 plays an essential role in GnRH-activated aGSU tran- mediates a phosphorylation-induced conformational change in the
scription. However, this gene is not regulated by CREB, and TORC target protein, which leads to altered activity, providing additional
does not appear to be recruited to the aGSU promoter by CREB. insight into the role of this post-translational modification (77,78).
Because we were unable to discern synergistic effects of TORC with Notably, Ser/Thr-Pro is a common motif that is phosphorylated by
any transcription factor in the activation of this gene, the factor various kinases, particularly by the MAPKs which form an integral
responsible for recruiting TORC remains to be identified, although it part of GnRH-induced signal transduction, and Pin1-catalysed prolyl
may already be found at the promoter in untreated cells (50). isomerisation has been shown to be involved in a wide variety of
CREB clearly does play a role in GnRH-mediated activation of contexts including the regulation of transcription, protein–protein
the FSHb gene (74), and it acts synergistically with TORC1 (50). The interaction and/or protein subcellular localisation (77,78).
CREB-TORC1 synergy is further enhanced by Nur77, although Pin1 activity is tightly regulated via several mechanisms that
Nur77 and TORC also exert a powerful synergistic effect without determine its ability to bind the substrate, its catalytic activity and/
the addition of CREB. TORC1 over-expression also increased the or its cellular localisation (Fig. 1). Phosphorylation of Pin1 at S16,
effect of GnRH, although its knockdown revealed more of an effect in the pSer/Thr-Pro binding pocket, is a key mechanism of control
on basal promoter activity than on that induced by GnRH. Notably, because it prevents the WW domain from interacting with pSer/
the GnRH-induced phosphorylation of CREB at S133 prevents its Thr-Pro motifs on its substrates and may facilitate Pin1 nuclear

Journal of Neuroendocrinology, 2012, 24, 1463–1475 © 2012 The Authors.


Journal of Neuroendocrinology © 2012 British Society for Neuroendocrinology
1468 P. Melamed et al.

export (79). In the gonadotrophs, intriguingly, GnRH increases levels expression of cyclin D1, although not cdk4 (Fig. 2B,C). Paradoxically,
of phosphoPin1, as does forskolin or, to a lesser extent, phorbol the cdk4 promoter is reportedly inhibited by calcineurin-regulated
12-myristate 13-acetate (PMA) treatment. This suggests a role for NFATc2, while calcineurin is also able to dephosphorylate cdk4
GnRH-activated PKC, or protein kinase A which has been implicated directly, leading to its inactivation (85,86). GnRH-induced NFAT
in Pin1 phosphorylation in the past (79). Conversely however, Pin1 activity does not appear to play a similar role in GnRH-activated
and calcineurin co-precipitate and this interaction increases after proliferation of the immature gonadotrophs because these initial
GnRH treatment. The GnRH-activated calcineurin acts to dephos- studies reveal that cdk4 expression is reduced by cyclosporin A but
phorylate Pin1, thus keeping it in its active state; this may be cru- not affected by GnRH (Fig. 2C). Any role for GnRH-activated post-
cial to counteract the apparently paradoxical increase in Pin1 translational regulation of cdk4 activity via calcineurin remains to
phosphorylation after GnRH treatment (49). be investigated.
One of the targets of Pin1 in the gonadotrophs is the transcrip- GnRH likely also affects cell cycle progression via its calcineurin-
tion factor Sf-1. Pin1 facilitates the phosphorylation-to-ubiquitina- mediated dephosphorylation of Pin1, which regulates transcription
tion transition of Sf-1 (Fig. 1), which is required for Sf-1 of cyclin D1, as well as the accumulation of both cyclin D1 and
interaction with Pitx-1; this interaction is essential for the synergis- b-catenin in the nucleus (78,87–89). Pin1 stimulates cyclin D1 gene
tic transcriptional activation of the LHb gene (49,80). However, Sf-1 expression via enhancing the activity of JNK, as well as stabilising
is required for the full GnRH response of all three gonadotrophin c-jun, both of which are activated downstream of GnRH in a man-
subunit genes, while many of the other GnRH-activated transcrip- ner that is dependent on b-catenin (4,52,82,90–92). GnRH and Pin1
tion factors that regulate gene expression in the gonadotrophs also have been shown to stimulate translocation of b-catenin to the
contain pSer/Thr-Pro motifs and are activated through phosphoryla- nucleus (91,93), and we show here in a novel study that b-catenin
tion by MAPKs, making them likely Pin1 targets. plays a role in GnRH-induced cyclin D1 gene expression, as well as
In other contexts, Pin1 has been shown to bind and regulate the its induction of the proliferation of aT3-1 cells (Fig. 2D–E).
activity of additional modulators of gonadotrophin gene expression, Additional anti-proliferative effects of calcineurin-inhibition, as
including ERa, c-Jun, b-catenin, SMADs 2 and 3, SMRT, NFAT and shown in various cell types, are mediated via growth factor-medi-
TORCs (2,3,43,44,50,78,81-84). For NFAT and the TORCs, the effect ated increases in p21 and/or the accumulation of p27, which are
of the Pin1 is to repress transcriptional activity of the factor by both cyclin-dependent kinase inhibitors (CKIs) that prevent activa-
keeping it in the cytoplasm; moreover, the interaction of NFAT with tion of the cyclin/cdk complexes (25,94). In our preliminary studies,
Pin1 specifically prevents NFAT activation by calcineurin (81,83). GnRH treatment led to a marginal increase in p27 and p21 levels
Although the role of Pin1 in regulating NFAT and/or TORC activity in aT3-1 cells, and this was inhibited by cyclosporin A treatment
in the gonadotrophs has yet to be verified, these studies indicate (Fig. 2F-G), suggesting an involvement of calcineurin in their tran-
that cytoplasmic, presumably phosphorylated, Pin1 also plays a role scription, as seen previously in other cell contexts (95,96). At low
in determining the signalling pathways to differential gene expres- levels, these CKIs may not suffice to inactivate the cyclin/cdk com-
sion, and may help contribute an explanation for the GnRH-induc- plex and may actually promote their activation through facilitating
tion of Pin1 phosphorylation. This paradoxical regulation further assembly of the complexes, increasing their nuclear import and by
highlights the important role of calcineurin in the regulation of stabilising cyclin D (25).
Pin1 function by phosphorylation/dephosphorylation and indicates In LbT2 cells, GnRH no longer induces cell proliferation but reduces
that Pin1 plays a likely widespread role not only in the gonado- cell numbers through inhibiting proliferation and/or inducing apopto-
troph, but also in MAPK signalling cascades in more diverse sis (1,6). GnRH is seen to have anti-proliferative/pro-apoptotic effects
contexts. in a variety of other cells as well, although, for the most part, the
mechanisms have either not been elucidated or appear to vary
between the different cell types (97–100). We are currently investi-
Effects of GnRH via Ca2+/CaM signalling on the cell cycle
gating the mechanisms through which GnRH reduces cell number of
and gonadotroph proliferation
the LbT2 cells, and the reasons for the apparent shift in response with
Ca2+/CaM signalling plays a variety of roles in the regulation of the differentiation. However, GnRH was reported to increase p21 mRNA
cell cycle, and CaM levels vary through the cell cycle, increasing levels quite significantly (three- to six-fold) in LbT2 cells (4,63,101),
just before the S-phase. CaM over-expression accelerates prolifera- suggesting that this CKI induction might play a role in blocking cell
tion, apparently due to a shortening of progression through G1, cycle progression. GnRH was also shown to reduce expression of a
whereas, conversely, a reduction of CaM levels inhibits cell prolifer- number of the cyclins, cyclin-associated kinases and several subunits
ation (25). We have already shown that GnRH induces an increase of the anaphase-promoting complex (4). Although these cells were
in the number of aT3-1 cells (6) and we now report preliminary immortalised using the SV40 large T antigen, which could potentially
experiments showing that this effect is abolished by treatment with alter their proliferative response, the vast majority of large T-respon-
cyclosporin A (Fig. 2A), indicating that it is likely mediated primarily sive genes are not affected by GnRH and/or activin treatments (4),
by the Ca2+/CaM activation of calcineurin. indicating that the presence of SV40 large T antigen is unlikely to
It has been suggested that Ca2+ and CaM are required for the confound these conclusions. Confirmation of this effect of GnRH in
activation of cyclin D1/cdk4 and pRb hyperphosphorylation and, primary partially-differentiated embryonic gonadotroph cells in
indeed, we show here in a pilot study that GnRH increases the embryos is technically difficult, however GnRH was seen to increase

© 2012 The Authors. Journal of Neuroendocrinology, 2012, 24, 1463–1475


Journal of Neuroendocrinology © 2012 British Society for Neuroendocrinology
GnRH signalling downstream of calmodulin 1469

(A) (B) (C) Cdk4


1.8 GnRH (h) 0 3 5 8 1.20

treated (normalised to RPLPO)


C B Control

mRNA levels fold over non


1.6 B GnRH
Absorbance (fold over

Cyclin D1 1.00
nontreated control)

1.4 Control
1.2 B 0.80
GnRH β -actin
1 A A
0.60
0.8
0.6 0.40
A A
0.4 0.20
0.2
0 0.00
NT CSA
(D) NT CSA
(E) 2
GnRH: – + – + B
1.8 Control
siRNA: GFP GFP β -catenin β -catenin siRNA: GFP β -catenin GnRH
1.6

Absorbance (fold over


β -catenin

nontreated control)
Cyclin D1 1.4
A
GAPDH 1.2
β -actin A
1
A
Normalised 1 2.93 2.79 1.28 1.06 1.34 1.22 0.8
values 0.6

(F) p27 (G) p21 0.4


B 2.4 B Control 0.2
Control
mRNA levels fold over nontreated

1.4
mRNA levels fold over nontreated

GnRH 0
A GnRH
1.2 2.0 siGFP siβ -catenin
(normalised to RPLPO)
(normalised to RPLPO)

A
1.0 A 1.6 A
0.8 A
1.2 A
0.6
0.8
0.4
0.2 0.4

0.0 0.0
NT CSA NT CSA

Fig. 2. Calcineurin plays a role in gonadotrophin-releasing hormone (GnRH)-activated aT3-1 cell proliferation. (A) An XTT assay to assess cell numbers was car-
ried out in aT3-1 cells after some were exposed to GnRH for 24 h, with or without 30 min cyclosporine A (CsA) pretreatment. Absorbance is expressed relative
to that in nontreated (NT) cells without GnRH; mean ± SEM (n = 7–9). ANOVA followed by a Bonferroni t-test compared all means and those that are similar
(P > 0.05) are designated the same letter. (B) Cyclin D1 protein levels (with b-actin as internal control) were assessed by western analysis in aT3-1 cells treated
with GnRH (10 nM) for 0–8 h. (C) Real-time quantitative polymerase chain reaction (PCR) analysis was performed to measure the effects of GnRH (8 h) with
and without CsA pre-treatment (10 min before GnRH) on mRNA levels of Cdk4. Transcript levels were quantified using a standard curve and are represented
relative to levels in untreated controls, after normalisation to levels of large ribosomal protein (RPLPO). Mean ± SEM (n = 3–4); statistical analysis as in Fig. 2(A).
(D) Small interfering (si)RNA targeting b-catenin, or green fluorescent protein (GFP) as control (both in pSUPER) was transfected 48 h before GnRH treatment
(3 h), after which RNA was extracted, reverse transcribed and PCR amplification carried out for cyclin D1 and b-actin. PCR products were resolved in agarose
gel and the intensity of each band quantified relative to untreated siGFP-control, after normalisation with b-actin levels. Western analysis for the b-catenin
knockdown is also shown. (E) The siRNA targeting b-catenin was transfected and, after 40 h, some cells were treated with GnRH (20 nM for 16 h) before a
bromodeoxyuridine assay was performed to assess cell proliferation. Absorbance is expressed and the data are presented as in Fig 2(A); mean ± SEM (n = 4).
(F, G) Real-time quantitative PCR analysis was carried out as in Fig. 2(C) to assess effects of CsA and/or GnRH on mRNA levels of (F) p27 and (G) p21. Experimental
details and data presentation are as in Fig. 2(C). All experiments were carried out on at least three independent occasions and representative results are shown.

gonadotrope proliferation in adult mice and rats under certain cir- cytoplasm, allowing activation of the caspases and also of the nuc-
cumstances, including at specific stages of the oestrous cycle or after leases required for the apoptotic response (16,105–107). However,
castration (102–104), suggesting a latent ability to proliferate in other forms of cell death triggered by Ca2+-mobilisation are CaM-
response to GnRH that can be moderated by other factors including dependent, including the activation of death-associated protein
steroids. kinase and AMPK, which are involved in autophagic cell death
Calcineurin is clearly not the only way in which calcium influx (108). AMPK is activated in gonadotrophs in response to GnRH,
stimulates an apoptotic response, which is driven in many cases although neither protein has yet been linked to cell death after
by CaM-independent mechanisms via caspase activation. The GnRH treatment (29). A specialised form of apoptosis, anoikis,
Ca2+-influx into the mitochondria increases cytochrome C in the involving detachment of cells from their surrounding matrix, is also

Journal of Neuroendocrinology, 2012, 24, 1463–1475 © 2012 The Authors.


Journal of Neuroendocrinology © 2012 British Society for Neuroendocrinology
1470 P. Melamed et al.

regulated by Ca2+-CaM, in which changes in Ca2+ homeostasis lead differ structurally from other GTPases in that they do not contain
to the activation of Rho kinase signalling (108). Although the lipid modification for anchorage to the membrane. Gem and other
induction of this form of apoptosis by GnRH has yet to be members of this group have several roles, including inhibiting Ca2+
reported, GnRH does induce major cytoskeletal rearrangement and channels, while Gem also affects rearrangement of the cytoskeleton
gonadotroph cell migration, likely involving Rho signalling cascades through altering Rho signalling. The activity and localisation of
(109,110). GnRH-induced Rho signalling may be regulated in part Gem are regulated through CaM and 14-3-3 proteins (111,112).
by a small GTP-binding protein, Gem, which is activated by GnRH Gem is expressed at higher levels in aT3-1 than in LbT2 cells,
and regulated by CaM, and was shown, in other contexts, to bind while in the LbT2 cells, its mRNA levels increase after GnRH treat-
Rho kinase to alter downstream signalling (111). ment (4,6,52). Given that Gem inactivates the L-type VSCC that are
utilised by GnRH for Ca2+ influx, it was suggested that Gem might
reduce external calcium entry as part of a negative-feedback loop,
A role for calmodulin in moderating GnRH-induced
so controlling pathways downstream of calcium (8,52,113). Consis-
signalling through negative-feedback?
tent with this, we found that Gem, which is initially found
throughout aT3-1 cells, moves out of the nucleus after exposure to
Gem and its regulation by GnRH
GnRH for 15–30 min (Fig. 3A). This effect appears to be indepen-
Gem belongs to the RGK (comprising Rad, Gem and Kir, as well as dent of CaM because a mutant Gem that cannot bind CaM
Rem1 and Rem2) subfamily of small Ras-related GTP-binding (W269G) (113) translocated to the cytoplasm in a manner similar
proteins that moderate various G-protein coupled pathways. These to the wild-type protein (Fig. 3A). The export is likely therefore

(A) (B)
WT Gem HDAC7 W269G Gem HDAC7 6 Control a
Firefly luciferase activity (fold over +GnRH
Before 5
normalised to Renilla)
nontreated control,
GnRH b
4
c

15 min 3
GnRH
2
A A B
30 min 1
GnRH
0
GSU Gem WT Gem W269G

(C) siGem
4 Control D – +
Firefly luciferase activity (fold over

Gem
3.5 +GnRH
Tubulin
normalised to Renilla)
nontreated control,

3
2.5 C

2
1.5 B
1
A
0.5
0
GSU + siGem

Fig. 3. Gonadotrophin-releasing hormone (GnRH)/calmodulin (CaM)-regulated Gem in gonadotrophs. (A) aT3-1 cells plated on glass cover-slips were co-trans-
fected with expression vectors for yellow fluorescent protein-tagged histone deacetylase (HDAC) 7 together with enhanced green fluorescemt protein (EGFP)-
tagged wild-type (WT) Gem (first and second columns) or EGFP-tagged W269G Gem mutant (third and fourth columns). After 48 h, GnRH in phenol-red free
medium was added. Confocal images of cells were captured initially and after 15 and 30 min. A single representative cell is shown for each treatment/time
point; scale bars = 10 lm. (B) aT3-1 cells were transfected with mouse glycoprotein hormone a-subunit promoter-luciferase reporter gene (aGSU), and WT
Gem or the W269G mutant were overexpressed, and samples exposed to GnRH treatment (100 nM, 8 h). After 60 h, luciferase assays were performed, and lev-
els normalised to those of Renilla; the results are expressed as the fold induction over the mouse aGSU-luciferase control-transfected and untreated cells. Sta-
tistical analysis (ANOVA followed by Bonferroni t-test) compared mean values separately for untreated (upper case) and GnRH-treated (lower case) cells, and
those designated by different letters are significantly different (P < 0.05); mean ± SEM (n = 5–6). (C) aT3-1 cells were transfected with mouse aGSU-luciferase
(as above) and some with small interfering (si)RNA targeting Gem (in pSUPER). Some of the cells were treated with GnRH, luciferase assays performed, values
normalised and presented all as above (n = 6). All experiments were carried out on at least three independent occasions and representative results are shown.
Insert: western blot showing levels of Gem in aT3-1 cells 60 h after transfection of siGEM.

© 2012 The Authors. Journal of Neuroendocrinology, 2012, 24, 1463–1475


Journal of Neuroendocrinology © 2012 British Society for Neuroendocrinology
GnRH signalling downstream of calmodulin 1471

driven by GnRH-activated phosphorylation of Gem, which facilitates signalling complex into lipid rafts together with the GnRHR was
recognition by 14-3-3 proteins and can be targeted by a number of proposed to integrate Ca2+ and ERK signalling and poise them for
kinases, as reported in other cell contexts (111,114,115). Both the GnRH-activated response. Both CaM and 14-3-3b colocalise in
groups of cells also underwent morphological changes after expo- these membrane-associated lipid rafts together with the GnRHR, Gq
sure to GnRH (Fig. 3A), which has already been reported in these and c-Raf kinase, and it was suggested that CaM serves as a link
cells (5), and is in keeping with actions of Gem on cytoskeletal between the VSCC, Ca2+ and ERK (13,53). Indeed, inhibition of CaM
rearrangement (115). blocks GnRH- induced ERK1/2 but not JNK activation in aT3-1 cells,
Although GnRH regulates Gem localisation, our preliminary stud- and it also attenuated ERK-dependent gene reporter activity of c-
ies did not provide any evidence of Gem exerting a negative effect fos, aGSU and MAPK phosphatase-2 promoters, in a manner that is
on gonadotrophin gene expression. GnRH-induced aGSU promoter at least partially CaMK-independent (53). Although RGS2 has yet to
activity in aT3-1 cells is highly dependent on calcium influx (60), examined in the context of these gonadotroph lipid rafts, it appears
yet, in our preliminary studies using aGSU-promoter-driven lucifer- likely that it is part of this signalling complex, being localised and/or
ase assays, over-expression of wild-type Gem did not reduce regulated in the rafts by CaM, to provide a mechanism of Ca2+-
GnRH-stimulated promoter activity but, instead, had a minor stimu- sensing in GnRH-induced ERK signalling.
latory effect, although the W269G mutant reduced promoter activ- Additional signalling complexes associated with ERK-activation
ity by approximately 30% (Fig. 3B). Furthermore, small interfering by GnRH are expected also to be regulated by CaM-activated path-
RNA-mediated partial knockdown of Gem also had an inhibitory ways (12,13,124–127). GnRH stimulates the rapid assembly of an
effect on the promoter activity (Fig. 3C). Taken together, these ERK-activating complex that contains the proline-rich tyrosine
results indicate that Gem does not exert negative-feedback on the kinase, Pyk2, which serves as a scaffold for c-Src, Grb2 and Sos
effect of GnRH but may perhaps play a novel positive role, possibly and is essential for ERK-dependent gene transcription. The catalytic
being regulated through its interaction with CaM in the cytoplasm. domain of Pyk2 interacts directly with Ca2+-CaM, and this is
required for its phosphorylation after GnRH exposure, as well as its
functional activity. The transmission of the ERK-activating signal to
RGS proteins
the nucleus by the Pyk complex appears to be via focal adhesion
RGS proteins, which act as GTPase-activating proteins of Ga, complexes and is dependent on a functional actin cytoskeleton. Its
including Gaq that transduces much of the effect of GnRH, have role in ERK activation and translocation to the nucleus led to the
also been implicated in modulating GnRH-induced signalling and suggestion that this complex also may form a link between VSCC
these too are regulated by CaM. RGS2 expression is increased by Ca2+ signals and the ERK activation pathway (12,13,125,126).
GnRH and it was proposed that the ability of RGS2 to down-regu- The GnRHR, c-Src, and focal adhesion kinase (FAK) are found
late Gaq activity might contribute to the frequency-dependent localised together in an additional signalling complex, likely also in
downstream gene activation (8,52). In keeping with this, RGS2 lipid rafts, that is restructured upon GnRH treatment. It was pro-
over-expression in aT3-1 cells reduced GnRH signalling via Gaq/11, posed that this complex acts to restrict the amount of active ERK
resulting in a marked reduction in phospholipase C activation, while from moving to the nucleus, and suggested that the ERK might
also decreasing GnRH-induced mRNA levels of Egr-1 and c-fos direct phosphorylation of FAK and paxillin to mediate some of the
(116,117). GnRH-induced changes in cell morphology (127,128). GnRH has
Although RGS repression of Gaq/11 can be inhibited by various been shown to promote cytoskeletal rearrangement, cell morphol-
kinases including PKC, as well as by phosphatidylinositol-3,4,5,-tris- ogy and adhesion, and Rho-kinase pathways were implicated in this
phosphate (PIP3), it is restored by Ca2+/CaM (118,119). It was sug- effect (110). It is an interesting speculation that Gem, which is reg-
gested that, in cardiac myocytes, the cyclical inhibition of Gaq/11 is ulated by GnRH and known to alter cell morphology by affecting
determined by CaM-stimulated RGS4, which inactivates Gaq/11 Rho kinase substrate specificity, might play a role in this complex.
leading to a reduction in the formation of PIP3; this results in a
drop in Ca2+ levels that leads to CaM dissociation from RGS4, after
Conclusions and future directions
which PIP3-mediated repression of RGS4 is restored and once more
inhibits Gaq/11 (118,120). Such a role for RGS2 in negative-feed- GnRH has far-reaching effects on the gonadotrophs beyond merely
back during GnRH-induced signalling in the gonadotroph appears inducing release and synthesis of the gonadotrophin hormones, and
feasible, although this has yet to be demonstrated. CaM-driven pathways, which are activated in response to increases
in Ca2+, clearly play a major role in many of these effects, as high-
lighted in this review. Although we have focused on recent literature
Ca2+/CaM-sensitive GnRHR-ERK signalling complexes
describing the effects of calcineurin and CaMKs, this is likely only a
RGS proteins are localised in membrane-associated lipid rafts where subset of the pathways activated downstream of CaM in response to
they interact with various scaffold proteins to concentrate these GnRH. Hundreds of proteins are known to be bound by CaM, includ-
proteins with the G-protein coupled receptor for regulation of ing various DEAD/H box proteins and enzymes involved in protein
downstream signalling, and disruption of these rafts was seen to ubiquitination and degradation, and there is particularly strong
inhibit the interaction between RGS4 and CaM (118,121–123). In the evidence for its dominant cytoplasmic roles in both signalling and
gonadotrophs, compartmentalisation of a putative Ca2+-sensitive vesicular trafficking (129–131). However, CaM also translocates to

Journal of Neuroendocrinology, 2012, 24, 1463–1475 © 2012 The Authors.


Journal of Neuroendocrinology © 2012 British Society for Neuroendocrinology
1472 P. Melamed et al.

the nucleus in response to various stimuli, and although its nuclear 12 Naor Z. Signaling by G-protein-coupled receptor (GPCR): studies on
functions have barely been studied, it appears to be involved in sig- the GnRH receptor. Front Neuroendocrinol 2009; 30: 10–29.
nalling to transcription; in neuronal cells, it was suggested to provide 13 Bliss SP, Navratil AM, Xie JJ, Roberson MS. GnRH signaling, the go-
nadotrope and endocrine control of fertility. Front Neuroendocrinol
a cellular ‘memory’ in facilitating repeated neural activity (132).
2010; 31: 322–340.
Moreover, a novel role for CaM was demonstrated recently, in which 14 Stojilkovic SS. Pituitary cell type-specific electrical activity, calcium sig-
it binds the signal peptides of small secretory proteins, and acts as a naling and secretion. Biol Res 2006; 39: 403–423.
chaperone for their translocation through the cytosol and into the 15 Stojilkovic SS, Catt KJ. Novel aspects of GnRH-induced intracellular sig-
endoplasmic reticulum (133). We have likely only just begun to naling and secretion in pituitary gonadotrophs. J Neuroendocrinol
understand the role of CaM in GnRH signalling in the gonadotroph. 1995; 7: 739–757.
16 Berridge MJ, Lipp P, Bootman MD. The versatility and universality of
calcium signalling. Nat Rev Mol Cell Biol 2000; 1: 11–21.
Acknowledgements 17 Berridge MJ, Bootman MD, Roderick HL. Calcium signalling: dynamics,
homeostasis and remodelling. Nat Rev Mol Cell Biol 2003; 4: 517–529.
This research was supported by the Technion VPR fund-Mallat family 18 Haisenleder DJ, Yasin M, Marshall JC. Gonadotropin subunit and gona-
research fund, and the European Framework programme Marie Curie dotropin-releasing hormone receptor gene expression are regulated by
Actions. alterations in the frequency of calcium pulsatile signals. Endocrinology
1997; 138: 5227–5230.
Received 15 March 2012, 19 Chin D, Means AR. Calmodulin: a prototypical calcium sensor. Trends
revised 24 June 2012, Cell Biol 2000; 10: 322–328.
accepted 3 July 2012 20 Means AR. The year in basic science: calmodulin kinase cascades. Mol
Endocrinol 2008; 22: 2759–2765.
21 Swulius MT, Waxham MN. Ca(2+)/calmodulin-dependent protein kinas-
References es. Cell Mol Life Sci 2008; 65: 2637–2657.
22 Izumi S, Iwashita M, Makino T, Saito S, Sakamoto S, Takeda Y, Nozawa
1 Miles LE, Hanyaloglu AC, Dromey JR, Pfleger KD, Eidne KA. Gonadotro-
S. Phorbol ester-induced LH release in pituitary gonadotropes: effects
pin-releasing hormone receptor-mediated growth suppression of
of antagonists of calmodulin and GnRH. Endocrinol Jpn 1991; 38: 195
immortalized LbT2 gonadotrope and stable HEK293 cell lines. Endocri-
–204.
nology 2004; 145: 194–204.
23 Conn PM, Chafouleas JG, Rogers D, Means AR. Gonadotropin releasing
2 Luo M, Koh M, Feng J, Wu Q, Melamed P. Cross-talk in hormonally-
hormone stimulates calmodulin redistribution in rat pituitary. Nature
regulated gene transcription through induction of estrogen receptor
1981; 292: 264–265.
ubiquitylation. Mol Cell Biol 2005; 25: 7386–7398.
24 Cesnjaj M, Catt KJ, Stojilkovic SS. Coordinate actions of calcium and
3 Melamed P, Abdul Kadir MN, Wijeweera A, Seah S. Transcription of
protein kinase-C in the expression of primary response genes in pitui-
gonadotropin b subunit genes involves cross-talk between the tran-
tary gonadotropes. Endocrinology 1994; 135: 692–701.
scription factors and co-regulators that mediate actions of the regula-
25 Kahl CR, Means AR. Regulation of cell cycle progression by calcium/
tory hormones. Mol Cell Endocrinol 2006a; 252: 167–183.
calmodulin-dependent pathways. Endocr Rev 2003; 24: 719–736.
4 Zhang H, Bailey JS, Coss D, Lin B, Tsutsumi R, Lawson MA, Mellon PL,
26 Lim S, Luo M, Koh M, Yang M, Abdul Kadir MN, Tan JH, Ye Z, Wang W,
Webster NJG. Activin modulates the transcriptional response of LbT2
Melamed P. Distinct mechanisms involving diverse histone deacetylases
cells to gonadotropin-releasing hormone and alters cellular prolifera-
repress expression of the two gonadotropin b-subunit genes in imma-
tion. Mol Endocrinol 2006; 20: 2909–2930.
ture gonadotropes, and their actions are overcome by GnRH. Mol Cell
5 Navratil AM, Knoll JG, Whitesell JD, Tobet SA, Clay CM. Neuroendocrine
Biol 2007; 27: 4105–4120.
plasticity in the anterior pituitary: gonadotropin-releasing hormone-
27 Tokumitsu H, Hatano N, Fujimoto T, Yurimoto S, Kobayashi R. Generation
mediated movement in vitro and in vivo. Endocrinology 2007; 148:
of autonomous activity of Ca(2+)/calmodulin-dependent protein kinase
1736–1744.
kinase b by autophosphorylation. Biochemistry 2011; 50: 8193–8201.
6 Feng J, Lawson MA, Melamed P. A proteomic comparison of immature
28 Green MF, Anderson KA, Means AR. Characterization of the CaMKK
and mature mouse gonadotropes reveals novel differentially expressed
b-AMPK signaling complex. Cell Signal 2011; 23: 2005–2012.
nuclear proteins that regulate gonadotropin gene transcription and
29 Andrade J, Quinn J, Rodriguez AL, Shupnik MA. AMPK is a key interme-
RNA splicing. Biol Reprod 2008; 79: 546–561.
diary in GnRH- and Insulin-stimulated LHb gene transcription. Endocr
7 Burger LL, Haisenleder DJ, Dalkin AC, Marshall JC. Regulation of gonado-
Rev 2010; 31: S1175.
tropin subunit gene transcription. J Mol Endocrinol 2004; 33: 559–584.
30 McKinsey TA, Zhang CL, Lu JR, Olson EN. Signal-dependent nuclear
8 Ferris HA, Shupnik MA. Mechanisms for pulsatile regulation of the
export of a histone deacetylase regulates muscle differentiation. Nature
gonadotropin subunit genes by GnRH1. Biol Reprod 2006; 74: 993–
2000; 408: 106–111.
998.
31 McKinsey TA, Zhang CL, Olson EN. Identification of a signal-responsive
9 Burger LL, Haisenleder DJ, Aylor KW, Marshall JC. Regulation of intra-
nuclear export sequence in class II histone deacetylases. Mol Cell Biol
cellular signaling cascades by GnRH pulse frequency in the rat pitui-
2001; 21: 6312–6321.
tary: roles for CaMK II, ERK, and JNK activation. Biol Reprod 2008; 79:
32 Verdin E, Dequiedt F, Kasler HG. Class II histone deacetylases: versatile
947–953.
regulators. Trends Genet 2003; 19: 286–293.
10 Lim S, Pnueli L, Tan JH, Naor Z, Rajagopal G, Melamed P. Negative
33 Melamed P. The role of histone deacetylases in expression of gonado-
feedback governs gonadotrope frequency-decoding of gonadotropin
tropin subunit genes. Trends Endocrinol Metab 2008; 19: 25–31.
releasing hormone pulse-frequency. PLoS ONE 2009; 4: e7244.
34 Soderling TR, Chang B, Brickey D. Cellular signaling through multifunc-
11 Plant TM, Gay VL, Marshall GR, Arslan M. Puberty in monkeys is trig-
tional Ca2+/calmodulin-dependent protein kinase II. J Biol Chem 2001;
gered by chemical stimulation of the hypothalamus. Proc Natl Acad
276: 3719–3722.
Sci U S A 1989; 86: 2506–2510.

© 2012 The Authors. Journal of Neuroendocrinology, 2012, 24, 1463–1475


Journal of Neuroendocrinology © 2012 British Society for Neuroendocrinology
GnRH signalling downstream of calmodulin 1473

35 De Koninck P, Schulman H. Sensitivity of CaM kinase II to the fre- 53 Roberson MS, Bliss SP, Xie JJ, Navratil AM, Farmerie TA, Wolfe MW,
quency of Ca2+ oscillations. Science 1998; 279: 227–230. Clay CM. Gonadotropin-releasing hormone induction of extracellular-
36 Dupont G, Houart G, De Koninck P. Sensitivity of CaM kinase II to the signal regulated kinase is blocked by inhibition of calmodulin. Mol
frequency of Ca2+ oscillations: a simple model. Cell Calcium 2003; 34: Endocrinol 2005; 19: 2412–2423.
485–497. 54 Armstrong SP, Caunt CJ, Finch AR, McArdle CA. Using automated
37 Haisenleder DJ, Ferris HA, Shupnik MA. The calcium component of gon- imaging to interrogate gonadotropin-releasing hormone receptor traf-
adotropin-releasing hormone-stimulated luteinizing hormone subunit ficking and function. Mol Cell Endocrinol 2011; 331: 194–204.
gene transcription is mediated by calcium/calmodulin-dependent pro- 55 Marantz Y, Reiss N, Przedecki F, Naor Z. Involvement of protein phos-
tein kinase type II. Endocrinology 2003; 144: 2409–2416. phatases in gonadotropin-releasing-hormone regulated gonadotropin
38 Haisenleder DJ, Burger LL, Aylor KW, Dalkin AC, Marshall JC. Gonado- secretion. Mol Cell Endocrinol 1995; 111: 7–11.
tropin-releasing hormone stimulation of gonadotropin subunit tran- 56 Purwana IN, Kanasaki H, Oride A, Miyazaki K. Induction of dual specificity
scription: evidence for the involvement of calcium/calmodulin- phosphatase 1 (DUSP1) by gonadotropin-releasing hormone (GnRH) and
dependent kinase II (Ca/CAMK II) activation in rat pituitaries. Endocri- the role for gonadotropin subunit gene expression in mouse pituitary
nology 2003; 144: 2768–2774. gonadotrope LbT2 cells. Biol Reprod 2010; 82: 352–362.
39 Krakauer DC, Page KM, Sealfon S. Module dynamics of the GnRH signal 57 Armstrong SP, Caunt CJ, Fowkes RC, Tsaneva-Atanasova K, McArdle
transduction network. J Theor Biol 2002; 218: 457–470. CA. Pulsatile and sustained gonadotropin-releasing hormone (GnRH)
40 Nguyen KA, Intriago RE, Upadhyay HC, Santos SJ, Webster NJ, Lawson receptor signaling does the Ca2+/NFAT signaling pathway decode GnRH
MA. Modulation of gonadotropin-releasing hormone-induced extracellu- pulse frequency? J Biol Chem 2009; 284: 35746–35757.
lar signal-regulated kinase activation by dual-specificity protein phos- 58 Wu H, Peisley A, Graef IA, Crabtree GR. NFAT signaling and the inven-
phatase 1 in LbT2 gonadotropes. Endocrinology 2010; 151: 4882–4893. tion of vertebrates. Trends Cell Biol 2007; 17: 251–260.
41 Lawson MA, Tsutsumi R, Zhang H, Talukdar I, Butler BK, Santos SJ, 59 Mueller MR, Rao A. NFAT, immunity and cancer: a transcription factor
Mellon PL, Webster NJ. Pulse sensitivity of the luteinizing hormone b comes of age. Nat Rev Immunol 2010; 10: 645–656.
promoter is determined by a negative feedback loop involving early 60 Holdstock JG, Aylwin SJB, Burrin JM. Calcium and glycoprotein hor-
growth response-1 and Ngfi-A binding protein 1 and 2. Mol Endocrinol mone a-subunit gene expression and secretion in aT3-1 gonadotropes.
2007; 21: 1175–1191. Mol Endocrinol 1996; 10: 1308–1317.
42 Ciccone NA, Xu S, Lacza CT, Carroll RS, Kaiser UB. Frequency-dependent 61 Weck J, Anderson AC, Jenkins S, Fallest PC, Shupnik MA. Divergent and
regulation of follicle-stimulating hormone b by pulsatile gonadotropin- composite gonadotropin-releasing hormone-responsive elements in the
releasing hormone is mediated by functional antagonism of bZIP tran- rat luteinizing hormone subunit genes. Mol Endcrinol 2000; 14: 472–485.
scription factors. Mol Cell Biol 2010; 30: 1028–1040. 62 Binder AK, Grammer JC, Herndon MK, Stanton JD, Nilson JH. GnRH
43 Jorgensen JS, Quirk CC, Nilson JH. Multiple and overlapping combina- regulation of Jun and Atf3 requires calcium, calcineurin, and NFAT.
torial codes orchestrate hormonal responsiveness and dictate cell-spe- Mol Endocrinol 2012; 26: 873–886.
cific expression of the genes encoding luteinizing hormone. Endocr Rev 63 Kakar SS, Winters SJ, Zacharias W, Miller DM, Flynn S. Identification of
2004; 25: 521–542. distinct gene expression profiles associated with treatment of LbT2
44 Melamed P. Hormonal signaling to FSHb gene expression. Mol Cell cells with gonadotropin-releasing hormone agonist using microarray
Endocrinol 2010; 314: 204–212. analysis. Gene 2003; 308: 67–77.
45 Ely HA, Mellon PL, Coss D. GnRH Induces the c-fos gene via phosphor- 64 Youn HD, Chatila TA, Liu JO. Integration of calcineurin and MEF2 sig-
ylation of SRF by the calcium/calmodulin kinase II pathway. Mol Endo- nals by the coactivator p300 during T-cell apoptosis. EMBO J 2000;
crinol 2011; 25: 669–680. 19: 4323–4331.
46 Mistry DS, Tsutsumi R, Fernandez M, Sharma S, Cardenas SA, Lawson 65 Li Y, Lau LF. Adrenocorticotropic hormone regulates the activities of
MA, Webster NJG. Gonadotropin-releasing hormone pulse sensitivity of the orphan nuclear receptor Nur77 through modulation of phosphory-
follicle-stimulating hormone-b gene is mediated by differential expres- lation. Endocrinology 1997; 138: 4138–4146.
sion of positive regulatory activator protein 1 factors and corepressors 66 Sohn YC, Kwak E, Na YJ, Lee JW, Lee SK. Silencing mediator of retinoid
SKIL and TGIF1. Mol Endocrinol 2011; 25: 1387–1403. and thyroid hormone receptors and activating signal cointegrator-2 as
47 Crivici A, Ikura M. Molecular and structural basis of target recognition transcriptional coregulators of the orphan nuclear receptor Nur77.
by calmodulin. Annu Rev Biophys Biomol Struct 1995; 24: 85–116. J Biol Chem 2001; 276: 43734–43739.
48 Shen XR, Li HM, Ou Y, Tao WB, Dong A, Kong JL, Ji CN, Yu SN. The 67 Kelly SN, McKenna TJ, Young LS. Coregulatory protein-orphan nuclear
secondary structure of calcineurin regulatory region and conforma- receptor interactions in the human adrenal cortex. J Endocrinol 2005;
tional change induced by calcium/calmodulin binding. J Biol Chem 186: 33–42.
2008; 283: 11407–11413. 68 Amelio AL, Miraglia LJ, Conkright JJ, Mercer BA, Batalov S, Cavett V,
49 Luo ZJ, Wijeweera A, Oh YZ, Liou YC, Melamed P. Pin1 regulates gona- Orth AP, Busby J, Hogenesch JB, Conkright MD. A coactivator trap
dotropin b-subunit gene transcription through facilitating the phos- identifies NONO (p54nrb) as a component of the cAMP-signaling path-
phorylation-dependent ubiquitination of SF-1. Mol Cell Biol 2010; 30: way. Proc Natl Acad Sci USA 2007; 104: 20314–20319.
745–763. 69 Canettieri G, Coni S, Della Guardia M, Nocerino V, Antonucci L,
50 Pnueli L, Luo M, Wang S, Naor Z, Melamed P. Calcineurin regulates Di Magno L, Screaton R, Screpanti I, Giannini G, Gulino A. The coacti-
expression of both subunits of the follicle-stimulating hormone vator CRTC1 promotes cell proliferation and transformation via AP-1.
through distinct mechanisms. Mol Cell Biol 2011; 31: 5023–5036. Proc Natl Acad Sci USA 2009; 106: 1445–1450.
51 Zhang T, Mulvaney JM, Roberson MS. Activation of mitogen-activated 70 Conkright MD, Canettieri G, Screaton R, Guzman E, Miraglia L, Hoge-
protein kinase phosphatase 2 by gonadotropin-releasing hormone. Mol nesch JB, Montminy M. TORCs: transducers of regulated CREB activity.
Cell Endocrinol 2001; 172: 79–89. Mol Cell 2003; 12: 413–423.
52 Wurmbach E, Yuen T, Ebersole BJ, Sealfon SC. Gonadotropin-releasing 71 Kasper LH, Lerach S, Wang JM, Wu S, Jeevan T, Brindle PK. CBP/p300
hormone receptor-coupled gene network organization. J Biol Chem double null cells reveal effect of coactivator level and diversity on
2001; 276: 47195–47201. CREB transactivation. EMBO J 2010; 29: 3660–3672.

Journal of Neuroendocrinology, 2012, 24, 1463–1475 © 2012 The Authors.


Journal of Neuroendocrinology © 2012 British Society for Neuroendocrinology
1474 P. Melamed et al.

72 Screaton RA, Conkright MD, Katoh Y, Best JL, Canettieri G, Jeffries S, 89 Liou YC, Ryo A, Huang HK, Lu PJ, Bronson R, Fujimori F, Uchida T,
Guzman E, Niessen S, Yates JR, Takemori H, Okamoto M, Montminy M. Hunter T, Lu KP. Loss of Pin1 function in the mouse causes phenotypes
The CREB coactivator TORC2 functions as a calcium- and cAMP-sensi- resembling cyclin D1-null phenotypes. Proc Natl Acad Sci USA 2002;
tive coincidence detector. Cell 2004; 119: 61–74. 99: 1335–1340.
73 Takemori H, Okamoto M. Regulation of CREB-mediated gene expression 90 Wulf GM, Ryo A, Wulf GG, Lee SW, Niu T, Petkova V, Lu KP. Pin1 is
by salt inducible kinase. J Steroid Biochem Mol Biol 2008; 108: 287– overexpressed in breast cancer and cooperates with Ras signaling in
291. increasing the transcriptional activity of c-Jun towards cyclin D1.
74 Ciccone NA, Lacza CT, Hou MY, Gregory SJ, Kam KY, Xu S, Kaiser UB. A EMBO J 2001; 20: 3459–3472.
composite element that binds basic helix loop helix and basic leucine 91 Salisbury TB, Binder AK, Grammer JC, Nilson JH. GnRH-regulated
zipper transcription factors is important for gonadotropin-releasing expression of Jun and JUN target genes in gonadotropes requires a
hormone regulation of the follicle stimulating hormone b gene. Mol functional interaction between TCF/LEF family members and b-catenin.
Endocrinol 2008; 22: 1908–1923. Mol Endocrinol 2009; 23: 402–411.
75 Breuillaud L, Halfon O, Magistrett PJ, Pralong FP, Cardinaux JR. Mouse 92 Park JE, Lee JA, Park SG, Lee DH, Kim SJ, Kim HJ, Uchida C, Uchida T,
fertility is not dependent on the CREB coactivator Crtc1. Nature Med Park BC, Cho S. A critical step for JNK activation: isomerization by the
2009; 15: 989–990. prolyl isomerase Pin1. Cell Death Differ 2012; 19: 153–161.
76 Eto A, Akita Y, Saido TC, Suzuki K, Kawashima S. The role of the cal- 93 Gardner S, Maudsley S, Millar RP, Pawson AJ. Nuclear stabilization of
pain-calpstatin system in thyrotropin releasing hormone-induced selec- b-catenin and inactivation of glycogen synthase kinase-3b by gonado-
tive down-regulation of a protein kinase-C isozyme, nPKC-epsilon, in tropin-releasing hormone: targeting Wnt signaling in the pituitary go-
rat pituitary GH(4)C(1) cells. J Biol Chem 1995; 270: 25115–25120. nadotrope. Mol Endocrinol 2007; 21: 3028–3038.
77 Lu KP, Zhou XZ. The prolyl isomerase PIN1: a pivotal new twist in 94 Santella L, Ercolano E, Nusco GA. The cell cycle: a new entry in the
phosphorylation signalling and disease. Nat Rev Mol Cell Biol 2007; 8: field of Ca2+ signaling. Cell Mol Life Sci 2005; 62: 2405–2413.
904–916. 95 Santini MP, Talora C, Seki T, Bolgan L, Dotto GP. Cross talk among cal-
78 Liou YC, Zhou XZ, Lu KP. Prolyl isomerase Pin 1 as a molecular switch cineurin, Sp1/Sp3, and NFAT in control of p21(WAF1/CIP1) expression
to determine the fate of phosphoproteins. Trends Biochem Sci 2011; in keratinocyte differentiation. Proc Natl Acad Sci USA 2001; 98: 9575
36: 501–514. –9580.
79 Lu PJ, Zhou XZ, Liou YC, Noel JP, Lu KP. Critical role of WW domain 96 Gafter-Gvili A, Sredni B, Gal R, Gafter U, Kalechman Y. Cyclosporin A-
phosphorylation in regulating phosphoserine binding activity and Pin1 induced hair growth in mice is associated with inhibition of calcineu-
function. J Biol Chem 2002; 277: 2381–2384. rin-dependent activation of NFAT in follicular keratinocytes. Am J
80 Tremblay JJ, Drouin J. Egr-1 is a downstream effector of GnRH and Physiol-Cell Biol 2003; 284: C1593–C1603.
synergizes by direct interaction with Ptx1 and SF-1 to enhance lutein- 97 Kraus S, Levy G, Hanoch T, Naor Z, Seger R. Gonadotropin-releasing
izing hormone b gene transcription. Mol Cell Biol 1999; 19: 2567– hormone induces apoptosis of prostate cancer cells: role of c-Jun
2576. NH2-terminal kinase, protein kinase B, and extracellular signal-regu-
81 Liu WF, Youn HD, Zhou XZ, Lu KP, Liu JO. Binding and regulation of lated kinase pathways. Cancer Res 2004; 64: 5736–5744.
the transcription factor NFAT by the peptidyl prolyl cis-trans isomerase 98 Maiti K, Oh DY, Moon JS, Acharjee S, Li JH, Bai DG, Park HS, Lee K, Lee
Pin1. FEBS Let 2001; 496: 105–108. YC, Jung NC, Kim K, Vaudry H, Kwon HB, Seong JY. Differential effects
82 Melamed P, Zhu Y, Tan SH, Xie M, Koh M. Gonadotropin releasing hor- of gonadotropin-releasing hormone (GnRH)-I and GnRH-II on prostate
mone activation of c-jun, but not Egr-1, stimulates transcription of a cancer cell signaling and death. J Clin Endocrinol Metab 2005; 90:
luteinizing hormone b subunit gene. Endocrinology 2006; 147: 3598– 4287–4298.
3605. 99 White CD, Stewart AJ, Lu ZL, Millar RP, Morgan K. Antiproliferative
83 Nakatsu Y, Sakoda H, Kushiyama A, Ono H, Fujishiro M, Horike N, Yon- effects of GnRH agonists: prospects and problems for cancer therapy.
eda M, Ohno H, Tsuchiya Y, Kamata H, Tahara H, Isobe T, Nishimura F, Neuroendocrinology 2008; 88: 67–79.
Katagiri H, Oka Y, Fukushima T, Takahashi SI, Kurihara H, Uchida T, 100 Wu HM, Cheng JC, Wang HS, Huang HY, MacCalman CD, Leung PC.
Asanoa T. Pin1 associates with and induces translocation of CRTC2 to Gonadotropin-releasing hormone type II induces apoptosis of human
the cytosol, thereby suppressing cAMP-responsive element transcrip- endometrial cancer cells by activating GADD45a. Cancer Res 2009; 69:
tional activity. J Biol Chem 2010; 285: 33018–33027. 4202–4208.
84 Rajbhandari P, Finn G, Solodin NM, Singarapu KK, Sahu SC, Markley JL, 101 Mazhawidza W, Winters SJ, Kaiser UB, Kakar SS. Identification.of gene
Kadunc KJ, Ellison-Zelski SJ, Kariagina A, Haslam SZ, Lu KP, Alarid ET. networks modulated by activin in LbT2 cells using DNA microarray
Regulation of estrogen receptor a N-terminus conformation and func- analysis. Histol Histopathol 2006; 21: 167–178.
tion by peptidyl prolyl isomerase. Mol Cell Biol 2012; 32: 445–457. 102 Sakai T, Inoue K, Hasegawa Y, Kurosumi K. Effect of passive immuniza-
85 Baksh S, DeCaprio JA, Burakoff SJ. Calcineurin regulation of the mam- tion to gonadotropin-releasing hormone (GnRH) using GnRH antiserum
malian G0/G1 checkpoint element, cyclin dependent kinase 4. Onco- on the mitotic activity of gonadotrophs in castrated male rats. Endo-
gene 2000; 19: 2820–2827. crinology 1988; 122: 2803–2808.
86 Baksh S, Widlund HR, Frazer-Abel AA, Du J, Fosmire S, Fisher DE, DeCa- 103 Childs GV, Unabia G. Epidermal growth factor and gonadotropin-
prio JA, Modiano JF, Burakoff SJ. NFATc2-mediated repression of cy- releasing hormone stimulate proliferation of enriched population of
clin-dependent kinase 4 expression. Mol Cell 2002; 10: 1071–1081. gonadotropes. Endocrinology 2001; 142: 847–853.
87 Zacchi P, Gostissa M, Uchida T, Salvagno C, Avolio F, Volinia S, Ronai 104 Lewy H, Ashkenazi IE, Naor Z. Gonadotropin releasing hormone (GnRH)
Z, Blandino G, Schneider C, Del Sal G. The prolyl isomerase Pin1 reveals and estradiol (E(2)) regulation of cell cycle in gonadotrophs. Mol Cell
a mechanism to control p53 functions after genotoxic insults. Nature Endocrinol 2003; 203: 25–32.
2002; 419: 853–857. 105 Mattson MP, Chan SL. Calcium orchestrates apoptosis. Nat Cell Biol
88 Zheng H, You H, Zhou XZ, Murray SA, Uchida T, Wulf G, Gu L, Tang X, 2003; 5: 1041–1043.
Lu KP, Xiao ZX. The prolyl isomerase Pin1 is a regulator of p53 in 106 Orrenius S, Zhivotovsky B, Nicotera P. Regulation of cell death: the cal-
genotoxic response. Nature 2002; 419: 849–853. cium-apoptosis link. Nat Rev Mol Cell Biol 2003; 4: 552–565.

© 2012 The Authors. Journal of Neuroendocrinology, 2012, 24, 1463–1475


Journal of Neuroendocrinology © 2012 British Society for Neuroendocrinology
GnRH signalling downstream of calmodulin 1475

107 Clapham DE. Calcium signaling. Cell 2007; 131: 1047–1058. 122 Nini L, Waheed AA, Panicker LM, Czapiga M, Zhang JH, Simonds WF.
108 Zhivotovsky B, Orrenius S. Calcium and cell death mechanisms: a per- R7-binding protein targets the G protein b 5/R7-regulator of G protein
spective from the cell death community. Cell Calcium 2011; 50: 211– signaling complex to lipid rafts in neuronal cells and brain. BMC Bio-
221. chem 2007; 8: 18.
109 Cheung LW, Wong AS. Gonadotropin-releasing hormone: GnRH recep- 123 Grabowska D, Jayaraman M, Kaltenbronn KM, Sandiford SL, Wang Q,
tor signaling in extrapituitary tissues. FEBS J 2008; 275: 5479–5495. Jenkins S, Slepak VZ, Smith Y, Blumer KJ. Postnatal induction and
110 Godoy J, Nishimura M, Webster NJ. Gonadotropin-releasing hormone localization of R7BP, a membrane-anchoring protein for regulator of G
induces miR-132 and miR-212 to regulate cellular morphology and protein signaling 7 family-Gb5 complexes in brain. Neuroscience 2008;
migration in immortalized LbT2 pituitary gonadotrope cells. Mol Endo- 151: 969–982.
crinol 2011; 25: 810–820. 124 Bliss SP, Navratil AM, Breed M, Skinner DC, Clay CM, Roberson MS.
111 Ward Y, Kelly K. Gem protein signaling and regulation. Methods Enzy- Signaling complexes associated with the type I gonadotropin-releasing
mol 2006; 407: 468–483. hormone (GnRH) receptor: colocalization of extracellularly regulated
112 Correll RN, Pang C, Niedowicz DM, Finlin BS, Andres DA. The RGK fam- kinase 2 and GnRH receptor within membrane rafts. Mol Endocrinol
ily of GTP-binding proteins: regulators of voltage-dependent calcium 2007; 21: 538–549.
channels and cytoskeleton remodeling. Cell Signal 2008; 20: 292–300. 125 Maudsley S, Naor Z, Bonfil D, Davidson L, Karali D, Pawson AJ, Larder
113 Béguin P, Nagashima K, Gonoi T, Shibasaki T, Takahashi K, Kashima Y, R, Pope C, Nelson N, Millar RP, Brown P. Proline-rich tyrosine kinase 2
Ozaki N, Geering K, Iwanaga T, Seino S. Regulation of Ca2+ channel mediates gonadotropin-releasing hormone signaling to a specific extra-
expression at the cell surface by the small G-protein kir/Gem. Nature cellularly regulated kinase-sensitive transcriptional locus in the lutein-
2001; 411: 701–706. izing hormone b-subunit gene. Mol Endocrinol 2007; 21: 1216–1233.
114 Mahalakshmi RN, Nagashima K, Ng MY, Inagaki N, Hunziker W, Béguin 126 Xie JJ, Allen KH, Marguet A, Berghorn KA, Bliss SP, Navratil AM, Guan
P. Nuclear transport of Kir/Gem requires specific signals and importin JL, Roberson MS. Analysis of the calcium-dependent regulation of pro-
a5 and is regulated by calmodulin and predicted serine phosphoryla- line-rich tyrosine kinase 2 by gonadotropin-releasing hormone. Mol
tions. Traffic 2007; 8: 1150–1163. Endocrinol 2008; 22: 2322–2335.
115 Béguin P, Mahalakshmi RN, Nagashima K, Cher DH, Takahashi A, Ya- 127 Dobkin-Bekman M, Naidich M, Rahamim L, Przedecki F, Almog T, Lim S,
mada Y, Seino Y, Hunziker W. 14-3-3 and calmodulin control subcellu- Melamed P, Liu P, Wohland T, Yao Z, Seger R, Naor Z. A preformed sig-
lar distribution of Kir/Gem and its regulation of cell shape and calcium naling complex mediates GnRH activated ERK phosphorylation of paxil-
channel activity. J Cell Sci 2005; 118: 1923–1934. lin and FAK at focal adhesions in LbT2 gonadotrope cells. Mol
116 Karakoula A, Tovey SC, Brighton PJ, Willars GB. Lack of receptor-selec- Endocrinol 2009; 23: 1850–1864.
tive effects of either RGS2, RGS3 or RGS4 on muscarinic M3- and gon- 128 Davidson L, Pawson AJ, Millar RP, Maudsley S. Cytoskeletal reorganiza-
adotropin-releasing hormone receptor-mediated signalling through tion dependence of signaling by the gonadotropin releasing hormone
Gaq/11. Eur J Pharmacol 2008; 587: 16–24. receptor. J Biol Chem 2004; 279: 1980–1993.
117 Fernandez MO, Webster NJG. Regulator of G protein signaling 2 (RGS2) 129 Shen X, Valencia CA, Szostak JW, Dong B, Liu R. Scanning the human
is induced by pulsatile GnRH and alters GnRH-induced gene expression proteome for calmodulin-binding proteins. Proc Natl Acad Sci USA
in LbT2 Cells. Endocr Rev 2010; 31: S1924. 2005; 102: 5969–5974.
118 Willars GB. Mammalian RGS proteins: multifunctional regulators of cel- 130 Berggård T, Arrigoni G, Olsson O, Fex M, Linse S, James P. 140 mouse
lular signalling. Semin Cell Dev Biol 2006; 17: 363–376. brain proteins identified by Ca2+-calmodulin affinity chromatography
119 Cunningham ML, Waldo GL, Hollinger S, Hepler JR, Harden TK. Protein and tandem mass spectrometry. J Proteome Res 2006; 5: 669–687.
kinase C phosphorylates RGS2 and modulates its capacity for negative 131 Saucerman JJ, Bers DM. Calmodulin binding proteins provide domains
regulation of G a(11) signaling. J Biol Chem 2001; 276: 5438–5444. of local Ca(2+) signaling in cardiac myocytes. J Mol Cell Cardiol 2012;
120 Ishii M, Inanobe A, Kurachi Y. PIP3 inhibition of RGS protein and its 52: 312–316.
reversal by Ca2+/calmodulin mediate voltage-dependent control of the 132 Mermelstein PG, Deisseroth K, Dasgupta N, Isaksen AL, Tsien RW. Cal-
G protein cycle in a cardiac K+ channel. Proc Natl Acad Sci USA 2002; modulin priming: nuclear translocation of a calmodulin complex and
99: 4325–4330. the memory of prior neuronal activity. Proc Natl Acad Sci USA 2001;
121 Ishii M, Ikushima M, Kurachi Y. In vivo interaction between RGS4 and 98: 15342–15347.
calmodulin visualized with FRET techniques: possible involvement of 133 Shao S, Hegde RS. A calmodulin-dependent translocation pathway for
lipid raft. Biochem Biophys Res Commun 2005; 338: 839–846. small secretory proteins. Cell 2011; 147: 1576–1588.

Journal of Neuroendocrinology, 2012, 24, 1463–1475 © 2012 The Authors.


Journal of Neuroendocrinology © 2012 British Society for Neuroendocrinology

You might also like