g-protein-coupled receptor signaling and the egf network in endocrine systems

7
G-protein-coupled receptor signaling and the EGF network in endocrine systems Minnie Hsieh and Marco Conti Division of Reproductive Biology, Department of Obstetrics and Gynecology, Stanford University, Stanford, CA 94305-5317, USA The epidermal growth factor (EGF) network is com- posed of a complex array of growth factors synthesized as precursors and expressed on the cell surface. These latent growth factors are activated by cleavage and shedding from the cell surface and act by binding to various homo- and hetero-dimers of the EGF receptors (ErbBs). Although the exact molecular steps are poorly understood, ligand binding to G-protein-coupled receptors as diverse as the b-adrenoceptors or the lysophosphatidic acid receptors leads to shedding of EGF growth factors and activation of EGF receptors. Recent observations from the pituitary and in the ovary are providing new insight into the role of this network in endocrine systems. Introduction Given the array of extracellular cues to which they are exposed, cells have developed complex machinery for the reception and interpretation of external stimuli. Multiple intracellular signaling pathways are required to receive these signals and translate them into changes in cellular functions. A common theme in the arrangement of these pathways is the integration and crosstalk between contiguous cascades to fine tune cellular functions as diverse as cell proliferation, differentiation and migration. The transactivation of receptor tyrosine kinases by G-protein-coupled receptors (GPCRs) is one example of communication and cooperation between different signal- ing networks. In a variety of cellular contexts, agonist binding to GPCRs results in transactivation of the epidermal growth factor (EGF) receptor (EGFR; also known as ErbB1 or HER) and activation of the extra- cellular signal-regulated kinase/mitogen-activated pro- tein kinase (ERK/MAPK) cascade. Traditionally, the EGF network has been viewed as a major orchestrator of cell replication under physiological and pathological conditions. However, this signaling network has much broader functions in the body, for instance in early development and in organogenesis, and in tissue homeo- stasis in the adult. Here, we review evidence supporting involvement of the EGF network in the regulation of endocrine and reproductive processes. These studies, together with those examining GPCR-mediated EGFR transactivation, provide new insight into the mechanism by which signals arising from GPCRs can be transduced via the EGF network to affect crucial endocrine functions. GPCR signaling to the ERK/MAPK pathway GPCRs are the largest family of cell surface receptors, with more than 1000 genes encoding this class of proteins. They are characterized by a conserved structural motif of seven transmembrane-spanning regions, and undergo agonist-induced conformational changes that expose intra- cellular binding sites for the heterotrimeric GTP-binding proteins (G proteins) (reviewed in [1]). Interaction of GPCRs with G proteins leads to the exchange of GDP for GTP in a binding pocket of the Ga subunit, followed by dissociation of G-protein complexes into activated a subunits and bg dimers. These, in turn, act on effector molecules to promote second messenger-generating sig- naling cascades, including the cAMP-, inositol trisphos- phate- and Ca 2C -dependent pathways. Signaling through these pathways usually culminates in the activation of kinases and changes in protein phosphorylation. Some of these kinases are directly regulated by second mes- sengers, but many constitute modules further down- stream of the signaling cascade, such as the RAF–MAPK kinase–ERK (RAF–MEK–ERK) module. The mechanism by which GPCRs activate the ERK/ MAPK cascade is complex and often cell specific. Some of the biochemical steps linking GPCRs to MAPK activation include the kinases protein kinase A (PKA) [2,3], protein kinase C (PKC) [4] or Src/Pyk2 [5,6], and might involve regulation of small G proteins and guanine nucleotide exchange factors [(e.g. EPAC (exchange protein directly activated by cAMP)] [7,8]. In other instances, more elabor- ate circuits are involved in activation of this signaling module. Daub et al. [9] were the first to demonstrate that the GPCR ligands lysophosphatidic acid (LPA), endothelin-1 and thrombin induced rapid tyrosine phosphorylation of the EGFR and ErbB2 in Rat-1 fibroblasts, and that the specific EGFR inhibitor tyrphostin AG1478 or a dominant- negative EGFR mutant abrogated GPCR-induced ERK/ MAPK activation. Further studies confirmed that many GPCRs utilize EGFR transactivation to couple to the ERK/MAPK cascade in diverse cell types (reviewed in [4,10]). For example, it has been proposed that activation of the b 2 -adrenoceptor in COS7 cells induces receptor complex Corresponding author: Conti, M. ([email protected]). Available online 28 July 2005 Review TRENDS in Endocrinology and Metabolism Vol.16 No.7 September 2005 www.sciencedirect.com 1043-2760/$ - see front matter Q 2005 Elsevier Ltd. All rights reserved. doi:10.1016/j.tem.2005.07.005

Upload: minnie-hsieh

Post on 29-Oct-2016

214 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: G-protein-coupled receptor signaling and the EGF network in endocrine systems

G-protein-coupled receptor signalingand the EGF network in endocrinesystemsMinnie Hsieh and Marco Conti

Division of Reproductive Biology, Department of Obstetrics and Gynecology, Stanford University, Stanford, CA 94305-5317, USA

The epidermal growth factor (EGF) network is com-

posed of a complex array of growth factors synthesized

as precursors and expressed on the cell surface. These

latent growth factors are activated by cleavage and

shedding from the cell surface and act by binding to

various homo- and hetero-dimers of the EGF receptors

(ErbBs). Although the exact molecular steps are poorly

understood, ligand binding to G-protein-coupled

receptors as diverse as the b-adrenoceptors or the

lysophosphatidic acid receptors leads to shedding of

EGF growth factors and activation of EGF receptors.

Recent observations from the pituitary and in the ovary

are providing new insight into the role of this network in

endocrine systems.

Introduction

Given the array of extracellular cues to which they areexposed, cells have developed complex machinery for thereception and interpretation of external stimuli. Multipleintracellular signaling pathways are required to receivethese signals and translate them into changes in cellularfunctions. A common theme in the arrangement of thesepathways is the integration and crosstalk betweencontiguous cascades to fine tune cellular functions asdiverse as cell proliferation, differentiation and migration.The transactivation of receptor tyrosine kinases byG-protein-coupled receptors (GPCRs) is one example ofcommunication and cooperation between different signal-ing networks. In a variety of cellular contexts, agonistbinding to GPCRs results in transactivation of theepidermal growth factor (EGF) receptor (EGFR; alsoknown as ErbB1 or HER) and activation of the extra-cellular signal-regulated kinase/mitogen-activated pro-tein kinase (ERK/MAPK) cascade. Traditionally, theEGF network has been viewed as a major orchestrator ofcell replication under physiological and pathologicalconditions. However, this signaling network has muchbroader functions in the body, for instance in earlydevelopment and in organogenesis, and in tissue homeo-stasis in the adult. Here, we review evidence supportinginvolvement of the EGF network in the regulation ofendocrine and reproductive processes. These studies,together with those examining GPCR-mediated EGFR

Corresponding author: Conti, M. ([email protected]).Available online 28 July 2005

www.sciencedirect.com 1043-2760/$ - see front matter Q 2005 Elsevier Ltd. All rights reserved

transactivation, provide new insight into the mechanismby which signals arising from GPCRs can be transducedvia the EGF network to affect crucial endocrine functions.

GPCR signaling to the ERK/MAPK pathway

GPCRs are the largest family of cell surface receptors,with more than 1000 genes encoding this class of proteins.They are characterized by a conserved structural motif ofseven transmembrane-spanning regions, and undergoagonist-induced conformational changes that expose intra-cellular binding sites for the heterotrimeric GTP-bindingproteins (G proteins) (reviewed in [1]). Interaction ofGPCRs with G proteins leads to the exchange of GDPfor GTP in a binding pocket of the Ga subunit, followedby dissociation of G-protein complexes into activateda subunits and bg dimers. These, in turn, act on effectormolecules to promote second messenger-generating sig-naling cascades, including the cAMP-, inositol trisphos-phate- and Ca2C-dependent pathways. Signaling throughthese pathways usually culminates in the activation ofkinases and changes in protein phosphorylation. Some ofthese kinases are directly regulated by second mes-sengers, but many constitute modules further down-stream of the signaling cascade, such as the RAF–MAPKkinase–ERK (RAF–MEK–ERK) module.

The mechanism by which GPCRs activate the ERK/MAPK cascade is complex and often cell specific. Some ofthe biochemical steps linking GPCRs to MAPK activationinclude the kinases protein kinase A (PKA) [2,3], proteinkinase C (PKC) [4] or Src/Pyk2 [5,6], and might involveregulation of small G proteins and guanine nucleotideexchange factors [(e.g. EPAC (exchange protein directlyactivated by cAMP)] [7,8]. In other instances, more elabor-ate circuits are involved in activation of this signalingmodule. Daub et al. [9] were the first to demonstrate thattheGPCRligands lysophosphatidicacid (LPA), endothelin-1and thrombin induced rapid tyrosine phosphorylation ofthe EGFR and ErbB2 in Rat-1 fibroblasts, and that thespecific EGFR inhibitor tyrphostin AG1478 or a dominant-negative EGFR mutant abrogated GPCR-induced ERK/MAPK activation.

Further studies confirmed that many GPCRs utilizeEGFR transactivation to couple to the ERK/MAPKcascade in diverse cell types (reviewed in [4,10]). Forexample, it has been proposed that activation of theb2-adrenoceptor in COS7 cells induces receptor complex

Review TRENDS in Endocrinology and Metabolism Vol.16 No.7 September 2005

. doi:10.1016/j.tem.2005.07.005

Page 2: G-protein-coupled receptor signaling and the EGF network in endocrine systems

Review TRENDS in Endocrinology and Metabolism Vol.16 No.7 September 2005 321

formation with the EGFR, with subsequent EGFRtransactivation and MAPK activation [11]. Stimulationof head and neck squamous cell carcinoma (HNSCC) cellsby the GPCR agonists LPA, carbachol, bradykinin orthrombin causes the rapid tyrosine phosphorylation ofEGFR, illustrating the convergence of different activatingsignals on a common pathway [12]. As in other cells, EGFRtyrosine kinase activity was required for LPA-inducedMAPK activation, and inhibition of EGFR signalingabrogated LPA-stimulated proliferation and migration ofHNSCC cells. The pathophysiological relevance of thisregulation is underscored by the finding that activation ofrat neonatal cardiomyocytes by phenylephrine, angio-tensin II and endothelin-1, known inducers of cardiachypertrophy, resulted in enhanced EGFR tyrosine phos-phorylation [13]. The discovery that GPCR-inducedcardiac hypertrophy was attenuated by a specific inhibitorof ADAM12 (a disintegrin and metalloprotease 12), whichis involved in processing of one of the EGFR ligands(discussed below), further established the requirementfor crosstalk in mediating pathophysiological events.Additional GPCR ligands shown to mediate EGFR trans-activation and stimulation of the ERK/MAPK pathwayinclude oxytocin [14] and prostaglandin F2a [15,16],raising the possibility that this circuit operates inreproductive and endocrine systems.

EGF signaling network

The ErbB receptor family comprises four members: EGFR(ErbB1, HER), ErbB2 (Neu, HER2), ErbB3 (HER3) andErbB4 (HER4) [17,18]. These receptors have in common acysteine-rich extracellular domain, a single membrane-spanning region and a cytoplasmic domain containing atyrosine kinase domain and tyrosine autophosphorylationsites. Ligand binding to specific ErbB receptors promotesreceptor homo- or heterodimerization, activation of theintrinsic tyrosine kinase, tyrosine phosphorylation ofseveral substrates and the ErbB receptor itself, andstimulation of downstream signaling cascades [19].

ErbB receptors are activated by members of theEGF-like growth factor family. Each EGF-like growthfactor contains an EGF-like motif characterized by sixcysteine residues that form three intramolecular disul-fide-bonded loops, and that confer binding specificity forthe ErbB receptors [17,18]. Group 1 ligands bind speci-fically to EGFR, and consist of EGF, transforming growthfactor a (TGF-a) and amphiregulin. The neuregulins thatcomprise group 2 bind ErbB3 and ErbB4. Group 3members include heparin-binding EGF-like factor(HB-EGF), betacellulin and epiregulin, and bind bothEGFR and ErbB4. Although ErbB2 has no known ligand,it heterodimerizes with other ErbB receptors to form apotent signaling complex. ErbB3 that has impaired kinaseactivity only becomes phosphorylated and forms an activereceptor complex when partnered with another ErbBreceptor.

Although not fully understood, the presence of multiplereceptors activated by a ligand, in addition to thepossibility of homo- and heterodimerization, undoubtedlyhas important implications for generating differentsignals and different outcomes. The specific ligand–

www.sciencedirect.com

receptor complex formed determines which tyrosineresidues are autophosphorylated, the cytoplasmicmediators that are engaged and hence which pathwaysare activated. Pathways activated include the Ras- andShc-activated MAPK pathway that is downstream of allErbB receptors, and the phosphatidyl inositol 3-kinasepathway that is directly or indirectly activated by mostErbB dimers [17,18]. Activation of intracellular signalingcascades culminates in events such as cell proliferation,differentiation, migration and survival, which are import-ant for pathological processes such as inflammation,tissue remodeling and wound healing. Some of theseprocesses bear striking similarities to the changes thatoccur in the ovarian follicle during ovulation.

EGF-like growth factor processing

The EGF-like growth factors are synthesized as trans-membrane precursors that can be released or ‘shed’ assoluble mature peptides by proteolytic cleavage of theectodomains at the membrane surface [20]. In particular,members of the ADAM family of proteases are thought tobe the major regulators of EGF-like growth factorshedding from the cell surface [21,22]. Tumor necrosisfactor a-converting enzyme (TACE/ADAM17) has beenimplicated in the shedding of amphiregulin, HB-EGF andTGF-a in several different cell lines [21–25], and inphorbol myristate acetate-induced shedding of epiregulinin mouse embryonic fibroblasts [21,23]. Consistent withthis view, TACE-deficient mice exhibit phenotypes similarto TGF-a-, HB-EGF- and EGFR-deficient mice [25–30]. Bycontrast, EGF shedding appears to be TACE independent[31], whereas ADAM10 but not TACE appears to be themajor convertase for betacellulin [21,23]. However, morethan one ADAMmight regulate proteolytic processing of aspecific EGFR ligand. For example, in addition to TACE,ADAM-9, -10 and -12 have been implicated in HB-EGFprocessing [13,24,32–34].

Although the unprocessed pro-forms of EGF-likegrowth factors have been shown to have biological activity[35–37], several lines of evidence suggest a crucial role forgrowth factor shedding in the activation of EGFRsignaling. For example, normal mouse development isdependent on soluble TGF-a rather than on the integralmembrane form, and requires functional TACE; therelease of soluble TGF-a was severely reduced inembryonic fibroblasts from TACE-deficient mice thatpresented with eye, hair and skin defects, abnormalitiesalso found in TGFa-deficient mice [25]. In HNSCC cells,cleavage of pro-amphiregulin at the cell surface to releasemature amphiregulin was required for GPCR-inducedEGFR activation and stimulation of cell proliferation andmigration [38].

Ligand-dependent EGFR transactivation by GPCRs

A model for the mechanism of GPCR-induced EGFRtransactivation has emerged and is referred to as the‘triple-membrane-passing-signaling’ pathway (Figure 1)[4,39]. In short, agonist binding to GPCRs leads tointracellular stimulation of specific metalloproteinases,cleavage and release of EGFR ligands at the cell surface,ligand binding and activation of the EGFR and

Page 3: G-protein-coupled receptor signaling and the EGF network in endocrine systems

TRENDS in Endocrinology & Metabolism

RasRafMEK

?

PP

PPα

β γ

LigandPro-

ligand

GPCR(active)

MMP orADAMs

EGF-likefactor

Soluble

EGFR(active)

ERK/MAPK

Second-messengercascades

(a)

(b)

(c)

Figure 1. Model of EGFR transactivation. After GPCR stimulation, (a) undefined

intracellular signals activate specific transmembrane-bound metalloproteinases,

leading to (b) proteolytic cleavage of the ectodomain of EGF-like ligands at

the cell surface. The released or ‘shed’ growth factors activate the EGFR and

(c) subsequently the ERK/MAPK cascade. Because signals cross the membrane

three times, this pathway is referred to as the ‘triple-membrane-passing-signaling’

mechanism [4,39].

Review TRENDS in Endocrinology and Metabolism Vol.16 No.7 September 2005322

subsequent stimulation of the ERK/MAPK cascade. Thespecific metalloproteinase(s) involved in ligand sheddingis cell-type dependent. For example, in HNSCC cells,inhibition of TACE impaired the ability of LPA- orcarbachol-stimulated GPCRs to induce amphiregulinrelease and EGFR activation, demonstrating a functionalrole for TACE in mediating this event [38]. TACE is alsoimplicated in angiotensin II-stimulated HB-EGF shed-ding, EGFR activation and cell proliferation in ACHNkidney tumor cells [40]. However, in TccSup bladdercarcinoma cells, ADAM15 and not TACE mediatedLPA-induced EGFR transactivation that also involvedboth amphiregulin and TGF-a [40]. In other cells, matrixmetalloproteinase 2 (MMP2) or MMP9 might be respon-sible for the GPCR-mediated shedding [41].

Although it is clear that shedding is activated by GPCRoccupancy, the steps leading to metalloprotease activationare not well defined. In some cells, ligand shedding isdependent on PKC-induced activation of the metallo-protease [20,21]. In other instances, Src family kinasesappear to mediate EGFR pro-ligand cleavage [42], oralternatively might directly phosphorylate the EGFRand downstream adaptor proteins, such as Shc [6,43,44].Finally, in other systems the regulation of the synthesis ofthe EGF precursors might be a determining factor, as ingranulosa cells (see below).

GPCR–EGFR crosstalk in the neuroendocrine control of

reproduction

Studies of gonadotropin-releasing hormone (GnRH)-inducedsignals in hypothalamic- and pituitary-derived cellsindicate a role for MAPK and EGFR transactivation intransducing GPCR effects. GnRH is the master regulatorof the reproductive system, and pulsatile release of GnRHfrom hypothalamic neurons is crucial for regulating thesynthesis and release of the pituitary gonadotropins,follicle-stimulating hormone (FSH) and luteinizing

www.sciencedirect.com

hormone (LH), and for exerting autocrine actions in theregulation of GnRH secretion [45]. FSH and LH, in turn,are essential for steroidogenesis, spermatogenesis, folli-culogenesis and ovulation.

Recently, Roelle et al. [41] demonstrated a requirementfor the gelatinases MMP2 and MMP9 in mediatingGnRH-induced EGFR transactivation and ERK/MAPKactivation in aT3-1 and LbT2 gonadotroph cells. These arethe first members of the MMP family reported toparticipate in this signaling mechanism. Roelle et al.[41] also showed that GnRH-induced EGFR transactiva-tion resulted in Fos and Jun expression in aT3-1 cells.Treatment of aT3-1 cells with the EGFR-specific inhibitorAG1478, the gelatinase inhibitor Ro28-2653 or the Srckinase inhibitor PP2 abrogated GnRH induction of c-Fosand c-Jun protein accumulation. Furthermore, ribozyme-targeted downregulation of either MMP2 or MMP9resulted in reduced c-Fos and c-Jun protein levels.

GnRH-induced synthesis of the a- and b-subunits ofFSH and LH in pituitary cells was also shown to involveMAPK activity. Treatment with PD098059, the specificinhibitor of MEK (MAPK activator), completely blockedGnRH-induced activation of the human a-subunit pro-moter in aT3-1 cells [46], and of ovine FSHb- and ratLHb-subunit promoters in LbT2 cells [47,48]. GnRH-regulated LHb-subunit protein accumulation alsooccurred via MAPK signaling in LbT2 cells [49]. WhetherGnRH-mediated MAPK activation and synthesis of thesefactors in the pituitary involves the EGFR is not yet clear.However, it is tempting to speculate that GnRH-inducedEGFR transactivation might be important during FSHand LH production.

Consistentwitha role forEGFRtransactivation inGnRHsignaling, GnRH induced metalloproteinase-dependentHB-EGF shedding in immortalized GT1-7 hypothalamicneuronal cells [50]. Interestingly, EGFR-induced signal-ing in hypothalamic astroglia, which are active partners ofneurons, is crucial for the production of glial-derivedmolecules that stimulate GnRH neurons to release GnRH[51]. Targeted disruption of EGFR signaling in astroglia ofadult female mice resulted in irregular estrous cycles anddecreased LH secretion, probably because of alterations inGnRH release, thus compromising reproductive function[52]. Taken together, these studies suggest a crucial rolefor interplay between the GPCR and EGFR pathways inthe neuroendocrine regulation of female reproduction.

GPCR–EGFR crosstalk in the regulation of ovulation

The importance of crosstalk between the GPCR and EGFRsignaling networks for reproductive processes is furthersupported by recent studies that implicate selectedmembers of the EGF-like growth factor family asmediators of LH action in the ovulatory follicle [53,54].In the ovary, the LH surge triggers a cascade of events inthe preovulatory follicle that are crucial for ovulation of afertilizable oocyte [55]. LH binds to its Gs-coupled receptoron theca and granulosa cells to activate adenylyl cyclase,increase intracellular cAMP levels and activate down-stream signaling pathways [56]. This leads to the spatialand temporal expression of specific genes that promoteoocyte meiotic resumption, cumulus expansion and

Page 4: G-protein-coupled receptor signaling and the EGF network in endocrine systems

Review TRENDS in Endocrinology and Metabolism Vol.16 No.7 September 2005 323

breakdown of the follicle wall – events necessary for therelease of a productive oocyte [55]. Because oocytes andcumulus cells express few or no LH receptors, LH effectson these cells are thought to be indirect [57]. Themechanism(s) by which LH signals are transduced tocumulus–oocyte complexes has not been fully elucidated,and is an area of great interest.

Recent studies show that amphiregulin, epiregulin andbetacellulin are rapidly and transiently induced by anovulatory dose of LH/human chorionic gonadotropin(hCG) in mouse and rat ovaries within 1–3 h of hormonestimulation (Figure 2) [53,54]. Furthermore, the LH surgeinduces a large increase inmRNA encoding epiregulin andamphiregulin in naturally cycling rats [58] and mice(M. Hsieh, unpublished). Transcripts for these growthfactors localized specifically to mural granulosa cells ofpreovulatory follicles, but not to the cumulus cells thatsurround the oocyte (with the exception of betacellulin)[53]. LH-induced synthesis of amphiregulin and epiregulinwas also detected in primary cultures of human granulosacells [59]. In addition, expression of the gene encodingepiregulin has emerged from several screenings of LH-regulated genes [60,61]. Park et al. [53] showed that thecorrespondingprotein is indeedproducedbygranulosa cells,as measured by immunological means and metaboliclabeling. Given that the presence of bioactive EGF-likegrowth factors in the follicular fluid has been previouslyreported [62], a substantial amount of data points to theLH-regulatedproduction ofEGF-like growth factors in vivo.

These EGF-like factors mimic many of the effects of LHin vitro. In a follicle culture model that recapitulates most

TRENDS in Endocrinology & Metabolism

AC

cAMP

PKA

LHR

LH

Areg Ereg Btc

Gs

mRNA

DNA

EGF-likegenes

Figure 2. Model of LH regulation of EGF-like growth factor expression. The LH surge

or hCG stimulation causes a large increase inmRNA encoding amphiregulin (Areg),

epiregulin (Ereg) and betacellulin (Btc) via a cAMP-mediated process. Biochemical

and immunological evidence confirms the presence of these proteins on granulosa

cells. Abbreviations: AC, adenylyl cyclase; LHR, LH receptor.

www.sciencedirect.com

of the events that occur in vivo, exogenously appliedamphiregulin and epiregulin induced oocyte meioticresumption to the same extent as did LH, but with afaster time course of induction [53]. Betacellulin waspartially effective in promoting this effect. These findingsare reminiscent of early studies showing that in vitrotreatment with EGF promotes oocyte maturation and is apotent stimulus of cumulus expansion [63–65]. Thus,epiregulin, amphiregulin and betacellulin are probablythe physiological ligands for the EGFR in the follicle. Thethree growth factors stimulated cumulus expansion inintact follicles, and the expression of the PTGS2 (prosta-glandinendoperoxidesynthase2, alsocalledcyclooxygenase2 or COX2), HAS2 (hyaluronan synthase 2), and TNFAIP6(tumor necrosis factor-a-induced protein 6, also calledTNF-stimulated gene 6 or TSG6) genes that are associatedwith this event [53,54]. In cultures of isolated cumulus–oocyte complexes where LH no longer has an effect, solubleamphiregulin, epiregulin or betacellulin were effective inpromoting cumulus expansion and oocyte maturation [53].Notably, these factors were unable to induce maturation ofdenuded oocytes, indicating a requirement for interactionsbetween theoocyteandcumuluscells [53].A similar absenceof direct effects on the isolated oocyte has been reportedwhen using EGF [64,66,67].

That LH effects are dependent on the activation of theEGF network is supported by the following observations.LH-induced maturation of cumulus–oocyte complexesrequires the activity of metalloproteases, because thepotent broad-spectrum metalloprotease inhibitor GM6001(also called Galardin or Ilomastat), but not a specificMMP2/MMP9 inhibitor, blocked LH-induced but notepiregulin-induced oocyte maturation in rat follicles [54].In addition, LH effects appear to involve activation of theEGFR found on granulosa and cumulus cells. LH/hCGinduced EGFR phosphorylation in mouse ovaries and incultured follicles; inhibition of EGFR activity by AG1478prevented LH-induced oocyte maturation and cumulusexpansion in vitro [53,54]. Moreover, injection of AG1478into rat ovarian bursas resulted in a significant decreasein LH-induced ovulation in the treated ovary, comparedwith the untreated controlateral ovaries and vehicle-treated ovaries [54]. An increased incidence of largefollicles containing entrapped oocytes, many of whichhad not undergone meiotic resumption and were sur-rounded by unexpanded cumulus cells, was observed intreated ovaries.

Based on the above findings, EGF-like growth factorsprobably function as mediators of LH action in theovulatory follicle. Because LH regulates mRNA andprotein synthesis, the transregulation of EGFR occurs ina time frame longer than the classic rapid regulation oflatent growth factor precursors. It remains to be deter-mined whether LH-stimulated shedding of pre-existinggrowth factor precursors has a role in the early stages ofgonadotropin signaling. A model of GPCR-mediatedEGFR transactivation can be presented, in which LHactivation of its specific GPCR leads to regulation ofEGF-like growth factors at the transcriptional level and atthe level of processing of ligands to their mature forms(Figure 3). The soluble EGF-like growth factors then act in

Page 5: G-protein-coupled receptor signaling and the EGF network in endocrine systems

TRENDS in Endocrinology & Metabolism

LHR

LH

?

PKA

AC

Granulosa cell

Gs

ERK/MAPKP P

P P P P

P P

cAMP

EGFRdimer

(Solubleligand)

Cumulus expansion

Cumulus cell

Oocyte meioticresumption

COX2TSG6HAS2

ERK/MAPK

AregEregBtc

MMP orADAMs

Pro-ligand

Figure 3. Model of EGFR transactivation in the regulation of LH-induced ovulation. LH activates the LHR on granulosa cells of preovulatory follicles, leading to stimulation of

cAMP-dependent signal cascades and the expression of mRNAs encoding the EGF-like growth factors amphiregulin (Areg), epiregulin (Ereg) and betacellulin (Btc). LH also

inducesmetalloproteinase-dependent processing of the pro-forms of these growth factors for the release ofmature peptides from the cell surface. The soluble growth factors

exert autocrine and paracrine effects through activation of the EGFR and ERK/MAPK signaling in granulosa cells and cumulus cells. This leads to oocyte meiotic resumption

and cumulus expansion, events crucial for the ovulatory process. Abbreviations: AC, adenylyl cyclase; LHR, LH receptor.

Review TRENDS in Endocrinology and Metabolism Vol.16 No.7 September 2005324

either an autocrine or paracrine manner to activate theEGFR, with subsequent activation of the MAPK cascade.Indeed, MAPK activity in cumulus cells has been shown tobe necessary for LH-induced cumulus expansion andoocyte maturation [68], and preliminary results suggestthat LH-induced MAPK activation in cumulus–oocytecomplexes is dependent on EGFR activity (M. Hsieh,unpublished).

Concluding remarks

The above studies show that EGFR transactivation is acommon mechanism by which an initial stimulus ispropagated to heterogeneous signal transduction path-ways. This transactivation was studied initially incultured cells in the realm of cancer biology or woundhealing. However, studies under more physiologicalconditions, particularly in the reproductive tract, supporta broader function for this signaling module. Thepossibility that activation of the EGF network plays animportant role in propagating the LH signal is an exampleof the involvement of this module in a physiologicalprocess. Understanding how this transactivation func-tions in the context of the follicle should provide a newperspective of this process. However, several major issuesstill need to be addressed. Most notably, the mechanism bywhich GPCRs activate metalloproteases involved inEGF-like growth factor shedding remains to be elucidated.Which metalloprotease(s) processes these growth factorsin the follicle is also unclear. Potential candidates can beinferred from what is known in other cell systems. This isan important issue because it might provide the specificityrequired for selective pharmacological manipulation.Finally, further studies are necessary to understand howEGFR transactivation in cumulus cells impacts on oocytemeiotic resumption. A better understanding of the role ofGPCR transactivation of the EGF network should open

www.sciencedirect.com

new doors for the manipulation of physiological processes,including ovulation, or the improvement of in vitro cultureconditions of human oocytes.

AcknowledgementsThe work done in our laboratory was supported by National Institute ofChild Health and Human Development NIH grant HD20788 and by acooperative agreement (U54-HD31398) as part of the SpecializedCooperative Centers Program in Reproduction Research.

References

1 Pierce, K.L. et al. (2002) Seven-transmembrane receptors. Nat. Rev.Mol. Cell Biol. 3, 639–650

2 Vossler, M.R. et al. (1997) cAMP activates MAP kinase and Elk-1through a B-Raf- and Rap1-dependent pathway. Cell 89, 73–82

3 Daaka, Y. et al. (1997) Switching of the coupling of the b2-adrenergicreceptor to different G proteins by protein kinase A.Nature 390, 88–91

4 Fischer, O.M. et al. (2003) EGFR signal transactivation in cancer cells.Biochem. Soc. Trans. 31, 1203–1208

5 Dikic, I. et al. (1996) A role for Pyk2 and Src in linking G-protein-coupled receptors with MAP kinase activation. Nature 383, 547–550

6 Keely, S.J. et al. (2000) Carbachol-stimulated transactivation ofepidermal growth factor receptor and mitogen-activated proteinkinase in T(84) cells is mediated by intracellular Ca2C, PYK-2, andp60src. J. Biol. Chem. 275, 12619–12625

7 Keiper, M. et al. (2004) Epac- and Ca2C -controlled activation of Rasand extracellular signal-regulated kinases by Gs-coupled receptors.J. Biol. Chem. 279, 46497–46508

8 Enserink, J.M. et al. (2002) A novel Epac-specific cAMP analoguedemonstrates independent regulation of Rap1 and ERK. Nat. CellBiol. 4, 901–906

9 Daub, H. et al. (1996) Role of transactivation of the EGF receptor insignalling by G-protein-coupled receptors. Nature 379, 557–560

10 Wetzker, R. and Bohmer, F.D. (2003) Transactivation joins multipletracks to the ERK/MAPK cascade.Nat. Rev. Mol. Cell Biol. 4, 651–657

11 Maudsley, S. et al. (2000) The b2-adrenergic receptor mediatesextracellular signal-regulated kinase activation via assembly of amulti-receptor complex with the epidermal growth factor receptor.J. Biol. Chem. 275, 9572–9580

12 Gschwind, A. et al. (2002) Lysophosphatidic acid-induced squamouscell carcinoma cell proliferation andmotility involves epidermal growthfactor receptor signal transactivation. Cancer Res. 62, 6329–6336

Page 6: G-protein-coupled receptor signaling and the EGF network in endocrine systems

Review TRENDS in Endocrinology and Metabolism Vol.16 No.7 September 2005 325

13 Asakura, M. et al. (2002) Cardiac hypertrophy is inhibited byantagonism of ADAM12 processing of HB-EGF: metalloproteinaseinhibitors as a new therapy. Nat. Med. 8, 35–40

14 Zhong, M. et al. (2003) Extracellular signal-regulated kinase 1/2activation by myometrial oxytocin receptor involves Ga(q)Gbg andepidermal growth factor receptor tyrosine kinase activation. Endo-crinology 144, 2947–2956

15 Ahmed, I. et al. (2003) Transactivation of the epidermal growthfactor receptor mediates parathyroid hormone and prostaglandinF2a-stimulated mitogen-activated protein kinase activation incultured transgenic murine osteoblasts. Mol. Endocrinol. 17,1607–1621

16 Sales, K.J. et al. (2004) Expression, localization, and signaling ofprostaglandin F2 a receptor in human endometrial adenocarcinoma:regulation of proliferation by activation of the epidermal growth factorreceptor and mitogen-activated protein kinase signaling pathways.J. Clin. Endocrinol. Metab. 89, 986–993

17 Yarden, Y. and Sliwkowski, M.X. (2001) Untangling the ErbBsignalling network. Nat. Rev. Mol. Cell Biol. 2, 127–137

18 Holbro, T. and Hynes, N.E. (2004) ErbB receptors: directing keysignaling networks throughout life. Annu. Rev. Pharmacol. Toxicol.44, 195–217

19 Schlessinger, J. (2000) Cell signaling by receptor tyrosine kinases.Cell103, 211–225

20 Massague, J. and Pandiella, A. (1993) Membrane-anchored growthfactors. Annu. Rev. Biochem. 62, 515–541

21 Sahin, U. et al. (2004) Distinct roles for ADAM10 and ADAM17in ectodomain shedding of six EGFR ligands. J. Cell Biol. 164,769–779

22 Sunnarborg, S.W. et al. (2002) Tumor necrosis factor-a convertingenzyme (TACE) regulates epidermal growth factor receptor ligandavailability. J. Biol. Chem. 277, 12838–12845

23 Hinkle, C.L. et al. (2004) Selective roles for tumor necrosis factora-converting enzyme/ADAM17 in the shedding of the epidermalgrowth factor receptor ligand family: the juxtamembrane stalkdetermines cleavage efficiency. J. Biol. Chem. 279, 24179–24188

24 Lemjabbar, H. and Basbaum, C. (2002) Platelet-activating factorreceptor and ADAM10 mediate responses to Staphylococcus aureus inepithelial cells. Nat. Med. 8, 41–46

25 Peschon, J.J. et al. (1998) An essential role for ectodomain shedding inmammalian development. Science 282, 1281–1284

26 Jackson, L.F. et al. (2003) Defective valvulogenesis in HB-EGF andTACE-null mice is associated with aberrant BMP signaling. EMBO J.22, 2704–2716

27 Mann, G.B. et al. (1993) Mice with a null mutation of the TGF a genehave abnormal skin architecture, wavy hair, and curly whiskers andoften develop corneal inflammation. Cell 73, 249–261

28 Miettinen, P.J. et al. (1995) Epithelial immaturity and multiorganfailure in mice lacking epidermal growth factor receptor. Nature 376,337–341

29 Threadgill, D.W. et al. (1995) Targeted disruption of mouse EGFreceptor: effect of genetic background on mutant phenotype. Science269, 230–234

30 Sibilia, M. and Wagner, E.F. (1995) Strain-dependent epithelialdefects in mice lacking the EGF receptor. Science 269, 234–238

31 Le Gall, S.M. et al. (2003) Regulated cell surface pro-EGF ectodomainshedding is a zinc metalloprotease-dependent process. J. Biol. Chem.278, 45255–45268

32 Izumi, Y. et al. (1998) Ametalloprotease-disintegrin, MDC9/meltrin-g/ADAM9 and PKCd are involved in TPA-induced ectodomain sheddingof membrane-anchored heparin-binding EGF-like growth factor.EMBO J. 17, 7260–7272

33 Yan, Y. et al. (2002) The metalloprotease Kuzbanian (ADAM10)mediates the transactivation of EGF receptor by G protein-coupledreceptors. J. Cell Biol. 158, 221–226

34 Kurisaki, T. et al. (2003) Phenotypic analysis of Meltrin a (ADAM12)-deficient mice: involvement of Meltrin a in adipogenesis andmyogenesis. Mol. Cell. Biol. 23, 55–61

35 Brachmann, R. et al. (1989) Transmembrane TGF-a precursorsactivate EGF/TGF-a receptors. Cell 56, 691–700

36 Wong, S.T. et al. (1989) The TGF-a precursor expressed on the cellsurface binds to the EGF receptor on adjacent cells, leading to signaltransduction. Cell 56, 495–506

www.sciencedirect.com

37 Takemura, T. et al. (1997) The membrane-bound form of heparin-binding epidermal growth factor-like growth factor promotes survivalof cultured renal epithelial cells. J. Biol. Chem. 272, 31036–31042

38 Gschwind, A. et al. (2003) TACE cleavage of proamphiregulinregulates GPCR-induced proliferation and motility of cancer cells.EMBO J. 22, 2411–2421

39 Prenzel, N. et al. (1999) EGF receptor transactivation by G-protein-coupled receptors requires metalloproteinase cleavage of proHB-EGF.Nature 402, 884–888

40 Schafer, B. et al. (2004) Distinct ADAMmetalloproteinases regulate Gprotein-coupled receptor-induced cell proliferation and survival.J. Biol. Chem. 279, 47929–47938

41 Roelle, S. et al. (2003) Matrix metalloproteinases 2 and 9 mediateepidermal growth factor receptor transactivation by gonadotropin-releasing hormone. J. Biol. Chem. 278, 47307–47318

42 Zhang, Q. et al. (2004) SRC family kinases mediate epidermal growthfactor receptor ligand cleavage, proliferation, and invasion of headand neck cancer cells. Cancer Res. 64, 6166–6173

43 Luttrell, L.M. et al. (1996) Role of c-Src tyrosine kinase in G protein-coupled receptor- and Gbg subunit-mediated activation of mitogen-activated protein kinases. J. Biol. Chem. 271, 19443–19450

44 Biscardi, J.S. et al. (1999) c-Src-mediated phosphorylation of theepidermal growth factor receptor on Tyr845 and Tyr1101 is associatedwith modulation of receptor function. J. Biol. Chem. 274, 8335–8343

45 Krsmanovic, L.Z. et al. (2003) An agonist-induced switch in G proteincoupling of the gonadotropin-releasing hormone receptor regulatespulsatile neuropeptide secretion. Proc. Natl. Acad. Sci. U. S. A. 100,2969–2974

46 Harris, D. et al. (2003) Extracellular signal-regulated kinase andc-Src, but not Jun N-terminal kinase, are involved in basal andgonadotropin-releasing hormone-stimulated activity of the glyco-protein hormone a-subunit promoter. Endocrinology 144, 612–622

47 Bonfil, D. et al. (2004) Extracellular signal-regulated kinase, JunN-terminal kinase, p38, and c-Src are involved in gonadotropin-releasing hormone-stimulated activity of the glycoprotein hormonefollicle-stimulating hormone b-subunit promoter. Endocrinology 145,2228–2244

48 Harris, D. et al. (2002) Activation of MAPK cascades by GnRH: ERKand Jun N-terminal kinase are involved in basal and GnRH-stimulated activity of the glycoprotein hormone LHb-subunitpromoter. Endocrinology 143, 1018–1025

49 Liu, F. et al. (2002) GnRH activates ERK1/2 leading to the induction ofc-fos and LHb protein expression in LbT2 cells. Mol. Endocrinol. 16,419–434

50 Shah, B.H. et al. (2004) Neuropeptide-induced transactivation of aneuronal epidermal growth factor receptor is mediated by metallo-protease-dependent formation of heparin-binding epidermal growthfactor. J. Biol. Chem. 279, 414–420

51 Ma, Y.J. et al. (1997) Hypothalamic astrocytes respond to transform-ing growth factor-a with the secretion of neuroactive substances thatstimulate the release of luteinizing hormone-releasing hormone.Endocrinology 138, 19–25

52 Li, B. et al. (2003) Compromised reproductive function in adult femalemice selectively expressing mutant ErbB-1 tyrosine kinase receptorsin astroglia. Mol. Endocrinol. 17, 2365–2376

53 Park, J.Y. et al. (2004) EGF-like growth factors as mediators of LHaction in the ovulatory follicle. Science 303, 682–684

54 Ashkenazi, H. et al. (2005) Epidermal growth factor family members:endogenous mediators of the ovulatory response. Endocrinology 146,77–84

55 Richards, J.S. et al. (2002) Ovulation: new dimensions and newregulators of the inflammatory-like response. Annu. Rev. Physiol. 64,69–92

56 Richards, J.S. (2001) New signaling pathways for hormones and cyclicadenosine 3 0,5 0-monophosphate action in endocrine cells. Mol.

Endocrinol. 15, 209–21857 Peng, X.R. et al. (1991) Localization of luteinizing hormone receptor

messenger ribonucleic acid expression in ovarian cell types duringfollicle development and ovulation. Endocrinology 129, 3200–3207

58 Sekiguchi, T. et al. (2004) Expression of epiregulin and amphiregulinin the rat ovary. J. Mol. Endocrinol. 33, 281–291

59 Freimann, S. et al. (2004) EGF-like factor epiregulin and

Page 7: G-protein-coupled receptor signaling and the EGF network in endocrine systems

Review TRENDS in Endocrinology and Metabolism Vol.16 No.7 September 2005326

amphiregulin expression is regulated by gonadotropins/cAMP inhuman ovarian follicular cells. Biochem. Biophys. Res. Commun.324, 829–834

60 Espey, L.L. and Richards, J.S. (2002) Temporal and spatial patterns ofovarian gene transcription following an ovulatory dose of gonado-tropin in the rat. Biol. Reprod. 67, 1662–1670

61 Robert, C. et al. (2001) Differential display and suppressivesubtractive hybridization used to identify granulosa cell messengerRNA associated with bovine oocyte developmental competence. Biol.Reprod. 64, 1812–1820

62 Hsu, C.J. et al. (1987) Ovarian epidermal growth factor-like activity.Concentrations in porcine follicular fluid during follicular enlarge-ment. Biochem. Biophys. Res. Commun. 147, 242–247

63 Dekel, N. and Sherizly, I. (1985) Epidermal growth factor inducesmaturation of rat follicle-enclosed oocytes. Endocrinology 116,406–409

64 Downs, S.M. et al. (1988) Induction of maturation in cumulus cell-

Endea

the quarterly magaziand philosophy

You can access EndeScienceDirect, whecollection of beaut

articles on the historreviews and edito

featuri

Selling the silver: country house libraries and the hisCarl Schmidt – a chemical tourist in V

The rise, fall and resurrection of gMary Anning: the fossilist as

Caroline Herschel: ‘the unqScience in the 19th-century

The melancholy of an

and comin

Etienne Geoffroy St-Hillaire, Napoleon’s Egyptian camLosing it in New Guinea: The voyage o

The accidental conservatPowering the porter bre

Female scientists in fi

and much, muc

Locate Endeavour on ScienceDirect

www.sciencedirect.com

enclosed mouse oocytes by follicle-stimulating hormone and epidermalgrowth factor: evidence for a positive stimulus of somatic cell origin.J. Exp. Zool. 245, 86–96

65 Prochazka, R. et al. (2000) Developmental regulation of effect ofepidermal growth factor on porcine oocyte–cumulus cell complexes:nuclear maturation, expansion, and F-actin remodeling. Mol. Reprod.Dev. 56, 63–73

66 Downs, S.M. (1989) Specificity of epidermal growth factor action onmaturation of the murine oocyte and cumulus oophorus in vitro. Biol.Reprod. 41, 371–379

67 Ben-Yosef, D. et al. (1992) Rat oocytes induced to mature by epidermalgrowth factor are successfully fertilized. Mol. Cell. Endocrinol. 88,135–141

68 Su, Y.Q. et al. (2002) Mitogen-activated protein kinase activity incumulus cells is essential for gonadotropin-induced oocyte meioticresumption and cumulus expansion in the mouse. Endocrinology 143,2221–2232

vour

ne for the historyof science

avour online viare you’ll find aifully illustratedy of science, bookrial comment.

ng

tory of science by Roger Gaskell and Patricia Faraictorian Britain by R. Stefan Ross

roup selection by M.E. Borelloexegete by T.W. Goodhueuiet heart’ by M. Hoskinzoo by Oliver Hochadelatomy by P. Fara

g soon

paign and a theory of everything by P. Humphriesf HMS Rattlesnake by J. Goodmanionist by M.A. Andreiwery by J. Sumner

lms by B.A. Jones

h more . . .

(http://www.sciencedirect.com)