MINIREVIEW

Minireview: Role of Intracellular Scaffolding Proteins in the Regulation of Endocrine G Protein-Coupled Receptor Signaling Cornelia Walther and Stephen S. G. Ferguson J. Allyn Taylor Centre for Cell Biology (C.W., S.S.G.F.), Robarts Research Institute, and Department of Physiology and Pharmacology (S.S.G.F.), University of Western Ontario, London, Ontario, Canada N6A 5K8

The majority of hormones stimulates and mediates their signal transduction via G protein-coupled receptors (GPCRs). The signal is transmitted into the cell due to the association of the GPCRs with heterotrimeric G proteins, which in turn activates an extensive array of signaling pathways to regulate cell physiology. However, GPCRs also function as scaffolds for the recruitment of a variety of cytoplasmic protein-interacting proteins that bind to both the intracellular face and protein interaction motifs encoded by GPCRs. The structural scaffolding of these proteins allows GPCRs to recruit large functional complexes that serve to modulate both G protein-dependent and -independent cellular signaling pathways and modulate GPCR intracellular trafficking. This review focuses on GPCR interacting PSD95-disc large-zona occludens domain containing scaffolds in the regulation of endocrine receptor signaling as well as their potential role as therapeutic targets for the treatment of endocrinopathies. (Molecular Endocrinology 29: 814 – 830, 2015)

ndocrine function is regulated by a myriad of hormones with the majority signaling their targets through G protein-coupled receptors (GPCRs). In humans, the superfamily GPCRs represents the largest family of cell surface proteins with approximately 800 GPCR genes identified to date (1–3). Even though the members of the GPCR family share a common molecular architecture consisting of seven-transmembrane domains connected by three extracellular and three intracellular loops, they display significant sequence and functional diversity (4 – 6). GPCRs are the target of a vast variety of ligands and thus function as molecular machines and transmit an incredible wide range of extracellular signals across the cell membrane as intracellular signals that are recognized by the cell. GPCRs are involved in the regulation of virtually all essential physiological processes in the human body. Thus, it is easy to appreciate that the dysfunction of GPCR-mediated signaling pathways can cause pathological states (7). Although GPCRs comprise only approximately 3% of all the genes encoded by the human ge-

E

nome, they represent the target for 40%–50% of all prescription drugs (3, 8), and endocrine GPCRs play a pivotal role as sensors regulating endocrine function (9). GPCRs act as ligand-regulated guanine nucleotide exchange factors for heterotrimeric GTP-binding proteins (G proteins) and thereby transduce extracellular signals into intracellular second messengers. Upon ligand binding GPCRs undergo a conformational change to adopt an active conformation. This facilitates the exchange of GDP for GTP on the G protein G␣-subunit and leads to the dissociation of the G␣-subunit from the receptor and the G␤␥-subunit. The G protein subunits then regulate a variety of downstream effectors, such as ion channels, phospholipases, and adenylyl cyclases (10). However, over the past 2 decades, new concepts for GPCR signal transduction have been described making the regulation of GPCR signaling more diverse and complicated than previously anticipated. In addition to the classical paradigm of G protein-mediated signal transduction, GPCRs have been shown to signal via G protein-independent mechanisms

ISSN Print 0888-8809 ISSN Online 1944-9917 Printed in USA Copyright © 2015 by the Endocrine Society Received April 1, 2015. Accepted April 29, 2015. First Published Online May 5, 2015

Abbreviations: ␣1/2AR, ␣1/2-adrenergic receptor; ␤1/2AR, ␤1/2-adrenergic receptor; AKAP, A kinase anchor proteins; AT1-i2m, AT1R second intracellular loop mutant; AT1R, angiotensin II type 1a receptor; CAL/PIST/GOPC, Golgi-associated PDZ and coiled-coil motif-containing protein; CRF, corticotropin-releasing factor; CRFR1, corticotropinreleasing factor receptor 1; D2R, dopamine receptor 2; EE, early endosome;

814

press.endocrine.org/journal/mend

Mol Endocrinol, June 2015, 29(6):814 – 830

doi: 10.1210/me.2015-1091

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

doi: 10.1210/me.2015-1091

such as ␤-arrestin-mediated signaling pathways (2, 11– 14). This has revealed that heterotrimeric G proteins are not the only GPCR interacting proteins (GIPs) that modulate GPCR signal transduction. It is now appreciated that the nature, kinetics, and strength of GPCR signaling is modulated by a vastly growing number of GIPs (see reviews in references 15–19). GPCR scaffolded proteins are capable of associating with multiple proteins that enable the fine-tuning of GPCR signaling (20). Consequently, the GPCR-dependent scaffolding of intracellular regulatory proteins provides the potential for the development of novel strategies that target specific intracellular protein interactions with GPCR that regulate specific GPCR intracellular signaling pathways. Specifically, this type of strategy may be of therapeutic value for the treatment of endocrine-related diseases. This review will focus on the current, yet evolving, understanding of the regulation of endocrine signaling by GPCR-scaffolded proteins and the potential targeting GPCR-scaffolded complexes for the treatment of endocrine diseases. GPCR signal transduction Upon agonist stimulation, in addition to heterotrimeric G protein coupling, the intracellular domains of the GPCR serves as substrates for phosphorylation by either second messenger-dependent protein kinases or G protein-coupled receptor kinases (GRKs) (2, 11–14). GPCR agonist activation and GRK phosphorylation subsequently results in the recruitment of ␤-arrestin proteins that contribute to the waning of GPCR responsiveness to repeated agonist stimulation; an important physiological mechanism that is termed desensitization. In addition to terminating G protein signaling, ␤-arrestin proteins also function as adaptor proteins that target GPCRs for clathrin-mediated endocytosis via their ability to recruit both clathrin and ␤-adaptin (11, 21–26). After internalization various different subcellular trafficking events take place EP, endogenous prostanoid; ER, endoplasmatic reticulum; GAIP/RGS, regulator of G protein signaling; GHSR1, GH secretagogue receptor or ghrelin receptor 1; GIP, G proteincoupled receptor interacting protein; GIPC, GAIP-interacting protein C terminus; GPCR, G protein-coupled receptor; GPR15, G protein-coupled receptor 15; GPR30, G proteincoupled estrogen receptor 1/G protein-coupled receptor 30; GRK, G protein-coupled receptor kinase; HPA, hypothalamic-pituitary-adrenal; 5-HT2A/2CR, serotonin 2A/2C receptor; IP3, inositol 1,4,5-trisphosphate; Jak, Janus kinase; LHR, LH receptor; LPA, lysophosphatidic acid; m3 AChR, M3-muscarinic acetylcholine receptor; MAGI, membraneassociated guanylate kinase, WW, and PDZ domain-containing proteins; MT1R, melatonin 1 receptor; MUPP-1, multiple PDZ domain protein; NHERF, Na⫹/H⫹ exchanger regulatory factor; PAR, protease-activated receptor; PDZ, postsynaptic density 95/disc large/zona occludens-1; PDZK, PDZ kinase; PDZ protein, PDZ-domain-containing proteins; PGE2, prostaglandin E2; PICK1, PRKCA-binding protein; PKA, protein kinase A; PKC, protein kinase C; PLC, phospholipase C; PSD-95, postsynaptic-density protein 95; PTH1R, PTH receptor; RGS, regulator of G protein signaling; SAP97, synapse-associated protein 97; SNX27, sorting nexin 27; SSTR, somatostatin receptor; STAT, signal transducers and activators of transcription; T2DM, type 2 diabetes mellitus; TGN, trans-Golgi network; TSHR, TSH receptor.

press.endocrine.org/journal/mend

815

to either restore GPCR responsiveness or to target the down-regulation of GPCRs (11, 27). The activation of a GPCR due to agonist binding leads to a conformational change in the receptor (transmembrane helix rearrangement) (28), which promotes the direct interaction of the GPCR with heterotrimeric G proteins via the four intracellular domains. Heterotrimeric G proteins are composed of ␣-, ␤-, and ␥-subunits (29). There are 16 known ␣-subunits, which are divided into four subfamilies based on sequence and functional similarities: G␣S, G␣i, G␣q, and G␣12/13. Gi subfamily members decrease the level of the intracellular second messenger cAMP by inhibiting adenylyl cyclase activity or activating phosphodiesterases. Members of the Gs family activate adenylyl cyclase, which results in the production of cAMP and the subsequent stimulation of protein kinase A (PKA), a process that is antagonized by G␣i-coupled GPCRs. The activation of G␣q proteins leads to the activation of phospholipase C (PLC), the formation of inositol 1,4,5 trisphosphate (IP3) and diacylglycerol, the release of intracellular Ca2⫹ stores, and the subsequent activation of protein kinase C (PKC). Members of the G12/13 family are well known to regulate small GTPases like Rho (30 –32). GPCR/G protein binding leads to the activation of G proteins due to the release of GDP and binding of GTP to the G␣ subunit (20, 33). The G␣- and G␤␥-subunits then dissociate and mediate the regulation of the various downstream effector systems mentioned above which results in changes in cellular physiology. This pathway, comprising monomeric/dimeric GPCRs, heterotrimeric G proteins, and multiple effector systems, is referred to as the canonical signaling pathway. However, this basic concept is somewhat outdated and has been revised over the past decades of GPCR research. We now know that there are various other GIPs other than G proteins that enable fine-tuning of signaling in a receptor subtype- and tissue-specific manner (5). GIPs play crucial roles in receptor signaling as they can do one of the following: 1) directly mediate receptor signaling, 2) control receptor localization and/or trafficking, 3) act as allosteric modulators, or 4) act as scaffold proteins that link the receptor to downstream effectors (20). This has led to the discovery of potential mechanisms for GPCR signaling that extend beyond classical paradigms. Because GPCRs can interact with multiple G protein and non-G protein effectors, it is now appreciated that GPCR signaling is pluridimensional (34). In past years, several receptors have been identified that are either able to couple to multiple G proteins or differentially activate G proteins different tissues thereby enable a single GPCR to engage in several signaling pathways simultaneously, ie, muscarinic acetylcholine recep-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

816

Walther and Ferguson

Endocrine GPCR Interacting Proteins

Mol Endocrinol, June 2015, 29(6):814 – 830

tors, human TSH receptor (TSHR), and D1 dopamine receptor (35–38). However, G protein signaling alone does not qualify for the term pluridimensional GPCR signaling. It was the discovery of the underlying robust G protein-independent signaling network that revealed the true GPCR efficacy (13, 37, 39). The key players in G protein-independent signaling pathways and most likely the best characterized non-G protein effectors are the arrestins. This family of cytosolic scaffolding proteins is well known for its critical role in GPCR desensitization. As indicated above, ␤-arrestin binding not only precludes G protein activation and targets GPCRs for endocytosis, the nonvisual arrestins also initiate a second wave of signaling that takes place as soon as G protein signaling vanishes and as receptors redistribute to intracellular compartments (39). Less than 20 years ago, it was believed that ␤-arrestins’ exclusive role in GPCR signaling is binding to the receptor to arrest G protein-mediated signaling (11). However, in the original studies demonstrating additional roles for ␤-arrestin function showed that they also scaffold protein complexes required for the initiation of endocytosis (clathrin and ␤-adaptin) and signaling (c-Src) (13, 21, 23, 25). The first example of ␤-arrestin signaling was reported in 1999 by Luttrell et al (13), who discovered ␤-arrestin-dependent formation of a c-Src signaling complex with the ␤2-adrenergic receptor (␤2AR). Furthermore, recent evidence suggests that not only GPCR desensitization but also internalization is a signaling event (11, 24). Particularly, the introduction of new techniques, such as conformational biosensors (40), has generated the hypothesis that cell signaling events occur within intracellular compartments (41). Such signaling processes from endosomes either include dynamic sites of GPCR/G protein activation or comprise signal control in endosomes independent of G proteins (reviewed in references 41– 43). Calebiro et al (44) and Ferrandon et al (45) discovered the requirement of GPCR/G protein internalization for sustained or persistent G␣s signaling from an endosomal compartment. Although the TSHR was found in a preGolgi compartment in close association with the G␣s subunit and adenylyl cyclase III (44), the internalization of the PTH receptor occurred in association with PTH (1– 34) and G␣s, most likely as an active ternary complex (45). Furthermore, a distinct mechanism of sustained G protein activation in which the GPCR, ␤-arrestin, and G protein ␤␥-subunits form a complex has been reported in a study of the PTH receptor signaling (46). A comparable observation has been reported for the vasopressin 2 receptor, in which a similar complex seems to be involved in a sustained cAMP generation (47).

G protein-independent signaling comprises not only ␤-arrestin-mediated signaling processes (after G protein signaling) but also signaling events independent of G protein activation. One example is the well-studied angiotensin II type 1a receptor (AT1R). Studies with cardiomyocytes demonstrated that strain, pressure, and shear stress induced AT1R activation and subsequent downstream ERK activation (48). Another study with AT1R mutants incapable to couple to heterotrimeric G proteins showed an angiotensin II-mediated activation of Scr and ERK (49). Furthermore, Zhai et al (50) studied the distinct physiological consequences of G protein-independent signal transduction via the AT1R in the heart. They generated transgenic mice with cardiac-specific overexpression of a wild-type AT1R or an AT1R second intracellular loop mutant (AT1-i2m). AT1-i2m mice displayed a pronounced cardiac phenotype and found impaired AT1i2m-mediated hypertrophy in in vitro assays upon pharmacological inhibition of ERK and Src activity. However, it is unclear what underlying mechanism is mediating the G protein-independent activation of cytoplasmic effectors such as ERK and Scr. It remains to be elucidated what signaling (or other) molecule could potentially serve proximal functions similar to G proteins, but ␤-arrestin represents the most intriguing candidate (14, 50). Of note, ␤-arrestin-mediated signaling events can also take place in the absence of G protein activation. This has been demonstrated for the decoy receptor chemokine receptor-7 in which ligand binding activates MAPKs through ␤-arrestins in transiently transfected cells but does not result in activation of signaling pathways typical of G proteins (51). Pharmacologically, GPCRs can be exploited in different ways, one of which is by targeting the orthosteric ligand binding site, which is the primary ligand binding site for the endogenous agonist (52, 53). The basic concept, that agonist binding induces a conformational change and switches the GPCR from an off to an on state, has been revised over the past several years because it is now appreciated that there is no equilibrium between on-off conformations. GPCRs rather adopt an ensemble of tertiary conformations that allow for new strategies that make use of other, spatially distinct allosteric binding sites to regulate diverse aspects of GPCR signaling (37, 54 –56). Furthermore, the active state of a receptor is specified by the ligand-receptor complex and other small molecule or protein-protein interaction that allosterically constrains the conformations available to the receptor (57). Consequently, GPCRs can adopt multiple active conformations (58). Ligands that selectively bind and stabilize different GPCR conformations can modulate either extracellular (other ligands) or intracellular (signal trans-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

doi: 10.1210/me.2015-1091

ducing molecules) interactions and are called either orthosteric or allosteric modulators. They have evolved as important pharmacological tools to couple GPCRs to selectively activate distinct and/or overlapping signal transduction pathways. This phenomenon is termed biased signaling (52, 59 – 63). The phenomenon of signaling bias from the inside of the cell rather than from the ligandbinding side of the receptor is the newest concept (64). Key components for the coordination of signal transduction cascades are scaffolding proteins. The importance of scaffolding proteins for GPCR signaling (Figure 1) Scaffolding proteins are defined as proteins that bring together multiple proteins to assemble functional complexes thus modulating cellular signaling pathways. This is achieved by regulating numerous GPCR regulatory processes such as receptor stabilization, subcellular localization, and trafficking (sorting and recycling), which ultimately modulate downstream signaling events (65). Cy-

Figure 1. Schematic of GPCR/scaffolding protein interactions involved in GPCR signaling pathways (individual examples are discussed in the text). Key proteins in the regulation of GPCR signaling are arrestins, PDZ proteins, and non-PDZ proteins, which all interact with the carboxyl terminal tail or the intracellular loops (mainly, but not exclusively, the third loop), respectively. Agonist activation of GPCRs leads to receptor phosphorylation which enables, yet is not always required, arrestin binding. A, Arrestin serves as adapter to link GPCRs to the endocytic machinery. Arrestin per se can also interact with multiple MAPKs and therefore is an important regulator of GPCR signaling. Furthermore, GPCR signaling from endosomes has been reported recently. B, The second group of proteins important for GPCR signaling is PDZ proteins. PDZ proteins have been shown to regulate various signaling pathways, such as the activation of ERK1/2, enhance or abolish the interaction with G proteins, hence modulating G protein-mediated signaling, facilitate the interaction with arrestin and PLC, and can activate Rho GTPase. The third group of scaffolding proteins that control GPCR signaling is the group of non-PDZ proteins. DAG, diacylglycerol. C, Non-PDZ proteins that interact with GPCRs have the ability to bind PDZ proteins but can also associate with PKA, Jak2 (to activate the Jak/STAT signaling pathway) and orchestrate Ras/ Raf signaling cascades.

press.endocrine.org/journal/mend

817

toplasmatic scaffolding proteins can be divided into three broad categories: arrestins, postsynaptic density 95/disc large/zona occludens-1 (PDZ) domain-containing proteins (PDZ proteins), and non-PDZ proteins (Table 1). GPCRs interact with such scaffolding proteins via their three intracellular loop domains and their carboxyl-terminus (20). As for the scaffolding proteins, the interaction with GPCRs is enabled, yet not exclusively, by multiple protein-protein interaction modules such as Scr homology 3, PDZ domains (66), or other critical determinants, ie, phosphorylation sites for ␤-arrestin binding. Arrestin family With a family of only four proteins in mammals and the lack of any recognizable sequence motif known to mediate protein-protein interactions, it is remarkable that ␤-arrestins bind to an astounding several hundred GPCRs to orchestrate cell signaling (67, 68). The arrestin family comprises the two visual arrestins (arrestin-1 and arrestin-4) that are confined to retinal cones and rods, the ubiquitously expressed nonvisual arrestins, ␤-arrestin-1 and -2 (arrestin-2 and -3) and was recently extended with a new class of arrestins called ␣-arrestins (69). ␣-Arrestins represent a fairly new member of the large family of scaffolding proteins known to be involved in the regulation of GPCR signaling. The members of the ␣-arrestin family comprise arrestin domain-containing1–5 and TXNIP/thioredoxin binding protein-2 (69). So far, ␣-arrestins have been shown to be involved only in the regulation of ␤2AR ubiquitylation. Two possible models emerged from contradictory data; hence, the underlying mechanisms remains to be elucidated (69 –72). On the other hand, the role of ␤-arrestins in the regulation of GPCR signaling has been well documented. The selectivity of ␤-arrestins for GRK phosphorylated GPCRs is considered to be regulated by two sensors for receptor activation and phosphorylation in arrestins that engage simultaneously (73– 75). However, evidence suggests that even though many GPCRs must be phosphorylated to bind ␤-arrestins with high affinity, phosphorylation-independent ␤-arrestin binding is being reported for a growing number of GPCRs (76). Of note, it has been hypothesized that arrestins can form ␣-arrestin-␤-arrestin heterodimers (69, 71). This may represent a potential new mechanism in, but also adds a new level of complexity to, the regulation of GPCR signaling. There is now a perpetually growing list of nonreceptor ␤-arrestin-interaction partners that have been discovered over the past years, which revolutionized the role of ␤-arrestin within the cell (reviewed in references 77 and 78). Many of the identified ␤-arrestin-interacting proteins are themselves signaling molecules and include, but are not

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

818

Walther and Ferguson

Table 1.

Endocrine GPCR Interacting Proteins

Mol Endocrinol, June 2015, 29(6):814 – 830

Partial List of GPCR/Non-PDZ Scaffold Interaction Involved in GPCR Signaling

Scaffold Protein AKAP79 AKAP250 AKAP5 Jak2 14-3-3

GPCR ␤2AR ␤2AR ␤1AR GPR30 AT1R FSHR PAR4 ␣2AR GPR15

Effect Phosphorylation 2 Association with PKA 1 Receptor recycling 1 cAMP accumulation 2 STAT phosphorylation 1 cAMP accumulation 2 Plasma membrane transport and signaling 1 Ras/Raf signaling 1 Stability and cell surface expression 1

Reference 142–144 143–145 96, 146 –147 148 149, 155–157 159 162 164, 165 163

Abbreviation: FSHR, FSH receptor.

limited to the following: cAMP PDE4 (79), Yes (80), cJun N-terminal kinase 3, ERK1 and ERK2, cRaf1 (13, 81– 84), phosphatidylinositol-3-kinase, Akt, and protein phosphatase 2A (77). The regulation of MAPK cascades by GPCRs involves a variety of mechanisms, including second messenger-dependent pathways, transactivation of classical receptor tyrosine kinases, and ␤-arrestin-scaffolded MAPK kinase complexes (78). ERK1/2 signaling represents the best-studied G protein-independent and ␤-arrestin-dependent signaling pathway (81– 83, 85). The relative stability of the receptor-␤-arrestin complex regulates the duration of ␤-arrestin-dependent ERK activation and also influences the subcellular localization of ERK1/2 signals (86, 87). For example, in cell lines transfected with recombinant the PTH receptor (PTH1R) stimulates ERK1/2 via multiple mechanisms. PTH activation of the PTH1R results in ERK1/2 activation via a rapid and early G protein-dependent pathway meditated by PKA and PKC and a delayed and prolong G protein-independent pathway mediated by ␤-arrestins in human embryonic kidnay-293 cells (88). In nondifferentiated expression systems, the activation of ␤-arrestin-dependent and G protein-independent ERK1/2 signaling is also regulated by inverse agonists, indicating that multiple GPCR activation states contribute to G protein-dependent vs G protein-independent pathways (89). However, it is of note that differentiated cells exhibit a different mechanism of ERK1/2 activation independent of arrestin (90). PDZ proteins The human genome encodes for more than a hundred PDZ domain-containing proteins (PDZ proteins) (65). PDZ proteins consist of multiple PDZ domains that facilitate the binding to specific sequences within the intracellular domains of GPCRs, preferably but not exclusively, the C terminus. Such PDZ binding motifs comprise three to four amino acids and can be divided into three classes: class I (x-S/T-x-V/I/L), class II(-␾-x-␾), and class III(␺-

x-␾) (␾-hydrophobic amino acid, ␺-acidic amino acid) (66, 91). The GPCR PDZ binding motif allows for PDZ proteins to associate and scaffold multiprotein complexes to modulate GPCR function. PDZ proteins have been implicated in the regulation of virtually all receptor properties (16 –17, 92). However, this review focuses on their implications in GPCR signaling/desensitization (Table 2). Prototypical PDZ proteins that have been shown to interact with various GPCRs include the following: the multiple PDZ domain protein (MUPP-1), members of the membrane-associated guanylate kinase family, such as synapse-associated protein 97 (SAP97), postsynaptic-density protein 95 (PSD-95), synapse-associated protein 102, WW and PDZ domain-containing proteins (MAGIs), sorting nexin 27 (SNX27), and members of the Na⫹/H⫹ exchanger regulatory factor (NHERF) family like NHERF1– 4. The ␤1AR is by far the best-studied receptor when it comes to PDZ protein interactions and their implication in the regulation of receptor activity. ␤1AR is known to bind to various PDZ proteins such as cystic fibrosis transmembrane conductance regulator-associated ligand (CAL), PDZ protein interacting specifically with TC10 (PIST)/golgi-associated PDZ and coiled-coil domainscontaining protein (GOPC), PSD-95, MAGI-2, GAIP-interacting protein C terminus (GIPC), MAGI-3, SAP97, and SNX27 (93–100). Although the interaction with MAGI-3 terminates ␤1AR-mediated ERK1/2 activation with no effect on agonist-stimulated cAMP formation or agonist-mediated internalization, the strong binding of MAGI-2 promotes ␤1AR internalization (100). The association of the Golgi-associated PDZ protein CAL to the ␤1AR results in intracellular receptor retention (101). PSD-95, another PDZ protein, can compete with CAL for the binding to the ␤1AR, which allows for ␤1AR transport to the cell surface but also antagonizes ␤1AR endocytosis (95, 101).

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

doi: 10.1210/me.2015-1091

Table 2. GPCR mGluR1a mGluR5 ␤1AR

␤2AR ␣2AR LPA2 MT1R PTH1R

D1R D2R P2Y1R 5-HT2AR GPR30 SSTR5 CRFR1 LHR GHRHR M3R ␮-OR IP

press.endocrine.org/journal/mend

819

Nonexhaustive List of GPCR/PDZ Scaffold Interaction Involved in GPCR Signaling PDZ Protein PIST (CAL; GOPC) NHERF2 GIPC MAGI-3 SAP-97 PIST SNX27 MAGI-3 NHERF1 SNX27 Spinophilin

Effect ERK activation 2 Ca2⫹ signaling 1 ERK activation 2 ERK activation 2 PKA-mediated receptor phosphorylation 1 ERK activation 2 Receptor recycling 1 ERK activation 2 Plasma membrane recycling 1 Functional resensitization 1 Cell surface expression 1, phosphorylation 2, MAPK signaling 2, Ca2⫹ signaling MAGI-3 ERK activation 1, ␳ activation 1 NHERF2 Interaction with PLC␤3 1, IP3 signaling 1, ERK activation 1 MUPP-1 Coupling with Gi protein 1, inhibition of adenylyl cyclase activity 1 NHERF1 (EBP50) Coupling to and activation of Gq protein 1, interaction with ␤-arrestin-2 2, desensitization 2, cAMP signaling 1 NHERF2 Interaction with and activation of PLC␤ 1, adenylyl cyclase activity through stimulation of Gi/o proteins PSD-95 cAMP signaling 2 GIPC G protein coupling2 Spinophilin Signaling 1 NHERF2 Interaction with PLC␤ 1, agonist-induced Ca2⫹ signaling 1 PDS-95 IP3 signaling 1 PDZK1/NHERF3 IP3 signaling 1 SAP97 IP3 signaling 1, ERK activation 1 SAP97/PSD-95 cAMP signaling 2 PDZK1/NHERF3 Interaction with PLC␤3 1 PIST (CAL, GOPC) Trafficking, cell surface expression 2 SNX27 Receptor recycling 1 PDZK1/NHERF3 ERK activation 1 SAP97 ERK activation 1 GIPC ERK activation 1 PICK1 Cell surface expression 1, cAMP signaling 2 Spinophilin Signaling 2 Spinophilin Endocytosis 1 PDZK1/NHERF4 Cell surface expression 1, cAMP signaling 1

Reference 210 211 212 97 96 104 98 213 93, 94 106, 107 132–134, 136, 137 214 119, 120 109 122, 215, 216 121, 215 217 115, 116 133 124 128 127 207 148 118 102 102 127 126 114 218 139 140 129

Abbreviations: D1R, D1 dopamine receptor; EBP50, ezrin-radixin-moesin-binding phosphoprotein-50; GHRHR, GHRH receptor; IP, I prostanoid receptor; LPA2, lysophosphatidic acid receptor 2; mGluR1/5, metabotropic glutamate receptor 1/5; P2Y1R, purinergic receptor type 1.

A similar phenomenon is reported for the somatostatin receptor (SSTR)-5, but in this case, NHERF released SSTR5 from trans-Golgi network (TGN) retention (102, 103). More recent studies analyzed these interactions in more detail and determined that CAL/PIST retains ␤1AR in the TGN and that CAL/PIST overexpression reduces the MAPK pathway by ␤1AR agonists. Upon agonist stimulation CAL/PIST relocates from the TGN to an endosomal compartment, thus contributing to ␤1AR sorting through anterograde and retrograde trafficking (104). A third PDZ protein involved in SSTR5 trafficking, particularly recycling, is SNX27. The functional resensitization of SSTR5 is shown to be regulated by SNX27 (102). Consequently, the interaction of SSTR5 with PIST/PDZ kinase PDZ domain-containing 1 (PDZK1)/SNX27 may contribute to an on-demand cell surface delivery of the receptor (105). This process might strongly influence the

ability of somatostatin to regulate insulin secretion from the pancreas or GH secretion from the pituitary. SNX27 also interacts with both the ␤1AR and ␤2AR (98, 106, 107). SNX27 mainly functions to receptor recycling back to the plasma membrane, thereby increasing the availability of functional receptors for agonist binding on the cell surface. Thus, recycling and resensitization represent additional critical steps in the regulation of GPCR signaling. Another SSTR subtype, SSTR3, was shown to interact with MUPP-1, which enables the incorporation of SSTR3 at tight junctions in a PDZ-dependent manner, and allows SSTR3 to control transepithelial conductance (108). Furthermore, MUPP-1 binds to the melatonin 1 receptor (MT1R) and abolishes G␣i-mediated signaling by interfering with MT1R-G␣i protein interaction (109). Another binding partner of MUPP-1 is the serotonin 2C receptor. This interaction facilitates receptor phosphory-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

820

Walther and Ferguson

Endocrine GPCR Interacting Proteins

Mol Endocrinol, June 2015, 29(6):814 – 830

lation and resensitization in a PDZ motif-dependent manner (110 –112). Signal specificity and diversity for distinct receptor families, such as GPCRs, can potentially be regulated by membrane trafficking. Receptors targeted to either lysosomal or recycling pathways generate different signaling profiles that result in either the permanent termination of G protein signal or resensitization of G protein signals. Thus is due to postendocytic sorting of receptors back to the cell surface, a process that is regulated by PDZ proteins (113). In this respect, GIPC represents a PDZ protein that plays an important role in regulating LH receptor (LHR) trafficking through the endocytic compartment as a consequence of its interaction with the C-terminal tail of the receptor. Specifically, GIPC routes the LHR to preearly endosomes for recycling rather than to early endosomes (114). This rerouting of the LHR from early endosomes to pre-early endosomes or vice versa appears to spatially reprogram MAPK signaling responses (114). In addition, dopamine receptor (D2R) and dopamine receptor 3 signaling is regulated by GIPC. For example, in neuroendocrine cells the D2R recruits GAIP [regulator of G protein signaling (RGS)-19], which is required for GIPC binding. Thus, GIPC might serve as a molecular adaptor between GPCRs and RGS proteins to facilitate the attenuation of D2R signaling (115, 116). NHERF proteins have been linked to the regulation of endocrine GPCR signaling and trafficking processes. One example comprises the formation of a ternary complex of the PTH1R, NHERF1, and ␤-arrestin-2 (117). This study suggests a model by which NHERF1 serves as an adaptor protein to recruit ␤-arrestin-2 into close proximity to the PTH1R, thus enabling ␤-arrestin-2 recruitment upon PTH1R activation. NHERFs also appear to be critical for the interaction with PLC-␤. For example, NHERF3 (PDZK1) enables SSTR5 coupling to PLC-␤3 (118), NHERF2 connects lipophosphatidic acid receptor 2 to the activation of PLC-␤3 (119, 120), and NHERF1 interactions with PTH1R link the receptor to PLC-␤ (121). This interaction causes a shift in the second-messenger response from adenylyl cyclase activation to the hydrolysis of phospholipids (121) and inhibits PTH1R desensitization (122). For the thromboxane A2 receptor, agonist stimulation promotes the interaction of NHERF1 (ezrin-radixinmoesin-binding phosphoprotein-50) with G␣q (123) and antagonizes thromboxane A2 receptor-mediated activation of G␣q and PLC-␤1. NHERFs not only connect receptors to PLC-␤ but also regulate calcium signaling by the purinergic receptor type 1. This is accomplished by the interaction of NHERF2 (124). NHERF1 is also required for efficient plasma membrane recycling of the ␤2AR (93,

125). Furthermore, GPCR/PDZ protein interactions play a crucial role in the regulation of inositol phosphate signaling (126 –128). This has been extensively studied for the 5-hydroxytryptamine receptor (5-HT2AR) with reports on multiple PDZ proteins, including NHERF3/ PDZK1, SAP97, and PSD-95, being involved in regulating G␣q signaling. The interplay between these three PDZ proteins and the 5-HT2AR is still unclear and remains to be elucidated. NHERF4/PDZK2 also interacts with the human I prostanoid receptor and agonist activation of the receptor leads to PKA- and PKC-mediated phosphorylation of NHERF4/PDZK2 (129). Spinophilin is another PDZ domain-containing protein that has been shown to interact with more than 30 diverse partner proteins, including GPCRs (130). However, most the GPCR-mediated interactions with spinophilin are mediated by a spinophilin receptor-interacting domain, which is described to associate with an interaction site within the third cytoplasmic loop of the ␣2-adrenergic receptor (␣2AR) and D2R (131–133). This interaction contributes to the cell surface stabilization of adrenergic receptors (134). Agonist occupancy of ␣2A/ 2BAR enhances spinophilin but also antagonizes GRK2 and ␤-arrestin binding to the receptors (135). Spinophilin blocks receptor phosphorylation by recognizing the complex of ␣2AR with G␤␥ and thereby competes with GRK2 for the binding to this complex. Consequently, spinophilin reduces receptor endocytosis and the stimulation of MAPKs (136). It has also been shown that spinophilin can actively regulate ␣1BAR function because it recruits RGS2 to the ␣1BAR/Gq signaling complex to form a heterotetrameric complex that is implicated in the regulation of Ca2⫹ signaling by ␣-agonists in cultured cells (137). In vivo studies with the ␣2AAR in spinophilin-null mice reveal the impact of spinophilin on ␣2AR-evoked cardiovascular responses (138). In mice lacking spinophilin, ␣2AR stimulation leads to an enhanced and prolonged hypotensive, bradycardic, and sedative-hypnotic responses. Eliminating spinophilin expression in native tissue also allowed for enhanced receptor/G protein coupling efficiency, which contributes to the sensitization of ␣2AR-mediated responses in vivo. Furthermore, spinophilin is a negative regulator of the M3-muscarinic acetylcholine receptor (m3 AchR)-mediated signaling because it is able to recruit RGS4 in an agonist-dependent manner to the m3R signaling complex (139) and promotes ␮-opioid receptor endocytosis (140). Non-PDZ proteins A-kinase anchor proteins (AKAPs) AKAPs were first identified as scaffolding proteins in 1999 (141). Because AKAPs coordinated the subcellular

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

doi: 10.1210/me.2015-1091

localization of second messenger-regulated enzymes such as PKA, PKC, and protein phosphatases, they orchestrate the regulation of these signaling pathways. AKAP79 and AKAP250 were identified as interaction partners for the ␤2AR (142–145). Specifically, AKAP79 promoted ␤2AR phosphorylation and downstream mitogenic signaling, whereas AKAP250 regulated ␤2AR desensitization via its association with PKA. AKAP79 was also found to interact with the ␤1AR/SAP97 protein complex (96), and SAP97 was previously shown to play a critical role in ␤1AR recycling (93, 94). This effect on ␤1AR desensitization was found to be dependent on the formation of a SAP97-AKAP79-PKA complex that was tethered by SAP97 to the type I PDZ motif of the ␤1AR (96, 146, 147). Recently another GPCR, GPR30, was added to the list of AKAP-interacting GPCRs. GPR30 was shown to form a PDZ motif-dependent plasma membrane complex with a membrane-associated guanylate kinase (likely SAP97 or PSD-95) and AKAP5, which led to the constitutive attenuation of cAMP production and retained the receptor at the plasma membrane (148). Janus kinase (Jak)-2 The Jak signal transducers and activators of the transcription (STAT) signaling pathway is typically activated by cytokine or growth factor receptors but not by GPCRs. However, stimulation of the AT1R has been found to activate not only G protein-dependent pathways but also the Jak/STAT signaling pathway (149 –155). The ability of the AT1R to regulate Jak/STAT signaling is dependent on a direct interaction of AT1R with the tyrosine kinase Jak2 (149, 155–157). The scaffolding of Jak2 on the AT1R was dependent on a distinct C-terminal tyrosine motif (YIPP) in the AT1R C terminus (156) that is required for Jak2 phosphorylation of the transcription factor STAT and the recruitment of STAT1 into a complex with the AT1R (149, 155). 14 –3-3 proteins Isoforms of 14 –3-3 proteins have been shown to play key roles in signal transduction pathways. 14 –3-3 proteins exhibit the characteristics of scaffolding proteins (158) and represent a potential mechanism by which GPCR-mediated signaling is modulated. Examples include the association of 14 –3-3 with the FSH receptor in a follitropin-dependent manner that results in a modest decrease of follitropin accumulation (159). Furthermore, 14 –3-3 proteins recognize and mask retinoid X receptor motifs to direct endoplasmic reticulum (ER)/Golgi export of multimeric proteins (160, 161). The protease-activated receptor (PAR)-4 encodes an ER retention motif and is usually retained in the ER (162). However, PAR4 evades

press.endocrine.org/journal/mend

821

ER retention by forming a heterodimer with PAR2, which allows interaction with 14 –3-3␨. Hence, the receptor is transported to the plasma membrane and exhibits substantially enhanced signaling properties. Similarly, phosphorylation-dependent 14 –3-3 binding to G protein-coupled receptor 15 (GPR15) has been shown to increase GPR15 stability and increased cell surface expression due to the suppression of the dibasic (retinoid X receptor) ER retention signal (163). The 14 –3-3␨ isoform also interacts with the ␣2AR and couples the ␣2AR to the activation of the Ras/Raf cascade via the G protein ␤␥-subunit (164, 165). Moreover, phosphorylated Raf functions to antagonize receptor-14 –3-3␨ interactions, indicating that the 14 –3-3 proteins interact with inactive ␣2ARs to facilitate the activation of the Ras/Raf cascade after the agonist binding. After the agonist activation, the ␤␥-subunit and the 14 –3-3 protein subsequently dissociate to enable Ras/ Raf signaling (164, 165). GPCRs in the endocrine system: implications in pathological states Major endocrine systems comprise the hypothalamicpituitary-thyroid axis, the hypothalamic-pituitary-gonadal axis, the hypothalamic-pituitary-adrenal (HPA) axis, the renin-angiotensin system, and energy homeostasis. Key players in these systems are the GPCRs that, upon hormone binding, mediate the downstream response in the target organ as well the GIPs that modulate and/or control downstream signaling profiles. Either inappropriate hormone release or response to signaling, usually caused by mutations, results in endocrine disorders (reviewed in references 3 and 166). A plethora of GPCRs and their associated interacting proteins have been implicated in endocrine diseases. Thus, it is impossible to address all of them within the scope of this review. Herein we will focus on a few selected examples, such as osteoporosis, cancer, diabetes, obesity stress, and anxiety because these diseases represent prevalent human health risks with hundreds of millions of people affected worldwide. Obesity and diabetes Obesity represents a critical predisposition to insulin resistance and the subsequent development of type 2 diabetes mellitus (T2DM). Targets for therapeutic interventions include several members of the GPCR superfamily and their ligands as well as various GPCR-scaffolding proteins (167). Ghrelin, with its critical role in energy homeostasis, and its receptor, the GH secretagogue receptor [ghrelin receptor (GHSR1)] are of great significance for the treatment of obesity (168). GHSR1 signals through G␣q/11, G␣i/o, and G␣12/13 as well as ␤-arrestinbased scaffolds. GHSR1 enhances appetite, GH release,

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

822

Walther and Ferguson

Endocrine GPCR Interacting Proteins

Mol Endocrinol, June 2015, 29(6):814 – 830

and modulation of reward and cognitive functions. Mutagenesis studies revealed different receptor conformations that support signaling bias at the GHSR1 and could prove beneficial for the development of future antiobesity drugs (168, 169). The metabolic syndrome is a condition that can lead to diabetes. Normal blood glucose levels in a healthy individual depend on the regulation of insulin secretion from pancreatic ␤-cells and a malfunction in insulin secretion causes T2DM (170). One GPCR involved in the maintenance of proper blood glucose levels is, among many others (171), the m3 AChR. Novel strategies in the treatment of T2DM include the enhancement of ␤-cell m3 AChR signaling, which involves G protein-dependent and G protein-independent such as ␤-arrestin-dependent signaling pathways (172). Studies with transgenic mice that expressed an m3 AChR-based (designer receptor exclusively activated by designer drugs, a receptor unable to bind the endogenous muscarinic receptor ligand ACh but can be efficiently activated by clozapine-N-oxide, an otherwise pharmacologically inert compound) (173–175). Designer receptors exclusively activated by designer drugs provide great potential in understanding the contribution of GPCR signaling, particularly biased signaling and scaffolding proteins as critical determinants of disease pathology. Because it has been discovered that m3 receptors can potentiate insulin release through a ␤-arrestin-dependent mechanism, targeting muscarinic receptor signaling with biased agonists selective for the ␤-arrestindependent pathways could prove therapeutically beneficial. Importantly, reduced side effects due to minimized G protein-dependent calcium signaling would be expected. Consequently, biased ligands that direct signaling of the m3 AChR via ␤-arrestin signaling would be expected to potentiate the insulin release at ␤-islets (176). In addition to ␤-arrestin, spinophilin represents another scaffolding protein capable of regulating m3 AChR signaling (139, 177). It has been identified as a negative regulator due to its selective inhibition of m3 AChR-mediated signaling and insulin release, which is mediated by RGS4 (178). Another GPCR expressed in pancreatic ␤-cells is the SSTR5. The availability of SSTR5 at the plasma membrane in these cells has been shown to be regulated by the scaffolding protein PIST (179). Accordingly, PIST plays a critical role in the SSTR5-mediated regulation of insulin secretion. In summary, scaffolding proteins, may serve as potential targets for therapeutic intervention aimed to modulate ␤-cell m3 AChR or SSTR5 function for the treatment of T2DM (139, 177–179).

Osteoporosis Reduced bone mineral density in elderly people leads to increased susceptibility to fracture, an age-related metabolic disorder, called osteoporosis (180). One of the two GPCRs involved in calcium homeostasis is the PTH1R, mainly expressed in kidney and bone. PTH signaling exerts multifaceted effects on bone (181, 182). It simultaneously promotes new bone formation by osteoblasts and bone loss through osteoclastic bone resorption (183). The effect of PTH on bone is complex and critically regulated by the kinetics of PTH1R activation because anabolic and catabolic effects of PTH are coupled. Hence, the PTH1R demonstrates the immense diversity of the signaling mechanisms that can originate from a single GPCR (182, 184). Specifically, the PTH1R represents an important target for the treatment of osteoporosis by using novel therapeutic strategies that selectively bias the activation of one specific signaling pathway over another. Thus, targeting exclusively the ␤-arrestin pathway will allow for selective targeting of anabolic bone formation yet at the same time exclude the catabolic/calcitrophic effects of PTH1R activation, such as bone resorption (184, 185). In conclusion, the PTH-␤-arrestin signaling pathway may represent a novel pharmaceutical opportunity in the battle against osteoporosis. Another regulatory component of PTH1R signaling is its association with the PDZ proteins NHERF1 and NHERF2 (121). NHERF1/2 association influences the specificity of G protein and/or ␤-arrestin coupling. Although NHERF1 binding to PTH1R allows for Gq and PLC activation without affecting its ability to stimulate Gs and adenylyl cyclase, NHERF2 enables coupling to Gi and Gq but prevents coupling to Gs. Consequently, NHERF2 enables the PTH1R to switch from activating AC to PLC (121). In summary, NHERF-mediated signaling provides a mechanism by which cells regulate responses to PTH in a tissue-specific manner but may also be a mechanism by which the cells specify sites of second messenger generation. Endocrine tumors GPCRs and their signaling networks have recently emerged as crucial players in aberrant tumor growth and metastasis. In fact, most gain-of-function mutations in GPCRs and the G␣ subunit of G proteins were identified in endocrine tumors, which cause uncontrolled hormone secretion (reviewed in reference 186). One prototypical example is the TSHR, found in thyroid carcinomas and almost all thyroid adenomas. A characteristic of these active TSHR mutants is a constant G␣s-dependent activation of adenylyl cyclase and G␤␥-dependent stimulation of phosphatidylinositol-3-kinase and MAPKs in thyrocytes (187). Because GPCRs regulate second messengers and their down-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

doi: 10.1210/me.2015-1091

stream targets, they can control cell survival, growth, and migration by activating MAPK cascades. It is appreciated that such MAPKs are critical for cell proliferation and metastasis. Consequently, their dysregulation is closely linked to human malignancies (188). In addition to the activation of second-messenger signals, GPCR-scaffolded proteins such as ␤-arrestin and PDZ proteins may represent potential targets for cancer treatment. ␤-Arrestins are involved in several cancer-related signal networks via multiple different receptor subtypes such as the endogenous prostanoid (EP)1– 4 receptors, endothelin type A receptor, lysophosphatidic acid (LPA) receptor, and ␤2AR (189 –192). The EP4 receptors have been demonstrated to mediate prostaglandin-induced A549 lung cancer cell migration (190). Prostaglandin E2 (PGE2) stimulation enhances c-Scr activation, which suggests that blockage of c-Scr activity may be a target for the inhibition of PGE2-mediated lung cancer cell migration. Direct EP4 effectors of migration include ␤-arrestins and heterotrimeric Gs proteins. Notably, PGE2-mediated c-Scr activation and A549 cell migration is abolished when ␤-arrestin was knocked down. Consequently, the EP4-␤arrestin1 signalosome represents a promising drug target for the treatment of lung cancer. Specifically, selective EP4 antagonists may prove beneficial (190). Furthermore, EP receptors are also involved in the transactivation of epithelial growth factor receptor, which is mediated by PGE2-stimulated EP2 and EP4 receptors. EP receptors are implicated in mouse skin papilloma development (189) and colorectal carcinoma cell migration and metastasis (193), which not only involves both ␤-arrestin1 and c-Src. Likewise, ␤-arrestins mediate endothelin type A receptor-stimulated epithelial growth factor receptor transactivation via c-Src, as well as effects on ␤-catenin, in ovarian cancer cells (192). LPA receptors represent yet another example for ␤-arrestin-mediated signaling in cancer cells (191). In a recent study, Li et al (191) have determined a novel role for ␤-arrestin/Ral signaling in mediating LPA-induced breast cancer cell metastasis. Prostate cancer cell progression has been shown to be induced by ␤-arrestin2-mediated ␤2AR signaling (194). Herein ␤2AR activation promoted increasing levels of cAMP and ERK1/2 activation, resulting in enhanced cell proliferation and migration in prostate cancer cells. Furthermore, ␤2AR activation augmented the formation of a ␤-arrestin2/c-Scr complex, which enables the use of a c-Scr inhibitor as a potential therapeutic tool to block the aforementioned complex formation as well as subsequent cell proliferation (194). However, ␤-arrestins not only activate proliferative signaling pathways but also engage in various pathways that suppress tumor growth and metastasis. This highlights their important therapeutic potential for cancer therapy (195). The androgen-dependent prostate cancer cells represent

press.endocrine.org/journal/mend

823

an example in which ␤-arrestin2 recruits the E3 ligase mouse double minute-2, which subsequently leads to receptor ubiquitylation and down-regulation (196, 197). PDZ proteins have also been closely connected to human malignancies (198, 199). Spinophilin expression has been shown to determine cellular growth in colorectal cancer (198). However, little, if anything, is known about GPCRs and their interactions with spinophilin in colon cancer growth. Nevertheless, because various GPCRs such as EP2, EP4, LPA1, endothelin, and PAR1 receptors have been implicated in colon cancer (186) and spinophilin is known to interact with them, it is likely that spinophilin interactions may be important for the antagonism of cancer cell growth (132–134, 136, 137, 139). Another PDZ protein that is linked to cancer is NHERF1 (200, 201). Mutations in NHERF1 have also been associated with breast cancer (200), and NHERF1 is downregulated during the development of colon cancer (201). In pancreatic cancers, the PDZ protein GIPC1/synectin is often overexpressed (202). Importantly, blocking synectin’s PDZ-binding potential in a mouse model of pancreatic tumor invasion results in the inhibition of tumor growth (203). Although the roles of the PDZ interactions of NHERF1, GIPC, and their cognate GPCRs have yet to be determined, it is easy to envisage that such GPCR/PDZ protein interactions are likely of importance in cancer growth and development. Our rapidly growing knowledge of the expression status and functional activity of GPCRs along with their associated interacting proteins in various cancer types and their exceptional drugability make them ideal candidates for cancer prevention and treatment in the near future (186). Stress and anxiety Corticotropin-releasing factor (CRF) is a neuropeptide that regulates the physiological response to stress by initiating the HPA axis stress response as well as regulating prefrontal cortical stress responses. Consequently, CRF is also implicated in the manifestation of depression (204). Its involvement in the regulation of stress and anxiety presents a functional link between endocrine signaling and neurobehavioral responses (205). Of note, serotonergic dysfunction is also well established in mood disorders because the serotonergic system is regulated by the HPA axis. CRF and 5-hydroxytryptamine exert their biological effects by activating a specific group of GPCRs, the CRF receptors (CRFRs) and 5-hydroxytryptamine receptors. These receptors represent promising targets for the treatment of mood disorders (206). Recently it has been reported that the preactivation of CRFR1 sensitizes of 5-HT2AR inositol phosphate signaling by a mechanism that requires intact receptor endocy-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

824

Walther and Ferguson

Endocrine GPCR Interacting Proteins

Mol Endocrinol, June 2015, 29(6):814 – 830

tosis, recycling, and increased 5-HT2AR expression at the cell surface (207). Both CRFR1 and 5-HT2AR encode a PDZ binding motif at their carboxyl-termini, and deleting of either PDZ motif resulted in a loss of heterologous sensititization of 5-HT2AR signaling that is associated with the inability of the 5-HT2AR to be mobilized to the cell surface. Even though multiple PDZ proteins (PDZK1, PSD-95, SAP97) have been identified as CRFR1 and/or 5-HT2AR interacting partners, the precise PDZ protein involved in the CRFR1/5-HT2AR cross talk remains to be determined (127, 128, 208). Nevertheless, the interaction of PDZ proteins with GPCRs represents a novel regulatory mechanism for functional receptor cross talk and highlights the importance of PDZ proteins in the regulation of GPCR function. Importantly, the down-regulation of 5-HT2AR-mediated signaling is a critical process achieved by second generation atypical antipsychotics (209). Thus, the selective prevention of a PDZ protein/5HT2AR interaction may result in a down-regulation of 5-HT2AR surface expression and subsequently decreased 5-HT2AR signaling. This may provide a novel therapeutic approach for the treatment of mood disorders.

whether one binding partner modulates the association/ dissociation of other proteins. Over the past few decades, it has become increasingly evident that GPCR activity is critically regulated by a plethora of interacting proteins, including proteins that are required for the scaffolding of macromolecular complexes. The formation of these complexes is not only essential for regulating the normal cell biology of GPCRs but are also important for the modulation of the normal and pathophysiological responses that contribute to the regulation of human health. A growing number of human diseases have been found to result from a dysfunction of such GPCR-scaffolding protein interactions. Therefore, delineating the mechanism of GPCR dysfunction represents an important strategy to either therapeutically restore or inhibit receptor activity. The current decade of GPCR research has revolutionized our understanding of GPCR signaling and provides new avenues for therapeutic molecule intervention in endocrine disease processes.

Conclusion The role of agonist stimulation for the association of a GPCR with a scaffold protein still remains poorly understood because it likely plays a critical role for the regulation of receptor signaling. Agonist stimulation causes a major change in GPCR conformation which leads to a significantly enhanced association of G proteins with the receptor’s intracellular domains. However, although some receptor/scaffold interactions have been found to be enhanced by agonist stimulation (␤2AR/NHERF, AT1R/ Jak), others do not require agonist binding (␤2AR/ AKAP). The underlying mechanism contributing to these differences exist, and the physiological significance of these interactions has yet to be fully explored and will require the transgenic overexpression and knockout of these proteins. Because scaffolding proteins have the ability to enhance protein-protein interactions yet do not possess an intrinsic activity of their own, studying their activity is nearly impossible in a nonphysiological setting. Furthermore, it still remains very challenging to fully understand cellular functions of scaffolding proteins. This is due to their ability to bind multiple proteins and generate very complex signal transduction complexes. In general, studying the interaction of more than two proteins can be extremely complicated. However, future research aimed at understanding how various binding partner bind simultaneously to a scaffold is essential. If these proteins do not bind simultaneously, it will be particularly interesting to understand both the order and kinetics of binding and

Address all correspondence and requests for reprints to: Stephen S. G. Ferguson, PhD, Department of Physiology and Pharmacology Medical Sciences, The University of Western Ontario, Building Room 216, Ontario Canada N6A 5C1. E-mail: [email protected]. Review criteria included the following: PubMed was searched using the following terms: GPCR and signaling, GPCR and scaffolding protein, arrestin and GPCR and signaling, and PDZ protein and GPCR. Original articles, reviews, and their reference lists were considered. Articles published between 1991 and 2015 were included. This work was supported by Canadian Institutes of Health Research Grant MOP-62738 (to S.S.G.F.), and S.S.G.F. is a Career Investigator of the Heart and Stroke Foundation of Ontario, Canada. C.W. is the recipient of a Bell Mental Health Research Training Award from the Brain Canada Foundation and Bell Canada. Disclosure Summary: The authors having nothing to disclose.

Acknowledgments

References 1. Jassal B, Jupe S, Caudy M, et al. The systematic annotation of the three main GPCR families in Reactome. Database (Oxford). 2010; 2010:baq018. 2. Luttrell LM. Reviews in molecular biology and biotechnology: transmembrane signaling by G protein-coupled receptors. Mol Biotechnol. 2008;39:239 –264. 3. Schoneberg T, Schulz A, Biebermann H, Hermsdorf T, Rompler H, Sangkuhl K. Mutant G-protein-coupled receptors as a cause of human diseases. Pharmacol Ther. 2004;104:173–206. 4. Fredriksson R, Lagerstrom MC, Lundin LG, Schioth HB. The

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

doi: 10.1210/me.2015-1091

5.

6.

7.

8.

9. 10. 11.

12.

13.

14.

15. 16.

17.

18.

19.

20. 21.

22.

23.

24.

25.

26.

27.

G-protein-coupled receptors in the human genome form five main families. Phylogenetic analysis, paralogon groups, and fingerprints. Mol Pharmacol. 2003;63:1256 –1272. Kamal M, Jockers R. Biological significance of GPCR heteromerization in the neuro-endocrine system. Front Endocrinol (Lausanne). 2011;2:2. Takeda S, Kadowaki S, Haga T, Takaesu H, Mitaku S. Identification of G protein-coupled receptor genes from the human genome sequence. FEBS Lett. 2002;520:97–101. Thompson MD, Cole DE, Jose PA. Pharmacogenomics of G protein-coupled receptor signaling: insights from health and disease. Methods Mol Biol. 2008;448:77–107. Lundstrom K. An overview on GPCRs and drug discovery: structure-based drug design and structural biology on GPCRs. Methods Mol Biol. 2009;552:51– 66. Vassart G, Costagliola S. G protein-coupled receptors: mutations and endocrine diseases. Nat Rev Endocrinol. 2011;7:362–372. Neer EJ. Heterotrimeric G proteins: organizers of transmembrane signals. Cell. 1995;80:249 –257. Ferguson SS. Evolving concepts in G protein-coupled receptor endocytosis: the role in receptor desensitization and signaling. Pharmacol Rev. 2001;53:1–24. Hall RA, Premont RT, Lefkowitz RJ. Heptahelical receptor signaling: beyond the G protein paradigm. J Cell Biol. 1999;145: 927–932. Luttrell LM, Ferguson SS, Daaka Y, et al. ␤-Arrestin-dependent formation of ␤2 adrenergic receptor-Src protein kinase complexes. Science. 1999;283:655– 661. Rajagopal K, Lefkowitz RJ, Rockman HA. When 7 transmembrane receptors are not G protein-coupled receptors. J Clin Invest. 2005;115:2971–2974. Bockaert J, Fagni L, Dumuis A, Marin P. GPCR interacting proteins (GIP). Pharmacol Ther. 2004;103:203–221. Bockaert J, Perroy J, Becamel C, Marin P, Fagni L. GPCR interacting proteins (GIPs) in the nervous system: roles in physiology and pathologies. Annu Rev Pharmacol Toxicol. 2010;50:89 –109. Magalhaes AC, Dunn H, Ferguson SS. Regulation of GPCR activity, trafficking and localization by GPCR-interacting proteins. Br J Pharmacol. 2012;165:1717–1736. Maurice P, Guillaume JL, Benleulmi-Chaachoua A, Daulat AM, Kamal M, Jockers R. GPCR-interacting proteins, major players of GPCR function. Adv Pharmacol. 2011;62:349 –380. Dunn HA, Ferguson SS. PDZ protein regulation of GPCR trafficking and signaling pathways [published online March 25, 2015]. Mol Pharmacol. doi:10.1124/mol.115.098509. Hall RA, Lefkowitz RJ. Regulation of G protein-coupled receptor signaling by scaffold proteins. Circ Res. 2002;91:672– 680. Ferguson SS, Downey WE 3rd, Colapietro AM, Barak LS, Menard L, Caron MG. Role of ␤-arrestin in mediating agonist-promoted G protein-coupled receptor internalization. Science. 1996;271:363– 366. Freedman NJ, Lefkowitz RJ. Desensitization of G protein-coupled receptors. Recent Prog Horm Res. 1996;51:319 –351; discussion 352–353. Goodman OB Jr, Krupnick JG, Santini F, et al. ␤-Arrestin acts as a clathrin adaptor in endocytosis of the ␤2-adrenergic receptor. Nature. 1996;383:447– 450. Jean-Alphonse F, Hanyaloglu AC. Regulation of GPCR signal networks via membrane trafficking. Mol Cell Endocrinol. 2011; 331:205–214. Laporte SA, Oakley RH, Zhang J, et al. The ␤2-adrenergic receptor/␤arrestin complex recruits the clathrin adaptor AP-2 during endocytosis. Proc Natl Acad Sci USA. 1999;96:3712–3717. Lefkowitz RJ. G protein-coupled receptors. III. New roles for receptor kinases and ␤-arrestins in receptor signaling and desensitization. J Biol Chem. 1998;273:18677–18680. Walther C, Ferguson SS. Arrestins: role in the desensitization,

press.endocrine.org/journal/mend

28.

29.

30. 31. 32. 33. 34.

35.

36.

37.

38.

39.

40.

41. 42.

43.

44.

45.

46.

47.

48.

49.

825

sequestration, and vesicular trafficking of G protein-coupled receptors. Prog Mol Biol Transl Sci. 2013;118:93–113. Rosenbaum DM, Rasmussen SG, Kobilka BK. The structure and function of G-protein-coupled receptors. Nature. 2009;459:356 – 363. Lambright DG, Sondek J, Bohm A, Skiba NP, Hamm HE, Sigler PB. The 2.0 A crystal structure of a heterotrimeric G protein. Nature. 1996;379:311–319. Hamm HE, Gilchrist A. Heterotrimeric G proteins. Curr Opin Cell Biol. 1996;8:189 –196. Milligan G, Kostenis E. Heterotrimeric G-proteins: a short history. Br J Pharmacol. 2006;147(suppl 1):S46 –S55. Offermanns S. G-proteins as transducers in transmembrane signalling. Prog Biophys Mol Biol. 2003;83:101–130. Gether U, Kobilka BK. G protein-coupled receptors. II. Mechanism of agonist activation. J Biol Chem. 1998;273:17979 –17982. Galandrin S, Bouvier M. Distinct signaling profiles of ␤1 and ␤2 adrenergic receptor ligands toward adenylyl cyclase and mitogenactivated protein kinase reveals the pluridimensionality of efficacy. Mol Pharmacol. 2006;70:1575–1584. Jin LQ, Wang HY, Friedman E. Stimulated D(1) dopamine receptors couple to multiple G␣ proteins in different brain regions. J Neurochem. 2001;78:981–990. Laugwitz KL, Allgeier A, Offermanns S, et al. The human thyrotropin receptor: a heptahelical receptor capable of stimulating members of all four G protein families. Proc Natl Acad Sci USA. 1996;93:116 –120. Luttrell LM. Minireview: more than just a hammer: ligand “bias” and pharmaceutical discovery. Mol Endocrinol. 2014;28:281– 294. Offermanns S, Wieland T, Homann D, et al. Transfected muscarinic acetylcholine receptors selectively couple to Gi-type G proteins and Gq/11. Mol Pharmacol. 1994;45:890 – 898. Luttrell LM, Gesty-Palmer D. Beyond desensitization: physiological relevance of arrestin-dependent signaling. Pharmacol Rev. 2010;62:305–330. Irannejad R, Tomshine JC, Tomshine JR, et al. Conformational biosensors reveal GPCR signalling from endosomes. Nature. 2013;495:534 –538. Irannejad R, von Zastrow M. GPCR signaling along the endocytic pathway. Curr Opin Cell Biol. 2014;27:109 –116. Tsvetanova NG, Irannejad R, von Zastrow M. G Protein-coupled receptor (GPCR) signaling via heterotrimeric G proteins from endosomes. J Biol Chem. 2015;290:6689 – 6696. Vilardaga JP, Jean-Alphonse FG, Gardella TJ. Endosomal generation of cAMP in GPCR signaling. Nat Chem Biol. 2014;10:700 – 706. Calebiro D, Nikolaev VO, Gagliani MC, et al. Persistent cAMPsignals triggered by internalized G-protein-coupled receptors. PLoS Biol. 2009;7:e1000172. Ferrandon S, Feinstein TN, Castro M, et al. Sustained cyclic AMP production by parathyroid hormone receptor endocytosis. Nat Chem Biol. 2009;5:734 –742. Wehbi VL, Stevenson HP, Feinstein TN, Calero G, Romero G, Vilardaga JP. Noncanonical GPCR signaling arising from a PTH receptor-arrestin-G␤␥ complex. Proc Natl Acad Sci USA. 2013; 110:1530 –1535. Feinstein TN, Yui N, Webber MJ, et al. Noncanonical control of vasopressin receptor type 2 signaling by retromer and arrestin. J Biol Chem. 2013;288:27849 –27860. Barauna VG, Magalhaes FC, Campos LC, et al. Shear stress-induced Ang II AT1 receptor activation: G-protein dependent and independent mechanisms. Biochem Biophys Res Commun. 2013; 434:647– 652. Seta K, Nanamori M, Modrall JG, Neubig RR, Sadoshima J. AT1 receptor mutant lacking heterotrimeric G protein coupling acti-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

826

50.

51.

52.

53.

54.

55. 56.

57.

58.

59.

60.

61.

62.

63.

64. 65. 66. 67.

68.

69. 70.

71.

Walther and Ferguson

Endocrine GPCR Interacting Proteins

Mol Endocrinol, June 2015, 29(6):814 – 830

vates the Src-Ras-ERK pathway without nuclear translocation of ERKs. J Biol Chem. 2002;277:9268 –9277. Zhai P, Yamamoto M, Galeotti J, et al. Cardiac-specific overexpression of AT1 receptor mutant lacking G␣q/G␣i coupling causes hypertrophy and bradycardia in transgenic mice. J Clin Invest. 2005;115:3045–3056. Rajagopal S, Kim J, Ahn S, et al. ␤-Arrestin- but not G proteinmediated signaling by the “decoy” receptor CXCR7. Proc Natl Acad Sci USA. 2010;107:628 – 632. Christopoulos A. Advances in G protein-coupled receptor allostery: from function to structure. Mol Pharmacol. 2014;86:463– 478. Neubig RR, Spedding M, Kenakin T, Christopoulos A. International Union of Pharmacology Committee on Receptor Nomenclature and Drug Classification. XXXVIII. Update on terms and symbols in quantitative pharmacology. Pharmacol Rev. 2003;55: 597– 606. Christopoulos A. Allosteric binding sites on cell-surface receptors: novel targets for drug discovery. Nat Rev Drug Discov. 2002;1: 198 –210. Christopoulos A, Kenakin T. G protein-coupled receptor allosterism and complexing. Pharmacol Rev. 2002;54:323–374. Wootten D, Christopoulos A, Sexton PM. Emerging paradigms in GPCR allostery: implications for drug discovery. Nat Rev Drug Discov. 2013;12:630 – 644. Kenakin T, Miller LJ. Seven transmembrane receptors as shapeshifting proteins: the impact of allosteric modulation and functional selectivity on new drug discovery. Pharmacol Rev. 2010; 62:265–304. Luttrell LM, Kenakin TP. Refining efficacy: allosterism and bias in G protein-coupled receptor signaling. Methods Mol Biol. 2011; 756:3–35. Kenakin T, Christopoulos A. Signalling bias in new drug discovery: detection, quantification and therapeutic impact. Nat Rev Drug Discov. 2013;12:205–216. Reiter E, Ahn S, Shukla AK, Lefkowitz RJ. Molecular mechanism of ␤-arrestin-biased agonism at seven-transmembrane receptors. Annu Rev Pharmacol Toxicol. 2012;52:179 –197. Shonberg J, Lopez L, Scammells PJ, Christopoulos A, Capuano B, Lane JR. Biased agonism at G protein-coupled receptors: the promise and the challenges—a medicinal chemistry perspective. Med Res Rev. 2014;34:1286 –1330. Stallaert W, Christopoulos A, Bouvier M. Ligand functional selectivity and quantitative pharmacology at G protein-coupled receptors. Expert Opin Drug Discov. 2011;6:811– 825. Urban JD, Clarke WP, von Zastrow M, et al. Functional selectivity and classical concepts of quantitative pharmacology. J Pharmacol Exp Ther. 2007;320:1–13. Shukla AK. Biasing GPCR signaling from inside. Sci Signal. 2014; 7:pe3. Garbett D, Bretscher A. The surprising dynamics of scaffolding proteins. Mol Biol Cell. 2014;25:2315–2319. Sheng M, Sala C. PDZ domains and the organization of supramolecular complexes. Annu Rev Neurosci. 2001;24:1–29. Shukla AK, Xiao K, Lefkowitz RJ. Emerging paradigms of ␤-arrestin-dependent seven transmembrane receptor signaling. Trends Biochem Sci. 2011;36:457– 469. Gurevich VV, Gurevich EV. Extensive shape shifting underlies functional versatility of arrestins. Curr Opin Cell Biol. 2014;27: 1–9. Puca L, Brou C. ␣-Arrestins—new players in Notch and GPCR signaling pathways in mammals. J Cell Sci. 2014;127:1359 –1367. Nabhan JF, Pan H, Lu Q. Arrestin domain-containing protein 3 recruits the NEDD4 E3 ligase to mediate ubiquitination of the ␤2-adrenergic receptor. EMBO Rep. 2010;11:605– 611. Shea FF, Rowell JL, Li Y, Chang TH, Alvarez CE. Mammalian ␣ arrestins link activated seven transmembrane receptors to Nedd4

family e3 ubiquitin ligases and interact with ␤ arrestins. PLoS One. 2012;7:e50557. Han SO, Kommaddi RP, Shenoy SK. Distinct roles for ␤-arrestin2 and arrestin-domain-containing proteins in ␤2 adrenergic receptor trafficking. EMBO Rep. 2013;14:164 –171. Gurevich VV, Benovic JL. Visual arrestin interaction with rhodopsin. Sequential multisite binding ensures strict selectivity toward light-activated phosphorylated rhodopsin. J Biol Chem. 1993; 268:11628 –11638. Gurevich VV, Gurevich EV. The molecular acrobatics of arrestin activation. Trends Pharmacol Sci. 2004;25:105–111. Palczewski K, Buczylko J, Kaplan MW, Polans AS, Crabb JW. Mechanism of rhodopsin kinase activation. J Biol Chem. 1991; 266:12949 –12955. Gurevich VV, Gurevich EV. The structural basis of arrestin-mediated regulation of G-protein-coupled receptors. Pharmacol Ther. 2006;110:465–502. DeFea KA. ␤-arrestins as regulators of signal termination and transduction: how do they determine what to scaffold? Cell Signal. 2011;23:621– 629. Lefkowitz RJ, Whalen EJ. ␤-Arrestins: traffic cops of cell signaling. Curr Opin Cell Biol. 2004;16:162–168. Perry SJ, Baillie GS, Kohout TA, et al. Targeting of cyclic AMP degradation to ␤2-adrenergic receptors by ␤-arrestins. Science. 2002;298:834 – 836. Imamura T, Huang J, Dalle S, et al. ␤-Arrestin-mediated recruitment of the Src family kinase Yes mediates endothelin-1-stimulated glucose transport. J Biol Chem. 2001;276:43663– 43667. DeFea KA, Vaughn ZD, O’Bryan EM, Nishijima D, Dery O, Bunnett NW. The proliferative and antiapoptotic effects of substance P are facilitated by formation of a ␤-arrestin-dependent scaffolding complex. Proc Natl Acad Sci USA. 2000;97:11086 –11091. DeFea KA, Zalevsky J, Thoma MS, Dery O, Mullins RD, Bunnett NW. ␤-Arrestin-dependent endocytosis of proteinase-activated receptor 2 is required for intracellular targeting of activated ERK1/2. J Cell Biol. 2000;148:1267–1281. Luttrell LM, Roudabush FL, Choy EW, et al. Activation and targeting of extracellular signal-regulated kinases by beta-arrestin scaffolds. Proc Natl Acad Sci USA. 2001;98:2449 –2454. McDonald PH, Chow CW, Miller WE, et al. ␤-Arrestin 2: a receptor-regulated MAPK scaffold for the activation of JNK3. Science. 2000;290:1574 –1577. Coffa S, Breitman M, Hanson SM, et al. The effect of arrestin conformation on the recruitment of c-Raf1, MEK1, and ERK1/2 activation. PLoS One. 2011;6:e28723. Tohgo A, Choy EW, Gesty-Palmer D, et al. The stability of the G protein-coupled receptor-␤-arrestin interaction determines the mechanism and functional consequence of ERK activation. J Biol Chem. 2003;278:6258 – 6267. Wei H, Ahn S, Barnes WG, Lefkowitz RJ. Stable interaction between ␤-arrestin 2 and angiotensin type 1A receptor is required for ␤-arrestin 2-mediated activation of extracellular signal-regulated kinases 1 and 2. J Biol Chem. 2004;279:48255– 48261. Gesty-Palmer D, Chen M, Reiter E, et al. Distinct ␤-arrestin- and G protein-dependent pathways for parathyroid hormone receptor-stimulated ERK1/2 activation. J Biol Chem. 2006;281: 10856 –10864. Azzi M, Charest PG, Angers S, Rousseau G, Kohout T, Bouvier M, Pineyro G. ␤-Arrestin-mediated activation of MAPK by inverse agonists reveals distinct active conformations for G protein-coupled receptors. Proc Natl Acad Sci USA. 2003;100:11406 –11411. Datta NS, Kolailat R, Fite A, Pettway G, Abou-Samra AB. Distinct roles for mitogen-activated protein kinase phosphatase-1 (MKP-1) and ERK-MAPK in PTH1R signaling during osteoblast proliferation and differentiation. Cell Signal. 2010;22:457– 466. Teyra J, Sidhu SS, Kim PM. Elucidation of the binding preferences

72.

73.

74. 75.

76.

77.

78. 79.

80.

81.

82.

83.

84.

85.

86.

87.

88.

89.

90.

91.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

doi: 10.1210/me.2015-1091

92.

93.

94.

95.

96.

97.

98.

99.

100.

101.

102.

103.

104.

105.

106.

107.

108.

of peptide recognition modules: SH3 and PDZ domains. FEBS Lett. 2012;586:2631–2637. Romero G, von Zastrow M, Friedman PA. Role of PDZ proteins in regulating trafficking, signaling, and function of GPCRs: means, motif, and opportunity. Adv Pharmacol. 2011;62:279 – 314. Cao TT, Deacon HW, Reczek D, Bretscher A, von Zastrow M. A kinase-regulated PDZ-domain interaction controls endocytic sorting of the ␤2-adrenergic receptor. Nature. 1999;401:286 –290. Hall RA, Premont RT, Chow CW, et al. The ␤2-adrenergic receptor interacts with the Na⫹/H⫹-exchanger regulatory factor to control Na⫹/H⫹ exchange. Nature. 1998;392:626 – 630. Hu LA, Tang Y, Miller WE, et al. ␤1-Adrenergic receptor association with PSD-95. Inhibition of receptor internalization and facilitation of ␤1-adrenergic receptor interaction with N-methyl-Daspartate receptors. J Biol Chem. 2000;275:38659 –38666. Gardner LA, Naren AP, Bahouth SW. Assembly of an SAP97AKAP79-cAMP-dependent protein kinase scaffold at the type 1 PSD-95/DLG/ZO1 motif of the human ␤(1)-adrenergic receptor generates a receptosome involved in receptor recycling and networking. J Biol Chem. 2007;282:5085–5099. He J, Bellini M, Inuzuka H, et al. Proteomic analysis of ␤1-adrenergic receptor interactions with PDZ scaffold proteins. J Biol Chem. 2006;281:2820 –2827. Nakagawa T, Asahi M. ␤1-Adrenergic receptor recycles via a membranous organelle, recycling endosome, by binding with sorting nexin27. J Membr Biol. 2013;246:571–579. Nooh MM, Chumpia MM, Hamilton TB, Bahouth SW. Sorting of ␤1-adrenergic receptors is mediated by pathways that are either dependent on or independent of type I PDZ, protein kinase A (PKA), and SAP97. J Biol Chem. 2014;289:2277–2294. Xu J, Paquet M, Lau AG, Wood JD, Ross CA, Hall RA. ␤1Adrenergic receptor association with the synaptic scaffolding protein membrane-associated guanylate kinase inverted-2 (MAGI-2). Differential regulation of receptor internalization by MAGI-2 and PSD-95. J Biol Chem. 2001;276:41310 – 41317. He J, Bellini M, Xu J, Castleberry AM, Hall RA. Interaction with cystic fibrosis transmembrane conductance regulator-associated ligand (CAL) inhibits ␤1-adrenergic receptor surface expression. J Biol Chem. 2004;279:50190 –50196. Bauch C, Koliwer J, Buck F, Honck HH, Kreienkamp HJ. Subcellular sorting of the G-protein coupled mouse somatostatin receptor 5 by a network of PDZ-domain containing proteins. PLoS One. 2014;9:e88529. Wente W, Stroh T, Beaudet A, Richter D, Kreienkamp HJ. Interactions with PDZ domain proteins PIST/GOPC and PDZK1 regulate intracellular sorting of the somatostatin receptor subtype 5. J Biol Chem. 2005;280:32419 –32425. Koliwer J, Park M, Bauch C, von Zastrow M, Kreienkamp HJ. The golgi-associated PDZ domain protein PIST/GOPC stabilizes the ␤1-adrenergic receptor in intracellular compartments after internalization. J Biol Chem. 2015;290:6120 – 6129. Stroh T, Jackson AC, Sarret P, et al. Intracellular dynamics of sst5 receptors in transfected COS-7 cells: maintenance of cell surface receptors during ligand-induced endocytosis. Endocrinology. 2000;141:354 –365. Lauffer BE, Melero C, Temkin P, et al. SNX27 mediates PDZdirected sorting from endosomes to the plasma membrane. J Cell Biol. 2010;190:565–574. Temkin P, Lauffer B, Jager S, Cimermancic P, Krogan NJ, von Zastrow M. SNX27 mediates retromer tubule entry and endosome-to-plasma membrane trafficking of signalling receptors. Nat Cell Biol. 2011;13:715–721. Liew CW, Vockel M, Glassmeier G, et al. Interaction of the human somatostatin receptor 3 with the multiple PDZ domain protein MUPP1 enables somatostatin to control permeability of epithelial tight junctions. FEBS Lett. 2009;583:49 –54.

press.endocrine.org/journal/mend

827

109. Guillaume JL, Daulat AM, Maurice P, et al. The PDZ protein mupp1 promotes Gi coupling and signaling of the Mt1 melatonin receptor. J Biol Chem. 2008;283:16762–16771. 110. Becamel C, Figge A, Poliak S, et al. Interaction of serotonin 5-hydroxytryptamine type 2C receptors with PDZ10 of the multi-PDZ domain protein MUPP1. J Biol Chem. 2001;276:12974 –12982. 111. Parker LL, Backstrom JR, Sanders-Bush E, Shieh BH. Agonistinduced phosphorylation of the serotonin 5-HT2C receptor regulates its interaction with multiple PDZ protein 1. J Biol Chem. 2003;278:21576 –21583. 112. Ullmer C, Schmuck K, Figge A, Lubbert H. Cloning and characterization of MUPP1, a novel PDZ domain protein. FEBS Lett. 1998;424:63– 68. 113. Hanyaloglu AC, von Zastrow M. Regulation of GPCRs by endocytic membrane trafficking and its potential implications. Annu Rev Pharmacol Toxicol. 2008;48:537–568. 114. Jean-Alphonse F, Bowersox S, Chen S, Beard G, Puthenveedu MA, Hanyaloglu AC. Spatially restricted G protein-coupled receptor activity via divergent endocytic compartments. J Biol Chem. 2014;289:3960 –3977. 115. Jeanneteau F, Diaz J, Sokoloff P, Griffon N. Interactions of GIPC with dopamine D2, D3 but not D4 receptors define a novel mode of regulation of G protein-coupled receptors. Mol Biol Cell. 2004; 15:696 –705. 116. Jeanneteau F, Guillin O, Diaz J, Griffon N, Sokoloff P. GIPC recruits GAIP (RGS19) to attenuate dopamine D2 receptor signaling. Mol Biol Cell. 2004;15:4926 – 4937. 117. Klenk C, Vetter T, Zurn A, et al. Formation of a ternary complex among NHERF1, ␤-arrestin, and parathyroid hormone receptor. J Biol Chem. 2010;285:30355–30362. 118. Kim JK, Kwon O, Kim J, et al. PDZ domain-containing 1 (PDZK1) protein regulates phospholipase C-␤3 (PLC-␤3)-specific activation of somatostatin by forming a ternary complex with PLC-␤3 and somatostatin receptors. J Biol Chem. 2012;287: 21012–21024. 119. Choi JW, Lim S, OH YS, et al. Subtype-specific role of phospholipase C-␤ in bradykinin and LPA signaling through differential binding of different PDZ scaffold proteins. Cell Signal. 2010;22: 1153–1161. 120. Oh YS, Jo NW, Choi JW, et al. NHERF2 specifically interacts with LPA2 receptor and defines the specificity and efficiency of receptor-mediated phospholipase C-␤3 activation. Mol Cell Biol. 2004; 24:5069 –5079. 121. Mahon MJ, Donowitz M, Yun CC, Segre GV. Na(⫹)/H(⫹) exchanger regulatory factor 2 directs parathyroid hormone 1 receptor signalling. Nature. 2002;417:858 – 861. 122. Wang B, Yang Y, Abou-Samra AB, Friedman PA. NHERF1 regulates parathyroid hormone receptor desensitization: interference with ␤-arrestin binding. Mol Pharmacol. 2009;75:1189 –1197. 123. Rochdi MD, Watier V, LA Madeleine C, Nakata H, Kozasa T, Parent JL. Regulation of GTP-binding protein alpha q (G␣q) signaling by the ezrin-radixin-moesin-binding phosphoprotein-50 (EBP50). J Biol Chem. 2002;277:40751– 40759. 124. Fam SR, Paquet M, Castleberry AM, et al. P2Y1 receptor signaling is controlled by interaction with the PDZ scaffold NHERF-2. Proc Natl Acad Sci USA. 2005;102:8042– 8047. 125. Hall RA, Ostedgaard LS, Premont RT, et al. A C-terminal motif found in the ␤2-adrenergic receptor, P2Y1 receptor and cystic fibrosis transmembrane conductance regulator determines binding to the Na⫹/H⫹ exchanger regulatory factor family of PDZ proteins. Proc Natl Acad Sci USA. 1998;95:8496 – 8501. 126. Dunn HA, Walther C, Godin CM, Hall RA, Ferguson SS. Role of SAP97 protein in the regulation of corticotropin-releasing factor receptor 1 endocytosis and extracellular signal-regulated kinase 1/2 signaling. J Biol Chem. 2013;288:15023–15034. 127. Walther C, Caetano FA, Dunn HA, Ferguson SS. PDZK1/NHERF3 differentially regulates corticotropin-releasing factor recep-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

828

128.

129.

130. 131.

132.

133.

134.

135.

136.

137.

138.

139.

140.

141. 142.

143.

144.

145.

146.

Walther and Ferguson

Endocrine GPCR Interacting Proteins

Mol Endocrinol, June 2015, 29(6):814 – 830

tor 1 and serotonin 2A receptor signaling and endocytosis. Cell Signal. 2015;27:519 –531. Xia Z, Gray JA, Compton-Toth BA, Roth BL. A direct interaction of PSD-95 with 5-HT2A serotonin receptors regulates receptor trafficking and signal transduction. J Biol Chem. 2003;278: 21901–21908. Reid HM, Turner EC, Mulvaney EP, Hyland PB, McLean C, Kinsella BT. Interaction of the human prostacyclin receptor and the NHERF4 family member intestinal and kidney enriched PDZ protein (IKEPP). Biochim Biophys Acta. 2012;1823:1998 –2012. Sarrouilhe D, di Tommaso A, Metaye T, Ladeveze V. Spinophilin: from partners to functions. Biochimie. 2006;88:1099 –1113. Brady AE, Wang Q, Allen PB, Rizzo M, Greengard P, Limbird LE. ␣2-Adrenergic agonist enrichment of spinophilin at the cell surface involves ␤␥ subunits of Gi proteins and is preferentially induced by the ␣2A-subtype. Mol Pharmacol. 2005;67:1690 –1696. Richman JG, Brady AE, Wang Q, Hensel JL, Colbran RJ, Limbird LE. Agonist-regulated Interaction between ␣2-adrenergic receptors and spinophilin. J Biol Chem. 2001;276:15003–15008. Smith FD, Oxford GS, Milgram SL. Association of the D2 dopamine receptor third cytoplasmic loop with spinophilin, a protein phosphatase-1-interacting protein. J Biol Chem. 1999;274: 19894 –19900. Brady AE, Wang Q, Colbran RJ, Allen PB, Greengard P, Limbird LE. Spinophilin stabilizes cell surface expression of ␣2B-adrenergic receptors. J Biol Chem. 2003;278:32405–32412. Wang Q, Limbird LE. Regulated interactions of the ␣2A adrenergic receptor with spinophilin, 14 –3–3␨, and arrestin 3. J Biol Chem. 2002;277:50589 –50596. Wang Q, Zhao J, Brady AE, et al. Spinophilin blocks arrestin actions in vitro and in vivo at G protein-coupled receptors. Science. 2004;304:1940 –1944. Wang X, Zeng W, Soyombo AA, et al. Spinophilin regulates Ca2⫹ signalling by binding the N-terminal domain of RGS2 and the third intracellular loop of G-protein-coupled receptors. Nat Cell Biol. 2005;7:405– 411. Lu R, Chen Y, Cottingham C, et al. Enhanced hypotensive, bradycardic, and hypnotic responses to ␣2-adrenergic agonists in spinophilin-null mice are accompanied by increased G protein coupling to the ␣2A-adrenergic receptor. Mol Pharmacol. 2010;78: 279 –286. Ruiz de Azua I, Nakajima K, Rossi M, et al. Spinophilin as a novel regulator of M3 muscarinic receptor-mediated insulin release in vitro and in vivo. FASEB J. 2012;26:4275– 4286. Charlton JJ, Allen PB, Psifogeorgou K, et al. Multiple actions of spinophilin regulate mu opioid receptor function. Neuron. 2008; 58:238 –247. Colledge M, Scott JD. AKAPs: from structure to function. Trends Cell Biol. 1999;9:216 –221. Cong M, Perry SJ, Lin FT, et al. Regulation of membrane targeting of the G protein-coupled receptor kinase 2 by protein kinase A and its anchoring protein AKAP79. J Biol Chem. 2001;276:15192– 15199. Fan G, Shumay E, Wang H, Malbon CC. The scaffold protein gravin (cAMP-dependent protein kinase-anchoring protein 250) binds the ␤2-adrenergic receptor via the receptor cytoplasmic Arg329 to Leu-413 domain and provides a mobile scaffold during desensitization. J Biol Chem. 2001;276:24005–24014. Fraser ID, Cong M, Kim J, et al. Assembly of an A kinase-anchoring protein-␤(2)-adrenergic receptor complex facilitates receptor phosphorylation and signaling. Curr Biol. 2000;10:409 – 412. Shih M, Lin F, Scott JD, Wang HY, Malbon CC. Dynamic complexes of ␤2-adrenergic receptors with protein kinases and phosphatases and the role of gravin. J Biol Chem. 1999;274:1588 – 1595. Li X, Nooh MM, Bahouth SW. Role of AKAP79/150 protein in

␤1-adrenergic receptor trafficking and signaling in mammalian cells. J Biol Chem. 2013;288:33797–33812. Nooh MM, Naren AP, Kim SJ, Xiang YK, Bahouth SW. SAP97 controls the trafficking and resensitization of the ␤1-adrenergic receptor through its PDZ2 and I3 domains. PLoS One. 2013;8: e63379. Broselid S, Berg KA, Chavera TA, et al. G protein-coupled receptor 30 (GPR30) forms a plasma membrane complex with membrane-associated guanylate kinases (MAGUKs) and protein kinase A-anchoring protein 5 (AKAP5) that constitutively inhibits cAMP production. J Biol Chem. 2014;289:22117–22127. Ali MS, Sayeski PP, Bernstein KE. Jak2 acts as both a STAT1 kinase and as a molecular bridge linking STAT1 to the angiotensin II AT1 receptor. J Biol Chem. 2000;275:15586 –15593. Kodama H, Fukuda K, Pan J, et al. Biphasic activation of the JAK/STAT pathway by angiotensin II in rat cardiomyocytes. Circ Res. 1998;82:244 –250. Marrero MB, Schieffer B, Paxton WG, et al. Direct stimulation of Jak/STAT pathway by the angiotensin II AT1 receptor. Nature. 1995;375:247–250. McWhinney CD, Dostal D, Baker K. Angiotensin II activates Stat5 through Jak2 kinase in cardiac myocytes. J Mol Cell Cardiol. 1998;30:751–761. McWhinney CD, Hunt RA, Conrad KM, Dostal DE, Baker KM. The type I angiotensin II receptor couples to Stat1 and Stat3 activation through Jak2 kinase in neonatal rat cardiac myocytes. J Mol Cell Cardiol. 1997;29:2513–2524. Pan J, Fukuda K, Kodama H, et al. Role of angiotensin II in activation of the JAK/STAT pathway induced by acute pressure overload in the rat heart. Circ Res. 1997;81:611– 617. Sayeski PP, Ali MS, Frank SJ, Bernstein KE. The angiotensin IIdependent nuclear translocation of Stat1 is mediated by the Jak2 protein motif 231YRFRR. J Biol Chem. 2001;276:10556 –10563. Ali MS, Sayeski PP, Dirksen LB, Hayzer DJ, Marrero MB, Bernstein KE. Dependence on the motif YIPP for the physical association of Jak2 kinase with the intracellular carboxyl tail of the angiotensin II AT1 receptor. J Biol Chem. 1997;272:23382–23388. Marrero MB, Venema VJ, Ju H, Eaton DC, Venema RC. Regulation of angiotensin II-induced JAK2 tyrosine phosphorylation: roles of SHP-1 and SHP-2. Am J Physiol. 1998;275:C1216 – C1223. Van Der Hoeven PC, Van Der Wal JC, Ruurs P, Van Dijk MC, Van Blitterswijk J. 14 –3-3 isotypes facilitate coupling of protein kinase C␨ to Raf-1: negative regulation by 14 –3-3 phosphorylation. Biochem J. 2000;345(Pt 2):297–306. Cohen BD, Nechamen CA, Dias JA. Human follitropin receptor (FSHR) interacts with the adapter protein 14 –3-3tau. Mol Cell Endocrinol. 2004;220:1–7. Shikano S, Coblitz B, Wu M, Li M. 14 –3-3 proteins: regulation of endoplasmic reticulum localization and surface expression of membrane proteins. Trends Cell Biol. 2006;16:370 –375. Yuan H, Michelsen K, Schwappach B. 14 –3-3 dimers probe the assembly status of multimeric membrane proteins. Curr Biol. 2003;13:638 – 646. Cunningham MR, McIntosh KA, Pediani JD, et al. Novel role for proteinase-activated receptor 2 (PAR2) in membrane trafficking of proteinase-activated receptor 4 (PAR4). J Biol Chem. 2012; 287:16656 –16669. Okamoto Y, Shikano S. Phosphorylation-dependent C-terminal binding of 14 –3-3 proteins promotes cell surface expression of HIV co-receptor GPR15. J Biol Chem. 2011;286:7171–7181. Prezeau L, Richman JG, Edwards SW, Limbird LE. The ␨ isoform of 14 –3-3 proteins interacts with the third intracellular loop of different ␣2-adrenergic receptor subtypes. J Biol Chem. 1999;274: 13462–13469. Wang Q, Limbird LE. Regulation of ␣2AR trafficking and signal-

147.

148.

149.

150.

151.

152.

153.

154.

155.

156.

157.

158.

159.

160.

161.

162.

163.

164.

165.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

doi: 10.1210/me.2015-1091

166.

167.

168.

169.

170.

171.

172.

173.

174.

175.

176.

177.

178.

179.

180.

181.

182. 183.

184.

185.

186.

ing by interacting proteins. Biochem Pharmacol. 2007;73:1135– 1145. Lania AG, Mantovani G, Spada A. Mechanisms of disease: mutations of G proteins and G-protein-coupled receptors in endocrine diseases. Nat Clin Pract Endocrinol Metab. 2006;2:681– 693. Kimple ME, Neuman JC, Linnemann AK, Casey PJ. Inhibitory G proteins and their receptors: emerging therapeutic targets for obesity and diabetes. Exp Mol Med. 2014;46:e102. Sivertsen B, Holliday N, Madsen AN, Holst B. Functionally biased signalling properties of 7TM receptors— opportunities for drug development for the ghrelin receptor. Br J Pharmacol. 2013; 170:1349 –1362. Evron T, Peterson SM, Urs NM, et al. G Protein and ␤-arrestin signaling bias at the ghrelin receptor. J Biol Chem. 2014;289: 33442–33455. Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. 2006;444:840 – 846. Ahren B. Islet G protein-coupled receptors as potential targets for treatment of type 2 diabetes. Nat Rev Drug Discov. 2009;8:369 – 385. Kong KC, Tobin AB. The role of M(3)-muscarinic receptor signaling in insulin secretion. Commun Integr Biol. 2011;4:489 – 491. Guettier JM, Gautam D, Scarselli M, et al. A chemical-genetic approach to study G protein regulation of beta cell function in vivo. Proc Natl Acad Sci USA. 2009;106:19197–19202. Jain S, Ruiz de Azua I, Lu H, White MF, Guettier JM, Wess J. Chronic activation of a designer G(q)-coupled receptor improves ␤ cell function. J Clin Invest. 2013;123:1750 –1762. Nakajima K, Wess J. Design and functional characterization of a novel, arrestin-biased designer G protein-coupled receptor. Mol Pharmacol. 2012;82:575–582. Kong KC, Butcher AJ, McWilliams P, et al. M3-muscarinic receptor promotes insulin release via receptor phosphorylation/arrestin-dependent activation of protein kinase D1. Proc Natl Acad Sci USA. 2010;107:21181–21186. Nakajima K, Jain S, Ruiz de Azua I, McMillin SM, Rossi M, Wess J. Minireview: novel aspects of M3 muscarinic receptor signaling in pancreatic ␤-cells. Mol Endocrinol. 2013;27:1208 –1216. Ruiz de Azua I, Scarselli M, Rosemond E, et al. RGS4 is a negative regulator of insulin release from pancreatic ␤-cells in vitro and in vivo. Proc Natl Acad Sci USA. 2010;107:7999 – 8004. Wente W, Efanov AM, Treinies I, et al. The PDZ/coiled-coil domain containing protein PIST modulates insulin secretion in MIN6 insulinoma cells by interacting with somatostatin receptor subtype 5. FEBS Lett. 2005;579:6305– 6310. Curtis JR, Safford MM. Management of osteoporosis among the elderly with other chronic medical conditions. Drugs Aging. 2012; 29:549 –564. Qin L, Raggatt LJ, Partridge NC. Parathyroid hormone: a doubleedged sword for bone metabolism. Trends Endocrinol Metab. 2004;15:60 – 65. Taylor CW, Tovey SC. From parathyroid hormone to cytosolic Ca2⫹ signals. Biochem Soc Trans. 2012;40:147–152. Silva BC, Costa AG, Cusano NE, Kousteni S, Bilezikian JP. Catabolic and anabolic actions of parathyroid hormone on the skeleton. J Endocrinol Invest. 2011;34:801– 810. Gesty-Palmer D, Luttrell LM. ’Biasing’ the parathyroid hormone receptor: a novel anabolic approach to increasing bone mass? Br J Pharmacol. 2011;164:59 – 67. Gesty-Palmer D, Flannery P, Yuan L, et al. A ␤-arrestin-biased agonist of the parathyroid hormone receptor (PTH1R) promotes bone formation independent of G protein activation. Sci Transl Med. 2009;1:1ra1. Dorsam RT, Gutkind JS. G-protein-coupled receptors and cancer. Nat Rev Cancer. 2007;7:79 –94.

press.endocrine.org/journal/mend

829

187. Rodien P, Ho SC, Vlaeminck V, Vassart G, Costagliola S. Activating mutations of TSH receptor. Ann Endocrinol (Paris). 2003; 64:12–16. 188. O’Hayre M, Degese MS, Gutkind JS. Novel insights into G protein and G protein-coupled receptor signaling in cancer. Curr Opin Cell Biol. 2014;27:126 –135. 189. Chun KS, Lao HC, Trempus CS, Okada M, Langenbach R. The prostaglandin receptor EP2 activates multiple signaling pathways and ␤-arrestin1 complex formation during mouse skin papilloma development. Carcinogenesis. 2009;30:1620 –1627. 190. Kim JI, Lakshmikanthan V, Frilot N, Daaka Y. Prostaglandin E2 promotes lung cancer cell migration via EP4-␤Arrestin1-c-Src signalsome. Mol Cancer Res. 2010;8:569 –577. 191. Li TT, Alemayehu M, Aziziyeh AI, et al. ␤-Arrestin/Ral signaling regulates lysophosphatidic acid-mediated migration and invasion of human breast tumor cells. Mol Cancer Res. 2009;7:1064 – 1077. 192. Rosano L, Bagnato A. Convergent pathways link the endothelin A receptor to the ␤-catenin: the beta-arrestin connection. Cell Cycle. 2009;8:1462–1463. 193. Buchanan FG, Gorden DL, Matta P, Shi Q, Matrisian LM, DuBois RN. Role of ␤-arrestin 1 in the metastatic progression of colorectal cancer. Proc Natl Acad Sci USA. 2006;103:1492–1497. 194. Zhang P, He X, Tan J, Zhou X, Zou L. ␤-Arrestin2 mediates ␤-2 adrenergic receptor signaling inducing prostate cancer cell progression. Oncol Rep. 2011;26:1471–1477. 195. Whalen EJ, Rajagopal S, Lefkowitz RJ. Therapeutic potential of ␤-arrestin- and G protein-biased agonists. Trends Mol Med. 2011; 17:126 –139. 196. Lakshmikanthan V, Zou L, Kim JI, et al. Identification of ␤Arrestin2 as a corepressor of androgen receptor signaling in prostate cancer. Proc Natl Acad Sci USA. 2009;106:9379 –9384. 197. Purayil HT, Zhang Y, Dey A, Gersey Z, Espana-Serrano L, Daaka Y. Arrestin2 modulates androgen receptor activation [published online August 11, 2014]. Oncogene. doi:10.1038/onc.2014.252. 198. Ress AL, Stiegelbauer V, Schwarzenbacher D, et al. Spinophilin expression determines cellular growth, cancer stemness and 5-flourouracil resistance in colorectal cancer. Oncotarget. 2014; 5:8492– 8502. 199. Subbaiah VK, Kranjec C, Thomas M, Banks L. PDZ domains: the building blocks regulating tumorigenesis. Biochem J. 2011;439: 195–205. 200. Dai JL, Wang L, Sahin AA, Broemeling LD, Schutte M, Pan Y. NHERF (Na⫹/H⫹ exchanger regulatory factor) gene mutations in human breast cancer. Oncogene. 2004;23:8681– 8687. 201. Hayashi Y, Molina JR, Hamilton SR, Georgescu MM. NHERF1/ EBP50 is a new marker in colorectal cancer. Neoplasia. 2010;12: 1013–1022. 202. Muders MH, Dutta SK, Wang L, et al. Expression and regulatory role of GAIP-interacting protein GIPC in pancreatic adenocarcinoma. Cancer Res. 2006;66:10264 –10268. 203. Muders MH, Vohra PK, Dutta SK, et al. Targeting GIPC/synectin in pancreatic cancer inhibits tumor growth. Clin Cancer Res. 2009;15:4095– 4103. 204. Aubry JM. CRF system and mood disorders. J Chem Neuroanat. 2013;54:20 –24. 205. Leonard BE. The HPA and immune axes in stress: the involvement of the serotonergic system. Eur Psychiatry. 2005;20(suppl 3): S302–S306. 206. Catapano LA, Manji HK. G protein-coupled receptors in major psychiatric disorders. Biochim Biophys Acta. 2007;1768:976 – 993. 207. Magalhaes AC, Holmes KD, Dale LB, et al. CRF receptor 1 regulates anxiety behavior via sensitization of 5-HT2 receptor signaling. Nat Neurosci. 2010;13:622– 629. 208. Dunn HA, Walther C, Yuan GY, Caetano FA, Godin CM, Fergu-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

830

209.

210.

211.

212.

213.

Walther and Ferguson

Endocrine GPCR Interacting Proteins

Mol Endocrinol, June 2015, 29(6):814 – 830

son SS. Role of SAP97 in the regulation of 5-HT2AR endocytosis and signaling. Mol Pharmacol. 2014;86:275–283. Fribourg M, Moreno JL, Holloway T, et al. Decoding the signaling of a GPCR heteromeric complex reveals a unifying mechanism of action of antipsychotic drugs. Cell. 2011;147:1011–1023. Zhang J, Cheng S, Xiong Y, et al. A novel association of mGluR1a with the PDZ scaffold protein CAL modulates receptor activity. FEBS Lett. 2008;582:4117– 4124. Paquet M, Asay MJ, Fam SR, et al. The PDZ scaffold NHERF-2 interacts with mGluR5 and regulates receptor activity. J Biol Chem. 2006;281:29949 –29961. Hu LA, Chen W, Martin NP, Whalen EJ, Premont RT, Lefkowitz RJ. GIPC interacts with the ␤1-adrenergic receptor and regulates ␤1-adrenergic receptor-mediated ERK activation. J Biol Chem. 2003;278:26295–26301. Yang X, Zheng J, Xiong Y, et al. ␤2 Adrenergic receptor mediated ERK activation is regulated by interaction with MAGI-3. FEBS Lett. 2010;584:2207–2212.

214. Zhang H, Wang D, Sun H, Hall RA, Yun CC. MAGI-3 regulates LPA-induced activation of Erk and RhoA. Cell Signal. 2007;19: 261–268. 215. Wang B, Ardura JA, Romero G, Yang Y, Hall RA, Friedman PA. Na/H exchanger regulatory factors control parathyroid hormone receptor signaling by facilitating differential activation of G␣ protein subunits. J Biol Chem. 2010;285:26976 –26986. 216. Wheeler D, Garrido JL, Bisello A, Kim YK, Friedman PA, Romero G. Regulation of parathyroid hormone type 1 receptor dynamics, traffic, and signaling by the Na⫹/H⫹ exchanger regulatory factor-1 in rat osteosarcoma ROS 17/2.8 cells. Mol Endocrinol. 2008;22:1163–1170. 217. Zhang J, Vinuela A, Neely MH, et al. Inhibition of the dopamine D1 receptor signaling by PSD-95. J Biol Chem. 2007;282:15778 – 15789. 218. Katsushima Y, Sato T, Yamada C, et al. Interaction of PICK1 with C-terminus of growth hormone-releasing hormone receptor (GHRHR) modulates trafficking and signal transduction of human GHRHR. J Pharmacol Sci. 2013;122:193–204.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 11 July 2015. at 08:48 For personal use only. No other uses without permission. . All rights reserved.

Minireview: Role of intracellular scaffolding proteins in the regulation of endocrine G protein-coupled receptor signaling.

The majority of hormones stimulates and mediates their signal transduction via G protein-coupled receptors (GPCRs). The signal is transmitted into the...
332KB Sizes 2 Downloads 7 Views