+ All Categories
Home > Documents > Novel Allosteric Modulators of G Protein-coupled Receptors*

Novel Allosteric Modulators of G Protein-coupled Receptors*

Date post: 05-Jan-2017
Category:
Upload: vuhuong
View: 226 times
Download: 1 times
Share this document with a friend
12
Novel Allosteric Modulators of G Protein-coupled Receptors * Published, JBC Papers in Press, June 22, 2015, DOI 10.1074/jbc.R115.662759 Patrick R. Gentry 1 , Patrick M. Sexton 2 , and Arthur Christopoulos 2,3 From Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia G protein-coupled receptors (GPCRs) are allosteric proteins, because their signal transduction relies on interactions between topographically distinct, yet conformationally linked, domains. Much of the focus on GPCR allostery in the new millennium, however, has been on modes of targeting GPCR allosteric sites with chemical probes due to the potential for novel therapeutics. It is now apparent that some GPCRs possess more than one targetable allosteric site, in addition to a growing list of putative endogenous modulators. Advances in structural biology are also shedding new insights into mechanisms of allostery, although the complexities of candidate allosteric drugs necessitate rigor- ous biological characterization. G protein-coupled receptors (GPCRs), 4 also known as 7-transmembrane (7TM) receptors, are the largest superfamily of cell surface receptor proteins encoded by the human genome (1). These integral membrane proteins are highly dynamic and exist in an equilibrium between various functionally distinct conformational states (2). GPCRs fulfil the vital biological func- tion of transducing a wide range of extracellular signals (e.g. photons, lipids, neurotransmitters, hormones, peptides, enzymes, ions, odorants) across the cell membrane into the cytosolic space. Physiologically, the process begins when an endogenous extracellular signal interacts with the primary (“orthosteric”) binding site of GPCR, resulting in a conforma- tional rearrangement that conveys the signal through the plasma membrane-spanning 7TM region and subsequently triggering intracellular signaling cascades via heterotrimeric G proteins and other accessory proteins (3). Because GPCR-me- diated signaling systems are involved in regulating a multitude of physiological and pathophysiological processes, it is not sur- prising that the GPCR superfamily encompasses the targets of more actual and potential drugs than any other family of pro- teins (4, 5). To date, the majority of probe compounds and marketed drugs that target GPCRs are small molecules, but it is notewor- thy that there is a growing interest in utilizing biologics and antibodies to target these receptors as well (6). In addition, although the mode of action of the bulk of GPCR-targeting agents remains orthosteric, the turn of the millennium has wit- nessed substantial efforts in alternative methods of modulating GPCR activity, specifically by targeting topographically distinct allosteric sites. This minireview discusses some of the key char- acteristics associated with GPCR allostery and ongoing chal- lenges and opportunities in understanding and exploiting the phenomenon. Characteristics of GPCR Allostery Allostery is a widespread biological phenomenon that describes the ability of interactions occurring at one site of a macromolecule to modulate interactions at a spatially distinct binding site on the same macromolecule in a reciprocal man- ner. Since allosteric effects were first described in archetypal examples, such as the heme-heme interactions of hemoglobin, allostery has been acknowledged as a means by which proteins and other molecules (e.g. DNA) may amplify, attenuate, bias, and otherwise fine-tune their physiological functions (2, 7–9). Initial observations of allosteric phenomena in enzymes were mechanistically summarized first in the Monod-Wyman- Changeux (MWC) and subsequently in the Koshland-Nem- ethy-Filmer (KNF) models (10, 11). Although the MWC model depicts allostery as a concerted process (i.e. conformational selection), and the KNF model describes it as a sequential pro- cess (i.e. conformational induction), each model reflects valid key aspects of the nature of allostery, which involves ligand- mediated shifts in the population of pre-existing macromolec- ular conformational ensembles and resulting changes in the interactive properties of the new ensembles (12). In addition to enzymes, it became apparent that other protein classes, includ- ing GPCRs, possess many of the characteristics associated with allosteric proteins (13). GPCRs are conformationally dynamic proteins that act as conduits for the transfer of energy over a distance. Indeed, GPCR signal transduction is intrinsically allosteric as it involves the binding of an extracellular stimulus and subsequent propagation of the signal through the protein to a topographically distinct (e.g. 50 Å) intracellular site rec- ognized by G proteins, -arrestins, and others. Moreover, because of the broad diversity of endogenous activators of GPCRs, an orthosteric region on one type of receptor (e.g. class A biogenic amine receptor) may represent an allosteric domain in another type of receptor (e.g. class B secretin family or class C glutamate family receptors) (14) (Fig. 1). * This work was supported by Program Grant APP1055134 of the National Health and Medical Research Council (NHMRC) of Australia. This is the sec- ond article in the Thematic Minireview series “New Directions in G Protein- coupled Receptor Pharmacology.” The authors declare that they have no conflicts of interest with the contents of this article. 1 A 2015 Sir Keith Murdoch Fellow of the American Australian Association. 2 Principal Research Fellows of the NHMRC. 3 To whom correspondence should be addressed. Tel.: 03-9903-9067; Fax: 03-9903–9581; E-mail: [email protected]. 4 The abbreviations used are: GPCR, G protein-coupled receptor; 7TM, 7-transmembrane; 5-HT, 5-hydroxytryptamine; CCh, carbachol (2-[(amino- carbonyl)oxy]-N,N,N-trimethylethanaminium chloride); CCR, chemokine CC motif receptor; CRF 1 , corticotropin-releasing factor 1; CXCR, CXC motif chemokine receptor; DHPG, dihydroxyphenylglycine; FFAR, free fatty acid receptor; GnRH, gonadotropin-releasing hormone; mAChR, muscarinic acetylcholine receptor; mGlu, metabotropic glutamate receptor; PGF2, prostaglandin F2; MWC, Monod-Wyman-Changeux; KNF, Koshland- Nemethy-Filmer; PAM, positive allosteric modulator; NAL, neutral allo- steric ligand; NAM, negative allosteric modulator; MPEP, 2-methyl-6- (phenylethynyl)pyridine. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 290, NO. 32, pp. 19478 –19488, August 7, 2015 © 2015 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A. 19478 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 290 • NUMBER 32 • AUGUST 7, 2015 MINIREVIEW by guest on February 20, 2018 http://www.jbc.org/ Downloaded from
Transcript
Page 1: Novel Allosteric Modulators of G Protein-coupled Receptors*

Novel Allosteric Modulators of GProtein-coupled Receptors*Published, JBC Papers in Press, June 22, 2015, DOI 10.1074/jbc.R115.662759

Patrick R. Gentry1, Patrick M. Sexton2,and Arthur Christopoulos2,3

From Drug Discovery Biology, Monash Institute of PharmaceuticalSciences, Monash University, 399 Royal Parade,Parkville, Victoria 3052, Australia

G protein-coupled receptors (GPCRs) are allosteric proteins,because their signal transduction relies on interactions betweentopographically distinct, yet conformationally linked, domains.Much of the focus on GPCR allostery in the new millennium,however, has been on modes of targeting GPCR allosteric siteswith chemical probes due to the potential for novel therapeutics.It is now apparent that some GPCRs possess more than onetargetable allosteric site, in addition to a growing list of putativeendogenous modulators. Advances in structural biology are alsoshedding new insights into mechanisms of allostery, althoughthe complexities of candidate allosteric drugs necessitate rigor-ous biological characterization.

G protein-coupled receptors (GPCRs),4 also known as7-transmembrane (7TM) receptors, are the largest superfamilyof cell surface receptor proteins encoded by the human genome(1). These integral membrane proteins are highly dynamic andexist in an equilibrium between various functionally distinctconformational states (2). GPCRs fulfil the vital biological func-tion of transducing a wide range of extracellular signals(e.g. photons, lipids, neurotransmitters, hormones, peptides,enzymes, ions, odorants) across the cell membrane into thecytosolic space. Physiologically, the process begins when anendogenous extracellular signal interacts with the primary(“orthosteric”) binding site of GPCR, resulting in a conforma-tional rearrangement that conveys the signal through theplasma membrane-spanning 7TM region and subsequentlytriggering intracellular signaling cascades via heterotrimeric G

proteins and other accessory proteins (3). Because GPCR-me-diated signaling systems are involved in regulating a multitudeof physiological and pathophysiological processes, it is not sur-prising that the GPCR superfamily encompasses the targets ofmore actual and potential drugs than any other family of pro-teins (4, 5).

To date, the majority of probe compounds and marketeddrugs that target GPCRs are small molecules, but it is notewor-thy that there is a growing interest in utilizing biologics andantibodies to target these receptors as well (6). In addition,although the mode of action of the bulk of GPCR-targetingagents remains orthosteric, the turn of the millennium has wit-nessed substantial efforts in alternative methods of modulatingGPCR activity, specifically by targeting topographically distinctallosteric sites. This minireview discusses some of the key char-acteristics associated with GPCR allostery and ongoing chal-lenges and opportunities in understanding and exploiting thephenomenon.

Characteristics of GPCR Allostery

Allostery is a widespread biological phenomenon thatdescribes the ability of interactions occurring at one site of amacromolecule to modulate interactions at a spatially distinctbinding site on the same macromolecule in a reciprocal man-ner. Since allosteric effects were first described in archetypalexamples, such as the heme-heme interactions of hemoglobin,allostery has been acknowledged as a means by which proteinsand other molecules (e.g. DNA) may amplify, attenuate, bias,and otherwise fine-tune their physiological functions (2, 7–9).Initial observations of allosteric phenomena in enzymes weremechanistically summarized first in the Monod-Wyman-Changeux (MWC) and subsequently in the Koshland-Nem-ethy-Filmer (KNF) models (10, 11). Although the MWC modeldepicts allostery as a concerted process (i.e. conformationalselection), and the KNF model describes it as a sequential pro-cess (i.e. conformational induction), each model reflects validkey aspects of the nature of allostery, which involves ligand-mediated shifts in the population of pre-existing macromolec-ular conformational ensembles and resulting changes in theinteractive properties of the new ensembles (12). In addition toenzymes, it became apparent that other protein classes, includ-ing GPCRs, possess many of the characteristics associated withallosteric proteins (13). GPCRs are conformationally dynamicproteins that act as conduits for the transfer of energy over adistance. Indeed, GPCR signal transduction is intrinsicallyallosteric as it involves the binding of an extracellular stimulusand subsequent propagation of the signal through the proteinto a topographically distinct (e.g. �50 Å) intracellular site rec-ognized by G proteins, �-arrestins, and others. Moreover,because of the broad diversity of endogenous activators ofGPCRs, an orthosteric region on one type of receptor (e.g. classA biogenic amine receptor) may represent an allosteric domainin another type of receptor (e.g. class B secretin family or class Cglutamate family receptors) (14) (Fig. 1).

* This work was supported by Program Grant APP1055134 of the NationalHealth and Medical Research Council (NHMRC) of Australia. This is the sec-ond article in the Thematic Minireview series “New Directions in G Protein-coupled Receptor Pharmacology.” The authors declare that they have noconflicts of interest with the contents of this article.

1 A 2015 Sir Keith Murdoch Fellow of the American Australian Association.2 Principal Research Fellows of the NHMRC.3 To whom correspondence should be addressed. Tel.: 03-9903-9067; Fax:

03-9903–9581; E-mail: [email protected] The abbreviations used are: GPCR, G protein-coupled receptor; 7TM,

7-transmembrane; 5-HT, 5-hydroxytryptamine; CCh, carbachol (2-[(amino-carbonyl)oxy]-N,N,N-trimethylethanaminium chloride); CCR, chemokineCC motif receptor; CRF1, corticotropin-releasing factor 1; CXCR, CXC motifchemokine receptor; DHPG, dihydroxyphenylglycine; FFAR, free fatty acidreceptor; GnRH, gonadotropin-releasing hormone; mAChR, muscarinicacetylcholine receptor; mGlu, metabotropic glutamate receptor; PGF2�,prostaglandin F2�; MWC, Monod-Wyman-Changeux; KNF, Koshland-Nemethy-Filmer; PAM, positive allosteric modulator; NAL, neutral allo-steric ligand; NAM, negative allosteric modulator; MPEP, 2-methyl-6-(phenylethynyl)pyridine.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 290, NO. 32, pp. 19478 –19488, August 7, 2015© 2015 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A.

19478 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 290 • NUMBER 32 • AUGUST 7, 2015

MINIREVIEW

by guest on February 20, 2018http://w

ww

.jbc.org/D

ownloaded from

Page 2: Novel Allosteric Modulators of G Protein-coupled Receptors*

Perhaps not surprisingly, therefore, the attractiveness andtractability of GPCRs as drug targets, coupled with advances indrug screening at these receptors, have uncovered differenttypes of allosteric GPCR modulators (Table 1) that are generallyclassified operationally based on their modes of pharmacology.An allosteric ligand that potentiates an agonist-mediatedreceptor response is referred to as a “positive allosteric modu-lator” (PAM), whereas one that attenuates activity is known as a“negative allosteric modulator” (NAM) (15). Mechanistically,these effects can be achieved through modulation of the bind-ing affinity of orthosteric ligands and/or through changes in theability of the orthosteric ligand-occupied receptor complex tointeract with intracellular transducer proteins. In contrast aligand that binds at the allosteric site without affecting receptoror orthosteric ligand activity (at equilibrium) is classed as “neu-tral allosteric ligand” (NAL). “Allosteric agonists” are ligandsthat are capable of directly activating the receptor from anallosteric site even in the absence of an orthosteric agonist (15),whereas “PAM agonists” and “NAM agonists” display mixedmodes of modulation and direct GPCR activation depending onthe cellular context (15). More recently, “bitopic ligands” havealso been described, which are defined as hybrid molecules pos-sessing separate orthosteric and allosteric pharmacophoresthat concomitantly engage with their respective sites on a singleGPCR to mediate novel pharmacology; several molecules ini-tially classified as allosteric agonists in the literature have sincebeen reclassified as bitopic ligands (15–18).

There are a number of general characteristics associated withallosteric GPCR modulators that present both unique advan-tages over orthosteric ligands as well as challenges to successfuldetection or validation of allosteric compounds. The first char-acteristic is the potential for allosteric ligands to exhibit greaterreceptor subtype selectivity. This property has two potential

origins: i) a decreased evolutionary pressure for sequence con-servation within allosteric sites relative to the orthosteric sitebetween GPCR subtypes (assuming there is no endogenousallosteric ligand for such a site, but see the next section) and/orii) selective cooperativity with an orthosteric site at one recep-tor subtype while exhibiting neutral cooperativity at other sub-types of that receptor family (19). The nature of the cooperativ-ity between orthosteric and allosteric sites on a GPCR alsorepresents a second important characteristic of allostery thathas practical and therapeutic implications. If the modulatordisplays minimal direct allosteric agonism in its own right, thenit will act as a PAM or NAM only when and where the endog-enous ligand is released, thus maintaining the natural spatio-temporal “rhythms” of the endogenous orthosteric ligand.Furthermore, very subtle degrees of positive or negative coop-erativity (which may be all that is necessary for certain GPCRsand disease states) result in an allosteric “effect ceiling” thatincreases the likelihood of on-target safety in overdose situa-tions, although this also poses a challenge for the screening ofmodulators with low degrees of cooperativity (20). The abilityto achieve unprecedented modes of on-target selectivity and/orfine-tune endogenous responses as a consequence of pure PAMor NAM activity may prove particularly important in diseaseswhere tight physiological regulation is vital, such as neurode-generation, schizophrenia, diabetes, and endocrine disorders.

A particularly interesting phenomenon associated with allos-tery that has been most noted at GPCRs is the property of“probe dependence,” wherein the magnitude and direction ofan allosteric effect can change depending on the nature of theinteracting ligands (15, 21). Probe dependence has substantialimplications for GPCR drug discovery and GPCR biology. Forinstance, many GPCRs have more than one endogenousorthosteric agonist, and totally different effects can be observed

FIGURE 1. Potential allosteric ligand-binding regions on GPCRs; some representative allosteric modulators recognizing each region are also listed.RAMP, receptor activity-modifying protein; MRAP, melanocortin receptor accessory protein; Cmpd, compound; AZ, AstraZeneca; Fz4, frizzled4 receptor; CaSR,calcium-sensing receptor; PCEP, 3-amino-3-carboxypropyl-2�-carboxyethyl phosphinic acid.

MINIREVIEW: Novel Allosteric Modulators of GPCRs

AUGUST 7, 2015 • VOLUME 290 • NUMBER 32 JOURNAL OF BIOLOGICAL CHEMISTRY 19479

by guest on February 20, 2018http://w

ww

.jbc.org/D

ownloaded from

Page 3: Novel Allosteric Modulators of G Protein-coupled Receptors*

TABLE 1Representative allosteric modulators of G protein-coupled receptorsAZ, AstraZeneca; BCQA, benzyl quinolone carboxylic acid; BMS, Bristol-Myers Squibb; Cmpd, compound; CGRP, calcitonin gene-related peptide; FSHR, follicle-stimulating hormone receptor; GH, growth hormone; GLP1R, glucagon-like peptide 1 receptor; GnRH, gonadotropin-releasing hormone; HMA, 5-(N,N-hexamethyl-ene)amiloride; P2Y12, purine P2Y12; RXFP, relaxin family peptide.

Receptor Exogenous modulator(s) Endogenous modulator(s)a

Class A5-HT1A Anandamide; cholesterol5-HT1B 5-HT-moduline; sodium5-HT1D 5-HT-moduline5-HT2A Anandamide; oleamide; sodium5-HT2C PNU-69176E Anandamide; oleamide5-HT4 IgG5-HT7 AnandamideAdenosine A1 LUF5484; PD 71,605; PD 81,723; PD 117,975 SodiumAdenosine A2A Amilorides SodiumAdenosine A3 DU124183; VUF5455; VUF8504 2-arachidonylglycerol; sodiumAdrenergic �1 Amilorides; benzodiazepines; conopeptide; �-TIA IgG; sodiumAdrenergic �2A Amilorides Cholesterol; sodiumAdrenergic �2B Amilorides SodiumAdrenergic �2D Agmatine; sodiumAdrenergic �1 Magnesium; manganese; IgGAdrenergic �2 Cholesterol; IgG; zincAngiotensin AT1 IgG; sodiumCannabinoid CB1 Org 27569; Org 27759; Org 29647; PSNCBAM-1; RTI-371 Lipoxin A4; Pepcan-12; pregnenolone;

sodiumChemokine CCK1 BenzodiazepinesChemokine CXCR1 AZ Cmpds. A & B; Reparixin; SCH 527123Chemokine CXCR2 AZ Cmpds. A & B; Reparixin; SCH 527123; SB 656933Chemokine CXCR3 IP-10; I-TACChemokine CXCR4 ASLW; RSVM; trichosanthin; plerixaforChemokine CXCR7 AMD3100; GSLWChemokine CCR1 BX-471; CP-481715; UCB35625Chemokine CCR2 CCR2-RA-[R]; JNJ-27141491; S.D.-24Chemokine CCR3 TAK779; UCB35625Chemokine CCR4 Pyrazinyl sulfonamidesChemokine CCR5 AK602; AK530; ancriviroc; aplaviroc; maraviroc SCH 351125; TAKK220;

TAK779; Trichosanthin; vicriviroc;Cholecystokinin CCK1 Devazepide; T-0632; GI181771X; Bdz-1Cholecystokinin CCK2 GI181771X; Bdz-2Dopamine D1 ZincDopamine D2 Amilorides; L-prolyl-L-leucyl-glycinamide; PAOPA SB269652; Homocysteine; IgG; melanotropin release

inhibiting factor 1; zincEndothelin ETA Acetylsalicylic acid; sodium salicylate IgG, sodiumFree fatty acid FFAR1 AM-1638; TAK-875 Docosahexaenoic acidFree fatty acid FFAR2 Phenylacetamides 1 & 2Free fatty acid FFAR3 hexahydroquinolone-3-carboxamide Cmpd 1FSHR BMS cmpds. 2–7GnRH receptor Furan-1; FD-1; HMAGH secretagogue L-692,429; GH-releasing peptide 6Luteinizing hormone Org 41841; [3H]-Org 43553M1 mAChR Brucine; BQCA; MK7622; ML169; MT3; MT7; staurosporine; tacrine

VU0029627; VU0119498Arachidonic acid; IgG; sodium

M2 mAChR Alcuronium; C7/3-phth; DUO3; gallamine; LY2033298; LY2119620; tacrine;W84

Arachidonic acid; dynorphin-A; IgG;myelin basic protein; major basicprotein; protamine; sodium

M3 mAChR Brucine; N-chloromethyl; WIN62577; VU0119498 Arachidonic acid; IgG; sodiumM4 mAChR Alcuronium; LY2033298; LY2119620; MT3; Thiochrome; VU0010010;

VU0152099; VU0152100Arachidonic acid; sodium

M5 mAChR ML326; ML375; ML380; ML381; VU0119498; VU0238429 Arachidonic acidNeurokinin 1 HeparinNeurokinin 2 [N-Benzyl, N- (2-naphthylmethyl)-amino]-acetonitrile�-opioid BMS-986121; BMS-986122; Cannabidiol Magnesium; manganese; sodium�-opioid BMS-986187; BMS-986188; Cannabidiol Manganese; sodium�-opioid Magnesium; manganese; sodiumOxytocin Cholesterol; Progesterone (rat); 5-

dihydroprogesterone (human)Purine P2Y12 2,2�-Pyridylsatogen tosylateRhodopsin CholesterolRXFP1 ML290RXFP3 135PAM1S1PR2 CYM-5520S1PR3 CYM-5541Thyrotropin receptor IgG

Class BCalcitonin receptor Pyrazolopyrimidines 2d, 2e, 2f, 2gCGRP receptor BIBN4096BSCRF1 receptor Antalarmin; CP-376395; NBI 35965; DMP696; NBI 27914Glucagon Bay27–9955; L-168,049GLP1R Novo Nordisk Compounds 1–6; BETP

MINIREVIEW: Novel Allosteric Modulators of GPCRs

19480 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 290 • NUMBER 32 • AUGUST 7, 2015

by guest on February 20, 2018http://w

ww

.jbc.org/D

ownloaded from

Page 4: Novel Allosteric Modulators of G Protein-coupled Receptors*

(or missed altogether) depending on which agonist is used toactivate the receptor in the presence of a given allosteric mod-ulator (22). In addition, the allosteric nature of GPCR signaltransduction means that the ability of a ligand-bound receptorto recognize cellular effector molecules is also subject to probedependence, this time directed intracellularly rather thanbetween two ligands. Specifically, the ability of orthosteric orallosteric GPCR ligands to stabilize different functionally rele-vant conformations of the same GPCR can give rise to the phe-nomenon of “biased agonism,” whereby only a subset of thepossible signaling repertoire of the receptor is recruited at therelative expense of other pathways (21, 23, 24).

Although beyond the scope of the current minireview, theallosteric pharmacology of bitopic ligands also presents aunique set of characteristics. Because of their dual pharmaco-phore nature, bitopic ligands can provide both greater selectiv-ity through interaction with an allosteric site and higher affinitythrough concomitant engagement of the orthosteric site. Ajudicious choice of orthosteric and allosteric building blockscan also yield bitopic ligands that display novel biased agonism(25). Although the spatiotemporal control of endogenous sig-naling afforded by pure allosteric modulator ligands is lost withbitopic ligands, the latter may prove particularly useful in situ-ations where endogenous agonist tone is progressively lost (e.g.neurodegenerative disorders) (18).

Endogenous Allosteric Modulators

Although most studies of GPCR allostery have traditionallyfocused on the actions of exogenous allosteric modulatorsbecause of the implications for novel drug discovery (seebelow), these receptors can also be modulated by a variety ofendogenous substances (26). As mentioned above, the bestcharacterized allosteric interaction at GPCRs is the positivecooperativity exhibited between the intracellular G protein-binding site with the orthosteric site (3). The GPCR intracellu-lar face can also interact with �-arrestins: endogenous GPCRaccessory proteins that were originally characterized as scaf-folding proteins involved in the termination of GPCR signaling,internalization, and recycling of receptors. It is now recognized

that �-arrestins can be involved in G protein-independent sig-nal transduction (27–30). These unique signaling propertiesemerge as a result of specific receptor conformations that havebeen shown to interact more readily with �-arrestins, leading toincreased �-arrestin binding and biased signaling through non-canonical signaling pathways (31–33). Although beyond thescope of the current review, it is also widely acknowledged thatdifferent types of GPCRs have the potential to associate withother proteins, such as receptor activity-modifying proteins(RAMPs) or melanocortin receptor accessory proteins(MRAPs), or with each other in the form of homo- or hetero-dimers (or higher order oligomers) with novel pharmacologicalproperties (26). In a number of such instances, cooperativeinteractions have been noted between orthosteric ligandswithin such complexes (34).

In addition to the ubiquitous allosteric sites utilized by Gproteins and �-arrestins, individual GPCR classes or subclassespossess more specific allosteric sites that may be targeted byendogenous allosteric modulators. For example, pharmacolog-ical and crystallographic evidence has shown that sodium isvital for stabilizing the inactive state of many class A GPCRs viaan allosteric site centered on a highly conserved aspartate resi-due, Asp2.50 (26). Additionally, aromatic amino acids (e.g.L-phenylalanine, L-tryptophan, and L-tyrosine) bind at a sitenear to, but spatially distinct from, the orthosteric site withinthe “Venus flytrap” (VFT) N-terminal domain of the calcium-sensing class C GPCR to potentiate the actions of extracellularcalcium at a number of intracellular signaling pathways(35–37). A similar allosteric site has been shown to be located inthe Venus flytrap of the metabotropic glutamate (mGlu) class CGPCRs. This site was initially reported as an allosteric chloride-binding site in the mGlu1 subtype, but some synthetic smallmolecule agonists have since been reported to bind this site inmGlu4 (38 – 40).

As summarized in Table 1, a growing number of substancesencompassing not only amino acids and ions, but also lipids,peptides, and proteins, have been proposed to act as putativeendogenous allosteric modulators of different types of GPCRs

TABLE I—continued

Receptor Exogenous modulator(s) Endogenous modulator(s)a

Class CCalcium-sensing receptor Fendeline; cinacalcet; NPS 467; NPS 568; NPS 2143; Calhex 231 L-Phe; L-Trp; L-Tyr; Glutathione; IgGGABAB CGP7930; CGP13501; GS39783 L-Leu; L-Ile; L-Phe; IgGmGlu1 (�)-CPCCOEt; [3H]R214127; BAY36–7620; cis-64a; EM-TBPC; JNJ16259685;

NPS2390; PHCCC; Ro01–6128; Ro67–7476; Ro0711401; YM-298198IgG

mGlu2 BINA; CBiPES; JNJ-40068782; LY181837; LY2607540; LY487379; MNI-137;RO4491533; RO4988546; RO5488608

mGlu3 AZD8529; MNI-137; RO4491533; RO4988546; RO5488608mGlu4 SIB-1893; MPEP; PHCCC; VU0080421; VU0155041; VU0155094; VU0422288mGlu5 5-MPEP; 5PAM523; ADX-47273; CDPPB; CPPHA; DFB; DMeOB; DCB;

DPFE; Fenobam; Mavoglurant; M-5MPEP; MPEP; MTEP; VU-29;VU0357121; VU0360172

IgG

mGlu6 VU0155041; VU0422288; PHCCCmGlu7 ADX71743; AMN082; MMPIP; VU0155041; VU0155094; VU0422288mGlu8 AZ12216052; VU0155041; VU0155094; VU0422288Taste T1R1 S807; IMPTaste T1R2 S819; S.E.-2; S.E.-3; SenomyxTaste T1R3 Cyclamate, lactisole

Class FSmoothened Sant1; Sant2; nat-20(S)-OHC OxysterolsFrizzled4 FzM1

a Note that some of these examples are more appropriately considered putative endogenous allosteric modulators.

MINIREVIEW: Novel Allosteric Modulators of GPCRs

AUGUST 7, 2015 • VOLUME 290 • NUMBER 32 JOURNAL OF BIOLOGICAL CHEMISTRY 19481

by guest on February 20, 2018http://w

ww

.jbc.org/D

ownloaded from

Page 5: Novel Allosteric Modulators of G Protein-coupled Receptors*

(26). It should be noted that in many of these instances, conclu-sive validation of an allosteric mechanism remains to be estab-lished, but the study of endogenous GPCR modulators mayprove to be a fertile ground for uncovering novel biology inhealth and disease. For instance, the nature and composition ofendogenous lipidic substances and numerous types of peptidesand proteins can vary dramatically in inflammation; if it can beshown that some of these substances are bona fide allostericmodulators of specific GPCRs, then this may represent a novelavenue for understanding and targeting GPCR functionality ininflammatory disease (26). In addition, GPCR-directed autoan-tibodies have been identified as potential endogenous allostericligands related to a number of chronic disease states. In partic-ular, autoantibodies have been identified in patients with a vari-ety of cardiovascular diseases, including autoantibodies againstthe AT1 receptor, the �1-adrenergic receptor, the M2 musca-rinic acetylcholine receptor (mAChR), the �1-adrenergicreceptor, the ETA receptor, and the 5-HT4 receptor (26). Addi-tionally, the central nervous system can be affected by autoan-tibody activity. For instance, autoantibodies against GABABhave been found in the cerebrospinal fluid of limbic encephali-tis patients, whereas patients with basal ganglia encephalitiswith dominant movement and psychiatric disease present withautoantibodies against the dopamine D2 receptor (26).

Exogenous Allosteric Modulators

Numerous synthetic small molecules with an allosteric modeof action have been reported for GPCRs (Table 1). The mAChRfamily is arguably the most well studied class A GPCR system inthis regard (41). Indeed, the first examples of GPCR allostericmodulators, the alkane bis-ammonium family of ligands, wereidentified at the mAChRs (42). Since that time, allosteric mod-ulators of nearly every mode of action have been found to targetthe mAChRs, including PAMs, NAMs, PAM and NAM ago-nists, and bitopic ligands (17, 43– 48). Interestingly, most ofthese ligands bind to the mAChRs at a shared site (albeit withdifferent affinities depending on the subtype), often referred toas the “common” allosteric site and highlighting how the tar-geting of a common allosteric domain can yield markedly dif-ferent biological behaviors (49, 50). However, evidence hasbeen provided for the existence of a second allosteric site on themAChRs, recognized by indolocarbazole analogues of stauro-sporine (e.g. KT5720 and KT5823) and the benzimidazolederivatives WIN63577 and WIN51708. As noted with the“common site” modulators, the second-site compounds alsodemonstrated positive, negative, or neutral interactions withacetylcholine depending on the mAChR subtype, but showedlargely neutral interactions with the binding of common sitemodulators, further supporting the presence of at least twoallosteric sites on a single GPCR (51–54).

The notion that a single GPCR may possess more than oneallosteric site for exogenous small molecules is likely morewidespread. For instance, a number of allosteric modulatorshave been described for the gonadotropin-releasing hormonereceptor, including Furan-1 and its derivative, FD-1 (55), as wellas a series of amiloride derivatives (56). Interestingly, interac-tion studies between the two different classes of allostericligand revealed neutral cooperativity, despite each class indi-

vidually modulating orthosteric ligands (56), again suggestive ofmore than one allosteric site on this receptor family. The intra-cellular face of GPCRs may also harbor allosteric sites for selec-tive small molecules. For instance, binding sites for allostericantagonists of the CXCR2, CCR4, and CCR5 chemokine recep-tors have been identified among the intracellular loops (57–59).Intracellular allosteric sites are also utilized by pepducins: cell-penetrating, lipidated peptides that target the intracellularloops. By tailoring the peptide’s design to the intracellulardomain of a particular GPCR, researchers have been successfulin producing pepducins selectively targeting a number ofGPCRs involved in inflammatory diseases, including protease-activated receptors (PAR1, -2, -4) and chemokine receptors(CXCR1, -2, -4) (60, 61).

Class B GPCRs, such as the secretin, glucagon, and gluca-gon-like peptide 1 receptors, have proven notoriously intrac-table to small molecule discovery and have remained an areaof intense research with regard to allosteric drugs. One com-mon observation is the discovery of direct-acting allostericagonists, in addition to PAM agonists or NAMs (62). Pureclass B GPCR PAMs have thus far remained relatively moreelusive, but whether this reflects a fundamental property ofthis class of GPCR or the relative immaturity of detailedstudies of Class B allostery remains to be determined. Incommon with the preceding examples, however, there isclear evidence suggesting the presence of more than oneallosteric site on Class B GPCRs (63, 64).

Within the class C GPCRs, the mGlu receptors have a veryrich allosteric pharmacology. In particular, allosteric modu-lation of the mGlu5 receptor has been of interest as a targetfor the treatment of schizophrenia and Alzheimer disease(65, 66). As such, multiple small molecule PAMs have beendeveloped for mGlu5 (67–72), with the bulk acting at a com-mon site in the upper region of the 7TM domain oftentermed the “MPEP-binding site” after the prototypicalmGlu5 NAM, MPEP (65, 66). However, several ligands havebeen identified that are non-competitive with the MPEP site,including the PAMs VU357121 and CPPHA, indicating thepresence of at least two distinct allosteric sites (67, 69, 73,74). Further investigations have shown that PAMs acting oneach of these sites may have varied effects on different sig-naling pathways, indicating that allosteric modulation of dis-crete allosteric sites may have significant effects on theresponse of the GPCR (75, 76).

Finally, the Class F GPCR family, which includes theSmoothened and Frizzled receptors, has also been considereddifficult to target with small molecules, but recent break-throughs have revealed that these receptors possess allostericbinding sites for exogenous ligands. For example, a series ofsmall molecules that were originally designed to act as pharma-cological chaperones for a misfolded mutant of Frizzled4 weresubsequently identified as novel allosteric modulators of thewild-type form of Frizzled4; the binding site for these com-pounds was proposed to be located in the vicinity of intracellu-lar loop 3 of the receptor (77). The recent crystal structure ofthe Smoothened receptor bound to the allosteric modulator,Sant1, also revealed an allosteric pocket that is located deepwithin the transmembrane-spanning cavity of the receptor,

MINIREVIEW: Novel Allosteric Modulators of GPCRs

19482 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 290 • NUMBER 32 • AUGUST 7, 2015

by guest on February 20, 2018http://w

ww

.jbc.org/D

ownloaded from

Page 6: Novel Allosteric Modulators of G Protein-coupled Receptors*

toward the cytosolic end; this is in contrast to the binding sitethat is closer to the extracellular entrance and utilized bycanonical ligands of this receptor (78).

It is important to note that although small molecules havemade an undeniable impact on the study of allostery, there isgrowing interest in utilizing biologics to target allosteric sites.Thus far, allosteric antibodies have shown the most promise intargeting receptor tyrosine kinases, such as the insulin receptor,where the monoclonal antibody XMetA allosterically binds andactivates the receptor (79, 80). A subcategory of allosteric anti-bodies known as “allosteric ligand-modifying antibodies”(ALMA) acts by binding the endogenous ligand prior to inter-acting with the receptor (15). One example is gevokizumab, ananti-interleukin-1� antibody that binds interleukin and altersthe conformation of the endogenous ligand. Subsequently, thegevokizumab-interleukin-1� complex binds in a characteristicternary complex with the receptor (81, 82). Although examplesof rationally designed allosteric antibodies targeting GPCRshave yet to be described, the novel mechanisms of allostericantibodies and allosteric ligand-modifying antibodies offer avaluable new strategy for the study of GPCR allostery in thefuture.

Chemical Biology Challenges in Designing AllostericModulators of GPCRs

The production of highly site-specific allosteric probe mole-cules is a crucial factor in the study of allosteric interactions.Structure-based drug design is still a relatively nascent fieldwith respect to GPCRs, although substantial progress hasoccurred in recent years (see below). Within both industry andacademia, a large number of resources have been dedicated tothe generation, collection, and curation of large libraries of nat-ural and synthetic small molecules from which to screen for andoptimize novel allosteric ligands, and this remains the majorsource of such compounds (83, 84). Given this significantinvestment, the methodologies with which these novel ligandsare interrogated are central to any research campaign. The fieldof chemical biology has provided a number of tools to probeallosteric mechanisms of GPCR structure and function. Thesetechniques may provide the biochemical and biophysicalinformation necessary to garner a clear understanding of thestructural dynamics and signaling behavior of allostericinteractions on a chemical level. Techniques such as biolu-minescent and Förster resonance energy transfer as well assingle-molecule detection fluorescence have gained popu-larity for the study of GPCRs and validated binding partners.However, there are limitations for using these techniques inthe process of screening for novel ligands, not least of whichis the challenge of applying these methods to a high through-put screening format (85, 86). Measurement of second mes-sengers of G protein signaling (e.g. calcium and cAMP) or�-arrestin recruitment has proven more successful in thisrespect; nevertheless, it remains challenging to developselective small molecule allosteric modulators.

For example, allosteric ligands often possess delicate struc-ture-activity relationships. That is, in a given chemical series, aseemingly minor modification to a molecule’s steric or elec-tronic properties often leads to the complete ablation of its

activity. Similarly, certain chemical series demonstrate “modeswitching” whereby small modifications to the structure canresult in dramatically changed pharmacological profiles (87).These concerns are particularly relevant when further modifi-cations are applied to the probes, for instance in the generationof irreversible or photoactivatable allosteric molecules (88, 89),or through the use of these chemical probes in vivo, where bio-chemical transformations undertaken by metabolic processesmay alter a modulator’s potency, cooperativity, receptor selec-tivity, or mode of action.

Novel chemotypes of allosteric modulators also pose chal-lenges to pharmacological characterization. For instance, manyallosteric modulators display phenomena such as probe depen-dence, and can impose biased signaling on the actions oforthosteric ligands (Table 2). These may go unnoticed with theuse of a single screening methodology. Only through the mea-surement of an allosteric ligand’s effect on multiple down-stream pathways in the presence of all relevant endogenousorthosteric ligands can a compound’s properties be fully eluci-dated, and this must be factored into all allosteric discoveryprograms (90). Further complexities in characterizing a novelallosteric ligand may be caused by differences between species’receptor isoforms, differential effects on orthosteric ligandaffinity and efficacy, varied kinetics between different signalingpathways, and the possibility of undetected endogenous allos-teric interactions. Therefore efforts must be made to fully char-acterize and validate novel chemical probes in as many experi-mental paradigms as possible to compose the clearest picture ofthe compound’s properties.

What Can Structural Biology Reveal About AllostericMechanisms?

Although the use of selective allosteric probe molecules andfunctional assays can reveal much about allosteric pharmacol-ogy, the information provided by crystal structures and otherhigh resolution approaches is invaluable for more direct,molecular level insights into GPCR allostery. Since the turn ofthe millennium, when the first high resolution GPCR crystalstructure was solved for bovine rhodopsin (91), there has been aseemingly exponential growth in the number of crystal struc-tures solved for a range of class A, B, and C GPCRs (Fig. 2),although the majority of these rely on some form of proteinengineering to improve stability and crystal formation. This hasallowed the observation of conformational snapshots adoptedby structures co-bound to ligands and/or interacting proteins,offering an unprecedented view into the fundamental struc-tural basis for receptor function.

Of note, there have been a number of inactive state crystalstructures of GPCRs in binary complexes with small mole-cule NAMs. These include the chemokine receptor CCR5bound to maraviroc, the corticotropin-releasing factorreceptor CRF1 bound to CP-376395, the mGlu1 bound toFITM, the mGlu5 bound to mavoglurant, and the aforemen-tioned Smoothened receptor bound to Sant1 (78, 92–95).Despite providing new insights into the binding behavior ofthese NAMs, the inactive and binary complex natures ofthese structures do not capture the complete structuralmechanisms that underlie cooperativity between the allos-

MINIREVIEW: Novel Allosteric Modulators of GPCRs

AUGUST 7, 2015 • VOLUME 290 • NUMBER 32 JOURNAL OF BIOLOGICAL CHEMISTRY 19483

by guest on February 20, 2018http://w

ww

.jbc.org/D

ownloaded from

Page 7: Novel Allosteric Modulators of G Protein-coupled Receptors*

teric and orthosteric sites. Indeed, the CRF1 and mGlu struc-tures lack the key N-terminal domains that constitute much(or all) of the orthosteric binding site.

A major breakthrough in recent years has been the crystalstructure depicting the classic ternary complex of anorthosteric agonist binding the �2-adrenergic receptor coupledto a Gs protein (96). This structure offered the first observationof how a GPCR orthosteric site is allosterically coupled to Gprotein activation. In terms of small molecule allostery, therecent structure of an active M2 mAChR in complex with thehigh efficacy orthosteric agonist, iperoxo, and the PAM,LY02119620 (97), also represented a major structural biologyadvance in understanding allostery at a GPCR. For example, thestructure is consistent with the predictions of the MWC modelof allostery in that the PAM preferentially recognizes and sta-bilizes a preformed active state of the receptor. Nonetheless,these structures still represent first steps in our molecular levelunderstanding of mechanisms underlying GPCR allostery. Phe-nomena such as biased allosteric modulation, probe depen-dence, and the actual mechanisms underlying transmission ofcooperativity remain challenging as they require the ability tocapture multiple states in the absence and presence of multipleligands. In the meantime, additional insights into the structuraland dynamic mechanisms of allostery are being obtained viaother methods. For example, the inactive M2 mAChR structurehas been subjected to long timescale molecular dynamics sim-ulations with a diverse range of small molecule allosteric mod-

ulators, in the absence or presence of an orthosteric ligand, toidentify binding poses and mechanisms underlying cooperativ-ity for a broad set of NAMs (98). The use of NMR has alsoallowed researchers to study the conformational flexibility ofthe receptor as a whole, providing more information on recep-tor dynamics than can be obtained through static crystal struc-tures, although no study has directly applied this approach toGPCR allosteric modulators to date (99, 100).

Conclusions

Allosteric modulation of GPCRs is now a widely acceptedphenomenon with substantial implications for novel drug dis-covery, yet many fundamental issues remain to be addressed.For example, it is now clear that a single GPCR can possessmore than one allosteric site, but whether the targeting of suchsites can lead to differential behaviors remains unknown. Theprevalence of endogenous allosteric modulators remains to bedetermined, but they may prove to be a mechanism of tissue-specific regulation in normal physiology or disease that may beamenable to chemical manipulation. The ascendance of biolog-ics as therapeutics opens new vistas for targeting of GPCRs witha greater degree of specificity than previously possible, yet theextent with which such substances can interact allostericallywith GPCRs is largely unexplored. As more detailed GPCRstructural information becomes available, it too will profoundlyaffect our understanding of allostery in GPCRs as well as themanner by which allosteric molecules are designed. In prepar-

TABLE 2Representative examples of biased allosteric modulationCaSR, calcium-sensing receptor; Oxo, oxotremorine (1-(4-pyrrolidin-1-ylbut-2-yn-1-yl)pyrrolidin-2-one); Oxo-M, oxotremorine methiodide (N,N,N-trimethyl-4-(2-oxo-1-pyrolidinyl)-2-butyn-1-ammonium iodide); PLD, phospholipase D; TMA, tetramethylammonium.

Ligand Receptor Bias profile (and orthosteric agonist probe)

Cinacalcet CaSR PAM for intracellular Ca2� mobilization (Ca2��)NAL for ERK1/2 phosphorylation (Ca2�)

LPI805 Neurokinin 2 NAM for cAMP production (Neurokinin A)NAL for intracellular Ca2� mobilization (Neurokinin A)

2-amino-3-benzothiophene derivative (MIPS#?) Adenosine A1 Allosteric agonist for cAMP productionVU0029767 M1 PAM for intracellular Ca2� mobilization (ACh)

NAL for PLD activation (ACh)Weak PAM for PI hydrolysis (ACh)

LY203398 M2 PAM for ERK1/2 phosphorylation (ACh, Oxo, Oxo-M,TMA, or McN-343)

NAM for ERK1/2 phosphorylation (Pilocarpine orXanomeline)

Brucine (mutant) M3K7.32E Allosteric PAM agonist for G�q (CCh)PAM for G�12 (CCh)NAL for G�i (CCh)

1-(4-Ethoxyphenyl)-5-methoxy-2-methylindole-3-carboxylic acid and N�-tosyltryptophan

CTRH2 NAMs for arrestin recruitment (prostaglandin D2)

NALs for G protein couplingCPPHA mGlu5 PAM for intracellular Ca2� mobilization (Glutamate or

DHPG)NAM for ERK1/2 phosphorylation (Glutamate or

DHPG, low concentrations)NAM for ERK1/2 phosphorylation (Glutamate or

DHPG, high concentrations)M-5MPEP mGlu5 NAM for intracellular Ca2� mobilization (quisqualate

or DHPG)Weak NAM for PI hydrolysis (quisqualate or DHPG)

PDC113.824 PGF2� NAM for G�12 (PGF2�)PAM for G�q (PGF2�)

ORG27569 CB1 NAM for G�i (CP55940)Allosteric agonist for ERK1/2 phosphorylation

MINIREVIEW: Novel Allosteric Modulators of GPCRs

19484 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 290 • NUMBER 32 • AUGUST 7, 2015

by guest on February 20, 2018http://w

ww

.jbc.org/D

ownloaded from

Page 8: Novel Allosteric Modulators of G Protein-coupled Receptors*

ing for such an eventuality, the rigorous biological character-ization of each new allosteric site and allosteric ligand discov-ered remains paramount.

Author Contributions—All authors wrote the manuscript.

References1. Fredriksson, R., Lagerström, M. C., Lundin, L.-G., and Schiöth, H. B.

(2003) The G-protein-coupled receptors in the human genome form fivemain families: phylogenetic analysis, paralogon groups, and fingerprints.Mol. Pharmacol. 63, 1256 –1272

2. Kenakin, T., and Miller, L. J. (2010) Seven transmembrane receptors asshapeshifting proteins: the impact of allosteric modulation and func-tional selectivity on new drug discovery. Pharmacol. Rev. 62, 265–304

3. Oldham, W. M., and Hamm, H. E. (2006) Structural basis of function inheterotrimeric G proteins. Q. Rev. Biophys. 39, 117–166

4. Rask-Andersen, M., Almén, M. S., and Schiöth, H. B. (2011) Trends in theexploitation of novel drug targets. Nat. Rev. Drug Discov. 10, 579 –590

5. Conn, P. J., Lindsley, C. W., Meiler, J., and Niswender, C. M. (2014)Opportunities and challenges in the discovery of allosteric modulators ofGPCRs for treating CNS disorders. Nat. Rev. Drug Discov. 13, 692–708

6. Overington, J. P., Al-Lazikani, B., and Hopkins, A. L.(2006) How manydrug targets are there? Nat. Rev. Drug Discov. 5, 993–996

7. Monod, J., Changeux, J.-P., and Jacob, F. (1963) Allosteric proteins andcellular control systems. J. Mol. Biol. 6, 306 –329

8. Christopoulos, A., and Kenakin, T. (2002) G protein-coupled receptorallosterism and complexing. Pharmacol. Rev. 54, 323–374

9. Fenton, A. W. (2008) Allostery: an illustrated definition for the “secondsecret of life.” Trends Biochem. Sci. 33, 420 – 425

10. Monod, J., Wyman, J., and Changeux, J.-P. (1965) On the nature of allos-teric transitions: a plausible model. J. Mol. Biol. 12, 88 –118

11. Koshland, D. E., Jr., Némethy, G., and Filmer, D. (1966) Comparison ofexperimental binding data and theoretical models in proteins containingsubunits. Biochemistry 5, 365–385

12. Motlagh, H. N., Wrabl, J. O., Li, J., and Hilser, V. J. (2014) The ensemblenature of allostery. Nature 508, 331–339

13. Canals, M., Sexton, P. M., and Christopoulos, A. (2011) Allostery in GP-CRs: “MWC” revisited. Trends Biochem. Sci. 36, 663– 672

14. Conn, P. J., Christopoulos, A., and Lindsley, C. W. (2009) Allosteric mod-ulators of GPCRs: a novel approach for the treatment of CNS disorders.Nat. Rev. Drug Discov. 8, 41–54

15. Christopoulos, A., Changeux, J.-P., Catterall, W. A., Fabbro, D., Burris,T. P., Cidlowski, J. A., Olsen, R. W., Peters, J. A., Neubig, R. R., Pin, J.-P.,Sexton, P. M., Kenakin, T. P., Ehlert, F. J., Spedding, M., and Langmead,C. J. (2014) International union of basic and clinical pharmacology. XC.Multisite pharmacology: recommendations for the nomenclature of re-ceptor allosterism and allosteric ligands. Pharmacol. Rev. 66, 918 –947

16. Mohr, K., Tränkle, C., Kostenis, E., Barocelli, E., De Amici, M., and Hol-zgrabe, U. (2010) Rational design of dualsteric GPCR ligands: quests andpromise. Br. J. Pharmacol. 159, 997–1008

17. Valant, C., Gregory, K. J., Hall, N. E., Scammells, P. J., Lew, M. J., Sexton,P. M., and Christopoulos, A. (2008) A novel mechanism of G protein-coupled receptor functional selectivity. Muscarinic partial agonist McN-A-343 as a bitopic orthosteric/allosteric ligand. J. Biol. Chem. 283,29312–29321

FIGURE 2. Timeline of representative GPCR crystal structures, 2000 –2015. N/OFQ, nociception/orphanin FQ receptor; SMO, Smoothened receptor; OR,opioid receptor; H1, histamine H1 receptor; S1P1, sphingosine-1-phosphate 1 receptor.

MINIREVIEW: Novel Allosteric Modulators of GPCRs

AUGUST 7, 2015 • VOLUME 290 • NUMBER 32 JOURNAL OF BIOLOGICAL CHEMISTRY 19485

by guest on February 20, 2018http://w

ww

.jbc.org/D

ownloaded from

Page 9: Novel Allosteric Modulators of G Protein-coupled Receptors*

18. Valant, C., Robert Lane, J., Sexton, P. M., and Christopoulos, A. (2012)The best of both worlds? Bitopic orthosteric/allosteric ligands of G pro-tein-coupled receptors. Annu. Rev. Pharmacol. Toxicol. 52, 153–178

19. Lazareno, S., Dolezal, V., Popham, A., and Birdsall, N. J. M. (2004) Thio-chrome enhances acetylcholine affinity at muscarinic M4 receptors: re-ceptor subtype selectivity via cooperativity rather than affinity. Mol.Pharmacol. 65, 257–266

20. Christopoulos, A. (2002) Allosteric binding sites on cell-surface recep-tors: novel targets for drug discovery. Nat. Rev. Drug Discov. 1, 198 –210

21. Kenakin, T. (2005) New concepts in drug discovery: collateral efficacyand permissive antagonism. Nat. Rev. Drug Discov. 4, 919 –927

22. Wootten, D., Christopoulos, A., and Sexton, P. M. (2013) Emerging par-adigms in GPCR allostery: implications for drug discovery. Nat. Rev.Drug. Discov. 12, 630 – 644

23. Leach, K., Sexton, P. M., and Christopoulos, A. (2007) Allosteric GPCRmodulators: taking advantage of permissive receptor pharmacology.Trends Pharmacol. Sci. 28, 382–389

24. Kenakin, T., and Christopoulos, A. (2013) Signalling bias in new drugdiscovery: detection, quantification and therapeutic impact. Nat. Rev.Drug Discov. 12, 205–216

25. Valant, C., May, L. T., Aurelio, L., Chuo, C. H., White, P. J., Baltos, J.-A.,Sexton, P. M., Scammells, P. J., and Christopoulos, A. (2014) Separationof on-target efficacy from adverse effects through rational design of abitopic adenosine receptor agonist. Proc. Natl. Acad. Sci. U.S.A. 111,4614 – 4619

26. van der Westhuizen, E. T., Valant, C., Sexton, P. M., and Christopoulos,A. (2015) Endogenous allosteric modulators of G protein-coupled recep-tors. J. Pharmacol. Exp. Ther. 353, 246 –260

27. Luttrell, L. M., Ferguson, S. S., Daaka, Y., Miller, W. E., Maudsley, S., DellaRocca, G. J., Lin, F., Kawakatsu, H., Owada, K., Luttrell, D. K., Caron,M. G., and Lefkowitz, R. J. (1999) �-Arrestin-dependent formation of �2

adrenergic receptor-Src protein kinase complexes. Science 283, 655– 66128. Lefkowitz, R. J., and Shenoy, S. K. (2005) Transduction of receptor signals

by �-arrestins. Science 308, 512–51729. Luttrell, L. M., and Gesty-Palmer, D. (2010) Beyond desensitization:

physiological relevance of arrestin-dependent signaling. Pharmacol. Rev.62, 305–330

30. Reiter, E., Ahn, S., Shukla, A. K., and Lefkowitz, R. J. (2012) Molecularmechanism of �-arrestin-biased agonism at seven-transmembrane re-ceptors. Annu. Rev. Pharmacol. Toxicol. 52, 179 –197

31. Gurevich, V. V., Pals-Rylaarsdam, R., Benovic, J. L., Hosey, M. M., andOnorato, J. J. (1997) Agonist-receptor-arrestin, an alternative ternarycomplex with high agonist affinity. J. Biol. Chem. 272, 28849 –28852

32. Galandrin, S., Oligny-Longpré, G., Bonin, H., Ogawa, K., Galés, C., andBouvier, M. (2008) Conformational rearrangements and signaling cas-cades involved in ligand-biased mitogen-activated protein kinase signal-ing through the �1-adrenergic receptor. Mol. Pharmacol. 74, 162–172

33. Tang, W., Strachan, R. T., Lefkowitz, R. J., and Rockman, H. A. (2014)Allosteric modulation of �-arrestin-biased angiotensin II type 1 receptorsignaling by membrane stretch. J. Biol. Chem. 289, 28271–28283

34. Milligan, G. (2009) G protein-coupled receptor hetero-dimerization:contribution to pharmacology and function. Br. J. Pharmacol. 158, 5–14

35. Conigrave, A. D., Quinn, S. J., and Brown, E. M. (2000) L-Amino acidsensing by the extracellular Ca2�-sensing receptor. Proc. Natl. Acad. Sci.U.S.A. 97, 4814 – 4819

36. Zhang, Z., Qiu, W., Quinn, S. J., Conigrave, A. D., Brown, E. M., and Bai,M. (2002) Three adjacent serines in the extracellular domains of the CaRare required for L-amino acid-mediated potentiation of receptor func-tion. J. Biol. Chem. 277, 33727–33735

37. Mun, H. C., Culverston, E. L., Franks, A. H., Collyer, C. A., Clifton-Bligh,R. J., and Conigrave, A. D. (2005) A double mutation in the extracellularCa2�-sensing receptor’s Venus flytrap domain that selectively disablesL-amino acid sensing. J. Biol. Chem. 280, 29067–29072

38. Ogawa, H., Qiu, Y., Philo, J. S., Arakawa, T., Ogata, C. M., and Misono,K. S. (2010) Reversibly bound chloride in the atrial natriuretic peptidereceptor hormone-binding domain: possible allosteric regulation and aconserved structural motif for the chloride-binding site. Protein Sci. 19,544 –557

39. Selvam, C., Oueslati, N., Lemasson, I. A., Brabet, I., Rigault, D., Courtiol,T., Cesarini, S., Triballeau, N., Bertrand, H. O., Goudet, C., Pin, J.-P., andAcher, F. C. (2010) A virtual screening hit reveals new possibilities fordeveloping group III metabotropic glutamate receptor agonists. J. Med.Chem. 53, 2797–2813

40. Acher, F. C., Selvam, C., Pin, J.-P., Goudet, C., and Bertrand, H. O. (2011)A critical pocket close to the glutamate binding site of mGlu receptorsopens new possibilities for agonist design. Neuropharmacology 60,102–107

41. Gregory, K. J., Sexton, P. M., and Christopoulos, A. (2007) Allostericmodulation of muscarinic acetylcholine receptors. Curr. Neuropharma-col. 5, 157–167

42. Lüllmann, H., Ohnesorge, F. K., Schauwecker, G. C., and Wassermann,O. (1969) Inhibition of the actions of carbachol and DFP on guinea pigisolated atria by alkane-bis-ammonium compounds. Eur. J. Pharmacol.6, 241–247

43. Clark, A. L., and Mitchelson, F. (1976) The inhibitory effect of gallamineon muscarinic receptors. Br. J. Pharmacol. 58, 323–331

44. Stockton, J. M., Birdsall, N. J., Burgen, A. S., and Hulme, E. C. (1983)Modification of the binding properties of muscarinic receptors by gal-lamine. Mol. Pharmacol. 23, 551–557

45. Proska, J., and Tucek, S. (1994) Mechanisms of steric and cooperativeactions of alcuronium on cardiac muscarinic acetylcholine receptors.Mol. Pharmacol. 45, 709 –717

46. Chan, W. Y., McKinzie, D. L., Bose, S., Mitchell, S. N., Witkin, J. M.,Thompson, R. C., Christopoulos, A., Lazareno, S., Birdsall, N. J. M., By-master, F. P., and Felder, C. C. (2008) Allosteric modulation of the mus-carinic M4 receptor as an approach to treating schizophrenia. Proc. Natl.Acad. Sci. U.S.A. 105, 10978 –10983

47. Leach, K., Loiacono, R. E., Felder, C. C., McKinzie, D. L., Mogg, A., Shaw,D. B., Sexton, P. M., and Christopoulos, A. (2010) Molecular mechanismsof action and in vivo validation of an M4 muscarinic acetylcholine recep-tor allosteric modulator with potential antipsychotic properties. Neuro-psychopharmacology 35, 855– 869

48. Steinfeld, T., Mammen, M., Smith, J. A. M., Wilson, R. D., and Jasper, J. R.(2007) A novel multivalent ligand that bridges the allosteric and or-thosteric binding sites of the M2 muscarinic receptor. Mol. Pharmacol.72, 291–302

49. Christopoulos, A., Lanzafame, A., and Mitchelson, F. (1998) Allostericinteractions at muscarinic cholinoceptors. Clin. Exp. Pharmacol. Physiol.25, 185–194

50. Ellis, J., and Seidenberg, M. (2000) Interactions of alcuronium, TMB-8,and other allosteric ligands with muscarinic acetylcholine receptors:studies with chimeric receptors. Mol. Pharmacol. 58, 1451–1460

51. Lazareno, S., Gharagozloo, P., Kuonen, D., Popham, A., and Birdsall, N. J.(1998) Subtype-selective positive cooperative interactions between bruc-ine analogues and acetylcholine at muscarinic receptors: radioligandbinding studies. Mol. Pharmacol. 53, 573–589

52. Lazareno, S., Popham, A., and Birdsall, N. J. (2000) Allosteric interactionsof staurosporine and other indolocarbazoles with N-[methyl-3H]scopol-amine and acetylcholine at muscarinic receptor subtypes: identificationof a second allosteric site. Mol. Pharmacol. 58, 194 –207

53. Lazareno, S., Popham, A., and Birdsall, N. J. M. (2002) Analogs of WIN62,577 define a second allosteric site on muscarinic receptors. Mol. Phar-macol. 62, 1492–1505

54. Lanzafame, A. A., Sexton, P. M., and Christopoulos, A. (2006) Interactionstudies of multiple binding sites on M4 muscarinic acetylcholine recep-tors. Mol. Pharmacol. 70, 736 –746

55. Sullivan, S. K., Brown, M. S., Gao, Y., Loweth, C. J., Lio, F. M., Crowe,P. D., Struthers, R. S., and Betz, S. F. (2006) Allosteric and orthostericbinding modes of two nonpeptide human gonadotropin-releasing hor-mone receptor antagonists. Biochemistry 45, 15327–15337

56. Heitman, L. H., Ye, K., Oosterom, J., and Ijzerman, A. P. (2008) Amiloridederivatives and a nonpeptidic antagonist bind at two distinct allostericsites in the human gonadotropin-releasing hormone receptor. Mol.Pharmacol. 73, 1808 –1815

57. Nicholls, D. J., Tomkinson, N. P., Wiley, K. E., Brammall, A., Bowers, L.,Grahames, C., Gaw, A., Meghani, P., Shelton, P., Wright, T. J., and

MINIREVIEW: Novel Allosteric Modulators of GPCRs

19486 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 290 • NUMBER 32 • AUGUST 7, 2015

by guest on February 20, 2018http://w

ww

.jbc.org/D

ownloaded from

Page 10: Novel Allosteric Modulators of G Protein-coupled Receptors*

Mallinder, P. R. (2008) Identification of a putative intracellular allostericantagonist binding-site in the CXC chemokine receptors 1 and 2. Mol.Pharmacol. 74, 1193–1202

58. Andrews, G., Jones, C., and Wreggett, K. A. (2008) An intracellular allos-teric site for a specific class of antagonists of the CC chemokine G pro-tein-coupled receptors CCR4 and CCR5. Mol. Pharmacol. 73, 855– 867

59. Salchow, K., Bond, M. E., Evans, S. C., Press, N. J., Charlton, S. J., Hunt,P. A., and Bradley, M. E. (2010) A common intracellular allosteric bindingsite for antagonists of the CXCR2 receptor. Br. J. Pharmacol. 159,1429 –1439

60. Hollenberg, M. D., Saifeddine, M., Sandhu, S., Houle, S., and Vergnolle,N. (2004) Proteinase-activated receptor-4: evaluation of tethered ligand-derived peptides as probes for receptor function and as inflammatoryagonists in vivo. Br. J. Pharmacol. 143, 443– 454

61. Kaneider, N. C., Agarwal, A., Leger, A. J., and Kuliopulos, A. (2005) Re-versing systemic inflammatory response syndrome with chemokine re-ceptor pepducins. Nat. Med. 11, 661– 665

62. Bortolato, A., Doré, A. S., Hollenstein, K., Tehan, B. G., Mason, J. S., andMarshall, F. H. (2014) Structure of Class B GPCRs: new horizons for drugdiscovery. Br. J. Pharmacol. 171, 3132–3145

63. Chen, D., Liao, J., Li, N., Zhou, C., Liu, Q., Wang, G., Zhang, R., Zhang, S.,Lin, L., Chen, K., Xie, X., Nan, F., Young, A. A, and Wang, M.-W. (2007)A nonpeptidic agonist of glucagon-like peptide 1 receptors with efficacyin diabetic db/db mice. Proc. Natl. Acad. Sci. U.S.A. 104, 943–948

64. Nolte, W. M., Fortin, J.-P., Stevens, B. D., Aspnes, G. E., Griffith, D. A.,Hoth, L. R., Ruggeri, R. B., Mathiowetz, A. M., Limberakis, C., Hepworth,D., and Carpino, P. A. (2014) A potentiator of orthosteric ligand activityat GLP-1R acts via covalent modification. Nat. Chem. Biol. 10, 629 – 631

65. Gregory, K. J., Dong, E. N., Meiler, J., and Conn, P. J. (2011) Allostericmodulation of metabotropic glutamate receptors: structural insights andtherapeutic potential. Neuropharmacology 60, 66 – 81

66. Gregory, K. J., and Conn, P. J. (2015) Molecular insights into metabo-tropic glutamate receptor allosteric modulation. Mol. Pharmacol. 88,188 –202

67. O’Brien, J. A., Lemaire, W., Wittmann, M., Jacobson, M. A., Ha, S. N.,Wisnoski, D. D., Lindsley, C. W., Schaffhauser, H. J., Rowe, B., Sur, C.,Duggan, M. E., Pettibone, D. J., Conn, P. J., and Williams, D. L. (2004) Anovel selective allosteric modulator potentiates the activity of nativemetabotropic glutamate receptor subtype 5 in rat forebrain. J. Pharma-col. Exp. Ther. 309, 568 –577

68. Chen, Y., Nong, Y., Goudet, C., Hemstapat, K., de Paulis, T., Pin, J.-P., andConn, P. J. (2007) Interaction of novel positive allosteric modulators ofmetabotropic glutamate receptor 5 with the negative allosteric antago-nist site is required for potentiation of receptor responses. Mol. Pharma-col. 71, 1389 –1398

69. Hammond, A. S., Rodriguez, A. L., Townsend, S. D., Niswender, C. M.,Gregory, K. J., Lindsley, C. W., and Conn, P. J. (2010) Discovery of a novelchemical class of mGlu5 allosteric ligands with distinct modes of phar-macology. ACS Chem. Neurosci. 1, 702–716

70. Rodriguez, A. L., Grier, M. D., Jones, C. K., Herman, E. J., Kane, A. S.,Smith, R. L., Williams, R., Zhou, Y., Marlo, J. E., Days, E. L., Blatt, T. N.,Jadhav, S., Menon, U. N., Vinson, P. N., Rook, J. M., Stauffer, S. R., Nis-wender, C. M., Lindsley, C. W., Weaver, C. D., and Conn, P. J. (2010)Discovery of novel allosteric modulators of metabotropic glutamate re-ceptor subtype 5 reveals chemical and functional diversity and in vivoactivity in rat behavioral models of anxiolytic and antipsychotic activity.Mol. Pharmacol. 78, 1105–1123

71. Lamb, J. P., Engers, D. W., Niswender, C. M., Rodriguez, A. L., Venable,D. F., Conn, P. J., and Lindsley, C. W. (2011) Discovery of molecularswitches within the ADX-47273 mGlu5 PAM scaffold that modulatemodes of pharmacology to afford potent mGlu5 NAMs, PAMs and par-tial antagonists. Bioorg. Med. Chem. Lett. 21, 2711–2714

72. Noetzel, M. J., Rook, J. M., Vinson, P. N., Cho, H. P., Days, E., Zhou, Y.,Rodriguez, A. L., Lavreysen, H., Stauffer, S. R., Niswender, C. M., Xiang,Z., Daniels, J. S., Jones, C. K., Lindsley, C. W., Weaver, C. D., and Conn,P. J. (2012) Functional impact of allosteric agonist activity of selectivepositive allosteric modulators of metabotropic glutamate receptor sub-type 5 in regulating central nervous system function. Mol. Pharmacol.

81, 120 –13373. Zhao, Z., Wisnoski, D. D., O’Brien, J. A., Lemaire, W., Williams, D. L., Jr.,

Jacobson, M. A., Wittman, M., Ha, S. N., Schaffhauser, H., Sur, C., Petti-bone, D. J., Duggan, M. E., Conn, P. J., Hartman, G. D., and Lindsley,C. W. (2007) Challenges in the development of mGluR5 positive allos-teric modulators: the discovery of CPPHA. Bioorg. Med. Chem. Lett. 17,1386 –1391

74. Chen, Y., Goudet, C., Pin, J.-P., and Conn, P. J. (2008)N-{4-Chloro-2-[(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)methyl]phe-nyl}-2-hydroxybenzamide (CPPHA) acts through a novel site as apositive allosteric modulator of group 1 metabotropic glutamate re-ceptors. Mol. Pharmacol. 73, 909 –918

75. Zhang, Y., Rodriguez, A. L., and Conn, P. J. (2005) Allosteric potentiatorsof metabotropic glutamate receptor subtype 5 have differential effects ondifferent signaling pathways in cortical astrocytes. J. Pharmacol. Exp.Ther. 315, 1212–1219

76. Noetzel, M. J., Gregory, K. J., Vinson, P. N., Manka, J. T., Stauffer, S. R.,Lindsley, C. W., Niswender, C. M., Xiang, Z., and Conn, P. J. (2013) Anovel metabotropic glutamate receptor 5 positive allosteric modulatoracts at a unique site and confers stimulus bias to mGlu5 signaling. Mol.Pharmacol. 83, 835– 847

77. Generoso, S. F., Giustiniano, M., La Regina, G., Bottone, S., Passacantilli,S., Di Maro, S., Cassese, H., Bruno, A., Mallardo, M., Dentice, M., Silves-tri, R., Marinelli, L., Sarnataro, D., Bonatti, S., Novellino, E., and Stornai-uolo, M. (2015) Pharmacological folding chaperones act as allosteric li-gands of Frizzled4. Nat. Chem. Biol. 11, 280 –286

78. Wang, C., Wu, H., Evron, T., Vardy, E., Han, G. W., Huang, X.-P., Hu-feisen, S. J., Mangano, T. J., Urban, D. J., Katritch, V., Cherezov, V., Caron,M. G., Roth, B. L., and Stevens, R. C. (2014) Structural basis for Smooth-ened receptor modulation and chemoresistance to anticancer drugs.Nat. Commun. 5, 4355

79. Bhaskar, V., Goldfine, I. D., Bedinger, D. H., Lau, A., Kuan, H. F., Gross,L. M., Handa, M., Maddux, B. A., Watson, S. R., Zhu, S., Narasimha, A. J.,Levy, R., Webster, L., Wijesuriya, S. D., Liu, N., Wu, X., Chemla-Vogel,D., Tran, C., Lee, S. R., Wong, S., Wilcock, D., White, M. L., and Corbin,J. A. (2012) A fully human, allosteric monoclonal antibody that activatesthe insulin receptor and improves glycemic control. Diabetes 61,1263–1271

80. Bhaskar, V., Lau, A., Goldfine, I. D., Narasimha, A. J., Gross, L. M., Wong,S., Cheung, B., White, M. L., and Corbin, J. A. (2013) XMetA, an allostericmonoclonal antibody to the insulin receptor, improves glycaemic controlin mice with diet-induced obesity. Diabetes Obes. Metab. 15, 272–275

81. Roell, M. K., Issafras, H., Bauer, R. J., Michelson, K. S., Mendoza, N.,Vanegas, S. I., Gross, L. M., Larsen, P. D., Bedinger, D. H., Bohmann, D. J.,Nonet, G. H., Liu, N., Lee, S. R., Handa, M., Kantak, S. S., Horwitz, A. H.,Hunter, J. J., Owyang, A. M., Mirza, A. M., Corbin, J. A., and White, M. L.(2010) Kinetic approach to pathway attenuation using XOMA 052, aregulatory therapeutic antibody that modulates interleukin-1� activity.J. Biol. Chem. 285, 20607–20614

82. Issafras, H., Corbin, J. A., Goldfine, I. D., and Roell, M. K. (2014) Detailedmechanistic analysis of gevokizumab, an allosteric anti-IL-1� antibodywith differential receptor-modulating properties. J. Pharmacol. Exp.Ther. 348, 202–215

83. Schultz Kirkegaard, H., and Valentin, F. (2014) Academic drug discoverycentres: the economic and organisational sustainability of an emergingmodel. Drug Discov. Today 19, 1699 –1710

84. Nicolaou, K. C. (2014) Advancing the drug discovery and developmentprocess. Angew Chem. Int. Ed. Engl. 53, 9128 –9140

85. Bouvier, M., Heveker, N., Jockers, R., Marullo, S., and Milligan, G. (2007)BRET analysis of GPCR oligomerization: newer does not mean better.Nat. Methods 4, 3– 4; author reply 4, 10.1038/nmeth0107-3

86. Huber, T., and Sakmar, T. P. (2014) Chemical Biology Methods for In-vestigating G Protein-Coupled Receptor Signaling. Chem. Biol. 21,1224 –1237

87. Wood, M. R., Hopkins, C. R., Brogan, J. T., Conn, P. J., and Lindsley, C. W.(2011) “Molecular switches” on mGluR allosteric ligands that modulatemodes of pharmacology. Biochemistry 50, 2403–2410

88. Davie, B. J., Valant, C., White, J. M., Sexton, P. M., Capuano, B., Christo-

MINIREVIEW: Novel Allosteric Modulators of GPCRs

AUGUST 7, 2015 • VOLUME 290 • NUMBER 32 JOURNAL OF BIOLOGICAL CHEMISTRY 19487

by guest on February 20, 2018http://w

ww

.jbc.org/D

ownloaded from

Page 11: Novel Allosteric Modulators of G Protein-coupled Receptors*

poulos, A., and Scammells, P. J. (2014) Synthesis and pharmacologicalevaluation of analogues of benzyl quinolone carboxylic acid (BQCA)designed to bind irreversibly to an allosteric site of the M1 muscarinicacetylcholine receptor. J. Med. Chem. 57, 5405–5418

89. Pittolo, S., Gómez-Santacana, X., Eckelt, K., Rovira, X., Dalton, J., Gou-det, C., Pin, J.-P., Llobet, A., Giraldo, J., Llebaria, A., and Gorostiza, P.(2014) An allosteric modulator to control endogenous G protein-cou-pled receptors with light. Nat. Chem. Biol. 10, 813– 815

90. Wootten, D., Savage, E. E., Willard, F. S., Bueno, A. B., Sloop, K. W.,Christopoulos, A., and Sexton, P. M. (2013) Differential activation andmodulation of the glucagon-like peptide-1 receptor by small moleculeligands. Mol. Pharmacol. 83, 822– 834

91. Palczewski, K., Kumasaka, T., Hori, T., Behnke, C. A., Motoshima, H.,Fox, B. A., Le Trong, I., Teller, D. C., Okada, T., Stenkamp, R. E.,Yamamoto, M., and Miyano, M. (2000) Crystal structure of rhodopsin: aG protein-coupled receptor. Science 289, 739 –745

92. Tan, Q., Zhu, Y., Li, J., Chen, Z., Han, G. W., Kufareva, I., Li, T., Ma, L.,Fenalti, G., Li, J., Zhang, W., Xie, X., Yang, H., Jiang, H., Cherezov, V., Liu,H., Stevens, R. C., Zhao, Q., and Wu, B. (2013) Structure of the CCR5chemokine receptor-HIV entry inhibitor maraviroc complex. Science341, 1387–1390

93. Hollenstein, K., Kean, J., Bortolato, A., Cheng, R. K. Y., Doré, A. S.,Jazayeri, A., Cooke, R. M., Weir, M., and Marshall, F. H. (2013) Structureof class B GPCR corticotropin-releasing factor receptor 1. Nature 499,438 – 443

94. Wu, H., Wang, C., Gregory, K. J., Han, G. W., Cho, H. P., Xia, Y., Nis-wender, C. M., Katritch, V., Meiler, J., Cherezov, V., Conn, P. J., andStevens, R. C. (2014) Structure of a class C GPCR metabotropic gluta-

mate receptor 1 bound to an allosteric modulator. Science 344, 58 – 6495. Doré, A. S., Okrasa, K., Patel, J. C., Serrano-Vega, M., Bennett, K., Cooke,

R. M., Errey, J. C., Jazayeri, A., Khan, S., Tehan, B., Weir, M., Wiggin,G. R., and Marshall, F. H. (2014) Structure of class C GPCR metabotropicglutamate receptor 5 transmembrane domain. Nature 511, 557–562

96. Rasmussen, S. G. F., DeVree, B. T., Zou, Y., Kruse, A. C., Chung, K. Y.,Kobilka, T. S., Thian, F. S., Chae, P. S., Pardon, E., Calinski, D., Mathiesen,J. M., Shah, S. T. A., Lyons, J. A., Caffrey, M., Gellman, S. H., Steyaert, J.,Skiniotis, G., Weis, W. I., Sunahara, R. K., and Kobilka, B. K. (2011)Crystal structure of the �2-adrenergic receptor-Gs protein complex. Na-ture 477, 549 –555

97. Kruse, A. C., Ring, A. M., Manglik, A., Hu, J., Hu, K., Eitel, K., Hübner, H.,Pardon, E., Valant, C., Sexton, P. M., Christopoulos, A., Felder, C. C.,Gmeiner, P., Steyaert, J., Weis, W. I., Garcia, K. C., Wess, J., and Kobilka,B. K. (2013) Activation and allosteric modulation of a muscarinic acetyl-choline receptor. Nature 504, 101–106

98. Dror, R. O., Green, H. F., Valant, C., Borhani, D. W., Valcourt, J. R., Pan,A. C., Arlow, D. H., Canals, M., Lane, J. R., Rahmani, R., Baell, J. B.,Sexton, P. M., Christopoulos, A., and Shaw, D. E. (2013) Structural basisfor modulation of a G-protein-coupled receptor by allosteric drugs. Na-ture 503, 295–299

99. Didenko, T., Liu, J. J., Horst, R., Stevens, R. C., and Wüthrich, K. (2013)Fluorine-19 NMR of integral membrane proteins illustrated with studiesof GPCRs. Curr. Opin. Struct. Biol. 23, 740 –747

100. Liu, J. J., Horst, R., Katritch, V., Stevens, R. C., and Wüthrich, K. (2012)Biased signaling pathways in �2-adrenergic receptor characterized by19F-NMR. Science 335, 1106 –1110

MINIREVIEW: Novel Allosteric Modulators of GPCRs

19488 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 290 • NUMBER 32 • AUGUST 7, 2015

by guest on February 20, 2018http://w

ww

.jbc.org/D

ownloaded from

Page 12: Novel Allosteric Modulators of G Protein-coupled Receptors*

Patrick R. Gentry, Patrick M. Sexton and Arthur ChristopoulosNovel Allosteric Modulators of G Protein-coupled Receptors

doi: 10.1074/jbc.R115.662759 originally published online June 22, 20152015, 290:19478-19488.J. Biol. Chem. 

  10.1074/jbc.R115.662759Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

  http://www.jbc.org/content/290/32/19478.full.html#ref-list-1

This article cites 100 references, 46 of which can be accessed free at

by guest on February 20, 2018http://w

ww

.jbc.org/D

ownloaded from


Recommended