+ All Categories
Home > Documents > Structure of G i1 Bound to a GDP-Selective Peptide Provides … · 2017. 2. 14. · 752 GEF...

Structure of G i1 Bound to a GDP-Selective Peptide Provides … · 2017. 2. 14. · 752 GEF...

Date post: 04-Sep-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
12
Structure, Vol. 13, 1069–1080, July, 2005, ©2005 Elsevier Ltd All rights reserved. DOI 10.1016/j.str.2005.04.007 Structure of G i1 Bound to a GDP-Selective Peptide Provides Insight into Guanine Nucleotide Exchange Christopher A. Johnston, 1 Francis S. Willard, 1,2,3 membrane domain G protein-coupled receptors (GPCRs) are associated with inactive, membrane-tethered G Mark R. Jezyk, 4 Zoey Fredericks, 5,7 Erik T. Bodor, 1 protein heterotrimers consisting of Gα,GDP bound to Miller B. Jones, 1,2,3 Rainer Blaesius, 5,8 Val J. Watts, 6 Gβγ.Gβγ facilitates the receptor coupling of Gα,GDP, T. Kendall Harden, 1 John Sondek, 1,2,4 stabilizes its GDP bound state, and prevents spontane- J. Kevin Ramer, 5,9 and David P. Siderovski 1,2,3, * ous nucleotide exchange, thus serving as a guanine nu- 1 Department of Pharmacology cleotide dissociation inhibitor (GDI) (Higashijima et al., 2 Lineberger Comprehensive Cancer Center 1987). Ligand-activated GPCRs serve as guanine nu- 3 UNC Neuroscience Center cleotide exchange factors (GEFs), catalyzing exchange 4 Department of Biochemistry & Biophysics of GDP for GTP on Gα. GTP binding alters the confor- The University of North Carolina at Chapel Hill mation of three flexible “switch” regions within Gα, Chapel Hill, North Carolina 27599 leading to Gβγ dissociation. Both Gα,GTP and Gβγ 5 Karo*Bio USA subsequently regulate several downstream effectors, Durham, North Carolina 27703 including adenylyl cyclases, phospholipases, kinases, 6 Department of Medicinal Chemistry and ion channels (Cabrera-Vera et al., 2003; McCudden and Molecular Pharmacology et al., 2005). Based on sequence similarity and func- School of Pharmacy tional differences in effector regulation, G proteins are Purdue University grouped into four distinct families: Gα i/o ,Gα s ,Gα q/11 , West Lafayette, Indiana 47907 and Gα 12/13 (Cabrera-Vera et al., 2003; McCudden et al., 2005). Signal termination is achieved by the intrinsic GTP hydrolysis activity of Gα and is accelerated by Summary “regulators of G protein signaling” proteins (RGS pro- teins; Neubig and Siderovski, 2002). Formation of Heterotrimeric G proteins are molecular switches that Gα,GDP causes heterotrimer reassociation, thereby regulate numerous signaling pathways involved in preventing further effector interactions by either Gα or cellular physiology. This characteristic is achieved by Gβγ. Accordingly, the duration of G protein signaling is the adoption of two principal states: an inactive, GDP determined by the lifetime of Gα in the GTP bound state bound state and an active, GTP bound state. Under (Sprang, 1997). Thus, G proteins serve as temporal reg- basal conditions, G proteins exist in the inactive, GDP ulators of signaling pathways, and understanding the bound state; thus, nucleotide exchange is crucial to molecular determinants of their guanine nucleotide cy- the onset of signaling. Despite our understanding of cle is of particular interest. G protein signaling pathways, the mechanism of nu- Structures of Gα subunits, including Gα i1 , in both in- cleotide exchange remains elusive. We employed active and activated states have revealed critical con- phage display technology to identify nucleotide state- formational changes that occur during GTP binding and dependent G binding peptides. Herein, we report a hydrolysis (Sprang, 1997). Gα consists of a Ras-like do- GDP-selective G binding peptide, KB-752, that en- main, a structural fold present in many GTPases, and a hances spontaneous nucleotide exchange of G i sub- unique all-helical domain. Bound nucleotide resides in units. Structural determination of the G i1 /peptide a cleft between these two domains. Although flexibility complex reveals unique changes in the G switch re- between these domains is thought to govern the rate gions predicted to enhance nucleotide exchange by of spontaneous nucleotide exchange (Remmers et al., creating a GDP dissociation route. Our results cast 1999), the mechanism whereby Gα GEFs induce nucle- light onto a potential mechanism by which G sub- otide exchange is not yet clear. Two distinct types of Gα GEFs are now known: membrane bound GPCRs units adopt a conformation suitable for nucleotide ex- and the soluble cytoplasmic RIC-8 proteins. The struc- change. ture of the prototypical GPCR rhodopsin provided the first structural glimpse of the most prominent class of Introduction Gα GEFs (Palczewski et al., 2000); however, the recep- tor was in an inactive form and was not bound to het- Heterotrimeric G proteins are crucial intracellular medi- erotrimer. Thus, little direct information was gained ators of a diverse array of extracellular signals, includ- about the mechanism for G protein activation. The non- ing hormones, photons, odorants, and small molecules receptor Gα GEF RIC-8 is widely conserved across (Cabrera-Vera et al., 2003; McCudden et al., 2005). In metazoa as a critical determinant (along with Gα i sub- the standard model of G protein signaling, seven trans- units) in mitotic spindle force generation during mitosis (reviewed in McCudden et al., 2005). Unlike the GEFs *Correspondence: [email protected] for Ras superfamily GTPases, such as the RhoGEF 7 Present address: Amgen, Inc., 1201 Amgen Court West, Seattle, family (Rossman et al., 2005), that have no preference Washington 98119. for nucleotide state (GDP or GTP bound), RIC-8 exhib- 8 Present address: Becton Dickinson, 21 Davis Drive, Research Tri- its selective interaction with the GDP bound state of angle Park, North Carolina 27709. Gα subunits and does not bind nor act as a GEF toward 9 Present address: Hemocellular Therapeutics, 5312 Farrington Road, Chapel Hill, North Carolina 27517. Gα,GTP (Afshar et al., 2004; Tall et al., 2003). As RIC-8
Transcript
Page 1: Structure of G i1 Bound to a GDP-Selective Peptide Provides … · 2017. 2. 14. · 752 GEF activity, we determined the crystal structure of 50% maximal response (EC 50) for KB-752

Structure, Vol. 13, 1069–1080, July, 2005, ©2005 Elsevier Ltd All rights reserved. DOI 10.1016/j.str.2005.04.007

Structure of G�i1 Bound to a GDP-Selective PeptideProvides Insight into Guanine Nucleotide Exchange

Christopher A. Johnston,1 Francis S. Willard,1,2,3

Mark R. Jezyk,4 Zoey Fredericks,5,7 Erik T. Bodor,1

Miller B. Jones,1,2,3 Rainer Blaesius,5,8 Val J. Watts,6

T. Kendall Harden,1 John Sondek,1,2,4

J. Kevin Ramer,5,9 and David P. Siderovski1,2,3,*1Department of Pharmacology2Lineberger Comprehensive Cancer Center3UNC Neuroscience Center4Department of Biochemistry & BiophysicsThe University of North Carolina at Chapel HillChapel Hill, North Carolina 275995Karo*Bio USADurham, North Carolina 277036Department of Medicinal Chemistry

and Molecular PharmacologySchool of PharmacyPurdue UniversityWest Lafayette, Indiana 47907

Summary

Heterotrimeric G proteins are molecular switches thatregulate numerous signaling pathways involved incellular physiology. This characteristic is achieved bythe adoption of two principal states: an inactive, GDPbound state and an active, GTP bound state. Underbasal conditions, G proteins exist in the inactive, GDPbound state; thus, nucleotide exchange is crucial tothe onset of signaling. Despite our understanding ofG protein signaling pathways, the mechanism of nu-cleotide exchange remains elusive. We employedphage display technology to identify nucleotide state-dependent G� binding peptides. Herein, we report aGDP-selective G� binding peptide, KB-752, that en-hances spontaneous nucleotide exchange of G�i sub-units. Structural determination of the G�i1/peptidecomplex reveals unique changes in the G� switch re-gions predicted to enhance nucleotide exchange bycreating a GDP dissociation route. Our results castlight onto a potential mechanism by which G� sub-units adopt a conformation suitable for nucleotide ex-change.

Introduction

Heterotrimeric G proteins are crucial intracellular medi-ators of a diverse array of extracellular signals, includ-ing hormones, photons, odorants, and small molecules(Cabrera-Vera et al., 2003; McCudden et al., 2005). Inthe standard model of G protein signaling, seven trans-

*Correspondence: [email protected]

7 Present address: Amgen, Inc., 1201 Amgen Court West, Seattle,Washington 98119. 8 Present address: Becton Dickinson, 21 Davis Drive, Research Tri-angle Park, North Carolina 27709. 9 Present address: Hemocellular Therapeutics, 5312 FarringtonRoad, Chapel Hill, North Carolina 27517.

membrane domain G protein-coupled receptors (GPCRs)are associated with inactive, membrane-tethered Gprotein heterotrimers consisting of Gα,GDP bound toGβγ. Gβγ facilitates the receptor coupling of Gα,GDP,stabilizes its GDP bound state, and prevents spontane-ous nucleotide exchange, thus serving as a guanine nu-cleotide dissociation inhibitor (GDI) (Higashijima et al.,1987). Ligand-activated GPCRs serve as guanine nu-cleotide exchange factors (GEFs), catalyzing exchangeof GDP for GTP on Gα. GTP binding alters the confor-mation of three flexible “switch” regions within Gα,leading to Gβγ dissociation. Both Gα,GTP and Gβγsubsequently regulate several downstream effectors,including adenylyl cyclases, phospholipases, kinases,and ion channels (Cabrera-Vera et al., 2003; McCuddenet al., 2005). Based on sequence similarity and func-tional differences in effector regulation, G proteins aregrouped into four distinct families: Gαi/o, Gαs, Gαq/11,and Gα12/13 (Cabrera-Vera et al., 2003; McCudden etal., 2005). Signal termination is achieved by the intrinsicGTP hydrolysis activity of Gα and is accelerated by“regulators of G protein signaling” proteins (RGS pro-teins; Neubig and Siderovski, 2002). Formation ofGα,GDP causes heterotrimer reassociation, therebypreventing further effector interactions by either Gα orGβγ. Accordingly, the duration of G protein signaling isdetermined by the lifetime of Gα in the GTP bound state(Sprang, 1997). Thus, G proteins serve as temporal reg-ulators of signaling pathways, and understanding themolecular determinants of their guanine nucleotide cy-cle is of particular interest.

Structures of Gα subunits, including Gαi1, in both in-active and activated states have revealed critical con-formational changes that occur during GTP binding andhydrolysis (Sprang, 1997). Gα consists of a Ras-like do-main, a structural fold present in many GTPases, and aunique all-helical domain. Bound nucleotide resides ina cleft between these two domains. Although flexibilitybetween these domains is thought to govern the rateof spontaneous nucleotide exchange (Remmers et al.,1999), the mechanism whereby Gα GEFs induce nucle-otide exchange is not yet clear. Two distinct types ofGα GEFs are now known: membrane bound GPCRsand the soluble cytoplasmic RIC-8 proteins. The struc-ture of the prototypical GPCR rhodopsin provided thefirst structural glimpse of the most prominent class ofGα GEFs (Palczewski et al., 2000); however, the recep-tor was in an inactive form and was not bound to het-erotrimer. Thus, little direct information was gainedabout the mechanism for G protein activation. The non-receptor Gα GEF RIC-8 is widely conserved acrossmetazoa as a critical determinant (along with Gαi sub-units) in mitotic spindle force generation during mitosis(reviewed in McCudden et al., 2005). Unlike the GEFsfor Ras superfamily GTPases, such as the RhoGEFfamily (Rossman et al., 2005), that have no preferencefor nucleotide state (GDP or GTP bound), RIC-8 exhib-its selective interaction with the GDP bound state ofGα subunits and does not bind nor act as a GEF towardGα,GTP (Afshar et al., 2004; Tall et al., 2003). As RIC-8

Page 2: Structure of G i1 Bound to a GDP-Selective Peptide Provides … · 2017. 2. 14. · 752 GEF activity, we determined the crystal structure of 50% maximal response (EC 50) for KB-752

Structure1070

proteins have only been recently discovered, structural tostudies of these proteins have yet to be reported. Thus,

the structural determinants of Gα activation by GEFs tsremain largely unknown.

Phage display is a powerful technique in identifying tbsmall peptides capable of binding desired targets in an

unbiased manner. Identified peptides can then serve as �stools for studying target protein binding surfaces, pro-

tein-protein interaction sites, and protein function and mfregulation (reviewed in Rodi et al., 2002). This technol-

ogy has identified peptide modulators of a variety of r(enzyme classes and signaling molecules (e.g., Ashraf et

al., 2003; Hyde-DeRuyscher et al., 2000). In particular, dsphage display and similar approaches have been used

to investigate G protein binding interfaces on GPCRs Ki(Gilchrist et al., 1998; Martin et al., 1996) and effector

binding regions on Gβγ subunits (Scott et al., 2001), as Gwell as to identify peptides with G protein regulatoryproperties, including both GEF and GDI activities (Hes- Ksling et al., 2003; Ja and Roberts, 2004). In the present Nstudy, we have identified guanine nucleotide-depen- Tdent Gα binding peptides from a phage display peptide clibrary. In particular, we describe the guanine nucleotide pexchange factor activity of a GDP-selective peptide, otermed KB-752. To understand the mechanism of KB- r752 GEF activity, we determined the crystal structure of 5the peptide bound to Gαi1. To our knowledge, these ostudies are the first to describe the structure of a Gα isubunit in complex with a GEF and provide direct struc- btural evidence in support of a previously proposed tmechanism for the GPCR-catalyzed nucleotide ex- schange reaction. t

nResults(rIdentification of Nucleotide-DependentsG� Binding PeptidesGWe used phage display to obtain peptides that recog-nnize the distinct conformations of Gα when bound toiGDP versus GTPγS (Cabrera-Vera et al., 2003; Sprang,m1997). Biotinylated Gαi1,GDP and Gαi1,GTPγS were in-edependently immobilized onto streptavidin-coated mi-fcrotiter plates for selection of phage-displayed pep-ctides. Phage selectivity was monitored by comparingmphage ELISA signals between wells containing Gαi1tand wells blocked with albumin. After four iterative

rounds of selection, clonal bacteriophage isolates wereGpurified, amplified, and screened for selective bindingito Gαi1 in GDP or GTPγS bound states (e.g., Figure 1A).aIn total, we isolated 51 GDP-dependent, 12 GTPγS-odependent, and 5 nucleotide state-independent phage-odisplayed peptides. Extensive database searchesKsuggest that none of these peptides has sequence sim-(ilarity to known Gα interacting proteins (data notfshown).TA representative group of GDP-dependent phagess(Figure 1B) showed strong sequence similarity around

the motif TWXE/DFL. Of these GDP-selective peptides,we focused initially on KB-752. Nucleotide-dependent S

TGα binding was quantitated by surface plasmon reso-nance (SPR) measurements on a streptavidin biosensor a

Gchip coated with biotinylated KB-752 (e.g., Figure 1Cfor Gα ). Dissociation constants (K values) were ob- a

i1 d

ained by simultaneous kinetic analysis of on (ka) andff (kd) rates obtained by injecting increasing concen-rations of Gα in GDP, GDP,AlF4

−, and GTPγS boundtates (e.g., Figure 1D for Gαi1,GDP). In agreement withhe phage selection, KB-752 displayed highest-affinityinding to Gαi1 in its GDP bound form (Kd of 3.9 ± 0.6M). No appreciable binding was observed to the tran-ition state-mimetic form of Gαi1,GDP,AlF4

−, althougheasurable (albeit low-affinity) binding was observed

or Gαi1,GTPγS (Kd of 28.0 ± 3.2 �M); given the slowate of spontaneous nucleotide exchange of Gαi1

Fields and Casey, 1997), this observed binding may beue to residual GDP bound protein. KB-752 demon-trated lower affinity for the closely related Gαo, with ad of 18.2 ± 3.0 �M for Gαo,GDP, but no measurable

nteraction with GDP,AlF4− nor GTPγS bound forms of

αo (data not shown).

B-752 Binding Affects Guanineucleotide Exchangeo examine the effects of KB-752 on nucleotide ex-hange by Gα binding partners, [35S]GTPγS binding tourified Gα was quantified in the absence or presencef peptide. KB-752 enhanced the nucleotide exchangeate of Gαi1 (Figure 2A); the effective concentration for0% maximal response (EC50) for KB-752 GEF activityn Gαi1 was 5.6 ± 1.1 �M (Figure 2B), comparable to

ts observed Kd. Equipotent GEF activity was found foroth Gαi2 and Gαi3 (Figure 2B). KB-752 did not affect

he nucleotide exchange rate of Gαi2β1γ2 (Figure 2C),uggesting that KB-752 cannot disrupt a native hetero-rimer and interacts solely with free Gα.

Despite binding to Gαo,GDP, KB-752 did not affectucleotide exchange even at saturating concentrations

Figure 2D). We hypothesized that the higher intrinsicate of spontaneous nucleotide exchange of Gαo ver-us Gαi contributes to the lack of KB-752 activity onαo. To test this, we purified Gαi1 containing an argi-ine 144 to alanine (R144A) mutation that disrupts an

nteraction between the all-helical and Ras-like do-ains and thus renders the spontaneous exchange rate

quivalent to that of Gαo (Remmers et al., 1999) (con-irmed in Figure 3A). KB-752 did not enhance the ex-hange rate of Gαi1(R144A) (Figure 3B), highlighting theechanism of KB-752 as enhancing GDP release from

he intrinsically slow-exchanger Gαi.To validate these results, we employed steady-stateTPase assays. Given that GDP release is the rate-lim-

ting step of the Gα guanine nucleotide cycle, any alter-tion of GDP release, either positively (i.e., GEF activity)r negatively (i.e., GDI activity), will be reflected in theverall steady-state rate of GTP hydrolysis (Ross, 2002).B-752 enhanced steady-state GTP hydrolysis by Gαi1

Figure 3C), further indicating that it has GEF activityor Gαi subunits. No effect of KB-752 was seen on Gαo.hese results support the conclusion that KB-752 pos-esses Gαi-selective GEF activity.

tructure of KB-752 Bound to G�i1

o ascertain the molecular mechanism of KB-752 GEFctivity, we determined the structure of KB-752 boundαi1,GDP (PDB ID 1Y3A; Figure 4 and Table 1). KB-752ssumes a partial α-helical structure and binds Gα be-

i1
Page 3: Structure of G i1 Bound to a GDP-Selective Peptide Provides … · 2017. 2. 14. · 752 GEF activity, we determined the crystal structure of 50% maximal response (EC 50) for KB-752

Gαi Binding Peptide with GEF Activity1071

Figure 1. Identification of Nucleotide-Depen-dent Gαi Binding Peptides via Phage Display

(A) Representative phage ELISA results indi-cating the identification of (i) GDP-selective,(ii) GTPγS-selective, and (iii) nucleotidestate-independent Gαi1 binding peptides.(B) Sequences of 12 isolated peptides withGDP-selective binding to Gαi1, sharing aconsensus TWXE/DFL motif with the particu-lar peptide used in this study: KB-752.(C) Nucleotide-dependent binding of KB-752as measured by surface plasmon resonance(SPR). 5 �M Gαi1 protein (“analyte”), in eachof three nucleotide bound states, was in-jected over immobilized, biotinylated KB-752. Nonspecific binding to a control peptidewas subtracted from each curve.(D) GDP bound Gαi1 was injected at each in-dicated concentration over immobilized KB-752 to determine the dissociation constant(Kd) for this interaction pair. SPR-deriveddissociation constants for the interaction ofKB-752 with Gαi1 and Gαo, in their ground

state (GDP bound), transition state-mimetic (GDP,AlF4− bound), and activated state (GTPγS bound) forms, were obtained from analyses (n =

4–6 for each state) similar to that shown in (D). Dissociation constants of >1000 �M were obtained for both Gα subunits in their GDP,AlF4−

bound form, and for Gαo bound to GTPγS.

Gαi1,GDP/KB-752 structure and creates part of a criti- all-helical domain of Gαo (Remmers et al., 1999); KB-

Figure 2. KB-752 Is a Selective Guanine Nu-cleotide Exchange Factor for Gαi Subunits

(A) KB-752 (10 �M) enhances the GTPγSbinding rate of Gαi1,GDP (50 nM); rate con-stants at 30°C: Gαi1 alone = 0.029 ± 0.006min−1, Gαi1 + KB-752 = 0.086 ± 0.008 min−1.(B) KB-752 is equipotent as a GEF on allthree Gαi members. 50 nM Gαi1, Gαi2, or Gαi3

was incubated with the indicated concen-trations of KB-752, and the amount of[35S]GTPγS binding was measured after 10min at 30°C and expressed as a percentageof maximal GTPγS binding.(C and D) KB-752 does not alter the rate ofGTPγS binding by (C) Gαi-heterotrimer Gαi2,GDP/Gβ1γ2 (peptide and protein amounts asin [A]), nor (D) isolated Gαo,GDP. For thedose-response curve of (D), 50 nM Gαi1 orGαo was incubated in the presence of theindicated concentrations of KB-752, and theamount of [35S]GTPγS binding was mea-sured (after 10 min at 30°C for Gαi1; after 5min at 20°C for Gαo) as described in the

Experimental Procedures and is expressed as the percentage of GTPγS bound in the absence of KB-752. The EC50 value for GEF activity onGαi1 was 5.6 ± 1.1 �M.Data shown in (A) and (C) are the mean ± standard deviation of triplicate samples from a representative experiment of 3–5 independentexperiments conducted.

tween switch II and the α3 helix of the Ras-like domain(Figures 4 and 5A). The repositioning of switch II affordsthe binding groove for KB-752, as the α3 helix is notsignificantly altered in conformation compared to otherstructures of Gαi1. Indeed, the ability to repositionswitch II likely defines the nucleotide specificity ofKB-752 binding, given predicted steric hindrance be-tween the N terminus of KB-752 and switch II withinGαi1,GTPγS and Gαi1,GDP,AlF4

− (Figure 5B). In partic-ular, the positioning of tryptophan 211 of switch IIwould not accommodate tryptophan 5 of KB-752 (Fig-ure 5B); however, tryptophan 211 is repositioned in the

cal hydrophobic pocket used by KB-752 for binding(see below).

The switch II/α3 helix binding pocket for KB-752 issimilar to that of the N-terminal α helix of the RGS14GoLoco motif, a short polypeptide that displays GDIactivity toward Gαi1 (PDB ID 1KJY) (Kimple et al., 2002);however, the GoLoco motif binding site extends intothe all-helical domain (Figure 5C), whereas KB-752makes no contacts with this region of Gαi1 (Figure 5A).The lack of functional contacts made between KB-752and the all-helical domain was validated by using a chi-mera (“Gαioi”) with the Ras-like domain of Gαi1 but the

Page 4: Structure of G i1 Bound to a GDP-Selective Peptide Provides … · 2017. 2. 14. · 752 GEF activity, we determined the crystal structure of 50% maximal response (EC 50) for KB-752

Structure1072

7otafvGGUi(mstdprrm

STFGwhGdFwti(C(u(rtdmwc(nS

Figure 3. KB-752 GEF Activity Increases Steady-State GTP Hydrol- dysis by Gαi1, but Does Not Act on a Gαi1 Point Mutant with Ac- mcelerated Spontaneous Nucleotide Release

c(A) 200 nM wild-type (wt) Gαi1, R144A Gαi1, or wt Gαo was added to

mcuvettes containing 1 �M BODIPY-FL-GTPγS. Real-time nucleotidedbinding (Kimple et al., 2004) was measured at 25°C as an increaseKin fluorescence response (λex = 485 nm; λem = 530 nm; slit widths

of 3.0 nm) upon binding BODIPY-FL-GTPγS. Mutation of arginine w144 to alanine (R144A) resulted in Gαi1 nucleotide binding kineticsindistinguishable from that of Gαo, as previously reported by gRemmers and colleagues (Remmers et al., 1999).

c(B) KB-752 does not alter the rate of GTPγS binding by the mutantcGαi1 subunit (R144A) with accelerated spontaneous nucleotide ex-

change comparable to that of wild-type Gαo (experiment performedas in Figure 2A, except conducted at 20°C). Data shown are themean ± standard deviation of triplicate samples from a representa-

ttive experiment of three independent experiments conducted.s(C) Confirming the GEF activity of KB-752 on wild-type Gαi1, addi-Gtion of KB-752 (100 �M) to Gα (200 nM) enhances the steady-stateihydrolysis of [γ-32P]GTP by Gαi1, but has no effect on Gαo. Note

ine to eliminate the ionic interaction with R208. W5

hat the rate-limiting step in steady-state hydrolysis of GTP by Gαubunits is release of product (i.e., GDP) and not the hydrolysis ofTP per se (Ross, 2002). Data shown are the mean ± SEM for five

ndependent experiments conducted in triplicate.

52 displays GEF activity on this chimera equal to thatn wild-type Gαi1 (Figure 5D), suggesting that interac-ions with the Ras-like domain are sufficient for GEFctivity. The use of a switch II/α3 helix binding pocketor both KB-752 and GoLoco motif peptides was alsoalidated biochemically. The GEF activity of KB-752 onαi1 was found to be competitively antagonized by theoLoco motif of the RGS14 paralog, RGS12 (Figure 5E).nlike the GoLoco motif, which lies over the GDP bind-

ng pocket and uses an arginine finger to stabilize GDPKimple et al., 2002), KB-752 does not occlude nor

ake contact with GDP (Figure 5A versus Figure 5C),uggesting that its GEF activity relies on conforma-ional changes induced within Gαi1. In support of theseistinct modes of interaction about the GDP bindingocket, KB-752 binding has almost no effect on theate by which Gαi1 is activated by aluminum tetrafluo-ide (Figure 5F), unlike the inhibitory effect of GoLocootif peptides (Willard et al., 2004).

tructural Basis for the ConservedWXE/DFL Binding Motifigure 6 shows specific contacts between KB-752 andαi1. Glutamate 11 (E11) of KB-752 forms a salt bridgeith R208 of Gαi1. Tryptophan 5 (W5) is found within aydrophobic pocket formed by F215, L249, and I253 ofαi1. Phenylalanine 8 (F8) is also placed within a hy-rophobic environment established by W211, I212, and215 of Gαi1. The burial of large hydrophobic residuesithin the hydrophobic groove between switch II and

he α3 helix is common among several known Gα bind-ng partners: p115RhoGEF-RGS inserts a methionineM165) into the Gαi/13 chimera (Chen et al., 2005), the2 domain of adenylyl cyclase inserts a phenylalanine

F991) into Gαs (Tesmer et al., 1997b), and the γ sub-nit of cGMP-phosphodiesterase inserts a tryptophan

W70) into Gαt (Slep et al., 2001). Burial of the peptideesidues W5 and F8 within Gαi1 validates the results ofhe phage selection, as these two hydrophobic resi-ues figure prominently within the TWXE/DFL bindingotif (Figure 1B). An intramolecular hydrogen bond net-ork between threonine 4 (T4) and both the side chainarboxylate and peptide bond nitrogen of aspartate 7D7) (Figure 6B) underscores the conservation of threo-ine and acidic residues within the TWXE/DFL motif.pecifically, the side chain hydroxyl of T4 forms a hy-rogen bond with both the side chain carboxylate andain chain amide nitrogen of D7, and the main chain

arbonyl oxygen of T4 forms a hydrogen bond with theain chain amide nitrogen of D7. Additionally, this hy-rogen bonding network within the α-helical portion ofB-752 serve to orient both W5 and F8 side chains to-ard the Gα binding face of the peptide.Based on contacts between KB-752 and Gαi1, we

enerated three KB-752 variants to validate biochemi-ally the structural model. E11 was replaced with leu-

Page 5: Structure of G i1 Bound to a GDP-Selective Peptide Provides … · 2017. 2. 14. · 752 GEF activity, we determined the crystal structure of 50% maximal response (EC 50) for KB-752

Gαi Binding Peptide with GEF Activity1073

Figure 4. Stereoview of Experimental Electron Density for the KB-752 Peptide Bound to Gαi1

The region highlighted is the entire peptide density (model in red; labels in white) found between switch II (α2 helix) and the α3 helix of Gαi1

(model in green; labels in yellow). Shown is a 2Fo − Fc simulated annealing composite omit map (generated with 5% overall model omitted)contoured at 1σ with electron density shown in white cage.

and F8 of the TWXE/DFL motif were each independentlyreplaced with alanine to reduce the potential for burialwithin hydrophobic environments created by switch IIand the α3 helix. We first confirmed by SPR that eachmutation abrogated Gαi1 binding. Gαi1,GDP was capa-ble of interacting with the E11L peptide, but bindingwas significantly attenuated compared to wild-type(Figure 6C). Both W5A and F8A peptides displayed anear complete loss of binding to Gαi1,GDP. We thentested the ability of each peptide to enhance nucleotideexchange by Gαi1,GDP. Wild-type KB-752 resultedin an approximately 3-fold increase in the rate of[35S]GTPγS binding. The E11L peptide had diminishedGEF activity compared to wild-type (Figure 6D), whileW5A and F8A peptides lacked significant GEF activity.These results corroborate the critical contacts madebetween Gαi1 and these residues of KB-752 in thestructural model.

Structural Basis for KB-752 GEF ActivityExchange of GDP for GTP results in movement ofthe three switch regions to stabilize bound GTP andadopt the conformation responsible for effector bind-ing (Sprang, 1997). Gαi1,GDP/KB-752 possesses sig-nificant alterations in each switch region compared toGα,GDP/Gβ1γ2 (Figure 7A). Most apparent is switch II,which is displaced down and outward compared to theGαi1,GDP/Gβ1γ2 structure (Wall et al., 1995). This move-ment results in the lip of switch II, normally ordered andhelical in the GTPγS bound state (Coleman et al., 1994;Sunahara et al., 1997), being displaced away from thenucleotide binding pocket and GDP. This conformation

in Gαi1,GDP/KB-752 contrasts with the movement ofswitch II toward the nucleotide pocket when GTPγS isbound. Switch III is also slightly displaced from GDPwithin KB-752 bound Gαi1 compared to the hetero-trimer (Figure 7A). However, of the four Gαi1/KB-752 di-mers in the asymmetric unit (Table 1), only one Gαi1

molecule (chain B of PDB ID 1Y3A) had sufficientelectron density to accurately model the switch III loop,suggesting that this region of Gαi1 is inherently flexibleeven when bound to KB-752. Similar alterations toboth switch regions II and III are seen in GoLoco boundGαi1 (Gαi1,GDP/R14GL) (Kimple et al., 2002); however,switch II is more dramatically displaced in Gαi1,GDP/KB-752 (Figure 7B). Interestingly, despite movement inswitch II, the β3/α2 loop at the entry to switch II is notsignificantly displaced in the Gαi1,GDP/R14GL struc-ture compared to the Gαi1,GDP/Gβ1γ2 structure (Figure7A versus Figure 7B). In contrast, this β3/α2 loop is re-moved from the guanine nucleotide pocket along withswitch II in the Gαi1,GDP/KB-752 structure (Figure 7B).Displacement of the β3/α2 loop is stabilized throughseveral interactions with KB-752, including hydrogenbonding between the carbonyl oxygen of glycine 202of the β3/α2 loop and the indole nitrogen of tryptophan5 in KB-752 (Figure 7C), indicating an additional role forthis key peptide residue. The displacement of switch IIpositions the catalytic glutamine 204 residue far fromthe nucleotide binding pocket compared to structuresof Gαi1,GTPγS and Gαi1,GDP,AlF4

− (Figure 8), and thisresidue makes an intramolecular bond with valine 201(Figure 7C).

Switch I within Gα ,GDP/KB-752 adopts a confor-

i1
Page 6: Structure of G i1 Bound to a GDP-Selective Peptide Provides … · 2017. 2. 14. · 752 GEF activity, we determined the crystal structure of 50% maximal response (EC 50) for KB-752

Structure1074

uTable 1. Data Collection and Refinement Statisticst

Data Collectional

Space group P21 nNumber of molecules per asymmetric unit 4 tUnit cell dimensions e

a, b, c (Å) 72.9, 112.8, 109.5 tα, β, γ (°) 90, 93.8, 90GWavelength (Å) 1.0093aResolution (Å) 50–2.5 (2.59–2.5)

Rsymm (%) 26.6 cLinear R factorb 0.072 (0.266) nSquare R factorc 0.065 (0.232) t<I/σI>d 24 (3.6) uCompleteness (%) 96.4 (89.6)

MRedundancy 3.5s

Refinement

Resolution (Å) 20–2.5 (2.53–2.5) DNumber of reflections (working/test) 29,795/1,561Rwork/Rfree (%)e 24.9/28.1

DNumber of nonhydrogen protein atoms 10,584gGDP molecules 4

Water molecules 136 eRms deviations s

Bonds (Å) 0.062 iAngles (°) 1.9

pOverall B factors (chain B:chain F dimer)pG alpha 41.9 (38.2)GKB-752 peptide 52.4 (43.9)

GDP 35.6 (32.6) hWater 32.5 t

Ramachandran plot (% in region) oMost favored 88.6

aAllowed 9.0Generously allowed 2.4

pDisallowed 0.0aa Numbers in parentheses pertain to the highest resolution shell.rb Linear R factor = Σ(|I − <I>|)/Σ(I).

c Square R factor = Σ(|I − <I>|)2/Σ(I)2. vd <I/σI>, mean signal-to-noise, where I is the integrated intensity of 1a measured reflection and σI is the estimated error in measure- Gment. he Rwork = Σ(|Fp − Fp(calc)|)/ΣFp, where Fp and Fp(calc) are the observed

iand calculated structure factor amplitudes, respectively. Rfree iskcalculated similarly by using test set reflections never used during

refinement. (D

p

tmation more similar to the activated states of Gαi1,

GTPγS and Gαi1,GDP,AlF4− (Figure 8), moving in closer v

hproximity to GDP (compared to Gβγ and GoLoco boundstates) and affecting the position of arginine 178 (R178) s

l(Figure 8A). In the Gαi1,GDP/Gβ1γ2 heterotrimer (PDB1GP2) (Wall et al., 1995), R178 of switch I forms a salt n

Fbridge interaction with glutamate 43 (E43) across thebound GDP (Figure 8A). This “seatbelt” conformation, t

sresulting from reoriented coordinating residues N149and D150 due to Gβ1γ2 binding (Wall et al., 1998), is i

lproposed to stabilize bound GDP (Lambright et al.,1996; Wall et al., 1995). This same interaction occurs in t

tGαi1,GDP/R14GL (Kimple et al., 2002) (Figure 8A), butnot in the structures of free Gαi1,GDP (Wall et al., 1998), t

csuggesting that the formation of this R178/E43 saltbridge represents a common mechanism used by GDIs ofor Gαi1. Interestingly, in the KB-752 bound structure,the seatbelt interaction is not present (Figures 8A and b

u8B); the conformation of R178 is nearly identical to thatof the Gα ,GDP,AlF − transition state (PDB 1GFI) (Fig- 7

i1 4

re 8C) (Coleman et al., 1994) in which R178 is orientedo participate in GTP hydrolysis by stabilization of theeaving γ phosphate group as mimicked by the alumi-um tetrafluoride anion. These findings suggest thathe R178/E43 interaction is broken during nucleotidexchange and that an “unbuckled seatbeat” conforma-ion may be essential for GDP release in addition toTP hydrolysis. Thus, KB-752 appears to alter switch Ind II to create a feasible exit route for GDP (see Dis-ussion below). Magnesium was not observed in theucleotide binding pocket of Gαi1,GDP/KB-752 (al-hough its coordinating residue T181 is unaltered; Fig-res 8C and 8D), consistent with studies showing thatg2+ has no effect on GDP binding to Gα (e.g., Higa-

hijima et al., 1987).

iscussion

espite many biochemical and structural studies of theuanine nucleotide cycle, the mechanism of heterotrim-ric G protein activation remains elusive. Mutagenesistudies have highlighted several determinants govern-ng the G protein coupling and nucleotide exchangeroperties of GPCRs (Bourne, 1997; Hamm, 2001), butrecisely how a Gα subunit is induced to exchangeDP for GTP has remained unanswered, given the in-erent difficulty in obtaining structural information onhe GPCR/G protein complex. Structural determinantsf recently described GEF activity of RIC-8 (Afshar etl., 2004; Tall et al., 2003) are also not known.An alternative approach has been the use of small

eptides that possess nucleotide-dependent bindingnd biochemical properties akin to known G proteinegulators. Mastoparan, a 14 aa peptide found in waspenom, is a GEF for Gαi and Gαo (Higashijima et al.,990). The solution structure of mastoparan bound toαi indicates a helical conformation for this peptide;owever, biochemical studies suggest that its binding

nterface resides at the extended N terminus of Gα (Su-umar and Higashijima, 1992). Moreover, mastoparanINLKALAALAKKIL) shows no similarity to the TWXE/FL motif found in KB-752 and other Gα,GDP bindingeptides from our screen. Synthetic peptides from thehird intracellular loop of several GPCRs, a region in-olved in G protein coupling and activation by receptor,ave also been used to study G protein activation. Aolution structure of a peptide from the third intracellu-

ar loop of the CB1 cannabinoid receptor suggests theecessity for a helical conformation (Ulfers et al., 2002).inally, phage display has identified short heptapep-ides with biochemical activity at specific G proteinubunits (Hessling et al., 2003), although no structuralnformation has been reported. Our results further high-ight the power of phage display as a useful techniqueo identify conformation-dependent binding peptideshat can be useful tools in investigating protein func-ion. The structure of the Gαi1,GDP/KB-752 complexlearly demonstrates the basis of nucleotide specificityf this peptide.Furthermore, our structural determination of KB-752

ound to Gαi1 represents the first glimpse of a Gα sub-nit bound to a GEF. As with other Gα regulators, KB-52 modulates the conformation of the switch regions

Page 7: Structure of G i1 Bound to a GDP-Selective Peptide Provides … · 2017. 2. 14. · 752 GEF activity, we determined the crystal structure of 50% maximal response (EC 50) for KB-752

Gαi Binding Peptide with GEF Activity1075

Figure 5. Biochemical Confirmation of theOverall Structural Features of the Gαi1/KB-752 Interaction

(A) Ribbon trace of KB-752 (red) bound be-tween the α2 (“switch II”) and α3 helices ofthe Gαi1 Ras-like domain (blue). No contactsare made between KB-752 and the all-helicaldomain (yellow) or bound GDP (magenta).Switch regions are denoted in green.(B) Structural basis for nucleotide selectivebinding of KB-752 to Gαi1. KB-752 peptide(red, translucent) binds Gαi1 between switchII and the α3 helix; the conformations ofthese two helices are shown for Gαi1,GDP/KB-752 (green), Gαi1,GTPγS (yellow), andGαi1,GDP,AlF4

− (magenta). Whereas the α3helix is not significantly altered, switch II isdisplaced to accommodate KB-752 binding.Switch II in both Gαi1,GTPγS and Gαi1,GDP,AlF4

− assumes an extended α-helicalconformation that is stabilized relative toGαi1,GDP (Mixon et al., 1995; Sprang, 1997).This conformation of switch II is not permis-sive to KB-752 binding, as it creates extens-ive steric hindrance. In particular, W211 ofswitch II (shown in space filling) is in a re-strictive position relative to W5 of KB-752.(C) The GoLoco motif of RGS14 (orange) isalso seen to bind, in an α-helical conforma-tion, between switch II and the α3 helix ofGαi1 (PDB ID 1KJY); the critical arginine fin-ger which contacts GDP is highlighted withinthe Cα carbon ribbon trace of the GoLocopeptide. Other features are colored as in (A).(D) KB-752 GEF activity does not rely on theall-helical domain. 100 nM Gαi1 or a chimeric

Gα containing the Ras-like domain of Gαi1 and the all-helical domain of Gαo (“Gαioi” [Remmers et al., 1999]) was incubated in the absenceor presence of 50 �M KB-752, and [35S]GTPγS binding after 10 min at 30°C was measured as described in the Experimental Procedures.Data are expressed as a percentage of GTPγS bound relative to Gα protein in the absence of KB-752 (“Control”) and are the average ± SEMof four independent experiments. Data shown are the mean ± SEM for five independent experiments conducted in triplicate.(E) The KB-752 binding site on Gαi1 overlaps that of GoLoco motif peptides. Gαi1 (50 nM) was incubated in the absence or presence of theindicated concentrations of a peptide representing the GoLoco motif of RGS12 (R12GL) (Kimple et al., 2002). GTPγS binding was thenmeasured in the presence of the indicated concentrations of KB-752. Data are expressed as fmol of GTPγS bound above that measured inthe absence of KB-752 and are from a representative experiment of three independent experiments.(F) The binding of KB-752 has no effect on the kinetics of Gαi1 activation by AlF4

−, unlike the slowed activation rate seen upon GoLocopeptide binding. Gαi1-CFP (200 nM) and YFP-RGS4 (280 nM) fusion proteins, previously shown to generate increased fluorescence resonanceenergy transfer (FRET) upon Gαi1 activation by AlF4

− and subsequent RGS-box binding (Willard et al., 2004), were mixed together andpreincubated with either 10 �M KB-752 peptide or 5 �M GoLoco consensus peptide (AGS3Con [Kimple et al., 2002]), prior to the addition ofNaF and AlCl3 to final concentrations of 20 mM and 30 �M, respectively, at the 150 s mark.

critical to the guanine nucleotide cycle (Sprang, 1997).Previous structures of uncomplexed Gαi1,GDP have re-vealed structural disorder in these switch regions, par-ticularly switch II and III (Coleman and Sprang, 1998).However, in structures in which Gαi1 is bound to regula-tors (Gβγ, RGS4, GoLoco motif) or is in the activatedstate (GTPγS or GDP,AlF4

− bound), the switch regionsbecome ordered in specific, defined conformations(Coleman et al., 1994; Kimple et al., 2002; Tesmer etal., 1997a; Wall et al., 1995). Similarly, our structure ofGαi1,GDP/KB-752 reveals order in the switch regions,suggesting that the peptide stabilizes this conforma-tion resulting in its GEF activity—specifically by creat-ing a stabilized route for GDP egress.

Since the Gα nucleotide binding pocket is buried farfrom the proposed Gα/receptor interacting surface, it isthought that GPCRs use Gβγ as a lever to “pull open”Gα, creating a GDP exit route. By modeling onto Gα thestructural changes in EF-Tu induced by EF-Ts during

nucleotide exchange, Bourne and colleagues havepointed to the β3/α2 loop as a potential “lip” that oc-cludes GDP release (Iiri et al., 1998). Gβγ makes severalcontacts with this region and has been proposed to useadditional contacts within the α2 helix (switch II),namely, D228 of Gβ1 contacting K210 in Gαi1 (K206 inGαs), to lever open the lip to induce GDP release (Ron-dard et al., 2001). GPCRs are thought to use the Gα Nterminus to tilt Gβγ (making extensive contacts with theGα N terminus) away from Gα, thereby opening the β3/α2 lip (Iiri et al., 1998). Our structure of the GEF peptideKB-752 bound to Gαi1 supports the Bourne model. Bybinding between the switch II and α3 helices, KB-752pushes the α2 helix away from nucleotide, similar tothe proposed levering action of Gβγ. Displacement ofswitch II results in the β3/α2 loop (part of the proposedocclusive lip [Iiri et al., 1998]) also being pulled awayfrom nucleotide in a way that might allow more efficientGDP egress. Whereas switch II is displaced by the

Page 8: Structure of G i1 Bound to a GDP-Selective Peptide Provides … · 2017. 2. 14. · 752 GEF activity, we determined the crystal structure of 50% maximal response (EC 50) for KB-752

Structure1076

Figure 6. Biochemical Confirmation of Spe-cific Interactions between KB-752 and Gαi1

(A) Positions of KB-752 residues W5, F8, andE11 relative to residues in the switch II andα3 helices of Gαi1. W5 and F8 are placedwithin hydrophobic pockets formed by Gαi1

residues F215, L249, and I253, and W211,I212, and F215, respectively. E11 forms a saltbridge with R208 of Gαi1.(B) Peptide residues T4 and D7 of the con-served TWXE/DFL binding motif form an in-trapeptide hydrogen bond network thathelps to orient W5 and F8.(C and D) Effects of W5A, F8A, and E11L mu-tations on KB-752 activity. (C) Indicated KB-752 mutant or wild-type (wt) peptides wereeach immobilized to a density of w1000 RUson separate streptavidin-coated flow cells,and 50 �M GDP bound Gαi1 (“analyte”) wasinjected simultaneously over all four sur-faces. (D) Compared to the increase inGTPγS binding observed by addition of 50�M wild-type KB-752 to 100 nM Gαi1,GDP,substantial reduction of GEF activity is seenupon mutation to the W5, F8, or E11 residueof KB-752. Data shown in (D) are the mean ±standard deviation of triplicate samples froma representative experiment of three inde-pendent experiments conducted.

which presumably serve to stabilize its reorientation.both Gβγ and GoLoco (each with GDI activity) position

Figure 7. Conformational Changes in GαSwitch Regions Induced by KB-752 Binding

(A and B) Relative orientations of the threeswitch regions (SI–III) in heterotrimer (PDBcode 1GP2; blue in [A]), Gαi1,GDP/R14GL(PDB code 1KJY; orange in [B]), andGαi1,GDP/KB-752 (green). Movement ofswitch II (α2 helix) and the connected β3/α2loop away from the nucleotide bindingpocket in Gαi1,GDP/KB-752 is thought tocontribute to GEF activity by creating a routefor GDP release. The RGS14 GoLoco motifpeptide (R14GL) displaces switch II but doesnot significantly alter the position of the β3/α2 loop.(C) Binding of KB-752 displaces switch II, re-sulting in a reorientation of the β3/α2 loopaway from the guanine nucleotide pocket.KB-752 (red) stabilizes the β3/α2 loop(green) via several hydrogen bonds (indi-cated as yellow dashes), including the car-bonyl oxygen of G202 (Gα) with the indolenitrogen of W5 (KB-752), and the side chainhydroxyl and main chain amide nitrogen ofS206 (Gα) with the main chain amide nitro-gen and carbonyl oxygen of V3 (KB-752), re-spectively. A water molecule (magenta ball)is coordinated by both Gα and KB-752 con-tacts.

binding of the RGS14 GoLoco peptide, the β3/α2 loop trremains essentially unaltered in conformation com-

pared to the Gβγ bound, heterotrimeric structure. These l7results further highlight the potential role of the β3/α2

loop as an occlusive lip preventing GDP release, as t

he β3/α2 loop to block the proposed GDP egressoute, whereas KB-752 (with GEF activity) removes theoop from this position. Importantly, not only does KB-52 displace the β3/α2 loop from its occlusive orienta-ion, but it also makes several contacts with this loop,

Page 9: Structure of G i1 Bound to a GDP-Selective Peptide Provides … · 2017. 2. 14. · 752 GEF activity, we determined the crystal structure of 50% maximal response (EC 50) for KB-752

Gαi Binding Peptide with GEF Activity1077

Figure 8. Comparison of Switch Regions andCore Catalytic Residues of KB-752 BoundGαi1 with Other States of Gαi1

(A) Movement of switch I in the Gαi1,GDP/KB-752 complex (green), versus its positionin the Gαi1β1γ2 heterotrimer (blue) and theGαi1,GDP/R14GL complex (orange), resultsin disruption of a salt bridge (black dottedline) between R178 and E43 that normallystabilizes bound GDP (magenta) within Gαi1

when complexed to a GDI (Gβγ or GoLocopeptide).(B) Electron density of the R178 side chain inthe Gαi1,GDP/KB-752 complex (from a 2Fo −Fc simulated annealing composite omit mapcontoured to a level of 1σ) is denoted bywhite mesh. In the background is the β phos-phate of the bound GDP (βP).(C and D) Switch region comparisons withactivated Gαi1 states. Switch regions ofGαi1,GDP/KB-752 (green), Gαi1,GDP,AlF4

(PDB code 1GFI; magenta; [C]), and Gαi1,GTPγS (PDB code 1GIA; yellow; [D]) areshown along with the residues critical forGTP hydrolysis (R178 and T181 within switchI and Q204 within switch II). GDP from theGαi1,GDP/KB-752 structure is shown for ref-erence in each case. Overall conformation of

the switch regions of Gαi1,GDP,AlF4− and Gαi1,GTPγS are very similar, save for key changes in the position of catalytic residue side chains

(Wall et al., 1998). Whereas switch I of Gαi1,GDP/KB-752 is very similar to that of the activated forms, both switch II and III are dramaticallyremoved from the guanine nucleotide to allow for GDP release. The catalytic Q204 residue within switch II is far removed from the boundnucleotide and active site for GTP hydrolysis in the Gαi1,GDP/KB-752 structure. However, R178 and T181 of switch I are in a strikingly similarposition to that of the Gαi1,GDP,AlF4

− structure.

Although the precise structural determinants of GPCR-mediated GEF activity will clearly be distinct from thatof our artificial phage-displayed peptide GEF, the struc-tural changes in Gαi1 induced by KB-752 provide sup-port for the Gβγ-levering model of receptor GEF func-tion by suggesting that repositioning of switch II andthe β3/α2 loop is critical for GDP release. In this model,the proposed egress route for GDP is toward the Gβγbinding face of Gα, which is more accessible followingthe displacement of the occlusive β3/α2 loop.

An alternative opinion on receptor-mediated hetero-trimer activation (Cherfils and Chabre, 2003) suggeststhat GPCRs use the Gα N terminus to maneuver Gβγ inan opposite fashion to that proposed in the Bournemodel. In this “gear-shift” model, Gβγ is shifted towardGα, resulting in a closely packing Gα-Gβ interface sta-bilized by a proposed binding of the Gγ N terminus tothe Gα helical domain (Cherfils and Chabre, 2003). ThisGβγ shift is proposed to alter the conformation of theα5 helix, previously implicated in the receptor-cata-lyzed nucleotide exchange reaction (Marin et al., 2002).Our structure of Gαi1,GDP/KB-752, while not invalidat-ing the receptor GEF model of Cherfils and Chabregiven lack of sequence similarity between KB-752 andknown Gα regulators, certainly does not support theirmodel of GPCR GEF activity for three reasons: (i) KB-752 causes switch II to be displaced away from theGDP pocket rather than being packed more tightly, (ii)the proposed GDP exit route induced by KB-752 bind-ing is on the Gβ binding face, and (iii) KB-752 does notcause significant alterations in α5 helix conformation.

In addition to affecting switch II (α2 helix) and maneu-vering the β3/α2 loop in a manner consistent with the

Gβγ-lever model (Iiri et al., 1998), KB-752 binding alsoalters switch I. In contrast to displacement of switch IIaway from the nucleotide binding pocket, switch I isdisplaced slightly toward this pocket into a similar con-formation to that of GTPγS and GDP,AlF4

− bound Gα.In the Gαi1,GDP/Gβ1γ2 heterotrimer and Gαi1,GDP/R14GL complex, R178 of switch I forms a salt bridge withE43 (an interaction not observed in free Gαi1,GDP), form-ing a “seatbelt” over bound GDP thought to aid in thestabilization of Gα,GDP by Gβγ or GoLoco binding(Kimple et al., 2002; Lambright et al., 1996; Wall et al.,1995). Switch I in the Gαi1,GDP/KB-752 structure re-veals an R178 conformation out of bonding distance toE43, similar to that seen in the structure of freeGαi1,GDP, in which R178 is thought to be quite flexible(Mixon et al., 1995). The loss of the R178/E43 interac-tion in both the Gαi1,GDP/KB-752 (GEF) structure aswell as in free Gαi1,GDP (which has higher spontane-ous nucleotide exchange compared to Gβγ bound) sup-ports the loss of this interaction as coinciding with nu-cleotide exchange. Thus, breaking the R178/E43 “GDPseatbelt” is a potentially crucial step in GDP releaseand subsequent GTP binding. Surprisingly, the R178side chain is in a nearly identical conformation in theGαi1,GDP/KB-752 structure compared to the Gαi1,GDP,AlF4

− structure (Figure 7B), indicating that thisresidue potentially adopts a conformation that is suit-able for both GDP/GTP exchange and GTP hydrolysis.Having the R178/E43 interaction disrupted, along withcreating a feasible exit route by modulating the switchII helix, may contribute to enhanced GDP release and,thus, an enhanced nucleotide exchange rate observedupon KB-752 binding.

Page 10: Structure of G i1 Bound to a GDP-Selective Peptide Provides … · 2017. 2. 14. · 752 GEF activity, we determined the crystal structure of 50% maximal response (EC 50) for KB-752

Structure1078

ADespite the GEF activity of KB-752 toward Gαi1, theastructure of the complex contains bound GDP. Thisbseemingly paradoxical observation is explained by sev-p

eral considerations. The nucleotide-free state of iso- tlated Gα is very unstable, likely reflecting an instantan- f

Seous conformation as nucleotide binding is extremelyrapid (Ferguson et al., 1986; Sprang, 1997); stable trap-

Pping of the nucleotide-free state has only recently beenHsuccessfully described following binding to the non-areceptor GEF RIC-8 (Tall and Gilman, 2004). The Gα/B

RIC-8 interface is likely more extensive than with the asmall KB-752 peptide, which would add substantial 3

bstability to the nucleotide-free conformation. Similarly,sestablishing the nucleotide-free state of small GTPasesaalso necessitates a large stabilizing interface with re-sspective GEFs (e.g., Worthylake et al., 2000) that can-c

not be provided by the small KB-752 peptide. Alongwith the fact that the Gαi1/KB-752 complex was crystal- Slized in the presence of 5 �M GDP, these factors likely S

aimpeded the chances of capturing Gαi1 in a nucleotide-lfree state.HIn conclusion, our identification and structural analy-csis of a Gα,GDP binding peptide with GEF activity to-b

ward Gαi1–3 subunits provides support of the “Gβγ le- wver” hypothesis of GPCR GEF activity. The activity of i

AKB-752 as a GEF for Gαi suggests a future utility of this2peptide as a new molecular tool to study heterotrimeric(G protein signaling in vitro and in vivo.d(Experimental ProcedurescwUnless otherwise noted, all reagents were from Sigma. PeptidesNwere synthesized by Anaspec (San Jose, CA). Biotinylated peptidesdwere synthesized by Dr. Michael Berne and colleagues of the TuftsGUniversity Core Facility: biotinylation was performed on resinubound, Fmoc group-protected synthetic peptides that were selec-stively deprotected only at their N termini, assuring that biotin conju-

gation occurred solely at the free amine.GGPhage SelectionfBiotinylated Gαi1 was purified from E. coli as described in Kimpleaet al., (2004): the presence of an N-terminal AviTag sequence((GLNDIFEAQKIEWHE) allowed for selective in vivo biotinylation onothe lysine residue during expression in E. coli strain AVB 100 that2also expresses biotin ligase (BirA) and fermentation in free biotin-3containing medium as per manufacturer’s instructions (Avidity LCC,

Denver, CO). A total of 19 different random peptide bacteriophagelibraries were obtained from New England Biolabs (PhD7, PhD12) C

Cor prepared by Karo*Bio USA by using published methods (Sparkset al., 1996). Immulon 4 plates (96-well; Dynatech) were coated with f

tstreptavidin in 0.1 M NaHCO3, blocked with 1.0% BSA in 0.1 MNaHCO3, then incubated for 1 hr at 25°C with 10 pmol/well of bio- m

Dtin-Gαi1 in buffer A (20 mM HEPES [pH 7.5], 1 mM EDTA, 16 mMMgCl2, 1 mM DTT, 0.05% Tween-20) with either 5 �M GDP or 1

dGTPγS. Iterative selection of binding phage was performed byusing published methods (Sparks et al., 1996). Briefly, after incubat- 1

hing phage libraries with immobilized biotin-Gαi1 for 3 hr at 25°C,nonspecifically bound phages were removed by washing with t

iTBST buffer (10 mM Tris-HCl [pH 8.0], 150 mM NaCl, 0.05% Tween-20) with 0.5 mM biotin. Bound phages were eluted sequentially with b

aa low-pH glycine buffer and a high-pH ethanolamine buffer; afterneutralizing the pH, phages were amplified and subjected to repeat G

Nrounds of selection (Sparks et al., 1996).After four iterations, clonal phage isolates were purified, ampli- 2

rfied, and sequenced as described (Sparks et al., 1996). To detectbound phage by ELISA, biotin-Gαi1 was incubated overnight in i

nbuffer A with either 100 �M GDP or GTPγS, and then 1 pmol Gαi1/well (or buffer A alone) was immobilized onto plates as previously c

cdescribed. A total of 5 �l phage was added to each well in buffer

with either 100 �M GDP or GTPγS and was incubated for 30 mint 25°C. Unbound phage was removed by TBST washes, andound phage was detected with an anti-M13 antibody/horseradisheroxidase conjuguate. Assays were developed for 10 min at room

emperature by adding 2,2-azinobis(3-ethylbenzothiazoline)-6 sul-onic acid and H2O2. Signal development was stopped by addingDS to a final concentration of 1%.

rotein Purificationis6-tagged human Gαi1 (full-length, R144A mutant, and N-end 25a truncated) and human GαoA (full-length) were purified fromL21(DE3) E. coli as previously described (Kimple et al., 2004). Gαi1

nd Gαo were induced at OD600 = 0.8 with 1 mM IPTG for 4 hr at7°C. Gαi2β1γ2 was purified from Sf9 insect cells coinfected withaculoviruses encoding Gαi2, Gβ1, and His6-Gγ2 as previously de-cribed (Hooks et al., 2003). Proteins were purified by Ni2+-NTA,nion exchange, and size exclusion chromatographies as de-cribed (Hooks et al., 2003; Kimple et al., 2004). All proteins wereoncentrated by using YM-10 centrifugal filters (Millipore).

urface Plasmon Resonance Biosensor MeasurementsPR binding assays were performed at 25°C on a BIAcore 3000. Tonalyze nucleotide-dependent Gα binding, N-terminally biotiny-

ated KB-752 (diluted to 0.1 �g/ml in BIA running buffer [10 mMEPES (pH 7.4), 150 mM NaCl, 10 mM MgCl2, 0.005% NP40]) wasoupled to separate flow cells of streptavidin biosensors (Biacore)y using MANUAL INJECT to a surface density of w250, w500, or1000 resonance units. Prior to injection, Gα subunits were diluted

n BIA running buffer with 100 �M GDP, 100 �M GDP plus 30 �MlCl3 and 10 mM NaF, or 100 �M GTPγS and were incubated at5°C for 2–3 hr. 30 �l Gα subunit was then simultaneously injected

by using KINJECT) over flow cells at 10 �l/min, followed by 300 sissociation. Binding to a non-Gα interacting, biotinylated peptide

mNOTCH1; Snow et al., 2002) was subtracted from all bindingurves to correct for nonspecific binding and buffer shifts. Surfacesere regenerated with two 10 �l pulses of 500 mM NaCl/25 mMaOH at 20 �l/min. Binding curves and kinetic analyses were con-ucted by using BIAevaluation ver. 3.0 and were plotted by usingraphPad Prism ver. 4.0b. Binding affinities were calculated bysing the simultaneous association (ka) and dissociation (kd) analy-is parameter with generated sensorgram curves.

� Nucleotide Cycle AssaysTPγS exchange assays were conducted by using a nitrocellulose

ilter binding method (Afshar et al., 2004), with GTPγS binding re-ctions performed at either 20°C (Gαo and Gαi1-R144A) or 30°C

Gαi1, i2, i3 and Gαi2β1γ2). Steady-state GTPase assays were carriedut by using a charcoal precipitation-based method (Afshar et al.,004), with reactions incubated at 20°C (Gαo) or 30°C (Gαi) for0 min.

rystallization and Structure Determinationrystals of KB-752 bound to Gαi1 were obtained by vapor diffusion

rom hanging drops (3 �l) containing a 1:1 (v/v) ratio of protein solu-ion (6 mg/ml Gαi1�N25 and 1.3-fold molar excess KB-752 in 20M Tris [pH 7.5], 20 mM NaCl, 1 mM MgCl2, 10 �M GDP, 1 mMTT, 5% glycerol) to well solution (50 mM sodium citrate [pH 5.0],0% (w/w) PEG-8000, 10% (w/w) sucrose). Crystals formed in 3–5ays at 4°C in the space group P21 (a = 72.9 Å, b = 112.8 Å, c =09.5 Å, α = 90°, β = 93.7°, γ = 90°), with four Gαi1,GDP/KB-752eterodimers in the asymmetric unit. To collect data at 100 K, crys-als were cryoprotected in 30% glycerol for 1 min, then submergedn liquid N2. A native data set was collected at the SER-CAT 22-IDeamline at APS, Argonne National Laboratory. Data were scalednd indexed by using the program HKL2000. The structure ofαi1,GDP,Mg2+ (PDB accession code 1BOF), excluding the 25 aaterminus, aa 177–184 (switch I), aa 200–218 (switch II), aa 233–

39 (switch III), and waters and sulphates, was used for moleculareplacement with AMoRe (Navaza, 1994). Model building was donen O (Jones et al., 1991), with successive rounds of simulated an-ealing, minimization, B group, and torsion angle refinements beingompleted by using CNS (Brunger et al., 1998). All refinement wasompleted with noncrystallographic symmetry restraints, and each

Page 11: Structure of G i1 Bound to a GDP-Selective Peptide Provides … · 2017. 2. 14. · 752 GEF activity, we determined the crystal structure of 50% maximal response (EC 50) for KB-752

Gαi Binding Peptide with GEF Activity1079

of the four Gαi1,GDP/KB-752 dimers are essentially identical.Electron density maps for model building as well as the simulatedannealing composite omit map were generated with CNS. Gαi1 resi-dues 26–33 (extreme N terminus), 113–116 (αB-αC loop, dubbed“switch IV,” within the all-helical domain; Mixon et al., 1995), and345–354 (extreme C terminus) were not included in the final modelgiven incomplete electron density; prior to removal, each regionhad refined B factors of >150, indicative of low statistical certaintyand relative disorder. Additionally, in three of the four Gαi1 subunits(molecules “A,” “C,” and “D”) in the asymmetric unit, residues 234–239 of switch III were removed from the final model. All structuralimages were made with PyMol (DeLano Scientific, San Carlos, CA).

Acknowledgments

We thank Dr. Richard Neubig for Gαioi and Drs. Dana Fowlkes andElliott Ross for early guidance on this project. F.S.W. and M.B.J.are postdoctoral fellows of the American Heart Association and thePharmaceutical Research and Manufacturers of America Founda-tion, respectively. This work was supported in part by Karo*BioUSA and by National Institutes of Health grants R01 GM062338 (toD.P.S.) and P01 GM065533 (to T.K.H., J.S., and D.P.S.). Use of theAdvanced Photon Source was supported by the U.S. Departmentof Energy, Office of Basic Energy Sciences, under Contract No.W-31-109-Eng-38.

Received: January 16, 2005Accepted: April 14, 2005Published: July 12, 2005

References

Afshar, K., Willard, F.S., Colombo, K., Johnston, C.A., McCudden,C.R., Siderovski, D.P., and Gonczy, P. (2004). RIC-8 is required forGPR-1/2-dependent Galpha function during asymmetric division ofC. elegans embryos. Cell 119, 219–230.

Ashraf, S.S., Anderson, E., Duke, K., Hamilton, P.T., and Fredericks,Z. (2003). Identification and characterization of peptide probes di-rected against PKCα conformations. J. Pept. Res. 61, 263–273.

Bourne, H.R. (1997). How receptors talk to trimeric G proteins. Curr.Opin. Cell Biol. 9, 134–142.

Brunger, A.T., Adams, P.D., Clore, G.M., DeLano, W.L., Gros, P.,Grosse-Kunstleve, R.W., Jiang, J.S., Kuszewski, J., Nilges, M.,Pannu, N.S., et al. (1998). Crystallography & NMR system: a newsoftware suite for macromolecular structure determination. ActaCrystallogr. D Biol. Crystallogr. 54, 905–921.

Cabrera-Vera, T.M., Vanhauwe, J., Thomas, T.O., Medkova, M.,Preininger, A., Mazzoni, M.R., and Hamm, H.E. (2003). Insights intoG protein structure, function, and regulation. Endocr. Rev. 24,765–781.

Chen, Z., Singer, W.D., Sternweis, P.C., and Sprang, S.R. (2005).Structure of the p115RhoGEF rgRGS domain-Gα13/i1 chimeracomplex suggests convergent evolution of a GTPase activator. Nat.Struct. Mol. Biol. 12, 191–197.

Cherfils, J., and Chabre, M. (2003). Activation of G-protein Gα sub-units by receptors through Gα-Gβ and Gα-Gγ interactions. TrendsBiochem. Sci. 28, 13–17.

Coleman, D.E., and Sprang, S.R. (1998). Crystal structures of the Gprotein Gi α 1 complexed with GDP and Mg2+: a crystallographictitration experiment. Biochemistry 37, 14376–14385.

Coleman, D.E., Berghuis, A.M., Lee, E., Linder, M.E., Gilman, A.G.,and Sprang, S.R. (1994). Structures of active conformations of Gi α1 and the mechanism of GTP hydrolysis. Science 265, 1405–1412.

Ferguson, K.M., Higashijima, T., Smigel, M.D., and Gilman, A.G.(1986). The influence of bound GDP on the kinetics of guanine nu-cleotide binding to G proteins. J. Biol. Chem. 261, 7393–7399.

Fields, T.A., and Casey, P.J. (1997). Signalling functions and bio-chemical properties of pertussis toxin-resistant G-proteins. Bio-chem. J. 321, 561–571.

Gilchrist, A., Mazzoni, M.R., Dineen, B., Dice, A., Linden, J., Proctor,W.R., Lupica, C.R., Dunwiddie, T.V., and Hamm, H.E. (1998). Antag-onists of the receptor-G protein interface block Gi-coupled signaltransduction. J. Biol. Chem. 273, 14912–14919.

Hamm, H.E. (2001). How activated receptors couple to G proteins.Proc. Natl. Acad. Sci. USA 98, 4819–4821.

Hessling, J., Lohse, M.J., and Klotz, K.N. (2003). Peptide G proteinagonists from a phage display library. Biochem. Pharmacol. 65,961–967.

Higashijima, T., Ferguson, K.M., Sternweis, P.C., Smigel, M.D., andGilman, A.G. (1987). Effects of Mg2+ and the beta gamma-subunitcomplex on the interactions of guanine nucleotides with G pro-teins. J. Biol. Chem. 262, 762–766.

Higashijima, T., Burnier, J., and Ross, E.M. (1990). Regulation of Giand Go by mastoparan, related amphiphilic peptides, and hy-drophobic amines. Mechanism and structural determinants of ac-tivity. J. Biol. Chem. 265, 14176–14186.

Hooks, S.B., Waldo, G.L., Corbitt, J., Bodor, E.T., Krumins, A.M.,and Harden, T.K. (2003). RGS6, RGS7, RGS9, and RGS11 stimulateGTPase activity of Gi family G-proteins with differential selectivityand maximal activity. J. Biol. Chem. 278, 10087–10093.

Hyde-DeRuyscher, R., Paige, L.A., Christensen, D.J., Hyde-DeRuy-scher, N., Lim, A., Fredericks, Z.L., Kranz, J., Gallant, P., Zhang, J.,Rocklage, S.M., et al. (2000). Detection of small-molecule enzymeinhibitors with peptides isolated from phage-displayed combinato-rial peptide libraries. Chem. Biol. 7, 17–25.

Iiri, T., Farfel, Z., and Bourne, H.R. (1998). G-protein diseases fur-nish a model for the turn-on switch. Nature 394, 35–38.

Ja, W.W., and Roberts, R.W. (2004). In vitro selection of state-spe-cific peptide modulators of G protein signaling using mRNA display.Biochemistry 43, 9265–9275.

Jones, T.A., Zou, J.Y., and Cowan, S.W. (1991). Improved methodsfor building protein models in electron density maps and the loca-tion of errors in these models. Acta Crystallogr. A 47 (Pt 2), 110–119.

Kimple, R.J., Kimple, M.E., Betts, L., Sondek, J., and Siderovski,D.P. (2002). Structural determinants for GoLoco-induced inhibitionof nucleotide release by Gα subunits. Nature 416, 878–881.

Kimple, R.J., Willard, F.S., Hains, M.D., Jones, M.B., Nweke, G.K.,and Siderovski, D.P. (2004). Guanine nucleotide dissociation inhibi-tor activity of the triple GoLoco motif protein G18: alanine-to-aspar-tate mutation restores function to an inactive second GoLoco mo-tif. Biochem. J. 378, 801–808.

Lambright, D.G., Sondek, J., Bohm, A., Skiba, N.P., Hamm, H.E.,and Sigler, P.B. (1996). The 2.0 Å crystal structure of a heterotrim-eric G protein. Nature 379, 311–319.

Marin, E.P., Krishna, A.G., and Sakmar, T.P. (2002). Disruption ofthe α5 helix of transducin impairs rhodopsin-catalyzed nucleotideexchange. Biochemistry 41, 6988–6994.

Martin, E.L., Rens-Domiano, S., Schatz, P.J., and Hamm, H.E.(1996). Potent peptide analogues of a G protein receptor-bindingregion obtained with a combinatorial library. J. Biol. Chem. 271,361–366.

McCudden, C.R., Hains, M.D., Kimple, R.J., Siderovski, D.P., andWillard, F.S. (2005). G-protein signaling: back to the future. Cell.Mol. Life Sci. 62, 551–577.

Mixon, M.B., Lee, E., Coleman, D.E., Berghuis, A.M., Gilman, A.G.,and Sprang, S.R. (1995). Tertiary and quaternary structural changesin Gi α 1 induced by GTP hydrolysis. Science 270, 954–960.

Navaza, J. (1994). AMoRe: an automated package for molecularreplacement. Acta Crystallogr. A 50, 157–163.

Neubig, R.R., and Siderovski, D.P. (2002). Regulators of G-proteinsignalling as new central nervous system drug targets. Nat. Rev.Drug Discov. 1, 187–197.

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.,et al. (2000). Crystal structure of rhodopsin: a G protein-coupledreceptor. Science 289, 739–745.

Page 12: Structure of G i1 Bound to a GDP-Selective Peptide Provides … · 2017. 2. 14. · 752 GEF activity, we determined the crystal structure of 50% maximal response (EC 50) for KB-752

Structure1080

Remmers, A.E., Engel, C., Liu, M., and Neubig, R.R. (1999). Interdo- srmain interactions regulate GDP release from heterotrimeric G pro-

teins. Biochemistry 38, 13795–13800.ARodi, D.J., Makowski, L., and Kay, B.K. (2002). One from column A

and two from column B: the benefits of phage display in molecular-recognition studies. Curr. Opin. Chem. Biol. 6, 92–96. A

tRondard, P., Iiri, T., Srinivasan, S., Meng, E., Fujita, T., and Bourne, uH.R. (2001). Mutant G protein α subunit activated by Gβ gamma:a model for receptor activation? Proc. Natl. Acad. Sci. USA 98,6150–6155.

Ross, E.M. (2002). Quantitative assays for GTPase-activating pro-teins. Methods Enzymol. 344, 601–617.

Rossman, K.L., Der, C.J., and Sondek, J. (2005). GEF means go:turning on RHO GTPases with guanine nucleotide-exchange fac-tors. Nat. Rev. Mol. Cell Biol. 6, 167–180.

Scott, J.K., Huang, S.F., Gangadhar, B.P., Samoriski, G.M., Clapp,P., Gross, R.A., Taussig, R., and Smrcka, A.V. (2001). Evidence thata protein-protein interaction ‘hot spot’ on heterotrimeric G proteinβγ subunits is used for recognition of a subclass of effectors. EMBOJ. 20, 767–776.

Slep, K.C., Kercher, M.A., He, W., Cowan, C.W., Wensel, T.G., andSigler, P.B. (2001). Structural determinants for regulation of phos-phodiesterase by a G protein at 2.0 Å. Nature 409, 1071–1077.

Snow, B.E., Brothers, G.M., and Siderovski, D.P. (2002). Molecularcloning of regulators of G-protein signaling family members andcharacterization of binding specificity of RGS12 PDZ domain.Methods Enzymol. 344, 740–761.

Sparks, A.B., Adey, N.B., Cwirla, S., and Kay, B.K. (1996). Screeningphage-displayed random peptide libraries. In Phage Display ofPeptides and Proteins, A Laboratory Manual, B.K. Kay, J. Winter,and J. McCafferty, eds. (San Diego: Academic Press), pp. 227–253.

Sprang, S.R. (1997). G protein mechanisms: insights from structuralanalysis. Annu. Rev. Biochem. 66, 639–678.

Sukumar, M., and Higashijima, T. (1992). G protein-bound confor-mation of mastoparan-X, a receptor-mimetic peptide. J. Biol.Chem. 267, 21421–21424.

Sunahara, R.K., Tesmer, J.J., Gilman, A.G., and Sprang, S.R. (1997).Crystal structure of the adenylyl cyclase activator Gsα. Science278, 1943–1947.

Tall, G.G., and Gilman, A.G. (2004). Purification and functionalanalysis of Ric-8A: a guanine nucleotide exchange factor for G-pro-tein α subunits. Methods Enzymol. 390, 377–388.

Tall, G.G., Krumins, A.M., and Gilman, A.G. (2003). Mammalian Ric-8A (synembryn) is a heterotrimeric Gα protein guanine nucleotideexchange factor. J. Biol. Chem. 278, 8356–8362.

Tesmer, J.J., Berman, D.M., Gilman, A.G., and Sprang, S.R. (1997a).Structure of RGS4 bound to AlF4-activated G(i α1): stabilization ofthe transition state for GTP hydrolysis. Cell 89, 251–261.

Tesmer, J.J., Sunahara, R.K., Gilman, A.G., and Sprang, S.R.(1997b). Crystal structure of the catalytic domains of adenylyl cy-clase in a complex with Gsα.GTPγS. Science 278, 1907–1916.

Ulfers, A.L., McMurry, J.L., Miller, A., Wang, L., Kendall, D.A., andMierke, D.F. (2002). Cannabinoid receptor-G protein interactions:G(α1)-bound structures of IC3 and a mutant with altered G proteinspecificity. Protein Sci. 11, 2526–2531.

Wall, M.A., Coleman, D.E., Lee, E., Iniguez-Lluhi, J.A., Posner, B.A.,Gilman, A.G., and Sprang, S.R. (1995). The structure of the G pro-tein heterotrimer Gi α 1 β 1 γ 2. Cell 83, 1047–1058.

Wall, M.A., Posner, B.A., and Sprang, S.R. (1998). Structural basisof activity and subunit recognition in G protein heterotrimers. Struc-ture 6, 1169–1183.

Willard, F.S., Kimple, R.J., Kimple, A.J., Johnston, C.A., and Sider-ovski, D.P. (2004). Fluorescence-based assays for RGS box func-tion. Methods Enzymol. 389, 56–71.

Worthylake, D.K., Rossman, K.L., and Sondek, J. (2000). Crystal

tructure of Rac1 in complex with the guanine nucleotide exchangeegion of Tiam1. Nature 408, 682–688.

ccession Numbers

tomic coordinates and structure factors for Gαi1 in complex withhe KB-752 peptide have been deposited in the Protein Data Banknder accession code 1Y3A.


Recommended