Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Jul 12.
Published in final edited form as: Methods Cell Biol. 2021 Jul 12;166:133–159. doi: 10.1016/bs.mcb.2021.06.001

Purinergic GPCR transmembrane residues involved in ligand recognition and dimerization

Veronica Salmaso 1, Shanu Jain 1, Kenneth A Jacobson 1,*
PMCID: PMC8620127  NIHMSID: NIHMS1758124  PMID: 34752329

Abstract

We compare the GPCR-ligand interactions and highlight important residues for recognition in purinergic receptors—from both X-ray crystallographic and cryo-EM structures. These include A1 and A2A adenosine receptors, and P2Y1 and P2Y12 receptors that respond to ADP and other nucleotides. These receptors are important drug discovery targets for immune, metabolic and nervous system disorders. In most cases, orthosteric ligands are represented, except for one allosteric P2Y1 antagonist. This review catalogs the residues and regions that engage in contacts with ligands or with other GPCR protomers in dimeric forms. Residues that are in proximity to bound ligands within purinergic GPCR families are correlated. There is extensive conservation of recognition motifs between adenosine receptors, but the P2Y1 and P2Y12 receptors are each structurally distinct in their ligand recognition. Identifying common interaction features for ligand recognition within a receptor class that has multiple structures available can aid in the drug discovery process.

1. Introduction

GPCR structural biology has accelerated the discovery of new drug molecules, for example, enabling the de novo design of A2A adenosine receptor (AR) antagonist AZD4635 (Fig. 1), which is in clinical trials for cancer (Borodovsky et al., 2020; Congreve, de Graaf, Swain, & Tate, 2020). X-ray crystallography has been used to determine most of these structures (Qu, Wang, & Wu, 2020; Wacker, Stevens, & Roth, 2017). In addition, cryogenic electron microscopy (cryo-EM) is increasingly used and has the advantage of more readily including protein partners such as the associated G proteins and detecting multiple conformations of the same protein (Safdari, Pandey, Shukla, & Dutta, 2018). GPCRs contain seven transmembrane helical domains (TMs), but curiously not all 7TM proteins are GPCRs (Vasiliauskaite-Brooks, Healey, & Granier, 2019). In most of the known structures of GPCR-ligand complexes, the recognition of small molecules involves amino acid residues in the TM region facing the centroid within the heptahelical bundle. The canonical, orthosteric ligand binding site of Class A (rhodopsin-like) GPCRs has largely conserved features (Ballesteros and Weinstein, 1995; van Rhee & Jacobson, 1996; Venkatakrishnan et al., 2013). This conserved structure-function aids in predicting the modes of interaction of competitive small molecule ligands, both for cases in which a protein structure is lacking (Jung et al., 2020) and when a known receptor structure is applied to predicting the interactions of different ligand classes (Rodriguez et al., 2016). The correspondence of key TM residues between receptor subtypes and their shared functionality in ligand binding and activation has been analyzed (Lebon et al., 2011; Wacker et al., 2017; Warne, Edwards, Dore, Leslie, & Tate, 2019; Xu et al., 2011) for Class A GPCRs. There is structural information available, as well, for non-class A GPCRs, in which the degree of homology with Class A is low, for example, a recently determined Class D Ste2 receptor dimer (Velazhahan et al., 2021). Key water molecules, H-bonding networks and molecular (micro)switches for activation within the TM region that are shared among various Class A GPCRs have been defined (Filipek, 2019; Venkatakrishnan et al., 2019; White et al., 2018; Zhou et al., 2019). GPCRmd (http://gpcrmd.org/) is an open source online platform for molecular dynamics (MD) simulations and visualization of dynamic ligand and interhelical interactions, conserved structural motifs, water molecules, H-bonding and other interactions in GPCRs (Rodriguez-Espigares et al., 2020).

FIG. 1.

FIG. 1

FIG. 1

Structures of pharmacological probes for ARs (A) and P2YRs (B) shown in Table 1, including ligands present in reported structures. The structures of adenosine, ADP and ATP are not shown. Theophylline and caffeine (shown under A2AAR) are nonselective antagonists.

Allosteric small molecule modulators of GPCRs have been shown to bind in nearly all regions of the general structure (Wold, Chen, Cunningham, & Zhou, 2019). Nevertheless, many of the allosteric binding regions that are distal or distinct from the canonical binding site still may involve TM residues. This review does not cover the GPCR intracellular loops (ILs), which interact with the C-terminus of the alpha subunit of the G protein or with β-arrestin (Carpenter & Tate, 2017; Ciancetta, Rubio, Lieberman, & Jacobson, 2019), but it emphasizes the role of TM residues. The extracellular loops (ELs) that have their own effects on GPCR ligand binding and activation (Cao et al., 2018; Marullo et al., 2020), and data for some EL residues are included in this review. EL2, in particular is typically in direct contact with the orthosteric ligands of Class A GPCRs.

GPCR dimerization is also now recognized as being important biologically (García-Recio et al., 2020; Pin, Kniazeff, Prezeau, Liu, & Rondard, 2019). Class C GPCRs form obligatory functional dimers, but there is increasing evidence for functional dimerization and its pharmacological consequences within Class A receptors, as well. It appears that the monomers and dimers may exist in an equilibrium. Thus, we discuss information about the TM regions that are engaged in putative contact during dimerization with other GPCR protomers.

2. AR and P2YR ligands and structures

In this review, we focus on residues of purinergic GPCRs, i.e., ARs and PY receptors (P2YRs), that are involved in ligand and protein recognition. These represent 12 Class A GPCRs (4 ARs and 8 P2YRs), which couple to various G proteins: Gi/o, Gs or Gq (Table 1). Representative members of these receptor families have been determined structurally by X-ray or cryo-EM methods (Table 2): specifically, A1AR, A2AAR, P2Y1R and P2Y12R (all human). In particular, the A2AAR has served as a model system for structural probing in many studies. An abundance of ligands has been discovered for these receptors, which have important drug discovery potential (Jacobson et al., 2021). These receptors are important drug discovery targets for immune, inflammatory, cardiovascular, metabolic and nervous system disorders. Some of these orthosteric ligands have been tested in clinical trials or are being developed for mainly chronic, but also some acute conditions, such as stroke (Pedata et al., 2014). Thus, it is valuable for future drug discovery efforts to analyze the commonality between ARs and within the two subfamilies of the P2YRs (Attah et al., 2020; Ciancetta & Jacobson, 2018; Gutierrez-de-Teran, Sallander, & Sotelo, 2017; Jespers et al., 2017; Jung et al., 2020).

Table 1.

G protein coupling of ARs and P2YRs and their ligands used as pharmacological probes (Jacobson et al., 2020; Jacobson, IJzerman, & Müller, 2021).

Receptor family Subtype (agonist) G protein Synthetic agonists; antagonists (Ki in nM at human receptor, unless noted)
ARs A1 Gi, Go Cl-ENBA (0.51), CPA (2.3), MRS7469 (2.14), SPA (7.92); DPCPX (3.9), PSB-36 (0.7)
A2A Gs, Golf CGS21680 (27), UK432097 (4), PSB-0777 (360); SCH442416 (4.1)
A2B Gs, Gq Bay60–6583 (114); MRS1754 (1.97), PSB-603 (0.55)
A3 Gi IB-MECA (1.8), Cl-IB-MECA (1.4), MRS5980 (0.70), MRS5698 (3.5); MRS1523 (19), MRS1191 (31)
P2YRs P2Y1-like P2Y1 (ADP) Gq MRS2365 (0.40); 2-MeS-ADP (2510)a; MRS2179 (331), MRS2500 (0.79), BPTU (6.0)
P2Y2 (ATP, UTP) Gq, Gi 2-thio-UTP (35), PBS-1114 (135); AR-C118925XX (58)
P2Y4 (UTP) Gq, Gi MRS4062 (23); PSB-16133 (234), PSB-1699 (407)
P2Y6 (UDP) Gq MRS4383 (493)b, PSB-0474 (71), 5-OMe-UDP (79); MRS2578 (372)b, TIM-38 (4300)b
P2Y11 (ATP) Gq, Gs NF546 (537); NF157 (45)c
P2Y12-like P2Y12 (ADP) Gi 2-MeS-ADP (5)a; cangrelor (0.40), ticagrelor (13), AZD1283 (31.6), PSB-0739 (0.16)
P2Y13 (ADP) Gi 2-MeS-ADP (19)a; MRS2211 (1620)
P2Y14 (UDPG, UDP) Gi MRS2905 (0.92), MRS2690 (49); PPTN (0.44), MRS4625 (27.6)b

Adenosine is the principal native agonist for the AR subtypes, while the more diverse endogenous P2YR ligands are indicated above. P2YR affinities are found in Jacobson et al. (2020). Structures of AR ligands are shown in Fig. 1.

a

Activates P2Y1, P2Y12 and P2Y13 receptors.

b

IC50 value.

c

Antagonist of P2Y1 and P2Y11 receptors.

Table 2.

Reported AR and P2YR experimental structures (in the Protein Data Bank (PDB)) and their characteristics.

Receptor Activation state Ligand PDB ID (References)
ARs
A1AR Inactive PSB36 5N2S (Cheng et al., 2017)
DU172 (covalent) 5UEN (Glukhova et al., 2017)
Active (Gi) Adenosinea 6D9H (Draper-Joyce et al., 2018)
A2AAR Inactive ZM241384 3EML (Jaakola et al., 2008), 3PWH (Doré et al., 2011), 3VG9 & 3VGA (Hino et al., 2012), 5UVI (Martin-Garcia et al., 2017), 5JTB (Melnikov et al., 2017), 6AQF (Eddy et al., 2018), 6MH8 (Martin-Garcia et al., 2019), 6S0L & 6S0Q (Nass et al., 2020)
ZM241384, Na+ allosteric mod 4EIY (Liu et al., 2012), 5IU4 (Segala et al., 2016), 5K2A & 5K2B & 5K2C & 5K2D (Batyuk et al., 2016), 5NLX & 5NM2 & 5NM4 (Weinert et al., 2017), 5VRA (Broecker et al., 2018), 5OLG (Rucktooa et al., 2018), 6JZH (Shimazu et al., 2019), 6LPJ & 6LPK & 6LPL (Ihara et al., 2020), 6PS7 (Ishchenko et al., 2019), 6WQA (Lee et al., 2020)
12b, Na+ allosteric mod 5IUA (Segala et al., 2016)
12x, Na+ allosteric mod 5IUB (Segala etal., 2016)
12c, Na+ allosteric mod 5IU7 (Segala et al., 2016)
12f, Na+ allosteric mod 5IU8 (Segala et al., 2016)
XAC 3REY (Doré et al., 2011)
Caffeine 3RFM (Doré et al., 2011)
Caffeine, Na+ allosteric mod 5MZP (Cheng et al., 2017)
Theophylline, Na+ allosteric mod 5MZJ (Cheng et al., 2017)
PSB36, Na+ allosteric mod 5N2R (Cheng et al., 2017)
T4G 3UZA (Congreve et al., 2012)
T4E 3UZC (Congreve et al., 2012)
T4E, Na+ allosteric mod 5OLZ & 5OM1 & 5OM4 (Rucktooa et al., 2018)
AZD4635, Na+ allosteric mod 6GT3 (Borodovsky et al., 2020)
Tozadenant, Na+ allosteric mod 5OLO (Rucktooa et al., 2018)
Cmpd-1 5UIG (Sun et al., 2017)
LUAA47070, Na+ allosteric mod 5OLV (Rucktooa et al., 2018)
Vipadenant, Na+ allosteric mod 5OLH (Rucktooa et al., 2018)
Chromone 4d, Na+ allosteric mod 6ZDR (Jespers et al., 2020)
Chromone 5d, Na+ allosteric mod 6ZDR (Jespers et al., 2020)
Intermediate UK432097 3QAK (Xu et al., 2011), 5WF5 & 5WF6 (White et al., 2018)
Adenosine 2YDO (Lebon et al., 2011)
NECA 2YDV (Lebon et al., 2011)
CGS21680 4UG2 & 4UHR (Lebon, Edwards, Leslie, & Tate, 2015)
Active (bound to mini-Gs) NECA 5G53 (Carpenter, Nehmé, Warne, Leslie, & Tate, 2016)
NECAa 6GDG (García-Nafría, Lee, Bai, Carpenter, &Tate, 2018)
P2YRs
P2Y1R Inactive MRS2500 4XNV (Zhang et al., 2015)
BPTU 4XNW (Zhang et al., 2015)
P2Y12R Inactive AZD1283 4NTJ (Zhang et al., 2014)
Intermediate 2-MeSADP 4PXZ (Zhang et al., 2014)
2-MeSATP 4PY0 (Zhang, Zhang, Gao, Zhang, et al., 2014)

The main technique used to solve the structures was by X-ray crystallography, except where noted.

a

Solved by cryo-EM.

Numerous selective agonist and antagonist ligands (Jacobson et al., 2021) have been reported for the ARs (Fig. 1A) and P2YRs (Fig. 1B), and receptor complexes have been determined structurally for some of these agonists/antagonists, including both subtype selective and nonselective ligands (Table 2). In the case of ARs, three-dimensional structures have been solved for two receptor subtypes with either agonists or antagonists: A1AR and A2AAR. Ligand binding in the orthosteric binding site has many parallels between the A1 and A2AARs structures and with the theoretical models of A2B and A3ARs (Kose et al., 2018; Tosh et al., 2020; Tosh et al., 2020). There are also many ligand-interacting amino acid residues in common between nucleoside agonists and diverse, non-nucleoside antagonists of the ARs. Although most AR agonists are adenosine derivatives, pyridine-3,5-dicarbonitrile agonists and partial agonists have been extensively explored (Dal Ben et al., 2019). Structural information on P2YRs is also available for two receptor subtypes, P2Y1R and P2Y12R, with only antagonists in the former example, but agonists and antagonists are represented in the latter. The parallelism that can be observed in the case of ARs, is missing in the case of P2YRs, where more variability has been observed among three-dimensional structures.

We have categorized GPCR-ligand interactions, i.e., identified important residues needed to coordinate the ligands—either from X-ray crystallographic or cryo-EM structures—for Gi/o-coupled A1 and Gs-coupled A2A receptors (Glukhova et al., 2017; Jaakola et al., 2008; Lebon et al., 2011; Xu et al., 2011), Gq-coupled P2Y1 and Gi-coupled P2Y12 receptors (Zhang et al., 2015; Zhang, Zhang, Gao, Paoletta, et al., 2014; Zhang, Zhang, Gao, Zhang, et al., 2014). Often, there is site-directed mutagenesis (SDM) data gathered in multiple studies to support these structures. We have investigated the tridimensional structures deposited in the Protein Data Bank (PDB) (rcsb.org; Berman et al., 2000), analyzed the contacts between ligands and receptor residues and collected the results in bar plots (Fig. 2) enabling the highlighting of most interacting residues. An automatic procedure was employed using a Schrödinger (Schrödinger Release 2020–4: Maestro, Schrödinger, LLC, New York, NY, 2020) command line script to detect ligand-receptor interactions on the PDB complexes, previously prepared with the Protein Preparation Wizard of the Schrödinger suite. Contacts defined as “good contacts” were taken into account, described by the distance of two atoms divided by the sum of their van der Waals radii within the range of 1.30 and 0.89. Furthermore, H-bonds, water mediated H-bonds, π–π interactions (face-to-face or edge-to-face), salt bridges and halogen bonds were examined, and these interaction patterns are discussed in the text. Chain A or R of the PDB files and the alternate state with higher occupancy were employed. TM residues are highlighted using the Ballesteros-Weinstein notation (Ballesteros & Weinstein, 1995) in addition to their number in the protein sequence (x·yz, where x is the helix number and yz is the amino acid residue number relative to the most conserved residue in that helix set as 50).

FIG. 2.

FIG. 2

The purinergic GPCR structures deposited in the Protein Data Bank (PDB) so far were surveyed to compare the interaction patterns of different ligands bound to ARs and P2YRs. Maestro by Schrödinger (Schrödinger Release 2020–4: Maestro, Schrödinger, LLC, New York, NY, 2020) was employed to prepare (Protein Preparation Wizard) and analyze the PDB structures, and the data were then plotted with matplotlib using python scripts. Contacts defined as “good contacts” in Maestro were taken into account (distance of two atoms divided by the sum of their van der Waals radii within the range of 1.30 and 0.89). Panels (A–D) show residues in contact with the ligand in the complexes deposited in the PDB. The height of the bar plots represents the number of structures where the contact is observed, with a possible maximum height, corresponding to the number of deposited structures, equal to 3, 57, 2, 3 in the case of A1AR, A2AAR, P2Y1R and P2Y12R, respectively (panels A–D). Different colors highlight structures with agonists and antagonists in the case of A1AR, A2AAR and P2Y12R (panels A, B and D), while discriminate the orthosteric antagonist MRS2500 from the allosteric BPTU in the case of P2Y1R (panel C). A focus on the H-bonds between A2AAR residues and agonists (panel E) or antagonists (panel F) is further provided. Ligand abbreviations: ADN, adenosine; UKA, UK432097; CGS, CGS21680; ZMA, ZM241385.

3. Analysis of small molecule recognition by TM residues of adenosine receptors

3.1. AR structures and models

X-ray and cryo-EM structures have been reported for two AR subtypes, specifically A1AR and A2AAR, for a total of 3 structures in the first case and 57 in the second (Table 2). The structures of the two subtypes show high similarity, with major differences confined to the loops. In particular, EL2 has a completely different conformation in the two receptors, and even the same receptor (A2AAR) with different bound ligands can show a large reorganization of EL2 (Xu et al., 2011). Human ARs have high sequence identity, ranging from a maximum of 46% for the A1AR-A3AR pair to a minimum of 30% for the A2AAR-A3AR pair (sequence identity data retrieved from GPCRdb; https://gpcrdb.org/similaritymatrix/render; Kooistra et al., 2021). The similarities among the TM structures of the two subtypes, together with their high sequence identity among ARs, makes structural information for A1AR and A2AAR useful also to build homology models for A2BAR and A3AR.

Experimental structures and models have been used extensively in medicinal chemistry to design new ligands of the same chemical family and to discover novel chemotypes for a given receptor by virtual screening. For example, the discovery of a sub-micromolar A2AAR antagonist by virtually screening around 40,000 lead-like compounds in the dark chemical matter was accomplished by Carlsson et al. (Ballante et al., 2020). The use of purinergic structure for rational drug design has been recently reviewed (Salmaso & Jacobson, 2020b).

3.1.1. A1AR structures

Three structures have been reported for A1AR, one in the agonist-bound active state (PDB ID: 6D9H, bound to adenosine) and two in the antagonist-bound inactive state (PDB IDs: 5N2S and 5UEN, respectively bound to PSB36 and the covalent antagonist DU172), where both antagonists have a xanthine structure (Fig. 3A and C). A total of 25 residues have been found in contact with a ligand in at least one of the three tridimensional structures, and among them 12 residues are common to all the three ligands (Fig. 2A): V873.32, L883.33, T913.36 (mutated into A91 in structure 5N2S), F171EL2, E172EL2, M1775.35, M1805.38, W2476.48, L2506.51, N2546.55, I2747.39 and H2787.43. A contact between adenosine and T2777.42 distinguishes this agonist from the two antagonists, which instead make additional contacts with L2536.54, T2576.58, T2707.35 (and with I692.64, N1845.42, H2516.52 and K265EL3 in the case of PSB36 in structure 5N2S, and with Y121.35, A662.61, N702.65 and Y2717.36 in the case of DU172 in structure 5UEN).

FIG. 3.

FIG. 3

(A) and (C) Experimental binding mode of the agonist adenosine (orange sticks) and of the covalent antagonist DU172 (green) to A1AR (dark and light gray when bound to agonist and antagonist, in structures 6D9H and 5UEN, respectively). (B) and (D) Experimental binding mode of the agonist adenosine (orange sticks) and of the antagonist ZM241385 (green) to A2AAR (dark and light gray when bound to agonist and antagonist, in structures 2YDO and 4EIY, respectively). Relevant receptor residues discussed in the text are shown by sticks.

AR-conserved N2546.55 is engaged in a bidentate H-bond with the exocyclic amine at position 6, N6H, and with the position 7 of the adenine scaffold in the case of adenosine (agonist), and with the carbonyl at position 6 and the NH at position 7 of the xanthine scaffold of the antagonists. The aromatic base scaffolds are furthermore involved in a π–π stacking interaction with F171EL2. Adenosine is involved in additional H-bonds with T913.36 through the 5′-hydroxy group and H2787.43 through the 3′-hydroxy group, and is in contact with T2777.42, whose equivalent position in A2AAR, i.e., S2777.42, interacts through H-bonds with agonists. The antagonists are engaged in fewer H-bonds compared to agonists, with only the additional involvement of Y121.35 in the case of the covalently bound xanthine DU172, but the cyclohexyl (DU172) and 3-tricyclo[3.3.1.03,7]nonanyl (of xanthine derivative PSB36) substitution at position 8 face the extracellular portion of the receptor making hydrophobic contacts with F171EL2, E172EL2, M1775.35, L2506.51, L2536.54, T2576.58 and T2707.35 (and K265EL3 in structure 5N2S). Moreover, the long propyl-carbamoyl-benzene-1-sulfonyl substituent at position 3 of DU172, covalently bound to Y2717.36, makes further interactions with TM1 (residue Y121.35) and TM2 (residues A662.61 and N702.65). However, the shorter 3-hydroxypropyl group of PSB36 engages TM2 only through I692.64. Differently, PSB36 presents a butyl substituent at position 1 which is bulkier than the propyl substituent of DU172, and probes the orthosteric pocket more deeply, entering in contact with N1845.42 and H2516.52.

3.1.2. A2AAR structures

In the case of A2AAR, 57 experimental structures have been deposited in the PDB so far. An ensemble of 38 residues appears in contact with a ligand in at least one PDB structure, and among them, 10 residues are common to all (i.e., F168EL2, L2496.51, N2536.55, M2707.35, I2747.39) or nearly all (~90% or more) (i.e., L853.33, E169EL2, M1775.38, W2466.48, H2506.52) structures. Thus, these 10 residues define the orthosteric binding pocket of A2AAR regardless of the agonist or antagonist character of the ligand (Fig. 2B). Moreover, seven residues are only in proximity to antagonists (i.e., Y91.35, I642.62, I803.28, A813.29, K153EL2, A265EL3, P2667.31). However, they interact in a small fraction of all the PDB structures (less than 10%), so they cannot be considered a fingerprint for this category of compounds. Four residues are only in proximity to agonists (i.e., Q893.37, which is mutated into A89 in some structures, I923.40, C1855.46, and V1865.47). These residues define a pocket between TM3 and TM5 that is the deepest region of the orthosteric site, which typically hosts the ethyl-amido group at position 5′ of most of the agonists, but it is not explored by any of the antagonists.

In 9 PDB structures, the A2AAR is bound to an agonist, in particular to adenosine (PDB ID: 2YDO), NECA (PDB IDs: 2YDV, 5G53 and 6GDG), CGS21680 (PDB IDs: 4UG2, 4UHR) and UK432097 (PDB IDs: 3QAK, 5WF5, 5WF6). Among these structures, 5G53 and 6GDG are in the active state, with the receptor bound to an engineered G protein, mini-Gs (truncated Gαs) alone or in a heterotrimeric state, respectively. The agonists share a similar binding mode (Fig. 3B), where the ligands are surrounded by V843.32, L853.33, T883.36, F168EL2, M1775.38, W2466.48, L2496.51, N2536.55, M2707.35, I2747.39, S2777.42, H2787.43 in all the structures, and by E169EL2, Q893.37 (mutated into A89 in some structures), N1815.42, H2506.52 in nearly all (~90%) of the structures (Fig. 2B). Particularly, agonists are involved in a π–π stacking interaction between F168EL2 and the adenine base, in a bidentate H-bond between N2536.55 and the endocyclic N7 position of the adenine base and the exocyclic N6H, which interacts also with E169EL2 (missing only in structure 6GDG), in a H-bond between S2777.42 and the 3′-hydroxyl group and between H2787.43 and the 2′-hydroxyl group (Fig. 2E).

Few differences can be observed among the agonists: T883.36 makes a H-bond with all the ligands’ 5′-amido substituent, while adenosine’s 5′-hydroxyl group is involved in a water mediated H-bond with N1815.42 and H2506.52 (PDB ID: 2YDO), with H2506.52 interacting instead with the carbonyl of the 5′-amido substituent of NECA, CGS21680 and UK432097 in some of the reported structures. E169EL2, involved in a salt bridge with H264EL3, interacts through a H-bond with the unsubstituted N6H moiety of adenosine, NECA (not in 6GDG) and CGS21680. However, in the case of UK432097, the E169EL2–H264EL3 salt bridge is broken, and E169EL2 makes a bidentate H-bond with the ureidic group of the substituent at position 2, interacting also with Y2717.36. The ethylamide substituent at position 5′ distinguishes the other agonists from adenosine, and enters the binding pocket more deeply, making contacts with Q893.37 (mutated into A89 in some structures), I923.40, C1855.46, and V1865.47 in different structures. As compared to adenosine and NECA, agonists CGS21680 and UK432097 bear a long substituent at position 2, which fills the orthosteric binding pocket entering in contact with residues of TM2 (A632.61, I662.64 and S672.65), EL2 (L167EL2), EL3 (H264EL3) and TM7 (L2677.32 and Y2717.36) in some of the PDB structures. In addition, UK432097’s bulky N6-2, 2-diphenylethyl substituent also engages the tip of TM6 (I2526.54 and T2566.58). Several conserved water molecules are noted to surround the AR agonists. For example, the stabilizing contribution of the conserved water bridging the adenine nitrogen at position 3 and the 2′-hydroxyl has been explored using a combination of free energy perturbation (FEP) and chemical synthesis (Matricon et al., 2021). The penalty for driving this water out of the binding site is a dramatic reduction of affinity at all AR subtypes.

Forty-eight structures of antagonist-bound A2AARs are present in the PDB, with [1,2,4]triazolo[2,3-a][1,3,5]triazine derivative ZM241385 in 27 cases (PDB IDs: 3EML, 3PWH, 3VG9, 3VGA, 4EIY, 5IU4, 5JTB, 5K2A, 5K2B, 5K2C, 5K2D, 5NLX, 5NM2, 5NM4, 5OLG, 5UVI, 5VRA, 6AQF, 6JZH, 6LPJ, 6LPK, 6LPL, 6MH8, 6PS7, 6S0L, 6S0Q, 6WQA, a ZM241385-related compound in 4 cases (PDB IDs: 5IU7, 5IU8, 5IUA, 5IUB), a xanthine derivative in 5 cases (PDB IDs: 3REY, 3RFM, 5MZJ, 5MZP, 5N2R), a 1,2,4-triazin-3-amine derivative, with an aryl substituent at position 5 and 6, in 6 cases (PDB IDs: 3UZA, 3UZC, 5OLZ, 5OM1, 5OM4, 6GT3), vipadenant (PDB ID: 5OLH), tozadenant (PDB ID: 5OLO), LUAA47070 (PDB ID: 5OLV), a triazole-carboximidamide, Compound-1 (PDB ID: 5UIG), chromone 14 (4d) (PDB ID: 6ZDR), chromone 5d (PDB ID: 6ZDV) in one case each. Nearly all the antagonists are in contact (in at least 90% of the PDB structures) with the pattern of 10 residues cited at the beginning of the paragraph, comprising: L853.33, F168EL2, E169EL2, M1775.38, W2466.48, L2496.51, H2506.52, N2536.55, M2707.35 and I2747.39 (Fig. 2B). N2536.55 is involved in a direct H-bond with the ligand in all the structures, and a π–π stacking interaction with F168EL2 is often observed as well (Fig. 3D). A π–π edge-to-face interaction with H2506.52 is made by several ligands (ZM241385 and ZM241385-related compounds, 1,2,4-triazin-3-amine derivatives, vipadenant and LUAA47070), and a H-bond with E169EL2 is encountered in most complexes with ZM241385, ZM241385-related compounds and 1,2,4-triazin-3-amine derivatives, and in the case of vipadenant. However, some ligand-specific interactions are observed for these antagonists.

In the case of ZM241385 and ZM241385-related compounds, the [1,2,4]triazolo [1,5-a][1,3,5]triazine aromatic scaffold participates in the π–π face-to-face interaction with F168EL2, and N1 (or sometimes the furan oxygen atom) and the 7-amino group interacts with N2536.55 through bidentate H-bonding (even if with a deviation from ideal geometry in the case of structures 3VG9 and 3VGA) (Fig. 2F). In addition, a π–π edge-to-face interaction is observed between the furan ring and H2506.52, and a H-bond is formed between the 7-amino group and E169EL2 (missing only in structures 3PWH, 3VG9, 3VGA). A water molecule bridges a H-bond between Y2717.36 and the 5-amino group in the 75% of the PDB structures, and often co-crystalized water molecules interact also with nitrogen atoms at positions 3 and 4 of the ligand. Moreover, in the case of ZM241385-related compounds bearing a piperidine or piperazine moiety in the 5-substituent, a salt bridge with E169EL2 can be observed.

X-ray structures for A2AAR in complex with four different xanthine antagonists have been reported so far, in particular XAC (PDB ID: 3REY), caffeine (PDB IDs: 3RFM, 5MZP), theophylline (PDB ID: 5MZJ), PSB36 (PDB ID: 5N2R). Caffeine and theophylline are naturally-occurring prototypical AR antagonists, and antagonist XAC was introduced in 1985 as a high affinity, functionalized congener for coupling to carrier or reporter moieties (Jacobson, 2009). The extended position 8 ethylamino chain of XAC was later confirmed in an A2AAR structure to be reaching toward the extracellular region, thus explaining its ability to be acylated without preventing receptor binding. Xanthine bases are involved in the π–π face-to-face interaction with F168EL2, while the H-bond interaction with N2536.55 is fulfilled by the carbonyl at position 6 (maintained in both the alternate states of caffeine observed in structure 5MZP), accompanied by the free NH at position 7 in theophylline and PSB36. In order to maintain a H-bond between N2536.55 and the carbonyl of XAC in structure 3REY, N2536.55 is flipped as compared to its common conformation. Moreover, a water molecule mediates an interaction between carbonyl at position 2 of theophylline, caffeine and PSB36 with A813.29 in structures 5MZJ, 5MZP and 5N2R, respectively.

In the case of the 1,2,4-triazin-3-amine A2AAR antagonists such as T4E (PDB ID: 3UZC, 5OLZ, 5OM1, 5OM4), T4G (PDB ID: 3UZA) and AZD4635 (PDB ID: 6GT3), the triazine moiety participates in the π–π interaction with F168EL2. This interaction is face-to-face in most cases, apart from structure 3UZA where it is closer to an edge-to-face interaction, and in structure 3UZC where the conformation of F168EL2 does not permit an ideal π–π interaction. The exocyclic 3-amino group and the endocyclic N4 are involved in the typical bidentate H-bond with N2536.55 (conserved across all ARs), and the exocyclic 3-amino group also interacts with E169EL2 in all structures except 3UZA and 3UZC, where the salt bridge between E169EL2 and H264EL3 is not formed (Fig. 2F). The 5-phenyl group makes a π–π edge-to-face interaction with H2506.52, while the 6-aryl group, consisting in a phenol in the case of compound T4E (ODB IDs: 3UZC, 5OLZ, 5OM1, 5OM4), interacts though its p-hydroxyl group with H2787.43, and with A592.57 through a water mediated H-bond (not present in structure 3UZC).

Triazolo[4,5-d]pyrimidin-5-amine derivative vipadenant (PDB ID: 5OLH) interacts through its [1,2,3]triazolo[4,5-d]pyrimidine structure with F168EL2 and through its 5-amine and N6H with N2532.55, making the typical π–π stacking interaction and bidentate H-bond. The furan ring attached to the position 7 makes a π–π edge-to-face interaction with H2506.52 and an H-bond with A813.29.

In the case of tozadenant (PDB ID: 5OLO), the typical π–π interaction with F168EL2 is granted by the benzothiazole structure, which contributes through its nitrogen atom to the bidentate hydrogen with N2536.55, together with the exocyclic nitrogen attached to position 2 of the scaffold. T2566.58 is engaged in an additional H-bond with the hydroxyl group at the position 4 of the piperidine moiety.

Thiazole derivative LUAA47070 (PDB ID: 5OLV) lacks the π–π stacking with F168EL2, even if its benzamide structure assumes a position not far from enabling that interaction. The amide nitrogen makes the H-bond with N2536.55, accompanied by the nitrogen of the attached thiazole group, which is also engaged in the π–π edge-to-face interaction with H2506.52.

In the case of structure 5UIG (Sun et al., 2017), the interactions of antagonist Compound-1 with F168EL2 and N2536.55 are present with the 5-amino-1,2,3-triazole scaffold; while in the case of chromones 4d and 5d in structures 6ZDR and 6ZDV, the π–π face-to-face interaction with F168EL2 is accomplished by the chromone group, while N2536.55 makes a single H-bond with the nitrogen of the thiazole ring (Jespers et al., 2020).

3.1.3. Examples of AR models

The overall sequence identity between A3AR and A1AR is 46%, and it goes up to 56% focusing on TM regions; while the overall sequence identity between A3AR and A2AAR is 30%, with 47% value for the TM regions (sequence identity data retrieved from GPCRdb; https://gpcrdb.org/similaritymatrix/render; Kooistra et al., 2021). The high sequence similarity, especially of the TM region, among ARs makes A1AR and A2AAR good templates for homology modeling of the other AR subtypes. For instance, a hybrid model of A3AR has been recently built using an A2AAR structure as template for the TM helices and an A1AR structure for TM2 (Tosh, Salmaso, Rao, Bitant, et al., 2020; Tosh, Salmaso, Rao, Campbell, et al., 2020). The upper part of TM2 is observed to move outward from the TM bundle in the case of antagonist-bound A1AR, and this is in agreement with an established hypothesis for A3AR, where the outwardly displaced TM2 would allow binding of agonists with bulky substituents at position 2. A similar model was previously built using the TM2 of opsin, outward displaced as well (Tosh et al., 2012). An A3AR homology model enabled understanding of agonism by NECA-like amides vs 5′-CH2OH derivatives (Dal Ben et al., 2014). These models enable docking of agonists in a conformation analogous to that experimentally assumed by agonists at A1AR and A2AAR binding sites and described in the previous paragraphs. In particular, the H-bonds of hydroxyl groups 2′ and 3′, and of the hydroxyl or amidic moiety at 5′, the π–π interaction of the aromatic base, the bidentate H-bonds of the adenine 6-amino group and N7 might occur in the A3AR with residues at equivalent positions to the other ARs, in particular with H2727.43, S2717.42, T943.36, F168EL2 and N2506.55. An A2BAR homology model based on a ZM241385-A2AAR complex (PDB ID: 3EML) was reported (Kose et al., 2018), which features a close similarity to other AR subtypes. The proposed binding mode of a fluorescent A2BAR antagonist derived from a 3-propylxanthine bore a close resemblance to the xanthine pose in the XAC-A2AAR complex (PDB ID: 3REY).

4. Analysis of small molecule recognition by TM residues of P2Y receptors

4.1. P2YR structures and models

There are two subfamilies of P2YRs, based on both sequence identity and on G protein coupling. The eight P2Y receptors have much more sequence diversity than the four AR subtypes, with sequence identity values below 20% between human P2Y1-like and P2Y12-like P2YRs. The X-ray structures of two P2Y receptors (Table 2) have been reported (P2Y1 and P2Y12), representative of each of the two subfamilies (Zhang et al., 2015; Zhang, Zhang, Gao, Paoletta, et al., 2014; Zhang, Zhang, Gao, Zhang, et al., 2014). These two P2YR subtypes are much less similar to each other than the intrafamily similarity of ARs, with a sequence identity of 19% (sequence identity data retrieved from GPCRdb; https://gpcrdb.org/similaritymatrix/render; Kooistra et al., 2021). Ligand binding in the orthosteric binding sites have identified common residues in the recognition of nucleotide agonists, which are completely different between P2Y1 and P2Y12Rs. However, several features are in common between the P2Y1 and P2Y12Rs. There is a predominance of positively charged Lys and Arg residues in the EL regions, which coordinate the negatively charged phosphate moieties of the nucleotide ligands. Also, an unusual disulfide bridge is formed between the N-terminus and EL3 that is common to all P2YRs. This disulfide bridge was first discovered by SDM and led to early modeling suggesting that the ELs are important in the path taken by nucleotide ligands (Moro, Hoffmann, & Jacobson, 1999). Modeling of other P2YR subtypes based on the X-ray structures has been reported (Attah et al., 2020; Jung et al., 2020; Toti et al., 2017).

4.1.1. P2Y1R structures

Two X-ray structures have been deposited in the PDB so far for P2Y1R. The receptor is co-crystallized with an orthosteric antagonist, MRS2500 (PDB ID: 4XNW), in one structure, and to one allosteric antagonist, BPTU (PDB ID: 4XNV), in the other, and the two ligands bind to two different binding pockets, with no common residues (Fig. 4A and C). MRS2500 is a nucleotide derivative, but its ribose ring has been sterically constrained by a bicyclic ring system (methanocarba) in a receptor-preferred North conformation, unlike native ribose which is freely twisting between North and South conformations. 18 residues in P2Y1R appear in contact only with the nucleotide antagonist, while 11 residues are in proximity only to the allosteric antagonist, bound on the outer surface in contact with the phospholipid bilayer (Fig. 2C).

FIG. 4.

FIG. 4

(A) and (C) Experimental binding mode of the orthosteric antagonist MRS2500 (orange sticks) and of the negative allosteric modulator BPTU (green) to P2Y1R (dark and light gray when bound to MRS2500 and BPTU, in structure 4XNW and 4XNV, respectively). (B) and (D) Experimental binding mode of the agonist 2MeSATP (orange sticks) and of the antagonist AZD1283 (green) to P2Y12R (dark and light gray when bound to agonist and antagonist, in structures 4PY0 and 4NTJ, respectively). Relevant receptor residues discussed in the text are shown by sticks.

Bicyclic nucleotide derivative MRS2500 binds the receptor close to the extracellular region, at the interface among the N-terminus, extracellular loops and tip of TM6 and TM7. This antagonist is surrounded by C42N-ter, L44N-ter and K46N-ter of the N-terminus, Y1102.63, R195EL2, T201EL2, Y203EL2, D204EL2 and T205EL2 of EL2, N2836.58, A2866.61, R2876.62, Q291EL3 of EL3, N2997.28, V3027.31, Y3037.32, Y3067.35, R3107.39 (Fig. 2C). Among these residues, N2836.58 interacts though a bidentate H-bond with the endocyclic N7 and exocyclic N6H positions of the adenine scaffold, D204EL2, T205EL2, Y3067.35 and R3107.39 are involved in H-bonds with the 5′-phosphate group, which interacts also with R2876.62 though a water mediated H-bond, K46N-ter, R195EL2, T201EL2 and Y3037.32 are involved in H-bonds with the 3′-phosphate group, with K46N-ter and R195EL2 contributing also through salt bridges (Fig. 4A). In addition, Y3037.32 makes an edge-to-face π–π interaction with the adenine aromatic scaffold, and the C42N-ter carbonyl is involved in a halogen bond (at a ~180 degree angle) with the iodine atom at position 2 of the base. Differently, BPTU binds at the interface between TM1, TM2 and TM3 on the membrane side, out of the TM bundle, in a binding site defined by F621.43, F661.47, L1022.55, T1032.56, P1052.58, A1062.59, F119EL2, M1233.24, L1263.27, Q1273.28, I1303.31 (Fig. 2C), with the backbone carbonyl of L1022.55 interacting through a H-bond with the ureidic potion of the ligand, and F621.43 and F119EL2 through an edge-to-face π–π interaction with the phenyl and pyridine substituents of the urea group of BPTU (Fig. 4C).

4.1.2. P2Y12R structures

In the case of the P2Y12R, two agonist-bound (2MeSATP and 2MeSADP, in PDB ID: 4PY0 and 4PXZ, respectively) and one non-nucleotide antagonist bound (AZD1283, PDB ID: 4NTJ) X-ray structures are reported. The three ligands bind the receptor in a similar region, sharing the contacts with residues V1023.30, Y1053.33, F1063.34, Y1093.37, M1524.53, L1554.56, S1564.57, N1594.60, H1875.36, V1905.39, N1915.40, C1945.43, R2566.55, Y2596.58, and K2807.35. Thus, 15 residues in P2Y12R appear in proximity of both agonist and antagonist ligands, while 6 residues are in contact only with the antagonist, and 12 are in contact only with agonists (Fig. 2D). However, it is to be noted that the ELs in the P2Y12R structures are mostly undefined in the antagonist-bound structure but well-defined with nucleotide agonists bound.

Y1053.33 is involved in a π–π stacking interaction in all the structures, interacting with the adenine base in the case of the agonists and with the pyridine in the case of the antagonists; R2566.55 and K2807.35 contribute through H-bonds with the 5′-phosphate in the case of the agonists and with the sulfonyl-carbamoyl substituent in the case of AZD1283. However, ligand binding of the non-nucleotide antagonist AZD1283 is unlike other purinergic receptors and has no similar structure among other GPCRs. The antagonist-bound structure consistently deviates from the agonist-bound P2Y12R structures: the tips of TM5, TM6 and TM7 bent toward the membrane environment, and the disulfide bond between C175EL2 and C973.25, linking EL2 to TM3, is broken. This is unusual for a GPCR structure, as this same disulfide is considered a conserved structural feature in other Class A receptors. AZD1283 in fact spans the TM bundle, going from a cleft defined by TM3, TM4 and TM5, where the ethyl ester engages N1594.60 in a H-bond and occupies the same hydrophobic pocket that hosts the 2-methylsulfanyl substituent of 2MeSADP and 2MeSATP in the agonist-bound structures, to the interface between TM7 and TM6, where it is surrounded by T2837.38, K2807.35, V2797.34, L2767.31, Y2596.58, R2566.55, A2556.54, F2526.51, stabilizing the ligand’s benzylic group through an edge-to-face π–π interaction (Fig. 4D). Differently, the TM bundle of the agonist-bound structures is more compact, and the two agonists share the same binding mode where the adenine base, involved in the π–π stacking interaction with Y1053.33, directs the exocyclic amino group toward the bottom of the pocket, where it is engaged in a H-bond with N1915.40, and the ribose ring toward the extracellular region, where it makes H-bonds with H1875.36 through the 2′-hydroxyl group and K179EL2 and C973.25 through the 3′-hydroxyl group (Fig. 4B). The α-phosphate interacts through H-bonds with Y1053.33, Q2636.62, in addition to the aforementioned R2566.55 and K2807.35, which have also a salt bridge character. The β-phosphate interacts via H-bonds with Y2596.58, K2807.35 and R933.21 (also salt bridge in the case of the last two), while the γ-phosphate of 2MeSATP interacts with R933.21, C175EL2, S176EL2.

4.1.3. Example of P2YR models

As an example of modeling for P2YRs, we report P2Y14R subtype, whose antagonists may have therapeutic interest in treating inflammatory diseases, asthma, acute kidney injury, etc. P2Y14R share with P2Y12R 44% sequence (44% overall sequence identity, 52% sequence identity of TM regions) (sequence identity data retrieved from GPCRdb; https://gpcrdb.org/similaritymatrix/render; Kooistra et al., 2021), so X-ray structures of the second can be used as templates for homology modeling. Specifically, a model was built using the 2-MeSADP-bound P2Y12R structure as a template and then refined using MD (Salmaso & Jacobson, 2020a) simulations after docking of the known biaryl antagonist PPTN (Junker et al., 2016). This model has been used to drive the design and synthesis of PPTN analogs as P2Y14R antagonists. The suggested binding mode of PPTN and related compounds involves a salt bridge between the ligand’s carboxylate and K772.60 and K2777.35, which behave also as H-bonds donor together with Y1023.33 (Jung et al., 2020; Yu et al., 2018). Cation-π interactions stabilize the aromatic moieties of the ligands, involving R2536.55 and R2747.32. Although the pose of P2Y14R antagonists does not resemble the experimental binding mode of P2Y12R ligands, and even though PPTN is selective for P2Y14R subtype, some of the residues involved in binding are equivalent to those enumerated for P2Y12R in the previous paragraph, i.e., Y1023.33, K2777.35 and R2536.55.

5. Receptor domains involved in dimerization

There is still some controversy about the stoichiometric nature of class A GPCRs (Felce et al., 2017). While class C GPCRs are known to dimerize, there is no consensus in the case of class A receptors. Evidence suggests that various ARs and P2YRs can form putative dimers, either homo- or heterodimers, with other receptor protomers (Borroto-Escuela et al., 2020; Hill, May, Kellam, & Woolard, 2014; Hinz et al., 2018; Navarro et al., 2016; Neumann, Müller, & Namasivayam, 2020). The contact regions for this dimerization have been determined experimentally, in limited cases, and through modeling (Al-Shar’i & Al-Balas, 2019). In some cases, the association between protomers is facilitated by loop regions of the receptors, for example, intracellular loops of the A2AAR (Fernandez-Duenas et al., 2012), but also a critical role of TM residues is proposed (Johnston, Wang, Provasi, & Filizola, 2012; Townsend-Nicholson, Altwaijry, Potterton, Morao, & Heifetz, 2019). A tool for exploring the location of residues involved in GPCR dimerization based on reported structures is available on the web (DIMERBOW, http://lmc.uab.es/dimerbow/; García-Recio et al., 2020).

An early modeling study of potential A3AR homodimerization pointed to a likely TM4-TM5 interface (Kim & Jacobson, 2006). A3AR homodimerization was later demonstrated pharmacologically (May, Bridge, Stoddart, Briddon, & Hill, 2011), and the TM4-TM5 interface was confirmed as an interface also for A2AAR-D2R heterodimerization, by means of peptide segments from the sequences of A2AAR’s TM4 and TM5 which interfered with heterodimerization (Borroto-Escuela et al., 2018). The same interface, which was experimentally observed in the X-ray structure of the oligomeric β1-adrenergic receptor (Huang, Chen, Zhang, & Huang, 2013), was proposed for homodimerization of A1AR and A2AAR, while the same work suggested the TM5-TM6 interface for a A1AR-A2AAR heterotetramer (Navarro et al., 2016). Moreover, A1AR crystallized in dimeric form in one of the X-ray structures (PDB ID: 5UEN), resulting in the burial of hydrophobic areas of TM4 and TM5 (Glukhova et al., 2017). The role of TM5 in A2AAR dimerization was previously anticipated in a study exploring the self-assembly of TM5 in oligomeric structures, which also found that a mutation of M193 (5.54) alters the dimerization capability of the whole length A2AAR (Thevenin, Lazarova, Roberts, & Robinson, 2005).

6. Summary

We compare the GPCR-ligand interactions and highlight important residues for recognition—from both X-ray crystallographic or cryo-EM structures for purinergic receptors. These include A1 and A2AARs and P2Y1 and P2Y12Rs that respond to nucleotides. Molecular modeling, including MD simulation, has been successful in predicting ligand interactions across most of this family of 12 purinergic GPCRs (Salmaso & Jacobson, 2020a). This review catalogs the residues of the binding pocket, with specific interest in transmembrane helical (TM) regions, that engage in contacts with ligands or with other GPCR protomers in dimeric forms. In most cases, orthosteric ligands are represented, except for one allosteric P2Y1 antagonist. There is extensive conservation of recognition motifs within the adenosine receptor, but the P2Y1 and P2Y12 receptors are each structurally distinct in their ligand recognition.

Acknowledgment

We thank the NIDDK Intramural Research Program for support (ZIADK-031126).

References

  1. Al-Shar’i NA, & Al-Balas QA (2019). Molecular dynamics simulations of adenosine receptors: Advances, applications and trends. Current Pharmaceutical Design, 25, 783–816. [DOI] [PubMed] [Google Scholar]
  2. Attah IY, Neumann A, Al-Hroub H, Rafehi M, Baqi Y, Namasivayam V, et al. (2020). Ligand binding and activation of UTP-activated G protein-coupled P2Y2 and P2Y4 receptors elucidated by mutagenesis, pharmacological and computational studies. Biochimica et Biophysica Acta—General Subjects, 1864(3), 129501. 10.1016/j.bbagen.2019.129501. [DOI] [PubMed] [Google Scholar]
  3. Ballante F, Rudling A, Zeifman A, Luttens A, Vo DD, Irwin JJ, et al. (2020). Docking finds GPCR ligands in dark chemical matter. Journal of Medicinal Chemistry, 63(2), 613–620. 10.1021/acs.jmedchem.9b01560. [DOI] [PubMed] [Google Scholar]
  4. Ballesteros JA, & Weinstein H (1995). Integrated methods for the construction of three dimensional models and computational probing of structure function relations in G protein-coupled receptors. Methods in Neurosciences, 25, 366–428. [Google Scholar]
  5. Batyuk A, Galli L, Ishchenko A, Han GW, Gati C, Popov PA, et al. (2016). Native phasing of x-ray free-electron laser data for a G protein-coupled receptor. Science Advances, 2, e1600292. 10.1126/sciadv.1600292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, Weissig H, et al. (2000). The Protein Data Bank. Nucleic Acids Research, 28(1), 235–242. 10.1093/nar/28.1.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Borodovsky A, Barbon CM, Wang Y, Ye M, Prickett L, Chandra D, et al. (2020). Small molecule AZD4635 inhibitor of A2AR signaling rescues immune cell function including CD103(+) dendritic cells enhancing anti-tumor immunity. Journal for Immunotherapy of Cancer, 8(2). 10.1136/jitc-2019-000417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Borroto-Escuela DO, Ferraro L, Narvaez M, Tanganelli S, Beggiato S, Liu F, et al. (2020). Multiple adenosine-dopamine (A2A-D2 like) heteroreceptor complexes in the brain and their role in schizophrenia. Cell, 9(5). 10.3390/cells9051077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Borroto-Escuela DO, Rodriguez D, Romero-Fernandez W, Kapla J, Jaiteh M, Ranganathan A, et al. (2018). Mapping the Interface of a GPCR dimer: A structural model of the A2A adenosine and D2 dopamine receptor heteromer. Frontiers in Pharmacology, 9, 829. 10.3389/fphar.2018.00829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Broecker J, Morizumi T, Ou W-L, Klingel V, Kuo A, Kissick DJ, et al. (2018). High-throughput in situ X-ray screening of and data collection from protein crystals at room temperature and under cryogenic conditions. Nature Protocols, 13, 260–292. 10.1038/nprot.2017.135. [DOI] [PubMed] [Google Scholar]
  11. Cao R, Giorgetti A, Bauer A, Neumaier B, Rossetti G, & Carloni P (2018). Role of extracellular loops and membrane lipids for ligand recognition in the neuronal adenosine receptor type 2A: An enhanced sampling simulation study. Molecules, 23(10). 10.3390/molecules23102616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Carpenter B, Nehm e R, Warne T, Leslie AGW, & Tate CG (2016). Structure of the adenosine A(2A) receptor bound to an engineered G protein. Nature, 536, 104–107. 10.1038/nature18966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Carpenter B, & Tate CG (2017). Active state structures of G protein-coupled receptors highlight the similarities and differences in the G protein and arrestin coupling interfaces. Current Opinion in Structural Biology, 45, 124–132. 10.1016/j.sbi.2017.04.010. [DOI] [PubMed] [Google Scholar]
  14. Cheng RKY, Segala E, Robertson N, Deflorian F, Doré AS, Errey JC, et al. (2017). Structures of human A1 and A2A adenosine receptors with xanthines reveal determinants of selectivity. Structure, 25, 1275–1285.e4. 10.1016/j.str.2017.06.012. [DOI] [PubMed] [Google Scholar]
  15. Ciancetta A, & Jacobson KA (2018). Breakthrough in GPCR crystallography and its impact on computer-aided drug design. Methods in Molecular Biology, 1705, 45–72. 10.1007/978-1-4939-7465-8_3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Ciancetta A, Rubio P, Lieberman DI, & Jacobson KA (2019). A3 adenosine receptor activation mechanisms: Molecular dynamics analysis of inactive, active, and fully active states. Journal of Computer-Aided Molecular Design, 33(11), 983–996. 10.1007/s10822-019-00246-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Congreve M, Andrews SP, Doré AS, Hollenstein K, Hurrell E, Langmead CJ, et al. (2012). Discovery of 1,2,4-triazine derivatives as adenosine A(2A) antagonists using structure based drug design. Journal of Medicinal Chemistry, 55, 1898–1903. 10.1021/jm201376w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Congreve M, de Graaf C, Swain NA, & Tate CG (2020). Impact of GPCR structures on drug discovery. Cell, 181(1), 81–91. 10.1016/j.cell.2020.03.003. [DOI] [PubMed] [Google Scholar]
  19. Dal Ben D, Buccioni M, Lambertucci C, Kachler S, Falgner N, Marucci G, et al. (2014). Different efficacy of adenosine and NECA derivatives at the human A3 adenosine receptor: Insight into the receptor activation switch. Biochemical Pharmacology, 87(2), 321–331. [DOI] [PubMed] [Google Scholar]
  20. Dal Ben D, Lambertucci C, Buccioni M, Martí Navia A, Marucci G, Spinaci A, et al. (2019). Non-nucleoside agonists of the adenosine receptors: An overview. Pharmaceuticals (Basel, Switzerland), 12(4), 150. 10.3390/ph12040150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Doré AS, Robertson N, Errey JC, Ng I, Hollenstein K, Tehan B, et al. (2011). Structure of the adenosine A(2A) receptor in complex with ZM241385 and the xanthines XAC and caffeine. Structure, 19, 1283–1293. 10.1016/j.str.2011.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Draper-Joyce CJ, Khoshouei M, Thal DM, Liang Y-L, Nguyen ATN, Furness SGB, et al. (2018). Structure of the adenosine-bound human adenosine A1 receptor-Gi complex. Nature, 558, 559–563. 10.1038/s41586-018-0236-6. [DOI] [PubMed] [Google Scholar]
  23. Eddy MT, Lee M-Y, Gao Z-G, White KL, Didenko T, Horst R, et al. (2018). Allosteric coupling of drug binding and intracellular signaling in the A2A adenosine receptor. Cell, 172, 68–80.e12. 10.1016/j.cell.2017.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Felce JH, Latty SL, Knox RG, Mattick SR, Lui Y, Lee SF, et al. (2017). Receptor quaternary organization explains G protein-coupled receptor family structure. Cell Reports, 20(11), 2654–2665. 10.1016/j.celrep.2017.08.072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Fernandez-Duenas V, Gomez-Soler M, Jacobson KA, Kumar ST, Fuxe K, Borroto-Escuela DO, et al. (2012). Molecular determinants of A2AR-D2R allosterism: Role of the intracellular loop 3 of the D2R. Journal of Neurochemistry, 123(3), 373–384. 10.1111/j.1471-4159.2012.07956.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Filipek S (2019). Molecular switches in GPCRs. Current Opinion in Structural Biology, 55, 114–120. 10.1016/j.sbi.2019.03.017. [DOI] [PubMed] [Google Scholar]
  27. García-Nafría J, Lee Y, Bai X, Carpenter B, & Tate CG (2018). Cryo-EM structure of the adenosine A2A receptor coupled to an engineered heterotrimeric G protein. elife, 7. 10.7554/eLife.35946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. García-Recio A, Navarro G, Franco R, Olivella M, Guixà-González R, & Cordomí A (2020). DIMERBOW: Exploring possible GPCR dimer interfaces. Bioinformatics, 36, 3271–3272. [DOI] [PubMed] [Google Scholar]
  29. Glukhova A, Thal DM, Nguyen AT, Vecchio EA, Jorg M, Scammells PJ, et al. (2017). Structure of the adenosine A1 receptor reveals the basis for subtype selectivity. Cell, 168(5), 867–877.e813. 10.1016/j.cell.2017.01.042. [DOI] [PubMed] [Google Scholar]
  30. Gutierrez-de-Teran H, Sallander J, & Sotelo E (2017). Structure-based rational design of adenosine receptor ligands. Current Topics in Medicinal Chemistry, 17(1), 40–58. 10.2174/1568026616666160719164207. [DOI] [PubMed] [Google Scholar]
  31. Hill SJ, May LT, Kellam B, & Woolard J (2014). Allosteric interactions at adenosine A (1) and A(3) receptors: New insights into the role of small molecules and receptor dimerization. British Journal of Pharmacology, 171(5), 1102–1113. 10.1111/bph.12345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Hino T, Arakawa T, Iwanari H, Yurugi-Kobayashi T, Ikeda-Suno C, Nakada-Nakura Y, et al. (2012). G-protein-coupled receptor inactivation by an allosteric inverse-agonist antibody. Nature, 482, 237–240. 10.1038/nature10750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Hinz S, Navarro G, Borroto-Escuela D, Seibt BF, Ammon YC, de Filippo E, et al. (2018). Adenosine A2A receptor ligand recognition and signaling is blocked by A2B receptors. Oncotarget, 9(17), 13593–13611. 10.18632/oncotarget.24423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Huang J, Chen S, Zhang JJ, & Huang XY (2013). Crystal structure of oligomeric beta1-adrenergic G protein-coupled receptors in ligand-free basal state. Nature Structural & Molecular Biology, 20(4), 419–425. 10.1038/nsmb.2504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Ihara K, Hato M, Nakane T, Yamashita K, Kimura-Someya T, Hosaka T, et al. (2020). Isoprenoid-chained lipid EROCOC17+4: A new matrix for membrane protein crystallization and a crystal delivery medium in serial femtosecond crystallography. Scientific Reports, 10, 19305. 10.1038/s41598-020-76277-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Ishchenko A, Stauch B, Han GW, Batyuk A, Shiriaeva A, Li C, et al. (2019). Toward G protein-coupled receptor structure-based drug design using X-ray lasers. IUCrJ, 6, 1106–1119. 10.1107/S2052252519013137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Jaakola VP, Griffith MT, Hanson MA, Cherezov V, Chien EY, Lane JR, et al. (2008). The 2.6 angstrom crystal structure of a human A2A adenosine receptor bound to an antagonist. Science, 322(5905), 1211–1217. 10.1126/science.1164772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Jacobson KA (2009). Functionalized congener approach to the design of ligands for G protein–coupled receptors (GPCRs). Bioconjugate Chemistry, 20, 1816–1835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Jacobson KA, Delicado EG, Gachet C, Kennedy C, von Kügelgen I, Li B, et al. (2020). Update of P2Y receptor pharmacology: IUPHAR Review 27. British Journal of Pharmacology, 177, 2413–2433. 10.1111/bph.15005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Jacobson KA, IJzerman AP, & Müller CE (2021). Medicinal chemistry of P2 and adenosine receptors: Common scaffolds adapted for multiple targets. Biochemical Pharmacology, 187, 114311. 10.1016/j.bcp.2020.114311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Jespers W, Oliveira A, Prieto-Diaz R, Majellaro M, Aqvist J, Sotelo E, et al. (2017). Structure-based design of potent and selective ligands at the four adenosine receptors. Molecules, 22(11). 10.3390/molecules22111945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Jespers W, Verdon G, Azuaje J, Majellaro M, Keranen H, Garcia-Mera X, et al. (2020). X-ray crystallography and free energy calculations reveal the binding mechanism of A2A adenosine receptor antagonists. Angewandte Chemie (International Ed. in English), 59(38), 16536–16543. 10.1002/anie.202003788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Johnston JM, Wang H, Provasi D, & Filizola M (2012). Assessing the relative stability of dimer interfaces in g protein-coupled receptors. PLoS Computational Biology, 8(8). 10.1371/journal.pcbi.1002649, e1002649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Jung YH, Yu J, Wen Z, Salmaso V, Karcz TP, Phung NB, et al. (2020). Exploration of alternative scaffolds for P2Y14 receptor antagonists containing a biaryl core. Journal of Medicinal Chemistry, 63(17), 9563–9589. 10.1021/acs.jmedchem.0c00745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Junker A, Balasubramanian R, Ciancetta A, Uliassi E, Kiselev E, Martiriggiano C, et al. (2016). Structure-based design of 3-(4-aryl-1H-1,2,3-triazol-1-yl)-biphenyl derivatives as P2Y14 receptor antagonists. Journal of Medicinal Chemistry, 59(13), 6149–6168. 10.1021/acs.jmedchem.6b00044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Kim SK, & Jacobson KA (2006). Computational prediction of homodimerization of the A3 adenosine receptor. Journal of Molecular Graphics & Modelling, 25(4), 549–561. 10.1016/j.jmgm.2006.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Kooistra AJ, Mordalski S, Pandy-Szekeres G, Esguerra M, Mamyrbekov A, Munk C, et al. (2021). GPCRdb in 2021: Integrating GPCR sequence, structure and function. Nucleic Acids Research, 49(D1), D335–D343. 10.1093/nar/gkaa1080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Kose M, Gollos S, Karcz T, Fiene A, Heisig F, Behrenswerth A, et al. (2018). Fluorescent-labeled selective adenosine A2B receptor antagonist enables competition binding assay by flow cytometry. Journal of Medicinal Chemistry, 61(10), 4301–4316. 10.1021/acs.jmedchem.7b01627. [DOI] [PubMed] [Google Scholar]
  49. Lebon G, Edwards PC, Leslie AGW, & Tate CG (2015). Molecular determinants of CGS21680 binding to the human adenosine A2A receptor. Molecular Pharmacology, 87, 907–915. 10.1124/mol.114.097360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Lebon G, Warne T, Edwards PC, Bennett K, Langmead CJ, Leslie AG, et al. (2011). Agonist-bound adenosine A2A receptor structures reveal common features of GPCR activation. Nature, 474(7352), 521–525. 10.1038/nature10136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Lee M-Y, Geiger J, Ishchenko A, Han GW, Barty A, White TA, et al. (2020). Harnessing the power of an X-ray laser for serial crystallography of membrane proteins crystallized in lipidic cubic phase. IUCrJ, 7, 976–984. 10.1107/S2052252520012701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Liu W, Chun E, Thompson AA, Chubukov P, Xu F, Katritch V, et al. (2012). Structural basis for allosteric regulation of GPCRs by sodium ions. Science, 337, 232–236. 10.1126/science.1219218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Martin-Garcia JM, Conrad CE, Nelson G, Stander N, Zatsepin NA, Zook J, et al. (2017). Serial millisecond crystallography of membrane and soluble protein microcrystals using synchrotron radiation. IUCrJ, 4, 439–454. 10.1107/S205225251700570X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Martin-Garcia JM, Zhu L, Mendez D, Lee M-Y, Chun E, Li C, et al. (2019). High-viscosity injector-based pink-beam serial crystallography of microcrystals at a synchrotron radiation source. IUCrJ, 6, 412–425. 10.1107/S205225251900263X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Marullo S, Doly S, Saha K, Enslen H, Scott MGH, & Coureuil M (2020). Mechanical GPCR activation by traction forces exerted on receptor N-glycans. ACS Pharmacology and Translational Science, 3(2), 171–178. 10.1021/acsptsci.9b00106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Matricon P, Suresh RR, Gao ZG, Panel N, Jacobson KA, & Carlsson J (2021). Ligand design by targeting a binding site water. Chemical Science, 12, 960–968. 10.1039/d0sc04938g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. May LT, Bridge LJ, Stoddart LA, Briddon SJ, & Hill SJ (2011). Allosteric interactions across native adenosine-A3 receptor homodimers: Quantification using single-cell ligand-binding kinetics. The FASEB Journal, 25(10), 3465–3476. 10.1096/fj.11-186296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Melnikov I, Polovinkin V, Kovalev K, Gushchin I, Shevtsov M, Shevchenko V, et al. (2017). Fast iodide-SAD phasing for high-throughput membrane protein structure determination. Science Advances, 3, e1602952. 10.1126/sciadv.1602952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Moro S, Hoffmann C, & Jacobson KA (1999). Role of the extracellular loops of G protein-coupled receptors in ligand recognition: A molecular modeling study of the human P2Y1 receptor. Biochemistry, 38, 3498–3507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Nass K, Cheng R, Vera L, Mozzanica A, Redford S, & Ozerov D (2020). Advances in long-wavelength native phasing at X-ray free-electron lasers. IUCrJ, 7, 965–975. 10.1107/S2052252520011379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Navarro G, Cordomi A, Zelman-Femiak M, Brugarolas M, Moreno E, Aguinaga D, et al. (2016). Quaternary structure of a G-protein-coupled receptor heterotetramer in complex with Gi and Gs. BMC Biology, 14, 26. 10.1186/s12915-016-0247-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Neumann A, Müller CE, & Namasivayam V (2020). P2Y1-like nucleotide receptors—Structures, molecular modeling, mutagenesis, and oligomerization. WIREs Computational Molecular Science, 10, e1464. 10.1002/wcms.1464. [DOI] [Google Scholar]
  63. Pedata F, Pugliese AM, Coppi E, Dettori I, Maraula G, Cellai L, et al. (2014). Adenosine A2A receptors modulate acute injury and neuroinflammation in brain ischemia. Mediators of Inflammation, 2014, 805198. 10.1155/2014/805198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Pin JP, Kniazeff J, Prezeau L, Liu JF, & Rondard P (2019). GPCR interaction as a possible way for allosteric control between receptors. Molecular and Cellular Endocrinology, 486, 89–95. 10.1016/j.mce.2019.02.019. [DOI] [PubMed] [Google Scholar]
  65. Qu X, Wang D, & Wu B (2020). Chapter 1—Progress in GPCR structure determination. In Jastrzebska B, & Park PSH (Eds.), GPCRs (pp. 3–22). Academic Press. [Google Scholar]
  66. Rodriguez D, Chakraborty S, Warnick E, Crane S, Gao ZG, O’Connor R, et al. (2016). Structure-based screening of uncharted chemical space for atypical adenosine receptor agonists. ACS Chemical Biology, 11(10), 2763–2772. 10.1021/acschembio.6b00357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Rodriguez-Espigares I, Torrens-Fontanals M, Tiemann JKS, Aranda-Garcia D, Ramirez-Anguita JM, Stepniewski TM, et al. (2020). GPCRmd uncovers the dynamics of the 3D-GPCRome. Nature Methods, 17(8), 777–787. 10.1038/s41592-020-0884-y. [DOI] [PubMed] [Google Scholar]
  68. Rucktooa P, Cheng RKY, Segala E, Geng T, Errey JC, Brown GA, et al. (2018). Towards high throughput GPCR crystallography: In meso soaking of adenosine A2A receptor crystals. Scientific Reports, 8, 41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Safdari HA, Pandey S, Shukla AK, & Dutta S (2018). Illuminating GPCR signaling by cryo-EM. Trends in Cell Biology, 28(8), 591–594. 10.1016/j.tcb.2018.06.002. [DOI] [PubMed] [Google Scholar]
  70. Salmaso V, & Jacobson KA (2020a). In silico drug design for purinergic GPCRs: Overview on molecular dynamics applied to adenosine and P2Y receptors. Biomolecules, 10(6). 10.3390/biom10060812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Salmaso V, & Jacobson KA (2020b). Purinergic signaling: Impact of GPCR structures on rational drug design. ChemMedChem, 15(21), 1958–1973. 10.1002/cmdc.202000465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Segala E, Guo D, Cheng RKY, Bortolato A, Deflorian F, Doré AS, et al. (2016). Controlling the dissociation of ligands from the adenosine A2A receptor through modulation of salt bridge strength. Journal of Medicinal Chemistry, 59, 6470–6479. 10.1021/acs.jmedchem.6b00653. [DOI] [PubMed] [Google Scholar]
  73. Shimazu Y, Tono K, Tanaka T, Yamanaka Y, Nakane T, Mori C, et al. (2019). High-viscosity sample-injection device for serial femtosecond crystallography at atmospheric pressure. Journal of Applied Crystallography, 52, 1280–1288. 10.1107/S1600576719012846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Sun B, Bachhawat P, Chu ML, Wood M, Ceska T, Sands ZA, et al. (2017). Crystal structure of the adenosine A2A receptor bound to an antagonist reveals a potential allosteric pocket. Proceedings of the National Academy of Sciences of the United States of America, 114(8), 2066–2071. 10.1073/pnas.1621423114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Thevenin D, Lazarova T, Roberts MF, & Robinson CR (2005). Oligomerization of the fifth transmembrane domain from the adenosine A2A receptor. Protein Science, 14(8), 2177–2186. 10.1110/ps.051409205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Tosh DK, Deflorian F, Phan K, Gao ZG, Wan TC, Gizewski E, et al. (2012). Structure-guided design of A(3) adenosine receptor-selective nucleosides: Combination of 2-arylethynyl and bicyclo[3.1.0]hexane substitutions. Journal of Medicinal Chemistry, 55(10), 4847–4860. 10.1021/jm300396n. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Tosh DK, Salmaso V, Rao H, Bitant A, Fisher CL, Lieberman DI, et al. (2020). Truncated (N)-methanocarba nucleosides as partial agonists at mouse and human A3 adenosine receptors: Affinity enhancement by N(6)-(2-phenylethyl) substitution. Journal of Medicinal Chemistry, 63(8), 4334–4348. 10.1021/acs.jmedchem.0c00235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Tosh DK, Salmaso V, Rao H, Campbell R, Bitant A, Gao ZG, et al. (2020). Direct comparison of (N)-methanocarba and ribose-containing 2-arylalkynyladenosine derivatives as A3 receptor agonists. ACS Medicinal Chemistry Letters, 11, 1935–1941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Toti KS, Jain S, Ciancetta A, Balasubramanian R, Chakraborty S, Surujdin R, et al. (2017). Pyrimidine nucleotides containing a (S)-methanocarba ring as P2Y6 receptor agonists. Medicinal Chemistry Communications, 8(10), 1897–1908. 10.1039/C7MD00397H. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Townsend-Nicholson A, Altwaijry N, Potterton A, Morao I, & Heifetz A (2019). Computational prediction of GPCR oligomerization. Current Opinion in Structural Biology, 55, 178–184. 10.1016/j.sbi.2019.04.005. [DOI] [PubMed] [Google Scholar]
  81. van Rhee AM, & Jacobson KA (1996). Molecular architecture of G protein-coupled receptors. Drug Development Research, 37(1), 1–38. . [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Vasiliauskaite-Brooks I, Healey RD, & Granier S (2019). 7TM proteins are not necessarily GPCRs. Molecular and Cellular Endocrinology, 491. 10.1016/j.mce.2019.02.009, 110397. [DOI] [PubMed] [Google Scholar]
  83. Velazhahan V, Ma N, Pandy-Szekeres G, Kooistra AJ, Lee Y, Gloriam DE, et al. (2021). Structure of the class D GPCR Ste2 dimer coupled to two G proteins. Nature, 589(7840), 148–153. 10.1038/s41586-020-2994-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Venkatakrishnan AJ, Deupi X, Lebon G, Tate CG, Schertler GF, & Babu MM (2013). Molecular signatures of G-protein-coupled receptors. Nature, 494(7436), 185–194. 10.1038/nature11896. [DOI] [PubMed] [Google Scholar]
  85. Venkatakrishnan AJ, Ma AK, Fonseca R, Latorraca NR, Kelly B, Betz RM, et al. (2019). Diverse GPCRs exhibit conserved water networks for stabilization and activation. Proceedings of the National Academy of Sciences of the United States of America, 116(8), 3288–3293. 10.1073/pnas.1809251116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Wacker D, Stevens RC, & Roth BL (2017). How ligands illuminate GPCR molecular pharmacology. Cell, 170(3), 414–427. 10.1016/j.cell.2017.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Warne T, Edwards PC, Dore AS, Leslie AGW, & Tate CG (2019). Molecular basis for high-affinity agonist binding in GPCRs. Science, 364(6442), 775–778. 10.1126/science.aau5595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Weinert T, Olieric N, Cheng R, Brünle S, James D, & Ozerov D (2017). Serial millisecond crystallography for routine room-temperature structure determination at synchrotrons. Nature Communications, 8, 542. 10.1038/s41467-017-00630-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. White KL, Eddy MT, Gao ZG, Han GW, Lian T, Deary A, et al. (2018). Structural connection between activation microswitch and allosteric sodium site in GPCR signaling. Structure, 26(2), 259–269.e255. 10.1016/j.str.2017.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Wold EA, Chen J, Cunningham KA, & Zhou J (2019). Allosteric modulation of class A GPCRs: Targets, agents, and emerging concepts. Journal of Medicinal Chemistry, 62(1), 88–127. 10.1021/acs.jmedchem.8b00875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Xu F, Wu H, Katritch V, Han GW, Jacobson KA, Gao ZG, et al. (2011). Structure of an agonist-bound human A2A adenosine receptor. Science, 332(6027), 322–327. 10.1126/science.1202793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Yu J, Ciancetta A, Dudas S, Duca S, Lottermoser J, & Jacobson KA (2018). Structure-guided modification of heterocyclic antagonists of the P2Y14 receptor. Journal of Medicinal Chemistry, 61(11), 4860–4882. 10.1021/acs.jmedchem.8b00168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Zhang D, Gao ZG, Zhang K, Kiselev E, Crane S, Wang J, et al. (2015). Two disparate ligand-binding sites in the human P2Y1 receptor. Nature, 520(7547), 317–321. 10.1038/nature14287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Zhang J, Zhang K, Gao ZG, Paoletta S, Zhang D, Han GW, et al. (2014). Agonist-bound structure of the human P2Y12 receptor. Nature, 509(7498), 119–122. 10.1038/nature13288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Zhang K, Zhang J, Gao ZG, Zhang D, Zhu L, Han GW, et al. (2014). Structure of the human P2Y12 receptor in complex with an antithrombotic drug. Nature, 509(7498), 115–118. 10.1038/nature13083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Zhou Q, Yang D, Wu M, Guo Y, Guo W, Zhong L, et al. (2019). Common activation mechanism of class A GPCRs. eLife, 8. 10.7554/eLife.50279. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES