Abstract

Endothelins and their receptors, type A (ETA) and type B (ETB), modulate vital cellular processes, including growth, survival, invasion and angiogenesis, through multiple G proteins. This review highlights the structural determinations of these receptors by X-ray crystallography and cryo-electron microscopy, and their activation mechanisms by endothelins. Explorations of the conformational changes upon receptor activation have provided insights into the unique G-protein coupling feature of the endothelin receptors. The review further delves into the binding modes of the clinical antagonist and the inverse agonists. These findings significantly contribute to understanding the mechanism of G-protein activation and have potential implications for drug development, particularly in the context of vasodilatory antagonists and agonists targeting the endothelin receptors.

Endothelins are 21 amino acid peptides primarily derived from the endothelium (1,) and play a crucial role in maintaining vascular homeostasis. Among these peptides, the potent vasoconstrictor ET-1 was the first discovered (2,3,). The transmission of signals by ET-1 is mediated by two endothelin receptor (ETR) subtypes, ETA and ETB, which are class A G-protein-coupled receptors (GPCRs) (4,5,). The ETA and ETB receptors are promiscuous GPCRs capable of activating multiple types of G proteins (6,7,) and regulate essential cellular processes such as growth, survival, invasion and angiogenesis (8,9,). In the vascular system, both receptors are coupled to Gq and have opposing functions. ETA primarily mediates vasoconstriction, which is prolonged upon irreversible binding to ET-1. Conversely, ETB induces vasorelaxation through the nitric oxide-mediated pathway and functions as a clearance receptor by removing circulating ET-1 via lysosomal degradation (10,). In astrocytes, ETB stimulates a Gi signalling pathway and thereby inhibits intercellular communication mediated by gap junctions (11,). Moreover, activation of the Rho signalling pathway in astrocytes prompts cytoskeletal reorganization and promotes the proliferation of adhesion-dependent cells (12,). This process subsequently leads to the induction of reactive astrocytes, ultimately facilitating neuroprotection (13).

Drug development for the ETRs is primarily focused on vasodilatory antagonists (8,9,). Bosentan, the first non-peptide antagonist for ETA and ETB, is clinically used for the treatment of pulmonary arterial hypertension. ETA-selective antagonists are also used as therapeutic agents with fewer side effects. Endothelin-1 primarily acts through the ETA receptor and is implicated in the neoplastic growth of multiple tumour types. Thus, ETR antagonists such as atrasentan and zibotentan have exhibited potential anticancer activities in preclinical studies (9,14,). In addition, the development of ETB agonists is underway, as they provide therapeutic benefits such as vasodilation and neuroprotection (3,13,). IRL1620, the smallest ETB-selective agonist and a truncated analogue of ET-1, selectively and transiently increases tumour blood flow (15,), making it a potential adjuvant cancer therapy for enhancing the delivery of anti-tumour drugs and a treatment for acute ischemic stroke (16,). However, IRL1620 is a linear peptide with exposed N- and C-termini, which pose challenges in terms of pharmacokinetics and drug delivery. Our previous crystallographic studies have elucidated the structure–activity relationships of peptide agonists and small-molecule clinical antagonists of the ETB receptor (1721,). Based on this structural information, researchers have developed new ETR antagonists. Moreover, cryo-electron microscopy structures of the ETB and ETA receptors in complex with G proteins have recently been reported (22,23). This article focuses on the mechanisms by which endothelin transmits signals via ETRs, and antagonists competitively block these signals.

Structural determination

Structural studies of the ETRs commenced with the X-ray crystallographic analyses of the ETB receptor. By innovating methods to crystallize difficult targets (24,), a set of crystal structures of GPCRs was reported in the 2010s. Regarding those, Okuta et al. (25,26,) established a thermostabilized ETB receptor containing five mutations (ETB-Y5) that could be functionally expressed in a cell-free system (26,). Using the modified constructs, we determined the crystal structures of the ETB receptor in complex with various compounds (1721,) (Fig. 1 and Table 1). Recently, our group and others have determined the structures of wild-type ETRs in complex with G proteins (Gi or Gq) by cryo-electron microscopy (22,23,). For the structural determination of the ET-1–ETB–Gi complex, we developed a “fusion G-system” that facilitates the efficient expression and purification of GPCR–G-protein complexes. So far, the structures of 12 different ETRs have been reported (Fig. 1 and Table 1), revealing a series of conformational changes from the inactive to fully activated state (1723).

ETR structures determined to date. The receptors, G proteins and agonist peptides are shown as ribbon representations. The compounds are shown as sticks.
Fig. 1

ETR structures determined to date. The receptors, G proteins and agonist peptides are shown as ribbon representations. The compounds are shown as sticks.

Table 1

List of the endothelin receptor structures determined to date

ReceptorCompoundMethodConstructG proteinPDB IDResolution (Å)Paper
ETBAgonistET-1X-rayETB-Y5-T4L5GLH2.8Shihoya et al., 2016
Cryo-EMETB-WTGi8I2Z2.8Sano et al., 2023
ETB-WTminiGsq8HCX3.5Yujie et al., 2023
ET-3X-rayETB-Y5-T4L6IGK2Shihoya et al., 2018
Srafotoxin S6bETB-Y5-T4L6LRY3Izume et al., 2020
Parital agonistIRL1620ETB-Y5-T4L6IGL2.7Shihoya et al., 2018
Cryo-EMETB-WTGi8HBD3Yujie et al., 2023
ApoX-rayETB-Y5-mT4L5GLI2.5Shihoya et al., 2016
AntagonistBosentanETB-Y4-mT4L5XPR3.6Shihoya et al., 2017
Inverse agonistK-8794ETB-Y5-mT4L5X932.2
IRL2500ETB-Y4-mT4L6K1Q2.7Nagiri et al., 2019
ETAAgonistET-1Cryo-EMETA-WTminiGsq8HCQ3Yujie et al., 2023
ReceptorCompoundMethodConstructG proteinPDB IDResolution (Å)Paper
ETBAgonistET-1X-rayETB-Y5-T4L5GLH2.8Shihoya et al., 2016
Cryo-EMETB-WTGi8I2Z2.8Sano et al., 2023
ETB-WTminiGsq8HCX3.5Yujie et al., 2023
ET-3X-rayETB-Y5-T4L6IGK2Shihoya et al., 2018
Srafotoxin S6bETB-Y5-T4L6LRY3Izume et al., 2020
Parital agonistIRL1620ETB-Y5-T4L6IGL2.7Shihoya et al., 2018
Cryo-EMETB-WTGi8HBD3Yujie et al., 2023
ApoX-rayETB-Y5-mT4L5GLI2.5Shihoya et al., 2016
AntagonistBosentanETB-Y4-mT4L5XPR3.6Shihoya et al., 2017
Inverse agonistK-8794ETB-Y5-mT4L5X932.2
IRL2500ETB-Y4-mT4L6K1Q2.7Nagiri et al., 2019
ETAAgonistET-1Cryo-EMETA-WTminiGsq8HCQ3Yujie et al., 2023
Table 1

List of the endothelin receptor structures determined to date

ReceptorCompoundMethodConstructG proteinPDB IDResolution (Å)Paper
ETBAgonistET-1X-rayETB-Y5-T4L5GLH2.8Shihoya et al., 2016
Cryo-EMETB-WTGi8I2Z2.8Sano et al., 2023
ETB-WTminiGsq8HCX3.5Yujie et al., 2023
ET-3X-rayETB-Y5-T4L6IGK2Shihoya et al., 2018
Srafotoxin S6bETB-Y5-T4L6LRY3Izume et al., 2020
Parital agonistIRL1620ETB-Y5-T4L6IGL2.7Shihoya et al., 2018
Cryo-EMETB-WTGi8HBD3Yujie et al., 2023
ApoX-rayETB-Y5-mT4L5GLI2.5Shihoya et al., 2016
AntagonistBosentanETB-Y4-mT4L5XPR3.6Shihoya et al., 2017
Inverse agonistK-8794ETB-Y5-mT4L5X932.2
IRL2500ETB-Y4-mT4L6K1Q2.7Nagiri et al., 2019
ETAAgonistET-1Cryo-EMETA-WTminiGsq8HCQ3Yujie et al., 2023
ReceptorCompoundMethodConstructG proteinPDB IDResolution (Å)Paper
ETBAgonistET-1X-rayETB-Y5-T4L5GLH2.8Shihoya et al., 2016
Cryo-EMETB-WTGi8I2Z2.8Sano et al., 2023
ETB-WTminiGsq8HCX3.5Yujie et al., 2023
ET-3X-rayETB-Y5-T4L6IGK2Shihoya et al., 2018
Srafotoxin S6bETB-Y5-T4L6LRY3Izume et al., 2020
Parital agonistIRL1620ETB-Y5-T4L6IGL2.7Shihoya et al., 2018
Cryo-EMETB-WTGi8HBD3Yujie et al., 2023
ApoX-rayETB-Y5-mT4L5GLI2.5Shihoya et al., 2016
AntagonistBosentanETB-Y4-mT4L5XPR3.6Shihoya et al., 2017
Inverse agonistK-8794ETB-Y5-mT4L5X932.2
IRL2500ETB-Y4-mT4L6K1Q2.7Nagiri et al., 2019
ETAAgonistET-1Cryo-EMETA-WTminiGsq8HCQ3Yujie et al., 2023

Endothelin binding modes

The ETB receptor adopts the typical GPCR architecture, comprising seven transmembrane (TM) helices and intracellular helix 8 (H8). The extracellular loop (ECL)2 forms long anti-parallel β-strands with a short hairpin, which is a common feature of the peptide receptors (Fig. 2A). In addition to the TM3-ECL2 disulphide bond that is highly conserved among the class A GPCRs, ETB has an additional disulphide bond between C90N-ter and C3587.25 (superscripts indicate Ballesteros–Weinstein numbering (27,)) (Fig. 2B). This disulphide bond links the receptor N-terminus and the extracellular end of TM7. The extracellular portion of the receptor is extensively involved in ET-1 binding, with the N-terminal tail, the three ECLs (ECL1–ECL3) and the six TM helices (TM2–TM7) all contributing to the formation of the orthosteric pocket, which is entirely occupied by ET-1. Furthermore, the N-terminal tail and the ECL2 β-sheet form a lid-like structure that covers the orthosteric pocket (Fig. 2B), resulting in a closed conformation that accounts for the virtually irreversible binding of ET-1 (2830).

Binding modes of endothelin-related peptides. (A) Overall structure of ETB in complex with ET-1, viewed from within the membrane plane. ETB is depicted by ribbons, with the ECL2 β-sheet highlighted. The disulphide bonds at the N-terminus and ECL2 of ETB are shown as sticks. ET-1 is shown as a transparent surface representation and a ribbon model. The side chains of ET-1 are shown as sticks. (B) Surface representation of the ET-1-bound ETB receptor, viewed from the extracellular side. (C) Comparison of the amino acid sequences of the endothelin-related peptides. (D) The architecture of ET-1 is shown in a ribbon representation, with its side chains, N-terminal amino group (N-t) and C-terminal carboxyl group (C-t) shown as sticks. (E–G) Detailed interactions between ET-1 and ETB at the C-terminal region (E), the α-helical region (F) and the N-terminal region (G). Dashed lines indicate hydrogen bonds. (H) Superimposition of the agonist-bound ETR structures, viewed from the extracellular side. (I) Electrostatic surface presentations of the extracellular regions of ET-1-bound ETA and IRL1620-bound ETB. ET-1 in the ET-1-ETA complex structure is omitted for clarity. The electrostatic potential was calculated and presented using CueMol.
Fig. 2

Binding modes of endothelin-related peptides. (A) Overall structure of ETB in complex with ET-1, viewed from within the membrane plane. ETB is depicted by ribbons, with the ECL2 β-sheet highlighted. The disulphide bonds at the N-terminus and ECL2 of ETB are shown as sticks. ET-1 is shown as a transparent surface representation and a ribbon model. The side chains of ET-1 are shown as sticks. (B) Surface representation of the ET-1-bound ETB receptor, viewed from the extracellular side. (C) Comparison of the amino acid sequences of the endothelin-related peptides. (D) The architecture of ET-1 is shown in a ribbon representation, with its side chains, N-terminal amino group (N-t) and C-terminal carboxyl group (C-t) shown as sticks. (E–G) Detailed interactions between ET-1 and ETB at the C-terminal region (E), the α-helical region (F) and the N-terminal region (G). Dashed lines indicate hydrogen bonds. (H) Superimposition of the agonist-bound ETR structures, viewed from the extracellular side. (I) Electrostatic surface presentations of the extracellular regions of ET-1-bound ETA and IRL1620-bound ETB. ET-1 in the ET-1-ETA complex structure is omitted for clarity. The electrostatic potential was calculated and presented using CueMol.

ET-1 adopts a bicyclic structure with two intrachain disulphide bond pairs (C1–C15 and C3–C11) and can be divided into three regions (Fig. 2C, D). In the C-terminal region, residues D18 to W21 adopt extended conformations and penetrate deeply into the receptor core, with the C-terminal W21 side chain directed towards the bottom of the pocket (Fig. 2E). Notably, the C-terminal carboxylate and the D18 side chain form an electrostatic interaction network that involves the charged residues of ETB (K1823.33, K2735.38, R3436.55 and D3687.35). The C-terminal W21 side chain interacts not only with K1823.33 via a π–cation interaction but also directly with W3366.48 in the C6.47W6.48xP6.50 motif, which is the essential motif for the signalling function of class A GPCRs. In the α-helical region, the central residues (D8 to L17) primarily consist of hydrophobic residues and are sandwiched between ECL2 and TM6–7 (Fig. 2F), where they form extensive van der Waals interactions with the receptor. Both ends of the α-helical region form hydrogen bonds with the bulky residues (K1612.64, Y247ECL2, Y350ECL3, R3577.24 and Y3697.36). In contrast, the N-terminal region of ET-1, with the residues C1 to M7, is exposed to the solvent and interacts poorly with the receptor compared with the other regions (Fig. 2G).

The α-helical and C-terminal domains are remarkably conserved among endothelins and related peptides (Fig. 2C), creating the principal elements of receptor interactions. Conversely, the amino acid sequences of the N-terminal region are diverse and exhibit sparse receptor interactions. In the structures of endothelin B receptors bound to endothelin-1, endothelin-3 and sarafotoxin S6b, the agonist peptides exhibit a high degree of superimposition. Moreover, the structure of ETA superimposes well on that of ETB (Fig. 2H), suggesting that the binding modes of endothelins are highly conserved in both subtypes. The ETB-selective agonist IRL1620 lacks the N-terminal region but retains the α-helical and C-terminal regions. Its negative charges, originating from Nα-succinylation and E9, are better suited to the positively charged extracellular region of ETB compared with ETA (Fig. 2I), resulting in preferential ETB binding (23).

Conformational changes upon receptor activation

In the absence of ligands, the TM helices in the ETB receptor are loosely packed against each other, resulting in a large cavity that extends to the receptor core (Fig. 3A). This broad entrance may facilitate the access of large peptide agonists to the orthosteric pocket. Upon ET-1 binding, TM2, TM6 and TM7 move inwards by 2.6, 4.1 and 4.9 Å (Fig. 3A), respectively, whereas TM1 moves outwards by about 4.4 Å, despite having no direct interaction with ET-1. These movements cause the orthosteric pocket to contract and adopt a compact conformation, enabling tight interactions with ET-1. At the intermembrane region, D1472.50 moves downward by about 3 Å and forms a hydrogen bond with N3827.49 (Fig. 3B), squeezing the intermembrane region. Essentially, W21 of ET-1 directly induces the downward rotation of the W3366.48 side chain by 2.5 Å (Fig. 3B), whose nitrogen atom forms a hydrogen bond with N3877.45. The downward motions of W3366.48 and N3877.45 push and induce the outward rotation of F3326.44 in the P5.50I/V3.40F6.44 motif (Fig. 3C, D), resulting in the 6.8-Å outward displacement of the intracellular portion of TM6 (Fig. 4A, B). The degree of opening is smaller than that observed in other Gi-coupled receptors (μOR: 10 Å) (31), reflecting the unique structural features of the G-protein coupling to the ETB receptor.

Structural changes upon endothelin binding. (A) Superimposition of the ET-1-bound and ligand-free structures of the ETB receptor. The extracellular view shows the ET-1-induced movement of the TM helices that constitute the orthosteric pocket. Arrows indicate the movement of helices, with the distances of the residues at each end of the helix. (B) The ET-1-bound and apo structures are superimposed to show the structural differences propagated from the ligand-binding pocket. (C) Packing interactions that stabilize the inactive state are observed between P2855.50, V1893.40, F3326.44 and N3787.45. (D) The inward movement of TM6, including W3366.48, upon agonist binding destabilizes the packing of V1893.40, P2855.50 and F3326.44, resulting in the downward rotation of F3326.44. These changes contribute to the rotation and outward movement of the intracellular portion of TM6 and the inward movement of TM7.
Fig. 3

Structural changes upon endothelin binding. (A) Superimposition of the ET-1-bound and ligand-free structures of the ETB receptor. The extracellular view shows the ET-1-induced movement of the TM helices that constitute the orthosteric pocket. Arrows indicate the movement of helices, with the distances of the residues at each end of the helix. (B) The ET-1-bound and apo structures are superimposed to show the structural differences propagated from the ligand-binding pocket. (C) Packing interactions that stabilize the inactive state are observed between P2855.50, V1893.40, F3326.44 and N3787.45. (D) The inward movement of TM6, including W3366.48, upon agonist binding destabilizes the packing of V1893.40, P2855.50 and F3326.44, resulting in the downward rotation of F3326.44. These changes contribute to the rotation and outward movement of the intracellular portion of TM6 and the inward movement of TM7.

G-protein coupling of ETRs. (A and B) Structural comparison of the apo and Gi-complexed ETB receptor, viewed from the membrane plane (A) and intracellular side (B). (C) Conformational changes on the intracellular side upon Gi coupling. The residues corresponding to the D3.49R3.50Y3.51 and N7.49P7.50xxY7.53 motifs are shown as sticks. The dashed line represents a hydrogen bond. (D) Comparison of the Gα positions in the GPCR–G-protein complexes. The structures are superimposed on the receptor structure of the NTS1 C state. (E) Comparison of the Gα positions in the ETR-G-protein complexes.
Fig. 4

G-protein coupling of ETRs. (A and B) Structural comparison of the apo and Gi-complexed ETB receptor, viewed from the membrane plane (A) and intracellular side (B). (C) Conformational changes on the intracellular side upon Gi coupling. The residues corresponding to the D3.49R3.50Y3.51 and N7.49P7.50xxY7.53 motifs are shown as sticks. The dashed line represents a hydrogen bond. (D) Comparison of the Gα positions in the GPCR–G-protein complexes. The structures are superimposed on the receptor structure of the NTS1 C state. (E) Comparison of the Gα positions in the ETR-G-protein complexes.

In most class A GPCRs, the highly conserved D3.49R3.50Y3.51 and N7.49P7.50xxY7.53 motifs on the intracellular side play an essential role in G-protein coupling (32). Upon ETB activation, the ionic lock between D1983.49 and R1993.50 is broken, and R1993.50 becomes oriented towards the intracellular cavity (Fig. 4C). However, in the ETB receptor, the N7.49P7.50xxY7.53 motif is altered to N7.49P7.50xxL7.53Y7.54, where Y7.53 is replaced by L3867.53. The activation of class A GPCRs induces the inward movement of Y7.53 to form a water-mediated hydrogen bond with the highly conserved Y5.58. In contrast, L3867.53 is a hydrophobic residue and cannot form a polar interaction, leading to the downward, instead of inward, displacement of the intracellular portion of TM7 by 2.8 Å in the ETB–Gi complex (Fig. 4C). Y7.54 is directed towards the membrane plane, and its rotamer does not change upon receptor activation. The substitution of Y7.53 with L3867.53 in ETB affects the movement of TM7 upon receptor activation, thereby distinguishing it from other class A GPCRs.

The atypical downward motion of TM7 results in the distinctive Gi coupling mode of the ETB receptor. L3867.53 forms a hydrophobic contact with G352G.H5.24 (superscript indicates the common Gα numbering system) (33,), and TM7 and H8 extensively interact with the α5-helix, which is absent in other GPCR–Gi complexes. These interactions allow the α5-helix of ETB–Gi to assume the shallowest position relative to the receptor among the Gs-, Gi- and Gq-coupled class A GPCR structures (31,3438) (Fig. 4D). This characteristic is commonly observed in the other Gi- or Gq-complexed structures of ETRs (Fig. 4E). Thus, the amino acid sequence of the α5-helix coupled to the ETRs would be less restricted, accounting for their G-protein promiscuity. This mechanism of G-protein promiscuity may be exclusive to ETRs, as other promiscuous GPCRs retain the N7.49P7.50xxY7.53 motif. Accordingly, this information is critical for understanding the mechanism of G-protein activation by GPCRs.

Antagonist binding modes

The crystal structures of ETB bound to the antagonist bosentan, and the inverse agonists K-8794 and IRL2500 have been determined (18,21,) (Fig. 5A–D). The antagonist-bound structures superimpose well on the apo form (17,) (Fig. 5A), and these compounds are located within the positively charged TM cleft of the receptor. Bosentan and its analogues, as well as K-8794, have many aromatic moieties consisting of a central pyrimidine template with four substituents, including 2-pyrimidyl, 4-sulfonamide, 5-phenol and 6-hydroxyl groups. The sulfonamide moiety of bosentan is specifically recognized by positively charged residues (Fig. 5B), whereas the other moieties occupy the space within the TM binding pocket surrounded by TM2–7, facilitating interactions with the receptor. Previous studies have shown that the sulfonamide substitution in bosentan analogues abolishes binding to ETRs (39,), indicating it is a pharmacophore. Bosentan superimposes well on K-8794 (Fig. 5A), and their binding modes are essentially the same (Fig. 5C). The larger substituent on the 6-position of the central pyrimidine in K-8794 leads to stronger interactions with the receptor, accounting for its higher affinity than bosentan (21,). Moreover, the K-8794-bound structure revealed the detailed water-mediated hydrogen-bonding network around D1472.50, the putative Na+ binding site in class A GPCRs (40). Our structural and functional analyses suggested that, instead of the Na+ ion, the water molecule networks connect TMs 2, 3, 6 and 7 and stabilize the inactive conformation of the receptor.

Structural basis for antagonist binding and receptor inactivation. (A) Superimposition of the ETB structures bound to ET-1, bosentan and K-8794, viewed from the extracellular side. The drugs are shown as sticks, colour-coded as in Figure 1. (B–D) Detailed interactions of bosentan (B), K-8794 (C) and IRL2500 (D) with the receptor, viewed from the extracellular side. Black dashed lines indicate hydrogen bonds. (E) The structural changes upon compound binding, focused on TM6. Black arrows indicate the inward movements of TM6, TM7 and W3366.48 upon ET-1 binding.
Fig. 5

Structural basis for antagonist binding and receptor inactivation. (A) Superimposition of the ETB structures bound to ET-1, bosentan and K-8794, viewed from the extracellular side. The drugs are shown as sticks, colour-coded as in Figure 1. (B–D) Detailed interactions of bosentan (B), K-8794 (C) and IRL2500 (D) with the receptor, viewed from the extracellular side. Black dashed lines indicate hydrogen bonds. (E) The structural changes upon compound binding, focused on TM6. Black arrows indicate the inward movements of TM6, TM7 and W3366.48 upon ET-1 binding.

Schematic representation of ETR activation. TM6, TM7 and H8 are highlighted. The residues involved in signal transduction (N1.50, D2.50, F6.44, W6.48 and N7.45) are represented with sticks. Black dashed lines indicate hydrogen bonds. Black arrows indicate the conformational changes of the receptor, and black double arrows indicate the conformational equilibrium. Black dashed double arrows mean fewer structural transitions, resulting in the basal activity or partial agonistic activity.
Fig. 6

Schematic representation of ETR activation. TM6, TM7 and H8 are highlighted. The residues involved in signal transduction (N1.50, D2.50, F6.44, W6.48 and N7.45) are represented with sticks. Black dashed lines indicate hydrogen bonds. Black arrows indicate the conformational changes of the receptor, and black double arrows indicate the conformational equilibrium. Black dashed double arrows mean fewer structural transitions, resulting in the basal activity or partial agonistic activity.

Unexpectedly, bosentan adopts a similar binding mode to the agonists, even though it was developed without mimicking endogenous agonist peptides (21). The ETB receptor similarly interacts with both bosentan and the C-terminal tripeptide of ET-1 (Fig. 5A), suggesting the importance of ionic and hydrophobic interactions within the TM core for antagonist and agonist binding. However, in contrast to the ET-1-induced 4-Å inward movements of both TM6 and TM7 (Fig. 5E), bosentan induces only a minor inward movement of TM6 and no movement of TM7. The 2-methoxyphenoxy group of bosentan sterically prevents the inward motion of W3366.48, which is critical for receptor activation, and thus bosentan functions as an antagonist.

IRL2500 comprises a tryptophan, a biphenyl group and a 3,5-dimethylbenzoyl group. The biphenyl group forms a peptide bond with the tryptophan and a peptoid bond with the dimethylbenzoyl group. The carboxylate group of the tryptophan moiety in IRL2500 forms a salt bridge with R3436.55, whereas the other aromatic moieties occupy the space within the TM binding pocket (Fig. 5D). The binding mode of IRL2500 is similar to that of bosentan. Notably, the biphenyl group penetrates deeply into the receptor core proximal to D1472.50. Thus, the dimethylbenzoyl and biphenyl groups of IRL2500 sandwich the W3366.48 side chain, securely preventing its inward rotation (Fig. 5E). These observations suggest that IRL2500 rigidly prevents the transition to the active state, compared with bosentan, and functions as an inverse agonist that reduces basal activity, as shown by pharmacological studies using the constitutive ETB mutant (18).

The residues constituting the antagonist binding site, including the positively charged residues that coordinate the sulfonamide in the current structures, are highly conserved between the ETA and ETB receptors, suggesting that bosentan and its analogues have common binding modes for both ETRs. Furthermore, other ETR antagonists that are unlike bosentan, including the ETA-selective antagonists ambrisentan (41,) and atrasentan (42), also possess a negatively charged group, such as a sulfonamide or a carboxylate, surrounded by two or three hydrophobic (aromatic or acyl) groups. The negatively charged group could occupy the centre of the TM pocket and form ionic interactions with the positively charged residues of the ETRs, whereas the other hydrophobic groups might fit within the pocket and facilitate these interactions, as observed in the current ETB structures.

Future Perspectives

Over the past decade, significant advances in the structural studies of ETRs have elucidated their fundamental compositions, mechanisms of activation and diverse drug binding modes (Fig. 6). This structural information has led to the development of several ETR drugs (43,44,). Yet, two major challenges remain in the structure-based drug discovery targeting ETRs. The first is the rational design of subtype-selective ETR antagonists. The ETA selectivity is purportedly crucial for effectiveness against pulmonary arterial hypertension and cancer (9,14,4548,). However, the mechanism underlying ETA selectivity remains enigmatic because of the high conservation of the residues constituting the binding pockets in both ETB and ETA(21,). In 2023, the first agonist-bound ETA structure was reported (23,). Thus, the inactive ETA structure will be determined in the future and significantly clarify the ETA selectivities of druggable antagonists. The second challenge is the creation of small-molecule agonists for ETRs. Because ETA activation leads to potent vasoconstriction, ETB activation holds therapeutic potential owing to its vasodilatory effects (9,15,16,). The linear peptide IRL1620 is an ETB-selective agonist, but small-molecule agonists have not been discovered. As evidenced by the structures, ETB activation by endothelin-related peptides entails substantial structural changes, including the 4-Å inward displacements of TMs 6 and 7 on the extracellular side (17). It would be difficult for small-molecule drugs to induce these specific structural changes precisely. Our structural investigations propose that the binding and movement of W3366.48 and R3436.55 with the C-terminal region of ET-1 are of utmost importance for receptor activation. Consequently, drugs that emulate this C-terminal region may offer potential as small-molecule ETR agonists.

Funding

This work was supported by grants from the Platform for Drug Discovery, Informatics and Structural Life Science by the Ministry of Education, Culture, Sports, Science and Technology (MEXT), and JSPS KAKENHI grants 21H05037 (O.N.), 22K19371 and 22H02751 (W.S.); ONO Medical Research Foundation (W.S.); The Kao Foundation for Arts and Sciences (W.S.); The Takeda Science Foundation (W.S.); The Uehara Memorial Foundation (W.S.); and the Platform Project for Supporting Drug Discovery and Life Science Research (Basis for Supporting Innovative Drug Discovery and Life Science Research (BINDS)) from AMED, under grant number JP23ama121012.

Conflict of interest

None declared.

References

1.

Yanagisawa
,
M.
,
Kurihara
,
H.
,
Kimura
,
S.
,
Tomobe
,
Y.
,
Kobayashi
,
M.
,
Mitsui
,
Y.
,
Yazaki
,
Y.
,
Goto
,
K.
, and
Masaki
,
T.
(
1988
)
A novel potent vasoconstrictor peptide produced by vascular endothelial cells
.
Nature
332
,
411
415

2.

Maguire
,
J.J.
and
Davenport
,
A.P.
(
2014
)
Endothelin@25—new agonists, antagonists, inhibitors and emerging research frontiers: IUPHAR Review 12
.
Br. J. Pharmacol.
171
,
5555
5572

3.

Davenport
,
A.P.
,
Hyndman
,
K.A.
,
Dhaun
,
N.
,
Southan
,
C.
,
Kohan
,
D.E.
,
Pollock
,
J.S.
,
Pollock
,
D.M.
,
Webb
,
D.J.
, and
Maguire
,
J.J.
(
2016
)
Endothelin
.
Pharmacol. Rev.
68
,
357
418

4.

Arai
,
H.
,
Hori
,
S.
,
Aramori
,
I.
,
Ohkubo
,
H.
, and
Nakanishi
,
S.
(
1990
)
Cloning and expression of a cDNA encoding an endothelin receptor
.
Nature
348
,
730
732

5.

Sakurai
,
T.
,
Yanagisawa
,
M.
,
Takuwat
,
Y.
,
Miyazakit
,
H.
,
Kimura
,
S.
,
Goto
,
K.
, and
Masaki
,
T.
(
1990
)
Cloning of a cDNA encoding a non-isopeptide-selective subtype of the endothelin receptor
.
Nature
348
,
732
735

6.

Inoue
,
A.
,
Raimondi
,
F.
,
Kadji
,
F.M.N.
,
Singh
,
G.
,
Kishi
,
T.
,
Uwamizu
,
A.
,
Ono
,
Y.
,
Shinjo
,
Y.
,
Ishida
,
S.
,
Arang
,
N.
,
Kawakami
,
K.
,
Gutkind
,
J.S.
,
Aoki
,
J.
, and
Russell
,
R.B.
(
2019
)
Illuminating G-protein-coupling selectivity of GPCRs
.
Cell
177
,
1933
1947.e25

7.

Doi
,
T.
,
Sugimoto
,
H.
,
Arimoto
,
I.
,
Hiroaki
,
Y.
, and
Fujiyoshi
,
Y.
(
1999
)
Interactions of endothelin receptor subtypes A and B with Gi, Go, and Gq in reconstituted phospholipid vesicles
.
Biochemistry
38
,
3090
3099

8.

Haryono
,
A.
,
Ramadhiani
,
R.
,
Ryanto
,
G.R.T.
, and
Emoto
,
N.
(
2022
)
Endothelin and the cardiovascular system: the long journey and where we are going
.
Biology
11
,
759

9.

Barton
,
M.
and
Yanagisawa
,
M.
(
2019
)
Endothelin: 30 years from discovery to therapy
.
Hypertension
74
,
1232
1265

10.

Bremnes
,
T.
,
Paasche
,
J.D.
,
Mehlum
,
A.
,
Sandberg
,
C.
,
Bremnes
,
B.
, and
Attramadal
,
H.
(
2000
)
Regulation and intracellular trafficking pathways of the endothelin receptors
.
J. Biol. Chem.
275
,
17596
17604

11.

Tencé
,
M.
,
Ezan
,
P.
,
Amigou
,
E.
, and
Giaume
,
C.
(
2012
)
Increased interaction of connexin43 with zonula occludens-1 during inhibition of gap junctions by G protein-coupled receptor agonists
.
Cell. Signal.
24
,
86
98

12.

Koyama
,
Y.
and
Baba
,
A.
(
1996
)
Endothelin-induced cytoskeletal actin re-organization in cultured astrocytes: inhibition by C3 ADP-ribosyltransferase
.
Glia
16
,
342
350

13.

Koyama
,
Y.
(
2021
)
Endothelin ETB receptor-mediated astrocytic activation: pathological roles in brain disorders
.
Int. J. Mol. Sci.
22
,
4333

14.

Rosanò
,
L.
,
Spinella
,
F.
, and
Bagnato
,
A.
(
2013
)
Endothelin 1 in cancer: biological implications and therapeutic opportunities
.
Nat. Rev. Cancer
13
,
637
651

15.

Gulati
,
A.
,
Sunila
,
E.S.
, and
Kuttan
,
G.
(
2012
)
IRL-1620, an endothelin-B receptor agonist, enhanced radiation induced reduction in tumor volume in Dalton’s lymphoma ascites tumor model
.
Arzneimittelforschung
62
,
14
17

16.

Ranjan
,
A.K.
and
Gulati
,
A.
(
2022
)
Sovateltide mediated endothelin B receptors agonism and curbing neurological disorders
.
Int. J. Mol. Sci.
23
,
3146

17.

Shihoya
,
W.
,
Nishizawa
,
T.
,
Okuta
,
A.
,
Tani
,
K.
,
Dohmae
,
N.
,
Fujiyoshi
,
Y.
,
Nureki
,
O.
, and
Doi
,
T.
(
2016
)
Activation mechanism of endothelin ETB receptor by endothelin-1
.
Nature
537
,
363
368

18.

Nagiri
,
C.
,
Shihoya
,
W.
,
Inoue
,
A.
,
Kadji
,
F.M.N.
,
Aoki
,
J.
, and
Nureki
,
O.
(
2019
)
Crystal structure of human endothelin ETB receptor in complex with peptide inverse agonist IRL2500
.
Commun. Biol.
2
,
236

19.

Izume
,
T.
,
Miyauchi
,
H.
,
Shihoya
,
W.
, and
Nureki
,
O.
(
2020
)
Crystal structure of human endothelin ETB receptor in complex with sarafotoxin S6b
.
Biochem. Biophys. Res. Commun.
528
,
383
388

20.

Shihoya
,
W.
,
Izume
,
T.
,
Inoue
,
A.
,
Yamashita
,
K.
,
Kadji
,
F.M.N.
,
Hirata
,
K.
,
Aoki
,
J.
,
Nishizawa
,
T.
, and
Nureki
,
O.
(
2018
)
Crystal structures of human ETB receptor provide mechanistic insight into receptor activation and partial activation
.
Nat. Commun.
9
,
4711

21.

Shihoya
,
W.
,
Nishizawa
,
T.
,
Yamashita
,
K.
,
Inoue
,
A.
,
Hirata
,
K.
,
Kadji
,
F.M.N.
,
Okuta
,
A.
,
Tani
,
K.
,
Aoki
,
J.
,
Fujiyoshi
,
Y.
,
Doi
,
T.
, and
Nureki
,
O.
(
2017
)
X-ray structures of endothelin ETB receptor bound to clinical antagonist bosentan and its analog
.
Nat. Struct. Mol. Biol.
24
,
758
764

22.

Sano
,
F.K.
,
Akasaka
,
H.
,
Shihoya
,
W.
, and
Nureki
,
O.
(
2023
)
Cryo-EM structure of the endothelin-1–ETB–Gi complex
.
Elife
12
,
706
731

23.

Ji
,
Y.
,
Duan
,
J.
,
Yuan
,
Q.
,
He
,
X.
,
Yang
,
G.
,
Zhu
,
S.
,
Wu
,
K.
,
Hu
,
W.
,
Gao
,
T.
,
Cheng
,
X.
,
Jiang
,
H.
,
Eric Xu
,
H.
, and
Jiang
,
Y.
(
2023
)
Structural basis of peptide recognition and activation of endothelin receptors
.
Nat. Commun.
14
,
1268

24.

Caffrey
,
M.
and
Cherezov
,
V.
(
2009
)
Crystallizing membrane proteins using lipidic mesophases
.
Nat. Protoc.
4
,
706
731

25.

Okuta
,
A.
,
Tani
,
K.
,
Nishimura
,
S.
,
Fujiyoshi
,
Y.
, and
Doi
,
T.
(
2016
)
Thermostabilization of the human endothelin type B receptor
.
J. Mol. Biol.
428
,
2265
2274

26.

Nakai
,
H.
,
Isshiki
,
K.
,
Hattori
,
M.
,
Maehira
,
H.
,
Yamaguchi
,
T.
,
Masuda
,
K.
,
Shimizu
,
Y.
,
Watanabe
,
T.
,
Hohsaka
,
T.
,
Shihoya
,
W.
,
Nureki
,
O.
,
Kato
,
Y.
,
Watanabe
,
H.
, and
Matsuura
,
T.
(
2022
)
Cell-free synthesis of human endothelin receptors and its application to ribosome display
.
Anal. Chem.
94
,
3831
3839

27.

Ballesteros
,
J.A.
and
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 Neurosci
.
25
,
366
428

28.

Mey
,
J.
,
Compeer
,
M.G.
, and
Meens
,
M.
(
2009
)
Endothelin-1, an endogenous irreversible agonist in search of an allosteric inhibitor
.
Mol. Cell. Pharmacol.
1
,
246
257

29.

Takasuka
,
T.
,
Sakurai
,
T.
,
Goto
,
K.
,
Furuichi
,
Y.
, and
Watanabe
,
T.
(
1994
)
Human endothelin receptor ETB. Amino acid sequence requirements for super stable complex formation with its ligand
.
J. Biol. Chem.
269
,
7509
7513

30.

Hilal-Dandan
,
R.
,
Villegas
,
S.
,
Gonzalez
,
A.
, and
Brunton
,
L.L.
(
1997
)
The quasi-irreversible nature of endothelin binding and G protein-linked signaling in cardiac myocytes
.
J. Pharmacol. Exp. Ther.
281
,
267
273

31.

Zhuang
,
Y.
,
Wang
,
Y.
,
He
,
B.
,
He
,
X.
,
Zhou
,
X.E.
,
Guo
,
S.
,
Rao
,
Q.
,
Yang
,
J.
,
Liu
,
J.
,
Zhou
,
Q.
,
Wang
,
X.
,
Liu
,
M.
,
Liu
,
W.
,
Jiang
,
X.
,
Yang
,
D.
,
Jiang
,
H.
,
Shen
,
J.
,
Melcher
,
K.
,
Chen
,
H.
,
Jiang
,
Y.
,
Cheng
,
X.
,
Wang
,
M.W.
,
Xie
,
X.
, and
Xu
,
H.E.
(
2022
)
Molecular recognition of morphine and fentanyl by the human μ-opioid receptor
.
Cell
185
,
4361
4375.e19

32.

Venkatakrishnan
,
A.J.
,
Deupi
,
X.
,
Lebon
,
G.
,
Tate
,
C.G.
,
Schertler
,
G.F.
, and
Babu
,
M.M.
(
2013
)
Molecular signatures of G-protein-coupled receptors
.
Nature
494
,
185
194

33.

Flock
,
T.
,
Ravarani
,
C.N.J.
,
Sun
,
D.
,
Venkatakrishnan
,
A.J.
,
Kayikci
,
M.
,
Tate
,
C.G.
,
Veprintsev
,
D.B.
, and
Babu
,
M.M.
(
2015
)
Universal allosteric mechanism for Gα activation by GPCRs
.
Nature
524
,
173
179

34.

Kim
,
K.
,
Che
,
T.
,
Panova
,
O.
,
DiBerto
,
J.F.
,
Lyu
,
J.
,
Krumm
,
B.E.
,
Wacker
,
D.
,
Robertson
,
M.J.
,
Seven
,
A.B.
,
Nichols
,
D.E.
,
Shoichet
,
B.K.
,
Skiniotis
,
G.
, and
Roth
,
B.L.
(
2020
)
Structure of a hallucinogen-activated Gq-coupled 5-HT2A serotonin receptor
.
Cell
182
,
1574
1588.e19

35.

Yuan
,
Y.
,
Jia
,
G.
,
Wu
,
C.
,
Wang
,
W.
,
Cheng
,
L.
,
Li
,
Q.
,
Li
,
Z.
,
Luo
,
K.
,
Yang
,
S.
,
Yan
,
W.
,
Su
,
Z.
, and
Shao
,
Z.
(
2021
)
Structures of signaling complexes of lipid receptors S1PR1 and S1PR5 reveal mechanisms of activation and drug recognition
.
Cell Res.
31
,
1263
1274

36.

Hua
,
T.
,
Li
,
X.
,
Wu
,
L.
,
Iliopoulos-Tsoutsouvas
,
C.
,
Wang
,
Y.
,
Wu
,
M.
,
Shen
,
L.
,
Brust
,
C.A.
,
Nikas
,
S.P.
,
Song
,
F.
,
Song
,
X.
,
Yuan
,
S.
,
Sun
,
Q.
,
Wu
,
Y.
,
Jiang
,
S.
,
Grim
,
T.W.
,
Benchama
,
O.
,
Stahl
,
E.L.
,
Zvonok
,
N.
,
Zhao
,
S.
,
Bohn
,
L.M.
,
Makriyannis
,
A.
, and
Liu
,
Z.J.
(
2020
)
Activation and signaling mechanism revealed by cannabinoid receptor–Gi complex structures
.
Cell
180
,
655
665.e18

37.

Rasmussen
,
S.G.F.
,
DeVree
,
B.T.
,
Zou
,
Y.
,
Kruse
,
A.C.
,
Chung
,
K.Y.
,
Kobilka
,
T.S.
,
Thian
,
F.S.
,
Chae
,
P.S.
,
Pardon
,
E.
,
Calinski
,
D.
,
Mathiesen
,
J.M.
,
Shah
,
S.T.A.
,
Lyons
,
J.A.
,
Caffrey
,
M.
,
Gellman
,
S.H.
,
Steyaert
,
J.
,
Skiniotis
,
G.
,
Weis
,
W.I.
,
Sunahara
,
R.K.
, and
Kobilka
,
B.K.
(
2011
)
Crystal structure of the β2 adrenergic receptor-Gs protein complex
.
Nature
477
,
549
555

38.

Kato
,
H.E.
,
Zhang
,
Y.
,
Hu
,
H.
,
Suomivuori
,
C.M.
,
Kadji
,
F.M.N.
,
Aoki
,
J.
,
Krishna Kumar
,
K.
,
Fonseca
,
R.
,
Hilger
,
D.
,
Huang
,
W.
,
Latorraca
,
N.R.
,
Inoue
,
A.
,
Dror
,
R.O.
,
Kobilka
,
B.K.
, and
Skiniotis
,
G.
(
2019
)
Conformational transitions of a neurotensin receptor 1–Gi1 complex
.
Nature
572
,
80
85

39.

Neidhart
,
W.
,
Breu
,
V.
,
Bur
,
D.
,
Burri
,
K.
,
Clozel
,
M.
,
Hirth
,
G.
,
Müller
,
M.
,
Wessel
,
H.P.
, and
Ramuz
,
H.
(
1996
)
The discovery of nonpeptide endothelin receptor antagonists. Progression towards Bosentan
.
Chimia
50
,
519
519

40.

Katritch
,
V.
,
Fenalti
,
G.
,
Abola
,
E.E.
,
Roth
,
B.L.
,
Cherezov
,
V.
, and
Stevens
,
R.C.
(
2014
)
Allosteric sodium in class a GPCR signaling
.
Trends Biochem. Sci.
39
,
233
244

41.

Vatter
,
H.
and
Seifert
,
V.
(
2006
)
Ambrisentan, a non-peptide endothelin receptor antagonist
.
Cardiovasc. Drug Rev.
24
,
63
76

42.

Winn
,
M.
,
von Geldern
,
T.W.
,
Opgenorth
,
T.J.
,
Jae
,
H.S.
,
Tasker
,
A.S.
,
Boyd
,
S.A.
,
Kester
,
J.A.
,
Mantei
,
R.A.
,
Bal
,
R.
,
Sorensen
,
B.K.
,
Wu-Wong
,
J.R.
,
Chiou
,
W.J.
,
Dixon
,
D.B.
,
Novosad
,
E.I.
,
Hernandez
,
L.
, and
Marsh
,
K.C.
(
1996
)
2,4-Diarylpyrrolidine-3-carboxylic Acids—potent ETA selective endothelin receptor antagonists. 1. Discovery of A-127722
.
J. Med. Chem.
39
,
1039
1048

43.

Gu
,
X.
,
Wang
,
Y.
,
Wang
,
H.
,
Wu
,
H.
,
Li
,
W.
,
Wang
,
J.
, and
Li
,
N.
(
2021
)
Homology modeling, molecular dynamics and virtual screening of endothelin-a receptor for the treatment of pulmonary arterial hypertension
.
J. Biomol. Struct. Dyn.
39
,
3912
3923

44.

Intagliata
,
S.
,
Helal
,
M.A.
,
Materia
,
L.
,
Pittalà
,
V.
,
Salerno
,
L.
,
Marrazzo
,
A.
,
Cagnotto
,
A.
,
Salmona
,
M.
,
Modica
,
M.N.
, and
Romeo
,
G.
(
2020
)
Synthesis and molecular modelling studies of new 1,3-diaryl-5-oxo-proline derivatives as endothelin receptor ligands
.
Molecules
25
,
1851

45.

Kohan
,
D.E.
,
Cleland
,
J.G.
,
Rubin
,
L.J.
,
Theodorescu
,
D.
, and
Barton
,
M.
(
2012
)
Clinical trials with endothelin receptor antagonists: what went wrong and where can we improve?
Life Sci.
91
,
528
539

46.

Boss
,
C.
,
Bolli
,
M.H.
, and
Gatfield
,
J.
(
2016
)
From bosentan (Tracleer®) to macitentan (Opsumit®): the medicinal chemistry perspective
.
Bioorg. Med. Chem. Lett.
26
,
3381
3394

47.

Newman
,
J.H.
,
Kar
,
S.
, and
Kirkpatrick
,
P.
(
2007
)
Ambrisentan
.
Nat. Rev. Drug Discov.
6
,
697
698

48.

Heerspink
,
H.J.L.
,
SONAR Committees and Investigators
. (
2019
)
Atrasentan and renal events in patients with type 2 diabetes and chronic kidney disease (SONAR): a double-blind, randomised, placebo-controlled trial
.
Lancet
393
,
1937
1947

Author notes

Osamu Nureki, Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo 113-0033, Japan. Tel.: +81-48-467-9547, Fax: +81-48-462-4679, email: [email protected]

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact [email protected]