Abstract
Apelin is an endogenous peptide that is involved in many diseases such as cardiovascular diseases, obesity, and cancer, which has made it an attractive target for drug discovery. Herein, we explore the penultimate and final sequence positions of [Pyr1]-apelin-13 (Ape13) via C-terminal Nα-alkylated amide bonds and the introduction of positive charges, potentially targeting the allosteric sodium pocket, by assessing the binding affinity and signaling profiles at the apelin receptor (APJ). Synthetic analogues modified within this segment of Ape13 showed high affinity (Ki 0.12–0.17 nM vs Ape13 Ki 0.7 nM), potent Gαi1 activation (EC50 Gαi1 0.4–0.9 nM vs Ape13 EC50 1.1 nM), partial agonist behavior disfavoring β-arrestin 2 recruitment for positively charged ligands (e.g., 49 (SBL-AP-058), EC50 β-arr2 275 nM, Emax 54%) and high plasma stability for N-alkyl ligands (t1/2 > 7 h vs Ape13 t1/2 0.5 h). Combining the benefits of the Nα-alkylated amide bond with the guanidino substitution in a constrained ligand led to 63 (SBL-AP-049), which displayed increased plasma stability (t1/2 5.3 h) and strong reduction of β-arrestin 2 signaling with partial maximal efficacy (EC50 β-arr 864 nM, Emax 48%), significantly reducing the hypotensive effect in vivo.
Keywords: apelin, APJ receptor, side-chain-constrained amino acids, GPCR, biased signaling, ACE2, cardiovascular effects
The apelin receptor (APJ) was discovered in 1993 and deorphanized in 1998 by Tatemoto and co-workers when they isolated its first endogenous ligand, the apelin peptide.1,2 The second endogenous ligand, ELABELA, was reported 15 years later by two independent groups while studying the implications of peptides in the embryogenesis process.3,4 However, these two endogenous ligands have distinct signaling profiles and recent evidence also suggests that they have different mechanisms of action, such as in the cardiovascular system.5,6 Of the two ligands, apelin is studied more extensively for historical reasons, but also because it is widely distributed in many tissue types and is involved in multiple physiological processes.7 Several isoforms of the apelin peptide exist after post-translational processing of prepro-apelin (77-aa long) into apelin-36, apelin-17, apelin-13, apelin-12, and [Pyr1]-apelin-13 (I, Figure 1). The latter is the predominant isoform in human plasma since the N-terminal glutamine cyclizes to pyroglutamic acid (Pyr).8−11 [Pyr1]-apelin-13 (Ape13) is a full agonist and has been extensively studied in diabetes,12−14 obesity,12 cancer,15−18 and cardiovascular diseases.19−23 Recent reviews focusing on biological applications and signaling are available,24−27 but it is noteworthy that several compounds targeting the apelin receptor were clinically tested in phase I or II to treat cardiovascular complications, such as heart failure.28−30 The apelin-13 peptide has a very short in vivo half-life (<5 min) and is known to be degraded at the C-terminus between Pro12-Phe13 by the metalloproteases angiotensin converting enzyme II (ACE2) and proline carboxy peptidase (PRCP) (Figure 1).31,32 More recently, neprilysin (NEP) has been identified, which has Arg4-Leu5 and Leu5-Ser6 as cleavage sites (Figure 1).33,34
Figure 1.

Linear and macrocyclic peptide-based apelin analogues in literature. NEP: neprilysin; ACE2: angiotensin-converting enzyme 2; PRCP: prolyl carboxypeptidase. IV–VI are modifications of [Pyr1]-apelin-13 at positions 12 and 13, and these analogues have Nle instead of Met11 to avoid oxidation of the Met side chain.
As a member of class A GPCRs, the apelin receptor triggers the G protein and β-arrestin signaling pathways upon activation.35 Two of the G proteins involved are Gαi, which inhibits adenylate cyclase and reduces cyclic AMP production,36 and Gα12, which leads to RhoGEF recruitment and ROCK activation.37,38 β-arrestin is also recruited to the cell membrane after receptor activation.39 This protein is mainly associated with receptor desensitization and endocytosis, but also activates other downstream signaling pathways and effectors, such as ERK1/2 and Raf.39 Altogether, the above signaling pathways trigger physiological responses such as vasodilation, cardiac contraction, and angiogenesis.35,40,41
Related to the present work, the literature has demonstrated that apelin administration is beneficial in heart failure models and has cardioprotective properties. However, the activation of the β-arrestin signaling pathway seems to cause cardiomyocyte hypertrophy.6 Therefore, APJ ligands that favor G protein activation and block β-arrestin signaling appear promising for patients with heart failure and other cardiovascular diseases. Interestingly, Li and co-workers recently demonstrated that APJ transgenic mutant mice (APJ I107A), which are unable to recruit β-arrestins after receptor activation do not seem to exhibit impairment of cardiac function after exercise or in response to pressure overload-induced cardiac hypertrophy.42 Altogether, these results suggest that intracellular signaling pathways may be subtly altered under pathological conditions, thus emphasizing the need for a comprehensive analysis of ligands with unique or unbalanced signaling profiles.43
Although many advances have been made in apelin research, it remains unclear why certain modifications of [Pyr1]-apelin-13 allow the triggering of different signaling profiles following APJ activation, a phenomenon called functional selectivity or biased agonism. Biased agonism is now accepted as representing a handle to prevent receptor desensitization, due to β-arrestin recruitment, and thus prolong the drug’s therapeutic effect.44,45 Brame et al. reported a macrocyclic analogue of apelin-13 (II, Figure 1) which showed biased G protein activation over β-arrestin recruitment.46 More recently, we have also reported the design and synthesis of novel macrocyclic analogues of apelin-13.47 These structures (e.g., III, Figure 1), containing a C-terminal macrocycle, showed potent bias toward G protein activation.47−49 Additionally, we showed that conformationally constrained or unnatural amino acids (e.g., IV, V, VI, Figure 1) at the 12th (Pro) or 13th (Phe) position had beneficial effects, both in terms of affinity and stability, compared to the apelin-13 peptide.50,51 Unfortunately, no biased ligand was obtained in these latter efforts. Recently, Vederas’ group has worked on modifications of the NEP cleavage site. They obtained agonists with good affinity and stability, but without studying the potential for signaling bias.52 In addition, many efforts have been made in recent years to develop small molecule agonists of the APJ receptor.28,53−60 Nevertheless, the development of peptide analogues allows an easy and rapid approach for structure–activity relationship (SAR) studies and functional selectivity, which is still poorly understood.
Only a handful of publications exist describing the rational approach to design a biased GPCR ligand. Among them, Roth and co-workers postulated that modulating the interaction of ligands with the transmembrane domain 5 (TM5) and extracellular loop 2 (ECL2) of the GPCR could impact signaling, as successfully demonstrated by the development of β-arrestin biased agonists of the dopamine D2 receptor.61 The interaction of orthosteric ligands with ECL2 has also been used to explain the bias for other GPCRs, in casu the serotonin receptor 5-HT2R.62 Although the X-ray structure of an apelin analogue in complex with a mutant form of the receptor has been solved and critical interactions of Ape13 binding to APJ have been suggested for agonist activity, it remains speculative that analogous interactions could be applied to produce a similar result.63 The C-terminus of Ape13 occupies the binding site between the TM1, TM2, TM6, and TM7 of the receptor and its SAR has been extensively studied in the past, in which certain modifications, especially on Phe13, resulted in high affinity and biased ligands.51,64 In contrast, modifications at the Nα-amide bond between residues Pro12-Phe13 have been scarcely investigated, although they may help to establish new interactions and signaling profiles, and potentially increase proteolytic stability.50,65
Herein, we describe the synthesis of a series of [Pyr1]-apelin-13 analogues in which the amide bond at the native Pro12-Phe13 peptide bond was substituted with different alkyl groups. The Nle11-1-Nal12-Phe13 segment (V, Figure 1) was taken as a starting point given its recently reported increased affinity and signaling potency for Gα12.50 A wide variety of substituents varying in length, nature (aliphatic vs aromatic), polarity, and steric bulk was applied. These substituents induce conformational shifts by steric interactions, potentially influencing any eventual signaling. On the other hand, unnatural amino acids bearing a positive charge were also designed and introduced into the C-terminus of [Pyr1]-apelin-13 to test their ability to modulate the signaling profile. The insertion of a positive charge was inspired by the work of the Yokoyama group, which demonstrated an inverse agonist mechanism on the leukotriene receptor BLT1 (class A GPCR) by targeting the allosteric sodium ion binding site with positively charged analogues.66 Other studies have also shown the potential of the sodium pocket for a pronounced signaling bias for class A GPCRs, including the angiotensin 1 receptor (AT1R) which has the highest homology to APJ.1,67−69 Several class A GPCRs share the same amino acid sequence for the allosteric sodium pocket, and the APJ receptor holds this sequence, suggesting the presence of this pocket.67
In contrast to the previously reported macrocyclic analogues, the ligands in this study only encompass minimal backbone modifications compared to the endogenous ligand and are intended to provide additional insights into the SAR of APJ and apelin. The affinity of the modified peptides for APJ was measured by a competitive binding assay against [125I][Nle75,Tyr77]Pyr-apelin-13, and their ability to activate the canonical signaling pathways Gαi1 and β-arrestin 2 was evaluated using bioluminescence resonance energy transfer (BRET)-based biosensors in HEK293 cells overexpressing APJ. Proteolytic stability was assessed for all analogues in rat plasma. Finally, synthetic analogues that showed beneficial pharmacological properties in vitro were selected for continuous in vivo blood pressure measurements.
Results and Discussion
Recently published results by our group have shown that insertion of sterically or covalently constrained unnatural amino acids produces metabolically stable [Pyr1]-apelin-13 analogues, but without bias in signaling.50,51 Hence, we further studied C-terminal modifications by adding alkyl groups ranging from a methyl to bulky benzyl ethers in the 1-Nal12-Phe13 dipeptide segment. The introduction of the additional substituent further inhibits protease access and is expected to increase hydrophobic interactions with the receptor. To avoid difficult resin couplings and to ensure easy peptide purifications, all Nα-alkylated dipeptides and phenylalanine derivatives were synthesized in solution prior to assembly by solid-phase peptide synthesis (SPPS).
Synthesis of N-Substituted Dipeptides
C-Terminal dipeptides of type 3 were synthesized by first monoalkylating the Nα of phenylalanine methyl ester 1 according to three different procedures from the literature (Scheme 1).70,71 For aromatic aldehydes (Conditions A, Scheme 1), phenylalanine methyl ester 1 was first dissolved in dry DCM under an inert atmosphere, to which aldehyde was added. After imine formation, the hydride donor NaBH4 was added, which allowed complete reduction to the desired secondary amine. For aliphatic aldehydes, a one-step reductive amination gave the best results. Here, NaBH(OAc)3 was added to a solution of phenylalanine and aldehyde (Conditions B, Scheme 1). Upon insertion of the 4-nitrobenzyl substituent, a third route was applied, in which mono-Nα alkylation was achieved using 4-nitrobenzyl bromide and Cs2CO3 in N,N-dimethylformamide (DMF) (Conditions C, Scheme 1). This alternative route was followed due to the observed epimerization of Phe when performing reductive amination conditions with 4-nitrobenzaldehyde on phenylalanine. All final products were concentrated in vacuo after extraction and precipitated in a solution of 2 N HCl in Et2O to afford the white HCl salt. Next, to prevent prolonged or repeated coupling using expensive coupling reagent mixtures, an acyl chloride-based coupling was used. Commercially available Fmoc-1-Nal-OH was treated with SOCl2 in dry DLM at room temperature. Full conversion was achieved after overnight reaction. This procedure enabled the synthesis of the targeted dipeptides (Conditions D, Scheme 1). However, in selected cases, the HATU reagent was sufficiently reactive to allow coupling between Fmoc-1-Nal-OH and alkylated phenylalanine bearing an aliphatic chain such as methyl, ethyl, n-propyl, etc. (Conditions E, Scheme 1).
Scheme 1. Synthesis of Fmoc-1-Nal-(N-R)-Phe-OH 3.
Conditions: A) 1) RCHO, Et3N, dry DCM, rt, 2h, 2) NaBH4, 3) 2 N HCl Et2O; B) 1) RCHO, Et3N, NaBH(OAc)3, dry DCM, rt, 2 h, 2) 2 N HCl Et2O; C) 1) 4-NO2-benzyl-bromide, Cs2CO3, DMF, rt, 16 h, 2) 2 N HCl Et2O; D) Fmoc-1-Nal-Cl, DIPEA, dry DCM, rt, 16 h; E) Fmoc-1-Nal-OH, HATU, DIPEA, dry DMF, rt, 3 h; F) SnMe3(OH), DCE, 60 °C, 16 h; G) 1 N HCl/1,4-dioxane (1:1), 90 °C, 16 h.
After dipeptide synthesis, orthogonal hydrolysis was needed to obtain the free C-terminus in compounds of type 3. Standard aqueous acidic conditions (e.g., 1 N HCl/acetone (1:1 v/v) or 1 N HCl/dioxane (1:1 v/v)) at reflux temperature resulted in good conversion for aliphatic substitutions, but poor conversion and compound degradation for most aromatic substitutions. Shifting to basic conditions (LiOH in methanol at elevated temperature) improved the conversion significantly, but with epimerization as a byproduct. However, the applying conditions reported by Nicolaou et al. afforded good conversion and low degradation (Conditions F, Scheme 1).72 All synthetic details can be found in the Supporting Information.
Synthesis of Substituted Phenylalanine Derivatives
Scheme 2 depicts the synthetic route toward the 4-amino-methylated compound 11, starting from natural phenylalanine amino acid 6. First, commercially available amide 4 was treated with formalin, followed by acyl iminium formation from 5 in H2SO4, and allowing substitution at the para-position of 6. Protection of amine 7 with Boc2O resulted in 8 which was orthogonally deprotected with a 20% NaOH solution (in H2O/EtOH) to afford 9. Alloc protection, giving compound 10, followed by Boc-to-Fmoc interconversion, resulted in the final unnatural amino acid 11.
Scheme 2. Synthetic Pathway towards Fmoc-(4-CH2NHAlloc)-Phe-OH 11.

Previous work involving the Tyr(OBn) substitution of Phe13 led to a substantial increase in [Pyr1]-apelin-13 binding to the pM level and still has, to our knowledge, the highest reported affinity for the APJ-receptor.51 For this reason, benzylated tyrosine was used as a scaffold to implement an additional, positively charged functional group. Two types of residues (15a and 15b; Scheme 3) were envisioned, with charged benzyloxy and alkoxy extensions built into the Tyr residue. The unnatural amino acid 14a was prepared from commercially available 4-cyanobenzyl bromide 12. First, the bromide was converted to the alcohol, using BaCO3 in water under reflux conditions, followed by nitrile reduction applying standard Pd/C under H2-atmosphere conditions.73 Compound 13, obtained after Alloc-protection, underwent a Mitsunobu reaction with Boc-L-Tyr-OMe to cleanly deliver 14a in good yield. Simultaneous Boc-deprotection and ester hydrolysis resulted in the unprotected amino acid whose amine was Fmoc protected to yield 15a. Second, Alloc protection of commercially available 3-aminopropyl-bromide 16 resulted in 17, which was used to alkylate the phenol group of Boc-L-Tyr-OMe in DMF with Cs2CO3 in excellent yield, to present compound 14b. The protective group switch, as described above, resulted in 15b. The Alloc-protected building blocks eventually served as precursors for the aminated and guanidinylated peptide ligands.
Scheme 3. Synthetic Pathway towards the Substituted and SPPS-Compatible Tyrosine Residues.

Solid-Phase Synthesis of [Pyr1]-Apelin-13 Analogues
Once available, the (un)natural amino acids and prepared Fmoc-1-Nal-(Nα-R)-Phe-OH dipeptide analogues were used to synthesize the envisaged analogues of [Pyr1]-apelin-13 using standard Fmoc-based SPPS. Dipeptide anchoring and elongation were performed on 2-chlorotrityl chloride resin to prevent the formation of diketopiperazines (DKPs), which is a common problem in the synthesis of N-alkylated peptides by Fmoc-SPPS (Scheme 4, Conditions A).74 Resin loadings (ca. 0.4–0.7 mmol/g resin) were determined by UV absorbance measurements of dibenzofulvene after Fmoc cleavage of the first anchored residue (see Supporting Information). Peptide elongation was performed using standard Fmoc-based SPPS with DMF/HATU/DIPEA as the coupling mixture. To prevent oxidation side products due to the presence of Met,11 this residue was replaced by Nle for all peptides. This unnatural amino acid does not affect the affinity of the apelin-13 analogues, as previously reported.51,75 To access amine- or guanidine-functionalized peptides, the Alloc side chain protecting group in compound 18b was removed by using a catalytic amount of Pd(PPh3)4 in the presence of PhSiH3 as a scavenger. Thereupon, to obtain guanylated analogues, the free amine was converted to guanidine with the guanylating reagent N,N′-bis-Boc-1H-pyrazole-1-carboxamidine on solid support (Scheme 4, Conditions C). As a result, the impact of amine and guanidine functional groups on affinity and signaling could be studied.
Scheme 4. SPPS Assembly of [Pyr1]-Apelin-13 Analogues.

Conditions: A) Standard deprotection and cleavage; B) Deprotection and cleavage strategy for [Pyr1]-apelin-13 analogues with an amine on the C-terminal amino acid; C) Deprotection, guanylation and cleavage strategy for [Pyr1]-apelin-13 analogues with a guanidine on the C-terminal amino acid.
Binding Affinity and APJ Signaling of the N-Alkylated [Pyr1]-Apelin-13 Analogues
Impact of Aliphatic Nα-Substituents on Affinity and Signaling
First, aliphatic substituents were introduced, but this type of modification showed no significant effect on ligand binding compared to the unalkylated reference (Table 1). Insertion of a methyl, ethyl, and propyl group provided good binding affinity (19 (SBL-AP-022), Ki 0.15 nM; 20 (SBL-AP-023), Ki 0.12 nM; 21 (SBL-AP-024), Ki 0.17 nM vs ref with Ki 0.12 nM) and longer alkyl groups, such as isobutyl, cyclohexylmethyl and n-heptyl, were also well tolerated (22 (SBL-AP-025), Ki 0.26 nM; 23 (SBL-AP-027), Ki 0.6 nM; 24 (SBL-AP-026), Ki 0.5 nM vs ref with Ki 0.12 nM). Interestingly, most of these alkyl groups resulted in a slight increase in Gαi1 activation, compared to the reference compound and the native ligand. Isobutyl alkylation delivered one of the most potent Gαi1 activators in our study (22, EC50 Gαi1 0.4 nM vs ref, EC50 Gαi1 4.4 nM; Ape13, EC50 Gαi1 1.1 nM). Only the cyclohexylmethyl substitution resulted in a slight loss of Gαi1 activation compared to the reference sequence and a 6-fold loss compared to Ape13 (23, EC50 Gαi1 7 nM vs ref, EC50 Gαi1 4.4 nM; Ape13, EC50 Gαi1 1.1 nM). Moreover, among the analogues with aliphatic alkyl substituents, insertion of the cyclohexylmethyl group was the only case in which a significant decrease in β-arrestin 2 recruitment was observed, compared to the reference and the endogenous ligand (23 (SBL-AP-027), EC50 β-arr2 184 nM vs ref, EC50 β-arr2 41 nM; Ape13, EC50 β-arr2 40 nM). Altogether, although some differences in signaling were observed, no significant impact was noted for this series when compared with Ape13.
Table 1. Chemical Structures of the Alkylated Substituents (1-Nal12-N(R)-Phe13), Affinity and Functional Activities of C-Terminally Modified [Pyr1]-Apelin-13 Analogues.

| compound no. | compound code | peptide sequence | Ki binding (nM)a | Gαi1 (nM)b | β-arrestin 2 (nM) (Emax%)c | rat plasma t1/2 (h)d |
|---|---|---|---|---|---|---|
| Ape13 | Pyr-R-P-R-L-S-H-K-G-P-Nle-Pro-Phe-OH | 0.7 ± 0.1 | 1.1 ± 0.1 | 40 ± 4 | 0.5 ± 0.1e | |
| ref | KT04-39 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-F-OH | 0.12 ± 0.01 | 4.4 ± 0.6 | 41 ± 4 (113%) | 0.4 ± 0.1e |
| 19 | SBL-AP-022 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-Me)Phe-OH | 0.15 ± 0.01 | 1.8 ± 0.7 | 28 ± 7 (103%) | >7 |
| 20 | SBL-AP-023 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-Et)Phe-OH | 0.12 ± 0.02 | 0.7 ± 0.3 | 38 ± 7 (105%) | >7 |
| 21 | SBL-AP-024 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-nPr)Phe-OH | 0.17 ± 0.02 | 1.0 ± 0.4 | 38 ± 8 (103%) | >7 |
| 22 | SBL-AP-025 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-iBu)Phe-OH | 0.26 ± 0.06 | 0.4 ± 0.1 | 52 ± 17 (101%) | >7 |
| 23 | SBL-AP-027 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-cyclohexylmethyl)Phe-OH | 0.6 ± 0.2 | 7 ± 2 | 184 ± 55 (88%) | >7 |
| 24 | SBL-AP-026 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-nheptyl)Phe-OH | 0.50 ± 0.07 | 1.4 ± 0.3 | 62 ± 7 (94%) | >7 |
| 25 | SBL-AP-028 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-Bn)Phe-OH | 0.25 ± 0.03 | 3 ± 1 | 61 ± 14 (101%) | >7 |
| 26 | SBL-AP-029 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-PhEt)Phe-OH | 0.4 ± 0.1 | 0.9 ± 0.2 | 70 ± 34 (91%) | >7 |
| 27 | SBL-AP-030 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-Me-1′-naph)Phe-OH | 0.57 ± 0.08 | 5 ± 2 | 115 ± 35 (97%) | >7 |
| 28 | SBL-AP-031 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-Me-2′-naph)Phe-OH | 1.1 ± 0.3 | 7 ± 3 | 139 ± 52 (87%) | >7 |
| 29 | SBL-AP-036 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-(4′-Et)Bn)Phe-OH | 0.5 ± 0.2 | 4 ± 2 | 89 ± 16 (94%) | >7 |
| 30 | SBL-AP-035 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-(4′-tBu)Bn)Phe-OH | 7 ± 2 | 7 ± 3 | 312 ± 93 (87%) | >7 |
| 31 | SBL-AP-032 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-(4′-I)Bn)Phe-OH | 0.3 ± 0.1 | 0.75 ± 0.09 | 82 ± 45 (83%) | >7 |
| 32 | SBL-AP-043 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-(4′-NO2)Bn)Phe-OH | 0.17 ± 0.03 | 0.9 ± 0.1 | 34 ± 3 (96%) | >7 |
| 33 | SBL-AP-064 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-(4′-CF3)Bn)Phe-OH | 0.9 ± 0.2 | 0.6 ± 0.2 | 22 ± 3 (83%) | >7 |
| 34 | SBL-AP-034 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-(4′-OH)Bn)Phe-OH | 0.14 ± 0.09 | 5 ± 3 | 108 ± 52 (92%) | >7 |
| 35 | SBL-AP-033 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-(4′-MeO)Bn)Phe-OH | 0.27 ± 0.03 | 5 ± 3 | 97 ± 34 (96%) | >7 |
| 36 | SBL-AP-041 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-MediOBn)Phe-OH | 0.21 ± 0.08 | 5 ± 1 | 114 ± 55 (93%) | >7 |
| 37 | SBL-AP-037 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-(4′-BnO)Bn)Phe-OH | 1.4 ± 0.3 | 7 ± 1 | 119 ± 33 (92%) | |
| 38 | SBL-AP-038 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-(3′-BnO)Bn)Phe-OH | 0.9 ± 0.3 | 11 ± 7 | 258 ± 74 (95%) | >7 |
| 39 | SBL-AP-039 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-(4′-PhO)Bn)Phe-OH | 1.3 ± 0.7 | 9 ± 2 | 252 ± 118 (84%) | >7 |
| 40 | SBL-AP-040 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-(3′-PhO)Bn)Phe-OH | 3 ± 2 | 24 ± 11 | 365 ± 130 (87%) | >24 |
| 41 | SBL-AP-042 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(N-methyldibenzofurane)Phe-OH | 4.8 ± 0.4 | 0.8 ± 0.2 | 121 ± 29 (89%) | >24 |
Ki was calculated from experimental IC50 values (the concentration of ligand that displaces 50% of radiolabeled [125I][Nle75,Tyr77]Pyr-apelin-13) using the Cheng–Prusoff equation.78 Values represent the mean ± SEM of two or three experiments, each performed in duplicate.
EC50 corresponds to the concentration of ligand that produces 50% dissociation of Gαi1 from the Gβγ subunits. All compounds showed Emax Gαi1 > 96%.
EC50 is the concentration of ligand that produces 50% recruitment of β-arrestin 2 to APJ. EC50 values represent the mean ± SEM of three experiments, each performed in triplicate. All Emax values can be found in Table S2 in the Supporting Information.
Values represent the mean ± SEM of three experiments. “>7” means between 7 and 24 h.
Reported in ref (50).
Impact of Aromatic Nα-Substituents on Affinity and Signaling
To further explore the effect of the substituent, we synthesized analogues with increased bulk by implementing benzyl, phenethyl, 1/2-naphthyl and 4-ethyl/4-tbutyl-benzyl groups (i.e., compounds 25 to 30; Table 1). Overall, the additional bulk did not significantly impact the binding affinity of these ligands. Only analogue 30 (SBL-AP-035) led to a significant decrease in affinity (30 (SBL-AP-035), Ki 7 nM vs ref, Ki 0.12 nM). This suggests that a tert-butyl group represents the steric hindrance threshold to impact affinity at the para-position of benzyl. Phenethyl alkylation showed a 4-fold increase in Gαi1 activation (26 (SBL-AP-029), EC50 Gαi1 0.9 nM vs ref, EC50 Gαi1 4.4 nM), but did not impact β-arrestin 2 recruitment. Hence, the series of aromatic Nα-substituents was extended even further to potentially modulate the signaling profile.
Addition of polar groups (e.g., hydroxyl or nitro substituents; Table 1) on the para-position of the benzyl moiety resulted in a maintained binding affinity (32 (SBL-AP-043), Ki 0.17 nM; 34 (SBL-AP-034), Ki 0.14 nM) compared to the unsubstituted benzyl group (25 (SBL-AP-028), Ki 0.25 nM. Remarkably, an electron-withdrawing group on the benzyl moiety (as in 32) favors Gαi1 activation, unlike an electron-donating group (34-36), which has no significant effect (32 EC50 Gαi1 0.9 nM; vs 34–36 EC50 Gαi1 5 nM). However, the recruitment of β-arrestin 2 seems to be independent of these electronic effects. The addition of iodine (31 (SBL-AP-032)) had the same effect as the nitro group on affinity and signaling. To reduce potential toxicity risks, the nitro group, known to be toxicophoric, was replaced with an electron-withdrawing trifluoromethyl bioisostere, resulting in slightly reduced affinity (32 (SBL-AP-043), Ki 0.17 nM vs 33 (SBL-AP-064), Ki 0.9 nM) and similar signaling potency.76 Finally, electron-donating alkoxy groups (35 (SBL-AP-033) and 36 (SBL-AP-041)) induced no improvement.
Additional substitution on the benzyl group via extensions with a benzyloxy and phenoxy unit resulted in a slight loss of affinity compared to the reference and unsubstituted benzyl group, but values similar to those of Ape13 were noted (Table 1). A decrease in signaling potency was observed for Gαi1 (37 (SBL-AP-037)–40 (SBL-AP-040), EC50 Gαi1 7–24 nM), but it was even more pronounced for β-arrestin 2 recruitment (37 (SBL-AP-037)–40 (SBL-AP-040), EC50 β-arr2 119–365 nM), compared to the reference (ref, EC50 Gαi1 4.4 nM and EC50 β-arr2 41 nM). As expected, the most bulky and rigid compound with the methyldibenzofuran extension presented the lowest measured affinity (41 (SBL-AP-042), Ki 4.8 nM vs ref, Ki 0.12 nM). In contrast, Gαi1 signaling increased, compared to the reference peptide (41 (SBL-AP-042), EC50 Gαi1 0.8 nM vs ref, EC50 Gαi1 4.4 nM).
It is noteworthy that all analogues behave as full agonists of the Gαi1 pathway (Emax Gαi1 > 96%). Interestingly, some compounds are partial agonists of β-arrestin 2 recruitment, but their efficacy remained high (Emax β-arr2 > 83%). A trend is observable between the size of the Nα-substituents and Emax: the larger the substituent, the lower the Emax.
To summarize, the results showed that the binding pocket displays a high tolerance to a wide variety of bulky substituents. Our observations are in agreement with recent X-ray structures of APJ complexed with an apelin-17 mimetic peptide that revealed that Pro12 and Phe13 are surrounded in the orthosteric binding pocket by hydrophobic residues with plenty of space available to diversify.63 In previous modifications of the Pro12 position of linear Ape13 analogues, we only noticed improvements in affinity and stability, but signaling remained unbiased.61,62,77 In this subseries, we did not obtain biased analogues, but a decrease in potency and maximum efficacy of β-arrestin 2 recruitment was observed.
Binding Affinity and APJ Signaling of the Positively Charged [Pyr1]-Apelin-13 Analogues
Impact of Positively Charged Residues at the C-Terminus on Affinity and Signaling
The above results showed that adding purely apolar aliphatic and aromatic groups at the C-terminus of [Pyr1]-apelin-13 analogues had no significant effect on affinity and signaling. However, the addition of polar and electron-withdrawing functionality had a significantly impact on signaling patterns. Hence, substitution of the para-position of Phe13 with a cyano group was carried out first. Compound 42 (SBL-AP-018) provided no significant change in binding affinity compared to the reference ligand (42, Ki 0.13 nM vs ref, Ki 0.12 nM), but a 10- and 3-fold increase was obtained in Gαi1 activation and β-arrestin 2 recruitment, respectively, compared to the reference (Table 2). Moreover, analogue 42 was found to be the best β-arrestin 2 recruiter in this study 42, (EC50 β-arr2 14 nM). Introduction of a positively charged aminomethyl group via a nitrile reduction (43 (SBL-AP-019)) resulted in a 20-fold loss of binding affinity compared to the reference peptide, a 6-fold loss of Gαi1 activation, and a significant reduction in β-arrestin 2 recruitment was observed, compared to the nitrile analogue 42. Guanylating the primary amine (44 (SBL-AP-020); Table 2) led not only to a further loss of affinity but also a loss of signaling efficacy (Ki 4.6 nM; EC50 Gαi1 10 nM; EC50 β-arr2 423 nM with Emax of 59%). The guadinylated analogue of Phe13 (45 (SBL-AP-051); Table 2) further decreased affinity, but Gαi1 signaling remained nevertheless in the low nM range, while the β-arrestin 2 pathway was attenuated (with Emax of 67%) to a similar extent of the guanidinomethyl analogue 44 (SBL-AP-020). Interestingly, the positively charged compounds are the only ones in this series that are partial agonists in β-arrestin 2 recruitment. Thus, the presence of guanidine moieties seemed promising, and we therefore altered the position of this positive charge relative to the peptide backbone via extension of the side chain (see entries 48 (SBL-AP-057) to 51 (SBL-AP-048); Table 2).
Table 2. Chemical Structures of Ligands Bearing a Positive Charge, Affinity and Functional Activities of C-Terminally Modified [Pyr1]-Apelin-13.
| compound no. | compound code | peptide sequence | Ki binding (nM)a | Gαi1 (nM)b | β-arrestin 2 (nM) (Emax%)c | rat plasma t1/2 (h)d |
|---|---|---|---|---|---|---|
| Ape13 | Pyr-R-P-R-L-S-H-K-G-P-Nle-Pro-Phe-OH | 0.7 ± 0.1 | 1.1 ± 0.1 | 40 ± 4 | 0.5 ± 0.1e | |
| ref | KT04-39 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-F-OH | 0.12 ± 0.01 | 4.4 ± 0.6 | 41 ± 4 | 0.4 ± 0.1e |
| 42 | SBL-AP-018 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-Phe(4-CN)-OH | 0.13 ± 0.02 | 0.4 ± 0.2 | 14 ± 5 (100%) | <1 |
| 43 | SBL-AP-019 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-Phe(4-CH2-NH2)-OH | 3.4 ± 1.0 | 2.4 ± 0.9 | 105 ± 39 (80%) | 1.0 ± 0.01 |
| 44 | SBL-AP-020 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-Phe(4-CH2-Gn)-OH | 4.6 ± 0.1 | 10 ± 4 | 423 ± 58 (59%) | <1 |
| 45 | SBL-AP-051 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-Phe(4-Gn)-OH | 7 ± 2 | 12 ± 5 | 303 ± 130 (67%) | <1 |
| 46 | SBL-AP-045 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-Tyr(O-4-NO2-Bn)-OH | 0.15 ± 0.004 | 0.9 ± 0.4 | 60 ± 5 (99%) | 5.4 ± 0.9 |
| 47 | SBL-AP-046 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-Tyr(O-4-CN-Bn)-OH | 0.12 ± 0.03 | 0.4 ± 0.2 | 28 ± 5 (99%) | 3.0 ± 0.4 |
| 48 | SBL-AP-057 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-Tyr(O-4-Bn-CH2-NH2)-OH | 3.0 ± 0.4 | 10 ± 3 | 324 ± 49 (80%) | 1.3 ± 0.1 |
| 49 | SBL-AP-058 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-Tyr(O-4-Bn-CH2-Gn)-OH | 1.1 ± 0.4 | 11 ± 4 | 275 ± 14 (54%) | 1.1 ± 0.02 |
| 50 | SBL-AP-047 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-Tyr(O-4-npropyl-NH2)-OH | 0.30 ± 0.08 | 1.2 ± 0.5 | 53 ± 2 (97%) | <1 |
| 51 | SBL-AP-048 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-Tyr(O-4-npropyl-Gn)-OH | 3 ± 1 | 1.4 ± 0.5 | 197 ± 18 (70%) | <1 |
| 52 | SBL-AP-021 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-Arg-OH | 1.2 ± 0.3 | 6 ± 2 | 116 ± 19 (76%) | <1 |
| 53 | SBL-AP-054 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-d-Arg-OH | 25 ± 5 | 10 ± 2 | 906 ± 229 (66%) | 2.8 ± 0.1 |
| 54 | SBL-AP-052 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-Lys-OH | 7.8 ± 0.4 | 14 ± 4 | 708 ± 185 (67%) | <1 |
| 55 | SBL-AP-053 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-hArg-OH | 3.2 ± 0.2 | 6 ± 3 | 412 ± 89 (72%) | <1 |
Ki was calculated from experimental IC50 values (the concentration of ligand that displaces 50% of radiolabeled [125I][Nle75,Tyr77]Pyr-apelin-13) using the Cheng–Prusoff equation.78 Values represent the mean ± SEM of two or three experiments, each performed in duplicate.
EC50 corresponds to the concentration of ligand that produces 50% dissociation of Gαi1 from the Gβγ subunits. All compounds showed an Emax Gαi1 > 99%.
EC50 is the concentration of ligand that produces 50% recruitment of β-arrestin 2 to the apelin receptor. EC50 values represent the mean ± SEM of three experiments, each performed in triplicate. All Emax values can be found in Table S2 in the Supporting Information.
Values represent the mean ± SEM of three experiments.
Reported in ref (50).
The introduction of 4′-substituted benzyl groups on Tyr, replacing Phe,13 was performed. Replacement of Phe13 with 4-nitro- and 4-cyanobenzyl-Tyr (46 (SBL-AP-045) and 47 (SBL-AP-046), respectively) led to similar binding affinity and β-arrestin 2 recruitment to the reference ligand (Table 2). However, higher Gαi1 potencies were observed (46, EC50 Gαi1 0.9 nM; 47, EC50 Gαi1 0.4 nM vs ref, EC50 Gαi1 4.4 nM). Reduction of nitrile toward aminomethyl (48 (SBL-AP-057)) provided very similar data compared to the previous aminomethylated analogue (43, Ki 3.4 nM vs 48, Ki 3.0 nM; with β-arr2 Emax of 80% for both), again showing the benefit of positive charges in finding biased ligands. Additionally, the corresponding guanylated analogue 49 (SBL-AP-058) recovered some affinity (49, Ki 1.1 nM), compared with the aminomethyl derivative 48, but more importantly, a maximum β-arrestin 2 recruitment efficacy of only 54% could be reached.
In a next step, we expanded the series with different side chain lengths (50-55; Table 2) to identify more precisely where the positive charge could be placed relative to the backbone to optimize the signaling bias. Substitutions on Tyr13 with a linear alkyl chain bearing amine or guanidine functionality were investigated, alongside the use of more standard basic amino acids, such as Arg and Lys, or variants thereof. Altogether, this subset of analogues provided ligands with high affinity (e.g., 50 (SBL-AP-047) and 52 (SBL-AP-021)), high Gαi1 signaling potency (e.g., 50 and 51 (SBL-AP-048)) and some with partial agonism, strongly disfavoring β-arrestin 2 recruitment (with a range between 66% and 76%). A reduction of the β-arrestin 2 signaling was observed, especially for the analogues possessing d-Arg13 and l-Lys13 (53 (SBL-AP-054), EC50 β-arr2 906 nM, Emax 66%; 54 (SBL-AP-052), β-arr2 708 nM, Emax 67%). Therefore, this suggests that, in addition to a positive charge, a shorter aliphatic side chain seems to decrease β-arrestin 2 recruitment and its maximum efficacy.
Synthesis of Constrained Analogues Bearing a Positive Charge
In an attempt to further optimize ligand binding, bias, and stability as well as to pinpoint more precisely in which direction the charged group should be oriented, we combined the N-alkyl substituent and the positive charge in this series of constrained analogues. We first opted to design tetrahydroisoquinoline-based constraint type 59 (Scheme 5), which correspond to more rigid versions of derivatives 50 (SBL-AP-047) and 51 (SBL-AP-048) (Table 2). Such custom residues not only allow χ-space screening but also represent tools for increasing the affinity and metabolic stability of more flexible peptide analogues.50,79
Scheme 5. Synthetic Pathway Towards Constrained Amino Acids and Dipeptides Bearing a Positive Charge, 59 and 61.

Panels: (A) Synthetic pathway toward (Fmoc-Tic(6-O-propyl-NHAlloc)-OH 59; (B) Synthetic pathway toward Fmoc-L-Aia-Orn(Alloc)-OH 61.
To obtain scaffold 59, treatment of meta-tyrosine 56 under Pictet–Spengler conditions gave the expected 6-hydroxy-tetrahydroxyisoquinoline Tic(6-OH) in excellent yield (Scheme 5, panel A). Next, the amine and acid were protected to deliver 57. The free phenol was engaged in a substitution reaction with previously synthesized Alloc-protected amino-propyl-bromide 17, to afford 58, which was Boc-deprotected and saponified in a one-pot protocol. Finally, Fmoc protection resulted in 59. Additionally, constrained building block 61 was prepared from Fmoc-2-formyl-L-Trp-OH 60, following a literature procedure (Scheme 5, panel B).80 Compound 61 served as a precursor for the Aia-Arg-bearing sequence 64.
Binding Affinity and APJ Signaling of the Constrainded and Positively Charged [Pyr1]-Apelin-13 Analogues
Impact of Constrained Analogues Bearing a Positive Charge at the C-Terminus on Binding Affinity and Receptor Signaling
With the combination of both modifications, we observed for the analogue carrying Tic amino acid (62 (SBL-AP-059)) native-like affinity and Gαi1 signaling, whereas β-arrestin 2 recruitment is decreased 4-fold, with the maximum efficacy of 87% (Table 3, Figure 2). Replacing 1-Nal12 with the amino-indoloazepinone (Aia) template and introducing a C-terminal Orn (63 (SBL-AP-049); Table 3, Figure 2) produced a decrease in receptor binding affinity and Gαi1 signaling, but more importantly in β-arrestin 2 recruitment (63, Ki 24 nM; EC50 Gαi1 22 nM; EC50 β-arr2 864 nM). However, replacement of 1-Nal12 in 52 with the constrained Aia residue resulted in a 4-fold loss of binding affinity (52, Ki 1.2 nM vs 64 (SBL-AP-050), Ki 4.7 nM), a 2-fold loss in Gαi1 activation and a 9-fold loss in β-arrestin 2 recruitment (Table 3, Figure 2). Interestingly, 63 and 64 behaved as partial agonists on the β-arrestin 2 pathway with Emax values of 48% and 82%, respectively. These data suggest that the substituted Tic analogue (62) does not provide optimal positive charge orientation to optimize signaling bias, but that the Aia moiety, used as a 1-Nal surrogate, provides one of the lowest maximal β-arrestin 2 recruitment efficacies. Noteworthily, both types of constraints improved proteolytic stability (vide infra).
Table 3. Chemical Structures of Constrained Ligands Bearing a Positive Charge and Affinity, Functional Activities of C-Terminally Modified [Pyr1]-Apelin-13 Analogues.
| compound no. | compound code | peptide sequence | Ki binding (nM)a | Gαi1 (nM)b | β-arrestin 2 (nM) (Emax%)c | rat plasma t1/2 (h)d |
|---|---|---|---|---|---|---|
| Ape13 | Pyr-R-P-R-L-S-H-K-G-P-Nle-Pro-Phe-OH | 0.7 ± 0.1 | 1.1 ± 0.1 | 40 ± 4 | 0.5 ± 0.1e | |
| ref | KT04-39 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-F-OH | 0.12 ± 0.01 | 4.4 ± 0.6 | 41 ± 4 | 0.4 ± 0.1e |
| 62 | SBL-AP-059 | Pyr-R-P-R-L-S-H-K-G-P-Nle-1-Nal-(O-6-npropyl-Gn)Tic-OH | 0.74 ± 0.01 | 1.3 ± 0.5 | 110 ± 28 (87%) | >7 |
| 63 | SBL-AP-049 | Pyr-R-P-R-L-S-H-K-G-P-Nle-Aia-Orn-OH | 24 ± 4 | 22 ± 13 | 864 ± 321 (48%) | 5.3 ± 0.1 |
| 64 | SBL-AP-050 | Pyr-R-P-R-L-S-H-K-G-P-Nle-Aia-Arg-OH | 4.7 ± 0.1 | 13 ± 6 | 993 ± 616 (82%) | 4.8 ± 0.4 |
Ki was calculated from experimental IC50 values (the concentration of ligand that displaces 50% of radiolabeled [125I][Nle75,Tyr77]Pyr-apelin-13) using the Cheng–Prusoff equation.78 Values represent the mean ± SEM of two or three experiments, each performed in duplicate.
EC50 corresponds to the concentration of ligand that produces 50% dissociation of Gαi1 from the Gβγ subunits. All compounds showed an Emax Gαi1 > 97%.
EC50 is the concentration of ligand that produces 50% recruitment of β-arrestin 2 to the apelin receptor. EC50 values represent the mean ± SEM of three experiments, each performed in triplicate. All Emax values can be found in Table S2 in the Supporting Information.
Values represent the mean ± SEM of three experiments. > 7 h means between 7 and 24 h.
Reported in ref (50).
Figure 2.
Functional assay and plasma stability of 62, 63, and 64 compared to Ape13. BRET concentration–response curves monitoring β-arrestin 2 recruitment (A) and degradation curves in rat plasma (B). Data are represented as the mean ± SEM of at least three separated experiments.
In Vitro Stability in Rat Plasma
The [Pyr1]-Apelin-13 analogues in this study were evaluated for in vitro proteolytic stability in rat plasma to test the impact of amide Nα-substituents and insertion of various phenylalanine derivates on degradation. The Nα-substitution led to a substantial increase in plasma stability (19-41, t1/2 > 7 h, Table 1), compared with the reference peptide and Ape13 (ref, t1/2 0.4 h; Ape13, t1/2 0.5 h). It is well-known that ACE2 is responsible for the main degradation of Ape13 analogues by hydrolysis between the last two C-terminal amino acids (Figure 1). Therefore, as expected, the addition of substituents on the backbone amide bond between 1-Nal12 and Phe13 significantly increased plasma stability, presumably by blocking cleavage by ACE2. Furthermore, incorporation of small polar substituents on the para-position of Phe13 (42–45) did not significantly impact plasma stability (Table 2). The same observation was made for the charged aliphatic substituents in compounds 50-51. In contrast, the addition of benzyloxy derivates of Phe13 (46-49), creating much bulkier side chains, increased plasma stability up to 10-fold in comparison with the reference and Ape13 (46, t1/2 5.4 h vs ref, t1/2 0.4 h; Ape13, t1/2 0.5 h). Additionally, aliphatic side chains at the 13th position (as in 52, 54, 55, Table 2) did not elicit any change in plasma stability, with the exception of analogue 53 bearing D-Arg13 (t1/2 2.8 h, Table 2) which exhibits inverted stereochemistry, compared to the other analogues (with an L-configuration). Moreover, constrained analogues (62-64) with backbone-anchored side chains increased plasma stability (62, t1/2 > 7 h; 63, t1/2 5.3 h; 64, t1/2 4.8 h, Table 3). Overall, the alkylated peptide bonds, increased side chain size and inverted stereochemistry at the C-terminus of [Pyr1]-Apelin-13 analogues decreased degradation in rat plasma. It is noteworthy that the positive charge had no influence on the stability of the compounds.
In Vivo Blood Pressure Measurement
To complete this study, we investigated the impact of applied chemical modifications on blood pressure in anesthetized rats. Apelin analogues were administered to rats via a single intravenous (i.v.) injection at a dose of 19.6 nmol/kg, while the mean arterial blood pressure (MABP) was continuously monitored. As recently reported by our group, the drop in blood pressure reaches the maximum hypotensive effect when [Pyr1]-Apelin-13 is injected at this bolus dose.81 Moreover, we have previously shown that the β-arrestin recruitment upon APJ activation correlates with the physiological hypotension effect observed in rats.40 Therefore, different analogues harboring an Nα-substituent, a positive charge at the C-terminal, or both, with maximal and reduced β-arrestin 2 recruitment potencies and efficacies were selected for in vivo blood pressure measurements. Analogues 19 and 47 produced a drop in blood pressure comparable to that of Ape13 (19, Max ΔMABP −37 mmHg; 47, Max ΔMABP −34 mmHg vs Ape13, Max ΔMABP −36 mmHg), which could be rationalized by their similarity in terms of β-arrestin 2 recruitment. Analogue 40, the N-alkylated compound with the lowest β-arrestin 2 recruitment potency (EC50 β-arr2 365 nM, Emax 87%), produced a 2-fold lower hypotensive effect than the endogenous ligand (40, Max ΔMABP −17 mmHg). The conformationally constrained and positively charged analogues 63 and 64 were the two most promising compounds in this study due to their good affinity for APJ, strong activation of the Gαi1 pathway, greatly increased proteolytic stability and limited recruitment of β-arrestin 2, with partial maximum efficacy (63, EC50 β-arr2 864 nM, Emax 48%; 64, EC50 β-arr2 993 nM, Emax 82%). Analogue 64 induced a 2-fold smaller pressure drop than Ape13 (64, Max ΔMABP −18 mmHg vs Ape13, Max ΔMABP −36 mmHg) and, interestingly, analogue 63 did not even affect blood pressure (63, Max ΔMABP −3 mmHg vs Saline, Max ΔMABP −2 mmHg). At a very higher dose (65.0 nmol/kg), however, analogue 63 demonstrated a blood pressure lowering effect that was nevertheless less significant than for Ape13 at this dose (63, Max ΔMABP −22 mmHg vs Ape13, Max ΔMABP −36 mmHg). Both analogues 63 and 64 showed the benefit of partial agonism in the β-arrestin 2 recruitment pathway. All analogues tested exhibit a longer plasma half-life than Ape13, yet no difference was observed in the duration of blood pressure effects.
Conclusion
In this study, 40 analogues of [Pyr1]-apelin-13, including backbone-alkylated dipeptides as well as modified amino acids, were synthesized. The general observation of this study is that the addition of various alkyl groups (e.g., Et, 4-nitrobenzyl, 4-benzyloxy) onto the Nα of Phe13 is well tolerated and produces excellent APJ ligands with affinities in the very low nM range (0.12–0.9 nM). Moreover, potent Gαi1 activators were obtained (EC50 Gαi1 0.4–0.9 nM). Interestingly, the introduction of longer and bulkier substituents (e.g., N-(3′-BnO)Bn (38) and N-(3′-PhO)Bn (40)) resulted in compounds with similar binding affinities to Ape13, but significant losses were observed in the signaling potency for the β-arrestin 2 pathway (EC50 β-arr2 258–365 nM) with a small decrease in the maximum efficacy (Emax 87–895%). These modifications also drastically increased stability in rat plasma with half-lives greater than 7 h. IWhen tested in vivo, the compound characterized by a reduced ability to recruit β-arrestin 2 also induced a decreased hypotensive effect (cfr. 19 vs 40 in Figure 3). Incorporation of a positive charge on the side chain of the C-terminal residue (herein a modified Phe or Tyr) provided (near-)native binding affinities, but at the same time unique signaling profiles where partial agonism for β-arrestin 2 recruitment was obtained (e.g., 44, 45, 49, 51–53, 63, 64). Among these, the constrained and positively charged C-terminal dipeptide Aia-Orn in analogue 63 gave rise to a promising analogue with high affinity (Ki 24 nM), high Gαi1 activation (EC50 Gαi1 22 nM), low β-arrestin 2 recruitment potency with partial maximum efficacy (EC50 β-arr2 864 nM, Emax 48%), improved stability (t1/2 5.3 h) and a greatly decreased hypotensive effect. In conclusion, we report the useful design and synthesis of apelin-13 analogues with high binding affinities and improved plasma stability by means of minimal structural modifications at the apelin C-terminus. These bioactive molecules represent novel pharmacological tools that may help shed new light on apelin-related pathologies.
Figure 3.
Effects of Ape13 and selected analogues on blood pressure in anesthetized rats. Recorded mean arterial blood pressure (ΔMABP) after intravenous injection of (A) Ape13 and selected analogues at 19.6 nmol/kg and (B) Ape13 and 63 at 65.0 nmol/kg. Maximal reduction of mean arterial pressure (Max ΔMAP) of (C) Ape13 and selected analogues at 19.6 nmol/kg and (D) Ape13 and 63 at 65.0 nmol/kg. Data are represented as the mean ± SEM (n = 5). Statistical analyses were performed with a one-way ANOVA followed by a Dunnett’s multiple comparisons test. **, p < 0.01, ***, p < 0.001, and ****, p < 0.0001 vs Ape13.
Experimental Section
General Information and Compound Characterization
Unless stated otherwise, all commercial chemicals were used without further purification. 1H and 13C NMR spectra were recorded using a Bruker Avance II 500 MHz at ambient temperature with the frequencies of 1H and 13C set, respectively, at 500.03 and 125.74 MHz. The chemical shifts were reported in delta (δ) units in parts per million (ppm) relative to the signal of the deuterated solvent. For CDCl3, the singlet in 1H NMR was calibrated at 7.26 ppm and the central line of the triplet in 13C NMR at 77.0 ppm. When recording in MeOD-d4 or DMSO-d6, the calibration was performed, respectively, at 3.31 and 2.50 ppm for 1H NMR and 49.0 and 39.5 ppm for 13C NMR. Assignments were made using one-dimensional (1D) 1H and 13C spectra and two-dimensional (2D) HSQC, HMBC, and COSY spectra. Multiplicities were described as singlet (s), doublet (d), triplet (t), quartet (q), multiplet (m), and broad (br), or a combination thereof. The corresponding coupling constants (J values) were reported in Hertz (Hz). Analytical RP-HPLC was performed on a VWR-Hitachi Chromaster HPLC with a Chromolith HighResolution RP-18C column from Merck (150 × 4.6 mm, 1.1 μm, 150 Å). The flow rate was 3 mL/min and UV detection occurred at 214 nm. The solvent system used consisted of 0.1% TFA in ultrapure water (A) and 0.1% TFA in acetonitrile (B) with a gradient from 3% B to 100% B over a 6 min runtime. For LC/MS analysis, the HPLC unit used was a Waters 600 system combined with a Waters 2487 UV detector at 215 nm and as stationary phase an EC 150/2 NUCLEODUR 300-5 C18 ec-column (150 × 2.1 mm2, 3 μm, 300 Å). The solvent system used was 0.1% formic acid in water (A) and 0.1% formic acid in acetonitrile (B) with a gradient going from 3% to 100% B over 20 min with a flow rate of 0.3 mL/min. The MS unit, coupled to the HPLC system, was a Micromass QTOF-micro system. For the high-resolution mass spectroscopy, the same MS system was used with reserpine (2.10–3 mg/mL) solution in H2O:CH3CN (1:1) as reference. Microwave syntheses were performed using a Biotage Initiator+ SP Wave. Automated flash chromatography was performed using Grace Reveleris X2 system equipped with an ELSD and UV detector (254 or 280 nm). The normal phase columns used for the systems were Grace Silica Flash Cartridges of 40 g with a flow rate of 40 mL/min unless stated otherwise. Semipreparative RP-HPLC-purifications were done using a Gilson HPLC system with Gilson 322 pump equipped with a INTERCHIM Vydac 150HC C18 (250 × 22 mm2, 10 μm) column and Waters UV/vis-156 detector at 215 nm. The same solvent system was used as applied for the analytical RP-HPLC with a flow rate of 20 mL/min.
Binding Experiments
Binding experiments were carried out on the cell membranes of HEK293 stably expressing the YFP-tagged human APJ receptor. Cells were frozen at −80 °C for storage and only thawed right before the experiments. For membrane extraction, cells were suspended in EDTA solution (1 mM EDTA and 10 mM Tris-HCl), transferred to a falcon tube, and centrifuged at 1600 g for 15 min at 4 °C. The precipitate was suspended in binding buffer (50 mM Tris-HCl, 0.2% BSA, pH 7.4). Binding assay was carried out in 96-well plates: 15 μg of membrane proteins was incubated with 0.2 nM radiolabeled [125I][Nle75,Tyr77]Pyr-apelin-1335 (820 Ci/mmol) and the test ligand with a range of concentrations from 10–5 to 10–11 M in a total volume of 200 μL for 1 h at room temperature. The incubation mixtures were filtered through a glass fiber filter (Millipore, preabsorbed of PEI 0.5% for 2 h at 4 °C) to remove unbound ligands, and the filtered membranes were washed three times with 150 μL of cold binding buffer (4 °C). The γ emission was measured using a γ-counter 1470 Wizard from PerkinElmer (Waltham) (80% efficiency). Nonspecific binding did not exceed over 5% of the total signal (determined by incubation with 10–5 M unlabeled [Pyr1]-apelin-13). IC50 values, determined from those results using GraphPad Prism 9, represent the concentration of the tested ligand displacing 50% of the radiolabeled ligand from the receptor. The KD of [Pyr1]-apelin-13 is 1.8 nM, determined by the saturation binding assay. The dissociation constant Ki value was calculated from IC50 using the Cheng–Prusoff equation, and results were displayed as mean ± SEM of two to three independent experiments, each done in duplicate.78
BRET Assays for Gαi1 Activation and β-Arrestin 2 Recruitment
HEK293 cells seeded in T175 flasks were allowed to grow in high-glucose DMEM supplemented with 10% FBS, 100 U/mL penicillin/streptomycin at 37 °C in a humidified chamber at 5% CO2. All transfections were carried out with PEI. After 24 h, cells were transfected with the plasmids coding for hAPJ, Gαi1-RlucII(91), GFP10-Gγ2, and Gβ1 (for the BRET-based Gαi1 activation assay) or coding for hAPJ, CAAX-GFP10 and RlucII-βarrestin 2.51,82,83 To perform BRET assays, cells were transferred into white 96-well plates BD Bioscience (Mississauga, Canada) at a concentration of 50 000 cells/well 24 h after transfection and incubated at 37 °C overnight. Cells were then washed with phosphate-buffered saline (PBS), and 90 μL of Hank’s balanced salt solution (HBSS) was added to each well. Then, cells were stimulated with analogues at concentrations ranging from 10–5 to 10–11 M for 5 min at 37 °C (Gαi1) or for 30 min at room temperature (β-arrestin 2). After stimulation, 5 μM coelanterazine 400a was added to each well and the plate was read using the BRET2 monochromatic filter set of a MithrasLB943 plate reader (Berthold Technologies, Germany). The BRET ratio was determined as GFP10em/RlucIIem. Data were plotted, and EC50 values were determined using GraphPad Prism 9. Each data point represents the mean ± SEM of at least three different experiments each done in triplicate.
Rat Plasma Stability
In a 96-well plate, 6 μL of compound (1 mM aqueous solution) was incubated with 27 μL of plasma (from male Sprague–Dawley rat) at 37 °C for 0, 1, 2, 4, 7, and 24 h. Degradation was quenched by adding 140 μL of 1% formic acid in acetonitrile-ethanol (1:1) solution containing N,N-dimethylbenzamide 0.25 mM (internal standard). This mixture was transferred to a filter plate Impact Protein Precipitation (Phenomenex, California), and another 96-well UPLC plate was placed at the bottom to collect filtrates. Both plates were centrifuged at 800 g for 10 min at 4 °C. The filtrates were diluted with 30 μL of distilled water and analyzed using an Acquity UPLC-MS system class H (column Acquity UPLC CSH C18 (2.1 mm × 50 mm), 1.7 μm particles with pores 130 Å). The degradation was calculated by comparing the mass spectrum of each point with those at 0 min (plasma-inactivated before adding the compound). Peptide half-life was calculated from the degradation curve using the exponential one-phase decay function in GraphPad Prism 9. Data were presented as mean ± SEM when the data are >1 h and <7 h of at least three different experiments each done in simplicate.
In Vivo Blood Pressure Measurement
Adult male Sprague–Dawley rats, 8–10 weeks of age (Charles River Laboratories, St-Constant, Quebec, Canada), were kept on a 12 h light/12 h dark cycle with access to food and water ad libitum. The animal experimental protocols in this study were approved by the Animal Care Committee of Université de Sherbrooke and complied with policies and directives of the Canadian Council on Animal Care.
Male Sprague–Dawley rats (250–300 g) anesthetized with ketamine/xylazine injection (87/13 mg/kg intramuscular (i.m.)) were placed in supine position on a thermostatic pad. A blood pressure monitoring catheter (PE 50 filled with heparinized saline) was inserted into the right carotid artery and connected to a Micro-Med transducer (model TDX-300, Calabasas) and a MicroMed blood pressure analyzer (model BPA-100c). Bolus injection of vehicle (isotonic saline) followed 5 min later by the injection of Ape13, saline, or modified analogues, 19, 40, 47, 63, or 64, (given at 19.6 or 65 nmol/kg; volume of 0.25 mL over 10 s) was carried out through another catheter (PE10) inserted into the left jugular vein. To remove residual injected substances, this i.v. catheter was flushed with saline (0.2 mL) immediately after each injection.
Acknowledgments
R.V.D.H. and L.T. pay special gratitude to late professor Eric Marsault for all the support and the many inspiring discussions.
Glossary
Abbreviations
- 1-Nal
3-(1-naphthyl)-l-alanine
- AA
amino acid
- ACE2
angiotensin-converting enzyme 2
- ACN
acetonitrile
- Aia
amino-indoloazepinone
- AMP
adenosine monophosphate
- Ape13
[Pyr1]-apelin-13
- Arg
arginine
- Atm
atmosphere
- Bn
benzyl
- Boc
tert-butyloxycarbonyl
- BRET
bioluminescence resonance energy transfer
- BSA
bovine serum albumin
- CSH
charged surface hybrid
- DCM
dichloromethane
- DIAD
diisopropyl azodicarboxylate
- DIPEA
N,N-diisopropylethylamine
- DKP
diketopiperazine
- DMEM
Dulbecco’s modified eagle medium
- DMF
N,N-dimethylformamide
- ECL2
extracellular loop
- EDC
1-ethyl3-(3-(dimethylamino)propyl)carbodiimide
- EDTA
ethylenediaminetetraacetic acid
- ERK
extracellular-signal-regulated kinase
- Fmoc
9-fluorenylmethoxycarbonyl
- GFP
green fluorescent protein
- Gn
guanidine
- GPCR
G-protein-coupled receptor
- hAPJ
human APJ
- HATU
1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate
- HBSS
Hank’s balanced salt solution
- HEK
human embryonic kidney
- HRMS
high-resolution mass spectrometry
- Lys
lysine
- MABP
mean arterial blood pressure
- Met
methionine
- MS
mass spectrometry
- NEP
neprilysin
- Nle
norleucine
- NMM
N-methylmorpholine
- Orn
ornithine
- PBS
phosphate-buffered saline
- PEI
polyethylenimine
- Ph
phenyl
- Phe
phenylalanine
- PRCP
proline carboxypeptidase
- Pro
proline
- Pyr
pyroglutamic acid
- Raf
rapidly accelerated fibrosarcoma
- RhoGEF
Rho-guanine nucleotide exchange factor
- RlucII
Renilla luciferase
- rt
room temperature
- SAR
structure–activity relationship
- SEM
standard error of the mean
- SPPS
solid-phase peptide synthesis
- TFA
trifluoroacetic acid
- THF
tetrahydrofuran
- Tic
tetrahydroisoquinoline carboxylic acid
- TIPS
triisopropylsilane
- TM
transmembrane domain
- Tris
tris(hydroxymethyl)aminomethane
- Trp
tryptophan
- Tyr
tyrosine
- UPLC
ultrahigh-performance liquid chromatography
- YFP
yellow fluorescent protein
Supporting Information Available
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsptsci.2c00219.
Synthetic procedures for unnatural amino acids and compound characterization; SPPS procedures; HRMS spectra; analytical UPLC-MS spectra; structures and molecular formula strings of the investigated compounds; competitive binding curves, saturation BRET curves monitoring Gαi1 activation and β-arrestin 2 recruitment; degradation curves in rat plasma (PDF)
Author Contributions
∥ L.T. and R.V.D.H. contributed equally to this study. S.B., P.-L.B. and P.S. contributed equally to directing this study.
Financial support from Université de Sherbrooke, the Canadian Institutes of Health Research, the Canada Foundation for Innovation, the Fonds de la Recherche du Québec en Santé (FRQS) is gratefully acknowledged. The Canadian Francophonie Scholarship Program (PCBF) is also acknowledged for scholarship grants to K.T. P.S. is the recipient of the Canada Research Chair in Neurophysiopharmacology of Chronic Pain. E.M. and P.L.B. are members of the FRQNT-funded Proteo Network. R.V.D.H. and S.B. thank the Strategic Research Program (SRP50) of the Research Council of VUB for the financial support.
The authors declare no competing financial interest.
Special Issue
Published as part of the ACS Pharmacology & Translational Science virtual special issue “GPCR Signaling”.
Supplementary Material
References
- O’Dowd B. F.; Heiber M.; Chan A.; Heng H. H.; Tsui L. C.; Kennedy J. L.; Shi X.; Petronis A.; George S. R.; Nguyen T. A human gene that shows identity with the gene encoding the angiotensin receptor is located on chromosome 11. Gene 1993, 136 (1–2), 355–360. 10.1016/0378-1119(93)90495-O. [DOI] [PubMed] [Google Scholar]
- Tatemoto K.; Hosoya M.; Habata Y.; Fujii R.; Kakegawa T.; Zou M. X.; Kawamata Y.; Fukusumi S.; Hinuma S.; Kitada C.; Kurokawa T.; Onda H.; Fujino M. Isolation and characterization of a novel endogenous peptide ligand for the human APJ receptor. Biochem. Biophys. Res. Commun. 1998, 251 (2), 471–476. 10.1006/bbrc.1998.9489. [DOI] [PubMed] [Google Scholar]
- Chng S. C.; Ho L.; Tian J.; Reversade B. ELABELA: a hormone essential for heart development signals via the apelin receptor. Dev Cell 2013, 27 (6), 672–680. 10.1016/j.devcel.2013.11.002. [DOI] [PubMed] [Google Scholar]
- Pauli A.; Norris M. L.; Valen E.; Chew G. L.; Gagnon J. A.; Zimmerman S.; Mitchell A.; Ma J.; Dubrulle J.; Reyon D.; Tsai S. Q.; Joung J. K.; Saghatelian A.; Schier A. F. Toddler: an embryonic signal that promotes cell movement via Apelin receptors. Science 2014, 343 (6172), 1248636. 10.1126/science.1248636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang Y.; Yan M.; Wang C.; Wang Q.; Chen X.; Zhang R.; Wan L.; Ji B.; Dong B.; Wang H.; Chen J. The Effects of Apelin and Elabela Ligands on Apelin Receptor Distinct Signaling Profiles. Front Pharmacol 2021, 12, 630548. 10.3389/fphar.2021.630548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu W.; Yan J.; Pan W.; Tang M. Apelin/Elabela-APJ: a novel therapeutic target in the cardiovascular system. Ann. Transl Med. 2020, 8 (5), 243. 10.21037/atm.2020.02.07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mughal A.; O’Rourke S. T. Vascular effects of apelin: Mechanisms and therapeutic potential. Pharmacol Ther 2018, 190, 139–147. 10.1016/j.pharmthera.2018.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hosoya M.; Kawamata Y.; Fukusumi S.; Fujii R.; Habata Y.; Hinuma S.; Kitada C.; Honda S.; Kurokawa T.; Onda H.; Nishimura O.; Fujino M. Molecular and functional characteristics of APJ. Tissue distribution of mRNA and interaction with the endogenous ligand apelin. J. Biol. Chem. 2000, 275 (28), 21061–21067. 10.1074/jbc.M908417199. [DOI] [PubMed] [Google Scholar]
- Kawamata Y.; Habata Y.; Fukusumi S.; Hosoya M.; Fujii R.; Hinuma S.; Nishizawa N.; Kitada C.; Onda H.; Nishimura O.; Fujino M. Molecular properties of apelin: tissue distribution and receptor binding. Biochim. Biophys. Acta 2001, 1538 (2–3), 162–171. 10.1016/S0167-4889(00)00143-9. [DOI] [PubMed] [Google Scholar]
- Langelaan D. N.; Bebbington E. M.; Reddy T.; Rainey J. K. Structural insight into G-protein coupled receptor binding by apelin. Biochemistry 2009, 48 (3), 537–548. 10.1021/bi801864b. [DOI] [PubMed] [Google Scholar]
- Lee D. K.; Saldivia V. R.; Nguyen T.; Cheng R.; George S. R.; O’Dowd B. F. Modification of the terminal residue of apelin-13 antagonizes its hypotensive action. Endocrinology 2005, 146 (1), 231–236. 10.1210/en.2004-0359. [DOI] [PubMed] [Google Scholar]
- Kotanidou E. P.; Kalinderi K.; Kyrgios I.; Efraimidou S.; Fidani L.; Papadopoulou-Alataki E.; Eboriadou-Petikopoulou M.; Galli-Tsinopoulou A. Apelin and G212A apelin receptor gene polymorphism in obese and diabese youth. Pediatr Obes 2015, 10 (3), 213–219. 10.1111/ijpo.251. [DOI] [PubMed] [Google Scholar]
- Ringstrom C.; Nitert M. D.; Bennet H.; Fex M.; Valet P.; Rehfeld J. F.; Friis-Hansen L.; Wierup N. Apelin is a novel islet peptide. Regul. Pept. 2010, 162 (1–3), 44–51. 10.1016/j.regpep.2010.03.005. [DOI] [PubMed] [Google Scholar]
- Bertrand C.; Valet P.; Castan-Laurell I. Apelin and energy metabolism. Front Physiol 2015, 6, 115. 10.3389/fphys.2015.00115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berta J.; Kenessey I.; Dobos J.; Tovari J.; Klepetko W.; Jan Ankersmit H.; Hegedus B.; Renyi-Vamos F.; Varga J.; Lorincz Z.; Paku S.; Ostoros G.; Rozsas A.; Timar J.; Dome B. Apelin expression in human non-small cell lung cancer: role in angiogenesis and prognosis. J. Thorac Oncol 2010, 5 (8), 1120–1129. 10.1097/JTO.0b013e3181e2c1ff. [DOI] [PubMed] [Google Scholar]
- Hall C.; Ehrlich L.; Venter J.; O’Brien A.; White T.; Zhou T.; Dang T.; Meng F.; Invernizzi P.; Bernuzzi F.; Alpini G.; Lairmore T. C.; Glaser S. Inhibition of the apelin/apelin receptor axis decreases cholangiocarcinoma growth. Cancer Lett. 2017, 386, 179–188. 10.1016/j.canlet.2016.11.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng M.; Yao G.; Yu H.; Qing Y.; Wang K. Tumor apelin, not serum apelin, is associated with the clinical features and prognosis of gastric cancer. BMC Cancer 2016, 16 (1), 794. 10.1186/s12885-016-2815-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoffmann M.; Fiedor E.; Ptak A. Bisphenol A and its derivatives tetrabromobisphenol A and tetrachlorobisphenol A induce apelin expression and secretion in ovarian cancer cells through a peroxisome proliferator-activated receptor gamma-dependent mechanism. Toxicol. Lett. 2017, 269, 15–22. 10.1016/j.toxlet.2017.01.006. [DOI] [PubMed] [Google Scholar]
- Szokodi I.; Tavi P.; Foldes G.; Voutilainen-Myllyla S.; Ilves M.; Tokola H.; Pikkarainen S.; Piuhola J.; Rysa J.; Toth M.; Ruskoaho H. Apelin, the novel endogenous ligand of the orphan receptor APJ, regulates cardiac contractility. Circ. Res. 2002, 91 (5), 434–440. 10.1161/01.RES.0000033522.37861.69. [DOI] [PubMed] [Google Scholar]
- Tatemoto K.; Takayama K.; Zou M. X.; Kumaki I.; Zhang W.; Kumano K.; Fujimiya M. The novel peptide apelin lowers blood pressure via a nitric oxide-dependent mechanism. Regul. Pept. 2001, 99 (2–3), 87–92. 10.1016/S0167-0115(01)00236-1. [DOI] [PubMed] [Google Scholar]
- Japp A. G.; Cruden N. L.; Amer D. A.; Li V. K.; Goudie E. B.; Johnston N. R.; Sharma S.; Neilson I.; Webb D. J.; Megson I. L.; Flapan A. D.; Newby D. E. Vascular effects of apelin in vivo in man. J. Am. Coll Cardiol 2008, 52 (11), 908–913. 10.1016/j.jacc.2008.06.013. [DOI] [PubMed] [Google Scholar]
- Japp A. G.; Cruden N. L.; Barnes G.; van Gemeren N.; Mathews J.; Adamson J.; Johnston N. R.; Denvir M. A.; Megson I. L.; Flapan A. D.; Newby D. E. Acute cardiovascular effects of apelin in humans: potential role in patients with chronic heart failure. Circulation 2010, 121 (16), 1818–1827. 10.1161/CIRCULATIONAHA.109.911339. [DOI] [PubMed] [Google Scholar]
- Zhong J. C.; Zhang Z. Z.; Wang W.; McKinnie S. M. K.; Vederas J. C.; Oudit G. Y. Targeting the apelin pathway as a novel therapeutic approach for cardiovascular diseases. Biochim Biophys Acta Mol. Basis Dis 2017, 1863 (8), 1942–1950. 10.1016/j.bbadis.2016.11.007. [DOI] [PubMed] [Google Scholar]
- de Oliveira A. A.; Vergara A.; Wang X.; Vederas J. C.; Oudit G. Y. Apelin pathway in cardiovascular, kidney, and metabolic diseases: Therapeutic role of apelin analogs and apelin receptor agonists. Peptides 2022, 147, 170697. 10.1016/j.peptides.2021.170697. [DOI] [PubMed] [Google Scholar]
- Wysocka M. B.; Pietraszek-Gremplewicz K.; Nowak D. The Role of Apelin in Cardiovascular Diseases, Obesity and Cancer. Front Physiol 2018, 9, 557. 10.3389/fphys.2018.00557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu S.; Tsao P. S.; Yue P. Apelin and insulin resistance: another arrow for the quiver?. J. Diabetes 2011, 3 (3), 225–231. 10.1111/j.1753-0407.2011.00132.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kurowska P.; Barbe A.; Rozycka M.; Chmielinska J.; Dupont J.; Rak A. Apelin in Reproductive Physiology and Pathology of Different Species: A Critical Review. Int. J. Endocrinol 2018, 2018, 9170480. 10.1155/2018/9170480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson J. A.; Kim S. H.; Jiang J.; Phillips M.; Schumacher W. A.; Bostwick J. S.; Gargalovic P. S.; Onorato J. M.; Luk C. E.; Generaux C.; He Y.; Chen X. Q.; Xu C.; Galella M. A.; Wang T.; Gordon D. A.; Wexler R. R.; Finlay H. J. Discovery of a Hydroxypyridinone APJ Receptor Agonist as a Clinical Candidate. J. Med. Chem. 2021, 64 (6), 3086–3099. 10.1021/acs.jmedchem.0c01878. [DOI] [PubMed] [Google Scholar]
- Ason B.; Chen Y.; Guo Q.; Hoagland K. M.; Chui R. W.; Fielden M.; Sutherland W.; Chen R.; Zhang Y.; Mihardja S. Cardiovascular response to small-molecule APJ activation. JCI Insight 2020, 5 (8), e132898. 10.1172/jci.insight.132898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pinilla-Vera M.; Hahn V. S.; Kass D. A. Leveraging Signaling Pathways to Treat Heart Failure With Reduced Ejection Fraction. Circ. Res. 2019, 124 (11), 1618–1632. 10.1161/CIRCRESAHA.119.313682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vickers C.; Hales P.; Kaushik V.; Dick L.; Gavin J.; Tang J.; Godbout K.; Parsons T.; Baronas E.; Hsieh F.; Acton S.; Patane M.; Nichols A.; Tummino P. Hydrolysis of biological peptides by human angiotensin-converting enzyme-related carboxypeptidase. J. Biol. Chem. 2002, 277 (17), 14838–14843. 10.1074/jbc.M200581200. [DOI] [PubMed] [Google Scholar]
- Kehoe K.; Van Elzen R.; Verkerk R.; Sim Y.; Van der Veken P.; Lambeir A. M.; De Meester I. Prolyl carboxypeptidase purified from human placenta: its characterization and identification as an apelin-cleaving enzyme. Biochim. Biophys. Acta 2016, 1864 (11), 1481–1488. 10.1016/j.bbapap.2016.07.004. [DOI] [PubMed] [Google Scholar]
- Murza A.; Belleville K.; Longpre J. M.; Sarret P.; Marsault E. Stability and degradation patterns of chemically modified analogs of apelin-13 in plasma and cerebrospinal fluid. Biopolymers 2014, 102 (4), 297–303. 10.1002/bip.22498. [DOI] [PubMed] [Google Scholar]
- McKinnie S. M.; Fischer C.; Tran K. M.; Wang W.; Mosquera F.; Oudit G. Y.; Vederas J. C. The Metalloprotease Neprilysin Degrades and Inactivates Apelin Peptides. Chembiochem 2016, 17 (16), 1495–1498. 10.1002/cbic.201600244. [DOI] [PubMed] [Google Scholar]
- Read C.; Nyimanu D.; Williams T. L.; Huggins D. J.; Sulentic P.; Macrae R. G. C.; Yang P.; Glen R. C.; Maguire J. J.; Davenport A. P. International Union of Basic and Clinical Pharmacology. CVII. Structure and Pharmacology of the Apelin Receptor with a Recommendation that Elabela/Toddler Is a Second Endogenous Peptide Ligand. Pharmacol Rev. 2019, 71 (4), 467–502. 10.1124/pr.119.017533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Masri B.; Morin N.; Pedebernade L.; Knibiehler B.; Audigier Y. The apelin receptor is coupled to Gi1 or Gi2 protein and is differentially desensitized by apelin fragments. J. Biol. Chem. 2006, 281 (27), 18317–18326. 10.1074/jbc.M600606200. [DOI] [PubMed] [Google Scholar]
- Kang Y.; Kim J.; Anderson J. P.; Wu J.; Gleim S. R.; Kundu R. K.; McLean D. L.; Kim J. D.; Park H.; Jin S. W.; Hwa J.; Quertermous T.; Chun H. J. Apelin-APJ signaling is a critical regulator of endothelial MEF2 activation in cardiovascular development. Circ. Res. 2013, 113 (1), 22–31. 10.1161/CIRCRESAHA.113.301324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang Y. M.; Kuen D. S.; Chung Y.; Kurose H.; Kim S. G. Galpha(12/13) signaling in metabolic diseases. Exp Mol. Med. 2020, 52 (6), 896–910. 10.1038/s12276-020-0454-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gurevich V. V.; Gurevich E. V. GPCR Signaling Regulation: The Role of GRKs and Arrestins. Front Pharmacol 2019, 10, 125. 10.3389/fphar.2019.00125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Besserer-Offroy E.; Berube P.; Cote J.; Murza A.; Longpre J. M.; Dumaine R.; Lesur O.; Auger-Messier M.; Leduc R.; Marsault E.; Sarret P. The hypotensive effect of activated apelin receptor is correlated with beta-arrestin recruitment. Pharmacol. Res. 2018, 131, 7–16. 10.1016/j.phrs.2018.02.032. [DOI] [PubMed] [Google Scholar]
- Helker C. S.; Eberlein J.; Wilhelm K.; Sugino T.; Malchow J.; Schuermann A.; Baumeister S.; Kwon H. B.; Maischein H. M.; Potente M. Apelin signaling drives vascular endothelial cells toward a pro-angiogenic state. eLife 2020, 9, e55589 10.7554/eLife.55589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li N.; Ma X.; Ban T.; Xu S.; Ma Y.; Ason B.; Hu L. A. Loss of APJ mediated beta-arrestin signalling improves high-fat diet induced metabolic dysfunction but does not alter cardiac function in mice. Biochem. J. 2020, 477 (17), 3313–3327. 10.1042/BCJ20200343. [DOI] [PubMed] [Google Scholar]
- Davenport A. P.; Scully C. C. G.; de Graaf C.; Brown A. J. H.; Maguire J. J. Advances in therapeutic peptides targeting G protein-coupled receptors. Nat. Rev. Drug Discov 2020, 19 (6), 389–413. 10.1038/s41573-020-0062-z. [DOI] [PubMed] [Google Scholar]
- Rajagopal S.; Rajagopal K.; Lefkowitz R. J. Teaching old receptors new tricks: biasing seven-transmembrane receptors. Nat. Rev. Drug Discov 2010, 9 (5), 373–386. 10.1038/nrd3024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang P.; Maguire J. J.; Davenport A. P. Apelin, Elabela/Toddler, and biased agonists as novel therapeutic agents in the cardiovascular system. Trends Pharmacol. Sci. 2015, 36 (9), 560–567. 10.1016/j.tips.2015.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brame A. L.; Maguire J. J.; Yang P.; Dyson A.; Torella R.; Cheriyan J.; Singer M.; Glen R. C.; Wilkinson I. B.; Davenport A. P. Design, characterization, and first-in-human study of the vascular actions of a novel biased apelin receptor agonist. Hypertension 2015, 65 (4), 834–840. 10.1161/HYPERTENSIONAHA.114.05099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tran K.; Sainsily X.; Cote J.; Coquerel D.; Couvineau P.; Saibi S.; Haroune L.; Besserer-Offroy E.; Flynn-Robitaille J.; Resua Rojas M.; Murza A.; Longpre J. M.; Auger-Messier M.; Lesur O.; Bouvier M.; Marsault E.; Boudreault P. L.; Sarret P. Size-Reduced Macrocyclic Analogues of [Pyr(1)]-apelin-13 Showing Negative Galpha(12) Bias Still Produce Prolonged Cardiac Effects. J. Med. Chem. 2022, 65 (1), 531–551. 10.1021/acs.jmedchem.1c01708. [DOI] [PubMed] [Google Scholar]
- Murza A.; Sainsily X.; Cote J.; Bruneau-Cossette L.; Besserer-Offroy E.; Longpre J. M.; Leduc R.; Dumaine R.; Lesur O.; Auger-Messier M.; Sarret P.; Marsault E. Structure-activity relationship of novel macrocyclic biased apelin receptor agonists. Org. Biomol Chem. 2017, 15 (2), 449–458. 10.1039/C6OB02247B. [DOI] [PubMed] [Google Scholar]
- Tran K.; Murza A.; Sainsily X.; Coquerel D.; Cote J.; Belleville K.; Haroune L.; Longpre J. M.; Dumaine R.; Salvail D.; Lesur O.; Auger-Messier M.; Sarret P.; Marsault E. A Systematic Exploration of Macrocyclization in Apelin-13: Impact on Binding, Signaling, Stability, and Cardiovascular Effects. J. Med. Chem. 2018, 61 (6), 2266–2277. 10.1021/acs.jmedchem.7b01353. [DOI] [PubMed] [Google Scholar]
- Tran K.; Van Den Hauwe R.; Sainsily X.; Couvineau P.; Cote J.; Simard L.; Echevarria M.; Murza A.; Serre A.; Theroux L.; Saibi S.; Haroune L.; Longpre J. M.; Lesur O.; Auger-Messier M.; Spino C.; Bouvier M.; Sarret P.; Ballet S.; Marsault E. Constraining the Side Chain of C-Terminal Amino Acids in Apelin-13 Greatly Increases Affinity, Modulates Signaling, and Improves the Pharmacokinetic Profile. J. Med. Chem. 2021, 64 (9), 5345–5364. 10.1021/acs.jmedchem.0c01941. [DOI] [PubMed] [Google Scholar]
- Murza A.; Besserer-Offroy E.; Cote J.; Berube P.; Longpre J. M.; Dumaine R.; Lesur O.; Auger-Messier M.; Leduc R.; Sarret P.; Marsault E. C-Terminal modifications of apelin-13 significantly change ligand binding, receptor signaling, and hypotensive action. J. Med. Chem. 2015, 58 (5), 2431–2440. 10.1021/jm501916k. [DOI] [PubMed] [Google Scholar]
- Fernandez K. X.; Fischer C.; Vu J.; Gheblawi M.; Wang W.; Gottschalk S.; Iturrioz X.; Llorens-Cortes C.; Oudit G. Y.; Vederas J. C. Metabolically stable apelin-analogues, incorporating cyclohexylalanine and homoarginine, as potent apelin receptor activators. RSC Med. Chem. 2021, 12 (8), 1402–1413. 10.1039/D1MD00120E. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Myers M. C.; Bilder D. M.; Cavallaro C. L.; Chao H. J.; Su S.; Burford N. T.; Nayeem A.; Wang T.; Yan M.; Langish R. A.; Dabros M.; Li Y. X.; Rose A. V.; Behnia K.; Onorato J. M.; Gargalovic P. S.; Wexler R. R.; Lawrence R. M. Discovery and SAR of aryl hydroxy pyrimidinones as potent small molecule agonists of the GPCR APJ.. Bioorg. Med. Chem. Lett. 2020, 30 (7), 126955. 10.1016/j.bmcl.2020.126955. [DOI] [PubMed] [Google Scholar]
- Narayanan S.; Vasukuttan V.; Rajagopal S.; Maitra R.; Runyon S. P. Identification of potent pyrazole based APELIN receptor (APJ) agonists. Bioorg. Med. Chem. 2020, 28 (4), 115237. 10.1016/j.bmc.2019.115237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pi Z.; Johnson J. A.; Meng W.; Phillips M.; Schumacher W. A.; Bostwick J. S.; Gargalovic P. S.; Onorato J. M.; Generaux C. N.; Wang T.; He Y.; Gordon D. A.; Wexler R. R.; Finlay H. J. Identification of 6-Hydroxypyrimidin-4(1H)-one-3-carboxamides as Potent and Orally Active APJ Receptor Agonists. ACS Med. Chem. Lett. 2021, 12 (11), 1766–1772. 10.1021/acsmedchemlett.1c00385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meng W.; Pi Z.; Brigance R.; Rossi K. A.; Schumacher W. A.; Bostwick J. S.; Gargalovic P. S.; Onorato J. M.; Luk C. E.; Generaux C. N.; Wang T.; Wexler R. R.; Finlay H. J. Identification of a Hydroxypyrimidinone Compound (21) as a Potent APJ Receptor Agonist for the Potential Treatment of Heart Failure. J. Med. Chem. 2021, 64 (24), 18102–18113. 10.1021/acs.jmedchem.1c01504. [DOI] [PubMed] [Google Scholar]
- Gargalovic P.; Wong P.; Onorato J.; Finlay H.; Wang T.; Yan M.; Crain E.; St-Onge S.; Heroux M.; Bouvier M.; Xu C.; Chen X. Q.; Generaux C.; Lawrence M.; Wexler R.; Gordon D. In Vitro and In Vivo Evaluation of a Small-Molecule APJ (Apelin Receptor) Agonist, BMS-986224, as a Potential Treatment for Heart Failure. Circ Heart Fail 2021, 14 (3), e007351 10.1161/CIRCHEARTFAILURE.120.007351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Narayanan S.; Wang S.; Vasukuttan V.; Vyas Devambatla R. K.; Dai D.; Jin C.; Snyder R.; Laudermilk L.; Runyon S. P.; Maitra R. Pyrazole Agonist of the Apelin Receptor Improves Symptoms of Metabolic Syndrome in Mice. J. Med. Chem. 2021, 64 (6), 3006–3025. 10.1021/acs.jmedchem.0c01448. [DOI] [PubMed] [Google Scholar]
- Trivedi A.; Kiang Y. H.; Saw R. E.; Cheng G. C.; Mather O.; Vega S.; Hellawell J.; Lee E. Evaluation of the Pharmacokinetics and Safety of AMG 986 Tablet and Capsule Formulations in Healthy Adult Subjects: A Phase I, Open-Label, Randomized Study. Drugs R D 2022, 22 (2), 147–154. 10.1007/s40268-022-00388-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Narayanan S.; Dai D.; Vyas Devambatla R. K.; Albert V.; Bruneau-Latour N.; Vasukuttan V.; Ciblat S.; Rehder K.; Runyon S. P.; Maitra R. Synthesis and characterization of an orally bioavailable small molecule agonist of the apelin receptor. Bioorg. Med. Chem. 2022, 66, 116789. 10.1016/j.bmc.2022.116789. [DOI] [PubMed] [Google Scholar]
- McCorvy J. D.; Butler K. V.; Kelly B.; Rechsteiner K.; Karpiak J.; Betz R. M.; Kormos B. L.; Shoichet B. K.; Dror R. O.; Jin J.; Roth B. L. Structure-inspired design of beta-arrestin-biased ligands for aminergic GPCRs. Nat. Chem. Biol. 2018, 14 (2), 126–134. 10.1038/nchembio.2527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wacker D.; Wang S.; McCorvy J. D.; Betz R. M.; Venkatakrishnan A. J.; Levit A.; Lansu K.; Schools Z. L.; Che T.; Nichols D. E.; Shoichet B. K.; Dror R. O.; Roth B. L. Crystal Structure of an LSD-Bound Human Serotonin Receptor. Cell 2017, 168 (3), 377–389. 10.1016/j.cell.2016.12.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma Y.; Yue Y.; Ma Y.; Zhang Q.; Zhou Q.; Song Y.; Shen Y.; Li X.; Ma X.; Li C.; Hanson M. A.; Han G. W.; Sickmier E. A.; Swaminath G.; Zhao S.; Stevens R. C.; Hu L. A.; Zhong W.; Zhang M.; Xu F. Structural Basis for Apelin Control of the Human Apelin Receptor. Structure 2017, 25 (6), 858–866. 10.1016/j.str.2017.04.008. [DOI] [PubMed] [Google Scholar]
- Ceraudo E.; Galanth C.; Carpentier E.; Banegas-Font I.; Schonegge A. M.; Alvear-Perez R.; Iturrioz X.; Bouvier M.; Llorens-Cortes C. Biased signaling favoring gi over beta-arrestin promoted by an apelin fragment lacking the C-terminal phenylalanine. J. Biol. Chem. 2014, 289 (35), 24599–24610. 10.1074/jbc.M113.541698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Juhl C.; Els-Heindl S.; Schonauer R.; Redlich G.; Haaf E.; Wunder F.; Riedl B.; Burkhardt N.; Beck-Sickinger A. G.; Bierer D. Development of Potent and Metabolically Stable APJ Ligands with High Therapeutic Potential. ChemMedChem. 2016, 11 (21), 2378–2384. 10.1002/cmdc.201600307. [DOI] [PubMed] [Google Scholar]
- Hori T.; Okuno T.; Hirata K.; Yamashita K.; Kawano Y.; Yamamoto M.; Hato M.; Nakamura M.; Shimizu T.; Yokomizo T.; Miyano M.; Yokoyama S. Na(+)-mimicking ligands stabilize the inactive state of leukotriene B(4) receptor BLT1. Nat. Chem. Biol. 2018, 14 (3), 262–269. 10.1038/nchembio.2547. [DOI] [PubMed] [Google Scholar]
- Katritch V.; Fenalti G.; Abola E. E.; Roth B. L.; Cherezov V.; Stevens R. C. Allosteric sodium in class A GPCR signaling. Trends Biochem. Sci. 2014, 39 (5), 233–244. 10.1016/j.tibs.2014.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bihoreau C.; Monnot C.; Davies E.; Teutsch B.; Bernstein K. E.; Corvol P.; Clauser E. Mutation of Asp74 of the rat angiotensin II receptor confers changes in antagonist affinities and abolishes G-protein coupling. Proc. Natl. Acad. Sci. U. S. A. 1993, 90 (11), 5133–5137. 10.1073/pnas.90.11.5133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonde M. M.; Hansen J. T.; Sanni S. J.; Haunso S.; Gammeltoft S.; Lyngso C.; Hansen J. L. Biased signaling of the angiotensin II type 1 receptor can be mediated through distinct mechanisms. PLoS One 2010, 5 (11), e14135 10.1371/journal.pone.0014135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Veeraswamy V.; Goswami G.; Mukherjee S.; Ghosh K.; Saha M. L.; Sengupta A.; Ghorai M. K. Memory of Chirality Concept in Asymmetric Intermolecular Michael Addition of alpha-Amino Ester Enolates to Enones and Nitroalkenes. J. Org. Chem. 2018, 83 (3), 1106–1115. 10.1021/acs.joc.7b02315. [DOI] [PubMed] [Google Scholar]
- Liu S.; Yosief H. O.; Dai L.; Huang H.; Dhawan G.; Zhang X.; Muthengi A. M.; Roberts J.; Buckley D. L.; Perry J. A.; Wu L.; Bradner J. E.; Qi J.; Zhang W. Structure-Guided Design and Development of Potent and Selective Dual Bromodomain 4 (BRD4)/Polo-like Kinase 1 (PLK1) Inhibitors. J. Med. Chem. 2018, 61 (17), 7785–7795. 10.1021/acs.jmedchem.8b00765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nicolaou K. C.; Estrada A. A.; Zak M.; Lee S. H.; Safina B. S. A mild and selective method for the hydrolysis of esters with trimethyltin hydroxide. Angew. Chem., Int. Ed. Engl. 2005, 44 (9), 1378–1382. 10.1002/anie.200462207. [DOI] [PubMed] [Google Scholar]
- Vassiliou S.; Weglarz-Tomczak E.; Berlicki L.; Pawelczak M.; Nocek B.; Mulligan R.; Joachimiak A.; Mucha A. Structure-guided, single-point modifications in the phosphinic dipeptide structure yield highly potent and selective inhibitors of neutral aminopeptidases. J. Med. Chem. 2014, 57 (19), 8140–8151. 10.1021/jm501071f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Teixido M.; Albericio F.; Giralt E. Solid-phase synthesis and characterization of N-methyl-rich peptides. J. Pept Res. 2005, 65 (2), 153–166. 10.1111/j.1399-3011.2004.00213.x. [DOI] [PubMed] [Google Scholar]
- Vogt W. Oxidation of methionyl residues in proteins: tools, targets, and reversal. Free Radic Biol. Med. 1995, 18 (1), 93–105. 10.1016/0891-5849(94)00158-G. [DOI] [PubMed] [Google Scholar]
- Nepali K.; Lee H. Y.; Liou J. P. Nitro-Group-Containing Drugs. J. Med. Chem. 2019, 62 (6), 2851–2893. 10.1021/acs.jmedchem.8b00147. [DOI] [PubMed] [Google Scholar]
- Murza A.; Parent A.; Besserer-Offroy E.; Tremblay H.; Karadereye F.; Beaudet N.; Leduc R.; Sarret P.; Marsault E. Elucidation of the structure-activity relationships of apelin: influence of unnatural amino acids on binding, signaling, and plasma stability. ChemMedChem. 2012, 7 (2), 318–325. 10.1002/cmdc.201100492. [DOI] [PubMed] [Google Scholar]
- Cheng Y.; Prusoff W. H. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50% inhibition (I50) of an enzymatic reaction. Biochem. Pharmacol. 1973, 22 (23), 3099–3108. 10.1016/0006-2952(73)90196-2. [DOI] [PubMed] [Google Scholar]
- Van der Poorten O.; Van Den Hauwe R.; Eiselt E.; Betti C.; Guillemyn K.; Chung N. N.; Halle F.; Bihel F.; Schiller P. W.; Tourwe D.; Sarret P.; Gendron L.; Ballet S. chi-Space Screening of Dermorphin-Based Tetrapeptides through Use of Constrained Arylazepinone and Quinolinone Scaffolds. ACS Med. Chem. Lett. 2017, 8 (11), 1177–1182. 10.1021/acsmedchemlett.7b00347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van der Poorten O.; Legrand B.; Vezenkov L. L.; Garcia-Pindado J.; Bettache N.; Knuhtsen A.; Pedersen D. S.; Sanchez-Navarro M.; Martinez J.; Teixido M.; Garcia M.; Tourwe D.; Amblard M.; Ballet S. Indoloazepinone-Constrained Oligomers as Cell-Penetrating and Blood-Brain-Barrier-Permeating Compounds. Chembiochem 2018, 19 (7), 696–705. 10.1002/cbic.201700678. [DOI] [PubMed] [Google Scholar]
- Murza A.; Sainsily X.; Coquerel D.; Cote J.; Marx P.; Besserer-Offroy E.; Longpre J. M.; Laine J.; Reversade B.; Salvail D.; Leduc R.; Dumaine R.; Lesur O.; Auger-Messier M.; Sarret P.; Marsault E. Discovery and Structure-Activity Relationship of a Bioactive Fragment of ELABELA that Modulates Vascular and Cardiac Functions. J. Med. Chem. 2016, 59 (7), 2962–2972. 10.1021/acs.jmedchem.5b01549. [DOI] [PubMed] [Google Scholar]
- Zimmerman B.; Beautrait A.; Aguila B.; Charles R.; Escher E.; Claing A.; Bouvier M.; Laporte S. A. Differential beta-arrestin-dependent conformational signaling and cellular responses revealed by angiotensin analogs. Sci. Signal 2012, 5 (221), ra33. 10.1126/scisignal.2002522. [DOI] [PubMed] [Google Scholar]
- Gales C.; Van Durm J. J.; Schaak S.; Pontier S.; Percherancier Y.; Audet M.; Paris H.; Bouvier M. Probing the activation-promoted structural rearrangements in preassembled receptor-G protein complexes. Nat. Struct Mol. Biol. 2006, 13 (9), 778–786. 10.1038/nsmb1134. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.






