Abstract
NMDA receptors mediate glutamatergic neurotransmission and are therapeutic targets due to their involvement in a variety of psychiatric and neurological disorders. Here, we describe the design and synthesis of a series of (R)-3-(5-fyranyl)carboxamido-2-aminopropanoic acid analogs 8a-s as agonists at the glycine (Gly) binding site in the GluN1 subunit, but not GluN3 subunits, of NMDA receptors. These novel analogs display high variation in potencies and agonist efficacies among the NMDA receptor subtypes (GluN1/2A-D) in a manner dependent on the GluN2 subunit. Notably, compound 8p is identified as a potent partial agonist at GluN1/2C (EC50 = 0.074 μM) with agonist efficacy of 28% relative to activation by Gly and virtually no agonist activity at GluN1/2A, GluN1/2B and GluN1/2D. Thus, these novel agonists can modulate the activity of specific NMDA receptor subtypes by replacing the full endogenous agonists Gly or d-serine (d-Ser), thereby providing new opportunities in the development of novel therapeutic agents.
INTRODUCTION
L-Glutamate (Glu, 1, Figure 1) is the major excitatory neurotransmitter in the central nervous system, and is involved in most aspects of normal brain function including cognition, memory and learning.1 Glutamatergic neurotransmission is mediated by ionotropic Glu receptors (iGluRs) that are ligand-gated ion channels2, 3 and metabotropic Glu receptors (mGluRs) that are G protein-coupled receptors (GPCRs).4–6 The iGluRs are divided into N-methyl-d-aspartic acid (NMDA) receptors, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, and kainate receptors.2, 3 While AMPA and kainate receptors only bind Glu, the activation of NMDA receptors requires the simultaneous binding of both Glu and Gly or d-Ser.2, 3 However, NMDA receptors mainly rely on synaptic release of Glu for activation, since Gly (or d-Ser) is present at concentrations estimated at 0.5–10 μM7–10, which will produce partial to full occupancy at GluN1 subunits11, 12. NMDA receptors have been implicated in several psychiatric and neurological diseases, including Alzheimer’s disease, Parkinson’s disease, schizophrenia, epilepsy, depression and neuropathic pain.2, 3, 13–19
Figure 1.
Chemical structures of L-Glu and agonists targeting the NMDA receptor Gly binding site.
NMDA receptors are tetrameric subunit complexes composed of two Gly-binding GluN1, and two Glu-binding GluN2 subunits (GluN2A-D) or two Gly-binding GluN3 subunits (GluN3A-B).2, 3 The GluN2 subunits have high sequence identity, and their Glu binding sites are highly conserved.2, 3 For this reason, the development of fully subtype-selective NMDA receptor ligands targeting the Glu binding site is profoundly challenging and has thus far been unsuccessful. Since synaptic NMDA receptors depend on Glu release for activation and the strength of Glu activation can be modulated by GluN1 agonists, therapeutic strategies to target the GluN1 agonist site have received considerable attention and have led to clinical trials with a number of Gly site ligands.20–23
The endogenous agonists for GluN1, Gly (2, Figure 1) and d-Ser (3), are considered full agonists at GluN1/2A-D subtypes.11, 12 d-Cycloserine (DCS, 4), a Gly site agonist,24 is a partial agonist at GluN1/2A, GluN1/2B, and GluN1/2D receptors with lower maximal response compared to Gly (Rmax), whereas DCS has a higher Rmax at GluN1/2C receptors compared to Gly (i.e. DCS is a superagonist at GluN1/2C).25–27 Administration of DCS can enhance extinction of fear in both rodents and humans, and this effect might be relevant in the treatment of some psychiatric disorders.28–30 (R)-2-Amino-3-(4-(2-ethylphenyl)-1H-indole-2-carboxamido)propanoic acid (AICP, 5) was reported by Urwyler et al. (2009) as a NMDA receptor Gly site agonist.31 Further investigation demonstrated superagonism at GluN1/2C (EC50 = 0.0017 μM, Rmax = 353%), full agonism at GluN1/2A (EC50 = 0.066 μM, Rmax = 100%), and partial agonist activity at GluN1/2B (EC50 = 0.025 μM, Rmax = 10%) and GluN1/2D (EC50 = 0.025 μM, Rmax = 27%).26 Compared to DCS, AICP can reduce the spike frequency and burst firing of nucleus reticularis of the thalamus (nRT) neurons in wild type, but not GluN2C knockout mice.32 The nRT neurons are enriched with GluN2C-containing NMDA receptors that are proposed to regulate corticothalamic and thalamocortical communication.32 Moreover, the administration of DCS or AICP into the globus pallidus externa (GPe) was shown to improve motor function in a mouse model of Parkinson’s disease.33 These observations demonstrate the potential for GluN2-specific Gly site agonists as modulators of NMDA receptors.
Apart from AICP, analogs 6 and 7 are GluN1/2C superagonists with moderate potencies (EC50 = 1.97 μM and 0.32 μM at GluN1/2C, respectively) and different agonist efficacy profiles across NMDA receptor subtypes compared to AICP.11
These results spurred our interest to develop new compound classes that would explore potency, efficacy, and GluN2 subunit-specific activity and facilitate further investigations of neuronal NMDA receptor subtypes. In this study, we maintained the essential α-amino acid part and amide part of AICP (5, Figure 2), whereas the N-acyl aromatic part was simplified to explore scaffold diversities. The fused phenyl ring was removed and the pyrrole ring was replaced by a furan ring, and substituted phenyl rings were introduced (8a-s). Thus, we exchanged the N-acyl aromatic scaffold in order to provide molecular flexibility that can enable new modes of agonist accommodation in the Gly binding pocket.
Figure 2.
Design of novel furanylamide derivatives as NMDA receptor Gly site agonists. R indicates substitutions on the phenyl ring.
RESULTS AND DISCUSSION
Chemistry
The synthesis of newly designed compounds 8a-p began with a Suzuki cross-coupling between 5-bromofuran-2-carbaldehyde 9 and a series of arylboronic acids/esters using the catalyst Pd(PPh3)4 and base Na2CO3 to give aldehydes 10d-p, which were then oxidized by AgNO3 to yield key acid intermediates 11d-p (Scheme 1). For the synthesis of carboxylic acid 11q-s with substitutions on the ortho-position of phenyl ring, the route commenced with diazotization of 2-bromoaniline 13 to give the aldehyde 10q. After Suzuki coupling with styrylboronic acid or 3-fluorostyrylboronic acid, the aldehyde 10r-s were produced, which was then oxidized by AgNO3 to give carboxylic acids 11q-s. The corresponding amides 12a-s were obtained by coupling of the corresponding carboxylic acid 11d-o, 11q-s or commercially available 11a-c and 11p with 3-amino-N-Boc-d-alanine. Carboxylic acid protection in this reaction would prevent byproduct formation,34 but also add extra steps with concomitant material loss, resulting in minimal improvement to the overall yield. The Boc groups of 12a-s were removed with TFA and the final pure products 8a-s were obtained after purification by prep-HPLC.
Scheme 1.
Synthesis route of analogs 8a-sa
aReagents and conditions: (i) arylboronic acid/ester, Pd(PPh3)4, Na2CO3, N2, Toluene, EtOH, H2O, 85°C; (ii) AgNO3, NaOH, EtOH, H2O, 55°C; (iii) 3-amino-N-Boc-d-alanine, EDCI, HOBt, NEt3, DCM, rt; (iv) TFA, DCM, (1:1 v/v), rt; (v) NaNO2, HCl, H2O, 0–5°C, then furfural, CuCl2, 0°C to rt; (vi) styrylboronic acid/3-fluorostyrylboronic acid, Pd(PPh3)4, Na2CO3, N2, Toluene, EtOH, H2O, 85°C.
Pharmacological Evaluation
The pharmacological properties of analogs 8a-s were evaluated using two-electrode voltage-clamp (TEVC) recordings of responses from recombinant NMDA receptor subtypes expressed in Xenopus oocytes (Table 1, Figure 3). None of the analogs display discernable agonist activity at GluN1/2B. However, all compounds activate the GluN1/2C subtype with potencies in the low micromolar to nanomolar range. The agonists display partial, full, or superagonism ranging from <10% to 157% relative to the maximal response compared to Gly (Rmax). In general, the compounds display a preference for GluN1/2C over the other three GluN1/2 NMDA receptor subtypes in terms of Rmax (Table 1).
Table 1.
Agonist Activities at Recombinant GluN1/2A-D NMDA Receptors
![]() |
|||||||||
---|---|---|---|---|---|---|---|---|---|
Compound | R | GluN1/2A | GluN1/2B | GluN1/2C | GluN1/2D | ||||
EC50 (μM) |
Rmax (%) |
EC50 (μM) |
Rmax (%) |
EC50 (μM) |
Rmax (%) |
EC50 (μM) |
Rmax (%) |
||
Gly | - | 0.99 | 100 | 0.24 | 100 | 0.21 | 100 | 0.091 | 100 |
d-Ser | - | 1.0 | 96 | 0.62 | 100 | 0.19 | 111 | 0.15 | 93 |
8a |
![]() |
4.3 | 14 | NRa | 1.7 | 44 | NRa | ||
8b |
![]() |
ND | NRa | 1.3 | 26 | NRa | |||
8c |
![]() |
4.8 | 22 | NRb | 2.9 | 77 | 4.3 | 16 | |
8d |
![]() |
0.13 | 24 | NRa | 0.040 | 36 | NRa | ||
8e |
![]() |
23 | 20 | NRb | 4.2 | 74 | 31 | 24 | |
8f |
![]() |
6.6 | 17 | NRb | 1.8 | 36 | 7.7 | 35 | |
8g |
![]() |
4.9 | 27 | NRb | 1.3 | 68 | 5.0 | 38 | |
8h |
![]() |
0.093 | 40 | NRb | 0.063 | 52 | 0.15 | 11 | |
8i |
![]() |
3.1 | 32 | NRb | 1.6 | 114 | 3.2 | 11 | |
8j |
![]() |
10 | 22 | NRb | 4.8 | 119 | NRb | ||
8k |
![]() |
0.18 | 29 | NRb | 0.11 | 53 | 0.23 | 12 | |
8l |
![]() |
ND | NRb | 3.9 | 17 | NDb | |||
8m |
![]() |
0.26 | 38 | NRb | 0.12 | 70 | 0.27 | 11 | |
8n |
![]() |
0.22 | 58 | NRc | 0.14 | 103 | 0.26 | 17 | |
8o |
![]() |
NRb | NRb | 4.0 | 157 | ND | |||
8p |
![]() |
ND | NRa | 0.074 | 28 | NRa | |||
8q |
![]() |
0.053 | 34 | NRa | 0.036 | 49 | 0.080 | 9 | |
8r |
![]() |
ND | NRa | 0.75 | 22 | ND | |||
8s |
![]() |
NRd | NRd | 5.8 | 16 | NRd |
EC50 values in the presence of 300 μM L-Glu measured using two-electrode voltage-clamp electrophysiology from Xenopus oocytes expressing recombinant rat NMDA receptor subtypes. Relative efficacy is the fitted maximal response to the agonist compared to the maximal response to 100 μM Gly in the same recording. Measurements of the full concentration-response data were limited by solubility for some compounds. Compound stock solutions were prepared in DMSO. The DMSO concentration was kept constant in all ligand solutions and never exceeded 0.5%. ND indicates that the EC50 could not be determined, but some agonist activity was observed. NR indicates less than 5% response at a 100μM, b 300μM, c 10 μM, or d 30 μM of the agonist. Data are from 3–14 oocytes. See Table S1 in the Supporting Information for sample size, Hill slopes, and standard deviations.
Figure 3.
(A-B) Representative recordings of 8k and 8n concentration-response data in the continuous presence of 300 μM L-Glu at recombinant GluN1/2A-D receptors measured using two-electrode voltage-clamp electrophysiology. (C) Concentration-response data measured using two-electrode voltage-clamp recordings for Gly and novel agonists with nanomolar potencies (8h, 8k, 8m, 8n, 8p, 8r, and 8s) at recombinant GluN1/2A-D receptors. The fitted maximal response to agonist is normalized to the maximal response to Gly measured in the same recording. Data are from 4–14 oocytes and data points are mean ± SEM. See Table 1 for values.
Analogs 8b, 8l, 8o, 8p, 8r and 8s are selective GluN1/2C agonists with little to no agonist activity at GluN1/2A, GluN1/2B, and GluN1/2D (Table 1). Specifically, analog 8o is a superagonist (EC50 = 4.0 μM, Rmax = 157%) at GluN1/2C, while analogs 8p (EC50 = 0.074 μM, Rmax = 28%) and 8r (EC50 = 0.75 μM, Rmax = 22%) are partial agonists. Analog 8h is a potent GluN1/2A (EC50 = 0.093 μM, Rmax = 40%), GluN1/2C (EC50 = 0.063 μM, Rmax = 52%), and GluN1/2D (EC50 = 0.15 μM, Rmax = 11%) partial agonist with no agonist activity at GluN1/2B. Compared to 8h, analog 8q shows improved agonist potency, but similar agonist efficacy at GluN1/2A (EC50 = 0.053 μM, Rmax = 34%), GluN1/2C (EC50 = 0.036 μM, Rmax = 49%), and GluN1/2D (EC50 = 0.080 μM, Rmax = 9%). Compounds 8k, 8m and 8n display pharmacological profiles similar to 8h and 8q, but with slightly lower potencies. The ortho-CF3 analog 8d is a partial agonist at GluN1/2A (EC50 = 0.13 μM, Rmax = 24%) and GluN1/2C (EC50 = 0.040 μM, Rmax = 36%) with no agonist activity at GluN1/2B and GluN1/2D.
The structure-activity relationship at GluN1/2A and GluN1/2C for chloro-substituted analogs 8a, 8g, 8m, and CF3-substitued analogs 8d and 8f demonstrates that the introduction of ortho-substitution on the phenyl ring improves agonist potency (Table 1, Figure 4). This observation is consistent with the potencies of other ortho-substituted analogs 8h, 8n, and 8q. Similar relationship between ortho-substitution and agonist potency is observed at GluN1/2D (Table 1, Figure 4). However, larger substitutions on the ortho-position for compounds 8o, 8s and 8r result in a decrease in potency at GluN1/2C and is not tolerated in GluN1/2A and GluN1/2D. In addition, compounds 8i, 8j and 8l with hydrophilic groups on the ortho-position only show moderate potencies, compared to 8m and 8n. Therefore, the hydrophobic and small ortho-substitutions might be more favorable for this series of analogs and thereby obtain high potency at GluN1/2A and GluN1/2C.
Figure 4.
LogEC50 values ± SEM for analogs with agonist activity are plotted for the indicated NMDA receptor subtypes. There is significant correlation for agonist potencies at GluN1/2A vs. GluN1/2C, GluN1/2D vs. GluN1/2C, and GluN1/2D vs. GluN1/2A. EC50 values are lognormally distributed35 and LogEC50 values were therefore used in the calculation of the Pearson correlation. See Table 1 for values.
There is a significant correlation for agonist potencies at GluN1/2A, GluN1/2C, and GluN1/2D (Figure 4). That is, the most potent GluN1/2C agonists are also the most potent GluN1/2A (Pearson r = 0.9856, N = 13, P < 0.05) and GluN1/2D agonists (Pearson r = 0.9780, N = 10, P < 0.05). Similarly, the most potent GluN1/2A agonists are also the most potent GluN1/2D agonists (Pearson r = 0.9964, N = 10, P < 0.05). Agonist efficacies are not displaying any significant correlation among the NMDA receptor subtypes (2A vs. 2C, Pearson r = 0.335, N = 13, P > 0.05; 2D vs. 2C, Pearson r = −0.231, N = 10, P > 0.05; 2A vs. 2D, Pearson r = −0.470, N = 10, P > 0.05). Interestingly, there is a significant inverse correlation between agonist potencies and efficacies at GluN1/2A (Pearson r = −0.6264, N = 13, P < 0.05) such that the least potent agonists tend to have the highest agonist efficacy. By contrast, there is a significant correlation between agonist potencies and efficacies at GluN1/2D (Pearson r = 0.6811, N = 10, P < 0.05) such that the most potent agonists tend to have the highest agonist efficacy. Unlike GluN1/2A and GluN1/2D, there is no significant correlation between agonist potencies and efficacies at GluN1/2C (Pearson r = 0.2333, N = 19, P > 0.05). The strong correlation for agonist potencies suggest that the ligand-receptor interactions are shared among the NMDA receptor subtypes, consistent with agonists binding to the Gly binding site in the conserved GluN1 subunit. On the other hand, the lack of correlation for agonist efficacies among NMDA receptor subtypes and the different trends for agonist potencies and efficacies at individual subtypes suggest that the mechanisms governing agonist efficacy is vastly different among NMDA receptor subtypes in a GluN2 subunit-specific manner.
The apparent lack of agonist activity of all analogs at GluN1/2B receptors could be due to binding without any agonist efficacy (i.e. competitive antagonism), binding with very low agonist efficacy that produces no discernable current response, or no binding to GluN1 at all in GluN1/2B receptors. To determine if the analogs are binding the GluN1 subunit in GluN1/2B receptors, we evaluated whether compounds 8h and 8p inhibit responses to Gly (Figure 5). As expected, the analogs alone (10 μM) in the continues presence of 300 μM L-Glu were unable to activate any current responses at GluN1/2B. However, both 8h and 8p (10 μM) inhibited responses to Gly (100 μM) by 47 ± 6% (n =12) and 57 ± 2% (n =12), respectively. Using the Cheng-Prusoff relationship36 and IC50 values of ~10 μM, these inhibitions correspond to estimated binding affinities (Ki) of ~0.1 μM for 8h and 8p at the Gly binding site in GluN1/2B receptors. These results further corroborate that the analogs bind to the conserved Gly binding site in the GluN1 subunit of GluN1/2A-D subtypes, but that agonist efficacies are controlled by the GluN2 subunit.
Figure 5.
(A) Representative recordings of responses to 8h and 8p in the absence and presence of Gly and in the continuous presence of 300 μM L-Glu at recombinant GluN1/2B receptors measured using two-electrode voltage-clamp electrophysiology. No responses to 8h and 8p (10 μM) are observed at GluN1/2B receptors, while both 8h and 8p inhibit responses to Gly. Data for each analog are from 12 oocytes and bar graphs are mean ± SEM. (B) Representative recordings of response to 8h and 8p in the absence and presence of Gly at recombinant GluN1FATL/3A and GluN1FATL/3B receptors that include F484A and T518L mutations in the GluN1 agonist binding site to prevent agonist binding to GluN1, thereby enabling evaluation of the pharmacology of GluN3 subunits. No responses to 8h and 8p are observed, and 8h and 8p did not inhibit responses to Gly. Data for each analog are from 12–13 oocytes and bar graphs are mean ± SEM.
The GluN3A and GluN3B NMDA receptor subunits also bind Gly,37, 38 but structural differences between GluN1 and GluN3 agonist binding sites can affect binding of Gly site agonists.39, 40 To determine if 8h and 8p bind GluN3A and GluN3B subunits, we evaluated these analogs at GluN1FATL/3A and GluN1FATL/3B receptors that include F484A and T518L mutations in the GluN1 agonist binding site (Figure 5). These mutations prevent agonist binding to GluN1 and enable evaluation of the pharmacology of GluN3 subunits.40 Furthermore, agonist binding to GluN1 triggers strong desensitization of GluN1/3 receptors, while agonist binding to GluN3 mediates activation.40–42 Both compounds 8h and 8p (10 μM) did not activate current responses by themselves and did not inhibit responses to Gly at GluN1FATL/3A and GluN1FATL/3B receptors (Figure 5). Thus, 8h and 8p (10 μM) selectively bind the agonist binding site in GluN1 over those in GluN3A and GluN3B subunits.
Molecular modeling
The structure-activity relationship was investigated in silico by induced-fit docking for compounds 8h, 8k and 8p into the Gly binding site of GluN1 using the crystal structure of the isolated GluN1/2A agonist binding domain (ABD) heterodimer (PDB ID: 5I57)43 as template (See Supporting Information for PDB files with docking poses).
According to induced-fit docking, the key interactions of the α-amino acid part for analogs 8h, 8k and 8p (Figure 6A–D) are conserved compared to Gly and d-Ser bound structures. The amide spacers of these analogs form hydrogen bonds and are accommodated in the narrow tunnel shaped by GluN1 residues T518, S688 and D732, which is consistent with previous molecular dynamics simulations of compound 6 docked into the GluN1 ABD.11 The substituted phenyl ring of these analogs interact with GluN1 residues Y692 and F754 and orient toward the interface of GluN1 and GluN2 subunits. These additional interactions with the surrounding residues might contribute to the improved binding affinity in the series. Furthermore, the docking poses are consistent with the observed selectivity of 8h and 8p for GluN1 over GluN3 subunits (Figure 5), since binding would be sterically occluded by GluN3A E871 and GluN3B E771 residues. The smaller GluN1 S756 at this position permits occupancy in the narrow tunnel toward the GluN1-GluN2 interface (Figure 6). Unlike 8h and 8k, the ortho-nitro substitution on the phenyl ring of 8p orients to the opposite direction and the phenyl ring shifts. Consequently, the para-Cl substituent on the phenyl ring is buried deeper into the GluN1-GluN2 subunit interface, thereby causing additional conformational change for surrounding residues. The distinct interactions formed by the ortho- and para- substitutions may, in part, mediate the remarkable GluN1/2C subtype preference of compound 8p.
Figure 6.
(A) Overview of GluN1/2A agonist binding domain heterodimer crystal structure (PDB ID: 5I57)43 with 8h docked into Gly binding site in GluN1. (B-D) Detailed views of induced-fit docking models for compounds 8h, 8k, and 8p in the GluN1/2A agonist binding domain structure. GluN1 and GluN2A are depicted as grey and blue cartoons, respectively. The pink oval highlights a possible steric clash that could influence agonist activity in a GluN2-specific manner, since GluN2A V783 is a non-conserved residue (Phe in GluN2B and Leu in GluN2C/D). See Supporting Information for PDB files with docking poses.
Considerable movement of GluN1 L654, V689, Y692, F753 and F754 is required for binding of all furanylamide analogs, while the position of the GluN2A ABD is not shifted compared to the Gly bound GluN1/2A structure. We hypothesize that this conformational change for residues in the GluN1 ABD influences interactions with non-conserved residues in GluN2 subunits (e.g. GluN2A V783), and that the precise orientation of substituents on the furan ring dictates agonist potency and efficacy. To determine if the non-conserved residues at the position of GluN2A V783 influences GluN2-specific agonist activity, we mutated this residue in GluN2A and GluN2B and evaluated responses to 8h and 8p (Figure 7A,B). Increasing the sidechain bulk from valine to leucine (as in GluN2C/2D) or phenylalanine (as in GluN2B) reduced the maximal response of 8h and 8p relative to Gly in GluN1/2A-V783L and GluN1/2A-V783F. By contrast, reducing sidechain bulk from phenylalanine to leucine (as in GluN2C/2D) and valine (as in GluN2A) did not significantly increase the maximal response to 8h or 8p relative to Gly in GluN1/2B-F784L and GluN1/2B-F784V. Consistent with smaller relative agonist efficacies, the potency of 8h was reduced at GluN1/2A-V783L (EC50 = 0.21 ± 0.07 μM, Rmax = 30 ± 2%, n = 6) and GluN1/2A-V783F (EC50 = 0.41 ± 0.06 μM, Rmax = 9 ± 1%, n = 4) compared to GluN1/2A wild type (EC50 = 0.11 ± 0.01 μM, Rmax = 45 ± 7%, n = 7) (Figure 7C). These results demonstrate that the non-conserved residue at position GluN2A V783 in the GluN1-GluN2 ABD dimer interface (Figure 6) can influence GluN2-specific agonist efficacy. However, the superagonism of the novel agonist series at GluN1/2C combined with lack of responses at GluN1/2B suggest that the GluN2-specific agonist efficacy is primarily governed by additional GluN2-dependent mechanisms that presumably include other regions of the ABD or other receptor domains.
Figure 7.
(A) Maximal responses to 8h or 8p (10 μM) at GluN1/2A wild type (WT) and mutated GluN1/2A-V783L and GluN1/2A-V783F receptors measured using two-electrode voltage-clamp electrophysiology in the continuous presence of 300 μM L-Glu. Data are mean ± SEM from 6–8 oocytes for 8h and 3–6 oocytes for 8p. For each analog, * indicates significant difference from wild type (P < 0.05, two-way ANOVA with Tukey’s multiple comparisons test). (B) Maximal responses to 8h or 8p (10 μM) at GluN1/2B wild type (WT) and mutated GluN1/2B receptors in the continuous presence of 300 μM L-Glu. Data are mean ± SEM from 4–6 oocytes for 8h and 3–6 oocytes for 8p. Agonist efficacies relative to Gly are not significantly changed at mutated GluN1/2B receptors compared to wild type (P > 0.05, two-way ANOVA with Tukey’s multiple comparisons test). (C) Concentration-response data for 8h at GluN1/2A wild type (EC50 = 0.11 ± 0.01 μM, Rmax = 45 ± 7%, n = 7), GluN1/2A-V783L (EC50 = 0.21 ± 0.07 μM, Rmax = 30 ± 2%, n = 6), and GluN1/2A-V783F (EC50 = 0.41 ± 0.06 μM, Rmax = 9 ± 1%, n = 4). The fitted maximal response to 8h is normalized to the maximal response to Gly measured in the same recording.
CONCLUSION
In the present work, a series of (R)-3-(5-furanyl)carboxamido-2-aminopropanoic acid derivatives (8a-s) were designed, synthesized and characterized in a functional assay at GluN1/2A-D NMDA receptor subtypes. Six analogs (8b, 8l, 8o, 8p, 8r and 8s) were identified as functionally selective GluN1/2C agonists with 8p and 8r displaying nanomolar potency. Compound 8h and 8q showed potent agonist activity at GluN1/2A, GluN1/2C and GluN1/2D, while the ortho-CF3 substituted compound 8d was shown to be a dual partial agonist at GluN1/2A and GluN1/2C subtypes (EC50 = 0.13 μM and 0.040 μM, respectively). Our in silico study suggested binding poses of 8h, 8k and 8p and illustrated possible structural mechanisms of agonist binding. However, more work is needed to appreciate the mechanisms by which these novel Gly site agonists engage in GluN2-specific interactions, thereby enabling variation in activity among NMDA receptor subtypes. The notable variation in potency, efficacy, and selectivity among these analogs at GluN1/2A-D receptor subtypes may provide new therapeutic strategies for neurological and psychiatric disorders, in which each NMDA receptor subtype can be distinctly modulated to balance therapeutic and potential adverse effects.
EXPERIMENTAL SECTION
Chemistry.
All reactions requiring inert conditions were performed in flame-dried glassware under an inert atmosphere of N2 using standard Schlenk and syringe-septum technique. Dichloromethane (DCM), tetrahydrofuran (THF) and dimethylformamide (DMF) were dried using a glass contour solvent system (SG Water, USA LCC). All other solvents for reaction, extraction, and purification were purchased in HPLC grade and used as received. Normal phase column chromatography was performed on silica gel 60 (Merck, 0.015–0.040 mm). Reactions and chromatography fractions were monitored by analytical thin layer chromatography (TLC), which was carried out using silica gel 60 F254 aluminum plates (Merck), and the compounds were visualized by using UV light (254 or 365 nm or both). All starting materials as well as the reagents were obtained from commercial sources and used without further purification.
Proton (1H) and carbon (13C) NMR spectra were recorded at 300 K in the solvent indicated. Bruker 400 and Bruker 600 spectrometers were operated at 400 and 600 MHz for proton and at 100 and 150 MHz for carbon nuclei. Chemical shifts (δ) are given in parts per million (ppm) using signals of residual nondeuterated solvent as internal standards.
LC/MS was performed on an Agilent 1200 system with a Zorbax Eclipse XBD-C18 column (4.6 mm × 50 mm) and UV detector using a linear gradient of the binary solvent system of H2O/MeCN/formic acid (A = 95/5/0.1 and B = 5/95/0.1) with a flow rate of 1 mL/min (0–100% B over 4 min, then 0.5 min 100% B) equipped with an API ion source (HP 1100 MSD).
Analytical HPLC was performed on a system consisting of an Ultimate 3000 pump and PDA detector, and a TSP AS-3000 autosampler with a Gemini-NX C18 column (4.6 mm × 250 mm), or on an Agilent 1260 system with Innoval C18 column (4.6 mm × 250 mm) equipped with a 254 nm UV detector using a linear gradient elution of the binary solvent system of H2O/MeCN/TFA (v/v/v; A = 95/5/0.1 and B = 5/95/0.1) with the concentration of B from 0% to 100% over 20 min with a flow rate of 1.0 mL/min. Prep-HPLC for the purification of final compounds was performed on an Ultimate 3000 system with a Gemini-NX C18 column (21 mm × 250 mm), or on a Waters instrument with an Innoval C18 column (21.2 mm × 250 mm) equipped with a 254 nm UV detector with the same binary solvent system with a flow rate of 20 mL/min. Purities of the tested compounds were determined by analytical HPLC to be >95%.
Procedure A: General procedure for Suzuki coupling between arylboronic acids and aryl halogenides. 5-bromofuran-2-carbaldehyde (175.0 mg, 1.0 mmol), appropriate arylboronic acid (1.5 mmol), Pd(PPh3)4 (57.8 mg, 0.05 mmol) and K2CO3 (414.6 mg, 3.0 mmol) in toluene/EtOH/H2O (10:3:2, v/v/v, 10 mL) were heated to 85°C overnight under a nitrogen atmosphere. The mixture was cooled to rt, diluted with EtOAc, filtered on Celite and evaporated to dryness. The crude was purified by column chromatography to afford the pure product.
Procedure B: General procedure for oxidation reaction from aldehyde to carboxyl acid. NaOH (320.0 mg, 4.0 mmol) was dissolved in H2O/EtOH (1:1, v/v, 12 mL) and heated to 60°C. AgNO3 (679.5 mg, 4.0 mmol) and the appropriate aldehyde (1.0 mmol) was added to the stirred solution. The reaction mixture was kept stirring for 2 h at 60°C and then filtered through Celite. The filtrate was acidified to pH~2 with 6 M HCl. The precipitate was then filtered off, washed with H2O and dried to afford the product without further purification.
Procedure C: General procedure for acylation and Boc deprotection. The appropriate acid (1.0 mmol), EDCI (191.7 mg, 1.0 mmol), HOBt (135.1 mg, 1.0 mmol), NEt3 (418.1 μL, 3.0 mmol) were dissolved in 10 mL anhydrous DCM and stirred for 20 min at rt. The solution of Boc-d-2,3-diaminopropionic acid (204.2 mg, 1.0 mmol) in 10 mL anhydrous DCM was added and stirred overnight at rt. The reaction mixture was washed with H2O (2 × 25 mL) and the organic phase was then dried with MgSO4. After concentration, the residue was treated with TFA/DCM (1:1, v/v, 10 mL). The resulting mixture was then stirred for another 4 h at rt and the solvent was evaporated to dryness. The final product was obtained as TFA salt after purification by prep-HPLC and 0.1% of TFA was added into the eluent. The yield and activity data were calculated considering the TFA salt.
5-(2-Ethylphenyl)furan-2-carbaldehyde (10h).
Procedure A. Yield: 75%. 1H NMR (600 MHz, methanol-d4) δ 1.21 (3H, t, J = 7.5 Hz), 2.87 (2H, q, J = 7.5 Hz), 6.83 (1H, d, J = 3.7 Hz), 7.27 (1H, td, J = 7.5, 1.6 Hz), 7.30–7.39 (2H, m), 7.50 (1H, d, J = 3.7 Hz), 7.66 (1H, dd, J = 7.7, 1.4 Hz), 9.59 (1H, s).
5-(2-(Methoxymethyl)phenyl)furan-2-carbaldehyde (10i).
Procedure A. Yield: 89%. 1H NMR (600 MHz, methanol-d4) δ 3.39 (3H, s), 4.58 (2H, s), 6.93 (1H, d, J = 3.7 Hz), 7.36–7.42 (2H, m), 7.47 (1H, d, J = 3.7 Hz), 7.49–7.53 (1H, m), 7.73– 7.81 (1H, m), 9.58 (1H, s).
2-(5-Formylfuran-2-yl)-N, N-dimethylbenzenesulfonamide (10j)
Procedure A. Yield: 93%. 1H NMR (400 MHz, methanol-d4) δ 2.62 (6H, s), 6.85–6.97 (1H, m), 7.34–7.45 (1H, m), 7.55–7.70 (3H, m), 7.91 (1H, d, J = 8.5 Hz), 9.56 (1H, s).
5-(Naphthalen-1-yl)furan-2-carbaldehyde (10k).
Procedure A. Yield: 63%. 1H NMR (600 MHz, chloroform-d) δ 6.87 (1H, d, J = 3.6 Hz), 7.36 (1H, d, J = 3.6 Hz), 7.46–7.50 (2H, m), 7.51–7.54 (1H, m), 7.80 (1H, d, J = 7.2 Hz), 7.83–7.88 (2H, m), 8.33 (1H, d, J = 8.4 Hz), 9.67 (1H, s).
5-(o-Tolyl)furan-2-carbaldehyde (10n).
Procedure A. Yield: 68%. 1H NMR (400 MHz, chloroform-d) δ 2.46 (3H, s), 6.65 (1H, d, J = 3.7 Hz), 7.18 (1H, d, J = 3.4 Hz), 7.21 (2H, d, J = 2.5 Hz), 7.25 (1H, d, J = 3.7 Hz), 7.70 (1H, dd, J = 7.3, 2.2 Hz), 9.58 (1H, s).
5-(2-(Benzyloxy)phenyl)furan-2-carbaldehyde (10o).
Procedure A. Yield: 85%. 1H NMR (400 MHz, chloroform-d) δ 5.13 (2H, s), 6.97– 7.03 (3H, m), 7.23–7.44 (7H, m), 8.00 (1H, dd, J = 7.8, 1.7 Hz), 9.56 (1H, s).
5-(2-bromophenyl)furan-2-carbaldehyde (10q).
2-Bromoaniline (109.0 μL, 1.0 mmol) was dissolved in 30 mL 6 M HCl. The solution was heated until all material is dissolved completely. The solution was then cooled to 0°C with ice bath and NaNO2 (75.89 mg, 1.1 mmol, 1.1 eq) in 10 mL H2O was added into the solution dropwise while keeping the temperature lower than 5°C during addition. The reaction was stirred for 10 min at 0°C. Furfural (99.39 μL, 1.2 mmol, 1.2 eq) and CuCl2 (40.34 mg, 0.3 mmol, 0.3 eq) dissolved in 10 mL H2O was added into the reaction mixture and stirred for 48h at rt. The precipitation was then filtered, washed with H2O and dried to afford the product without further purification. Yield: 11%. 1H NMR (600 MHz, chloroform-d) δ 9.69 (s, 1H), 7.93 (dd, J = 7.9, 1.7 Hz, 1H), 7.69 (dd, J = 8.0, 1.2 Hz, 1H), 7.41 (td, J = 7.6, 1.3 Hz, 1H), 7.35 (d, J = 3.7 Hz, 1H), 7.34 (d, J = 3.8 Hz, 1H), 7.24 (td, J = 7.7, 1.7 Hz, 1H). 13C NMR (151 MHz, chloroform-d) δ 177.69, 156.71, 151.88, 134.49, 130.55, 130.07, 129.79, 127.83, 122.63, 120.92, 113.08.
(E)-5-(2-styrylphenyl)furan-2-carbaldehyde (10s).
5-(2-Bromophenyl)furan-2-carbaldehyde (10q, 251.1 mg, 1.0 mmol), styrylboronic acid (222.0 mg, 1.5 mmol), Pd(PPh3)4 (57.8 mg, 0.05 mmol) and K2CO3 (414.6 mg, 3.0 mmol) were dissolved in toluene, EtOH and H2O (20 mL, 10:3:2, v/v/v). The reaction mixture was purged with N2 for three times and heated to reflux overnight. The reaction mixture was then filtered through celite and further purified by column to afford the title product. Yield: 83%. 1H NMR (600 MHz, chloroform-d) δ 9.71 (s, 1H), 7.83 (dd, J = 7.7, 1.3 Hz, 1H), 7.69 (dd, J = 7.7, 1.2 Hz, 1H), 7.52 (dd, J = 8.0, 1.1 Hz, 2H), 7.46 (td, J = 7.8, 1.4 Hz, 1H), 7.45 (d, J = 16.5 Hz, 1H), 7.39 (td, J = 7.7, 1.6 Hz, 2H), 7.35 (d, J = 3.7 Hz, 1H), 7.30 (dd, J = 7.4, 1.1 Hz, 2H), 7.05 (d, J = 16.2 Hz, 1H), 6.71 (d, J = 3.7 Hz, 1H). 13C NMR (151 MHz, chloroform-d) δ 177.65, 158.59, 152.29, 137.27, 136.60, 131.95, 129.90, 129.00, 128.55, 128.21, 127.95, 127.87, 127.57, 127.31, 126.87, 112.78, 112.75.
5-(3,4-Dichlorophenyl)furan-2-carboxylic acid (11e).
Procedure B. Yield: 85%. 1H NMR (600 MHz, DMSO-d6) δ 7.36–7.43 (2H, m), 7.80–7.87 (2H, m), 8.12 (1H, d, J = 2.0 Hz), 13.31 (1H, s).
5-(3-(Trifluoromethyl)phenyl)furan-2-carboxylic acid (11f).
Procedure B. Yield: 97%. 1H NMR (400 MHz, DMSO-d6) δ 7.38 (2H, q, J = 3.6 Hz), 7.72–7.79 (2H, m), 8.12 (2H, d, J = 7.3 Hz), 13.27 (1H, s).
5-(3-Chlorophenyl)furan-2-carboxylic acid (11j).
Procedure B. Yield: 92%. 1H NMR (400 MHz, DMSO-d6) δ 7.26–7.37 (2H, m), 7.45– 7.56 (2H, m), 7.77–7.90 (2H, m).
5-(2-Ethylphenyl)furan-2-carboxylic acid (11h).
Procedure B. Yield: 47%. 1H NMR (600 MHz, methanol-d4) δ 1.09–1.14 (3H, m), 2.78 (2H, q, J = 7.4 Hz), 6.61–6.65 (1H, m), 7.14–7.20 (1H, m), 7.19–7.29 (3H, m), 7.54 (1H, d, J = 7.5 Hz).
5-(2-(N,N-Dimethylsulfamoyl)phenyl)furan-2-carboxylic acid (11j).
Procedure B. Yield: 89%. 1H NMR (400 MHz, methanol-d4) δ 2.74 (6H, s), 6.88–7.05 (1H, m), 7.28–7.43 (1H, m), 7.65–7.86 (3H, m), 8.04 (1H, d, J = 7.7 Hz).
5-(Naphthalen-1-yl)furan-2-carboxylic acid (11k).
Procedure B. Yield: 50%. 1H NMR (400 MHz, chloroform-d) δ 6.81 (1H, d, J = 3.5 Hz), 7.41– 7.58 (4H, m), 7.76–7.92 (3H, m), 8.30–8.38 (1H, m).
5-(2,3-Dihydrobenzo[b][1,4]dioxin-5-yl)furan-2-carboxylic acid (11l).
Procedure B. Yield: 51%. 1H NMR (400 MHz, methanol-d4) δ 4.18 (4H, s), 6.66 (1H, d, J = 3.6 Hz), 6.79 (1H, d, J = 8.1 Hz), 7.14–7.22 (3H, m).
5-(2-Chlorophenyl)furan-2-carboxylic acid (11m).
Procedure B. Yield: 82%. 1H NMR (400 MHz, methanol-d4) δ 7.15 (1H, d, J = 3.7 Hz), 7.22 (1H, d, J = 3.7 Hz), 7.26 (1H, td, J = 7.7, 1.7 Hz), 7.33 (1H, td, J = 7.7, 1.3), 7.42 (1H, dd, J = 7.9, 1.2 Hz), 7.86–7.90 (1H, m).
5-(o-Tolyl)furan-2-carboxylic acid (11n).
Procedure B. Yield: 64%. 1H NMR (400 MHz, DMSO-d6) δ 2.51 (3H, s), 6.93 (1H, dd, J = 3.5, 1.8 Hz), 7.33–7.36 (4H, m), 7.72 (1H, dt, J = 5.0, 2.3 Hz), 13.09 (1H, s).
5-(2-(Benzyloxy)phenyl)furan-2-carboxylic acid (11o).
Procedure B. Yield: 97%. 1H NMR (400 MHz, chloroform-d) δ 5.13 (2H, s), 6.95 (1H, d, J = 3.7 Hz), 6.96–7.04 (2H, m), 7.24–7.41 (7H, m), 7.99 (1H, dd, J = 7.8, 1.6 Hz).
5-(2-bromophenyl)furan-2-carboxylic acid (11q).
Procedure B. Yield: 67%. 1H NMR (600 MHz, chloroform-d) δ 7.92 (dd, J = 7.9, 1.7 Hz, 1H), 7.69 (dd, J = 8.0, 1.3 Hz, 1H), 7.42 (td, J = 7.8, 1.2 Hz, 1H), 7.41 (d, J = 3.7 Hz, 1H), 7.29 (d, J = 3.7 Hz, 1H), 7.23 (ddd, J = 8.1, 7.4, 1.7 Hz, 1H). 13C NMR (151 MHz, chloroform-d) δ 161.34, 156.00, 142.69, 134.41, 130.27, 130.05, 130.01, 127.81, 121.42, 120.74, 112.74.
(E)-5-(2-styrylphenyl)furan-2-carboxylic acid (11s).
Procedure B. Yield: 83%. 1H NMR (600 MHz, DMSO-d6) δ 13.16 (s, 1H), 7.83 (d, J = 7.7 Hz, 1H), 7.73 (dd, J = 7.7, 1.5 Hz, 1H), 7.62 (dd, J = 7.6, 1.5 Hz, 2H), 7.59 (dd, J = 16.5, 1.3 Hz, 1H), 7.47 (ddd, J = 22.6, 7.5, 1.5 Hz, 1H), 7.43 (td, J = 7.5, 1.3 Hz, 1H), 7.39 (t, J = 7.6 Hz, 2H), 7.37 (d, J = 3.6 Hz, 1H), 7.30 (t, J = 7.3 Hz, 1H), 7.21 (d, J = 16.2 Hz, 1H), 6.85 (d, J = 3.6 Hz, 1H). 13C NMR (151 MHz, DMSO-d6) δ 159.31, 155.64, 144.43, 137.02, 135.07, 131.13, 129.32, 128.78, 128.76, 128.12, 127.92, 127.59, 127.01, 126.70, 126.60, 119.43, 111.77.
(R)-2-Amino-3-(5-(4-chlorophenyl)furan-2-carboxamido)propanoic acid (8a).
Procedure C. Yield for two steps: 5%. 1H NMR (400 MHz, DMSO-d6) δ 8.84 (t, J = 5.8 Hz, 1H), 7.92 (d, J = 8.4 Hz, 2H), 7.55 (d, J = 8.4 Hz, 2H), 7.21 (d, J = 3.7 Hz, 1H), 7.17 (d, J = 3.6 Hz, 1H), 3.78 (td, J = 11.2, 9.3, 4.8 Hz, 1H), 3.72 (q, J = 4.6 Hz, 1H), 3.64 (dt, J = 14.1, 6.9 Hz, 1H). 13C NMR (101 MHz, DMSO-d6) δ 169.35, 158.72, 153.83, 147.45, 133.56, 129.47, 128.65, 126.45, 116.53, 108.83, 53.63, 40.00. MS calcd for C14H13ClN2O4: 308.1, found: 309.5 [M+H]+.
(R)-2-Amino-3-(5-(4-methoxyphenyl)furan-2-carboxamido)propanoic acid (8b).
Procedure C. Yield for two steps: 5%. 1H NMR (400 MHz, DMSO-d6) δ 8.74 (t, J = 6.0 Hz, 1H), 8.41 (s, 2H), 7.84 (d, J = 8.8 Hz, 2H), 7.18 (d, J = 3.5 Hz, 1H), 7.03 (d, J = 8.8 Hz, 2H), 6.96 (d, J = 3.6 Hz, 1H), 4.07 (dd, J = 7.0, 4.9 Hz, 1H), 3.81 (s, 3H), 3.80 – 3.67 (m, 2H). 13C NMR (101 MHz, DMSO-d6) δ 169.69, 160.13, 159.05, 155.41, 146.34, 126.44, 122.58, 116.80, 114.83, 106.41, 55.75, 52.98, 39.96. MS calcd for C15H16N2O5: 304.1, found: 305.5 [M+H]+.
(R)-2-Amino-3-(5-phenylfuran-2-carboxamido)propanoic acid (8c).
Procedure C. Yield for two steps: 6%. 1H NMR (400 MHz, DMSO-d6) δ 8.76 (t, J = 6.0 Hz, 1H), 8.38 (s, 2H), 7.96 – 7.87 (m, 2H), 7.48 (dd, J = 8.4, 6.9 Hz, 2H), 7.43 – 7.35 (m, 1H), 7.22 (d, J = 3.6 Hz, 1H), 7.13 (d, J = 3.6 Hz, 1H), 4.11 (dd, J = 6.8, 5.0 Hz, 1H), 3.82 – 3.68 (m, 2H). 13C NMR (101 MHz, DMSO-d6) δ 169.66, 159.00, 155.14, 146.99, 129.74, 129.37, 129.16, 124.79, 116.73, 108.14, 52.83, 39.99. MS calcd for C14H14N2O4: 274.1, found: 275.5 [M+H]+.
(R)-2-Amino-3-(5-(2-(trifluoromethyl)phenyl)furan-2-carboxamido)propanoic acid (8d).
Procedure C. Yield for two steps: 10%. 1H NMR (600 MHz, DMSO-d6) δ 8.84 (t, J = 5.9 Hz, 1H), 8.61 (s, 2H), 7.99 (d, J = 7.8 Hz, 1H), 7.89 (d, J = 7.8 Hz, 1H), 7.80 (t, J = 7.6 Hz, 1H), 7.67 (t, J = 7.7 Hz, 1H), 7.38 (d, J = 3.6 Hz, 1H), 6.89 (d, J = 3.6 Hz, 1H), 4.09 (t, J = 5.7 Hz, 1H), 3.78 (t, J = 5.9 Hz, 2H). 13C NMR (151 MHz, DMSO-d6) δ 169.50, 158.51, 151.64, 148.01, 133.29, 131.37, 130.07, 128.33, 127.25, 127.21, 127.18, 127.14, 127.00, 116.24, 112.50, 112.48, 52.73, 39.10. MS calcd for C15H13F3N2O4: 342.1, found: 343.5 [M+H]+.
(R)-2-Amino-3-(5-(3,4-dichlorophenyl)furan-2-carboxamido)propanoic acid (8e).
Procedure C. Yield for two steps: 41%. 1H NMR (600 MHz, methanol-d4) δ 8.10 (d, J = 2.0 Hz, 1H), 7.78 (dd, J = 8.4, 2.0 Hz, 1H), 7.60 (d, J = 8.4 Hz, 1H), 7.27 (d, J = 3.6 Hz, 1H), 7.06 (d, J = 3.7 Hz, 1H), 4.17 (dd, J = 6.6, 3.9 Hz, 1H), 4.00 (dd, J = 14.7, 3.9 Hz, 1H), 3.89 (dd, J = 14.7, 6.7 Hz, 1H). 13C NMR (151 MHz, methanol-d4) δ 168.62, 160.50, 153.73, 146.67, 132.78, 132.04, 130.77, 129.74, 125.95, 123.88, 116.83, 108.51, 53.73, 39.18. MS calcd for C14H12Cl2N2O4: 342.0, found: 343.4 [M+H]+.
(R)-2-Amino-3-(5-(3-(trifluoromethyl)phenyl)furan-2-carboxamido)propanoic acid (8f).
Procedure C. Yield for two steps: 32%. 1H NMR (600 MHz, methanol-d4) δ 8.09 (s, 1H), 7.99 (d, J = 6.8 Hz, 1H), 7.54 (d, J = 7.3 Hz, 2H), 7.17 (d, J = 3.6 Hz, 1H), 6.99 (d, J = 3.6 Hz, 1H), 3.85 (dt, J = 11.9, 3.6 Hz, 2H), 3.76 (dt, J = 15.3, 7.1 Hz, 1H). 13C NMR (151 MHz, methanol-d4) δ 160.60, 146.75, 131.43, 131.22, 131.00, 130.79, 130.46, 126.76, 124.96, 124.81, 124.79, 124.76, 124.73, 123.16, 121.36, 120.79, 116.76, 108.35, 54.65, 39.58. MS calcd for C15H13F3N2O4: 342.1, found: 343.5 [M+H]+.
(R)-2-Amino-3-(5-(3-chlorophenyl)furan-2-carboxamido)propanoic acid (8g).
Procedure C. Yield for two steps: 46%. 1H NMR (600 MHz, methanol-d4) δ 7.91 (t, J = 1.9 Hz, 1H), 7.76 (dt, J = 7.7, 1.3 Hz, 1H), 7.41 (t, J = 7.9 Hz, 1H), 7.35 (ddd, J = 8.0, 2.1, 1.0 Hz, 1H), 7.26 (d, J = 3.6 Hz, 1H), 6.99 (d, J = 3.6 Hz, 1H), 4.28 (dd, J = 6.8, 4.0 Hz, 1H), 4.02 (dd, J = 14.7, 4.0 Hz, 1H), 3.91 (dd, J = 14.7, 6.9 Hz, 1H). 13C NMR (151 MHz, methanol-d4) δ 168.57, 160.53, 154.72, 146.38, 134.66, 131.26, 130.12, 128.27, 124.03, 122.58, 116.85, 108.01, 53.45, 39.12. MS calcd for C14H13ClN2O4: 308.1, found: 309.4 [M+H]+.
(R)-2-Amino-3-(5-(2-ethylphenyl)furan-2-carboxamido)propanoic acid (8h).
Procedure C. Yield for two steps: 41%. 1H NMR (600 MHz, methanol-d4) δ 7.63 – 7.58 (m, 1H), 7.27 – 7.21 (m, 2H), 7.20 – 7.15 (m, 2H), 6.62 (d, J = 3.6 Hz, 1H), 3.89 (dd, J = 6.8, 3.8 Hz, 1H), 3.83 (dd, J = 14.6, 3.9 Hz, 1H), 3.75 (dd, J = 14.7, 6.9 Hz, 1H), 2.77 (q, J = 7.6 Hz, 2H), 1.11 (t, J = 7.5 Hz, 3H). 13C NMR (151 MHz, methanol-d4) δ 169.58, 160.76, 156.16, 145.87, 141.89, 129.15, 128.97, 128.41, 128.39, 125.75, 116.38, 110.18, 54.66, 39.55, 26.58, 14.12. MS calcd for C16H18N2O4: 302.1, found: 303.5 [M+H]+.
(R)-2-Amino-3-(5-(2-(methoxymethyl)phenyl)furan-2-carboxamido)propanoic acid (8i).
Procedure C. Yield for two steps: 44%. 1H NMR (400 MHz, methanol-d4) δ 7.91 – 7.86 (m, 1H), 7.53 (dd, J = 7.1, 1.9 Hz, 1H), 7.46 – 7.39 (m, 2H), 7.30 (t, J = 3.5 Hz, 1H), 6.85 (d, J = 3.6 Hz, 1H), 4.60 (s, 2H), 4.26 (dd, J = 6.7, 4.0 Hz, 1H), 4.02 (dd, J = 14.7, 4.1 Hz, 1H), 3.91 (dd, J = 14.7, 6.7 Hz, 1H), 3.41 (s, 3H). 13C NMR (101 MHz, methanol-d4) δ 168.59, 160.70, 154.99, 146.19, 134.83, 130.08, 129.06, 128.52, 128.01, 127.88, 116.61, 110.94, 72.62, 56.83, 53.43, 39.08. MS calcd for C16H18N2O5: 318.1, found: 319.5 [M+H]+.
(R)-2-Amino-3-(5-(2-(N,N-dimethylsulfamoyl)phenyl)furan-2-carboxamido)propanoic acid (8j).
Procedure C. Yield for two steps: 29%. 1H NMR (600 MHz, methanol-d4) δ 7.98 (d, J = 7.9 Hz, 1H), 7.81 (d, J = 7.6 Hz, 1H), 7.74 (t, J = 7.5 Hz, 1H), 7.68 (t, J = 7.7 Hz, 1H), 7.28 (d, J = 3.6 Hz, 1H), 6.98 (d, J = 3.6 Hz, 1H), 4.24 (dd, J = 6.6, 4.0 Hz, 1H), 4.00 (dd, J = 14.7, 3.9 Hz, 1H), 3.88 (dd, J = 14.7, 6.7 Hz, 1H), 2.74 (s, 6H). 13C NMR (151 MHz, methanol-d4) δ 168.53, 160.54, 152.85, 146.68, 136.71, 132.42, 132.21, 129.50, 129.22, 128.88, 116.03, 113.16, 53.44, 39.05, 36.18. MS calcd for C16H19N3O6S: 381.1, found: 382.5 [M+H]+.
(R)-2-Amino-3-(5-(naphthalen-1-yl)furan-2-carboxamido)propanoic acid (8k).
Procedure C. Yield for two steps: 51%. 1H NMR (400 MHz, methanol-d4) δ 8.22 (d, J = 8.1 Hz, 1H), 7.84 (d, J = 7.9 Hz, 2H), 7.76 (d, J = 7.2 Hz, 1H), 7.45 (tt, J = 7.6, 4.1 Hz, 3H), 7.27 (d, J = 3.5 Hz, 1H), 6.84 (d, J = 3.6 Hz, 1H), 4.15 (dd, J = 6.7, 4.1 Hz, 1H), 3.90 (dd, J = 14.7, 4.0 Hz, 1H), 3.80 (dd, J = 15.1, 7.2 Hz, 1H). 13C NMR (101 MHz, methanol-d4) δ 168.50, 160.77, 155.92, 146.43, 134.02, 130.18, 129.54, 128.40, 127.00, 126.84, 126.83, 125.92, 124.90, 124.55, 116.55, 111.26, 53.44, 39.08. MS calcd for C18H16N2O4: 324.1, found: 325.5 [M+H]+.
(R)-2-Amino-3-(5-(2,3-dihydrobenzo[b][1,4]dioxin-5-yl)furan-2-carboxamido) propanoic acid (8l).
Procedure C. Yield for two steps: 39%. 1H NMR (400 MHz, DMSO-d6) δ 8.72 (t, J = 5.8 Hz, 1H), 7.45 (d, J = 2.1 Hz, 1H), 7.37 (dd, J = 8.4, 2.1 Hz, 1H), 7.16 (d, J = 3.6 Hz, 1H), 6.99 – 6.92 (m, 2H), 4.29 (s, 4H), 3.88 (dd, J = 7.3, 4.9 Hz, 1H), 3.77 – 3.61 (m, 2H). 13C NMR (101 MHz, DMSO-d6) δ 169.40, 158.92, 154.95, 146.51, 144.48, 144.18, 123.29, 118.24, 118.04, 116.67, 113.61, 106.90, 64.74, 64.61, 53.41, 40.68. MS calcd for C16H16N2O6: 332.1, found: 333.5 [M+H]+.
(R)-2-Amino-3-(5-(2-chlorophenyl)furan-2-carboxamido)propanoic acid (8m).
Procedure C. Yield for two steps: 34%. 1H NMR (400 MHz, DMSO-d6) δ 8.84 (t, J = 5.6 Hz, 1H), 8.11 (d, J = 7.8 Hz, 1H), 7.60 (d, J = 7.9 Hz, 1H), 7.50 (t, J = 7.6 Hz, 1H), 7.43 (t, J = 7.7 Hz, 1H), 7.26 (q, J = 3.8 Hz, 2H), 3.72 (dd, J = 9.6, 4.5 Hz, 2H), 3.67 – 3.62 (m, 1H). 13C NMR (101 MHz, DMSO-d6) δ 169.19, 158.57, 151.07, 131.32, 130.44, 130.17,129.10, 128.08, 127.98, 116.12, 113.18, 53.67, 40.44. MS calcd for C14H13ClN2O4: 308.1, found: 309.5 [M+H]+.
(R)-2-Amino-3-(5-(o-tolyl)furan-2-carboxamido)propanoic acid (8n).
Procedure C. Yield for two steps: 22%. 1H NMR (400 MHz, methanol-d4) δ 7.86 (td, J = 7.4, 3.5 Hz, 1H), 7.31 (td, J = 8.7, 4.5 Hz, 4H), 6.84 – 6.78 (m, 1H), 4.25 (tt, J = 6.5, 3.8 Hz, 1H), 4.02 (dq, J = 15.0, 4.4 Hz, 1H), 3.92 (dq, J = 14.5, 6.9 Hz, 1H), 2.53 (s, 3H). 13C NMR (101 MHz, methanol-d4) δ 168.58, 160.78, 156.12, 145.62, 135.31, 130.90, 128.83, 128.55, 127.43, 125.82, 116.54, 110.46, 53.56, 39.12, 20.47. MS calcd for C15H16N2O4: 288.1, found: 289.5 [M+H]+.
(R)-2-Amino-3-(5-(2-(benzyloxy)phenyl)furan-2-carboxamido)propanoic acid (8o).
Procedure C. Yield for two steps: 40%. 1H NMR (400 MHz, methanol-d4) δ 7.39 (dd, J = 7.7, 1.7 Hz, 1H), 7.33 – 7.15 (m, 8H), 6.99 (d, J = 7.4 Hz, 1H), 6.96 – 6.92 (m, 1H), 4.17 (dd, J = 6.9, 3.8 Hz, 1H), 3.96 (d, J = 4.7 Hz, 1H), 3.92 (d, J = 3.7 Hz, 1H), 3.85 (s, 2H). 13C NMR (101 MHz, methanol-d4) δ 168.56, 160.95, 155.19, 150.67, 145.32, 140.31, 130.71, 130.48, 128.31, 128.00, 125.76, 124.21, 119.22, 117.81, 117.15, 115.79, 53.60, 39.00, 31.08. MS calcd for C21H20N2O5: 380.1, found: 381.6 [M+H]+.
(R)-2-amino-3-(5-(4-chloro-2-nitrophenyl)furan-2-carboxamido)propanoic acid (8p).
Procedure C. Yield for two steps: 42%. 1H NMR (400 MHz, DMSO-d6) δ 8.79 (t, J = 6.0 Hz, 1H), 8.43 (s, 3H), 8.16 (d, J = 2.1 Hz, 1H), 8.02 (d, J = 8.5 Hz, 1H), 7.86 (dd, J = 8.5, 2.2 Hz, 1H), 7.26 (d, J = 3.7 Hz, 1H), 6.90 (d, J = 3.7 Hz, 1H), 4.10 (t, J = 5.7 Hz, 1H), 3.75 (ddt, J = 25.3, 14.3, 6.3 Hz, 2H). 13C NMR (101 MHz, DMSO-d6) δ 169.61, 158.53, 148.66, 148.60, 147.83, 134.65, 133.06, 131.33, 124.56, 121.32, 116.33, 112.36, 52.67, 39.29. MS calcd for C14H12ClN3O4: 353.0, found: 354.4 [M+H]+.
(R)-2-Amino-3-(5-(2-bromophenyl)furan-2-carboxamido)propanoic acid (8q).
Procedure C. Yield for two steps: 3%. 1H NMR (400 MHz, DMSO-d6) δ 8.77 (t, J = 5.8 Hz, 1H), 8.02 (dd, J = 7.9, 1.7 Hz, 1H), 7.79 (dd, J = 8.0, 1.2 Hz, 1H), 7.54 (td, J = 7.6, 1.3 Hz, 1H), 7.35 (td, J = 7.7, 1.7 Hz, 1H), 7.30 (d, J = 3.7 Hz, 1H), 7.26 (d, J = 3.6 Hz, 1H), 3.82 (dd, J = 7.5, 4.9 Hz, 1H), 3.73 (dt, J = 13.9, 5.0 Hz, 1H), 3.66 (dt, J = 14.0, 7.0 Hz, 1H). 13C NMR (151 MHz, DMSO-d6) δ 169.30, 158.64, 152.34, 147.43, 134.65, 130.77, 129.99, 129.98, 128.51, 119.92, 115.92, 112.83, 53.46, 40.00. MS calcd for C14H13BrN2O4: 352.0, found: 353.5 [M+H]+.
(R,E)-2-Amino-3-(5-(2-(3-fluorostyryl)phenyl)furan-2-carboxamido)propanoic acid (8r).
Procedure C. Yield for two steps: 4%. 1H NMR (400 MHz, DMSO-d6) δ 8.80 (t, J = 5.8 Hz, 1H), 7.90 – 7.83 (m, 1H), 7.81 – 7.74 (m, 1H), 7.59 (d, J = 16.2 Hz, 1H), 7.46 (dd, J = 8.4, 3.8 Hz, 5H), 7.29 (d, J = 3.6 Hz, 1H), 7.20 (d, J = 16.2 Hz, 1H), 7.13 (tt, J = 6.7, 2.7 Hz, 1H), 6.75 (d, J = 3.6 Hz, 1H), 3.92 – 3.88 (m, 1H), 3.78 (dt, J = 15.8, 7.9 Hz, 1H), 3.68 (dt, J = 13.7, 6.7 Hz, 1H). 13C NMR (151 MHz, DMSO-d6) δ 169.68, 164.36, 161.84, 158.90, 153.86, 147.42, 140.09, 140.01, 135.15, 131.18, 131.10, 130.61, 130.59, 129.50, 128.72, 128.57, 128.42, 128.39, 127.71, 123.49, 123.47, 116.31, 115.12, 114.91, 113.54, 113.33, 112.55, 53.41, 39.97. MS calcd for C22H19FN2O4: 394.1, found: 395.6 [M+H]+.
(R,E)-2-Amino-3-(5-(2-styrylphenyl)furan-2-carboxamido)propanoic acid (8s).
Procedure C. Yield for two steps: 36%. 1H NMR (600 MHz, DMSO-d6) δ 8.73 (t, J = 5.9 Hz, 1H), 7.84 (dd, J = 7.4, 1.8 Hz, 1H), 7.80 (dd, J = 7.5, 1.6 Hz, 1H), 7.61 (dd, J = 8.5, 1.2 Hz, 2H), 7.52 (d, J = 16.2 Hz, 1H), 7.46 (dt, J = 7.5, 2.8, 1.6 Hz, 2H), 7.40 (td, J = 7.7, 1.5 Hz, 2H), 7.30 (tt, J = 7.2, 2.4, 1.3 Hz, 1H), 7.28 (d, J = 3.5 Hz, 1H), 7.20 (d, J = 16.2 Hz, 1H), 6.75 (d, J = 3.5 Hz, 1H), 3.96 (dd, J = 7.1, 5.0 Hz, 1H), 3.75 (dt, J = 14.1, 5.2 Hz, 1H), 3.69 (dt, J = 13.9, 6.8 Hz, 1H). 13C NMR (151 MHz, DMSO-d6) δ 169.05, 158.47, 153.58, 146.86, 136.92, 135.11, 131.34, 129.11, 128.83, 128.01, 127.97, 127.83, 127.09, 126.75, 126.57, 115.92, 112.01, 52.70, 40.06. MS calcd for C22H20N2O4: 376.1, found: 377.6 [M+H]+.
Pharmacological evaluation
DNA Constructs.
cDNAs encoding GluN1–1a (Genbank accession number U11418 and U08261), GluN2A (D13211), GluN2B (U11419), GluN2C (M91563), GluN2D (L31611), GluN3A (U29873), and GluN3B (AF440691) were generously provided by Dr. S. Heinemann (Salk Institute, La Jolla, CA), Dr. S. Nakanishi (Osaka Bioscience Institute, Osaka, Japan), Dr. P. Seeburg (University of Heidelberg, Germany), and Dr. D. Zhang (Sanford-Burnham Medical Research Institute, La Jolla, CA). The GluN2B open reading frame was edited without changing the amino acid sequence to remove a T7 RNA polymerase termination site.44
Two-Electrode Voltage-Clamp Recordings.
For expression in Xenopus laevis oocytes, cDNAs were linearized using restriction enzymes and used as templates to synthesize cRNA using the mMessage mMachine kit (Ambion, Life Technologies, Paisley, UK). Xenopus oocytes were obtained from Rob Weymouth (Xenopus 1, Dexter, MI). The oocytes were injected with cRNAs encoding GluN1 and GluN2 in a 1:2 ratio and maintained as previously described.45
Two-electrode voltage-clamp recordings were performed 2–4 days following cRNA injection at room temperature and at a holding potential of −40 mV essentially as previously described.45 Oocytes were perfused with extracellular recording solution comprised of 90 mM NaCl, 1 mM KCl, 10 mM HEPES, 0.5 mM BaCl2 and 0.01 mM EDTA (pH 7.4 with NaOH). Oocytes expressing GluN1/2A or GluN1/2B were injected with 50 nL and 30 nL, respectively, of 50 mM BAPTA approximately 10–30 min before recordings to prevent activity-dependent increases in response amplitude.46 Compounds were dissolved in DMSO to make 20–100 mM stock solutions and the concentration of DMSO was kept constant (< 0.5%) in all recording solutions.
Data were analyzed using GraphPad Prism (GraphPad Software, La Jolla, CA). Agonist concentration-response data for individual oocytes were fitted to the Hill equation, I = Rmax/(1+10^((logEC50-log[A])*nH)), where Rmax is the maximum current in response to the agonist relative to the maximal response to 100 μM Gly in the same recording, nH is the Hill slope, [A] is the agonist concentration, and EC50 is the agonist concentration that produces half-maximum response.
Molecular modeling
Marvin and JChem for Office were used for drawing, characterizing, and organizing a subset of the chemical structures, Marvin 20.15.0 and JChem 20.14.0.668, ChemAxon (https://www.chemaxon.com). All modeling was carried out in the Schrodinger Maestro program, Maestro version 11.9.011, Release 2019–1. The GluN1/2A ABD crystal structure (PDB ID: 5I57) was prepared using the Protein Preparation Wizard with default parameters. The protein bond order was reassigned with CCD database. Protein hydrogens were added, all H2O molecules were deleted, and the protonation state for the ligands and receptors was set to pH 7.0. Restrained minimization was carried out with OPLS3e force field, and the resulting protein structure was used for next step induced-fit docking. Compounds 8h, 8k and 8p were prepared by LigPrep with default parameters. Ligands were generated at pH 7.0 with OPLS3e force field to neutralize the molecules. For each ligand, the highest scoring conformation in LigPrep was used for docking.
Induced-fit docking of compound 8h was performed with default parameters. Standard protocol was used and up to 20 docking poses were generated. The Gly binding site in the GluN1/2A ABD complex (PDB ID: 5I57) was identified as the binding box center and the box size was set as similar in size to 8h. The Glide redocking precision was set as XP Glide Redocking Precision and the other parameters was set as default. The docking model of 8h was used for the induced-fit docking of 8k and 8p with the box center as 8h and the same docking parameters. The binding model of each compound was selected based on the overlay with Gly and the IFDScore. All docking results were treated with energy minimization.
Supplementary Material
ACKNOWLEDGMENTS
The authors acknowledge financial support from the China Scholarship Council to F.Z. and National Institutes of Health [NS097536, NS116055, GM103546] to K.B.H.
K.B.H. is principal investigator on a research grant from Janssen Research and Development to the University of Montana. All other authors have no conflicts of interest to declare.
ABBREVIATIONS USED
- AMPA
α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
- ABD
agonist binding domain
- Boc
tert-butyloxycarbonyl
- DCM
dichloromethane
- DIPEA
N, N-diisopropylethylamine
- d-Ser
, d-serine
- EDCI
N-(3-Dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride
- EtOH
ethanol
- Glu
glutamate
- Gly
glycine
- HOBt
hydroxybenzotriazole
- NMDA
N-methyl-d-aspartic acid
- Pd(PPh3)4
Tetrakis(triphenylphosphine)palladium
- prep-HPLC
preparative-high performance liquid chromatography
- rt
room temperature
- TEVC
two-electrode voltage-clamp
Footnotes
Supporting Information
Supporting information includes an extended version of Table 1 with sample size, Hill slopes, and standard deviations, the docking poses of 8h, 8k and 8p, and purity traces of all final compounds.
REFERENCES
- 1.Lemoine D; Jiang R; Taly A; Chataigneau T; Specht A; Grutter T, Ligand-gated ion channels: new insights into neurological disorders and ligand recognition. Chem Rev 2012, 112 (12), 6285–6318. [DOI] [PubMed] [Google Scholar]
- 2.Traynelis SF; Wollmuth LP; McBain CJ; Menniti FS; Vance KM; Ogden KK; Hansen KB; Yuan H; Myers SJ; Dingledine R, Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev 2010, 62 (3), 405–496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Hansen KB; Wollmuth LP; Bowie D; Furukawa H; Menniti FS; Sobolevsky AI; Swanson GT; Swanger SA; Greger IH; Nakagawa T; McBain CJ; Jayaraman V; Low CM; Dell’Acqua ML; Diamond JS; Camp CR; Perszyk RE; Yuan H; Traynelis SF, Structure, Function, and Pharmacology of Glutamate Receptor Ion Channels. Pharmacol Rev 2021, 73 (4), 298–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Pin JP; Duvoisin R, The metabotropic glutamate receptors: structure and functions. Neuropharmacology 1995, 34 (1), 1–26. [DOI] [PubMed] [Google Scholar]
- 5.Conn PJ; Pin JP, Pharmacology and functions of metabotropic glutamate receptors. Annu Rev Pharmacol Toxicol 1997, 37 (1), 205–237. [DOI] [PubMed] [Google Scholar]
- 6.Niswender CM; Conn PJ, Metabotropic glutamate receptors: physiology, pharmacology, and disease. Annu Rev Pharmacol Toxicol 2010, 50, 295–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Bae M; Roh JD; Kim Y; Kim SS; Han HM; Yang E; Kang H; Lee S; Kim JY; Kang R; Jung H; Yoo T; Kim H; Kim D; Oh H; Han S; Kim D; Han J; Bae YC; Kim H; Ahn S; Chan AM; Lee D; Kim JW; Kim E, SLC6A20 transporter: a novel regulator of brain glycine homeostasis and NMDAR function. EMBO Mol Med 2021, 13 (2), e12632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bergeron R; Meyer TM; Coyle JT; Greene RW, Modulation of N-methyl-D-aspartate receptor function by glycine transport. Proc Natl Acad Sci U S A 1998, 95 (26), 15730–15734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Billups D; Attwell D, Active release of glycine or D-serine saturates the glycine site of NMDA receptors at the cerebellar mossy fibre to granule cell synapse. Eur J Neurosci 2003, 18 (11), 2975–2980. [DOI] [PubMed] [Google Scholar]
- 10.Ishiwata S; Umino A; Balu DT; Coyle JT; Nishikawa T, Neuronal serine racemase regulates extracellular D-serine levels in the adult mouse hippocampus. J Neural Transm (Vienna) 2015, 122 (8), 1099–1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Maolanon AR; Risgaard R; Wang SY; Snoep Y; Papangelis A; Yi F; Holley D; Barslund AF; Svenstrup N; Hansen KB; Clausen RP, Subtype-Specific Agonists for NMDA Receptor Glycine Binding Sites. ACS Chem Neurosci 2017, 8 (8), 1681–1687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Chen PE; Geballe MT; Katz E; Erreger K; Livesey MR; O’Toole KK; Le P; Lee CJ; Snyder JP; Traynelis SF; Wyllie DJ, Modulation of glycine potency in rat recombinant NMDA receptors containing chimeric NR2A/2D subunits expressed in Xenopus laevis oocytes. J Physiol 2008, 586 (1), 227–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Farlow MR, NMDA receptor antagonists. A new therapeutic approach for Alzheimer’s disease. Geriatrics 2004, 59 (6), 22–27. [PubMed] [Google Scholar]
- 14.Olivares D; Deshpande VK; Shi Y; Lahiri DK; Greig NH; Rogers JT; Huang X, N-methyl D-aspartate (NMDA) receptor antagonists and memantine treatment for Alzheimer’s disease, vascular dementia and Parkinson’s disease. Curr Alzheimer Res 2012, 9 (6), 746–758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Mishizen-Eberz AJ; Rissman RA; Carter TL; Ikonomovic MD; Wolfe BB; Armstrong DM, Biochemical and molecular studies of NMDA receptor subunits NR1/2A/2B in hippocampal subregions throughout progression of Alzheimer’s disease pathology. Neurobiol Dis 2004, 15 (1), 80–92. [DOI] [PubMed] [Google Scholar]
- 16.Mohn AR; Gainetdinov RR; Caron MG; Koller BH, Mice with reduced NMDA receptor expression display behaviors related to schizophrenia. Cell 1999, 98 (4), 427–436. [DOI] [PubMed] [Google Scholar]
- 17.Coyle JT, NMDA receptor and schizophrenia: a brief history. Schizophr Bull 2012, 38 (5), 920–926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Snyder MA; Gao WJ, NMDA receptor hypofunction for schizophrenia revisited: Perspectives from epigenetic mechanisms. Schizophr Res 2020, 217, 60–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hanada T, Ionotropic Glutamate Receptors in Epilepsy: A Review Focusing on AMPA and NMDA Receptors. Biomolecules 2020, 10 (3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Balu DT; Coyle JT, The NMDA receptor ‘glycine modulatory site’ in schizophrenia: D-serine, glycine, and beyond. Curr Opin Pharmacol 2015, 20, 109–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Goff DC, Drug development in schizophrenia: are glutamatergic targets still worth aiming at? Curr Opin Psychiatry 2015, 28 (3), 207–215. [DOI] [PubMed] [Google Scholar]
- 22.Schade S; Paulus W, D-Cycloserine in Neuropsychiatric Diseases: A Systematic Review. Int J Neuropsychopharmacol 2016, 19 (4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Szakacs R; Janka Z; Kalman J, The “blue” side of glutamatergic neurotransmission: NMDA receptor antagonists as possible novel therapeutics for major depression. Neuropsychopharmacol Hung 2012, 14 (1), 29–40. [PubMed] [Google Scholar]
- 24.Hood WF; Compton RP; Monahan JB, D-cycloserine: a ligand for the N-methyl-D-aspartate coupled glycine receptor has partial agonist characteristics. Neurosci Lett 1989, 98 (1), 91–95. [DOI] [PubMed] [Google Scholar]
- 25.Dravid SM; Burger PB; Prakash A; Geballe MT; Yadav R; Le P; Vellano K; Snyder JP; Traynelis SF, Structural determinants of D-cycloserine efficacy at the NR1/NR2C NMDA receptors. J Neurosci 2010, 30 (7), 2741–2754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Jessen M; Frederiksen K; Yi F; Clausen RP; Hansen KB; Brauner-Osborne H; Kilburn P; Damholt A, Identification of AICP as a GluN2C-Selective N-Methyl-d-Aspartate Receptor Superagonist at the GluN1 Glycine Site. Mol Pharmacol 2017, 92 (2), 151–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Sheinin A; Shavit S; Benveniste M, Subunit specificity and mechanism of action of NMDA partial agonist D-cycloserine. Neuropharmacology 2001, 41 (2), 151–158. [DOI] [PubMed] [Google Scholar]
- 28.Davis M; Ressler K; Rothbaum BO; Richardson R, Effects of D-cycloserine on extinction: translation from preclinical to clinical work. Biol Psychiatry 2006, 60 (4), 369–375. [DOI] [PubMed] [Google Scholar]
- 29.Hofmann SG; Otto MW; Pollack MH; Smits JA, D-cycloserine augmentation of cognitive behavioral therapy for anxiety disorders: an update. Curr Psychiatry Rep 2015, 17 (1), 532. [DOI] [PubMed] [Google Scholar]
- 30.Singewald N; Schmuckermair C; Whittle N; Holmes A; Ressler KJ, Pharmacology of cognitive enhancers for exposure-based therapy of fear, anxiety and trauma-related disorders. Pharmacol Ther 2015, 149, 150–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Urwyler S; Floersheim P; Roy BL; Koller M, Drug design, in vitro pharmacology, and structure-activity relationships of 3-acylamino-2-aminopropionic acid derivatives, a novel class of partial agonists at the glycine site on the N-methyl-D-aspartate (NMDA) receptor complex. J Med Chem 2009, 52 (16), 5093–5107. [DOI] [PubMed] [Google Scholar]
- 32.Liu J; Shelkar GP; Zhao F; Clausen RP; Dravid SM, Modulation of burst firing of neurons in nucleus reticularis of the thalamus by GluN2C-containing NMDA receptors. Mol Pharmacol 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Liu J; Shelkar GP; Sarode LP; Gawande DY; Zhao F; Clausen RP; Ugale RR; Dravid SM, Facilitation of GluN2C-containing NMDA receptors in the external globus pallidus increases firing of fast spiking neurons and improves motor function in a hemiparkinsonian mouse model. Neurobiol Dis 2021, 150, 105254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zhao F; Rouzbeh N; Hansen KB; Clausen RP, Improved synthetic route for the GluN2-specific NMDA receptor glycine site agonist AICP. Tetrahedron Lett 2020, 61 (12), 151653. [Google Scholar]
- 35.Christopoulos A, Assessing the distribution of parameters in models of ligand-receptor interaction: to log or not to log. Trends Pharmacol Sci 1998, 19 (9), 351–357. [DOI] [PubMed] [Google Scholar]
- 36.Cheng Y; Prusoff WH, Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem Pharmacol 1973, 22 (23), 3099–3108. [DOI] [PubMed] [Google Scholar]
- 37.Chatterton JE; Awobuluyi M; Premkumar LS; Takahashi H; Talantova M; Shin Y; Cui J; Tu S; Sevarino KA; Nakanishi N; Tong G; Lipton SA; Zhang D, Excitatory glycine receptors containing the NR3 family of NMDA receptor subunits. Nature 2002, 415 (6873), 793–798. [DOI] [PubMed] [Google Scholar]
- 38.Yao Y; Mayer ML, Characterization of a soluble ligand binding domain of the NMDA receptor regulatory subunit NR3A. J Neurosci 2006, 26 (17), 4559–4566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Yao Y; Harrison CB; Freddolino PL; Schulten K; Mayer ML, Molecular mechanism of ligand recognition by NR3 subtype glutamate receptors. EMBO J 2008, 27 (15), 2158–2170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kvist T; Greenwood JR; Hansen KB; Traynelis SF; Brauner-Osborne H, Structure-based discovery of antagonists for GluN3-containing N-methyl-D-aspartate receptors. Neuropharmacology 2013, 75, 324–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Awobuluyi M; Yang J; Ye Y; Chatterton JE; Godzik A; Lipton SA; Zhang D, Subunit-specific roles of glycine-binding domains in activation of NR1/NR3 N-methyl-D-aspartate receptors. Mol Pharmacol 2007, 71 (1), 112–122. [DOI] [PubMed] [Google Scholar]
- 42.Madry C; Mesic I; Bartholomaus I; Nicke A; Betz H; Laube B, Principal role of NR3 subunits in NR1/NR3 excitatory glycine receptor function. Biochem Biophys Res Commun 2007, 354 (1), 102–108. [DOI] [PubMed] [Google Scholar]
- 43.Yi F; Mou TC; Dorsett KN; Volkmann RA; Menniti FS; Sprang SR; Hansen KB, Structural Basis for Negative Allosteric Modulation of GluN2A-Containing NMDA Receptors. Neuron 2016, 91 (6), 1316–1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Hansen KB; Ogden KK; Yuan H; Traynelis SF, Distinct functional and pharmacological properties of Triheteromeric GluN1/GluN2A/GluN2B NMDA receptors. Neuron 2014, 81 (5), 1084–1096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Hansen KB; Tajima N; Risgaard R; Perszyk RE; Jorgensen L; Vance KM; Ogden KK; Clausen RP; Furukawa H; Traynelis SF, Structural determinants of agonist efficacy at the glutamate binding site of N-methyl-D-aspartate receptors. Mol Pharmacol 2013, 84 (1), 114–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Williams K, Ifenprodil discriminates subtypes of the N-methyl-D-aspartate receptor: selectivity and mechanisms at recombinant heteromeric receptors. Mol Pharmacol 1993, 44 (4), 851–859. [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.