Abstract
Some inverse agonists of cannabinoid receptor type 1 (CB1) have been demonstrated to be anorectic antiobesity drug candidates. However, the first generation of CB1 inverse agonists, represented by rimonabant (SR141716A), otenabant, and taranabant, are centrally active, with a high level of psychiatric side effects. Hence, the discovery of CB1 inverse agonists with a chemical scaffold distinct from these holds promise for developing peripherally active CB1 inverse agonists with fewer side effects. We generated a new CB1 inverse agonist, (4-(bis(4-fluorophenyl)methyl)piperazin-1-yl)(cyclohexyl)methanone hydrochloride (LDK1229), from the class of benzhydryl piperazine analogs. This compound binds to CB1 more selectively than cannabinoid receptor type 2, with a Ki value of 220 nM. Comparable CB1 binding was also observed by analogs 1-[bis(4-fluorophenyl)methyl]-4-cinnamylpiperazine dihydrochloride (LDK1203) and 1-[bis(4-fluorophenyl)methyl]-4-tosylpiperazine hydrochloride (LDK1222), which differed by the substitution on the piperazine ring where the piperazine of LDK1203 and LDK1222 are substituted by an alkyl group and a tosyl group, respectively. LDK1229 exhibits efficacy comparable with SR141716A in antagonizing the basal G protein coupling activity of CB1, as indicated by a reduction in guanosine 5′-O-(3-thio)triphosphate binding. Consistent with inverse agonist behavior, increased cell surface localization of CB1 upon treatment with LDK1229 was also observed. Although docking and mutational analysis showed that LDK1229 forms similar interactions with the receptor as SR141716A does, the benzhydryl piperazine scaffold is structurally distinct from the first-generation CB1 inverse agonists. It offers new opportunities for developing novel CB1 inverse agonists through the optimization of molecular properties, such as the polar surface area and hydrophilicity, to reduce the central activity observed with SR141716A.
Introduction
The cannabinoid receptors are members of the class A superfamily of G protein–coupled receptors (GPCRs). The cannabinoid receptor 1 (CB1) is present in high abundance throughout the central nervous system (Howlett, 1995) but is also expressed in a number of peripheral tissues, such as the cardiovascular and reproductive systems as well as the gastrointestinal tract (Croci et al., 1998; Batkai et al., 2001; Engeli et al., 2005), and is involved in substance addiction, chronic pain, memory, and metabolic and inflammatory disorders (Howlett et al., 2004; Mackie, 2006; Pertwee, 2006). A second subtype of the cannabinoid receptors, the cannabinoid receptor 2 (CB2), is predominantly found in immune cells and non-neuronal tissues (Galiegue et al., 1995) and is implicated in a variety of modulatory functions, including immune suppression, induction of apoptosis, and induction of cell migration (Basu and Dittel, 2011).
The CB1 receptor preferentially couples to the Gi/o type of G proteins (Howlett and Fleming, 1984) and has been functionally linked to the inhibition of adenylate cyclase (Slipetz et al., 1995) and the activation of mitogen-activated protein kinases, including extracellular signal-regulated kinase-1 and -2, p38 mitogen-activated protein kinase, and c-Jun N-terminal kinase (Bouaboula et al., 1996; Turu and Hunyady, 2010; Ahn et al., 2012). In addition, it is associated with the inhibition of N- and P/Q-type voltage-dependent Ca2+ channels and the stimulation of A-type and inwardly rectifying K+ channels (Howlett, 2005).
As is common for many GPCRs (De Lean et al., 1980), the CB1 receptor may exist in multiple activation states that are promoted by its binding to different ligands. Upon binding to receptor agonists, such as the endogenous arachidonylethanolamide and 2-arachidonyl glycerol, or the synthetic agonists 5-(1,1-dimethylheptyl)-2-(5-hydroxy-2-(3-hydroxypropyl)cyclohexyl)phenol (CP55,940) and (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl)pyrrolo-[1,2,3-d,e]-1,4-benzoxazin-6-yl]-1-naphthalenyl-methanone (WIN55,212-2), the active form of the receptor predominates. Interestingly, the CB1 receptor possesses agonist-independent constitutive or basal activity that can be inhibited by inverse agonists (Pertwee, 2005). This ligand-independent activity led to a receptor model that accounts for multiple activation states (Gether and Kobilka, 1998; Ghanouni et al., 2001) with distinct biochemical characteristics, including extent and selectivity of G protein or β-arrestin coupling (Kenakin, 1995).
Inverse agonists of the CB1 receptor have attracted considerable attention in drug discovery because of their ability to regulate appetite and manage substance addiction (Janero, 2012; Janero and Makriyannis, 2009; Silvestri and Di Marzo, 2012). Consequently, considerable effort has been invested in discovering compounds that can regulate the constitutive activity of the CB1 receptor. However, SR141716A (rimonabant) (Rinaldi-Carmona et al., 1994), the only CB1 inverse agonist to be briefly clinically marketed in Europe for the control of obesity (Moreira and Crippa, 2009), was removed from use due to its severe neuropsychiatric side effects, including mood-depressant actions (Despres et al., 2005; Traynor, 2007). The first generation of CB1 inverse agonists are commonly derived from diaryl analogs of pyrazole (e.g., SR141716A) or pyrazole bioisosteres, such as imidazole, triazole, thiazole, and pyrazoline (Lange and Kruse, 2005, 2008; Muccioli and Lambert, 2005). However, most of these are brain penetrant due to their physicochemical nature (Chorvat, 2013) and will likely generate unwanted side effects in the central nervous system. In an effort to develop new CB1 inverse agonist scaffolds, we analyzed the common pharmacophore of this class of compounds, which was proposed by Lange and Kruse (2005). This showed that the biaryl groups connecting to a central heteroaromatic ring are pivotal in forming aromatic stacking interactions with CB1 receptors (McAllister et al., 2004; Shim et al., 2012). This brought our attention to the benzhydryl piperazine scaffold, which exhibits a similar structure to the common pharmacophore of CB1 inverse agonists in lieu of the biaryl heteroaromatic ring moiety. Hence, we synthesized a group of benzhydryl piperazine analogs, and demonstrated that some of the synthesized analogs, i.e., LDK1203, LDK1222, and LDK1229, exhibited inverse agonist binding profiles for the CB1 receptor. In addition to its binding profile to the CB1 receptor, LDK1229’s inverse agonism is evident from its effect on basal as well as agonist-induced G protein coupling and its impact on the internalization and cell surface expression of CB1. Docking studies using a model of the inactive CB1 receptor showed that LDK1229 forms interactions with the receptor that were consistent with the inverse agonist SR141716A. Discovering new and improved means for inhibiting the activity of CB1 is critical for understanding the constitutive activity of CB1 and developing new therapeutic agents for treating substance addiction and disorders associated with CB1 activity.
Materials and Methods
Synthesis
The benzhydrylpiperazine analogs LDK1203, LDK1222, and LDK1229 were synthesized by alkylation, tosylation, and acylation of the 1-(4,4′-difluorobenzhydryl)piperazine (Fig. 1A). The employed 1-(4,4′-difluorobenzhydryl)piperazine (III) was prepared by monoalkylation of piperazine with 4,4′-difluorobenzhydryl chloride (II) that was obtained by halogenation of 4,4′-difluorobenzhydryl methanol (I) with oxalyl chloride, as previously reported (Weïwer et al., 2012). The obtained free bases of the benzhydrylpiperazine analogs were then converted to their corresponding hydrochloric salts by reacting with an ethereal solution of HCl. The chemical identity of the newly synthesized compounds is as follows: LDK1203 is 1-[bis(4-fluorophenyl)methyl]-4-cinnamylpiperazine dihydrochloride, LDK1222 is 1-[bis(4-fluorophenyl)methyl]-4-tosylpiperazine hydrochloride, and LDK1229 is (4-(bis(4-fluorophenyl)methyl)piperazin-1-yl)(cyclohexyl)methanone hydrochloride. The structure confirmation data for the free bases of LDK1203, LDK1222, and LDK1229 are as follows. LDK1203: 1H NMR (500 MHz, chloroform-d) δ 7.28–7.40 (m, 8H), 7.23 (t, J = 7.5 Hz, 1H), 6.99 (t, J = 7.5 Hz, 4H), 6.53 (d, J = 15 Hz, 1H), 6.28 (d, J = 15 Hz, 1H), 4.26 (s, 1H), 3.22 (d, J = 6 Hz, 2H), 2.24–2.78 (m, 8H). Mass (MS) (electron impact [EI]): m/z = 404.21 (M+). LDK1222: 1H NMR (500 MHz, chloroform-d) 7.64 (d, J = 7.9 Hz, 2H), 7.36 (d, J = 7.9 Hz, 2H), 7.22–7.31 (m, 4H), 6.94 (t, J = 8.4 Hz, 4H), 4.22 (s, 1H), 2.91–3.10 (m, 4H), 2.48 (s, 3H), 2.38–2.47 (m, 4H). MS (EI): m/z = 442.15 (M+). LDK1229: 1H NMR (500 MHz, chloroform-d) δ 7.35 (dd, J = 8.2, 5.5 Hz, 4H), 6.99 (t, J = 8.2 Hz, 4H), 4.22 (s, 1H), 3.61 (t, J = 5 Hz, 2H), 3.42 (t, J = 5 Hz, 2H), 2.43 (t, J = 5 Hz, 2H), 2.42 (t, J = 5 Hz, 2H), 2.31–2.40 (m, 4H), 1.74–1.82 (m, 2H), 1.65–1.73 (m, 3H), 1.44–1.56 (m, 2H). MS (EI): m/z = 398.2 (M+).
Fig. 1.
Compound structures. (A) Synthesis of benzhydryl piperazine analogs LDK1203, LDK1222, and LDK1229. (a) Oxalyl chloride, dichloromethane (DCM), catalytic N,N-dimethylformamide (DMF). (b) Piperazine, CH3CN. (c) 3-bromo-1-phenyl-1-propene, K2CO3, DMF. (d) TsCl, N,N-diisopropylethylamine (DIPEA), DCM. (e) Cyclohexanecarbonyl chloride, DIPEA, DCM; (f) 1.0 M HCl in ether. (B) Representative members of the first generation of CB1 inverse agonists are shown for comparison.
Plasmid Construction
All mutants were generated by site-directed mutagenesis (QuickChange Kit; Stratagene, La Jolla, CA) using the human CB1 cDNA cloned into pcDNA3.1 as a template, according to the manufacturer’s instructions. All mutations were confirmed by DNA sequencing.
CB1 Expression and Membrane Preparation
Human embryonic kidney 293 (HEK293) cells were maintained in Dulbecco’s modified Eagle’s medium supplemented with 10% fetal bovine serum and 3.5 mg/ml glucose at 37°C in 5% CO2. For transient expression of the receptors, HEK293 cells were seeded at 800,000 cells/100-mm dish on the day prior to transfection and transfected with 5–10 µg of the wild-type or mutant human CB1 receptor cloned into pcDNA3.1 using the calcium phosphate precipitation method (Chen and Okayama, 1987). At 24 hours post-transfection, membranes of transfected cells expressing either the wild-type or mutant receptors were prepared as described previously (Ahn et al., 2009).
Radioligand Binding Assay
In the homologous and heterologous competition binding experiments, approximately 7.5 μg of wild-type CB1, CB2, or mutant CB1 membrane was incubated at 30°C for 60 minutes with a fixed tracer ([3H]CP55,940, 141.2 Ci/mmol; [3H]SR141716A, 56 Ci/mmol; or [3H]WIN55,212-2, 52.2 Ci/mmol, PerkinElmer Life Sciences, Boston, MA) concentration typically at its Kd, which was determined from saturation-binding isotherms (see Results for details). Binding assays were performed with at least nine concentrations of unlabeled competitor ligand (ranging between 100 pM and 100 μM), as described previously (Ahn et al., 2012). Nonspecific binding was determined in the presence of 1 μM unlabeled CP55,940, SR141716A, or WIN55,212-2 (Tocris, Bristol, UK). Reactions were terminated by adding 300 μl of Tris-Mg2+-EDTA buffer containing 5% bovine serum albumin followed by filtration with a Brandel cell harvester (Gaithersburg, MD) through Whatman GF/C filter paper. Radioactivity was measured by liquid scintillation counting. The total assay volume and amount of membrane samples were adjusted to avoid ligand depletion by keeping the bound ligand at less than 10% of the total.
Guanosine 5′-O-(3-Thio)Triphosphate Binding Assay
Guanosine 5′-O-(3-thio)triphosphate (GTPγS) binding assays were performed as described previously (Ahn et al., 2012). Briefly, 7.5 μg of membranes were incubated for 60 minutes at 30°C in a total volume of 200 μl GTPγS binding assay buffer (50 mM Tris-HCl, pH 7.4, 3 mM MgCl2, 0.2 mM EGTA, and 100 mM NaCl), with unlabeled test compounds as indicated: 0.1 nM guanosine 5′-O-(3-[35S]thio)triphosphate ([35S]GTPγS) (1250 Ci/mmol; PerkinElmer Life Sciences, Boston, MA), 3 μM GDP (Sigma-Aldrich, St. Louis, MO), and 0.1% (w/v) bovine serum albumin. Three micromolar GDP was used to increase the window of basal activity. Nonspecific binding was determined with 10 μM unlabeled GTPγS (Sigma-Aldrich). The reaction was terminated by rapid filtration through Whatman GF/C filters. The radioactivity trapped in the filters was determined by liquid scintillation counting.
Confocal Microscopy and Image Quantification
HEK293 cells were transfected with the CB1 receptor carboxyl terminally fused to green fluorescent protein (GFP) using Lipofectamine (Invitrogen, Carlsbad, CA) according to the manufacturer’s instructions. The CB1-GFP expressing cells were seeded onto 35-mm glass-bottomed dishes (MatTek, Ashland, MA) precoated with poly(d-lysine). Cells were treated with different compounds for various lengths of time and then washed twice with phosphate-buffered saline, followed by fixation with 4% paraformaldehyde for 10 minutes at room temperature. Images were acquired using a Leica confocal laser scanning microscope (Leica, Buffalo Grove, IL), and detection of GFP was carried out following excitation at 488 nm. Quantification of the fluorescence intensity was achieved by using the Quantitative Imaging of Membrane Proteins software (http://go.warwick.ac.uk/bretschneider/quimp) (Dormann et al., 2002; Bosgraaf et al., 2009), a set of plug-ins for the open source program ImageJ (http://rsb.info.nih.gov/ij/). The Boa plug-in was used to detect the cell surface and checked against the cell edge in the transmitted image of each cell. The Ana plug-in was then used to read the cell contours produced by the Boa plug-in and compute the ratio of fluorescence intensity on the cell surface to the average intensity of the cell interior fluorescence. The results are representative of at least four independent transfections and six different images for each condition. Untransfected cells exhibited no apparent fluorescence under the experimental conditions that were used. The parameters for all of the acquired images and their consequent analysis were kept constant throughout.
Ligand and GTPγS Binding Data Analysis
All ligand-binding assays and GTPγS-binding assays were carried out in duplicate. Data are presented as the mean ± S.E. value or the mean with the corresponding 95% confidence limits from at least three independent experiments. Ki values were calculated using the Cheng-Prusoff equation (Cheng and Prusoff, 1973) based on Kd values obtained from saturation-binding analyses. The binding constants for the wild-type and mutant receptors were compared using analysis of variance followed by Bonferroni’s post hoc test for significance. P values of < 0.05 were considered to be statistically significant.
Computational Methods
Conformational Analysis of LDK1229.
To generate a library of low-energy conformers of LDK1229, the Spartan Conformation Distribution protocol was used (Wavefunction, Inc., Irvine, CA). In this protocol, the algorithm systematically searches through all rotatable bonds and ring conformations (e.g., alternate chair conformations for flexible rings). The energy of each conformer generated was calculated using the Merck Molecular Force Field (MMFF94S). This calculation yielded 68 unique conformations of LDK1229. The geometry and energy of these 68 conformations was refined by performing ab initio HF-6-31G* energy minimizations on each conformer. To calculate the difference in energy between the global minimum energy conformer and its final docked conformation, rotatable bonds in the global minimum energy conformer were driven to their corresponding value in the final docked conformation and the single-point energy of the resultant structure was calculated at the HF 6-31G* level.
Template Rationale.
Our CB1 inactive state model was initially constructed by using the 2.8-Å X-ray crystal structure of bovine rhodopsin as a template (Palczewski et al., 2000). We chose rhodopsin for several reasons: 1) Rhodopsin has an intact ionic lock (R3.50214-E/D6.30338), which is the hallmark of the class A GPCR inactive state. 2) The cannabinoid receptors and rhodopsin have very hydrophobic-binding pockets. Crystal structures reveal that the N-terminus of rhodopsin/opsin is closed over the binding pocket, preventing access from the extracellular milieu (Palczewski et al., 2000; Park et al., 2008; Scheerer et al., 2008). It is very likely that CB1, with its 112-residue N-terminus, is also closed off to the extracellular milieu. Instead, rhodopsin/opsin have been reported to have lipid portals that are used for entry and exit via the lipid bilayer for 11-cis-retinal/trans-retinal as they are shuttled into and out of the receptor (Hildebrand et al., 2009). There is evidence from simulations (Hurst et al., 2010) and from experimental covalent labeling studies (Picone et al., 2005; Pei et al., 2008) that the cannabinoid receptors also possess a portal between transmembrane helix (TMH) 6 and TMH7 through which ligands enter. 3) In addition, the cannabinoid receptors and rhodopsin share an unusual GWNC amino acid sequence motif at the extracellular end of TMH4. Here, a Trp forms an aromatic stacking interaction with Y5.39275. This interaction influences the extracellular positions of TMH3-4-5.
CB1 Inactive State Model.
A sequence alignment between the sequence of the human CB1 receptor (Gerard et al., 1991) and the sequence of bovine rhodopsin was constructed using highly conserved residues as an alignment guide. In addition, the hydrophobicity profile of the sequence was also considered when constructing the sequence alignment. Residues in the bovine rhodopsin structure were then mutated to those of human CB1. The Monte Carlo–simulated annealing technique Conformational Memories was used to sample the conformational space of TMH6 (Barnett-Norris et al., 2002). This is because TMH6 is known to undergo a functionally necessary conformational change for G protein–mediated signaling. For the inactive state model, the chosen TMH6 conformer was one that enabled the formation of a salt bridge between the highly conserved TMH6 residue D6.30338 and the highly conserved TMH3 residue R3.50214. This salt bridge (also termed the ionic lock) has been shown to be important in maintaining the inactive state in the β2-adrenergic (Ballesteros et al., 2001) and serotonin receptor type 2α (5HT-2α) receptors (Visiers et al., 2002). Extracellular and intracellular loops were added to the model using Modeller (Marcu et al., 2013). SR141716A was docked (within the TMH3-4-5-6 region) in this CB1 inactive state model, and the energy of the complex was minimized, as previously described (Hurst et al., 2006).
Glide Docking of LDK1229 at the CB1 Receptor (Inactive State Conformation).
The docking program Glide (version 5.7, Schrödinger, New York, NY), was used to explore possible receptor binding modes of LDK1229. First, the SR141716A-CB1 binding site was chosen as a starting point for Glide docking studies because of the similar pharmacology between SR141716A and LDK1229. Second, LDK1229 displaces SR141617A in competitive binding experiments, suggesting some commonality between their binding sites. Thus, Glide was used to generate a grid centered on the center of mass of our previously reported binding site for SR141716A at the CB1 receptor (Hurst et al., 2002; McAllister et al., 2003, 2004). The grid dimensions were 26 × 26 × 26 Å. This grid size allowed Glide to thoroughly explore the receptor for possible binding site(s). In addition, the results of previously reported mutagenesis and synthetic studies suggest that SR141716A forms an important interaction with K3.28192 that is necessary for its ability to act as an inverse agonist of G protein–mediated signaling (Hurst et al., 2002, 2006). Therefore, Glide was required to dock LDK1229 in such a way so that it formed a hydrogen bond with K3.28192. The only other constraint used was the requirement that LDK1229 must be docked within the exploration grid. Standard precision was selected for the docking setup. A total of 68 conformations of LDK1229 was flexibly docked using Glide. The best Glide dock was chosen for subsequent calculations. The chosen Glide dock was minimized using the minimization protocol described below.
Receptor Model Energy Minimization Protocol.
The energy of the LDK1229-CB1 complex, including loop regions, was minimized using the Optimized Potentials for Liquid Simulations 2005 force field in Macromodel 9.9 (Schrödinger). An 8.0-Å extended nonbonded cutoff (updated every 10 steps), a 20.0-Å electrostatic cutoff, and a 4.0-Å hydrogen bond cutoff were used in the calculation. The minimization was performed in two stages. In the first stage, the TMH backbone was frozen. This constraint was used to preserve the secondary structure while allowing the ligand and TMH side chains to relax. In addition, loop residues were frozen until they could be minimized using an appropriate dielectric (in the next stage of the minimization). No constraints were placed on the ligands during this stage. The minimization consisted of a conjugate gradient minimization using a distance-dependent dielectric of 2.0, performed in 1000-step increments until the bundle reached the 0.05 kJ/mol gradient. In the second stage of the calculation, the helices and ligand were frozen, but the loops were allowed to relax. The generalized Born/surface area continuum solvation model for water as implemented in Macromodel was used. This stage of the calculation consisted of a Polak-Ribier conjugate gradient minimization in 1000-step increments until the bundle reached the 0.05 kJ/mol gradient.
Results
Chemistry.
The free base forms of LDK1203, LDK1222, and LDK1229 were synthesized according to the scheme illustrated in Fig. 1A, and then purified by Combiflash chromatography (Teledyne Isco, Lincoln, NE) followed by conversion to hydrochloric salts to increase shelf life and aqueous solubility. Confirmation data for their structure by mass spectroscopy and 1H NMR are presented in Materials and Methods. The representative first-generation inverse agonists rimonabant, otenabant, taranabant, and ibipinabant are shown for comparison (Fig. 1B).
LDK1229 and Its Analogs Exhibit Inverse Agonist Binding Profiles.
To elucidate the nature of these new compounds, we performed ligand-binding studies using membrane preparations from HEK293 cells transfected with CB1. All three compounds competed with CP55,940 and SR141716A, an agonist and inverse agonist of CB1, respectively. Using [3H]CP55,940 (Kd = 2 nM) as a tracer, LDK1203, LDK1222, and LDK1229 bound wild-type CB1 with Ki values of 260, 331, and 220 nM, respectively. Similarly, while using [3H]SR141716A as a tracer (Kd = 5 nM), the compounds also bound with comparable affinities (Ki = 297, 366, and 246 nM, respectively; Tables 1–3). The structures of LDK1203 and LDK1222 differed from that of LDK1229 by the substitution groups on the piperazine ring where the piperazine ring of LDK1203 was substituted by an alkyl group and the piperazine of LDK1222 was substituted by a tosyl group.
TABLE 1.
LDK1203 and LDK1222 binding to the wild-type CB1 receptor
|
Kia |
|
|---|---|
| nM | |
| Versus [3H]CP55,940 | |
| CP55,940 | 2.56 (1.45–4.51)b |
| LDK1203 | 260 (80.7–837) |
| LDK1222 | 331 (177–619) |
| Versus [3H]SR141716A | |
| SR141716A | 5.23 (3.29–8.32)b |
| LDK1203 | 297 (192–458) |
| LDK1222 | 366 (144–928) |
Data are the median and corresponding 95% confidence limits of three independent experiments performed in duplicate. Ki values were determined from competition binding assays using [3H]CP55,940 or [3H]SR141716A as tracers at their respective Kd values, as described in Results.
Binding values are from Ahn et al. (2009) for comparison.
TABLE 3.
LDK1229 binding to the T210A, wild-type, and T210I CB1 receptors using [3H]CP55,940 as a tracer
| Kia | ||||
|---|---|---|---|---|
| CP55,940 | Ki Ratiob | LDK1229 | Ki Ratiob | |
| nM | ||||
| CB1 T210A | 7.00 (2.59–18.9) | 1:3 | NB | — |
| CB1 wild type | 2.56 (1.45–4.51)c | 1:1 | 220 (106–457) | 1:1 |
| CB1 T210I | 0.835 (0.542–1.29) | 3:1 | 5831 (907–7510) | 1:27 |
NB, no binding using up to 32 μM of LDK compound.
Data are the median and corresponding 95% confidence limits of three independent experiments performed in duplicate. Ki values were determined from competition binding assays using [3H]CP55,940 as tracer at its respective Kd values, as described in Results.
Wild type:mutant.
Binding values are from Ahn et al. (2009) for comparison.
Since LDK1229 exhibited the strongest binding affinity among the compounds, it was further investigated for its capability to bind CB1 receptors in the active and inactive states to confirm its nature as a CB1 inverse agonist. The CB1 receptor in the active and inactive states can be readily mimicked by our previously engineered mutant CB1 receptor models (T210I and T210A) (D’Antona et al., 2006). CB1 with the T210I mutation was shown to adopt a fully active form in comparison with the CB1 wild type. In contrast, CB1 with the T210A mutation adopts an inactive state. A GPCR in its active state displays enhanced affinity toward agonists but decreased affinity toward inverse agonists, whereas the inactive state of the receptor exhibits the opposite binding properties such that inverse agonists show higher binding affinity than agonists (Cotecchia et al., 1990; McWhinney et al., 2000; Wade et al., 2001; D’Antona et al., 2006). Hence, our mutant CB1 receptor models provide ideal tools to evaluate the inverse agonism of LDK1229.
As expected for an inverse agonist, using [3H]SR141716A as the tracer, the binding affinity of LDK1229 to the inactive T210A receptor was enhanced (Ki = 68 nM) relative to its affinity to the wild-type CB1 receptor (Ki = 246 nM; Table 2). A comparable trend is observed with the inverse agonist SR141716A, which binds T210A with the highest affinity (Ki = 1.47 nM) and a progressively weaker affinity for the wild-type CB1 (Ki = 5.23 nM) and then the T210I receptor (Ki = 14.7 nM). The ratio of binding affinity for the wild-type receptor relative to the mutant receptor for LDK1229 and SR141716A also reflects the preference of these two compounds for the inactive T210A receptor (Table 2).
TABLE 2.
LDK1229 binding to the T210A, wild-type, and T210I CB1 receptors using [3H]SR141716A as a tracer
| Kia | ||||
|---|---|---|---|---|
| SR141716A | Ki Ratiob | LDK1229 | Ki Ratiob | |
| nM | ||||
| CB1 T210A | 1.47 (1.14–1.90) | 4:1 | 67.4 (34.7–131) | 3:1 |
| CB1 wild type | 5.23 (3.29–8.32)c | 1:1 | 246 (164–367) | 1:1 |
| CB1 T210I | 14.7 (9.61–23.1) | 1:3 | NB | — |
NB, no binding using up to 32 μM of LDK compound.
Data are the median and corresponding 95% confidence limits of three independent experiments performed in duplicate. Ki values were determined from competition binding assays using [3H]SR141716A as tracers at its respective Kd values, as described in Results.
Wild type:mutant.
Binding values are from Ahn et al. (2009) for comparison.
Using [3H]CP55,940 as the tracer and examining binding of the agonist CP55,940, the pattern is reversed (Table 3). CP55,940 binds T210A most weakly and then progressively exhibits enhanced affinity for the wild-type CB1 and the T210I receptors. This is consistent with its agonist properties. In contrast, LDK1229 binds CB1 wild type more tightly than the T210I receptor, which is consistent with inverse agonist binding profiles (D’Antona et al., 2006). For the constitutively active T210I receptor, a large decrease in the binding affinity of LDK1229 compared with the binding affinity of the agonist CP55,940 (5831 nM versus 0.835 nM) is observed. The ratio of binding affinity for the wild-type receptor relative to the mutant receptor for LDK1229 and CP55,940 is also given for comparison (Table 3). At the extremes, when using [3H]CP55,940 as a tracer, we cannot detect any specific binding with T210A, nor can we detect any specific binding with T210I up to 32 µM of LDK1229 when using [3H]SR141716A as a tracer. Taken together, these data suggest that LDK1229 increased binding affinity to T210A over wild type, and its decreased binding affinity to T210I results from the compounds’ inverse agonist mode of action.
LDK1229 Inhibits Basal and Agonist-Induced G Protein Coupling in a Concentration-Dependent Manner.
Given the inverse agonist properties observed in the binding profile of LDK1229 to the wild-type and mutant CB1 receptors, [35S]GTPγS binding assays were performed in its presence. This assay monitors the level of G protein activation by determining the extent of binding of the nonhydrolyzable GTP analog to Gα subunits. We investigated the effects of LDK1229 on the basal G protein coupling activity levels of the wild-type CB1 receptor (Fig. 2A). Interestingly, using 1 μM of LDK1229 in the absence of CP55,940 substantially reduced the basal level of [35S]GTPγS binding from 110 to 70 fmol/mg. We also evaluated its impact on agonist CP55,940-induced [35S]GTPγS binding in the presence of various concentrations of LDK1229. We observed a progressive decrease in the specific GTPγS binding, with increasing concentration of LDK1229 up to 32 μM for the CB1 wild-type receptor (Fig. 2B).
Fig. 2.
Effect of LDK1229 and LDK1203 on the stimulation of [35S]GTPγS binding to HEK293 cell membranes expressing the CB1 wild-type receptor. (A) The basal level of [35S]GTPγS binding was measured for the CB1 wild-type receptor. The stimulatory effect of CP55,940 (CP) and the inhibitory effects of SR141716A (SR), LDK1229, and LDK1203 on [35S]GTPγS binding were measured at the indicated concentrations. Statistical significance of the differences compared with basal was assessed using one-way analysis of variance and Bonferroni’s post hoc test. ***P < 0.001. (B) The inhibitory effects of both LDK1229 and LDK1203 on CP55,940-induced [35S]GTPγS binding in membrane preparations. Statistical significance of the differences compared with 0.1 μM CP55,940 alone was assessed using one-way analysis of variance and Bonferroni’s post hoc test. †††P < 0.001. Data are presented as specific binding of GTPγS to the membranes. Nonspecific binding was determined in the presence of 10 µM unlabeled GTPγS. Each data point represents the mean ± S.E. (error bars) of at least three independent experiments performed in duplicate. The dotted line indicates the level of non-CB1–mediated GTPγS binding obtained from GTPγS binding to the mock-transfected membrane sample.
Like LDK1229, LDK1203 produced an antagonizing effect on the basal and agonist-induced levels of [35S]GTPγS and is included for comparison (Fig. 2). The antagonizing effect is consistent with the properties of an inverse agonist that promotes the inactive form of the receptor and suggests that this compound inhibits G protein coupling.
LDK1229 Promotes Cell Surface Expression of the CB1 Receptor.
Upon prolonged exposure to an agonist, GPCRs become desensitized and subsequently internalize in the cell. Inverse agonists, however, act in the opposite manner by promoting GPCR localization to the cell surface (Rinaldi-Carmona et al., 1998b; Marion et al., 2004). To assess the effect of LDK1229 on CB1 cellular localization, we determined the cellular response of CB1 upon LDK1229 treatment using confocal microscopy of cells expressing GFP-tagged CB1 receptors. Previous observations (Leterrier et al., 2004; Martini et al., 2007; Rozenfeld and Devi, 2008) indicate that CB1 is partially constitutive and that much of the wild-type CB1 receptor (∼85%) is localized on intracellular vesicles. Using the CB1-GFP chimera expressed in HEK293 cells and treated with vehicle alone (0.03% dimethylsulfoxide), we found that about 15% of CB1 is present at the plasma membrane but that the pattern of receptor fluorescence within the cells was intracellularly punctate, suggesting that a substantial proportion of receptors is internalized, which is consistent with its basal activity (Fig. 3). Treatment with 0.1 µM of the agonist CP55,940 resulted in a further shift toward internalization and a punctate appearance of the CB1-GFP receptor within the cells (data not shown). In contrast, treatment with 1 µM SR141716A or 10 μM LDK1229 resulted in approximately 20% cell surface localization evident within 3 hours and an increase to about 65% of cell surface localization after 5 hours (Fig. 3B). This is strikingly different from the cell surface localization of 13% for basal cells at 5 hours. Taken together, these data further support the inverse agonist activity of LDK1229.
Fig. 3.
Effect of LDK1229 on internalization of the CB1 receptor. (A) HEK293 cells expressing the CB1-GFP receptor were incubated with vehicle alone (0.03% dimethylsulfoxide [DMSO]), 0.5 µM SR141716A (SR), or 10 µM LDK1229. SR141716A is shown for comparison. (B) Quantification of CB1 receptors on the cell surface. The Quantitative Imaging of Membrane Proteins software with plug-ins was used as described in Materials and Methods. Images are representative of at least four independent transfections. The scale bar is 5 μm.
LDK1229 Docked in the CB1 Receptor (Inactive State) Model.
Since LDK1229 exhibited the strongest binding affinity among the compounds, Glide docking studies were performed and suggest that LDK1229 binds in the TMH3-4-5-6 region of CB1. This is the same region that forms the binding site for SR141716A at CB1. Figure 4, A and C illustrates SR141716A (shown in cyan) docked in the CB1 inactive state model. The hydrogen bonding interaction is consistent with our prior CB1 mutant cycle studies, which indicated that the amide oxygen of SR141716A interacts directly with K3.28192. This interaction is critical for the inverse agonist properties of SR141716A (Hurst et al., 2002, 2006).
Fig. 4.
Model of LDK1229 or SR141716A docked in the inactive CB1 receptor model. (A and C) illustrate SR141716A (shown in cyan) docked in the CB1 inactive state model. (B and D) illustrate the final docked conformation of LDK1229 (shown in lavender) in the CB1 inactive state model. Residues that form hydrogen bonds are shown in yellow, aromatic microdomain residues are shown in orange, and residues important for hydrophobic interactions are shown in lime. A PDB homology model illustrating the final docked conformation of LDK1229 in the CB1 inactive state model can be found in the Supplemental Data.
At its binding site, SR141716A also forms several aromatic-stacking interactions. First, the SR141716A dichlorophenyl ring forms aromatic T-stacks with F3.36200 and W5.43279 (shown in orange; see Fig. 4A). In addition, the SR141716A chlorophenyl ring forms offset parallel aromatic stacks with Y5.39275 and W5.43279 (shown in orange). In addition, SR141716A forms hydrophobic interactions with W6.48356. These aromatic interactions are consistent with our prior mutagenesis studies (McAllister et al., 2003, 2004), which indicated that SR141716A interacts within an aromatic microdomain in CB1, which is comprised of F3.36200, Y5.39275, W5.43279, and W6.48356. Finally, SR141716A forms hydrophobic interactions with L3.29193, V3.32196, and L6.51359 (shown in lime; see Fig. 4C). In the ligand-free CB1 receptor, the F3.36200/W6.48356 aromatic stacking interaction is instrumental to the maintenance of the CB1-inactive state (McAllister et al., 2004). The binding of SR141716A stabilizes the F3.36200/W6.48356 aromatic stacking interaction through the formation of an extensive network of aromatic stacks within the ligand-receptor complex (Shim et al., 2012).
Figure 4, B and D illustrates the final docked conformation of LDK1229 (shown in lavender) in the CB1 inactive state model (Supplemental Material). Like SR141716A, the LDK1229 amide oxygen forms a hydrogen bond with K3.28192 (shown in yellow). The geometry of this hydrogen bond is not optimal, however, because the hydrogen bond distance is longer than that found for SR141716A (2.8 Å versus 2.6 Å) and because the hydrogen bond angle deviates more from linearity than that formed with SR141716A (157° versus 171°). This suggests that the interaction between LDK1229 and K3.28192 is weaker than the interaction between SR141716A and K3.28192. The importance of this difference between SR141716A and LDK1229 is discussed in more detail below.
Like SR141716A, LDK1229 also docks in the CB1 TMH3-4-5-6 aromatic microdomain, but the geometry of its interactions differ somewhat from SR141716A. The fluorophenyl ring of LDK1229 forms aromatic T-stacking interactions with W6.48356 and W5.43279 (shown in orange; see Fig. 3B). In addition, the other fluorophenyl ring of LDK1229 forms offset parallel aromatic stacking interactions with Y5.39275 and W5.43279 (shown in orange). However, LDK1229 does not form an aromatic stacking interaction with F3.36200. This is due to a difference in how the two compounds are oriented within the receptor. Finally, LDK1229 forms several hydrophobic interactions with residues F3.36200 (shown in orange), L3.29193, V3.32196, and L6.51359 (shown in lime).
Mutational Analysis of Computationally Predicted Residues in the LDK1229 Binding Pocket.
To test the LDK1229 binding-site hypothesis generated by the receptor model, K3.28192A, W5.43279A, W6.48356A, and C7.42386M mutant CB1 receptors were generated. Our results showed that the K3.28192A mutation resulted in total ablation of [3H]CP55,940 binding at wild-type CB1 and that this residue is critical for CP55,940 binding consistent with previous data (Song and Bonner, 1996). Therefore, competition binding assays on the K3.28192A mutant were performed using [3H]WIN55,212-2 as a tracer since [3H]WIN55,212-2 binding to the K3.28192A mutant was not significantly different than its binding to the wild-type receptor (Kd = 8.6 nM versus 4.5 nM). Competition binding analysis revealed that the binding of LDK1229 (Ki = 1316 nM) was affected 6-fold by this mutation compared with wild type (Ki = 324 nM; Table 4), suggesting that this residue may be important for a hydrogen bonding interaction with LDK1229. These results alone cannot tell us the specific region on LDK1229 with which K3.28192 interacts, but modeling studies reported here suggest that K3.28192 hydrogen bonds with the carboxyl oxygen of LDK1229. The K3.28192A mutation results reported here are consistent with previous CB1 K3.28192A mutation studies, which showed that K3.28192A is an important interaction site (Hurst et al., 2002) for the carboxamide oxygen of SR141716A (Hurst et al., 2006).
TABLE 4.
LDK1229 binding to the wild-type or mutant CB1 receptors using [3H]CP55,940 or [3H]WIN55,212-2 as a tracer
| Receptor | [3H]CP55,940 | |
|---|---|---|
| Kia | Ki Ratiob | |
| nM | ||
| CB1 wild type | 220 (106–457) | 1:1 |
| K3.28192A | 1316 (1115–1626)c | 1:4c |
| W5.43279A | ND | — |
| W6.48356A | 1987 (895–2736) | 1:9 |
| C7.42386M | 1010 (832–1154) | 1:5 |
ND, not detectable; 39% displacement at 32 µM.
Data are the median and corresponding 95% confidence limits of three independent experiments performed in duplicate. Ki values were determined from competition binding assays using [3H]CP55,940 as tracer at its respective Kd values, as described in Results.
Wild type:mutant.
Binding was performed with [3H]WIN55,212-2 as a tracer. LDK1229 binding to CB1 wild type using [3H]WIN55,212-2 as a tracer yielded a Ki of 324 nM (205–524).
Saturation (equilibrium) binding analysis for [3H]CP55,940 at the other three mutant receptors, W5.43279A, W6.48356A, and C7.42386M, showed that the affinity parameters obtained for the mutant receptors were not significantly different from the Kd at the wild-type CB1 receptor (Kd for mutants = 6, 4, and 3 nM, respectively), suggesting that these receptors were properly folded. Therefore, competition binding assays on these mutants were performed using [3H]CP55,940 as a tracer. The W5.43279A mutation had the most profound effect on LDK1229 binding compared with any of the other mutations and showed only 39% displacement of [3H]CP55,940 when using 32 µM of LDK1229. This result is consistent with the modeling studies that suggest that W5.43279A is central in the formation of the aromatic T-stacking interactions with the two fluorophenyl rings in LDK1229. The W6.48356A mutant affected LDK1229 binding 9-fold (Ki = 1987 nM) and the C7.42386M mutant 5-fold (Ki = 1010 nM; Table 4). The result for the W6.48356A mutation further supports the modeling results, which suggest that this residue participates in direct aromatic stacking interactions with one of the fluorophenyl rings of LDK1229, consistent with prior studies (McAllister et al., 2003). As shown in Table 4, we found here that enlarging the residue at position 7.42 via the C7.42386M mutation results in a 5-fold loss in affinity for LDK1229, which is consistent with previous studies by Farrens and colleagues (Fay et al., 2005). Taken together, the results of all mutation studies are consistent with the modeling results reported here, specifically that LDK1229 occupies the same general binding region as SR1417167A.
LDK1229 Has Diminished Activity for CB2.
Based on both binding and functional data, LDK1229 showed some selectivity for the CB1 receptor over the CB2 receptor (Fig. 5). This is evident by LDK1229 reducing the binding affinity 3-fold (Ki= 633 nM; Fig. 5A) to the CB2 receptor compared with the CB1 receptor (Ki= 220 nM; Table 3) using [3H]CP55,940 as a tracer. This selectivity is also evident by the small magnitude of the reduction in basal and agonist-induced G protein coupling using CB2. Using 1 μM of LDK1229 in the absence of CP55,940 only reduced the basal level of [35S]GTPγS binding from 95 to 88 fmol/mg (Fig. 5B) compared with the 2-fold decrease observed with CB1 (Fig. 2A). We also observed a small decrease in the CP55,940-induced [35S]GTPγS binding with an increasing concentration of LDK1229 (Fig. 4B), with up to 32 μM of LDK1229 (to 110 fmol/mg from the original 120 fmol/mg). The CB2 selective inverse agonist N-[(1S)-endo-1,3,3-trimethylbicycloheptan-2-yl)-5-(4-chloro-3-methylphenyl)-1-(4-methylbenzyl)-pyrazole-3-carboxamide] (SR144528) (Rinaldi-Carmona et al., 1998a) antagonized the basal level of [35S]GTPγS binding and is shown for comparison. We also found that LDK1229 can dock in the CB2 inactive state model but in a higher energy conformation (data not shown). This is likely the origin of the 3-fold loss of affinity at CB2.
Fig. 5.
Binding parameters of LDK1229 and LDK1203 to CB2. (A) Binding of LDK1229 and LDK1203 to CB2 wild-type receptors and (B) their effect on the stimulation of [35S]GTPγS binding. Statistical significance of the differences (*compared with basal and †compared with 0.1 μM CP55,940 alone) was assessed using one-way analysis of variance and Bonferroni’s post hoc test. ***P < 0.001; ††P < 0.01.
Discussion
In an effort to develop new modulators of the CB1 receptor, we synthesized a group of benzhydryl piperazine analogs, including the compounds LDK1203, LDK1222, and LDK1229, and describe their inverse agonist properties in this study. In addition to their inverse agonist binding profiles to the CB1 receptor and their preference to bind the inactive T210A CB1 receptor over the constitutively active wild-type CB1 or fully active T210I receptor, the inverse agonism exhibited by LDK1229 was also evident from its antagonistic effect on basal and agonist-induced G protein coupling and its ability to increase the CB1 localization to the cell surface. LDK1229 exhibited a lower affinity for the CB2 receptor, with a 3-fold relative selectivity for the CB1 receptor. Because the CB1 receptor is constitutively active both in vitro and in vivo (Landsman et al., 1997; Meschler et al., 2000), discovering new and improved means for inhibiting the activity of the receptor is therapeutically useful and relevant for modulating activity of the CB1 receptor system in the brain.
Our results show that the benzhydryl piperazine analogs represented by LDK1229 behave as inverse agonists of the CB1 receptor. Structurally, the benzhydryl piperazine analogs are distinct from the first generation of CB1 inverse agonists, which generally possess nitrogen-containing five- or six-member aromatic rings as their central connecting units (Lange and Kruse, 2005; Vemuri et al., 2008; Chorvat, 2013). In contrast, benzhydryl piperazine analogs have a central core of piperazine, which is nonaromatic and possesses basic amino group(s). A pharmacophore model of the first generation of CB1 inverse agonists was proposed (Lange and Kruse, 2005), of which the biaryls (e.g., SR141716A) form favorable aromatic stacking interactions with two subpockets surrounded by residues Y5.39275-W4.64255-F5.42278 and residues W5.43279-F3.36200-W6.48356 of CB1. The central core (e.g., pyrazole in SR141716A) then connects to a lipophilic moiety through a hydrogen bond acceptor (e.g., the carbonyl of SR141716A). The hydrogen bond acceptor stabilizes the D6.58366-K3.28192 salt bridge in the inactive state of the CB1 receptor. The lipophilic moiety (e.g., the methylene groups of piperidine ring in SR141716A) fits in a pocket formed by V3.32196-F2.57170-L7.43387 and M7.44384 of the CB1 receptor (Lange and Kruse, 2005). In spite of the structural difference between our benzhydryl piperazine analogs and the first generation of CB1 inverse agonists, docking SR141716A and LDK1229 into the CB1 inactive state model suggest that both compounds bind in similar receptor regions and form similar interactions with the receptor. This is also consistent with the results of competitive displacement assays that suggest LDK1229 displaces SR141716A, implying an overlapping binding site. However, the results of the docking studies suggest one major difference between SR141716A and LDK1229. The geometry of the SR141716A hydrogen bond with K3.28192 is much better than that of LDK1229. Conformational analysis of LDK1229 suggests that it does not have the conformational freedom to adopt a conformation that would allow it to improve its hydrogen bond geometry. In addition, the extra ring hydrogen (cyclohexyl ring versus piperidine ring) introduced in LDK1229 forces the ligand to position itself differently. Together, these effects lead to a less favorable hydrogen bond with K3.28192. This is consistent with the results of the binding experiments that show that SR141716A has a higher affinity for CB1 than does LDK1229. Notably, LDK1229 exhibits an efficacy comparable to SR141716A in antagonizing basal GTPγS binding to the CB1 receptor, although it shows a weaker binding affinity to the receptor (Fig. 2; Tables 2 and 3). One advantage of the benzhydryl piperazine analogs is that the piperazine ring provides two amino groups that can be readily used in further derivatization. This opens up rich opportunities for structural modifications that may lead to the improvement of the pharmacokinetic properties of the drug molecules, such as their lipophilicity and polar surface area, which are critical factors influencing the brain barrier penetration of these compounds.
Evidence has begun to emerge on the involvement of the endocannabinoid system in the regulation of metabolism in several peripheral organs crucial to energy storage and utilization (Di Marzo and Matias, 2005; Silvestri et al., 2011). Elevated peripheral endocannabinoid levels and increased CB1 expression in these tissues have been observed in several studies of obese mice (Pagotto et al., 2006) as well as obese humans compared with leaner controls (Engeli et al., 2005). In light of the above, it has proven difficult to diminish the adverse central nervous system effects of brain-penetrant CB1 inverse agonists (Jones, 2008). Not surprisingly, significant effort has been put forth to develop CB1 neutral antagonists (Janero, 2012) or peripherally restricted CB1 inverse agonists (Silvestri and Di Marzo, 2012; Chorvat, 2013) that do not produce undesired central nervous system side effects. LDK1229 suggests a new lead for developing novel CB1 inverse agonists and is attractive for the design and synthesis of peripherally active CB1 inverse agonists, a class of potential therapeutic agents for the treatment of obesity and other related metabolic syndromes. In general, there are several strategies to reduce a ligand’s permeability to the blood brain barrier (Clark 2005). These include increasing H-bonding capacity, molecular weight, and polar surface area as well as introducing acidic functional groups or increasing hydrophilicity. In comparison with the conventional scaffold of CB1 inverse agonists (i.e., biaryl substituted heteroaromatic ring), the benzhydryl piperazine scaffold offers two basic nitrogen atoms, which provide extra opportunities to increase the overall H-bonding capability. Additionally, the two basic nitrogen functionalities are considerably easy to modify to introduce various substitutions so that the molecular weight and polar surface area properties can be readily manipulated to gain suitable pharmacokinetic properties that reduce blood brain barrier penetration. Hence, this scaffold enriches the chances for pharmacokinetic-driven drug optimization to achieve the desired peripheral restriction of CB1 inverse agonists.
Supplementary Material
Acknowledgments
The authors thank Sushma Samala and Hamed I. Ali for providing some technical assistance in the synthesis and chemical characterization of the benzhydryl piperazine analogs.
Abbreviations
- [35S]GTPγS
guanosine 5′-O-(3-[35S]thio)triphosphate
- CB1
cannabinoid receptor type 1
- CB2
cannabinoid receptor type 2
- CP55,940
5-(1,1-dimethylheptyl)-2-(5-hydroxy-2-(3-hydroxypropyl)cyclohexyl)phenol
- EI
electron impact
- GFP
green fluorescent protein
- GPCR
G-protein–coupled receptor
- GTPγS
guanosine 5′-O-(3-thio)triphosphate
- HEK293
human embryonic kidney 293 cells
- MS
mass
- SR141716A
rimonabant
- TMH
transmembrane helix
- WIN55,212-2
(R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl)pyrrolo-[1,2,3-d,e]-1,4-benzoxazin-6-yl]-1-naphthalenyl-methanone
Authorship contributions
Participated in research design: Mahmoud, Kendall, Lu, Olszewska, Shore, Hurst, Reggio.
Conducted experiments: Mahmoud, Olszewska, Liu, Lu, Shore.
Performed data analysis: Mahmoud, Kendall, Olszewska, Lu, Shore, Hurst, Reggio.
Wrote or contributed to the writing of the manuscript: Mahmoud, Lu, Kendall, Shore, Hurst, Reggio.
Footnotes
This work was supported in part by the National Institutes of Health [Grants R01-DA020763, R01-DA003934, and K05-DA021358] and Faculty Development Fund of Texas A&M Health Sciences Center. The authors declare no conflict of interest.
This article has supplemental material available at molpharm.aspetjournals.org.
References
- Ahn KH, Bertalovitz AC, Mierke DF, Kendall DA. (2009) Dual role of the second extracellular loop of the cannabinoid receptor 1: ligand binding and receptor localization. Mol Pharmacol 76:833–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ahn KH, Mahmoud MM, Kendall DA. (2012) Allosteric modulator ORG27569 induces CB1 cannabinoid receptor high affinity agonist binding state, receptor internalization, and Gi protein-independent ERK1/2 kinase activation. J Biol Chem 287:12070–12082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ballesteros JA, Jensen AD, Liapakis G, Rasmussen SG, Shi L, Gether U, Javitch JA. (2001) Activation of the beta 2-adrenergic receptor involves disruption of an ionic lock between the cytoplasmic ends of transmembrane segments 3 and 6. J Biol Chem 276:29171–29177. [DOI] [PubMed] [Google Scholar]
- Barnett-Norris J, Hurst DP, Buehner K, Ballesteros JA, Guarnieri F, Reggio PH. (2002) Agonist alkyl tail interaction with cannabinoid CB1 receptor V6.43/I6.46 groove induces a helix 6 active conformation. Int J Quantum Chem 88:76–86. [Google Scholar]
- Basu S, Dittel BN. (2011) Unraveling the complexities of cannabinoid receptor 2 (CB2) immune regulation in health and disease. Immunol Res 51:26–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bátkai S, Járai Z, Wagner JA, Goparaju SK, Varga K, Liu J, Wang L, Mirshahi F, Khanolkar AD, Makriyannis A, et al. (2001) Endocannabinoids acting at vascular CB1 receptors mediate the vasodilated state in advanced liver cirrhosis. Nat Med 7:827–832. [DOI] [PubMed] [Google Scholar]
- Bosgraaf L, van Haastert PJ, Bretschneider T. (2009) Analysis of cell movement by simultaneous quantification of local membrane displacement and fluorescent intensities using Quimp2. Cell Motil Cytoskeleton 66:156–165. [DOI] [PubMed] [Google Scholar]
- Bouaboula M, Poinot-Chazel C, Marchand J, Canat X, Bourrié B, Rinaldi-Carmona M, Calandra B, Le Fur G, Casellas P. (1996) Signaling pathway associated with stimulation of CB2 peripheral cannabinoid receptor. Involvement of both mitogen-activated protein kinase and induction of Krox-24 expression. Eur J Biochem 237:704–711. [DOI] [PubMed] [Google Scholar]
- Chen C, Okayama H. (1987) High-efficiency transformation of mammalian cells by plasmid DNA. Mol Cell Biol 7:2745–2752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng Y, Prusoff WH. (1973) Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem Pharmacol 22:3099–3108. [DOI] [PubMed] [Google Scholar]
- Chorvat RJ. (2013) Peripherally restricted CB1 receptor blockers. Bioorg Med Chem Lett 23:4751–4760. [DOI] [PubMed] [Google Scholar]
- Clark DE. (2005) Computational prediction of blood-brain barrier permeation. Annu Rep Med Chem 40:403–415. [Google Scholar]
- Cotecchia S, Exum S, Caron MG, Lefkowitz RJ. (1990) Regions of the alpha 1-adrenergic receptor involved in coupling to phosphatidylinositol hydrolysis and enhanced sensitivity of biological function. Proc Natl Acad Sci USA 87:2896–2900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Croci T, Manara L, Aureggi G, Guagnini F, Rinaldi-Carmona M, Maffrand JP, Le Fur G, Mukenge S, Ferla G. (1998) In vitro functional evidence of neuronal cannabinoid CB1 receptors in human ileum. Br J Pharmacol 125:1393–1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- D’Antona AM, Ahn KH, Kendall DA. (2006) Mutations of CB1 T210 produce active and inactive receptor forms: correlations with ligand affinity, receptor stability, and cellular localization. Biochemistry 45:5606–5617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Lean A, Stadel JM, Lefkowitz RJ. (1980) A ternary complex model explains the agonist-specific binding properties of the adenylate cyclase-coupled beta-adrenergic receptor. J Biol Chem 255:7108–7117. [PubMed] [Google Scholar]
- Després JP, Golay A, Sjöström L, Rimonabant in Obesity-Lipids Study Group (2005) Effects of rimonabant on metabolic risk factors in overweight patients with dyslipidemia. N Engl J Med 353:2121–2134. [DOI] [PubMed] [Google Scholar]
- Di Marzo V, Matias I. (2005) Endocannabinoid control of food intake and energy balance. Nat Neurosci 8:585–589. [DOI] [PubMed] [Google Scholar]
- Dormann D, Libotte T, Weijer CJ, Bretschneider T. (2002) Simultaneous quantification of cell motility and protein-membrane-association using active contours. Cell Motil Cytoskeleton 52:221–230. [DOI] [PubMed] [Google Scholar]
- Engeli S, Böhnke J, Feldpausch M, Gorzelniak K, Janke J, Bátkai S, Pacher P, Harvey-White J, Luft FC, Sharma AM, et al. (2005) Activation of the peripheral endocannabinoid system in human obesity. Diabetes 54:2838–2843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fay JF, Dunham TD, Farrens DL. (2005) Cysteine residues in the human cannabinoid receptor: only C257 and C264 are required for a functional receptor, and steric bulk at C386 impairs antagonist SR141716A binding. Biochemistry 44:8757–8769. [DOI] [PubMed] [Google Scholar]
- Galiègue S, Mary S, Marchand J, Dussossoy D, Carrière D, Carayon P, Bouaboula M, Shire D, Le Fur G, Casellas P. (1995) Expression of central and peripheral cannabinoid receptors in human immune tissues and leukocyte subpopulations. Eur J Biochem 232:54–61. [DOI] [PubMed] [Google Scholar]
- Gérard CM, Mollereau C, Vassart G, Parmentier M. (1991) Molecular cloning of a human cannabinoid receptor which is also expressed in testis. Biochem J 279:129–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gether U, Kobilka BK. (1998) G protein-coupled receptors. II. Mechanism of agonist activation. J Biol Chem 273:17979–17982. [DOI] [PubMed] [Google Scholar]
- Ghanouni P, Steenhuis JJ, Farrens DL, Kobilka BK. (2001) Agonist-induced conformational changes in the G-protein-coupling domain of the beta 2 adrenergic receptor. Proc Natl Acad Sci USA 98:5997–6002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hildebrand PW, Scheerer P, Park JH, Choe HW, Piechnick R, Ernst OP, Hofmann KP, Heck M. (2009) A ligand channel through the G protein coupled receptor opsin. PLoS ONE 4:e4382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Howlett AC. (1995) Pharmacology of cannabinoid receptors. Annu Rev Pharmacol Toxicol 35:607–634. [DOI] [PubMed] [Google Scholar]
- Howlett AC. (2005) Cannabinoid receptor signaling. Handb Exp Pharmacol 168:53–79. [DOI] [PubMed] [Google Scholar]
- Howlett AC, Breivogel CS, Childers SR, Deadwyler SA, Hampson RE, Porrino LJ. (2004) Cannabinoid physiology and pharmacology: 30 years of progress. Neuropharmacology 47 (Suppl 1):345–358. [DOI] [PubMed] [Google Scholar]
- Howlett AC, Fleming RM. (1984) Cannabinoid inhibition of adenylate cyclase. Pharmacology of the response in neuroblastoma cell membranes. Mol Pharmacol 26:532–538. [PubMed] [Google Scholar]
- Hurst D, Umejiego U, Lynch D, Seltzman H, Hyatt S, Roche M, McAllister S, Fleischer D, Kapur A, Abood M, et al. (2006) Biarylpyrazole inverse agonists at the cannabinoid CB1 receptor: importance of the C-3 carboxamide oxygen/lysine3.28(192) interaction. J Med Chem 49:5969–5987. [DOI] [PubMed] [Google Scholar]
- Hurst DP, Grossfield A, Lynch DL, Feller S, Romo TD, Gawrisch K, Pitman MC, Reggio PH. (2010) A lipid pathway for ligand binding is necessary for a cannabinoid G protein-coupled receptor. J Biol Chem 285:17954–17964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hurst DP, Lynch DL, Barnett-Norris J, Hyatt SM, Seltzman HH, Zhong M, Song ZH, Nie J, Lewis D, Reggio PH. (2002) N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide (SR141716A) interaction with LYS 3.28(192) is crucial for its inverse agonism at the cannabinoid CB1 receptor. Mol Pharmacol 62:1274–1287. [DOI] [PubMed] [Google Scholar]
- Janero DR. (2012) Cannabinoid-1 receptor (CB1R) blockers as medicines: beyond obesity and cardiometabolic disorders to substance abuse/drug addiction with CB1R neutral antagonists. Expert Opin Emerg Drugs 17:17–29. [DOI] [PubMed] [Google Scholar]
- Janero DR, Makriyannis A. (2009) Cannabinoid receptor antagonists: pharmacological opportunities, clinical experience, and translational prognosis. Expert Opin Emerg Drugs 14:43–65. [DOI] [PubMed] [Google Scholar]
- Jones D. (2008) End of the line for cannabinoid receptor 1 as an anti-obesity target? Nat Rev Drug Discov 7:961–962. [DOI] [PubMed] [Google Scholar]
- Kenakin T. (1995) Agonist-receptor efficacy. II. Agonist trafficking of receptor signals. Trends Pharmacol Sci 16:232–238. [DOI] [PubMed] [Google Scholar]
- Landsman RS, Burkey TH, Consroe P, Roeske WR, Yamamura HI. (1997) SR141716A is an inverse agonist at the human cannabinoid CB1 receptor. Eur J Pharmacol 334:R1–R2. [DOI] [PubMed] [Google Scholar]
- Lange JH, Kruse CG. (2005) Keynote review: medicinal chemistry strategies to CB1 cannabinoid receptor antagonists. Drug Discov Today 10:693–702. [DOI] [PubMed] [Google Scholar]
- Lange JH, Kruse CG. (2008) Cannabinoid CB1 receptor antagonists in therapeutic and structural perspectives. Chem Rec 8:156–168. [DOI] [PubMed] [Google Scholar]
- Leterrier C, Bonnard D, Carrel D, Rossier J, Lenkei Z. (2004) Constitutive endocytic cycle of the CB1 cannabinoid receptor. J Biol Chem 279:36013–36021. [DOI] [PubMed] [Google Scholar]
- Mackie K. (2006) Cannabinoid receptors as therapeutic targets. Annu Rev Pharmacol Toxicol 46:101–122. [DOI] [PubMed] [Google Scholar]
- Marcu J, Shore DM, Kapur A, Trznadel M, Makriyannis A, Reggio PH, Abood ME. (2013) Novel insights into CB1 cannabinoid receptor signaling: a key interaction identified between the extracellular-3 loop and transmembrane helix 2. J Pharmacol Exp Ther 345:189–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marion S, Weiner DM, Caron MG. (2004) RNA editing induces variation in desensitization and trafficking of 5-hydroxytryptamine 2c receptor isoforms. J Biol Chem 279:2945–2954. [DOI] [PubMed] [Google Scholar]
- Martini L, Waldhoer M, Pusch M, Kharazia V, Fong J, Lee JH, Freissmuth C, Whistler JL. (2007) Ligand-induced down-regulation of the cannabinoid 1 receptor is mediated by the G-protein-coupled receptor-associated sorting protein GASP1. FASEB J 21:802–811. [DOI] [PubMed] [Google Scholar]
- McAllister SD, Hurst DP, Barnett-Norris J, Lynch D, Reggio PH, Abood ME. (2004) Structural mimicry in class A G protein-coupled receptor rotamer toggle switches: the importance of the F3.36(201)/W6.48(357) interaction in cannabinoid CB1 receptor activation. J Biol Chem 279:48024–48037. [DOI] [PubMed] [Google Scholar]
- McAllister SD, Rizvi G, Anavi-Goffer S, Hurst DP, Barnett-Norris J, Lynch DL, Reggio PH, Abood ME. (2003) An aromatic microdomain at the cannabinoid CB(1) receptor constitutes an agonist/inverse agonist binding region. J Med Chem 46:5139–5152. [DOI] [PubMed] [Google Scholar]
- McWhinney C, Wenham D, Kanwal S, Kalman V, Hansen C, Robishaw JD. (2000) Constitutively active mutants of the alpha(1a)- and the alpha(1b)-adrenergic receptor subtypes reveal coupling to different signaling pathways and physiological responses in rat cardiac myocytes. J Biol Chem 275:2087–2097. [DOI] [PubMed] [Google Scholar]
- Meschler JP, Kraichely DM, Wilken GH, Howlett AC. (2000) Inverse agonist properties of N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2, 4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide HCl (SR141716A) and 1-(2-chlorophenyl)-4-cyano-5-(4-methoxyphenyl)-1H-pyrazole-3-carboxyl ic acid phenylamide (CP-272871) for the CB(1) cannabinoid receptor. Biochem Pharmacol 60:1315–1323. [DOI] [PubMed] [Google Scholar]
- Moreira FA, Crippa JA. (2009) The psychiatric side-effects of rimonabant. Rev Bras Psiquiatr 31:145–153. [DOI] [PubMed] [Google Scholar]
- Muccioli GG, Lambert DM. (2005) Current knowledge on the antagonists and inverse agonists of cannabinoid receptors. Curr Med Chem 12:1361–1394. [DOI] [PubMed] [Google Scholar]
- Pagotto U, Marsicano G, Cota D, Lutz B, Pasquali R. (2006) The emerging role of the endocannabinoid system in endocrine regulation and energy balance. Endocr Rev 27:73–100. [DOI] [PubMed] [Google Scholar]
- Palczewski K, Kumasaka T, Hori T, Behnke CA, Motoshima H, Fox BA, Le Trong I, Teller DC, Okada T, Stenkamp RE, et al. (2000) Crystal structure of rhodopsin: a G protein-coupled receptor. Science 289:739–745. [DOI] [PubMed] [Google Scholar]
- Park JH, Scheerer P, Hofmann KP, Choe HW, Ernst OP. (2008) Crystal structure of the ligand-free G-protein-coupled receptor opsin. Nature 454:183–187. [DOI] [PubMed] [Google Scholar]
- Pei Y, Mercier RW, Anday JK, Thakur GA, Zvonok AM, Hurst D, Reggio PH, Janero DR, Makriyannis A. (2008) Ligand-binding architecture of human CB2 cannabinoid receptor: evidence for receptor subtype-specific binding motif and modeling GPCR activation. Chem Biol 15:1207–1219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pertwee RG. (2005) Inverse agonism and neutral antagonism at cannabinoid CB1 receptors. Life Sci 76:1307–1324. [DOI] [PubMed] [Google Scholar]
- Pertwee RG. (2006) Cannabinoid pharmacology: the first 66 years. Br J Pharmacol 147 (Suppl 1):S163–S171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Picone RP, Khanolkar AD, Xu W, Ayotte LA, Thakur GA, Hurst DP, Abood ME, Reggio PH, Fournier DJ, Makriyannis A. (2005) (-)-7′-Isothiocyanato-11-hydroxy-1′,1′-dimethylheptylhexahydrocannabinol (AM841), a high-affinity electrophilic ligand, interacts covalently with a cysteine in helix six and activates the CB1 cannabinoid receptor. Mol Pharmacol 68:1623–1635. [DOI] [PubMed] [Google Scholar]
- Rinaldi-Carmona M, Barth F, Héaulme M, Shire D, Calandra B, Congy C, Martinez S, Maruani J, Néliat G, Caput D, et al. (1994) SR141716A, a potent and selective antagonist of the brain cannabinoid receptor. FEBS Lett 350:240–244. [DOI] [PubMed] [Google Scholar]
- Rinaldi-Carmona M, Barth F, Millan J, Derocq JM, Casellas P, Congy C, Oustric D, Sarran M, Bouaboula M, Calandra B, et al. (1998a) SR 144528, the first potent and selective antagonist of the CB2 cannabinoid receptor. J Pharmacol Exp Ther 284:644–650. [PubMed] [Google Scholar]
- Rinaldi-Carmona M, Le Duigou A, Oustric D, Barth F, Bouaboula M, Carayon P, Casellas P, Le Fur G. (1998b) Modulation of CB1 cannabinoid receptor functions after a long-term exposure to agonist or inverse agonist in the Chinese hamster ovary cell expression system. J Pharmacol Exp Ther 287:1038–1047. [PubMed] [Google Scholar]
- Rozenfeld R, Devi LA. (2008) Regulation of CB1 cannabinoid receptor trafficking by the adaptor protein AP-3. FASEB J 22:2311–2322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scheerer P, Park JH, Hildebrand PW, Kim YJ, Krauss N, Choe HW, Hofmann KP, Ernst OP. (2008) Crystal structure of opsin in its G-protein-interacting conformation. Nature 455:497–502. [DOI] [PubMed] [Google Scholar]
- Shim JY, Bertalovitz AC, Kendall DA. (2012) Probing the interaction of SR141716A with the CB1 receptor. J Biol Chem 287:38741–38754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Silvestri C, Di Marzo V. (2012) Second generation CB1 receptor blockers and other inhibitors of peripheral endocannabinoid overactivity and the rationale of their use against metabolic disorders. Expert Opin Investig Drugs 21:1309–1322. [DOI] [PubMed] [Google Scholar]
- Silvestri C, Ligresti A, Di Marzo V. (2011) Peripheral effects of the endocannabinoid system in energy homeostasis: adipose tissue, liver and skeletal muscle. Rev Endocr Metab Disord 12:153–162. [DOI] [PubMed] [Google Scholar]
- Slipetz DM, O’Neill GP, Favreau L, Dufresne C, Gallant M, Gareau Y, Guay D, Labelle M, Metters KM. (1995) Activation of the human peripheral cannabinoid receptor results in inhibition of adenylyl cyclase. Mol Pharmacol 48:352–361. [PubMed] [Google Scholar]
- Song ZH, Bonner TI. (1996) A lysine residue of the cannabinoid receptor is critical for receptor recognition by several agonists but not WIN55212-2. Mol Pharmacol 49:891–896. [PubMed] [Google Scholar]
- Traynor K. (2007) Panel advises against rimonabant approval. Am J Health Syst Pharm 64:1460–1461. [DOI] [PubMed] [Google Scholar]
- Turu G, Hunyady L. (2010) Signal transduction of the CB1 cannabinoid receptor. J Mol Endocrinol 44:75–85. [DOI] [PubMed] [Google Scholar]
- Vemuri VK, Janero DR, Makriyannis A. (2008) Pharmacotherapeutic targeting of the endocannabinoid signaling system: drugs for obesity and the metabolic syndrome. Physiol Behav 93:671–686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Visiers I, Ebersole BJ, Dracheva S, Ballesteros J, Sealfon SC, Weinstein H. (2002) Structural motifs as functional microdomains in G-protein-coupled receptors: energetic considerations in the mechanism of activation of the serotonin 5-HT2A receptor by disruption of the ionic lock of the arginine cage. Int J Quantum Chem 88:65–75. [Google Scholar]
- Wade SM, Lan K, Moore DJ, Neubig RR. (2001) Inverse agonist activity at the alpha(2A)-adrenergic receptor. Mol Pharmacol 59:532–542. [DOI] [PubMed] [Google Scholar]
- Weïwer M, Bittker JA, Lewis TA, Shimada K, Yang WS, MacPherson L, Dandapani S, Palmer M, Stockwell BR, Schreiber SL, et al. (2012) Development of small-molecule probes that selectively kill cells induced to express mutant RAS. Bioorg Med Chem Lett 22:1822–1826. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.





