Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Nov 22.
Published in final edited form as: J Recept Signal Transduct Res. 2024 Nov 22;44(4):151–159. doi: 10.1080/10799893.2024.2431986

The Cannabinoid CB2 Receptor Positive Allosteric Modulator EC21a Exhibits Complicated Pharmacology In Vitro

Aidong Qi 1, Xueqing Han 2, Marc Quitalig 3, Jessica Wu 4, Plamen P Christov 5, KyuOk Jeon 6, Somnath Jana 7, Kwangho Kim 8, Darren W Engers 9, Craig W Lindsley 10,11, Alice L Rodriguez 12, Colleen M Niswender 13,14,15
PMCID: PMC11636628  NIHMSID: NIHMS2038606  PMID: 39575892

Abstract

Schizophrenia is a complex disease involving the dysregulation of numerous brain circuits and patients exhibit positive symptoms (hallucinations, delusions), negative symptoms (anhedonia), and cognitive impairments. We have shown that the antipsychotic efficacy of positive allosteric modulators (PAMs) of both the M4 muscarinic receptor and metabotropic glutamate receptor 1 (mGlu1) involve the retrograde activation of the presynaptic cannabinoid type-2 (CB2) receptor, indicating that CB2 activation or potentiation could result in a novel therapeutic strategy for schizophrenia. We used two complementary assays, receptor-mediated phosphoinositide hydrolysis and GIRK channel activation, to characterize a CB2 PAM scaffold, represented by the compound EC21a, to explore its potential as a starting point to optimize therapeutics for schizophrenia. These studies revealed that EC21a acts as an allosteric inverse agonist at CB2 in both assays and exhibits a mixed allosteric agonist/negative allosteric modulator profile at CB1 depending upon the assay used for profiling. A series of compounds related to EC21a also functioned as CB2 inverse agonists. Overall, these results suggest that EC21a exhibits complicated and potentially assay-dependent pharmacology, which may impact interpretation of in vivo studies.

Keywords: Cannabinoid receptors, Allosteric modulator, Assay development, Dopamine signaling, Schizophrenia

Graphical Abstract

graphic file with name nihms-2038606-f0001.jpg

Introduction

The endocannabinoid system is composed of two main receptor subtypes, cannabinoid type-1 (CB1) and type-2 receptors (CB2), which belong to the Class A family of G Protein-Coupled Receptors (GPCRs; [1,2]). While CB1 is the most widely expressed GPCR in the brain [3], CB2 exhibits high expression in the immune system [46] and CB2 receptors are important in immune responses and neuroinflammation via modulation of microglia [710]. CB2 receptors are also expressed in neuronal circuits and evidence suggests therapeutic potential for CB2 modulation in the treatment of several brain disorders including Alzheimer’s disease, pain and epilepsy (reviewed in [11]). Recently, we have found that cholinergic signaling regulates dopamine release in the striatum via a CB2-dependent mechanism that involves the activation of M4 muscarinic receptors and metabotropic glutamate receptor 1 (mGlu1) [12,13]. Potentiation of M4 or mGlu1 with highly selective positive allosteric modulators (PAMs) induces antipsychotic-like activity; a mechanism key to these effects involves the retrograde signaling of the endocannabinoid, 2-AG, which acts on CB2 receptors to decrease local dopamine release [1113].

While there have been a number of agonists of CB2 that have been reported in clinical development [14,15], modulation of receptor activity can also be achieved using ligands that bind to allosteric sites and either enhance or inhibit activity of endogenous agonists. Allosteric ligands represent a unique approach, particularly for CNS indications, as they have the ability to more subtly modulate neurotransmission, maintain spatial and temporal effects of endogenous neurotransmitters, and also provide enhanced selectivity for specific receptor isoforms [16]. In the case of the CB receptors, this point is paramount, as activation of the CB1 receptor results in psychotomimetic effects [17,18].

To search for and profile new CB2-selective PAMs, we are employing two orthogonal assays for compound evaluation: PI hydrolysis pathway via a chimeric G protein, and thallium flux induced by CB1- and CB2-mediated opening of G protein-coupled inwardly rectifying potassium channels (GIRKs). Using these assays, we evaluated the profile of the recently described CB2-selective PAM, EC21a [19,20], anticipating moving the compound into rodent models of psychosis. Until the development of EC21a, the only compound with reported CB2 PAM activity was a peptide termed pepcan-12, which also acts as a negative allosteric modulator (NAM) of CB1 [21]. EC21a was originally reported as a selective CB2 PAM based on in vitro pharmacology studies with [35S]GTPγS binding and [3H]CP55940 competition binding assays (Gado et al, 2019). In these studies, EC21a enhanced [35S]GTPγS binding induced by the agonist CP55940 and increased binding of [3H]CP55940 at low concentrations; this effect reversed at a 10 μM concentration. Additionally, Gado et al. used kinetic binding to show that EC21a significantly altered the dissociation rate of [3H]CP55940; all of these studies are consistent with an allosteric interaction between the agonist CP55940 and EC21a.

We confirmed here that EC21a only weakly and incompletely displaces the binding of the radioligand [3H]CP55940, suggesting it interacts at a distinct receptor binding site. Functional profiling of EC21a, however, showed that the compound functions as an allosteric agonist of CB2 and a mixed allosteric agonist/NAM of CB1 depending upon the assay used for profiling. Synthesis of a range of analogs based upon the EC21a scaffold showed that all of them blocked baseline and agonist-mediated responses at CB2. Overall, these results suggest that the EC21a series of compounds exhibits complicated pharmacology at the cannabinoid receptors.

Materials and Methods

Drugs

JWH133, 2-Arachidonylglycerol (2-AG), N-(2-Chloroethyl)-5Z,8Z,11Z,14Z-eicosatetraenamide (arachidonyl 2’-chloroethylamide, ACEA), AM251, AM630, and CP55940 were purchased from Tocris Bioscience (Minneapolis, MN). [3H]-CP55940 was obtained from Perkin Elmer (Boston, MA). EC21a (VU6043295) and structural analogs were synthesized by the Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, USA (Supplemental Methods).

Cell Lines and Cell Culture

Human Embryonic Kidney (HEK) 293 cells stably expressing Gqi9 (Gladstone Institute) were maintained in Dulbecco’s modified Eagle media (DMEM, Invitrogen. Waltham MA) containing 10% FBS, 100 units/mL penicillin/streptomycin, 20 mM HEPES, 1 mM sodium pyruvate, 2 mM L-glutamine, 1x non-essential amino acids, 700 μg/mL G418 sulfate. HEK cells stably expressing G protein inwardly rectifying potassium channels (GIRK1 and GIRK 2 subunits, HEK/GIRK [22]) were maintained in DMEM/F12 containing 10% FBS, 100 units/mL penicillin/streptomycin, 20 mM HEPES, 1 mM sodium pyruvate, 2 mM L-glutamine, 1x non-essential amino acids, and 700 μg/mL G418 sulfate. Cells were monitored by mycoplasma testing every two to three months.

Construction of human and rat CB1 and CB2 in pIRESneo3

The coding sequences of the rat CB1 (ENSRNOG00000008223), rat CB2 (ENSRNOG00000009260), human CB1 (ENSG00000118432), human CB2 (ENSG00000188822) receptors were cloned into the pIRESpuro3 expression vector (Takara Bio USA, San Jose, CA) using AgeI and EcoRI restriction sites. All cDNA sequences were confirmed by sequencing at Azenta Life Sciences (Chelmsford, MA).

Expression of human and rat CB1 and CB2 in HEK293 cells

Transient transfections were performed with ViaFect Transfection Reagent (Promega, Madison, WI). Briefly, 5 X 105 HEK293 cells stably expressing Gqi9 (HEK/Gqi9) or GIRK (HEK/GIRK) per well in 6-well dish grown overnight at 37 °C. Cells were incubated with 4.5 μL of ViaFect Transfection Reagent and 1.5 μg pIRESpuro3-based plasmid construct in Opti-MEM I Reduced Serum Medium. Cells were selected with DMEM growth medium containing 1 μg/mL of puromycin.

Assay of Inositol Phosphate Accumulation by chromatography

Data in Figures 2B and 3A were generated using this assay. Selected HEK/Gqi9 cells stably expressing CB1 or CB2 were seeded in 24-well plates at 1.5 X 105 cells/well and assayed after 48 hours. Inositol lipids were radiolabeled by overnight incubation of cells with 200 μL inositol-free DMEM containing 0.4 μCi myo-[3H]inositol (specific activity = 20 Ci/mmol, PerkinElmer) and 5% dialyzed FBS. Agonists were added at 5 x concentration in 50 μL of 50 mM LiCl and 250 mM HEPES, pH 7.25. Following a 30-minute incubation at 37 °C, the medium was aspirated, and the reaction was terminated by adding 0.75 mL boiling EDTA, pH 8.0. [3H]Inositol phosphates were resolved on Dowex AG1-X8 (Sigma-Aldrich) columns and counted by scintillation counting as described previously [23].

Figure 2. EC21a only weakly displaces [3H]CP55940 and blocks basal signaling in both the rat CB2 PI hydrolysis and GIRK assays.

Figure 2.

A. Increasing concentrations of EC21a or AM630 were added to membranes and competition with [3H]CP99540 was performed. Data are representative of three experiments performed in triplicate or duplicate (pIC50 values = 6.08+/− 0.86 for EC21a, and 7.14 +/−0.12 for AM630, respectively). B. Increasing concentrations of JWH133 alone (gray) or 10 μM EC21a plus JWH133 (red) were applied to CB2/HEK/Gqi9 cells and PI hydrolysis responses were measured ([3H]inositol radiolabeled chromatography assays). EC21a decreased the basal level of inositol phosphates and shifted the potency of JWH133 to the right (pEC50 value = 7.54 ± 0.16 for JWH133 alone, > 5μM for JWH133 + EC21a,). When applied alone (black), EC21a induced a concentration-dependent decrease in PI hydrolysis (pEC50 value = 6.66 ± 0.23). C. When assessed in the rat CB2/HEK/GIRK assay, EC21a blocked the response to JWH133 (red versus gray). As a control, the known inverse agonist AM630 was also applied with JWH133 and similarly blocked the response. D. When assessed alone in the rat CB2/HEK/GIRK thallium flux assay, EC21a decreased basal thallium flux levels concentration-dependently (pEC50 of 7.00 ± 0.11). Data are the Mean ± SEM of three independent experiments performed in triplicate.

Figure 3. EC21a exhibits agonist activity at rat CB1 in PI hydrolysis and negative allosteric modulator activity at rat CB1 in thallium flux.

Figure 3.

A. Increasing concentrations of ACEA were applied in either the absence (gray, pEC50 = 5.28 ± 0.64) or presence (black, pEC50 = > 5 μM; note that maximal responses were not achieved in this assay) of 10 μM EC21a; additionally, increasing concentrations of EC21a were applied to rat CB1/HEK/Gqi9 cells alone (white, pEC50 = 5.77 ± 0.36, [3H]inositol radiolabeled chromatography assays). B. ACEA-induced thallium flux responses in rat CB1/HEK/GIRK cells were blocked by increasing concentrations of EC21a (Emax = 100 ± 2 and 22 ± 2 in the absence and presence 10 μM EC21a, p< 0.0001). Data are the Mean ± SEM of three independent experiments performed in triplicate. Increasing concentrations of EC21a did not significantly shift the ACEA potencies in the thallium flux assay (B, pEC50 = 7.67 ± 0.05, DMSO versus 6.93 ± 0.28, ACEA + 10 μM EC21a, p = 0.06).

Assay of Inositol Phosphate Accumulation by IP-ONE kit

Due to a lack of routine availability of some reagents for radiolabeling chromatography assays, IP-ONE kits were used in some PI hydrolysis experiments; data in Table 1 were generated using this assay. This method produced comparable data to chromatography (data not shown). One day prior to experimentation, selected HEK/Gqi9 monoclonal cells stably expressing rat CB1 or rat CB2 were plated onto poly-D-lysine coated clear-bottom 384-well plates (20,000 cells per well) in DMEM supplemented with 10% FBS and 20 mM HEPES. Ten minutes prior to the assay, cell culture medium was replaced with 20 μL 37 °C Hank’s balanced salt solution (HBSS, with Ca2+, Mg2+). IP1 stimulation was then initiated by adding 5 μL of 5x agonists dissolved in HBSS plus 50 mM Li+, and cells were incubated for an additional hour before aspiration and addition of Lysis Buffer. IP1 levels were determined using the Cisbio HTRF IP-ONE assay kit per the manufacturer’s instructions, and fluorescence was measured using an Envision plate reader (PerkinElmer). Data were acquired as HTRF ratio (665/620) and expressed as nanomolar levels of IP1.

Table 1.

Agonist potencies (pEC50) in HEK/Gqi9 cells expressing cannabinoid receptors.

Rat CB1
pEC50 ± SEM
Rat CB2
pEC50 ± SEM
2-AG 6.07 ± 0.09 5.69 ± 0.01
ACEA 6.53 ± 0.05 NA
JWH133 NA 7.64 ± 0.06

Agonist-promoted PI hydrolysis was measured in HEK/Gqi9 cells expressing rat CB1 or CB2 receptors. Data are Mean ± SEM of three to five independent experiments performed in triplicate using IP-One assays. NA=not assessed.

Thallium Flux Assay

Thallium flux assays were performed as previously described (Niswender et al., 2008). HEK/GIRK cells stably expressing cannabinoid receptors (15,000 cells/20 μL/well) were seeded in 384-well, amine-coated assay plates (Greiner BioOne, Monroe, NC) and incubated overnight at 5% CO2 cell and 37 °C. Media was replaced with 20 μL/well of a dye loading solution containing assay buffer (Hanks Balanced Salt Solution plus 20 mM HEPES, pH 7.3), and 1.2 μM solution of the thallium-sensitive dye Thallos-AM (ION biosciences, San Marcos, TX), prepared as a DMSO stock and mixed first in a 1:1 ratio with (w/v) Pluronic F-127 (Sigma-Aldrich, St. Louis, MO). Following 1 hour at RT, dye solution was replaced with 20 μL/well assay buffer and the plates were loaded into a Hamamatsu FDSS 7000 (Bridgewater, NJ). Data were acquired at 1 Hz (excitation 470 ± 20 nm, emission 540 ± 30 nm) for ten seconds, followed by the addition of 20 μL/well of compound or DMSO (to test for PAM activity), followed by an additional 4 minutes of data collection. 140 seconds after compound addition, 10 μL/well of a thallium stimulus buffer (125 mM NaHCO3, 1.8 mM CaSO4, 1 mM MgSO4, 5 mM glucose, 12 mM Tl2SO4, 10 mM HEPES pH 7.4), along with serial dilution of agonist (5X), was added and data collection continued for an additional 2 minutes.

Raw kinetic data were analyzed in a multi-step process (see Supplemental Figure 1). 1) Fluorescence readings for each time point in a well were divided by the fluorescence reading at the initial time point to account for differences in cell number, non-uniform illumination, and dye-loading. 2) The slope value for each kinetic trace was calculated for the time window of 145–155 seconds, a window occurring directly after addition of agonist. 3) The average slope was calculated for wells containing vehicle and this value was subtracted from all wells. 4) Vehicle-subtracted slope was normalized to the relevant maximal agonist signal for each assay. Normalized data were fit to a four-parameter logistic equation using GraphPad Prism. Data shown represent the mean ± standard error of the pEC50 or maximal response. Experiments were performed in triplicate and repeated three to four separate times. To assess effects on inverse agonism, slope values were calculated after compound addition alone.

Radioligand Binding Assays

Membranes were made from HEK/GIRK cells stably expressing rat CB2. Radioligand competition binding assays were performed as previously described [24] with minor modifications. In brief, compounds were serially diluted into assay buffer with 0.1% bovine serum albumin (BSA) and added to each well of a 96-well plate, along with 20 μg/well cell membrane and approximately 500 pM [3H]-CP55940 (specific activity = 104 Ci/mmol, Perkin Elmer, Boston, MA). Following a 3-hour incubation period on shaker at room temperature, the membrane-bound ligand was separated from free ligand by filtration through glass fiber 96-well filter plates (Unifilter-96, GF/B; PerkinElmer, Boston, MA). Forty microliters of scintillation fluid were added to each well, and the membrane-bound radioactivity was determined by scintillation counting (Microbeta2; Revvity, Hopkinton, MA). Nonspecific binding was determined using 10 μM of cold CP55940.

Chemistry

Compounds were prepared using previously published procedures in the case of compound EC21a, compound 1 [19], and compound 9 [25]. Several closely related analogs, compounds 5–8 and 10, were prepared by similar procedures reported in [19] (see supplemental information for chemical synthesis).

Results

The compound EC21a acts as an inverse agonist of CB2 receptors in both the PI hydrolysis and thallium flux assays.

To explore the pharmacology of the CB receptors, we employed two assays, phosphoinositide hydrolysis in the presence of the chimeric G protein, Gqi9, and thallium flux through G protein inwardly rectifying potassium (GIRK) channels [22]. These assays revealed the expected pharmacology for a range of receptor ligands for both the rat and human receptors (Tables 1 and 2), and agonist activities were blocked by the antagonists AM251 (for CB1) and AM630 (for CB2).

Table 2.

Agonist potencies (pEC50) in HEK/GIRK cells expressing cannabinoid receptors.

Rat CB1 Rat CB2 Human CB1 Human CB2
pEC50 Emax (%) pEC50 Emax (%) pEC50 Emax (%) pEC50 Emax (%)
2-AG 7.77±0.03 91±9 7.31±0.14 130±12 7.18±0.09 110±2 6.84±0.08 117±9
ACEA 7.70±0.07 97±2 6.60±0.12 98±21 7.22±0.04 98±1 5.77±0.21 76±12
JWH133 5.70±0.17 47±10 6.27±0.09 103±2 5.91±0.18 23±4 6.56±0.08 104±3

Agonist-promoted thallium influx signals were assayed in HEK/GIRK cells expressing the rat CB1, rat CB2, human CB1, and human CB2 receptors. Data are Mean ± SEM of three to five independent experiments performed in triplicate. The maximum efficacies (Emax) were normalized to ACEA (for CB1) and JWH 133 (for CB2) respectively. Data are Mean ± SEM of three independent experiments performed in triplicate.

We next evaluated the activity of the newly reported CB2 PAM, EC21a (Figure 1, (Gado et al, 2019; 2021)). We performed binding with [3H]CP55940 and observed that AM630 almost completely displaced the binding of [3H]CP55940 (84.9 ± 6.9%); in contrast, EC21a only exhibited partial displacement (37.6 ± 4.9%), consistent with compound binding to an alternate site compared to AM630 and CP55940 (pIC50 = 6.08 ± 0.86 for EC21a, and 7.14 ± 0.12 for AM630, respectively) (Figure 2A). We next evaluated EC21a functional activity in the PI hydrolysis and thallium flux assays (Figures 2BD). Surprisingly, EC21a did not potentiate agonist responses in either assay. As shown in Figure 2B, addition of a 10 μM concentration of EC21a to rCB2/HEK/Gqi9 cells in the presence of the agonist JWH133 decreased basal levels of inositol phosphates and shifted the concentration-response of JWH133 to the right compared to the response of JWH133 alone (gray, pEC50 JWH133 alone = 7.54 ± 0.16; red, pEC50 JWH133 + EC21a = EC50 > 5μM). Application of increasing concentrations of EC21a alone resulted in a concentration-dependent decrease in basal PI hydrolysis with a pEC50 of 6.66 ± 0.23 (black, n=3, Figure 2B), consistent with blockade of constitutive activity/allosteric inverse agonist activity. In the rCB2/HEK/GIRK assay (Figure 2C), EC21a again significantly decreased basal responses (30.0 ± 5.9%, n=3 experiments) and shifted the concentration-response of JWH133 to the right with a nearly full blockade of the maximal signal (red versus gray). This response was similar to the response observed when the known inverse agonist AM630 was applied with JWH133 (black, Figure 2C). Application of increasing concentrations of EC21a alone to rCB2/HEK/GIRK cells resulted in a concentration-dependent decrease in basal thallium flux with a pEC50 of 7.00 ± 0.11 (Figure 2D). To eliminate the possibility that EC21a directly interacts with GIRK channels, increasing concentrations of EC21a were applied prior to the addition of the group III metabotropic glutamate receptor agonist L-AP4 to cells expressing the human mGlu7 receptor in the HEK/GIRK background (Niswender et al, 2008). As shown in Supplemental Figure 2, EC21a had no effect on thallium flux responses promoted by the group III metabotropic glutamate receptor agonist L-AP4 in hmGlu7/HEK/GIRK cells, indicating that effects of E21a are not due to non-specific effects.

Figure 1.

Figure 1.

Structure of EC21a.

EC21a was originally reported as a selective CB2 receptor PAM [19]. As shown in Figure 3A, EC21a induced IP accumulation in rCB1/HEK/Gqi9 cells when applied alone with a pEC50 of 5.77 ± 0.36 (white). When 10 μM EC21a was applied with a concentration-response of the agonist ACEA, EC21a raised the baseline PI hydrolysis response without an apparent shift in the pEC50 of ACEA (Figure 3A, black), which is consistent with distinct binding sites for EC21a and ACEA. We would note, however, that we did not achieve maximal PI hydrolysis responses in this assay. In the rCB1/HEK/GIRK assay, EC21a significantly and concentration-dependently decreased the maximal response to ACEA (Emax = 100 ± 2, DMSO versus 22 ± 2, ACEA + 10μM EC21a; p<0.0001) in a concentration-dependent manner without significantly shifting ACEA potency (pEC50 = 7.67 ± 0.05, DMSO versus 6.93 ± 0.28, ACEA + 10 μM EC21a, p = 0.06, Figure 3B), consistent with the profile of a negative allosteric modulator (NAM). These findings suggest that EC21a is a mixed agonist/NAM of CB1 in these assays and its activity is consistent with activity via an allosteric site.

EC21a analogs all inhibit, rather than potentiate, thallium flux in rCB2/HEK/GIRK cells.

Within scaffolds of allosteric modulators, subtle chemical modifications can induce dramatic shifts in pharmacology, from positive to negative to silent allosteric modulators (e.g., [2628]). We synthesized a variety of compounds based on the EC21a scaffold (Figure 4) and tested them in single point format in either the absence or the presence of a 10 μM concentration of the CB2 agonist JWH133. As shown in Figure 5, all of these compounds decreased both basal and JWH133-promoted thallium flux responses, suggesting that, similar to EC21a, these analogs block constitutive receptor activity; based on structural similarity with EC21a, we would anticipate that these effects are mediated by allosteric binding sites.

Figure 4.

Figure 4.

Structures of EC21a analogs

Figure 5. EC21a analogs block agonist-induced and constitutive thallium flux in the rat CB2/HEK/GIRK assay.

Figure 5.

10 μM of each EC21a analog was applied prior to vehicle (white) or 10 μM JWH133 (gray). Similar to EC21a itself, none of these compounds induced potentiation; rather, they all decreased basal levels of thallium flux and blocked JWH133 responses. Data are the Mean ± SEM of three independent experiments performed in triplicate.

Discussion

The CB1 and CB2 receptors have been studied for years as the targets of cannabis and both receptors have been investigated for their therapeutic potential in neurological and psychiatric diseases. While CB1 is thought to mediate psychoactive effects, CB2 is an intriguing target that is highly expressed in cells of the immune system and microglia [46], as well as in neuronal populations in hippocampal, cortical, and dopaminergic circuits [710,2936].

Recently, CB2 has been genetically linked to schizophrenia; for example, reduced CB2 receptor expression and/or activity is correlated with an increased risk for the manifestation of schizophrenia [3740]. We have recently shown that striatal cholinergic signaling regulates dopamine release via a CB2-dependent mechanism [12]. Potentiation of the M4 muscarinic receptor or mGlu1 with highly selective PAMs induces antipsychotic-like activity in numerous rodent behavioral assays [12,4143]. One important process by which M4 and mGlu1 PAMs mediate antipsychotic-like efficacy is by reducing striatal extracellular dopamine [13,44], and M4 and mGlu1-mediated effects on striatal dopamine release and antipsychotic efficacy are blocked by the CB2-selective antagonist AM630 and lost in CB2 knockout animals, indicating that CB2 receptors are required for the efficacy of these compounds [12,13]. CB2 activation or potentiation may represent a potential alternate, downstream strategy to M4 or mGlu1 potentiation that engages similar circuity but may have additional advantages. For example, CB2 activation has been shown to be cardioprotective [45] and anti-inflammatory by suppressing microglia activation and the release of inflammatory cytokines (reviewed in [4648]). Here, we have focused on profiling a PAM of CB2 as PAMs are predicted to avoid receptor desensitization that may occur with direct agonists and maintain spatial and temporal signaling of endogenous neurotransmitters’ plasticity [16,4951].

Our work provides further evidence demonstrating that CB receptor signaling pathways are complex and diverse. For example, the CB1 receptor has been shown to couple to Gi/o, Gs, β-arrestin, phospholipase C and inositol phosphate pathways, p38 and p42 Mitogen Activated Proteins Kinases (MAPKs), Phosphoinositide 3 Kinase (PI3K), potassium channels (including GIRKs), calcium channels (including N-, L-, and P/Q-type), and β-arrestins [5256]. In addition, CB signaling pathways are often biased depending on the cellular context and ligands themselves [53]. Here, we used two assays to interrogate the activity of ligands at CB1 and CB2 receptors: PI hydrolysis and thallium flux mediated by the βγ subunit stimulation of the GIRK channels. The latter assay complements studies in neurons which endogenously express the GIRKs or oocytes in which the channel is co-expressed with cannabinoid receptors showing that activation of the CB receptors induces GIRK channel opening [53,5760].

In this study, we examined the activity of EC21a, a recently reported PAM of CB2 [19,61], as we anticipated advancement of the compound to in vivo studies to evaluate the therapeutic potential of CB2 PAMs in rodent schizophrenia models. EC21a was originally described in a GTPγS assay as a PAM in the presence of the agonists CP55940 or 2-AG, but not anandamide, an endogenous cannabinoid [19]. It was also proposed to bind to the receptor at an allosteric site based on increased binding of the ligand [3H]CP55940 at low concentrations and displacement at only high concentrations (~10 μM [19]). We confirmed partial displacement of [3H]CP55940 at high concentrations of EC21a (Figure 2A). Functionally, however, EC21a behaved as an inverse agonist, presumably via an allosteric site based on its binding profile [19], of the CB2 receptor in both PI hydrolysis and thallium flux assays. In addition, despite its reported selectivity for the CB2 receptor, EC21a displayed agonist activity in PI hydrolysis and negative allosteric modulation in thallium flux at the CB1 receptor; all analogs of EC21a also decreased GIRK channel activity when applied alone to CB2/GIRK/HEK cells. Overall, our findings suggest that EC21a exhibits complicated pharmacology in vitro at both CB receptor isoforms.

EC21a has been shown to exhibit efficacy in a neuropathic pain model, and effects were lost when the animals were pretreated with a CB2 antagonist but not a CB1 antagonist [19]. EC21a was also shown to potentiate the activity of a CB2 agonist, B2, in an in vitro model of lipopolysaccharide (LPS)-induced cytokine release and the effect was also blocked by a CB2 antagonist [62]. EC21a had no efficacy alone in this model. EC21a was also shown to be effective in reducing seizures and these effects were blocked by the CB2 antagonist AM630 [63]; additionally, in a genetic model of seizures (R1648H SCN1A mice), EC21a was also effective. This efficacy was not observed with AM630 [63]. These findings suggest that the activity of EC21a in vivo is distinct from AM630 and suggests that additional systematic studies are warranted to understand the signaling pathways that underlie the in vivo efficacy of EC21a, AM630, and CB2 modulators.

Supplementary Material

Supp 1

Acknowledgements

We thank the William K. Warren Foundation for their funding toward the Warren Center for Neuroscience Drug Discovery.

Funding Sources

This work was supported by National Institutes of Health under Grant No. MH119673 and Grant No. MH062646.

Footnotes

Ethical Approval

This article does not contain any studies involving animals or human participants performed by any of the authors.

Disclosure Statement

No author has an actual or perceived conflict of interest with the contents of this article.

Contributor Information

Aidong Qi, Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.

Xueqing Han, Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.

Marc Quitalig, Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.

Jessica Wu, Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.

Plamen P. Christov, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, USA

KyuOk Jeon, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, USA.

Somnath Jana, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, USA.

Kwangho Kim, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, USA.

Darren W. Engers, Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA

Craig W. Lindsley, Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, USA.

Alice L. Rodriguez, Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA

Colleen M. Niswender, Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, USA; Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA, Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA.

Data Availability Statement

The authors declare that all processed data supporting the findings of this study are available within the paper and its Supplemental Data. Raw data are available on request from the corresponding author.

References

  • [1].Pertwee RG. Receptors and channels targeted by synthetic cannabinoid receptor agonists and antagonists. Curr Med Chem. 2010;17(14):1360–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Pertwee RG, Howlett AC, Abood ME, et al. International Union of Basic and Clinical Pharmacology. LXXIX. Cannabinoid receptors and their ligands: beyond CB(1) and CB(2). Pharmacol Rev. 2010;62(4):588–631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Tsou K, Brown S, Sanudo-Pena MC, et al. Immunohistochemical distribution of cannabinoid CB1 receptors in the rat central nervous system. Neuroscience. 1998;83(2):393–411. [DOI] [PubMed] [Google Scholar]
  • [4].Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a peripheral receptor for cannabinoids. Nature. 1993;365(6441):61–5. [DOI] [PubMed] [Google Scholar]
  • [5].Brown SM, Wager-Miller J, Mackie K. Cloning and molecular characterization of the rat CB2 cannabinoid receptor. Biochim Biophys Acta. 2002;1576(3):255–64. [DOI] [PubMed] [Google Scholar]
  • [6].Atwood BK, Mackie K. CB2: a cannabinoid receptor with an identity crisis. British journal of pharmacology. 2010;160(3):467–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Komorowska-Muller JA, Schmole AC. CB2 Receptor in Microglia: The Guardian of Self-Control. Int J Mol Sci. 2020;22(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Duffy SS, Hayes JP, Fiore NT, et al. The cannabinoid system and microglia in health and disease. Neuropharmacology. 2021;190:108555. [DOI] [PubMed] [Google Scholar]
  • [9].de Oliveira DR, Maia RC, de Carvalho Franca PR, et al. 2-Arylpropionic Acid Pyrazolamides as Cannabinoid CB2 Receptor Inverse Agonists Endowed with Anti-Inflammatory Properties. Pharmaceuticals (Basel). 2022;15(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Reusch N, Ravichandran KA, Olabiyi BF, et al. Cannabinoid receptor 2 is necessary to induce toll-like receptor-mediated microglial activation. Glia. 2022;70(1):71–88. [DOI] [PubMed] [Google Scholar]
  • [11].Ferranti AS, Foster DJ. Cannabinoid type-2 receptors: An emerging target for regulating schizophrenia-relevant brain circuits. Front Neurosci. 2022;16:925792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Foster DJ, Wilson JM, Remke DH, et al. Antipsychotic-like Effects of M4 Positive Allosteric Modulators Are Mediated by CB2 Receptor-Dependent Inhibition of Dopamine Release. Neuron. 2016;91(6):1244–1252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Yohn SE, Foster DJ, Covey DP, et al. Activation of the mGlu1 metabotropic glutamate receptor has antipsychotic-like effects and is required for efficacy of M4 muscarinic receptor allosteric modulators. Mol Psychiatry. 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].An D, Peigneur S, Hendrickx LA, et al. Targeting Cannabinoid Receptors: Current Status and Prospects of Natural Products. Int J Mol Sci. 2020;21(14). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Whiting ZM, Yin J, de la Harpe SM, et al. Developing the Cannabinoid Receptor 2 (CB2) pharmacopoeia: past, present, and future. Trends in pharmacological sciences. 2022;43(9):754–771. [DOI] [PubMed] [Google Scholar]
  • [16].Lindsley CW, Emmitte KA, Hopkins CR, et al. Practical Strategies and Concepts in GPCR Allosteric Modulator Discovery: Recent Advances with Metabotropic Glutamate Receptors. Chem Rev. 2016;116(11):6707–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Martin RS, Secchi RL, Sung E, et al. Effects of cannabinoid receptor ligands on psychosis-relevant behavior models in the rat. Psychopharmacology (Berl). 2003;165(2):128–35. [DOI] [PubMed] [Google Scholar]
  • [18].Gobira PH, LaMar J, Marques J, et al. CB1 Receptor Silencing Attenuates Ketamine-Induced Hyperlocomotion Without Compromising Its Antidepressant-Like Effects. Cannabis Cannabinoid Res. 2023;8(5):768–778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Gado F, Di Cesare Mannelli L, Lucarini E, et al. Identification of the First Synthetic Allosteric Modulator of the CB(2) Receptors and Evidence of Its Efficacy for Neuropathic Pain Relief. J Med Chem. 2019;62(1):276–287. [DOI] [PubMed] [Google Scholar]
  • [20].Gado F, Arena C, Fauci C, et al. Modification on the 1,2-dihydro-2-oxo-pyridine-3-carboxamide core to obtain multi-target modulators of endocannabinoid system. Bioorg Chem. 2020;94:103353. [DOI] [PubMed] [Google Scholar]
  • [21].Petrucci V, Chicca A, Glasmacher S, et al. Pepcan-12 (RVD-hemopressin) is a CB2 receptor positive allosteric modulator constitutively secreted by adrenals and in liver upon tissue damage. Sci Rep. 2017;7(1):9560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Niswender CM, Johnson KA, Luo Q, et al. A novel assay of Gi/o-linked G protein coupled receptor coupling to potassium channels provides new insights into the pharmacology of the group III metabotropic glutamate receptors. Mol Pharmacol. 2008;73(4):1213–1224. [DOI] [PubMed] [Google Scholar]
  • [23].Qi AD, Harden TK, Nicholas RA. Is GPR17 a P2Y/leukotriene receptor? examination of uracil nucleotides, nucleotide sugars, and cysteinyl leukotrienes as agonists of GPR17. J Pharmacol Exp Ther. 2013;347(1):38–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Anderson JJ, Rao SP, Rowe B, et al. [3H]Methoxymethyl-3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine binding to metabotropic glutamate receptor subtype 5 in rodent brain: in vitro and in vivo characterization. J Pharmacol Exp Ther. 2002;303(3):1044–51. [DOI] [PubMed] [Google Scholar]
  • [25].Jourdan T, Szanda G, Rosenberg AZ, et al. Overactive cannabinoid 1 receptor in podocytes drives type 2 diabetic nephropathy. Proc Natl Acad Sci U S A. 2014;111(50):E5420–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Sharma S, Rodriguez AL, Conn PJ, et al. Synthesis and SAR of a mGluR5 allosteric partial antagonist lead: unexpected modulation of pharmacology with slight structural modifications to a 5-(phenylethynyl)pyrimidine scaffold. Bioorg Med Chem Lett. 2008;18(14):4098–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Sharma S, Kedrowski J, Rook JM, et al. Discovery of molecular switches that modulate modes of metabotropic glutamate receptor subtype 5 (mGlu5) pharmacology in vitro and in vivo within a series of functionalized, regioisomeric 2- and 5-(phenylethynyl)pyrimidines. J Med Chem. 2009;52(14):4103–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Gregory KJ, Kufareva I, Keller AN, et al. Dual Action Calcium-Sensing Receptor Modulator Unmasks Novel Mode-Switching Mechanism. ACS Pharmacol Transl Sci. 2018;1(2):96–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Sagar DR, Kelly S, Millns PJ, et al. Inhibitory effects of CB1 and CB2 receptor agonists on responses of DRG neurons and dorsal horn neurons in neuropathic rats. Eur J Neurosci. 2005;22(2):371–9. [DOI] [PubMed] [Google Scholar]
  • [30].Van Sickle MD, Duncan M, Kingsley PJ, et al. Identification and functional characterization of brainstem cannabinoid CB2 receptors. Science. 2005;310(5746):329–32. [DOI] [PubMed] [Google Scholar]
  • [31].Ashton JC, Friberg D, Darlington CL, et al. Expression of the cannabinoid CB2 receptor in the rat cerebellum: an immunohistochemical study. Neurosci Lett. 2006;396(2):113–6. [DOI] [PubMed] [Google Scholar]
  • [32].Gong JP, Onaivi ES, Ishiguro H, et al. Cannabinoid CB2 receptors: immunohistochemical localization in rat brain. Brain research. 2006;1071(1):10–23. [DOI] [PubMed] [Google Scholar]
  • [33].Onaivi ES, Ishiguro H, Gong JP, et al. Discovery of the presence and functional expression of cannabinoid CB2 receptors in brain. Ann N Y Acad Sci. 2006;1074:514–36. [DOI] [PubMed] [Google Scholar]
  • [34].Onaivi ES. Neuropsychobiological evidence for the functional presence and expression of cannabinoid CB2 receptors in the brain. Neuropsychobiology. 2006;54(4):231–46. [DOI] [PubMed] [Google Scholar]
  • [35].Brusco A, Tagliaferro PA, Saez T, et al. Ultrastructural localization of neuronal brain CB2 cannabinoid receptors. Ann N Y Acad Sci. 2008;1139:450–7. [DOI] [PubMed] [Google Scholar]
  • [36].Brusco A, Tagliaferro P, Saez T, et al. Postsynaptic localization of CB2 cannabinoid receptors in the rat hippocampus. Synapse. 2008;62(12):944–9. [DOI] [PubMed] [Google Scholar]
  • [37].Ishiguro H, Kibret BG, Horiuchi Y, et al. Potential Role of Cannabinoid Type 2 Receptors in Neuropsychiatric and Neurodegenerative Disorders. Front Psychiatry. 2022;13:828895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Kibret BG, Ishiguro H, Horiuchi Y, et al. New Insights and Potential Therapeutic Targeting of CB2 Cannabinoid Receptors in CNS Disorders. Int J Mol Sci. 2022;23(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Legge SE, Jones HJ, Kendall KM, et al. Association of Genetic Liability to Psychotic Experiences With Neuropsychotic Disorders and Traits. JAMA Psychiatry. 2019;76(12):1256–1265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Ishiguro H, Horiuchi Y, Ishikawa M, et al. Brain cannabinoid CB2 receptor in schizophrenia. Biol Psychiatry. 2010;67(10):974–82. [DOI] [PubMed] [Google Scholar]
  • [41].Bubser M, Bridges TM, Dencker D, et al. Selective activation of M4 muscarinic acetylcholine receptors reverses MK-801-induced behavioral impairments and enhances associative learning in rodents. ACS Chem Neurosci. 2014;5(10):920–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Brady AE, Jones CK, Bridges TM, et al. Centrally active allosteric potentiators of the M4 muscarinic acetylcholine receptor reverse amphetamine-induced hyperlocomotor activity in rats. J Pharmacol Exp Ther. 2008;327(3):941–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Conn PJ, Jones CK, Lindsley CW. Subtype-selective allosteric modulators of muscarinic receptors for the treatment of CNS disorders. Trends in pharmacological sciences. 2009;30(3):148–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Byun NE, Grannan M, Bubser M, et al. Antipsychotic drug-like effects of the selective M4 muscarinic acetylcholine receptor positive allosteric modulator VU0152100. Neuropsychopharmacology. 2014;39(7):1578–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].More SA, Deore RS, Pawar HD, et al. CB2 Cannabinoid Receptor as a Potential Target in Myocardial Infarction: Exploration of Molecular Pathogenesis and Therapeutic Strategies. Int J Mol Sci. 2024;25(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Magham SV, Thaggikuppe Krishnamurthy P, Shaji N, et al. Cannabinoid receptor 2 selective agonists and Alzheimer’s disease: An insight into the therapeutic potentials. J Neurosci Res. 2021;99(11):2888–2905. [DOI] [PubMed] [Google Scholar]
  • [47].Morris G, Walder K, Kloiber S, et al. The endocannabinoidome in neuropsychiatry: Opportunities and potential risks. Pharmacol Res. 2021;170:105729. [DOI] [PubMed] [Google Scholar]
  • [48].Bie B, Wu J, Foss JF, et al. An overview of the cannabinoid type 2 receptor system and its therapeutic potential. Curr Opin Anaesthesiol. 2018;31(4):407–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Tschammer N Allosteric Modulators of the Class A G Protein Coupled Receptors. Adv Exp Med Biol. 2016;917:185–207. [DOI] [PubMed] [Google Scholar]
  • [50].Kenakin T Allostery: The Good, the Bad, and the Ugly. J Pharmacol Exp Ther. 2024;388(1):110–120. [DOI] [PubMed] [Google Scholar]
  • [51].Kenakin TP. Biased signalling and allosteric machines: new vistas and challenges for drug discovery. Br J Pharmacol. 2012;165(6):1659–1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Demuth DG, Molleman A. Cannabinoid signalling. Life Sci. 2006;78(6):549–63. [DOI] [PubMed] [Google Scholar]
  • [53].Soethoudt M, Grether U, Fingerle J, et al. Cannabinoid CB(2) receptor ligand profiling reveals biased signalling and off-target activity. Nat Commun. 2017;8:13958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Howlett AC, Abood ME. CB(1) and CB(2) Receptor Pharmacology. Adv Pharmacol. 2017;80:169–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Gasperi V, Guzzo T, Topai A, et al. Recent Advances on Type-2 Cannabinoid (CB(2)) Receptor Agonists and their Therapeutic Potential. Curr Med Chem. 2023;30(12):1420–1457. [DOI] [PubMed] [Google Scholar]
  • [56].Legare CA, Raup-Konsavage WM, Vrana KE. Therapeutic Potential of Cannabis, Cannabidiol, and Cannabinoid-Based Pharmaceuticals. Pharmacology. 2022;107(3–4):131–149. [DOI] [PubMed] [Google Scholar]
  • [57].Guo J, Ikeda SR. Endocannabinoids modulate N-type calcium channels and G-protein-coupled inwardly rectifying potassium channels via CB1 cannabinoid receptors heterologously expressed in mammalian neurons. Mol Pharmacol. 2004;65(3):665–74. [DOI] [PubMed] [Google Scholar]
  • [58].Robbe D, Alonso G, Duchamp F, et al. Localization and mechanisms of action of cannabinoid receptors at the glutamatergic synapses of the mouse nucleus accumbens. J Neurosci. 2001;21(1):109–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].McAllister SD, Griffin G, Satin LS, et al. Cannabinoid receptors can activate and inhibit G protein-coupled inwardly rectifying potassium channels in a xenopus oocyte expression system. J Pharmacol Exp Ther. 1999;291(2):618–26. [PubMed] [Google Scholar]
  • [60].Henry DJ, Chavkin C. Activation of inwardly rectifying potassium channels (GIRK1) by co-expressed rat brain cannabinoid receptors in Xenopus oocytes. Neurosci Lett. 1995;186(2–3):91–4. [DOI] [PubMed] [Google Scholar]
  • [61].Gado F, Mohamed KA, Meini S, et al. Variously substituted 2-oxopyridine derivatives: Extending the structure-activity relationships for allosteric modulation of the cannabinoid CB2 receptor. Eur J Med Chem. 2021;211:113116. [DOI] [PubMed] [Google Scholar]
  • [62].Polini B, Cervetto C, Carpi S, et al. Positive Allosteric Modulation of CB1 and CB2 Cannabinoid Receptors Enhances the Neuroprotective Activity of a Dual CB1R/CB2R Orthosteric Agonist. Life (Basel). 2020;10(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Shapiro L, Gado F, Manera C, et al. Allosteric modulation of the cannabinoid 2 receptor confers seizure resistance in mice. Neuropharmacology. 2021;188:108448. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supp 1

Data Availability Statement

The authors declare that all processed data supporting the findings of this study are available within the paper and its Supplemental Data. Raw data are available on request from the corresponding author.

RESOURCES