Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
Elife ; 122023 Dec 12.
Article En | MEDLINE | ID: mdl-38085667

Free fatty acid receptor 2 (FFAR2) is activated by short-chain fatty acids and expressed widely, including in white adipocytes and various immune and enteroendocrine cells. Using both wild-type human FFAR2 and a designer receptor exclusively activated by designer drug (DREADD) variant we explored the activation and phosphorylation profile of the receptor, both in heterologous cell lines and in tissues from transgenic knock-in mouse lines expressing either human FFAR2 or the FFAR2-DREADD. FFAR2 phospho-site-specific antisera targeting either pSer296/pSer297 or pThr306/pThr310 provided sensitive biomarkers of both constitutive and agonist-mediated phosphorylation as well as an effective means to visualise agonist-activated receptors in situ. In white adipose tissue, phosphorylation of residues Ser296/Ser297 was enhanced upon agonist activation whilst Thr306/Thr310 did not become phosphorylated. By contrast, in immune cells from Peyer's patches Thr306/Thr310 become phosphorylated in a strictly agonist-dependent fashion whilst in enteroendocrine cells of the colon both Ser296/Ser297 and Thr306/Thr310 were poorly phosphorylated. The concept of phosphorylation bar-coding has centred to date on the potential for different agonists to promote distinct receptor phosphorylation patterns. Here, we demonstrate that this occurs for the same agonist-receptor pairing in different patho-physiologically relevant target tissues. This may underpin why a single G protein-coupled receptor can generate different functional outcomes in a tissue-specific manner.


Fatty Acids, Nonesterified , Receptors, G-Protein-Coupled , Animals , Humans , Mice , Cell Line , Fatty Acids, Volatile/metabolism , Mice, Transgenic , Phosphorylation , Receptors, G-Protein-Coupled/metabolism
2.
Elife ; 112022 03 01.
Article En | MEDLINE | ID: mdl-35229717

Volatile small molecules, including the short-chain fatty acids (SCFAs), acetate and propionate, released by the gut microbiota from the catabolism of nondigestible starches, can act in a hormone-like fashion via specific G-protein-coupled receptors (GPCRs). The primary GPCR targets for these SCFAs are FFA2 and FFA3. Using transgenic mice in which FFA2 was replaced by an altered form called a Designer Receptor Exclusively Activated by Designer Drugs (FFA2-DREADD), but in which FFA3 is unaltered, and a newly identified FFA2-DREADD agonist 4-methoxy-3-methyl-benzoic acid (MOMBA), we demonstrate how specific functions of FFA2 and FFA3 define a SCFA-gut-brain axis. Activation of both FFA2/3 in the lumen of the gut stimulates spinal cord activity and activation of gut FFA3 directly regulates sensory afferent neuronal firing. Moreover, we demonstrate that FFA2 and FFA3 are both functionally expressed in dorsal root- and nodose ganglia where they signal through different G proteins and mechanisms to regulate cellular calcium levels. We conclude that FFA2 and FFA3, acting at distinct levels, provide an axis by which SCFAs originating from the gut microbiota can regulate central activity.


Brain-Gut Axis , Receptors, Cell Surface , Animals , Fatty Acids, Volatile/metabolism , Mice , Propionates/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism
3.
Curr Opin Endocr Metab Res ; 16: 1-9, 2021 Feb.
Article En | MEDLINE | ID: mdl-32835130

FFA2 and FFA3 are receptors for short-chain fatty acids which are produced in prodigious amounts by fermentation of poorly digested carbohydrates by gut bacteria. Understanding the roles of these receptors in regulating enteroendocrine, metabolic and immune functions has developed with the production and use of novel pharmacological tools and animal models. A complex (patho)physiological scenario is now emerging in which strategic expression of FFA2 and FFA3 in key cell types and selective modulation of their signalling might regulate body weight management, energy homoeostasis and inflammatory disorders.

4.
Nat Chem Biol ; 15(5): 489-498, 2019 05.
Article En | MEDLINE | ID: mdl-30992568

Differentiating actions of short chain fatty acids (SCFAs) at free fatty acid receptor 2 (FFA2) from other free fatty acid-responsive receptors and from non-receptor-mediated effects has been challenging. Using a novel chemogenetic and knock-in strategy, whereby an engineered variant of FFA2 (FFA2-DREADD) that is unresponsive to natural SCFAs but is instead activated by sorbic acid replaced the wild-type receptor, we determined that activation of FFA2 in differentiated adipocytes and colonic crypt enteroendocrine cells of mouse accounts fully for SCFA-regulated lipolysis and release of the incretin glucagon-like peptide-1 (GLP-1), respectively. In vivo studies confirmed the specific role of FFA2 in GLP-1 release and also demonstrated a direct role for FFA2 in accelerating gut transit. Thereby, we establish the general principle that such a chemogenetic knock-in strategy can successfully define novel G-protein-coupled receptor (GPCR) biology and provide both target validation and establish therapeutic potential of a 'hard to target' GPCR.


Fatty Acids, Volatile/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Humans , Mice , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled/genetics
5.
Sci Rep ; 9(1): 1861, 2019 02 12.
Article En | MEDLINE | ID: mdl-30755705

Many members of the G protein-coupled receptor family, including examples with clear therapeutic potential, remain poorly characterised. This often reflects limited availability of suitable tool ligands with which to interrogate receptor function. In the case of GPR84, currently a target for the treatment of idiopathic pulmonary fibrosis, recent times have seen the description of novel orthosteric and allosteric agonists. Using 2-(hexylthiol)pyrimidine-4,6 diol (2-HTP) and di(5,7-difluoro-1H-indole-3-yl)methane (PSB-16671) as exemplars of each class, in cell lines transfected to express either human or mouse GPR84, both ligands acted as effective on-target activators and with high co-operativity in their interactions. This was also the case in lipopolysaccharide-activated model human and mouse immune cell lines. However in mouse bone-marrow-derived neutrophils, where expression of GPR84 is particularly high, the capacity of PSB-16671 but not of 2-HTP to promote G protein activation was predominantly off-target because it was not blocked by an antagonist of GPR84 and was preserved in neutrophils isolated from GPR84 deficient mice. These results illustrate the challenges of attempting to study and define functions of poorly characterised receptors using ligands that have been developed via medicinal chemistry programmes, but where assessed activity has been limited largely to the initially identified target.


Receptors, G-Protein-Coupled/chemistry , Allosteric Site , Animals , Binding Sites , Bone Marrow Cells/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Ligands , Lipopolysaccharides/metabolism , Mice , Mice, Knockout , Neutrophils/metabolism , RAW 264.7 Cells , Receptors, G-Protein-Coupled/agonists , THP-1 Cells
6.
Gastroenterology ; 156(5): 1354-1367.e6, 2019 Apr.
Article En | MEDLINE | ID: mdl-30550821

BACKGROUND & AIMS: Exclusive enteral nutrition (EEN) is the only established dietary treatment for Crohn's disease (CD), but its acceptability is limited. There is a need for novel dietary treatments for CD. METHODS: We evaluated the effects of an individualized food-based diet (CD-TREAT), with similar composition to EEN, on the gut microbiome, inflammation, and clinical response in a rat model, healthy adults, and children with relapsing CD. Twenty-five healthy adults randomly received EEN or CD-TREAT for 7 days, followed by a 14-day washout period, followed by the alternate diet. Fecal microbiome and metabolome were assessed before and after each diet. HLA-B7 and HLA-B27 transgenic rats with gut inflammation received EEN, CD-TREAT, or standard chow for 4 weeks. Fecal, luminal, and tissue microbiome, fecal metabolites, and gut inflammation were assessed. Five children with active CD activity received CD-TREAT and their clinical activity and calprotectin were evaluated after 8 weeks of treatment. RESULTS: For healthy adults, CD-TREAT was easier to comply with and more acceptable than EEN. CD-TREAT induced similar effects to EEN (EEN vs CD-TREAT) on fecal microbiome composition, metabolome, mean total sulfide (increase 133.0 ± 80.5 vs 54.3 ± 47.0 nmol/g), pH (increase 1.3 ± 0.5 vs 0.9 ± 0.6), and the short-chain fatty acids (µmol/g) acetate (decrease 27.4 ± 22.6 vs 21.6 ± 20.4), propionate (decrease 5.7 ± 7.8 vs 5.2 ± 7.9), and butyrate (decrease 7.0 ± 7.4 vs 10.2 ± 8.5). In the rat model, CD-TREAT and EEN produced similar changes in bacterial load (decrease 0.3 ± 0.3 log10 16S rRNA gene copies per gram), short-chain fatty acids, microbiome, and ileitis severity (mean histopathology score decreases of 1.25 for EEN [P = .015] and 1.0 for CD-TREAT [P = .044] vs chow). In children receiving CD-TREAT, 4 (80%) had a clinical response and 3 (60%) entered remission, with significant concurrent decreases in fecal calprotectin (mean decrease 918 ± 555 mg/kg; P = .002). CONCLUSION: CD-TREAT replicates EEN changes in the microbiome, decreases gut inflammation, is well tolerated, and is potentially effective in patients with active CD. ClinicalTrials.gov, numbers NCT02426567 and NCT03171246.


Bacteria/growth & development , Crohn Disease/diet therapy , Enteral Nutrition , Gastrointestinal Microbiome , Nutritive Value , Adolescent , Adult , Animals , Bacteria/isolation & purification , Bacteria/metabolism , Bacterial Load , Child , Crohn Disease/diagnosis , Crohn Disease/microbiology , Crohn Disease/physiopathology , Disease Models, Animal , Feces/microbiology , Female , HLA-B27 Antigen/genetics , HLA-B7 Antigen/genetics , Humans , Male , Nutritional Status , Rats, Transgenic , Recurrence , Remission Induction , Scotland , Time Factors , Treatment Outcome , Young Adult
7.
J Med Chem ; 61(21): 9534-9550, 2018 11 08.
Article En | MEDLINE | ID: mdl-30247908

Free fatty acid receptor 2 (FFA2/GPR43) is a receptor for short-chain fatty acids reported to be involved in regulation of metabolism, appetite, fat accumulation, and inflammatory responses and is a potential target for treatment of various inflammatory and metabolic diseases. By bioisosteric replacement of the central pyrrolidine core of a previously disclosed FFA2 agonist with a synthetically more tractable thiazolidine, we were able to rapidly synthesize and screen analogues modified at both the 2- and 3-positions on the thiazolidine core. Herein, we report SAR exploration of thiazolidine FFA2 agonists and the identification of 31 (TUG-1375), a compound with significantly increased potency (7-fold in a cAMP assay) and reduced lipophilicity (50-fold reduced clog P) relative to the pyrrolidine lead structure. The compound has high solubility, high chemical, microsomal, and hepatocyte stability, and favorable pharmacokinetic properties and was confirmed to induce human neutrophil mobilization and to inhibit lipolysis in murine adipocytes.


Drug Discovery , Receptors, Cell Surface/agonists , Thiazolidines/pharmacology , Thiazolidines/pharmacokinetics , Animals , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Models, Molecular , Molecular Conformation , Receptors, Cell Surface/chemistry , Thiazolidines/chemistry , Tissue Distribution
8.
Sci Rep ; 7(1): 13741, 2017 10 23.
Article En | MEDLINE | ID: mdl-29061999

Free Fatty Acid Receptor 2 is a GPCR activated by short chain fatty acids produced in high levels in the lower gut by microbial fermentation of non-digestible carbohydrates. A major challenge in studying this receptor is that the mouse ortholog does not have significant affinity for antagonists that are able to block the human receptor. Docking of exemplar antagonists from two chemical series to homology models of both human and mouse Free Fatty Acid Receptor 2 suggested that a single lysine - arginine variation at the extracellular face of the receptor might provide the basis for antagonist selectivity and mutational swap studies confirmed this hypothesis. Extending these studies to agonist function indicated that although the lysine - arginine variation between human and mouse orthologs had limited effect on G protein-mediated signal transduction, removal of positive charge from this residue produced a signalling-biased variant of Free Fatty Acid Receptor 2 in which Gi-mediated signalling by both short chain fatty acids and synthetic agonists was maintained whilst there was marked loss of agonist potency for signalling via Gq/11 and G12/13 G proteins. A single residue at the extracellular face of the receptor thus plays key roles in both agonist and antagonist function.


Extracellular Space/metabolism , GTP-Binding Proteins/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Amino Acid Substitution , Animals , HEK293 Cells , Humans , Mice , Models, Molecular , Protein Conformation , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Substrate Specificity
9.
Handb Exp Pharmacol ; 236: 17-32, 2017.
Article En | MEDLINE | ID: mdl-27757758

A large number of reviews and commentaries have highlighted the potential role of the short-chain fatty acid receptors GPR41 (FFA3) and, particularly, GPR43 (FFA2) as an interface between the intestinal microbiota and metabolic and inflammatory disorders. However, short-chain fatty acids have very modest potency and display limited selectivity between these two receptors, and studies on receptor knockout mice have resulted in non-uniform conclusions; therefore, selective and high-potency/high-affinity synthetic ligands are required to further explore the contribution of these receptors to health and disease. Currently no useful orthosteric ligands of FFA3 have been reported and although a number of orthosteric FFA2 agonists and antagonists have been described, a lack of affinity of different chemotypes of FFA2 antagonists at the mouse and rat orthologs of this receptor has hindered progress. Selective allosteric regulators of both FFA2 and FFA3 have provided tools to address a number of basic questions in both in vitro and ex vivo preparations, but at least some of the positive modulators appear to be biased and able to regulate only a subset of the functional capabilities of the short-chain fatty acids. Significant further progress is required to provide improved tool compounds to better assess potential translational opportunities of these receptors for short-chain fatty acids.


Receptors, Cell Surface/physiology , Receptors, G-Protein-Coupled/physiology , Allosteric Regulation , Animals , Humans , Ligands , Receptors, Cell Surface/agonists , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/drug effects , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/drug effects
10.
J Biol Chem ; 291(36): 18915-31, 2016 09 02.
Article En | MEDLINE | ID: mdl-27385588

The short chain fatty acid receptor FFA2 is able to stimulate signaling via both Gi- and Gq/G11-promoted pathways. These pathways are believed to control distinct physiological end points but FFA2 receptor ligands appropriate to test this hypothesis have been lacking. Herein, we characterize AZ1729, a novel FFA2 regulator that acts as a direct allosteric agonist and as a positive allosteric modulator, increasing the activity of the endogenously produced short chain fatty acid propionate in Gi-mediated pathways, but not at those transduced by Gq/G11 Using AZ1729 in combination with direct inhibitors of Gi and Gq/G11 family G proteins demonstrated that although both arms contribute to propionate-mediated regulation of phospho-ERK1/2 MAP kinase signaling in FFA2-expressing 293 cells, the Gq/G11-mediated pathway is predominant. We extend these studies by employing AZ1729 to dissect physiological FFA2 signaling pathways. The capacity of AZ1729 to act at FFA2 receptors to inhibit ß-adrenoreceptor agonist-promoted lipolysis in primary mouse adipocytes and to promote chemotaxis of isolated human neutrophils confirmed these as FFA2 processes mediated by Gi signaling, whereas, in concert with blockade by the Gq/G11 inhibitor FR900359, the inability of AZ1729 to mimic or regulate propionate-mediated release of GLP-1 from mouse colonic preparations defined this physiological response as an end point transduced via activation of Gq/G11.


Depsipeptides/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go , GTP-Binding Protein alpha Subunits, Gq-G11 , MAP Kinase Signaling System/drug effects , Receptors, Cell Surface , Receptors, G-Protein-Coupled , Allosteric Regulation/drug effects , Animals , Colon/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Lipolysis/drug effects , Lipolysis/genetics , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neutrophils/metabolism , Receptors, Cell Surface/agonists , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
11.
Mol Pharmacol ; 89(3): 388-98, 2016 Mar.
Article En | MEDLINE | ID: mdl-26719580

Despite some blockbuster G protein-coupled receptor (GPCR) drugs, only a small fraction (∼ 15%) of the more than 390 nonodorant GPCRs have been successfully targeted by the pharmaceutical industry. One way that this issue might be addressed is via translation of recent deorphanization programs that have opened the prospect of extending the reach of new medicine design to novel receptor types with potential therapeutic value. Prominent among these receptors are those that respond to short-chain free fatty acids of carbon chain length 2-6. These receptors, FFA2 (GPR43) and FFA3 (GPR41), are each predominantly activated by the short-chain fatty acids acetate, propionate, and butyrate, ligands that originate largely as fermentation by-products of anaerobic bacteria in the gut. However, the presence of FFA2 and FFA3 on pancreatic ß-cells, FFA3 on neurons, and FFA2 on leukocytes and adipocytes means that the biologic role of these receptors likely extends beyond the widely accepted role of regulating peptide hormone release from enteroendocrine cells in the gut. Here, we review the physiologic roles of FFA2 and FFA3, the recent development and use of receptor-selective pharmacological tool compounds and genetic models available to study these receptors, and present evidence of the potential therapeutic value of targeting this emerging receptor pair.


Fatty Acids, Volatile/pharmacology , Fatty Acids, Volatile/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Fatty Acids, Volatile/chemistry , Humans , Immunity, Cellular/physiology , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/physiology , Receptors, G-Protein-Coupled/chemistry
12.
Proc Natl Acad Sci U S A ; 112(28): 8774-9, 2015 Jul 14.
Article En | MEDLINE | ID: mdl-26124120

Activation of the CB2 receptor is apparently an endogenous protective mechanism. Thus, it restrains inflammation and protects the skeleton against age-related bone loss. However, the endogenous cannabinoids, as well as Δ(9)-tetrahydrocannabinol, the main plant psychoactive constituent, activate both cannabinoid receptors, CB1 and CB2. HU-308 was among the first synthetic, selective CB2 agonists. HU-308 is antiosteoporotic and antiinflammatory. Here we show that the HU-308 enantiomer, designated HU-433, is 3-4 orders of magnitude more potent in osteoblast proliferation and osteoclast differentiation culture systems, as well as in mouse models, for the rescue of ovariectomy-induced bone loss and ear inflammation. HU-433 retains the HU-308 specificity for CB2, as shown by its failure to bind to the CB1 cannabinoid receptor, and has no activity in CB2-deficient cells and animals. Surprisingly, the CB2 binding affinity of HU-433 in terms of [(3)H]CP55,940 displacement and its effect on [(35)S]GTPγS accumulation is substantially lower compared with HU-308. A molecular-modeling analysis suggests that HU-433 and -308 have two different binding conformations within CB2, with one of them possibly responsible for the affinity difference, involving [(35)S]GTPγS and cAMP synthesis. Hence, different ligands may have different orientations relative to the same binding site. This situation questions the usefulness of universal radioligands for comparative binding studies. Moreover, orientation-targeted ligands have promising potential for the pharmacological activation of distinct processes.


Cannabinoid Receptor Agonists/pharmacology , Cannabinoids/pharmacology , Receptor, Cannabinoid, CB2/agonists , Animals , CHO Cells , Cannabinoid Receptor Agonists/chemistry , Cannabinoid Receptor Agonists/metabolism , Cannabinoids/chemistry , Cannabinoids/metabolism , Cricetinae , Cricetulus , Mice , Mice, Inbred C57BL , Stereoisomerism
13.
PLoS One ; 8(10): e76918, 2013.
Article En | MEDLINE | ID: mdl-24204703

In the present study, we found that CBD inhibited U87-MG and T98G cell proliferation and invasiveness in vitro and caused a decrease in the expression of a set of proteins specifically involved in growth, invasion and angiogenesis. In addition, CBD treatment caused a dose-related down-regulation of ERK and Akt prosurvival signaling pathways in U87-MG and T98G cells and decreased hypoxia inducible factor HIF-1α expression in U87-MG cells. Taken together, these results provide new insights into the antitumor action of CBD, showing that this cannabinoid affects multiple tumoral features and molecular pathways. As CBD is a non-psychoactive phytocannabinoid that appears to be devoid of side effects, our results support its exploitation as an effective anti-cancer drug in the management of gliomas.


Antineoplastic Agents/pharmacology , Cannabidiol/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioma/drug therapy , Glioma/metabolism , Glioma/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasm Invasiveness , Proteome/metabolism , Proteomics/methods , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
14.
Mol Pharmacol ; 83(2): 322-38, 2013 Feb.
Article En | MEDLINE | ID: mdl-23160940

We have previously identified allosteric modulators of the cannabinoid CB(1) receptor (Org 27569, PSNCBAM-1) that display a contradictory pharmacological profile: increasing the specific binding of the CB(1) receptor agonist [(3)H]CP55940 but producing a decrease in CB(1) receptor agonist efficacy. Here we investigated the effect one or both compounds in a broad range of signaling endpoints linked to CB(1) receptor activation. We assessed the effect of these compounds on CB(1) receptor agonist-induced [(35)S]GTPγS binding, inhibition, and stimulation of forskolin-stimulated cAMP production, phosphorylation of extracellular signal-regulated kinases (ERK), and ß-arrestin recruitment. We also investigated the effect of these allosteric modulators on CB(1) agonist binding kinetics. Both compounds display ligand dependence, being significantly more potent as modulators of CP55940 signaling as compared with WIN55212 and having little effect on [(3)H]WIN55212 binding. Org 27569 displays biased antagonism whereby it inhibits: agonist-induced guanosine 5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTPγS) binding, simulation (Gα(s)-mediated), and inhibition (Gα(i)-mediated) of cAMP production and ß-arrestin recruitment. In contrast, it acts as an enhancer of agonist-induced ERK phosphorylation. Alone, the compound can act also as an allosteric agonist, increasing cAMP production and ERK phosphorylation. We find that in both saturation and kinetic-binding experiments, the Org 27569 and PSNCBAM-1 appeared to influence only orthosteric ligand maximum occupancy rather than affinity. The data indicate that the allosteric modulators share a common mechanism whereby they increase available high-affinity CB(1) agonist binding sites. The receptor conformation stabilized by the allosterics appears to induce signaling and also selectively traffics orthosteric agonist signaling via the ERK phosphorylation pathway.


Allosteric Regulation/drug effects , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/metabolism , Animals , Arrestins/metabolism , Benzoxazines/pharmacology , Brain/drug effects , Brain/metabolism , CHO Cells , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Colforsin/pharmacology , Cricetinae , Cyclic AMP/metabolism , Cyclohexanols/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , HEK293 Cells , Humans , Indoles/pharmacology , Kinetics , Ligands , MAP Kinase Signaling System/drug effects , Male , Mice , Morpholines/pharmacology , Naphthalenes/pharmacology , Phosphorylation/drug effects , Piperidines/pharmacology , Protein Binding/drug effects , Signal Transduction/drug effects , beta-Arrestins
15.
Eur J Med Chem ; 58: 30-43, 2012 Dec.
Article En | MEDLINE | ID: mdl-23085772

Within our studies on structure-activity relationships of 4-quinolone-3-carboxamides as cannabinoid ligands, a new series of compounds characterized by a fluoro or phenylthio group at 7-position and different substituents at N1 and carboxamide nitrogen were synthesized and evaluated for their binding ability to cannabinoid type 1 (CB1) and type 2 (CB2) receptors. Most of the compounds showed affinity for one or both cannabinoid receptors at nanomolar concentration, with K(i)(CB1) and K(i)(CB2) values ranging from 2.45 to >10,000 nM and from 0.09 to 957 nM, respectively. The N-(3,4-dichlorobenzyl)amide derivatives 27 and 40 displayed relatively low affinity, but high selectivity towards the CB1 receptor. Compounds 4 and 40, a CB2 and a CB1 ligand, respectively, behaved as partial agonists in the [(35)S]GTPγS assay. They showed very low permeability through (MDCK-MDR1) cells and might, therefore, represent possible lead structures for further optimization in the search for cannabinoid ligands unable to cross the blood-brain barrier.


Quinolones/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Animals , CHO Cells , Cell Line , Cricetinae , Dogs , Dose-Response Relationship, Drug , Humans , Ligands , Molecular Structure , Quinolones/chemical synthesis , Quinolones/chemistry , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Structure-Activity Relationship
16.
ChemMedChem ; 7(5): 920-34, 2012 May.
Article En | MEDLINE | ID: mdl-22383251

Three heterocyclic systems were selected as potential bioisosteres of the amide linker for a series of 1,6-disubstituted-4-quinolone-3-carboxamides, which are potent and selective CB2 ligands that exhibit poor water solubility, with the aim of improving their physicochemical profile and also of clarifying properties of importance for amide bond mimicry. Among the newly synthesized compounds, a 1,2,3-triazole derivative (1-(adamantan-1-yl)-4-[6-(furan-2-yl)-1,4-dihydro-4-oxo-1-pentylquinolin-3-yl]-1H-1,2,3-triazole) emerged as the most promising in terms of both physicochemical and pharmacodynamic properties. When assayed in vitro, this derivative exhibited inverse agonist activity, whereas, in the formalin test in mice, it produced analgesic effects antagonized by a well-established inverse agonist. Metabolic studies allowed the identification of a side chain hydroxylated derivative as its only metabolite, which, in its racemic form, still showed appreciable CB2 selectivity, but was 150-fold less potent than the parent compound.


4-Quinolones/chemistry , Carboxylic Acids/chemistry , Computer Simulation , Receptor, Cannabinoid, CB2/agonists , Animals , CHO Cells , Cell Line, Tumor , Chromatography, High Pressure Liquid , Cricetinae , Cricetulus , Humans , Ligands , Lipids/chemistry , Mice , Models, Molecular , Molecular Structure , Protein Structure, Tertiary , Solubility , Structure-Activity Relationship
17.
Br J Pharmacol ; 165(8): 2561-74, 2012 Apr.
Article En | MEDLINE | ID: mdl-21615724

BACKGROUND AND PURPOSE: We have investigated how pre-incubating hCB(2) CHO cells with the CB(2) receptor antagonists/inverse agonists, AM630 and SR144528, affects how these and other ligands target hCB(2) receptors in these cells or their membranes. EXPERIMENTAL APPROACH: We tested the ability of AM630, SR144528 and of the CB(1) /CB(2) receptor agonists, CP55940 and R-(+)-WIN55212, to modulate forskolin-stimulated cAMP production in hCB(2) CHO cells or [(35) S]-GTPγS binding to membranes prepared from these cells, or to displace [(3) H]-CP55940 from whole cells and membranes. Assays were also performed with the CB(2) receptor partial agonist, Δ(9) -tetrahydrocannabivarin. Some cells were pre-incubated with AM630 or SR144528 and then washed extensively. KEY RESULTS: AM630 behaved as a low-potency neutral competitive antagonist in AM630-pre-incubated cells, a low-potency agonist in SR144528-pre-incubated cells, and a much higher-potency inverse agonist/antagonist in vehicle-pre-incubated cells. AM630 pre-incubation (i) reduced the inverse efficacy of SR144528 without abolishing it; (ii) increased the efficacy of Δ(9) -tetrahydrocannabivarin; and (iii) did not affect the potency with which AM630 displaced [(3) H]-CP55940 from whole cells or its inverse agonist potency and efficacy in the [(35) S]-GTPγS membrane assay. CONCLUSIONS AND IMPLICATIONS: These results suggest that AM630 is a protean ligand that can target a constitutively active form of the hCB(2) receptor (R*) with low affinity to produce agonism or neutral antagonism and a constitutively inactive form of this receptor (R) with much higher affinity to produce inverse agonism, and that the constitutive activity of whole cells is decreased less by pre-incubation with AM630 than with the higher-efficacy inverse agonist, SR144528. LINKED ARTICLES: This article is part of a themed section on Cannabinoids in Biology and Medicine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.165.issue-8. To view Part I of Cannabinoids in Biology and Medicine visit http://dx.doi.org/10.1111/bph.2011.163.issue-7.


Indoles/pharmacology , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Animals , Benzoxazines/pharmacology , CHO Cells , Camphanes/pharmacology , Cannabinoids/pharmacology , Cricetinae , Cricetulus , Cyclohexanols/pharmacology , Dronabinol/analogs & derivatives , Dronabinol/pharmacology , Humans , Ligands , Morpholines/pharmacology , Naphthalenes/pharmacology , Pyrazoles/pharmacology , Radioligand Assay , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/metabolism
18.
Eur J Pharmacol ; 651(1-3): 96-105, 2011 Jan 25.
Article En | MEDLINE | ID: mdl-21114999

Rimonabant, the prototypic antagonist of cannabinoid CB(1) receptors, has been reported to have inverse agonist properties at higher concentrations, which may complicate its use as a tool for mechanistic evaluation of cannabinoid pharmacology. Consequently, recent synthesis efforts have concentrated on discovery of a neutral antagonist using a variety of structural templates. The purpose of this study was to evaluate the pharmacological properties of the putative neutral cannabinoid CB(1) receptor antagonist O-2050, a sulfonamide side chain analog of Δ(8)-tetrahydrocannabinol. O-2050 and related sulfonamide cannabinoids exhibited good affinity for both cannabinoid CB(1) and CB(2) receptors. While the other sulfonamide analogs produced cannabinoid agonist effects in vivo (e.g., activity suppression, antinociception, and hypothermia), O-2050 stimulated activity and was inactive in the other two tests. O-2050 also decreased food intake in mice, an effect that was reminiscent of that produced by rimonabant. Unlike rimonabant, however, O-2050 did not block the effects of cannabinoid agonists in vivo, even when administered i.c.v. In contrast, O-2050 antagonized the in vitro effects of cannabinoid agonists in [(35)S]GTPγS and mouse vas deferens assays without having activity on its own in either assay. Further evaluation revealed that O-2050 fully and dose-dependently substituted for Δ(9)-tetrahydrocannabinol in a mouse drug discrimination procedure (a cannabinoid agonist effect) and that it inhibited forskolin-stimulated cyclic AMP signaling with a maximum efficacy of approximately half that of the full agonist CP55,940 [(-)-cis-3-[2-hydroxy-4(1,1-dimethyl-heptyl)phenyl]-trans-4-(3-hydroxy-propyl)cyclohexanol]. Together, these results suggest that O-2050 is not a viable candidate for classification as a neutral cannabinoid CB(1) receptor antagonist.


Dronabinol/analogs & derivatives , Pyrans/chemistry , Pyrans/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Animals , Cyclic AMP/metabolism , Dronabinol/chemistry , Dronabinol/metabolism , Dronabinol/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Male , Mice , Pyrans/metabolism , Receptor, Cannabinoid, CB1/metabolism , Vas Deferens/drug effects , Vas Deferens/metabolism
19.
J Med Chem ; 53(16): 5915-28, 2010 Aug 26.
Article En | MEDLINE | ID: mdl-20718492

A set of quinolone-3-carboxamides 2 bearing diverse substituents at position 1, 3, and 6 of the bicyclic nucleus was prepared. Except for six compounds exhibiting Ki>100 nM, all the quinolone-3-carboxamides 2 proved to be high affinity CB2 ligands, with Ki values ranging from 73.2 to 0.7 nM and selectivity [SI=Ki(CB1)/Ki(CB2)] varying from >14285 to 1.9, with only 2ah exhibiting a reverse selectivity (SI<1). In the formalin test of peripheral acute and inflammatory pain in mice, 2ae showed analgesic activity that was antagonized by a selective CB2 antagonist. By contrast, 2e was inactive per se and antagonized the effect of a selective CB2 agonist. Finally, 2g and 2p exhibited CB2 inverse agonist-like behavior in this in vivo test. However, two different functional assays carried out in vitro on 2e and 2g indicated for both compounds an overall inverse agonist activity at CB2 receptors.


Amides/chemical synthesis , Analgesics/chemical synthesis , Quinolones/chemical synthesis , Receptor, Cannabinoid, CB2/agonists , Amides/chemistry , Amides/pharmacology , Analgesics/chemistry , Analgesics/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Inverse Agonism , Humans , Ligands , Mice , Pain Measurement , Quinolones/chemistry , Quinolones/pharmacology , Radioligand Assay , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Stereoisomerism , Structure-Activity Relationship
20.
Br J Pharmacol ; 160(3): 677-87, 2010 Jun.
Article En | MEDLINE | ID: mdl-20590571

BACKGROUND AND PURPOSE: The phytocannabinoid, Delta(9)-tetrahydrocannabivarin (THCV), can block cannabinoid CB(1) receptors. This investigation explored its ability to activate CB(2) receptors, there being evidence that combined CB(2) activation/CB(1) blockade would ameliorate certain disorders. EXPERIMENTAL APPROACH: We tested the ability of THCV to activate CB(2) receptors by determining whether: (i) it inhibited forskolin-stimulated cyclic AMP production by Chinese hamster ovary (CHO) cells transfected with human CB(2) (hCB(2)) receptors; (ii) it stimulated [(35)S]GTPgammaS binding to hCB(2) CHO cell and mouse spleen membranes; (iii) it attenuated signs of inflammation/hyperalgesia induced in mouse hind paws by intraplantar injection of carrageenan or formalin; and (iv) any such anti-inflammatory or anti-hyperalgesic effects were blocked by a CB(1) or CB(2) receptor antagonist. KEY RESULTS: THCV inhibited cyclic AMP production by hCB(2) CHO cells (EC(50)= 38 nM), but not by hCB(1) or untransfected CHO cells or by hCB(2) CHO cells pre-incubated with pertussis toxin (100 ng.mL(-1)) and stimulated [(35)S]GTPgammaS binding to hCB(2) CHO and mouse spleen membranes. THCV (0.3 or 1 mg.kg(-1) i.p.) decreased carrageenan-induced oedema in a manner that seemed to be CB(2) receptor-mediated and suppressed carrageenan-induced hyperalgesia. THCV (i.p.) also decreased pain behaviour in phase 2 of the formalin test at 1 mg.kg(-1), and in both phases of this test at 5 mg.kg(-1); these effects of THCV appeared to be CB(1) and CB(2) receptor mediated. CONCLUSIONS AND IMPLICATIONS: THCV can activate CB(2) receptors in vitro and decrease signs of inflammation and inflammatory pain in mice partly via CB(1) and/or CB(2) receptor activation.


Dronabinol/analogs & derivatives , Inflammation/drug therapy , Pain/drug therapy , Receptor, Cannabinoid, CB2/agonists , Animals , CHO Cells , Cell Line, Transformed , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Dronabinol/pharmacology , Dronabinol/therapeutic use , Edema/drug therapy , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Inflammation/complications , Male , Membranes/metabolism , Mice , Mice, Inbred C57BL , Pain/complications , Pain Measurement/methods , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Spleen/drug effects , Spleen/metabolism
...