Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Pharmacol Rev ; 75(5): 885-958, 2023 09.
Article in English | MEDLINE | ID: mdl-37164640

ABSTRACT

The cannabis derivative marijuana is the most widely used recreational drug in the Western world and is consumed by an estimated 83 million individuals (∼3% of the world population). In recent years, there has been a marked transformation in society regarding the risk perception of cannabis, driven by its legalization and medical use in many states in the United States and worldwide. Compelling research evidence and the Food and Drug Administration cannabis-derived cannabidiol approval for severe childhood epilepsy have confirmed the large therapeutic potential of cannabidiol itself, Δ9-tetrahydrocannabinol and other plant-derived cannabinoids (phytocannabinoids). Of note, our body has a complex endocannabinoid system (ECS)-made of receptors, metabolic enzymes, and transporters-that is also regulated by phytocannabinoids. The first endocannabinoid to be discovered 30 years ago was anandamide (N-arachidonoyl-ethanolamine); since then, distinct elements of the ECS have been the target of drug design programs aimed at curing (or at least slowing down) a number of human diseases, both in the central nervous system and at the periphery. Here a critical review of our knowledge of the goods and bads of the ECS as a therapeutic target is presented to define the benefits of ECS-active phytocannabinoids and ECS-oriented synthetic drugs for human health. SIGNIFICANCE STATEMENT: The endocannabinoid system plays important roles virtually everywhere in our body and is either involved in mediating key processes of central and peripheral diseases or represents a therapeutic target for treatment. Therefore, understanding the structure, function, and pharmacology of the components of this complex system, and in particular of key receptors (like cannabinoid receptors 1 and 2) and metabolic enzymes (like fatty acid amide hydrolase and monoacylglycerol lipase), will advance our understanding of endocannabinoid signaling and activity at molecular, cellular, and system levels, providing new opportunities to treat patients.


Subject(s)
Cannabidiol , Cannabinoids , Cannabis , Hallucinogens , Humans , Child , Endocannabinoids/metabolism , Cannabidiol/therapeutic use , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Cannabinoids/metabolism , Dronabinol , Cannabis/chemistry , Cannabis/metabolism , Carrier Proteins , Cannabinoid Receptor Agonists
2.
J Pharmacol Exp Ther ; 372(1): 119-127, 2020 01.
Article in English | MEDLINE | ID: mdl-31641018

ABSTRACT

Despite a growing acceptance that withdrawal symptoms can emerge following discontinuation of cannabis products, especially in high-intake chronic users, there are no Food and Drug Administration (FDA)-approved treatment options. Drug development has been hampered by difficulties studying cannabis withdrawal in laboratory animals. One preclinical approach that has been effective in studying withdrawal from drugs in several pharmacological classes is antagonist drug discrimination. The present studies were designed to examine this paradigm in squirrel monkeys treated daily with the long-acting CB1 agonist AM2389 (0.01 mg/kg) and trained to discriminate the CB1 inverse agonist/antagonist rimonabant (0.3 mg/kg) from saline. The discriminative-stimulus effects of rimonabant were both dose and time dependent and, importantly, could be reproduced by discontinuation of agonist treatment. Antagonist substitution tests with the CB1 neutral antagonists AM4113 (0.03-0.3 mg/kg), AM6527 (0.03-1.0 mg/kg), and AM6545 (0.03-1.0 mg/kg) confirmed that the rimonabant discriminative stimulus also could be reproduced by CB1 antagonists lacking inverse agonist action. Agonist substitution tests with the phytocannabinoid ∆9-tetrahydrocannabinol (0.1-1.0 mg/kg), synthetic CB1 agonists nabilone (0.01-0.1 mg/kg), AM4054 (0.01-0.03 mg/kg), K2/Spice compound JWH-018 (0.03-0.3 mg/kg), FAAH-selective inhibitors AM3506 (0.3-5.6 mg/kg), URB597 (3.0-5.6 mg/kg), and nonselective FAAH/MGL inhibitor AM4302 (3.0-10.0 mg/kg) revealed that only agonists with CB1 affinity were able to reduce the rimonabant-like discriminative stimulus effects of withholding daily agonist treatment. Although the present studies did not document physiologic disturbances associated with withdrawal, the results are consistent with the view that the cannabinoid antagonist drug discrimination paradigm provides a useful screening procedure for examining the ability of candidate medications to attenuate the interoceptive stimuli provoked by cannabis discontinuation. SIGNIFICANCE STATEMENT: Despite a growing acceptance that withdrawal symptoms can emerge following the discontinuation of cannabis products, especially in high-intake chronic users, there are no FDA-approved pharmacotherapies to assist those seeking treatment. The present studies systematically examined cannabinoid antagonist drug discrimination, a preclinical animal model that is designed to appraise the ability of candidate medications to attenuate the interoceptive effects that accompany abrupt cannabis abstinence.


Subject(s)
Cannabinoid Receptor Antagonists/therapeutic use , Discrimination, Psychological , Disease Models, Animal , Substance Withdrawal Syndrome/drug therapy , Animals , Benzopyrans/administration & dosage , Benzopyrans/adverse effects , Benzopyrans/therapeutic use , Cannabinoid Receptor Agonists/administration & dosage , Cannabinoid Receptor Agonists/adverse effects , Cannabinoid Receptor Agonists/therapeutic use , Cannabinoid Receptor Antagonists/administration & dosage , Cannabinoid Receptor Antagonists/adverse effects , Drug Evaluation, Preclinical/methods , Drug Substitution/methods , Male , Rimonabant/administration & dosage , Rimonabant/adverse effects , Rimonabant/therapeutic use , Saimiri , Substance Withdrawal Syndrome/psychology
3.
Lancet Psychiatry ; 6(1): 35-45, 2019 01.
Article in English | MEDLINE | ID: mdl-30528676

ABSTRACT

BACKGROUND: Cannabis is one of the most widely used drugs worldwide. Cannabis use disorder is characterised by recurrent use of cannabis that causes significant clinical and functional impairment. There are no approved pharmacological treatments for cannabis use disorder. One approach is to potentiate endocannabinoid signalling by inhibiting fatty acid amide hydrolase (FAAH), the enzyme that degrades the endocannabinoid anandamide. We aimed to test the efficacy and safety of the FAAH-inhibitor PF-04457845 in reduction of cannabis withdrawal and cannabis use in men who were daily cannabis users. METHODS: We did a double-blind, placebo-controlled, parallel group phase 2a trial at one site in men aged 18-55 years with cannabis dependence according to DSM-IV criteria (equivalent to cannabis use disorder in DSM-5). After baseline assessments, participants were randomly assigned (2:1) to receive PF-04457845 (4 mg per day) or placebo using a fixed block size of six participants, stratified by severity of cannabis use and desire to quit. Participants were admitted to hospital for 5 days (maximum 8 days) to achieve abstinence and precipitate cannabis withdrawal, after which they were discharged to continue the remaining 3 weeks of treatment as outpatients. The primary endpoints were treatment-related differences in cannabis withdrawal symptoms during hospital admission, and week 4 (end of treatment) self-reported cannabis use and urine THC-COOH concentrations in the intention-to-treat population. The study is registered at ClinicalTrials.gov, number NCT01618656. FINDINGS: Between Sept 12, 2012, and Jan 18, 2016, 46 men were randomly assigned to PF-04457845 and 24 to placebo. Adherence to study medication was 88%, as confirmed by video-calling and pill count, and corroborated by corresponding drug and anandamide concentrations in blood. Relative to placebo, treatment with PF-04457845 was associated with reduced symptoms of cannabis withdrawal (first day of treatment mean symptom score 11·00 [95% CI 7·78-15·57] vs 6·04 [4·43-8·24]; difference 4·96 [0·71-9·21]; padj=0·048; second day of treatment 11·74 [8·28-16·66] vs 6·02 [4·28-8·47]; difference 5·73 [1·13-10·32]; padj=0·035) and related mood symptoms during the inpatient phase. Additionally, treatment with PF-04457845 was associated with lower self-reported cannabis use at 4 weeks (mean 1·27 joints per day [95% CI 0·82-1·97] vs 0·40 [0·25-0·62]; difference 0·88 [0·29-1·46]; p=0·0003) and lower urinary THC-COOH concentrations (mean 657·92 ng/mL [95% CI 381·60-1134·30] vs 265·55 [175·60-401·57]; difference 392·37 [17·55-767·18)]; p=0·009). Eight (17%) patients in the PF-04457845 group and four (17%) in the placebo group discontinued during the treatment period. During the 4-week treatment phase, 20 (43%) of 46 participants in the PF-04457845 group and 11 (46%) of 24 participants in the placebo group had an adverse event. There were no serious adverse events. INTERPRETATION: PF-04457845, a novel FAAH inhibitor, reduced cannabis withdrawal symptoms and cannabis use in men, and might represent an effective and safe approach for the treatment of cannabis use disorder. FUNDING: United States National Institute of Drug Abuse (NIDA).


Subject(s)
Cannabis , Marijuana Abuse/drug therapy , Pyridazines/administration & dosage , Substance Withdrawal Syndrome , Urea/analogs & derivatives , Adolescent , Adult , Amidohydrolases , Double-Blind Method , Humans , Male , Marijuana Smoking , Middle Aged , Treatment Outcome , Urea/administration & dosage , Young Adult
4.
Neuropharmacology ; 144: 327-336, 2019 01.
Article in English | MEDLINE | ID: mdl-30439418

ABSTRACT

Tobacco smoke is the leading preventable cause of death in the world and treatments aimed to increase success rate in smoking cessation by reducing nicotine dependence are sought. Activation of peroxisome proliferator-activated receptor-alpha (PPARα) by synthetic or endogenous agonists was shown to suppress nicotine-induced activation of mesolimbic dopamine system, one of the major neurobiological substrates of nicotine dependence, and nicotine-seeking behavior in rats and monkeys. An alternative indirect way to activate PPARα is inhibition of N-acylethanolamine acid amidase (NAAA), one of the major hydrolyzing enzyme for its endogenous agonists palmitoylethanolamide (PEA) and oleoylethanolamide (OEA). We synthetized a novel specific brain permeable NAAA inhibitor, AM11095. We administered AM11095 to rats and carried out brain lipid analysis, a functional observational battery (FOB) to assess toxicity, in vivo electrophysiological recording from dopamine cells in the ventral tegmental area, brain microdialysis in the nucleus accumbens shell and behavioral experiments to assess its effect on nicotine -induced conditioned place preference (CPP). AM11095 (5 and 25 mg/kg, i.p.) was devoid of neurotoxic and behavioral effects and did not affect motor behavior and coordination. This NAAA inhibitor (5 mg/kg i.p.) increased OEA and PEA levels in the hippocampus and cortex, prevented nicotine-induced activation of mesolimbic dopamine neurons in the ventral tegmental area, nicotine-induced elevation of dopamine levels in the nucleus accumbens shell and decreased the expression of nicotine CPP. Our results indicate that NAAA inhibitors represent a new class of pharmacological tools to modulate brain PEA/PPARα signalling and show potential in the treatment of nicotine dependence.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Dopamine/metabolism , Nicotine/pharmacology , Psychotropic Drugs/pharmacology , Reward , Amidohydrolases/metabolism , Animals , Brain/drug effects , Brain/metabolism , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Male , Mice , Nicotinic Agonists/pharmacology , Random Allocation , Rats, Sprague-Dawley , Spatial Behavior/drug effects , Spatial Behavior/physiology
5.
Nutrients ; 10(9)2018 Sep 12.
Article in English | MEDLINE | ID: mdl-30213124

ABSTRACT

Recognized as the gold standard, human milk (HM) is an extremely complex yet fascinating biofluid tailored to meet an infant's nutritional requirements throughout development. Endocannabinoids and endocannabinoid-like compounds (endocannabinoid metabolome, ECM) are endogenous lipid mediators derived from long-chain polyunsaturated fatty acids that have been identified in HM. Previous research has shown that arachidonoylglycerol might play a role in establishing the infant's suckling response during lactation by activating the type 1 cannabinoid receptor in the infant's brain. The mechanisms of action and the role of the ECM in HM are not fully understood. Transitional and mature milk samples were collected from lactating women (n = 24) for ECM characterization, quantification, and to evaluate differences among the two stages. HM samples were analyzed by liquid chromatography-mass spectrometry. Identified members of the ECM were: arachidonoylethanolamine, palmitoylethanolamine, oleoylethanolamine, docosahexaenoylethanolamine, eicoapentaenoylethanolamine, eicosenoylethanolamine, arachidonoylglycerol, palmitoyglycerol, oleoylglycerol, docosahexaenoylglycerol, eicosapentaenoylglycerol, eiconenooylglycerol, arachidonic acid, docosahexaenoic acid, and eicosapentaenoic acid. Only docosahexaenoylglycerol was different across transitional and mature milk (p ≤ 0.05). Data from this cohort suggest that bioactive constituents in HM may also play a role in infant health and development. Future studies can be developed based on this study's data to help elucidate specific roles for each ECM member in addition to understanding how the ECM modulates infant health.


Subject(s)
Breast Milk Expression , Endocannabinoids/metabolism , Lactation , Metabolome , Metabolomics/methods , Milk, Human/metabolism , Adolescent , Adult , Breast Feeding , Child Development , Chromatography, Liquid , Female , Humans , Infant , Infant Nutritional Physiological Phenomena , Infant, Newborn , Mass Spectrometry , Nutritive Value , Young Adult
6.
Nephrol Dial Transplant ; 32(10): 1655-1665, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28387811

ABSTRACT

BACKGROUND: The endocannabinoid system has been implicated in the pathogenesis of diabetic nephropathy (DN). We investigated the effect of combined therapy with AM6545, a 'peripherally' restricted cannabinoid receptor type 1 (CB1R) neutral antagonist, and AM1241, a cannabinoid receptor type 2 (CB2R) agonist, in experimental DN. METHODS: Renal function and structure, podocyte proteins and markers of both fibrosis and inflammation were studied in streptozotocin-induced diabetic mice treated for 14 weeks with vehicle, AM6545, AM1241 and AM6545-AM1241. RESULTS: Single treatment with either AM6545 or AM1241 alone reduced diabetes-induced albuminuria and prevented nephrin loss both in vivo and in vitro in podocytes exposed to glycated albumin. Dual therapy performed better than monotherapies, as it abolished albuminuria, inflammation, tubular injury and markedly reduced renal fibrosis. Converging anti-inflammatory mechanisms provide an explanation for this greater efficacy as dual therapy abolished diabetes-induced renal monocyte infiltration and M1/M2 macrophage imbalance in vivo and abrogated the profibrotic effect of M1 macrophage-conditioned media on cultured mesangial cells. CONCLUSION: 'Peripheral' CB1R blockade is beneficial in experimental DN and this effect is synergically magnified by CB2R activation.


Subject(s)
Cannabinoid Receptor Agonists/administration & dosage , Cannabinoid Receptor Antagonists/administration & dosage , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/prevention & control , Morpholines/administration & dosage , Pyrazoles/administration & dosage , Albuminuria/prevention & control , Animals , Anti-Inflammatory Agents/administration & dosage , Cannabinoids/administration & dosage , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Drug Combinations , Drug Evaluation, Preclinical , Male , Mice , Mice, Inbred C57BL , Neutrophil Activation/drug effects , Podocytes/drug effects , Podocytes/metabolism
7.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(5): 474-484, 2017 May.
Article in English | MEDLINE | ID: mdl-28065729

ABSTRACT

N-acylethanolamines (NAEs) such as N-palmitoylethanolamine and anandamide are endogenous bioactive lipids having numerous functions, including the control of inflammation. Their levels and therefore actions can be controlled by modulating the activity of two hydrolytic enzymes, N-acylethanolamine-hydrolyzing acid amidase (NAAA) and fatty acid amide hydrolase (FAAH). As macrophages are key to inflammatory processes, we used lipopolysaccharide-activated J774 macrophages, as well as primary mouse alveolar macrophages, to study the effect of FAAH and NAAA inhibition, using PF-3845 and AM9053 respectively, on macrophage activation and NAE levels measured by HPLC-MS. Markers of macrophage activation were measured by qRT-PCR and ELISA. Activation of macrophages decreased NAAA expression and NAE hydrolytic activity. FAAH and NAAA inhibition increased the levels of the different NAEs, although with different magnitudes, whether in control condition or following LPS-induced macrophage activation. Both inhibitors reduced several markers of macrophage activation, such as mRNA expression of inflammatory mediators, as well as cytokine and prostaglandin production, with however some differences between FAAH and NAAA inhibition. Most of the NAEs tested - including N-docosatetraenoylethanolamine and N-docosahexaenoylethanolamine - also reduced LPS-induced mRNA expression of inflammatory mediators, again with differences depending on the marker and the NAE, thus offering a potential explanation for the differential effect of the inhibitors on macrophage activation markers. In conclusion, we show different and complementary effects of NAE on lipopolysaccharide-induced macrophage activation. Our results support an important role for inhibition of NAE hydrolysis and NAAA inhibition in particular in controlling macrophage activation, and thus inflammation.


Subject(s)
Amidohydrolases/metabolism , Ethanolamines/metabolism , Inflammation/drug therapy , Palmitic Acids/metabolism , Amides , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/chemistry , Animals , Arachidonic Acids/metabolism , Endocannabinoids/metabolism , Ethanolamines/chemistry , Gene Expression Regulation, Enzymologic/drug effects , Humans , Inflammation/enzymology , Inflammation/metabolism , Lipopolysaccharides/toxicity , Macrophage Activation/drug effects , Macrophage Activation/genetics , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/enzymology , Mice , Palmitic Acids/chemistry , Piperidines/administration & dosage , Polyunsaturated Alkamides/metabolism , Pyridines/administration & dosage
8.
ACS Chem Neurosci ; 6(8): 1400-10, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-25978068

ABSTRACT

The cannabinoid 1 receptor (CB1R) is one of the most abundant G protein-coupled receptors (GPCRs) in the central nervous system. CB1R involvement in multiple physiological processes, especially neurotransmitter release and synaptic function, has made this GPCR a prime drug discovery target, and pharmacological CB1R activation has been demonstrated to be a tenable therapeutic modality. Accordingly, the design and profiling of novel, drug-like CB1R modulators to inform the receptor's ligand-interaction landscape and molecular pharmacology constitute a prime contemporary research focus. For this purpose, we report utilization of AM3677, a designer endocannabinoid (anandamide) analogue derivatized with a reactive electrophilic isothiocyanate functionality, as a covalent, CB1R-selective chemical probe. The data demonstrate that reaction of AM3677 with a cysteine residue in transmembrane helix 6 of human CB1R (hCB1R), C6.47(355), is a key feature of AM3677's ligand-binding motif. Pharmacologically, AM3677 acts as a high-affinity, low-efficacy CB1R agonist that inhibits forskolin-stimulated cellular cAMP formation and stimulates CB1R coupling to G protein. AM3677 also induces CB1R endocytosis and irreversible receptor internalization. Computational docking suggests the importance of discrete hydrogen bonding and aromatic interactions as determinants of AM3677's topology within the ligand-binding pocket of active-state hCB1R. These results constitute the initial identification and characterization of a potent, high-affinity, hCB1R-selective covalent agonist with utility as a pharmacologically active, orthosteric-site probe for providing insight into structure-function correlates of ligand-induced CB1R activation and the molecular features of that activation by the native ligand, anandamide.


Subject(s)
Arachidonic Acids/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Isothiocyanates/pharmacology , Animals , Arachidonic Acids/chemistry , Cannabinoid Receptor Agonists/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Colforsin , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Endocytosis/drug effects , HEK293 Cells , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Hydrogen Bonding , Isothiocyanates/chemistry , Mice , Molecular Docking Simulation , Molecular Structure , Mutation , Radioligand Assay , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Transfection
9.
J Pain ; 16(8): 707-16, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25958314

ABSTRACT

UNLABELLED: Omega-3 and omega-6 fatty acids are biosynthetic precursors of endocannabinoids with antinociceptive, anxiolytic, and neurogenic properties. We recently reported that targeted dietary manipulation-increasing omega-3 fatty acids while reducing omega-6 linoleic acid (the H3-L6 intervention)-reduced headache pain and psychological distress among chronic headache patients. It is not yet known whether these clinical improvements were due to changes in endocannabinoids and related mediators derived from omega-3 and omega-6 fatty acids. We therefore used data from this trial (N = 55) to investigate 1) whether the H3-L6 intervention altered omega-3- and omega-6-derived endocannabinoids in plasma and 2) whether diet-induced changes in these bioactive lipids were associated with clinical improvements. The H3-L6 intervention significantly increased the omega-3 docosahexaenoic acid derivatives 2-docosahexaenoylglycerol (+65%, P < .001) and docosahexaenoylethanolamine (+99%, P < .001) and reduced the omega-6 arachidonic acid derivative 2-arachidonoylglycerol (-25%, P = .001). Diet-induced changes in these endocannabinoid derivatives of omega-3 docosahexaenoic acid, but not omega-6 arachidonic acid, correlated with reductions in physical pain and psychological distress. These findings demonstrate that targeted dietary manipulation can alter endocannabinoids derived from omega-3 and omega-6 fatty acids in humans and suggest that 2-docosahexaenoylglycerol and docosahexaenoylethanolamine could have physical and/or psychological pain modulating properties. TRIAL REGISTRATION: ClinicalTrials.gov (NCT01157208) PERSPECTIVE: This article demonstrates that targeted dietary manipulation can alter endocannabinoids derived from omega-3 and omega-6 fatty acids and that these changes are related to reductions in headache pain and psychological distress. These findings suggest that dietary interventions could provide an effective, complementary approach for managing chronic pain and related conditions.


Subject(s)
Diet , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/metabolism , Headache/diet therapy , Stress, Psychological/diet therapy , Adolescent , Adult , Aged , Endocannabinoids/administration & dosage , Fatty Acids/administration & dosage , Female , Headache/metabolism , Humans , Male , Middle Aged , Psychological Tests , Quality of Life , Stress, Psychological/metabolism , Young Adult
10.
Bioorg Med Chem Lett ; 21(19): 5999-6002, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21880487

ABSTRACT

To obtain information on the pharmacophoric requirements of the CB1/CB2 partial agonist BAY 59-3074 we have synthesized a series of new conformationally constrained dibenzofuran (4a-d) and dibenzopyran analogs (5). All constrained analogs exhibited reduced binding affinity at both cannabinoid receptor subtypes, suggesting that planar conformations of these ligands are less favored by both receptors. We also found that 4c, 4d, and 5 exhibited 3- to 12-fold selectivity for hCB2 over rCB1 receptors and may serve as new chemotypes for the development of CB2-selective cannabinergics.


Subject(s)
Alkanesulfonates/metabolism , Analgesics/metabolism , Benzofurans/metabolism , Benzopyrans/metabolism , Drug Design , Nitriles/metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Alkanesulfonates/chemistry , Alkanesulfonates/pharmacology , Analgesics/chemical synthesis , Analgesics/chemistry , Analgesics/pharmacology , Animals , Benzofurans/chemical synthesis , Benzofurans/chemistry , Benzofurans/pharmacology , Benzopyrans/chemical synthesis , Benzopyrans/chemistry , Benzopyrans/pharmacology , Chronic Pain/drug therapy , Chronic Pain/physiopathology , Computer Simulation , Drug Evaluation, Preclinical , Humans , Ligands , Mice , Molecular Conformation , Molecular Targeted Therapy , Neuralgia/drug therapy , Neuralgia/physiopathology , Nitriles/chemistry , Nitriles/pharmacology , Protein Binding , Radioligand Assay , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Software , Structure-Activity Relationship , Substrate Specificity
11.
Biochim Biophys Acta ; 1808(9): 2095-101, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21129361

ABSTRACT

Two key commonly used cannabinergic agonists, CP55940 and WIN55212-2, are investigated for their effects on the lipid membrane bilayer using (2)H solid state NMR, and the results are compared with our earlier work with delta-9-tetrahydrocannabinol (Δ(9)-THC). To study the effects of these ligands we used hydrated bilayers of dipalmitoylphosphatidylcholine (DPPC) deuterated at the 2' and 16' positions of both acyl chains with deuterium atoms serving as probes for the dynamic and phase changes at the membrane interface and at the bilayer center respectively. All three cannabinergic ligands lower the phospholipid membrane phase transition temperature, increase the lipid sn-2 chain order parameter at the membrane interface and decrease the order at the center of the bilayer. Our studies show that the cannabinoid ligands induce lateral phase separation in the lipid membrane at physiological temperatures. During the lipid membrane phase transition, the cooperative dynamic process whereby the C-(2)H segments at the interface and center of the bilayer spontaneously reach the fast exchange regime ((2)H NMR timescale) is distinctively modulated by the two cannabinoids. Specifically, CP55940 is slightly more efficient at inducing liquid crystalline-type (2)H NMR spectral features at the membrane interface compared to WIN55212-2. In contrast, WIN55212-2 has a far superior ability to induce liquid crystalline-type spectral features at the center of the bilayer, and it increases the order parameter of the sn-1 chain in addition to the sn-2 chain of the lipids. These observations suggest the cannabinoid ligands may influence lipid membrane domain formations and there may be contributions to their cannabinergic activities through lipid membrane microdomain related mechanisms. Our work demonstrates that experimental design strategies utilizing specifically deuterium labeled lipids yield more detailed insights concerning the properties of lipid bilayers.


Subject(s)
Benzoxazines/pharmacology , Cyclohexanols/pharmacology , Lipid Bilayers/metabolism , Magnetic Resonance Spectroscopy/methods , Morpholines/pharmacology , Naphthalenes/pharmacology , Phospholipids/chemistry , Cannabinoids , Cell Membrane/metabolism , Chemistry, Pharmaceutical/methods , Crystallization , Drug Evaluation, Preclinical/methods , Immunosuppressive Agents/pharmacology , Ligands , Lipid Bilayers/chemistry , Models, Chemical , Protein Conformation , Temperature
12.
J Lipid Res ; 51(6): 1416-23, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20071693

ABSTRACT

The endocannabinoid metabolome consists of a growing, (patho)physiologically important family of fatty-acid derived signaling lipids. Diet is a major source of fatty acid substrate for mammalian endocannabinoid biosynthesis. The principal long-chain PUFA found in mammalian brain, docosahexaenoic acid (DHA), supports neurological function, retinal development, and overall health. The extent to which dietary DHA supplementation influences endocannabinoid-related metabolites in brain, within the context of the circulating endocannabinoid profile, is currently unknown. We report the first lipidomic analysis of acute 2-week DHA dietary supplementation effects on the physiological state of 15 fatty-acid, N-acylethanolamine, and glycerol-ester endocannabinoid metabolome constituents in murine plasma and brain. The DHA-rich diet markedly elevated DHA, eicosapentaenoic acid, 2-eicosapentanoylglycerol (EPG), and docosahexanoylethanolamine in both compartments. Dietary DHA enhancement generally affected the synthesis of the N-acyl-ethanolamine and glycerol-ester metabolites to favor the docosahexaenoic and eicosapentaenoic vs. arachidonoyl and oleoyl homologs in both brain and plasma. The greater overall responsiveness of the endocannabinoid metabolome in plasma versus brain may reflect a more circumscribed homeostatic response range of brain lipids to dietary DHA supplementation. The ability of short-term DHA enhancement to modulate select constituents of the physiological brain and plasma endocannabinoid metabolomes carries metabolic and therapeutic implications.


Subject(s)
Brain/drug effects , Brain/metabolism , Cannabinoid Receptor Modulators/blood , Cannabinoid Receptor Modulators/metabolism , Dietary Supplements , Docosahexaenoic Acids/pharmacology , Endocannabinoids , Animals , Body Weight/drug effects , Chromatography, Liquid , Ethanolamines/metabolism , Fatty Acids, Unsaturated/metabolism , Glycerol/chemistry , Glycerol/metabolism , Lipid Metabolism/drug effects , Male , Metabolome/drug effects , Mice , Tandem Mass Spectrometry , Time Factors
13.
Expert Opin Emerg Drugs ; 14(1): 43-65, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19249987

ABSTRACT

The endogenous cannabinoid (CB) (endocannabinoid) signaling system is involved in a variety of (patho)physiological processes, primarily by virtue of natural, arachidonic acid-derived lipids (endocannabinoids) that activate G protein-coupled CB1 and CB2 receptors. A hyperactive endocannabinoid system appears to contribute to the etiology of several disease states that constitute significant global threats to human health. Consequently, mounting interest surrounds the design and profiling of receptor-targeted CB antagonists as pharmacotherapeutics that attenuate endocannabinoid transmission for salutary gain. Experimental and clinical evidence supports the therapeutic potential of CB1 receptor antagonists to treat overweight/obesity, obesity-related cardiometabolic disorders, and substance abuse. Laboratory data suggest that CB2 receptor antagonists might be effective immunomodulatory and, perhaps, anti-inflammatory drugs. One CB1 receptor antagonist/inverse agonist, rimonabant, has emerged as the first-in-class drug approved outside the United States for weight control. Select follow-on agents (taranabant, otenabant, surinabant, rosonabant, SLV-319, AVE1625, V24343) have also been studied in the clinic. However, rimonabant's market withdrawal in the European Union and suspension of rimonabant's, taranabant's, and otenabant's ongoing development programs have highlighted some adverse clinical side effects (especially nausea and psychiatric disturbances) of CB1 receptor antagonists/inverse agonists. Novel CB1 receptor ligands that are peripherally directed and/or exhibit neutral antagonism (the latter not affecting constitutive CB1 receptor signaling) may optimize the benefits of CB1 receptor antagonists while minimizing any risk. Indeed, CB1 receptor-neutral antagonists appear from preclinical data to offer efficacy comparable to or better than that of prototype CB1 receptor antagonists/inverse agonists, with less propensity to induce nausea. Continued pharmacological profiling, as the prelude to first-in-man testing of CB1 receptor antagonists with unique modes of targeting/pharmacological action, represents an exciting translational frontier in the critical path to CB receptor blockers as medicines.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Animals , Clinical Trials as Topic , Drug Delivery Systems , Drug Design , Drug Evaluation, Preclinical , Humans , Obesity/drug therapy , Obesity/physiopathology , Signal Transduction/drug effects
14.
Trends Pharmacol Sci ; 30(1): 1-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19042036

ABSTRACT

Endocannabinoids, endogenous lipid ligands of cannabinoid receptors, mediate a variety of effects similar to those of marijuana. Cannabinoid CB(1) receptors are highly abundant in the brain and mediate psychotropic effects, which limits their value as a potential therapeutic target. There is growing evidence for CB(1) receptors in peripheral tissues that modulate a variety of functions, including pain sensitivity and obesity-related hormonal and metabolic abnormalities. In this review we propose that selective targeting of peripheral CB(1) receptors has potential therapeutic value because it would help to minimize addictive, psychoactive effects in the case of CB(1) agonists used as analgesics, or depression and anxiety in the case of CB(1) antagonists used in the management of cardiometabolic risk factors associated with the metabolic syndrome.


Subject(s)
Metabolic Diseases/drug therapy , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Animals , Drug Design , Humans , Inflammation/physiopathology , Inflammation/prevention & control , Metabolic Diseases/physiopathology , Metabolic Diseases/prevention & control , Pain/drug therapy , Pain/physiopathology , Pain/prevention & control , Receptor, Cannabinoid, CB1/physiology , Terminology as Topic
15.
Curr Psychiatry Rep ; 9(5): 365-73, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17915075

ABSTRACT

The endogenous endocannabinoid system encompasses a family of natural signaling lipids ("endocannabinoids") functionally related to (9)-tetrahydrocannabinol, the psychoactive ingredient of marijuana (cannabis), along with proteins that modulate the endocannabinoids, including enzymes, transporters, and receptors. The endocannabinoid system's ubiquitous regulatory actions in health and disease underscore its importance to mammalian (patho)physiology and suggest discrete targets through which it may be modulated for therapeutic gain. Medications based on the endocannabinoid system are an important focus of contemporary translational research, particularly with respect to substance abuse and obesity, two prevalent disorders with a pathogenic component of endocannabinoid system hyperactivity. Pressing health care needs have made the rational design of targeted CB1 cannabinoid-receptor modulators a promising route to future medications with significant therapeutic impact against psychobehavioral and metabolic disturbances having a reward-supported appetitive component.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Cannabinoid Receptor Modulators/therapeutic use , Obesity/rehabilitation , Receptors, Cannabinoid/drug effects , Substance-Related Disorders/rehabilitation , Humans , Receptor, Cannabinoid, CB1/drug effects
16.
Curr Top Med Chem ; 6(13): 1401-26, 2006.
Article in English | MEDLINE | ID: mdl-16918457

ABSTRACT

Habitual cannabis use has been shown to affect the human immune system, and recent advances in endocannabinoid research provide a basis for understanding these immunomodulatory effects. Cell-based experiments or in vivo animal testing suggest that regulation of the endocannabinoid circuitry can impact almost every major function associated with the immune system. These studies were assisted by the development of numerous novel molecules that exert their biological effects through the endocannabinoid system. Several of these compounds were tested for their effects on immune function, and the results suggest therapeutic opportunities for a variety of inflammatory diseases such as multiple sclerosis, rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, allergic asthma, and autoimmune diabetes through modulation of the endocannabinoid system.


Subject(s)
Anti-Inflammatory Agents , Cannabinoid Receptor Modulators , Drug Design , Receptors, Cannabinoid , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cannabinoid Receptor Modulators/immunology , Cannabinoid Receptor Modulators/metabolism , Cannabinoid Receptor Modulators/physiology , Humans , Ligands , Molecular Structure , Receptors, Cannabinoid/immunology , Receptors, Cannabinoid/metabolism , Receptors, Cannabinoid/physiology , Structure-Activity Relationship
17.
Curr Pharm Des ; 12(14): 1751-69, 2006.
Article in English | MEDLINE | ID: mdl-16712486

ABSTRACT

The cannabinoid receptors CB1 and CB2 are family A, G-protein Coupled Receptors that mediate the effects of cannabinoids, a class of compounds that are so named because the first members were isolates of the cannabis plant. In recent history, there has been much anecdotal evidence that the potent and diverse physiological responses produced by these compounds can be turned to therapeutic benefit for a wide variety of maladies. The remarkable abundance of cannabinoid receptors and the discovery of several endogenous ligands along with enzyme and transporter proteins for which they are substrates, suggests that an endogenous cannabinoid neuromodulatory system is an important mediator of biological function. For these reasons CB1 and CB2 receptors are attractive targets for the design of therapeutic ligands. The action of these receptors, however, may also be modulated by manipulating the enzymes and membrane transporters that regulate the endogenous ligands. Despite the range of physiological processes and activities that are mediated by cannabinoid receptors, it is clear that it is possible to produce ligands that result in differential responses. In this paper, we review the pharmacophoric elements that lead to these differential responses and in order to discuss them in context we present an overview of structural aspects governing cannabinoid receptor function, the cannabinergic system and its physiological functions.


Subject(s)
Receptors, Cannabinoid/drug effects , Animals , Cannabinoid Receptor Agonists , Cannabinoid Receptor Antagonists , Humans , Ligands
18.
Synapse ; 60(2): 93-101, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16715483

ABSTRACT

G-protein coupled receptors exist in both high and low agonist affinity conformations, with tracer levels of agonist radioligands preferentially binding to the former. The goal of the present study was to characterize the in vivo binding of the aminoalkyindole-based, CB1 receptor agonist, R-[125/131I]AM2233 ((2-[125/131I]iodo-phenyl)-[1-(1-methyl-piperidin-2-yl-methyl)-1H-indol-3-yl]-methanone), and to use this radiotracer to selectively measure the receptor occupancy by the related CB1 receptor agonist, WIN55212-2, to the agonist-preferring affinity state of the receptor. In mouse locomotor assays, both WIN55212-2 and AM2233 (racemic) produced an approximately 60% reduction in activity at 1 mg/kg, (i.v.) and completely inhibited activity at 3 mg/kg, confirming their agonist nature. In ex vivo autoradiography, preferential uptake of R-[131I]AM2233 was apparent in CB1 receptor-rich areas, including globus pallidus, substantia nigra, striatum, cerebellum, and hippocampus. Overall brain uptake of R-[131I]AM2233 was 1.3% injected activity/g at 5 min in mice. Coinjection of 3 mg/kg (i.v.) SR141716A, a CB1 receptor antagonist, with R-[125I]AM2233 inhibited the radiotracer binding almost to nonspecific levels in the striatum, globus pallidus, and substantia nigra, although residual binding to a non-CB1 receptor remained in the hippocampus. In contrast to the effect of SR141716A, coinjection of 10 mg/kg (i.v.) WIN55212-2, a high dose that produced an immediate and profound immobility and catalepsy in the mice, reduced CB1 receptor-specific binding of R-[125I]AM2233 in CB1 receptor-rich areas by only 21-43%. These observations suggest that the behavioral effects of CB1 receptor agonists are manifested with a relatively small fraction of the agonist-preferring affinity state of the receptor occupied.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Cannabinoids/pharmacology , Neurons/drug effects , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Animals , Behavior, Animal/physiology , Benzoxazines , Binding Sites/drug effects , Binding Sites/physiology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Brain/anatomy & histology , Brain/metabolism , Cannabinoids/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Indoles/metabolism , Indoles/pharmacology , Iodine Radioisotopes/metabolism , Male , Mice , Morpholines/metabolism , Morpholines/pharmacology , Motor Activity/drug effects , Motor Activity/physiology , Naphthalenes/metabolism , Naphthalenes/pharmacology , Neurons/metabolism , Piperidines/metabolism , Piperidines/pharmacology , Protein Conformation/drug effects , Pyrazoles/pharmacology , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/metabolism , Rimonabant
19.
Planta Med ; 71(8): 701-5, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16142631

ABSTRACT

Alkamides are the major lipophilic constituents of Echinacea angustifolia roots. Due to their structural similarity with anandamide, we have evaluated their ability to bind to rodent cannabinoid receptors CB1 and CB2 by a standard receptor binding assay using [(3)H]CP-55,940 as a radioligand. The alkamides exhibited selective affinity especially to CB2 receptors and can therefore be considered as CB ligands. Most of the alkamides showed good metabolic stability as indicated by the similarity between affinity to CB1 determined in the presence/absence of the protease inhibitor PMSF. It is suggested that CB2 interactions may be the molecular mode of action of Echinacea alkamides as immunomodulators.


Subject(s)
Cannabinoid Receptor Modulators/pharmacology , Echinacea , Endocannabinoids , Phytotherapy , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB2/drug effects , Animals , Brain/metabolism , Cannabinoid Receptor Modulators/administration & dosage , Cannabinoid Receptor Modulators/therapeutic use , Mice , Plant Roots , Rats , Spleen/metabolism , Structure-Activity Relationship
20.
J Pharmacol Exp Ther ; 308(2): 446-53, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14610224

ABSTRACT

The present studies were conducted to test the hypothesis that activation of peripheral cannabinoid CB(2) receptors would suppress hyperalgesia evoked by intradermal administration of capsaicin, the pungent ingredient in hot chili peppers. The CB(2)-selective cannabinoid agonist (2-iodo-5-nitro-phenyl)-[1-(1-methyl-piperidin-2-ylmethyl)-1H-indol-3-yl]-methanone (AM1241) (33, 330 microg/kg i.p.) suppressed the development of capsaicin-evoked thermal and mechanical hyperalgesia and allodynia. AM1241 also produced a dose-dependent suppression of capsaicin-evoked nocifensive behavior. The AM1241-induced suppression of each parameter of capsaicin-evoked pain behavior was completely blocked by the CB(2) antagonist N-[(1S)-endo-1,3,3-trimethyl bicycle [2.2.1] heptan-2-yl]-5-(4-chloro-3-methylphenyl)-1-(4-methylbenzyl)-pyrazole-3-carboxamide (SR144528) but not by the CB(1) antagonist N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamidehydrochloride (SR141716A). AM1241 (33 microg/kg i.pl.) suppressed capsaicin-evoked thermal and mechanical hyperalgesia and allodynia after local administration to the capsaicin-treated (ipsilateral) paw but was inactive after administration to the capsaicin-untreated (contralateral) paw. Our data indicate that AM1241 suppresses capsaicin-evoked hyperalgesia and allodynia through a local site of action. These data provide evidence that actions at cannabinoid CB(2) receptors are sufficient to normalize nociceptive thresholds and produce antinociception in persistent pain states.


Subject(s)
Analgesics/therapeutic use , Capsaicin/therapeutic use , Hyperalgesia/drug therapy , Pain/drug therapy , Receptor, Cannabinoid, CB2/metabolism , Animals , Cannabinoids/metabolism , Disease Models, Animal , Injections, Intradermal , Male , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB2/agonists
SELECTION OF CITATIONS
SEARCH DETAIL