Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 170(3): 679-92, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23902406

ABSTRACT

BACKGROUND AND PURPOSE: Epilepsy is the most prevalent neurological disease and is characterized by recurrent seizures. Here, we investigate (i) the anticonvulsant profiles of cannabis-derived botanical drug substances (BDSs) rich in cannabidivarin (CBDV) and containing cannabidiol (CBD) in acute in vivo seizure models and (ii) the binding of CBDV BDSs and their components at cannabinoid CB1 receptors. EXPERIMENTAL APPROACH: The anticonvulsant profiles of two CBDV BDSs (50-422 mg·kg(-1) ) were evaluated in three animal models of acute seizure. Purified CBDV and CBD were also evaluated in an isobolographic study to evaluate potential pharmacological interactions. CBDV BDS effects on motor function were also investigated using static beam and grip strength assays. Binding of CBDV BDSs to cannabinoid CB1 receptors was evaluated using displacement binding assays. KEY RESULTS: CBDV BDSs exerted significant anticonvulsant effects in the pentylenetetrazole (≥100 mg·kg(-1) ) and audiogenic seizure models (≥87 mg·kg(-1) ), and suppressed pilocarpine-induced convulsions (≥100 mg·kg(-1) ). The isobolographic study revealed that the anticonvulsant effects of purified CBDV and CBD were linearly additive when co-administered. Some motor effects of CBDV BDSs were observed on static beam performance; no effects on grip strength were found. The Δ(9) -tetrahydrocannabinol and Δ(9) -tetrahydrocannabivarin content of CBDV BDS accounted for its greater affinity for CB1 cannabinoid receptors than purified CBDV. CONCLUSIONS AND IMPLICATIONS: CBDV BDSs exerted significant anticonvulsant effects in three models of seizure that were not mediated by the CB1 cannabinoid receptor and were of comparable efficacy with purified CBDV. These findings strongly support the further clinical development of CBDV BDSs for the treatment of epilepsy.


Subject(s)
Anticonvulsants/pharmacology , Brain/drug effects , Cannabinoids/pharmacology , Cannabis , Plant Extracts/pharmacology , Seizures/prevention & control , Animals , Anticonvulsants/metabolism , Brain/metabolism , Brain/physiopathology , Cannabidiol/pharmacology , Cannabinoids/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Hand Strength , Male , Mice , Mice, Inbred DBA , Motor Activity/drug effects , Noise/adverse effects , Pentylenetetrazole , Phytotherapy , Pilocarpine , Plant Extracts/metabolism , Plants, Medicinal , Protein Binding , Rats , Rats, Inbred WKY , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/metabolism , Seizures/etiology , Seizures/metabolism , Seizures/physiopathology
2.
Br J Pharmacol ; 168(6): 1456-70, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23121618

ABSTRACT

BACKGROUND AND PURPOSE: To evaluate the ability of cannabidiolic acid (CBDA) to reduce nausea and vomiting and enhance 5-HT(1A) receptor activation in animal models. EXPERIMENTAL APPROACH: We investigated the effect of CBDA on (i) lithium chloride (LiCl)-induced conditioned gaping to a flavour (nausea-induced behaviour) or a context (model of anticipatory nausea) in rats; (ii) saccharin palatability in rats; (iii) motion-, LiCl- or cisplatin-induced vomiting in house musk shrews (Suncus murinus); and (iv) rat brainstem 5-HT(1A) receptor activation by 8-hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT) and mouse whole brain CB(1) receptor activation by CP55940, using [³5S]GTPγS-binding assays. KEY RESULTS: In shrews, CBDA (0.1 and/or 0.5 mg·kg⁻¹ i.p.) reduced toxin- and motion-induced vomiting, and increased the onset latency of the first motion-induced emetic episode. In rats, CBDA (0.01 and 0.1 mg·kg⁻¹ i.p.) suppressed LiCl- and context-induced conditioned gaping, effects that were blocked by the 5-HT(1A) receptor antagonist, WAY100635 (0.1 mg·kg⁻¹ i.p.), and, at 0.01 mg·kg⁻¹ i.p., enhanced saccharin palatability. CBDA-induced suppression of LiCl-induced conditioned gaping was unaffected by the CB1 receptor antagonist, SR141716A (1 mg·kg⁻¹ i.p.). In vitro, CBDA (0.1-100 nM) increased the E(max) of 8-OH-DPAT. CONCLUSIONS AND IMPLICATIONS: Compared with cannabidiol, CBDA displays significantly greater potency at inhibiting vomiting in shrews and nausea in rats, and at enhancing 5-HT(1A) receptor activation, an action that accounts for its ability to attenuate conditioned gaping in rats. Consequently, CBDA shows promise as a treatment for nausea and vomiting, including anticipatory nausea for which no specific therapy is currently available.


Subject(s)
Antiemetics/therapeutic use , Brain/drug effects , Cannabinoids/therapeutic use , Nausea/prevention & control , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin 5-HT1 Receptor Agonists/therapeutic use , Vomiting/prevention & control , Animals , Antiemetics/antagonists & inhibitors , Behavior, Animal/drug effects , Brain/metabolism , Brain Stem/drug effects , Brain Stem/metabolism , Cannabinoid Receptor Antagonists/pharmacology , Cannabinoids/antagonists & inhibitors , Female , Male , Mice , Motion Sickness/physiopathology , Motion Sickness/prevention & control , Nausea/chemically induced , Nausea/etiology , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/metabolism , Receptor, Serotonin, 5-HT1A/chemistry , Serotonin 5-HT1 Receptor Agonists/chemistry , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Shrews , Vomiting/chemically induced , Vomiting/etiology
3.
Br J Pharmacol ; 165(8): 2620-34, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21827451

ABSTRACT

BACKGROUND AND PURPOSE: To evaluate the hypothesis that activation of somatodendritic 5-HT(1A) autoreceptors in the dorsal raphe nucleus (DRN) produces the anti-emetic/anti-nausea effects of cannabidiol (CBD), a primary non-psychoactive cannabinoid found in cannabis. EXPERIMENTAL APPROACH: The potential of systemic and intra-DRN administration of 5-HT(1A) receptor antagonists, WAY100135 or WAY100635, to prevent the anti-emetic effect of CBD in shrews (Suncus murinus) and the anti-nausea-like effects of CBD (conditioned gaping) in rats were evaluated. Also, the ability of intra-DRN administration of CBD to produce anti-nausea-like effects (and reversal by systemic WAY100635) was assessed. In vitro studies evaluated the potential of CBD to directly target 5-HT(1A) receptors and to modify the ability of the 5-HT(1A) agonist, 8-OH-DPAT, to stimulate [(35) S]GTPγS binding in rat brainstem membranes. KEY RESULTS: CBD suppressed nicotine-, lithium chloride (LiCl)- and cisplatin (20 mg·kg(-1) , but not 40 mg·kg(-1) )-induced vomiting in the S. murinus and LiCl-induced conditioned gaping in rats. Anti-emetic and anti-nausea-like effects of CBD were suppressed by WAY100135 and the latter by WAY100635. When administered to the DRN: (i) WAY100635 reversed anti-nausea-like effects of systemic CBD, and (ii) CBD suppressed nausea-like effects, an effect that was reversed by systemic WAY100635. CBD also displayed significant potency (in a bell-shaped dose-response curve) at enhancing the ability of 8-OH-DPAT to stimulate [(35) S]GTPγS binding to rat brainstem membranes in vitro. Systemically administered CBD and 8-OH-DPAT synergistically suppressed LiCl-induced conditioned gaping. CONCLUSIONS AND IMPLICATIONS: These results suggest that CBD produced its anti-emetic/anti-nausea effects by indirect activation of the somatodendritic 5-HT(1A) autoreceptors in the DRN. 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.


Subject(s)
Antiemetics/therapeutic use , Cannabidiol/therapeutic use , Raphe Nuclei/physiology , Receptor, Serotonin, 5-HT1A/physiology , Serotonin 5-HT1 Receptor Agonists/pharmacology , Vomiting/drug therapy , 8-Hydroxy-2-(di-n-propylamino)tetralin/metabolism , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Behavior, Animal/drug effects , Cannabis , Female , Male , Nausea/drug therapy , Nausea/physiopathology , Piperazines/pharmacology , Pyridines/pharmacology , Raphe Nuclei/drug effects , Rats , Rats, Sprague-Dawley , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Shrews , Vomiting/physiopathology
4.
Article in English | MEDLINE | ID: mdl-21995886

ABSTRACT

Omega-3 (n-3) fatty acids inhibit breast and prostate cancer cell growth. We previously showed that N-acylethanolamine derivatives of n-3 (n-3-NAE) are endocannabinoids, which regulate cancer cell proliferation. These n-3-NAE are synthesised in certain cells/tissues, after supplementing with fatty acids, however, no one has assessed whether and to what extent this occurs in cancer cells. We determined levels of endogenous n-3-NAEs in hormone sensitive and insensitive prostate and breast cancer cells and subsequent effects on other endocannabinoids (anandamide and 2-arachidonoylglycerol), before and after supplementing with DHA and EPA fatty acids, using HPLC tandem mass spectrometry. This is the first study reporting that n-3-NAEs are synthesised from their parent n-3 fatty acids in cancer cells, regardless of tumour type, hormone status or the presence of fatty acid amide hydrolase. This could have important implications for the use of n-3 fatty acids as therapeutic agents in breast and prostate cancers expressing cannabinoid receptors.


Subject(s)
Breast Neoplasms/metabolism , Cannabinoid Receptor Modulators/biosynthesis , Ethanolamines/metabolism , Fatty Acids, Omega-3/metabolism , Prostatic Neoplasms/metabolism , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Cannabinoid Receptor Modulators/chemistry , Cell Line, Tumor , Female , Humans , Male , Tandem Mass Spectrometry
5.
Pharmacol Rev ; 62(4): 588-631, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21079038

ABSTRACT

There are at least two types of cannabinoid receptors (CB(1) and CB(2)). Ligands activating these G protein-coupled receptors (GPCRs) include the phytocannabinoid Δ(9)-tetrahydrocannabinol, numerous synthetic compounds, and endogenous compounds known as endocannabinoids. Cannabinoid receptor antagonists have also been developed. Some of these ligands activate or block one type of cannabinoid receptor more potently than the other type. This review summarizes current data indicating the extent to which cannabinoid receptor ligands undergo orthosteric or allosteric interactions with non-CB(1), non-CB(2) established GPCRs, deorphanized receptors such as GPR55, ligand-gated ion channels, transient receptor potential (TRP) channels, and other ion channels or peroxisome proliferator-activated nuclear receptors. From these data, it is clear that some ligands that interact similarly with CB(1) and/or CB(2) receptors are likely to display significantly different pharmacological profiles. The review also lists some criteria that any novel "CB(3)" cannabinoid receptor or channel should fulfil and concludes that these criteria are not currently met by any non-CB(1), non-CB(2) pharmacological receptor or channel. However, it does identify certain pharmacological targets that should be investigated further as potential CB(3) receptors or channels. These include TRP vanilloid 1, which possibly functions as an ionotropic cannabinoid receptor under physiological and/or pathological conditions, and some deorphanized GPCRs. Also discussed are 1) the ability of CB(1) receptors to form heteromeric complexes with certain other GPCRs, 2) phylogenetic relationships that exist between CB(1)/CB(2) receptors and other GPCRs, 3) evidence for the existence of several as-yet-uncharacterized non-CB(1), non-CB(2) cannabinoid receptors; and 4) current cannabinoid receptor nomenclature.


Subject(s)
Receptors, Cannabinoid/metabolism , Cannabinoid Receptor Agonists , Cannabinoid Receptor Antagonists , Cannabinoid Receptor Modulators/metabolism , Cannabinoids/metabolism , Humans , Ligands , Phylogeny , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptor, Cannabinoid, CB2/metabolism , Terminology as Topic
6.
Br J Pharmacol ; 160(3): 549-60, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20590565

ABSTRACT

BACKGROUND AND PURPOSE: Inhibitors of fatty acid amide hydrolase (FAAH), the enzyme responsible for the metabolism of the endogenous cannabinoid (CB) receptor ligand anandamide (AEA), are effective in a number of animal models of pain. Here, we investigated a series of isoflavones with respect to their abilities to inhibit FAAH. EXPERIMENTAL APPROACH: In vitro assays of FAAH activity and affinity for CB receptors were used to characterize key compounds. In vivo assays used were biochemical responses to formalin in anaesthetized mice and the 'tetrad' test for central CB receptor activation. KEY RESULTS: Of the compounds tested, biochanin A was adjudged to be the most promising. Biochanin A inhibited the hydrolysis of 0.5 microM AEA by mouse, rat and human FAAH with IC(50) values of 1.8, 1.4 and 2.4 microM respectively. The compound did not interact to any major extent with CB(1) or CB(2) receptors, nor with FAAH-2. In anaesthetized mice, URB597 (30 microg i.pl.) and biochanin A (100 microg i.pl.) both inhibited the spinal phosphorylation of extracellular signal-regulated kinase produced by the intraplantar injection of formalin. The effects of both compounds were significantly reduced by the CB(1) receptor antagonist/inverse agonist AM251 (30 microg i.pl.). Biochanin A (15 mg.kg(-1) i.v.) did not increase brain AEA concentrations, but produced a modest potentiation of the effects of 10 mg.kg(-1) i.v. AEA in the tetrad test. CONCLUSIONS AND IMPLICATIONS: It is concluded that biochanin A, in addition to its other biochemical properties, inhibits FAAH both in vitro and peripherally in vivo.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Genistein/pharmacology , Isoflavones/pharmacology , Animals , Arachidonic Acids/metabolism , Behavior, Animal/drug effects , Benzamides/antagonists & inhibitors , Benzamides/pharmacology , Brain/drug effects , Brain/enzymology , Brain/metabolism , COS Cells , Cannabinoid Receptor Antagonists , Carbamates/antagonists & inhibitors , Carbamates/pharmacology , Cell Line, Transformed , Chlorocebus aethiops , Drug Interactions , Endocannabinoids , Extracellular Signal-Regulated MAP Kinases/metabolism , Formaldehyde/antagonists & inhibitors , Genistein/antagonists & inhibitors , Humans , Liver/enzymology , Mice , Mice, Inbred ICR , Piperidines/pharmacology , Polyunsaturated Alkamides/metabolism , Pyrazoles/pharmacology , Rats
7.
Br J Pharmacol ; 160(3): 712-23, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20590574

ABSTRACT

BACKGROUND AND PURPOSE: Cannabis is taken as self-medication by patients with inflammatory bowel disease for symptomatic relief. Cannabinoid receptor agonists decrease inflammation in animal models of colitis, but their effects on the disturbed motility is not known. (-)-Cannabidiol (CBD) has been shown to interact with Delta(9)-tetrahydrocannabinol (THC) in behavioural studies, but it remains to be established if these cannabinoids interact in vivo in inflammatory disorders. Therefore the effects of CBD and THC alone and in combination were investigated in a model of colitis. EXPERIMENTAL APPROACH: The 2,4,6-trinitrobenzene sulphonic acid (TNBS) model of acute colitis in rats was used to assess damage, inflammation (myeloperoxidase activity) and in vitro colonic motility. Sulphasalazine was used as an active control drug. KEY RESULTS: Sulphasalazine, THC and CBD proved beneficial in this model of colitis with the dose-response relationship for the phytocannabinoids showing a bell-shaped pattern on the majority of parameters (optimal THC and CBD dose, 10 mg.kg(-1)). THC was the most effective drug. The effects of these phytocannabinoids were additive, and CBD increased some effects of an ineffective THC dose to the level of an effective one. THC alone and in combination with CBD protected cholinergic nerves whereas sulphasalazine did not. CONCLUSIONS AND IMPLICATIONS: In this model of colitis, THC and CBD not only reduced inflammation but also lowered the occurrence of functional disturbances. Moreover the combination of CBD and THC could be beneficial therapeutically, via additive or potentiating effects.


Subject(s)
Cannabidiol/administration & dosage , Colitis/drug therapy , Colon/drug effects , Dronabinol/administration & dosage , Inflammation/drug therapy , Animals , Cannabidiol/pharmacology , Colitis/metabolism , Colitis/pathology , Colitis/physiopathology , Colon/pathology , Colon/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Dronabinol/pharmacology , Drug Therapy, Combination , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , In Vitro Techniques , Inflammation/pathology , Inflammation/physiopathology , Male , Peroxidase/metabolism , Rats , Rats, Wistar , Sulfasalazine/therapeutic use , Trinitrobenzenesulfonic Acid
8.
Curr Med Chem ; 17(14): 1360-81, 2010.
Article in English | MEDLINE | ID: mdl-20166927

ABSTRACT

It is widely accepted that non-endogenous compounds that target CB(1) and/or CB(2) receptors possess therapeutic potential for the clinical management of an ever growing number of disorders. Just a few of these disorders are already treated with Delta(9)-tetrahydrocannabinol or nabilone, both CB(1)/CB(2) receptor agonists, and there is now considerable interest in expanding the clinical applications of such agonists and also in exploiting CB(2)-selective agonists, peripherally restricted CB(1)/CB(2) receptor agonists and CB(1)/CB(2) antagonists and inverse agonists as medicines. Already, numerous cannabinoid receptor ligands have been developed and their interactions with CB(1) and CB(2) receptors well characterized. This review describes what is currently known about the ability of such compounds to bind to, activate, inhibit or block non-CB(1), non- CB(2) G protein-coupled receptors such as GPR55, transmitter gated channels, ion channels and nuclear receptors in an orthosteric or allosteric manner. It begins with a brief description of how each of these ligands interacts with CB(1) and/or CB(2) receptors.


Subject(s)
Cannabinoid Receptor Agonists , Cannabinoid Receptor Antagonists , Dronabinol/analogs & derivatives , Dronabinol/pharmacology , Ion Channels/metabolism , Ligands , Piperidines/chemistry , Piperidines/pharmacology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Receptors, Cannabinoid/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism , Rimonabant
9.
Br J Pharmacol ; 159(1): 129-41, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20002104

ABSTRACT

BACKGROUND AND PURPOSE: Cannabis is the source of at least seventy phytocannabinoids. The pharmacology of most of these has been little investigated, three notable exceptions being Delta(9)-tetrahydrocannabinol, cannabidiol and Delta(9)-tetrahydrocannabivarin. This investigation addressed the question of whether the little-studied phytocannabinoid, cannabigerol, can activate or block any G protein-coupled receptor. EXPERIMENTAL APPROACH: The [(35)S]GTPgammaS binding assay, performed with mouse brain membranes, was used to test the ability of cannabigerol to produce G protein-coupled receptor activation or blockade. Its ability to displace [(3)H]CP55940 from mouse CB(1) and human CB(2) cannabinoid receptors and to inhibit electrically evoked contractions of the mouse isolated vas deferens was also investigated. KEY RESULTS: In the brain membrane experiments, cannabigerol behaved as a potent alpha(2)-adrenoceptor agonist (EC(50)= 0.2 nM) and antagonized the 5-HT(1A) receptor agonist, R-(+)-8-hydroxy-2-(di-n-propylamino)tetralin (apparent K(B)= 51.9 nM). At 10 microM, it also behaved as a CB(1) receptor competitive antagonist. Additionally, cannabigerol inhibited evoked contractions of the vas deferens in a manner that appeared to be alpha(2)-adrenoceptor-mediated (EC(50)= 72.8 nM) and displayed significant affinity for mouse CB(1) and human CB(2) receptors. CONCLUSIONS AND IMPLICATIONS: This investigation has provided the first evidence that cannabigerol can activate alpha(2)-adrenoceptors, bind to cannabinoid CB(1) and CB(2) receptors and block CB(1) and 5-HT(1A) receptors. It will now be important to investigate why cannabigerol produced signs of agonism more potently in the [(35)S]GTPgammaS binding assay than in the vas deferens and also whether it can inhibit noradrenaline uptake in this isolated tissue and in the brain.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Cannabinoids/pharmacology , Serotonin 5-HT1 Receptor Antagonists , Serotonin Antagonists/pharmacology , Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-Agonists/administration & dosage , Animals , CHO Cells , Cannabinoids/administration & dosage , Cannabis/chemistry , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction/drug effects , Protein Binding , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/drug effects , Receptor, Cannabinoid, CB2/metabolism , Serotonin Antagonists/administration & dosage , Vas Deferens/drug effects , Vas Deferens/metabolism
10.
Br J Pharmacol ; 153(2): 199-215, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17828291

ABSTRACT

Cannabis sativa is the source of a unique set of compounds known collectively as plant cannabinoids or phytocannabinoids. This review focuses on the manner with which three of these compounds, (-)-trans-delta9-tetrahydrocannabinol (delta9-THC), (-)-cannabidiol (CBD) and (-)-trans-delta9-tetrahydrocannabivarin (delta9-THCV), interact with cannabinoid CB1 and CB2 receptors. Delta9-THC, the main psychotropic constituent of cannabis, is a CB1 and CB2 receptor partial agonist and in line with classical pharmacology, the responses it elicits appear to be strongly influenced both by the expression level and signalling efficiency of cannabinoid receptors and by ongoing endogenous cannabinoid release. CBD displays unexpectedly high potency as an antagonist of CB1/CB2 receptor agonists in CB1- and CB2-expressing cells or tissues, the manner with which it interacts with CB2 receptors providing a possible explanation for its ability to inhibit evoked immune cell migration. Delta9-THCV behaves as a potent CB2 receptor partial agonist in vitro. In contrast, it antagonizes cannabinoid receptor agonists in CB1-expressing tissues. This it does with relatively high potency and in a manner that is both tissue and ligand dependent. Delta9-THCV also interacts with CB1 receptors when administered in vivo, behaving either as a CB1 antagonist or, at higher doses, as a CB1 receptor agonist. Brief mention is also made in this review, first of the production by delta9-THC of pharmacodynamic tolerance, second of current knowledge about the extent to which delta9-THC, CBD and delta9-THCV interact with pharmacological targets other than CB1 or CB2 receptors, and third of actual and potential therapeutic applications for each of these cannabinoids.


Subject(s)
Cannabidiol/pharmacology , Dronabinol/analogs & derivatives , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB2/drug effects , Animals , Dronabinol/pharmacology , Drug Tolerance , Humans , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Synaptic Transmission/drug effects
11.
Br J Pharmacol ; 152(7): 984-6, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17876300

ABSTRACT

In this issue of the British Journal of Pharmacology, Ryberg et al. present convincing in vitro evidence that the orphan GPCR, GPR55, is a cannabinoid receptor. GPR55 was activated by a range of plant, synthetic and endogenous cannabinoids and blocked by the non-psychoactive phytocannabinoid, cannabidiol. Their experiments have revealed several differences between the pharmacology of GPR55 and the established cannabinoid CB1 and CB2 receptors. For example, the CB1 receptor antagonist, AM251, activated GPR55 and the main psychoactive constituent of cannabis, Delta9-tetrahydrocannabinol, displayed greater efficacy at GPR55 than at CB1 or CB2 receptors. They also compared the distribution of GPR55 and CB1 mRNA in mouse and report that GPR55 couples to Galpha13, that it is activated by virodhamine, palmitoylethanolamide and oleoylethanolamide, and that virodhamine displays relatively high efficacy as a GPR55 agonist. Still to be identified are the main roles played by GPR55 in health and disease and any potential therapeutic benefits of activating or blocking this receptor.


Subject(s)
Receptors, G-Protein-Coupled/physiology , Amides , Animals , Arachidonic Acids/pharmacology , Cannabinoids , Dronabinol/pharmacology , Endocannabinoids , Ethanolamines , Humans , Oleic Acids/pharmacology , Palmitic Acids/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptors, Cannabinoid , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/drug effects
12.
Br J Pharmacol ; 152(5): 583-93, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17641667

ABSTRACT

A meta-analysis, unlike a literature review, synthesizes previous studies into new results. Pooled data from 211 studies measured ligand binding affinities at human (Hs) or rat (Rn) cannabinoid receptors CB1 and CB2. Cochrane methods were modified for this non-clinical analysis. Meta-regression detected data heterogeneity arising from methodological factors: use of sectioned tissues, lack of PMSF and choice of radioligand. Native brain tissues exhibited greater affinity (lower nM) than transfected cells, but the trend fell short of significance, as did the trend between centrifugation and filtration methods. Correcting for heterogeneity, mean Ki values for delta 9-tetrahydrocannabinol differed significantly between HsCB1 and RnCB1 (25.1 and 42.6 nM, respectively) but not between HsCB1 and HsCB2 (25.1 and 35.2). Mean Kd values for HsCB1, RnCB1 and HsCB2 of CP55,940 (2.5, 0.98, 0.92) and WIN55,212-2 (16.7, 2.4, 3.7) differed between HsCB1 and RnCB1 and between HsCB1 and HsCB2. SR141716A differed between HsCB1 and RnCB1 (2.9 and 1.0 nM). Anandamide at HsCB1, RnCB1 and HsCB2 (239.2, 87.7, 439.5) fell short of statistical differences due to heterogeneity. We consider these Kd and Ki values to be the most valid estimates in the literature. Sensitivity analyses did not support the numerical validity of cannabidiol, cannabinol, 2-arachidonoyl glycerol and all ligands at RnCB2. Aggregate rank order analysis of CB(1) distribution in the brain (pooled from 119 autoradiographic, immunohistochemical and in situ hybridization studies) showed denser HsCB1 expression in cognitive regions (cerebral cortex) compared to RnCB1, which was relatively richer in movement-associated areas (cerebellum, caudate-putamen). Implications of interspecies differences are discussed.


Subject(s)
Cannabinoids/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Animals , Binding, Competitive , Cannabinoids/metabolism , Humans , Ligands , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Species Specificity
13.
Br J Pharmacol ; 151(5): 688-700, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17502849

ABSTRACT

BACKGROUND AND PURPOSE: Previous work implied that the hippocampal cannabinoid system was particularly important in some forms of learning, but direct evidence for this hypothesis is scarce. We therefore assessed the effects of the synthetic cannabinoid HU210 on memory and hippocampal activity. EXPERIMENTAL APPROACH: HU210 (100 microg kg(-1)) was administered intraperitoneally to rats under three experimental conditions. One group of animals were pre-trained in spatial working memory using a delayed-matching-to-position task and effects of HU210 were assessed in a within-subject design. In another, rats were injected before acquisition learning of a spatial reference memory task with constant platform location. Finally, a separate group of animals was implanted with electrode bundles in CA1 and CA3 and single unit responses were isolated, before and after HU210 treatment. KEY RESULTS: HU210 treatment had no effect on working or short-term memory. Relative to its control Tween 80, deficits in acquisition of a reference memory version of the water maze were obtained, along with drug-related effects on anxiety, motor activity and spatial learning. Deficits were not reversed by the CB(1) receptor antagonists SR141716A (3 mg kg(-1)) or AM281 (1.5 mg kg(-1)). Single unit recordings from principal neurons in hippocampal CA3 and CA1 confirmed HU210-induced attenuation of the overall firing activity lowering both the number of complex spikes fired and the occurrence of bursts. CONCLUSIONS AND IMPLICATIONS: These data provide the first direct evidence that the underlying mechanism for the spatial memory deficits induced by HU210 in rats is the accompanying abnormality in hippocampal cell firing.


Subject(s)
Cannabinoids/toxicity , Dronabinol/analogs & derivatives , Hippocampus/drug effects , Memory Disorders/chemically induced , Space Perception/drug effects , Animals , Behavior, Animal/drug effects , Dronabinol/toxicity , Electrophysiology , Hippocampus/cytology , Hippocampus/physiopathology , Male , Maze Learning/drug effects , Memory/drug effects , Memory Disorders/physiopathology , Memory Disorders/psychology , Morpholines/pharmacology , Motor Activity/drug effects , Neurons/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Receptor, Cannabinoid, CB1/drug effects , Rimonabant
14.
Izv Akad Nauk Ser Biol ; (1): 35-44, 2007.
Article in Russian | MEDLINE | ID: mdl-17352198

ABSTRACT

Intravenous administration of cannabinoid (CB) receptor agonists (HU-210, 0.1 mg/kg; ACPA, 0.125 mg/kg; methanandamide, 2.5 mg/kg; and anandamide, 2.5 mg/kg) induced bradycardia in chloralose-anesthetized rats irrespective of the solubilization method. Methanandamide, HU-210, and ACPA had no effect on the electrophysiological activity in the heart, while anandamide increased the duration of the QRS complex. The negative chronotropic effect of HU-210 was due to CB1 receptor activation since is was not observed after CB1 receptor blockade by SR141716A (1 mg/kg intravenously) but was present after pretreatment with CB2 receptor antagonist SR144528 (1 mg/kg intravenously). CB receptor antagonists SR141716A and SR144528 had no effect on cardiac rhythm or ECG indices. Hence, in the intact heart, endogenous CB receptor agonists are not involved in the regulation of cardiac rhythm and electrophysiological processes. The chronotropic effect of CBs was independent of the autonomic nervous system because it remained significant after autonomic ganglion blockade by hexamethonium (1 mg/kg intravenously). CB receptor activation by HU-210 (0.1 and 1 microM) in vitro decreased the rate and force of isolated heart contractions, the rates of contraction and relaxation, and end diastolic pressure. The negative chronotropic effect of HU-210 was less pronounced in vitro than in vivo. The maximum inotropic effect of HU-210 was reached at the concentration of 0.1 microM.


Subject(s)
Heart Rate , Heart/physiology , Myocardial Contraction , Receptors, Cannabinoid/physiology , Animals , Cannabinoid Receptor Agonists , Cannabinoid Receptor Antagonists , Cannabinoids/antagonists & inhibitors , Cannabinoids/pharmacology , Electrophysiology , Heart Rate/drug effects , Male , Myocardial Contraction/drug effects , Rats , Rats, Wistar
15.
Br J Pharmacol ; 150(5): 613-23, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17245363

ABSTRACT

BACKGROUND AND PURPOSE: A nonpsychoactive constituent of the cannabis plant, cannabidiol has been demonstrated to have low affinity for both cannabinoid CB1 and CB2 receptors. We have shown previously that cannabidiol can enhance electrically evoked contractions of the mouse vas deferens, suggestive of inverse agonism. We have also shown that cannabidiol can antagonize cannabinoid receptor agonists in this tissue with a greater potency than we would expect from its poor affinity for cannabinoid receptors. This study aimed to investigate whether these properties of cannabidiol extend to CB1 receptors expressed in mouse brain and to human CB2 receptors that have been transfected into CHO cells. EXPERIMENTAL APPROACH: The [35S]GTPS binding assay was used to determine both the efficacy of cannabidiol and the ability of cannabidiol to antagonize cannabinoid receptor agonists (CP55940 and R-(+)-WIN55212) at the mouse CB1 and the human CB2 receptor. KEY RESULTS: This paper reports firstly that cannabidiol displays inverse agonism at the human CB2 receptor. Secondly, we demonstrate that cannabidiol is a high potency antagonist of cannabinoid receptor agonists in mouse brain and in membranes from CHO cells transfected with human CB2 receptors. CONCLUSIONS AND IMPLICATIONS: This study has provided the first evidence that cannabidiol can display CB2 receptor inverse agonism, an action that appears to be responsible for its antagonism of CP55940 at the human CB2 receptor. The ability of cannabidiol to behave as a CB2 receptor inverse agonist may contribute to its documented anti-inflammatory properties.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Benzoxazines/antagonists & inhibitors , Brain/drug effects , Cannabidiol/pharmacology , Cannabinoid Receptor Agonists , Cyclohexanes/antagonists & inhibitors , Morpholines/antagonists & inhibitors , Naphthalenes/antagonists & inhibitors , Phenols/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/metabolism , Benzoxazines/pharmacology , Brain/metabolism , CHO Cells , Camphanes/pharmacology , Cannabidiol/metabolism , Cannabinoid Receptor Antagonists , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetinae , Cricetulus , Cyclohexanes/pharmacology , Cyclohexanols , Dose-Response Relationship, Drug , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Morpholines/pharmacology , Naphthalenes/pharmacology , Phenols/pharmacology , Piperidines/pharmacology , Protein Binding , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Receptors, Cannabinoid/genetics , Receptors, Cannabinoid/metabolism , Rimonabant , Transfection
16.
Br J Pharmacol ; 150(5): 586-94, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17245367

ABSTRACT

BACKGROUND AND PURPOSE: To follow up in vitro evidence that Delta(9)-tetrahydrocannabivarin extracted from cannabis (eDelta(9)-THCV) is a CB(1) receptor antagonist by establishing whether synthetic Delta(9)-tetrahydrocannabivarin (O-4394) and Delta(8)-tetrahydrocannabivarin (O-4395) behave as CB(1) antagonists in vivo. EXPERIMENTAL APPROACH: O-4394 and O-4395 were compared with eDelta(9)-THCV as displacers of [(3)H]-CP55940 from specific CB(1) binding sites on mouse brain membranes and as antagonists of CP55940 in [(35)S]GTPgammaS binding assays performed with mouse brain membranes and of R-(+)-WIN55212 in mouse isolated vasa deferentia. Their ability to antagonize in vivo effects of 3 or 10 mg kg(-1) (i.v.) Delta(9)-tetrahydrocannabinol in mice was then investigated. KEY RESULTS: O-4394 and O-4395 exhibited similar potencies to eDelta(9)-THCV as displacers of [(3)H]-CP55940 (K (i)=46.6 and 64.4 nM, respectively) and as antagonists of CP55940 in the [(35)S]GTPgammaS binding assay (apparent K (B)=82.1 and 125.9 nM, respectively) and R-(+)-WIN55212 in the vas deferens (apparent K (B)=4.8 and 3.9 nM respectively). At i.v. doses of 0.1, 0.3, 1.0 and/or 3 mg kg(-1) O-4394 and O-4395 attenuated Delta(9)-tetrahydrocannabinol-induced anti-nociception (tail-flick test) and hypothermia (rectal temperature). O-4395 but not O-4394 also antagonized Delta(9)-tetrahydrocannabinol-induced ring immobility. By themselves, O-4395 and O-4394 induced ring immobility at 3 or 10 mg kg(-1) (i.v.) and antinociception at doses above 10 mg kg(-1) (i.v.). O-4395 also induced hypothermia at 3 mg kg(-1) (i.v.) and above. CONCLUSIONS AND IMPLICATIONS: O-4394 and O-4395 exhibit similar in vitro potencies to eDelta(9)-THCV as CB(1) receptor ligands and as antagonists of cannabinoid receptor agonists and can antagonize Delta(9)-tetrahydrocannabinol in vivo.


Subject(s)
Brain/drug effects , Cannabinoid Receptor Antagonists , Dronabinol/analogs & derivatives , Dronabinol/antagonists & inhibitors , Psychotropic Drugs/antagonists & inhibitors , Vas Deferens/drug effects , Analgesics, Non-Narcotic/antagonists & inhibitors , Animals , Benzoxazines/pharmacology , Binding, Competitive , Body Temperature/drug effects , Brain/metabolism , Cannabinoid Receptor Agonists , Cyclohexanes/metabolism , Cyclohexanes/pharmacology , Cyclohexanols , Dose-Response Relationship, Drug , Dronabinol/metabolism , Dronabinol/pharmacology , Electric Stimulation , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , In Vitro Techniques , Locomotion/drug effects , Male , Mice , Mice, Inbred ICR , Morpholines/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Naphthalenes/pharmacology , Pain Measurement , Pain Threshold/drug effects , Phenols/metabolism , Phenols/pharmacology , Protein Binding , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptors, Cannabinoid/metabolism , Vas Deferens/metabolism
17.
Br J Pharmacol ; 149(4): 431-40, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16953186

ABSTRACT

BACKGROUND AND PURPOSE: We have previously reported the development of CB-25 and CB-52, two ligands of CB1 and CB2 cannabinoid receptors. We assessed here their functional activity. EXPERIMENTAL APPROACH: The effect of the two compounds on forskolin-induced cAMP formation in intact cells or GTP-gamma-S binding to cell membranes, and their action on nociception in vivo was determined. KEY RESULTS: CB-25 enhanced forskolin-induced cAMP formation in N18TG2 cells (EC50 approximately 20 nM, max. stimulation = 48%), behaving as an inverse CB1 agonist, but it stimulated GTP-gamma-S binding to mouse brain membranes, behaving as a partial CB1 agonist (EC50 =100 nM, max. stimulation = 48%). At human CB1 receptors, CB-25 inhibited cAMP formation in hCB1-CHO cells (EC50 = 1600 nM, max. inhibition = 68% of CP-55,940 effect). CB-52 inhibited forskolin-induced cAMP formation by N18TG2 cells (IC50 = 450 nM, max. inhibition = 40%) and hCB1-CHO cells (EC50 = 2600 nM, max. inhibition = 62% of CP-55,940 effect), and stimulated GTP-gamma-S binding to mouse brain membranes (EC50 = 11 nM, max. stimulation approximately 16%). Both CB-25 and CB-52 showed no activity in all assays of CB2-coupled functional activity and antagonized CP55940-induced stimulation of GTP-gamma-S binding to hCB2-CHO cell membranes. In vivo, both compounds, administered i.p., produced dose-dependent nociception in the plantar test carried out in healthy rats, and antagonised the anti-nociceptive effect of i.p. WIN55,212-2. In the formalin test in mice, however, the compounds counteracted both phases of formalin-induced nociception. CONCLUSIONS AND IMPLICATIONS: CB-25 and CB-52 behave in vitro mostly as CB1 partial agonists and CB2 neutral antagonists, whereas their activity in vivo might depend on the tonic activity of cannabinoid receptors.


Subject(s)
Amides/pharmacology , Analgesics/pharmacology , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB2/drug effects , Resorcinols/pharmacology , Action Potentials/drug effects , Adenylyl Cyclases/metabolism , Animals , Brain Stem/cytology , Brain Stem/drug effects , Brain Stem/metabolism , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Ligands , Male , Mice , Pain/chemically induced , Pain/metabolism , Pain/prevention & control , Pain Measurement , Pain Threshold/drug effects , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism , Transfection
18.
Int J Obes (Lond) ; 30 Suppl 1: S13-8, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16570099

ABSTRACT

Mammalian tissues express at least two cannabinoid receptor types, CB1 and CB2, both G protein coupled. CB1 receptors are found predominantly at nerve terminals where they mediate inhibition of transmitter release. CB2 receptors occur mainly on immune cells, one of their roles being to modulate cytokine release. Endogenous agonists for cannabinoid receptors also exist, and are all eicosanoids. The first-discovered of these 'endocannabinoids' was arachidonoylethanolamide and there is convincing evidence that this ligand and some of its metabolites can activate vanilloid VRI (TRPV1) receptors. Certain cannabinoids also appear to have TRPV1-like and/or non-CB1, non-CB2, non-TRPV1 targets. Several CB1- and CB2-selective agonists and antagonists have been developed. Antagonists include the CB1-selective SR141716A, AM251, AM281 and LY320135, and the CB2-selective SR144528 and AM630. These all behave as inverse agonists, one indication that CB1 and CB2 receptors can exist in a constitutively active state. 'Neutral' cannabinoid receptor antagonists have also been developed. CB1 and/or CB2 receptor activation appears to ameliorate inflammatory and neuropathic pain and certain multiple sclerosis symptoms. This might be exploited clinically by using CB1, CB2 or CB1/CB2 agonists, or inhibitors of the membrane transport or catabolism of endocannabinoids that are released in increased amounts, at least in animal models of pain and multiple sclerosis. We have recently discovered the presence of an allosteric site on the CB1 receptor. Consequently, it may also prove possible to enhance 'autoprotective' effects of released endocannabinoids with CB1 allosteric enhancers or, indeed, to reduce proposed 'autoimpairing' effects of released endocannabinoids such as excessive food intake with CB1 allosteric antagonists.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Nerve Endings/metabolism , Receptors, Cannabinoid/metabolism , Animals , Cannabinoid Receptor Agonists , Cannabinoid Receptor Antagonists , Humans , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Pain/drug therapy , Pain/metabolism , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors
19.
Bull Exp Biol Med ; 142(5): 557-61, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17415461

ABSTRACT

We studied the effect of selective ligands of cannabinoid (CB) receptors on contractility of isolated Langendorff-perfused rat heart under conditions of 45-min total ischemia and 30-min reperfusion. Perfusion with a solution containing selective CB receptor agonist HU-210 for 10 min before ischemia increased the severity of reperfusion contractile dysfunction. This drug decreased left ventricular developed pressure and maximum rates of contraction and relaxation, but had no effect on heart rate and end-diastolic pressure. The negative inotropic effect of the drug was transitory and disappeared after 5-min reperfusion. Pretreatment with selective CB1 receptor antagonist SR141716A and selective CB2 receptor antagonist SR144528 had no effect on heart rate and myocardial contractility during reperfusion. Our results indicate that stimulation of CB receptors can increase the degree of reperfusion-induced cardiac contractile dysfunction. However, endogenous cannabinoids are not involved in the development of myocardial contractile dysfunction during ischemia/reperfusion of the isolated heart.


Subject(s)
Myocardial Contraction/physiology , Myocardial Reperfusion Injury/physiopathology , Receptors, Cannabinoid/physiology , Animals , Blood Pressure/drug effects , Camphanes/pharmacology , Dronabinol/analogs & derivatives , Dronabinol/pharmacology , Heart Rate/drug effects , In Vitro Techniques , Male , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptors, Cannabinoid/drug effects , Rimonabant
20.
Bull Exp Biol Med ; 139(5): 558-61, 2005 May.
Article in English, Russian | MEDLINE | ID: mdl-16224548

ABSTRACT

We studied the effect of selective cannabinoid receptor ligands on contractility of isolated Langendorff-perfused rat heart. It was found that 10-min perfusion of rat heart with a solution containing selective agonist of CB1 and CB2 receptors HU-210 (10 nM) decreased left ventricular developed pressure and maximum rates of contraction and relaxation. However, HU-210 had no effect on heart rate and end-diastolic pressure. Treatment with selective CB1 receptor antagonist SR141716 (1 microM) and selective CB2 receptor antagonist SR144528 (1 microM) decreased left ventricular developed pressure and maximum rates of contraction and relaxation, but had no effect on heart rate and end-diastolic pressure. Ten-minute perfusion of rat heart with a solution containing selective agonist of CB1 and CB2 receptors HU-210 (10 nM) decreased cAMP concentration in the heart. CB receptor antagonists had little effect on cAMP concentration in the heart. The negative inotropic effect of HU-210 and CB receptor antagonists is probably mediated by activation of CB1 receptors. It can be hypothesized that the decrease in heart cAMP concentration is related to stimulation of CB2 receptors. Our results suggest that selective CB receptor antagonists SR141716 and SR144528 in a final concentration of 1 microM exhibit properties of partial CB receptor agonists.


Subject(s)
Camphanes/pharmacology , Heart/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptors, Cannabinoid , Animals , Anti-Arrhythmia Agents/pharmacology , Blood Pressure/drug effects , Cannabinoid Receptor Agonists , Cannabinoid Receptor Antagonists , Cannabinoids/antagonists & inhibitors , Dronabinol/analogs & derivatives , Dronabinol/pharmacology , Heart Rate/drug effects , In Vitro Techniques , Myocardial Contraction/drug effects , Rats , Rimonabant
SELECTION OF CITATIONS
SEARCH DETAIL
...