Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
Behav Brain Res ; 378: 112297, 2020 01 27.
Article En | MEDLINE | ID: mdl-31626848

Although a lot of information can be found on the specific dual role of the endocannabinoid system in the emotional-related responses, little is known whether stimulation or inhibition of the cannabinoid (CB) receptors may affect the activity of the frequently prescribed antidepressant drugs. Our interests have been particularly focused on the potential influence of the CB2 receptors, as the ones whose central effects are relatively poorly documented when compared to the central effects of the CB1 receptors. Therefore, we evaluated the potential interaction between the CB2 receptor ligands (i.e., JWH133 - CB2 receptor agonist and AM630 - CB2 receptor inverse agonist) and several common antidepressant drugs that influence the monoaminergic system (i.e., imipramine, escitalopram, reboxetine). In order to assess the antidepressant-like effects we used two widely recognized behavioural tests, the mouse forced swim test (FST) and the tail suspension test (TST). Brain concentrations of the tested antidepressants were evaluated by the HPLC method. Intraperitoneal co-administration of per se ineffective doses of JWH133 (0.25 mg/kg) or AM630 (0.25 mg/kg) with imipramine (15 mg/kg), escitalopram (2 mg/kg), and reboxetine (2.5 mg/kg) significantly shortened the immobility time of mice in the FST and the TST, whereas it did not disturb their spontaneous locomotor activity. Furthermore, the brain levels of antidepressants were not changed. Summarizing, the results of the present study revealed that both activation and inhibition of the CB2 receptor function have a potential to strengthen the antidepressant activity of drugs targeting the monoaminergic system. Most probably, the described interaction has a pharmacodynamic background.


Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Cannabinoid Receptor Modulators/pharmacology , Locomotion/drug effects , Receptor, Cannabinoid, CB2/drug effects , Animals , Antidepressive Agents/administration & dosage , Antidepressive Agents/pharmacokinetics , Cannabinoid Receptor Agonists/administration & dosage , Cannabinoid Receptor Modulators/administration & dosage , Cannabinoid Receptor Modulators/pharmacokinetics , Cannabinoids/administration & dosage , Citalopram/administration & dosage , Drug Synergism , Imipramine/administration & dosage , Indoles/administration & dosage , Male , Mice , Reboxetine/administration & dosage , Receptor, Cannabinoid, CB2/agonists
2.
Neuropsychopharmacology ; 43(1): 34-51, 2018 Jan.
Article En | MEDLINE | ID: mdl-28811670

Recent policy changes have led to significant increases in the use of cannabis for both medical and recreational purposes. Although men are more likely to endorse past month cannabis use and are more frequently diagnosed with Cannabis Use Disorder relative to women, a growing proportion of medical cannabis users are reported to be women. The increased popularity of cannabis for medical purposes and the narrowing gap in prevalence of use between men and women raises questions regarding sex-dependent effects related to therapeutic efficacy and negative health effects of cannabis and cannabinoids. The objective of this review is to provide a translational perspective on the sex-dependent effects of cannabis and cannabinoids by synthesizing findings from preclinical and clinical studies focused on sex comparisons of their therapeutic potential and abuse liability, two specific areas that are of significant public health relevance. Hormonal and pharmacological mechanisms that may underlie sex differences in the effects of cannabis and cannabinoids are highlighted.


Cannabinoid Receptor Modulators/pharmacology , Cannabinoids/pharmacology , Cannabis , Sex Characteristics , Animals , Cannabinoid Receptor Modulators/adverse effects , Cannabinoid Receptor Modulators/pharmacokinetics , Cannabinoid Receptor Modulators/therapeutic use , Cannabinoids/adverse effects , Cannabinoids/pharmacokinetics , Cannabinoids/therapeutic use , Humans , Marijuana Abuse/physiopathology , Medical Marijuana/adverse effects , Medical Marijuana/pharmacokinetics , Medical Marijuana/pharmacology , Medical Marijuana/therapeutic use
3.
Neuropsychopharmacology ; 40(13): 2948-59, 2015 Dec.
Article En | MEDLINE | ID: mdl-26052038

The CB1 receptor represents a promising target for the treatment of several disorders including pain-related disease states. However, therapeutic applications of Δ(9)-tetrahydrocannabinol and other CB1 orthosteric receptor agonists remain limited because of psychoactive side effects. Positive allosteric modulators (PAMs) offer an alternative approach to enhance CB1 receptor function for therapeutic gain with the promise of reduced side effects. Here we describe the development of the novel synthetic CB1 PAM, 6-methyl-3-(2-nitro-1-(thiophen-2-yl)ethyl)-2-phenyl-1H-indole (ZCZ011), which augments the in vitro and in vivo pharmacological actions of the CB1 orthosteric agonists CP55,940 and N-arachidonoylethanolamine (AEA). ZCZ011 potentiated binding of [(3)H]CP55,940 to the CB1 receptor as well as enhancing AEA-stimulated [(35)S]GTPγS binding in mouse brain membranes and ß-arrestin recruitment and ERK phosphorylation in hCB1 cells. In the whole animal, ZCZ011 is brain penetrant, increased the potency of these orthosteric agonists in mouse behavioral assays indicative of cannabimimetic activity, including antinociception, hypothermia, catalepsy, locomotor activity, and in the drug discrimination paradigm. Administration of ZCZ011 alone was devoid of activity in these assays and did not produce a conditioned place preference or aversion, but elicited CB1 receptor-mediated antinociceptive effects in the chronic constriction nerve injury model of neuropathic pain and carrageenan model of inflammatory pain. These data suggest that ZCZ011 acts as a CB1 PAM and provide the first proof of principle that CB1 PAMs offer a promising strategy to treat neuropathic and inflammatory pain with minimal or no cannabimimetic side effects.


Analgesics, Non-Narcotic/pharmacology , Cannabinoid Receptor Modulators/pharmacology , Indoles/pharmacology , Neuralgia/drug therapy , Neuralgia/metabolism , Receptor, Cannabinoid, CB1/metabolism , Thiophenes/pharmacology , Allosteric Regulation , Amidohydrolases/genetics , Amidohydrolases/metabolism , Analgesics, Non-Narcotic/adverse effects , Analgesics, Non-Narcotic/pharmacokinetics , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/drug effects , Brain/metabolism , CHO Cells , Cannabinoid Receptor Modulators/adverse effects , Cannabinoid Receptor Modulators/pharmacokinetics , Carrageenan , Cricetulus , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Indoles/adverse effects , Indoles/pharmacokinetics , Male , Mice, Inbred C57BL , Mice, Knockout , Thiophenes/adverse effects , Thiophenes/pharmacokinetics
4.
Acta Neurobiol Exp (Wars) ; 74(2): 197-210, 2014.
Article En | MEDLINE | ID: mdl-24993629

Paediatric traumatic brain injury (TBI) is a leading cause of death and disability. Previous studies showed neuroprotection after TBI by (endo)cannabinoid mechanisms, suggesting involvement of cannabinoid receptors (CBR). We therefore determined CBR densities and expression of the translocator protein 18 kDA (TSPO) in newborn piglets after experimental TBI. Newborn female piglets were subjected to sham operation (n=6) or fluid-percussion (FP) injury (n=7) under controlled physiological conditions. After six hours, brains were frozen, sagittally cut and incubated with radioligands for CBR ([3HCP-55,940, [3H]SR141716A) and TSPO ([3H]PK11195), an indicator of gliosis/brain injury. Early after injury, FP-TBI elicited a significant ICP increase at a temporary reduced cerebral perfusion pressure; however, CBF and CMRO2 remained within physiological range. At 6 hours post injury, we found a statistically significant increase in binding of the non-selective agonist [3H]CP-55,940 in 15 of the 24 investigated brain regions of injured animals. By contrast, no significant changes in binding of the CB1R-selective antagonist [3H]SR141716A were observed. A non-significant trend towards increased binding of [3H]PK11195 was observed, suggesting an incipient microglial activation. We therefore conclude that in this model and time span after injury, the increase in [3H]CP-55,940 binding reflects changes in CB2R density, while CB1R density is not affected. The results may provide explanation for the neuroprotective properties of cannabinoid ligands and future therapeutic strategies of TBI.


Brain Injuries/metabolism , Brain Injuries/pathology , Brain/metabolism , Receptors, Cannabinoid/metabolism , Analysis of Variance , Animals , Animals, Newborn , Autoradiography , Brain/diagnostic imaging , Brain/drug effects , Brain Injuries/diagnostic imaging , Cannabinoid Receptor Modulators/pharmacokinetics , Cyclohexanols/pharmacokinetics , Disease Models, Animal , Female , Isoquinolines , Piperidines/pharmacokinetics , Protein Binding/drug effects , Pyrazoles/pharmacokinetics , Radionuclide Imaging , Receptors, GABA/metabolism , Rimonabant , Swine , Time Factors , Tritium/pharmacokinetics
5.
Behav Pharmacol ; 25(2): 182-5, 2014 Apr.
Article En | MEDLINE | ID: mdl-24603340

Several allosteric modulators (AMs) of the CB1 receptor have been characterized in vitro, including Org27569, which enhances CB1-specific binding of [H]CP55,940, but behaves as an insurmountable CB1-receptor antagonist in several biochemical assays. Although a growing body of research has investigated the molecular actions of this unusual AM, it is unknown whether these actions translate to the whole animal. The purpose of the present study was to determine whether Org27569 would produce effects in well-established mouse behavioral assays sensitive to CB1 orthosteric agonists and antagonists. Similar to the orthosteric CB1 antagonist/inverse agonist rimonabant, Org27569 reduced food intake; however, this anorectic effect occurred independently of the CB1 receptor. Org27569 did not elicit CB1-mediated effects alone and lacked efficacy in altering antinociceptive, cataleptic, and hypothermic actions of the orthosteric agonists anandamide, CP55,940, and Δ-tetrahydrocannabinol. Moreover, it did not alter the discriminative stimulus effects of anandamide in FAAH-deficient mice or Δ-tetrahydrocannabinol in wild-type mice in the drug discrimination paradigm. These findings question the utility of Org27569 as a 'gold standard' CB1 AM and underscore the need for the development of CB1 AMs with pharmacology that translates from the molecular level to the whole animal.


Cannabinoid Receptor Modulators/pharmacology , Indoles/pharmacology , Piperidines/pharmacology , Receptor, Cannabinoid, CB1/metabolism , Allosteric Regulation , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , Arachidonic Acids/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Cannabinoid Receptor Modulators/pharmacokinetics , Catalepsy/chemically induced , Catalepsy/drug therapy , Catalepsy/metabolism , Cyclohexanols/pharmacology , Dronabinol/pharmacology , Drug Evaluation , Eating/drug effects , Eating/physiology , Endocannabinoids/pharmacology , Female , Hypothermia/chemically induced , Hypothermia/drug therapy , Hypothermia/metabolism , Indoles/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Nociception/drug effects , Nociception/physiology , Piperidines/pharmacokinetics , Polyunsaturated Alkamides/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/genetics , Rimonabant
6.
J Neurosci ; 32(20): 7091-101, 2012 May 16.
Article En | MEDLINE | ID: mdl-22593077

Painful peripheral neuropathy is a dose-limiting complication of chemotherapy. Cisplatin produces a cumulative toxic effect on peripheral nerves, and 30-40% of cancer patients receiving this agent experience pain. By modeling cisplatin-induced hyperalgesia in mice with daily injections of cisplatin (1 mg/kg, i.p.) for 7 d, we investigated the anti-hyperalgesic effects of anandamide (AEA) and cyclohexylcarbamic acid 3'-carbamoyl-biphenyl-3-yl ester (URB597), an inhibitor of AEA hydrolysis. Cisplatin-induced mechanical and heat hyperalgesia were accompanied by a decrease in the level of AEA in plantar paw skin. No changes in motor activity were observed after seven injections of cisplatin. Intraplantar injection of AEA (10 µg/10 µl) or URB597 (9 µg/10 µl) transiently attenuated hyperalgesia through activation of peripheral CB1 receptors. Co-injections of URB597 (0.3 mg/kg daily, i.p.) with cisplatin decreased and delayed the development of mechanical and heat hyperalgesia. The effect of URB597 was mediated by CB1 receptors since AM281 (0.33 mg/kg daily, i.p.) blocked the effect of URB597. Co-injection of URB597 also normalized the cisplatin-induced decrease in conduction velocity of Aα/Aß-fibers and reduced the increase of ATF-3 and TRPV1 immunoreactivity in dorsal root ganglion (DRG) neurons. Since DRGs are a primary site of toxicity by cisplatin, effects of cisplatin were studied on cultured DRG neurons. Incubation of DRG neurons with cisplatin (4 µg/ml) for 24 h decreased the total length of neurites. URB597 (100 nM) attenuated these changes through activation of CB1 receptors. Collectively, these results suggest that pharmacological facilitation of AEA signaling is a promising strategy for attenuating cisplatin-associated sensory neuropathy.


Arachidonic Acids/therapeutic use , Benzamides/therapeutic use , Carbamates/therapeutic use , Cisplatin/antagonists & inhibitors , Hyperalgesia/drug therapy , Neurotoxicity Syndromes/drug therapy , Peripheral Nervous System Diseases/drug therapy , Polyunsaturated Alkamides/therapeutic use , Receptor, Cannabinoid, CB1/agonists , Activating Transcription Factor 3/metabolism , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/antagonists & inhibitors , Arachidonic Acids/pharmacokinetics , Arachidonic Acids/pharmacology , Benzamides/antagonists & inhibitors , Benzamides/pharmacology , Cannabinoid Receptor Modulators/pharmacokinetics , Cannabinoid Receptor Modulators/pharmacology , Cannabinoid Receptor Modulators/therapeutic use , Carbamates/antagonists & inhibitors , Carbamates/pharmacology , Cells, Cultured , Cisplatin/adverse effects , Disease Models, Animal , Drug Interactions , Endocannabinoids , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hyperalgesia/chemically induced , Male , Mice , Mice, Inbred C3H , Morpholines/pharmacology , Motor Activity/drug effects , Neurites/drug effects , Peripheral Nervous System Diseases/chemically induced , Polyunsaturated Alkamides/pharmacokinetics , Polyunsaturated Alkamides/pharmacology , Pyrazoles/pharmacology , TRPV Cation Channels/metabolism
7.
Nat Rev Drug Discov ; 11(4): 292-310, 2012 Apr.
Article En | MEDLINE | ID: mdl-22460123

Chronic pain remains unsatisfactorily treated, and few novel painkillers have reached the market in the past century. Increasing the levels of the main endogenous opioid peptides - enkephalins - by inhibiting their two inactivating ectopeptidases, neprilysin and aminopeptidase N, has analgesic effects in various models of inflammatory and neuropathic pain. Stemming from the same pharmacological concept, fatty acid amide hydrolase (FAAH) inhibitors have also been found to have analgesic effects in pain models by preventing the breakdown of endogenous cannabinoids. Dual enkephalinase inhibitors and FAAH inhibitors are now in early-stage clinical trials. In this Review, we compare the effects of these two potential classes of novel analgesics and describe the progress in their rational design. We also consider the challenges in their clinical development and opportunities for combination therapies.


Analgesics/pharmacology , Analgesics/pharmacokinetics , Cannabinoid Receptor Modulators/antagonists & inhibitors , Cannabinoid Receptor Modulators/pharmacokinetics , Chronic Pain/drug therapy , Enkephalins/antagonists & inhibitors , Amidohydrolases/antagonists & inhibitors , Animals , Cannabinoid Receptor Modulators/pharmacology , Chronic Pain/metabolism , Humans
8.
Trends Pharmacol Sci ; 33(4): 181-5, 2012 Apr.
Article En | MEDLINE | ID: mdl-22297258

The endocannabinoids anandamide (AEA) and 2-arachidonoylglycerol are removed from the extracellular space by a process of cellular uptake followed by metabolism. Although the enzymes responsible for endocannabinoid metabolism have been well characterised, the processes involved in uptake have been the subject of much controversy. Recent studies, however, have identified intracellular transport proteins (fatty acid binding proteins 5 and 7, heat shock protein 70, albumin, and fatty acid amide hydrolase-like AEA transporter protein) that shuttle AEA from the plasma membrane to its metabolic enzymes. Proteins such as the fatty acid amide hydrolase-like anandamide transporter protein may be useful targets for novel therapeutic strategies aimed at potentiating AEA signalling. In this article I review the current state of the art of endocannabinoid uptake.


Arachidonic Acids/metabolism , Cannabinoid Receptor Modulators/metabolism , Carrier Proteins/metabolism , Polyunsaturated Alkamides/metabolism , Animals , Arachidonic Acids/pharmacokinetics , Cannabinoid Receptor Modulators/pharmacokinetics , Cell Membrane/metabolism , Endocannabinoids , Humans , Polyunsaturated Alkamides/pharmacokinetics , Protein Transport
9.
Eur J Pharmacol ; 600(1-3): 50-8, 2008 Dec 14.
Article En | MEDLINE | ID: mdl-18762181

Lack of involvement of the opioid system with the endocannabinoid, arachidonylethanolamide (anandamide) was possibly due to hydrolysis by fatty acid amide hydrolase (FAAH). Cyclohexylcarbamic acid 3'-carbamoyl-biphenyl-3-yl ester (URB597) is an inhibitor of FAAH, increases brain anandamide levels and enhances anandamide-induced antinociception in male ICR mice (25-30 g). The combination of URB597 (10 mg/kg, i.p.) and anandamide (40 mg/kg, i.p.) produced maximal antinociception in the mouse tail-flick test [68.7+/-16.8 percent maximum possible effect (%MPE)], versus either substance alone (27.3+/-7.9%MPE and 4.6+/-2.3%MPE, respectively) and is significantly blocked (p<0.05) by the cannabinoid CB(1) receptor antagonist, SR141716A (rimonabant), the kappa opioid receptor-selective antagonist, nor-Binaltorphimine (10 microg i.t.; 12.7+/-4.0%MPE) and the mu opioid receptor antagonist, naloxone (1 mg/kg, s.c.; 6.0+/-3.8%MPE), but not by the delta opioid receptor-selective antagonist, naltrindole (2 mg/kg, s.c.; 29.7+/-8.2%MPE) or the cannabinoid CB(2) receptor antagonist, SR144528. In addition, nor-BNI (10 microg i.t) administration to FAAH(-/-) knockout mice produced a nociceptive response. The URB597/anandamide combination was not active in the CB(1)(-/-) knockout mice, but retained activity in the MOR(-/-) knockout mice. The sub-active combination of (URB597 10 mg/kg, i.p/anandamide 10 mg/kg, i.p.; 15.5+/-4.3%MPE) shifted the dose response curve of morphine to the left (morphine alone ED(50)=4.6 mg/kg [3.7-5.6] versus morphine/URB597/anandamide (ED(50)=2.5 mg/kg [1.9-3.4]). These data are the first demonstration that anandamide, if protected from degradation, acts via the CB(1) receptor to interact with kappa opioid receptor systems in opioid analgesia.


Arachidonic Acids/pharmacology , Cannabinoid Receptor Modulators/pharmacology , Polyunsaturated Alkamides/pharmacology , Receptor, Cannabinoid, CB1/drug effects , Receptors, Opioid, kappa/drug effects , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Animals , Arachidonic Acids/administration & dosage , Arachidonic Acids/pharmacokinetics , Benzamides/administration & dosage , Benzamides/pharmacology , Cannabinoid Receptor Modulators/administration & dosage , Cannabinoid Receptor Modulators/pharmacokinetics , Carbamates/administration & dosage , Carbamates/pharmacology , Dose-Response Relationship, Drug , Drug Therapy, Combination , Endocannabinoids , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Male , Mice , Mice, Inbred ICR , Mice, Knockout , Morphine/administration & dosage , Morphine/pharmacology , Pain/drug therapy , Polyunsaturated Alkamides/administration & dosage , Polyunsaturated Alkamides/pharmacokinetics , Receptor, Cannabinoid, CB1/genetics , Receptors, Opioid, kappa/metabolism
10.
Br J Pharmacol ; 152(5): 624-32, 2007 Nov.
Article En | MEDLINE | ID: mdl-17704819

Cannabinoid CB1 and CB2 receptors are located at key sites involved in the relaying and processing of noxious inputs. Both CB1 and CB2 receptor agonists have analgesic effects in a range of models of inflammatory and neuropathic pain. Importantly, clinical trials of cannabis-based medicines indicate that the pre-clinical effects of cannabinoid agonists may translate into therapeutic potential in humans. One of the areas of concern with this pharmacological approach is that CB1 receptors have a widespread distribution in the brain and that global activation of CB1 receptors is associated with adverse side effects. Studies of the endogenous cannabinoids (endocannabinoids) have demonstrated that they are present in most tissues and that in some pain states, such as neuropathic pain, levels of endocannabinoids are elevated at key sites involved in pain processing. An alternative approach that can be used to harness the potential therapeutic effects of cannabinoids is to maximise the effects of the endocannabinoids, the actions of which are terminated by re-uptake and metabolism by various enzymes, including fatty acid amide hydrolase (FAAH), monoacylglycerol lipase (MAGL) and cyclooxygenase type 2 (COX2). Preventing the metabolism, or uptake, of endocannabinoids elevates levels of these lipid compounds in tissue and produces behavioural analgesia in models of acute pain. Herein we review recent studies of the effects of inhibition of metabolism of endocannabinoids versus uptake of endocannabinoids on nociceptive processing in models of inflammatory and neuropathic pain.


Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Inflammation/physiopathology , Pain/physiopathology , Animals , Cannabinoid Receptor Antagonists , Cannabinoid Receptor Modulators/pharmacokinetics , Humans , Inflammation/metabolism , Inflammation/prevention & control , Models, Biological , Pain/metabolism , Pain/prevention & control , Receptors, Cannabinoid/physiology
11.
Br J Pharmacol ; 139(4): 805-15, 2003 Jun.
Article En | MEDLINE | ID: mdl-12813004

1 The purpose of this study was to determine whether endocannabinoids can protect the heart against ischaemia and reperfusion. 2 Rat isolated hearts were exposed to low-flow ischaemia (0.5-0.6 ml min(-1)) and reperfusion. Functional recovery as well as CK and LDH overflow into the coronary effluent were monitored. Infarct size was determined at the end of the experiments. Phosphorylation levels of p38, ERK1/2, and JNK/SAPK kinases were measured by Western blots. 3 None of the untreated hearts recovered from ischaemia during the reperfusion period. Perfusion with either 300 nM palmitoylethanolamide (PEA) or 300 nM 2-arachidonoylglycerol (2-AG), but not anandamide (up to 1 micro M), 15 min before and throughout the ischaemic period, improved myocardial recovery and decreased the levels of coronary CK and LDH. PEA and 2-AG also reduced infarct size. 4 The CB(2)-receptor antagonist, SR144528, blocked completely the cardioprotective effect of both PEA and 2-AG, whereas the CB(1)-receptor antagonist, SR141716A, blocked partially the effect of 2-AG only. In contrast, both ACEA and JWH015, two selective agonists for CB(1)- and CB(2)- receptors, respectively, reduced infarct size at a concentration of 50 nM. 5 PEA enhanced the phosphorylation level of p38 MAP kinase during ischaemia. PEA perfusion doubled the baseline phosphorylation level of ERK1/2, and enhanced its increase upon reperfusion. The cardioprotective effect of PEA was completely blocked by the p38 MAP kinase inhibitor, SB203580, and significantly reduced by the ERK1/2 inhibitor, PD98059, and the PKC inhibitor, chelerythrine. 6 In conclusion, endocannabinoids exert a strong cardioprotective effect in a rat model of ischaemia-reperfusion that is mediated mainly through CB(2)-receptors, and involves p38, ERK1/2, as well as PKC activation.


Cannabinoid Receptor Modulators/pharmacokinetics , Endocannabinoids , Heart/drug effects , Myocardial Ischemia/prevention & control , Amides , Animals , Arachidonic Acids/antagonists & inhibitors , Arachidonic Acids/pharmacology , Arachidonic Acids/therapeutic use , Biomarkers , Blotting, Western , Camphanes/pharmacology , Cannabinoid Receptor Modulators/administration & dosage , Cannabinoid Receptor Modulators/therapeutic use , Ethanolamines , Glycerides/antagonists & inhibitors , Glycerides/pharmacology , Glycerides/therapeutic use , Heart/physiopathology , Imidazoles/pharmacology , L-Lactate Dehydrogenase/analysis , L-Lactate Dehydrogenase/drug effects , L-Lactate Dehydrogenase/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Myocardial Infarction/complications , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardial Ischemia/complications , Myocardial Ischemia/drug therapy , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/prevention & control , Palmitic Acids/antagonists & inhibitors , Palmitic Acids/pharmacology , Palmitic Acids/therapeutic use , Piperidines/pharmacology , Protein Kinase C/analysis , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Rimonabant , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases
...