Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 13: 803331, 2022.
Article in English | MEDLINE | ID: mdl-35529434

ABSTRACT

Acutely, non-selective cannabinoid (CB) agonists have been shown to increase morphine antinociceptive effects, and we and others have also demonstrated that non-selective CB agonists attenuate morphine antinociceptive tolerance. Activation of cannabinoid CB2 receptors reverses allodynia and hyperalgesia in models of chronic pain, and co-administration of morphine with CB2 receptor selective agonists has been shown to be synergistic. CB2 receptor activation has also been shown to reduce morphine-induced hyperalgesia in rodents, an effect attributed to CB2 receptor modulation of inflammation. In the present set of experiments, we tested both the acute and chronic interactions between morphine and the CB2 receptor selective agonist O-1966 treatments on antinociception and antinociceptive tolerance in C57Bl6 mice. Co-administration of morphine and O-1966 was tested under three dosing regimens: simultaneous administration, morphine pre-treated with O-1966, and O-1966 pre-treated with morphine. The effects of O-1966 on mu-opioid receptor binding were determined using [3H]DAMGO and [35S]GTPγS binding assays, and these interactions were further examined by FRET analysis linked to flow cytometry. Results yielded surprising evidence of interactions between the CB2 receptor selective agonist O-1966 and morphine that were dependent upon the order of administration. When O-1966 was administered prior to or simultaneous with morphine, morphine antinociception was attenuated and antinociceptive tolerance was exacerbated. When O-1966 was administered following morphine, morphine antinociception was not affected and antinociceptive tolerance was attenuated. The [35S]GTPγS results suggest that O-1966 interrupts functional activity of morphine at the mu-opioid receptor, leading to decreased potency of morphine to produce acute thermal antinociceptive effects and potentiation of morphine antinociceptive tolerance. However, O-1966 administered after morphine blocked morphine hyperalgesia and led to an attenuation of morphine tolerance, perhaps due to well-documented anti-inflammatory effects of CB2 receptor agonism.

2.
Br J Pharmacol ; 176(10): 1552-1567, 2019 05.
Article in English | MEDLINE | ID: mdl-29338068

ABSTRACT

BACKGROUND AND PURPOSE: It has been suggested that the non-euphorogenic phytocannabinoid cannabidiol (CBD) can ameliorate adverse effects of Δ9 -tetrahydrocannabinol (THC). We determined whether CBD ameliorates cognitive deficits and withdrawal signs induced by cannabinoid CB1 /CB2 receptor agonists or produces these pharmacological effects on its own. EXPERIMENTAL APPROACH: The effects of THC or the CB1 /CB2 receptor full agonist WIN55212 alone, CBD alone or their combination were tested across a range of doses. Cognitive effects were assessed in C57BL/6 mice in a conditional discrimination task and in the Barnes maze. Cannabinoid withdrawal signs were assessed following precipitated withdrawal by acute administration of the CB1 receptor antagonist SR141716, the 5-HT1A receptor antagonist WAY100635, the TRPV1 receptor antagonist capsazepine or the adenosine A2A receptor antagonist SCH58261. KEY RESULTS: THC produced significant motor and cognitive impairment in the Barnes maze task, none of which were attenuated by the addition of CBD. CBD alone did not affect cognitive performance. Precipitation of withdrawal signs by SR141716 occurred in mice chronically treated with THC or WIN55,212. These withdrawal signs were not attenuated by addition of chronic CBD. Chronic treatment with CBD alone did not induce withdrawal signs precipitated by SR141716 or WAY100635. Chronic CBD treatment also produced anxiolysis, which was not altered by attempting to precipitate withdrawal-induced anxiety with a range of antagonists. CONCLUSIONS AND IMPLICATIONS: CBD as a monotherapy may prove to be a safer pharmacological agent, than CB1 receptor agonists alone or in combination with CBD, for the treatment of several disorders. LINKED ARTICLES: This article is part of a themed section on 8th European Workshop on Cannabinoid Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.10/issuetoc.


Subject(s)
Cannabidiol/pharmacology , Cannabinoid Receptor Agonists/adverse effects , Cannabis/chemistry , Cognition/drug effects , Dronabinol/adverse effects , Substance Withdrawal Syndrome/prevention & control , Animals , Cannabidiol/administration & dosage , Cannabidiol/isolation & purification , Cannabinoid Receptor Agonists/administration & dosage , Cannabinoid Receptor Agonists/chemical synthesis , Cannabinoid Receptor Antagonists/pharmacology , Dronabinol/administration & dosage , Dronabinol/chemical synthesis , Mice , Mice, Inbred C57BL , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/agonists , Substance Withdrawal Syndrome/etiology , Substance Withdrawal Syndrome/metabolism
3.
Neurobiol Dis ; 117: 28-41, 2018 09.
Article in English | MEDLINE | ID: mdl-29859319

ABSTRACT

The molecular substrates underlying cocaine reinforcement and addiction have been studied for decades, with a primary focus on signaling molecules involved in modulation of neuronal communication. Brain-derived neurotrophic factor (BDNF) is an important signaling molecule involved in neuronal dendrite and spine modulation. Methyl CpG binding protein 2 (MeCP2) binds to the promoter region of BDNF to negatively regulate its expression and cocaine can recruit MeCP2 to alter the expression of genes such as BDNF that are involved in synaptic plasticity. For several decades, BDNF has been implicated in mediating synaptic plasticity associated with cocaine abuse, and most studies report that neurons are the primary source for BDNF production in the brain. The current study assessed the effects of intravenous cocaine self-administration on microglial activation, and MeCP2 and BDNF expression in reward regions of the brain in vivo, as well as determined specific effects of cocaine exposure on MeCP2 and BDNF expression in human primary neurons and microglia. The results from this study highlight a distinct molecular pathway in microglia through which cocaine increases BDNF, including the phosphorylation of MeCP2 its subsequent translocation from the nucleus to the cytosol, which frees the BDNF promoter and permits its transcriptional activation. Results from these studies show for the first time that cocaine self-administration increases microglial activation, and that microglial MeCP2 is a sensitive target of cocaine resulting in increased release of BDNF from microglia, and possibly contributing to cocaine-induced synaptic plasticity.


Subject(s)
Brain-Derived Neurotrophic Factor/biosynthesis , Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Methyl-CpG-Binding Protein 2/biosynthesis , Microglia/drug effects , Microglia/metabolism , Animals , Brain-Derived Neurotrophic Factor/agonists , Cells, Cultured , Humans , Male , Methyl-CpG-Binding Protein 2/agonists , Rats , Rats, Sprague-Dawley , Self Administration
4.
Cell Immunol ; 329: 1-9, 2018 07.
Article in English | MEDLINE | ID: mdl-29784129

ABSTRACT

We evaluated the effects of the non-psychoactive cannabinoid cannabidiol (CBD) on the inflammatory response and recovery of function following spinal cord injury (SCI). Female C57Bl/6 mice were exposed to spinal cord contusion injury (T9-10) and received vehicle or CBD (1.5 mg/kg IP) injections for 10 weeks following injury. The effect of SCI and CBD treatment on inflammation was assessed via microarray, qRT-PCR and flow cytometry. Locomotor and bladder function and changes in thermal and mechanical hind paw sensitivity were also evaluated. There was a significant decrease in pro-inflammatory cytokines and chemokines associated with T-cell differentiation and invasion in the SCI-CBD group as well as a decrease in T cell invasion into the injured cord. A higher percentage of SCI mice in the vehicle-treated group (SCI-VEH) went on to develop moderate to severe (0-65.9% baseline thermal threshold) thermal sensitivity as compared with CBD-treated (SCI-CBD) mice. CBD did not affect recovery of locomotor or bladder function following SCI. Taken together, CBD treatment attenuated the development of thermal sensitivity following spinal cord injury and this effect may be related to protection against pathological T-cell invasion.


Subject(s)
Cannabidiol/pharmacology , Spinal Cord Injuries/drug therapy , T-Lymphocytes/drug effects , Animals , Cannabidiol/metabolism , Cannabinoids/metabolism , Cannabinoids/pharmacology , Chemokines/drug effects , Cytokines/drug effects , Disease Models, Animal , Female , Hot Temperature , Hyperalgesia , Inflammation/drug therapy , Mice , Mice, Inbred C57BL , Spinal Cord Injuries/complications
5.
Life Sci ; 195: 1-5, 2018 Feb 15.
Article in English | MEDLINE | ID: mdl-29288767

ABSTRACT

AIMS: We tested the hypothesis that CB1/CB2 receptor double knockout would produce significant increases in infarct size and volume and significant worsening in clinical score, using two mouse models, one of permanent ischemia and one of ischemia/reperfusion. MAIN METHODS: Focal cerebral infarcts were created using either photo induced permanent injury or transient middle cerebral artery occlusion. Infarct volume and motor function were evaluated in cannabinoid receptor 1/cannabinoid receptor 2 double knockout mice. KEY FINDINGS: The results surprisingly revealed that CB1/CB2 double knockout mice showed improved outcomes, with the most improvements in the mouse model of permanent ischemia. SIGNIFICANCE: Although the number of individuals suffering from stroke in the United States and worldwide will continue to grow, therapeutic intervention for treatment following stroke remains frustratingly limited. Both the cannabinoid 1 receptor (CB1R) and the cannabinoid 2 receptor (CB2R) have been studied in relationship to stroke. Deletion of the CB2R has been shown to worsen outcome, while selective CB2R agonists have been demonstrated to be neuroprotective following stroke. Although initial studies of CB1R knockout mice demonstrated increased injury following stroke, indicating that activation of the CB1R was neuroprotective, later studies of selective antagonists of the CB1R also demonstrated a protective effect. Surprisingly the double knockout animals had improved outcome. Since the phenotype of the double knockout is not dramatically changed, significant changes in the contribution of other homeostatic pathways in compensation for the loss of these two important receptors may explain these apparently contradictory results.


Subject(s)
Brain Ischemia/genetics , Brain Ischemia/physiopathology , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/genetics , Animals , Brain/pathology , Brain Ischemia/therapy , Cerebral Infarction/etiology , Cerebral Infarction/genetics , Cerebral Infarction/pathology , Cerebrovascular Circulation/genetics , Infarction, Middle Cerebral Artery , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroprotective Agents/therapeutic use , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Stroke/genetics , Stroke/physiopathology , Stroke/therapy , Treatment Outcome
6.
Br J Pharmacol ; 174(17): 2832-2841, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28548225

ABSTRACT

BACKGROUND AND PURPOSE: The non-psychoactive phytocannabinoid cannabidiol (CBD) can affect the pharmacological effects of Δ9 -tetrahydrocannabinol (THC). We tested the possible synergy between CBD and THC in decreasing mechanical sensitivity in a mouse model of paclitaxel-induced neuropathic pain. We also tested the effects of CBD on oxaliplatin- and vincristine-induced mechanical sensitivity. EXPERIMENTAL APPROACH: Paclitaxel-treated mice (8.0 mg·kg-1 i.p., days 1, 3, 5 and 7) were pretreated with CBD (0.625-20.0 mg·kg-1 i.p.), THC (0.625-20.0 mg·kg-1 i.p.) or CBD + THC (0.04 + 0.04-20.0 + 20.0 mg·kg-1 i.p.), and mechanical sensitivity was assessed on days 9, 14 and 21. Oxaliplatin-treated (6.0 mg·kg-1 i.p., day 1) or vincristine-treated mice (0.1 mg·kg-1 i.p. days 1-7) were pretreated with CBD (1.25-10.0 mg·kg-1 i.p.), THC (10.0 mg·kg-1 i.p.) or THC + CBD (0.16 mg·kg-1 THC + 0.16 mg·kg-1 CBD i.p.). KEY RESULTS: Both CBD and THC alone attenuated mechanical allodynia in mice treated with paclitaxel. Very low ineffective doses of CBD and THC were synergistic when given in combination. CBD also attenuated oxaliplatin- but not vincristine-induced mechanical sensitivity, while THC significantly attenuated vincristine- but not oxaliplatin-induced mechanical sensitivity. The low dose combination significantly attenuated oxaliplatin- but not vincristine-induced mechanical sensitivity. CONCLUSIONS AND IMPLICATIONS: CBD may be potent and effective at preventing the development of chemotherapy-induced peripheral neuropathy, and its clinical use may be enhanced by co-administration of low doses of THC. These treatment strategies would increase the therapeutic window of cannabis-based pharmacotherapies.


Subject(s)
Analgesics/therapeutic use , Cannabidiol/therapeutic use , Dronabinol/therapeutic use , Hyperalgesia/drug therapy , Neuralgia/drug therapy , Animals , Antineoplastic Agents, Phytogenic , Disease Models, Animal , Drug Therapy, Combination , Hyperalgesia/chemically induced , Male , Mice, Inbred C57BL , Neuralgia/chemically induced , Organoplatinum Compounds , Oxaliplatin , Paclitaxel , Vincristine
7.
Concussion ; 2(4): CNC49, 2017 Dec.
Article in English | MEDLINE | ID: mdl-30202590

ABSTRACT

Post-traumatic headache is the most common symptom of postconcussion syndrome and becomes a chronic neurological disorder in a substantial proportion of patients. This review provides a brief overview of the epidemiology of postconcussion headache, research models used to study this disorder, as well as the proposed mechanisms. An objective of this review is to enhance the understanding of how the endogenous cannabinoid system is essential for maintaining the balance of the CNS and regulating inflammation after injury, and in turn making the endocannabinoid system a potential modulator of the trigeminal response to concussion. The review describes the role of endocannabinoid modulation of pain and the potential for use of phytocannabinoids to treat pain, migraine and concussion.

8.
Neurosci Lett ; 630: 9-15, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27453059

ABSTRACT

Modulation of the endocannabinoid system has been shown to have a significant impact on outcomes in animal models of stroke. We have previously reported a protective effect of the CB1 antagonist, SR141716A, in a transient reperfusion mouse model of cerebral ischemia. This protective effect was in part mediated by activation of the 5HT1A receptor. Here we have examined its effect in a mouse model of permanent ischemia induced by photoinjury. The CB1 antagonist was found to be protective in this model. As was the case following transient ischemia reperfusion, SR141716A (5mg/kg) resulted in smaller infarct fractions and stroke volumes when utilized both as a pretreatment and as a post-treatment. In contrast to the effect in a transient ischemia model, the pretreatment effect did not depend on the 5HT1A receptor. Neurological function correlated favorably to the reduction in stroke size when SR141716A was given as a pretreatment. With the incidence of stroke predicted to rise in parallel with an ever aging population, understanding mechanisms underlying ischemia and therapeutics remains a paramount goal of research.


Subject(s)
Brain Ischemia/prevention & control , Cannabinoid Receptor Antagonists/administration & dosage , Neuroprotective Agents/administration & dosage , Piperidines/administration & dosage , Pyrazoles/administration & dosage , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Stroke/prevention & control , Animals , Behavior, Animal/drug effects , Body Weight/drug effects , Brain/drug effects , Brain/pathology , Brain Ischemia/complications , Brain Ischemia/pathology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Rimonabant , Stroke/complications , Stroke/pathology
9.
Life Sci ; 138: 72-7, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26032254

ABSTRACT

AIMS: We have recently demonstrated that treatment with a cannabinoid CB2 agonist was protective in a mouse middle cerebral artery occlusion model of cerebral ischemia/reperfusion injury. The present study aimed to determine whether these protective effects of CB2 agonism would extend to a mouse photoinjury model of permanent ischemia and determine associated alterations in cognition and infarct size. MAIN METHODS: Mice received three injections of the CB2 selective agonist O-1966 or vehicle 1h prior to and 2 and 5days following induction of stroke. Infarct size was assessed at 1, 3, or 7days post-injury and learning and memory effects of injury and O-1966 treatment were assessed on days 6 and 7 using a novel object recognition task and an operant acquisition and retention procedure. KEY FINDINGS: O-1966 treated mice had significantly smaller infarct volumes compared with vehicle treated mice. Photoinjury was also associated with a significant memory impairment on day 7 post-injury, and this deficit was reversed with O-1966 treatment. Surprisingly, sham-operated mice receiving O-1966 treatment showed a significant learning deficit in both the recognition and operant tasks compared with vehicle treated sham mice. SIGNIFICANCE: We conclude that CB2 activation is protective against cognitive deficits and tissue damage following permanent ischemia, but may dysregulate glial or neuronal function of learning and memory circuits in the absence of injury and/or inflammation.


Subject(s)
Anisoles/therapeutic use , Cannabinoid Receptor Agonists/therapeutic use , Memory Disorders/drug therapy , Memory Disorders/psychology , Receptor, Cannabinoid, CB2/drug effects , Stroke/drug therapy , Stroke/psychology , Animals , Cerebral Infarction/pathology , Conditioning, Operant/drug effects , Cyclohexanols , Male , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Psychomotor Performance/drug effects , Recognition, Psychology/drug effects , Stroke/pathology
10.
Behav Pharmacol ; 26(3): 304-14, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25485642

ABSTRACT

Cannabinoid and opioid agonists can display overlapping behavioral effects and the combination of these agonists is known to produce enhanced antinociception in several rodent models of acute and chronic pain. The present study investigated the antinociceptive effects of the nonpsychoactive cannabinoid, cannabidiol (CBD) and the µ-opioid agonist morphine, both alone and in combination, using three behavioral models in mice, to test the hypothesis that combinations of morphine and CBD would produce synergistic effects. The effects of morphine, CBD, and morphine/CBD combinations were assessed in the following assays: (a) acetic acid-stimulated stretching; (b) acetic acid-decreased operant responding for palatable food; and (c) hot plate thermal nociception. Morphine alone produced antinociceptive effects in all three models of acute nociception, whereas CBD alone produced antinociception only in the acetic acid-stimulated stretching assay. The nature of the interactions between morphine and CBD combinations were assessed quantitatively based on the principle of dose equivalence. Combinations of CBD and morphine produced synergistic effects in reversing acetic acid-stimulated stretching behavior, but subadditive effects in the hot plate thermal nociceptive assay and the acetic acid-decreased operant responding for palatable food assay. These results suggest that distinct mechanisms of action underlie the interactions between CBD and morphine in the three different behavioral assays and that the choice of appropriate combination therapies for the treatment of acute pain conditions may depend on the underlying pain type and stimulus modality.


Subject(s)
Acute Pain/drug therapy , Analgesics, Opioid/pharmacology , Cannabidiol/pharmacology , Morphine/pharmacology , Acetic Acid/pharmacology , Acute Pain/physiopathology , Analgesics, Opioid/administration & dosage , Animals , Behavior, Animal/drug effects , Cannabidiol/administration & dosage , Disease Models, Animal , Drug Synergism , Male , Mice , Mice, Inbred C57BL , Morphine/administration & dosage
11.
J Neuroinflammation ; 11: 191, 2014 Nov 22.
Article in English | MEDLINE | ID: mdl-25416141

ABSTRACT

BACKGROUND: Immunomodulatory therapies have been identified as interventions for secondary injury after traumatic brain injury (TBI). The cannabinoid receptor type-2 (CB2R) is proposed to play an important, endogenous role in regulating inflammation. The effects of CB2R stimulation, blockade, and deletion on the neurovascular inflammatory responses to TBI were assessed. METHODS: Wild-type C57BL/6 or CB2R knockout mice were randomly assigned to controlled cortical impact (CCI) injury or to craniotomy control groups. The effects of treatment with synthetic, selective CB2R agonists (0-1966 and JWH-133), a selective CB2R antagonist, or vehicle solution administered to CCI groups were assessed at 1-day after injury. Changes in TNF-α, intracellular adhesion molecule (ICAM-1), inducible nitric oxide synthase (iNOS), macrophage/microglial ionized calcium-binding adaptor molecule, and blood-brain-barrier (BBB) permeability were assessed using ELISA, quantitative RT-PCR, immunohistochemistry, and fluorometric analysis of sodium fluorescein uptake. CB2R knockouts and wild-type mice with CCI injury were treated with a CB2R agonist or vehicle treatment. RESULTS: TNF-α mRNA increased at 6 hours and 1 to 3 days after CCI; a CB2R antagonist and genetic knockout of the CB2R exacerbated TNF-α mRNA expression. Treatment with a CB2R agonist attenuated TNF-α protein levels indicating post-transcriptional mechanisms. Intracellular adhesion molecule (ICAM-1) mRNA was increased at 6 hours, and at 1 to 2 days after CCI, reduced in mice treated with a CB2R agonist, and increased in CB2R knockout mice with CCI. Sodium fluorescein uptake was increased in CB2R knockouts after CCI, with and without a CB2R agonist. iNOS mRNA expression peaked early (6 hours) and remained increased from 1 to 3 days after injury. Treatment with a CB2R agonist attenuated increases in iNOS mRNA expression, while genetic deletion of the CB2R resulted in substantial increases in iNOS expression. Double label immunohistochemistry confirmed that iNOS was expressed by macrophage/microglia in the injured cortex. CONCLUSION: Findings demonstrate that the endogenous cannabinoid system and CB2R play an important role in regulating inflammation and neurovascular responses in the traumatically injured brain. CB2R stimulation with two agonists (0-1966 and JWH-133) dampened post-traumatic inflammation, while blockade or deletion of the CB2R worsened inflammation. Findings support previous evidence that modulating the CB2R alters infiltrating macrophages and activated resident microglia. Further investigation into the role of the CB2R on specific immune cell populations in the injured brain is warranted.


Subject(s)
Brain Injuries/metabolism , Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Gene Deletion , Receptor, Cannabinoid, CB2/deficiency , Vasculitis, Central Nervous System/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain Injuries/drug therapy , Cannabinoid Receptor Agonists/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Random Allocation , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Treatment Outcome , Vasculitis, Central Nervous System/drug therapy
12.
Cell Immunol ; 287(1): 1-17, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24342422

ABSTRACT

CB2, the cannabinoid receptor expressed primarily on hematopoietic cells and activated microglia, mediates the immunoregulatory functions of cannabinoids. The involvement of CB2 in EAE has been demonstrated by using both endogenous and exogenous ligands. We showed previously that CB2 selective agonists inhibit leukocyte rolling and adhesion to CNS microvasculature and ameliorate clinical symptom in both chronic and remitting-relapsing EAE models. Here we showed that Gp1a, a highly selective CB2 agonist, with a four log higher affinity for CB2 than CB1, reduced clinical scores and facilitated recovery in EAE in conjunction with long term reduction in demyelination and axonal loss. We also established that Gp1a affected EAE through at least two different mechanisms, i.e. an early effect on Th1/Th17 differentiation in peripheral immune organs, and a later effect on the accumulation of pathogenic immune cells in the CNS, associated with reductions in the expression of CNS and T cell chemokine receptors, chemokines and adhesion molecules. This is the first report on the in vivo CB2-mediated Gp1a inhibition of Th17/Th1 differentiation. We also confirmed the Gp1a-induced inhibition of Th17/Th1 differentiation in vitro, both in non-polarizing and polarizing conditions. The CB2-induced inhibition of Th17 differentiation is highly relevant in view of recent studies emphasizing the importance of pathogenic self-reactive Th17 cells in EAE/MS. In addition, the combined effect on Th17 differentiation and immune cell accumulation into the CNS, emphasize the relevance of CB2 selective ligands as potential therapeutic agents in neuroinflammation.


Subject(s)
Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Indenes/administration & dosage , Pyrazoles/administration & dosage , Receptor, Cannabinoid, CB2/metabolism , Th17 Cells/drug effects , Animals , Cell Adhesion Molecules/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Chemokines/metabolism , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Indenes/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/immunology , Neuroimmunomodulation , Peptide Fragments/immunology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/genetics , Receptors, Chemokine/metabolism , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/immunology
13.
J Biochem ; 153(2): 221-31, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23172304

ABSTRACT

The kunitz protease inhibitor domain of PN2 (PN2KPI) is a potent and specific inhibitor (K(i) 0.5-2 nM) of factor XIa (FXIa) and inhibits cerebrovascular thrombosis in mice. To determine whether the antithrombotic properties of PN2KPI arise from its FXIa-inhibitory activity, we have now prepared mutant forms of PN2KPI. Mutations at the P1 (Arg(15)) site in combination with P2' (Met(17)) mutations profoundly affect inhibition of FXIa, plasmin, kallikrein, factor Xa and thrombin. The mutant proteins PN2KPI-R(15)K, -M(17)K, -R(15)K,M(17)K and -R(15)K,M(17)R lost inhibitory activity against FXIa (K(i) 34, 94, 3081 and 707 nM, respectively) and kallikrein (no inhibition) and gained inhibitory activity against plasmin (K(i) 108, 7, 8 and 8 nM, respectively). The intravenous administration of rPN2KPI into mice dramatically decreased thrombus formation in a murine model of FeCl(3)-induced carotid injury, whereas rPN2KPI-R(15)K,M(17)K failed to inhibit thrombus formation. Molecular modelling studies showed that fine structural variations explain the observed functional differences in FXIa and plasmin inhibition. PN2KPI has potent antithrombotic activity due to its specific FXIa anticoagulant activity, whereas PN2KPI-R(15)K,M(17)K and PN2KPI-R(15)K,M(17)R have potent antifibrinolytic (antiplasmin) activity without anticoagulant or antithrombotic activity.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/pharmacology , Antifibrinolytic Agents/metabolism , Antifibrinolytic Agents/pharmacology , Factor XIa/antagonists & inhibitors , Amyloid beta-Protein Precursor/genetics , Animals , Female , Humans , Mice , Mice, Inbred C57BL , Mutation
14.
Blood ; 120(18): 3741-9, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22972984

ABSTRACT

Administration of cannabinoid receptor 2 (CB2R) agonists in inflammatory and autoimmune disease and CNS injury models results in significant attenuation of clinical disease, and reduction of inflammatory mediators. Previous studies reported that CB2R signaling also reduces leukocyte migration. Migration of dendritic cells (DCs) to various sites is required for their activation and for the initiation of adaptive immune responses. Here, we report for the first time that CB2R signaling affects DC migration in vitro and in vivo, primarily through the inhibition of matrix metalloproteinase 9 (MMP-9) expression. Reduced MMP-9 production by DCs results in decreased migration to draining lymph nodes in vivo and in vitro in the matrigel migration assay. The effect on Mmp-9 expression is mediated through CB2R, resulting in reduction in cAMP levels, subsequent decrease in ERK activation, and reduced binding of c-Fos and c-Jun to Mmp-9 promoter activator protein 1 sites. We postulate that, by dampening production of MMP-9 and subsequent MMP-9-dependent DC migration, cannabinoids contribute to resolve acute inflammation and to reestablish homeostasis. Selective CB2R agonists might be valuable future therapeutic agents for the treatment of chronic inflammatory conditions by targeting activated immune cells, including DCs.


Subject(s)
Cell Movement , Chemotaxis, Leukocyte/immunology , Dendritic Cells/immunology , Matrix Metalloproteinase 9/immunology , Receptor, Cannabinoid, CB2/immunology , Signal Transduction/immunology , Animals , Blotting, Western , Dendritic Cells/cytology , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Receptor, Cannabinoid, CB2/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
J Neurosci Res ; 90(12): 2293-305, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22903455

ABSTRACT

After traumatic brain injury (TBI), inflammation participates in both the secondary injury cascades and the repair of the CNS, both of which are influenced by the endocannabinoid system. This study determined the effects of repeated treatment with a cannabinoid type 2 receptor (CB(2) R) agonist on blood-brain barrier integrity, neuronal degeneration, and behavioral outcome in mice with TBI. We also looked for the presence of a prolonged treatment effect on the macrophage/microglial response to injury. C57BL/6 mice underwent controlled cortical impact (CCI) and received repeated treatments with a CB(2) R agonist, 0-1966, or vehicle. After euthanasia at 6 hr or 1, 2, 3, or 7 days postinjury, brains were removed for histochemical analysis. Blood-brain barrier permeability changes were evaluated by using sodium fluorescein (NaF). Perilesional degenerating neurons, injury volumes, and macrophage/microglia cells were quantified by stereological methods. Rota-rod and open-field testing were performed to evaluate motor function and natural exploratory behavior in mice. 0-1966 Treatment resulted in a significant reduction in NaF uptake and number of degenerating neurons compared with the vehicle-treated group. 0-1966-Treated mice demonstrated improvement on rota-rod and open-field testing compared with vehicle-treated mice. These changes in CCI mice treated with 0-1966 were associated with a prolonged reduction in macrophage/microglia cell counts. In conclusion, repeated treatments with a CB(2) R agonist, 0-1966, result in attenuated blood-brain barrier disruption and neuronal degeneration. In addition, repeated treatment with 0-1966 shows prolonged treatment effects on behavior and the macrophage/microglia cell response over several days.


Subject(s)
Anisoles/therapeutic use , Blood-Brain Barrier/drug effects , Brain Injuries/drug therapy , Nerve Degeneration/prevention & control , Neuroprotective Agents/therapeutic use , Receptor, Cannabinoid, CB2/agonists , Animals , Brain Injuries/pathology , Brain Injuries/physiopathology , Cyclohexanols , Drug Evaluation, Preclinical , Exploratory Behavior , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Neurons/drug effects , Neurons/pathology , Receptor, Cannabinoid, CB2/physiology , Rotarod Performance Test , Wounds, Nonpenetrating/drug therapy , Wounds, Nonpenetrating/pathology , Wounds, Nonpenetrating/physiopathology
16.
Blood ; 120(3): 671-7, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22674803

ABSTRACT

Coagulation factor XI (FXI) plays an important part in both venous and arterial thrombosis, rendering FXIa a potential target for the development of antithrombotic therapy. The kunitz protease inhibitor (KPI) domain of protease nexin-2 (PN2) is a potent, highly specific inhibitor of FXIa, suggesting its possible role in the inhibition of FXI-dependent thrombosis in vivo. Therefore, we examined the effect of PN2KPI on thrombosis in the murine carotid artery and the middle cerebral artery. Intravenous administration of PN2KPI prolonged the clotting time of both human and murine plasma, and PN2KPI inhibited FXIa activity in both human and murine plasma in vitro. The intravenous administration of PN2KPI into WT mice dramatically decreased the progress of FeCl(3)-induced thrombus formation in the carotid artery. After a similar initial rate of thrombus formation with and without PN2KPI treatment, the propagation of thrombus formation after 10 minutes and the amount of thrombus formed were significantly decreased in mice treated with PN2KPI injection compared with untreated mice. In the middle cerebral artery occlusion model, the volume and fraction of ischemic brain tissue were significantly decreased in PN2KPI-treated compared with untreated mice. Thus, inhibition of FXIa by PN2KPI is a promising approach to antithrombotic therapy.


Subject(s)
Amyloid beta-Protein Precursor/pharmacology , Carotid Artery Thrombosis/blood , Carotid Artery Thrombosis/drug therapy , Factor XIa/antagonists & inhibitors , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/drug therapy , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Animals , Anticoagulants/pharmacology , Bleeding Time , Blood Coagulation/drug effects , Blood Coagulation/physiology , Disease Models, Animal , Drug Design , Factor XIa/metabolism , Female , Humans , Injections, Intravenous , Mice , Mice, Inbred C57BL , Protein Structure, Tertiary/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
17.
Curr Pharm Biotechnol ; 13(1): 46-58, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21470162

ABSTRACT

Coronary and carotid arterial occlusion due to thrombosis after atherosclerotic plaque rupture is the major cause of myocardial and cerebral infarction. Together these acute events represent the leading cause of death worldwide. Early reperfusion is the best method to salvage the ischemic organ; however, it leads to additional damage known as reperfusion injury. A large number of experimental studies has been performed in the past aimed at targeting individual mediators of reperfusion injury such as treatment with anti-oxidants or anti-inflammatory agents. Although many agents proved beneficial in animal models of myocardial or cerebral ischemia/reperfusion, the attempts to translate these protective effects into clinical practice were mostly disappointing. Elucidating the complex cellular and molecular mechanisms involved in ischemic cell death is crucial for the development of more efficient drugs in order to improve current treatment strategies. The aim of this review is to discuss cannabinoid and endocannabinoid-mediated effects in the pathogenesis of myocardial infarction and reperfusion injury, post-myocardial infarction remodeling, as well as ischemic stroke and reperfusion injury. We report experimental evidence suggesting that targeting the endocannabinoid system might evolve as a novel therapeutic concept to limit the devastating consequences of these acute vascular events through a wide variety of mechanisms, including lowering inflammation, oxidative stress, fibrosis, and excitotoxicity, and enhanced blood flow.


Subject(s)
Brain Ischemia/drug therapy , Cannabinoid Receptor Modulators/physiology , Myocardial Infarction/drug therapy , Reperfusion Injury/drug therapy , Animals , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebrovascular Circulation , Humans , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Receptor, Cannabinoid, CB1/physiology , Receptor, Cannabinoid, CB2/physiology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology
18.
Transl Stroke Res ; 3(3): 348-56, 2012 Sep.
Article in English | MEDLINE | ID: mdl-24323810

ABSTRACT

We reported previously that both a cannabinoid receptor 2 (CB2R) agonist and a cannabinoid receptor 1 (CB1R) antagonist were protective in the treatment of transient middle cerebral artery occlusion/reperfusion injury (MCAO/R) and that they acted in a synergistic manner when administered in combination. The goal of the current study was to determine which of the potential cannabinoid receptors participate in the protective effects of this drug combination in a mouse model of MCAO/R. The effects of administration of the CB2R agonist/CB1R antagonist combination on infarct size and cerebral blood flow during a 1-h occlusion were tested in CB1R-deficient animals, CB2R-deficient animals, and animals treated with capsazepine, the antagonist for the vanilloid receptor type I (TRPV1) and WAY100135, the antagonist for the hydroxytryptamine1A receptor (5-HT1A). The protective effect of the CB2R agonist/CB1R antagonist combination on infarct size was not influenced by the absence of the CB1R nor by blocking the TRPV1 receptor, but was attenuated by the absence of CB2R and by blocking the 5-HT1A receptor. Increases in cerebral blood flow and arteriolar diameter were also found to be independent of the CB1R and TRPV1 receptor. In conclusion, administration of the CB2R agonist/CB1R antagonist combination causes a significant reduction in infarct size in the MCAO/R model. The protective effect involves both the CB2R and the 5-HT1A receptor. Neither the CB1R nor the TRPV1 receptors appear to participate in this response.

19.
J Neurotrauma ; 28(12): 2417-27, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21970496

ABSTRACT

The goal of the current investigation was to evaluate the mechanisms through which administration of a selective cannabinoid-2 (CB2) agonist (O-1966) modifies inflammatory responses and helps to improve function following spinal cord injury. A comparison of motor function, autonomic function, and inflammatory responses was made between animals treated with O-1966 (5 mg/kg IP) and animals treated with vehicle 1 h and 24 h following contusion injury to the spinal cord. Motor function was significantly improved in the treated animals at each time point during the 14 days of evaluation. The percentage of animals able to spontaneously void their bladder was also greater over the entire study period in the group treated with the selective CB2 agonist. Seven days following injury there was a significant reduction in both hematopoietic and myeloid cell invasion of the spinal cord, and a reduction in the number of immunoreactive microglia. The results of the evaluation of chemokine/cytokine expression and inflammatory cell invasion also demonstrated a significant effect of treatment on inflammatory reactions following injury. Two days after injury, animals treated with O-1966 had significant reductions in CXCL-9 and CXCL-11, and dramatic reductions in IL-23p19 expression and its receptor IL-23r. Treatment with O-1966 also caused inhibition of toll-like receptor expression (TLR1, TLR4, TLR6 and TLR7) following injury. These results demonstrate that the improvement in motor and autonomic function resulting from treatment with a selective CB2 agonist is associated with a significant effect on inflammatory responses in the spinal cord following injury.


Subject(s)
Anisoles/pharmacology , Inflammation Mediators/pharmacology , Receptor, Cannabinoid, CB2/agonists , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Animals , Anisoles/therapeutic use , Cyclohexanols , Female , Inflammation/drug therapy , Inflammation/pathology , Inflammation Mediators/therapeutic use , Mice , Mice, Inbred C57BL , Receptor, Cannabinoid, CB2/physiology
20.
J Neurotrauma ; 28(6): 973-81, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21332427

ABSTRACT

Proposed therapeutic strategies for attenuating secondary traumatic brain injury (TBI) include modulation of acute neuroimmune responses. The goal of this study was to examine the acute effects of cannabinoid-2 receptor (CB(2)R) modulation on behavioral deficits, cerebral edema, perivascular substance P, and macrophage/microglial activation in a murine model of TBI. Thirty male C57BL/6 mice underwent sham surgery, or cortical contusion impact injury (CCI). CCI mice received vehicle or the CB(2)R agonist 0-1966 at 1 and 24 h after injury. Performance on the rotarod, forelimb cylinder, and open-field tests were evaluated before and at 48 h after sham or CCI surgery. Cerebral edema was evaluated using the wet-dry weight technique. Immunohistochemical analysis was used to examine changes in substance P and macrophage/microglia-specific Iba1 protein immunoreactivity. Locomotor performance and exploratory behavior were significantly improved in mice receiving 0-1966 (CB(2)R agonist) compared to vehicle-treated mice. Significant reductions were found for cerebral edema, number of perivascular areas of substance P immunoreactivity, and number of activated macrophages/microglial cells in the injured brains of 0-1966-treated mice compared to vehicle-treated mice. The findings show that the effects of the CB(2)R agonist 0-1966 on edema, substance P immunoreactivity, and macrophage/microglial activation, were associated with recovery of acute motor and exploratory deficits. This study provides evidence of acute neuroprotective effects derived from selective CB(2)R activation that may represent an avenue for further development of novel therapeutic agents in the treatment of TBI.


Subject(s)
Anisoles/pharmacology , Brain Injuries/drug therapy , Brain Injuries/metabolism , Cannabinoid Receptor Modulators/pharmacology , Inflammation Mediators/pharmacology , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/physiology , Animals , Brain Injuries/complications , Cannabinoid Receptor Modulators/therapeutic use , Cyclohexanols , Disease Models, Animal , Inflammation/complications , Inflammation/drug therapy , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/pathology , Resorcinols/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...