Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Headache ; 2024 May 27.
Article in English | MEDLINE | ID: mdl-38800847

ABSTRACT

OBJECTIVE: To report the cardiovascular (CV) safety of dihydroergotamine mesylate (DHE) administered by precision olfactory delivery (INP104) from two clinical trials. BACKGROUND: Although the absolute risk is low, migraine is associated with an increased risk of CV events. DHE is a highly effective acute treatment for migraine, but due to its theoretical risk of promoting arterial vasoconstriction, DHE is contraindicated in patients with CV disease or an unfavorable risk factor profile. The INP104 is a novel drug-device combination product approved for acute treatment of migraine that delivers DHE to the upper nasal space using precision olfactory delivery (POD®). METHODS: The STOP 101 was a Phase 1 open-label study that assessed the safety, tolerability, and bioavailability of INP104 1.45 mg, intravenous DHE 1.0 mg, and MIGRANAL (nasal DHE) 2.0 mg in healthy participants. The STOP 301 was a pivotal Phase 3, open-label study that assessed the safety, tolerability, and exploratory efficacy of INP104 1.45 mg over 24 and 52 weeks in patients with migraine. In both studies, active or a history of CV disease, as well as significant CV risk factors, were exclusion criteria. RESULTS: In STOP 101, 36 participants received one or more doses of investigational product. Treatment with intravenous DHE, but not INP104 or nasal DHE, resulted in clinically relevant changes from baseline in systolic blood pressure (BP; 11.4 mmHg, 95% confidence interval [CI] 7.9-15.0) and diastolic BP (13.3 mmHg, 95% CI 9.4-17.1) at 5 min post-dose, persisting up to 30 min post-dose for systolic BP (6.3 mmHg; 95% CI 3.0-9.5) and diastolic BP (7.9 mmHg, 95% CI 3.9-11.9). None of the treatments produced any clinically meaningful electrocardiogram (ECG) changes. In STOP 301, 354 patients received one or more doses of INP104. Over 24 weeks, five patients (1.4%) experienced a non-serious, vascular treatment-emergent adverse event (TEAE). Minimal changes were observed for BP and ECG parameters over 24 or 52 weeks. Off-protocol concomitant use of triptans and other ergot derivatives did not result in any TEAEs. CONCLUSION: In two separate studies, INP104 demonstrated a favorable CV safety profile when used in a study population without CV-related contraindications.

2.
Front Neurol ; 14: 1282846, 2023.
Article in English | MEDLINE | ID: mdl-38073648

ABSTRACT

Introduction: Dihydroergotamine mesylate (DHE) is an established effective acute therapy for migraine and is often characterized by its broad receptor pharmacology. Knowledge of DHE pharmacology largely comes from studies employing older methodologies. Objective: To assess DHE receptor activity using high-throughput methods to screen for functional ß-arrestin activity at G protein-coupled receptors (GPCRs). Methods: Functional receptor activities of DHE and sumatriptan succinate (both 10 µM) were screened against 168 GPCRs using the gpcrMAX assay. Agonist and antagonist effects were considered significant if receptor activity was >30% or inhibited by >50%, respectively. Radiolabeled ligand binding assays were performed for DHE (0.01-300 nM for 5-HT3 and 4E; 0.3-10,000 nM for 5-HT1B, α-adrenergic2B [i.e., α2B-adrenoceptor], D2, and D5) to assess specific binding to select receptors. Results: DHE (10 µM) exhibited agonist activity at α-adrenergic2B, CXC chemokine receptor 7 (CXCR7), dopamine (D)2/5, and 5-hydroxytryptamine (5-HT)1A/1B/2A/2C/5A receptors and antagonist activity at α-adrenergic1B/2A/2C (i.e., α1B/2A/2C-adrenoceptors), calcitonin receptor-receptor activity modifying protein 2 (CTR-RAMP2) or amylin 2 (AMY2), D1/3/4/5, and 5-HT1F receptors. Sumatriptan succinate (10 µM) exhibited agonist activity at the 5-HT1B/1E/1F/5A receptors. DHE demonstrated a half-maximal inhibitory concentration (IC50) of 149 nM at the 5-HT1F receptor and a half-maximal effective concentration (EC50) of 6 µM at the CXCR7 receptor. DHE did not bind to the 5-HT3 receptor at concentrations up to 300 nM and bound poorly to 5-HT4E and D5 receptors (IC50 of 230 and 370 nM, respectively). DHE bound strongly to the D2, 5-HT1B, and α-adrenergic2B receptors (IC50 of 0.47, 0.58, and 2.8 nM, respectively). Conclusion: By using a high-throughput ß-arrestin recruitment assay, this study confirmed the broad receptor profile of DHE and provided an update on DHE receptor pharmacology as it relates to migraine.

3.
Curr Pain Headache Rep ; 27(11): 765-774, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37792173

ABSTRACT

PURPOSE OF REVIEW: Historical evidence suggests a shared underlying etiology for migraine and gastrointestinal (GI) disorders that involves the gut-brain axis. Here we provide narrative review of recent literature on the gut-brain connection and migraine to emphasize the importance of tailoring treatment plans for patients with episodic migraine who experience GI comorbidities and symptoms. RECENT FINDINGS: Recent population-based studies report the prevalence of migraine and GI disorders as comorbidities as well as overlapping symptomology. American Headache Society (AHS) guidelines have integrated GI symptoms as part of migraine diagnostic criteria and recommend nonoral therapies for patients with GI symptoms or conditions. Nasal delivery is a recommended nonoral alternative; however, it is important to understand potential adverse events that may cause or worsen GI symptoms in some patients due to the site of drug deposition within the nasal cavity with some nasal therapies. Lastly, clinical perspectives emphasize the importance of identifying GI symptoms and comorbidities in patients with episodic migraine to best individualize migraine management. Support for an association between the gut-brain axis and migraine continues to prevail in recent literature; however, the relationship remains complex and not well elucidated. The presence of GI comorbidities and symptoms must be carefully considered when making treatment decisions for patients with episodic migraine.


Subject(s)
Migraine Disorders , Humans , Migraine Disorders/diagnosis , Migraine Disorders/epidemiology , Migraine Disorders/drug therapy , Brain , Headache/epidemiology , Comorbidity
4.
J Neuroimmune Pharmacol ; 10(3): 425-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25850894

ABSTRACT

Haloperidol is an effective antipsychotic drug for treatment of schizophrenia, but prolonged use can lead to debilitating side effects. To better understand the effects of long-term administration, we measured global metabolic changes in mouse brain following 3 mg/kg/day haloperidol for 28 days. These conditions lead to movement-related side effects in mice akin to those observed in patients after prolonged use. Brain tissue was collected following microwave tissue fixation to arrest metabolism and extracted metabolites were assessed using both liquid and gas chromatography mass spectrometry (MS). Over 300 unique compounds were identified across MS platforms. Haloperidol was found to be present in all test samples and not in controls, indicating experimental validity. Twenty-one compounds differed significantly between test and control groups at the p < 0.05 level. Top compounds were robust to analytical method, also being identified via partial least squares discriminant analysis. Four compounds (sphinganine, N-acetylornithine, leucine and adenosine diphosphate) survived correction for multiple testing in a non-parametric analysis using false discovery rate threshold < 0.1. Pathway analysis of nominally significant compounds (p < 0.05) revealed significant findings for sphingolipid metabolism (p = 0.015) and protein biosynthesis (p = 0.024). Altered sphingolipid metabolism is suggestive of disruptions to myelin. This interpretation is supported by our observation of elevated N-acetyl-aspartyl-glutamate in the haloperidol-treated mice (p = 0.004), a marker previously associated with demyelination. This study further demonstrates the utility of murine neurochemical metabolomics as a method to advance understanding of CNS drug effects.


Subject(s)
Antipsychotic Agents/adverse effects , Basal Ganglia Diseases/chemically induced , Brain/drug effects , Haloperidol/adverse effects , Lipid Metabolism/drug effects , Metabolic Networks and Pathways/drug effects , Sphingolipids/metabolism , Animals , Antipsychotic Agents/administration & dosage , Gas Chromatography-Mass Spectrometry , Haloperidol/administration & dosage , Male , Metabolomics , Mice , Mice, Inbred C57BL , Sphingolipids/biosynthesis
5.
Neuropharmacology ; 93: 237-42, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25698527

ABSTRACT

A number of studies have examined the ability of the endogenous cannabinoid anandamide to elicit Δ(9)-tetrahydrocannabinol (THC)-like subjective effects, as modeled through the THC discrimination paradigm. In the present study, we compared transgenic mice lacking fatty acid amide hydrolase (FAAH), the enzyme primarily responsible for anandamide catabolism, to wildtype counterparts in a THC discrimination procedure. THC (5.6 mg/kg) served as a discriminative stimulus in both genotypes, with similar THC dose-response curves between groups. Anandamide fully substituted for THC in FAAH knockout, but not wildtype, mice. Conversely, the metabolically stable anandamide analog O-1812 fully substituted in both groups, but was more potent in knockouts. The CB1 receptor antagonist rimonabant dose-dependently attenuated THC generalization in both groups and anandamide substitution in FAAH knockouts. Pharmacological inhibition of monoacylglycerol lipase (MAGL), the primary catabolic enzyme for the endocannabinoid 2-arachidonoylglycerol (2-AG), with JZL184 resulted in full substitution for THC in FAAH knockout mice and nearly full substitution in wildtypes. Quantification of brain endocannabinoid levels revealed expected elevations in anandamide in FAAH knockout mice compared to wildtypes and equipotent dose-dependent elevations in 2-AG following JZL184 administration. Dual inhibition of FAAH and MAGL with JZL195 resulted in roughly equipotent increases in THC-appropriate responding in both groups. While the notable similarity in THC's discriminative stimulus effects across genotype suggests that the increased baseline brain anandamide levels (as seen in FAAH knockout mice) do not alter THC's subjective effects, FAAH knockout mice are more sensitive to the THC-like effects of pharmacologically induced increases in anandamide and MAGL inhibition (e.g., JZL184).


Subject(s)
Amidohydrolases/deficiency , Cannabinoid Receptor Agonists/pharmacology , Discrimination, Psychological/drug effects , Dronabinol/pharmacology , Amidohydrolases/genetics , Analysis of Variance , Animals , Arachidonic Acids/metabolism , Benzodioxoles/pharmacology , Brain/drug effects , Brain/metabolism , Cannabinoid Receptor Antagonists/pharmacology , Carbamates/pharmacology , Discrimination, Psychological/physiology , Dose-Response Relationship, Drug , Endocannabinoids/metabolism , Enzyme Inhibitors/pharmacology , Glycerides/metabolism , Male , Mice , Mice, Knockout , Piperazines/pharmacology , Piperidines/pharmacology , Polyunsaturated Alkamides/metabolism , Pyrazoles/pharmacology , Rimonabant
6.
J Pharmacol Exp Ther ; 352(2): 195-207, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25398241

ABSTRACT

A growing body of evidence implicates endogenous cannabinoids as modulators of the mesolimbic dopamine system and motivated behavior. Paradoxically, the reinforcing effects of Δ(9)-tetrahydrocannabinol (THC), the primary psychoactive constituent of cannabis, have been difficult to detect in preclinical rodent models. In this study, we investigated the impact of THC and inhibitors of the endocannabinoid hydrolytic enzymes fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL) on operant responding for electrical stimulation of the medial forebrain bundle [intracranial self-stimulation (ICSS)], which is known to activate the mesolimbic dopamine system. These drugs were also tested in assays of operant responding for food reinforcement and spontaneous locomotor activity. THC and the MAGL inhibitor JZL184 (4-[bis(1,3-benzodioxol-5-yl)hydroxymethyl]-1-piperidinecarboxylic acid 4-nitrophenyl ester) attenuated operant responding for ICSS and food, and also reduced spontaneous locomotor activity. In contrast, the FAAH inhibitor PF-3845 (N-3-pyridinyl-4-[[3-[[5-(trifluoromethyl)-2-pyridinyl]oxy]phenyl]methyl]-1-piperidinecarboxamide) was largely without effect in these assays. Consistent with previous studies showing that combined inhibition of FAAH and MAGL produces a substantially greater cannabimimetic profile than single enzyme inhibition, the dual FAAH-MAGL inhibitor SA-57 (4-[2-(4-chlorophenyl)ethyl]-1-piperidinecarboxylic acid 2-(methylamino)-2-oxoethyl ester) produced a similar magnitude of ICSS depression as that produced by THC. ICSS attenuation by JZL184 was associated with increased brain levels of 2-arachidonoylglycerol (2-AG), whereas peak effects of SA-57 were associated with increased levels of both N-arachidonoylethanolamine (anandamide) and 2-AG. The cannabinoid receptor type 1 receptor antagonist rimonabant, but not the cannabinoid receptor type 2 receptor antagonist SR144528, blocked the attenuating effects of THC, JZL184, and SA-57 on ICSS. Thus, THC, MAGL inhibition, and dual FAAH-MAGL inhibition not only reduce ICSS, but also decrease other reinforced and nonreinforced behaviors.


Subject(s)
Dronabinol/pharmacology , Endocannabinoids/metabolism , Enzyme Inhibitors/pharmacology , Medial Forebrain Bundle/drug effects , Reinforcement, Psychology , Self Stimulation , Amidohydrolases/antagonists & inhibitors , Animals , Behavior, Animal/drug effects , Benzodioxoles/pharmacology , Biphenyl Compounds/pharmacology , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Electric Stimulation , Male , Medial Forebrain Bundle/enzymology , Medial Forebrain Bundle/metabolism , Mice, Inbred C57BL , Monoacylglycerol Lipases/antagonists & inhibitors , Motor Activity/drug effects , Piperidines/pharmacology , Pyridinium Compounds/pharmacology
7.
Eur J Pharmacol ; 737: 97-105, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24858366

ABSTRACT

The mechanism through which marijuana produces its psychoactive effects is Δ(9)-tetrahydrocannabinol (THC)-induced activation of cannabinoid CB1 receptors. These receptors are normally activated by endogenous lipids, including anandamide and 2-arachidonoyl glycerol (2-AG). A logical "first step" in determination of the role of these endocannabinoids in THC׳s psychoactive effects is to investigate the degree to which pharmacologically induced increases in anandamide and/or 2-AG concentrations through exogenous administration and/or systemic administration of inhibitors of their metabolism, fatty acid amide hydrolase (FAAH) or monoacylglycerol lipase (MAGL), respectively, share THC׳s discriminative stimulus effects. To this end, adult male mice and rats were trained to discriminate THC (5.6 and 3mg/kg, respectively). In Experiment 1, exogenous administration of anandamide or 2-AG did not substitute for THC in mice nor was substitution enhanced by co-administration of the FAAH or MAGL inhibitors, URB597 and N-arachidonyl maleimide (NAM), respectively. Significant decreases in responding may have prevented assessment of adequate endocannabinoid doses. In mice trained at higher baseline response rates (Experiment 2), the FAAH inhibitor PF3845 (10mg/kg) enhanced anandamide substitution for THC without producing effects of its own. The MAGL inhibitor JZL184 increased brain levels of 2-AG in vitro and in vivo, increased THC-like responding without co-administration of 2-AG. In rats, neither URB597 nor JZL184 engendered significant THC-appropriate responding, but co-administration of these two enzyme inhibitors approached full substitution. The present results highlight the complex interplay between anandamide and 2-AG and suggest that endogenous increases of both endocannabinoids are most effective in elicitation of THC-like discriminative stimulus effects.


Subject(s)
Arachidonic Acids/metabolism , Discrimination, Psychological/drug effects , Dronabinol/pharmacology , Endocannabinoids/metabolism , Glycerides/metabolism , Polyunsaturated Alkamides/metabolism , Amidohydrolases/antagonists & inhibitors , Animals , Brain/drug effects , Brain/metabolism , Enzyme Inhibitors/pharmacology , Male , Mice , Monoacylglycerol Lipases/antagonists & inhibitors , Rats , Receptor, Cannabinoid, CB1/metabolism
8.
Metabolomics ; 9(2): 392-402, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23554582

ABSTRACT

Methamphetamine (MA) is an illegal stimulant drug of abuse with serious negative health consequences. The neurochemical effects of MA have been partially characterized, with a traditional focus on classical neurotransmitter systems. However, these directions have not yet led to novel drug treatments for MA abuse or toxicity. As an alternative approach, we describe here the first application of metabolomics to investigate the neurochemical consequences of MA exposure in the rodent brain. We examined single exposures at 3 mg/kg and repeated exposures at 3 mg/kg over 5 days in eight common inbred mouse strains. Brain tissue samples were assayed using high-throughput gas and liquid chromatography mass spectrometry, yielding quantitative data on >300 unique metabolites. Association testing and false discovery rate control yielded several metabolome-wide significant associations with acute MA exposure, including compounds such as lactate (p = 4.4 × 10-5, q = 0.013), tryptophan (p = 7.0 × 10-4, q = 0.035) and 2-hydroxyglutarate (p = 1.1 × 10-4, q = 0.022). Secondary analyses of MA-induced increase in locomotor activity showed associations with energy metabolites such as succinate (p = 3.8 × 10-7). Associations specific to repeated (5 day) MA exposure included phosphocholine (p = 4.0 × 10-4, q = 0.087) and ergothioneine (p = 3.0 × 10-4, q = 0.087). Our data appear to confirm and extend existing models of MA action in the brain, whereby an initial increase in energy metabolism, coupled with an increase in behavioral locomotion, gives way to disruption of mitochondria and phospholipid pathways and increased endogenous antioxidant response. Our study demonstrates the power of comprehensive MS-based metabolomics to identify drug-induced changes to brain metabolism and to develop neurochemical models of drug effects.

9.
Drug Alcohol Depend ; 126(3): 316-23, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22776442

ABSTRACT

BACKGROUND: Use of synthetic "marijuana" has increased in recent years, produced adverse effects and prompted the temporary DEA ban of five specific cannabinoid analogs, including JWH-018. The objectives of the current study include determining the chemical content of the herbal product, Buzz, assessing its behavioral effects upon inhalation exposure to mice, determining whether CB(1) receptors mediate its pharmacological activity, and ascertaining its biodisposition in blood and various organs. METHODS: Using a nose-only exposure system, mice were exposed to smoke produced from combustion of an herbal incense product, Buzz, which contained 5.4% JWH-018. Cannabimimetic effects following smoke exposure were evaluated using the tetrad procedure, consisting of the following indices: hypomotility, antinociception, catalepsy, and hypothermia. Additionally, blood and tissues were collected for JWH-018 quantification. RESULTS: Inhalation exposure to Buzz produced dose-related tetrad effects similar to marijuana as well as dose-related increased levels of JWH-018 in the blood, brain, heart, kidney, liver, lung, and spleen. The behavioral effects were blocked by rimonabant, a CB(1) receptor antagonist. Effects produced by Buzz were similar in magnitude and time-course to those produced by marijuana, though equipotent doses of Buzz and marijuana yielded considerably lower brain levels of JWH-018 than THC for the respective materials. CONCLUSIONS: Inhalation exposure to a product containing JWH-018 penetrates into the brain and other organs and produces CB(1) receptor-mediated behavioral pharmacological effects in mice. The increased potency of JWH-018 compared to THC, the variable amount of drug added to various herbal products, and unknown toxicity, undoubtedly contribute to public health risks of synthetic cannabinoids.


Subject(s)
Cannabinoids/pharmacology , Illicit Drugs/pharmacology , Animals , Behavior, Animal/drug effects , Body Temperature/drug effects , Catalepsy/chemically induced , Dose-Response Relationship, Drug , Illicit Drugs/pharmacokinetics , Indoles/pharmacokinetics , Indoles/pharmacology , Inhalation Exposure , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Naphthalenes/pharmacokinetics , Naphthalenes/pharmacology , Nociception/drug effects
10.
Behav Pharmacol ; 22(5-6): 480-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21712709

ABSTRACT

Δ(9)-Tetrahydrocannabinol (THC) discrimination in rodents is a behavioral assay that has been used to probe differences among classes of cannabinoids in rats. The purpose of this study was to determine whether traditional and anandamide-like cannabinoids were distinguishable in cannabinoid discrimination procedures in mice. Male mice were trained to discriminate 30 mg/kg THC or 70 mg/kg methanandamide from vehicle in a two-lever milk-reinforced drug discrimination procedure. After acquisition, agonist tests with THC, methanandamide, CP 55940, and anandamide were conducted, as were antagonism tests with rimonabant. Substitution (agonism) and antagonism tests were also carried out in female mice trained to discriminate THC. THC and CP 55940 fully substituted in THC-trained mice of both sexes. Further, THC substitution was rimonabant reversible. In contrast, mice injected with methanandamide or anandamide failed to respond substantially on the THC lever, even up to doses that decreased overall responding. In methanandamide-trained mice, methanandamide fully generalized to the methanandamide training dose. Rimonabant did not reverse this generalization. Although THC, CP 55940, and anandamide also increased responding on the methanandamide lever, the magnitude of substitution was less than for methanandamide. These results suggest incomplete overlap in the underlying mechanisms mediating endocannabinoid pharmacology and marijuana intoxication. Further, they suggest that methanandamide discrimination may involve a non-CB(1) receptor mechanism that is particularly prominent at higher doses.


Subject(s)
Arachidonic Acids/pharmacology , Cyclohexanols/pharmacology , Discrimination Learning/drug effects , Dronabinol/pharmacology , Animals , Arachidonic Acids/administration & dosage , Cyclohexanols/administration & dosage , Dose-Response Relationship, Drug , Dronabinol/administration & dosage , Female , Generalization, Stimulus/drug effects , Male , Mice , Mice, Inbred C57BL , Piperidines/pharmacology , Psychotropic Drugs/administration & dosage , Psychotropic Drugs/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Rimonabant , Sex Factors
11.
Eur J Pharmacol ; 656(1-3): 63-7, 2011 Apr 10.
Article in English | MEDLINE | ID: mdl-21300050

ABSTRACT

The endogenous cannabinoid system has been noted for its therapeutic potential, as well as the psychoactivity of cannabinoids such as Δ9-tetrahydrocannabinol (THC). However, less is known about the psychoactivity of anandamide (AEA), an endocannabinoid ligand. Thus, the goals of this study were to establish AEA as a discriminative stimulus in transgenic mice lacking fatty acid amide hydrolase (i.e., FAAH -/- mice unable to rapidly metabolize AEA), evaluate whether THC or oleamide, a fatty acid amide, produced AEA-like responding, and assess for CB(1) mediation of AEA's discriminative stimulus. Mice readily discriminated between 6mg/kg AEA and vehicle in a two-lever drug discrimination task. AEA dose-dependently generalized to itself. THC elicited full AEA-like responding, whereas oleamide failed to substitute. The CB(1) antagonist rimonabant attenuated AEA- and THC-induced AEA-appropriate responding, demonstrating CB(1) mediation of AEA's discriminative stimulus. These findings suggest that, in the absence of FAAH, AEA produces intoxication comparable to THC, and consequently to marijuana.


Subject(s)
Amidohydrolases/deficiency , Amidohydrolases/genetics , Arachidonic Acids/pharmacology , Discrimination, Psychological/drug effects , Dronabinol/pharmacology , Gene Knockout Techniques , Polyunsaturated Alkamides/pharmacology , Animals , Behavior, Animal/drug effects , Endocannabinoids , Male , Mice , Oleic Acids/pharmacology
12.
Eur J Pharmacol ; 651(1-3): 96-105, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21114999

ABSTRACT

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


Subject(s)
Dronabinol/analogs & derivatives , Pyrans/chemistry , Pyrans/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Animals , Cyclic AMP/metabolism , Dronabinol/chemistry , Dronabinol/metabolism , Dronabinol/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Male , Mice , Pyrans/metabolism , Receptor, Cannabinoid, CB1/metabolism , Vas Deferens/drug effects , Vas Deferens/metabolism
13.
Neuropsychopharmacology ; 36(3): 616-26, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21107309

ABSTRACT

Neurocognitive deficits are a core feature of schizophrenia and, therefore, represent potentially critical outcome variables for assessing antipsychotic treatment response. We performed genome-wide association studies (GWAS) with 492K single nucleotide polymorphisms (SNPs) in a sample of 738 patients with schizophrenia from the Clinical Antipsychotic Trials of Intervention Effectiveness study. Outcome variables consisted of a neurocognitive battery administered at multiple time points over an 18-month period, measuring processing speed, verbal memory, vigilance, reasoning, and working memory domains. Genetic mediation of improvements in each of these five domains plus a composite neurocognitive measure was assessed for each of five antipsychotics (olanzapine, perphenazine, quetiapine, risperidone, and ziprasidone). Six SNPs achieved genome-wide significance using a pre-specified threshold that ensures, on average, only 1 in 10 findings is a false discovery. These six SNPs were located within, or in close proximity to, genes EHF, SLC26A9, DRD2, GPR137B, CHST8, and IL1A. The more robust findings, that is those significant across multiple neurocognitive domains and having adjacent SNPs showing evidence for association, were rs286913 at the EHF gene (p-value 6.99 × 10(-8), q-value 0.034, mediating the effects of ziprasidone on vigilance), rs11240594 at SLC26A9 (p-value 1.4 × 10(-7), q-value 0.068, mediating the effects of olanzapine on processing speed), and rs11677416 at IL1A (p-value 6.67 × 10(-7), q-value 0.081, mediating the effects of olanzapine on working memory). This study has generated several novel candidate genes for antipsychotic response. However, our findings will require replication and functional validation. To facilitate replication efforts, we provide all GWAS p-values for download.


Subject(s)
Antipsychotic Agents/therapeutic use , Cognition Disorders , Genome-Wide Association Study , Pharmacogenetics , Schizophrenia , Adult , Antiporters/genetics , Antipsychotic Agents/pharmacology , Attention/drug effects , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Cognition Disorders/genetics , Female , Follow-Up Studies , Humans , Interleukin-1alpha/genetics , Male , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Neuropsychological Tests , Polymorphism, Single Nucleotide/genetics , Schizophrenia/complications , Schizophrenia/drug therapy , Schizophrenia/genetics , Sulfate Transporters , Time Factors , Transcription Factors/genetics
14.
J Pharmacol Exp Ther ; 334(3): 1087-95, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20576796

ABSTRACT

Bupropion is an atypical antidepressant that also has utility as a smoking cessation aid. Hydroxybupropions are major metabolites of bupropion and are believed to contribute to antidepressant and perhaps smoking cessation activities. Because bupropion metabolism is more similar in humans and mice than in humans and rats, the present study investigated effects of hydroxybupropion enantiomers in mouse behavioral models measuring various aspects of nicotine dependence. Bupropion and (2S,3S)-hydroxybupropion, but not (2R,3R)-hydroxybupropion, significantly decreased the development of nicotine reward as measured in the conditioned place preference and withdrawal paradigm in mice. Bupropion and both of its metabolites reversed affective and somatic withdrawal signs in nicotine-dependent mice, but the (2S,3S)-hydroxymetabolite had higher potency. Bupropion and (2S,3S)-, but not (2R,3R)-hydroxybupropion, produced partial substitution for nicotine in drug discrimination tests. Our findings support the hypothesis that the effects of bupropion on measures of nicotine dependence reflect actions of bupropion itself, its hydroxymetabolites, or a combination and suggest that the (2S,3S)-hydroxy isomer is the most active principle, making it a potentially better drug candidate for smoking cessation than bupropion.


Subject(s)
Behavior, Animal/drug effects , Bupropion/analogs & derivatives , Bupropion/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Tobacco Use Disorder/drug therapy , Tobacco Use Disorder/psychology , Animals , Anxiety/psychology , Bupropion/pharmacokinetics , Conditioning, Operant/drug effects , Discrimination Learning/drug effects , Discrimination, Psychological/drug effects , Dopamine Uptake Inhibitors/pharmacokinetics , Dose-Response Relationship, Drug , Emotions/drug effects , Hydroxylation , Hyperalgesia/psychology , Mice , Mice, Inbred ICR , Nicotine/administration & dosage , Nicotine/pharmacology , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/pharmacology , Reward , Substance Withdrawal Syndrome/prevention & control , Substance Withdrawal Syndrome/psychology
15.
Psychopharmacology (Berl) ; 210(2): 275-84, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20354680

ABSTRACT

RATIONALE: Salvinorin A, the primary psychoactive derivative of the hallucinogenic herb Salvia divinorum, is a potent and highly selective kappa-opioid receptor (KOR) agonist. Several recent studies, however, have suggested endocannabinoid system mediation of some of its effects. OBJECTIVES: This study represents a systematic examination of this hypothesis. METHODS: Salvinorin A was isolated from S. divinorum and was evaluated in a battery of in vitro and in vivo procedures designed to detect cannabinoid activity, including CB(1) receptor radioligand and [(35)S]GTPgammaS binding, calcium flux assay, in vivo cannabinoid screening tests, and drug discrimination. RESULTS: Salvinorin A did not bind to nor activate CB(1) receptors. In vivo salvinorin A produced pronounced hypolocomotion and antinociception (and to a lesser extent, hypothermia). These effects were blocked by the selective KOR antagonist, JDTic, but not by the CB(1) receptor antagonist rimonabant. Interestingly, however, rimonabant attenuated KOR activation stimulated by U69,593 in a [(35)S]GTPgammaS assay. Salvinorin A did not substitute for Delta(9)-tetrahydrocannabinol (THC) in mice trained to discriminate THC. CONCLUSIONS: These findings suggest that similarities in the pharmacological effects of salvinorin A and those of cannabinoids are mediated by its activation of KOR rather than by any direct action of salvinorin A on the endocannabinoid system. Further, the results suggest that rimonabant reversal of salvinorin A effects in previous studies may be explained in part by rimonabant attenuation of KOR activation.


Subject(s)
Diterpenes, Clerodane/pharmacology , Receptors, Opioid, kappa/agonists , Animals , Calcium/metabolism , Cannabinoid Receptor Modulators/physiology , Discrimination Learning , Dronabinol/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Hypothermia/chemically induced , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Pain Measurement , Piperidines/pharmacology , Pyrazoles/pharmacology , Radioligand Assay , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Receptors, Opioid, kappa/antagonists & inhibitors , Rimonabant , Salvia , Tetrahydroisoquinolines/pharmacology
16.
Proc Natl Acad Sci U S A ; 106(48): 20270-5, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19918051

ABSTRACT

Delta(9)-tetrahydrocannabinol (THC), the psychoactive component of marijuana, and other direct cannabinoid receptor (CB1) agonists produce a number of neurobehavioral effects in mammals that range from the beneficial (analgesia) to the untoward (abuse potential). Why, however, this full spectrum of activities is not observed upon pharmacological inhibition or genetic deletion of either fatty acid amide hydrolase (FAAH) or monoacylglycerol lipase (MAGL), enzymes that regulate the two major endocannabinoids anandamide (AEA) and 2-arachidonoylglycerol (2-AG), respectively, has remained unclear. Here, we describe a selective and efficacious dual FAAH/MAGL inhibitor, JZL195, and show that this agent exhibits broad activity in the tetrad test for CB1 agonism, causing analgesia, hypomotilty, and catalepsy. Comparison of JZL195 to specific FAAH and MAGL inhibitors identified behavioral processes that were regulated by a single endocannabinoid pathway (e.g., hypomotility by the 2-AG/MAGL pathway) and, interestingly, those where disruption of both FAAH and MAGL produced additive effects that were reversed by a CB1 antagonist. Falling into this latter category was drug discrimination behavior, where dual FAAH/MAGL blockade, but not disruption of either FAAH or MAGL alone, produced THC-like responses that were reversed by a CB1 antagonist. These data indicate that AEA and 2-AG signaling pathways interact to regulate specific behavioral processes in vivo, including those relevant to drug abuse, thus providing a potential mechanistic basis for the distinct pharmacological profiles of direct CB1 agonists and inhibitors of individual endocannabinoid degradative enzymes.


Subject(s)
Amidohydrolases/metabolism , Cannabinoid Receptor Modulators/metabolism , Carbamates/pharmacology , Endocannabinoids , Monoacylglycerol Lipases/metabolism , Piperazines/pharmacology , Piperidines/pharmacology , Amidohydrolases/antagonists & inhibitors , Animals , Arachidonic Acids/metabolism , Carbamates/chemical synthesis , Carboxylic Ester Hydrolases/metabolism , Glycerides/metabolism , Mice , Molecular Structure , Monoacylglycerol Lipases/antagonists & inhibitors , Motor Activity/drug effects , Pain Measurement , Piperazines/chemical synthesis , Piperidines/chemical synthesis , Polyunsaturated Alkamides/metabolism
17.
Pharmacol Biochem Behav ; 94(1): 114-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19660491

ABSTRACT

The introduction of transgenic and knockout mice has shaped new interest in developing novel and modified behavioral methods for mice that evaluate the various manifestations of nicotine withdrawal syndromes. This study assessed the disruption of operant baselines during drug withdrawal, an established rat model of nicotine dependence, in mice. Subjects were trained to lever press for food reinforcement during daily operant sessions. After stable operant baselines were established, mice were implanted with osmotic minipumps containing 0 (saline), 6, 12, 24, or 48 mg/kg/day nicotine base. Operant responding was assessed for disruptions in daily sessions throughout the experiment. Somatic signs of withdrawal were assessed after the operant session on day 7, following administration of mecamylamine (1 mg/kg), and on days 12, 13, and 14, following spontaneous removal of nicotine. Spontaneous removal of nicotine increased somatic signs of withdrawal but did not disrupt operant responding. Mecamylamine failed to produce signs of precipitated withdrawal in either procedure. This study demonstrated nicotine dependence in mice during spontaneous removal of nicotine. Moreover, since signs of behavioral withdrawal (i.e. disruptions in operant response rates) were not observed, these findings suggest the importance of considering differences in the apparent manifestations of withdrawal syndromes while evaluating nicotine dependence.


Subject(s)
Behavior, Animal/drug effects , Mecamylamine/pharmacology , Nicotine/adverse effects , Nicotinic Agonists/adverse effects , Nicotinic Antagonists/pharmacology , Substance Withdrawal Syndrome/diagnosis , Animals , Conditioning, Operant , Dose-Response Relationship, Drug , Habituation, Psychophysiologic , Infusion Pumps, Implantable , Infusions, Subcutaneous , Male , Mecamylamine/administration & dosage , Mice , Mice, Inbred ICR , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Nicotinic Antagonists/administration & dosage , Reinforcement, Psychology , Substance Withdrawal Syndrome/etiology , Time Factors
18.
Eur J Pharmacol ; 615(1-3): 102-7, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19470387

ABSTRACT

Primarily, rats have served as subjects in Delta(9)-tetrahydrocannabinol's (THC) discrimination studies although other species such as monkeys and pigeons have been used. While the introduction of the knockout and transgenic mice has vastly stimulated the study of the discriminative stimulus effects of drugs there is only a single published report of mice trained to discriminate THC. Thus, this study extended those results by providing a systematic replication that THC serves as an effective discriminative stimulus in mice and by further investigating the mechanisms of action involved in the THC discrimination model in the mouse. Male C57BL/6J mice were trained to discriminate 10 mg/kg THC from vehicle in 2-lever drug discrimination. THC fully and dose dependently substituted for itself. Cannabinoid indoles, except one with low cannabinoid CB(1) receptor affinity, substituted for THC. Anandamide failed to substitute for THC when administered alone but completely substituted when administered with the non-specific fatty acid amide hydrolase inhibitor, phenylmethylsulphonyl fluoride. As expected, nicotine failed to substitute for THC. Lastly, the cannabinoid CB(1) receptor antagonist rimonabant blocked THC's discriminative stimulus effects. Taken together these studies demonstrate THC's ability to produce discriminative stimulus effects as well as demonstrate its pharmacological specificity and mechanism of action in a two-lever drug discrimination mouse model.


Subject(s)
Discrimination, Psychological , Dronabinol/pharmacology , Hallucinogens/pharmacology , Amidohydrolases/antagonists & inhibitors , Animals , Arachidonic Acids/pharmacology , Discrimination Learning/drug effects , Dose-Response Relationship, Drug , Endocannabinoids , Male , Mice , Mice, Inbred C57BL , Nicotine/pharmacology , Phenylmethylsulfonyl Fluoride/pharmacology , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Rimonabant
19.
Drug Alcohol Depend ; 103(3): 124-30, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19443130

ABSTRACT

Replacement therapy with the synthetic mu-opioid agonist methadone is an efficacious treatment for opioid abuse. While much is known about methadone's pharmacology, its discriminative stimulus properties remain largely unexplored. The present study sought to establish methadone discrimination in rats. Moreover, some research suggests that route of administration alters the discriminative stimulus of methadone. Thus, the present study also compared intraperitoneal (i.p.) and subcutaneous (s.c.) routes of administration. Male Sprague-Dawley rats were trained to discriminate 3.0mg/kg methadone (i.p.) from vehicle in a two-lever discrimination procedure. Generalization tests were conducted with a variety of compounds administered i.p. and s.c. Methadone fully substituted for itself, yielding ED(50)s of 1.5mg/kg (i.p.) and 0.2mg/kg (s.c.). Naltrexone (i.p.), an opioid antagonist produced a dose-dependent reduction in methadone-appropriate responding. The methadone stereoisomers fully substituted for methadone when given s.c.; however, when administered i.p., (+) and (-) methadone produced partial and no substitution, respectively. Heroin fully generalized to methadone regardless of administration route, while morphine fully substituted when given s.c., but not i.p. The kappa-agonist U50-488 failed to generalize to methadone with either route of administration. These results demonstrated that methadone's discriminative stimulus is mediated through mu-opioid receptor activity and is similar to that of commonly abused opioids (heroin, morphine). Additionally, route of administration produced differential results for many of the drugs tested, suggesting decreased drug bioavailability following i.p. administration due to hepatic first pass metabolism. Taken together, these results suggest that methadone's shared subjective effects with abused opioids, as well as its unique metabolic properties contribute to its efficacy in opioid maintenance therapy.


Subject(s)
Choice Behavior/drug effects , Discrimination, Psychological/drug effects , Methadone/administration & dosage , Methadone/pharmacology , Pharmaceutical Vehicles/administration & dosage , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/administration & dosage , Analgesics, Non-Narcotic/administration & dosage , Animals , Conditioning, Operant , Heroin/administration & dosage , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Morphine/administration & dosage , Naltrexone/administration & dosage , Narcotic Antagonists/administration & dosage , Narcotics/administration & dosage , Narcotics/pharmacology , Rats , Rats, Sprague-Dawley , Stereoisomerism
20.
Pharmacol Biochem Behav ; 91(1): 150-4, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18644403

ABSTRACT

The Lewis (LEW) strain of rat appears more sensitive to nicotine than other strains in self-administration, conditioned place preference, and drug discrimination behavioral studies. The present study sought to further evaluate the behavioral effects of chronic nicotine treatment in the LEW strain by assessing spontaneous activity, which has consistently revealed sensitization to chronic nicotine administration in Sprague Dawley (SD) rats. High active and low active male and female LEW rats (N=8 per group) were treated twice daily with either nicotine (0.4 mg/kg, sc) or vehicle for 14 consecutive days. Regardless of baseline activity level or sex, spontaneous activity was significantly decreased, compared to saline-treated rats, after a single nicotine injection. However, spontaneous activity increased in both low- and high-activity rats (both sexes) over the two weeks of nicotine administration to levels that were significantly higher than saline-treated rats. Based on these findings, acute and chronic nicotine administration had greater suppressive and enhancing effects on spontaneous activity in LEW rats compared to other strains of rats previously studied. These results further clarify the behavioral sensitivity of the LEW strain of rat to nicotine exposure and lend credence to the role of genetics in the individual susceptibility to nicotine dependence.


Subject(s)
Motor Activity/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Analysis of Variance , Animals , Data Interpretation, Statistical , Female , Male , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Rats , Rats, Inbred Lew
SELECTION OF CITATIONS
SEARCH DETAIL
...