Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Cachexia Sarcopenia Muscle ; 10(4): 844-859, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31035309

RESUMEN

BACKGROUND: Muscle wasting, anorexia, and metabolic dysregulation are common side-effects of cytotoxic chemotherapy, having a dose-limiting effect on treatment efficacy, and compromising quality of life and mortality. Extracts of Cannabis sativa, and analogues of the major phytocannabinoid Δ9-tetrahydrocannabinol, have been used to ameliorate chemotherapy-induced appetite loss and nausea for decades. However, psychoactive side-effects limit their clinical utility, and they have little efficacy against weight loss. We recently established that the non-psychoactive phytocannabinoid cannabigerol (CBG) stimulates appetite in healthy rats, without neuromotor side-effects. The present study assessed whether CBG attenuates anorexia and/or other cachectic effects induced by the broad-spectrum chemotherapy agent cisplatin. METHODS: An acute cachectic phenotype was induced in adult male Lister-hooded rats by 6 mg/kg (i.p.) cisplatin. In total 66 rats were randomly allocated to groups receiving vehicle only, cisplatin only, or cisplatin and 60 or 120 mg/kg CBG (po, b.i.d.). Feeding behavior, bodyweight and locomotor activity were recorded for 72 hours, at which point rats were sacrificed for post-mortem analyses. Myofibre atrophy, protein synthesis and autophagy dysregulation were assessed in skeletal muscle, plasma metabolic profiles were obtained by untargeted 1H-NMR metabonomics, and levels of endocannabinoid-like lipoamines quantified in plasma and hypothalami by targeted HPLC-MS/MS lipidomics. RESULTS: CBG (120 mg/kg) modestly increased food intake, predominantly at 36-60hrs (p<0.05), and robustly attenuated cisplatin-induced weight loss from 6.3% to 2.6% at 72hrs (p<0.01). Cisplatin-induced skeletal muscle atrophy was associated with elevated plasma corticosterone (3.7 vs 13.1ng/ml, p<0.01), observed selectively in MHC type IIx (p<0.05) and IIb (p<0.0005) fibres, and was reversed by pharmacological rescue of dysregulated Akt/S6-mediated protein synthesis and autophagy processes. Plasma metabonomic analysis revealed cisplatin administration produced a wide-ranging aberrant metabolic phenotype (Q2Y=0.5380, p=0.001), involving alterations to glucose, amino acid, choline and lipid metabolism, citrate cycle, gut microbiome function, and nephrotoxicity, which were partially normalized by CBG treatment (Q2Y=0.2345, p=0.01). Lipidomic analysis of hypothalami and plasma revealed extensive cisplatin-induced dysregulation of central and peripheral lipoamines (29/79 and 11/26 screened, respectively), including reversible elevations in systemic N-acyl glycine concentrations which were negatively associated with the anti-cachectic effects of CBG treatment. CONCLUSIONS: Endocannabinoid-like lipoamines may have hitherto unrecognized roles in the metabolic side-effects associated with chemotherapy, with the N-acyl glycine subfamily in particular identified as a potential therapeutic target and/or biomarker of anabolic interventions. CBG-based treatments may represent a novel therapeutic option for chemotherapy-induced cachexia, warranting investigation in tumour-bearing cachexia models.


Asunto(s)
Caquexia/inducido químicamente , Cannabinoides/uso terapéutico , Hipotálamo/efectos de los fármacos , Espectroscopía de Resonancia Magnética/métodos , Animales , Cannabinoides/farmacología , Modelos Animales de Enfermedad , Humanos , Masculino , Proyectos Piloto , Ratas
2.
Epilepsy Behav ; 70(Pt B): 319-327, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28190698

RESUMEN

The isolation and identification of the discrete plant cannabinoids in marijuana revived interest in analyzing historical therapeutic claims made for cannabis in clinical case studies and anecdotes. In particular, sources as old as the 11th and 15th centuries claimed efficacy for crude marijuana extracts in the treatment of convulsive disorders, prompting a particularly active area of preclinical research into the therapeutic potential of plant cannabinoids in epilepsy. Since that time, a large body of literature has accumulated describing the effects of several of the >100 individual plant cannabinoids in preclinical models of seizures, epilepsy, epileptogenesis, and epilepsy-related neuroprotection. We surveyed the literature for relevant reports of such plant cannabinoid effects and critically reviewed their findings. We found that acute CB1R agonism in simple models of acute seizures in rodents typically produces anti-convulsant effects whereas CB1R antagonists exert converse effects in the same models. However, when the effects of such ligands are examined in more complex models of epilepsy, epileptogenesis and neuroprotection, a less simplistic narrative emerges. Here, the complex interactions between (i) brain regions involved in a given model, (ii) relative contributions of endocannabinoid signaling to modulation of synaptic transmission in such areas, (iii) multi-target effects, (iv) cannabinoid type 1 and type 2 receptor signaling interactions and, (v) timing, (vi) duration and (vii) localization of ligand administration suggest that there is both anti-epileptic therapeutic potential and a pro-epileptic risk in up- and down-regulation of endocannabinoid signaling in the central nervous system. Factors such receptor desensitization and specific pharmacology of ligands used (e.g. full vs partial agonists and neutral antagonists vs inverse agonists) also appear to play an important role in the effects reported. Furthermore, the effects of several plant cannabinoids, most notably cannabidiol (CBD) and cannabidavarin (CBDV), in models of seizures, epilepsy, epileptogenesis, and neuroprotection are less ambiguous, and consistent with reports of therapeutically beneficial effects of these compounds in clinical studies. However, continued paucity of firm information regarding the therapeutic molecular mechanism of CBD/CBDV highlights the continued need for research in this area in order to identify as yet under-exploited targets for drug development and raise our understanding of treatment-resistant epilepsies. The recent reporting of positive results for cannabidiol treatment in two Phase III clinical trials in treatment-resistant epilepsies provides pivotal evidence of clinical efficacy for one plant cannabinoid in epilepsy. Moreover, risks and/or benefits associated with the use of unlicensed Δ9-THC containing marijuana extracts in pediatric epilepsies remain poorly understood. Therefore, in light of these paradigm-changing clinical events, the present review's findings aim to drive future drug development for newly-identified targets and indications, identify important limitations of animal models in the investigation of plant cannabinoid effects in the epilepsies, and focuses future research in this area on specific, unanswered questions regarding the complexities of endocannabinoid signaling in epilepsy. This article is part of a Special Issue titled Cannabinoids and Epilepsy.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Cannabinoides/uso terapéutico , Modelos Animales de Enfermedad , Epilepsia/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Convulsiones/tratamiento farmacológico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Cannabidiol/uso terapéutico , Cannabis , Dronabinol/uso terapéutico , Combinación de Medicamentos , Epilepsia/fisiopatología , Humanos , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/antagonistas & inhibidores , Convulsiones/fisiopatología
3.
Behav Pharmacol ; 28(4): 280-284, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28125508

RESUMEN

Nonpsychoactive phytocannabinoids (pCBs) from Cannabis sativa may represent novel therapeutic options for cachexia because of their pleiotropic pharmacological activities, including appetite stimulation. We have recently shown that purified cannabigerol (CBG) is a novel appetite stimulant in rats. As standardized extracts from Cannabis chemotypes dominant in one pCB [botanical drug substances (BDSs)] often show greater efficacy and/or potency than purified pCBs, we investigated the effects of a CBG-rich BDS, devoid of psychoactive [INCREMENT]-tetrahydrocannabinol, on feeding behaviour. Following a 2 h prefeed satiation procedure, 16 male Lister-hooded rats were administered CBG-BDS (at 30-240 mg/kg) or vehicle. Food intake, meal pattern microstructure and locomotor activity were recorded over 2 h. The total food intake was increased by 120 and 240 mg/kg CBG-BDS (1.53 and 1.36 g, respectively, vs. 0.56 g in vehicle-treated animals). Latency to feeding onset was dose dependently decreased at all doses, and 120 and 240 mg/kg doses increased both the number of meals consumed and the cumulative size of the first two meals. No significant effect was observed on ambulatory activity or rearing behaviour. CBG-BDS is a novel appetite stimulant, which may have greater potency than purified CBG, despite the absence of [INCREMENT]-tetrahydrocannabinol in the extract.


Asunto(s)
Cannabinoides/farmacología , Cannabis/química , Hiperfagia/inducido químicamente , Extractos Vegetales/farmacología , Animales , Estimulantes del Apetito/administración & dosificación , Estimulantes del Apetito/farmacología , Caquexia/tratamiento farmacológico , Cannabinoides/administración & dosificación , Relación Dosis-Respuesta a Droga , Conducta Alimentaria/efectos de los fármacos , Locomoción , Masculino , Extractos Vegetales/administración & dosificación , Ratas
4.
Br J Pharmacol ; 173(22): 3196-3207, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27545646

RESUMEN

BACKGROUND AND PURPOSE: Essential tremor (ET) is a neurological disorder with unknown aetiology. Its symptoms include cerebellar motor disturbances, cognitive and personality changes, hearing and olfactory deficits. Hyperactivity of excitotoxic cerebellar climbing fibres may underlie essential tremor and has been induced in rodents by systemic harmaline administration. Cannabinoid (CB) receptor agonists can cause motor disturbances; although, there are also anecdotal reports of therapeutic benefits of cannabis in motor disorders. We set out to establish the effects of CB receptor agonism and antagonism on an established rodent model of ET using a battery of accepted behaviour assays in order to determine the risk and therapeutic potential of modulating the endocannabinoid system in ET. EXPERIMENTAL APPROACH: Behavioural effects of systemic treatment with a CB receptor agonist (0.1, 0.5 and 1 mg kg-1 WIN55, 212-2) or two CB1 receptor antagonists (1 mg kg-1  AM251 and 10 mg kg-1 rimonabant) on tremor induced in rats by harmaline (30 mg kg-1 ; i.p.), were assessed using tremor scoring, open field, rotarod, grip and gait tests. KEY RESULTS: Overall, harmaline induced robust tremor that was typically worsened across the measured behavioural domains by CB receptor agonism but ameliorated by CB1 receptor antagonism. CONCLUSIONS AND IMPLICATIONS: These results provide the first evidence of the effects of modulating the endocannabinoid system on motor function in the harmaline model of ET. Our data suggest that CB1 receptor manipulation warrants clinical investigation as a therapeutic approach to protection against behavioural disturbances associated with ET.


Asunto(s)
Antagonistas de Receptores de Cannabinoides/farmacología , Antagonistas de Receptores de Cannabinoides/uso terapéutico , Temblor Esencial/inducido químicamente , Temblor Esencial/tratamiento farmacológico , Harmalina/farmacología , Receptor Cannabinoide CB1/antagonistas & inhibidores , Animales , Masculino , Ratas , Ratas Wistar
5.
Neurotherapeutics ; 12(4): 699-730, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26264914

RESUMEN

Cannabis has a long history of anecdotal medicinal use and limited licensed medicinal use. Until recently, alleged clinical effects from anecdotal reports and the use of licensed cannabinoid medicines are most likely mediated by tetrahydrocannabinol by virtue of: 1) this cannabinoid being present in the most significant quantities in these preparations; and b) the proportion:potency relationship between tetrahydrocannabinol and other plant cannabinoids derived from cannabis. However, there has recently been considerable interest in the therapeutic potential for the plant cannabinoid, cannabidiol (CBD), in neurological disorders but the current evidence suggests that CBD does not directly interact with the endocannabinoid system except in vitro at supraphysiological concentrations. Thus, as further evidence for CBD's beneficial effects in neurological disease emerges, there remains an urgent need to establish the molecular targets through which it exerts its therapeutic effects. Here, we conducted a systematic search of the extant literature for original articles describing the molecular pharmacology of CBD. We critically appraised the results for the validity of the molecular targets proposed. Thereafter, we considered whether the molecular targets of CBD identified hold therapeutic potential in relevant neurological diseases. The molecular targets identified include numerous classical ion channels, receptors, transporters, and enzymes. Some CBD effects at these targets in in vitro assays only manifest at high concentrations, which may be difficult to achieve in vivo, particularly given CBD's relatively poor bioavailability. Moreover, several targets were asserted through experimental designs that demonstrate only correlation with a given target rather than a causal proof. When the molecular targets of CBD that were physiologically plausible were considered for their potential for exploitation in neurological therapeutics, the results were variable. In some cases, the targets identified had little or no established link to the diseases considered. In others, molecular targets of CBD were entirely consistent with those already actively exploited in relevant, clinically used, neurological treatments. Finally, CBD was found to act upon a number of targets that are linked to neurological therapeutics but that its actions were not consistent withmodulation of such targets that would derive a therapeutically beneficial outcome. Overall, we find that while >65 discrete molecular targets have been reported in the literature for CBD, a relatively limited number represent plausible targets for the drug's action in neurological disorders when judged by the criteria we set. We conclude that CBD is very unlikely to exert effects in neurological diseases through modulation of the endocannabinoid system. Moreover, a number of other molecular targets of CBD reported in the literature are unlikely to be of relevance owing to effects only being observed at supraphysiological concentrations. Of interest and after excluding unlikely and implausible targets, the remaining molecular targets of CBD with plausible evidence for involvement in therapeutic effects in neurological disorders (e.g., voltage-dependent anion channel 1, G protein-coupled receptor 55, CaV3.x, etc.) are associated with either the regulation of, or responses to changes in, intracellular calcium levels. While no causal proof yet exists for CBD's effects at these targets, they represent the most probable for such investigations and should be prioritized in further studies of CBD's therapeutic mechanism of action.


Asunto(s)
Cannabinoides/uso terapéutico , Endocannabinoides/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Animales , Endocannabinoides/genética , Humanos , Enfermedades del Sistema Nervioso/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo
6.
Neurotherapeutics ; 12(4): 747-68, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26282273

RESUMEN

Cannabis has been used for centuries to treat seizures. Recent anecdotal reports, accumulating animal model data, and mechanistic insights have raised interest in cannabis-based antiepileptic therapies. In this study, we review current understanding of the endocannabinoid system, characterize the pro- and anticonvulsive effects of cannabinoids [e.g., Δ9-tetrahydrocannabinol and cannabidiol (CBD)], and highlight scientific evidence from pre-clinical and clinical trials of cannabinoids in epilepsy. These studies suggest that CBD avoids the psychoactive effects of the endocannabinoid system to provide a well-tolerated, promising therapeutic for the treatment of seizures, while whole-plant cannabis can both contribute to and reduce seizures. Finally, we discuss results from a new multicenter, open-label study using CBD in a population with treatment-resistant epilepsy. In all, we seek to evaluate our current understanding of cannabinoids in epilepsy and guide future basic science and clinical studies.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Cannabinoides/uso terapéutico , Epilepsia/tratamiento farmacológico , Animales , Humanos
7.
ACS Chem Neurosci ; 5(11): 1131-41, 2014 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-25029033

RESUMEN

Epilepsy is the most common neurological disorder, with over 50 million people worldwide affected. Recent evidence suggests that the transient receptor potential cation channel subfamily V member 1 (TRPV1) may contribute to the onset and progression of some forms of epilepsy. Since the two nonpsychotropic cannabinoids cannabidivarin (CBDV) and cannabidiol (CBD) exert anticonvulsant activity in vivo and produce TRPV1-mediated intracellular calcium elevation in vitro, we evaluated the effects of these two compounds on TRPV1 channel activation and desensitization and in an in vitro model of epileptiform activity. Patch clamp analysis in transfected HEK293 cells demonstrated that CBD and CBDV dose-dependently activate and rapidly desensitize TRPV1, as well as TRP channels of subfamily V type 2 (TRPV2) and subfamily A type 1 (TRPA1). TRPV1 and TRPV2 transcripts were shown to be expressed in rat hippocampal tissue. When tested on epileptiform neuronal spike activity in hippocampal brain slices exposed to a Mg(2+)-free solution using multielectrode arrays (MEAs), CBDV reduced both epileptiform burst amplitude and duration. The prototypical TRPV1 agonist, capsaicin, produced similar, although not identical effects. Capsaicin, but not CBDV, effects on burst amplitude were reversed by IRTX, a selective TRPV1 antagonist. These data suggest that CBDV antiepileptiform effects in the Mg(2+)-free model are not uniquely mediated via activation of TRPV1. However, TRPV1 was strongly phosphorylated (and hence likely sensitized) in Mg(2+)-free solution-treated hippocampal tissue, and both capsaicin and CBDV caused TRPV1 dephosphorylation, consistent with TRPV1 desensitization. We propose that CBDV effects on TRP channels should be studied further in different in vitro and in vivo models of epilepsy.


Asunto(s)
Cannabinoides/farmacología , Potenciales de la Membrana/efectos de los fármacos , Canales Catiónicos TRPV/metabolismo , Animales , Capsaicina/farmacología , Diterpenos/farmacología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Hipocampo/citología , Humanos , Técnicas In Vitro , Magnesio/metabolismo , Potenciales de la Membrana/genética , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Ratas , Canal Catiónico TRPA1 , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética , Transfección , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo
8.
Nutr Rev ; 72(5): 319-33, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24666107

RESUMEN

Reviewed here is the existing evidence for the effects of ginseng extracts and isolated ginsenosides relevant to cognition in humans. Clinical studies in healthy volunteers and in patients with neurological disease or deficit, evidence from preclinical models of cognition, and pharmacokinetic data are considered. Conditions under which disease modification may indirectly benefit cognition but may not translate to cognitive benefits in healthy subjects are discussed. The number of chronic studies of ginseng effects in healthy individuals is limited, and the results from acute studies are inconsistent, making overall assessment of ginseng's efficacy as a cognitive enhancer premature. However, mechanistic results are encouraging; in particular, the ginsenosides Rg3 , Rh1 , Rh2 , Rb1 , Rd, Rg2 , and Rb3 , along with the aglycones protopanaxadiol and protopanaxatriol, warrant further attention. Compound K has a promising pharmacokinetic profile and can affect neurotransmission and neuroprotection. Properly conducted trials using standardized tests in healthy individuals reflecting the target population for ginseng supplementation are required to address inconsistencies in results from acute studies. The evidence summarized here suggests ginseng has potential, but unproven, benefits on cognition.


Asunto(s)
Trastornos del Conocimiento/tratamiento farmacológico , Cognición/efectos de los fármacos , Ginsenósidos/farmacología , Panax/química , Extractos Vegetales/farmacología , Cognición/fisiología , Humanos , Fármacos Neuroprotectores/farmacología , Fitoterapia , Sapogeninas/farmacología
9.
Phytother Res ; 28(5): 643-55, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-23897801

RESUMEN

Hypericum perforatum (HP) belongs to the Hypericaceae family and is one of the oldest used and most extensively investigated medicinal herbs. The medicinal form comprises the leaves and flowering tops of which the primary ingredients of interest are naphthodianthrones, xanthones, flavonoids, phloroglucinols (e.g. hyperforin), and hypericin. Although several constituents elicit pharmacological effects that are consistent with HP's antidepressant activity, no single mechanism of action underlying these effects has thus far been found. Various clinical trials have shown that HP has a comparable antidepressant efficacy as some currently used antidepressant drugs in the treatment of mild/moderate depression. Interestingly, low-hyperforin-content preparations are effective in the treatment of depression. Moreover, HP is also used to treat certain forms of anxiety. However, HP can induce various cytochrome P450s isozymes and/or P-glycoprotein, of which many drugs are substrates and which are the main origin of HP-drug interactions. Here, we analyse the existing evidence describing the clinical consequence of HP-drug interactions. Although some of the reported interactions are based on findings from in vitro studies, the clinical importance of which remain to be demonstrated, others are based on case reports where causality can, in some cases, be determined to reveal clinically significant interactions that suggest caution, consideration, and disclosure of potential interactions prior to informed use of HP.


Asunto(s)
Interacciones de Hierba-Droga , Hypericum/química , Extractos Vegetales/farmacología , Plantas Medicinales/química , Antracenos , Antidepresivos/farmacología , Antivirales/farmacología , Depresión/tratamiento farmacológico , Humanos , Perileno/análogos & derivados , Perileno/farmacocinética , Floroglucinol/análogos & derivados , Floroglucinol/farmacocinética , Extractos Vegetales/uso terapéutico , Terpenos/farmacocinética
10.
Epilepsy Res ; 106(1-2): 74-82, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23860329

RESUMEN

Drugs that modulate the endocannabinoid system and endocannabinoids typically play an anticonvulsant role although some proconvulsant effects have been reported both in humans and animal models. Moreover, no evidence for a role of the cannabinoid system in human absence epilepsy has been found although limited evidence of efficacy in relevant experimental animal models has been documented. This study aims to characterize the role of cannabinoids in specific areas of the cortico-thalamic network involved in oscillations that underlie seizures in a genetic animal model of absence epilepsy, the WAG/Rij rat. We assessed the effects of focal injection of the endogenous cannabinoid, anandamide (AEA), a non-selective CB receptor agonist (WIN55,212) and a selective CB1 receptor antagonist/inverse agonist (SR141716A) into thalamic nuclei and primary somatosensory cortex (S1po) of the cortico-thalamic network. AEA and WIN both reduced absence seizures independently from the brain focal site of infusion while, conversely, rimonabant increased absence seizures but only when focally administered to the ventroposteromedial thalamic nucleus (VPM). These results, together with previous reports, support therapeutic potential for endocannabinoid system modulators in absence epilepsy and highlight that attenuated endocannabinergic function may contribute to the generation and maintenance of seizures. Furthermore, the entire cortico-thalamic network responds to cannabinoid treatment, indicating that in all areas considered, CB receptor activation inhibits the pathological synchronization that subserves absence seizures. In conclusion, our result might be useful for the identification of future drug therapies in absence epilepsy.


Asunto(s)
Anticonvulsivantes , Agonistas de Receptores de Cannabinoides/farmacología , Corteza Cerebral/efectos de los fármacos , Epilepsia Tipo Ausencia/genética , Epilepsia Tipo Ausencia/prevención & control , Vías Nerviosas/efectos de los fármacos , Receptor Cannabinoide CB1/agonistas , Tálamo/efectos de los fármacos , Animales , Ácidos Araquidónicos/farmacología , Benzoxazinas/farmacología , Agonistas de Receptores de Cannabinoides/administración & dosificación , Electroencefalografía/efectos de los fármacos , Endocannabinoides/farmacología , Epilepsia/tratamiento farmacológico , Epilepsia/fisiopatología , Inyecciones Intraventriculares , Masculino , Morfolinas/farmacología , Naftalenos/farmacología , Piperidinas/farmacología , Alcamidas Poliinsaturadas/farmacología , Pirazoles/farmacología , Ratas , Rimonabant , Convulsiones/fisiopatología , Corteza Somatosensorial/fisiología
11.
Seizure ; 21(5): 344-52, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22520455

RESUMEN

Cannabis sativa has been associated with contradictory effects upon seizure states despite its medicinal use by numerous people with epilepsy. We have recently shown that the phytocannabinoid cannabidiol (CBD) reduces seizure severity and lethality in the well-established in vivo model of pentylenetetrazole-induced generalised seizures, suggesting that earlier, small-scale clinical trials examining CBD effects in people with epilepsy warrant renewed attention. Here, we report the effects of pure CBD (1, 10 and 100mg/kg) in two other established rodent seizure models, the acute pilocarpine model of temporal lobe seizure and the penicillin model of partial seizure. Seizure activity was video recorded and scored offline using model-specific seizure severity scales. In the pilocarpine model CBD (all doses) significantly reduced the percentage of animals experiencing the most severe seizures. In the penicillin model, CBD (≥ 10 mg/kg) significantly decreased the percentage mortality as a result of seizures; CBD (all doses) also decreased the percentage of animals experiencing the most severe tonic-clonic seizures. These results extend the anti-convulsant profile of CBD; when combined with a reported absence of psychoactive effects, this evidence strongly supports CBD as a therapeutic candidate for a diverse range of human epilepsies.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Cannabidiol/farmacología , Convulsiones/tratamiento farmacológico , Lóbulo Temporal/efectos de los fármacos , Animales , Anticonvulsivantes/farmacología , Cannabidiol/administración & dosificación , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Endogámicas WKY , Convulsiones/inducido químicamente , Resultado del Tratamiento
12.
Pharmacol Ther ; 133(1): 79-97, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21924288

RESUMEN

The Cannabis sativa herb contains over 100 phytocannabinoid (pCB) compounds and has been used for thousands of years for both recreational and medicinal purposes. In the past two decades, characterisation of the body's endogenous cannabinoid (CB) (endocannabinoid, eCB) system (ECS) has highlighted activation of central CB(1) receptors by the major pCB, Δ(9)-tetrahydrocannabinol (Δ(9)-THC) as the primary mediator of the psychoactive, hyperphagic and some of the potentially therapeutic properties of ingested cannabis. Whilst Δ(9)-THC is the most prevalent and widely studied pCB, it is also the predominant psychotropic component of cannabis, a property that likely limits its widespread therapeutic use as an isolated agent. In this regard, research focus has recently widened to include other pCBs including cannabidiol (CBD), cannabigerol (CBG), Δ(9)tetrahydrocannabivarin (Δ(9)-THCV) and cannabidivarin (CBDV), some of which show potential as therapeutic agents in preclinical models of CNS disease. Moreover, it is becoming evident that these non-Δ(9)-THC pCBs act at a wide range of pharmacological targets, not solely limited to CB receptors. Disorders that could be targeted include epilepsy, neurodegenerative diseases, affective disorders and the central modulation of feeding behaviour. Here, we review pCB effects in preclinical models of CNS disease and, where available, clinical trial data that support therapeutic effects. Such developments may soon yield the first non-Δ(9)-THC pCB-based medicines.


Asunto(s)
Cannabinoides/farmacología , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Fitoterapia , Preparaciones de Plantas/farmacología , Animales , Cannabinoides/biosíntesis , Cannabinoides/uso terapéutico , Cannabis/metabolismo , Enfermedades del Sistema Nervioso Central/metabolismo , Humanos , Preparaciones de Plantas/uso terapéutico
13.
Behav Pharmacol ; 23(1): 113-7, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22157176

RESUMEN

Cannabinoid type 1 receptor-mediated appetite stimulation by Δ9tetrahydrocannabinol (Δ9THC) is well understood. Recently, it has become apparent that non-Δ9THC phytocannabinoids could also alter feeding patterns. Here, we show definitively that non-Δ9THC phytocannabinoids stimulate feeding. Twelve male, Lister-Hooded rats were prefed to satiety prior to administration of a standardized cannabis extract or to either of two mixtures of pure phytocannabinoids (extract analogues) comprising the phytocannabinoids present in the same proportions as the standardized extract (one with and one without Δ9THC). Hourly intake and meal pattern data were recorded and analysed using two-way analysis of variance followed by one-way analysis of variance and Bonferroni post-hoc tests. Administration of both extract analogues significantly increased feeding behaviours over the period of the test. All three agents increased hour-one intake and meal-one size and decreased the latency to feed, although the zero-Δ9THC extract analogue did so to a lesser degree than the high-Δ9THC analogue. Furthermore, only the analogue containing Δ9THC significantly increased meal duration. The data confirm that at least one non-Δ9THC phytocannabinoid induces feeding pattern changes in rats, although further trials using individual phytocannabinoids are required to fully understand the observed effects.


Asunto(s)
Dronabinol/farmacología , Ingestión de Alimentos/efectos de los fármacos , Extractos Vegetales/farmacología , Animales , Relación Dosis-Respuesta a Droga , Masculino , Ratas
14.
Phytother Res ; 25(2): 170-88, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21213357

RESUMEN

The herb Cannabis sativa (C. sativa) has been used in China and on the Indian subcontinent for thousands of years as a medicine. However, since it was brought to the UK and then the rest of the western world in the late 19th century, its use has been a source of controversy. Indeed, its psychotropic side effects are well reported but only relatively recently has scientific endeavour begun to find valuable uses for either the whole plant or its individual components. Here, we discuss evidence describing the endocannabinoid system, its endogenous and exogenous ligands and their varied effects on feeding cycles and meal patterns. Furthermore we also critically consider the mounting evidence which suggests non-Δ(9) tetrahydrocannabinol phytocannabinoids play a vital role in C. sativa-induced feeding pattern changes. Indeed, given the wide range of phytocannabinoids present in C. sativa and their equally wide range of intra-, inter- and extra-cellular mechanisms of action, we demonstrate that non-Δ(9) tetrahydrocannabinol phytocannabinoids retain an important and, as yet, untapped clinical potential.


Asunto(s)
Regulación del Apetito/efectos de los fármacos , Cannabis/química , Dronabinol/farmacología , Animales , Sistema Nervioso Central/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Receptores de Cannabinoides/efectos de los fármacos
15.
J Neurosci Methods ; 185(2): 246-56, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19836417

RESUMEN

The acute hippocampal brain slice preparation is an important in vitro screening tool for potential anticonvulsants. Application of 4-aminopyridine (4-AP) or removal of external Mg(2+) ions induces epileptiform bursting in slices which is analogous to electrical brain activity seen in status epilepticus states. We have developed these epileptiform models for use with multi-electrode arrays (MEAs), allowing recording across the hippocampal slice surface from 59 points. We present validation of this novel approach and analyses using two anticonvulsants, felbamate and phenobarbital, the effects of which have already been assessed in these models using conventional extracellular recordings. In addition to assessing drug effects on commonly described parameters (duration, amplitude and frequency), we describe novel methods using the MEA to assess burst propagation speeds and the underlying frequencies that contribute to the epileptiform activity seen. Contour plots are also used as a method of illustrating burst activity. Finally, we describe hitherto unreported properties of epileptiform bursting induced by 100 microM 4-AP or removal of external Mg(2+) ions. Specifically, we observed decreases over time in burst amplitude and increase over time in burst frequency in the absence of additional pharmacological interventions. These MEA methods enhance the depth, quality and range of data that can be derived from the hippocampal slice preparation compared to conventional extracellular recordings. It may also uncover additional modes of action that contribute to anti-epileptiform drug effects.


Asunto(s)
Anticonvulsivantes/farmacología , Evaluación Preclínica de Medicamentos/instrumentación , Electrodos , 4-Aminopiridina/efectos adversos , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Electrofisiología/métodos , Epilepsia/tratamiento farmacológico , Epilepsia/etiología , Felbamato , Femenino , Hipocampo/efectos de los fármacos , Técnicas In Vitro , Deficiencia de Magnesio/complicaciones , Potenciales de la Membrana/efectos de los fármacos , Fenobarbital/farmacología , Fenilcarbamatos/farmacología , Glicoles de Propileno/farmacología , Ratas , Ratas Wistar , Factores de Tiempo
16.
Neurosci Lett ; 365(1): 58-63, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15234473

RESUMEN

Cannabis is a potential treatment for epilepsy, although the few human studies supporting this use have proved inconclusive. Previously, we showed that a standardized cannabis extract (SCE), isolated Delta9-tetrahydrocannabinol (Delta9-THC), and even Delta9-THC-free SCE inhibited muscarinic agonist-induced epileptiform bursting in rat olfactory cortical brain slices, acting via CB1 receptors. The present work demonstrates that although Delta9-THC (1 microM) significantly depressed evoked depolarizing postsynaptic potentials (PSPs) in rat olfactory cortex neurones, both SCE and Delta9-THC-free SCE significantly potentiated evoked PSPs (all results were fully reversed by the CB1 receptor antagonist SR141716A, 1 microM); interestingly, the potentiation by Delta9-THC-free SCE was greater than that produced by SCE. On comparing the effects of Delta9-THC-free SCE upon evoked PSPs and artificial PSPs (aPSPs; evoked electrotonically following brief intracellular current injection), PSPs were enhanced, whereas aPSPs were unaffected, suggesting that the effect was not due to changes in background input resistance. Similar recordings made using CB1 receptor-deficient knockout mice (CB1-/-) and wild-type littermate controls revealed cannabinoid or extract-induced changes in membrane resistance, cell excitability and synaptic transmission in wild-type mice that were similar to those seen in rat neurones, but no effect on these properties were seen in CB1-/- cells. It appears that the unknown extract constituent(s) effects over-rode the suppressive effects of Delta9-THC on excitatory neurotransmitter release, which may explain some patients' preference for herbal cannabis rather than isolated Delta9-THC (due to attenuation of some of the central Delta9-THC side effects) and possibly account for the rare incidence of seizures in some individuals taking cannabis recreationally.


Asunto(s)
Cannabinoides/farmacología , Neuronas/efectos de los fármacos , Vías Olfatorias/efectos de los fármacos , Extractos Vegetales/farmacología , Transmisión Sináptica/efectos de los fármacos , Animales , Estimulación Eléctrica , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Masculino , Ratones , Ratones Noqueados , Microelectrodos , Neuronas/fisiología , Vías Olfatorias/fisiología , Técnicas de Cultivo de Órganos , Ratas , Ratas Wistar , Receptor Cannabinoide CB1/deficiencia , Transmisión Sináptica/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA