Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Sci Rep ; 10(1): 20405, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33230154

RESUMEN

The Cannabis sativa plant contains more than 120 cannabinoids. With the exceptions of ∆9-tetrahydrocannabinol (∆9-THC) and cannabidiol (CBD), comparatively little is known about the pharmacology of the less-abundant plant-derived (phyto) cannabinoids. The best-studied transducers of cannabinoid-dependent effects are type 1 and type 2 cannabinoid receptors (CB1R, CB2R). Partial agonism of CB1R by ∆9-THC is known to bring about the 'high' associated with Cannabis use, as well as the pain-, appetite-, and anxiety-modulating effects that are potentially therapeutic. CB2R activation by certain cannabinoids has been associated with anti-inflammatory activities. We assessed the activity of 8 phytocannabinoids at human CB1R, and CB2R in Chinese hamster ovary (CHO) cells stably expressing these receptors and in C57BL/6 mice in an attempt to better understand their pharmacodynamics. Specifically, ∆9-THC, ∆9-tetrahydrocannabinolic acid (∆9-THCa), ∆9-tetrahydrocannabivarin (THCV), CBD, cannabidiolic acid (CBDa), cannabidivarin (CBDV), cannabigerol (CBG), and cannabichromene (CBC) were evaluated. Compounds were assessed for their affinity to receptors, ability to inhibit cAMP accumulation, ßarrestin2 recruitment, receptor selectivity, and ligand bias in cell culture; and cataleptic, hypothermic, anti-nociceptive, hypolocomotive, and anxiolytic effects in mice. Our data reveal partial agonist activity for many phytocannabinoids tested at CB1R and/or CB2R, as well as in vivo responses often associated with activation of CB1R. These data build on the growing body of literature showing cannabinoid receptor-dependent pharmacology for these less-abundant phytocannabinoids and are critical in understanding the complex and interactive pharmacology of Cannabis-derived molecules.


Asunto(s)
Analgésicos/farmacología , Ansiolíticos/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Cannabis/química , Psicotrópicos/farmacología , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB2/genética , Analgésicos/aislamiento & purificación , Animales , Ansiolíticos/aislamiento & purificación , Células CHO , Cannabidiol/aislamiento & purificación , Cannabidiol/farmacología , Agonistas de Receptores de Cannabinoides/aislamiento & purificación , Cannabinoides/aislamiento & purificación , Cannabinoides/farmacología , Cricetulus , Dronabinol/análogos & derivados , Dronabinol/aislamiento & purificación , Dronabinol/farmacología , Expresión Génica , Humanos , Ratones Endogámicos C57BL , Extractos Vegetales/química , Psicotrópicos/aislamiento & purificación , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/metabolismo , Transgenes , Arrestina beta 2/genética , Arrestina beta 2/metabolismo
2.
Molecules ; 25(17)2020 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-32854452

RESUMEN

Opioids such as morphine-acting at the mu opioid receptor-are the mainstay for treatment of moderate to severe pain and have good efficacy in these indications. However, these drugs produce a plethora of unwanted adverse effects including respiratory depression, constipation, immune suppression and with prolonged treatment, tolerance, dependence and abuse liability. Studies in ß-arrestin 2 gene knockout (ßarr2(-/-)) animals indicate that morphine analgesia is potentiated while side effects are reduced, suggesting that drugs biased away from arrestin may manifest with a reduced-side-effect profile. However, there is controversy in this area with improvement of morphine-induced constipation and reduced respiratory effects in ßarr2(-/-) mice. Moreover, studies performed with mice genetically engineered with G-protein-biased mu receptors suggested increased sensitivity of these animals to both analgesic actions and side effects of opioid drugs. Several new molecules have been identified as mu receptor G-protein-biased agonists, including oliceridine (TRV130), PZM21 and SR-17018. These compounds have provided preclinical data with apparent support for bias toward G proteins and the genetic premise of effective and safer analgesics. There are clinical data for oliceridine that have been very recently approved for short term intravenous use in hospitals and other controlled settings. While these data are compelling and provide a potential new pathway-based target for drug discovery, a simpler explanation for the behavior of these biased agonists revolves around differences in intrinsic activity. A highly detailed study comparing oliceridine, PZM21 and SR-17018 (among others) in a range of assays showed that these molecules behave as partial agonists. Moreover, there was a correlation between their therapeutic indices and their efficacies, but not their bias factors. If there is amplification of G-protein, but not arrestin pathways, then agonists with reduced efficacy would show high levels of activity at G-protein and low or absent activity at arrestin; offering analgesia with reduced side effects or 'apparent bias'. Overall, the current data suggests-and we support-caution in ascribing biased agonism to reduced-side-effect profiles for mu-agonist analgesics.


Asunto(s)
Analgésicos Opioides , Aprobación de Drogas , Dolor/tratamiento farmacológico , Analgésicos Opioides/química , Analgésicos Opioides/uso terapéutico , Animales , Evaluación Preclínica de Medicamentos , Humanos , Ratones , Ratones Noqueados , Dolor/genética , Dolor/metabolismo , Dolor/patología , Arrestina beta 2/agonistas , Arrestina beta 2/genética , Arrestina beta 2/metabolismo
3.
Biochem Pharmacol ; 177: 113934, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32224136

RESUMEN

The A3 adenosine receptor (A3AR) is a G protein-coupled receptor that is involved in a wide variety of physiological and pathological processes, such as cancer. However, the use of compounds pharmacologically targeting this receptor remains limited in clinical practice, despite extensive efforts for compound synthesis. Moreover, the possible occurrence of biased agonism further complicates the interpretation of the functional characteristics of compounds. Hence the need for simple assays, which are comparable in terms of the used cell lines and read-out technique. We previously established a stable ß-arrestin 2 (ßarr2) bioassay, employing a simple, luminescent read-out via functional complementation of a split nanoluciferase enzyme. Here, we developed a complementary, new bioassay in which coupling of an engineered miniGαi protein to activated A3AR is monitored using a similar approach. Application of both bioassays for the concurrent determination of the potencies and efficacies of a set of 19 N6-substituted adenosine analogues not only allowed for the characterization of structure-activity relationships, but also for the quantification of biased agonism. Although a broad distribution in potency and efficacy values was obtained within the test panel, no significant bias was observed toward either the ßarr2 or miniGαi pathway.


Asunto(s)
Agonistas del Receptor de Adenosina A3/farmacología , Evaluación Preclínica de Medicamentos/métodos , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Receptor de Adenosina A3/metabolismo , Arrestina beta 2/metabolismo , Adenosina/análogos & derivados , Agonistas del Receptor de Adenosina A3/síntesis química , Citometría de Flujo/métodos , Subunidades alfa de la Proteína de Unión al GTP/genética , Células HEK293 , Humanos , Ligandos , Mediciones Luminiscentes/métodos , Receptor de Adenosina A3/genética , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Transducción Genética/métodos , Transfección/métodos , Arrestina beta 2/genética
4.
Exp Neurol ; 290: 115-122, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28077335

RESUMEN

Omega-3 fatty acids (ω-3 FAs) attenuate inflammation and improve neurological outcome in response to traumatic brain injury (TBI), but the specific anti-inflammatory mechanisms remain to be elucidated. Here we found that NLRP3 inflammasome and subsequent pro-inflammatory cytokines were activated in human brains after TBI. Rats treated with ω-3 FAs had significantly less TBI-induced caspase-1 cleavage and IL-1ß secretion than those with vehicle. G protein-coupled receptor 40 (GPR40) was observed to be involved in this anti-inflammation. GW1100, a GPR40 inhibitor, eliminated the anti-inflammatory effect of ω-3 FAs after TBI. ß-Arrestin-2 (ARRB2), a downstream scaffold protein of GPR40, was activated to inhibit inflammation via directly binding with NLRP3 in the ω-3 FAs treatment group. Interestingly, we also observed that ω-3 FAs prevented NLRP3 mitochondrial localization, which was reversed by GW1100. Furthermore, ω-3 FAs markedly ameliorated neuronal death and behavioral deficits after TBI, while GW1100 significantly suppressed this effect. Collectively, these data indicate that the GPR40-mediated pathway is involved in the inhibitory effects of ω-3 FAs on TBI-induced inflammation and ARRB2 is activated to interact with NLRP3.


Asunto(s)
Conducta Animal/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/psicología , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Omega-3/uso terapéutico , Inflamasomas/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Animales , Benzoatos/uso terapéutico , Química Encefálica/efectos de los fármacos , Caspasa 1/biosíntesis , Caspasa 1/genética , Citocinas/líquido cefalorraquídeo , Inhibidores Enzimáticos/farmacología , Interleucina-1/biosíntesis , Interleucina-1/genética , Masculino , Mitocondrias/efectos de los fármacos , Pirimidinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Arrestina beta 2/biosíntesis , Arrestina beta 2/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA