Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 145
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Diabetes Obes Metab ; 18(6): 549-57, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26880114

RESUMEN

Visceral obesity is a major risk factor for the development of insulin resistance which can progress to overt type 2 diabetes (T2D) with loss of ß-cell function and, ultimately, loss of ß-cells. Insulin secretion by ß-cells of the pancreatic islets is tightly coupled to blood glucose concentration and modulated by a large number of blood-borne or locally released mediators, including endocannabinoids. Obesity and its complications, including T2D, are associated with increased activity of the endocannabinoid/CB1 receptor (CB1 R) system, as indicated by the therapeutic effects of CB1 R antagonists. Similar beneficial effects of CB1 R antagonists with limited brain penetrance indicate the important role of CB1 R in peripheral tissues, including the endocrine pancreas. Pancreatic ß-cells express all of the components of the endocannabinoid system, and endocannabinoids modulate their function via both autocrine and paracrine mechanisms, which influence basal and glucose-induced insulin secretion and also affect ß-cell proliferation and survival. The present brief review will survey available information on the modulation of these processes by endocannabinoids and their receptors, with an attempt to assess the contribution of such effects to glycaemic control in T2D and insulin resistance.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Endocannabinoides/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Animales , Glucemia/efectos de los fármacos , Cannabis/fisiología , Diabetes Mellitus Tipo 2/sangre , Endocannabinoides/fisiología , Humanos , Resistencia a la Insulina/fisiología , Células Secretoras de Insulina/fisiología , Receptor Cannabinoide CB1/fisiología
2.
Mol Psychiatry ; 18(7): 813-23, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22688188

RESUMEN

Endocannabinoids are released 'on-demand' on the basis of physiological need, and can be pharmacologically augmented by inhibiting their catabolic degradation. The endocannabinoid anandamide is degraded by the catabolic enzyme fatty acid amide hydrolase (FAAH). Anandamide is implicated in the mediation of fear behaviors, including fear extinction, suggesting that selectively elevating brain anandamide could modulate plastic changes in fear. Here we first tested this hypothesis with preclinical experiments employing a novel, potent and selective FAAH inhibitor, AM3506 (5-(4-hydroxyphenyl)pentanesulfonyl fluoride). Systemic AM3506 administration before extinction decreased fear during a retrieval test in a mouse model of impaired extinction. AM3506 had no effects on fear in the absence of extinction training, or on various non-fear-related measures. Anandamide levels in the basolateral amygdala were increased by extinction training and augmented by systemic AM3506, whereas application of AM3506 to amygdala slices promoted long-term depression of inhibitory transmission, a form of synaptic plasticity linked to extinction. Further supporting the amygdala as effect-locus, the fear-reducing effects of systemic AM3506 were blocked by intra-amygdala infusion of a CB1 receptor antagonist and were fully recapitulated by intra-amygdala infusion of AM3506. On the basis of these preclinical findings, we hypothesized that variation in the human FAAH gene would predict individual differences in amygdala threat-processing and stress-coping traits. Consistent with this, carriers of a low-expressing FAAH variant (385A allele; rs324420) exhibited quicker habituation of amygdala reactivity to threat, and had lower scores on the personality trait of stress-reactivity. Our findings show that augmenting amygdala anandamide enables extinction-driven reductions in fear in mouse and may promote stress-coping in humans.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/fisiología , Ácidos Araquidónicos/fisiología , Endocannabinoides/fisiología , Extinción Psicológica/fisiología , Miedo/fisiología , Adaptación Psicológica/fisiología , Adulto , Alcanosulfonatos/administración & dosificación , Alcanosulfonatos/farmacología , Amidohidrolasas/antagonistas & inhibidores , Amidohidrolasas/genética , Amígdala del Cerebelo/efectos de los fármacos , Animales , Ácidos Araquidónicos/metabolismo , Antagonistas de Receptores de Cannabinoides/administración & dosificación , Antagonistas de Receptores de Cannabinoides/farmacología , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Endocannabinoides/metabolismo , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Miedo/efectos de los fármacos , Miedo/psicología , Femenino , Neuroimagen Funcional , Estudios de Asociación Genética , Habituación Psicofisiológica/efectos de los fármacos , Habituación Psicofisiológica/fisiología , Humanos , Masculino , Ratones , Microinyecciones , Persona de Mediana Edad , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Personalidad/genética , Personalidad/fisiología , Fenoles/administración & dosificación , Fenoles/farmacología , Piperidinas/administración & dosificación , Piperidinas/farmacología , Polimorfismo de Nucleótido Simple , Alcamidas Poliinsaturadas/metabolismo , Pirazoles/administración & dosificación , Pirazoles/farmacología , Rimonabant
3.
Pharmacol Rev ; 62(4): 588-631, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21079038

RESUMEN

There are at least two types of cannabinoid receptors (CB(1) and CB(2)). Ligands activating these G protein-coupled receptors (GPCRs) include the phytocannabinoid Δ(9)-tetrahydrocannabinol, numerous synthetic compounds, and endogenous compounds known as endocannabinoids. Cannabinoid receptor antagonists have also been developed. Some of these ligands activate or block one type of cannabinoid receptor more potently than the other type. This review summarizes current data indicating the extent to which cannabinoid receptor ligands undergo orthosteric or allosteric interactions with non-CB(1), non-CB(2) established GPCRs, deorphanized receptors such as GPR55, ligand-gated ion channels, transient receptor potential (TRP) channels, and other ion channels or peroxisome proliferator-activated nuclear receptors. From these data, it is clear that some ligands that interact similarly with CB(1) and/or CB(2) receptors are likely to display significantly different pharmacological profiles. The review also lists some criteria that any novel "CB(3)" cannabinoid receptor or channel should fulfil and concludes that these criteria are not currently met by any non-CB(1), non-CB(2) pharmacological receptor or channel. However, it does identify certain pharmacological targets that should be investigated further as potential CB(3) receptors or channels. These include TRP vanilloid 1, which possibly functions as an ionotropic cannabinoid receptor under physiological and/or pathological conditions, and some deorphanized GPCRs. Also discussed are 1) the ability of CB(1) receptors to form heteromeric complexes with certain other GPCRs, 2) phylogenetic relationships that exist between CB(1)/CB(2) receptors and other GPCRs, 3) evidence for the existence of several as-yet-uncharacterized non-CB(1), non-CB(2) cannabinoid receptors; and 4) current cannabinoid receptor nomenclature.


Asunto(s)
Receptores de Cannabinoides/metabolismo , Agonistas de Receptores de Cannabinoides , Antagonistas de Receptores de Cannabinoides , Moduladores de Receptores de Cannabinoides/metabolismo , Cannabinoides/metabolismo , Humanos , Ligandos , Filogenia , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/antagonistas & inhibidores , Receptor Cannabinoide CB2/metabolismo , Terminología como Asunto
4.
Nat Med ; 7(7): 827-32, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11433348

RESUMEN

Advanced cirrhosis is associated with generalized vasodilation of unknown origin, which contributes to mortality. Cirrhotic patients are endotoxemic, and activation of vascular cannabinoid CB1 receptors has been implicated in endotoxin-induced hypotension. Here we show that rats with biliary cirrhosis have low blood pressure, which is elevated by the CB1 receptor antagonist SR141716A. The low blood pressure of rats with CCl4-induced cirrhosis was similarly reversed by SR141716A, which also reduced the elevated mesenteric blood flow and portal pressure. Monocytes from cirrhotic but not control patients or rats elicited SR141716A-sensitive hypotension in normal recipient rats and showed significantly elevated levels of anandamide. Compared with non-cirrhotic controls, in cirrhotic human livers there was a three-fold increase in CB1 receptors on isolated vascular endothelial cells. These results implicate anandamide and vascular CB1 receptors in the vasodilated state in advanced cirrhosis and indicate a novel approach for its management.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Cirrosis Hepática/fisiopatología , Receptores de Droga/metabolismo , Vasodilatación/fisiología , Animales , Moduladores de Receptores de Cannabinoides , Cromatografía Líquida de Alta Presión , Endocannabinoides , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Hipotensión/etiología , Masculino , Espectrometría de Masas , Monocitos/metabolismo , Alcamidas Poliinsaturadas , Ratas , Ratas Sprague-Dawley , Receptores de Cannabinoides
5.
Science ; 211(4477): 82-4, 1981 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-6108611

RESUMEN

Clonidine and L-alpha-methylnoradrenaline (but not D-alpha-methylnoradrenaline) increase the release of a substance with beta-endorphin immunoreactivity from slices of brainstem of spontaneously hypertensive rats, but not that of normotensive rats. It was reported earlier that opiate antagonists inhibit the hypotensive action of clonidine and alpha-methyldopa in spontaneously hypertensive but not in normotensive rats and that beta-endorphin has hypotensive effects of its own. Together, these findings indicate that release of beta-endorphin by central alpha-receptor agonists may contribute to the antihypertensive action of these drugs.


Asunto(s)
Agonistas alfa-Adrenérgicos/farmacología , Tronco Encefálico/metabolismo , Endorfinas/metabolismo , Hipertensión/fisiopatología , Animales , Clonidina/farmacología , Modelos Animales de Enfermedad , Inmunoensayo , Masculino , Naloxona/farmacología , Nordefrin/farmacología , Ratas
6.
Neuron ; 31(3): 453-62, 2001 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-11516401

RESUMEN

Endocannabinoids are retrograde messengers released by neurons to modulate the strength of their synaptic inputs. Endocannabinoids are thought to mediate the suppression of GABA release that follows depolarization of a hippocampal CA1 pyramidal neuron-termed "depolarization-induced suppression of inhibition" (DSI). Here, we report that DSI is absent in mice which lack cannabinoid receptor-1 (CB1). Pharmacological and kinetic evidence suggests that CB1 activation inhibits presynaptic Ca2+ channels through direct G protein inhibition. Paired recordings show that endocannabinoids selectively inhibit a subclass of synapses distinguished by their fast kinetics and large unitary conductance. Furthermore, cannabinoid-sensitive inputs are unusual among central nervous system synapses in that they use N- but not P/Q-type Ca2+ channels for neurotransmitter release. These results indicate that endocannabinoids are highly selective, rapid modulators of hippocampal inhibition.


Asunto(s)
Cannabinoides/metabolismo , Hipocampo/fisiología , Terminales Presinápticos/fisiología , Células Piramidales/fisiología , Receptores de Droga/fisiología , Receptores Presinapticos/fisiología , Animales , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/fisiología , Moduladores de Receptores de Cannabinoides , Colforsina/farmacología , Endocannabinoides , Femenino , Proteínas de Unión al GTP/metabolismo , Técnicas In Vitro , Interneuronas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Placa-Clamp , Terminales Presinápticos/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Cannabinoides , Receptores de Droga/deficiencia , Receptores de Droga/genética , Receptores Presinapticos/efectos de los fármacos , Transducción de Señal
7.
Neuroscience ; 152(4): 1032-9, 2008 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-18343585

RESUMEN

Tissue levels of anandamide (AEA) and 2-arachidonoylglycerol (2-AG) have been determined in 16 regions and nuclei from human brains, using liquid chromatography/in-line mass spectrometry. Measurements in brain samples stored at -80 degrees C for 2 months to 13 years indicated that endocannabinoids were stable under such conditions. In contrast, the postmortal delay had a strong effect on brain endocannabinoid levels, as documented in brain samples microdissected and frozen 1-6 h postmortem, and in neurosurgical samples 0, 5, 30, 60, 180 and 360 min after their removal from the brain. The tissue levels of AEA increased continuously and in a region-dependent manner from 1 h after death, increasing about sevenfold by 6 h postmortem. In contrast, concentrations of 2-AG, which were 10-100 times higher in human brain regions than those of AEA, rapidly declined: within the first hour, 2-AG levels dropped to 25-35% of the initial ('0 min') value, thereafter they remained relatively stable. As analyzed in samples removed 1-1.5 h postmortem, AEA levels ranged from a high of 96.3 fmol/mg tissue in the nucleus accumbens to a low of 25.0 fmol/mg in the cerebellum. 2-AG levels varied eightfold, from 8.6 pmol/mg in the lateral hypothalamus to 1.1 pmol/mg in the nucleus accumbens. Relative levels of AEA and 2-AG varied from region to region, with the 2-AG:AEA ratio being high in the sensory spinal trigeminal nucleus (140:1), the spinal dorsal horn (136:1) and the lateral hypothalamus (98:1) and low in the nucleus accumbens (16:1) and the striatum (31:1). The results highlight the pitfall of analyzing endocannabinoid content in brain samples of variable postmortal delay, and document differential distribution of the two main endocannabinoids in the human brain.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Química Encefálica/fisiología , Encéfalo/metabolismo , Glicéridos/metabolismo , Alcamidas Poliinsaturadas/metabolismo , Cambios Post Mortem , Encéfalo/anatomía & histología , Cromatografía Liquida/métodos , Endocannabinoides , Femenino , Humanos , Masculino , Espectrometría de Masas/métodos , Microdisección , Factores de Tiempo
8.
Mol Cell Biol ; 16(11): 5997-6008, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8887629

RESUMEN

The 5' upstream region from --490 to --540 (footprint II) within the dominant P2 promoter of the rat alpha(1b) adrenergic receptor (alpha(1b)AR) gene is recognized by a sequence-specific DNA-binding protein (B. Gao, M. S. Spector, and G. Kunos, J. Biol. Chem. 270:5614-5619, 1995). This protein, detectable in Southwestern (DNA-protein) blots of crude nuclear extracts as 32- and 34-kDa bands, has been purified 6,000-fold from rat livers by DEAE-Sepharose, heparin-Sepharose, and DNA affinity chromatography. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis and UV cross-linking of the purified protein indicated the same molecular mass as that in crude extracts. Methylation interference analysis revealed strong contact with a TTGGCT hexamer and weak contact with a TGGCGT hexamer in the 3' and 5' portions of footprint II, respectively. Nucleotide substitutions within these hexamers significantly reduced protein binding to footprint II and the promoter activity of P2 in Hep3B cells. The purified protein also bound to the nuclear factor 1 (NF1)/CTF consensus sequence, albeit with lower affinity. Gel mobility supershift and Western blotting (immunoblotting) analyses using an antibody against the NF1/CTF protein identified the purified 32- and 34-kDa polypeptides as NF1 or a related protein. Cotransfection into Hep3B cells or primary rat hepatocytes of cDNAs of the NF1-like proteins NF1/L, NF1/X, and NF1/Redl resulted in a three- to fivefold increase in transcription directed by wild-type P2 but not by the mutated P2. Partial hepatectomy markedly decreased the levels of NF1 in the remnant liver and its binding to P2, which paralleled declines in the rate of transcription of the alpha(1b)AR gene and in the steady-state levels of its mRNA. These observations indicate that NF1 activates transcription of the rat alpha(1b)AR gene via interacting with its P2 promoter and that a decline in the expression of NF1 is one of the mechanisms responsible for the reduced expression of the alpha(1b)AR gene during liver regeneration.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Regeneración Hepática , Hígado/metabolismo , Receptores Adrenérgicos alfa 1/biosíntesis , Animales , Secuencia de Bases , Línea Celular , Núcleo Celular/metabolismo , Células Cultivadas , Cloranfenicol O-Acetiltransferasa/biosíntesis , Secuencia de Consenso , Metilación de ADN , Cartilla de ADN , Proteínas de Unión al ADN/aislamiento & purificación , Desoxirribonucleasa I , Cinética , Masculino , Datos de Secuencia Molecular , Factores de Transcripción NFI , Proteínas Nucleares , Oligodesoxirribonucleótidos , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/biosíntesis , Factores de Transcripción/metabolismo , Transfección , Rayos Ultravioleta , Proteína 1 de Unión a la Caja Y
9.
Mol Cell Biol ; 17(7): 3556-65, 1997 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9199291

RESUMEN

To study the mechanisms by which catecholamines regulate hepatocyte proliferation after partial hepatectomy (PHX), hepatocytes were isolated from adult male rats 24 h after sham operation or two-thirds PHX and treated with catecholamines and other agonists. In freshly isolated sham cells, p42 mitogen-activated protein (MAP) kinase activity was stimulated by the alpha1-adrenergic agonist phenylephrine (PHE). Activation of p42 MAP kinase by growth factors was blunted by pretreatment of sham hepatocytes with glucagon but not by that with the beta2-adrenergic agonist isoproterenol (ISO). In PHX cells, the ability of PHE to activate p42 MAP kinase was dramatically reduced, whereas ISO became competent to inhibit p42 MAP kinase activation. PHE treatment of sham but not PHX and ISO treatment of PHX but not sham hepatocytes also activated the stress-activated protein (SAP) kinases p46/54 SAP kinase and p38 SAP kinase. These data demonstrate that an alpha1- to beta2-adrenergic receptor switch occurs upon PHX and results in an increase in SAP kinase versus MAP kinase signaling by catecholamines. In primary cultures of hepatocytes, ISO treatment of PHX but not sham cells inhibited [3H]thymidine incorporation. In contrast, PHE treatment of sham but not PHX cells stimulated [3H]thymidine incorporation, which was reduced by approximately 25 and approximately 95% with specific inhibitors of p42 MAP kinase and p38 SAP kinase function, respectively. Inhibition of the p38 SAP kinase also dramatically reduced basal [3H]thymidine incorporation. These data suggest that p38 SAP kinase plays a permissive role in liver regeneration. Alterations in the abilities of catecholamines to modulate the activities of protein kinase A and the MAP and SAP kinase pathways may represent one physiological mechanism by which these agonists can regulate hepatocyte proliferation after PHX.


Asunto(s)
Agonistas Adrenérgicos/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Regeneración Hepática , Hígado/fisiología , Proteínas Quinasas Activadas por Mitógenos , Receptores Adrenérgicos alfa 1/fisiología , Receptores Adrenérgicos beta 2/fisiología , Estrés Fisiológico/fisiopatología , Animales , División Celular , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos , Hígado/citología , Masculino , Fosforilasas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-raf , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos
10.
Br J Pharmacol ; 173(7): 1116-27, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26076890

RESUMEN

UNLABELLED: Increasing evidence suggests that an overactive endocannabinoid system (ECS) may contribute to the development of diabetes by promoting energy intake and storage, impairing both glucose and lipid metabolism, by exerting pro-apoptotic effects in pancreatic beta cells and by facilitating inflammation in pancreatic islets. Furthermore, hyperglycaemia associated with diabetes has also been implicated in triggering perturbations of the ECS amplifying the pathological processes mentioned above, eventually culminating in a vicious circle. Compelling evidence from preclinical studies indicates that the ECS also influences diabetes-induced oxidative stress, inflammation, fibrosis and subsequent tissue injury in target organs for diabetic complications. In this review, we provide an update on the contribution of the ECS to the pathogenesis of diabetes and diabetic microvascular (retinopathy, nephropathy and neuropathy) and cardiovascular complications. The therapeutic potential of targeting the ECS is also discussed. LINKED ARTICLES: This article is part of a themed section on Endocannabinoids. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v173.7/issuetoc.


Asunto(s)
Complicaciones de la Diabetes/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Endocannabinoides/metabolismo , Animales , Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Cannabidiol/uso terapéutico , Enfermedad Crónica , Complicaciones de la Diabetes/prevención & control , Diabetes Mellitus Tipo 2/prevención & control , Humanos , Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Obesidad/metabolismo , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/metabolismo
11.
J Am Coll Cardiol ; 38(7): 2048-54, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11738314

RESUMEN

OBJECTIVES: We sought to determine whether endocannabinoids influence hemodynamic variables in experimental models of acute myocardial infarction (MI). BACKGROUND: Hypotension and cardiogenic shock are common complications in acute MI. Cannabinoids are strong vasodilators, and endocannabinoids are involved in hypotension in hemorrhagic and septic shock. METHODS: The early effect of left coronary artery ligation on hemodynamic variables was measured in rats pretreated with the selective cannabinoid(1) receptor (CB(1)) antagonist SR141716A (herein referred to as SR, 6.45 micromol/kg body weight intravenously) or vehicle. Endocannabinoids produced in monocytes and platelets were quantified by liquid chromatography/mass spectrometry (LC/MS), and their effects on blood pressure and vascular reactivity were determined. RESULTS: After MI, mean arterial pressure (MAP) dropped from 126 +/- 2 mm Hg to 76 +/- 3 mm Hg in control rats, whereas the decline in blood pressure was smaller (from 121 +/- 3 mm Hg to 108 +/- 7 mm Hg, p < 0.01) in rats pretreated with SR. SR increased the tachycardia that follows MI (change [Delta] in heart rate [HR] = 107 +/- 21 beats/min vs. 49 +/- 9 beats/min in control rats, p < 0.05). The MI sizes were the same in control rats and SR-treated rats. Circulating monocytes and platelets isolated 30 min after MI only decreased MAP when injected into untreated rats (DeltaMAP = -20 +/- 5 mm Hg), but not in SR-pretreated rats. The endocannabinoids anandamide and 2-arachidonyl glycerol were detected in monocytes and platelets isolated after MI, but not in cells from sham rats. Survival rates at 2 h after MI were 70% for control rats and 36% for SR-treated rats (p < 0.05). Endothelium-dependent arterial relaxation was attenuated in SR-treated rats (maximal relaxation: 44 +/- 3% [p < 0.01] vs. 70 +/- 3% in control rats) and further depressed by SR treatment (24 +/- 5%, p < 0.01 vs. MI placebo). CONCLUSIONS: Cannabinoids generated in monocytes and platelets contribute to hypotension in acute MI. Cannabinoid(1) receptor blockade restores MAP but increases 2-h mortality, possibly by impairing endothelial function.


Asunto(s)
Ácidos Araquidónicos/fisiología , Glicéridos/fisiología , Hipotensión/fisiopatología , Infarto del Miocardio/fisiopatología , Choque Cardiogénico/fisiopatología , Animales , Moduladores de Receptores de Cannabinoides , Endocannabinoides , Femenino , Alcamidas Poliinsaturadas , Ratas , Ratas Wistar , Receptores de Cannabinoides , Receptores de Droga/fisiología , Vasodilatación/fisiología
12.
J Mol Med (Berl) ; 76(12): 824-36, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9846953

RESUMEN

Marijuana is a widely abused recreational drug well known for its psychoactive properties. Cannabinoids, the active ingredients of marijuana, elicit their neurobehavioral effects by interacting with the CB1 cannabinoid receptor subtype, expressed primarily in the brain but also present in some peripheral tissues. A second receptor subtype, the CB2 receptor, is expressed on cells of the immune system and is thought to be responsible for the immunosuppressant effects of cannabinoids. Recently, endogenous lipidlike substances have been identified, including arachidonyl ethanolamide (anandamide) and 2-arachidonyl glyceride, that bind to cannabinoid receptors and mimic many of the neurobehavioral effects of plant-derived cannabinoids. Both plant-derived cannabinoids and the endogenous ligands have been shown to elicit hypotension and bradycardia via activation of peripherally located CB1 receptors. Possible underlying mechanisms include presynaptic CB1 receptor mediated inhibition of norepinephrine release from peripheral sympathetic nerve terminals, and/or direct vasodilation via activation of vascular cannabinoid receptors. The latter may also be the target of endocannabinoids of vascular endothelial origin. Recent studies indicate that a peripheral endogenous cannabinoid system in circulating macrophages and platelets is activated in hemorrhagic and septic shock and may contribute to the hypotension associated with these conditions via activation of vascular cannabinoid receptors. The potential role of this mechanism in human shock conditions is under investigation.


Asunto(s)
Cannabinoides/farmacología , Corazón/efectos de los fármacos , Choque/metabolismo , Animales , Moduladores de Receptores de Cannabinoides , Cannabinoides/metabolismo , Humanos , Estructura Molecular , Plantas/química , Receptores de Cannabinoides , Receptores de Droga/metabolismo
13.
Handb Exp Pharmacol ; (168): 599-625, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16596789

RESUMEN

Cannabinoids and their synthetic and endogenous analogs affect a broad range of physiological functions, including cardiovascular variables, the most important component of their effect being profound hypotension. The mechanisms of the cardiovascular effects of cannabinoids in vivo are complex and may involve modulation of autonomic outflow in both the central and peripheral nervous systems as well as direct effects on the myocardium and vasculature. Although several lines of evidence indicate that the cardiovascular depressive effects of cannabinoids are mediated by peripherally localized CB1 receptors, recent studies provide strong support for the existence of as-yet-undefined endothelial and cardiac receptor(s) that mediate certain endocannabinoid-induced cardiovascular effects. The endogenous cannabinoid system has been recently implicated in the mechanism of hypotension associated with hemorrhagic, endotoxic, and cardiogenic shock, and advanced liver cirrhosis. Furthermore, cannabinoids have been considered as novel antihypertensive agents. A protective role of endocannabinoids in myocardial ischemia has also been documented. In this chapter, we summarize current information on the cardiovascular effects of cannabinoids and highlight the importance of these effects in a variety of pathophysiological conditions.


Asunto(s)
Cannabinoides/farmacología , Sistema Cardiovascular/efectos de los fármacos , Animales , Moduladores de Receptores de Cannabinoides/fisiología , Humanos , Hipertensión/etiología , Contracción Miocárdica/efectos de los fármacos , Daño por Reperfusión Miocárdica/etiología , Receptor Cannabinoide CB1/fisiología , Choque Séptico/etiología , Canales Catiónicos TRPV/fisiología , Vasodilatación/efectos de los fármacos
14.
Endocrinology ; 115(6): 2481-3, 1984 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-6094160

RESUMEN

In various forms of shock, TRH is equivalent to naloxone in reversing the hypotension and improving the survival rate. The present findings indicate that in spontaneously hypertensive rats (SHR), TRH has another naloxone-like effect in antagonizing the antihypertensive response to clonidine and alpha-methyldopa. When given during the hypotensive response to alpha-methyldopa, both naloxone and TRH produce a pressor response. While this effect of naloxone is blocked by prazosin, the effect of TRH is not influenced by prazosin or hexamethonium but is inhibited by a vasopressin pressor antagonist. This suggests that the pressor response to naloxone is mediated by the sympathetic nervous system, whereas the similar action of TRH is independent of sympatho-adrenomedullary functions and it is mediated by vasopressin.


Asunto(s)
Antihipertensivos/antagonistas & inhibidores , Clonidina/antagonistas & inhibidores , Metildopa/antagonistas & inhibidores , Naloxona/farmacología , Receptores Adrenérgicos alfa/efectos de los fármacos , Hormona Liberadora de Tirotropina/farmacología , Animales , Fenómenos Biomecánicos , Masculino , Ratas , Ratas Endogámicas SHR , Estimulación Química
15.
Endocrinology ; 118(5): 1814-22, 1986 May.
Artículo en Inglés | MEDLINE | ID: mdl-2938932

RESUMEN

The interaction between clonidine and opiate receptor antagonists on arterial blood pressure (BP) and heart rate were examined in normotensive Wistar Kyoto (WKY) and spontaneously hypertensive rats (SHR). In conscious SHR, the hypotension and bradycardia caused by clonidine, 5 micrograms/kg iv, were significantly attenuated by naltrexone, 2 mg/kg ip. In urethane-anesthetized SHR, the reduction in mean BP and heart rate in response to 5 nmol clonidine microinjected into the nucleus of the solitary tract (NTS), were similarly inhibited after intra-NTS microinjection of 100 ng DL-naloxone but not after the same dose of D-naloxone. Neonatal treatment of SHR by monosodium glutamate (MSG) markedly reduced the beta-endorphin (BE) but not the leucin-enkephalin content of the arcuate nucleus and the NTS. MSG treatment did not affect the basal BP of these animals, but significantly reduced the hypotensive effect of clonidine and eliminated its susceptibility to opiate antagonists in both conscious and anesthetized SHR. In conscious and anesthetized WKY, the cardiovascular effects of clonidine were smaller than in SHR and were unaffected by naloxone or naltrexone. Neonatal treatment of WKY with MSG reduced the BE content of the arcuate nucleus but not of the NTS. MSG treatment of WKY did not influence either basal BP or the cardiovascular effects of clonidine, and the latter remained unaffected by opiate antagonists. These findings support the hypothesis that in SHR, but not in WKY, the centrally mediated cardiovascular effects of clonidine are partially mediated by the release of a BE-like opioid. They also strongly suggest that the site of both the release and the action of this opioid is in the NTS.


Asunto(s)
Animales Recién Nacidos/fisiología , Clonidina/farmacología , Endorfinas/fisiología , Glutamatos/farmacología , Hemodinámica/efectos de los fármacos , Hipertensión/fisiopatología , Glutamato de Sodio/farmacología , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/fisiopatología , Presión Sanguínea/efectos de los fármacos , Endorfinas/metabolismo , Encefalina Leucina/metabolismo , Frecuencia Cardíaca/efectos de los fármacos , Bulbo Raquídeo/efectos de los fármacos , Bulbo Raquídeo/fisiopatología , Naloxona/farmacología , Naltrexona/farmacología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , betaendorfina
16.
Endocrinology ; 120(3): 1073-8, 1987 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-2948818

RESUMEN

In conscious, chronically cannulated, unrestrained rats, systemic administration of catecholamines increases the plasma levels of beta-endorphin-like immunoreactivity (beta Ei). In euthyroid rats, this effect is mediated by both alpha 1 and beta-adrenergic receptors; the rise in plasma beta Ei caused by isoproterenol is blocked by 1 mg/kg propranolol, and the similar effects of norepinephrine and phenylephrine are blocked by 0.1 mg/kg prazosin. Both types of responses are completely suppressed by a 4-h pretreatment of rats with 0.1 mg/kg dexamethasone, indicating the anterior pituitary origin of the beta Ei released. Prior sectioning of the pituitary stalk does not significantly reduce the response to either phenylephrine or isoproterenol, suggesting that both agents act directly on the pituitary. Hypothyroidism induced by surgical thyroidectomy does not influence the beta Ei response to isoproterenol, which remains sensitive to block by propranolol or suppression by dexamethasone. However, neither norepinephrine nor phenylephrine is able to increase plasma beta Ei in the hypothyroid animals. Both isoproterenol and phenylephrine remain fully effective in rats made hyperthyroid by daily injections of 40 micrograms/kg T3 for 4 days. We propose that in unstressed rats catecholamines increase plasma beta Ei by a direct action on the anterior pituitary via either alpha 1- or beta-adrenergic receptors, and that expression of the alpha 1-, but not the beta-adrenergic response is regulated by thyroid hormones.


Asunto(s)
Endorfinas/metabolismo , Isoproterenol/farmacología , Norepinefrina/farmacología , Fenilefrina/farmacología , Adenohipófisis/metabolismo , Prazosina/farmacología , Propranolol/farmacología , Glándula Tiroides/fisiología , Tiroidectomía , Animales , Dexametasona/farmacología , Endorfinas/sangre , Masculino , Adenohipófisis/efectos de los fármacos , Ratas , Ratas Endogámicas , betaendorfina
17.
Endocrinology ; 129(2): 1116-8, 1991 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1649740

RESUMEN

Steady state levels of the mRNAs for alpha 1B, beta 1- and beta 2-adrenergic receptors (alpha 1BAR, beta 1AR, beta 2AR) were quantified by DNA excess solution hybridization assays in the heart, lungs, and liver of rats. Tissues for RNA extraction were obtained from euthyroid and thyroidectomized rats and from thyroidectomized rats treated with a single dose of thyroxine. Thyroidectomy resulted in significant decreases in beta 1AR and beta 2AR mRNAs in heart and lung and alpha 1BAR mRNA in liver, whereas the levels of beta 2AR mRNA in liver and alpha 1BAR mRNA in heart and lung were significantly increased. All these changes were reversed within 20 hours of a single s.c. injection of 1 mg/kg thyroxine. These findings indicate for the first time that thyroid state regulates mRNA levels for adrenergic receptors, and that this regulation is tissue- and receptor-specific. The changes in adrenergic receptor mRNAs correlate with and probably underlie the well documented, thyroid-dependent changes in the cellular densities and physiological reactivities of adrenergic receptors.


Asunto(s)
Regulación de la Expresión Génica , Hipotiroidismo/metabolismo , ARN Mensajero/metabolismo , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos beta/genética , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Hígado/metabolismo , Pulmón/metabolismo , Masculino , Miocardio/metabolismo , Hibridación de Ácido Nucleico , Especificidad de Órganos , Ratas , Ratas Endogámicas , Tiroidectomía , Tiroxina/farmacología
18.
Hypertension ; 33(1 Pt 2): 429-34, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9931142

RESUMEN

Cannabinoids, including the endogenous ligand anandamide (arachidonyl ethanolamide), elicit pronounced hypotension in rats via activation of peripherally located CB1 cannabinoid receptors, which have been also implicated in endotoxin (lipopolysaccharide [LPS])-induced hypotension. The present study was designed to test the role of vascular CB1 receptors in cannabinoid- and endotoxin-induced mesenteric vasodilation. In the isolated, buffer-perfused rat mesenteric arterial bed precontracted with phenylephrine, anandamide induced long-lasting (up to 60 minutes) dose-dependent vasodilation (ED50: 79+/-3 nmol; maximal relaxation: 77+/-2%), inhibited by 0.5 to 5.0 micromol/L of the selective CB1 receptor antagonist SR141716A. Low doses of the calcium ionophore ionomycin also caused mesenteric vasodilation inhibited by SR141716A. The metabolically stable analogue R-methanandamide elicited mesenteric vasodilation (ED50: 286+/-29 nmol), whereas the potent synthetic CB1 receptor agonists WIN 55212-2 and HU-210 caused no change in vascular tone or only a minor dilator effect not affected by SR141716A, respectively. The endogenous ligand 2-arachidonyl glycerol caused no change in vascular tone, whereas Delta9-tetrahydrocannabinol and arachidonic acid caused mesenteric vasoconstriction. After endothelial denudation, the dilator response to anandamide was slightly reduced and was no longer inhibited by SR141716A. In preparations from LPS-pretreated rats, SR141716A alone caused a significant and prolonged increase in perfusion pressure, whereas it had no such effect in control preparations perfused in vitro with or without LPS or after endothelial denudation in preparations from rats pretreated with LPS. We conclude that anandamide-induced mesenteric vasodilation is mediated by an endothelially located SR141716A-sensitive "anandamide receptor" distinct from CB1 cannabinoid receptors and that activation of such receptors by an endocannabinoid, possibly anandamide, contributes to LPS-induced mesenteric vasodilation in vivo.


Asunto(s)
Ácidos Araquidónicos/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Endotelio Vascular/fisiología , Arterias Mesentéricas/fisiología , Músculo Liso Vascular/fisiología , Vasodilatación/fisiología , Animales , Ácido Araquidónico/farmacología , Moduladores de Receptores de Cannabinoides , Dronabinol/farmacología , Endocannabinoides , Glicéridos/farmacología , Ligandos , Masculino , Arterias Mesentéricas/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Piperidinas/farmacología , Alcamidas Poliinsaturadas , Pirazoles/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Droga/antagonistas & inhibidores , Receptores de Droga/fisiología , Rimonabant , Vasoconstricción , Vasodilatación/efectos de los fármacos
19.
Hypertension ; 29(5): 1204-10, 1997 May.
Artículo en Inglés | MEDLINE | ID: mdl-9149688

RESUMEN

We previously showed that in anesthetized rats anandamide elicits bradycardia and a triphasic blood pressure response: transient hypotension secondary to a vagally mediated bradycardia, followed by a brief pressor and prolonged depressor response, the latter two effects being similar to those of delta 9-tetrahydrocannabinol (THC). The prolonged depressor but not the pressor response was reduced after alpha-adrenergic receptor blockade or cervical spinal cord transection and was inhibited by the cannabinoid type 1 (CB1) receptor antagonist SR141716A, suggesting CB1 receptor-mediated sympathoinhibition as the underlying mechanism. Here we examined the relationship between sympathetic tone and the cardiovascular effects of anandamide by testing these effects in both conscious and anesthetized, normotensive and spontaneously hypertensive rats. In urethane-anesthetized normotensive rats, SR141716A inhibited the prolonged depressor and bradycardic effects of anandamide and THC with similar potency, whereas it did not affect the pressor response to either agent. Anadamide caused similar hypotension in spontaneously breathing and in paralyzed, mechanically ventilated rats, suggesting that the hypotension is not secondary to respiratory effects. In conscious normotensive rats, anandamide elicited transient vagal activation and a brief pressor response, but the prolonged hypotensive component was absent. SR141716A potentiated and prolonged the brief pressor response to anandamide, suggesting that the depressor response may have been masked by an increased pressor response. All three phases of the anadamide response were present in both anesthetized and conscious spontaneously hypertensive rats, and the hypotensive component, inhibited by SR141716A in both, was more prolonged in the absence (> 50 minutes) than the presence (10 to 15 minutes) of anesthesia. We conclude that anandamide causes a non-CB1 receptor-mediated pressor and a CB1 receptor-mediated prolonged depressor response. The depressor response can be elicited in both conscious and anesthetized animals, but its magnitude depends on preexisting sympathetic tone.


Asunto(s)
Ácidos Araquidónicos/administración & dosificación , Presión Sanguínea/efectos de los fármacos , Bloqueadores de los Canales de Calcio/administración & dosificación , Frecuencia Cardíaca/efectos de los fármacos , Hipertensión/fisiopatología , Anestesia , Animales , Relación Dosis-Respuesta a Droga , Endocannabinoides , Inyecciones Intravenosas , Masculino , Alcamidas Poliinsaturadas , Ratas , Ratas Endogámicas SHR , Ratas Sprague-Dawley
20.
Hypertension ; 28(4): 682-6, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8843898

RESUMEN

We studied the effects of the endogenous cannabinoid ligand anandamide on blood pressure, single unit activity of barosensitive neurons in the rostral ventrolateral medulla, and postganglionic splanchnic sympathetic nerve discharge in urethane-anesthetized rats. In rats with an intact baroreflex, an intravenous bolus of 4 mg/kg anandamide caused a triphasic blood pressure response: transient hypotension, followed by a brief pressor and more prolonged depressor phase. Anandamide evoked a "primary" increase in neuronal firing coincident with its pressor effect and a "secondary," baroreflex-mediated rise coincident with its depressor effect at both sites. Pretreatment of rats with phentolamine or trimethaphan did not inhibit either the pressor response or the primary increase in splanchnic nerve discharge elicited by anandamide. In barodenervated rats, electrical stimulation of the rostral ventrolateral medulla increased blood pressure and splanchnic nerve discharge. Anandamide treatment blunted the rise in blood pressure without affecting the increase in splanchnic nerve discharge. Anandamide did not affect the rise in blood pressure in response to an intravenous bolus dose of phenylephrine. The results indicate that (1) the brief pressor response to anandamide is not sympathetically mediated, and (2) the prolonged hypotensive response to anandamide is not initiated in the central nervous system, in ganglia, or at postsynaptic adrenergic receptors but is due to a presynaptic action that inhibits norepinephrine release from sympathetic nerve terminals in the heart and vasculature.


Asunto(s)
Ácidos Araquidónicos/farmacología , Presión Sanguínea/efectos de los fármacos , Cannabinoides/farmacología , Anestesia , Animales , Dronabinol/farmacología , Endocannabinoides , Masculino , Alcamidas Poliinsaturadas , Presorreceptores/fisiología , Ratas , Ratas Sprague-Dawley , Sistema Nervioso Simpático/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA