Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
1.
Curr Protoc Chem Biol ; 6(1): 39-51, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24652622

RESUMEN

Label-free cell-based assays offer a powerful approach to drug discovery and compound profiling for endogenously expressed receptors in a variety of cell types, including primary and stem cells. Dynamic mass redistribution (DMR) responses in whole cells following receptor stimulation provide phenotypic activity profiles that are readily amenable to evaluation of compound pharmacology. Protocols are provided in this unit to obtain DMR response profiles in adherent and suspension cells, and then to use known tool compounds to delineate the biology of the underlying signaling pathways from the information-rich kinetic traces that are recorded.


Asunto(s)
Bioensayo/métodos , Descubrimiento de Drogas/métodos , Animales , Células CHO , Línea Celular Tumoral , Cricetulus , Interpretación Estadística de Datos , Humanos , Cultivo Primario de Células , Receptores de Droga/agonistas , Receptores de Droga/antagonistas & inhibidores , Receptores de Droga/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
2.
J Thorac Cardiovasc Surg ; 140(5): 1153-9, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20804990

RESUMEN

OBJECTIVE: Animal and human myocytes demonstrate significant swelling and reduced contractility during exposure to stress (metabolic inhibition, hyposmotic stress, or hyperkalemic cardioplegia), and these detrimental consequences may be inhibited by the addition of diazoxide (adenosine triphosphate-sensitive potassium channel opener) via an unknown mechanism. Both SUR1 and SUR2A subunits have been localized to the heart, and mouse sarcolemmal adenosine triphosphate-sensitive potassium channels are composed of SUR2A/Kir6.2 subunits in the ventricle and SUR1/Kir6.2 subunits in the atria. This study was performed to localize the mechanism of diazoxide by direct probing of sarcolemmal adenosine triphosphate-sensitive potassium channel current and by genetic deletion of channel subunits. METHODS: Sarcolemmal adenosine triphosphate-sensitive potassium channel current was recorded in isolated wild-type ventricular mouse myocytes during exposure to Tyrode's solution, Tyrode's + 100 µmol/L diazoxide, hyperkalemic cardioplegia, cardioplegia + diazoxide, cardioplegia + 100 µmol/L pinacidil, or metabolic inhibition using whole-cell voltage clamp (N = 7-12 cells per group). Ventricular myocyte volume was measured from SUR1(-/-) and wild-type mice during exposure to control solution, hyperkalemic cardioplegia, or cardioplegia + 100 µmol/L diazoxide (N = 7-10 cells per group). RESULTS: Diazoxide did not increase sarcolemmal adenosine triphosphate-sensitive potassium current in wild-type myocytes, although they demonstrated significant swelling during exposure to cardioplegia that was prevented by diazoxide. SUR1(-/-) myocytes also demonstrated significant swelling during exposure to cardioplegia, but this was not altered by diazoxide. CONCLUSIONS: Diazoxide does not open the ventricular sarcolemmal adenosine triphosphate-sensitive potassium channel but provides volume homeostasis via an SUR1-dependent pathway in mouse ventricular myocytes, supporting a mechanism of action distinct from sarcolemmal adenosine triphosphate-sensitive potassium channel activation.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/agonistas , Cardiotónicos/farmacología , Tamaño de la Célula/efectos de los fármacos , Diazóxido/farmacología , Canales KATP/agonistas , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Canales de Potasio de Rectificación Interna/agonistas , Receptores de Droga/agonistas , Sarcolema/efectos de los fármacos , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Soluciones Cardiopléjicas/farmacología , Femenino , Homeostasis , Hiperpotasemia/metabolismo , Canales KATP/deficiencia , Canales KATP/genética , Canales KATP/metabolismo , Masculino , Potenciales de la Membrana , Ratones , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Presión Osmótica , Técnicas de Placa-Clamp , Pinacidilo/farmacología , Canales de Potasio de Rectificación Interna/deficiencia , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , Receptores de Droga/deficiencia , Receptores de Droga/genética , Receptores de Droga/metabolismo , Sarcolema/metabolismo , Receptores de Sulfonilureas
3.
J Cardiovasc Pharmacol ; 56(4): 345-53, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20505525

RESUMEN

We sought to explore new strategies targeting SUR2B/Kir6.1, a subtype of adenosine triphosphate (ATP)-sensitive potassium channels (KATP), against pressure overload-induced heart failure. The effects of natakalim, a SUR2B/Kir6.1 selective channel opener, on progression of cardiac remodeling were investigated. Pressure overload-induced heart failure was induced in Wistar rats by abdominal aortic banding. The effects of natakalim (1, 3, and 9 mg·kg⁻¹·d⁻¹ for 10 weeks) on myocardial hypertrophy and heart failure, cardiac histology, vasoactive compounds, and gene expression were assessed. Ten weeks after the onset of pressure overload, natakalim treatment potently inhibited cardiac hypertrophy and prevented heart failure. Natakalim remarkably inhibited the changes of left ventricular hemodynamic parameters and reversed the increase of heart mass index, left ventricular weight index, and lung weight index. Histological examination demonstrated that there was no significant hypertrophy or fibrosis in pressure-overloaded hearts of natakalim-treated rats. Ultrastructural examination of hearts revealed well-organized myofibrils with mitochondria grouped along the periphery of longitudinally oriented fibers in rats from the natakalim group. The content of serum nitric oxide and plasma prostacyclin was increased, whereas that of plasma endothelin-1 and cardiac tissue hydroxyproline and atrial and B-type natriuretic peptide messenger RNA was downregulated in natakalim-treated rats. Natakalim at 0.01-100 µM had no effects on isolated working hearts derived from Wistar rats; however, natakalim had endothelium-dependent vasodilatory effects on the isolated tail artery helical strips precontracted with norepinephrine. These results indicate that natakalim reduces heart failure caused by pressure overloading by activating the SUR2B/Kir6.1 KATP channel subtype and protecting against endothelial dysfunction.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Compuestos Alílicos/farmacología , Fármacos Cardiovasculares/farmacología , Endotelio Vascular/efectos de los fármacos , Insuficiencia Cardíaca/prevención & control , Hipertensión/complicaciones , Canales de Potasio de Rectificación Interna/metabolismo , Propilaminas/farmacología , Receptores de Droga/metabolismo , Remodelación Ventricular/efectos de los fármacos , Transportadoras de Casetes de Unión a ATP/agonistas , Compuestos Alílicos/uso terapéutico , Animales , Presión Sanguínea/efectos de los fármacos , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/etiología , Cardiomegalia/fisiopatología , Fármacos Cardiovasculares/uso terapéutico , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Endotelina-1/sangre , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Epoprostenol/sangre , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/fisiopatología , Hipertensión/fisiopatología , Técnicas In Vitro , Canales KATP , Masculino , Miocardio/metabolismo , Miocardio/patología , Óxido Nítrico/sangre , Canales de Potasio de Rectificación Interna/agonistas , Propilaminas/uso terapéutico , Ratas , Ratas Wistar , Receptores de Droga/agonistas , Receptores de Sulfonilureas , Cola (estructura animal)/irrigación sanguínea , Vasodilatadores/farmacología , Vasodilatadores/uso terapéutico
4.
Anesthesiology ; 112(3): 623-30, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20179498

RESUMEN

BACKGROUND: Adenosine triphosphate-sensitive potassium (KATP) channels in brain are involved in neuroprotective mechanisms. Pharmacologic activation of these channels is seen as beneficial, but clinical exploitation by using classic K channel openers is hampered by their inability to cross the blood-brain barrier. This is different with the inhalational anesthetic xenon, which recently has been suggested to activate KATP channels; it partitions freely into the brain. METHODS: To evaluate the type and mechanism of interaction of xenon with neuronal-type KATP channels, these channels, consisting of Kir6.2 pore-forming subunits and sulfonylurea receptor-1 regulatory subunits, were expressed in HEK293 cells and whole cell, and excised patch-clamp recordings were performed. RESULTS: Xenon, in contrast to classic KATP channel openers, acted directly on the Kir6.2 subunit of the channel. It had no effect on the closely related, adenosine triphosphate (ATP)-regulated Kir1.1 channel and failed to activate an ATP-insensitive mutant version of Kir6.2. Furthermore, concentration-inhibition curves for ATP obtained from inside-out patches in the absence or presence of 80% xenon revealed that xenon reduced the sensitivity of the KATP channel to ATP. This was reflected in an approximately fourfold shift of the concentration causing half-maximal inhibition (IC50) from 26 +/- 4 to 96 +/- 6 microm. CONCLUSIONS: Xenon represents a novel KATP channel opener that increases KATP currents independently of the sulfonylurea receptor-1 subunit by reducing ATP inhibition of the channel. Through this action and by its ability to readily partition across the blood-brain barrier, xenon has considerable potential in clinical settings of neuronal injury, including stroke.


Asunto(s)
Anestésicos por Inhalación/farmacología , Canales KATP/agonistas , Xenón/farmacología , Transportadoras de Casetes de Unión a ATP/agonistas , Transportadoras de Casetes de Unión a ATP/genética , Algoritmos , Animales , Línea Celular , Electrofisiología , Humanos , Canales KATP/genética , Ratones , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/agonistas , Canales de Potasio de Rectificación Interna/genética , Receptores de Droga/agonistas , Receptores de Droga/genética , Receptores de Sulfonilureas , Transfección
5.
J Biol Chem ; 284(12): 7951-9, 2009 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-19151370

RESUMEN

The beta-cell ATP-sensitive potassium (K(ATP)) channel composed of sulfonylurea receptor SUR1 and potassium channel Kir6.2 serves a key role in insulin secretion regulation by linking glucose metabolism to cell excitability. Mutations in SUR1 or Kir6.2 that decrease channel function are typically associated with congenital hyperinsulinism, whereas those that increase channel function are associated with neonatal diabetes. Here we report that two hyperinsulinism-associated SUR1 missense mutations, R74W and E128K, surprisingly reduce channel inhibition by intracellular ATP, a gating defect expected to yield the opposite disease phenotype neonatal diabetes. Under normal conditions, both mutant channels showed poor surface expression due to retention in the endoplasmic reticulum, accounting for the loss of channel function phenotype in the congenital hyperinsulinism patients. This trafficking defect, however, could be corrected by treating cells with the oral hypoglycemic drugs sulfonylureas, which we have shown previously to act as small molecule chemical chaperones for K(ATP) channels. The R74W and E128K mutants thus rescued to the cell surface paradoxically exhibited ATP sensitivity 6- and 12-fold lower than wild-type channels, respectively. Further analyses revealed a nucleotide-independent decrease in mutant channel intrinsic open probability, suggesting the mutations may reduce ATP sensitivity by causing functional uncoupling between SUR1 and Kir6.2. In insulin-secreting cells, rescue of both mutant channels to the cell surface led to hyperpolarized membrane potentials and reduced insulin secretion upon glucose stimulation. Our results show that sulfonylureas, as chemical chaperones, can dictate manifestation of the two opposite insulin secretion defects by altering the expression levels of the disease mutants.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Chaperonas Moleculares/farmacología , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , Receptores de Droga/genética , Receptores de Droga/metabolismo , Compuestos de Sulfonilurea/farmacología , Transportadoras de Casetes de Unión a ATP/agonistas , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Sustitución de Aminoácidos , Animales , Línea Celular , Hiperinsulinismo Congénito/genética , Hiperinsulinismo Congénito/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Glucosa/metabolismo , Humanos , Secreción de Insulina , Mutación , Mutación Missense , Canales de Potasio de Rectificación Interna/agonistas , Ratas , Receptores de Droga/agonistas , Receptores de Sulfonilureas
6.
Cell Physiol Biochem ; 18(4-5): 187-98, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17167224

RESUMEN

The interstitial cells of Cajal (ICC) are pacemaker cells in gastrointestinal tract and generate an electrical rhythm in gastrointestinal muscles. We investigated the possibility that PGE(2) might affect the electrical properties of cultured ICC by activating ATP-dependent K(+) channels and, the EP receptor subtypes and the subunits of ATP-dependent K(+) channels involved in these activities were identified. In addition, the regulation of intracellular Ca(2+) ([Ca(2+)](i)) mobilization may be involved the action of PGE(2) on ICC. Treatments of ICC with PGE(2) inhibited electrical pacemaker activities in the same manner as pinacidil, an ATP-dependent K(+) channel opener and PGE(2) had only a dose-dependent effect. Using RT-PCR technique, we found that ATP-dependent K(+) channels exist in ICC and that these are composed of K(ir) 6.2 and SUR 2B subunits. To characterize the specific membrane EP receptor subtypes in ICC, EP receptor agonists and RT-PCR were used: Butaprost (an EP(2) receptor agonist) showed the actions on pacemaker currents in the same manner as PGE(2). However sulprostone (a mixed EP(1) and EP(3) agonist) had no effects. In addition, RT-PCR results indicated the presence of the EP(2) receptor in ICC. To investigate cAMP involvement in the effects of PGE(2) on ICCs, SQ-22536 (an inhibitor of adenylate cyclase) and cAMP assays were used. SQ-22536 did not affect the effect of PGE(2) on pacemaker currents, and PGE(2) did not stimulate cAMP production. Also, we found PGE(2) inhibited the spontaneous [Ca(2+)](i) oscillations in cultured ICC. These observations indicate that PGE(2) alters pacemaker currents by activating the ATP-dependent K(+) channels comprised of K(ir) 6.2-SUR 2B in ICC and this action of PGE(2) are through EP(2) receptor subtype and also the activation of ATP-dependent K(+) channels involves intracellular Ca(2+) mobilization.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/agonistas , Dinoprostona/farmacología , Intestino Delgado/efectos de los fármacos , Canales de Potasio de Rectificación Interna/agonistas , Canales de Potasio/agonistas , Receptores de Droga/agonistas , Receptores de Prostaglandina E/metabolismo , Transportadoras de Casetes de Unión a ATP/análisis , Transportadoras de Casetes de Unión a ATP/genética , Adenina/análogos & derivados , Adenina/farmacología , Adenosina Trifosfato/metabolismo , Inhibidores de Adenilato Ciclasa , Alprostadil/análogos & derivados , Alprostadil/farmacología , Animales , Calcio/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Electrofisiología , Femenino , Intestino Delgado/química , Intestino Delgado/citología , Masculino , Potenciales de la Membrana , Ratones , Ratones Endogámicos BALB C , Canales de Potasio/análisis , Canales de Potasio/genética , Canales de Potasio de Rectificación Interna/análisis , Canales de Potasio de Rectificación Interna/genética , Receptores de Droga/análisis , Receptores de Droga/genética , Subtipo EP2 de Receptores de Prostaglandina E , Receptores de Sulfonilureas
7.
FEBS Lett ; 580(13): 3070-4, 2006 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-16678176

RESUMEN

Gene matching shows that Nischarin is a mouse homologue of human imidazoline receptor antisera-selective (IRAS) protein, a viable candidate of the imidazoline (I1) receptor. Nischarin and IRAS share the functions of enhancing cell survival, growth and migration. Bioinformatics modeling indicates that the IRAS and Nischarin may be transmembrane proteins and the convergence information raises the interesting possibility that Nischarin might serve as the I1-receptor. To test this hypothesis, we developed antibodies against the Nischarin protein, and conducted signal transduction (functional) studies with the I1-receptor agonist rilmenidine in the presence and absence of Nischarin antisense oligodeoxynucleotides (ODNs). NIH3T3 cells transfected with the Nischarin cDNA and incubated with the newly synthesized antibody expressed a 190 kD band. The antibody identified endogenous Nischarin in differentiated PC12 cells around 210 kD, which is consistent with reported findings in other cells of neuronal origin. The immunoflourescence findings showed the targeted protein to be associated with the cell membrane in PC12 cells. Nischarin ODNs abolished the expression of Nischarin in PC12 cells. Equally important, the Nischarin ODNs eliminated the production of MAPK(p42/44), a recognized signal transduction product generated by I1-receptor activation in differentiated PC12 cells. Together, the present findings suggest that Nischarin may serve as the functional I1-receptor or at least share a common signaling pathway in the differentiated PC12 cells.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/fisiología , Receptores de Droga/fisiología , Agonistas alfa-Adrenérgicos/farmacología , Animales , Anticuerpos/inmunología , Receptores de Imidazolina , Péptidos y Proteínas de Señalización Intracelular/análisis , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Células 3T3 NIH , Oligodesoxirribonucleótidos Antisentido/genética , Oligodesoxirribonucleótidos Antisentido/farmacología , Oxazoles/farmacología , Células PC12 , Ratas , Receptores de Droga/agonistas , Rilmenidina
8.
Curr Med Chem ; 13(4): 361-76, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16475928

RESUMEN

ATP sensitive potassium (K(ATP)) channels have important functions in neuroendocrine tissue, in smooth and skeletal muscle and in the heart. In pancreatic beta cells the K(ATP) channels, which are formed by 4 ion channels (Kir6.2) and 4 regulatory sulfonylurea receptors (SUR1), control the glucose stimulated release of insulin. The Kir6.2/SUR1 K(ATP) channels are also present in the brain and in other neuroendocrine tissues. Blockers of Kir6.2/SUR1 channels, e.g. glibenclamide and repaglinide stimulate release of insulin and are used for treatment of type 2 diabetes. Openers of Kir6.2/SUR1 channels, e.g. diazoxide, have in contrast only found limited clinical use in treatment of hypersecretion of insulin associated with certain tumours (insulinoma) and genetic disorders (persistent hyperinsulinemia and hypoglycemia of infancy, PHHI). Recent studies have however, indicated that openers of Kir6.2/SUR1 channels could be useful in treatment of e.g. metabolic disorders and diseases of the CNS. The clinical use of diazoxide has been hampered by its lack of potency and selectivity giving rise to side effects, such as oedema and hirsutism and new selective openers of Kir6.2/SUR1 channels have been pursued. This has provided several structurally diverse series, which include 1,2,4-thiadiazine 1,1-dioxide derivatives, like BPDZ 62, BPDZ 73, NNC 55-0462, NNC 55-0118 and NN414, cyanoguanidines, nitropyrazoles and 4-sulfamoylphenylbenzamides. NN414 has been shown to be a potent and Kir6.2/SUR1 selective K(ATP) channels opener, which inhibits glucose stimulated insulin release in vitro and in vivo and which has beneficial effects on glucose homeostasis in preclinical and clinical studies.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/agonistas , Transportadoras de Casetes de Unión a ATP/efectos de los fármacos , Diazóxido/uso terapéutico , Canales de Potasio de Rectificación Interna/agonistas , Canales de Potasio de Rectificación Interna/efectos de los fármacos , Canales de Potasio/agonistas , Canales de Potasio/efectos de los fármacos , Receptores de Droga/agonistas , Receptores de Droga/efectos de los fármacos , Vasodilatadores/uso terapéutico , Amidas/uso terapéutico , Animales , Benzopiranos/uso terapéutico , Benzotiadiazinas/uso terapéutico , Hiperinsulinismo Congénito/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/fisiopatología , Femenino , Guanidinas , Humanos , Nitrilos/uso terapéutico , Obesidad/tratamiento farmacológico , Obesidad/fisiopatología , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Receptores de Sulfonilureas , Tiadiazinas/uso terapéutico
9.
Eur J Pharmacol ; 514(2-3): 191-9, 2005 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-15910806

RESUMEN

Clonidine (a mixed alpha2-adrenoceptor and imidazoline I1 receptor agonist)-evoked hypotension was associated with dissimilar reductions in c-jun gene expression in the rostral ventrolateral medulla (RVLM) and the nucleus tractus solitarius (NTS) in normotensive rats. In the present study, we investigated the relative contribution of the alpha2-adrenoceptor vs. the imidazoline I1 receptor to the reduction in c-jun gene expression in these two brainstem areas. In conscious spontaneously hypertensive rats (SHRs), equihypotensive doses of three centrally acting hypotensive drugs with different selectivity for the two receptors were administered intracisternally (4 microl) to limit their actions to the brain. As a control, a similar hypotensive response was elicited by i.v. hydralazine. Clonidine (0.5 microg), or alpha-methylnorepinephrine (alpha-MNE, 4 microg), a highly selective alpha2-adrenoceptor agonist, similarly reduced c-jun mRNA expression in the NTS and rostral ventrolateral medulla. In contrast, a similar hypotensive response (-37+/-3.5 mm Hg) caused by the selective imidazoline I1 receptor agonist rilmenidine (25 microg) was associated with reduction in c-jun mRNA expression in the rostral ventrolateral medulla, but not in the NTS. Further, intra-rostral ventrolateral medulla rilmenidine (40 nmol) reduced c-Jun protein expression in rostral ventrolateral medulla and blood pressure and both responses were antagonized by selective imidazoline I1 receptor (efaroxan, 4 nmol), but not alpha2-adrenoceptor (SK&F 86466, 10 nmol) blockade. These results suggest: (1) the c-jun containing neurons in the brainstem are involved in the centrally mediated hypotension elicited by centrally acting antihypertensive agents, and (2) the alpha2-adrenoceptor modulates c-jun gene expression in the NTS and rostral ventrolateral medulla implicated in centrally mediated hypotension, and (3) the imidazoline I1 receptor mediated inhibition of c-jun gene expression in the rostral ventrolateral medulla, but not in the NTS, contributes to the centrally mediated hypotension by the second generation drugs.


Asunto(s)
Tronco Encefálico/metabolismo , Bulbo Raquídeo/metabolismo , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Receptores de Droga/fisiología , Agonistas de Receptores Adrenérgicos alfa 2 , Antagonistas de Receptores Adrenérgicos alfa 2 , Agonistas alfa-Adrenérgicos/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Animales , Antihipertensivos/farmacología , Benzazepinas/farmacología , Benzofuranos/farmacología , Presión Sanguínea/efectos de los fármacos , Tronco Encefálico/efectos de los fármacos , Líquido Cefalorraquídeo/fisiología , Clonidina/farmacología , Estado de Conciencia , Expresión Génica/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Hidralazina/farmacología , Hipotensión/inducido químicamente , Hipotensión/fisiopatología , Imidazoles/farmacología , Receptores de Imidazolina , Inmunohistoquímica , Masculino , Neuronas/efectos de los fármacos , Nordefrin/farmacología , Oxazoles/farmacología , Proteínas Proto-Oncogénicas c-jun/análisis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas SHR , Receptores Adrenérgicos alfa 2/fisiología , Receptores de Droga/agonistas , Receptores de Droga/antagonistas & inhibidores , Rilmenidina , Factores de Tiempo , Vasoconstrictores/farmacología , Vasodilatadores/farmacología
11.
Neurosci Lett ; 370(1): 55-60, 2004 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-15489017

RESUMEN

The Transient Receptor Potential cation channel V1 (TRPV1) is expressed in peripheral nociceptive neurons and is subject to polymodal activation via various agents including capsaicin, noxious heat, low extracellular pH, and direct phosphorylation by protein kinase C (PKC). We have cloned and heterologously expressed mouse TRPV1 (mTRPV1) and characterized its function utilizing FLIPR-based calcium imaging to measure functional responses to various small molecule agonists, low pH and direct phosphorylation via PKC. The various TRPV1 agonists activated mTRPV1 with a rank order of agonist potency of (resiniferatoxin (RTX) = arvanil > capsaicin = olvanil > OLDA > PPAHV) (EC50 values of 0.15+/-0.04 nM, 0.27+/-0.07 nM, 9.1+/-1.2 nM, 3.7+/-0.3 nM, 258+/-105 nM, and 667+/-151 nM, respectively). Additionally, mTRPV1 was activated by either low pH or with addition of the PKC activator phorbol 12-myristate 13-acetate (PMA). The TRPV1 antagonists iodinated-resiniferatoxin (I-RTX) or BCTC were both able to block capsaicin, pH and PKC-induced responses of mTRPV1 (IC50 (I-RTX) = 0.35+/-0.12 nM, 1.9+/-0.7 nM, and 0.80+/-0.68 nM, IC50 (BCTC) = 1.3+/-0.36 nM, 0.59+/-0.16 nM, and 0.37+/-0.15 nM, respectively). However, the antagonist capsazepine was only able to inhibit a capsaicin-evoked response of mTRPV1 with an IC50 of 1426+/-316 nM. Comparable results were achieved with rat TRPV1, while capsazepine blocked all modes of human TRPV1 activation. Thus, the mTRPV1 cation channel has a molecular pharmacological profile more akin to rat TRPV1 than either human or guinea pig TRPV1 and the molecular pharmacology suggests that capsazepine may be an ineffective TRPV1 antagonist for in vivo models of inflammatory pain in the mouse.


Asunto(s)
Canales Iónicos/genética , Receptores de Droga/fisiología , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Capsaicina/agonistas , Capsaicina/farmacología , Línea Celular , Clonación Molecular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Diterpenos/farmacología , Activación Enzimática/efectos de los fármacos , Cobayas , Humanos , Concentración de Iones de Hidrógeno , Concentración 50 Inhibidora , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Canales Iónicos/fisiología , Ratones , Ésteres del Forbol/farmacología , Fosforilación/efectos de los fármacos , ARN Mensajero/biosíntesis , Conejos , Ratas , Receptores de Droga/agonistas , Receptores de Droga/antagonistas & inhibidores , Receptores de Droga/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Canales Catiónicos TRPV , Transfección/métodos
12.
J Physiol ; 561(Pt 2): 459-69, 2004 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-15459245

RESUMEN

In this study we have investigated the voltage dependence of ATP-dependent K+ current (I(K(ATP))) in atrial and ventricular myocytes from hearts of adult rats and in CHO cells expressing Kir6.2 and SUR2A. The current-voltage relation of 2,4-dinitrophenole (DNP) -induced I(K(ATP)) in atrial myocytes and expressed current in CHO cells was linear in a voltage range between 0 and -100 mV. In ventricular myocytes, the background current-voltage relation of which is dominated by a large constitutive inward rectifier (I(K1)), the slope conductance of I(K(ATP)) was reduced at membrane potentials negative to E(K) (around -50 mV), resulting in an outwardly rectifying I-V relation. Overexpression of Kir2.1 by adenoviral gene transfer, a subunit contributing to I(K1) channels, in atrial myocytes resulted in a large I(K1)-like background current. The I-V relation of I(K(ATP)) in these cells showed a reduced slope conductance negative to E(K) similar to ventricular myocytes. In atrial myocytes with an increased background inward-rectifier current through Kir3.1/Kir3.4 channels (I(K(ACh))), irreversibly activated by intracellular loading with GTP-gamma-S, the I-V relation of I(K(ATP)) showed a reduced slope negative to E(K), as in ventricular myocytes and atrial myocytes overexpressing Kir2.1. It is concluded that the voltage dependencies of membrane currents are not only dependent on the molecular composition of the charge-carrying channel complexes but can be affected by the activity of other ion channel species. We suggest that the interference between inward I(K(ATP)) and other inward rectifier currents in cardiac myocytes reflects steady-state changes in K+ driving force due to inward K+ current.


Asunto(s)
Acetilcolina/farmacología , Adenosina Trifosfato/fisiología , Miocitos Cardíacos/fisiología , Canales de Potasio de Rectificación Interna/fisiología , Transportadoras de Casetes de Unión a ATP/agonistas , Transportadoras de Casetes de Unión a ATP/fisiología , Animales , Células CHO , Células Cultivadas , Cricetinae , Canales de Potasio de Conductancia Intermedia Activados por el Calcio , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Miocitos Cardíacos/efectos de los fármacos , Pinacidilo/farmacología , Canales de Potasio/agonistas , Canales de Potasio/fisiología , Canales de Potasio Calcio-Activados/agonistas , Canales de Potasio Calcio-Activados/fisiología , Canales de Potasio de Rectificación Interna/agonistas , Ratas , Receptores de Droga/agonistas , Receptores de Droga/fisiología , Receptores de Sulfonilureas
13.
Eur J Pharmacol ; 501(1-3): 31-9, 2004 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-15464060

RESUMEN

beta-Carbolines (including harmane and pinoline) stimulate insulin secretion by a mechanism that may involve interaction with imidazoline I(3)-receptors but which also appears to be mediated by actions that are additional to imidazoline receptor agonism. Using the MIN6 beta-cell line, we now show that both the imidazoline I(3)-receptor agonist, efaroxan, and the beta-carboline, harmane, directly elevate cytosolic Ca(2+) and increase insulin secretion but that these responses display different characteristics. In the case of efaroxan, the increase in cytosolic Ca(2+) was readily reversible, whereas, with harmane, the effect persisted beyond removal of the agonist and resulted in the development of a repetitive train of Ca(2+)-oscillations whose frequency, but not amplitude, was concentration-dependent. Initiation of the Ca(2+)-oscillations by harmane was independent of extracellular calcium but was sensitive to both dantrolene and high levels (20 mM) of caffeine, suggesting the involvement of ryanodine receptor-gated Ca(2+)-release. The expression of ryanodine receptor-1 and ryanodine receptor-2 mRNA in MIN6 cells was confirmed using reverse transcription-polymerase chain reaction (RT-PCR) and, since low concentrations of caffeine (1 mM) or thimerosal (10 microM) stimulated increases in [Ca(2+)](i), we conclude that ryanodine receptors are functional in these cells. Furthermore, the increase in insulin secretion induced by harmane was attenuated by dantrolene, consistent with the involvement of ryanodine receptors in mediating this response. By contrast, the smaller insulin secretory response to efaroxan was unaffected by dantrolene. Harmane-evoked changes in cytosolic Ca(2+) were maintained by nifedipine-sensitive Ca(2+)-influx, suggesting the involvement of L-type voltage-gated Ca(2+)-channels. Taken together, these data imply that harmane may interact with ryanodine receptors to generate sustained Ca(2+)-oscillations in pancreatic beta-cells and that this effect contributes to the insulin secretory response.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Harmina/análogos & derivados , Harmina/farmacología , Líquido Intracelular/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Receptores de Droga/agonistas , Animales , Señalización del Calcio/fisiología , Línea Celular Tumoral , Receptores de Imidazolina , Líquido Intracelular/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Receptores de Droga/metabolismo
14.
Proc Natl Acad Sci U S A ; 101(37): 13677-82, 2004 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-15365189

RESUMEN

We identified the previously unknown structures of ribosylated imidazoleacetic acids in rat, bovine, and human tissues to be imidazole-4-acetic acid-ribotide (IAA-RP) and its metabolite, imidazole-4-acetic acid-riboside. We also found that IAA-RP has physicochemical properties similar to those of an unidentified substance(s) extracted from mammalian tissues that interacts with imidazol(in)e receptors (I-Rs). ["Imidazoline," by consensus (International Union of Pharmacology), includes imidazole, imidazoline, and related compounds. We demonstrate that the imidazole IAA-RP acts at I-Rs, and because few (if any) imidazolines exist in vivo, we have adopted the term "imidazol(in)e-Rs."] The latter regulate multiple functions in the CNS and periphery. We now show that IAA-RP (i) is present in brain and tissue extracts that exhibit I-R activity; (ii) is present in neurons of brainstem areas, including the rostroventrolateral medulla, a region where drugs active at I-Rs are known to modulate blood pressure; (iii) is present within synaptosome-enriched fractions of brain where its release is Ca(2+)-dependent, consistent with transmitter function; (iv) produces I-R-linked effects in vitro (e.g., arachidonic acid and insulin release) that are blocked by relevant antagonists; and (v) produces hypertension when microinjected into the rostroventrolateral medulla. Our data also suggest that IAA-RP may interact with a novel imidazol(in)e-like receptor at this site. We propose that IAA-RP is a neuroregulator acting via I-Rs.


Asunto(s)
Imidazoles/farmacología , Receptores de Droga/agonistas , Ribosamonofosfatos/farmacología , Médula Suprarrenal/metabolismo , Animales , Anticuerpos/inmunología , Especificidad de Anticuerpos , Ácido Araquidónico/metabolismo , Tronco Encefálico/citología , Calcio/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Hipertensión/inducido químicamente , Imidazoles/química , Imidazoles/inmunología , Receptores de Imidazolina , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Isomerismo , Ligandos , Estructura Molecular , Neuronas/metabolismo , Células PC12 , Ratas , Receptores de Droga/metabolismo , Ribosamonofosfatos/química , Ribosamonofosfatos/inmunología
15.
Neurochem Int ; 45(7): 1005-10, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15337299

RESUMEN

Capsaicin, a pungent ingredient of chili pepper, activates vanilloid receptor subtype 1 (VR1), which is a nonselective cation channel with high Ca(2+) permeability. Although VR1 and its splice variant are highly expressed in sensory neurons, they are expressed in neuronal cells in brain and peripheral non-neuronal cells. In this study, we investigated whether VR1 is expressed in PC12 cells, rat pheochromocytoma. Capsaicin at concentrations above 100 microM induced an increase in intracellular free Ca(2+) concentrations by influx from extracellular spaces, and the effect was blocked by capsazepine, a selective antagonist of VR1. VR1 transcript and protein were detected by reverse transcription-polymerase chain reaction and Western blotting analysis, respectively. Immunocytochemical analysis revealed that VR1 protein was expressed in the cytosol and the plasma membrane of PC12 cells, and treatment with the antisense oligonucleotide for VR1 decreased the expression. VR1 in PC12 cells showed different characters from that in sensory neurons; capsaicin concentration-dependency and heat- and nerve growth factor-sensitivities. These results suggested that VR1 was functionally expressed in PC12 cells. The usefulness of PC12 cell line for studying functions and/or expression of VR1 is discussed.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Receptores de Droga/biosíntesis , Animales , Capsaicina/farmacología , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Células PC12 , Ratas , Receptores de Droga/agonistas , Receptores de Droga/genética
16.
Nature ; 431(7006): 316-20, 2004 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-15372035

RESUMEN

Antagonistic activities of glucagon and insulin control metabolism in mammals, and disruption of this balance underlies diabetes pathogenesis. Insulin-producing cells (IPCs) in the brain of insects such as Drosophila also regulate serum glucose, but it remains unclear whether insulin is the sole hormonal regulator of glucose homeostasis and whether mechanisms of glucose-sensing and response in IPCs resemble those in pancreatic islets. Here we show, by targeted cell ablation, that Drosophila corpora cardiaca (CC) cells of the ring gland are also essential for larval glucose homeostasis. Unlike IPCs, CC cells express Drosophila cognates of sulphonylurea receptor (Sur) and potassium channel (Ir), proteins that comprise ATP-sensitive potassium channels regulating hormone secretion by islets and other mammalian glucose-sensing cells. They also produce adipokinetic hormone, a polypeptide with glucagon-like functions. Glucose regulation by CC cells is impaired by exposure to sulphonylureas, drugs that target the Sur subunit. Furthermore, ubiquitous expression of an akh transgene reverses the effect of CC ablation on serum glucose. Thus, Drosophila CC cells are crucial regulators of glucose homeostasis and they use glucose-sensing and response mechanisms similar to islet cells.


Asunto(s)
Drosophila/citología , Drosophila/metabolismo , Glándulas Endocrinas/citología , Glándulas Endocrinas/fisiología , Glucosa/metabolismo , Homeostasis , Canales de Potasio de Rectificación Interna , Transportadoras de Casetes de Unión a ATP/agonistas , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Drosophila/genética , Drosophila/crecimiento & desarrollo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Genes de Insecto/genética , Hemolinfa/metabolismo , Hormonas de Insectos/genética , Hormonas de Insectos/metabolismo , Larva/metabolismo , Oligopéptidos/genética , Oligopéptidos/metabolismo , Canales de Potasio/agonistas , Canales de Potasio/metabolismo , Ácido Pirrolidona Carboxílico/análogos & derivados , Receptores de Droga/agonistas , Receptores de Droga/metabolismo , Compuestos de Sulfonilurea/farmacología , Receptores de Sulfonilureas , Transgenes/genética
17.
Nature ; 430(7001): 748-54, 2004 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-15306801

RESUMEN

The mammalian sensory system is capable of discriminating thermal stimuli ranging from noxious cold to noxious heat. Principal temperature sensors belong to the TRP cation channel family, but the mechanisms underlying the marked temperature sensitivity of opening and closing ('gating') of these channels are unknown. Here we show that temperature sensing is tightly linked to voltage-dependent gating in the cold-sensitive channel TRPM8 and the heat-sensitive channel TRPV1. Both channels are activated upon depolarization, and changes in temperature result in graded shifts of their voltage-dependent activation curves. The chemical agonists menthol (TRPM8) and capsaicin (TRPV1) function as gating modifiers, shifting activation curves towards physiological membrane potentials. Kinetic analysis of gating at different temperatures indicates that temperature sensitivity in TRPM8 and TRPV1 arises from a tenfold difference in the activation energies associated with voltage-dependent opening and closing. Our results suggest a simple unifying principle that explains both cold and heat sensitivity in TRP channels.


Asunto(s)
Frío , Calor , Activación del Canal Iónico , Canales Iónicos/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Droga/metabolismo , Capsaicina/farmacología , Línea Celular , Conductividad Eléctrica , Humanos , Activación del Canal Iónico/efectos de los fármacos , Canales Iónicos/agonistas , Canales Iónicos/genética , Ligandos , Potenciales de la Membrana/efectos de los fármacos , Mentol/farmacología , Modelos Biológicos , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/genética , Técnicas de Placa-Clamp , Receptores de Droga/agonistas , Receptores de Droga/genética , Canales Catiónicos TRPM
18.
Neurosci Lett ; 364(2): 94-7, 2004 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15196685

RESUMEN

In the present study, c-fos expression in the spinal cord has been used as a marker of neuronal activation induced by capsaicin-sensitive sensory afferents from the dorsal neck muscles in cats (n = 6). The number of Fos-immunoreactive neurons, which were revealed using the avidin-biotin-peroxidase method, was significantly increased in the cervical and lumbar spinal cord. In contrast to the control group (n = 3), 2 h after intramuscular capsaicin injection, c-fos expression was more extensive ipsilaterally to the injected side in the C3-C6 segments, and bilaterally in the L4-L6 segments. Most labeled neurons in the cervical spinal cord were small and giant cells, predominantly located in the middle and lateral parts of lamina I and, additionally, at the neck of the dorsal horn (lamina V), i.e., within the zones of termination of high-threshold muscle afferents. The widespread distribution of labeled cells throughout the cervical cord within the intermediate zone (lamina VII) coincided with the sites of last-order premotor interneurons and cells of origin of long crossed and uncrossed descending propriospinal pathways to the lumbar spinal cord. These findings suggest possible mechanisms for spreading of nociceptive signals between cervical and lumbar regions.


Asunto(s)
Genes fos/genética , Músculos del Cuello/fisiología , Receptores de Droga/agonistas , Médula Espinal/metabolismo , Animales , Capsaicina/administración & dosificación , Capsaicina/farmacología , Gatos , Tamaño de la Célula , Vértebras Cervicales , Femenino , Histocitoquímica , Inyecciones , Vértebras Lumbares , Masculino , Músculos del Cuello/inervación , Músculos del Cuello/metabolismo , Vías Nerviosas/fisiología , Dolor/fisiopatología , Estimulación Química
19.
J Neurosci ; 24(20): 4709-17, 2004 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-15152030

RESUMEN

Vanilloid (VR1) and purinergic (P2X) receptors are found in cranial afferent neurons in nodose ganglia and their central terminations within the solitary tract nucleus (NTS), but little is known about their function. We mechanically dissociated dorsomedial NTS neurons to preserve attached native synapses and tested for VR1 and P2X function primarily in spindle-shaped neurons resembling intact second-order neurons. All neurons (n = 95) exhibited spontaneous glutamate (EPSCs) and GABA (IPSCs)-mediated synaptic currents. VR1 agonist capsaicin (CAP; 100 nm) reversibly increased EPSC frequency, effects blocked by capsazepine. ATP (100 microm) increased EPSC frequency, actions blocked by P2X antagonist pyridoxalphosphate-6-azophenyl-2', 4'-disulfonic acid (PPADS; 20 microm). In all CAP-resistant neurons, P2X agonist alphabeta-methylene-ATP (alphabeta-m-ATP) increased EPSC frequency. Neither CAP nor alphabeta-m-ATP altered EPSC amplitudes, kinetics, or holding currents. Thus, activation of VR1 and P2X receptors selectively facilitated presynaptic glutamate release on different NTS neurons. PPADS and 2',3'-O-(2,4,6-trinitrophenyl)-ATP blocked alphabeta-m-ATP responses, but P2X1-selective antagonist NF023 (8,8'-[carbonylbis (imino-3,1-phenylene carbonylimino)]bis-1,3,5-naphthalenetrisulfonic acid) did not. The pharmacological profile and transient kinetics of ATP responses are consistent with P2X3 homomeric receptors. TTX and Cd(2+) did not eliminate agonist-evoked EPSC frequency increases, suggesting that voltage-gated sodium and calcium channels are not required. In nodose ganglia, CAP but not alphabeta-m-ATP evoked inward currents in slow conducting neurons and the converse pattern in myelinated, rapidly conducting neurons (n = 14). Together, results are consistent with segregation of glutamatergic terminals into either P2X sensitive or VR1 sensitive that correspondingly identify myelinated and unmyelinated afferent pathways at the NTS.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Vías Aferentes/fisiología , Capsaicina/análogos & derivados , Ácido Glutámico/metabolismo , Terminales Presinápticos/metabolismo , Fosfato de Piridoxal/análogos & derivados , Receptores de Droga/metabolismo , Receptores Purinérgicos P2/metabolismo , Núcleo Solitario/metabolismo , Adenosina Trifosfato/farmacología , Animales , Capsaicina/farmacología , Separación Celular , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Técnicas In Vitro , Masculino , Inhibición Neural/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Ganglio Nudoso/citología , Ganglio Nudoso/fisiología , Técnicas de Placa-Clamp , Agonistas del Receptor Purinérgico P2 , Antagonistas del Receptor Purinérgico P2 , Fosfato de Piridoxal/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Droga/agonistas , Receptores de Droga/antagonistas & inhibidores , Núcleo Solitario/citología , Núcleo Solitario/efectos de los fármacos
20.
Br J Pharmacol ; 140(6): 1035-44, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14530215

RESUMEN

1. Arvanil (N-arachidonoylvanillamine), a nonpungent capsaicin-anandamide hybrid molecule, has been shown to exert biological activities through VR1/CB1-dependent and -independent pathways. We have found that arvanil induces dose-dependent apoptosis in the lymphoid Jurkat T-cell line, but not in peripheral blood T lymphocytes. Apoptosis was assessed by DNA fragmentation through cell cycle and TUNEL analyses. 2. Arvanil-induced apoptosis was initiated independently of any specific phase of the cell cycle, and it was inhibited by specific caspase-8 and -3 inhibitors and by the activation of protein kinase C. In addition, kinetic analysis by Western blots and fluorimetry showed that arvanil rapidly activates caspase-8, -7 and -3, and induces PARP cleavage. 3. The arvanil-mediated apoptotic response was greatly inhibited in the Jurkat-FADDDN cell line, which constitutively expresses a negative dominant form of the adapter molecule Fas-associated death domain (FADD). This cell line does not undergo apoptosis in response to Fas (CD95) stimulation. 4. Using a cytofluorimetric approach, we have found that arvanil induced the production of reactive oxygen species (ROS) in both Jurkat-FADD+ and Jurkat-FADDDN cell lines. However, ROS accumulation only plays a residual role in arvanil-induced apoptosis. 5. These results demonstrate that arvanil-induced apoptosis is essentially mediated through a mechanism that is typical of type II cells, and implicates the death-inducing signalling complex and the activation of caspase-8. This arvanil-apoptotic activity is TRPV1 and CB-independent, and can be of importance for the development of potential anti-inflammatory and antitumoral drugs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Apoptosis/efectos de los fármacos , Capsaicina/análogos & derivados , Capsaicina/farmacología , Proteínas Portadoras/metabolismo , Caspasas/metabolismo , Transducción de Señal , Apoptosis/genética , Caspasa 8 , Inhibidores de Caspasas , Células Cultivadas , Inhibidores de Cisteína Proteinasa/farmacología , Fragmentación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Proteína Ligando Fas , Proteína de Dominio de Muerte Asociada a Fas , Humanos , Etiquetado Corte-Fin in Situ , Células Jurkat , Glicoproteínas de Membrana/genética , Modelos Biológicos , Especies Reactivas de Oxígeno/metabolismo , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/fisiología , Receptores de Droga/agonistas , Receptores de Droga/fisiología , Linfocitos T/efectos de los fármacos , Factores de Tiempo , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA