Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Bioorg Med Chem Lett ; 23(12): 3584-8, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23664879

RESUMEN

1-(1,3,5-Triazin-yl)piperidine-4-carboxamide inhibitors of soluble epoxide hydrolase were identified from high through-put screening using encoded library technology. The triazine heterocycle proved to be a critical functional group, essential for high potency and P450 selectivity. Phenyl group substitution was important for reducing clearance, and establishing good oral exposure. Based on this lead optimization work, 1-[4-methyl-6-(methylamino)-1,3,5-triazin-2-yl]-N-{[[4-bromo-2-(trifluoromethoxy)]-phenyl]methyl}-4-piperidinecarboxamide (27) was identified as a useful tool compound for in vivo investigation. Robust effects on a serum biomarker, 9, 10-epoxyoctadec-12(Z)-enoic acid (the epoxide derived from linoleic acid) were observed, which provided evidence of robust in vivo target engagement and the suitability of 27 as a tool compound for study in various disease models.


Asunto(s)
Amidas/química , Amidas/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Epóxido Hidrolasas/antagonistas & inhibidores , Piperidinas/química , Piperidinas/farmacología , Amidas/síntesis química , Descubrimiento de Drogas , Inhibidores Enzimáticos/síntesis química , Epóxido Hidrolasas/metabolismo , Humanos , Modelos Moleculares , Piperidinas/síntesis química , Relación Estructura-Actividad , Triazinas/síntesis química , Triazinas/química , Triazinas/farmacología
2.
J Enzyme Inhib Med Chem ; 28(1): 95-104, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22136506

RESUMEN

NADPH oxidase isoform-2 (NOX2) generates reactive oxygen species (ROS) that contribute to neurodegenerative and cardiovascular pathologies. However, validation of NOX2 as a pharmacotherapeutic target has been hampered by a lack of mechanistically-defined inhibitors. Using cellular and biochemical assays, we explored previously reported inhibitors of ROS production (perhexiline, suramin, VAS2870 and two Shionogi patent compounds) as direct NOX2 inhibitors. All but suramin, which presumably lacks cell penetrance, inhibit cellular ROS production. However, only perhexiline and suramin inhibit biochemical NOX2 activity. Indeed, our data suggest that NOX2 inhibition by perhexiline may contribute significantly to its demonstrated cardioprotective effects. Inhibition of protein kinase CßII explains the cellular activity of the Shionogi compounds, whereas VAS2870 inhibits by an as-yet unidentified mechanism unrelated to direct NOX2 function or subunit assembly. These data delineate the mechanisms of action of these compounds and highlight their strengths and limitations for use in future target validation studies.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Glicoproteínas de Membrana/antagonistas & inhibidores , NADPH Oxidasas/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Secuencia de Bases , Benzoxazoles/farmacología , Fármacos Cardiovasculares/farmacología , Células Cultivadas , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Datos de Secuencia Molecular , NADPH Oxidasa 2 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/enzimología , Neutrófilos/metabolismo , Perhexilina/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C beta , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Suramina/farmacología , Triazoles/farmacología
3.
Circ Res ; 105(7): 686-95, 19 p following 695, 2009 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-19696412

RESUMEN

RATIONALE: Expression of the vasoactive peptide Urotensin II (UII) is elevated in a number of cardiovascular diseases. OBJECTIVE: Here, we sought to determine the effect of UII receptor (UT) gene deletion in a mouse model of atherosclerosis. METHODS AND RESULTS: UT knockout (KO) mice were crossed with ApoE KO mice to generate UT/ApoE double knockout (DKO) mice. Mice were placed on a high-fat Western-type diet for 12 weeks. We evaluated the degree of atherosclerosis and hepatic steatosis by histology. In addition, serum glucose, insulin, and lipids were determined. DKO mice exhibited significantly increased atherosclerosis compared to ApoE KO mice (P<0.05). This was associated with a significant increase in serum insulin and lipids (P<0.001) but a decrease in hepatic steatosis (P<0.001). UT gene deletion led to a significant increase in systolic pressure and pulse pressure. RT-PCR and immunoblot analyses showed significant reductions in hepatic scavenger receptors, nuclear receptors, and acyl-CoA:cholesterol acyltransferase (ACAT1) expression in DKO mice. UII induced a significant increase in intracellular cholesteryl ester formation in primary mouse hepatocytes, which was blocked by the MEK inhibitor, PD98059. Hepatocytes of UTKO mice showed a significant reduction in lipoprotein uptake compared to wild-type mice. CONCLUSIONS: We propose that UT gene deletion in an ApoE-deficient background promotes downregulation of ACAT1, which in turn attenuates hepatic lipoprotein receptor-mediated uptake and lipid transporter expression. As the liver is the main organ for uptake of lipoprotein-derived lipids, DKO leads to an increase in hyperlipidemia, with a concomitant decrease in hepatic steatosis, and consequently increased atherosclerotic lesion formation. Furthermore, the hypertension associated with UT gene deletion is likely to contribute to the increased atherosclerotic burden.


Asunto(s)
Aorta/metabolismo , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Hiperlipidemias/metabolismo , Hígado/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Urotensinas/metabolismo , Acetil-CoA C-Acetiltransferasa/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Glucemia/metabolismo , Presión Sanguínea , Células Cultivadas , Ésteres del Colesterol/metabolismo , Grasas de la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Hígado Graso/metabolismo , Hígado Graso/prevención & control , Genotipo , Hiperlipidemias/genética , Hiperlipidemias/patología , Hiperlipidemias/fisiopatología , Insulina/sangre , Lípidos/sangre , Hígado/efectos de los fármacos , Hígado/patología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Depuradores/metabolismo , Factores de Tiempo
4.
J Pharmacol Exp Ther ; 328(1): 231-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18836067

RESUMEN

Epoxy- and dihydroxy-eicosatrienoic acids (EETs and DHETs) are vasoactive cytochrome P450 metabolites of arachidonic acid. Interestingly, however, the mechanism(s) by which EETs/DHETs mediate smooth muscle relaxation remains unclear. In contrast to previous reports, where dilation was purportedly large-conductance Ca(2+)-activated K(+) (BK(Ca)) and/or transient receptor potential cation channel, subfamily V, member 4 (TRPV4) channel-mediated, 14,15-EET-induced vasodilation [reversal of contractile tone established with the thromboxane receptor (TP) agonist 15-hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic acid (U-46619)] was unaltered in BK(Ca) and TRPV4 knockout mouse isolated aortae compared with wild-type controls, indicating a significant BK(Ca)/TRPV4-resistant mechanism. Whereas all EET and DHET regioisomers reversed U-46619 contraction in rat aortae and mouse mesenteric resistance arteries, these eicosanoids failed to alter phenylephrine-induced contraction, suggesting that they mediated dilation via a "TP-selective" mechanism. Competitive TP antagonism was also observed in nonvascular tissue, including rat fundus and tertiary bronchus, indicating that the effect is not specific to blood vessels. Such effects were TP-selective because 14,15-EET failed to inhibit "non-TP" prostanoid receptor-mediated function in multiple cell/tissue-based assays (K(b) > 10 microM). In accordance, 14,15-EET inhibited specific [(3)H]7-(3-((2-((phenylamino)carbonyl)hydrazino)-methyl)-7-oxabicyclo(2.2.1)hept-2-yl)-5-heptenoic acid (SQ-29548) binding to human recombinant TP receptor, with a K(i) value of 3.2 microM, and it showed weaker affinity for non-TP prostanoid receptors, including DP, FP, EP(1-4), and IP receptors (K(i) values of 6.1, 5.3, 42.6, 19.7, 13.2, 20.2, and >25 microM, respectively) and no appreciable affinity (K(i) values >10 microM) for a diverse array of pharmacologically distinct receptors, including the leukotriene receptors Cys-LT(1/2) and BLT(1). As such, EETs/DHETs represent a unique class of "endogenous" G protein-coupled receptor competitive antagonists, inducing vasodilation via direct TP inhibition. Thus, EETs/DHETs represent novel autoregulatory agents, directly modulating the actions of cyclooxygenase-derived eicosanoids following arachidonic acid mobilization.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/farmacología , Aorta Torácica/fisiología , Vasodilatación/fisiología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Animales , Aorta Torácica/efectos de los fármacos , Ácidos Araquidónicos/farmacología , Cobayas , Ácidos Hidroxieicosatetraenoicos/farmacología , Masculino , Ratones , Ratones Noqueados , Ratas , Ratas Sprague-Dawley , Receptores de Tromboxanos/agonistas , Receptores de Tromboxanos/efectos de los fármacos , Receptores de Tromboxanos/fisiología , Circulación Esplácnica/efectos de los fármacos , Circulación Esplácnica/fisiología , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/fisiología , Tráquea/efectos de los fármacos , Tráquea/fisiología , Resistencia Vascular/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vasodilatación/efectos de los fármacos
5.
Anesth Analg ; 108(4): 1177-81, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19299782

RESUMEN

BACKGROUND: Urotensin II (UII) and its receptor (UT) are implicated in mood disorders, such as stress and anxiety, and this may result, at least in part, from increased norepinephrine release from the cerebral cortex. Benzodiazepines have been widely used as hypnotics and anxiolytics, producing a decrease in cerebrocortical norepinephrine release. We hypothesized that there was some interaction between benzodiazepines and the UII system in the cerebral cortex. METHODS: In the present study, we have examined the effects of benzodiazepines on UII-increased norepinephrine release from rat cerebrocortical slices and intracellular Ca(2+) concentrations ([Ca(2+)]i) in HEK293 cells expressing rat UT receptor (HEK293-rUT cells). RESULTS: Midazolam, diazepam and flunitrazepam concentration-dependently inhibited UII-evoked norepinephrine release but did not affect [Ca(2+)]i. The IC(50) of midazolam for inhibition of UII-evoked norepinephrine release (0.32 microM, P < 0.01) was significantly lower than that of diazepam (187 microM) or flunitrazepam (40 microM). The inhibitory effects of midazolam on UII-evoked norepinephrine release were significantly attenuated by flumazenil, a benzodiazepine site antagonist. CONCLUSION: The present study suggests that midazolam, at clinically relevant concentration, significantly inhibited UII-evoked norepinephrine release. This inhibitory effect may be partially mediated via central benzodiazepine receptors.


Asunto(s)
Benzodiazepinas/farmacología , Corteza Cerebral/efectos de los fármacos , Hipnóticos y Sedantes/farmacología , Midazolam/farmacología , Norepinefrina/metabolismo , Urotensinas/metabolismo , Animales , Benzoxazoles/farmacología , Señalización del Calcio , Línea Celular , Corteza Cerebral/metabolismo , Diazepam/farmacología , Relación Dosis-Respuesta a Droga , Flumazenil/farmacología , Flunitrazepam/farmacología , Agonistas de Receptores de GABA-A , Humanos , Técnicas In Vitro , Masculino , Naftiridinas , Receptores de Orexina , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de GABA-A/metabolismo , Receptores de Neuropéptido/efectos de los fármacos , Receptores de Neuropéptido/metabolismo , Transfección , Urea/análogos & derivados , Urea/farmacología
6.
Neurosci Lett ; 440(3): 275-9, 2008 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-18572318

RESUMEN

Urotensin II (UII) has been reported to modulate rapid eye movement (REM) sleep via activation of brainstem cholinergic neurons and REM sleep is regulated by locus coerleus (LC)-cerebrocortical noradrenergic neurons. We hypothesized that UII may activate LC-cerebrocortical noradrenergic neurons. To test this hypothesis, we have examined the effects of UII on norepinephrine release from rat cerebrocortical slices. In addition, the effect of the putative UT receptor antagonist [Pen(5), DTrp(7), Dab(8)]UII(4-11) (UFP-803) was assessed. We have compared this with other wakefulness-promoting neurotransmitters such as dopamine, glutamate, serotonin and histamine. We also studied the effects of UII and UFP-803 on intracellular Ca(2+) ([Ca(2+)]i) in HEK293 cells stably expressing rat UT receptor (HEK293-rUT cells). UII produced a time- (peaking at approximately 10 min following stimulation with 10nM) and concentration-dependent increase in norepinephrine release with pEC(50) and E(max) (% of basal) values of 8.78+/-0.17 (1.65 nM) and 138+/-2%, respectively. UII also evoked dopamine, serotonin and histamine release with similar pEC(50) values. UII increased glutamate release but only at high concentrations (<100 nM) and this failed to saturate. UII markedly increased [Ca(2+)](i) in HEK293-rUT cells in a concentration-dependent manner with pEC(50) of 8.26+/-0.24. The UT antagonist UFP-803 reversed both UII-increased norepinephrine release from the cerebrocortical slices (pK(B)=8.98) and [Ca(2+)](i) (pK(B)=8.87) in HEK293-rUT cells. Collectively these data suggest that UII evokes the release of norepinephrine via UT receptor activation and produces similar effects on other wakefulness-promoting neurotransmitters: these neurochemical actions of UII may be important for the control of the sleep-wake cycle.


Asunto(s)
Cerebelo/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Neurotransmisores/metabolismo , Urotensinas/farmacología , Animales , Línea Celular Transformada , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Técnicas In Vitro , Masculino , Fragmentos de Péptidos/farmacología , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/metabolismo , Factores de Tiempo , Transfección
7.
Bioorg Med Chem Lett ; 18(18): 4936-9, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18768319

RESUMEN

High throughput screening of our compound collection led to the discovery of a novel series of N-alkyl-5H-pyrido[4,3-b]indol-1-amines as urotensin-II receptor antagonists. Synthesis, initial structure and activity relationships, functional and animal ortholog activities of the series are described.


Asunto(s)
Aminas/síntesis química , Aminas/farmacología , Piridinas/síntesis química , Piridinas/farmacología , Urotensinas/antagonistas & inhibidores , Aminas/química , Animales , Técnicas Químicas Combinatorias , Ratones , Estructura Molecular , Piridinas/química , Relación Estructura-Actividad
8.
Bioorg Med Chem Lett ; 18(14): 3950-4, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18573659

RESUMEN

SAR exploration of the central diamine, benzyl, and terminal aminoalkoxy regions of the N-cyclic azaalkyl benzamide series led to the identification of very potent human urotensin-II receptor antagonists such as 1a with a K(i) of 4 nM. The synthesis and structure-activity relationships (SAR) of N-cyclic azaalkyl benzamides are described.


Asunto(s)
Benzamidas/química , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Sitios de Unión , Química Farmacéutica/métodos , Diaminas/química , Diseño de Fármacos , Humanos , Concentración 50 Inhibidora , Cinética , Modelos Químicos , Relación Estructura-Actividad
9.
Bioorg Med Chem Lett ; 18(9): 2860-4, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18420409

RESUMEN

A series of 2-aminomethyl piperidines has been discovered as novel urotensin-II receptor antagonists. The synthesis, initial structure-activity relationships, and optimization of the initial hit that resulted in the identification of potent, cross-species active, and functional urotensin-II receptor antagonists such as 1a and 11a are described.


Asunto(s)
Metilaminas/farmacología , Piperidinas/farmacología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Vasoconstrictores/farmacología , Sitios de Unión , Humanos , Metilaminas/síntesis química , Modelos Químicos , Piperidinas/síntesis química , Estereoisomerismo , Relación Estructura-Actividad , Vasoconstrictores/síntesis química
10.
Bioorg Med Chem Lett ; 18(16): 4470-3, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18674898

RESUMEN

Aminomethylpiperazines, reported previously as being kappa-opioid receptor agonists, were identified as lead compounds in the development of selective urotensin receptor antagonists. Optimized substitution of the piperazine moiety has provided high affinity urotensin receptor antagonists with greater than 100-fold selectivity over the kappa-opioid receptor. Select compounds were found to inhibit urotensin-induced vasoconstriction in isolated rat aortic rings consistent with the hypothesis that an urotensin antagonist may be useful for the treatment of hypertension.


Asunto(s)
Química Farmacéutica/métodos , Piperazinas/farmacología , Receptores Opioides kappa/antagonistas & inhibidores , Taurina/análogos & derivados , Urotensinas/antagonistas & inhibidores , Acamprosato , Animales , Aorta/metabolismo , Diseño de Fármacos , Humanos , Hipertensión/tratamiento farmacológico , Modelos Químicos , Piperazinas/química , Ratas , Relación Estructura-Actividad , Taurina/efectos de los fármacos
11.
Bioorg Med Chem Lett ; 18(12): 3500-3, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18502123

RESUMEN

This work describes the development of potent and selective human Urotensin-II receptor antagonists starting from lead compound 1, (3,4-dichlorophenyl)methyl{2-oxo-2-[3-phenyl-2-(1-pyrrolidinylmethyl)-1-piperidinyl]ethyl}amine. Several problems relating to oral bioavailability, cytochrome P450 inhibition, and off-target activity at the kappa opioid receptor and cardiac sodium channel were addressed during lead development. hUT binding affinity relative to compound 1 was improved by more than 40-fold in some analogs, and a structural modification was identified which significantly attenuated both off-target activities.


Asunto(s)
Compuestos de Anilina/farmacología , Piperidonas/farmacología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Administración Oral , Compuestos de Anilina/síntesis química , Compuestos de Anilina/química , Animales , Disponibilidad Biológica , Línea Celular , Evaluación Preclínica de Medicamentos , Humanos , Estructura Molecular , Peso Molecular , Piperidonas/síntesis química , Piperidonas/química , Ratas , Bibliotecas de Moléculas Pequeñas , Estereoisomerismo , Relación Estructura-Actividad
12.
Bioorg Med Chem Lett ; 18(13): 3716-9, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18524591

RESUMEN

Lead compound 1 was successfully redesigned to provide compounds with improved pharmacokinetic profiles for this series of human urotensin-II antagonists. Replacement of the 2-pyrrolidinylmethyl-3-phenyl-piperidine core of 1 with a substituted N-methyl-2-(1-pyrrolidinyl)ethanamine core as in compound 7 resulted in compounds with improved oral bioavailability in rats. The relationship between stereochemistry and selectivity for hUT over the kappa-opioid receptor was also explored.


Asunto(s)
Química Farmacéutica/métodos , Urotensinas/antagonistas & inhibidores , Administración Oral , Animales , Encéfalo/metabolismo , Cromatografía Líquida de Alta Presión , Diaminas/química , Diseño de Fármacos , Humanos , Concentración 50 Inhibidora , Modelos Químicos , Ratas , Receptores Opioides kappa/química , Estereoisomerismo , Relación Estructura-Actividad , Urotensinas/química
13.
Br J Pharmacol ; 147(1): 92-100, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16273120

RESUMEN

The novel urotensin-II (U-II) receptor (UT) ligand, [Pen(5),DTrp(7),Dab(8)]U-II(4-11) (UFP-803), was pharmacologically evaluated and compared with urantide in in vitro and in vivo assays. In the rat isolated aorta, UFP-803 was inactive alone but, concentration dependently, displaced the contractile response to U-II to the right, revealing a competitive type of antagonism and a pA(2) value of 7.46. In the FLIPR [Ca(2+)](i) assay, performed at room temperature in HEK293(hUT) and HEK293(rUT) cells, U-II increased [Ca(2+)](i) with pEC(50) values of 8.11 and 8.48. Urantide and UFP-803 were inactive as agonists, but antagonized the actions of U-II by reducing, in a concentration-dependent manner, the agonist maximal effects with apparent pK(B) values in the range of 8.45-9.05. In a separate series of experiments performed at 37 degrees C using a cuvette-based [Ca(2+)](i) assay and CHO(hUT) cells, urantide mimicked the [Ca(2+)](i) stimulatory effect of U-II with an intrinsic activity (alpha) of 0.80, while UFP-803 displayed a small (alpha=0.21) but consistent residual agonist activity. When the same experiments were repeated at 22 degrees C (a temperature similar to that in FLIPR experiments), urantide displayed a very small intrinsic activity (alpha=0.11) and UFP-803 was completely inactive as an agonist. In vivo in mice, UFP-803 (10 nmol kg(-1)) antagonized U-II (1 nmol kg(-1))-induced increase in plasma extravasation in various vascular beds, while being inactive alone. In conclusion, UFP-803 is a potent UT receptor ligand which displays competitive/noncompetitive antagonist behavior depending on the assay. While UFP-803 is less potent than urantide, it displayed reduced residual agonist activity and as such may be a useful pharmacological tool.


Asunto(s)
Fragmentos de Péptidos/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Urotensinas/farmacología , Animales , Aorta/efectos de los fármacos , Células CHO , Línea Celular , Cricetinae , Cricetulus , Humanos , Ligandos , Ratones , Ratas
14.
Br J Pharmacol ; 148(2): 173-90, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16547525

RESUMEN

Several peptidic urotensin-II (UT) receptor antagonists exert 'paradoxical' agonist activity in recombinant cell- and tissue-based bioassay systems, likely the result of differential urotensin-II receptor (UT receptor) signal transduction/coupling efficiency between assays. The present study has examined this phenomenon in mammalian arteries and recombinant UT-HEK (human embryonic kidney) cells.BacMam-mediated recombinant UT receptor upregulation in HEK cells augmented agonist activity for all four peptidic UT ligands studied. The nominal rank order of relative intrinsic efficacy was U-II>urantide ([Pen(5)-DTrp(7)-Orn(8)]hU-II(4-11))>SB-710411 (Cpa-c[DCys-Pal-DTrp-Lys-Val-Cys]-Cpa-amide)>>GSK248451 (Cin-c[DCys-Pal-DTrp-Orn-Val-Cys]-His-amide) (the relative coupling efficiency of recombinant HEK cells was cat>human>>rat UT receptor). The present study further demonstrated that the use of high signal transduction/coupling efficiency isolated blood vessel assays (primate>cat arteries) is required in order to characterize UT receptor antagonism thoroughly. This cannot be attained simply by using the rat isolated aorta, an artery with low signal transduction/coupling efficiency in which low-efficacy agonists appear to function as antagonists. In contrast to the 'low-efficacy agonists' urantide and SB-710411, GSK248451 functioned as a potent UT receptor antagonist in all native isolated tissues studied (UT receptor selectivity was confirmed in the rat aorta). Further, GSK248451 exhibited an extremely low level of relative intrinsic activity in recombinant HEK cells (4-5-fold less than seen with urantide). Since GSK248451 (1 mg kg(-1), i.v.) blocked the systemic pressor actions of exogenous U-II in the anaesthetized cat, it represents a suitable peptidic tool antagonist for delineating the role of U-II in the aetiology of mammalian cardiometabolic diseases.


Asunto(s)
Fragmentos de Péptidos/farmacología , Péptidos Cíclicos/farmacología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Urotensinas/farmacología , Animales , Arterias/efectos de los fármacos , Arterias/fisiología , Unión Competitiva/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Calcio/metabolismo , Gatos , Línea Celular , Relación Dosis-Respuesta a Droga , Haplorrinos , Humanos , Técnicas In Vitro , Masculino , Fragmentos de Péptidos/metabolismo , Péptidos Cíclicos/química , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Urotensinas/metabolismo , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología
15.
Peptides ; 27(6): 1532-7, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16290260

RESUMEN

Urotensin-II (U-II), a ligand for the G-protein-coupled receptor UT, has been characterized as the most potent mammalian vasoconstrictor identified to date. Although circulating levels of U-II are altered in lower species (e.g., fish) upon exposure to hypo-osmotic stress, little is known about the actions of this cyclic undecapeptide within the kidney, an organ that plays a pivotal role in the control of cardiovascular homeostasis, influencing both cardiac preload (plasma volume) and after load (peripheral resistance). The present study reports the identification of specific, high affinity [125I]hU-II binding sites in Sprague-Dawley rat kidney outer medulla by autoradiography and also through membrane radioligand binding (Kd 1.9 +/- 0.9 nM and Bmax 408 +/- 47 amol mm(-2) and Kd 1.4 +/- 0.3 nM and Bmax 51.3 +/- 7.8 fmol mg(-1) protein, respectively). Differences were observed in the binding characteristics within rat strains. Compared to the Sprague-Dawley, Wistar Kyoto (WKY) and spontaneously hypertensive (SHR) rat kidney outer medulla displayed low density < 20 fmol mg(-1) protein and low affinity (> 1 microM) [125I]hU-II binding sites. Thus, the relative contribution of specific U-II binding sites to the physiological actions of U-II in the control of cardiorenal homeostasis is worthy of further investigation.


Asunto(s)
Médula Renal/metabolismo , Urotensinas/química , Animales , Autorradiografía , Sitios de Unión , Unión Competitiva , Humanos , Cinética , Ligandos , Unión Proteica , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Especificidad de la Especie , Urotensinas/metabolismo
16.
Peptides ; 27(11): 2919-26, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16919371

RESUMEN

It is now well established that urotensin-II (UII) levels are increased in several cardiovascular diseases. We previously demonstrated that UII and the UII receptor (UT) protein levels are significantly increased in the hearts of both humans and rats with congestive heart failure (CHF). We have also recently demonstrated that UII blockade, with a selective UII antagonist, improves heart function in a rat model of ischemic CHF. Here, we evaluated the attenuation of cardiac remodeling associated with UII antagonism in the same rat model of ischemic CHF. Animals were administered a specific UT receptor antagonist, SB-611812 (30 mg/kg/day, gavage), or vehicle 30 min prior to coronary artery ligation followed by daily treatment for 8 weeks. Myocardial interstitial fibrosis was analyzed by Masson's trichrome and picrosirius red staining. RT-PCR analysis was utilized for mRNA expression studies. We used Western blotting to assess levels of collagen types I and III. Mitogenic activity of UII on cultured neonatal cardiac fibroblasts was also evaluated. Following coronary ligation, SB-611812 significantly attenuated both myocardial and endocardial interstitial fibrosis, and reduced collagen type I:III ratio (P<0.01). UII induced proliferation of cardiac fibroblasts and this mitogenic effect was significantly inhibited with 1 microM of SB-611218 (P<0.05). We demonstrate here that selective blockade of UT reduces diastolic dysfunction by decreasing myocardial fibrosis post-coronary ligation in vivo, and inhibits UII-mediated fibroblast proliferation in vitro.


Asunto(s)
Isquemia Miocárdica/tratamiento farmacológico , Isquemia Miocárdica/fisiopatología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Sulfonamidas/farmacología , Remodelación Ventricular/efectos de los fármacos , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis/patología , Fibrosis/prevención & control , Perfilación de la Expresión Génica , Masculino , Isquemia Miocárdica/patología , Miocardio/patología , Ratas , Ratas Endogámicas Lew , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Urotensinas/farmacología
17.
Trends Pharmacol Sci ; 25(2): 76-85, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15102493

RESUMEN

The identification of a human homolog of urotensin-II (U-II) and a novel, specific G-protein-coupled receptor by Ames et al. in 1999 changed the perception that the U-II isopeptide family was an esoteric collection of 'somatostatin-like neuropeptides' present only in the nervous systems of an eclectic array of aquatic invertebrates, fish and amphibians. In this article, we review recent developments in the pharmacology of human U-II, focusing on the actions of this peptide in the mammalian cardiorenal system. The putative role of U-II in the etiology of hypertension, heart failure, renal dysfunction and diabetes is discussed, as are novel U-II receptor antagonists.


Asunto(s)
Corazón/efectos de los fármacos , Riñón/efectos de los fármacos , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Urotensinas , Animales , Humanos , Hipertensión/etiología , Riñón/fisiología , Especificidad de la Especie , Urotensinas/antagonistas & inhibidores , Urotensinas/farmacología , Urotensinas/fisiología
18.
Trends Pharmacol Sci ; 25(4): 225-33, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15063087

RESUMEN

The latter half of the 20th century has been characterized by pharmacologists as the 'age of the receptor', an era in which the bioassay, that stalwart of classical pharmacology, has played a seminal role in identifying novel cardiovascular medicines. In this article, we ask what, if anything, has changed in the pharmacologist's approach to discovering novel cardiovascular drugs on this, the 25th anniversary of the inaugural publication of Trends in Pharmacological Sciences.


Asunto(s)
Fármacos Cardiovasculares/uso terapéutico , Farmacología/tendencias , Receptores Purinérgicos/efectos de los fármacos , Animales , Humanos , Receptores Purinérgicos/genética , Receptores Purinérgicos/metabolismo
19.
Br J Pharmacol ; 145(5): 620-35, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15852036

RESUMEN

1. SB-706375 potently inhibited [(125)I]hU-II binding to both mammalian recombinant and 'native' UT receptors (K(i) 4.7+/-1.5 to 20.7+/-3.6 nM at rodent, feline and primate recombinant UT receptors and K(i) 5.4+/-0.4 nM at the endogenous UT receptor in SJRH30 cells). 2. Prior exposure to SB-706375 (1 microM, 30 min) did not alter [(125)I]hU-II binding affinity or density in recombinant cells (K(D) 3.1+/-0.4 vs 5.8+/-0.9 nM and B(max) 3.1+/-1.0 vs 2.8+/-0.8 pmol mg(-1)) consistent with a reversible mode of action. 3. The novel, nonpeptidic radioligand [(3)H]SB-657510, a close analogue of SB-706375, bound to the monkey UT receptor (K(D) 2.6+/-0.4 nM, B(max) 0.86+/-0.12 pmol mg(-1)) in a manner that was inhibited by both U-II isopeptides and SB-706375 (K(i) 4.6+/-1.4 to 17.6+/-5.4 nM) consistent with the sulphonamides and native U-II ligands sharing a common UT receptor binding domain. 4. SB-706375 was a potent, competitive hU-II antagonist across species with pK(b) 7.29-8.00 in HEK293-UT receptor cells (inhibition of [Ca(2+)](i)-mobilization) and pK(b) 7.47 in rat isolated aorta (inhibition of contraction). SB-706375 also reversed tone established in the rat aorta by prior exposure to hU-II (K(app) approximately 20 nM). 5. SB-706375 was a selective U-II antagonist with >/=100-fold selectivity for the human UT receptor compared to 86 distinct receptors, ion channels, enzymes, transporters and nuclear hormones (K(i)/IC(50)>1 microM). Accordingly, the contractile responses induced in isolated aortae by KCl, phenylephrine, angiotensin II and endothelin-1 were unaltered by SB-706375 (1 microM). 6. In summary, SB-706375 is a high-affinity, surmountable, reversible and selective nonpeptide UT receptor antagonist with cross-species activity that will assist in delineating the pathophysiological actions of U-II in mammals.


Asunto(s)
Pirrolidinas/farmacología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Sulfonamidas/farmacología , Algoritmos , Animales , Aorta Torácica/efectos de los fármacos , Unión Competitiva/efectos de los fármacos , Gatos , Línea Celular Tumoral , Membrana Celular/metabolismo , Haplorrinos , Humanos , Técnicas In Vitro , Ratones , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Ensayo de Unión Radioligante , Ratas , Proteínas Recombinantes/metabolismo , Rabdomiosarcoma/metabolismo , Especificidad de la Especie
20.
Biochem Pharmacol ; 69(7): 1069-79, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15763543

RESUMEN

Urotensin-II (U-II), acting through its G-protein-coupled receptor, UT, is a possible contributor to hypertension. Variable functional responses to U-II, both within and between species studied to date, complicate the characterization of UT antagonists. In the cat, however, U-II causes systemic hypertension and constricts arterial segments isolated from several vascular beds. The purpose of this study was to clone and pharmacologically characterize cat recombinant UT to determine whether this system represents a model for characterizing UT antagonists. Cloned cat UT displayed 74% identity to primate UT, and 77% identity to rodent UT. [(125)I] hU-II bound in a saturable manner to a single site on recombinant cat UT with high affinity (K(D) 288+/-13pM) and high density (B(max) 747+/-66fmol/mg protein). U-II isopeptides displayed equipotent, high affinity binding to cat UT (K(i) 1.8-5.3nM). Cat UT was coupled to intracellular [Ca(2+)] release (EC(50) 0.6+/-0.2nM) and total inositol phosphate (IP) formation (EC(50) 0.4+/-0.1nM). Protein kinase C activation desensitized cat, but not human, UT-mediated IP formation. UT mRNA expression was detected in cat blood vessels, trachea, lung, and kidney, where the medulla (K(D) 815+/-34) and cortex and (K(D) 316+/-39pM) displayed high affinity binding for human U-II (hU-II). The cat urotensin-II receptor represents a suitable in vitro model to examine the role of the U-II/UT system in the etiology of hypertension, assisting in the evaluation of the UT antagonists to help treat cardiovascular disease.


Asunto(s)
Clonación Molecular , Receptores Acoplados a Proteínas G/genética , Secuencia de Aminoácidos , Animales , Señalización del Calcio/fisiología , Gatos , Línea Celular , Membrana Celular/fisiología , Secuencia Conservada , Cartilla de ADN , Humanos , Fosfatos de Inositol/metabolismo , Ratones , Datos de Secuencia Molecular , Especificidad de Órganos , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Ratas , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA