Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Eur J Pharmacol ; 416(3): 213-21, 2001 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-11290371

RESUMEN

In the current study, two specific glycosaminoglycan lyases, chondroitinase AC and chondroitinase B, were utilized to examine the roles of chondroitin sulfates and dermatan sulfate in tumor metastasis and angiogenesis. Melanoma cells (SK-MEL) or endothelial cells were treated with either medium or chondroitinase enzyme. Chondroitinase AC inhibited melanoma invasion and proliferation as well as endothelial proliferation and angiogenesis. Apoptosis of melanoma and endothelial cells, as measured by the activity of caspase-3, was also increased by chondroitinase AC, but not by chondroitinase B. Chondroitinase B inhibited endothelial and melanoma proliferation and invasion, but to a lesser extent than chondroitinase AC. Neither chondroitinase had a detectable effect on gelatinase secretion by melanoma cells. These results indicate that both chondroitin and dermatan sulfates regulate many cellular activities related to metastasis.


Asunto(s)
División Celular , Sulfatos de Condroitina/fisiología , Condroitinasas y Condroitín Liasas/farmacología , Dermatán Sulfato/fisiología , Metástasis de la Neoplasia , Neovascularización Fisiológica , Animales , Apoptosis/efectos de los fármacos , Bovinos , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Gelatinasas/metabolismo , Humanos , Melanoma/patología , Neovascularización Patológica , Neovascularización Fisiológica/efectos de los fármacos , Células Tumorales Cultivadas
2.
Clin Appl Thromb Hemost ; 7(1): 58-64, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11190907

RESUMEN

A synthetic pentasaccharide (SR90107/ ORG31540) representing the antithrombin III (ATIII) binding sequence in heparin is under clinical development for the prophylaxis and management of venous thromboembolism. This pentasaccharide exhibits potent anti-factor Xa (AXa) effects (>750 IU/mg) and does not exhibit any anti-factor IIa (AIIa) activity. Previous reports have suggested that synthetic heparin pentasaccharides are resistant to the digestive effects of heparinase I. To investigate the effect of heparinase I on the AXa activity of pentasaccharide SR90107/ORG31540, graded concentrations (1.25-100 microg/ml) were incubated with a fixed amount of heparinase I (0.1 U/ml). Heparinase I produced a strong neutralizing effect on this pentasaccharide, as measured by AXa activity. This observation led to further studies where high performance liquid chromatography (HPLC) analysis was employed to determine the potential breakdown products of the pentasaccharide. The experiment with the pentasaccharide included incubation (37 degrees C) at 1 mg/ml and exposure to graded concentrations of heparinase I (0.125-1 U/ml). After 30 min of incubation, the enzymatic activity was stopped by heat treatment and the mixture was analyzed using high performance size exclusion chromatography (HPSEC). Heparinase I concentration-dependent cleavage of the pentasaccharide was evident. The breakdown products exhibited a mass of 1,034 d and 743 d, respectively, suggesting the generation of a trisaccharide and a disaccharide moiety. The extinction of a disaccharide moiety in the UV region was high, indicating the presence of a double bond in this molecule. These data clearly suggest that pentasaccharide SR90107/ORG31540 is digestible by heparinase I into its two components. Furthermore, these data support the hypothesis that heparinase I can be used as a neutralizing agent for pentasaccharide overdose. Additionally, a highly methylated analog of the previously mentioned synthetic pentasaccharide. SanOrg34006, which has also been subjected to similar experiments, has shown complete resistance to the depolymerizing function of heparinase I; therefore, its use may be appropriate in chronic situations as a long-acting form of the pentasaccharide.


Asunto(s)
Liasa de Heparina/metabolismo , Heparina/metabolismo , Oligosacáridos/metabolismo , Polímeros/metabolismo , Antitrombina III/metabolismo , Secuencia de Carbohidratos , Cromatografía en Gel , Cromatografía Líquida de Alta Presión , Disacáridos/química , Disacáridos/metabolismo , Relación Dosis-Respuesta a Droga , Factor Xa/metabolismo , Inhibidores del Factor Xa , Datos de Secuencia Molecular , Tromboembolia/tratamiento farmacológico , Tromboembolia/prevención & control , Trisacáridos/química , Trisacáridos/metabolismo , Trombosis de la Vena/tratamiento farmacológico , Trombosis de la Vena/prevención & control
3.
Eur J Pharmacol ; 400(2-3): 145-53, 2000 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-10988328

RESUMEN

In the current study, a glycosaminoglycan lyase, chondroitinase B, was used to study the role of dermatan sulfate proteoglycans on human dermal fibroblast proliferation. Pretreatment with chondroitinase B significantly decreased fibroblast proliferative responses to serum (20% to 55%). In contrast, heparinase III and chondroitinase AC were less effective in inhibiting fibroblast proliferation to serum. Analysis of glycosaminoglycans on chondroitinase B-treated fibroblasts confirmed that dermatan sulfate was removed from fibroblasts by this enzyme. Chondroitinase B treatment also decreased proliferation to basic fibroblast growth factor (bFGF) by 20% and reduced receptor binding by 25%. Heparinase III inhibited bFGF binding by 73%, but decreased proliferation to bFGF by only 21%. Chondroitinase AC had no effect on bFGF proliferation or binding. These data suggest that dermatan sulfate proteoglycans play a significant role in the control of human dermal fibroblast proliferation.


Asunto(s)
Dermatán Sulfato/fisiología , Fibroblastos/citología , Piel/citología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , División Celular/efectos de los fármacos , División Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Condroitín Liasas/farmacología , Proteoglicanos Tipo Condroitín Sulfato/deficiencia , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/fisiología , Dermatán Sulfato/deficiencia , Dermatán Sulfato/metabolismo , Interacciones Farmacológicas , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Polisacárido Liasas/metabolismo , Polisacárido Liasas/farmacología , Piel/efectos de los fármacos , Piel/metabolismo
4.
Pulm Pharmacol Ther ; 13(4): 175-80, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10930356

RESUMEN

Neutrophil elastase is involved in the pathogenesis of several pulmonary diseases; a strategy for monitoring in vivo elastase activity is to measure changes in biochemical markers. The objective of this study was to determine whether differences in the urinary excretion of the elastin crosslinks, desmosine and isodesmosine (which are unique amino acid products of elastase activity), could be discerned between groups of patients with chronic obstructive pulmonary disease (COPD) or cystic fibrosis (CF), and non-diseased, age-matched controls. Twenty-four-hour urine collections were analysed to eliminate variations in excretion throughout the day, and urine was collected on four separate days in 29-31 subjects/group to investigate the variability in desmosines excretion among the groups. Both sets of patient populations had significantly more variable desmosines readings (higher standard deviations) relative to their respective age-matched control group. The means for three adult groups (COPD, controls and a COPD-smoker subset) ranged from 28.4 to 35.5 pmol desmosines/mg creatinine and there were no differences among the groups. Values in children were higher: 55 pmol desmosines/mg creatinine in the non-CF children and 77 pmol desmosines/mg creatinine for the CF group (P<0.01 vs. age-matched controls). The results of this study show that urinary desmosines, as a surrogate marker for enhanced elastase activity, are more highly variant in both patient populations relative to age-matched controls, and an overall increase in the mean value is further observed in patients with cystic fibrosis.


Asunto(s)
Fibrosis Quística/orina , Desmosina/orina , Isodesmosina/orina , Elastasa de Leucocito/metabolismo , Enfermedades Pulmonares Obstructivas/orina , Biomarcadores/orina , Estudios de Casos y Controles , Niño , Fibrosis Quística/enzimología , Femenino , Humanos , Enfermedades Pulmonares Obstructivas/enzimología , Masculino , Persona de Mediana Edad
5.
J Cardiovasc Pharmacol ; 35(1): 29-36, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10630730

RESUMEN

Bucindolol is a nonselective beta-adrenergic receptor antagonist that has additional vasodilating properties. Because some beta-adrenergic receptor antagonists such as cyanopindolol are used as 5-HT1A/5-HT1B receptor antagonists, we tested the hypothesis that bucindolol can interact with 5-HT receptors. Both in vitro and in vivo methods were used to examine the interaction of bucindolol with 5-HT receptors relevant to the cardiovascular system-the 5-HT1A, 5-HT1D, 5-HT2A, and 5-HT2B receptors-and with alpha1-adrenergic receptors. In binding studies, bucindolol displayed high affinity for the 5-HT1A receptor (Ki, 11 nM), modest affinity for the 5-HT2A receptor (Ki, 382 nM), and no measurable affinity for the 5-HT1D receptor; binding affinity for the 5-HT2B receptor was not studied. Bucindolol also displayed significant binding affinity (Ki, 69 nM) for the alpha1-adrenergic receptors. Alpha1-Adrenergic receptor antagonist activity was confirmed by the ability of bucindolol (1 mg/kg) to act as a competitive antagonist against 0.01-30 microg/kg phenylephrine-induced pressor responses in conscious rats. In conscious permanently instrumented rats, bucindolol (0.1-3.0 mg/kg, i.v.) did not cause bradycardia similar to that elicited by the 5-HT1A-receptor agonist 8-OH-DPAT (3-300 microg/kg, i.v.), nor did bucindolol (1 mg/kg) block the 8-OH-DPAT-induced bradycardia. Bucindolol (10(-9)-10(-5) M) did not cause relaxation in the PGF2alpha-contracted, endothelium-intact porcine coronary artery, nor did bucindolol (10(-5) M) block 5-HT-induced coronary artery relaxation, indicating that bucindolol does not have significant interactions at the 5-HT1D receptor. Bucindolol also displayed no agonist activity at the 5-HT2A and 5-HT2B receptor (endothelium-denuded rat thoracic aorta and rat stomach fundus, respectively), but did act as a weak 5-HT2A-receptor antagonist (-log K(B) [M] = 5.4+/-0.1) and 5-HT2B-receptor antagonist (-log K(B) [M] = 7.8+/-0.1). Thus, these data suggest that bucindolol lacks the ability to activate the 5-HT1A, 5-HT1D, 5-HT2A, and 5-HT2B receptor, but can block alpha1-adrenergic receptors and act as a weak 5-HT2A- and 5-HT2B-receptor antagonist. The relevance of these serotoninergic effects as it pertains to the mechanism of bucindolol-induced vasodilation is unknown.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Propanolaminas/farmacología , Receptores de Serotonina/efectos de los fármacos , Agonistas alfa-Adrenérgicos/farmacología , Antagonistas Adrenérgicos beta/metabolismo , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Vasos Coronarios/efectos de los fármacos , Fundus Gástrico/efectos de los fármacos , Fundus Gástrico/metabolismo , Hemodinámica/efectos de los fármacos , Técnicas In Vitro , Masculino , Fenilefrina/farmacología , Propanolaminas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Receptores Adrenérgicos alfa 1/metabolismo , Receptores de Serotonina/metabolismo , Antagonistas de la Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología , Estereoisomerismo , Porcinos
6.
Thromb Res ; 91(3): 143-50, 1998 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-9733158

RESUMEN

Neutralase (heparinase I; E.C. 4.2.2.7) is a heparin-degrading enzyme undergoing clinical evaluation as an alternative to protamine for reversing the anticoagulant effects of heparin in coronary bypass surgery. The objective of this study was to assess the relative effects of Neutralase and protamine on reversal of heparin-dependent elevations in coagulation parameters and inhibition of clot formation in a rabbit vena caval stasis model. Rabbits were treated with saline or heparin (300 U/kg) for 10 minutes, followed by saline, protamine (2.6 mg/kg), or Neutralase (10 or 30 microg/kg, representing 1.23 IU/kg and 3.69 IU/kg, respectively). Twenty minutes later, venous stasis was induced, and vena caval clots were excised, weighed, and characterized. Coagulation parameters [activated partial thromboplastin time (aPTT) and thrombin clotting time (TCT)] and antiFactor IIa and Xa levels were measured throughout the protocol. Both protamine and Neutralase reversed heparin-mediated increases in aPTT (>300 seconds to 26-35 seconds) and TCT (>300 seconds to 29-56 seconds) to values that were not different from saline-treated, nonheparinized animals. Thrombus weight in the nonheparinized saline group was 62+/-7 mg; heparin-treated animals had no detectable clots. Protamine reversal of heparin was associated with clot formation (89+/-20 mg) while Neutralase reversal was not (no clots). Heparin-induced increases in antiFactor IIa activity were reversed similarly by protamine and Neutralase (from 4.3-8.8 U/ml to 0.2-0.3 U/ml) while antiFactor Xa activity was differentially reversed (from 3.9-5.9 U/ml to 0.7-1.3 U/ml Neutralase; 5.5 U/ml to 0.02 U/ml protamine). These results are consistent with a hypothesis that Neutralase cleaves heparin into fragments, which are devoid of antiFactor IIa activity that retain modest antiFactor Xa activity, resulting in reversal of anticoagulant, but not antithrombotic, heparin activity. This property of Neutralase may be beneficial in reducing post-surgical thrombotic events after reversal of heparin.


Asunto(s)
Anticoagulantes/farmacología , Antagonistas de Heparina/farmacología , Liasa de Heparina/farmacología , Heparina/farmacología , Protaminas/farmacología , Trombosis de la Vena/tratamiento farmacológico , Animales , Coagulación Sanguínea/efectos de los fármacos , Interacciones Farmacológicas , Heparina/uso terapéutico , Masculino , Conejos
7.
Eur J Pharmacol ; 351(1): 79-83, 1998 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-9698208

RESUMEN

Heparinase III degrades heparan sulfate proteoglycans, which are co-receptors for growth factors that stimulate arterial proliferation. We assessed the ability of locally-delivered heparinase III to limit medial vascular smooth muscle cell proliferation induced by balloon catheter injury in rat carotid arteries. Whereas vehicle-treated arteries showed 12% of smooth muscle cells proliferating after 2 days, heparinase III (0.022-5.7 mg/kg) treated arteries showed 0.8-4%. Chemically-inactivated heparinase III did not limit proliferation. In isolated rat A10 vascular smooth muscle cells, heparinase III (1 IU/ml) inhibited both PDGF-BB and bFGF mediated increases in proliferation and migration. These results suggest that heparinase III can limit proliferation by affecting heparan sulfate proteoglycan binding growth factors following arterial injury.


Asunto(s)
Arteria Carótida Externa/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Músculo Liso Vascular/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Polisacárido Liasas/farmacología , Animales , Becaplermina , Arteria Carótida Externa/patología , Estenosis Carotídea/prevención & control , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proteoglicanos de Heparán Sulfato/metabolismo , Inmunohistoquímica , Técnicas In Vitro , Masculino , Proteínas Proto-Oncogénicas c-sis , Ratas , Ratas Sprague-Dawley
8.
Eur J Pharmacol ; 349(2-3): 263-8, 1998 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-9671106

RESUMEN

This study describes the phosphodiesterase inhibitory potency and cardiovascular actions of WIN 65579 (1-cyclopentyl-3-ethyl-6-(3-ethoxy-4-pyrridyl)-1H-pyrazolo[3,4-d]p yrimidin-4-one), a potent, new cGMP phosphodiesterase 5 inhibitor. WIN 65579 is a competitive inhibitor of phosphodiesterase 5, with IC50 values of 2-3 nM for phosphodiesterase 5 from human or canine vascular sources. WIN 65579 has low affinity for phosphodiesterases 1, 2 and 3 (IC50 > 3-10 microM), and is somewhat selective for phosphodiesterase 4 (IC50 approximately 100 nM). WIN 65579 is an endothelial-dependent relaxant of rat aortic smooth muscle (EC50 = 60 nM) and lowers mean arterial blood pressure in conscious spontaneous hypertensive rats following intravenous or oral dosing. WIN 65579 also increases plasma cGMP levels, and reinstates vascular responsiveness to nitroglycerin in conscious rats that are nitroglycerin-tolerant. These data show that WIN 65579 is one of the more potent phosphodiesterase 5 inhibitors, and that WIN 65579 possesses cardiovascular activities consistent with vascular phosphodiesterase 5 inhibition in vivo.


Asunto(s)
3',5'-GMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Presión Sanguínea/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Purinonas/farmacología , Pirimidinas/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/fisiología , GMP Cíclico/sangre , Masculino , Relajación Muscular , Músculo Liso Vascular/fisiología , Nitroglicerina/farmacología , Ratas , Ratas Endogámicas SHR
9.
Expert Opin Investig Drugs ; 7(6): 1003-14, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15992012

RESUMEN

IBT 9302 (heparinase III, EC 4.2.2.8), purified from Flavobacterium heparinum, selectively cleaves heparan sulfate proteoglycans (HSPGs) from cellular surfaces and extracellular matrices. HSPGs serve as binding sites for P- and L-selectins, as well as for pro-inflammatory chemokines, such as interleukin (IL)-8. IBT 9302 reversibly removes these binding sites and inflammatory mediators, thereby limiting tissue damage following reperfusion of ischaemic areas by reducing leukocyte rolling, adhesion and extravasation. In models of myocardial ischaemia/reperfusion injury, infusion of IBT 9302 the time of transport and reperfusions, reduces the area of necrosis/area at risk ratios relative to vehicle-treated animals. Cardioprotection is accompanied by a reduction in serum creatine kinase levels and neutrophil adherence to coronary vessels, and the preservation of endothelial relaxation responsiveness to acetylcholine. HSPGs also serve as accumulation sites for most growth factors and IBT 9302 limits both proliferation and migration of vascular smooth muscle cells to platelet-derived growth factor (PDGF), basic fibroblast growth factor (bFGF) and endothelial growth factor (EGF). In vivo, the application of IBT 9302 at the time of vascular injury significantly reduces arterial medial proliferation. External application of IBT 9302 to wounds in a steroid-impaired wound healing model increases tensile strength by releasing mitogenic growth factors and HSPGs from the surrounding extracellular matrix. Pharmacokinetic studies show a simple monoexponential decay following iv. bolus dosing of IBT 9302, with a half-life of 5 - 6 min. The majority of [(125)I]-IBT 9302 goes to the liver (60%) and kidneys (25%), following iv. dosing. Preliminary toxicology studies in rats following single iv. bolus (10 mg/kg) or infusion (10 mug/kg/min) dosing show no untoward effects. These results suggest that IBT 9302 may have therapeutic utility in treating myocardial ischaemia/reperfusion injury, ischaemic stroke, restenosis or in healing diabetic ulcer wounds, by virtue of its ability to selectively cleave HSPGs.

10.
Eur J Pharmacol ; 313(1-2): 89-96, 1996 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-8905333

RESUMEN

Hemodynamic tolerance to nitroglycerin was developed in spontaneously hypertensive rats following 2-3 days of pretreatment with 100 mg/kg of nitroglycerin administered s.c. 3 times/day. Tolerance was evaluated both in vivo, by administering ascending bolus doses of nitroglycerin of 1-300 micrograms/kg i.v., and ex vivo in isolated, denuded aortic vascular rings by exposure to ascending concentrations of nitroglycerin of 0.0003-100 microM. Tolerance was observed as a significant blunting of the hypotensive and vasorelaxant effect of nitroglycerin. Co-incubation of tolerant aortic rings and pretreatment of tolerant SHR with 10 microM and 0.1-10 mg/kg zaprinast, respectively, resulted in full restoration of the vasorelaxant and hypotensive effect of nitroglycerin. Zaprinast partially reversed hemodynamic tolerance at 0.01 mg/kg. Conversely, dipyridamole (10 microM) reversed tolerance ex vivo, but was ineffective in reversing tolerance in vivo at pretreatment doses of 30 and 60 mg/kg. Following a 100-micrograms/kg i.v. challenge dose of nitroglycerin, aortic cyclic guanosine monophosphate (cGMP) levels were lower in nitroglycerin tolerant SHR when compared to non-tolerant SHR. Pretreatment of tolerant SHR with 10 mg/kg zaprinast restored the increase in cGMP levels to nitroglycerin to that seen in non-tolerant SHR. Conversely, dipyridamole (30 mg/kg) pretreatment was not effective in restoring cGMP levels. These data therefore suggest that reversal of hemodynamic tolerance in vivo is related to restoration of changes in vascular cGMP levels. Zaprinast, a selective cGMP phosphodiesterase inhibitor, effectively reverses tolerance and dipyridamole, a rather non-selective inhibitor, does not.


Asunto(s)
Aorta Abdominal/metabolismo , Presión Sanguínea/efectos de los fármacos , GMP Cíclico/metabolismo , Nitroglicerina/antagonistas & inhibidores , Purinonas/farmacología , Vasodilatadores/antagonistas & inhibidores , 3',5'-GMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Análisis de Varianza , Animales , Dipiridamol/farmacología , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Masculino , Relajación Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Nitroglicerina/farmacología , Ratas , Ratas Endogámicas SHR , Vasodilatadores/farmacología
11.
Biochem Pharmacol ; 51(12): 1631-8, 1996 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-8687478

RESUMEN

In this report, we describe the discovery and characterization of a novel biarylhydrazone series of platelet-derived growth factor (PDGF) receptor tyrosine kinase inhibitors typified by the prototype WIN 41662 (3-phenyl-N1-[1-(4-pytidyl)pyrimidine]hydrazone). WIN 41662 inhibited PDGF-stimulated autophosphorylation of PDGF receptors from human vascular smooth muscle cells (hVSMC) with an IC50 value of 60 nM. The inhibitor appeared to be competitive with respect to substrate (Mn(2+)-ATP), having a calculated Ki of 15 +/- 5 nM. WIN 41662 was approximately 500-fold more potent in inhibiting the PDGF receptor tyrosine kinase than the p56lck tyrosine kinase. It was inactive against other serine/threonine and tyrosine kinases tested. WIN 41662 produced concentration-dependent inhibition of PDGF-stimulated receptor autophosphorylation in intact hVSMC with an IC50 < 100 nM. Intracellular Ca2+ mobilization and cell proliferation were events that occurred in hVSMC subsequent to PDGF receptor activation. WIN 41662 inhibited PDGF-stimulated Ca2+ mobilization and cell proliferation ([3H]TdR incorporation) with IC50 values of 430 nM and 2.3 microM, respectively. These effects appeared to be specifically related to PDGF receptor tyrosine kinase inhibition since WIN 41662 was not cytotoxic (in vitro) and since WIN 72039, a close structural analog that does not inhibit PDGF receptor tyrosine kinase, also did not inhibit PDGF-stimulated receptor autophosphorylation, Ca2+ mobilization, or hVSMC proliferation. Thus, WIN 41662 is representative of a novel class of selective PDGF receptor tyrosine kinase inhibitors that inhibit PDGF-regulated secondary events in intact cells.


Asunto(s)
Aorta/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas Tirosina Quinasas/efectos de los fármacos , Piridinas/farmacología , Pirimidinas/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/efectos de los fármacos , Adenosina Trifosfato/farmacología , Calcio/metabolismo , Células Cultivadas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Técnicas In Vitro , Masculino , Persona de Mediana Edad
12.
J Med Chem ; 38(14): 2551-6, 1995 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-7629795

RESUMEN

A series of 4,5-dihydro-3-[2-(methanesulfonamidophenyl)ethyl]-1-phenyl- 1H-2,4-benzodiazepines has been identified as potential antiarrhythmic agents that interact at the delayed rectifier myocardial potassium channels (IKr) and prolong the ventricular effective refractory period (ERP) in rabbit isolated Langendorff heart preparations. Structure-activity relationship (SAR) studies based upon prolongation of ERP indicate that placement of the sulfonamido group is important for potent activity in this model. Furthermore, methanesulfonamido has enhanced activity over its ethyl or trifluoromethyl analogs. Slightly greater activity was observed in compounds that had a heteroatom in the ethyl bridge that connects the methanesulfonamidophenyl to the benzodiazepine. Further incremental improvements in activity were noted when the 1-phenyl ring was substituted with a variety of substituents. Chirality of the compounds of interest in this series does not appear to influence activity in this model. Several of these compounds were chosen for advanced evaluation, and all possess high selectivity for blockade of potassium current in hearts relative to other ion channels. In addition, these compounds prolong cardiac refractoriness in dogs following oral dosing. Thus, these agents may represent potential new class III agents, but with the potential liability of myocardial IKr blockers.


Asunto(s)
Antiarrítmicos/farmacología , Benzodiazepinas/farmacología , Animales , Antiarrítmicos/química , Antiarrítmicos/metabolismo , Benzodiazepinas/química , Benzodiazepinas/metabolismo , Canales de Calcio/metabolismo , Perros , Humanos , Técnicas In Vitro , Lactante , Espectroscopía de Resonancia Magnética , Masculino , Contracción Miocárdica/efectos de los fármacos , Conejos , Ensayo de Unión Radioligante , Canales de Sodio/metabolismo , Estereoisomerismo , Relación Estructura-Actividad
13.
J Cardiovasc Pharmacol ; 25(1): 14-21, 1995 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-7723343

RESUMEN

We describe the biochemical and pharmacologic effects of two novel fused pyridinones derived from milrinone: WIN 58993 and WIN 62005. Both WIN 58993 and WIN 62005 competitively inhibit cyclic GMP-inhibitable low Km cyclic AMP phosphodiesterase (PDE III) from rat heart and canine aorta with Ki values of 25 +/- 3 and 26 +/- 5 nM, respectively, and are selective (at least 160-fold) for PDE III inhibition relative to other PDE isozymes. WIN 58993 and WIN 62005 were given to conscious, chronically instrumented rats and dogs intravenously (i.v.) or perorally (p.o.). Because the doses of WIN 58993 and WIN 62005 required to decrease mean arterial blood pressure (MAP) by 20% were estimated to be 0.9 and 0.7 mg/kg, respectively, the compounds appear to be equipotent after acute i.v. administration in rats. However, the duration of the depressor responses in rats apparently differs since MAP remained significantly decreased 6 h after i.v. or p.o. administration of WIN 58993, but returned to control levels < or = 4 h after administration of WIN 62005. WIN 58993 may be slightly less potent than WIN 62005 after acute i.v. administration to dogs since significant increases in left ventricular (LV)dP/dtmax first occurred at doses of 0.1 and 0.03 mg/kg, respectively. LVdP/dtmax significantly increased in 30 min and returned to baseline 3 h after p.o. administration of 1 mg/kg WIN 58993. After p.o. administration of 1 mg/kg WIN 62005. LVdP/dtmax was significantly increased in 30 min and remained increased for at least 6 h. These data suggest that WIN 58993 and WIN 62005 are potent, selective, p.o.-active inhibitors of PDE III.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Cardiotónicos/farmacología , Imidazoles/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Piridinas/farmacología , Piridonas/farmacología , Tiazoles/farmacología , Administración Oral , Animales , Aorta/enzimología , Presión Sanguínea/efectos de los fármacos , GMP Cíclico/antagonistas & inhibidores , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3 , Perros , Relación Dosis-Respuesta a Droga , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Inyecciones Intravenosas , Isoenzimas/antagonistas & inhibidores , Masculino , Milrinona , Contracción Miocárdica/efectos de los fármacos , Miocardio/enzimología , Piridonas/química , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Relación Estructura-Actividad , Vasodilatadores/farmacología , Función Ventricular Izquierda/efectos de los fármacos
14.
J Pharmacol Exp Ther ; 271(3): 1143-9, 1994 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7996419

RESUMEN

The objectives of this study were to determine the potency and selectivity of the structurally novel cyclic nucleotide phosphodiesterase (PDE) inhibitor, WIN 58237 (1-cyclopentyl-3-methyl-6-(4- pyridyl)pyrazolo[3,4-d]pyrimidin-4-(5H)-one), and to determine if this compound possesses cyclic GMP (cGMP) PDE inhibitory activity in vitro and in vivo. WIN 58237 is a competitive inhibitor of cGMP PDE V from canine aorta, with a Ki value of 170 nM. It is a relatively less potent inhibitor of calmodulin-sensitive PDE I and cGMP-inhibitable cyclic AMP PDE III; but does inhibit cyclic AMP PDE IV with an IC50 value of approximately 300 nM. In vitro, WIN 58237 is a functional cGMP PDE inhibitor at submicromolar concentrations as evident by potentiation of both sodium nitroprusside- and atrial natriuretic factor-mediated vasorelaxation of contracted, endothelial-denuded rat aortic rings. Moreover, WIN 58237 possesses vasorelaxant activity in the presence of an intact endothelium or nitric oxide. Similar results are evident in vivo, as WIN 58237 (0.3-3.0 mg/kg i.v.) decreases mean arterial pressure in conscious spontaneously hypertensive rats with an associated increase in vascular (aortic) cGMP content in vivo. Both the decrease in mean arterial blood pressure and increase in aortic cGMP content are attenuated by the nitric oxide synthase inhibitor, N omega-nitro-l-arginine. However, WIN 58237 may possess an additional depressor mechanism of action. WIN 58237 restores vasorelaxation responsiveness to nitroglycerin in vitro and in vivo in models of vascular tolerance.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
3',5'-GMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Inhibidores de Fosfodiesterasa/farmacología , Pirimidinas/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Sinergismo Farmacológico , Guanilato Ciclasa/metabolismo , Técnicas In Vitro , Masculino , Nitroglicerina/farmacología , Nitroprusiato/farmacología , Purinonas/farmacología , Ratas , Ratas Endogámicas SHR , Ratas Sprague-Dawley , Vasodilatación/efectos de los fármacos
15.
J Cardiovasc Pharmacol ; 24(3): 403-10, 1994 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-7528296

RESUMEN

We describe the biochemical, pharmacologic, and in vivo pharmacodynamic profiles of two novel inhibitors of the cyclic GMP-inhibitable, low Km cyclic AMP phosphodiesterase (PDE) III; WIN 63291, a 6-quinolinyl analogue of the prototypic PDE III inhibitor milrinone and WIN 62582, an imidazopyridinone. Both WIN 62582 and WIN 63291 competitively inhibit PDE III from rat, dog, and human heart and from rat and canine aorta with IC50 values of 5-37 and 55-80 nM, respectively; the IC50 values for milrinone ranged from 300 to 520 nM. WIN 62582 and WIN 63291 are at least 1,000-fold selective for PDE III relative to inhibition of PDE isozymes I, II, IV, and V. We evaluated WIN 62582 and WIN 63291 in conscious rats and dogs after intravenous (i.v.) and oral (p.o.) administration. The dose of WIN 62582 required to reduce mean arterial blood pressure (MAP) by 20% (ED20) in rats was 1.8 mg/kg, with a pharmacodynamic duration of action of approximately 2 h. In comparison, the estimated i.v. ED20 for WIN 63291 in rats was 0.4 mg/kg, with a pharmacodynamic duration of action > 6 h. In conscious dogs, the i.v. doses of WIN 62582 and 63291 required to increase left ventricular (LV)dP/dtmax significantly were 0.1 and 0.01 mg/kg, respectively. In dogs, WIN 63291 0.1 mg/kg p.o. increased LVdP/dtmax by 86% in 30 min; LVdP/dtmax remained increased by 60% for at least 6 h. In comparison, WIN 62582, 0.3 mg/kg p.o., increased LVdP/dt by 56% in 30 min and remained increased by 40% at 6 h.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Corazón/efectos de los fármacos , Imidazoles/farmacología , Músculo Liso Vascular/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Piridinas/farmacología , Quinolinas/farmacología , 3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Administración Oral , Animales , Aorta/efectos de los fármacos , Aorta/enzimología , Presión Sanguínea/efectos de los fármacos , Perros , Relación Dosis-Respuesta a Droga , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Imidazoles/administración & dosificación , Imidazoles/farmacocinética , Inyecciones Intravenosas , Isoenzimas/antagonistas & inhibidores , Masculino , Milrinona , Músculo Liso Vascular/enzimología , Miocardio/enzimología , Inhibidores de Fosfodiesterasa/administración & dosificación , Inhibidores de Fosfodiesterasa/farmacocinética , Piridinas/administración & dosificación , Piridinas/farmacocinética , Piridonas/química , Piridonas/farmacología , Quinolinas/administración & dosificación , Quinolinas/farmacocinética , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
16.
Eur J Pharmacol ; 266(3): 245-54, 1994 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-8174607

RESUMEN

Pharmacological modulation of human sodium current was examined in Xenopus oocytes expressing human heart Na+ channels. Na+ currents activated near -50 mV with maximum current amplitudes observed at -20 mV. Steady-state inactivation was characterized by a V1/2 value of -57 +/- 0.5 mV and a slope factor (k) of 7.3 +/- 0.3 mV. Sodium currents were blocked by tetrodotoxin with an IC50 value of 1.8 microM. These properties are consistent with those of Na+ channels expressed in mammalian myocardial cells. We have investigated the effects of several pharmacological agents which, with the exception of lidocaine, have not been characterized against cRNA-derived Na+ channels expressed in Xenopus oocytes. Lidocaine, quinidine and flecainide blocked resting Na+ channels with IC50 values of 521 microM, 198 microM, and 41 microM, respectively. Use-dependent block was also observed for all three agents, but concentrations necessary to induce block were higher than expected for quinidine and flecainide. This may reflect differences arising due to expression in the Xenopus oocyte system or could be a true difference in the interaction between human cardiac Na+ channels and these drugs compared to other mammalian Na+ channels. Importantly, however, this result would not have been predicted based upon previous studies of mammalian cardiac Na+ channels. The effects of DPI 201-106, RWJ 24517, and BDF 9148 were also tested and all three agents slowed and/or removed Na+ current inactivation, reduced peak current amplitudes, and induced use-dependent block. These data suggest that the alpha-subunit is the site of interaction between cardiac Na+ channels and Class I antiarrhythmic drugs as well as inactivation modifiers such as DPI 201-106.


Asunto(s)
Cardiotónicos/farmacología , Miocardio/metabolismo , Canales de Sodio/efectos de los fármacos , Animales , Azetidinas/farmacología , Electrofisiología , Femenino , Flecainida/farmacología , Humanos , Lidocaína/farmacología , Mercaptopurina/análogos & derivados , Mercaptopurina/farmacología , Oocitos/metabolismo , Piperazinas/farmacología , Quinidina/farmacología , ARN Complementario/genética , Canales de Sodio/metabolismo , Tetrodotoxina/farmacología , Transcripción Genética , Xenopus laevis
17.
Eur J Pharmacol ; 249(3): 293-7, 1993 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-8287916

RESUMEN

The purpose of this study was to determine if significant relationships exist between plasma and aortic cyclic GMP (cGMP) levels and pharmacodynamic effect after the i.v. administration of the cGMP-selective phosphodiesterase inhibitor zaprinast to conscious, spontaneously hypertensive rats. Zaprinast dose-dependently increased plasma and aortic cGMP levels at 10, 18 and 30 mg/kg and decreased mean arterial blood pressure (MAP) at 18 and 30 mg/kg. The concentrations of cGMP in the plasma and in the aorta were significantly correlated (r = 0.765, P < 0.0001). The changes in MAP were significantly correlated to aortic (r = -0.750, P < 0.0001) and plasma (r = -0.762, P < 0.0001) cGMP levels. We conclude that plasma cGMP may be an index of cGMP-selective phosphodiesterase inhibition in vivo.


Asunto(s)
3',5'-GMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Aorta Abdominal/metabolismo , GMP Cíclico/sangre , GMP Cíclico/metabolismo , Purinonas/farmacología , Análisis de Varianza , Animales , Presión Sanguínea/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Hipertensión/fisiopatología , Inyecciones Intravenosas , Masculino , Purinonas/administración & dosificación , Radioinmunoensayo , Ratas , Ratas Endogámicas SHR
18.
Eur J Pharmacol ; 243(2): 141-7, 1993 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-8276063

RESUMEN

Following in vitro exposure of rat aortic rings to 550 microM nitroglycerin for 1 h, tolerance was demonstrated by a significant increase in EC50 values for nitroglycerin-induced relaxation. However, cross-tolerance to sodium nitroprusside was not observed. Co-incubation of aortic rings with the cGMP-phosphodiesterase (cGMP-PDE) inhibitor zaprinast (10 microM), during incubation with 550 microM nitroglycerin, did not prevent the development of tolerance. However, the addition of 0.30 or 10 microM zaprinast to tolerant aortic rings did restore responsiveness to nitroglycerin. The increase in cGMP in tolerant aortic rings in response to 300 nM nitroglycerin (2-4 fmol/micrograms) was significantly less than that observed for non-tolerant rings (6.6-12 fmol/micrograms), but cGMP levels were restored in tolerant rings by zaprinast (7-12 fmol/micrograms). These data suggest that inhibition of vascular cGMP-PDE activity does not prevent the development of tolerance in vitro, but does reverse the loss of vasorelaxant potency to nitroglycerin via restoration of intracellular cGMP levels.


Asunto(s)
3',5'-GMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Nitroglicerina/farmacología , Purinonas/farmacología , 3',5'-GMP Cíclico Fosfodiesterasas/aislamiento & purificación , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/enzimología , GMP Cíclico/metabolismo , Tolerancia a Medicamentos , Hidrólisis , Técnicas In Vitro , Masculino , Relajación Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Músculo Liso/enzimología , Músculo Liso/metabolismo , Ratas , Ratas Sprague-Dawley
19.
Methods Find Exp Clin Pharmacol ; 15(8): 527-34, 1993 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8309316

RESUMEN

Increases in intracellular cAMP levels have previously been shown to decrease intraocular pressure (IOP) and increase ocular blood flow (OBF). However, the ocular effects of milrinone, which increases intracellular cAMP levels via selective cAMP PDE III inhibition, have not been investigated. The purpose of this study was to investigate the ocular effects of topically administered milrinone at different concentrations in rabbits and cats. When compared to vehicle in conscious rabbits, topical administration of milrinone at 0.03% decreased IOP (-14.1 +/- 2.6% vs. -7.4 +/- 3.7%, max. changes expressed as mean +/- SEM), at 0.1% increased IOP (10.4 +/- 8.5% vs. -1.7 +/- 4.1%), and at 0.01% and 1% did not significantly affect IOP. Neither pupil size nor central corneal thickness were affected by milrinone. Additionally, there were no signs of inflammation and no effects on corneal clarity. In anesthetized cats, topical administration of milrinone at 0.01-0.3% increased OBF (38.9 +/- 6.0% for milrinone vs. -7.4 +/- 4.4% for vehicle), and at 0.03%-0.3% decreased mean arterial pressure (-19.0 +/- 5.6 vs. 3.0 +/- 4.1 mmHg) in a dose-related manner. The durations of OBF enhancement (1.5-2.5 h) and MAP reduction (less than 30 min to 2 h) were also dose-dependent. In conclusion, milrinone induced biphasic IOP effects: IOP was decreased at 0.03% but increased at 0.1%. Milrinone at 0.01% increased OBF, possibly via a local vasodilator effect, and at 0.03-0.3% increased OBF, possibly via local and systemic effects. These data suggest that cAMP PDE III inhibitors such as milrinone may have efficacy as agents which enhance ocular blood flow following topical ocular application.


Asunto(s)
Ojo/efectos de los fármacos , Presión Intraocular/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Piridonas/farmacología , Administración Tópica , Análisis de Varianza , Animales , Humor Acuoso/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Gatos , Relación Dosis-Respuesta a Droga , Ojo/irrigación sanguínea , Femenino , Masculino , Milrinona , Soluciones Oftálmicas , Inhibidores de Fosfodiesterasa/administración & dosificación , Conejos , Reflejo Pupilar/efectos de los fármacos , Flujo Sanguíneo Regional
20.
Eur J Pharmacol ; 234(1): 77-82, 1993 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-8386094

RESUMEN

The purpose of this study was to determine whether cross-tolerance develops between nitroglycerin and endothelium-derived relaxing factor (EDRF)-mediated vasoactive agents in vivo. Spontaneously hypertensive rats (SHR) were made tolerant by pretreatment with high doses of nitroglycerin (100 mg/kg s.c., 3 times/day, for 3 consecutive days). The hypotensive effect of challenge doses of nitroglycerin (1, 10, 300, 100 micrograms/kg i.v.) was completely abolished in nitroglycerin-pretreated SHR. To evaluate cross-tolerance, the effects of the following EDRF-dependent vasoactive agents on blood pressure were determined in groups of nitroglycerin-pretreated and vehicle-pretreated SHR: acetylcholine, bradykinin and L-arginine. In addition, the hypotensive effects of zaprinast (M & B 22,928), a cyclic guanosine monophosphate (cGMP) phosphodiesterase inhibitor, and the hypertensive effects of the nitric oxide-synthase inhibitor N omega-nitro-L-arginine were also evaluated. In all cases, there was no difference in the effects of these agents on blood pressure when compared in nitroglycerin-pretreated (tolerant) and vehicle-pretreated (non-tolerant) SHR. The use of a variety of agents which modulate EDRF release or its effects by several different mechanisms suggests that cross-tolerance does not occur between nitroglycerin and EDRF in vivo.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Hipertensión/fisiopatología , Óxido Nítrico/fisiología , Nitroglicerina/farmacología , Vasodilatadores/farmacología , 3',5'-GMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Acetilcolina/farmacología , Animales , Arginina/análogos & derivados , Arginina/farmacología , Bradiquinina/farmacología , Tolerancia a Medicamentos , Masculino , Nitroarginina , Purinonas/farmacología , Ratas , Ratas Endogámicas SHR
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...