Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Int J Immunopharmacol ; 17(7): 571-80, 1995 Jul.
Article in English | MEDLINE | ID: mdl-8586485

ABSTRACT

Multiple organ dysfunction (MOD) is the leading cause of mortality in septic patients with circulatory shock. Recent evidence suggests that the overproduction of the cytokine, tumor necrosis factor-alpha(TNF), and oxygen free radical molecules may mediate the progression of sepsis to MOD and death. In this study, we have examined the ability of MDL 101,002, a free radical scavenger, to reduce organ dysfunction and cytokine secretion induced by lipopolysaccharide (LPS) administration in rats. Treatment with MDL 101,002(10-60 ng/kg, i.p.) 30 min prior to an LPS challenge resulted in a dose-dependent reduction in several markers indicative of organ dysfunction and mortality. MDL 101,002 markedly decreased LPS-induced liver and kidney damage as indicated by serum levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) or urea and creatinine, respectively. MDL 101,002 also prevented LPS-induced pulmonary edema, but did not prevent leukopenia and only partially reduced thrombocytopenia. Associated with these improvements in organ dysfunction and survival was a modest decrease in LPS-stimulated interleukin-1 alpha (IL-1 alpha) and interleukin-1 beta (IL-1 beta) secretion and a marked ( > 90%) inhibition of TNF secretion by MDL 101,002. The data are consistent with a role for oxygen free radicals in the development of endotoxin-induced organ dysfunction and shock and suggest that free radical scavengers could reduce the mortality consequent to sepsis by decreasing organ dysfunction, at least in part, through a reduction in free radical stimulated cytokine secretion.


Subject(s)
Antioxidants/pharmacology , Cytokines/biosynthesis , Escherichia coli/metabolism , Isoquinolines/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Nitrogen Oxides/pharmacology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Cell Survival/drug effects , Interleukin-1/biosynthesis , Interleukin-2/biosynthesis , Male , Pulmonary Edema/chemically induced , Pulmonary Edema/metabolism , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/biosynthesis
2.
J Pharmacol Exp Ther ; 274(1): 444-9, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7616429

ABSTRACT

Perhexiline maleate is an antianginal drug that has been shown to have antiarrhythmic effects in humans. To examine whether some of these clinical observations could be caused by block of cardiac K+ channels, we examined the effects of perhexiline on a rapidly activating delayed rectifier K+ channel (Kv1.5) cloned from human heart and stably expressed in human embryonic kidney cells as well as a corresponding K+ current (the ultra-rapid delayed rectifier, IKur) in human atrial myocytes. With the use of inside-out macropatches, we found that perhexiline inhibited Kv1.5 current in a time- and voltage-dependent manner with an IC50 value of 1.5 x 10(-6) M at +50 mV. Perhexiline reduced Kv1.5 tail current amplitude and slowed its decay relative to control. These data are consistent with blockade of open channels, probably from the intracellular surface. Perhexiline (3 microM) also blocked IKur in human atrial myocytes. The block that was observed was both time- and voltage-dependent in qualitatively similar ways to block of Kv1.5 channels. However, the time-dependent block of IKur by perhexiline was somewhat slower and its voltage-dependence steeper relative to its effects on Kv1.5. These data indicate that perhexiline blocks both cloned and native human cardiac K+ channels. Blockade of one or more types of voltage-dependent K+ channels may explain some of the electrophysiological effects of perhexiline observed in humans.


Subject(s)
Heart Atria/drug effects , Perhexiline/pharmacology , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Atrial Function , Cells, Cultured , Cloning, Molecular , Heart Atria/metabolism , Humans , Kv1.5 Potassium Channel , Membrane Potentials , Potassium Channels/genetics
3.
J Cardiovasc Pharmacol ; 26(1): 107-13, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7564349

ABSTRACT

Angiotensin converting enzyme (ACE) and neutral endopeptidase (NEP), are two mechanistically similar enzymes involved in the metabolism of several vasoactive peptides. Selective inhibitors of ACE are effective antihypertensive agents in high-renin, renovascular rats and normal-renin, spontaneously hypertensive rats (SHR), but are not effective in the low-renin, deoxycorticosterone acetate (DOCA)-salt hypertensive rats. In contrast, NEP inhibitors are only effective in the low-renin model of hypertension. Treatment with a combination of selective inhibitors or with a dual inhibitor of both enzymes produces an antihypertensive response regardless of basal plasma renin activity. In this study, we compared the activities of MDL 100,173, a novel subnanomolar inhibitor of both ACE and NEP, with those of equimolar doses of captopril, a selective ACE inhibitor, following intravenous administration in these three rat models of hypertension. Treatment with MDL 100,173 significantly lowered blood pressure compared to vehicle treatment in all three models, whereas captopril treatment lowered blood pressure in the renovascular and SHR models only. Administration of MDL 100,173 also significantly elevated diuresis and natriuresis compared to either vehicle or captopril treatment in the SHR and DOCA-salt rats. Urinary excretion of atrial natriuretic peptide (ANP) was increased by MDL 100,173 treatment in all three models of hypertension. Treatment with captopril did not alter urine, sodium, or ANP excretion in any of the models. However, plasma-renin activity was elevated by both MDL 100,173 and captopril '''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''' ''''''''


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Benzazepines/therapeutic use , Hypertension/drug therapy , Neprilysin/antagonists & inhibitors , Protease Inhibitors/therapeutic use , Pyridines/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Atrial Natriuretic Factor/urine , Benzazepines/administration & dosage , Benzazepines/pharmacology , Blood Pressure/drug effects , Captopril/administration & dosage , Captopril/pharmacology , Captopril/therapeutic use , Disease Models, Animal , Diuresis/drug effects , Hypertension/enzymology , Hypertension, Renal/drug therapy , Male , Protease Inhibitors/administration & dosage , Protease Inhibitors/pharmacology , Pyridines/administration & dosage , Pyridines/pharmacology , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Sodium/urine
4.
J Cardiovasc Pharmacol ; 24(5): 761-72, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7532754

ABSTRACT

We wished to characterize MDL 28,133A (1-(4-fluorophenyl)-2-[4-[(4-methanesulfonamidophenyl)carbonyl]-1- piperidinyl-ethanone HCl), a potent 5-HT2 receptor antagonist, on platelet aggregation and platelet thrombosis and determined its effect on thrombolysis with streptokinase (SK) in dogs with coronary thrombosis. MDL 28,133A and ritanserin, 0.3-1 microM, competitively inhibited 5-HT-induced platelet aggregation in dog platelet-rich plasma (PRP) in vitro. The pA2 values and slopes were 6.29 +/- 0.09, -0.96 +/- 0.14, and 6.58 +/- 0.09, =1.64 +/- 0.34 for MDL 28,133A and ritanserin, respectively, suggesting that both agents are similar in potency to 5-HT2 receptor antagonists. MDL 28,133A (0.01 mg/kg, p.o.) produced inhibition of arachidonic acid-induced platelet aggregation in whole blood from conscious dogs ex vivo for > or = 2 h, indicating oral bioavailability. The magnitude and duration of the effect of MDL 28,133A on platelet aggregation in whole blood was similar to that of ketanserin (2.5 mg/kg, p.o.). MDL 28,133A (0.001-0.03 mg/kg, i.v.) completely abolished cyclic flow reductions (CFRs) in stenosed and partially deendothelialized left anterior descending coronary arteries of anesthetized dogs for at least 120 min without affecting systemic blood pressure (BP) and heart rate, thus indicating that MDL 28,133A is a potent antithrombotic agent. Ketanserin (0.5 mg/kg, i.v.) also abolished CFRs, but produced a significant decrease in systemic BP as well. MDL 28,133A (0.001 mg/kg, i.v.) shortened time to reperfusion (15 +/- 1 vs. 36 +/- 8 min), prolonged time to reocclusion (112 +/- 6 vs. 85 +/- 6 min), and increased total volume reflow (20 +/- 2 vs. 10 +/- 2%) in dogs with coronary artery thrombosis undergoing thrombolysis with SK (1,000 U/min, i.a.). Reocclusion rate was not affected by MDL 28,133A (4 of 5 vs. 4 of 6). Treatment with heparin (150 U/kg, i.v. every hour for 2 h) alone or in combination with MDL 28,133A did not improve time to reperfusion but enhanced total volume reflow and prevented reocclusion. MDL 28,133A is a potent 5-HT2 receptor antagonist that inhibits platelet thrombosis and facilitates thrombolysis in vivo. The impeding effect of MDL 28,133A in coronary thrombosis and the lack of hemodynamic effects suggests that MDL 28,133A may be of benefit in treatment of hyperthrombotic conditions without having hemodynamic side effects.


Subject(s)
Coronary Thrombosis/drug therapy , Piperidines/pharmacology , Platelet Aggregation/drug effects , Serotonin Antagonists/pharmacology , Administration, Oral , Animals , Biological Availability , Blood Platelets/drug effects , Blood Pressure/drug effects , Disease Models, Animal , Dogs , Drug Therapy, Combination , Electric Stimulation , Female , Heart Rate/drug effects , Heparin/administration & dosage , Heparin/pharmacology , Heparin/therapeutic use , In Vitro Techniques , Male , Piperidines/administration & dosage , Piperidines/therapeutic use , Ritanserin/administration & dosage , Ritanserin/pharmacology , Ritanserin/therapeutic use , Serotonin/metabolism , Serotonin/toxicity , Serotonin Antagonists/administration & dosage , Serotonin Antagonists/therapeutic use , Streptokinase/administration & dosage , Streptokinase/pharmacology , Streptokinase/therapeutic use
5.
Circ Shock ; 43(3): 130-6, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7850933

ABSTRACT

Evidence of a role for oxygen-derived free radicals in the pathophysiology of endotoxic shock has been found in animal models. However, the importance of free radicals in chronic models of bacterial infection has not been examined. In this study a novel nitrone radical spin trap is described and its activity in animal models of endotoxic shock and chronic bacteremia were explored. MDL 101,002 is a cyclized variant of alpha-phenyl N-tert-butyl nitrone (PBN), an established spin trap. MDL 101,002 can react with free radicals to form persistent adducts as demonstrated by electron paramagnetic resonance (EPR) spectroscopy. This agent is about 10 times more potent than PBN as an in vitro antioxidant and scavenger of hydroxyl radicals. In a rat endotoxic shock model MDL 101,002 (3-30 mg/kg, i.p.) administered 30 min prior to endotoxin (30 mg/kg, i.p.) treatment reduced mortality in a dose-dependent manner. Peroxide-enhanced chemiluminescence in hepatic homogenates from endotoxin treated rats was elevated indicating that oxidative stress and antioxidant depletion was increased. Importantly, treatment with MDL 101,002 (30 mg/kg, i.p.) 30 min prior to, and 120 min following endotoxin, minimized the increase in chemiluminescence. MDL 101,002 also reduced mortality in a model of chronic bacteremia employing implantation of infected fibrin clots into the peritoneal cavity of gentamicin-treated leukopenic rats. MDL 101,002 (2.5 mg/kg/hr) increased survival from 24% to 52% in these rats. These data are consistent with a role for free radicals in the pathophysiology of endotoxic shock and suggest free radicals are also important mediators in chronic models of sepsis.


Subject(s)
Antioxidants/pharmacology , Bacteremia/prevention & control , Free Radical Scavengers/pharmacology , Isoquinolines/pharmacology , Nitrogen Oxides/pharmacology , Shock, Septic/prevention & control , Animals , Antioxidants/therapeutic use , Bacteremia/metabolism , Chronic Disease , Free Radical Scavengers/therapeutic use , Free Radicals , Hydroxyl Radical , Isoquinolines/therapeutic use , Leukopenia/metabolism , Male , Nitrogen Oxides/therapeutic use , Rats , Rats, Sprague-Dawley , Shock, Septic/metabolism
6.
J Pharmacol Exp Ther ; 268(1): 180-6, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8301555

ABSTRACT

In the present study we characterize key activities of an agent designed to simultaneously inhibit angiotensin I-converting enzyme (ACE) and neutral endopeptidase (NEP). MDL 100,240 is a thioester prodrug of MDL 100,173, which is a potent competitive inhibitor of both ACE and NEP in vitro. MDL 100,240 was shown in an ex vivo study to inhibit both of these enzymes in rat kidney. When administered to anesthetized rats, MDL 100,240 enhanced the effect of infused ANP on blood pressure, diuresis and natriuresis and of infused bradykinin on blood pressure. Moreover, MDL 100,173 and MDL 100,240 inhibited the pressor response to angiotensin I. These results indicate that MDL 100,173 and its prodrug, MDL 100,240, produced effects, in vivo, consistent with inhibition of both ACE and NEP.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Benzazepines/pharmacology , Neprilysin/antagonists & inhibitors , Peptidyl-Dipeptidase A/metabolism , Pyridines/pharmacology , Animals , Atrial Natriuretic Factor/pharmacology , Bradykinin/pharmacology , Male , Molecular Structure , Neprilysin/metabolism , Prodrugs/pharmacology , Rats , Rats, Sprague-Dawley
7.
Mol Pharmacol ; 44(6): 1240-5, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8264561

ABSTRACT

Use of the nonsedating antihistamine terfenadine has been associated with altered cardiac repolarization in certain clinical settings. For this reason we examined the effects of terfenadine, and its metabolites, on a rapidly activating delayed rectifier K+ channel (fHK) cloned from human heart. fHK was stably expressed in human embryonic kidney cells, and both whole-cell currents and currents from excised inside-out patches were recorded. Terfenadine (3 microM) blocked whole-cell fHK current by 72 +/- 6%. In inside-out patches, terfenadine applied to the cytoplasmic surface blocked fHK with an IC50 value of 367 nM. The main effect of terfenadine was to enhance the rate of inactivation of fHK current and thereby reduce the current at the end of a prolonged voltage-clamp pulse. The blockade displayed a weak voltage dependence, increasing at more positive potentials. The mechanism of action of terfenadine is therefore consistent with blockade of open channels. In contrast, the metabolites of terfenadine were weakly active on fHK. IC50 values for all of the metabolites tested ranged from 27-fold to 583-fold higher than that obtained for terfenadine. It is concluded that terfenadine, but not its metabolites, blocks at least one type of human cardiac K+ channel at clinically relevant concentrations and that this activity may underlie the cardiac arrhythmias that have been associated with the use of this drug.


Subject(s)
Myocardium/metabolism , Potassium Channels/drug effects , Terfenadine/pharmacology , Cell Line , Cloning, Molecular , Humans , Kidney/cytology , Kidney/embryology , Kidney/metabolism , Potassium Channels/genetics , Potassium Channels/physiology , Terfenadine/metabolism
8.
Mol Pharmacol ; 44(3): 642-8, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8371716

ABSTRACT

Verapamil is an antagonist of L-type Ca2+ channels, and part of its binding site is located in the sixth transmembrane segment (S6) in the fourth repeat of the protein. Verapamil also blocks K+ channels, which are members of the same supergene family as Ca2+ channels. We examined the effects of verapamil on a rapidly activating delayed rectifier K+ channel (designated fHK) cloned from human heart. Verapamil inhibited 86Rb+ efflux from fHK-transfected human embryonic kidney cells with an EC50 of 4.5 x 10(-5) M. Whole-cell patch-clamp experiments revealed that verapamil induced a rapid component of fHK current inactivation but was without effect on activation. The effect was concentration and voltage dependent and was attributed to open channel blockade. The apparent association and dissociation rate constants measured at +50 mV were about 1.65 x 10(5) M-1 sec-1 and 3.48 sec-1, respectively. S6 of fHK has significant homology to that portion of the verapamil binding site identified in Ca2+ channels, and S6 is thought to form part of the inner mouth of K+ channel pores. The data support a role for verapamil as a blocker of the inner pore of voltage-dependent K+ channels in human myocardium.


Subject(s)
Myocardium/chemistry , Potassium Channels/drug effects , Verapamil/pharmacology , Amino Acid Sequence , Cell Line , Humans , Membrane Potentials/drug effects , Molecular Sequence Data , Potassium Channels/metabolism , Recombinant Proteins/drug effects , Rubidium Radioisotopes , Sequence Homology, Amino Acid
10.
J Pharmacol Exp Ther ; 265(3): 1125-30, 1993 Jun.
Article in English | MEDLINE | ID: mdl-7685384

ABSTRACT

We compared the cardiovascular effects of two structurally distinct L-type Ca++ channel activators, the 1,4-dihydropyridine Bay K 8644 and the benzoylpyrrole FPL 64176. Both compounds prolonged action potential duration and enhanced contractility in guinea pig papillary muscle with these responses being greater in the presence of FPL 64176 compared to (S)-Bay K 8644. (S)-Bay K 8644 (300 nM) and FPL 64176 (300 nM) increased whole-cell Ca++ channel current amplitude in neonatal rat ventricular cells by 249 +/- 14 and 484 +/- 100%, respectively. (S)-Bay K 8644 had little effect on Ca++ channel activation but significantly enhanced the rate of Ca++ channel current inactivation. FPL 64176 significantly slowed Ca++ channel current activation and inactivation. Tail current decay at -50 mV was monoexponential in the presence of (S)-Bay K 8644 and had a time constant of 4.59 +/- 0.16 msec. FPL 64176 produced biexponential tail current decays at -50 mV with fast and slow time constants of 4.30 +/- 0.30 and 44.52 +/- 4.56 msec, respectively. Intravenous administration (1-100 micrograms/kg) of Bay K 8644 and FPL 64176 produced large increases in cardiac contractile force and diastolic blood pressure in anesthetized dogs. Pretreatment with nifedipine attenuated the blood pressure response to FPL 64176 but not the effects on cardiac contractility. This study demonstrates that the benzoylpyrrole FPL 64176 defines a new and potent class of Ca++ channel agonist molecule and that this compound has pharmacological activity that differs, at least in some respects, from the 1,4-dihydropyridine group of agonists.


Subject(s)
3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Calcium Channels/drug effects , Heart/drug effects , Pyrroles/pharmacology , Action Potentials , Animals , Blood Pressure/drug effects , Calcium Channels/physiology , Guinea Pigs , Heart/physiology , In Vitro Techniques , Male , Myocardial Contraction/drug effects , Rats
11.
Eur J Pharmacol ; 233(1): 109-12, 1993 Mar 16.
Article in English | MEDLINE | ID: mdl-8472739

ABSTRACT

A fluorescently labeled ligand was utilized to establish the existence of an interleukin-1 (IL-1) receptor in vascular smooth muscle. The binding of the phycoerythrin-labeled IL-1 beta to the murine T cell line, EL-4, was examined as a positive control. The phycoerythrin-labeled IL-1 beta identified a specific IL-1 receptor in the EL-4 cells. Vascular smooth muscle cells were also positively stained by the fluorescent ligand. The binding of phycoerythrin-labeled IL-1 beta to these cells was saturable and reversed by 100-fold excess unlabeled IL-1 beta. Incubation of the vascular smooth muscle cells with IL-1 beta (25 ng/ml) or IL-6 (250 ng/ml) for 18 h increased and decreased, respectively, the percentage of cells positively stained by phycoerythrin-labeled IL-1 beta which suggests these cytokines regulate IL-1 receptor expression in these cells. These data indicate a specific receptor for IL-1 exists in vascular smooth muscle cells.


Subject(s)
Interleukin-1/pharmacology , Interleukin-6/pharmacology , Muscle, Smooth, Vascular/metabolism , Receptors, Interleukin-1/biosynthesis , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Fluorescent Dyes , Male , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Phycoerythrin/metabolism , Rats , Rats, Wistar , Receptors, Interleukin-1/drug effects
12.
Mol Pharmacol ; 41(4): 599-602, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1373795

ABSTRACT

We examined the interactions of two Ca2+ channel activators, (S)-Bay K 8644 and FPL 64176, on smooth muscle L-type Ca2+ channels. FPL 64176 (300 nM) caused a sustained contraction of rat tail artery strips. This contractile response was inhibited by approximately 70% by (S)-Bay K 8644 (EC50 = 14 nM). (S)-Bay K 8644 (100 nM) increased whole-cell Ca2+ currents in A7r5 smooth muscle cells but effectively blocked further stimulation by 1 microM FPL 64176. When added alone, 1 microM FPL 64176 increased Ca2+ channel current amplitude, slowed current activation, and prolonged tail current duration. Furthermore, no inactivation of current during step depolarizations was observed in the presence of FPL 64176. After subsequent addition of (S)-Bay K 8644, Ca2+ channel current activation was accelerated and tail current duration was shortened. Additionally, pronounced inactivation of the Ca2+ channel current became apparent. These results are consistent with a negative allosteric interaction between the (S)-Bay K 8644 binding site and that of FPL 64176, in smooth muscle.


Subject(s)
3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Calcium Channels/drug effects , Muscle, Smooth, Vascular/drug effects , Pyrroles/pharmacology , Animals , Drug Interactions , In Vitro Techniques , Kinetics , Male , Membrane Potentials , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Rats
13.
Am J Physiol ; 261(6 Pt 2): H1945-50, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1661094

ABSTRACT

We have tested the effects of the active 1-34 amino acid sequence of rat parathyroid hormone (PTH) on Ca2+ channel activity in neonatal rat ventricular cells. Rat PTH (30 pM to 10 nM) increased depolarization-induced Ca2+ influx into these cells, an effect that was abolished by 1 microM nifedipine. The 1-34 amino acid sequence of bovine PTH also stimulated Ca2+ influx in control cells but not in cells pretreated with cholera toxin. Rat PTH also elevated adenosine 3',5'-cyclic monophosphate accumulation in these ventricular myocytes. Whole cell voltage-clamp recordings confirmed a stimulatory effect of rat PTH on cardiac L-type Ca2+ channels. Cell-attached single channel recordings revealed an increase in the probability of channel opening as the primary mechanism for the enhancement of Ca2+ current. Taken together these results suggest an important role for PTH as an endogenous modulator of cardiac L-type Ca2+ channels.


Subject(s)
Calcium Channels/physiology , Heart/physiology , Parathyroid Hormone/pharmacology , Animals , Animals, Newborn , Calcium/metabolism , Calcium Channels/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Electric Conductivity , Heart/drug effects , Heart Ventricles/drug effects , Isoproterenol/pharmacology , Parathyroid Hormone/physiology , Potassium/pharmacology , Rats , Ventricular Function
14.
J Pharmacol Exp Ther ; 259(1): 260-4, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1717680

ABSTRACT

Interleukin-1 beta (IL-1) reduces vascular smooth muscle contractility. The purpose of the present study was to investigate the role of nitric oxide synthesis in mediating this effect of IL-1. We studied the influence of inhibitors of nitric oxide synthesis on the depression of norepinephrine-induced contractions of rat aortic rings by IL-1. Also, we examined the ability of IL-1 to increase the production of nitric oxide by rat aortic smooth muscle cells in culture as determined indirectly by measuring nitrite concentrations. NG-amino-L-arginine blocked the effect of IL-1 on norepinephrine-induced contractions of rat aortic rings whereas NG-monomethyl-L-arginine and NG-nitro-L-arginine were considerably less effective. In addition, this effect of IL-1 was prevented by coincubation of the rings with cycloheximide. IL-1 greatly elevated nitrite production by rat aortic smooth muscle cells, and this effect could also be blocked completely by the arginine analogs. NG-amino-L-arginine was the most potent inhibitor of nitrite synthesis (IC50 = 1.7 microM) whereas NG-monomethyl-L-arginine and NG-nitro-L-arginine were about 10-fold less potent (IC50 = 16 and 22 microM, respectively). These results suggest that IL-1-induced depression of norepinephrine-induced vascular contraction is mediated by the increased synthesis of nitric oxide synthase by vascular smooth muscle cells. The relative potency of the arginine analogs for the inhibition of nitrite synthesis suggests that the synthase in vascular smooth muscle is similar to the synthase in macrophages.


Subject(s)
Amino Acid Oxidoreductases/antagonists & inhibitors , Arginine/analogs & derivatives , Interleukin-1/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Nitric Oxide/pharmacology , Norepinephrine/pharmacology , Vasoconstriction/drug effects , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Arginine/pharmacology , Cells, Cultured , Interleukin-1/pharmacology , Male , Nitric Oxide Synthase , Nitroarginine , Norepinephrine/antagonists & inhibitors , Rats , Rats, Inbred Strains
15.
J Cardiovasc Pharmacol ; 17(3): 445-55, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1711607

ABSTRACT

The smooth muscle relaxant effect of MDL 27,032, 4-propyl-5-(4-pyridinyl)-2(3H)-oxazolone, was studied in vitro using strips of femoral arteries and saphenous veins from dogs and trachea from guinea pigs. MDL 27,032 (10(-6)-10(-3) M) produced a concentration-dependent relaxation of arterial and venous strips contracted by carbachol. MDL 27,032 also antagonized contractions of arterial and venous strips produced by phorbol 12,13-dibutyrate (PDB), a protein kinase C activator, both in normal-Ca2+ and zero-Ca2+ medium. The compound inhibited protein kinase C in soluble extracts prepared from saphenous veins of dogs, with an IC50 value of 36.6 microM. MDL 27,032 was more effective against the contractions produced by phenylephrine and serotonin than by KCl in arteries, but no such selectivity was noted in veins. MDL 27,032 (10(-3) M) also inhibited accumulation of inositol phosphates in femoral artery but not in saphenous vein, and this effect may have contributed to the arterial-relaxant effect. Because the vascular smooth muscle relaxant effect of MDL 27,032 was endothelium independent, did not involve blockade of alpha-adrenoceptors or inhibition of cyclic nucleotide phosphodiesterases, stimulation of beta-adrenergic receptors, stimulation of adenosine A2-receptors, or activation of K+ channels, these data suggest that the relaxant effects of MDL 27,032 primarily involve inhibition of protein kinase C.


Subject(s)
Muscle, Smooth, Vascular/drug effects , Oxazoles/pharmacology , Oxazolone/analogs & derivatives , Protein Kinase C/antagonists & inhibitors , Pyridines/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Dogs , Dose-Response Relationship, Drug , In Vitro Techniques , Inositol Phosphates/metabolism , Muscle, Smooth, Vascular/physiology , Phorbol 12,13-Dibutyrate/pharmacology , Potassium Chloride/pharmacology , Protein Kinase Inhibitors
16.
J Cardiovasc Pharmacol ; 17(2): 185-91, 1991 Feb.
Article in English | MEDLINE | ID: mdl-1709221

ABSTRACT

Postischemic myocardial dysfunction in canine myocardium has been reported to be reduced by scavengers of oxygen-derived free radicals. One potential source of oxygen-derived free radicals in canine myocardium is xanthine oxidase, but human and rabbit myocardium either lack or possess very low levels of this enzyme. Therefore, the effects of scavengers of oxygen-derived free radicals on postischemic myocardial dysfunction produced by 15 min of ischemia and 3 h of reperfusion were evaluated in vivo in the rabbit. Superoxide dismutase (SOD) (45,000 U/kg) and catalase (55,000 U/kg) were given into the left atrium 10 min before ischemia, and followed by an additional 45,000 U/kg of SOD and 55,000 U/kg of catalase given over 85 min. This treatment reduced postischemic myocardial dysfunction, as did sulfhydryl-containing free radical scavengers N-2-mercaptopropionyl glycine (4 mg/kg, i.v.) and captopril (3 mg/kg, i.v.) given 5 min before and 60 min after reperfusion. SOD given alone at the same dose was ineffective, as was enalaprilat (0.3 mg/kg, i.v.), an angiotensin-converting enzyme inhibitor that does not scavenge oxygen-derived free radicals. Thus, postischemic myocardial dysfunction was reduced by scavengers of oxygen-derived free radicals in vivo in a species that is deficient in myocardial xanthine oxidase. This suggests that oxygen-derived free radicals derived from a source other than xanthine oxidase play a role in postischemic myocardial dysfunction.


Subject(s)
Free Radical Scavengers , Myocardial Reperfusion Injury/prevention & control , Oxygen/metabolism , Anesthesia , Animals , Blood Pressure/drug effects , Captopril/pharmacology , Catalase/pharmacology , Enalaprilat/pharmacology , Endotoxins/pharmacology , Female , Heart Rate/drug effects , Myocardial Reperfusion Injury/metabolism , Rabbits , Superoxide Dismutase/pharmacology , Tiopronin/pharmacology , Ventricular Function, Left/drug effects
17.
J Pharmacol Exp Ther ; 256(1): 358-64, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1899120

ABSTRACT

We examined the binding of the antidiabetic sulfonylurea [3H] glibenclamide to rat brain and heart membranes. High affinity binding was observed in adult rat forebrain (Kd = 137.3 pM, maximal binding site density = 91.8 fmol/mg of protein) and ventricle (Kd = 77.1 pM, maximal binding site density = 65.1 fmol/mg of protein). Binding site density increased approximately 250% in forebrain membranes during postnatal development but was constant in ventricular membranes. Quantitative autoradiography was used to examine the regional distribution of [3H] glibenclamide binding sites in sections from rat brain, spinal cord and heart. The greatest density of binding in adult brain was found in the substantia nigra and globus pallidus, whereas the other areas displayed heterogenous binding. In agreement with the membrane binding studies, 1-day-old rat brain had significantly fewer [3H]glibenclamide binding sites than adult brain. Additionally, the pattern of distribution of these sites was qualitatively different from that of the adult. In adult rat spinal cord, moderate binding densities were observed in spinal cord gray and displayed a rostral to caudal gradient. In adult rat heart, moderate binding densities were observed and the sites were distributed homogeneously. In conclusion, significant development of [3H]glibenclamide binding sites was seen in the brain but not the heart during postnatal maturation. Furthermore, a heterogeneous distribution of binding sites was observed in both the brain and spinal cord of adult rats.


Subject(s)
Brain/metabolism , Glyburide/metabolism , Myocardium/metabolism , Spinal Cord/metabolism , Aging/metabolism , Aging/physiology , Animals , Autoradiography/methods , Binding Sites , Brain/anatomy & histology , Brain/growth & development , Brain/ultrastructure , Female , Heart/anatomy & histology , Heart/growth & development , Male , Membranes/metabolism , Myocardium/ultrastructure , Rats , Rats, Inbred Strains , Spinal Cord/growth & development , Spinal Cord/ultrastructure , Tritium
18.
J Cardiovasc Pharmacol ; 17(1): 158-65, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1708050

ABSTRACT

The effects of probucol, a lipophilic antioxidant, on the myocardial dysfunction (stunning) observed during reperfusion after 15-min ischemia in rabbit heart were studied. Rabbits received food with or without 1% probucol for 3 weeks. They were then anesthetized and prepared for recording of myocardial segment shortening, arterial blood pressure (BP), left ventricular pressure (LVP), rate of development of LVP (dP/dt), and a lead II ECG. Regional myocardial ischemia was produced by acute occlusion of the first marginal branch of the left coronary artery. Myocardial segment shortening was depressed after reperfusion in control rabbits. In comparison, myocardial segment shortening was significantly greater in probucol-treated rabbits than in control rabbits during reperfusion, indicating a beneficial effect. No hemodynamic or ECG changes measured could explain this difference. The number of premature ventricular contractions was reduced in the probucol-treated group, although the incidence of ventricular tachycardia (VT) and ventricular fibrillation (VF) were not. Concentrations of probucol in serum and heart of five rabbits were 15.0 +/- 1.2 micrograms/ml and 17.5 +/- 2.5 micrograms/g (mean +/- SEM), respectively. Only probucol concentrations in the serum were positively correlated with the improvement in myocardial segment shortening (r = 0.91, p = 0.03). We conclude that a clinically relevant serum concentration of probucol reduces ischemia-induced myocardial stunning in the rabbit.


Subject(s)
Probucol/pharmacology , Reperfusion Injury/drug therapy , Animals , Electrocardiography/drug effects , Female , Heart/drug effects , Heart/physiology , Hemodynamics/drug effects , Myocardium/metabolism , Probucol/blood , Probucol/metabolism , Probucol/therapeutic use , Rabbits , Reperfusion Injury/physiopathology , Tachycardia/drug therapy , Tachycardia/prevention & control , Ventricular Fibrillation/drug therapy , Ventricular Fibrillation/prevention & control , Ventricular Function/drug effects
19.
Neurochem Int ; 18(4): 497-501, 1991.
Article in English | MEDLINE | ID: mdl-20504732

ABSTRACT

Endothelin-induced contraction in dog femoral artery strips was relaxed by a protein kinase C inhibitor (MDL 27,032), cyclic nucleotide phosphodiesterase (PDE) inhibitors (enoximone, piroximone, rolipram, 3-isobutyl-1-methylxanthine), an adenylcyclase activator (forskolin), a guanylcyclase activator (nitroprusside), but only slightly relaxed by a calcium channel blocker (nifedipine). Endothelin-induced contractions were effectively relaxed by MDL 27,032, but were only slightly antagonized by nifedipine. Endothelin lost its contractile effect in the absence of external calcium whereas phorbol 12, 13-dibutyrate produced a contractile effect in the presence or absence of external calcium. Thus, endothelin-induced contraction requires external calcium ions which may enter vascular smooth muscle cells through nifedipine resistant channels. Endothelin-induced contraction also appears to involve activation of protein kinase C. PDE inhibitors, forskolin and nitroprusside all antagonized the contractile effect of endothelin in femoral arteries. These results indicate that protein kinase C inhibitors and compounds which increase cyclic nucleotide levels can be used to antagonize the vasoconstriction produced by endothelin.

20.
J Pharmacol Exp Ther ; 255(3): 1392-8, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2175806

ABSTRACT

MDL 27,032 [4-propyl-5-(4-pyridinyl)-2(3H)-oxazolone] is a novel vasodilator whose mechanism of action has not been elucidated. We investigated whether smooth muscle relaxation by MDL 27,032, in vitro, may involve an alteration in the activity of protein kinase C, cyclic AMP (cAMP)-dependent protein kinase or myosin light chain kinase by investigating the effects of MDL 27,032 on cyclic nucleotide phosphodiesterases (PDEs) and protein kinase activities. Strips of dog femoral artery or saphenous vein contracted with phorbol 12-myristate 13-acetate (PMA) were relaxed by 100 microM concentrations of MDL 27,032, as well as by other known inhibitors of PDEs [3-isobutyl-1-methylxanthine and papaverine], myosin light chain kinase (W-7) and protein kinase C (H-7 and polymyxin B). In contrast to 3-isobutyl-1-methylxanthine and papaverine, MDL 27,032 was either inactive or weak as an inhibitor of purified PDE types I, II, IVa and IVb. Similarly, it was a weak inhibitor of myosin light chain kinase. However, MDL 27,032 was a significantly more potent inhibitor of protein kinase C and cAMP-dependent protein kinase in cytosolic extracts of dog vein. Kinetic experiments utilizing purified rat brain protein kinase C revealed that inhibition with MDL 27,032 was competitive with Mg(++)-ATP (Ki 24 microM) and noncompetitive with phospholipid, diacylglycerol, PMA, calcium or substrate proteins. Inhibition of the catalytic subunit of cAMP-dependent protein kinase was also competitive with Mg(++)-ATP (Ki 14.3 microM). Similar results were obtained with MDL 27,032 and H-7 on both enzymes.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Muscle Relaxation/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine , 2',3'-Cyclic-Nucleotide Phosphodiesterases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Brain/enzymology , Chickens , Dogs , Female , Femoral Artery/drug effects , Femoral Artery/physiology , Isoenzymes/antagonists & inhibitors , Isoquinolines/pharmacology , Kinetics , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Piperazines/pharmacology , Rats , Saphenous Vein/drug effects , Saphenous Vein/physiology , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...