Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Thromb Res ; 91(3): 143-50, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9733158

ABSTRACT

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.


Subject(s)
Anticoagulants/pharmacology , Heparin Antagonists/pharmacology , Heparin Lyase/pharmacology , Heparin/pharmacology , Protamines/pharmacology , Venous Thrombosis/drug therapy , Animals , Blood Coagulation/drug effects , Drug Interactions , Heparin/therapeutic use , Male , Rabbits
2.
Am J Hypertens ; 9(5): 432-6, 1996 May.
Article in English | MEDLINE | ID: mdl-8735173

ABSTRACT

Brain natriuretic peptide (BNP) is a cardiac-derived peptide hormone with cardiovascular and renal actions that is structurally and functionally related to atrial natriuretic peptide (ANP). Previous studies using rat vascular tissue have demonstrated a direct vasorelaxant effect of BNP. However, species-specific potency issues have precluded an accurate measurement of the effect of human BNP. This report demonstrates the vasorelaxant effects of human BNP on human vascular tissue prepared from internal mammary artery and saphenous vein samples. The vasorelaxant effect of human BNP is compared to the other members of the natriuretic peptide family, human ANP and C-type natriuretic peptide (CNP). With regard to potency and magnitude of effect, human BNP and human ANP were similar in relaxing arterial tissue preconstricted with endothelin-1 (BNP ED50 = 1.9 nmol/L and ANP ED50 = 1.8 nmol/L) or phenylephrine (BNP ED50 = 10 nmol/L and ANP ED50 = 19 nmol/L), while CNP was significantly less effective. All three natriuretic peptides exhibited weak venodilating action. These data demonstrate that human BNP is a potent inhibitor of the vasoconstrictive actions of endothelin-1 and the alpha-adrenergic agonist phenylephrine on isolated human artery tissue preparations.


Subject(s)
Muscle, Smooth, Vascular/drug effects , Nerve Tissue Proteins/pharmacology , Atrial Natriuretic Factor/pharmacology , Endothelins/pharmacology , Humans , In Vitro Techniques , Mammary Arteries/drug effects , Muscle Relaxation/drug effects , Natriuretic Peptide, Brain , Phenylephrine/pharmacology , Saphenous Vein/drug effects , Vasoconstrictor Agents/pharmacology
3.
J Cardiovasc Pharmacol ; 23(5): 792-8, 1994 May.
Article in English | MEDLINE | ID: mdl-7521463

ABSTRACT

We wished to provide comparative information regarding the direct effects of flosequinan, a novel quinolone under development for treating heart failure, on isolated human arterial, venous, and cardiac muscle. A similar assessment was made for four other agents--milrinone, ouabain, captopril and diltiazem--that have been used to treat heart failure patients, as well as for flosequinoxan, which is the primary metabolite of flosequinan. Flosequinan produced a potent and balanced relaxant effect on norepinephrine (NE)-contracted human arterial and venous smooth muscle, with IC25 values of 0.32 and 0.50 microM, respectively. At higher concentrations, flosequinan also produced a positive inotropic effect on human cardiac muscle (EC25 = 32 microM). A similar pattern of responses was observed with flosequinoxan. The pharmacologic profile obtained for the other agents examined differed from that observed with flosequinan and flosequinoxan in the following ways: Milrinone produced both vascular relaxant and positive inotropic effects, but at comparable concentrations; ouabain produced both vasoconstrictor and positive inotropic effects; diltiazem exerted a vascular relaxant effect at low concentrations and a negative inotropic effect at higher concentrations; and captopril had slight arterial relaxant and negative inotropic effects. These results demonstrate that the pharmacologic profile of flosequinan and flosequinoxan is unique as compared with that of other agents that have been used to treat patients with heart failure.


Subject(s)
Muscle, Smooth, Vascular/drug effects , Myocardial Contraction/drug effects , Quinolines/pharmacology , Vasodilator Agents/pharmacology , Captopril/pharmacology , Diltiazem/pharmacology , Heart Failure/drug therapy , Humans , In Vitro Techniques , Milrinone , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/metabolism , Ouabain/pharmacology , Pyridones/pharmacology , Pyridones/therapeutic use , Quinolones/pharmacology
4.
Endocrinology ; 134(2): 899-905, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8299585

ABSTRACT

In this report, we demonstrate a significant inverse correlation between contractility and serum 1,25-dihydroxyvitamin D3 [1,25-(OH)2D3] levels and no correlation between contractility and serum levels of calcium, phosphate, or PTH. We also examined myosin isozyme distribution in vitamin D3-deficient rats, because myosin isozyme distribution can alter contractility. There was a significant increase in the levels of the V1 myosin isozyme in animals raised on a vitamin D3-deficient diet that maintained normal serum calcium and phosphate levels. There was no difference in the relative myosin isozyme distribution in animals raised on a hypocalcemia-yielding vitamin D3-deficient diet vs. animals raised on a control diet. As increased contractility has been observed in both groups of vitamin D3-deficient animals, a shift in myosin isozyme distribution cannot solely explain the increase in contractility previously observed in the vitamin D3-deficient rat heart. To determine whether 1,25-(OH)2D3 directly regulates myosin isozyme levels, we analyzed myosin isozyme distribution in primary cultures of ventricular myocytes. We found that 1,25-(OH)2D3 reduces total myosin levels, but does not alter myosin isozyme distribution. Thus, we show that the influence of vitamin D3 status on myosin isozyme expression in the intact rat involves a complex regulatory system of direct and indirect effects.


Subject(s)
Calcitriol/pharmacology , Gene Expression Regulation/physiology , Heart/physiology , Isoenzymes/biosynthesis , Myocardial Contraction , Myocardium/metabolism , Myosins/biosynthesis , Parathyroid Hormone/blood , Vitamin D Deficiency/metabolism , Animals , Calcium/blood , Calcium/metabolism , Cells, Cultured , Gene Expression Regulation/drug effects , Heart/drug effects , Heart/physiopathology , Isoenzymes/isolation & purification , Male , Myosins/isolation & purification , Rats , Rats, Sprague-Dawley , Vitamin D Deficiency/blood , Vitamin D Deficiency/physiopathology
5.
Eur J Pharmacol ; 236(3): 363-6, 1993 Jun 04.
Article in English | MEDLINE | ID: mdl-8395383

ABSTRACT

The goal of this study was to assess the relationship between the positive inotropic response to high concentrations of the vasodilators flosequinan and BTS 53 554 (the sulfone metabolite of flosequinan) and the effect of both compounds on different forms of cyclic nucleotide phosphodiesterase. In addition, the relationship between inotropic activity and phosphodiesterase inhibition for the cardiotonic milrinone was also evaluated. All three agents exerted a positive inotropic effect on human cardiac muscle fibers. The concentration of milrinone required to increase cardiac contractility was comparable to the concentration required to inhibit the milrinone-sensitive subclass of cyclic AMP-specific phosphodiesterase (type III phosphodiesterase). However, no such relationship was observed for flosequinan and BTS 53 554. These results suggest that the cardiac response to high concentrations of flosequinan and BTS 53 554 is not mediated by inhibition of type III phosphodiesterase.


Subject(s)
Myocardial Contraction/drug effects , Phosphodiesterase Inhibitors/pharmacology , Pyridones/pharmacology , Quinolines/pharmacology , Quinolones/pharmacology , Humans , In Vitro Techniques , Milrinone , Phosphoric Diester Hydrolases/metabolism , Vasodilation/drug effects
7.
J Med Chem ; 34(7): 2248-60, 1991 Jul.
Article in English | MEDLINE | ID: mdl-2066997

ABSTRACT

The synthesis and pharmacological evaluation of a series of 2-[(arylsulfonyl)methyl]-4-aryl-5-cyano-1,4-dihydropyridine-3-carboxylic acid esters and analogues are described. These compounds possess a unique profile namely, calcium channel blocking and positive inotropic activities in vitro. Compound 54 was selected as the best compound in the series and was studied in detail. The synthesis and biological profiles of enantiomers of 54 are also reported. The data indicate that although the calcium channel blocking property of 54 is stereospecific the positive inotropic activity is not. Examples of 3- and 6-cyano and other closely related 1,4-dihydropyridine derivatives are described and evaluated for comparison and were found to be devoid of dual activities mentioned above.


Subject(s)
Calcium Channel Blockers/chemical synthesis , Carboxylic Acids/chemical synthesis , Dihydropyridines/chemical synthesis , Nicotinic Acids/chemical synthesis , Sulfones/chemical synthesis , Animals , Calcium Channel Blockers/pharmacology , Carboxylic Acids/pharmacology , Chemical Phenomena , Chemistry , Dihydropyridines/pharmacology , Guinea Pigs , Muscle, Smooth, Vascular/drug effects , Myocardial Contraction/drug effects , Nicotinic Acids/pharmacology , Nitriles/chemical synthesis , Nitriles/pharmacology , Rabbits , Rats , Structure-Activity Relationship , Sulfones/pharmacology
8.
Circ Res ; 68(5): 1302-12, 1991 May.
Article in English | MEDLINE | ID: mdl-1850330

ABSTRACT

The effect of chronic therapy with quinapril on the temporal progression of left ventricular failure and survival was assessed in the CHF 146 cardiomyopathic (CM) hamster, which is an idiopathic model of congestive heart failure. Age-matched Golden Syrian (GS) hamsters served as normal controls. Quinapril was administered in the drinking water at average daily doses of 10.2, 112.4, and 222.4 mg/kg/day. In untreated CM hamsters, in vitro left ventricular performance progressively deteriorated with increasing age beginning at roughly 180 days. This decline in left ventricular performance was accompanied by a decrease in coronary flow and an increase in left ventricular volume. Administration of quinapril from 180 to 300 days of age prevented the decline of in vitro left ventricular contractile performance and coronary flow and also reduced the age-dependent increases in left ventricular volume. The cardioprotective effects of quinapril were observed at doses of 112.4 and 222.4 mg/kg/day but not at 10.2 mg/kg/day. Lung angiotensin converting enzyme activity was significantly inhibited by quinapril in GS and CM hamsters at 240 and 300 days of age at all dose levels. In contrast, significant inhibition of ventricular angiotensin converting enzyme activity was observed consistently at doses of 112.4 and 222.4 mg/kg/day quinapril but not at 10.2 mg/kg/day. In the survival protocol, CM and GS hamsters were treated with vehicle or quinapril (100 mg/kg/day) from 180 to 522 days of age. During the initial 210 days of treatment (from 180 to 390 days of age) 78.3% of the vehicle-treated CM hamsters died compared with 27.7% of quinapril-treated CM hamsters. Quinapril increased the median survival time of CM hamsters by 32.9% (112 days). It is concluded that chronic quinapril therapy exerts a significant cardioprotective effect and also increases survival.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Heart Failure/drug therapy , Isoquinolines/therapeutic use , Tetrahydroisoquinolines , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Atrial Natriuretic Factor/analysis , Coronary Circulation/drug effects , Cricetinae , Heart/drug effects , Heart Failure/metabolism , Heart Failure/physiopathology , Hemodynamics , In Vitro Techniques , Isoquinolines/pharmacology , Mesocricetus , Myocardial Contraction/drug effects , Peptidyl-Dipeptidase A/analysis , Perfusion , Quinapril
9.
J Pharmacol Exp Ther ; 256(2): 568-74, 1991 Feb.
Article in English | MEDLINE | ID: mdl-1847201

ABSTRACT

Electrical stimulation of the isolated rat mesenteric vascular bed resulted in a frequency-dependent pressor response, which could be potentiated by increasing concentrations of renin substrate (synthetic tetradecapeptide). This potentiating effect appeared to be mediated by tissue conversion of renin substrate to angiotensin II because the response 1) could be mimicked by angiotensin II, 2) was accompanied by an increase in angiotensin II production and 3) was blocked by the angiotensin converting enzyme (ACE) inhibitor quinaprilat and the angiotensin II receptor antagonist saralasin ([Sar1,Ile5,Ala8]angiotensin II). To assess the role of this tissue renin-angiotensin system in contributing to blood pressure regulation, spontaneously hypertensive rats were administered the prodrug ACE inhibitor quinapril at a dose of 10 mg/kg/day for 7 days. Such administration resulted in a reduction in systolic blood pressure of 48 +/- 3 mm Hg, a greater than 95% inhibition of serum ACE activity, and a significant attenuation of the potentiating effect of renin substrate on electrically evoked contractions of isolated mesenteric beds. Significant reductions in blood pressure and the potentiating effect of renin substrate on the isolated mesenteric vasculature were still observed 24 and 48 hr after the last dose of quinapril. In contrast, serum ACE activity returned to normal levels within 48 hr after the last dose of quinapril. These results suggest that the changes in tissue renin-angiotensin system, and not the circulating system, are closely related to the blood pressure lowering effect of the ACE inhibitor, quinapril.


Subject(s)
Blood Pressure , Mesentery/physiology , Renin-Angiotensin System/physiology , Tetrahydroisoquinolines , Angiotensin II/pharmacology , Angiotensinogen/pharmacology , Animals , Electric Stimulation , Isoquinolines/pharmacology , Peptidyl-Dipeptidase A/analysis , Quinapril , Rats , Rats, Inbred Strains , Vasoconstriction/drug effects
10.
Clin Exp Hypertens A ; 13(8): 1395-414, 1991.
Article in English | MEDLINE | ID: mdl-1760894

ABSTRACT

Hypertension was produced in cynomolgus monkeys by reducing blood flow to the left kidney by 60% via renal artery stenosis (2-kidney, 1-clip). Significant increases in mean arterial blood pressure (MABP) were observed within two to three weeks. Maximum increase (from 95 +/- 6 mmHg to 130 +/- 7 mmHg) occurred at about four to six weeks following renal artery stenosis and was sustained for more than 24 weeks. Plasma renin activity (PRA) was elevated concomitantly with the increase in MABP. PRA was raised to 42 +/- 3 ng angiotensin I/ml/hr six weeks after renal artery stenosis from a control PRA of 3 +/- 0.7 ng angiotensin I/ml/hr. At six months post renal artery stenosis, PRA was 33.4 +/- 4.2 ng angiotensin I/ml/hr. The angiotensin II (AII) receptor antagonist saralasin, the angiotensin I converting enzyme inhibitor captopril, and the renin inhibitor CGP 38,560 produced sustained reductions in MABP. The antihypertensive response to the renin inhibitor CGP 38,560 was associated with a reduction in PRA of greater than 99%, and a greater than 90% reduction in immunoreactive AII. These studies demonstrate that high-renin hypertension can be induced in the cynomolgus monkey. This pathological model provides a useful method for investigating the antihypertensive effects of agents which antagonize the renin-angiotensin system in a nonhuman primate.


Subject(s)
Hypertension, Renovascular/etiology , Renin/blood , Angiotensin II/blood , Animals , Blood Pressure/drug effects , Captopril/pharmacology , Disease Models, Animal , Hypertension, Renovascular/blood , Hypertension, Renovascular/physiopathology , Macaca fascicularis , Male , Oligopeptides/pharmacology , Renal Artery Obstruction/complications , Renin/antagonists & inhibitors , Saralasin/pharmacology
11.
Pharmacology ; 43(4): 187-98, 1991.
Article in English | MEDLINE | ID: mdl-1685246

ABSTRACT

alpha 2-Adrenoceptors were characterized on neural and vascular membranes from 2-kidney-1-clip renal hypertensive (RHT) and normotensive (NT) rats. Rats were sacrificed 6 weeks after induction of renal ischemia, and the specific binding of 3H-clonidine to smooth muscle membranes form tail arteries and neural membranes from various brain regions was examined. Additionally, isometric contractions of helically cut tail artery strips produced by various alpha-adrenoceptor agonists were measured. Scatchard analysis indicated an increased number of high-affinity binding sites on the smooth muscle membranes from RHT rats (Bmax = 43.5 +/- 1.4 fmol/mg protein) compared to that from the NT rats (25.4 +/- 3.8 fmol/mg protein). An increased contractile sensitivity to clonidine was also observed in tail artery strips from RHT rats (EC50 for RHT = 3.04 x 10(-8) M; NT = 1.52 x 10(-7) M). In neural tissue, the number of alpha 2-adrenoceptor-binding sites was significantly increased in the locus coeruleus from RHT rats, but not in the amygdala, hypothalamus, parietal cortex, hippocampus or lower brain stem. These results demonstrate that renal ischemia produces changes in both peripheral and neural alpha 2-adrenoceptor density. The increase in smooth muscle alpha 2-adrenoceptors might also provide a partial explanation for the supersensitivity to adrenergic agonists in this model of hypertension.


Subject(s)
Brain/ultrastructure , Hypertension, Renal/physiopathology , Ischemia/physiopathology , Kidney/blood supply , Muscle, Smooth, Vascular/ultrastructure , Receptors, Adrenergic, alpha/physiology , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Arteries/drug effects , Arteries/physiology , Brain/metabolism , Hypertension, Renal/etiology , Hypertension, Renal/metabolism , In Vitro Techniques , Ischemia/complications , Ischemia/metabolism , Kinetics , Male , Membranes/metabolism , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Inbred Strains , Receptors, Adrenergic, alpha/metabolism , Tail/blood supply
12.
Mol Pharmacol ; 38(3): 370-7, 1990 Sep.
Article in English | MEDLINE | ID: mdl-2402226

ABSTRACT

Specific binding sites for angiotensin II were identified in the rabbit adrenal gland and uterus and in the rat liver and were divided into two subclasses based on inhibition by nonpeptide ligands. Peptide ligands affected binding equally in all three tissues. However, the nonpeptide antagonists Dup 753 and Exp 6803 blocked angiotensin II binding to adrenal and liver homogenates at nanomolar concentrations but exerted only a minimal effect on binding to uterine homogenates. The nonpeptide PD 123319 potently blocked angiotensin II binding to uterine homogenates but had no effect on adrenal or liver homogenates at concentrations up to 10 microM. Further analysis of angiotensin II binding in uterus showed that both sites are present, with the PD 123319-sensitive site predominating. Additionally, the nonhydrolyzable GTP analogue 5'-guanylyl-imidodiphosphate was able to modulate binding to liver and to the Dup 753-sensitive site in uterus but not that to the PD 123319-sensitive site. Saralasin and the nonpeptide antagonists Dup 753 and Exp 6803 blocked angiotensin II-stimulated accumulation of inositol phosphates in cultured Clone 9 cells and also relaxed aortic rings previously contracted with angiotensin II. In contrast, PD 123319 had no effect on either angiotensin II-stimulated inositol phosphate accumulation or vasoconstriction. Saralasin and Exp 6803, but not PD 123319, lowered blood pressure in renal hypertensive rats following intravenous administration. These results suggest the existence of two subclasses of angiotensin II binding sites, which differ in their tissue distribution and affinity for the nonpeptide ligands Dup 763, Exp 6803, and PD 123319. Although no functional role for the PD 123319-sensitive subclass has yet been identified, the Dup 753/Exp 6803-sensitive subclass plays an important role in mediating inositol phosphate metabolism, vascular contractile activity, and blood pressure regulation.


Subject(s)
Angiotensin II/metabolism , Receptors, Angiotensin/analysis , Angiotensin II/antagonists & inhibitors , Angiotensin II/pharmacology , Angiotensin Receptor Antagonists , Animals , Binding Sites , Blood Pressure/drug effects , Female , Guanylyl Imidodiphosphate/pharmacology , Imidazoles/pharmacology , In Vitro Techniques , Inositol Phosphates/metabolism , Losartan , Male , Muscle Contraction/drug effects , Rabbits , Rats , Rats, Inbred Strains , Receptors, Angiotensin/physiology , Tetrazoles/pharmacology
13.
Biochem Pharmacol ; 40(4): 699-707, 1990 Aug 15.
Article in English | MEDLINE | ID: mdl-1696820

ABSTRACT

Multiple molecular forms of cyclic nucleotide phosphodiesterase have been characterized in various tissues and cells according to their substrate specificity, intracellular location, and calmodulin dependence. The purpose of this study was to evaluate the possible involvement of different molecular forms of phosphodiesterase in regulating the respiratory burst and lysosomal enzyme release responses of human neutrophils. Treatment with the selective cyclic AMP-specific, cyclic GMP-insensitive phosphodiesterase inhibitors Ro 20-1724 or rolipram, or the nonselective phosphodiesterase inhibitor 3-isobutyl-1-methylxanthine (IBMX), resulted in inhibition of respiratory burst stimulated by the chemoattractants formyl-L-methionyl-L-leucyl-L-phenylalanine (FMLP) (IC50 values: 0.71-17 microM) and complement fragment C5a (IC50 values: 61-93 microM), but did not inhibit phagocytosis-stimulated respiratory burst (less than 10% inhibition at 100 microM). Selective inhibitors of calmodulin-dependent phosphodiesterase (ICI 74,917), calmodulin-insensitive, cyclic GMP-specific phosphodiesterase (M & B 22,948), cyclic GMP-stimulated phosphodiesterase (AR-L 57), or cyclic AMP-specific, cyclic GMP-inhibited phosphodiesterase (amrinone and cilostamide) exhibited little or no inhibitory effect on FMLP- or phagocytosis-stimulated respiratory burst (0-42% inhibition at 100 microM). Regulation of neutrophil activation by phosphodiesterase was also response specific, as Ro 20-1724, rolipram and IBMX were less potent inhibitors of FMLP-induced lysosomal enzyme release (0-14% inhibition at 100 microM). Analysis of human neutrophil preparations confirmed the existence of a cyclic AMP-specific, cyclic GMP-insensitive phosphodiesterase, which was associated with the particulate fraction of the cell. These results demonstrate a role for the cyclic AMP-specific, cyclic GMP-insensitive phosphodiesterase in the regulation of human neutrophil functions, which appears to be both stimulus specific and response specific.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/physiology , Neutrophils/drug effects , Phosphodiesterase Inhibitors/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , 4-(3-Butoxy-4-methoxybenzyl)-2-imidazolidinone/pharmacology , Cyclic AMP/analysis , Cyclic Nucleotide Phosphodiesterases, Type 1 , Humans , Lysosomes/enzymology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/metabolism , Oxygen Consumption/drug effects , Pyrrolidinones/pharmacology , Rolipram
14.
Hypertension ; 15(5): 528-40, 1990 May.
Article in English | MEDLINE | ID: mdl-2158939

ABSTRACT

Two subclasses of cyclic guanosine monophosphate (GMP)-specific phosphodiesterases were identified in vascular tissue from several beds. The activity of one subclass (phosphodiesterase IB) was stimulated severalfold by calmodulin and selectively inhibited by the phosphodiesterase inhibitor TCV-3B. The activity of the other subclass (phosphodiesterase IC) was not stimulated by calmodulin and was selectively inhibited by the phosphodiesterase inhibitor M&B 22,948. To assess the involvement of both subclasses in regulating cyclic GMP-dependent responses, the ability of TCV-3B and M&B 22,948 to potentiate the in vitro and in vivo responses to the endogenous guanylate cyclase stimulator atrial natriuretic factor (ANF) was evaluated. Both TCV-3B and M&B 22,948 relaxed isolated rabbit aortic and pulmonary artery rings and also potentiated the relaxant effect of ANF. In addition, both inhibitors produced small increases in urine flow and sodium excretion in anesthetized rats and potentiated the diuretic and natriuretic responses to exogenous ANF. M&B 22,948 (30 micrograms/kg/min) produced a threefold increase in the natriuretic response to simultaneously administered ANF, and TCV-3B (10 micrograms/kg/min) produced a twofold increase in the response to ANF. The results of the present experiments suggest that both the calmodulin-sensitive and calmodulin-insensitive subclasses of cyclic GMP-specific phosphodiesterase play a role in regulating the in vitro and in vivo response to ANF.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/classification , Atrial Natriuretic Factor/pharmacology , Phosphodiesterase Inhibitors/pharmacology , 3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-GMP Phosphodiesterases/physiology , Animals , Blood Vessels/enzymology , Dogs , Drug Synergism , Kinetics , Male , Phosphodiesterase Inhibitors/classification , Purinones/pharmacology , Rabbits , Rats , Vinca Alkaloids/pharmacology
15.
Am J Physiol ; 258(1 Pt 1): E134-42, 1990 Jan.
Article in English | MEDLINE | ID: mdl-2154115

ABSTRACT

We have previously shown that depletion of vitamin D3 in rats results in a large increase in the contractile function of isolated hearts (R. E. Weishaar, J. Clin. Invest. 79: 1706-1712, 1987). To characterize the mechanism responsible for this increase, the effect of vitamin D3 depletion on key physical and morphological properties of cardiac muscle was examined. Depletion of vitamin D3 increased the heart weight/body weight ratio. This increase could neither be blocked by limiting hypocalcemia nor reversed by restoring increasing serum calcium levels. The cardiomegaly observed 9 wk after vitamin D3 depletion was not accompanied by an increase in myocardial water content or leakage of myocardial creatine phosphokinase and was not caused by myocardial cell hypertrophy. Histological examination of ventricular muscle from vitamin D3-deficient rats revealed a significant decrease in myofibrillar area and a significant increase in extracellular space. The increase in extracellular space was accompanied by a significant increase in myocardial collagen. Prevention of hypocalcemia in the vitamin D3-deficient rats did not prevent the increase in myocardial collagen. Such alterations in the physical and morphological properties of myocardial tissue might represent the basis for the change in myocardial contractile function that accompanies lengthy periods of vitamin D3 deficiency.


Subject(s)
Cholecalciferol/pharmacology , Heart/anatomy & histology , Myocardial Contraction , Vitamin D Deficiency/physiopathology , Animals , Body Weight/drug effects , Calcium/blood , Catecholamines/blood , Creatine Kinase/metabolism , Extracellular Space , Heart/drug effects , Hormones/blood , Magnesium/blood , Male , Microscopy, Electron , Myocardial Contraction/drug effects , Myocardium/enzymology , Myocardium/pathology , Myocardium/ultrastructure , Organ Size/drug effects , Rats , Rats, Inbred Strains
16.
Can J Physiol Pharmacol ; 67(11): 1460-3, 1989 Nov.
Article in English | MEDLINE | ID: mdl-2560676

ABSTRACT

The positive inotropic activity of the novel cardiotonic DPI 201-106 was investigated in rat and guinea pig isolated hearts. For comparative purposes, the adenylate cyclase stimulant forskolin and the sodium channel agonist veratridine were also evaluated in both species. DPI 201-106 and veratridine produced greater inotropic effects in rat hearts than in guinea pig hearts, whereas forskolin produced comparable effects. In both species the inotropic response to DPI 201-106 and veratridine, but not forskolin, was reversed by the sodium channel antagonist tetrodotoxin. These results confirm that the positive inotropic effect of DPI 201-106 is due to stimulation of the sodium channel and demonstrate for the first time that species differences exist in the inotropic response to this novel cardiotonic drug.


Subject(s)
Cardiotonic Agents/pharmacology , Myocardial Contraction/drug effects , Piperazines/pharmacology , Animals , Colforsin/pharmacology , Guinea Pigs , In Vitro Techniques , Male , Rats , Sodium Channels/drug effects , Species Specificity , Tetrodotoxin/pharmacology , Veratridine/pharmacology
17.
Eur J Pharmacol ; 167(3): 323-31, 1989 Aug 29.
Article in English | MEDLINE | ID: mdl-2572433

ABSTRACT

Contractions to transmural electrical stimulation and exogenous norepinephrine were recorded in isolated longitudinal segments of rat vas deferens. Electrical stimulation for 30 s produced a biphasic contraction in the vas deferens consisting of a rapid, transient response (Phase I), followed by a slowly developing, sustained contraction (Phase II). N6-Cyclohexyladenosine (CHA), a selective adenosine1 (A1)-receptor agonist, attenuated in a concentration-dependent manner the Phase I contractile response, while having little effect on the Phase II response. In contrast, 2-(phenylamino)adenosine (CV-1808), a selective adenosine2 (A2)-receptor agonist had little effect on either contractile phase. CHA did not inhibit the contraction to exogenous norepinephrine, suggesting that A1-receptors were located at a presynaptic site. The relatively selective alpha 2-receptor agonist clonidine produced the same pattern of contractile inhibition as CHA. The inhibitory effect of CHA on the Phase I contractile response in the vas deferens could be antagonized by the selective A1-receptor antagonist 8-cyclopentyltheophylline, while the selective alpha 2-receptor antagonist idazoxan preferentially antagonized the inhibitory effect of clonidine on the Phase I response. Both the Phase I and Phase II contractile responses were reduced by the selective alpha 1-adrenoceptor antagonist prazosin and the ATP analog alpha, beta-methylene adenosine triphosphate (alpha, beta-methylene ATP), suggesting that norepinephrine and ATP are coreleased as neurotransmitters for both responses. The results of the present study demonstrate that in the rat vas deferens the presynaptic inhibitory effects of adenosine is mediated by the A1-receptor subtype, and that both A1- and alpha 2-receptor agonists exert a selective inhibitory effect on the Phase I contractile response to electrical stimulation.


Subject(s)
Adenosine/physiology , Muscle, Smooth/physiology , Receptors, Adrenergic, alpha/physiology , Adenosine Triphosphate/pharmacology , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Electric Stimulation , In Vitro Techniques , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Neurotransmitter Agents/physiology , Norepinephrine/pharmacology , Rats , Rats, Inbred Strains , Receptors, Adrenergic, alpha/drug effects , Vas Deferens/drug effects
19.
J Pharmacol Exp Ther ; 250(1): 22-30, 1989 Jul.
Article in English | MEDLINE | ID: mdl-2473188

ABSTRACT

The cardiac and vascular activities of ethyl 5-cyano-1,4-dihydro-6-methyl-2-[(4-pyridinyl-sulfonyl)methyl]-4-[2- (trifluoromethyl)phenyl]-3-pyridine carboxylase (PD 122860), a novel dihydropyridine, were investigated in vitro using rat heart and rabbit aorta, and compared with reference inotropic and vasodilator agents. In the rat heart, PD 122860 increased left ventricular contractility, decreased coronary resistance and altered the shape of the electrocardiogram T-wave. All three effects were observed at comparable concentrations of PD 122860. The inotropic response to PD 122860 was reversed by the Na+ channel blocker tetrodotoxin and blunted by the Na+-Ca++ exchange inhibitor dichlorobenzamil. The effects of tetrodotoxin and dichlorobenzamil on the inotropic response to the reference Na+ channel stimulant veratridine were comparable to PD 122860, whereas tetrodotoxin and dichlorobenzamil had no inhibitory effect on the inotropic responses to the adenylate cyclase stimulator forskolin or the Ca++ channel stimulant BAY K 8644. PD 122860 selectively relaxed potassium-contracted aortic rings and inhibited [3H]nitrendipine binding to rat brain membranes, suggesting that the vasodilator activity of PD 122860 is due to Ca++ channel blockade. In contrast to BAY K 8644, PD 122860 did not contract partially depolarized aortic rings, suggesting an absence of Ca++ channel stimulant activity. PD 122860 is a racemic mixture and both the vasorelaxant and [3H]nitrendipine binding inhibitory activities selectively reside in the (+)-enantiomer [(+)-PD 122860]. In contrast, the inotropic response resides with both enantiomers of PD 122860. It is therefore concluded that PD 122860 represents a unique dihydropyridine derivative which possesses both Na+ channel stimulating and Ca++ channel blocking activities.


Subject(s)
Calcium Channels/physiology , Dihydropyridines/pharmacology , Heart/physiology , Sodium Channels/physiology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Calcium Channels/drug effects , Colforsin/pharmacology , Coronary Circulation/drug effects , Electrocardiography , Heart/drug effects , Heart Rate/drug effects , In Vitro Techniques , Male , Myocardial Contraction/drug effects , Ouabain/pharmacology , Rats , Rats, Inbred Strains , Reference Values , Sodium Channels/drug effects , Veratridine/pharmacology
20.
Cardiovasc Drugs Ther ; 3(1): 29-42, 1989 Mar.
Article in English | MEDLINE | ID: mdl-2487522

ABSTRACT

Intense efforts during the last decade to identify a useful positive inotropic agent to replace digitalis for the treatment of congestive heart failure have led to the discovery of several dozen potential substitutes, of which a number are currently undergoing clinical trials. In addition to producing a variety of new therapeutic entities, research in this area has also yielded valuable new information regarding the fundamental events that regulate calcium homeostasis and contractile function in the cardiac cell. For example, several of these new inotropic agents, including the calcium-channel stimulator BAY-k 8644, the sodium-channel stimulator DPI-201-186, and the sodium-calcium exchange inhibitor dichlorobenzamil, have provided considerable insight into the role of sodium and calcium in regulating contractility and the molecular events that mediate potential-dependent ion channels. Likewise, the discovery and development of agents like imazodan, amrinone, enoximone, and other selective type III phosphodiesterase inhibitors have provided new information regarding multiple molecular forms of cyclic nucleotide phosphodiesterase, compartmentation of cyclic AMP, and the importance of soluble vs. membrane-bound phosphodiesterases.


Subject(s)
Cardiotonic Agents/pharmacology , Pyridazines/pharmacology , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...