Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters










Publication year range
1.
Hypertension ; 37(3): 862-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11244009

ABSTRACT

In the present study, we sought to evaluate whether the antiadrenergic action of adenosine in the heart is altered in pressure overload hypertrophy produced in rats by suprarenal aortic banding. Epicardial and coronary effluent adenosine and inosine concentrations and release were significantly elevated in compensated pressure overload hypertrophy but not in hearts with left ventricular failure. In pressure overload hearts, the contractile response to beta-adrenergic stimulation was less inhibited by incremental concentrations of either adenosine or the selective A(1) receptor agonist chloro-N:(6)-cyclopentyl adenosine than in controls. Furthermore, the extent of desensitization to the antiadrenergic actions of adenosine in pressure overload hypertrophy appeared to be proportional to the extent of chamber dilation and dysfunction. A 60-minute infusion of adenosine produced a sustained antiadrenergic effect that lasted up to 45 minutes after the infusion was terminated in both controls and hearts with compensated hypertrophy. This effect was not observed in the decompensated left ventricular failure group. Subsequent infusion with adenosine of the A(2A) receptor antagonist 8-(3-chlorostyryl)-caffeine to counteract the proadrenergic effect of A(2A) receptor stimulation did not alter the decreased sensitivity to the antiadrenergic actions of adenosine in hypertrophied hearts. Finally, isolated myocytes from hypertrophied hearts demonstrated a decreased ability to suppress isoproterenol-elicited increases in [Ca(2+)](i) transients in the presence of adenosine and the A(2A) receptor antagonist compared with myocytes from control hearts. Myocardial adenosine concentrations increase during the compensated phase of pressure overload hypertrophy but then decrease when there is evidence of decompensation. The antiadrenergic actions of adenosine transduced via the myocardial A(1) receptor are diminished in pressure overload hypertrophied hearts. These factors may render these hearts more vulnerable to the detrimental effects of chronically increased sympathetic activity.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/pharmacology , Adrenergic Antagonists/pharmacology , Cardiomegaly/physiopathology , Adenosine/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Blood Pressure , Body Weight , Caffeine/analogs & derivatives , Caffeine/pharmacology , Calcium/metabolism , Cardiomegaly/etiology , Cells, Cultured , Coronary Circulation/drug effects , Disease Models, Animal , Echocardiography , In Vitro Techniques , Inosine/metabolism , Isoproterenol , Male , Myocardial Contraction/drug effects , Myocardium/metabolism , Organ Size , Perfusion , Purinergic P1 Receptor Agonists , Purinergic P1 Receptor Antagonists , Rats , Rats, Sprague-Dawley
2.
Am J Physiol Heart Circ Physiol ; 279(4): H1472-81, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11009431

ABSTRACT

Adenosine A(2a)-receptor activation enhances shortening of isolated cardiomyocytes. In the present study the effect of A(2a)-receptor activation on the contractile performance of isolated rat hearts was investigated by recording left ventricular pressure (LVP) and the maximal rate of LVP development (+dP/dt(max)). With constant-pressure perfusion, adenosine caused concentration-dependent increases in LVP and +dP/dt(max), with detectable increases of 4.1 and 4.8% at 10(-6) M and maximal increases of 12.0 and 11.1% at 10(-4) M, respectively. The contractile responses were prevented by the A(2a)-receptor antagonists chlorostyryl-caffeine and aminofuryltriazolotriazinyl-aminoethylphenol (ZM-241385) but were not affected by the beta(1)-adrenergic antagonist atenolol. The adenosine A(1)-receptor antagonist dipropylcyclopentylxanthine and pertussis toxin potentiated the positive inotropic effects of adenosine. The A(2a)-receptor agonists ethylcarboxamidoadenosine and dimethoxyphenyl-methylphenylethyl-adenosine also enhanced contractility. With constant-flow perfusion, 10(-5) M adenosine increased LVP and +dP/dt(max) by 5.5 and 6.0%, respectively. In the presence of the coronary vasodilator hydralazine, adenosine increased LVP and +dP/dt(max) by 7.5 and 7.4%, respectively. Dipropylcyclopentylxanthine potentiated the adenosine contractile responses with constant-flow perfusion in the absence and presence of hydralazine. These increases in contractile performance were also antagonized by chlorostyryl-caffeine and ZM-241385. The results indicate that adenosine increases contractile performance via activation of A(2a) receptors in the intact heart independent of beta(1)-adrenergic receptor activation or changes in coronary flow.


Subject(s)
Adenosine/analogs & derivatives , Myocardial Contraction/physiology , Receptors, Purinergic P1/physiology , Adenosine/pharmacology , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Atenolol/pharmacology , Drug Synergism , Heart/drug effects , In Vitro Techniques , Male , Myocardial Contraction/drug effects , Perfusion , Pertussis Toxin , Pressure , Purinergic P1 Receptor Agonists , Purinergic P1 Receptor Antagonists , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2A , Vasodilation , Vasodilator Agents/pharmacology , Virulence Factors, Bordetella/pharmacology , Xanthines/pharmacology
3.
J Vasc Surg ; 32(4): 804-13, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11013045

ABSTRACT

PURPOSE: Restenosis after angioplasty or bypass grafting to restore circulation to ischemic organs is still an unsolved problem. Thrombin generated in high concentrations at the sites of vascular injury plays a central role in thrombosis and hemostasis. alpha-Thrombin has also been implicated as a mitogen for smooth muscle cell (SMC) proliferation that contributes to arterial restenosis. Thrombomodulin has a high affinity of binding with thrombin and converts thrombin from a procoagulant to an anticoagulant. This study was designed to examine whether thrombomodulin could also moderate the thrombin-mediated SMC proliferative response. METHODS: Porcine carotid artery SMCs (passages 4-7) were plated onto 96-well plates and incubated for 3 days. After growth arrest in a defined serum-free medium for 2 to 3 days, SMCs were subjected to the reagents as follows: (1) human alpha-thrombin, (2) recombinant human soluble thrombomodulin containing a chondroitin sulfate moiety, (3) thrombin receptor agonist peptide (SFLLRNPNDKYEPF), and (4) alpha-thrombin or thrombin receptor agonist peptide combined with recombinant thrombomodulin (rTM). The viability and proliferation status of SMCs were quantified with MTT (thiazolyl blue) mitochondrial function and bromodeoxyuridine (BrdU)-DNA incorporation assays. RESULTS: Human alpha-thrombin increased SMC proliferation in a dose dependent manner by more than 25% and 30% with thrombin 1 U/mL to 3 U/mL compared with control groups on day 7 (P <.006). rTM concentrations from 0.5 microg/mL to 3 microg/mL have no significant effect on SMC growth. The stimulation of SMC proliferation induced by alpha-thrombin at 0.5 U/mL, 1 U/mL, and 2 U/mL was significantly inhibited with rTM at 2 microg/mL and 3 microg/mL on days 3, 7, and 10 as evaluated with MTT assay (P <.01 to <.05) and BrdU-DNA incorporation assay on day 3 (P <.008). Thrombin receptor agonist peptide increased SMC BrdU-DNA incorporation at 48 hours (P <.007), and its effect was not altered by rTM. CONCLUSION: rTM containing all of the extracellular domains of thrombomodulin inhibits the effect of thrombin on SMC proliferation in vitro. Because thrombin is a mitogenic mediator of SMC in vascular injury, inhibition of its function in vivo could help to prevent SMC hyperplasia. The success of further studies in vivo may lead to use of rTM for decreasing or preventing arterial restenosis.


Subject(s)
Muscle, Smooth, Vascular/cytology , Thrombin/physiology , Thrombomodulin/physiology , Animals , Animals, Newborn , Biological Assay , Cell Division/drug effects , Cells, Cultured , Culture Media, Serum-Free , DNA/biosynthesis , Humans , In Vitro Techniques , Muscle, Smooth, Vascular/drug effects , Recombinant Proteins/pharmacology , Swine
4.
J Mol Cell Cardiol ; 32(9): 1695-701, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10966832

ABSTRACT

The aim of this study was to establish whether epicardial transudates could be used to uncover small, but physiologically important changes in interstitial NE concentrations under normal and pathological conditions. Norepinephrine (NE) concentrations measured in epicardial transudate fluid were compared to NE levels in the coronary effluent in normal and pressure overload hypertrophied (POH) rat hearts. Hearts were isolated together with the stellate ganglion and perfused in the inverted position. Epicardial surface transudates, representative fluid of the interstitial myocardial compartment, and coronary effluents were collected for determination of NE levels in the presence and absence of stellate ganglion stimulation. The same protocol was repeated in the presence and absence of nisoxetine, a NE uptake blocker. NE concentrations in epicardial transudates were 16- and 19-fold higher than in the coronary effluent in both sham and POH groups, respectively. NE concentrations in the transudates but not in the coronary effluents were significantly higher (1.6-fold) in hearts with POH when compared to normal hearts. Likewise, nisoxetine (10(-5)m) increased (1.3-fold) NE concentrations in the transudates but not in the effluents of sham animals. As expected, stellate ganglion stimulation increased NE concentrations in both transudates and effluents in sham and POH hearts. In conclusion, determination of NE concentrations in epicardial transudates represents a simple, rapid and sensitive method to detect increases in adrenergic activity in normal and abnormal hearts.


Subject(s)
Heart/physiology , Norepinephrine/physiology , Receptors, Adrenergic/physiology , Animals , Blood Pressure , Cardiomegaly/physiopathology , Male , Myocardial Reperfusion , Pericardium/metabolism , Rats , Rats, Sprague-Dawley
5.
J Mol Cell Cardiol ; 32(7): 1371-5, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10860777

ABSTRACT

Multiple brief periods of ischemia in the mammalian heart elicits protection against morphologic and functional damage caused by longer-duration ischemia. Preconditioning-induced protection against post-ischemic contractile dysfunction has been reported to be depressed with aging of the adult heart. This study was undertaken to determine whether aging of the adult myocardium reduces the preconditioning-induced attenuation of necrosis observed with ischemia. Isolated, perfused hearts obtained from Fischer 344 rats of either 3 (young) or 22 (aged) months of age were paced and instrumented for determination of developed left ventricular pressure. Necrosis was determined with triphenyltetrazolium. In the absence of preconditioning, young and aged adult hearts made globally ischemic for 45 min developed necrosis involving 53+/-6% and 49+/-6% of the myocardium, respectively. Contractile function (+dP/dt(max)) at 90 min of reperfusion was depressed by 80% in young and 52% in aged hearts, compared to values obtained prior to preconditioning. Preconditioning with two 5 min ischemia/5 min reperfusion cycles significantly reduced necrosis development and enhanced reperfusion contractile function in young hearts. However, in aged adult hearts, the preconditioning did not significantly reduce the development of necrosis or enhance reperfusion contractile function. These data suggest that aging reduces the effectiveness of preconditioning in providing cardioprotection against ischemic-induced myocardial necrosis.


Subject(s)
Aging , Ischemic Preconditioning, Myocardial , Myocardium/pathology , Animals , Male , Myocardial Contraction , Necrosis , Perfusion , Rats , Rats, Inbred F344
6.
Can J Physiol Pharmacol ; 77(8): 579-88, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10543721

ABSTRACT

The contribution of neuronal ATP to interstitial adenosine levels was investigated in isolated perfused rat hearts. Ventricular surface transudates, representing interstitial fluid, were analyzed for norepinephrine, ATP, and adenosine. Exocytotic release of norepinephrine was induced by electrical stimulation of cardiac efferents emanating from the stellate ganglion. Ganglion stimulation increased contractility, interstitial norepinephrine, ATP, and adenosine. Interstitial adenosine was 11- to 27-fold higher than interstitial ATP, suggesting that the released ATP is unlikely the only source of adenosine. In the presence of AOPCP (alpha,beta-methyleneadenosine 5'-diphosphate), an ecto-5'-nucleotidase inhibitor, the ganglion-stimulated increase in interstitial ATP and adenosine reached levels similar to those in the absence of AOPCP, also suggesting that adenosine does not derive from extracellular ATP. The perfusate Ca2+ was raised from 1 to 4 mM to determine the importance of the enhanced contractile function on the levels of norepinephrine, ATP, and adenosine. The results were increases in contractility and interstitial norepinephrine, ATP, and adenosine, which were not suppressed with atenolol, indicating a norepinephrine-independent release of ATP and adenosine. Reserpine treatment and administration of guanethidine depleted the catecholamine stores and diminished the catecholamine release, respectively. However, neither agent altered Ca2+-induced increases in ATP and adenosine. It is concluded that the amount of neuronal-derived ATP is low and most likely does not contribute significantly to interstitial levels of adenosine. Furthermore, elevations in interstitial norepinephrine, ATP, and adenosine are associated with neuronal-independent increases in contractile function.


Subject(s)
Adenosine Triphosphate/metabolism , Adenosine/metabolism , Myocardium/metabolism , Animals , Calcium/metabolism , Male , Myocardial Contraction , Norepinephrine/metabolism , Perfusion , Rats , Rats, Sprague-Dawley , Stellate Ganglion/metabolism
7.
Am J Physiol ; 276(5): H1434-41, 1999 05.
Article in English | MEDLINE | ID: mdl-10330225

ABSTRACT

Adenosine A2a receptor (A2aR) stimulation enhances the shortening of ventricular myocytes. Whether the A2aR-mediated increase in myocyte contractility is associated with alterations in the amplitude of intracellular Ca2+ transients was investigated in isolated, contracting rat ventricular myocytes using the Ca2+-sensitive fluorescent dye fura 2-AM. In the presence of intact inhibitory G protein pathways, 10(-4) M 2-p-(2-carboxyethyl)phenethyl-amino-5'-N-ethylcarboxamidoadenosine (CGS-21680), an A2aR agonist, insignificantly increased Ca2+ transients by 8 +/- 5%, whereas myocyte shortening increased by 54 +/- 1%. In contrast, 2 x 10(-7) M isoproterenol, a beta-adrenergic receptor agonist, increased Ca2+ transients by 104 +/- 15% and increased myocyte shortening by 61 +/- 6%. When A2aR were stimulated in myocytes that had the antiadrenergic actions of adenosine (Ado) abolished by either treatment with pertussis toxin (PTx) or the presence of 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), an adenosine A1-receptor antagonist, the maximum increases in Ca2+ transients were similarly nominal (with PTx: 10(-4) M CGS-21680, 14 +/- 6% and 10(-4) M Ado, 15 +/- 4%; without PTx: 10(-5) M Ado + 2 x 10(-7) M DPCPX, 19 +/- 1%). These results indicate that compared with beta-adrenergic stimulation, which markedly increases myocyte Ca2+ transients and shortening, A2aR-mediated increases in myocyte shortening are accompanied by only modest increases in Ca2+ transients. These observations suggest that the A2aR-induced contractile effects are mediated predominantly by Ca2+-independent inotropic mechanisms.


Subject(s)
Calcium/metabolism , Muscle Fibers, Skeletal/physiology , Myocardial Contraction/physiology , Myocardium/cytology , Receptors, Adrenergic, alpha-2/physiology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adrenergic beta-Agonists/pharmacology , Animals , Antihypertensive Agents/pharmacology , Carotenoids/pharmacology , Isoproterenol/pharmacology , Male , Muscle Fibers, Skeletal/chemistry , Myocardium/chemistry , Oxygenases/pharmacology , Pertussis Toxin , Phenethylamines/pharmacology , Rats , Rats, Sprague-Dawley , Virulence Factors, Bordetella/pharmacology , Xanthines/pharmacology
8.
J Mol Cell Cardiol ; 31(2): 401-12, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10093052

ABSTRACT

Adenosine levels present in the interstitial fluid and coronary effluent of the aged heart exceed those of the young adult heart. The present study investigated mechanisms in the Fischer 344 rat heart which may be responsible for the observed differences. (1) Total production of adenosine was determined in isolated perfused hearts by measuring coronary effluent adenosine content while inhibiting adenosine deamination and rephosphorylation with erythrohydroxy-nonyladenosine (EHNA) and iodotubercidin (ITC), respectively. Total adenosine production was similar in both young (3-4 month) and aged (20-21 month) hearts at 31.8 +/- 6.6 and 38.4 +/- 3.3 nmol/min/g dry wt, respectively. However, stimulation with the beta-adrenergic agent, isoproterenol, elicited a significantly greater increase in adenosine production in the young vs. aged heart. (2) Adenosine transport was evaluated in isolated perfused hearts by determining 14C uptake by the myocardium after 20 min of 14C-adenosine perfusion. Adenosine uptake in the agent-free heart was found to be decreased 17 to 25% in aged compared to young adult hearts. (3) Adenosine transport characteristics were determined with nitrobenzylthioinosine saturation-binding studies in ventricular membrane preparations. The Bmax values were significantly lower in aged than young adult hearts (140.2 +/- 1.5 fmol/mg and 191.9 +/- 2.3 fmol/mg in aged and young hearts, respectively) indicating a decreased number of transporter sites in the aged heart. However, the values for Kd were decreased with aging, suggesting an increase in the affinity of the transporter for adenosine in the aged vs. young adult heart. (4) The activities and kinetics of adenosine kinase were determined in homogenates of aged and young adult ventricular myocardium. No statistical difference was found between the two activities. Taken together these results suggest that increased interstitial adenosine levels in the aged heart result from decreased uptake of adenosine by the ventricular myocardium.


Subject(s)
Adenosine Kinase/metabolism , Adenosine/biosynthesis , Aging/metabolism , Myocardium/metabolism , Animals , Carbon Radioisotopes , Heart/physiology , Inosine/biosynthesis , Male , Myocardial Contraction , Rats , Rats, Inbred F344 , Receptors, Adrenergic, beta/metabolism , Thioinosine/analogs & derivatives , Thioinosine/metabolism
9.
Am J Physiol ; 276(2): H341-9, 1999 02.
Article in English | MEDLINE | ID: mdl-9950832

ABSTRACT

Presently, the physiological significance of myocardial adenosine A2a receptor stimulation is unclear. In this study, the influence of adenosine A2a receptor activation on A1 receptor-mediated antiadrenergic actions was studied using constant-flow perfused rat hearts and isolated rat ventricular myocytes. In isolated perfused hearts, the selective A2a receptor antagonists 8-(3-chlorostyryl)caffeine (CSC) and 4-(2-[7-amino-2-(2-furyl)[1,2, 4]triazolo[2,3-a][1,3,5]triazin-5-ylamino]ethyl)phenol (ZM-241385) potentiated adenosine-mediated decreases in isoproterenol (Iso; 10(-8) M)-elicited contractile responses (+dP/dtmax) in a dose-dependent manner. The effect of ZM-241385 on adenosine-induced antiadrenergic actions was abolished by the selective A1 receptor antagonist 1,3-dipropyl-8-cyclopentylxanthine (10(-7) M), but not the selective A3 receptor antagonist 3-ethyl-5-benzyl-2-methyl-4-phenylethynyl-6-phenyl-1, 4-(+/-)-dihydropyridine-3,5-dicarboxylate (MRS-1191, 10(-7) M). The A2a receptor agonist carboxyethylphenethyl-aminoethyl-carboxyamido-adenosine (CGS-21680) at 10(-5) M attenuated the antiadrenergic effect of the selective A1 receptor agonist 2-chloro-N6-cyclopentyladenosine (CCPA), whereas CSC did not influence the antiadrenergic action of this agonist. In isolated ventricular myocytes, CSC potentiated the inhibitory action of adenosine on Iso (2 x 10(-7) M)-elicited increases in intracellular Ca2+ concentration ([Ca2+]i) transients but did not influence Iso-induced changes in [Ca2+]i transients in the absence of exogenous adenosine. These results indicate that adenosine A2a receptor antagonists enhance A1-receptor-induced antiadrenergic responses and that A2a receptor agonists attenuate (albeit to a modest degree) the antiadrenergic actions of A1 receptor activation. In conclusion, the data in this study support the notion that an important physiological role of A2a receptors in the normal mammalian myocardium is to reduce A1 receptor-mediated antiadrenergic actions.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Heart/drug effects , Myocardium/metabolism , Receptors, Purinergic P1/physiology , Adenosine/pharmacology , Animals , Calcium/metabolism , Drug Synergism , In Vitro Techniques , Intracellular Membranes/metabolism , Male , Myocardium/cytology , Osmolar Concentration , Rats , Rats, Sprague-Dawley , Triazines/antagonists & inhibitors , Triazines/pharmacology , Triazoles/antagonists & inhibitors , Triazoles/pharmacology
10.
J Hypertens ; 16(12 Pt 1): 1813-22, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9869016

ABSTRACT

BACKGROUND: Increased sympathetic activity contributes to the progression of heart failure. Adenosine counteracts sympathetic activity by inhibition of presynaptic norepinephrine release and attenuation of the metabolic and contractile responses to beta-adrenergic stimulation. In this study, we tested the hypothesis that the adenosinergic effects (uptake blockade) of dipyridamole may retard the progression of pressure overload hypertrophy in the rat. METHODS AND RESULTS: To verify that the administration of dipyridamole increases myocardial adenosine levels in the rat, epicardial adenosine concentrations were measured from 12 isolated, perfused rat hearts exposed to 10(-7) and 10(-6) mol/l dipyridamole. Adenosine concentrations were increased with both doses of dipyridamole. Also, 9 weeks of dipyridamole treatment resulted in decreased sensitivity to the adenosine A1-receptor agonist, 2-chloro-N6-cyclopentyl adenosine, suggesting that dipyridamole increases adenosine levels in the intact rat. In the second part of the study, rats were divided into either abdominal aortic-banded or sham-operated groups and were treated with either dipyridamole or saline. After 9 weeks of treatment, two-dimensional Doppler echocardiographic studies were performed and the adrenergic responsiveness to 10(-8) mol/l isoproterenol was assessed in vitro. The saline-treated banded group demonstrated concentric left ventricular hypertrophy, abnormal diastolic filling, increased wet lung weights and attenuation of adrenergic responsiveness. In contrast, the dipyridamole-treated banded rats exhibited more concentric geometry (higher relative wall thickness with similar left ventricular mass), normal left ventricular filling characteristics and preserved adrenergic responsiveness. Systolic left ventricular chamber and myocardial function, as assessed by stress-endocardial and midwall shortening relationships, were not significantly altered by banding or dipyridamole treatment. CONCLUSIONS: Dipyridamole treatment prevented the development of abnormal left ventricular chamber filling, preserved adrenergic responsiveness and appeared to attenuate detrimental chamber remodeling in rats with pressure overload hypertrophy.


Subject(s)
Adenosine/antagonists & inhibitors , Dipyridamole/pharmacology , Hypertrophy, Left Ventricular/metabolism , Receptors, Adrenergic, beta/metabolism , Ventricular Function, Left/drug effects , Adenosine/blood , Animals , Blood Pressure/drug effects , Body Weight , Dipyridamole/therapeutic use , Heart/drug effects , Hypertrophy, Left Ventricular/drug therapy , Lung , Male , Myocardium/chemistry , Myocardium/metabolism , Myocardium/pathology , Organ Size , Perfusion , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta/physiology , Receptors, Purinergic P1/physiology , Sensitivity and Specificity , Survival , Systole/drug effects , Systole/physiology , Ventricular Function, Left/physiology
11.
Circ Res ; 83(7): 761-71, 1998 Oct 05.
Article in English | MEDLINE | ID: mdl-9758647

ABSTRACT

Adenosine attenuates the myocardial metabolic and contractile responses induced by ss-adrenergic stimulation. Our study was conducted to investigate the longevity of this antiadrenergic action after adenosine exposure. Adenosine (33 micromol/L) was infused into isolated perfused rat hearts for 1, 5, 30, or 60 minutes, and the adrenergic responsiveness (AR) to isoproterenol (10(-8) mol/L) was determined at the end of each infusion period and during a 45-minute adenosine washout period. Interstitial levels of adenosine, as determined from epicardial surface transudates, returned to preinfusion levels within 10 minutes of washout. The duration of adenosine infusion had no effect on the extent of attenuation of AR at the end of the infusion. Whereas AR returned to preadenosine levels with washout of shorter adenosine infusions (1 and 5 minutes), there was a slow and incomplete recovery of AR after the longer exposures (30 and 60 minutes) to adenosine. The magnitude of this persistent antiadrenergic effect (PAE) of adenosine at 15 minutes of washout was proportional to the epicardial concentration of adenosine during infusion of the nucleoside. Infusion of adenosine either with the nonselective adenosine receptor antagonist 8-p-sulfophenyl theophylline or with the selective A1-receptor antagonist 1,3-dipropyl, 8-cyclopentylxanthine, abolished the PAE during the washout period. In addition, the PAE could be demonstrated only with the selective A1-receptor agonist 2-chloro-N6-cyclopentyladenosine and not with the selective A3-receptor agonist 4-aminobenzyl-5'-N methylcarboxamido-adenosine. When the protein kinase C (PKC) inhibitor chelerythrine was coadministered with adenosine, the PAE of adenosine was not apparent during adenosine washout. A 30-minute infusion of phenylephrine, an alpha-adrenergic agonist that enhances PKC activity, produced a PAE that lasted for up to 30 minutes of washout. This effect was prevented by the coinfusion of chelerythrine. Thus, it is concluded that the PAE of adenosine is determined by the myocardial concentration of this nucleoside and is manifested when myocardial concentrations of adenosine returned to baseline levels. Moreover, a 5-minute duration of adenosine exposure is required for the expression of the PAE. This latter effect seems to be dependent on adenosine-induced PKC activation via A1-receptors.


Subject(s)
Adenosine/pharmacology , Adrenergic beta-Antagonists/pharmacology , Heart/drug effects , Protein Kinase C/drug effects , Animals , Enzyme Activation , In Vitro Techniques , Male , Myocardium/enzymology , Pericardium/drug effects , Pericardium/enzymology , Rats , Rats, Sprague-Dawley
12.
Am J Physiol ; 274(2): H627-35, 1998 02.
Article in English | MEDLINE | ID: mdl-9486267

ABSTRACT

In the heart, endogenous adenosine attenuates the beta-adrenergic-elicited increase in contractile performance via activation of adenosine A1 receptors. It has been recently reported that this function of adenosine becomes more pronounced with myocardial maturation. The purpose of the present study was to determine whether mature hearts possess a greater sensitivity than immature hearts to this antiadrenergic effect of adenosine. Isolated perfused hearts or atria from immature (ca. 23 days) and mature (ca. 80 days) rats were stimulated with isoproterenol (Iso), a beta-adrenergic agonist, at 10(-8) M and concomitantly exposed to increasing concentrations of 2-chloro-N6-cyclopentyladenosine (CCPA), a highly selective and potent adenosine A1-receptor agonist, from 10(-12) to 10(-6) M. CCPA at 10(-10)-10(-6) M dose dependently reduced the Iso-elicited contractile response more in immature than in mature hearts or atria. At 10(-6) M, CCPA reduced the Iso-elicited contractile response by 103% in immature hearts and by 55% in mature hearts. These effects of CCPA were attenuated by the adenosine A1-receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine at 10(-7) M. In additional experiments, CCPA exhibited similar effectiveness in reducing the spontaneous heart rate of immature and mature hearts, an effect also mediated by activation of adenosine A1 receptors. Similar to CCPA, the adenosine A1-receptor agonist R-N6-(2-phenylisopropyl)adenosine reduced the Iso-elicited contractile response more in immature than in mature hearts, albeit with less effectiveness than CCPA. In agreement with these results, CCPA reduced Iso-elicited adenylyl cyclase activity more in immature than in mature hearts. Overall, in contrast with our original hypothesis, these results indicate that immature hearts display greater sensitivity than mature hearts to the antiadrenergic effect of adenosine A1-receptor activation.


Subject(s)
Heart/growth & development , Receptors, Purinergic P1/physiology , Adenosine/analogs & derivatives , Adenosine/antagonists & inhibitors , Adenosine/pharmacology , Adenylyl Cyclases/metabolism , Adrenergic beta-Agonists/pharmacology , Aging , Animals , Heart/drug effects , Heart/physiology , Heart Rate/drug effects , Isoproterenol/pharmacology , Male , Myocardial Contraction/drug effects , Purinergic P1 Receptor Agonists , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P1/drug effects , Vasodilator Agents/pharmacology , Xanthines/pharmacology
13.
J Surg Res ; 80(2): 357-64, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9878338

ABSTRACT

BACKGROUND: Adenosine is a potent vasodilator of vascular smooth muscle. Endothelium-derived nitric oxide (NO) elicits vasodilation. We have previously reported that adenosine stimulates the production of NO from porcine carotid arterial endothelial cells (PCAEC) via a receptor-mediated mechanism. This study was to determine whether adenosine also enhances NO production from human arterial endothelium and to define the involvement of adenosine A1 and A2 receptors. MATERIALS AND METHODS: Human iliac arterial endothelial cells (HIAEC) and PCAEC were harvested and cultured in dishes. NO production was evaluated with a NO electrode sensor which measured continuously real-time NO production. RESULTS: NO content of the medium bathing HIAEC and PCAEC was significantly increased with adenosine (100 micromol/L). Ethylcarboxamidoadenosine (NECA), a nonselective adenosine receptor agonist, and carboxyethyl-phenethylamino-ethylcarboxamidoadenosine (CGS-21680), a selective adenosine A2a receptor agonist, increased NO production by HIAEC and PCAEC with respective EC50 values of 3.32 and 6.96 nmol/L for NECA and 30.97 and 29.47 nmol/L for CGS-21680. Chlorofuryl-triazolo-quinazolinamine (CGS-15943; 1 micromol/L), an adenosine A1 and A2 receptor antagonist, and aminofuryltriazolotriazinyl-aminoethylphenol (ZM-241385; 1 micromol/L), a selective adenosine A2a receptor antagonist, inhibited the effect of CGS-21680. Chlorocyclopentyl-adenosine (CCPA; 1 micromol/L), an adenosine A1 receptor agonist, significantly depressed NO production by both HIAEC and PCAEC: This effect was inhibited by cyclopentyl-dipropylxanthine (DPCPX), a selective adenosine A1 receptor antagonist. CONCLUSIONS: The results demonstrate that adenosine A2a receptors increase, and adenosine A1 receptors decrease, the production of NO by human and porcine arterial endothelial cells.


Subject(s)
Endothelium, Vascular/metabolism , Nitric Oxide/biosynthesis , Receptors, Purinergic P1/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Animals , Carotid Arteries/cytology , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Iliac Artery/cytology , Iliac Artery/drug effects , Iliac Artery/metabolism , Phenethylamines/pharmacology , Purinergic P1 Receptor Agonists , Purinergic P1 Receptor Antagonists , Quinazolines/pharmacology , Receptor, Adenosine A2A , Swine , Triazines/pharmacology , Triazoles/pharmacology , Xanthines/pharmacology
14.
Cardiovasc Res ; 34(2): 337-47, 1997 May.
Article in English | MEDLINE | ID: mdl-9205548

ABSTRACT

OBJECTIVE: This study was undertaken to investigate the functional significance of adenosine A2 receptor stimulation in a mammalian ventricular myocyte preparation. METHODS: Isolated contracting rat ventricular myocytes were employed to assess the contractile, adenylyl cyclase and cyclic AMP responses to adenosine receptor stimulation. RESULTS: In single myocytes the presence of A1 receptors was confirmed, as indicated by the A1 receptor agonist, phenylisopropyladenosine (PIA), reducing by 60 and 74% the inotropic response and activation of adenylyl cyclase, respectively, elicited by the beta-adrenergic agonist, isoproterenol. An A1 receptor antagonist, dipropylcyclopentylxanthine (DPCPX), prevented the antiadrenergic action of PIA. The A2 receptor agonist, carboxyethylphenethyl-aminoethyl-carboxamido-adenosine (CGS-21680; 0.01-10 microM) increased myocyte inotropy in a concentration-dependent manner, reaching a maximum of 41-45%. Ethylcarboxamidoadenosine (NECA), naphthyl-substituted aralkoxy-adenosine (SHA-082) and adenosine in the presence of DPCPX also increased myocyte inotropy, as evidenced by increases in myocyte shortening, duration of shortening, time-to-peak shortening, time-to-75% relaxation and rate of maximal shortening. The agonists, however, did not effect the maximal rate of relaxation. The A2 receptor antagonists, chlorofuranyldihydrotri-azoloquinazolinimine (CGS-15943) and chlorostyrylcaffeine (CSC), the latter selective for the A2a receptor, prevented the contractile responses elicited by the A2 agonists. Compared to the concentrations of A2 receptor agonists necessary to increase myocyte contractile variables, 3-12 times greater concentrations of the agonist were required to increase myocyte adenylyl cyclase activity and cAMP levels. CONCLUSIONS: The results suggest the presence of adenosine A2a receptors in the rat ventricular myocyte that appear to be responsible for an increase in inotropy via cAMP-dependent and -independent mechanisms.


Subject(s)
Myocardial Contraction/drug effects , Myocardium/metabolism , Receptors, Purinergic P2/drug effects , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine-5'-(N-ethylcarboxamide) , Adenylyl Cyclases/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Antihypertensive Agents/pharmacology , Cells, Cultured , Cyclic AMP/metabolism , Heart Ventricles , Isoproterenol/pharmacology , Phenethylamines/pharmacology , Phenylisopropyladenosine/pharmacology , Purinergic P1 Receptor Agonists , Purinergic P2 Receptor Agonists , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2/metabolism , Stimulation, Chemical , Vasodilator Agents/pharmacology
15.
Am J Physiol ; 272(3 Pt 2): H1470-9, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9087626

ABSTRACT

We investigated adenosine stimulation of DNA synthesis in human endothelial cells by measuring [3H]thymidine incorporation in cultures derived from human umbilical veins. After 18 h of exposure to adenosine in serum-free medium, endothelial cell [3H]thymidine incorporation was increased by 30-64%. Adenosine-induced DNA synthesis was not mimicked by adenosine receptor agonists and was not inhibited by adenosine receptor antagonists. Adenosine-induced DNA synthesis was inhibited 81% by 100 microM 5'-(N,N-dimethyl)amiloride, an inhibitor of Na+/H+ exchange, and was totally inhibited by 10 microM 2',4'-dibromoacetophenone, an inhibitor of phospholipase A2 (PLA2). Adenosine increased adenosine 3',5'-cyclic monophosphate levels in endothelial cells, but adenosine-induced DNA synthesis was not inhibited by the protein kinase A (PKA) inhibitor Rp-cAMPS. Both ATP and the phorbol ester 4beta-phorbol 12-myristate 13-acetate (PMA) increased DNA synthesis in human endothelial cells. Stimulation by ATP was inhibited by the P2-receptor antagonist suramin, and PMA stimulation was inhibited by the protein kinase C (PKC) inhibitor H-7. Neither suramin nor H-7 inhibited adenosine-stimulated DNA synthesis. The results suggest that Na+/H+ exchange and PLA2 are involved in adenosine-induced DNA synthesis in cultures of human endothelial cells independently of adenosine receptor, PKA, or PKC activation.


Subject(s)
Adenosine/pharmacology , DNA/biosynthesis , Endothelium, Vascular/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , Adenosine Triphosphate/pharmacology , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/analogs & derivatives , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Kinetics , Poly A/pharmacology , Purinergic P1 Receptor Antagonists , Quinazolines/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Thioinosine/analogs & derivatives , Thioinosine/pharmacology , Thionucleotides/pharmacology , Thymidine/metabolism , Triazoles/pharmacology , Umbilical Veins
17.
Am J Physiol ; 271(1 Pt 2): H235-43, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8760180

ABSTRACT

Myocardial contractile responsiveness to beta-adrenoceptor stimulation is known to be reduced with maturation or aging. The present study was undertaken to determine the role of antiadrenergic A1 and stimulatory A2 adenosine receptors in the modulation of beta-adrenergic-elicited contractile performance of the heart at juvenile (approximately 25 days) and adult (approximately 79 days) stages of maturation. Isoproterenol, a beta-adrenergic agonist, at 10(-7) M produced a greater maximal increase in contractility, assessed as the maximal rate of left ventricular pressure development (+dP/dtmax), in immature than in mature hearts (104 and 80%, respectively), but produced a greater increase in venous adenosine concentration in the mature than in the immature hearts (738 and 277 nM, respectively). Isoproterenol at 10(-9) to 10(-8) M produced similar increases in contractility in the absence or presence of the A1 adenosine receptor antagonist xanthine amine congener (XAC; 0.5 microM) for both immature and mature hearts. In addition, XAC did not alter the isoproterenol-elicited contractile response in the immature heart during hypoperfusion induced by 50% reduction of coronary flow. However, in the mature heart, 10(-8) M isoproterenol elicited a significantly greater increase in +dP/dtmax during hypoperfusion in the presence (79%) vs. the absence (60%) of XAC. In both immature and mature hearts, hypoperfusion enhanced isoproterenol-elicited venous adenosine concentration by similar magnitudes of 76 and 72%, respectively. In further studies, the A2 adenosine receptor antagonist 9-chloro-2-(2-furyl)[1,2,4]-triazolo[1,5-c]quinazolin-5-amine (CGS-15943; 1 microM) reduced the isoproterenol-elicited contractile response of mature but not immature hearts during normal perfusion. These results suggest that myocardial adenosine modulates the beta-adrenergic-elicited contractile response of the adult heart via activation of both A1 and A2 adenosine receptors and that these functions of adenosine become expressed with myocardial maturation.


Subject(s)
Aging/metabolism , Myocardium/metabolism , Receptors, Purinergic P1/metabolism , Adenosine/blood , Animals , Coronary Vessels , Drug Synergism , Heart/drug effects , Heart/growth & development , Isoproterenol/pharmacology , Male , Myocardial Contraction/drug effects , Osmolar Concentration , Purinergic P1 Receptor Antagonists , Quinazolines/pharmacology , Rats , Rats, Sprague-Dawley , Triazoles/pharmacology , Veins , Xanthines/pharmacology
18.
Life Sci ; 58(6): 493-502, 1996.
Article in English | MEDLINE | ID: mdl-8569422

ABSTRACT

Interstitial levels and release of adenosine have been shown to be greater for aged adult hearts compared to young adult hearts. Furthermore, blockade of A1 adenosine receptors in the aged adult heart prevents the reduced contractile and metabolic response to isoproterenol. The aim of this study was to determine whether there is an enhanced antiadrenergic effect of adenosine in the aged adult heart. Ventricular membranes from young and aged adult hearts were incubated in the presence of isoproterenol (ISO) and phenylisopropyladenosine (PIA) either alone or in combination. Basal and ISO-enhanced adenylyl cyclase activity were significantly reduced in the membranes from aged rats. PIA alone, at 0.1 nM to 100 microM, had no direct effect on basal adenylyl cyclase activity in membranes from either group. In the presence of either 100 nM or 1 microM ISO, 100 microM PIA significantly attenuated ISO-enhanced adenylyl cyclase activity to a greater extent in the aged adult heart membranes (78 or 48% for the aged vs. 37 or 25% for the young). Moreover, in the presence of 100 nM ISO the IC50 for the PIA concentration response curve was shifted to the left for the aged ventricular membranes as compared to the membranes from young adults (1.62 x 10(-7) M vs 1.5 x 10(-6) M, aged vs young, respectively). The enhanced inhibition of adenylyl cyclase is associated with an increase in adenosine A1 receptor density (23.7 +/- 3.5 vs 14.7 +/- 1.7 fmol/mg, aged vs young) and Kd (6.1 +/- 1.7 vs 2.2 +/- 0.5 nM, aged vs young) in the aged adult heart membranes as determined by [3H]DPCPX binding. These results suggest that the reduced response to catecholamines in the aged adult heart may be due, at least in part, to an enhanced expression of the antiadrenergic effect of adenosine on beta-adrenergic receptor mediated activation of adenylyl cyclase.


Subject(s)
Adenosine/physiology , Adenylyl Cyclases/metabolism , Adrenergic beta-Agonists/pharmacology , Aging/metabolism , Heart/drug effects , Heart/growth & development , Isoproterenol/pharmacology , Myocardium/enzymology , Phenylisopropyladenosine/pharmacology , Adenylyl Cyclase Inhibitors , Animals , Drug Interactions , Male , Membranes/drug effects , Membranes/enzymology , Myocardium/ultrastructure , Purinergic P1 Receptor Agonists , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Receptors, Purinergic P1/metabolism , Stimulation, Chemical , Tritium , Xanthines/metabolism , Xanthines/pharmacology
19.
J Mol Cell Cardiol ; 27(10): 2373-83, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8576951

ABSTRACT

High levels of norepinephrine in the heart are cardiotoxic resulting in contractile dysfunction and arrhythmic activity via beta-adrenoceptor mediated mechanisms. A low flow heart model perfused with physiological saline containing glucose and bubbled with an O2 gas mixture was used to determine whether adenosine, a nucleoside with antiadrenergic properties, could reduce the functional manifestations of catecholamine cardiotoxicity. Isolated rat hearts were treated with dipropylcyclopentylxanthine (DPCPX; 0.1 microM; A1 receptor antagonist) to block endogenous adenosine. In DPCPX-treated hearts stimulated with isoproterenol (ISO; 1 microM) during 45 min of low flow (0.5 ml/min) perfusion, the recovery of contractile function (ConF) at 15 min after the restoration of normal flow was 64% of control (before low flow) values as compared to 110% recovery of ConF in the absence of ISO. The incidence of arrhythmias observed upon restoration of control flow was increased by ISO when the action of endogenous adenosine was blocked with DPCPX. In the absence of DPCPX both the functional depression and arrhythmias induced by ISO were prevented in the presence of phenylisopropyladenosine (PIA; 1 microM; A1 receptor agonist). At 15 min after normal flow was restored. ConF in ISO-treated hearts with PIA was 53% greater than in the absence of PIA and presence of DPCPX. This enhancement of ConF by PIA was significantly reduced by DPCPX. By 30 min after flow restoration, these significant differences were absent. DPCPX reversed the PIA-induced reduction in arrhythmias observed upon restoration of normal flow. PIA and DPCPX alone in the absence of ISO, and ISO in the absence of PIA and DPCPX, did not result in altered ConF upon restoration of normal flow. These findings indicate that intense beta-adrenergic stimulation of the heart during low-flow perfusion in the absence of adenosine A1 receptor activity induces contractile depression and arrhythmicity subsequent to restoration of control perfusion. It is concluded that endogenous adenosine protects the heart against catecholamine toxicity via stimulation of adenosine A1 receptors.


Subject(s)
Adenosine/physiology , Adrenergic beta-Agonists/pharmacology , Arrhythmias, Cardiac/chemically induced , Isoproterenol/pharmacology , Myocardial Contraction/drug effects , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/physiopathology , Receptors, Adrenergic, beta/drug effects , Receptors, Purinergic P1/physiology , Adrenergic beta-Agonists/toxicity , Animals , Arrhythmias, Cardiac/physiopathology , Coronary Circulation , Epinephrine/metabolism , Isoproterenol/antagonists & inhibitors , Isoproterenol/toxicity , Male , Perfusion , Purinergic P1 Receptor Antagonists , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta/physiology , Xanthines/pharmacology
20.
Am J Physiol ; 269(2 Pt 1): C519-23, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7653535

ABSTRACT

Adenosine per se is a potent vasodilator of vascular smooth muscle. Endothelial cells modulate vascular tone via the release of nitric oxide (NO), which also elicits vasodilation. This study was undertaken to determine whether adenosine could directly stimulate endothelial cells to enhance NO production, which could subsequently reduce vascular tone. NO production was evaluated in porcine carotid artery endothelial cells (PCAEC) and human saphenous vein endothelial cells (HSVEC) seeded on multiwell plates, grown to confluence, and treated with adenosine for 1 h. The bathing medium was collected, and the NO production was determined as reflected by the formation of NO2- and NO3-. NO production by PCAEC was significantly increased by adenosine in a dose-dependent manner, whereas there was only an insignificant tendency for an increase by HSVEC. The addition of the NO synthase competitive inhibitor, NG-monomethyl-L-arginine (NMMA), or the adenosine receptor antagonist, theophylline, prevented the increase in NO production by adenosine. The results suggest that adenosine stimulates, by a receptor-mediated mechanism, the production of NO by arterial, but not by venous, endothelial cells.


Subject(s)
Adenosine/pharmacology , Endothelium, Vascular/metabolism , Nitric Oxide/biosynthesis , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Carotid Arteries/cytology , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Saphenous Vein/cytology , Saphenous Vein/drug effects , Saphenous Vein/metabolism , Swine , Theophylline/pharmacology , omega-N-Methylarginine
SELECTION OF CITATIONS
SEARCH DETAIL
...