Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters










Publication year range
1.
Nucl Med Biol ; 34(2): 211-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17307129

ABSTRACT

This study evaluated novel potential dopamine transporter (DAT) inhibitors as ligands for positron emission tomography. Five new tropane analogs were synthesized and compared with the known ligand 2beta-carbomethoxy-3beta-(4-iodophenyl)tropane (beta-CIT) and the recently characterized ligands N-(3-iodoprop-2E-enyl)-2beta-carbomethoxy-3beta-(4-methylphenyl)-nortropane (PE2I) and 2beta-carbofluoroethoxy-3beta-(4-methylphenyl)tropane (FETT). Evaluation with autoradiography measured the ability to antagonize the binding of [(131)I]iodine-labeled beta-CIT and [(18)F]fluorine-labeled N-(3-fluoropropyl)-2beta-carbomethoxy-3beta-(4-iodo-phenyl) nortropane in rat and pig brains. The standards for comparison (PE2I and FETT) competed strongly in all regions investigated (striatum, cortex, superior colliculus and cerebellum). Of the new compounds, 2alpha-amido-fluoroethyl-3beta-(4-iodophenyl)tropane (4) and 2beta-amido-fluoroethyl-3beta-(4-iodophenyl)tropane (4a) competed strongly with [(131)I]beta-CIT in DAT-rich striatum, but also in other brain regions suggesting poor DAT selectivity. Because [(131)I]beta-CIT binds unselectively both to DAT and serotonin transporters, no definite conclusion about the selectivity of the new compounds is possible. However, preclinical studies using the compounds and labeled with fluorine-18 or iodine-131 are encouraged.


Subject(s)
Brain/metabolism , Cocaine/analogs & derivatives , Tropanes/pharmacokinetics , Animals , Brain/diagnostic imaging , Cocaine/pharmacokinetics , Fluorine Radioisotopes/pharmacokinetics , Iodine Radioisotopes/pharmacokinetics , Metabolic Clearance Rate , Organ Specificity , Protein Binding , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Wistar , Tissue Distribution , Tropanes/chemistry
2.
Nucl Med Biol ; 31(2): 173-7, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15013482

ABSTRACT

Autoradiography on rat brain using tritiated (1*), mono- (2*) and di-radioiodinated (3*) derivatives of the A(2A) adenosine receptor antagonist ZM241,385 showed high receptor density in striatum. K(D)s of 1*, 2* and 3* were 0.4, 2.2 and 15 nM and nonspecific binding was 5, 40 and 50% of total binding. Striatal uptake of 2* in mice was approximately 0.2% ID/g 60 min post-injection; blocking by 2 was insignificant. Poor penetration of the blood brain barrier and high nonspecific binding make 2* unsuitable for imaging striatal receptors.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Receptor, Adenosine A2A/metabolism , Triazines/pharmacokinetics , Triazoles/pharmacokinetics , Animals , Iodine Radioisotopes/pharmacokinetics , Isotope Labeling/methods , Metabolic Clearance Rate , Organ Specificity , Protein Binding , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Rats , Tissue Distribution , Tritium/pharmacokinetics
3.
Nucl Med Biol ; 30(6): 661-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12900292

ABSTRACT

8-Cyclopentyl-3-[(E)-3-[(131)I]iodoprop-2-en-1-yl]-1-propylxanthine (2*) was generated by iododestannylation of the tributyl-stannyl-precursor with [(131)I]NaI and chloramine T. The radiochemical yield of 2* was 82 +/- 4%, and the purity exceeded 98%. The specific activity was 33 +/- 19 GBq/micromol. Affinities for rat, pig and human A(1) adenosine receptors (A(1)ARs) were in the low nanomolar range, but poor selectivity for the human A(1)AR over the A(2A)AR was found. Additionally, in vitro and ex vivo autoradiographic studies revealed high unspecific binding which makes this ligand unsuitable for SPECT imaging.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Receptors, Purinergic P1/metabolism , Tomography, Emission-Computed, Single-Photon/methods , Xanthine/pharmacokinetics , Animals , Autoradiography , Cell Line , Female , Humans , In Vitro Techniques , Male , Metabolic Clearance Rate , Mice , Organ Specificity , Purinergic P1 Receptor Antagonists , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Wistar , Receptors, Purinergic P1/classification , Reproducibility of Results , Sensitivity and Specificity , Species Specificity , Swine , Tissue Distribution , Xanthine/chemical synthesis , Xanthines
4.
Am J Physiol Heart Circ Physiol ; 278(1): H74-84, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10644586

ABSTRACT

The purpose of this investigation was to quantitatively evaluate the role of adenosine in coronary exercise hyperemia. Dogs (n = 10) were chronically instrumented with catheters in the aorta and coronary sinus, and a flow probe on the circumflex coronary artery. Cardiac interstitial adenosine concentration was estimated from arterial and coronary venous plasma concentrations using a previously tested mathematical model. Coronary blood flow, myocardial oxygen consumption, heart rate, and aortic pressure were measured at rest and during graded treadmill exercise with and without adenosine receptor blockade with either 8-phenyltheophylline (8-PT) or 8-p-sulfophenyltheophylline (8-PST). In control vehicle dogs, exercise increased myocardial oxygen consumption 4.2-fold, coronary blood flow 3.8-fold, and heart rate 2.5-fold, whereas mean aortic pressure was unchanged. Coronary venous plasma adenosine concentration was little changed with exercise, and the estimated interstitial adenosine concentration remained well below the threshold for coronary vasodilation. Adenosine receptor blockade did not significantly alter myocardial oxygen consumption or coronary blood flow at rest or during exercise. Coronary venous and estimated interstitial adenosine concentration did not increase to overcome the receptor blockade with either 8-PT or 8-PST as would be predicted if adenosine were part of a high-gain, negative-feedback, local metabolic control mechanism. These results demonstrate that adenosine is not responsible for local metabolic control of coronary blood flow in dogs during exercise.


Subject(s)
Adenosine/physiology , Coronary Circulation/physiology , Motor Activity/physiology , Animals , Dogs , Dose-Response Relationship, Drug , Male , Purinergic P1 Receptor Antagonists , Pyridines/pharmacology , Receptors, Endothelin/physiology , Tetrazoles/pharmacology , Theophylline/analogs & derivatives , Theophylline/pharmacology
5.
J Med Chem ; 41(4): 555-63, 1998 Feb 12.
Article in English | MEDLINE | ID: mdl-9484505

ABSTRACT

The high affinity of 8-cyclopentyl-1,3-dipropylxanthine (CPX) for the A1 adenosine receptor (A1AR) provides a good lead for developing radioligands suitable for positron emission tomography (PET) and single-photon emission tomography (SPET). This study tested the hypothesis that the kinds of chemical modifications made in the synthesis of CPX analogues containing carbon-11, fluorine-18, or radioiodine will not alter affinity for the A1AR. This report describes the synthesis and radioligand binding assays of unlabeled CPX analogues having methyl, 2-methoxyethyl, 2-fluoropropyl, or 3-fluoropropyl substituents, respectively, at either N-1 (13a-d) or N-3 (8a-d) or an (E)-3-iodoprop-2-en-1-yl substituent at N-3 (8f). Compounds 8d,f and 13b,d antagonized the binding of [3H]CPX to the A1AR of rat brain with affinities similar to those of CPX; compound 8c was twice as potent as CPX. Analogues 8a,b and 13a were less potent than CPX, but for each the Ki of antagonism was > or = 0.5 nM. Attempts to iodinate the 8-(4-hydroxyphenyl) analogue of CPX failed, probably because the xanthine substituent strongly deactivated the phenol toward electrophilic iodination. In summary, several of the modifications of the propyl groups of CPX needed to produce ligands for imaging by PET and SPET preserve or enhance affinity for the A1AR.


Subject(s)
Brain/metabolism , Purinergic P1 Receptor Antagonists , Xanthines/chemical synthesis , Animals , Binding, Competitive , Cattle , Cell Membrane/metabolism , Cerebral Cortex/metabolism , Drug Design , Indicators and Reagents , Ligands , Radioligand Assay , Rats , Structure-Activity Relationship , Tomography, Emission-Computed , Tomography, Emission-Computed, Single-Photon , Tritium , Xanthines/pharmacokinetics
6.
J Pharmacol Exp Ther ; 284(3): 1066-73, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9495868

ABSTRACT

The coronary vasodilation caused by adenosine is due to activation of A2 adenosine receptors (A2AdoRs), but the subtype or subtypes of A2AdoR (A2A and/or A2B) that mediate this action are uncertain. The purpose of this study was to test the hypothesis that A2AAdoRs mediate coronary vasodilation caused by exogenous or endogenous adenosine in the guinea pig isolated perfused heart. The newly described A2AAdoR antagonist SCH58261 was used to selectively block A2AAdoRs. Attenuations by SCH58261 of increases in coronary conductance (A2 response) and of atrioventricular nodal conduction time (A1 response) caused by exogenous and endogenous adenosine and by agonists with relative selectivity for A2A and A1AdoRs were measured. The CGS21680-induced increase of coronary conductance was antagonized by SCH58261 in a concentration-dependent and competitive manner with a KB value of 5.01 nm. Also reversed by SCH58261 (60 nmol/L) were the increases in coronary conductance caused by the relatively selective A1AdoR agonists CCPA (70 nM), and (R)-(-)N(b)-(2-phenyl-isopropyl)adenosine (60 nM) but not those caused by sodium nitroprusside (1.2 microM) and diltiazem (0.4 microM). SCH58261 (< or = 100 nM) did not attenuate the A1AdoR-mediated prolongations of S-H interval caused by either adenosine or CCPA. SCH58261 attenuated the coronary vasodilation caused by 50 nM adenosine with an IC50 value of 6.8 +/- 0.6 nM. The coronary vasodilations caused by the nucleoside uptake inhibitor draflazine and the adenosine kinase inhibitor iodotubercidin were completely reversed by 60 nM SCH58261 or adenosine deaminase (7 U/ml). Thus, the A2AAdoR plays a major role as mediator of coronary vasodilation caused by exogenous and endogenous adenosine and by AdoR agonists.


Subject(s)
Coronary Vessels/physiology , Receptors, Purinergic P1/physiology , Vasodilation/drug effects , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Female , Guinea Pigs , Male , Perfusion , Phenethylamines/pharmacology , Piperazines/pharmacology , Pyrimidines/pharmacology , Triazoles/pharmacology , Tubercidin/analogs & derivatives , Tubercidin/pharmacology , Xanthines/pharmacology
7.
Am J Physiol ; 270(2 Pt 2): F263-74, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8779886

ABSTRACT

Adenosine A1 receptor densities were increased in cultured LLC-PK1 and OK cells by chronic treatment with the adenosine receptor antagonists 1,3,7-trimethylxanthine (caffeine, 1 mM) and 1,3-dimethyl-8-cyclopentylxanthine [cyclopentyltheophylline (CPT), < or = 0.4 mM], respectively. The A1 receptor number per cell was increased twofold by 10-day treatments with 1 mM caffeine or 0.1 mM CPT, and the sodium-coupled glucose uptake was augmented twofold by 1 mM caffeine and sevenfold by 0.1 microM CPT (higher doses of CPT were progressively less stimulatory). Glucose uptake was blocked by acute (2-h) treatment with CPT, adenosine deaminase, or calphostin C. Caffeine (1 mM) or CPT (> or = 0.1 mM) inhibited cell proliferation for the first 10 days, then cell growth assumed a normal proliferative rate despite continued presence of antagonist. Cytosolic protein kinase C (PKC) beta-isoform immunoactivity and PKC-beta II mRNA were elevated at least twofold during 10 days of 0.1 mM CPT or 1 mM caffeine treatment. The sustained elevation in sodium-glucose symport and PKC activity observed with adenosine receptor antagonists was similar to acute (2-h) effects of the adenosine A1 agonist R(-)-N6-phenylisopropyladenosine (R-PIA, 0.1-1 microM). Moreover, cell proliferation was increased by adenosine (0.1 microM R-PIA), whereas Na-K-adenosinetriphosphatase activity was unaltered with chronic antagonist or acute adenosine treatments. Caffeine treatment may have some non-adenosine A1 receptor-mediated actions, because it slightly (30%) augmented protein kinase A activity. It is concluded that chronic exposure of proximal tubule cells to caffeine or CPT augments PKC and sodium-glucose transport but retards cell proliferation mainly via adenosine A1 receptor-mediated mechanisms.


Subject(s)
Glucose/metabolism , Kidney/metabolism , Protein Kinase C/metabolism , Receptors, Purinergic P1/metabolism , Up-Regulation , Adenosine Deaminase/pharmacology , Animals , Biological Transport/physiology , Caffeine/pharmacology , Cell Division/drug effects , Cell Division/physiology , Cell Line , Kidney/cytology , LLC-PK1 Cells , Phenylisopropyladenosine/pharmacology , Purinergic P1 Receptor Agonists , Purinergic P1 Receptor Antagonists , Swine , Theophylline/analogs & derivatives , Theophylline/pharmacology
8.
Naunyn Schmiedebergs Arch Pharmacol ; 353(3): 250-60, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8692279

ABSTRACT

The effect of several adenosine analogues on cyclic AMP accumulation was examined in the rat phaeochromocytoma cell PC12 and in the human T-cell leukaemia cell Jurkat, selected as prototypes of cells predominantly expressing adenosine A2A or A2B receptors. Using the reverse transcription-polymerase chain reaction it was, however, demonstrated that the Jurkat cell and the PC12 cell express both A2A and A2B receptor mRNA, albeit in different relative proportions. In PC12 cells the concentration required for half-maximal response (EC50) for the full agonist 5'-N-ethyl-carboxamidoadenosine (NECA) was 30 times lower than in Jurkat cells. There was no significant difference in the pA2 for the antagonist 5-amino-9-chloro-2-(2-furanyl)- 1,2,4-triazolo(1,5-C)quinazolinemonomethanesulphonate (CGS 15943) between the two cell types. In the presence of forskolin (1 microM in PC12 cells; 10 microM in Jurkat cells) the EC50 value for NECA was reduced two-to sixfold. Forskolin also increased the maximal cAMP accumulation twofold in PC12 cells and sevenfold in Jurkat cells. A series of 2-substituted adenosine analogues CV 1808 (2-phenylamino adenosine), CV 1674 [2-(4-methoxyphenyl)adenosine], CGS 21680 ¿2-[p-(2-carbonylethyl)phenylethylamino]-5'-N-ethyl- carboxamido adenosine¿, and four 2-substituted isoguanosines, SHA 40 [2-(2-phenylethoxy)adenosine; PEA], SHA 91 [2-(2-cyclohexylethoxy)adenosine; CEA], SHA 118 ¿2-[2-(p-methylphenyl)ethoxy]adenosine; MPEA¿, and SHA 125 (2-hexyloxyadenosine; HOA), all raised cAMP accumulation in PC12 cells, but had minimal or no effect in Jurkat cells. In the PC12 cells the addition of forskolin (1 microM) reduced the EC50 by a factor of 2(CV 1808) to 12 (SHA 125). In Jurkat cells all the analogues gave a significant, but submaximal, cAMP response in the presence of forskolin (10 microM), but they were essentially inactive in its absence. The results show that a series of 2-substituted adenosine analogues can be used to discriminate between A2A and A2B receptors. The two receptor subtypes appear to coexist, even in clonal cells selected for typical pharmacology. A2 receptor pharmacology can therefore be complex.


Subject(s)
Adenosine/analogs & derivatives , Leukemia, T-Cell/pathology , PC12 Cells/drug effects , Purinergic P1 Receptor Agonists , Quinazolines/pharmacology , Triazoles/pharmacology , Adenosine/metabolism , Adenosine/pharmacology , Adenosine-5'-(N-ethylcarboxamide) , Amino Acid Sequence , Animals , Base Sequence , Binding, Competitive , Colforsin/pharmacology , Cyclic AMP/metabolism , DNA/biosynthesis , DNA/chemistry , Humans , Isotope Labeling , Lethal Dose 50 , Leukemia, T-Cell/metabolism , Molecular Sequence Data , PC12 Cells/cytology , PC12 Cells/metabolism , Phenethylamines/metabolism , Phenethylamines/pharmacology , Polymerase Chain Reaction , Quinazolines/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Receptors, Purinergic P1/classification , Receptors, Purinergic P1/genetics , Receptors, Purinergic P1/metabolism , Triazoles/metabolism , Tumor Cells, Cultured
9.
Mol Pharmacol ; 47(2): 307-13, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7870039

ABSTRACT

Adenosine analogs substituted in the 2-position with arylamino groups have been found to have high affinity and selectivity for A2a adenosine receptors. Two such compounds, 2-[2-(4-aminophenyl)ethylamino]adenosine and 2-[2-4-amino-3-iodophenyl)ethylamino]adenosine (I-APE), were synthesized and found to be potent coronary vasodilators (ED50 < 3 nm). These compounds bind weakly to A1 adenosine receptors of rat cortex (Ki > 150 nM). 125I-APE was synthesized and the new radioligand was found to bind to two affinity states of rat striatal A2a adenosine receptors (Kd = 1.3 +/- 0.1 nM and 19 +/- 4.5 nM). The high affinity site represents a previously unrecognized small (15-20%) fraction of A2a adenosine receptors coupled to G proteins. Guanosine 5'-O-(3-thio)triphosphate (GTP gamma S) reduces specific binding of 125I-APE half-maximally at a concentration of 45 +/- 2 nM. [3H]CGS21680 also binds to two affinity states of A2a receptors on striatal membranes (Kd = 3.9 +/- 0.9 and 51 +/- 5.5 nM), although in previous studies single Kd values ranging from 5 to 15 nM have been reported. This high affinity site is substantiated by the finding that the IC50 of CGS21680 in competition with 125I-APE binding to striatal membranes is shifted leftward in membranes diluted for 4 min before filtration, to selectively dissociate radioligand from low affinity receptors. Assuming that agonist radioligands bind to both coupled and uncoupled forms of striatal A2a adenosine receptors, we could simulate with the computer the finding that the decrease in specific binding induced by GTP gamma S (100 microM) is variable and depends on radioligand concentration, ranging from 20 to 90%. Unlike 125I-APE, [3H]CGS21680 is charged at physiological pH, and treatment of membranes with the pore-forming antibiotic alamethicin uncovers cryptic [3H]CGS21680 but not 125I-APE binding sites. We conclude that the GTP gamma S-sensitive high affinity form of the A2a adenosine receptor can be preferentially labeled by 125I-APE, due to both its high specific activity and its physicochemical properties. Possible functional manifestations of poor coupling of A2a adenosine receptors to G proteins are discussed.


Subject(s)
Adenosine/analogs & derivatives , Receptors, Purinergic P1/metabolism , Adenosine/metabolism , Animals , Cell Line , Iodine Radioisotopes , Phenethylamines/metabolism , Protein Binding , Radioligand Assay , Rats , Tritium
10.
J Pharmacol Exp Ther ; 271(1): 14-9, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7965706

ABSTRACT

Experiments in guinea pig heart Langendorff preparations assessed the effect of KATP channel blockade on the coronary vasoactivity of adenosine and 17 analogs chosen to represent a variety of purine and ribose modifications. Although glibenclamide is a functional antagonist that acts at the level of an effector rather than at a receptor, it caused parallel rightward shifts of agonist dose-response curves. The size of the shift of EC50 differed according to the kind of analog: the ranking was, generally, N6-phenethyladenosines > 2-aryl-aminoadenosines = 2-(1-alkyn-1-yl)adenosines > N6-cycloalkyladenosines = adenosine-5' -uronamides. The coronary vasoactivity ranking of agonists in the presence of supramaximal concentrations of glibenclamide was 2-(1-alkyn-1-yl)adenosines = 2-aralkoxyadenosines > 2-aralkylaminoadenosines > 2-arylaminoadenosines > N6-substituted adenosines. Glibenclamide did not affect the vasoactivity of adenosine itself, perhaps because avid uptake by endothelial cells prevented penetration of the agonist to receptors deeper in the vascular wall. The results exclude a model consisting of one kind of receptor acting exclusively through a KATP channel, argue against one kind of receptor coupled to a KATP channel as well as to an additional effector but is consistent with two kinds of vasodilatory adenosine receptors, one of which activates a KATP channel. The identity of the adenosine receptor coupled to the KATP channel is uncertain; the other receptor has the pharmacological profile of an A2a-adenosine receptor.


Subject(s)
Coronary Vessels/drug effects , Glyburide/pharmacology , Purinergic P1 Receptor Agonists , Adenosine/metabolism , Adenosine/pharmacology , Animals , Coronary Vessels/physiology , Dose-Response Relationship, Drug , Guinea Pigs , In Vitro Techniques , Potassium Channels/drug effects , Receptors, Purinergic P1/physiology
11.
J Med Chem ; 37(17): 2704-12, 1994 Aug 19.
Article in English | MEDLINE | ID: mdl-8064798

ABSTRACT

This report describes the synthesis of 29 xanthines containing a chemoreactive chloroaryl, beta-chloroethylamino, alpha,beta-unsaturated carbonyl, bromoacetyl, 3-(fluorosulfonyl)benzoyl, or 4-(fluorosulfonyl)benzoyl group as part of an exocyclic 1-, 3-, or 8-substituent. The xanthines inhibited the binding of [3H]-8-cyclopentyl-1,3-dipropylxanthine ([3H]CPX) to the A1 adenosine receptor (A1AR) of DDT1 MF2 cells at IC50s in the low-nanomolar to low-micromolar range. Seven of the 29 analogues irreversibly inhibited the binding of [3H]CPX without changing the KD of that ligand; five were 1,3-dipropylxanthines having the following reactive groups as 8-substituents: (bromoacetamido)methyl (24), (bromoacetamido)ethyl (25), (bromoacetamido)propyl (26), [4-(fluorosulfonyl)benzamido]methyl (33) or 3-[[4-(fluorosulfonyl)benzoyl]oxy]cyclopentyl (42). Both 8-cyclopentyl-3-[3-[[4- (fluorosulfonyl)benzoyl]oxy]propyl]-1-propylxanthine (53) and 8-cyclopentyl-1,3-bis[3-[[4- (fluorosulfonyl)benzoyl]oxy]propyl]xanthine (55) inhibited [3H]CPX binding irreversibly. Five of the ligands, including 26, 33 (IC50 = 49 microM), and 53 (IC50 = 9 microM), antagonized the binding of [3H]NECA to the A2aAR of PC12 cells, but unlike binding to the A1AR, binding to the A2aAR was completely reversible. The potency of 33 (IC50 = 2 microM, 72% loss of CPX binding at 1 microM) and 53 (IC50 = 0.01 microM, 74% loss of CPX binding at 0.05 microM) and their selectivity for the A1AR suggest that those two ligands may be useful in studies of the structure and function of that receptor.


Subject(s)
Purinergic P1 Receptor Antagonists , Xanthines/chemical synthesis , Xanthines/pharmacology , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine-5'-(N-ethylcarboxamide) , Animals , Cell Membrane/metabolism , Drug Design , Indicators and Reagents , Magnetic Resonance Spectroscopy , Molecular Structure , PC12 Cells , Radioligand Assay , Structure-Activity Relationship , Xanthines/chemistry , Xanthines/metabolism
12.
Cardiovasc Res ; 28(7): 1057-61, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7954592

ABSTRACT

OBJECTIVE: Agonists selective for the A1 adenosine receptor mimic the protective effect of ischaemic preconditioning against infarction in the rabbit heart. Unselective adenosine antagonists block this protection but, paradoxically, the A1 adenosine receptor selective antagonist 8-cyclopentyl- 1,3-dipropylxanthine (DPCPX) does not. The aim of this study was to test the hypothesis that the newly described A3 adenosine receptor, which has an agonist profile similar to the A1 receptor but is insensitive to DPCPX, might mediate preconditioning. METHODS: Isolated rabbit hearts perfused with Krebs buffer experienced 30 min of regional ischaemia followed by 120 min of reperfusion. Infarct size was measured by tetrazolium staining. RESULTS: In control hearts infarction was 32.2(SEM 1.5)% of the risk zone. Preconditioning by 5 min ischaemia and 10 min reperfusion reduced infarct size to 8.8(2.3)%. Replacing the regional ischaemia with 5 min perfusion with 10 microM adenosine or 65 nM N6-[2-(4-aminophenyl)ethyl]adenosine (APNEA), an adenosine A3 receptor agonist, was equally protective. The unselective antagonist 8-p-sulphophenyl theophylline at 100 microM abolished protection by preconditioning, adenosine, and APNEA, but 200 nM DPCPX did not block protection by any of the interventions. Likewise the potent but unselective A3 receptor antagonist 8-(4-carboxyethenylphenyl)-1,3-dipropylxanthine (BW A1433) completely blocked protection from ischaemic preconditioning. CONCLUSIONS: Because protection against infarction afforded by ischaemic preconditioning, adenosine, or the A3 receptor agonist APNEA could not be blocked by DPCPX and because the potent A3 receptor antagonist BW A1433 blocked protection from ischaemic preconditioning, these data indicate that the protection of preconditioning is not exclusively mediated by the adenosine A1 receptor in rabbit heart and could involve the A3 receptor.


Subject(s)
Myocardial Infarction/prevention & control , Myocardial Ischemia/metabolism , Receptors, Purinergic P1/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Antihypertensive Agents/pharmacology , Female , In Vitro Techniques , Male , Perfusion , Purinergic Antagonists , Rabbits , Theophylline/analogs & derivatives , Theophylline/pharmacology , Xanthines/pharmacology
13.
Naunyn Schmiedebergs Arch Pharmacol ; 349(6): 644-50, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7969516

ABSTRACT

2-(Ar)alkoxyadenosines, which are agonists selective for the A2AAR in PC 12 cell and rat striatum membranes, are also agonists at the A2AR coupled to adenylate cyclase (AC) that mediates the inhibition of platelet aggregation. A panel of twelve well-characterized adenosine analogues stimulated human platelet AC and inhibited ADP-induced platelet aggregation at sub- to low-micromolar concentrations with a potency ranking CGS 21680 > adenosine > R-PIA. There were significant correlations between the EC50 of anti-aggregatory activity and either the EC50 of stimulation of platelet and PC 12 cell AC (r2 = 0.66 and 0.67, respectively) or the Ki of inhibition of [3H]NECA binding to the rat striatum membranes (r2 = 0.75). Likewise, platelet AC stimulation correlated well with stimulation of PC 12 cell AC and with [3H]NECA binding (r2 = 0.94 and 0.91, respectively). Ten 2-(ar)alkoxyadenosines stimulated platelet AC at EC50s ranging between 0.16 and 2.3 microM and inhibited platelet aggregation at EC50s ranging between 2 and 30 microM. There were no correlations between the EC50s of anti-aggregatory activity and either the EC50s of the stimulation of platelet or PC 12 AC (r2 = 0.08 and 0.06, respectively) or with the Ki of the inhibition of [3H]NECA binding to the A2aAR in rat striatum (r2 = 0.02). The EC50s of the stimulation of platelet AC correlated with those of the stimulation of PC 12 AC (r2 = 0.48), and also with the Ki of [3H]NECA binding (r2 = 0.71). Each of the 23 adenosines completely inhibited platelet aggregation and thus, functionally, all behaved as full agonists.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Adenosine/analogs & derivatives , Adenosine/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Purinergic P1 Receptor Agonists , Adenosine/pharmacokinetics , Adenosine-5'-(N-ethylcarboxamide) , Adenylyl Cyclases/metabolism , Animals , Blood Platelets/drug effects , Blood Platelets/enzymology , Cell Membrane/drug effects , Cell Membrane/enzymology , Humans , In Vitro Techniques , Neostriatum/drug effects , Neostriatum/metabolism , PC12 Cells , Rats , Vasodilator Agents/pharmacology
14.
J Mol Cell Cardiol ; 26(3): 303-11, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8028014

ABSTRACT

Our aim was to determine whether adenosine A1 receptor-mediated protection could be maintained for a prolonged period of time by a continuous infusion of an A1-selective agonist. To produce myocardial infarction a branch of the left coronary artery of rabbit hearts was occluded for 30 min and reperfused for 3 h. Infarct size was determined with tetrazolium staining. Prior to the 30 min ischaemia, rabbits were subjected to one of the following six protocols: (1) 6 h i.v. saline infusion; (2) 6 h i.v. CCPA (0.043 mg/kg/h) infusion; (3) 72 h saline infusion; (4) 72 h CCPA infusion; (5) 72 h CCPA infusion plus preconditioning with 5 min ischaemia followed by 10 min reperfusion; (6) 72 h saline infusion plus preconditioning. The 6 h CCPA infusion group had significantly smaller infarct sizes than the 6 h vehicle group. 16.2 +/- 2.9% infarction of the ischaemic region v 39.5 +/- 2.6%, P < 0.01. Infarction in the 72 h CCPA infusion group (37.7 +/- 2.7%) was the same as in the 72 h vehicle group (35.2 +/- 3.1%). Ischaemic preconditioning could not limit infarct size in 72 h CCPA animals (%infarction; 29.1 +/- 4.6%) but did protect animals given vehicle for 72 h (8.4 +/- 1.2%, P < 0.01). After 72 h infusion of CCPA, both the cardioprotective effect of adenosine A1-selective agonist and ischaemic preconditioning were attenuated. These findings indicate that: (1) the myocytes become desensitized to the protective effect of CCPA with prolonged exposure; and (2) ischaemic preconditioning is no longer protective when tachyphylaxis to CCPA occurs.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/physiology , Hemodynamics/drug effects , Muscle Proteins/antagonists & inhibitors , Myocardial Infarction/prevention & control , Myocardial Ischemia/physiopathology , Purinergic P1 Receptor Antagonists , Acetylcholine/pharmacology , Adenosine/administration & dosage , Adenosine/blood , Adenosine/pharmacology , Adenosine/therapeutic use , Animals , Bradycardia/chemically induced , Drug Tolerance , Female , Hypotension/chemically induced , Infusions, Intravenous , Male , Muscle Proteins/physiology , Myocardial Infarction/pathology , Myocardium/pathology , Necrosis , Rabbits , Receptors, Muscarinic/drug effects , Receptors, Purinergic P1/physiology
15.
Eur J Pharmacol ; 243(1): 35-8, 1993 Oct 12.
Article in English | MEDLINE | ID: mdl-8253122

ABSTRACT

We examined the ability of four 2-(ar)alkoxyadenosines (2-(2-phenylethoxy)adenosine, PEA; 2-[2-(2-naphthyl)ethoxy]adenosine, NEA; 2-[2-(4-methylphenyl)ethoxy]adenosine, mPEA; 2-(1-hexyloxy)adenosine, HOA) to relax porcine coronary artery in vitro. All four compounds produced concentration-dependent relaxations in rings contracted with 30 mM KCl. The EC25 values are as follows (x 10(-9) mol/l): CGS21680, (2-[p-(2-carboxyethyl)phenethylamino]-5'-N-ethylcarboxamidoadenosi ne) (32.7) approximately NECA, 5'-N-ethylcarboxamidoadenosine (51.4) approximately mPEA (74.3) approximately NEA (160.7) > HOA (855.1) approximately PEA (1259) approximately 2-chloroadenosine (1871) > adenosine (9705). However, EC75 values for all the compounds except adenosine and 2-chloroadenosine converged to a range of 8.16 to 22.86 microM, suggesting a biphasic response. Furthermore, the responses were found to be independent of endothelial integrity. The unselective adenosine receptor antagonist 8-p-sulphophenyltheophylline (100 microM) attenuated the relaxant response to NEA (EC25 = 1172 nM), suggesting that adenosine receptors mediated relaxation. Structure-activity correlations suggest that the adenosine A2 receptor in porcine coronary artery contains a region of limited bulk tolerance juxtaposed to the region occupied by adenine C-2 and distal to that a large hydrophobic region.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/pharmacology , Coronary Vessels/drug effects , Receptors, Purinergic P1/physiology , Animals , Coronary Vessels/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , In Vitro Techniques , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Structure-Activity Relationship , Swine
16.
Naunyn Schmiedebergs Arch Pharmacol ; 347(5): 521-6, 1993 May.
Article in English | MEDLINE | ID: mdl-8321327

ABSTRACT

Experiments employing guinea pig heart Langendorff preparations compared the coronary vasoactivity of a functionalized congener of adenosine, 2-[(2-aminoethyl-aminocarbonylethyl)phenylethylamino]-5'-N-e thyl- carboxamidoadenosine, APEC, with the vasoactivity of the product of the reaction of APEC with 1,4-phenylene-diisothiocyanate, 4-isothiocyanatophenylaminothiocarbonyl-APEC (DITC-APEC). Previous experiments showed that whereas APEC binds reversibly to the A2A adenosine receptor of brain striatum, DITC-APEC binds irreversibly. APEC caused concentration-dependent coronary vasodilation that persisted unchanged when agonist administration continued for up to 165 min, but promptly faded when the agent was withdrawn. The unselective adenosine receptor antagonist 8-(4-sulfophenyl)theophyline (8-SPT) antagonized the vasoactivity of APEC. By contrast, DITC-APEC (0.125-1.0 nM) caused progressive, concentration-independent vasodilation that persisted unchanged for as long as 120 min after the agent was stopped and that was insensitive to antagonism by subsequently applied 8-SPT. However, perfusion of the heart with buffer containing 0.1 mM 8-SPT strongly antagonized the coronary vasodilatory action of DITC-APEC given subsequently. Such observations indicate that the covalent binding of DITC-APEC causes irreversible activation of the guinea pig coronary artery A2A adenosine receptor. Neither APEC nor DITC-APEC appeared to desensitize the coronary adenosine receptor during two or more hours of exposure to either agonist.


Subject(s)
Adenosine/analogs & derivatives , Coronary Vessels/metabolism , Phenethylamines/metabolism , Receptors, Purinergic/metabolism , Thiocyanates/metabolism , Adenosine/antagonists & inhibitors , Adenosine/metabolism , Adenosine/pharmacology , Animals , Blood Flow Velocity/drug effects , Coronary Vessels/drug effects , Coronary Vessels/physiology , Guinea Pigs , Isothiocyanates , Phenethylamines/antagonists & inhibitors , Phenethylamines/pharmacology , Theophylline/analogs & derivatives , Theophylline/pharmacology , Thiocyanates/antagonists & inhibitors , Thiocyanates/pharmacology , Vasodilation/drug effects
17.
J Pharmacol Exp Ther ; 265(1): 248-53, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8474010

ABSTRACT

Substituting a methyl group for the ribose moiety of N6-substituted adenosines that are selective agonists at the adenosine A1 receptor creates antagonists that are A1-selective. Inasmuch as 2-phenylethoxyadenosine is a selective agonist for the adenosine A2 receptor, 2-phenylethoxy-9-methyl-adenine (PEMA) was synthesized and tested as a potential adenosine A2 receptor antagonist. In guinea pig hearts, PEMA antagonized with the same potency (pKB approximately 6.1) the A1-mediated negative dromotropic and inotropic actions and the A2-mediated coronary vasoactivity of the nonselective adenosine receptor agonist 5'-N-ethylcarboxamidoadenosine (NECA). PEMA at concentrations up to 30 microM did not antagonize the NECA-induced relaxations in guinea pig aortic rings. At concentrations exceeding 10 microM, PEMA caused xanthine-insensitive relaxations of both the aorta and the coronary vessels. Pharmacological resultant analysis revealed A2 receptor antagonism by PEMA in the guinea pig aorta (pKB = 5.2). The nonselective adenosine receptor antagonist 8-p-sulfophenyl-theophylline antagonized NECA responses in all four assays with equal potency (pKB approximately 5.7). Thus, PEMA does not discriminate between A2 receptors in the coronary vessels and A1 receptors in the atria of the guinea pig, but it is 10-fold more potent at antagonizing the A2 receptor in coronaries than the A2 receptors in the aorta. The data suggest that the A2 receptors in the coronary vasculature may be of the A2a subtype, whereas those in the aorta may be of the A2b subtype.


Subject(s)
Adenine/analogs & derivatives , Aorta/drug effects , Coronary Vessels/drug effects , Purinergic Antagonists , Adenine/pharmacology , Adenosine/analogs & derivatives , Adenosine/antagonists & inhibitors , Adenosine/pharmacology , Adenosine-5'-(N-ethylcarboxamide) , Animals , Aorta/metabolism , Coronary Vessels/metabolism , Guinea Pigs , Heart Atria/drug effects , Heart Atria/metabolism , In Vitro Techniques , Male , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Vasodilator Agents/pharmacology
18.
Pharmacology ; 46(2): 91-100, 1993.
Article in English | MEDLINE | ID: mdl-8441759

ABSTRACT

A series of 55 2-alkyloxy-, 2-aryloxy- and 2-aralkyloxy-adenosines was screened as inhibitors of the binding of [3H]R-phenyl-isopropyladenosine to A1 adenosine receptors in rat cerebral cortical membranes, and of the binding of [3]N-ethylcarboxamidoadenosine to A2 adenosine receptors in rat striatal membranes and as agonists at A2 adenosine receptors coupled to adenylate cyclase in rat pheochromocytoma PC12 cell membranes. The activities are consonant with a hydrophobic binding site in the A2 receptors at a distance from the 2-position of the adenine ring corresponding to a spacer chain of -O-CH2-CH2-. These is little lateral steric tolerance in the region occupied by the spacer chain. Interaction with the hydrophobic binding site is greatest in the 2-alkyloxy series for 2-cyclohexylethoxy-, 2-cyclohexylpropoxy- and 2-cyclohexylbutoxyadenosines and in the 2-aralkoxy series for 2-phenylethoxy-, 2-(4-methylphenyl)ethoxy-, 2-(4-chlorophenyl)ethoxy-, and 2-naphthylethoxy-adenosine. The affinities of the 2-substituted adenosines for the rat cerebral cortical A1 receptors are not as markedly altered by structural changes and are in almost all cases two- to hundredfold less than the affinity of the 2-substituted adenosine for the rat striatal A2 receptor. There is excellent correspondence of the present data on rat A2 receptors with reported potencies of these 2-substituted adenosines as coronary vasodilators in guinea pig heart preparations.


Subject(s)
Adenosine/analogs & derivatives , Adenylyl Cyclases/metabolism , Receptors, Purinergic/drug effects , Adenosine/chemistry , Adenosine/pharmacology , Animals , Cell Membrane/metabolism , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Guinea Pigs , Myocardium/metabolism , PC12 Cells , Radioligand Assay , Rats , Species Specificity , Structure-Activity Relationship
19.
Circ Res ; 72(1): 194-205, 1993 Jan.
Article in English | MEDLINE | ID: mdl-8380261

ABSTRACT

In this study, the hypothesis that mammalian ventricular myocytes possess A2-adenosine receptors was tested. Electrophysiological, contractile, and cAMP responses to the selective A2-adenosine receptor agonists 2-[2-(4-methylphenyl)ethoxy]adenosine (WRC-0090) and 2-(2-cyclohexylethoxy)adenosine (WRC-0013) and the nonselective adenosine receptor agonist 5'-(N-ethylcarboxamido)adenosine (NECA) were measured using ventricular myocytes isolated from guinea pig, rabbit, and rat hearts. Pertussis toxin pretreatment and/or the selective A1-adenosine receptor antagonists 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) and (+/-)N6-endonorbornan-2-yl-9-methyladenine (N-0861) were used to prevent activation of A1-adenosine receptors in these cells. Action potential duration at 50% repolarization was not altered by WRC-0090, NECA, or WRC-0013 with or without 0.1 microM DPCPX or pertussis toxin pretreatment, and WRC-0090 and NECA failed to prolong the action potential duration of myocytes exposed to 0.1 or 1 microM forskolin. WRC-0090 alone or with 0.1 microM DPCPX did not increase the amplitude of shortening of pertussis toxin-treated or untreated myocytes, and WRC-0090 or NECA did not significantly increase cAMP accumulation. In contrast to these results with myocytes, in the smooth muscle cell line DDT1MF-2 the effect of both selective A2-agonists on cAMP accumulation was biphasic: low concentrations (< or = 0.3 microM) increased but higher concentrations decreased accumulation of cAMP. The decreased cAMP accumulation seen at higher agonist concentrations was completely abolished by either 0.1 microM DPCPX or pretreatment of cells with pertussis toxin. In summary, the results of the present study do not provide evidence for A2-adenosine receptors on mammalian ventricular cardiomyocytes but confirm reports of the coexistence of both A1 and A2 subtypes of adenosine receptors on DDT1MF-2 cells.


Subject(s)
Adenosine/analogs & derivatives , Cyclic AMP/metabolism , Heart/drug effects , Myocardium/metabolism , Receptors, Purinergic/analysis , Action Potentials/drug effects , Adenosine/pharmacology , Adenosine-5'-(N-ethylcarboxamide) , Animals , Cell Line/drug effects , Colforsin/pharmacology , Guinea Pigs , Rabbits , Rats
20.
J Med Chem ; 35(24): 4562-6, 1992 Nov 27.
Article in English | MEDLINE | ID: mdl-1469688

ABSTRACT

This study aimed at the development of 2-(N'-aralkylidenehydrazino)adenosines as coronary vasodilators. The reaction of aromatic aldehydes or ketones with 2-hydrazinoadenosine in refluxing methanol formed the target compounds 2-27 as crystalline products in good yields. Two kinds of receptors mediate the actions of adenosine on the heart. Retardation of impulse conduction through the atrioventricular node, the negative dromotropic action, is an example of adenosine's action at an A1 receptor (A1AR) and coronary vasodilation reflects adenosine's action at an A2 receptor (A2AR). Accordingly, bioassays employing guinea pig heart Langendorff preparations assessed the selectivity of 2-27 as coronary vasodilators. Analogues 2-27 were weak negative dromotropic agents; the EC50 of the most active analogue, 2-[N'-(1-naphthylmethylene)hydrazino]-adenosine, 23, was 0.8 microM, several orders of magnitude less than many A1AR agonists. Some of the analogues were quite active coronary vasodilators; 2-(N'-benzylidenehydrazino)adenosine, 2, and several of its para-substituted derivatives, namely, the fluoro (7), methyl (13), methoxy (16), and tert-butylcarbonylethyl, 31, had EC50s for coronary vasodilation in the range 1.7-3.2 nM. The selectivity ratios, EC50 (negative dromotropic)/EC50 (coronary vasodilatory), of these five analogues ranged between 5100 (analogue 31) and 43,000 (analogue 2). Phenyl ring substitutions of other kinds or at other positions, replacement of the phenyl ring by other aryl or heteroaryl groups, or the replacement of the benzylic H by a methyl group lowered coronary vasoactivity significantly. The unselective adenosine receptor antagonist 8-(p-sulfophenyl)theophylline raised the EC50 of the negative dromotropic activities of 2, 16, and 2-[N'-(2-naphthylmethylene)hydrazino]adenosine, 24, by 3-, 18-, and 7-fold, and raised the EC50s of coronary vasoactivity by 11-, 3-, and 30-fold, respectively evidence that vasoactivity was receptor-mediated.


Subject(s)
Adenosine/analogs & derivatives , Coronary Vessels/drug effects , Hydrazines/chemistry , Vasodilator Agents/chemical synthesis , Aldehydes/chemistry , Animals , Atrioventricular Node/drug effects , Atrioventricular Node/physiology , Coronary Vessels/physiology , Depression, Chemical , Electric Conductivity , Guinea Pigs , Hydrazines/pharmacology , Ketones/chemistry , Molecular Structure , Structure-Activity Relationship , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...