Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 150(3): 342-52, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17179945

ABSTRACT

BACKGROUND AND PURPOSE: The prostamides (prostaglandin-ethanolamides) and prostaglandin (PG) glyceryl esters are biosynthesized by COX-2 from the respective endocannabinoids anandamide and 2-arachidonyl glycerol. Agonist studies suggest that their pharmacologies are unique and unrelated to prostanoid receptors. This concept was further investigated using antagonists. EXPERIMENTAL APPROACH: The isolated feline iris was used as a key preparation, where prostanoid FP receptors and prostamide activity co-exist. Activity at human recombinant FP and other prostanoid receptors was determined using stable transfectants. KEY RESULTS: In the feline iris, AGN 204396 produced a rightward shift of the dose-response curves for prostamide F2alpha and the prostamide F2alpha analog bimatoprost but did not block the effects of PGF2alpha and synthetic FP receptor agonists. Studies on human recombinant prostanoid receptors confirmed that AGN 204396 did not behave as a prostanoid FP receptor antagonist. AGN 204396 exhibited no antagonism at DP and EP1-4, but was a highly effective TP receptor antagonist. Contrary to expectation, the FP receptor antagonist AL-8810 efficaciously contracted the cat iris. AGN 204396 did not affect AL-8810 induced contractions, demonstrating that AL-8810 and AGN 204396 are pharmacologically distinct. Unlike AL-8810, the ethylamide derivate of AL-8810 was not an agonist. Al-8810 did not block prostamide F2alpha activity. Finally, AGN 204396 did not block PGE2-glyceryl ester activity. CONCLUSIONS AND IMPLICATIONS: The ability of AGN 204396 to selectively block prostamide responses suggests the existence of prostamide sensitive receptors as entities distinct from receptors recognizing PGF2alpha and PGE2-glyceryl ester.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Dinoprost/analogs & derivatives , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Iris/drug effects , Oxazoles/pharmacology , Animals , Cats , Dinoprost/pharmacology , Dinoprost/physiology , Dinoprostone/physiology , Dose-Response Relationship, Drug , In Vitro Techniques , Receptors, Prostaglandin/drug effects , Recombinant Proteins
2.
J Pharmacol Exp Ther ; 309(2): 745-57, 2004 May.
Article in English | MEDLINE | ID: mdl-14757851

ABSTRACT

We investigated whether prostaglandin ethanolamides (prostamides) E(2), F(2alpha), and D(2) exert some of their effects by 1) activating prostanoid receptors either per se or after conversion into the corresponding prostaglandins; 2) interacting with proteins for the inactivation of the endocannabinoid N-arachidonoylethanolamide (AEA), for example fatty acid amide hydrolase (FAAH), thereby enhancing AEA endogenous levels; or 3) activating the vanilloid receptor type-1 (TRPV1). Prostamides potently stimulated cat iris contraction with potency approaching that of the corresponding prostaglandins. However, prostamides D(2), E(2), and F(2alpha) exhibited no meaningful interaction with the cat recombinant FP receptor, nor with human recombinant DP, EP(1-4), FP, IP, and TP prostanoid receptors. Prostamide F(2alpha) was also very weak or inactive in a panel of bioassays specific for the various prostanoid receptors. None of the prostamides inhibited AEA enzymatic hydrolysis by FAAH in cell homogenates, or AEA cellular uptake in intact cells. Furthermore, less than 3% of the compounds were hydrolyzed to the corresponding prostaglandins when incubated for 4 h with homogenates of rat brain, lung, or liver, and cat iris or ciliary body. Very little temperature-dependent uptake of prostamides was observed after incubation with rat brain synaptosomes or RBL-2H3 cells. We suggest that prostamides' most prominent pharmacological actions are not due to transformation into prostaglandins, activation of prostanoid receptors, enhancement of AEA levels, or gating of TRPV1 receptors, but possibly to interaction with novel receptors that seem to be functional in the cat iris.


Subject(s)
Amides/pharmacology , Amidohydrolases/metabolism , Arachidonic Acids/metabolism , Cannabinoid Receptor Modulators/metabolism , Prostaglandins/pharmacology , Amides/metabolism , Amidohydrolases/drug effects , Animals , Cats , Cell Line , Endocannabinoids , Ethanolamines/metabolism , Ethanolamines/pharmacology , Guinea Pigs , Humans , Hydrolysis , Iris/drug effects , Iris/physiology , Jugular Veins/drug effects , Jugular Veins/physiology , Mice , Polyunsaturated Alkamides , Prostaglandins/metabolism , Rabbits , Rats , Rats, Sprague-Dawley , Receptors, Drug/metabolism , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Recombinant Proteins/metabolism , Synaptosomes/metabolism , TRPV Cation Channels , Tumor Cells, Cultured
3.
J Pharmacol Exp Ther ; 305(2): 772-85, 2003 May.
Article in English | MEDLINE | ID: mdl-12606640

ABSTRACT

Replacement of the carboxylic acid group of prostaglandin (PG) F(2alpha) with a nonacidic moiety, such as hydroxyl, methoxy, or amido, results in compounds with unique pharmacology. Bimatoprost (AGN 192024) is also a pharmacologically novel PGF(2alpha) analog, where the carboxylic acid is replaced by a neutral ethylamide substituent. Bimatoprost potently contracted the feline lung parenchymal preparation (EC(50) value of 35-55 nM) but exhibited no meaningful activity in a variety of PG-sensitive tissue and cell preparations. Its activity seemed unrelated to FP receptor stimulation according to the following evidence. 1) Bimatoprost exhibited no meaningful activity in tissues and cells containing functional FP receptors. 2) Bimatoprost activity in the cat lung parenchyma is not species-specific because its potent activity in this preparation could not be reproduced in cells stably expressing the feline FP receptor. 3) Radioligand binding studies using feline and human recombinant FP receptors exhibited minimal competition versus [(3)H]17-phenyl PGF(2a) for Bimatoprost. 4) Bimatoprost pretreatment did not attenuate PGF(2alpha)-induced Ca(2+) signals in Swiss 3T3 cells. 5) Regional differences were apparent for Bimatoprost but not FP agonist effects in the cat lung. Bimatoprost reduced intraocular pressure in ocular normotensive and hypertensive monkeys over a 0.001 to 0.1% dose range. A single-dose and multiple-dose ocular distribution/metabolism studies using [(3)H]Bimatoprost (0.1%) were performed. Within the globe, bimatoprost concentrations were 10- to 100-fold higher in anterior segment tissues compared with the aqueous humor. Bimatoprost was overwhelmingly the predominant molecular species identified at all time points in ocular tissues, indicating that the intact molecule reduces intraocular pressure.


Subject(s)
Dinoprost/analogs & derivatives , Glaucoma/drug therapy , Lipids/pharmacology , Amides , Animals , Bimatoprost , Calcium Signaling/drug effects , Cats , Cloprostenol/analogs & derivatives , Colon/drug effects , Dinoprost/biosynthesis , Dinoprost/pharmacology , Eye/metabolism , Female , Gastric Fundus/drug effects , Genes, Reporter/drug effects , Gerbillinae , Humans , Ileum/drug effects , In Vitro Techniques , Inositol Phosphates/metabolism , Intraocular Pressure/drug effects , Lipids/pharmacokinetics , Luciferases/genetics , Mice , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Rats , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin/biosynthesis
4.
Br J Pharmacol ; 130(8): 1933-43, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10952685

ABSTRACT

Replacement of the carboxylic acid group of PGF(2alpha) with the non-acidic substituents hydroxyl (-OH) or methoxy (-OCH(3)) resulted in an unexpected activity profile. Although PGF(2alpha) 1-OH and PGF(2alpha) 1-OCH(3) exhibited potent contractile effects similar to 17-phenyl PGF(2alpha) in the cat lung parenchymal preparation, they were approximately 1000 times less potent than 17-phenyl PGF(2alpha) in stimulating recombinant feline and human FP receptors. In human dermal fibroblasts and Swiss 3T3 cells PGF(2alpha) 1-OH and PGF(2alpha) 1-OCH(3) produced no Ca(2+) signal until a 1 microM concentration was exceeded. Pretreatment of Swiss 3T3 cells with either 1 microM PGF(2alpha) 1-OH or PGF(2alpha) 1-OCH(3) did not attenuate Ca(2+) signal responses produced by PGF(2alpha) or fluprostenol. In the rat uterus, PGF(2alpha) 1-OH was about two orders of magnitude less potent than 17-phenyl PGF(2alpha) whereas PGF(2alpha) 1-OCH(3) produced only a minimal effect. Radioligand binding studies on cat lung parenchymal plasma membrane preparations suggested that the cat lung parenchyma does not contain a homogeneous population of receptors that equally respond to PGF(2alpha)1-OH, PGF(2alpha)1-OCH(3), and classical FP receptor agonists. Studies on smooth muscle preparations and cells containing DP, EP(1), EP(2), EP(3), EP(4), IP, and TP receptors indicated that the activity of PGF(2alpha) 1-OH and PGF(2alpha) 1-OCH(3) could not be ascribed to interaction with these receptors. The potent effects of PGF(2alpha) 1-OH and PGF(2alpha) 1-OCH(3) on the cat lung parenchyma are difficult to describe in terms of interaction with the FP or any other known prostanoid receptor.


Subject(s)
Dinoprost/analogs & derivatives , Dinoprost/chemistry , Dinoprost/pharmacology , 3T3 Cells , Animals , Binding, Competitive/drug effects , COS Cells , Calcium/metabolism , Cats , Cell Line , DNA, Recombinant , Dose-Response Relationship, Drug , Female , Guinea Pigs , Humans , In Vitro Techniques , Mice , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Prostaglandin D2/metabolism , Prostaglandins F, Synthetic/pharmacology , Rabbits , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Epoprostenol , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP1 Subtype , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP3 Subtype , Receptors, Prostaglandin E, EP4 Subtype , Receptors, Thromboxane/metabolism , Structure-Activity Relationship
5.
J Biol Chem ; 275(38): 29907-14, 2000 Sep 22.
Article in English | MEDLINE | ID: mdl-10893233

ABSTRACT

Prostaglandin F(2 alpha) (PGF(2 alpha)) receptors are G-protein-coupled receptors consisting of two alternative mRNA splice variants, named FP(A) and FP(B). As compared with the FP(A) isoform, the FP(B) isoform lacks the last 46 amino acids of the carboxyl terminus and, therefore, represents a truncated version of the FP(A). We recently found (Pierce, K. L., Fujino, H., Srinivasan, D., and Regan, J. W. (1999) J. Biol. Chem. 274, 35944-35949) that stimulation of both isoforms with PGF(2 alpha) leads to activation of a Rho signaling pathway, resulting in tyrosine phosphorylation of p125 focal adhesion kinase, formation of actin stress fibers, and cell rounding. Although the activation of Rho and subsequent cell rounding occur at a similar rate for both isoforms, we now report that following the removal of PGF(2 alpha) the reversal of cell rounding is much slower for cells expressing the FP(B) isoform as compared with the FP(A) isoform. Thus, in HEK-293 cells that stably express the FP(A) isoform, the reversal of cell rounding appears to be complete after 1 h, whereas for FP(B)-expressing cells there is essentially no reversal even after 2 h. Similarly, the disappearance of stress fibers and dephosphorylation of p125 focal adhesion kinase following removal of agonist are much slower in FP(B)-expressing cells than in FP(A)-expressing cells. The mechanism of this differential reversal appears to involve a difference in receptor resensitization following the removal of agonist. Based upon whole cell radioligand binding, agonist-induced stimulation of inositol phosphate formation, and mobilization of intracellular Ca(2+), the FP(B) isoform resensitizes more slowly than the FP(A) isoform. These findings suggest that the carboxyl terminus of the FP(A) is critical for resensitization and that the slower resensitization of the FP(B) isoform leads to prolonged signaling. This differential signaling distinguishes the FP(A) and FP(B) receptor isoforms and could be important toward understanding the physiological actions of PGF(2 alpha).


Subject(s)
Cell Size/physiology , Receptors, Prostaglandin/physiology , Calcium/physiology , Cell Line , Humans , Protein Isoforms/physiology , Signal Transduction/physiology
6.
J Ocul Pharmacol Ther ; 13(4): 303-12, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9261766

ABSTRACT

The pharmacological activity of two novel thromboxane A2 (TxA2)-mimetics, AGN191976 and AGN192093, was investigated in vitro, using standard organ bath assays and human platelets, to determine potency and selectivity at various prostanoid (PG-) receptors. The effects of these compounds on intraocular pressure in Beagle dogs were then compared with U-46619, a widely employed and structurally different TP-receptor agonist. AGN191976 and AGN192093 were highly potent TP-receptor agonists in the rat aorta (EC50 of 0.32 and 1.3 nM, respectively) and human myometrium. Both compounds were approximately 10 to 50 fold more potent than U-46619. These contractile responses could be blocked with a potent TP-receptor antagonist, SQ29548. In human platelets, AGN191976 (EC50 = 16.3 nM) and U-46619 (EC50 = 538.3 nM) potently stimulated aggregation (TP-receptor mediated effect), whereas AGN192093 was a much weaker agonist (EC50 = 37.9 microM). AGN192093 was not a partial agonist in platelets, since it did not antagonize aggregation induced by AGN191976, U-46619, arachidonic acid or ADP. These results provide evidence for a subdivision of TP-receptors, and AGN192093 appears to be able to distinguish between TP-receptors in smooth muscle and platelets. In the Beagle dog eye, both AGN191976 and AGN192093 were highly potent and efficacious ocular hypotensives. Single 2.5 micrograms doses of drug decreased IOP by 11.4 (AGN191976) and 7.7 mm Hg (AGN192093) relative to the contralateral control eye. In contrast, U-46619 did not lower IOP. AGN191976, but not U-46619, increased outflow facility in these animals, which is consistent with their effects on IOP. Neither compound caused miosis which is FP-receptor mediated in the dog. These studies suggest the existence of heterogeneous populations of TP-receptors. AGN191976 and AGN192093, two novel TP-receptor agonists, appear to be useful tools for the pharmacological distinction of TP-receptor subtypes.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Eye/metabolism , Muscle, Smooth/metabolism , Prostaglandin Endoperoxides, Synthetic/pharmacology , Receptors, Thromboxane/metabolism , Thromboxane A2/analogs & derivatives , Vasoconstrictor Agents/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Animals , Blood Platelets/drug effects , Capillary Permeability/drug effects , Cats , Chickens , Conjunctiva/blood supply , Dogs , Fatty Acids, Unsaturated , Guinea Pigs , Humans , Hydrazines/pharmacology , Intraocular Pressure/drug effects , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Ocular Hypotension/chemically induced , Platelet Aggregation/drug effects , Pupil/drug effects , Rats , Receptors, Thromboxane/agonists , Receptors, Thromboxane/antagonists & inhibitors , Thromboxane A2/pharmacology
7.
Br J Pharmacol ; 117(6): 1171-80, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8882612

ABSTRACT

1. The pharmacological activity of a novel series of 9,11-cyclic carbonate derivatives of prostaglandin F2 alpha (PGF2 alpha) was investigated in various isolated smooth muscle preparations possessing different prostanoid receptor subtypes as well as in human platelets. Since subdivision of thromboxane (TP-) receptors into vascular/smooth muscle and platelet subtypes is a controversial subject, our studies included a human smooth muscle preparation (myometrium) in addition to the widely used rat aorta and human platelets as TP-receptor preparations. 2. Two members of that series, AGN191976 and AGN192093 were found to be highly potent and selective thromboxane-mimetics. AGN191976 and AGN192093 contracted isolated tissues of the rat thoracic aorta with EC50 values of 0.32 +/- 0.08 and 1.30 +/- 0.53 nM, respectively. Both agonists were at least 10 times more potent than the benchmark TP-agonist, U-46619, in this preparation, whilst being at least 500 times less potent at other prostanoid receptors (DP, EP1, EP3, FP, IP) in vitro. 3. In human myometrial strips from pregnant and non-pregnant donors, both AGN191976 and AGN192093 were potent contractile agonists. The rank order of potency in myometrium of AGN191976 > AGN192093 > U-46619 correlated well with that in the rat aorta. In human platelet-rich plasma (PRP), however, AGN191976 had potent proaggregatory activity (EC50 = 16.3 +/- 1.4 nM), which is a TP-receptor-mediated event, whereas AGN192093 was a much weaker agonist (EC50 = 37.9 +/- 2.0 microM). AGN192093 did not behave as an antagonist in the platelets, since it did not antagonize platelet aggregation induced by ADP, arachidonic acid, U-46619 or AGN191976. In human washed platelets, the activity profile of AGN191976 (EC50 = 4.15 +/- 0.52 nM) and AGN192093 (no aggregation up to 10 microM) was similar to that obtained in PRP. 4. The involvement of TP-receptors was verified with the potent TP-antagonist, SQ29548. SQ29548 (0.1 microM in myometrium; 1 microM in aorta; 1 microM and 10 microM in platelets) antagonized responses to U-46619, AGN191976 and AGN192093 as expected. 5. In conclusion, AGN191976 and AGN192093, both 9,11-cyclic carbonate derivatives of PGF2 alpha, were found to be highly potent and selective thromboxane-mimetics in rat vascular and human myometrial smooth muscle. However, only AGN 191976 was a potent agonist at TP-receptors in human platelets. The differential activity of AGN192093 on TP-receptor-mediated events in platelets and smooth muscle provides further evidence for a subdivision of TP-receptors. AGN192093 appears to be a useful tool for the pharmacological distinction of TP-receptor subtypes.


Subject(s)
Blood Platelets/drug effects , Dinoprost/pharmacology , Muscle, Smooth/drug effects , Prostaglandins F, Synthetic/pharmacology , Receptors, Thromboxane/drug effects , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Animals , Blood Platelets/metabolism , Bridged Bicyclo Compounds, Heterocyclic , Dose-Response Relationship, Drug , Fatty Acids, Unsaturated , Humans , Hydrazines/pharmacology , In Vitro Techniques , Muscle Contraction , Muscle, Smooth/metabolism , Platelet Aggregation , Prostaglandin Endoperoxides, Synthetic/pharmacology , Thromboxane A2/analogs & derivatives , Thromboxane A2/pharmacology , Vasoconstrictor Agents/pharmacology
8.
J Pharmacol Exp Ther ; 273(1): 285-7, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7714778

ABSTRACT

The rat colon and Swiss 3T3 cells have been proposed as FP receptor preparations. However, the rank orders of potency for contraction of the rat colon and Ca++ signaling in Swiss 3T3 cells were found to be disparate. Although both appeared to be FP receptor preparations in that PGF2 alpha and FP receptor selective analogs were the most potent agonists, the potency ranking for other PGs and their analogs differed markedly. This presented two alternative major hypotheses for interpreting these data: (1) Swiss 3T3 cells and the rat colon possess different FP receptor subtypes and (2) the rat colon contains a heterogeneous population of prostanoid receptors. To further characterize prostanoid receptor populations in these two preparations, radioligand binding studies were performed with 3H-PGE2 and 3H-17-phenyl-PGF2 alpha. The rank order of potency for inhibition of 3H-PGE2 binding in the rat colon was consistent with EP3 receptor pharmacology. Thus, MB 28767, sulprostone and PGE2 were potent inhibitors, whereas PGF2 alpha, PGD2 and other analogs were substantially less potent. The rank order of potency for inhibition of 3H-17-phenyl-PGF2 alpha binding in the rat colon was consistent with the presence of an FP receptor. Thus, the potency rank order for the natural PGs was PGF2 alpha > PGD2 > PGE2 and among the synthetic analogs only PGF2 alpha analogs were potent competitors. In Swiss 3T3 cells an identical rank order of potency for eliciting a Ca++ transient signal and inhibition of 3H-17-phenyl-PGF2 alpha binding was obtained.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Colon/chemistry , Prostaglandins/pharmacology , Receptors, Prostaglandin/classification , 3T3 Cells , Animals , Colon/drug effects , Colon/physiology , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Dinoprostone/metabolism , Female , In Vitro Techniques , Mice , Radioligand Assay , Rats , Rats, Sprague-Dawley
9.
J Ocul Pharmacol Ther ; 11(3): 203-12, 1995.
Article in English | MEDLINE | ID: mdl-8590251

ABSTRACT

The possible subdivision of thromboxane A2-sensitive (TP) receptors is currently a controversial subject. We report herein on a novel thromboxane A2 mimetic, AGN 191976, which has almost identical pharmacological activity to the well-characterized prostaglandin H2/thromboxane A2 (PGH2/TxA2) mimetic U-46619, but its effects on intraocular pressure are quite distinct from U-46619. Prostanoid receptor activity was determined in vitro using different smooth muscle assays and platelets. Intraocular pressure was measured tonometrically in ocular normotensive Beagle dogs and Cynomolgus monkeys. Conjunctival microvascular permeability was determined in guinea pigs. Despite closely resembling U-46619 as a potent and selective TP receptor agonist, AGN 191976 was a potent ocular hypotensive in dogs and monkeys whereas U-46619 did not lower IOP in either species. The ocular hypotensive effect of AGN 191976 in dogs was attenuated by pretreatment with the TP receptor antagonist SQ 29548. Thus, the ocular hypotensive effects of AGN 191976 are consistent with TP receptor stimulation. Both TxA2-mimetics caused plasma leakage in the guinea pig conjunctiva. The disparate activities of U-46619 and AGN 191976 in our studies suggest the existence of heterogeneous populations of TP-receptors in the eye.


Subject(s)
Antihypertensive Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Intraocular Pressure/drug effects , Ocular Hypertension/chemically induced , Receptors, Thromboxane/metabolism , Thromboxane A2/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Aged , Animals , Blood Platelets/drug effects , Capillary Permeability/drug effects , Conjunctiva/blood supply , Dogs , Fatty Acids, Unsaturated , Female , Guinea Pigs , Humans , Hydrazines/pharmacology , Macaca fascicularis , Male , Muscle, Smooth/drug effects , Ophthalmic Solutions , Platelet Aggregation/drug effects , Prostaglandin Endoperoxides, Synthetic/pharmacology , Pupil/drug effects , Receptors, Thromboxane/antagonists & inhibitors , Thromboxane A2/analogs & derivatives , Thromboxane A2/pharmacology , Tonometry, Ocular , Vasoconstrictor Agents/pharmacology
10.
Br J Ophthalmol ; 78(7): 560-7, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7918269

ABSTRACT

A novel series of prostaglandin F2 alpha (PGF2 alpha) prodrugs, with acyl ester groups at the 9, 11, and 15 positions, was prepared in order to design clinically acceptable prostaglandins for treating glaucoma. Studies involving isolated esterases and ocular tissue homogenates indicated that 9-acyl esters cannot provide a prodrug since PGF2 alpha would not be formed as a product. In contrast, 11-mono, 15-mono, and 11, 15-diesters were converted to PGF2 alpha in ocular tissues and could, therefore, be considered as prodrugs of PGF2 alpha. Carboxylesterase (CE) appeared critically important for the hydrolytic conversion of those PGF2 alpha prodrugs where the 11 or 15-OH group was esterified and such prodrugs were not substrates for acetylcholinesterase (ACHE) or butyrylcholinesterase (BuCHE). The enzymatic hydrolysis of PGF2 alpha-1-isopropyl ester was also investigated for comparative purposes. This PGF2 alpha prodrug was a good substrate for CE, but was also hydrolysed by BuCHE, albeit at a much slower rate. The most striking feature of the enzymatic hydrolysis of PGF2 alpha-1-isopropyl ester in ocular tissue homogenates was that it was much faster than for prodrugs esterified at the 11 and/or 15 positions. In terms of ocular hypotensive activity, all prodrugs which showed detectable conversion to nascent PGF2 alpha were potent ocular hypotensives. Although no separation of ocular hypotensive and ocular surface hyperaemic effects was apparent for PGF2 alpha-1-isopropyl ester, a temporal separation of these effects was apparent for the novel PGF2 alpha ester series. This difference may reflect an unfavourably rapid conversion of PGF2 alpha-1-isopropyl ester in ocular surface tissues compared with anterior segment tissues.


Subject(s)
Dinoprost/metabolism , Drug Design , Prodrugs/metabolism , 3T3 Cells , Animals , Calcium/metabolism , Dinoprost/chemistry , Eye/metabolism , Female , Glaucoma , Hydrolysis , Hyperemia , Intraocular Pressure , Male , Mice , Prodrugs/chemistry , Rabbits
11.
Prostaglandins ; 46(4): 371-83, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8248550

ABSTRACT

The physiological significance of the formation of large quantities of 19(R)-hydroxy prostaglandin E's (19-OH PGE) from PGE1 and PGE2 in human seminal plasma is intriguing. The concept that prostaglandins exert their biological effects by interacting with specific receptors, according to the current working classification for prostanoid receptors, was employed as a conceptual framework to re-examine the activity of 19(R)-OH PG's. In contrast to PGE2, which may indiscriminately stimulate a variety of prostanoid receptor subtypes, 19(R)-OH PGE2 exhibited selectivity for the EP2-receptor subtype. In EP1 (guinea pig ileum contraction), EP2 (cat trachea relaxation), and EP3 (chick ileum contraction) preparations where PGE2 is equipotent, 19(R)-OH PGE2 exhibited greater potency in the EP2-receptor population. Moreover, unlike PGE2, 19(R)-OH PGE2 did not stimulate an FP-receptor preparation (cat iris). 19(R)-OH PGE2 was devoid of activity at thromboxane A2-(TP), prostaglandin D2-(DP) and prostacyclin-(IP) sensitive receptors as indicated by its inability to cause human platelet aggregation or inhibit ADP-induced platelet aggregation. 19(R)-OH PGE1 had an entirely converse profile of activity. As a myotropic agent in the guinea pig and chick ileal preparations, 19(R)-OH PGE1 was approximately 1.5 orders of magnitude more potent than 19(R)-OH PGE2 but it appeared devoid of EP2-receptor stimulant properties. 19(R)-OH PGF2 alpha possessed very little biological activity in a diverse variety of isolated tissue preparations, indicating that 19-hydroxylation represents a highly efficient inactivation step for PGF2 alpha. The implications of the formation of receptor selective PGE derivatives in human seminal fluid for human reproductive physiology remains to be established.


Subject(s)
Dinoprostone/analogs & derivatives , Receptors, Prostaglandin E/physiology , Adenosine Diphosphate/pharmacology , Animals , Cats , Chickens , Dinoprostone/pharmacology , Guinea Pigs , Humans , Ileum/drug effects , Ileum/physiology , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Platelet Aggregation/drug effects , Receptors, Prostaglandin E/drug effects , Trachea/drug effects , Trachea/physiology
12.
Eur J Pharmacol ; 230(3): 327-33, 1993 Jan 19.
Article in English | MEDLINE | ID: mdl-8440310

ABSTRACT

Prostaglandin D2 (PGD2) and the selective DP receptor agonist BW 245C have been previously shown to lower intraocular pressure in rabbits, while PGD2, but not BW 245C, caused plasma extravasation, eosinophil infiltration, and goblet cell depletion. In these present studies definition of the ocular pharmacology of prostaglandin D2 (PGD2) has been extended by using a further selective DP receptor agonist SQ 27986 and a potent and selective DP receptor antagonist BW A868C. In cats and rabbits SQ 27986 caused ocular hypotension. The ocular hypotensive effect of PGD2 in rabbits was blocked by pretreatment with the DP receptor antagonist BW A868C, whereas the activities of PGE2 and PGF2 alpha remained unaltered. The singular involvement of the DP receptor in changes in rabbit intraocular pressure evoked by PGD2 was thereby verified by using the antagonist BW A868C. In terms of effects on the ocular surface, SQ 27986 caused no increase in conjunctival microvascular permeability, no eosinophil infiltration, and no depletion of the goblet cell population. These findings reinforce the concept that selective DP receptor agonists may be useful for lowering intraocular pressure without causing ocular surface pathology. PGD2 induced increases in conjunctival microvascular permeability were inhibited by BW A868C, despite the fact that DP receptor agonists failed to evoke a plasma exudation response. This finding was unexpected and suggests a possible subdivision of the DP receptor designation.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic , Eye/drug effects , Prostaglandin D2/pharmacology , Receptors, Immunologic , Receptors, Prostaglandin/drug effects , Animals , Bridged Bicyclo Compounds/pharmacology , Capillary Permeability/drug effects , Cats , Conjunctiva/blood supply , Dinoprost/pharmacology , Dinoprostone/pharmacology , Fatty Acids, Unsaturated/pharmacology , Female , Guinea Pigs , Hydantoins/pharmacology , Intraocular Pressure/drug effects , Male , Rabbits
13.
Invest Ophthalmol Vis Sci ; 31(11): 2463-6, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2173691

ABSTRACT

Previous studies on prostanoid-induced blood-aqueous barrier disruption have been predominantly restricted to studies on natural prostanoids and the highly selective DP-receptor agonist BW245C, which was inactive. The pharmacology of prostanoid-induced blood-aqueous barrier breakdown in rabbits has been extended in these studies by determining the activity of additional prostanoid receptor agonists which are relatively selective according to the current classification for prostanoid receptors. Prostanoid effects were evaluated by measuring plasma albumin leakage into the aqueous humor as an indicator of blood-aqueous barrier breakdown. EP1 and EP3 receptor subtype agonists were ineffective in altering the blood-aqueous barrier, but the relatively selective EP2 agonist 11-deoxy prostaglandin E1 had a dose-dependent disruptive effect on the blood-aqueous barrier. In contrast, FP, IP and TP receptor agonists were ineffective in disrupting the blood-aqueous barrier. Taken together, these data imply that the EP2 receptor subtype, as a singular entity, may account for the rapid disruption of the rabbit blood-aqueous barrier that typically occurs in response to prostanoids.


Subject(s)
Aqueous Humor/drug effects , Capillary Permeability/drug effects , Prostaglandins/pharmacology , Receptors, Prostaglandin/drug effects , Animals , Biological Transport/drug effects , Dinoprostone/analogs & derivatives , Female , Male , Rabbits , Receptors, Prostaglandin E
14.
Invest Ophthalmol Vis Sci ; 31(1): 138-46, 1990 Jan.
Article in English | MEDLINE | ID: mdl-2298534

ABSTRACT

Prostaglandin D2 (PGD2) exerts a variety of biologic actions in the eye; these include ocular hypotension and inflammatory effects on the conjunctiva. The profile of activity of PGD2 in ocular tissues was compared to that of BW 245C, a selective agonist for the PGD2-sensitive (DP) receptor, and to that of the biologically active metabolites of PGD2, 9 alpha,11 beta-prostaglandin F2 (9 alpha,11 beta-PGF2) and prostaglandin J2 (PGJ2). PGD2 produced a dose-dependent decrease in intraocular pressure and in the conjunctiva it caused increased conjunctival microvascular permeability, eosinophil infiltration and goblet cell depletion. Although BW 245C was equipotent to PGD2 as an ocular hypotensive agent, it did not cause pathological effects in the conjunctiva. Thus, the ocular hypotensive effect of PGD2 may be separated from inflammatory effects on the conjunctiva by employing a selective DP-receptor agonist such as BW 245C. 9 alpha,11 beta-PGF2 was a weak ocular hypotensive and did not cause conjunctival inflammation. PGJ2 produced no significant effect on intraocular pressure. PGJ2 did not elicit a microvascular permeability response in the conjunctiva, but was inflammatory in other respects and caused eosinophil infiltration and goblet cell depletion similar to PGD2. Thus, both the ocular hypotensive actions and the conjunctival pathology of PGD2 may be replicated individually by employing PGD2 analogues and metabolites.


Subject(s)
Dinoprost/pharmacology , Eye/drug effects , Intraocular Pressure/drug effects , Animals , Aqueous Humor/drug effects , Capillary Permeability/drug effects , Chemotaxis, Leukocyte/drug effects , Conjunctiva/blood supply , Conjunctiva/drug effects , Dinoprost/analogs & derivatives , Eosinophils/drug effects , Evans Blue , Female , Guinea Pigs , Hydantoins/pharmacology , Inflammation/etiology , Male , Prostaglandin D2/pharmacology , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...