Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Mol Pharmacol ; 101(1): 56-67, 2022 01.
Article in English | MEDLINE | ID: mdl-34718225

ABSTRACT

The cation channel of sperm (CatSper) is the principal entry point for calcium in human spermatozoa and its proper function is essential for successful fertilization. As CatSper is potently activated by progesterone, we evaluated a range of steroids to define the structure-activity relationships for channel activation and found that CatSper is activated by a broad range of steroids with diverse structural modifications. By testing steroids that failed to elicit calcium influx as inhibitors of channel activation, we discovered that medroxyprogesterone acetate, levonorgestrel, and aldosterone inhibited calcium influx produced by progesterone, prostaglandin E1, and the fungal natural product l-sirenin, but these steroidal inhibitors failed to prevent calcium influx in response to elevated K+ and pH. In contrast to these steroid antagonists, we demonstrated for the first time that the T-type calcium channel blocker ML218 acts similarly to mibefradil, blocking CatSper channels activated by both ligands and alkalinization/depolarization. These T-type calcium channel blockers produced an insurmountable blockade of CatSper, whereas the three steroids produced antagonism that was surmountable by increasing concentrations of each activator, indicating that the steroids selectively antagonize ligand-induced activation of CatSper rather than blocking channel function. Both the channel blockers and the steroid antagonists markedly reduced hyperactivated motility of human sperm assessed by computer-aided sperm analysis, consistent with inhibition of CatSper activation. Unlike the channel blockers mibefradil and ML218, which reduced total and progressive motility, medroxyprogesterone acetate, levonorgestrel, and aldosterone had little effect on these motility parameters, indicating that these steroids are selective inhibitors of hyperactivated sperm motility. SIGNIFICANCE STATEMENT: The steroids medroxyprogesterone acetate, levonorgestrel, and aldosterone selectively antagonize progesterone- and prostaglandin E1-induced calcium influx through the CatSper cation channel in human sperm. In contrast to T-type calcium channel blockers that prevent all modes of CatSper activation, these steroid CatSper antagonists preferentially reduce hyperactivated sperm motility, which is required for fertilization. The discovery of competitive antagonists of ligand-induced CatSper activation provides starting points for future discovery of male contraceptive agents acting by this unique mechanism.


Subject(s)
Alprostadil/antagonists & inhibitors , Azabicyclo Compounds/pharmacology , Benzamides/pharmacology , Calcium Channels/metabolism , Progesterone/antagonists & inhibitors , Steroids/pharmacology , Aldosterone/chemistry , Aldosterone/pharmacology , Dose-Response Relationship, Drug , Humans , Levonorgestrel/chemistry , Levonorgestrel/pharmacology , Male , Semen/drug effects , Semen/metabolism , Sperm Motility/drug effects , Sperm Motility/physiology , Steroids/chemistry , Structure-Activity Relationship
2.
J Clin Invest ; 76(5): 1828-36, 1985 Nov.
Article in English | MEDLINE | ID: mdl-2997290

ABSTRACT

Confluent T84 monolayers grown on permeable supports and mounted in a modified Ussing chamber secrete chloride (Cl-) in response to prostaglandin E1. The threshold stimulation was observed at 10(-9) M and a maximal effect at 10(-6) M. Unidirectional flux studies showed an increase in both serosal to mucosal and mucosal to serosal Cl- fluxes with 10(-6) M prostaglandin E1; the increase in serosal to mucosal Cl- flux exceeded the increase in mucosal to serosal flux, resulting in net Cl- secretion. Na+ transport was not affected in either direction and the changes in net Cl- flux correlated well with the changes in short circuit current. To identify the electrolyte transport pathways involved in the Cl- secretory process, the effect of prostaglandin E1 on ion fluxes was tested in the presence of putative inhibitors. Bumetanide was used as an inhibitor for the basolaterally localized Na+,K+,Cl- cotransport system whose existence and bumetanide sensitivity have been verified in earlier studies (Dharmsathaphorn et al. 1984. J. Clin. Invest. 75:462-471). Barium was used as an inhibitor for the K+ efflux pathway on the basolateral membrane whose existence and barium sensitivity were demonstrated in this study by preloading the monolayers with 86Rb+ (as a tracer for K+) and simultaneously measuring 86Rb+ efflux into both serosal and mucosal reservoirs. Both bumetanide and barium inhibited the net chloride secretion induced by prostaglandin E1 suggesting the involvement of the Na+,K+,Cl- cotransport and a K+ efflux pathways on the basolateral membrane in the Cl- secretory process. The activation of another Cl- transport pathway on the apical membrane by prostaglandin E1 was suggested by Cl- uptake studies. Our findings indicate that the prostaglandin E1-stimulated Cl- secretion, which is associated with an increase in cyclic AMP level, intimately involves (a) a bumetanide-sensitive Na+,K+,Cl- cotransport pathway that serves as a Cl- uptake step across the basolateral membrane, (b) the stimulation of a barium-sensitive K+ efflux mechanism on the basolateral membrane that most likely acts to recycle K+, and (c) the activation of a Cl- transport pathway on the apical membrane that serves as a Cl- exit pathway.


Subject(s)
Alprostadil/pharmacology , Chlorides/metabolism , Intestinal Mucosa/metabolism , Alprostadil/antagonists & inhibitors , Anthracenes/pharmacology , Barium/pharmacology , Bumetanide/pharmacology , Cell Compartmentation , Cell Line , Cyclic AMP/biosynthesis , Epithelium/metabolism , Humans , Intestinal Mucosa/drug effects , Potassium/metabolism
3.
Prostaglandins Other Lipid Mediat ; 79(1-2): 101-13, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16516814

ABSTRACT

Prostaglandins are key regulators of ion transport in the kidney. In MDCK cells, which model distal tubule cells, the transcription of the Na,K-ATPase beta1 subunit is regulated by PGE1 and PGE2. To identify the EP receptors that mediate transcriptional regulation, transient transfection studies are conducted using the human beta1promoter/luciferase construct, pHbeta1-1141 Luc. The involvement of EP1 and EP2 receptors is indicated by studies with the EP1 selective agonist 17-phenyl trinor PGE2, and the EP2 selective agonist butaprost (which stimulate), as well as by studies with the antagonists SC-51089 (EP1 specific) and AH 6809 (EP1 and EP2 specific). Consistent with the involvement of Gs coupled EP2 receptors, is that the PGE1 stimulation is inhibited by the PKAI expression vector (encoding the protein kinase A (PKA) inhibitory protein), as well as by the myristolated PKA inhibitory peptide PKI. In addition to this evidence (for the involvement of EP2 receptors), evidence for the involvement of EP1 receptors in the PGE1 mediated stimulation of Na,K-ATPase beta subunit gene transcription includes the stimulatory effect of 17-phenyl trinor PGE2, as well as the inhibitory effects of SC-51089. Also consistent with the involvement of Gq coupled EP1 receptors, the PGE1 stimulation is inhibited by the PKCI vector (encoding the PKC inhibitory domain), the PKC inhibitor Go 6976, thapsigargin, as well as the calmodulin antagonists W7 and W13.


Subject(s)
Alprostadil/metabolism , Dinoprostone/metabolism , Kidney Tubules, Distal/enzymology , Receptors, Prostaglandin E/agonists , Sodium-Potassium-Exchanging ATPase/biosynthesis , Alprostadil/antagonists & inhibitors , Animals , Cell Line , Dinoprostone/antagonists & inhibitors , Dogs , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Hydrazines/pharmacology , Ion Transport/drug effects , Ion Transport/physiology , Oxazepines/pharmacology , Prostaglandin Antagonists/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP1 Subtype , Receptors, Prostaglandin E, EP2 Subtype , Xanthones/pharmacology
4.
Biochim Biophys Acta ; 972(1): 45-53, 1988 Oct 28.
Article in English | MEDLINE | ID: mdl-2902882

ABSTRACT

Activation of human blood platelet adenylate cyclase is initiated through the binding of prostaglandin E1 to the membrane receptors. Incubation of platelet membrane with [3H]prostaglandin E1 at pH 7.5 in the presence of 5 mM MgCl2 showed that the binding of the autacoid was rapid, reversible and highly specific. The binding was linearly proportional to the activation of adenylate cyclase. Although the membrane-bound radioligand could not be removed either by GTP or its stable analogue 5'-guanylylimido diphosphate, 150 nM cyclic AMP displaced about 40% of the bound agonist from the membrane. Scatchard analyses of the binding of the prostanoid to the membrane in the presence or absence of cyclic AMP showed that the nucleotide specifically inhibited the high-affinity binding sites without affecting the low-affinity binding sites. Incubation of the membrane with 150 mM cyclic AMP and varying amounts of prostaglandin E1 (25 nM to 1.0 microM) showed that the percent removal of the membrane-bound autacoid was similar to the percent inhibition of adenylate cyclase at each concentration of the agonist. At a concentration of 25 nM prostaglandin E1, both the binding of the agonist and the activity of adenylate cyclase were maximally inhibited by 40%. With the increase of the agonist concentration in the assay mixture, the inhibitory effects of the nucleotide gradually decreased and at a concentration of 1.0 microM prostaglandin E1 the effect of the nucleotide became negligible. These results show that cyclic AMP inhibits the activation of adenylate cyclase by low concentrations of prostaglandin E1 through the inhibition of the binding of the agonist to high-affinity binding sites.


Subject(s)
Adenylyl Cyclases/metabolism , Alprostadil/antagonists & inhibitors , Blood Platelets/enzymology , Cyclic AMP/blood , Receptors, Prostaglandin/blood , Binding, Competitive , Enzyme Activation/drug effects , GTP-Binding Proteins/blood , Guanosine Triphosphate/blood , Humans , In Vitro Techniques , Kinetics , Prostaglandins/metabolism , Receptors, Prostaglandin E
5.
J Bone Miner Res ; 11(6): 789-99, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8725176

ABSTRACT

Extracellular cations have paradoxical trophic and toxic effects on osteoblast function. In an effort to explain these divergent actions, we investigated in MC3T3-E1 osteoblasts if polyvalent cations differentially modulate the agonist-stimulated cyclic adenosine monophosphate (cAMP) pathway, an important regulator of osteoblastic function. We found that a panel of cations, including gadolinium, aluminum, calcium, and neomycin, inhibited prostaglandin E1 (PGE)-stimulated cAMP accumulation but paradoxically potentiated parathyroid hormone (PTH)-stimulated cAMP production. In contrast, these cations had no effect on forskolin- or cholera toxin-induced increases in cAMP, suggesting actions proximal to adenylate cyclase and possible modulation of receptor interactions with G proteins. Phorbol 12-myristate 13-acetated (PMA) mimicked the effects of cations on PGE1- and PTH-stimulated cAMP accumulation in MC3T3-E1 cells, respectively, diminishing and augmenting the responses. Moreover, down-regulation of protein kinase C (PKC) by overnight treatment with PMA prevented gadolinium (Gd3+) from attenuating PGE1- and enhancing PTH-stimulated cAMP production, indicating involvement of PKC-dependent pathways. Cations, however, activated signal transduction pathways not coupled to phosphatidylinositol-specific phospholipase C (PI-PLC), since there was no corresponding increase in inositol phosphate formation or intracellular calcium concentrations. In addition, pertussis toxin treatment failed to prevent Gd(3+)-mediated suppression of PGE1-stimulated cAMP, suggesting actions independent of Gm. Thus, polyvalent cations may either stimulate or inhibit hormone-mediated cAMP accumulation in osteoblasts. These differential actions provide a potential explanation for the paradoxical trophic and toxic effects of cations on osteoblast function that occur in vivo under different hormonal conditions.


Subject(s)
Cations/pharmacology , Cyclic AMP/biosynthesis , Osteoblasts/metabolism , Adenylate Cyclase Toxin , Alprostadil/antagonists & inhibitors , Analysis of Variance , Cells, Cultured , Cholera Toxin/antagonists & inhibitors , Cholera Toxin/pharmacology , Colforsin/antagonists & inhibitors , Colforsin/pharmacology , Gadolinium/pharmacology , Norepinephrine/antagonists & inhibitors , Parathyroid Hormone/agonists , Pertussis Toxin , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoinositide Phospholipase C , Phosphoric Diester Hydrolases/analysis , Phosphoric Diester Hydrolases/drug effects , Protein Kinase C/physiology , Virulence Factors, Bordetella/antagonists & inhibitors , Virulence Factors, Bordetella/pharmacology
6.
FEBS Lett ; 298(2-3): 199-202, 1992 Feb 24.
Article in English | MEDLINE | ID: mdl-1312024

ABSTRACT

Stimulation of human platelets with the thromboxane A2 analogue, U46619, after treatment with prostaglandin E1 or forskolin, reduced the inhibition of ADP-evoked Mn2+ influx and the release of Ca2+ from intracellular stores. U46619 decreased the elevated concentration of 3',5'-cyclic AMP in platelets that were pretreated with prostaglandin E1. These results suggest that occupation of prostaglandin H2/thromboxane A2 receptors, like those for other agonists, inhibits adenylate cyclase activity, which can contribute to the promotion of platelet activation.


Subject(s)
Adenylyl Cyclase Inhibitors , Alprostadil/antagonists & inhibitors , Cyclic AMP/metabolism , Platelet Activation , Prostaglandin Endoperoxides, Synthetic/pharmacology , Receptors, Prostaglandin/metabolism , Thromboxane A2/metabolism , Adenosine Diphosphate/antagonists & inhibitors , Adenosine Diphosphate/physiology , Calcium/metabolism , Colforsin/pharmacology , Fura-2 , Humans , Manganese/metabolism , Receptors, Thromboxane
7.
Biochem Pharmacol ; 48(7): 1319-26, 1994 Oct 07.
Article in English | MEDLINE | ID: mdl-7945429

ABSTRACT

The effects of extracellular divalent cations on the responses of human platelets to adenosine 5'-diphosphate (ADP) and on its inhibition by the competitive antagonist adenosine 5'-triphosphate (ATP) were investigated. Two responses were studied, shape change and the inhibition of prostaglandin E1 (PGE1)-stimulated adenylate cyclase, and experiments were carried out in the presence of divalent cations (Ca2+ and Mg2+, 1 mM) or in their absence. For each response there was a small leftward shift of the concentration-response curve to ADP in the absence of divalent cations compared to that in their presence, and this leftward shift disappeared when the results were plotted in terms of ADP3- rather than total ADP concentration. The shape change results were, however, complicated by a reduction in the maximal response to ADP in the absence of divalent cations. For each response there was also a marked increase in the pA2 value of ATP in the absence of divalent cations compared to that in their presence, and this difference disappeared if the results were calculated in terms of ATP4- instead of total ATP. These results suggest that the human platelet ADP receptor, in common with other receptors for adenine nucleotides, recognises predominantly the uncomplexed forms of ADP and ATP as ligands.


Subject(s)
Adenosine Diphosphate/pharmacology , Blood Platelets/drug effects , Cations, Divalent/pharmacology , Adenosine Diphosphate/antagonists & inhibitors , Adenosine Triphosphate/pharmacology , Adenylyl Cyclase Inhibitors , Alprostadil/antagonists & inhibitors , Calcium/pharmacology , Dose-Response Relationship, Drug , Humans , Magnesium/pharmacology , Platelet Aggregation/drug effects
8.
Article in English | MEDLINE | ID: mdl-7708815

ABSTRACT

Prostaglandin E1 (PGE1) is known to possess various actions in vivo. Of these actions, the contraction of the ileum and inflammation are undesirable side effects. We previously proposed a hypothesis concerning the receptors for human blood platelet aggregation and its inhibition, and the contraction of the ileum and uterus based on a study of structure-activity relationships. If the same principle can be applied to contraction of the ileum and inflammation induced by PGE1, compounds that suppress the side effects of PGE1 can be developed. The antihistamine diphenylpyraline and the anticholinergic atropine antagonized PGE1-induced contraction of the ileum in guinea pigs. Papaverine, which is a smooth muscle relaxant, also acted as an antagonist. Gabexate mesylate (FOY), a non-peptide proteinase inhibitor, inhibits guinea pig ileum contraction induced by PGE1, but epsilon-guanidinocaproic acid (GCA), a metabolite of FOY, does not. Increased microvascular permeability of the abdominal skin in rats induced by the local injection of PGE1 and histamine was suppressed by atropine, papaverine and diphenylpyraline. FOY, not GCA, had a weak inhibitory action. We demonstrate the possibility of suppressing the side effects of PGE1 based on the results obtained in the present and previous studies.


Subject(s)
Alprostadil/adverse effects , Alprostadil/antagonists & inhibitors , Alprostadil/pharmacology , Animals , Atropine/pharmacology , Capillary Permeability/drug effects , Gabexate/pharmacology , Guanidines/pharmacology , Guinea Pigs , Histamine/pharmacology , Ileum/physiology , Inflammation/chemically induced , Male , Muscle Contraction/drug effects , Papaverine/pharmacology , Piperidines/pharmacology , Rats
9.
Prostaglandins Leukot Essent Fatty Acids ; 50(6): 299-302, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7938079

ABSTRACT

Prostaglandin E1 induced increase in cytosolic immunoreactive rap 1-B G-protein in a time and concentration dependent manner in human platelets. However, this increase was completely blocked by simultaneous treatment of platelets with phorbol 12-myristate 13-acetate (PMA).


Subject(s)
Alprostadil/pharmacology , Blood Platelets/drug effects , GTP-Binding Proteins/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Alprostadil/antagonists & inhibitors , Animals , Biological Transport/drug effects , Blood Platelets/metabolism , Cytosol/metabolism , GTP-Binding Proteins/immunology , Humans , Immune Sera , Rabbits , Signal Transduction/drug effects , rap GTP-Binding Proteins
10.
Neuropeptides ; 14(1): 39-44, 1989 Jul.
Article in English | MEDLINE | ID: mdl-2550843

ABSTRACT

Non-selective enkephalin analogs, such as [D-Ala2, Met5]enkephalinamide, have been found to increase jejunal absorption in the rat and dog after their intracerebroventricular administration. In the present investigation, experiments were designed to characterize the brain opiate receptor subtype mediating this action in the rat proximal jejunum in situ and to assess the involvement of extrinsic sympathetic nerves innervating this gut region in opioid-induced absorption. Changes in jejunal water transport were examined in rats pretreated with bolus i.c.v. doses (less than or equal to 1 microgram) of the respective mu-, delta-, or kappa-opiate agonists [D-Ala2, N-Me-Phe4, Gly5-ol]enkephalin (DAGO), [D-Pen2, D-Pen5]enkephalin or U-50488 and infused intravenously with the secretagogue prostaglandin E1 (PGE1, 5 micrograms/kg-min). In saline-pretreated rats, PGE1 produced large, time-dependent decreases in jejunal fluid absorption. Of the opiate agonists examined, only DAGO in submicrogram doses significantly inhibited PGE1 actions 60 to 90 min after its administration. Extirpation of the celiac and superior mesenteric ganglia, major sources of sympathetic neurons innervating the upper gut, significantly attenuated both the antiabsorptive actions of PGE1 and the proabsorptive actions of DAGO (0.3 micrograms, i.cv.). These results suggest that CNS mu-opiate receptors modulate jejunal absorption in the rat, an action mediated through extrinsic sympathetic nerves innervating the upper small intestine.


Subject(s)
Alprostadil/antagonists & inhibitors , Intestinal Absorption , Jejunum/metabolism , Receptors, Opioid/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer , Alprostadil/metabolism , Alprostadil/pharmacology , Animals , Body Water/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enkephalin, D-Penicillamine (2,5)- , Enkephalins/pharmacology , Intestinal Absorption/drug effects , Jejunum/drug effects , Jejunum/innervation , Kinetics , Male , Pyrrolidines/pharmacology , Rats , Rats, Inbred Strains
11.
Eur J Pharmacol ; 237(1): 131-7, 1993 Jun 11.
Article in English | MEDLINE | ID: mdl-7689467

ABSTRACT

Isolated smooth muscle cells from the circular layer of pig and guinea-pig ileum were used to study the effect of prostaglandin E2 (PGE2) and three PGE2 receptor (EP) agonists; iloprost (EP1), butaprost (EP2) and enprostil (EP3). In pig cells, PGE2 and enprostil induced cell contraction (22.1 and 21.5% shortening of cell length, obtained at 10 nM for PGE2 and 1 nM for enprostil, respectively). Iloprost and butaprost had no contractile effect. However, the cholecystokinin octapeptide (CCK-8; 10 nM)-induced contraction was inhibited when cells were preincubated with iloprost or butaprost. In guinea-pig cells, PGE2, butaprost and iloprost induced cell contraction, whereas enprostil had no effect (23.1% for 10 nM PGE2, 22.8% for 1 nM butaprost and 22.6% for 10 nM iloprost). Preincubation with SC19220 (EP1 antagonist) inhibited the PGE2-, butaprost- and iloprost-induced contractions. When the contractile effect of PGE2, butaprost and iloprost was inhibited by addition of SC19220, these agents inhibited the cell contraction induced by CCK-8 (1 nM). Smooth muscle cells from guinea-pig and pig ileum express two PGE2 receptor subtypes that induce opposite effect. EP1 and EP3 receptors mediate cell contraction in guinea-pig and pig, respectively, whereas EP2 receptors mediate cell relaxation in both species.


Subject(s)
Dinoprostone/metabolism , Muscle, Smooth/drug effects , Receptors, Prostaglandin/drug effects , Alprostadil/analogs & derivatives , Alprostadil/antagonists & inhibitors , Alprostadil/pharmacology , Animals , Dibenz(b,f)(1,4)oxazepine-10(11H)-carboxylic acid, 8-chloro-, 2-acetylhydrazide/pharmacology , Dinoprostone/antagonists & inhibitors , Dinoprostone/pharmacology , Enprostil/antagonists & inhibitors , Enprostil/pharmacology , Guinea Pigs , Ileum/cytology , Ileum/drug effects , Iloprost/antagonists & inhibitors , Iloprost/pharmacology , In Vitro Techniques , Male , Muscle Contraction/drug effects , Prostaglandins E, Synthetic/antagonists & inhibitors , Prostaglandins E, Synthetic/pharmacology , Receptors, Prostaglandin/isolation & purification , Sincalide/pharmacology , Swine
12.
Life Sci ; 39(4): 303-11, 1986 Jul 28.
Article in English | MEDLINE | ID: mdl-3016438

ABSTRACT

The role of cyclic AMP in the control of vascular smooth muscle tone was studied by monitoring the effects of prostaglandin E1 (PGE1), isoproterenol and forskolin on cyclic AMP levels and tension in rabbit aortic rings. PGE1, isoproterenol and forskolin all increased cyclic AMP levels in rabbit aortic rings. Isoproterenol and forskolin relaxed phenylephrine-contracted aortic rings, but PGE1 contracted the rings in the presence or absence of phenylephrine. Isoproterenol relaxed these PGE1-contracted aortic rings without further change in total cyclic AMP levels, which were already elevated by the PGE1 alone. Pretreatment with forskolin potentiated the effects of PGE1 on cyclic AMP levels. PGE1 caused contractions in muscles partially relaxed by forskolin, even though very large increases in cyclic AMP levels (30 fold) were produced by PGE1 in the presence of forskolin. Isoproterenol was able to relax these forskolin-treated, PGE1-contracted muscles with no further increase in cyclic AMP levels. Thus, there does not appear to be a good correlation between total tissue levels of cyclic AMP and tension in these experiments. Our results suggest that, if cyclic AMP is responsible for relaxation of smooth muscle, some form of functional compartmentalization of cyclic AMP must exist in this tissue.


Subject(s)
Alprostadil/pharmacology , Colforsin/pharmacology , Isoproterenol/pharmacology , Muscle, Smooth, Vascular/drug effects , Alprostadil/antagonists & inhibitors , Animals , Cyclic AMP/metabolism , In Vitro Techniques , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/metabolism , Phenylephrine/antagonists & inhibitors , Phenylephrine/pharmacology , Rabbits
13.
J Pharm Pharmacol ; 49(11): 1109-13, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9401947

ABSTRACT

It is well-known that enteric, secreto-motor nerves mediate cholera toxin-induced fluid secretion in the rat small intestine. This notion is, in part, derived from experiments on anaesthetized animals in which the response to cholera toxin was antagonized by the ganglionic nicotinic receptor antagonist, hexamethonium. In the current study, such anti-secretory action of ganglionic blocking compounds was analysed in an experiment designed to minimize any possible negative effect of general anaesthesia on intestinal secretion. Rats were anaesthetized with ether for 5-10 min, during which time a jejunal loop (10-12 cm) was constructed. The loop was challenged with one of the secretagogues, cholera toxin, prostaglandin E1 (PGE1) or okadaic acid. Saline (control) or either of the ganglionic blockers, hexamethonium and chlorisondamine, was administered intravenously. The rats were killed 5 h (cholera toxin) or 1.5 h (PGE1 and okadaic acid) after challenge, and the amount of fluid accumulated in the loops was determined. Cholera toxin-induced secretion was unchanged by hexamethonium but reduced by approximately 80% by chlorisondamine. The difference in effect between the two blockers might relate to the duration of ganglionic blockade. Chlorisondamine blocked secretion induced by either PGE1 or okadaic acid by approximately 60%. It is suggested that the anti-secretory effect of ganglionic blocking compounds might be a result of blockade of secreto-motor nerves but other mechanisms, for example interference with haemodynamic factors, cannot be ruled out.


Subject(s)
Alprostadil/antagonists & inhibitors , Chlorisondamine/pharmacology , Ganglionic Blockers/pharmacology , Jejunum/drug effects , Vasodilator Agents/antagonists & inhibitors , Animals , Blood Pressure/drug effects , Cholera Toxin/pharmacology , Ionophores/antagonists & inhibitors , Jejunum/metabolism , Male , Okadaic Acid/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
14.
Med Hypotheses ; 28(3): 213-4, 1989 Mar.
Article in English | MEDLINE | ID: mdl-2523509

ABSTRACT

It is suggested that the mechanism for decreased incidence of dysmenorrhea in female cigarette smokers may lie in the possible inhibition of algesic prostaglandins smoking induced stimulation of beta-endorphin, nicotine, or acrolein.


Subject(s)
Acrolein/pharmacology , Aldehydes/pharmacology , Dysmenorrhea/blood , Nicotine/pharmacology , Prostaglandin Antagonists/pharmacology , Smoking/blood , beta-Endorphin/blood , Alprostadil/antagonists & inhibitors , Female , Humans
15.
Chin J Physiol ; 34(4): 355-69, 1991.
Article in English | MEDLINE | ID: mdl-1820842

ABSTRACT

Dispersed bovine luteal cells collected on day 7 or 13 postestrus were incubated in vitro with human chorionic gonadotropin (hCG) or prostaglandins E1 or E2 (PGE1; PGE2) in the presence or absence of adenosine, dipyridamole and/or prostaglandin F2 alpha (PGF2 alpha). Secretion of progesterone by day 7 or 13 bovine luteal cells was increased by hCG, PGE1 or PGE2 which was increased further when co-incubated with adenosine (p less than 0.05). Adenosine alone increased secretion of progesterone by day 7 luteal cells (p less than 0.05) but not by day 13 luteal cells (p greater than 0.05). PGF2 alpha inhibited the gonadotropic response to hCG, PGE1 or PGE2 (p less than 0.05) which was reduced by adenosine (p less than 0.05) by day 7 or 13 bovine luteal cell suspensions. Dipyridamole reduced the adenosine-facilitated response to hCG, PGE1 or PGE2 by day 7 or 13 luteal cell suspensions (p less than 0.05). It is concluded that adenosine enhances the steroidogenic response by functional bovine luteal cells to hCG, PGE1 or PGE2 and that adenosine reduces the luteolytic response to PGF2 alpha by gonadotropin-stimulated bovine luteal cells in vitro.


Subject(s)
Adenosine/pharmacology , Chorionic Gonadotropin/pharmacology , Corpus Luteum/metabolism , Dinoprost/antagonists & inhibitors , Gonadotropins/metabolism , Alprostadil/antagonists & inhibitors , Alprostadil/pharmacology , Animals , Cattle , Corpus Luteum/cytology , Corpus Luteum/drug effects , Dinoprost/pharmacology , Dinoprostone/antagonists & inhibitors , Dinoprostone/pharmacology , Dipyridamole/pharmacology , Drug Synergism , Estrus/physiology , Female , Humans , Progesterone/metabolism
16.
Article in English | MEDLINE | ID: mdl-22858445

ABSTRACT

The chick chorioallantoic membrane (CAM) subserves gas exchange in the developing embryo and shell-less culture affords a unique opportunity for direct observations over time of individual blood vessels to pharmacologic interventions. We tested a number of lipids including prostaglandins PGE(1&2) for vascular effects and signaling in the CAM. Application of PGE(1&2) induced a decrease in the diameter of large blood vessels and a concentration-dependent, localized, reversible loss of blood flow through small vessels. The loss of flow was also mimicked by misoprostol, an agonist for 3 of 4 known PGE receptors, EP(2-4), and by U46619, a thromboxane mimetic. Selective receptor antagonists for EP(3) and thromboxane each partially blocked the response. This is a first report of the effects of prostaglandins on vasoreactivity in the CAM. Our model allows the unique ability to examine simultaneous responses of large and small vessels in real time and in vivo.


Subject(s)
Alprostadil/pharmacology , Chorioallantoic Membrane/drug effects , Dinoprostone/pharmacology , Vasoconstrictor Agents/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Alprostadil/antagonists & inhibitors , Animals , Biphenyl Compounds/pharmacology , Blood Vessels/drug effects , Blood Vessels/physiology , Bridged Bicyclo Compounds, Heterocyclic , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/metabolism , Dibenz(b,f)(1,4)oxazepine-10(11H)-carboxylic acid, 8-chloro-, 2-acetylhydrazide/pharmacology , Dinoprostone/antagonists & inhibitors , Dose-Response Relationship, Drug , Estradiol/pharmacology , Estrogens/pharmacology , Fatty Acids, Unsaturated , Hydrazines/pharmacology , Microsomes/drug effects , Microsomes/metabolism , Misoprostol/pharmacology , Prostaglandin Antagonists/pharmacology , Rats , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin/metabolism , Receptors, Thromboxane/agonists , Receptors, Thromboxane/antagonists & inhibitors , Receptors, Thromboxane/metabolism , Vasoconstriction/drug effects , Vasoconstrictor Agents/antagonists & inhibitors , Xanthones/pharmacology , alpha-Linolenic Acid/pharmacology
18.
Biol Pharm Bull ; 31(6): 1288-91, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18520071

ABSTRACT

In this study erythrocyte transport of prostaglandin E1 (PGE1) was investigated by employing inside-out membrane vesicles prepared from rat erythrocytes. The uptake of [3H]PGE1 in the presence of ATP was significantly higher than that of AMP, suggesting the involvement of an ATP-dependent efflux system in PGE1 transport across the erythrocyte membrane. Coincubation of glutathione with ATP further stimulated the uptake of [3H]PGE1. The uptake of [3H]PGE1 in the presence of ATP and glutathione was temperature-sensitive, and various eicosanoids including PGE2 and PGF2alpha decreased the uptake. Multidrug resistance-associated protein (MRP) 4 substrates/inhibitors including methotrexate, indomethacin, taurocholic acid and indocyanine green significantly inhibited [3H]PGE1 uptake. Western blot analysis revealed that Mrp4 is expressed in rat erythrocyte membrane. These results suggest that the release of PGE1 from the erythrocyte into the blood circulation may be mediated by ATP-dependent efflux pump(s) such as Mrp4.


Subject(s)
Alprostadil/blood , Erythrocyte Membrane/metabolism , Adenosine Triphosphate/physiology , Alprostadil/antagonists & inhibitors , Animals , Biological Transport, Active , Blotting, Western , Eicosanoids/pharmacology , Glutathione/metabolism , Immunoblotting , In Vitro Techniques , Multidrug Resistance-Associated Proteins/metabolism , Prostaglandin Antagonists/pharmacology , Rats , Temperature , Transport Vesicles/metabolism
19.
Blood ; 105(12): 4635-41, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15705796

ABSTRACT

Cystic fibrosis (CF) is caused by a mutation of the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR). We examined platelet function in CF patients because lung inflammation is part of this disease and platelets contribute to inflammation. CF patients had increased circulating leukocyte-platelet aggregates and increased platelet responsiveness to agonists compared with healthy controls. CF plasma caused activation of normal and CF platelets; however, activation was greater in CF platelets. Furthermore, washed CF platelets also showed increased reactivity to agonists. CF platelet hyperreactivity was incompletely inhibited by prostaglandin E(1) (PGE(1)). As demonstrated by Western blotting and reverse-transcriptase-polymerase chain reaction (RT-PCR), there was neither CFTR nor CFTR-specific mRNA in normal platelets. There were abnormalities in the fatty acid composition of membrane fractions of CF platelets. In summary, CF patients have an increase in circulating activated platelets and platelet reactivity, as determined by monocyte-platelet aggregation, neutrophil-platelet aggregation, and platelet surface P-selectin. This increased platelet activation in CF is the result of both a plasma factor(s) and an intrinsic platelet mechanism via cyclic adenosine monophosphate (cAMP)/adenylate cyclase, but not via platelet CFTR. Our findings may account, at least in part, for the beneficial effects of ibuprofen in CF.


Subject(s)
Cystic Fibrosis/blood , Platelet Activation , Adenylyl Cyclases/metabolism , Adolescent , Adult , Alprostadil/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arachidonic Acid/metabolism , Blood Platelets/metabolism , Blotting, Western , Case-Control Studies , Cell Membrane/metabolism , Child , Cyclic AMP/metabolism , Cystic Fibrosis/drug therapy , Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Fatty Acids/metabolism , Genotype , Humans , Ibuprofen/pharmacology , Inflammation , Leukocytes/cytology , Monocytes/cytology , Monocytes/metabolism , Neutrophils/metabolism , P-Selectin/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thromboxane A2/metabolism , Time Factors , Vitamin E/metabolism
20.
Arch Biochem Biophys ; 292(1): 239-43, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1309294

ABSTRACT

Under aerobic conditions, 9,10-phenanthrenequinone and 5,6-chyrsenequinone undergo oxidation-reduction cycling in the presence of NADH and the NAD-linked 15-hydroxyprostaglandin dehydrogenase. This results in the formation of potentially hazardous semiquinones, the superoxide anion, and H2O2. Superoxide dismutase inhibits this cycling by destroying the free radical chain propagator, the superoxide anion. Four other polycyclic aromatic hydrocarbon quinones are not substrates of the enzyme and they cause it to undergo a time-dependent inactivation. This presumably results from alkylation of the enzyme. Glutathione fully protects the enzyme against inactivation by 1,2-naphthoquinone but is only partially effective against 7,8-benzo[a]pyrenequinone. These results suggest that in tissues which contain the NAD-linked 15-hydroxyprostaglandin dehydrogenase some polycyclic aromatic hydrocarbon quinones might produce deleterious effects by undergoing redox cycling. Others might cause such effects by irreversibly inhibiting the enzyme which catalyzes the first step in prostaglandin catabolism.


Subject(s)
Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Placenta/enzymology , Polycyclic Compounds/chemistry , Quinones/chemistry , Alprostadil/antagonists & inhibitors , Humans , Hydrogen Peroxide/chemistry , Kinetics , Oxidation-Reduction/drug effects , Substrate Specificity , Superoxide Dismutase/pharmacology , Superoxides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL