Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Prostaglandins Other Lipid Mediat ; 148: 106406, 2020 06.
Article in English | MEDLINE | ID: mdl-31945460

ABSTRACT

Androgen deprivation induces vascular dysfunction in which altered release and action of prostanoids has been extensively studied. On the other hand, the vascular organ-culture system has been reported as a valid model for phenotypic changes that occur in several cardiovascular pathologies. Since there are no studies analyzing the impact of androgenic loss on vascular vulnerability during induced vascular damage, the objective of this study was to analyze the possible preventive role of male sex hormones on the organ culture-induced vascular damage in rat aorta. The link to possible changes in gross structure was also analyzed. For this purpose, fresh and 20 h-cultured aortic arterial segments from intact and orchidectomized rats were used to analyze: (i) the release and vasomotor effect of the thromboxane A2 (TXA2), prostaglandin (PG) E2, PGF2α and PGI2; (ii) the vasodilator response induced by acetylcholine (ACh) as well as the involvement of prostanoids, in particular TXA2, in the ACh-induced response; (iii) the effect of activation of thromboxane/prostaglandin (TP) receptors on the ACh-induced response; and (iv) the vascular structure. The results showed that organ culture: i) increased production of prostanoids; ii) increased prostanoids-induced vasomotor responses; iii) decreased ACh-induced relaxation after incubation with indomethacin, a blocker of cyclooxygenases; iv) increased the ACh-induced relaxation after incubation with the TXA2 synthase inhibitor, furegrelate, more in arteries from orchidectomized rats than in those of intact rats; v) diminished ACh-induced relaxation after U-46619 incubation only in arteries from orchidectomized rats; and vi) preserved the integrity of the different vascular layers. These results showed the protective role of male sex hormones against the induced vascular damage, since a decreased deleterious effect of prostanoids, in particular that of TXA2, was observed in arteries from rats with intact gonadal function.


Subject(s)
Androgens/pharmacology , Aorta/drug effects , Endothelium, Vascular/drug effects , Orchiectomy/methods , Organ Culture Techniques/methods , Prostaglandins/toxicity , Thromboxane A2/toxicity , Animals , Aorta/metabolism , Aorta/pathology , Blood Pressure , Cyclooxygenase 2/chemistry , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Male , Models, Animal , Rats , Rats, Sprague-Dawley , Thromboxane-A Synthase/antagonists & inhibitors
2.
Anesthesiology ; 111(2): 279-86, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19568163

ABSTRACT

BACKGROUND: It is not known whether thromboxane A2 impairs adenosine triphosphate (ATP)-sensitive K channel function via increased production of superoxide in blood vessels and whether propofol as a nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitor restores this modification. METHODS: Rat aortas without endothelium were used for isometric force recording, measurements of membrane potential, and superoxide production and Western immunoblotting. Vasorelaxation to an ATP-sensitive K channel opener levcromakalim was obtained during contraction to phenylephrine (3 x 10(-7) M) or a thromboxane A2 analogue U46619 (3 x 10(-7) M). In some experiments, aortas were incubated with an ATP-sensitive K channel antagonist glibenclamide, a superoxide inhibitor Tiron, a nonselective NADPH oxidase inhibitor apocynin, a hydrogen peroxide scavenger catalase, a xanthine oxidase inhibitor allopurinol, a thromboxane receptor antagonist SQ29548 or propofol (3 x 10(-7) to 3 x 10(-6) M). RESULTS: Levcromakalim-induced vasorelaxation was abolished by glibenclamide in rings contracted with either vasoconstrictor agent. Tiron, apocynin, and propofol, but not catalase, augmented the vasodilator response as well as the hyperpolarization by levcromakalim in aortas contracted with U46619. Tiron, apocynin, SQ29548, and propofol, but not allopurinol, similarly reduced in situ levels of superoxide within aortic vascular smooth muscle exposed to U46619. Protein expression of a NADPH oxidase subunit p47phox increased in these arteries, and this augmentation was abolished by propofol. CONCLUSIONS: Thromboxane receptor activation induces vascular oxidative stress via NADPH oxidase, resulting in the impairment of ATP-sensitive K channel function. Propofol reduces this stress via inhibition of a NADPH oxidase subunit p47phox and, therefore, restores ATP-sensitive K channel function.


Subject(s)
Anesthetics, Intravenous/therapeutic use , KATP Channels/drug effects , Propofol/therapeutic use , Thromboxane A2/antagonists & inhibitors , Thromboxane A2/toxicity , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Aorta, Thoracic/drug effects , Blotting, Western , Cromakalim/pharmacology , Dose-Response Relationship, Drug , Electrophysiology , In Vitro Techniques , Male , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Phenylephrine/pharmacology , Rats , Rats, Wistar , Superoxides/metabolism , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology
3.
Cardiovasc Res ; 21(2): 119-23, 1987 Feb.
Article in English | MEDLINE | ID: mdl-3664541

ABSTRACT

Diffuse coronary artery vasoconstriction was provoked in the rabbit by a stable thromboxane A2 analogue, STA2 (9,11-epithio-11,12-methano thromboxane A2). Injection of 25 micrograms.kg-1 STA2 into the left main trunk caused complete occlusion of the left anterior descending artery and narrowing of the left circumflex artery. Two minutes after injection, however, the diameter of the coronary artery returned to the control value (n = 10). The right coronary artery was also temporarily occluded by an injection of 25 micrograms.kg-1 STA2. Left ventricular end diastolic pressure increased significantly, and ST segment elevation of the electrocardiogram occurred during vasoconstriction. The angiographic findings showed that the vasoconstriction in the coronary artery induced by STA2 was similar to the diffuse vasoconstriction seen clinically. Induction of the vasoconstriction by STA2 was prevented by the preadministration of 25 micrograms.kg-1 of either a calcium antagonist, diltiazem, or a thromboxane A2 receptor antagonist, ONO 3708 (n = 10). The relation of the calcium movement to this vasoconstriction was studied in vitro using the isolated left circumflex artery in the rabbit. STA2 (50 micrograms.litre-1 to 0.5 mg.litre-1) produced a concentration dependent contraction of helical strips of left circumflex artery. Diltiazem (50-100 g.litre-1) suppressed this contraction dose dependently. ONO 3708 (10 micrograms.litre-1 to 1 mg.litre-1) caused a significant rightward and downward shift of the dose-response curve.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Coronary Vasospasm/chemically induced , Thromboxane A2/analogs & derivatives , Animals , Calcium/pharmacology , Coronary Angiography , Coronary Vasospasm/diagnostic imaging , Diltiazem/pharmacology , Dose-Response Relationship, Drug , Rabbits , Thromboxane A2/antagonists & inhibitors , Thromboxane A2/toxicity
4.
J Cereb Blood Flow Metab ; 17(2): 210-9, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9040501

ABSTRACT

The role of the phosphodiesterase type IV isozyme (PDE IV) in the regulation of cerebrovascular tone was investigated in the canine basilar artery in vitro and in vivo. The PDE isozymes extracted from the canine basilar artery were isolated by diethylaminoethanol (DEAE)-Sepharose affinity chromatography and identified based on sensitivity to isozyme-selective PDE inhibitors. [3H]cAMP hydrolysis was observed in one major and one minor peak of activity. The predominant peak was inhibited by the addition of cGMP (25%), siguazodan (26%), rolipram (39%), and the combination of siguazodan and rolipram (95%). Selective PDE IV inhibitors BRL 61063, rolipram, and denbufylline were equieffective inhibitors of [3H]-ccAMP hydrolysis mediated by PDE IV isolated from the canine basilar artery [concentrations producing 50% inhibition (IC50S) = 0.21 +/- 0.05 microM, 0.67 +/- 0.23 microM, and 0.73 +/- 0.16 microM, respectively]. In precontracted isolated ring segments of the canine basilar artery, selective PDE IV inhibitors produced potent and complete relaxation (IC50S < 150 nM). In contrast, zaprinast (a selective PDE V inhibitor) and siguazodan (a selective PDE III inhibitor) produced only weak relaxation of the basilar artery (IC50S = 4.5 microM and > 10 microM, respectively). Vasorelaxation produced by PDE IV inhibitors was not altered by removing the endothelium, 1-NAME, or adenosine receptor antagonism. In a canine model of acute cerebral vasospasm, all three selective PDE IV inhibitors reversed basilar artery spasm produced by autologous blood without altering mean arterial blood pressure. In contrast, prolonged treatment with BRL 61063 failed to alter the development of basilar spasm in the two hemorrhage canine models of chronic cerebral vasospasm. Denbufylline-induced relaxation in vitro was also significantly impaired in basilar arteries obtained from the model of chronic vasospasm. In conclusion, PDE IV appears to be the predominant isozyme regulating vascular tone mediated by cAMP hydrolysis in cerebral vessels. In addition, vasorelaxation modulated by PDE IV is compromised in chronic cerebral vasospasm associated with subarachnoid hemorrhage.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases , Cerebral Arteries/enzymology , Isoenzymes/physiology , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Animals , Basilar Artery , Cyclic AMP/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4 , Dogs , Drug Evaluation, Preclinical , Ischemic Attack, Transient/complications , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/enzymology , Isoenzymes/isolation & purification , Male , Phosphodiesterase Inhibitors/therapeutic use , Phosphoric Diester Hydrolases/isolation & purification , Prostaglandin Endoperoxides, Synthetic/pharmacology , Prostaglandin Endoperoxides, Synthetic/toxicity , Pyrrolidinones/pharmacology , Pyrrolidinones/therapeutic use , Rolipram , Second Messenger Systems , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/etiology , Subarachnoid Hemorrhage/prevention & control , Thromboxane A2/analogs & derivatives , Thromboxane A2/pharmacology , Thromboxane A2/toxicity , Vasoconstrictor Agents/pharmacology , Vasoconstrictor Agents/toxicity , Vasodilator Agents/pharmacology , Vasodilator Agents/therapeutic use , Xanthines/pharmacology , Xanthines/therapeutic use
5.
J Appl Physiol (1985) ; 77(3): 1466-73, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7836154

ABSTRACT

Cardiorespiratory variables were measured continuously in five conscious goats before and after the infusion of U-46619 at a dose of either 2, 4, or 6 micrograms.kg-1.5 min-1. Infusion of U-46619 led to immediate increases in pulmonary arterial blood pressure (ABP) that were sustained for up to 15 min after the end of the infusion. Systemic ABP also increased, but the relative increase from control was less than the pulmonary pressor response. At the highest dose, U-46619 elicited a delayed tachypneic response that was greatest several minutes after the infusion was stopped. U-46619 was also infused simultaneously with sodium nitroprusside to clamp ABP pressure at baseline levels to determine whether stimulation of baroreceptors might contribute to the latency of the tachypneic response. Although sodium nitroprusside infusion prevented the increase in ABP, the increase in breathing frequency was still delayed 3-4 min from the start of the infusion. We conclude that U-46619 elicits pulmonary and systemic arterial hypertension in the conscious goat. At the higher dose U-46619 also elicits a delayed tachypnea that remains delayed even if ABP is normal.


Subject(s)
Hypertension, Pulmonary/chemically induced , Hypertension/chemically induced , Prostaglandin Endoperoxides, Synthetic/toxicity , Respiration Disorders/chemically induced , Thromboxane A2/analogs & derivatives , Vasoconstrictor Agents/toxicity , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Animals , Blood Gas Analysis , Blood Pressure/drug effects , Female , Goats , Hypertension/physiopathology , Hypertension, Pulmonary/physiopathology , Nitroprusside/pharmacology , Phenylephrine/pharmacology , Pressoreceptors/drug effects , Respiration Disorders/physiopathology , Respiratory Mechanics/drug effects , Thromboxane A2/toxicity
6.
Eur J Pharmacol ; 250(1): 189-91, 1993 Nov 30.
Article in English | MEDLINE | ID: mdl-8119318

ABSTRACT

The thromboxane A2 receptor agonist, U-46619 ((5Z, 9 alpha, 11 alpha, 13E, 15(S))-9,11-(methanoepoxy)prosta-5,13-dien-1-oic acid) (10 micrograms/kg), induced a typical ischemic change (ST elevation) in the electrocardiogram on intracoronary arterial administration in the rat. The elevation of the ST segment induced by U-46619 was significantly reduced by pretreatment with anti-endothelin-1 rabbit serum. The plasma concentration of endothelin-1 dose dependently increased at the time of ST segment elevation after U-46619. These results indicate that endogenous endothelin-1 partly contributes to coronary spasmodic angina induced by thromboxane A2 in rats.


Subject(s)
Angina Pectoris/chemically induced , Endothelins/physiology , Myocardial Ischemia/chemically induced , Prostaglandin Endoperoxides, Synthetic/toxicity , Thromboxane A2/analogs & derivatives , Vasoconstrictor Agents/toxicity , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Angina Pectoris/physiopathology , Animals , Electrocardiography/drug effects , Endothelins/blood , Injections, Intra-Arterial , Male , Myocardial Ischemia/physiopathology , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Thromboxane A2/toxicity
7.
Fundam Clin Pharmacol ; 10(5): 458-66, 1996.
Article in English | MEDLINE | ID: mdl-8902549

ABSTRACT

The effects of S 12370 (2-[4-benzhydryloxypiperidinoethyl]isoxindole), were studied in vitro. In guinea pig isolated tracheal rings, S 12370 induced a similar competitive inhibition of the contractile responses produced by acetylcholine, histamine and serotonin. However, it did not affect the contractions induced by leukotriene D4 (LTD4), substance P and U 46619, a stable analogue of thromboxane A2. S 12370 induced a concentration dependent inhibition of the cholinergic component of the contraction induced by electrical field stimulation, whereas it did not influence the sustained nonadrenergic noncholinergic (NANC) excitatory response observed in guinea pig isolated bronchi. S 12370 did not influence the relaxations induced by prostaglandin E2, isoprenaline and salbutamol, and did not modify the nonadrenergic noncholinergic inhibitory response induced by electrical field stimulation. In isolated left atria, the negative inotropic effect of acetylcholine was competitively inhibited by S 12370. In binding experiments, S 12370 exhibited similar affinity for M1, M2, M3, M4 muscarinic receptors and also recognized 5-HT2 serotonin and H1 histamine receptor subtypes. In ovalbumin-sensitized animals, the contractile response of isolated tracheal rings produced by exposure to the allergen was not influenced by S 12370. Tracheal rings from sensitized animals preexposed in vitro to the allergen developed a hyporesponsiveness to beta-adrenoceptor stimulation. S 12370 prevented the inhibitory effect caused by ovalbumin immune sensitization in the relaxation to isoprenaline. In rat polymorphonuclear neutrophil (PMN) cells, S 12370 up to 10(-5) M did not inhibit the arachidonic acid metabolism. These results suggest that in guinea pig tracheal smooth muscle, S 12370 is a competitive inhibitor of muscarinic, serotonin and histamine receptors and can modulate the beta-adrenergic dysfunction induced by immune sensitization. S 12370 may present some therapeutic interest in inflammatory airway diseases.


Subject(s)
Benzhydryl Compounds/pharmacology , Muscle, Smooth/drug effects , Ovalbumin/toxicity , Vasoconstrictor Agents/toxicity , Vasodilator Agents/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Acetylcholine/toxicity , Animals , Arachidonic Acid/metabolism , Benzhydryl Compounds/metabolism , Binding, Competitive , Electric Stimulation , Guinea Pigs , Heart/drug effects , Histamine/toxicity , Leukotriene D4/toxicity , Lung/drug effects , Lung/metabolism , Male , Muscle Contraction/drug effects , Muscle, Smooth/metabolism , Muscle, Smooth, Vascular/drug effects , Oxindoles , Prostaglandin Endoperoxides, Synthetic/toxicity , Rats , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta/metabolism , Serotonin/toxicity , Substance P/toxicity , Thromboxane A2/analogs & derivatives , Thromboxane A2/toxicity
8.
Early Hum Dev ; 55(2): 125-36, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10390088

ABSTRACT

IUGR was induced by maternal administration of synthetic thromboxane A2 (STA2) from the 13th day of gestation. Fetuses and neonates showed a markedly significant weight reduction. In E16 IUGR brain, no pathological abnormalities were found, but morphological changes appeared in the cortical plate of E18 IUGR brain. In E20 IUGR brain, ectopic clusters of differentiating cells cytologically mimicking neuroblasts were found in the neuroepithelial layer, but these abnormal clusters of cells in IUGR brain of late gestation were never observed in PN7. Morphometric analysis of coronal-sectional areas of the brain and cortical plate demonstrated that there were no differences between IUGR rats and controls in E16 and E18. These areas were, however, significantly reduced in E20 and PN7 growth-retarded rats compared with the control. Because the period of STA2 administration coincides with the neuro-developmental stage of cell migration and differentiation, reduction of the uteroplacental blood supply might cause a transient abnormal cytoarchitecture of the cerebral cortex resulting in brain growth retardation.


Subject(s)
Brain Diseases/pathology , Brain/pathology , Fetal Growth Retardation/pathology , Thromboxane A2/analogs & derivatives , Animals , Animals, Newborn , Body Weight/drug effects , Brain/drug effects , Brain/embryology , Brain Diseases/chemically induced , Disease Models, Animal , Embryonic and Fetal Development/drug effects , Female , Fetal Growth Retardation/chemically induced , Pregnancy , Rats , Rats, Sprague-Dawley , Thromboxane A2/toxicity
9.
J Pharm Pharmacol ; 46(7): 558-62, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7996382

ABSTRACT

The anti-anginal effect of nitroglycerin and prostacyclin was examined using, as an index, the ischaemic electrocardiogram (ECG) change (ST elevation) induced by intracoronary arterial injection of 9,11-dideoxy-11 alpha,9 alpha-epoxymethano-PGF2 alpha (U-46619), a stable thromboxane A2 agonist, in anaesthetized rats. The ST elevation induced by U-46619 (5-20 micrograms kg-1, i.c.a.) was dose-dependent and reproducible. U-46619-induced ST elevation was markedly prevented by the pretreatment of intravenous administration of prostacyclin (0.01 micrograms kg-1), and to a lesser extent by nitroglycerin (0.3 mg kg-1). Simultaneously, platelet count decreased significantly in the coronary arterial blood which indicated that platelet aggregation was enhanced by U-46619. The decrease of platelet count in coronary arterial blood at the time of ST elevation was significantly suppressed by prostacyclin (0.1 microgram kg-1, i.v.), but not by nitroglycerin (0.3 mg kg-1, i.v.). These results suggest that the ST elevation induced by intracoronary arterial injection of U-46619 may be derived from spasm of coronary artery and platelet aggregation in the intracoronary artery in rats.


Subject(s)
Electrocardiography/drug effects , Epoprostenol/therapeutic use , Myocardial Ischemia/prevention & control , Nitroglycerin/therapeutic use , Prostaglandin Endoperoxides, Synthetic/toxicity , Thromboxane A2/analogs & derivatives , Vasoconstrictor Agents/toxicity , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Analysis of Variance , Animals , Blood Platelets/drug effects , Blood Pressure/drug effects , Coronary Vessels/drug effects , Disease Models, Animal , Epoprostenol/administration & dosage , Epoprostenol/pharmacology , Heart Rate/drug effects , Injections, Intra-Arterial , Injections, Intravenous , Male , Myocardial Ischemia/chemically induced , Nitroglycerin/administration & dosage , Nitroglycerin/pharmacology , Platelet Aggregation/drug effects , Platelet Count/drug effects , Prostaglandin Endoperoxides, Synthetic/administration & dosage , Rats , Rats, Sprague-Dawley , Thromboxane A2/administration & dosage , Thromboxane A2/toxicity , Vasoconstrictor Agents/administration & dosage
10.
Methods Find Exp Clin Pharmacol ; 13(2): 105-10, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1906568

ABSTRACT

Injection of sodium arachidonate (NaAr) intravenously at a dose of 2 mg/kg is uniformly lethal in rabbits within 3 min. This sudden death is characterized by a precipitous drop in mean blood pressure within 2 min after injection of NaAr, a marked decrease in the circulating platelet count, a significant increase in intratracheal pressure and in plasma thromboxane A2 (TxA2) concentration as measured by radioimmunoassay of its stable breakdown product, TxB2. Pretreatment with Bay-u-3405, a new specific thromboxane receptor antagonist, at a dose of 1 or 10 mg/kg dramatically protected rabbits against sudden death induced by injection of NaAr. All of the rabbits treated with either of these two doses of Bay-u-3405 survived, and their thrombocytopenia, elevated plasma TxB2 concentration and bronchoconstriction were significantly attenuated. However, administration of 0.1 mg/kg Bay-u-3405 exerted no protective effect in this lethal model. Bay-u-3405 was shown to be a potent and specific inhibitor of thromboxane-mimetic induced platelet aggregation in vitro. Our data clearly show that Bay-u-3405 is a very effective protective agent against NaAr-induced sudden death in rabbits, blocking all of the known deleterious effects of TxA2.


Subject(s)
Arachidonic Acids/antagonists & inhibitors , Carbazoles/pharmacology , Death, Sudden , Sulfonamides/pharmacology , Thromboxanes/antagonists & inhibitors , Animals , Arachidonic Acid , Arachidonic Acids/toxicity , Blood Pressure/drug effects , Bronchi/drug effects , Electrocardiography , Male , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Rabbits , Radioimmunoassay , Thromboxane A2/toxicity
11.
Thromb Haemost ; 112(5): 1051-64, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25030617

ABSTRACT

High shear stress that develops in the arteriovenous fistula of chronic kidney diseases (CKD) may increase H2O2 and thromboxane A2 (TXA2) release, thereby exacerbating endothelial dysfunction, thrombosis, and neointimal hyperplasia. We investigated whether glucagon-like peptide-1 receptor agonist/exendin-4, a potentially cardiovascular protective agent, could improve TXA2-induced arteriovenous fistula injury in CKD. TXA2 administration to H2O2-exposed human umbilical vein endothelial cells increased apoptosis, senescence, and detachment; these phenotypes were associated with the downregulation of phosphorylated endothelial nitric oxide synthase/heme oxygenase-1 (eNOS/HO-1) signalling. Exendin-4 reduced H2O2/TXA2-induced endothelial injury via inhibition of apoptosis-related mechanisms and restoration of phosphorylated eNOS/HO-1 signalling. Male Wistar rats subjected to right common carotid artery-external jugular vein anastomosis were treated with exendin-4 via cervical implant osmotic pumps for 16-42 days. High shear stress induced by the arteriovenous fistula significantly increased venous haemodynamics, blood and tissue H2O2 and TXB2 levels, macrophage/monocyte infiltration, fibrosis, proliferation, and adhesion molecule-1 expression. Apoptosis was also increased due to NADPH oxidase gp91 activation and mitochondrial Bax translocation in the proximal end of the jugular vein of CKD rats. Exendin-4-treatment of rats with CKD led to the restoration of normal endothelial morphology and correction of arteriovenous fistula function. Exendin-4 treatment or thromboxane synthase gene deletion in CKD mice markedly reduced ADP-stimulated platelet adhesion to venous endothelium, and prevented venous occlusion in FeCl3-injured vessels by upregulation of HO-1. Together, these data reveal that the use of glucagon-like peptide-1 receptor agonists is an effective strategy for treatment of CKD-induced arteriovenous fistula failure.


Subject(s)
Arteriovenous Shunt, Surgical/adverse effects , Peptides/therapeutic use , Receptors, Glucagon/agonists , Renal Insufficiency, Chronic/therapy , Venoms/therapeutic use , Venous Thrombosis/prevention & control , Anastomosis, Surgical , Animals , Apoptosis/drug effects , Carotid Artery, Common/pathology , Carotid Artery, Common/surgery , Cell Adhesion/drug effects , Cellular Senescence/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Exenatide , Glucagon-Like Peptide-1 Receptor , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen Peroxide/toxicity , Jugular Veins/pathology , Jugular Veins/surgery , Male , Mice , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , Peptides/pharmacology , Rats , Rats, Wistar , Receptors, Glucagon/physiology , Thromboxane A2/toxicity , Venoms/pharmacology , Venous Thrombosis/etiology
12.
Exp Neurol ; 219(1): 81-92, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19393646

ABSTRACT

Fetal growth retardation (FGR) is a critical problem in the neonatal period, because a substantial population of infants born with FGR go on to develop various developmental disorders. In the present study, we produced FGR model rats by continuous administration of a synthetic thromboxane A2 analogue (STA2) to pregnant rats. The FGR pups exhibited a significant delay in postnatal neurological development. Moreover, behavioral analyses revealed the presence of a learning disability in juvenile FGR male rats. To investigate the mechanism underlying the neurological disorders, histological and biochemical analyses of the brain of FGR rats were performed. The density of neurons in the cortical plate of an FGR brain was low compared with the brains of a similarly aged, healthy rat. Consistent with this finding, the density of TUNEL-positive cells was higher in the cortical plate of FGR brains. Western blot analyses showed that the levels of three brain-specific chondroitin sulfate proteoglycans (CSPGs), neurocan, phosphacan, and neuroglycan C, were all significantly reduced in the brain of neonatal FGR rats compared with those of the control. The reduction of CSPG-levels and morphological changes in the brain may be relevant to neurological dysfunction in FGR.


Subject(s)
Brain/growth & development , Brain/metabolism , Developmental Disabilities/metabolism , Fetal Growth Retardation/metabolism , Proteoglycans/deficiency , Animals , Animals, Newborn , Brain/pathology , Cell Count , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Chondroitin Sulfate Proteoglycans/metabolism , Developmental Disabilities/etiology , Developmental Disabilities/physiopathology , Disease Models, Animal , Female , Fetal Growth Retardation/chemically induced , Fetal Growth Retardation/physiopathology , Growth Inhibitors/toxicity , Humans , Infant, Newborn , Learning Disabilities/etiology , Learning Disabilities/metabolism , Learning Disabilities/physiopathology , Male , Membrane Proteins/metabolism , Neurocan , Pregnancy , Proteoglycans/metabolism , Rats , Rats, Sprague-Dawley , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Thromboxane A2/analogs & derivatives , Thromboxane A2/toxicity
13.
J Lipid Mediat Cell Signal ; 11(1): 1-12, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7728413

ABSTRACT

Membrane-derived lipid mediators have been considered to play a major role in pathogenesis of bronchial asthma. However, the importance of and the interactions among each mediator are still unclear. We examined the role of thromboxane A2 (TXA2) and platelet-activating factor (PAF) in immediate asthmatic response (IAR) and interactions between these lipid mediators in guinea pig airway in vivo using a specific TXA2 antagonist S-1452 and a specific PAF antagonist Y-24180. We confirmed the activity of each antagonist, as S-1452 and Y-24180 significantly and dose-dependently inhibited bronchoconstriction induced by respective agonist inhalation. S-1452 inhibited IAR but Y-24180 did not, indicating that TXA2 plays a major role in IAR but PAF does not. S-1452 significantly inhibited PAF-induced bronchoconstriction but Y-24180 did not inhibit synthesized TXA2 (STA2)-induced bronchoconstriction, showing that the bronchoconstrictive effect of PAF is at least in part dependent on secondarily released TXA2, but TXA2 does not induce PAF production.


Subject(s)
Asthma/immunology , Hypersensitivity/physiopathology , Platelet Activating Factor/physiology , Thromboxane A2/physiology , Administration, Inhalation , Animals , Antigens/toxicity , Asthma/physiopathology , Azepines/pharmacology , Bridged Bicyclo Compounds/pharmacology , Bronchoconstriction/drug effects , Bronchoconstriction/immunology , Dose-Response Relationship, Drug , Fatty Acids, Monounsaturated/pharmacology , Guinea Pigs , Male , Platelet Activating Factor/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Thromboxane A2/analogs & derivatives , Thromboxane A2/antagonists & inhibitors , Thromboxane A2/toxicity , Triazoles/pharmacology
14.
J Cardiovasc Pharmacol ; 23(1): 113-9, 1994 Jan.
Article in English | MEDLINE | ID: mdl-7511722

ABSTRACT

Nitric oxide (NO), a labile humoral factor produced by vascular endothelial cells, is a potent vasodilator and an important mediator of pulmonary vascular tone. Nucleophile/NO adducts are a new class of compounds that spontaneously and predictively release NO. We investigated the hemodynamic effects of intravenous (i.v.) infusions of a recently developed NO-donor drug, the diethylamine-nitric oxide adduct (DEA/NO), in 17 intact newborn lambs. At rest, DEA/NO (1-2 microgram.kg-1.min-1) produced dose-dependent decreases in mean pulmonary (from 10.6 +/- 8.6 to 21.2 +/- 7.9%, p < 0.05) and systemic arterial pressure (from 13.2 +/- 11.7 to 31.0 +/- 15.4%, p < 0.05). Similarly, during pulmonary hypertension induced by infusion of U46619, DEA/NO (0.5-2.0 micrograms.kg-1.min-1) produced dose-dependent decreases in mean pulmonary (from 7.3 +/- 5.6 to 24.1 +/- 13.3%, p < 0.05) and systemic arterial pressure (from 2.2 +/- 3.8 to 20.3 +/- 12.9%, p < 0.05). Cardiac output (CO), heart rate (HR), systemic arterial blood gases, and pH were unchanged; atrial pressures decreased at higher doses. Equimolar infusions of S-nitroso-N-acetyl-penicillamine, nitroglycerin (NTG), and sodium nitroprusside (SNP) produced similar decreases in pulmonary and systemic arterial pressure. The nucleophile/NO adducts are potent vasodilators; their predictable and quantitative release of NO make them potentially useful research tools. In addition, because these compounds may decrease the incidence of tolerance and the risk from toxic metabolites associated with use of other nitrovasodilators, they may be clinically useful.


Subject(s)
Blood Pressure/drug effects , Diethylamines/pharmacology , Hypertension, Pulmonary/drug therapy , Nitric Oxide/pharmacology , Pulmonary Artery/drug effects , Vasodilation/drug effects , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Hemodynamics/drug effects , Hypertension, Pulmonary/chemically induced , Infusions, Intravenous , Prostaglandin Endoperoxides, Synthetic/toxicity , Sheep , Thromboxane A2/analogs & derivatives , Thromboxane A2/toxicity , Vascular Resistance/drug effects , Vasoconstrictor Agents/toxicity , Vasodilator Agents/pharmacology
15.
J Cardiovasc Pharmacol ; 25(2): 207-15, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7752646

ABSTRACT

The hemodynamic effects of endothelin-1 (ET-1) are mediated by at least two distinct receptors: ETa and ETb receptors. Recently, ETb receptor agonists (4 Ala ET-1 and IRL 1620) were developed. To investigate the role of ETb receptor activation on the pulmonary and systemic circulations, we studied the hemodynamic effects of intrapulmonary arterial injections of these receptor agonists in 10 intact newborn lambs. At rest, 4 Ala ET-1 (290-1,725 ng/kg) changed no hemodynamic variables. IRL 1620 (180-1,095 ng/kg) decreased mean pulmonary arterial pressure (PAP, 16.8% +/- 15.0 and 17.8% +/- 8.5, p < 0.05) and left pulmonary artery blood flow (21.6% +/- 22.1 and 33.4% +/- 27.7, p < 0.05) at the two highest doses only. During U46619-induced pulmonary hypertension, both 4 Ala ET-1 (3.2% +/- 8.0 to 15.9% +/- 6.4, p < 0.05) and IRL 1620 (8.7% +/- 6.3 to 21.9% +/- 4.1, p < 0.05) produced selective dose-dependent decreases in PAP. The decrease in mean PAP induced by 4 Ala ET-1 and IRL 1620 was attenuated by N omega-nitro-L-arginine [an inhibitor of endothelium-derived nitric oxide (EDNO) synthesis] (16.6% +/- 3.5 vs. 5.9% +/- 2.3 and 16.2% +/- 3.4 vs. 6.6% +/- 2.8, p < 0.05) and by glybenclamide (a blocker of ATP-dependent potassium channels) (18.2% +/- 7.9 vs. 7.5% +/- 8.3 and 14.7% +/- 3.6 vs. 6.3% +/- 3.2, p < 0.05). ETb receptor activation produces selective pulmonary vasodilation during pulmonary hypertension in intact newborn lambs. The vasodilating properties are mediated in part by release of ENDO and by potassium channel activation.


Subject(s)
Endothelins/pharmacology , Hypertension, Pulmonary/physiopathology , Peptide Fragments/pharmacology , Vasodilation/drug effects , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Animals , Animals, Newborn , Arginine/analogs & derivatives , Arginine/pharmacology , Blood Pressure/drug effects , Disease Models, Animal , Endothelins/administration & dosage , Endothelins/therapeutic use , Endothelium, Vascular/drug effects , Glyburide/pharmacology , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Injections, Intra-Arterial , Nitroarginine , Peptide Fragments/administration & dosage , Peptide Fragments/therapeutic use , Prostaglandin Endoperoxides, Synthetic/administration & dosage , Prostaglandin Endoperoxides, Synthetic/toxicity , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Circulation/drug effects , Regional Blood Flow/drug effects , Sheep , Thromboxane A2/administration & dosage , Thromboxane A2/analogs & derivatives , Thromboxane A2/toxicity , Vascular Resistance/drug effects , Vasoconstrictor Agents/administration & dosage , Vasoconstrictor Agents/toxicity
16.
J Cardiovasc Pharmacol ; 30(1): 102-9, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9268228

ABSTRACT

In the fetal lamb, oxygen-induced pulmonary vasodilation is attenuated by the combined use of purinergic receptor P1 and P2y antagonists, which block the effect of adenosine and adenosine triphosphate (ATP), respectively, and by N(omega)-nitro-L-arginine [an inhibitor of endothelium-derived nitric oxide (EDNO) synthesis]. In the newborn lamb, oxygen-induced pulmonary vasodilation is not blocked by N(omega)-nitro-L-arginine. We investigated the role of ATP and adenosine in oxygen-induced pulmonary vasodilation in eight newborn lambs with pulmonary hypertension induced by the thromboxane mimic, U46619. The hemodynamic effects of hyperoxia, ATP, adenosine, sodium nitroprusside (SNP), and acetylcholine (ACh) were compared before and after purinergic receptor blockade with Cibacron blue (CB, a P2y-receptor antagonist) and 8-phenyltheophylline (8PT, a P1-receptor antagonist) individually, together, and on a separate day, after infusion of N(omega)-nitro-L-arginine. During pulmonary hypertension, combined pretreatment with 8PT and CB attenuated the decrease in pulmonary arterial pressure caused by hyperoxia (11.3 vs. 35.2%), ATP (10.6 vs. 32.2%), and adenosine (1.9 vs. 33.7%) without change in the effect of ACh or SNP (p < 0.05). N(omega)-Nitro-L-arginine attenuated the pulmonary vasodilation caused by ATP and ACh but not by hyperoxia, adenosine, or SNP. In the newborn lamb, the pulmonary vasodilating effect of both oxygen and ATP are attenuated by combined P1 and P2y purinergic-receptor antagonists. Postnatally, oxygen-induced pulmonary vasodilation appears to be mediated by ATP through purinergic receptors.


Subject(s)
Adenosine Triphosphate/physiology , Hypertension, Pulmonary/physiopathology , Oxygen/pharmacology , Pulmonary Artery/physiopathology , Vasodilation/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Adenosine/physiology , Animals , Animals, Newborn , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Hemodynamics/drug effects , Hypertension, Pulmonary/chemically induced , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitroarginine/pharmacology , Prostaglandin Endoperoxides, Synthetic/toxicity , Pulmonary Artery/drug effects , Purinergic Antagonists , Sheep , Theophylline/analogs & derivatives , Theophylline/pharmacology , Thromboxane A2/analogs & derivatives , Thromboxane A2/toxicity , Triazines/pharmacology , Vasoconstrictor Agents/toxicity , Vasodilation/drug effects , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL