Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 209
Filter
1.
Allergol Int ; 69(2): 223-231, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31601467

ABSTRACT

BACKGROUND: Cough variant asthma (CVA) is recognized as a precursor of bronchial asthma (BA). However, the cough response to bronchoconstriction differs between these similar diseases. Repeated bronchoconstriction and the resulting imbalance of endogenous lipid mediators may impact the cough response. METHODS: We investigated the influence of repeated bronchoconstriction on the cough response to bronchoconstriction using naïve guinea pigs. Bronchoconstriction was induced for 3 consecutive days and changes in the cough response and lipid mediators, such as PGE2, PGI2, and cysteinyl-LTs (Cys-LTs), in BAL fluid (BALF) were assessed. We investigated the effect of endogenous PGI2 on the cough response by employing a PGI2 receptor antagonist. In order to investigate the cough response over a longer period, we re-evaluated the cough response 2 weeks after repeated bronchoconstriction. RESULTS: The number of coughs induced by bronchoconstriction were significantly decreased by repeated bronchoconstriction. The levels of PGE2, PGI2, and Cys-LTs, and the ratio of PGI2/PGE2 were significantly increased, following repeated bronchoconstriction. This decrease in the cough response was suppressed by pretreatment with a PGI2 receptor antagonist. Two weeks after repeated bronchoconstriction, the cough response returned to the same level as before repeated bronchoconstriction along with a concomitant return of lipid mediators, such as PGE2, PGI2, and Cys-LTs and the ratio of PGI2/PGE2. CONCLUSIONS: Our results suggest that repeated bronchoconstriction and the resulting imbalance of endogenous lipid mediators contribute to the difference in cough responses to bronchoconstriction in CVA and BA.


Subject(s)
Asthma/metabolism , Bronchi/physiology , Cough/metabolism , Animals , Asthma/physiopathology , Bronchial Provocation Tests , Bronchoconstriction , Cough/physiopathology , Cysteine/metabolism , Dinoprostone/metabolism , Disease Models, Animal , Epoprostenol/antagonists & inhibitors , Epoprostenol/metabolism , Guinea Pigs , Humans , Leukotrienes/metabolism , Lipid Metabolism , Male , Methacholine Chloride
2.
Br J Clin Pharmacol ; 85(2): 377-384, 2019 02.
Article in English | MEDLINE | ID: mdl-30378151

ABSTRACT

AIMS: Nitroglycerin (or glyceryl trinitrate, GTN) has been long considered an endothelium-independent vasodilator because GTN vasodilation is intact in the absence of the endothelium and in the presence of endothelial dysfunction. However, in animal and in vitro models, GTN has been shown to stimulate the release of certain endothelium-derived vasodilators such as nitric oxide (NO) and prostacyclin (PGI2 ). In addition, chronic GTN therapy leads to endothelial dysfunction. In this series of experiments, we explored how GTN might interact with the vascular endothelium in normal humans, without cardiovascular disease or risk factors associated with abnormalities in vascular function. METHODS: We examined the effect of inhibition of NO, PGI2 , and epoxyeicosatrienoic acids (EETs, a class of endothelium-derived hyperpolarizing factor) on GTN-mediated vasodilation. We measured arterial blood flow responses to brachial artery infusions of GTN in the absence and presence of L-NMMA (n = 13), ketorolac (n = 14) and fluconazole (n = 16), which are inhibitors of endothelium-derived NO, PGI2 and EETs, respectively, in healthy volunteers. RESULTS: Our results demonstrate that inhibition of endothelium-dependent vasodilator mechanisms does not alter forearm resistance vessel responses to GTN. CONCLUSION: We conclude that GTN-mediated dilation of forearm resistance vessels is largely independent of vascular endothelium.


Subject(s)
Endothelium, Vascular/drug effects , Nitroglycerin/pharmacology , Regional Blood Flow/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Adolescent , Adult , Animals , Endothelium, Vascular/metabolism , Epoprostenol/antagonists & inhibitors , Epoprostenol/metabolism , Female , Forearm/blood supply , Healthy Volunteers , Humans , Male , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Pilot Projects , Vascular Resistance/drug effects , Young Adult
3.
Circ Res ; 122(12): 1689-1702, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29545367

ABSTRACT

RATIONALE: Pulmonary arterial hypertension (PH) is a life-threatening condition associated with immune dysregulation and abnormal regulatory T cell (Treg) activity, but it is currently unknown whether and how abnormal Treg function differentially affects males and females. OBJECTIVE: To evaluate whether and how Treg deficiency differentially affects male and female rats in experimental PH. METHODS AND RESULTS: Male and female athymic rnu/rnu rats, lacking Tregs, were treated with the VEGFR2 (vascular endothelial growth factor receptor 2) inhibitor SU5416 or chronic hypoxia and evaluated for PH; some animals underwent Treg immune reconstitution before SU5416 administration. Plasma PGI2 (prostacyclin) levels were measured. Lung and right ventricles were assessed for the expression of the vasoprotective proteins COX-2 (cyclooxygenase 2), PTGIS (prostacyclin synthase), PDL-1 (programmed death ligand 1), and HO-1 (heme oxygenase 1). Inhibitors of these pathways were administered to athymic rats undergoing Treg immune reconstitution. Finally, human cardiac microvascular endothelial cells cocultured with Tregs were evaluated for COX-2, PDL-1, HO-1, and ER (estrogen receptor) expression, and culture supernatants were assayed for PGI2 and IL (interleukin)-10. SU5416-treatment and chronic hypoxia produced more severe PH in female than male athymic rats. Females were distinguished by greater pulmonary inflammation, augmented right ventricular fibrosis, lower plasma PGI2 levels, decreased lung COX-2, PTGIS, HO-1, and PDL-1 expression and reduced right ventricular PDL-1 levels. In both sexes, Treg immune reconstitution protected against PH development and raised levels of plasma PGI2 and cardiopulmonary COX-2, PTGIS, PDL-1, and HO-1. Inhibiting COX-2, HO-1, and PD-1 (programmed death 1)/PDL-1 pathways abrogated Treg protection. In vitro, human Tregs directly upregulated endothelial COX-2, PDL-1, HO-1, ERs and increased supernatant levels of PGI2 and IL-10. CONCLUSIONS: In 2 animal models of PH based on Treg deficiency, females developed more severe PH than males. The data suggest that females are especially reliant on the normal Treg function to counteract the effects of pulmonary vascular injury leading to PH.


Subject(s)
Hypertension, Pulmonary/prevention & control , Sex Factors , T-Lymphocytes, Regulatory/physiology , Angiogenesis Inhibitors/pharmacology , Animals , B7-H1 Antigen/analysis , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Chronic Disease , Cyclooxygenase 2/analysis , Cyclooxygenase 2/metabolism , Cytochrome P-450 Enzyme System/analysis , Cytochrome P-450 Enzyme System/metabolism , Epoprostenol/antagonists & inhibitors , Epoprostenol/blood , Epoprostenol/metabolism , Female , Heme Oxygenase (Decyclizing)/analysis , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/metabolism , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/etiology , Hypoxia/complications , Indoles/pharmacology , Intramolecular Oxidoreductases/analysis , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/metabolism , Lung/metabolism , Male , Prostaglandins I/biosynthesis , Pyrroles/pharmacology , Rats , Rats, Nude , Receptors, Estrogen/analysis , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , T-Lymphocytes, Regulatory/immunology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
4.
BMC Surg ; 17(1): 90, 2017 Aug 11.
Article in English | MEDLINE | ID: mdl-28800765

ABSTRACT

BACKGROUND: This study aimed to reveal the appropriate timing for the intravenous administration of flurbiprofen axetil for preventing mesenteric traction syndrome (MTS), caused by prostacyclin release. METHODS: In this prospective, randomized, clinical study, forty-five patients who were undergoing elective surgery for colorectal cancer via laparotomy were enrolled. Patients were randomly divided into 3 groups: a preoperative group (n = 16) receiving flurbiprofen axetil directly before surgery; a post-MTS group (n = 14) receiving following MTS onset; and a control group (n = 15) who were not administered flurbiprofen axetil. 6-keto-PGF1α, a stable metabolite of prostacyclin, levels were measured and mean blood pressures were recorded. RESULTS: In the preoperative group, 6-keto-PGF1α levels did not increase, blood pressure levels did not decrease, and no facial flushing was observed. In both the post-MTS and control groups, 6-keto-PGF1α levels increased markedly after mesenteric traction and blood pressure decreased significantly. The post-MTS group exhibited a faster decreasing trend in 6-keto-PGF1α levels and quick restore of the mean blood pressure, and the use of vasopressors and phenylephrine were lower than that in the control group. CONCLUSIONS: Even therapeutic administration of flurbiprofen axetil after the onset of MTS has also effects on MTS by suppressing prostacyclin production. TRIAL REGISTRATION: Clinical trial number: UMIN000009111 . (Registered 14 October 2012).


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Flurbiprofen/analogs & derivatives , Flushing/drug therapy , Hemodynamics/drug effects , Hypotension/drug therapy , Intraoperative Complications/drug therapy , Tachycardia/drug therapy , 6-Ketoprostaglandin F1 alpha/blood , Aged , Blood Pressure/drug effects , Colorectal Neoplasms/surgery , Epoprostenol/antagonists & inhibitors , Epoprostenol/biosynthesis , Female , Flurbiprofen/administration & dosage , Flushing/prevention & control , Humans , Hypotension/prevention & control , Infusions, Intravenous , Intraoperative Complications/prevention & control , Laparotomy , Male , Middle Aged , Prospective Studies , Syndrome , Tachycardia/prevention & control
5.
Nutrition ; 37: 74-78, 2017 May.
Article in English | MEDLINE | ID: mdl-28359367

ABSTRACT

High sodium, high glucose, and obesity are important risk factors for age-related diseases such as cardiovascular disease (CVDs), stroke, and cancer. Coupling factor 6 (CF6) is released from vascular endothelial cells and functions as a circulating peptide that inhibits prostacyclin and nitric oxide generation by intracellular acidosis. High glucose elevates CF6 by activation of protein kinase C and p38 mitogen-activated protein kinase, whereas CF6 causes type 2 diabetes mellitus, resulting in a high glucose vicious cycle. Low glucose increases inhibitory factor peptide 1, an endogenous inhibitor of CF6. High salt intake increases CF6 through nuclear factor κB signaling, whereas CF6 induces salt-sensitive hypertension and salt-induced congestive heart failure. Oral administration of vitamin C cancels salt-induced increase in CF6, and estrogen replacement leads to the delayed onset of CF6-induced salt-sensitive hypertension and the rescue from cardiac systolic dysfunction. Because CF6 contributes to the onset of CVDs, nutritional regulation of CF6 will shed light on the understanding of preventive strategy and mechanisms for CVDs and a target for therapy.


Subject(s)
Endothelial Cells/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Oxidative Phosphorylation Coupling Factors/metabolism , Administration, Oral , Ascorbic Acid/pharmacology , Diabetes Mellitus, Type 2/diagnosis , Epoprostenol/antagonists & inhibitors , Epoprostenol/metabolism , Heart Failure/diagnosis , Heart Failure/etiology , Humans , Hypertension/diagnosis , NF-kappa B/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Protein Kinase C/metabolism , Signal Transduction , Sodium Chloride, Dietary/administration & dosage , Sodium Chloride, Dietary/adverse effects , p38 Mitogen-Activated Protein Kinases/metabolism
6.
J Steroid Biochem Mol Biol ; 168: 118-126, 2017 04.
Article in English | MEDLINE | ID: mdl-28223151

ABSTRACT

Testosterone has endothelium-dependent vasodilatory effects on the coronary artery, with some reports suggesting endothelial ion channel involvement. This study employed the whole-cell patch clamp technique to investigate the effect of testosterone on ion channels in human coronary artery endothelial cells (HCAECs) and the mechanisms involved. We found that 0.03-3µM testosterone significantly induced a rapid, concentration-dependent increase in total HCAEC current (EC50, 71.96±1.66nM; maximum increase, 59.13±8.37%; mean±SEM). The testosterone-enhanced currents consisted of small- and large-conductance Ca2+-activated K+ currents (SKCa and BKCa currents), but not Cl- and nonselective cation currents. Either a non-permeant testosterone conjugate or the non-aromatizable androgen dihydrotestosterone (DHT) could increase HCAEC currents as well. The androgen receptor antagonist flutamide prevented this testosterone, testosterone conjugate, and DHT effect, while the estrogen receptor antagonist fulvestrant did not. Incubating HCAECs with pertussis toxin or protein kinase A inhibitor H-89 largely inhibited the testosterone effect, while pre-incubation with phospholipase C inhibitor U-73122, prostacyclin inhibitor indomethacin, nitric oxide synthase inhibitor L-NAME or cytochrome P450 inhibitor MS-PPOH, did not. Finally, testosterone application induced HCAEC hyperpolarization within minutes; this effect was prevented by SKCa and BKCa current inhibitors apamin and iberiotoxin. This is the first electrophysiological demonstration of androgen-induced KCa current increase, leading to hyperpolarization, in any endothelial cell, and the first report of SKCa as a testosterone target. Our data show that testosterone rapidly increased whole-cell HCAEC SKCa and BKCa currents via a surface androgen receptor, Gi/o protein, and protein kinase A. This mechanism may explain rapid testosterone-induced coronary vasodilation seen in vivo.


Subject(s)
Coronary Vessels/cytology , Endothelial Cells/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Potassium Channels, Calcium-Activated/metabolism , Testosterone/blood , Androgens/chemistry , Apamin/chemistry , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelial Cells/drug effects , Epoprostenol/antagonists & inhibitors , Estrenes/chemistry , Humans , Indomethacin/chemistry , NG-Nitroarginine Methyl Ester/chemistry , Nitric Oxide Synthase/chemistry , Pyrrolidinones/chemistry , Receptors, Androgen/metabolism , Signal Transduction , Testosterone/chemistry , Vasodilation
7.
BMC Anesthesiol ; 16(1): 121, 2016 12 03.
Article in English | MEDLINE | ID: mdl-27914476

ABSTRACT

BACKGROUND: Lidocaine is an approved local anesthetic and Class 1B antiarrhythmic with a number of ancillary properties. Our aim was to investigate lidocaine's vasoreactivity properties in intact versus denuded rat thoracic aortic rings, and the effect of inhibitors of nitric oxide (NO), prostenoids, voltage-dependent Kv and KATP channels, membrane Na+/K+ pump, and A2a and A2b receptors. METHODS: Aortic rings were harvested from adult male Sprague Dawley rats and equilibrated in an organ bath containing oxygenated, modified Krebs-Henseleit solution, pH 7.4, 37 °C. The rings were pre-contracted sub-maximally with 0.3 µM norepinephrine (NE), and the effect of increasing lidocaine concentrations was examined. Rings were tested for viability after each experiment with maximally dilating 100 µM papaverine. The drugs 4-aminopyridine (4-AP), glibenclamide, 5-hydroxydecanoate, ouabain, 8-(3-chlorostyryl) caffeine and PSB-0788 were examined. RESULTS: All drugs tested had no significant effect on basal tension. Lidocaine relaxation in intact rings was biphasic between 1 and 10 µM (Phase 1) and 10 and 1000 µM (Phase 2). Mechanical removal of the endothelium resulted in further relaxation, and at lower concentrations ring sensitivity (% relaxation per µM lidocaine) significantly increased 3.5 times compared to intact rings. The relaxing factor(s) responsible for enhancing ring relaxation did not appear to be NO- or prostacyclin-dependent, as L-NAME and indomethacin had little or no effect on intact ring relaxation. In denuded rings, lidocaine relaxation was completely abolished by Kv channel inhibition and significantly reduced by antagonists of the MitoKATP channel, and to a lesser extent the SarcKATP channel. Curiously, A2a subtype receptor antagonism significantly inhibited lidocaine relaxation above 100 µM, but not the A2b receptor. CONCLUSIONS: We show that lidocaine relaxation in rat thoracic aorta was biphasic and significantly enhanced by endothelial removal, which did not appear to be NO or prostacyclin dependent. The unknown factor(s) responsible for enhanced relaxation was significantly reduced by Kv inhibition, 5-HD inhibition, and A2a subtype inhibition indicating a potential role for crosstalk in lidocaine's vasoreactivity.


Subject(s)
Aorta, Thoracic/drug effects , KATP Channels/antagonists & inhibitors , Lidocaine/pharmacology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Vasodilation/drug effects , 4-Aminopyridine/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Caffeine/analogs & derivatives , Caffeine/pharmacology , Decanoic Acids/pharmacology , Dose-Response Relationship, Drug , Epoprostenol/antagonists & inhibitors , Glyburide/pharmacology , Hydroxy Acids/pharmacology , In Vitro Techniques , Indomethacin/pharmacology , Lidocaine/antagonists & inhibitors , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/antagonists & inhibitors , Norepinephrine/pharmacology , Ouabain/pharmacology , Papaverine/pharmacology , Rats , Receptor Cross-Talk/drug effects , Receptor, Adenosine A2A/drug effects
8.
Mediators Inflamm ; 2016: 9847840, 2016.
Article in English | MEDLINE | ID: mdl-27478311

ABSTRACT

Prostaglandin E2 (PGE2) is well known as a mediator of inflammatory symptoms such as fever, arthritis, and inflammatory pain. In the present study, we evaluated the analgesic effect of our selective PGE2 synthesis inhibitor, compound I, 2-methyl-2-[cis-4-([1-(6-methyl-3-phenylquinolin-2-yl)piperidin-4-yl]carbonyl amino)cyclohexyl] propanoic acid, in rat yeast-induced acute and adjuvant-induced chronic inflammatory pain models. Although this compound suppressed the synthesis of PGE2 selectively, no analgesic effect was shown in both inflammatory pain models. Prostacyclin (PGI2) also plays crucial roles in inflammatory pain, so we evaluated the involvement of PGI2 signaling in rat inflammatory pain models using prostacyclin receptor (IP) antagonist, RO3244019. RO3244019 showed no analgesic effect in inflammatory pain models, but concomitant administration of compound I and RO3244019 showed analgesic effects comparable to celecoxib, a specific cyclooxygenase- (COX-) 2 inhibitor. Furthermore, coadministration of PGE2 receptor 4 (EP4) antagonist, CJ-023423, and RO3244019 also showed an analgesic effect. These findings suggest that both PGE2 signaling, especially through the EP4 receptor, and PGI2 signaling play critical roles in inflammatory pain and concurrent inhibition of both signals is important for suppression of inflammatory hyperalgesia.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Dinoprostone/metabolism , Epoprostenol/metabolism , Hyperalgesia/drug therapy , Pain/drug therapy , Analgesics/therapeutic use , Animals , Anti-Inflammatory Agents/chemistry , Cyclooxygenase 2 Inhibitors/therapeutic use , Dinoprostone/antagonists & inhibitors , Epoprostenol/antagonists & inhibitors , Inflammation/drug therapy , Macrophages/drug effects , Male , Rats , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Signal Transduction/drug effects , Sulfonamides/therapeutic use
9.
Eur J Nutr ; 55(2): 749-758, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25862540

ABSTRACT

PURPOSE: The aims of this study were to evaluate the antihypertensive effectiveness of different doses of grape seed polyphenols in cafeteria diet-fed hypertensive rats (CHRs) and to establish the mechanism involved in the blood pressure (BP) lowering effect of these compounds in this experimental model of metabolic syndrome (MS). METHODS: Male 8-week-old Wistar rats were fed cafeteria or standard (ST) diet for 10 weeks. After this, the antihypertensive effect of a single oral administration of a polyphenol grape seed extract (GSPE) was tested at different doses (250, 375 and 500 mg/kg) in CHRs. BP was recorded before and 2, 4, 6, 8, 24 and 48 h post-administration. The hypotensive effect of GSPE was also proved in ST diet-fed rats. Additionally, in other experiment, CHRs were orally administered 375 mg/kg GSPE. Four hours post-administration, the rats were intraperitoneally administrated 30 mg/kg NG-nitro-L-arginine methyl ester (L-NAME) or 5 mg/kg indomethacin [inhibitors of nitric oxide (NO) and prostacyclin synthesis, respectively]. BP was recorded initially and 6 h post-administration. RESULTS: GSPE produced a decrease in SBP and DBP, the most effective dose (375 mg/kg) showing an antihypertensive effect in CHRs similar to the drug captopril, and did not affect BP of ST diet-fed rats. The antihypertensive effect was completely abolished by L-NAME and partially inhibited by indomethacin. CONCLUSIONS: GSPE acts as an antihypertensive agent in a rat model of hypertension associated with MS. The change in endothelium-derived NO availability is one of the mechanisms involved in the antihypertensive effect of GSPE in CHRs. Additionally, endothelial prostacyclin contributes to the effect of GSPE on arterial BP.


Subject(s)
Blood Pressure/drug effects , Epoprostenol/metabolism , Grape Seed Extract/pharmacology , Metabolic Syndrome/drug therapy , Nitric Oxide/metabolism , Polyphenols/pharmacology , Administration, Oral , Animals , Antihypertensive Agents/pharmacology , Diet/adverse effects , Dose-Response Relationship, Drug , Epoprostenol/antagonists & inhibitors , Hypertension/chemically induced , Hypertension/drug therapy , Indomethacin/pharmacology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/antagonists & inhibitors , Rats , Rats, Wistar , Seeds/chemistry , Vitis/chemistry
10.
Basic Clin Pharmacol Toxicol ; 113(4): 250-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23724868

ABSTRACT

Polyunsaturated fatty acid (PUFA)-activated two-pore domain potassium channels (K2P ) have been proposed to be expressed in the pulmonary vasculature. However, their physiological or pathophysiological roles are poorly defined. Here, we tested the hypothesis that PUFA-activated K2P are involved in pulmonary vasorelaxation and that alterations of channel expression are pathophysiologically linked to pulmonary hypertension. Expression of PUFA-activated K2P in the murine lung was investigated by quantitative reverse-transcription polymerase chain reaction (qRT-PCR), immunohistochemistry (IHC), by patch clamp (PC) and myography. K2P -gene expression was examined in chronic hypoxic mice. qRT-PCR showed that the K2P 2.1 and K2P 6.1 were the predominantly expressed K2P in the murine lung. IHC revealed protein expression of K2P 2.1 and K2P 6.1 in the endothelium of pulmonary arteries and of K2P 6.1 in bronchial epithelium. PC showed pimozide-sensitive K2P -like K(+) -current activated by docosahexaenoic acid (DHA) in freshly isolated endothelial cells as well as DHA-induced membrane hyperpolarization. Myography on pulmonary arteries showed that DHA induced concentration-dependent instantaneous relaxations that were resistant to endothelial removal and inhibition of NO and prostacyclin synthesis and to a cocktail of blockers of calcium-activated K(+) channels but were abolished by high extracellular (30 mM) K(+) -concentration. Gene expression and protein of K2P 2.1 were not altered in chronic hypoxic mice, while K2P 6.1 was up-regulated by fourfold. In conclusion, the PUFA-activated K2P 2.1 and K2P 6.1 are expressed in murine lung and functional K2P -like channels contribute to endothelium hyperpolarization and pulmonary artery relaxation. The increased K2P 6.1-gene expression may represent a novel counter-regulatory mechanism in pulmonary hypertension and suggest that arterial K2P 2.1 and K2P 6.1 could be novel therapeutic targets.


Subject(s)
Docosahexaenoic Acids/pharmacology , Hypertension, Pulmonary/pathology , Hypoxia/pathology , Potassium Channels, Tandem Pore Domain/metabolism , Animals , Electrophysiology , Epoprostenol/antagonists & inhibitors , Epoprostenol/biosynthesis , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/etiology , Hypoxia/complications , Hypoxia/drug therapy , Lung/drug effects , Lung/metabolism , Male , Mice , Potassium Channels, Tandem Pore Domain/genetics , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation , Vasodilation/drug effects
11.
Prostaglandins Other Lipid Mediat ; 104-105: 93-108, 2013.
Article in English | MEDLINE | ID: mdl-22975359

ABSTRACT

We have previously demonstrated that inhibition of vasodilator prostanoids, PGI2 and PGE2, and nitric oxide (NO) synthesis by a selective cyclooxygenase-2 (COX-2) inhibitor, NS-398, restores blood pressure as a result of increased systemic and renal levels of 20-hydroxyeicosatetraenoic acid (20-HETE) in endotoxemic rats. The aim of this study was to further investigate the effects of NS-398 on the changes in expression and/or activity of COX-2, cytochrome P450 4A1 (CYP4A1), inducible NO synthase (iNOS), and peroxynitrite formation in serum, renal, cardiac, and/or vascular tissues of lipopolysaccharide (LPS)-treated rats. LPS (10mg/kg, i.p.)-induced decrease in blood pressure was associated with increased protein levels of COX-2, iNOS, and nitrotyrosine in kidney, heart, thoracic aorta, and superior mesenteric artery. The activities of COX-2 and iNOS as well as levels of PGI2, PGE2, and nitrotyrosine were also increased in the systemic circulation and renal, cardiac, and vascular tissues of LPS-treated rats. In contrast, renal, cardiac, and vascular CYP4A1 protein expression as well as systemic and tissue levels of 20-HETE were decreased in endotoxemic rats. These effects of LPS, except COX-2 protein expression, were prevented by NS-398 (10 mg/kg, i.p.), given 1h after injection of LPS. These data suggest that COX-2-derived vasodilator prostanoids, PGI2 and PGE2, produced during endotoxemia increase iNOS protein expression and activity as well as peroxynitrite formation resulting in decreased CYP4A1 protein expression and 20-HETE synthesis. Taken together, we concluded that an increase in 20-HETE levels associated with a decrease in the production of vasodilator prostanoids and NO participates in the effect of NS-398 to prevent hypotension in the rat model of septic shock.


Subject(s)
Cyclooxygenase 2 Inhibitors/pharmacology , Endotoxemia/prevention & control , Hypotension/prevention & control , Nitric Oxide/metabolism , Nitrobenzenes/pharmacology , Peroxynitrous Acid/metabolism , Shock, Septic/prevention & control , Sulfonamides/pharmacology , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytochrome P-450 CYP4A/genetics , Cytochrome P-450 CYP4A/metabolism , Dinoprostone/antagonists & inhibitors , Dinoprostone/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endotoxemia/chemically induced , Endotoxemia/metabolism , Endotoxemia/physiopathology , Epoprostenol/antagonists & inhibitors , Epoprostenol/metabolism , Gene Expression , Heart/drug effects , Heart/physiopathology , Hydroxyeicosatetraenoic Acids/metabolism , Hypotension/chemically induced , Hypotension/metabolism , Hypotension/physiopathology , Kidney/drug effects , Kidney/metabolism , Kidney/physiopathology , Lipopolysaccharides , Male , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Peroxynitrous Acid/antagonists & inhibitors , Rats , Rats, Wistar , Shock, Septic/chemically induced , Shock, Septic/metabolism , Shock, Septic/physiopathology
12.
Yao Xue Xue Bao ; 48(11): 1657-64, 2013 Nov.
Article in Chinese | MEDLINE | ID: mdl-24475702

ABSTRACT

This study is to investigate the impairment and possible mechanism of endothelium-dependent relaxation of mice mesenteric arteries induced by mmLDL. Wire myography was employed to examine endothelial function of mesenteric arteries. Ultramicrostructure of mesenteric vascular beds were detected by transmission electron microscope. The results showed that endothelium cell edema and peeling, vascular elastic membrane fracture traces in mmLDL group. Endothelium-dependent relaxation was decreased in a time-dependent and dose-dependent manner by using mmLDL, compared with normal arteries. In endothelium-derived hyperpolarizing factor (EDHF)-mediated relaxation, the Rmax and pIC50 were decreased from (63 +/- 5) % and 6.42 +/- 0.09 of normal saline control to (31 +/- 3) % and 5.67 +/- 0.07 in mmLDL group (P < 0.001, P < 0.001), respectively. In nitric oxide (NO)-mediated relaxation, the Rmax and pIC50 were decreased from (45 +/- 4) % and 5.93 +/- 0.08 in normal saline control to (32 +/- 4) % and 5.43 +/- 0.11 in mmLDL group (P < 0.05, P < 0.01), respectively. There is no significant alteration of prostacyclin I2 (PGI2) pathway between these two groups. In conclusion, mmLDL induced the impairment of the ultramicrostructure of mesenteric vascular endothelium cell as well as the endothelium-dependent relaxation. The latter includes the dysfunction of NO- and EDHF pathway mediated endothelium-dependent relaxation.


Subject(s)
Lipoproteins, LDL/pharmacology , Mesenteric Arteries/physiology , Vasodilation/drug effects , Animals , Biological Factors/antagonists & inhibitors , Biological Factors/physiology , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/ultrastructure , Epoprostenol/antagonists & inhibitors , Epoprostenol/physiology , Female , Lipoproteins, LDL/administration & dosage , Male , Mesenteric Arteries/cytology , Mesenteric Arteries/ultrastructure , Mice , Mice, Inbred ICR , Microscopy, Electron, Transmission , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/physiology
13.
Can J Physiol Pharmacol ; 90(10): 1413-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23067376

ABSTRACT

Angiotensin may promote endothelial dysfunction through iron accumulation. To research this, bovine endothelial cells (ECs) were incubated with iron (30 µmol·L⁻¹) with or without angiotensin II (100 nmol·L⁻¹). After incubation for 6 h, it was observed that the addition of angiotensin enhanced EC iron accumulation by 5.1-fold compared with a 1.8-fold increase for cells incubated with iron only. This enhanced iron uptake was attenuated by losartan (100 nmol·L⁻¹), d-propranolol (10 µmol·L⁻¹), 4-HO-propranolol (5 µmol·L⁻¹), and methylamine, but not by vitamin E or atenolol. After 6 h of incubation, angiotensin plus iron provoked intracellular oxidant formation (2'7'-dichlorofluorescein diacetate (DCF-DA) fluorescence) and elevated oxidized glutathione; significant loss of cell viability occurred at 48 h. Stimulated prostacyclin release decreased by 38% (6 h) and NO synthesis was reduced by 41% (24 h). Both oxidative events and functional impairment were substantially attenuated by losartan or d-propranolol. It is concluded that angiotensin promoted non-transferrin-bound iron uptake via AT-1 receptor activation, leading to EC oxidative functional impairment. The protective effects of d-propranolol and 4-HO-propranolol may be related to their lysosomotropic properties.


Subject(s)
Angiotensin II/metabolism , Endothelium, Vascular/metabolism , Epoprostenol/metabolism , Iron/metabolism , Losartan/pharmacology , Nitric Oxide/metabolism , Propranolol/analogs & derivatives , Adrenergic beta-Antagonists/chemistry , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Angiotensin II/chemistry , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Animals , Aorta/cytology , Aorta/drug effects , Aorta/metabolism , Cattle , Cell Survival/drug effects , Cells, Cultured , Down-Regulation/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Epoprostenol/agonists , Epoprostenol/antagonists & inhibitors , Glutathione/metabolism , Iron/adverse effects , Iron Overload/prevention & control , Losartan/therapeutic use , Nitric Oxide/agonists , Nitric Oxide/antagonists & inhibitors , Oxidation-Reduction , Oxidative Stress/drug effects , Propranolol/pharmacology , Propranolol/therapeutic use , Receptor, Angiotensin, Type 1/metabolism
14.
J Pharmacol Exp Ther ; 341(1): 242-50, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22262921

ABSTRACT

Vascular cyclooxygenase (COX)-2-dependent prostacyclin (PGI(2)) may affect angiogenesis by preventing endothelial activation and platelet release of angiogenic factors present in platelet α-granules. Thus, a profound inhibition of COX-2-dependent PGI(2) might be associated with changes in circulating markers of angiogenesis. We aimed to address this issue by performing a clinical study with celecoxib in familial adenomatous polyposis (FAP). In nine patients with FAP and healthy controls, pair-matched for gender and age, we compared systemic biosynthesis of PGI(2), thromboxane (TX) A(2), and prostaglandin (PG) E(2), assessing their urinary enzymatic metabolites, 2,3-dinor-6-keto PGF(1α) (PGI-M), 11-dehydro-TXB(2) (TX-M), and 11-α-hydroxy-9,15-dioxo-2,3,4,5-tetranor-prostane-1,20-dioic acid (PGE-M), respectively. The impact of celecoxib (400 mg b.i.d. for 7 days) on prostanoid biosynthesis and 14 circulating biomarkers of angiogenesis was evaluated in FAP. Intestinal tumorigenesis was associated with enhanced urinary TX-M levels, but unaffected by celecoxib, suggesting the involvement of a COX-1-dependent pathway, presumably from platelets. This was supported by the finding that in cocultures of a human colon adenocarcinoma cell line (HT-29) and platelets enhanced TXA(2) generation was almost completely inhibited by pretreatment of platelets with aspirin, a preferential inhibitor of COX-1. In FAP, celecoxib profoundly suppressed PGE(2) and PGI(2) biosynthesis that was associated with a significant increase in circulating levels of most proangiogenesis proteins but also the antiangiogenic tissue inhibitor of metalloproteinase 2. Urinary PGI-M, but not PGE-M, was negatively correlated with circulating levels of fibroblast growth factor 2 and angiogenin. In conclusion, inhibition of tumor COX-2-dependent PGE(2) by celecoxib may reduce tumor progression. However, the coincident depression of vascular PGI(2), in a context of enhanced TXA(2) biosynthesis, may modulate the attendant angiogenesis, contributing to variability in the chemopreventive efficacy of COX-2 inhibitors such as celecoxib.


Subject(s)
Adenomatous Polyposis Coli/blood , Neovascularization, Physiologic/physiology , Prostaglandins/biosynthesis , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Adenomatous Polyposis Coli/drug therapy , Adult , Animals , Celecoxib , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , Epoprostenol/antagonists & inhibitors , Epoprostenol/biosynthesis , Female , HT29 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Neovascularization, Physiologic/drug effects , Prostaglandins/blood , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , Thromboxane A2/antagonists & inhibitors , Thromboxane A2/biosynthesis , Treatment Outcome , Vascular Endothelial Growth Factor A/blood
15.
Free Radic Biol Med ; 52(1): 182-90, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22067901

ABSTRACT

Benfotiamine, a lipid-soluble analogue of vitamin B1, is a potent antioxidant that is used as a food supplement for the treatment of diabetic complications. Our recent study (U.C. Yadav et al., Free Radic. Biol. Med. 48:1423-1434, 2010) indicates a novel role for benfotiamine in the prevention of bacterial endotoxin, lipopolysaccharide (LPS)-induced cytotoxicity and inflammatory response in murine macrophages. Nevertheless, it remains unclear how benfotiamine mediates anti-inflammatory effects. In this study, we investigated the anti-inflammatory role of benfotiamine in regulating arachidonic acid (AA) pathway-generated inflammatory lipid mediators in RAW264.7 macrophages. Benfotiamine prevented the LPS-induced activation of cPLA2 and release of AA metabolites such as leukotrienes, prostaglandin E2, thromboxane 2 (TXB2), and prostacyclin (PGI2) in macrophages. Further, LPS-induced expression of AA-metabolizing enzymes such as COX-2, LOX-5, TXB synthase, and PGI2 synthase was significantly blocked by benfotiamine. Furthermore, benfotiamine prevented the LPS-induced phosphorylation of ERK1/2 and expression of transcription factors NF-κB and Egr-1. Benfotiamine also prevented the LPS-induced oxidative stress and protein-HNE adduct formation. Most importantly, compared to specific COX-2 and LOX-5 inhibitors, benfotiamine significantly prevented LPS-induced macrophage death and monocyte adhesion to endothelial cells. Thus, our studies indicate that the dual regulation of the COX and LOX pathways in AA metabolism could be a novel mechanism by which benfotiamine exhibits its potential anti-inflammatory response.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Gene Expression/drug effects , Inflammation/drug therapy , Macrophages/drug effects , Signal Transduction/drug effects , Thiamine/analogs & derivatives , Animals , Arachidonic Acid/metabolism , Cell Adhesion/drug effects , Cell Death/drug effects , Cell Line , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/antagonists & inhibitors , Dinoprostone/biosynthesis , Dinoprostone/metabolism , Epoprostenol/antagonists & inhibitors , Epoprostenol/biosynthesis , Epoprostenol/metabolism , Extracellular Matrix Proteins/antagonists & inhibitors , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Inflammation/genetics , Inflammation/metabolism , Leukotrienes/biosynthesis , Leukotrienes/metabolism , Lipopolysaccharides/adverse effects , Macrophages/cytology , Macrophages/metabolism , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/metabolism , Thiamine/pharmacology , Thromboxane B2/antagonists & inhibitors , Thromboxane B2/biosynthesis , Thromboxane B2/metabolism
16.
J Physiol Pharmacol ; 62(1): 21-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21451206

ABSTRACT

Curcumin regulates prostaglandin (PG) synthesis in a variety of cells. PGE2 and PGI2 are generated from arachidonic acid (AA) by cyclooxygenases 1 and 2 (COX-1 and COX-2) and the synthase (PGES and PGI2S) pathways. This study evaluates the in vitro effect of curcumin on the expression of COX-1, COX-2, PGI2S and microsomal PGES-1 (mPGES-1), and the production of PGE2 and PGI2 in human coronary artery endothelial cells (HCAEC). HCAEC monolayers were incubated with curcumin and the expression of mRNA, protein and the production of PGI2 and PGE2 were quantified. Incubation of HCAEC with curcumin led to a time and concentration-dependent increases in COX-2 mRNA with a small but significant decrease in COX-1 mRNA expression. Curcumin also stimulated the expression of PGI2S and mPGES-1 mRNA. Although curcumin stimulated COX-2, PGI2S and mPGES-1 gene expression, it failed to increase PGI2 or PGE2 production. Interestingly, supplementation of the culture medium with AA increased prostanoid production by both quiescent and curcumin-treated cells. However, in comparison to the quiescent cells, the prostanoid production by curcumin-treated cells was markedly enhanced as AA concentrations in the medium were increased, and the enhanced prostanoid production was blocked by the presence of COX-2 specific inhibitor. Taken together, these results suggest that curcumin regulates prostanoid homeostasis in HCAEC by modulating multiple steps including the expression of COX-1, COX-2, PGI2S and mPGES-1.


Subject(s)
Coronary Vessels/metabolism , Curcumin/pharmacology , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 2/biosynthesis , Dinoprostone/biosynthesis , Endothelial Cells/metabolism , Epoprostenol/biosynthesis , Arachidonic Acid/pharmacology , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/drug effects , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Dinoprostone/antagonists & inhibitors , Dinoprostone/genetics , Dinoprostone/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Epoprostenol/antagonists & inhibitors , Epoprostenol/genetics , Epoprostenol/metabolism , Gene Expression Regulation/drug effects , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Prostaglandin-E Synthases
17.
Brain Res ; 1346: 121-31, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20529667

ABSTRACT

To analyze ischemia-reperfusion effects on the cerebrovascular adrenergic response, the left middle cerebral artery (MCA) of anesthetized goats was occluded for 120 min and reperfused for 60 min. Isolated segments from the left (ischemic) and right (control) MCA exhibited isometric constriction in response to noradrenaline (10(-8)-10(-4)M, in the presence of beta-adrenoceptors blockade), phenylephrine (alpha(1)-adrenoceptors agonist, 10(-8)-10(-4)M), B-HT-920 (alpha(2)-adrenoceptors agonist, 10(-7) - 3 x 10(-3)M) or tyramine (indirect sympatheticomimetic amine, 10(-8)-10(-4)M), but this constriction was greater in ischemic arteries. The cyclooxygenase (COX) inhibitor meclofenamate (10(-5)M) augmented the response to noradrenaline only in control arteries. The prostacyclin (PGI(2)) synthesis inhibitor tranylcypromine (TCP, 10(-5)M) increased the response to noradrenaline in control arteries and reduced it in ischemic arteries. The thromboxane A(2) (TXA(2)) synthase inhibitor furegrelate (10(-6)M) did not modify the noradrenaline effect in both types of arteries, whereas the TXA(2) receptor antagonist SQ 29 548 (10(-5)M) and the COX-2 inhibitor NS-398 (10(-6)M) decreased the response to noradrenaline only in ischemic arteries. PGI(2) caused a small relaxation in control arteries and a small contraction in ischemic arteries. alpha-Adrenoceptors and COX-2 protein expression and the metabolite of PGI(2) were augmented in ischemic arteries. Therefore, ischemia-reperfusion may increase the cerebrovascular responsiveness to noradrenaline, through upregulation of alpha-adrenoceptors and increased COX-2-derived PGI(2) exerting a vasoconstrictor action. After ischemia-reperfusion, noradrenaline might increase PGI(2) production thus contributing to adrenergic vasoconstriction and/or PGI(2) would potentiate the noradrenaline effects.


Subject(s)
Cerebral Arteries/physiology , Epoprostenol/physiology , Receptors, Adrenergic, alpha/physiology , Reperfusion Injury/physiopathology , Sympathetic Nervous System/physiopathology , Adrenergic alpha-Agonists/pharmacology , Animals , Blotting, Western , Cyclooxygenase 2 Inhibitors/pharmacology , Dose-Response Relationship, Drug , Epoprostenol/antagonists & inhibitors , Female , Goats , Hemodynamics/physiology , In Vitro Techniques , Middle Cerebral Artery/physiology , Muscle, Smooth, Vascular/drug effects , Potassium Chloride/pharmacology , Receptors, Adrenergic, alpha/drug effects , Receptors, Thromboxane A2, Prostaglandin H2/antagonists & inhibitors , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasoconstrictor Agents/pharmacology
18.
FEMS Yeast Res ; 9(3): 420-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19207291

ABSTRACT

Prostanoids are a large family of lipid mediators originating from prostaglandin H synthase (PGHS) activity on the 20-carbon polyunsaturated fatty acids dihomo-gamma-linolenic acid (DGLA), arachidonic acid (AA) and eicosapentaenoic acid. The two mouse PGHS isoforms, PGHS-1 and PGHS-2, were expressed in Saccharomyces cerevisiae (yeast), as was a signal-peptide-deleted version of PGHS-1 (PGHS-1MA). PGHS-1 showed high activity with both AA and DGLA as substrate, whereas PGHS-2 activity was high with DGLA but low with AA. Signal peptide removal reduced the activity of PGHS-1MA by >50% relative to PGHS-1, but the residual activity indicated that correct targeting to the lumen of the endoplasmic reticulum may not be necessary for enzyme function. Coexpression of PGHS-1 with cDNAs encoding mouse prostaglandin I synthase and thromboxane A synthase, and with Trypanosoma brucei genomic DNA encoding prostaglandin F synthase in AA-supplemented yeast cultures resulted in production of the corresponding prostanoids, prostaglandin I(2), thromboxane A(2) and prostaglandin F(2alpha). The inhibitory effects of nonsteroidal anti-inflammatory drugs (NSAIDs) on prostanoid production were tested on yeast cells expressing PGHS-1 in AA-supplemented culture. Dose-dependent inhibition of prostaglandin H(2) production by aspirin, ibuprofen and indomethacin demonstrated the potential utility of this simple expression system in screening for novel NSAIDs.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dinoprost/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Epoprostenol/antagonists & inhibitors , Saccharomyces cerevisiae/metabolism , Thromboxane A2/antagonists & inhibitors , Animals , Biosynthetic Pathways , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 1/genetics , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/genetics , Dinoprost/biosynthesis , Epoprostenol/biosynthesis , Intramolecular Oxidoreductases/biosynthesis , Intramolecular Oxidoreductases/genetics , Mice , Models, Biological , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Saccharomyces cerevisiae/genetics , Thromboxane A2/biosynthesis , Thromboxane-A Synthase/biosynthesis , Thromboxane-A Synthase/genetics , Trypanosoma brucei brucei/genetics
19.
Coll Antropol ; 33 Suppl 2: 25-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20120521

ABSTRACT

Prostaglandins (PGs) are lipid compounds that mediate the variety of physiological and pathological functions in almost all body tissues and organs. Prostacyclin (prostaglandin 12, PGI2), which is synthesized by the vascular endothelium, is a potent vasodilator, inhibits the aggregation of platelets in vitro and has cytoprotective effect on gastrointestinal mucosa. The aim of this study was to determine whether PGI2 is playing a role in host defense to toxic effect of acetaminophen (APAP). This was investigated in C57Black/6 mice which were intoxicated with single lethal or high sublethal dose of APAP. APAP was administered to mice by gastric lavage and PGI2 agonists or antagonists were given intraperitoneally (i.p.) 30 minutes before or 2 hours after administration of APAP. The toxicity of APAP was determined by observing the survival of mice during 48 hours, by measuring the concentration of alanine-aminotransferase (ALT) in plasma 20-24 hours after APAP administration, and by liver histology. Mice were given either pure PGI2 (PGI2 sodium salt), its stable agonist (iloprost) or inhibitor of prostacyclin (IP)-receptor (CAY-10441). The results have shown that PGI2 exibits a strong hepatoprotective effect when it was given to mice either before or after APAP (both increase of survival of mice and decrease of plasma ALT levels were statistical significant). Iloprost has not shown a similar effect and CAY-10441 increased toxic effect of APAP if given 2 hours after its administration. Histopathological changes in liver generally support these findings. These investigations support the view that PGI2 is involved in defense of organism to noxious effects of xenobiotics on liver.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Chemical and Drug Induced Liver Injury/etiology , Epoprostenol/physiology , Alanine Transaminase/blood , Alanine Transaminase/drug effects , Animals , Chemical and Drug Induced Liver Injury/physiopathology , Chemical and Drug Induced Liver Injury/prevention & control , Cytoprotection , Epoprostenol/agonists , Epoprostenol/antagonists & inhibitors , Epoprostenol/pharmacology , Female , Iloprost , Male , Mice , Mice, Inbred C57BL , Receptors, Epoprostenol
20.
J Am Coll Cardiol ; 52(20): 1628-36, 2008 Nov 11.
Article in English | MEDLINE | ID: mdl-18992652

ABSTRACT

OBJECTIVES: We studied the association between the frequency, dose, and duration of different nonsteroidal anti-inflammatory drugs (NSAIDs) and the risk of myocardial infarction (MI) in the general population. We verified whether the degree of inhibition of whole blood cyclooxygenase (COX)-2 by average circulating drug levels can be a surrogate biochemical predictor of the risk of MI by NSAIDs. BACKGROUND: There is evidence that both traditional NSAIDs and selective inhibitors of COX-2 may increase the risk of MI. METHODS: From the THIN (The Health Improvement Network) database, we identified 8,852 cases of nonfatal MI in patients 50 to 84 years old between 2000 and 2005 and conducted a nested case-control analysis. We correlated the risk of MI with the degree of inhibition of platelet COX-1 and monocyte COX-2 in vitro by average therapeutic concentrations of individual NSAIDs. RESULTS: The risk of MI was increased with current use of NSAIDs (relative risk [RR]: 1.35; 95% confidence interval [CI]: 1.23 to 1.48). The risk increased with treatment duration and daily dose. We found a significant correlation between the degree of inhibition in vitro of whole blood COX-2 (r(2) = 0.7458, p = 0.0027), but not whole blood COX-1 (r(2) = 0.0007, p = 0.947), and the risk of MI associated with individual NSAIDs that lacked complete suppression (> or =95%) of platelet COX-1 activity. Individual NSAIDs with a degree of COX-2 inhibition <90% at therapeutic concentrations presented an RR of 1.18 (95% CI: 1.02 to 1.38), whereas those with a greater COX-2 inhibition had an RR of 1.60 (95% CI: 1.41 to 1.81). CONCLUSIONS: Our findings suggest that the variable risk of MI among NSAIDs that do not inhibit platelet COX-1 completely and persistently is largely related to their extent of COX-2 inhibition.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Myocardial Infarction/chemically induced , Aged , Aged, 80 and over , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Epoprostenol/antagonists & inhibitors , Humans , Middle Aged , Retrospective Studies , Risk Assessment
SELECTION OF CITATIONS
SEARCH DETAIL
...