Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
1.
J Thromb Thrombolysis ; 51(2): 260-264, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33170486

ABSTRACT

Arachidonic acid (AA)-induced platelet aggregation (PA) and serum thromboxane B2 (TxB2) inhibition are widely used to indicate cyclooxygenase-1 activity and the antiplatelet effect of acetylsalicylic acid (ASA). Despite decades of investigations, the relation between these measurements remains unclear. We sought to evaluate the relation between AA-PA and serum TxB2 inhibition. We serially measured AA-PA (conventional aggregation), serum TxB2, plasma ASA and salicylic acid (SA) (liquid chromatography-mass spectrometry), and urinary 11-dehydro thromboxane B2 (u11-dh TxB2) (enzyme-linked immunosorbent assay) levels at 10 times over 24 hours in seventeen healthy volunteers receiving a single dose of 162 mg chewed and swallowed ASA (n = 6), 50 mg inhaled ASA (n = 6), or 100 mg inhaled ASA (n = 5) (ClinicalTrials.gov Identifier: NCT04328883, April 1, 2020). Baseline variability was more pronounced with serum TxB2 (31-680 ng/mL) as compared to maximal AA-PA (65-81%) and u11-dh TxB2 (1556-4440 pg/mg creatinine). The relation between serum TxB2 inhibition and AA-PA was stepwise; after 30-40% inhibition of serum TxB2, AA-PA fell to < 5%. By receiver operating characteristic curve analysis using AA-PA < 5% to define aspirin responsiveness, serum TxB2 inhibition > 49% and u11-dh TxB2 < 1520 pg/mg creatinine met the definition. Our study demonstrates a non-linear relation between serum TxB2 inhibition and AA-PA. Aggregation was nil once TxB2 inhibition reached > 49%. Moreover, these results suggest that the definition of > 95% inhibition of serum TxB2 to indicate the level of platelet COX-1 inhibition needed for clinical efficacy may be overestimated and should be re-considered in future translational research investigations that attempt to link the clinical efficacy of ASA with a laboratory measurement cutoff.


Subject(s)
Aspirin/pharmacology , Blood Platelets/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Thromboxane B2/antagonists & inhibitors , Adult , Aspirin/administration & dosage , Dose-Response Relationship, Drug , Female , Humans , Male , Pilot Projects , Platelet Aggregation Inhibitors/administration & dosage , Thromboxane B2/blood , Young Adult
2.
Biomolecules ; 10(9)2020 09 10.
Article in English | MEDLINE | ID: mdl-32927723

ABSTRACT

Pulmonary fibrosis is a severe lung disease with progressive worsening of dyspnea, characterized by chronic inflammation and remodeling of lung parenchyma. Carbonic anhydrases are a family of zinc-metallo-enzymes that catalyze the reversible interconversion of carbon-dioxide and water to bicarbonate and protons. Carbonic Anhydrase Inhibitor (CAI) exhibited anti-inflammatory effects in animals with permanent-middle-cerebral artery occlusion, arthritis and neuropathic pain. The pharmacological profile of a new class of hybrid compounds constituted by a CAI connected to a Nonsteroidal-Anti-Inflammatory Drug (NSAID) was studied in the modulation of inflammation and fibrosis. In-vitro tests were performed to assess their effects on cyclo-oxygenase enzyme (COX)-1 and COX-2, namely inhibition of platelet aggregation and thromboxane B2 production in the human-platelet-rich plasma, and reduction of Prostaglandin-E2 production in lipopolysaccharide-treated-RAW-264.7 macrophage cell line. The activity of compound 3, one of the most active, was studied in a model of bleomycin-induced lung fibrosis in C57BL/6 mice. The hybrid compounds showed a higher potency in inhibiting PGE2 production, but not in modifying the platelet aggregation and the TXB2 production in comparison to the reference molecules, indicating an increased activity in COX-2 inhibition. In the in-vivo murine model, the compound 3 was more effective in decreasing inflammation, lung stiffness and oxidative stress in comparison to the reference drugs given alone or in association. In conclusion, these CAI-NSAID hybrid compounds are promising new anti-inflammatory drugs for the treatment of lung chronic inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carbonic Anhydrase Inhibitors/pharmacology , Gene Expression/drug effects , Oxidative Stress/drug effects , Platelet Aggregation/drug effects , Pulmonary Fibrosis/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Bleomycin/administration & dosage , Blood Platelets/drug effects , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrases/genetics , Carbonic Anhydrases/metabolism , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/antagonists & inhibitors , Dinoprostone/biosynthesis , Disease Models, Animal , Drug Design , Humans , Inflammation/prevention & control , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , RAW 264.7 Cells , Structure-Activity Relationship , Thromboxane B2/antagonists & inhibitors , Thromboxane B2/biosynthesis
3.
J Thromb Thrombolysis ; 48(4): 554-562, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31420787

ABSTRACT

Aspirin (acetylsalicylic acid, ASA) can lead to gastrointestinal mucosal injury through disruption of its protective phospholipid bilayer. A liquid formulation of a novel pharmaceutical lipid-aspirin complex (PL-ASA) was designed to prevent this disruption. We sought to determine the pharmacokinetic (PK)/pharmacodynamic (PD) characteristics of PL-ASA compared with immediate release aspirin (IR-ASA). In this active-control crossover study, 32 healthy volunteers were randomized to receive 1 of 2 dose levels (a single dose of 325 mg or 650 mg) of either PL-ASA or IR-ASA. After a 2-week washout period between treatment assignments, subjects received a single dose of the alternative treatment, at the same dose level. The primary objectives of the study were to assess, for PL-ASA and IR-ASA at 325 mg and 650 mg dose levels, PK and PD bioequivalence, and safety, over a 24-h period after administration of both drugs. PK parameters were similar for PL-ASA and IR-ASA, and met FDA-criteria for bioequivalence. Regarding PD, both drugs also showed Cmin TxB2 values below 3.1 ng/mL (cut-off associated with decreased cardiovascular events) and > 99% inhibition of serum TxB2 ( ≥ 95% inhibition represents the cut-off for aspirin responders) along with similar results in several secondary PK/PD parameters. There were no serious adverse events or changes from baseline in vital signs or laboratory values in either of the 2 treatment groups. PL-ASA's novel liquid formulation has similar PK and PD performance compared with IR-ASA, supporting functional and clinical equivalence. These data coupled with the improved gastric safety of PL-ASA suggest that this novel formulation may exhibit an improved benefit-risk profile, warranting evaluation in future trials.Clinical trial registration: http://www.clinicaltrials.gov . Unique Identifier: NCT04008979.


Subject(s)
Aspirin/administration & dosage , Drug Carriers/chemistry , Lipids/therapeutic use , Adult , Aspirin/adverse effects , Aspirin/pharmacokinetics , Cross-Over Studies , Gastrointestinal Tract/pathology , Humans , Middle Aged , Mucous Membrane/injuries , Therapeutic Equivalency , Thromboxane B2/antagonists & inhibitors , Young Adult
4.
J Thromb Thrombolysis ; 45(1): 18-26, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29198079

ABSTRACT

Aspirin is the dominant antiplatelet therapy for cardiovascular disease. Naproxen is frequently used in aspirin-treated patients and may influence the antiplatelet effect of aspirin. We evaluated the pharmacodynamic interaction (lower bound of the one-sided 95% CI for serum TxB2 inhibition < 95%) between 220 mg immediate-release naproxen sodium (once or twice daily) and 81 mg daily immediate release aspirin at various dosing intervals. There was no interaction during the first day of concurrent treatment. After 10 days, irrespective of the timing and dose of naproxen in relation to aspirin dosing, a pharmacodynamic interaction occurred which persisted after discontinuing naproxen. In the control group (aspirin alone), the lower bound for serum TxB2 inhibition was > 98% at all time points. The clinical relevance of these observations remains unknown and merits further investigation since over-the-counter naproxen is widely used to relieve pain by individuals taking low dose aspirin for cardioprotection. CLINICAL TRIAL REGISTRATION: NCT02229461.


Subject(s)
Aspirin/administration & dosage , Naproxen/administration & dosage , Thromboxanes/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal , Dose-Response Relationship, Drug , Drug Interactions , Female , Humans , Male , Middle Aged , Platelet Aggregation Inhibitors , Thromboxane B2/antagonists & inhibitors
5.
Blood Coagul Fibrinolysis ; 27(2): 141-6, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26340455

ABSTRACT

Vitis labrusca is a grapevine that has antioxidant, neuroprotective, hepatoprotective, and anticarcinogenic activity. However, the antithrombotic effect of Vitis labrusca leaves on platelets is yet to be ascertained. We investigated the inhibitory effect of V. labrusca leaf extract (VLE) on platelet aggregation in vitro and ex vivo. The thromboxane B2 (TXB2) and serotonin concentrations were measured by ELISA. The flavonoids content was measured by ultraperformance liquid chromatography (UPLC). The antithrombotic activity of VLE was evaluated using various agonists in vitro. VLE strongly inhibited adenosine diphosphate (ADP)-induced platelet aggregation. In rats, VLE treatment (100 mg/kg) reduced ADP-stimulated platelet aggregation, without affecting tail bleeding and coagulation time. Moreover, VLE significantly suppressed TXB2 and serotonin secretion. UPLC analysis indicated that VLE contains quercetin, isorhamnetin, and rutin. Our results indicate that VLE possesses antiplatelet activity via the suppression of TXB2 and serotonin, without affecting bleeding. Further, we identified the flavonoids present in VLE. Thus, VLE may be a potential agent for the prevention of cardiovascular diseases.


Subject(s)
Blood Platelets/drug effects , Flavonoids/pharmacology , Plant Extracts/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Vitis/chemistry , Adenosine Diphosphate/pharmacology , Animals , Dose-Response Relationship, Drug , Flavonoids/chemistry , Flavonoids/isolation & purification , Male , Plant Extracts/chemistry , Plant Leaves/chemistry , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/isolation & purification , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Thromboxane B2/antagonists & inhibitors , Thromboxane B2/metabolism
6.
Platelets ; 27(2): 105-9, 2016.
Article in English | MEDLINE | ID: mdl-26083594

ABSTRACT

The aim of this study is to quantify the extent and recovery of platelet inhibition after administration of indobufen and aspirin in healthy volunteers. Indobufen inhibits platelet aggregation by reversibly inhibiting the platelet cyclooxygenase enzyme, thereby suppressing thromboxane synthesis. Twenty healthy volunteers completed the study and received aspirin (200 mg/day for 2 weeks) followed by a 4-week washout period and then indobufen (200 mg twice a day for 2 weeks). The percent (%) inhibition of platelet aggregation (IPA) was assessed using arachidonic acid (0.5 mg/ml) and adenosine diphosphate (5 µM) at 4, 12, 24 and 48 hours after last dose of each drug. IPA assessed using arachidonic acid as the agonist was similar at 4 hours after the last dose of indobufen (81.07 ± 9.36%) and aspirin (96.99 ± 0.29%, p = 0.10), but significantly lower at 12 hours (74.04 ± 9.55% vs. 97.94 ± 0.28%, p = 0.02), 24 hours (33.39 ± 11.13% vs. 97.48 ± 0.32%, p < 0.001) and 48 hours (14.12 ± 9.74% vs. 98.22 ± 0.31%, p < 0.001) after indobufen, compared to the relative values for aspirin. IPA assessed using adenosine diphosphate as the agonist was similar in the two groups at 4, 12 and 24 hours after the last dose, but significantly lower 48 hours after the last dose of indobufen, compared to the relative value for aspirin (1.98 ± 3.57% vs. 12.61 ± 2.71%, p = 0.002). Indobufen (200 mg twice a day) caused equivalent initial inhibition of platelet aggregation to aspirin (200 mg daily), and the anti-aggregation effect diminished faster than after aspirin.


Subject(s)
Aspirin/pharmacology , Blood Platelets/drug effects , Isoindoles/pharmacology , Phenylbutyrates/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Adenosine Diphosphate/pharmacology , Adolescent , Adult , Aged , Arachidonic Acid/antagonists & inhibitors , Arachidonic Acid/pharmacology , Cross-Over Studies , Drug Administration Schedule , Female , Healthy Volunteers , Humans , Male , Middle Aged , Platelet Function Tests , Prostaglandin-Endoperoxide Synthases/blood , Thromboxane B2/antagonists & inhibitors , Thromboxane B2/blood
7.
PLoS One ; 10(3): e0120157, 2015.
Article in English | MEDLINE | ID: mdl-25785937

ABSTRACT

Inflammation and altered immunity are recognized components of severe pulmonary arterial hypertension in human patients and in animal models of PAH. While eicosanoid metabolites of cyclooxygenase and lipoxygenase pathways have been identified in the lungs from pulmonary hypertensive animals their role in the pathogenesis of severe angioobliterative PAH has not been examined. Here we investigated whether a cyclooxygenase-2 (COX-2) inhibitor or diethylcarbamazine (DEC), that is known for its 5-lipoxygenase inhibiting and antioxidant actions, modify the development of PAH in the Sugen 5416/hypoxia (SuHx) rat model. The COX-2 inhibitor SC-58125 had little effect on the right ventricular pressure and did not prevent the development of pulmonary angioobliteration. In contrast, DEC blunted the muscularization of pulmonary arterioles and reduced the number of fully obliterated lung vessels. DEC treatment of SuHx rats, after the lung vascular disease had been established, reduced the degree of PAH, the number of obliterated arterioles and the degree of perivascular inflammation. We conclude that the non-specific anti-inflammatory drug DEC affects developing PAH and is partially effective once angioobliterative PAH has been established.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Diethylcarbamazine/pharmacology , Hypertension, Pulmonary/drug therapy , Hypoxia/drug therapy , Lipoxygenase Inhibitors/pharmacology , Lung/drug effects , Animals , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/metabolism , Arterioles/drug effects , Arterioles/enzymology , Arterioles/pathology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprost/antagonists & inhibitors , Dinoprost/biosynthesis , Dinoprostone/antagonists & inhibitors , Dinoprostone/biosynthesis , Humans , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/genetics , Hypoxia/enzymology , Hypoxia/genetics , Hypoxia/pathology , Inflammation , Leukotriene D4/antagonists & inhibitors , Leukotriene D4/biosynthesis , Lung/enzymology , Lung/pathology , Male , Prostaglandins F/antagonists & inhibitors , Prostaglandins F/biosynthesis , Pulmonary Artery/drug effects , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Thromboxane B2/antagonists & inhibitors , Thromboxane B2/biosynthesis , Ventricular Function, Right/drug effects
8.
N Z Vet J ; 63(2): 92-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25075617

ABSTRACT

AIMS: To determine the pharmacokinetics of cimicoxib and to assess the inhibition of cyclooxygenase (COX) after a 5 mg/kg, single oral administration in horses that were fasted or fed. METHODS: The study was conducted using an open, single dose (5 mg/kg), two treatment (fasted and fed), two-period, crossover design with a 2-week interval between dosages. Six healthy mares received 5 mg/kg of cimicoxib via nasogastric tube after fasting for 12 hours, or 2 hours after feeding. After administration, blood samples were collected for up to 24 hours and plasma used for pharmacokinetic analysis. Additional serum and plasma samples were used to measure concentrations of thromboxane B2 (TXB2) and prostaglandin E2 (PGE2), to assess COX-1 and -2 inhibition, respectively. RESULTS: Following cimicoxib administration, the mean maximum plasma concentration was 0.16 (SD 0.01) µg/mL and 0.14 (SD 0.03) µg/mL in fasted and fed groups, respectively. The mean time taken to reach maximum plasma concentration was longer in the fed group (5.91 (SD 3.23) hours) compared with the fasted group (3.25 (SD 1.17) hours), but this difference was not significant (p=0.12). The mean maximal inhibition of TXB2 was 62.4 (SD 13.8)% and 54.6 (SD 15.4)%, and of PGE2 was 72.1 (SD 43.3)% and 68.5 (SD 24.4)%, in fasted and fed horses, respectively. CONCLUSION: In the present study, although the COX-2 selective action of cimicoxib was not apparent, a relatively low concentration of cimicoxib resulted in both COX-1 and -2 inhibition in horses. Further investigations are required to establish an optimal dosage regimen and safety profile before clinical trials are initiated.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Food Deprivation , Horses/blood , Horses/metabolism , Imidazoles/pharmacokinetics , Sulfonamides/pharmacokinetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/blood , Area Under Curve , Dinoprostone/antagonists & inhibitors , Dinoprostone/blood , Dose-Response Relationship, Drug , Female , Half-Life , Imidazoles/blood , Pilot Projects , Sulfonamides/blood , Thromboxane B2/antagonists & inhibitors , Thromboxane B2/blood
9.
J Nutr Biochem ; 26(2): 130-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25458529

ABSTRACT

Dietary conjugated linoleic acid (CLA) reduces indicators of early renal disease progression and the associated elevated cyclooxygenase (COX) levels in young obese rats with obesity-associated nephropathy (OAN). Therefore, renal function and injury and COX and its metabolites were assessed in obese fa/fa Zucker rats with more advanced renal disease. Obese rats at 16 weeks of age were provided with either cis(c)9, trans(t)11 (fa/fa-9,11) or t10,c12 (fa/fa-10,12) CLA for 8 weeks, and compared to lean (lean-CTL) and obese (fa/fa-CTL) rats provided the control diet without CLA. Obese rats displayed significantly reduced renal function and increased renal injury compared to lean rats. In the obese rat groups, glomerular hypertrophy was reduced in both CLA-supplemented groups. While all other measures of renal function or injury were not different in fa/fa-9,11 compared to fa/fa-CTL rats, the fa/fa-10,12 rats had greater renal hypertrophy, glomerular fibrosis, fibrosis, tubular casts and macrophage infiltration compared to the fa/fa-CTL and fa/fa-9,11 groups. The fa/fa-10,12 group also had elevated levels of renal COX1, which was associated with increased levels of two oxylipins produced by this enzyme, 6-keto-prostaglandin F(1α), and thromboxane B2. Renal linoleic acid and its lipoxygenase products also were lower in obese compared to lean rats, but CLA supplementation had no effect on these or any other lipoxygenase oxylipins. In summary, supplementation with c9,t11 CLA did not improve more advanced OAN and t10,c12 CLA worsened the renal pathology. Altered production of select COX1 derived oxylipins was associated with the detrimental effect of the t10,c12 isomer.


Subject(s)
Aging , Dietary Supplements/adverse effects , Kidney/pathology , Linoleic Acids, Conjugated/adverse effects , Obesity/physiopathology , Oxylipins/agonists , Renal Insufficiency/etiology , 6-Ketoprostaglandin F1 alpha/agonists , 6-Ketoprostaglandin F1 alpha/antagonists & inhibitors , 6-Ketoprostaglandin F1 alpha/metabolism , Animals , Cyclooxygenase 1/metabolism , Disease Progression , Fibrosis , Hypertrophy , Kidney/immunology , Kidney/metabolism , Kidney/physiopathology , Macrophage Activation , Membrane Proteins/agonists , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Obesity/immunology , Oxylipins/antagonists & inhibitors , Oxylipins/metabolism , Rats, Zucker , Renal Insufficiency/metabolism , Renal Insufficiency/pathology , Renal Insufficiency/physiopathology , Severity of Illness Index , Thromboxane B2/agonists , Thromboxane B2/antagonists & inhibitors , Thromboxane B2/metabolism
10.
Bioorg Med Chem Lett ; 24(1): 344-8, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24279991

ABSTRACT

A new regular diterpene possessing an unusual 1,6-anti-3-methylcyclohex-2-en-1-ol ring system, dactyloditerpenol acetate (1), has been extracted from the tropical sea hare Aplysia dactylomela and its stereostructure elucidated by spectroscopic methods. The absolute configuration of 1 was determined as 1S, 6S, 7R, 10S, and 11R by application of Kishi's method for the assignment of absolute configuration of alcohols. The new diterpene potently inhibited in vitro thromboxane B2 (TXB2) (IC50 0.4µM) and superoxide anion (O2(-)) (IC50 1µM) generation from Escherichia coli lipopolysaccharide (LPS)-activated rat neonatal microglia, with concomitant low short-term toxicity.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aplysia/chemistry , Diterpenes/pharmacology , Superoxides/antagonists & inhibitors , Thromboxane B2/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Diterpenes/chemistry , Diterpenes/isolation & purification , Dose-Response Relationship, Drug , Escherichia coli/chemistry , Escherichia coli/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Microglia/drug effects , Microglia/metabolism , Models, Molecular , Molecular Conformation , Rats , Stereoisomerism , Structure-Activity Relationship , Superoxides/metabolism , Thromboxane B2/biosynthesis
11.
Clin Drug Investig ; 34(1): 19-25, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24132773

ABSTRACT

BACKGROUND: Low-dose acetylsalicylic acid (ASA; aspirin) is well-established as a platelet anti-aggregating agent for the secondary prevention of cardiovascular events. OBJECTIVES: The objective of this study was to investigate the non-inferiority of a novel ASA 75 mg soft-gel capsule formulation compared with a marketed powder for oral solution in terms of reduction in serum thromboxane B2 (TXB2), a surrogate for platelet aggregation. Pharmacokinetics and tolerability of the products were also investigated. METHODS: In this randomised, two-way crossover study, 46 male and female healthy subjects received a single dose of the investigational products in two periods separated by a 14-day washout. Serum TXB2 and plasma ASA were determined up to 24 h post-dose. Maximum percentage of TXB2 inhibition (I max) and area under the inhibition-time curve (AUICt) were calculated. Non-inferiority was assumed if the lower limits of the 95 % confidence intervals (CIs) for the two pharmacodynamic parameters were above 85 %. RESULTS: The 95 % CI lower limits were 95.35 % for I max and 86.12 % for AUICt, i.e. within the pre-specified delta. Time to achieve I max did not differ between treatments (p = 0.88). The two formulations were bioequivalent as regards the extent of ASA exposure (area under the plasma concentration-time curve from zero to time t [AUCt] 90 % CIs 96.67-113.37); a delayed ASA absorption (later time to reach maximum plasma concentration [t max], lower maximum plasma concentration [C max]) was observed for the test product. No treatment-related adverse events were reported. CONCLUSIONS: In healthy subjects, the 75 mg soft-gel capsules were not inferior to the oral solution in terms of serum TXB2 inhibition, indicating that the novel formulation could be an effective alternative in the secondary prevention of cardiovascular events.


Subject(s)
Aspirin/administration & dosage , Aspirin/pharmacokinetics , Platelet Aggregation/drug effects , Administration, Oral , Adult , Aspirin/adverse effects , Aspirin/pharmacology , Capsules , Cross-Over Studies , Female , Healthy Volunteers , Humans , Male , Middle Aged , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacology , Solutions , Therapeutic Equivalency , Thromboxane B2/antagonists & inhibitors , Thromboxane B2/blood , Young Adult
12.
Biol Pharm Bull ; 36(8): 1341-7, 2013.
Article in English | MEDLINE | ID: mdl-23739488

ABSTRACT

Rhamnetin is a naturally occurring polyphenolic compound. In this report, experimental evidence is presented on the suppression of melanogenesis by rhamnetin using B16 murine melanoma cells (B16 cells). To document the underlying anti-melanogenic action of rhamnetin, several key biochemical mediators were quantified: superoxide (O2(•-)), nitric oxide (·NO) and peroxynitrite (ONOO(-)) in vitro, and total reactive species (RS) generation, O2(•-), ·NO and ONOO(-), reduced glutathione (GSH)/GSH-to-oxidized glutathione (GSSG) ratio, prostaglandin E2 (PGE2) and thromboxane B2 (TXB2) in B16 cells. Results revealed that rhamnetin inhibited murine tyrosinase activity, suppressed melanin content and oxidative stress, reducing O2(•-),·NO and ONOO(-) in vitro and total RS generation, O2(•-), ·NO and ONOO(-) in B16 cells, while maintaining a well-balanced GSH/GSSG ratio in B16 cells. Results further revealed that rhamnetin suppressed key pro-inflammatory mediators such as PGE2 and TXB2. Thus, these results strongly indicate that rhamnetin has powerful anti-melanogenic properties through its anti-oxidative and anti-inflammatory actions.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Melanins/antagonists & inhibitors , Quercetin/analogs & derivatives , Animals , Cell Line, Tumor , Cell Survival/drug effects , Dinoprostone/antagonists & inhibitors , Dinoprostone/metabolism , Glutathione/metabolism , Glutathione Disulfide/metabolism , Melanoma, Experimental/metabolism , Mice , Monophenol Monooxygenase/metabolism , Oxidative Stress/drug effects , Quercetin/pharmacology , Reactive Oxygen Species/metabolism , Thromboxane B2/antagonists & inhibitors , Thromboxane B2/metabolism
13.
J Vet Pharmacol Ther ; 36(4): 320-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-22913421

ABSTRACT

A tissue cage model of inflammation in calves was used to determine the pharmacokinetic and pharmacodynamic properties of individual carprofen enantiomers, following the administration of the racemate. RS(±) carprofen was administered subcutaneously both alone and in combination with intramuscularly administered oxytetracycline in a four-period crossover study. Oxytetracycline did not influence the pharmacokinetics of R(-) and S(+) carprofen enantiomers, except for a lower maximum concentration (Cmax ) of S(+) carprofen in serum after co-administration with oxytetracycline. S(+) enantiomer means for area under the serum concentration-time curve (AUC0-96 h were 136.9 and 128.3 µg·h/mL and means for the terminal half-life (T(1/2) k10 ) were = 12.9 and 17.3 h for carprofen alone and in combination with oxytetracycline, respectively. S(+) carprofen AUC0-96 h in both carprofen treatments and T(1/2) k10 for carprofen alone were lower (P < 0.05) than R(-) carprofen values, indicating a small degree of enantioselectivity in the disposition of the enantiomers. Carprofen inhibition of serum thromboxane B2 ex vivo was small and significant only at a few sampling times, whereas in vivo exudate prostaglandin (PG)E2 synthesis inhibition was greater and achieved overall significance between 36 and 72 h (P < 0.05). Inhibition of PGE2 correlated with mean time to achieve maximum concentrations in exudate of 54 and 42 h for both carprofen treatments for R(-) and S(+) enantiomers, respectively. Carprofen reduction of zymosan-induced intradermal swelling was not statistically significant. These data provide a basis for the rational use of carprofen with oxytetracycline in calves and indicate that no alteration to carprofen dosage is required when the drugs are co-administered.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Carbazoles/pharmacokinetics , Cattle/metabolism , Oxytetracycline/pharmacology , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/blood , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/blood , Area Under Curve , Carbazoles/administration & dosage , Carbazoles/blood , Cattle/blood , Cross-Over Studies , Diffusion Chambers, Culture/veterinary , Dinoprostone/antagonists & inhibitors , Drug Interactions , Half-Life , Injections, Intramuscular/veterinary , Injections, Subcutaneous/veterinary , Male , Oxytetracycline/administration & dosage , Oxytetracycline/blood , Thromboxane B2/antagonists & inhibitors
14.
J Nutr Sci Vitaminol (Tokyo) ; 59(5): 470-3, 2013.
Article in English | MEDLINE | ID: mdl-24418882

ABSTRACT

Hesperidin (HES) and glucosyl hesperidin (GHES) have antihypertensive effects. In the present study, to clarify the antihypertensive mechanisms, we compared the effects of continuous ingestion of HES and GHES in spontaneously hypertensive rats (SHRs). HES and GHES ingestion for 8 wk significantly prevented hypertension and suppressed the mRNA expression of NADPH oxidase subunits and thromboxane A2 synthase in SHR aortas. Further, hesperetin, a common metabolite of HES and GHES, reduced thromboxane B2 release from SHR aortas. These findings indicate that continuous ingestion of HES and GHES prevents hypertension via regulating the gene expression related to the modulation of vascular tone.


Subject(s)
Antihypertensive Agents/therapeutic use , Aorta, Thoracic/enzymology , Dietary Supplements , Endothelium, Vascular/enzymology , Enzyme Repression , Hesperidin/therapeutic use , Hypertension/prevention & control , Animals , Antihypertensive Agents/adverse effects , Antihypertensive Agents/chemistry , Antihypertensive Agents/metabolism , Aorta, Thoracic/metabolism , Dietary Supplements/adverse effects , Endothelium, Vascular/metabolism , Glucosides/adverse effects , Glucosides/chemistry , Glucosides/metabolism , Glucosides/therapeutic use , Hesperidin/adverse effects , Hesperidin/analogs & derivatives , Hesperidin/chemistry , Hesperidin/metabolism , Hypertension/metabolism , In Vitro Techniques , Male , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Random Allocation , Rats , Rats, Inbred SHR , Solubility , Thromboxane B2/antagonists & inhibitors , Thromboxane B2/metabolism , Thromboxane-A Synthase/antagonists & inhibitors , Thromboxane-A Synthase/genetics , Thromboxane-A Synthase/metabolism , Vascular Resistance
15.
Am J Cardiovasc Drugs ; 12(4): 217-24, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22631032

ABSTRACT

BACKGROUND: The absence of a pharmacokinetic interaction between the proton pump inhibitor esomeprazole (40 mg) and acetylsalicylic acid (aspirin, ASA; 325 mg) has previously been established. OBJECTIVE: This study set out to investigate the potential for pharmacodynamic interaction between low-dose ASA and esomeprazole in healthy volunteers, by measuring ASA antiplatelet activity. STUDY DESIGN: This was a single-center, open-label, two-period, randomized crossover study. PARTICIPANTS: Healthy male and female volunteers aged 18-75 years were included. All volunteers received ASA 81 mg once daily for 5 days prior to the study (pre-screen). Subjects were eligible for inclusion if they had aspirin reactivity units (ARU, as measured by the VerifyNow ASA assay) of <550 on Day 6. INTERVENTION: After pre-screening and a washout period of at least 14 days, eligible volunteers received ASA 81 mg with or without esomeprazole 20 mg once daily for 5 days in randomized order, with a 14-day washout between treatments. MAIN OUTCOME MEASURE: The main outcome measure was the antiplatelet activity of ASA, as assessed by ARU ratio relative to baseline in the VerifyNow ASA assay; suppression of serum thromboxane B(2) (TXB(2)) was a secondary endpoint. Statistical comparisons were made using linear mixed models. RESULTS: A total of 29 volunteers (19 aged ≥50 years; 8 women; 21 men) were evaluable for pharmacodynamic analysis (per protocol). All volunteers on both treatments achieved ARU <550 at Day 6. The geometric mean ratio of Day 6 to Day 1 (baseline) platelet aggregation was 0.70 (95% confidence interval [CI] 0.68, 0.72) with ASA alone and 0.71 (95% CI 0.69, 0.74) with ASA + esomeprazole. The ratio of platelet aggregation (ASA + esomeprazole/ASA) was 1.02 (95% CI 0.99, 1.05). ASA administered alone or with esomeprazole reduced serum TXB(2) by more than 99.5%. The ratio of suppression of serum TXB(2) levels (ASA + esomeprazole/ASA) was 1.06 (95% CI 0.88, 1.29). The combination of ASA and esomeprazole was well tolerated. CONCLUSION: No pharmacodynamic interaction between low-dose ASA and esomeprazole was found with regard to platelet function. TRIAL REGISTRATION: Registered at ClinicalTrials. gov as NCT01199328.


Subject(s)
Aspirin/pharmacology , Esomeprazole/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Proton Pump Inhibitors/pharmacology , Adolescent , Adult , Aged , Aspirin/adverse effects , Cross-Over Studies , Drug Interactions , Female , Humans , Male , Middle Aged , Platelet Aggregation/drug effects , Thromboxane B2/antagonists & inhibitors , Thromboxane B2/blood , Young Adult
16.
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
17.
Prostaglandins Other Lipid Mediat ; 94(3-4): 124-32, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21397034

ABSTRACT

Atherosclerosis is the principal cause of mortality in industrialized countries. Its development is influenced by several mediators of which thromboxane A(2) (TXA(2)) and 8-iso-PGF(2α) have recently received a lot of attention. This study aimed to investigate the effect of a dual thromboxane synthase inhibitor and thromboxane receptor antagonist (BM-573) and ASA on lesion formation in apolipoprotein E-deficient mice. The combination of ASA and BM-573 was also studied. Plasma measurements demonstrated that the treatments did not affect body weight or plasma cholesterol levels. BM-573, but not ASA, significantly decreased atherogenic lesions as demonstrated by macroscopic analysis. Both treatments alone inhibited TXB(2) synthesis but only BM-573 and the combination therapy were able to decrease firstly, plasma levels of soluble intracellular adhesion molecule-1 (sICAM-1) and soluble vascular cell adhesion molecule-1 (sVCAM-1) and secondly, the expression of these proteins in the aortic root of Apo E. These results were confirmed in endothelial cell cultures derived from human saphenous vein endothelial cells (HSVECs). In these cells, BM-573 also prevented the increased mRNA expression of ICAM-1 and VCAM-1 induced by U-46619 and 8-iso-PGF(2α). Our results show that a molecule combining receptor antagonism and thromboxane synthase inhibition is more efficient in delaying atherosclerosis in Apo E(-/-) mice than sole inhibition of TXA(2) formation.


Subject(s)
Aspirin/therapeutic use , Atherosclerosis/prevention & control , Receptors, Thromboxane/antagonists & inhibitors , Sulfonylurea Compounds/therapeutic use , Thromboxane-A Synthase/antagonists & inhibitors , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/metabolism , Animals , Apolipoproteins E/genetics , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Drug Therapy, Combination , Endothelial Cells/cytology , Humans , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Knockout , Saphenous Vein/cytology , Thromboxane A2/antagonists & inhibitors , Thromboxane A2/biosynthesis , Thromboxane B2/antagonists & inhibitors , Thromboxane B2/biosynthesis , Vascular Cell Adhesion Molecule-1/metabolism
18.
Int J Cardiol ; 150(1): 39-44, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-20207433

ABSTRACT

BACKGROUND: Platelets, long believed to be incapable of de novo protein synthesis, may retain their ability to form the cyclooxygenase (COX) enzyme once it has been inactivated by aspirin. This may explain the inefficacy of the drug to induce sustained platelet inhibition in certain patients. We evaluated the stability of platelet inhibition following once-daily enteric-coated aspirin administration. METHODS: Platelet responsiveness to aspirin was evaluated in 11 stable coronary artery disease patients on chronic aspirin therapy before and 1, 3, 8, and 24h after observed ingestion of 80-mg enteric-coated aspirin. Inhibition of the COX pathway was measured pharmacologically through plasma thromboxane (Tx) B(2) levels, and functionally by light transmission aggregometry in response to arachidonic acid. COX-independent platelet activity was measured in response to adenosine diphosphate, epinephrine and collagen. RESULTS: Plasma TxB(2) levels showed profound inhibition of TxA(2) formation, which was stable throughout 24h, in all but 1 subject. This subject had optimal response to aspirin (inhibition of platelet TxA(2) production within 1h), but recovered the ability to synthesize TxA(2) within 24h of aspirin ingestion. Arachidonic acid-induced platelet aggregation closely mirrored TxB(2) formation in this patient, portraying a functional ability of the platelet to aggregate within 24h of aspirin ingestion. COX-independent platelet aggregation triggered TxA(2) production to a similar extent in all patients, likely through signal-dependent protein synthesis. CONCLUSIONS: COX-dependent platelet activity is recovered in certain individuals within 24h of aspirin administration. Further research should consider increasing aspirin dosing frequency to twice daily, to allow sustained inhibition in such subjects.


Subject(s)
Aspirin/therapeutic use , Coronary Artery Disease/drug therapy , Coronary Artery Disease/enzymology , Cyclooxygenase Inhibitors/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Prostaglandin-Endoperoxide Synthases , Adult , Aspirin/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Female , Genetic Heterogeneity/drug effects , Humans , Male , Middle Aged , Platelet Aggregation Inhibitors/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Thromboxane A2/antagonists & inhibitors , Thromboxane A2/metabolism , Thromboxane B2/antagonists & inhibitors , Thromboxane B2/metabolism
19.
Phytomedicine ; 18(4): 278-84, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-20864324

ABSTRACT

Glycyrrhiza glabra and its phytoconstituents have been known to possess widespread pharmacological properties as an anti-inflammatory, anti-viral, antitumour and hepatoprotective drug. In this study, we examined the inhibitory potential of extract of G. glabra (GutGard™) root and its phytoconstituents (glabridin, glycyrrhizin, and isoliquiritigenin) on both cyclooxygenase (COX) and lipoxygenase (LOX) products in order to understand the mechanism of its anti-inflammatory action. Inhibitory effect of GutGard™ and its phytoconstituents on lipopolysaccharide (LPS) induced prostaglandin E(2) (PGE(2)), calcimycin (A23187) induced thromboxane (TXB(2)), and leukotriene (LTB(4)) release was studied using murine macrophages (J774A.1) and human neutrophil (HL-60) cells. Results revealed that, G. glabra and glabridin significantly inhibited PGE(2), TXB(2) (COX) and LTB(4) (LOX), while, isoliquiritigenin exerted inhibitory effect only against COX products but failed to suppress LOX product. However, glycyrrhizin at the tested concentrations failed to exhibit inhibitory effect on both COX and LOX products. Here, we report for the first time that G. glabra (almost devoid of glycyrrhizin) exhibits anti-inflammatory property likely through the inhibition of PGE(2), TXB(2) and LTB(4) in mammalian cell assay system, which could be influenced in part by glabridin and isoliquiritigenin.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Glycyrrhiza/chemistry , Lipoxygenase/drug effects , Plant Extracts/pharmacology , Prostaglandin-Endoperoxide Synthases/drug effects , Animals , Anti-Inflammatory Agents/therapeutic use , Calcimycin/pharmacology , Cell Line , Chalcones/pharmacology , Chalcones/therapeutic use , Dinoprostone/antagonists & inhibitors , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/therapeutic use , HL-60 Cells , Humans , Isoflavones/pharmacology , Isoflavones/therapeutic use , Leukotriene B4/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mice , Phenols/pharmacology , Phenols/therapeutic use , Plant Extracts/therapeutic use , Plant Roots/chemistry , Thromboxane B2/antagonists & inhibitors
20.
Indian J Exp Biol ; 49(12): 904-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22403863

ABSTRACT

Platelets aggregation around migrating tumor cells offers protection against the cytotoxic activity of the natural killers cells (NKC). The ascorbic acid in 3 x 10(-3) M concentration completely inhibited platelet aggregation, decreased thromboxane B2 levels, and inhibited the expression of platelet membranic receptor GpIIb/IIIa in non stimulated platelets, and increased the NKC cytotoxicity in an average rate of 105, 61, and 285% in the NKC/targets cells ratios 12.5:1, 25:1 and 50:1 respectively. The results suggest the role of ascorbic acid in increasing the susceptibility of tumor cells to NKC; the ascorbic acid could be used as part of a multidrug therapy to treat diseases which up to now have been treated only through chemotherapy.


Subject(s)
Ascorbic Acid/pharmacology , Immunomodulation/drug effects , Killer Cells, Natural/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Blood Platelets/drug effects , Blood Platelets/immunology , Cell Survival/drug effects , Cell Survival/immunology , Dose-Response Relationship, Drug , Flow Cytometry , Humans , K562 Cells , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/biosynthesis , Thromboxane B2/antagonists & inhibitors , Thromboxane B2/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...