Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
J Immunol ; 184(4): 2140-7, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20065114

ABSTRACT

NO-donating aspirins consist of aspirin to which a NO-donating group is covalently linked via a spacer molecule. NCX 4040 and NCX 4016 are positional isomers with respect to the -CH(2)ONO(2) group (para and meta, respectively) on the benzene ring of the spacer. Because positional isomerism is critical for antitumor properties of NO-donating aspirins, we aimed to compare their anti-inflammatory effects with those of aspirin in vitro. Thus, we assessed their impacts on cyclooxygenase-2 activity (by measuring PGE(2) levels), protein expression, and cytokine generation(IL-1beta, IL-18, TNF-alpha, and IL-10) in human whole blood and isolated human monocytes stimulated with LPS. Interestingly, we found that micromolar concentrations of NCX 4040, but not NCX 4016 or aspirin, affected cyclooxygenase-2 expression and cytokine generation. We compared the effects of NCX 4040 with those of NCX 4016 or aspirin on IkappaB-alpha stabilization and proteasome activity in the LPS-stimulated human monocytic cell line THP1. Differently from aspirin and NCX 4016, NCX 4040, at a micromolar concentration range, inhibited IkappaB-alpha degradation. In fact, NCX 4040 caused concentration-dependent accumulation of IkappaB-alpha and its phosphorylated form. This effect was not reversed by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, an inhibitor of guanylyl cyclase, thus excluding the contribution of NO-dependent cGMP generation. In contrast, IkappaB-alpha accumulation by NCX 4040 may involve an inhibitory effect on proteasome functions. Indeed, NCX 4040 inhibited 20S proteasome activity when incubated with intact cells but not in the presence of cell lysate supernatants, thus suggesting an indirect inhibitory effect. In conclusion, NCX 4040 is an inhibitor of IkappaB-alpha degradation and proteasome function, and it should be taken into consideration for the development of novel anti-inflammatory and chemopreventive agents.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/analogs & derivatives , I-kappa B Proteins/antagonists & inhibitors , I-kappa B Proteins/metabolism , Monocytes/drug effects , Monocytes/metabolism , Nitric Oxide Donors/pharmacology , Nitro Compounds/pharmacology , Adult , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Aspirin/chemistry , Aspirin/pharmacology , Blood Platelets/drug effects , Blood Platelets/enzymology , Cell Line, Tumor , Cyclooxygenase 1/blood , Cyclooxygenase 2/blood , Dinoprostone/biosynthesis , Dinoprostone/blood , Humans , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/physiology , NF-KappaB Inhibitor alpha , Nitric Oxide Donors/chemistry , Nitro Compounds/chemistry , Signal Transduction/drug effects , Signal Transduction/immunology
2.
J Pharmacol Exp Ther ; 322(2): 453-60, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17473175

ABSTRACT

We compared the variability in degree and recovery from steady-state inhibition of cyclooxygenase (COX)-1 and COX-2 ex vivo and in vivo and platelet aggregation by naproxen sodium at 220 versus 440 mg b.i.d. and low-dose aspirin in healthy subjects. Six healthy subjects received consecutively naproxen sodium (220 and 440 mg b.i.d.) and aspirin (100 mg daily) for 6 days, separated by washout periods of 2 weeks. COX-1 and COX-2 inhibition was determined using ex vivo and in vivo indices of enzymatic activity: 1) the measurement of serum thromboxane (TX)B(2) levels and whole-blood lipopolysaccharide-stimulated prostaglandin (PG)E(2) levels, markers of COX-1 in platelets and COX-2 in monocytes, respectively; 2) the measurement of urinary 11-dehydro-TXB(2) and 2,3-dinor-6-keto-PGF(1alpha) levels, markers of systemic TXA(2) biosynthesis (mostly COX-1-derived) and prostacyclin biosynthesis (mostly COX-2-derived), respectively. Arachidonic acid (AA)-induced platelet aggregation was also studied. The maximal inhibition of platelet COX-1 (95.9 +/- 5.1 and 99.2 +/- 0.4%) and AA-induced platelet aggregation (92 +/- 3.5 and 93.7 +/- 1.5%) obtained at 2 h after dosing with naproxen sodium at 220 and 440 mg b.i.d., respectively, was indistinguishable from aspirin, but at 12 and 24 h after dosing, we detected marked variability, which was higher with naproxen sodium at 220 mg than at 440 mg b.i.d. Assessment of the ratio of inhibition of urinary 11-dehydro-TXB(2) versus 2,3-dinor-6-keto-PGF(1alpha) showed that the treatments caused a more profound inhibition of TXA(2) than prostacyclin biosynthesis in vivo throughout dosing interval. In conclusion, neither of the two naproxen doses mimed the persistent and complete inhibition of platelet COX-1 activity obtained by aspirin, but marked heterogeneity was mitigated by the higher dose of the drug.


Subject(s)
Aspirin/pharmacology , Naproxen/pharmacology , Platelet Aggregation/drug effects , 6-Ketoprostaglandin F1 alpha/analogs & derivatives , 6-Ketoprostaglandin F1 alpha/urine , Adult , Arachidonic Acid/pharmacology , Blood Platelets/drug effects , Blood Platelets/enzymology , Blood Platelets/metabolism , Cyclooxygenase 1/blood , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/blood , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/blood , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Epoprostenol/metabolism , Humans , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Monocytes/enzymology , Monocytes/metabolism , Naproxen/blood , Thromboxane B2/analogs & derivatives , Thromboxane B2/blood , Thromboxane B2/urine
3.
Prostaglandins Other Lipid Mediat ; 82(1-4): 85-94, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17164136

ABSTRACT

We provide comprehensive knowledge on the differential regulation of expression and catalysis of cyclooxygenase (COX)-1 and COX-2 in health and disease which represents an essential requirement to read out the clinical consequences of selective and nonselective inhibition of COX-isozymes in humans. Furthermore, we describe the pharmacodynamic and pharmacokinetic characteristics of major traditional nonsteroidal anti-inflammatory drugs (tNSAIDs) and coxibs (selective COX-2 inhibitors) which play a prime role in their efficacy and toxicity. Important information derived from our pharmacological studies has clarified that nonselective COX inhibitors should be considered the tNSAIDs with a balanced inhibitory effect on both COX-isozymes (exemplified by ibuprofen and naproxen). In contrast, the tNSAIDs meloxicam, nimesulide and diclofenac (which are from 18- to 29-fold more potent towards COX-2 in vitro) and coxibs (i.e. celecoxib, valdecoxib, rofecoxib, etoricoxib and lumiracoxib, which are from 30- to 433-fold more potent towards COX-2 in vitro) should be comprised into the cluster of COX-2 inhibitors. However, the dose and frequency of administration together with individual responses will drive the degree of COX-2 inhibition and selectivity achieved in vivo. The results of clinical pharmacology of COX inhibitors support the concept that the inhibition of platelet COX-1 may translate into an increased incidence of serious upper gastrointestinal bleeding but this effect on platelet COX-1 may mitigate the cardiovascular hazard associated with the profound inhibition of COX-2-dependent prostacyclin (PGI2).


Subject(s)
Cyclooxygenase 1/biosynthesis , Cyclooxygenase 2/biosynthesis , Cyclooxygenase Inhibitors/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biomarkers , Blood Platelets/enzymology , Cyclooxygenase Inhibitors/pharmacokinetics , Endothelium, Vascular/enzymology , Gastric Mucosa/enzymology , Gene Expression Regulation, Enzymologic , Humans , Therapeutic Equivalency
4.
Clin Pharmacol Ther ; 80(3): 264-74, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16952493

ABSTRACT

BACKGROUND AND OBJECTIVE: We performed a placebo-controlled, randomized study to address whether celecoxib or ibuprofen undermines the functional range of inhibition of platelet cyclooxygenase (COX)-1 activity by aspirin in patients with osteoarthritis and stable ischemic heart disease. METHODS: Twenty-four patients who were undergoing long-term treatment with aspirin (100 mg daily) for cardioprotection were coadministered celecoxib, 200 mg twice daily, ibuprofen, 600 mg 3 times daily, or placebo for 7 days. RESULTS: The coadministration of placebo or celecoxib did not undermine the aspirin-related inhibition of platelet COX-1 activity, as assessed by measurements of serum thromboxane B(2) (TXB(2)) levels, as well as platelet function. In contrast, a significant (P < .001) increase in serum TXB(2) level was detected on day 7 before drug administration (median, 19.13 ng/mL [range, 1-47.5 ng/mL]) and at 24 hours after the coadministration of aspirin and ibuprofen (median, 22.28 ng/mL [range, 4.9-44.4 ng/mL]) versus baseline (median, 1.65 ng/mL [range, 0.55-79.8 ng/mL]); this was associated with a significant increase in arachidonic acid-induced platelet aggregation (P < .01) and adenosine diphosphate-induced platelet aggregation (P < .05) and a decrease in the time to form an occlusive thrombus in the platelet function analyzer (P < .01). The urinary excretion of 11-dehydro-TXB(2), an index of systemic thromboxane biosynthesis, was not significantly affected by the coadministration of treatment drugs. At steady state, a comparable and persistent inhibition of lipopolysaccharide-stimulated prostaglandin E(2) generation, a marker of COX-2 activity ex vivo, was caused by ibuprofen (>or=80%) or celecoxib (>or=70%) but not placebo. CONCLUSIONS: Unlike ibuprofen, celecoxib did not interfere with the inhibition of platelet COX-1 activity and function by aspirin despite a comparable suppression of COX-2 ex vivo in patients with osteoarthritis and stable ischemic heart disease.


Subject(s)
Aspirin/therapeutic use , Ibuprofen/therapeutic use , Myocardial Ischemia/drug therapy , Osteoarthritis/drug therapy , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , Adenosine Diphosphate/pharmacology , Aged , Arachidonic Acid/pharmacology , Aspirin/administration & dosage , Aspirin/urine , Celecoxib , Double-Blind Method , Drug Administration Schedule , Drug Therapy, Combination , Female , Humans , Ibuprofen/administration & dosage , Ibuprofen/urine , Male , Middle Aged , Myocardial Ischemia/blood , Osteoarthritis/blood , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation Inhibitors/urine , Platelet Function Tests/methods , Pyrazoles/administration & dosage , Pyrazoles/urine , Sulfonamides/administration & dosage , Sulfonamides/urine , Thromboxane B2/analogs & derivatives , Thromboxane B2/blood , Thromboxane B2/urine , Treatment Outcome
5.
Clin Pharmacol Ther ; 80(2): 115-25, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16890573

ABSTRACT

BACKGROUND AND OBJECTIVES: Complete and persistent suppression of platelet thromboxane (TX) A(2) biosynthesis by aspirin is mandatory to fulfill its cardioprotection. We explored the determinants of heterogeneity of TXB2 generation in clotting whole blood, a capacity index of platelet cyclooxygenase (COX) activity, in patients with coronary heart disease (CHD) versus healthy subjects treated with low-dose aspirin on a long-term basis. METHODS: We studied 30 patients with CHD (ie, chronic stable angina, unstable angina, and acute myocardial infarction) and 10 healthy subjects, who were treated with low-dose aspirin (100 mg daily) on a long-term basis, 12 hours after the administration of 160 mg aspirin to ensure saturation of platelet COX-1 activity. Serum TXB2 levels were assessed. The contribution of blood COX-2 to TXA2 biosynthesis was explored by evaluation of the effect of a selective COX-2 inhibitor (L-745,337) added to heparinized whole blood stimulated with Ca++ ionophore A23187 (20 micromol/L) for 1 hour or lipopolysaccharide (0.1 microg/mL) for 4 hours. RESULTS: In healthy subjects serum TXB2 levels ranged from 0.6 to 7.9 ng/mL (median, 2.1 ng/mL; mean +/- SD, 3.2 +/- 2.6 ng/mL). In CHD patients we detected enhanced variability in serum TXB2 generation (median, 3.1 ng/mL [range, 0.15-47 ng/mL]; mean, 8.5 +/- 12.3 ng/mL), which in 8 patients (27%) exceeded the mean value + 2 SDs detected in healthy subjects (ie, 8.4 ng/mL), set as the limit value for an adequate inhibition of platelet COX-1 by aspirin. Elevated whole-blood TXB2 generation was not dependent on leukocyte count, COX-2 activity, or cigarette smoking but was plausibly a result of defective suppression of platelet COX-1 activity. CONCLUSIONS: Heterogeneity in the suppression of platelet COX-1 activity by aspirin occurred in CHD patients. The measurement of the serum TXB2 level seems to be an appropriate biomarker to identify patients who have an inadequate inhibition of platelet COX-1 activity by aspirin.


Subject(s)
Aspirin/pharmacology , Blood Platelets/enzymology , Coronary Disease/blood , Coronary Disease/enzymology , Cyclooxygenase 1/blood , Cyclooxygenase Inhibitors/pharmacology , Aged , Arachidonic Acid/metabolism , Calcimycin/pharmacology , Cyclooxygenase 2/biosynthesis , Female , Humans , Indans/pharmacology , Lipopolysaccharides , Male , Middle Aged , Neutrophils/enzymology , Thromboxane A2/biosynthesis , Thromboxane B2/metabolism
6.
Circ Res ; 98(5): 593-5, 2006 Mar 17.
Article in English | MEDLINE | ID: mdl-16484611

ABSTRACT

Aspirin affords cardioprotection through the acetylation of serine529 in human cyclooxygenase-1 (COX-1) of anucleated platelets, inducing a permanent defect in thromboxane A2 (TXA2)-dependent platelet function. However, heterogeneity of COX-1 suppression by aspirin has been detected in cardiovascular disease and may contribute to failure to prevent clinical events. The recent recognized capacity of platelets to make proteins de novo paves the way to identify new mechanisms involved in the variable response to aspirin. We found that in washed human platelets, the complete suppression of TXA2 biosynthesis by aspirin, in vitro, recovered in response to thrombin and fibrinogen in a time-dependent fashion (at 0.5 and 24 hours, TXB2 averaged 0.1+/-0.03 and 3+/-0.8 ng/mL; in the presence of arachidonic acid [10 micromol/L], it was 2+/-0.7 and 25+/-7 ng/mL, respectively), and it was blocked by translational inhibitors, by rapamycin, and by inhibitors of phosphatidylinositol 3-kinase. The results that COX-1 mRNA was readily detected in resting platelets and that [35S]-methionine was incorporated into COX-1 protein after stimulation strongly support the occurrence of de novo COX-1 synthesis in platelets. This process may interfere with the complete and persistent suppression of TXA2 biosynthesis by aspirin necessary for cardioprotection.


Subject(s)
Aspirin/pharmacology , Blood Platelets/drug effects , Cyclooxygenase 1/biosynthesis , Platelet Aggregation Inhibitors/pharmacology , Thromboxane A2/biosynthesis , Adult , Blood Platelets/metabolism , Cyclooxygenase 1/genetics , Dose-Response Relationship, Drug , Humans , Middle Aged , Phosphatidylinositol 3-Kinases/physiology , Protein Biosynthesis , RNA, Messenger/analysis
7.
Blood ; 107(9): 3572-4, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16397127

ABSTRACT

The recent demonstration that platelets express a functional toll-like receptor 4 (TLR4) prompted us to explore the influence of TLR4 polymorphisms (Asp299Gly alone or in combination with Thr399Ile) on thromboxane A(2) (TXA(2)) biosynthesis in vivo. In 17 subjects with TLR4 polymorphisms versus 17 wild type (untreated with aspirin, matched for age, sex, and cardiovascular risk factors), intima-media thickness in the common carotid arteries was significantly lower. Average urinary excretion of 11-dehydro-TXB(2), an index of systemic biosynthesis of TX, was significantly reduced by 65%. The urinary excretion of 2,3-dinor-6-keto-prostaglandin F(1alpha), an index of systemic biosynthesis of prostacyclin, was marginally depressed but the prostacyclin/TXA(2) biosynthesis ratio was significantly higher than in wild type. Selective inhibition of cyclooxygenase 2-dependent prostacyclin (by rofecoxib or etoricoxib) was associated with increased urinary excretion of 11-dehydro-TXB(2) in carriers of TLR4 polymorphisms, but not in wild-type, suggesting a restrainable effect of prostacyclin on platelet function in vivo in this setting. Reduced TXA(2) biosynthesis may contribute to the protective cardiovascular phenotype of TLR4 polymorphisms.


Subject(s)
Heterozygote , Polymorphism, Genetic , Thromboxane A2/biosynthesis , Toll-Like Receptor 4/genetics , Cardiovascular Diseases/genetics , Cardiovascular Diseases/prevention & control , Carotid Artery, Common/diagnostic imaging , Case-Control Studies , Epoprostenol/biosynthesis , Female , Humans , Male , Middle Aged , Phenotype , Risk Factors , Ultrasonography
8.
J Am Coll Cardiol ; 45(8): 1295-301, 2005 Apr 19.
Article in English | MEDLINE | ID: mdl-15837265

ABSTRACT

OBJECTIVES: We investigated the occurrence of pharmacodynamic interaction between low-dose aspirin and naproxen. BACKGROUND: The uncertainty of cardioprotection by naproxen has encouraged its combination with aspirin in patients with arthritis and cardiovascular disease. METHODS: The incubation of washed platelets with naproxen for 5 min before the addition of aspirin reduced the irreversible inhibition of thromboxane (TX)B(2) production by aspirin. The pharmacodynamic interaction between the two drugs was then investigated in four healthy volunteers who received aspirin (100 mg daily) for 6 days and then the combination of aspirin and naproxen for further 6 days: aspirin 2 h before naproxen (500 mg, twice-daily dosing). After 14 days of washout, naproxen was given 2 h before aspirin for further 6 days. RESULTS: The inhibition of serum TXB(2) production (index of platelet cyclooxygenase [COX]-1 activity) and platelet aggregation ex vivo and urinary 11-dehydro-TXB(2) levels (index of TXB(2) biosynthesis in vivo) by aspirin alone (99 +/- 0.2%, 95 +/- 0.6%, and 81 +/- 4%, respectively) was not significantly altered by the co-administration of naproxen, given either 2 h after aspirin or in reverse order. In a second study, the concurrent administration of a single dose of aspirin and naproxen did not affect platelet TXB(2) production and aggregation at 1 h after dosing, when aspirin alone causes maximal inhibitory effect. Moreover, the rapid recovery of platelet COX-1 activity and function supports the occurrence of a pharmacodynamic interaction between naproxen and aspirin. CONCLUSIONS: Naproxen interfered with the inhibitory effect of aspirin on platelet COX-1 activity and function. This pharmacodynamic interaction might undermine the sustained inhibition of platelet COX-1 that is necessary for aspirin's cardioprotective effects.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/administration & dosage , Naproxen/pharmacology , Adult , Arthritis/drug therapy , Aspirin/pharmacology , Cardiovascular Diseases/drug therapy , Cyclooxygenase 1 , Drug Interactions , Drug Therapy, Combination , Humans , In Vitro Techniques , Membrane Proteins , Platelet Aggregation/drug effects , Prostaglandin-Endoperoxide Synthases , Thromboxane B2/antagonists & inhibitors
9.
Pharmacol Rep ; 57 Suppl: 66-85, 2005.
Article in English | MEDLINE | ID: mdl-16415488

ABSTRACT

Prostanoids act leading roles in a myriad of physiologic and pathologic processes because these autacoids participate in the amplification of biological responses induced by innumerable stimuli. The formation of prostanoids is operated by two synthases named cyclooxygenase(COX)-1 and COX-2. Traditional nonsteroidal antiinflammatory drugs (tNSAIDs) and COX-2 inhibitors (coxibs) give rise to antipyretic, analgesic, and antiinflammatory actions, through their reversible clogging of the COX channel of COX-2 - apart from aspirin which modifies irreversibly the catalytic activity of COX-2. tNSAIDs and COX-2 inhibitors resulted clinically equivalent for the relief of acute pain and symptoms of arthropathies but they failed to modify disease progression. Clinical evidence of the possible contribution of COX-1 in inflammation and pain in some occasion - as suggested by experimental and pharmacology studies - is orphan because none efficacy trial with COX inhibitors was designed to establish it. COX-2 inhibitors were developed with the aim to reduce the incidence of serious gastrointestinal (GI) adverse effects associated with the administration of tNSAIDs ensued as a consequence of the inhibition of cytoprotective COX-1-derived prostanoids. However, the reduced incidence of serious GI adverse effects compared to tNSAIDs demonstrated for 2 COX-2 inhibitors (e.g. rofecoxib and lumiracoxib) has been countered by an increased incidence of myocardial infarction and stroke detected in 5 placebo controlled trials involving the COX-2 inhibitors celecoxib, rofecoxib and valdecoxib. The future of COX-2 inhibitors will be an example of personalised medicine as their use will be restricted to patients who do not respond to tNSAIDs or with increased risk of GI complications.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cyclooxygenase 2 Inhibitors/therapeutic use , Inflammation/drug therapy , Pain/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase 1/drug effects , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/adverse effects , Cyclooxygenase 2 Inhibitors/pharmacology , Humans , Inflammation/physiopathology , Pain/physiopathology , Prostaglandins/metabolism
10.
J Pathol ; 202(3): 305-12, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14991895

ABSTRACT

Helicobacter pylori up-regulates cyclo-oxygenase-2 (COX-2) expression, which in turn is involved in tumourigenesis. Recently, a causal link between COX-2 and multidrug resistance 1 (MDR-1) gene expression, implicated in cancer chemoresistance, has been demonstrated. Thus, the expression of COX-2 and the downstream enzyme involved in PGE2 biosynthesis, microsomal PGE-synthase1 (mPGES1), was correlated with P-gp, the product of MDR-1, and the anti-apoptotic protein, Bcl-xL, in gastric biopsies from patients with H pylori infection and in patients with gastric cancer. In a retrospective analysis of endoscopic and pathology files, 40 H pylori-negative patients (Hp-), 50 H pylori-positive patients who responded to eradication therapy (Hp+R), 84 H pylori-positive patients who did not respond to eradication therapy (Hp+NR), and 30 patients with gastric cancer (18 intestinal and 12 diffuse types) were selected. COX-2, mPGES1, P-gp, and Bcl-xL were detected by immunohistochemistry. COX-2, mPGES1, P-gp, and Bcl-xL expression was undetectable in gastric mucosa from Hp- patients. By contrast, COX-2 and mPGES1 expression was detected in 42% and 44% of Hp+R patients, respectively, and in up to 66% (range 63-66%) of Hp+NR patients (p < 0.05). The expression of COX-2 and mPGES1 correlated significantly (p < 0.0001) with that of P-gp and Bcl-xL. High levels of COX-2, mPGES1, P-gp, and Bcl-xL expression were found in intestinal-type gastric cancer samples. In conclusion, H pylori-dependent induction of COX-2 and mPGES1 is associated with enhanced production of P-gp and Bcl-xL that may contribute to gastric tumourigenesis and resistance to therapy.


Subject(s)
Intramolecular Oxidoreductases/analysis , Isoenzymes/analysis , Prostaglandin-Endoperoxide Synthases/analysis , Proto-Oncogene Proteins c-bcl-2/analysis , Stomach Neoplasms/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , Adult , Aged , Chi-Square Distribution , Cyclooxygenase 2 , Enzyme Activation , Female , Gastritis/drug therapy , Gastritis/metabolism , Helicobacter Infections/complications , Helicobacter Infections/drug therapy , Helicobacter Infections/metabolism , Helicobacter pylori , Humans , Immunohistochemistry/methods , Male , Membrane Proteins , Middle Aged , Prostaglandin-E Synthases , Stomach Neoplasms/drug therapy , Stomach Neoplasms/microbiology , bcl-X Protein
11.
Circulation ; 109(12): 1468-71, 2004 Mar 30.
Article in English | MEDLINE | ID: mdl-15037526

ABSTRACT

BACKGROUND: The current controversy on the potential cardioprotective effect of naproxen prompted us to evaluate the extent and duration of platelet, monocyte, and vascular cyclooxygenase (COX) inhibition by naproxen compared with low-dose aspirin. METHODS AND RESULTS: We performed a crossover, open-label study of low-dose aspirin (100 mg/d) or naproxen (500 mg BID) administered to 9 healthy subjects for 6 days. The effects on thromboxane (TX) and prostacyclin biosynthesis were assessed up to 24 hours after oral dosing. Serum TXB2, plasma prostaglandin (PG) E2, and urinary 11-dehydro-TXB2 and 2,3-dinor-6-keto-PGF(1alpha) were measured by previously validated radioimmunoassays. The administration of naproxen or aspirin caused a similar suppression of whole-blood TXB2 production, an index of platelet COX-1 activity ex vivo, by 94+/-3% and 99+/-0.3% (mean+/-SD), respectively, and of the urinary excretion of 11-dehydro-TXB2, an index of systemic biosynthesis of TXA2 in vivo, by 85+/-8% and 78+/-7%, respectively, that persisted throughout the dosing interval. Naproxen, in contrast to aspirin, significantly reduced systemic prostacyclin biosynthesis by 77+/-19%, consistent with differential inhibition of monocyte COX-2 activity measured ex vivo. CONCLUSIONS: The regular administration of naproxen 500 mg BID can mimic the antiplatelet COX-1 effect of low-dose aspirin. Naproxen, unlike aspirin, decreased prostacyclin biosynthesis in vivo.


Subject(s)
6-Ketoprostaglandin F1 alpha/analogs & derivatives , Aspirin/pharmacology , Blood Platelets/enzymology , Cyclooxygenase Inhibitors/pharmacology , Endothelial Cells/enzymology , Endothelium, Vascular/cytology , Isoenzymes/antagonists & inhibitors , Monocytes/enzymology , Naproxen/pharmacology , Thromboxane B2/analogs & derivatives , 6-Ketoprostaglandin F1 alpha/urine , Adult , Aspirin/administration & dosage , Blood Platelets/drug effects , Cardiotonic Agents/pharmacology , Cross-Over Studies , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Dinoprostone/blood , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Female , Humans , Isoenzymes/blood , Male , Membrane Proteins , Middle Aged , Monocytes/drug effects , Prostaglandin-Endoperoxide Synthases/blood , Prostaglandins I/biosynthesis , Thromboxane B2/blood , Thromboxane B2/urine
12.
Br J Pharmacol ; 138(4): 634-41, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12598417

ABSTRACT

1. Acetaminophen, an analgesic and antipyretic drug with weak antiinflammatory properties, has been suggested to act as a tissue-selective inhibitor of prostaglandin H synthases (PGHSs) (e.g. COX-1 and COX-2) through its reducing activity, that is influenced by the different cellular levels of peroxides. 2. We have studied the effects of acetaminophen on inducible and constitutive prostanoid biosynthesis in monocytes and platelets in vitro. To discriminate between the inhibitory effect of the drug on PGHS-isozymes vs PGE-synthases (PGESs), parallel measurements of PGE(2) and thromboxane (TX) B(2) were carried out. Since antioxidant enzymes and cofactors, present in plasma, may affect acetaminophen-dependent inhibition of prostanoids, comparative experiments in whole blood vs isolated monocytes were performed. 3. Acetaminophen inhibited LPS-induced whole blood PGE(2) and TXB(2) production, in a concentration-dependent fashion [IC(50) microM (95% confidence intervals): 44 (27-70) and 94 (79-112), respectively]. Therapeutic plasma concentrations (100 and 300 microM) of the drug more profoundly reduced PGE(2) than TXB(2) (71 +/- 3 vs 54 +/- 4 and 95 +/- 0.8 vs 78 +/- 2%, respectively, mean +/- s.e.mean, n = 6, P < 0.01). 4. Differently, in isolated monocytes stimulated with LPS, both PGE(2) and TXB(2) production was maximally reduced by only 60%. 5 At 100 and 300 microM, the drug caused a similar and incomplete inhibition of platelet PGE(2) and TXB(2) production during whole blood clotting (45 +/- 4 vs 54 +/- 4 and 75 +/- 2 vs 75 +/- 1%, respectively, mean +/- s.e.mean, n = 4). 6 In conclusion, therapeutic concentrations of acetaminophen caused an incomplete inhibition of platelet COX-1 and monocyte COX-2 but in the presence of plasma, the drug almost completely suppressed inducible PGE(2) biosynthesis through its inhibitory effects on both COX-2 and inducible PGES.


Subject(s)
Acetaminophen/pharmacology , Blood Cells/drug effects , Blood Cells/metabolism , Prostaglandins/biosynthesis , Adult , Dose-Response Relationship, Drug , Female , Humans , Male
13.
Curr Med Res Opin ; 18(8): 503-11, 2002.
Article in English | MEDLINE | ID: mdl-12564662

ABSTRACT

We have evaluated the biochemical selectivity of novel cyclo-oxygenase (COX)-2 inhibitors, etoricoxib, valdecoxib, DFU and DFP, vs rofecoxib and celecoxib, using the human whole blood assays of COX-isozyme activity, in vitro. Compounds were incubated with human whole blood samples, allowed to clot for 1 h at 37 degrees C, or stimulated with lipopolysaccharide (10 microg/ml) for 24 h at 37 degrees C. Serum thromboxane (TX) B2 and plasma prostaglandin (PG) E2 levels were measured by specific radioimmunoassays as indices of platelet COX-1 and monocyte COX-2 activity, respectively. Valdecoxib, etoricoxib, DFU and DFP inhibited platelet COX-1 and monocyte COX-2 with the following COX-1/COX-2 IC50 ratios: 61.5, 344, 660 and 1918, respectively. The reference compounds, celecoxib and rofecoxib had corresponding values of 29.6 and 272. In conclusion, a second wave of COX-2 inhibitors with higher biochemical selectivity than the existing coxibs has been developed. Whether their administration will be associated with improved clinical efficacy and/or safety vis-à-vis celecoxib and rofecoxib remains to be established.


Subject(s)
Cyclooxygenase Inhibitors/blood , Isoenzymes/drug effects , Isoxazoles/blood , Lactones/blood , Prostaglandin-Endoperoxide Synthases/drug effects , Pyridines/blood , Sulfonamides/blood , Sulfones/blood , Adult , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Etoricoxib , Female , Humans , In Vitro Techniques , Italy , Male , Membrane Proteins
SELECTION OF CITATIONS
SEARCH DETAIL