Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Anal Bioanal Chem ; 409(30): 7097-7103, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29018901

ABSTRACT

A method has been developed for the trace analysis of oxypurinol that is considered as an active pharmaceutical ingredient and an emerging environmental contaminant. The method achieved the identification and quantification of oxypurinol in surface water samples utilizing solid phase extraction and ultra high-performance liquid chromatography with diode array and fluorescence detection for the first time. Four principal parameters of solid phase extraction were optimized to obtain maximum extraction efficiency. Under the isocratic elution of methanol/water (5:95, v/v) and the excitation/emission wavelength of 254/359 nm, a rapid determination was achieved in 2.0 min with good linearity of 1.05-351 µg/L (coefficient of determination above 0.9998). The limit of detection and method detection limit were 0.210 µg/L and 1.34 ng/L, respectively. Precision of the method was evaluated and a relative standard deviation value of 3.3% was obtained for analyses of six replicate spiking blank samples (200 mL, 176 ng/L) according to the overall proposed procedure. The method showed a great anti-interference ability and average spiked recoveries of oxypurinol in five surface water samples were in the range of 94.5-111%. The ability of the method to detect and correctly identify oxypurinol can significantly promote investigation on the occurrence of oxypurinol in water and its potential (eco-)toxicological effects. Graphical abstract Quantification of the emerging contaminant oxypurinol in s urface water using SPE/UHPLC-FLD.


Subject(s)
Chromatography, High Pressure Liquid/methods , Oxypurinol/chemistry , Solid Phase Extraction/methods , Water Pollutants, Chemical/analysis , Fluorescence , Limit of Detection , Sensitivity and Specificity , Tandem Mass Spectrometry/methods , Water/chemistry
2.
J Immunol ; 192(7): 2984-93, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24591375

ABSTRACT

Allopurinol (ALP) hypersensitivity is a major cause of severe cutaneous adverse reactions and is strongly associated with the HLA-B*58:01 allele. However, it can occur in the absence of this allele with identical clinical manifestations. The immune mechanism of ALP-induced severe cutaneous adverse reactions is poorly understood, and the T cell-reactivity pattern in patients with or without the HLA-B*58:01 allele is not known. To understand the interactions among the drug, HLA, and TCR, we generated T cell lines that react to ALP or its metabolite oxypurinol (OXP) from HLA-B*58:01(+) and HLA-B*58:01(-) donors and assessed their reactivity. ALP/OXP-specific T cells reacted immediately to the addition of the drugs and bypassed intracellular Ag processing, which is consistent with the "pharmacological interaction with immune receptors" (p-i) concept. This direct activation occurred regardless of HLA-B*58:01 status. Although most OXP-specific T cells from HLA-B*58:01(+) donors were restricted by the HLA-B*58:01 molecule for drug recognition, ALP-specific T cells also were restricted to other MHC class I molecules. This can be explained by in silico docking data that suggest that OXP binds to the peptide-binding groove of HLA-B*58:01 with higher affinity. The ensuing T cell responses elicited by ALP or OXP were not limited to particular TCR Vß repertoires. We conclude that the drug-specific T cells are activated by OXP bound to HLA-B*58:01 through the p-i mechanism.


Subject(s)
HLA-B Antigens/immunology , Lymphocyte Activation/immunology , Oxypurinol/immunology , T-Lymphocytes/immunology , Allopurinol/chemistry , Allopurinol/immunology , Allopurinol/pharmacology , Binding, Competitive/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Calcium/immunology , Calcium/metabolism , Cells, Cultured , Flow Cytometry , HLA-B Antigens/chemistry , HLA-B Antigens/genetics , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lysosomal-Associated Membrane Protein 1/immunology , Lysosomal-Associated Membrane Protein 1/metabolism , Models, Molecular , Molecular Structure , Oxypurinol/chemistry , Oxypurinol/pharmacology , Protein Binding/immunology , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
3.
J Drug Target ; 19(10): 954-66, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22074217

ABSTRACT

Xanthine oxidase (XO) is the major source of superoxide anion (O(2)(-)) that is associated with various reactive oxygen species (ROS) related diseases. 4-amino-6-hydroxypyrazolo[3,4-d]pyrimidine (AHPP) is a potent XO inhibitor discovered in Maeda's laboratory, which is now being developed for the treatment of ischemia reperfusion injury and inflammatory diseases. However, the poor aqueous solubility of AHPP at physiological pH hampers its clinical development. To overcome this drawback, in the present study water soluble polyethyleneglycol conjugated AHPP (AHPP-PEG) was synthesized via two different approaches, which resulted in two derivatives of AHPP-PEG, namely, mono-AHPP-PEG and bis-(AHPP)-PEG depending on the number of AHPP on PEG chain. We characterized both conjugates by UV, FTIR spectroscopy and elemental analysis. Dynamic light scattering and Sephadex G-100 chromatography studies revealed mean particle size of 164.1 and 218.8 nm and Mw. equivalent to 107 and 126 kDa for mono-AHPP-PEG and bis-(AHPP)-PEG, respectively. Further, XO inhibitory activity for mono-AHPP-PEG and bis-(AHPP)-PEG were found with Ki of 0.23±0.03 and 0.21±0.03 µM, respectively. In vivo pharmacokinetic study showed longer circulation time of AHPP-PEG conjugates compared to free AHPP. These results indicate AHPP-PEG conjugates have better potentials with supramolecular assemblies in aqueous medium and may become a good candidate for the treatment of ROS related diseases.


Subject(s)
Drug Carriers/chemistry , Oxypurinol/analogs & derivatives , Polyethylene Glycols/chemistry , Xanthine Oxidase/antagonists & inhibitors , Animals , Male , Mice , Micelles , Oxypurinol/administration & dosage , Oxypurinol/chemistry , Oxypurinol/pharmacology , Particle Size , Solubility , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared , Water/chemistry
4.
Exp Biol Med (Maywood) ; 235(4): 487-96, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20407081

ABSTRACT

The detrimental role of superoxide anion (O(2)(-)) has been well documented in the pathogenesis of ischemia-reperfusion (I/R) injury. Our and other studies suggested that one critical source of O(2)(-) generation may be xanthine oxidase (XO). We thus hypothesized that I/R injury could be protected by inhibiting XO activity, which would reduce the amount of O(2)(-) and hence reduce pathogenic consequences. Among various XO inhibitors, we previously found 4-amino-6-hydroxypyrazolo[3,4-d]pyrimidine (AHPP) exhibited potent XO inhibitory activity. Here, we report that the covalent conjugate of AHPP with amphipathic styrene-maleic acid copolymer (SMA-AHPP) showed protective effect against I/R-induced injury in a rat hepatic I/R model. Liver ischemia was induced by occluding both the portal vein and the hepatic artery for 30 min, and followed by reperfusion. SMA-AHPP was administered via the tail vein two hours before ischemia was initiated. A remarkable increase of liver enzymes in plasma (aspartate aminotransferase, AST; alanine aminotransferase, ALT and lactate dehydrogenase, LDH) was detected three hours after reperfusion, whereas prior injection of SMA-AHPP greatly suppressed this increase of AST, ALT and LDH. Moreover, induction of inflammatory cytokines, i.e. tumor necrosis factor-alpha (TNF-alpha), interleukin-12 (IL-12) and monocyte chemotactic protein-1 (MCP-1) by I/R were significantly inhibited by SMA-AHPP treatment. Accordingly, cytotoxic effect or apoptosis in the liver caused by I/R was clearly reduced by SMA-AHPP pretreatment. Furthermore, thiobarbituric acid-reactive substance assay showed a significant decrease of lipid peroxidation in rat liver after the administration of SMA-AHPP, which is parallel with the decreased XO activity after SMA-AHPP treatment, indicating the involvement of reactive oxygen species generated by XO. In addition, SMA-AHPP was found to bind to albumin, thus to exhibit prolonged in vivo (plasma) half-life. These results suggest that SMA-AHPP exerted a potent cytoprotective effect against I/R injury in rat liver, by inhibiting XO activity and the subsequent generation of O(2)(-).


Subject(s)
Enzyme Inhibitors/pharmacology , Liver/blood supply , Maleates/pharmacology , Oxypurinol/analogs & derivatives , Polystyrenes/pharmacology , Reperfusion Injury/prevention & control , Xanthine Oxidase/antagonists & inhibitors , Albumins/metabolism , Animals , Apoptosis/drug effects , Cytokines/blood , Enzyme Inhibitors/chemistry , Heme Oxygenase-1/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Maleates/chemistry , Molecular Weight , Oxypurinol/chemistry , Oxypurinol/pharmacology , Polystyrenes/chemistry , Rats , Rats, Wistar , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Superoxides/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Tissue Distribution , Xanthine Oxidase/metabolism
5.
J Biol Chem ; 284(13): 8768-76, 2009 Mar 27.
Article in English | MEDLINE | ID: mdl-19109249

ABSTRACT

Rhodobacter capsulatus xanthine dehydrogenase (XDH) is an (alphabeta)(2) heterotetrameric cytoplasmic enzyme that resembles eukaryotic xanthine oxidoreductases in respect to both amino acid sequence and structural fold. To obtain a detailed understanding of the mechanism of substrate and inhibitor binding at the active site, we solved crystal structures of R. capsulatus XDH in the presence of its substrates hypoxanthine, xanthine, and the inhibitor pterin-6-aldehyde using either the inactive desulfo form of the enzyme or an active site mutant (E(B)232Q) to prevent substrate turnover. The hypoxanthine- and xanthine-bound structures reveal the orientation of both substrates at the active site and show the importance of residue Glu(B)-232 for substrate positioning. The oxygen atom at the C-6 position of both substrates is oriented toward Arg(B)-310 in the active site. Thus the substrates bind in an orientation opposite to the one seen in the structure of the reduced enzyme with the inhibitor oxypurinol. The tightness of the substrates in the active site suggests that the intermediate products must exit the binding pocket to allow first the attack of the C-2, followed by oxidation of the C-8 atom to form the final product uric acid. Structural studies of pterin-6-aldehyde, a potent inhibitor of R. capsulatus XDH, contribute further to the understanding of the relative positioning of inhibitors and substrates in the binding pocket. Steady state kinetics reveal a competitive inhibition pattern with a K(i) of 103.57 +/- 18.96 nm for pterin-6-aldehyde.


Subject(s)
Bacterial Proteins/chemistry , Enzyme Inhibitors/chemistry , Oxypurinol/chemistry , Protein Folding , Rhodobacter capsulatus/enzymology , Xanthine Dehydrogenase/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Catalytic Domain/physiology , Crystallography, X-Ray , Eukaryotic Cells/enzymology , Hypoxanthine/chemistry , Mutation , Protein Structure, Quaternary/physiology , Pterins/chemistry , Rhodobacter capsulatus/genetics , Xanthine/chemistry , Xanthine Dehydrogenase/antagonists & inhibitors , Xanthine Dehydrogenase/genetics
6.
Nucleosides Nucleotides Nucleic Acids ; 27(6): 888-93, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18600558

ABSTRACT

Inhibitors of xanthine oxidoreductase block conversion of xanthine to uric acid and are therefore potentially useful for treatment of hyperuricemia or gout. We determined the crystal structure of reduced bovine milk xanthine oxidoreductase complexed with oxipurinol at 2.0 A resolution. Clear electron density was observed between the N2 nitrogen of oxipurinol and the molybdenum atom of the molybdopterin cofactor, indicating that oxipurinol coordinated directly to molybdenum. Oxipurinol forms hydrogen bonds with glutamate 802, arginine 880, and glutamate 1261, which have previously been shown to be essential for the enzyme reaction. We discuss possible differences in the hypouricemic effect of inhibitors, including allopurinol and newly developed inhibitors, based on their mode of binding in the crystal structures.


Subject(s)
Allopurinol/pharmacology , Enzyme Inhibitors/pharmacology , Milk/enzymology , Oxypurinol/chemistry , Oxypurinol/metabolism , Xanthine Dehydrogenase/antagonists & inhibitors , Xanthine Dehydrogenase/chemistry , Animals , Cattle , Crystallography, X-Ray , Milk/metabolism , Oxidation-Reduction , Xanthine Dehydrogenase/metabolism
7.
Br J Clin Pharmacol ; 43(2): 197-200, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9131954

ABSTRACT

AIMS: In the present study we have investigated the use of caffeine, administered in the form of instant coffee, as a prodrug for 1MX to validate the use of the 1MU:1MX ratio following caffeine administration as a pharmacodynamic measure of oxypurinol effect on xanthine oxidase. METHODS: Five healthy volunteers took caffeine 75 mg 8 hourly administered as instant coffee over a 7 day period. They were given allopurinol 600 mg on day 4. Urine was collected in 8 h aliquots from day 1-day 7. The ratio of 1-methyluric acid (1MU) to 1-methylxanthuric (1MX) was determined. RESULTS: The relationship between the plasma oxypurinol (the active metabolite of allopurinol) concentration at the midpoint of each caffeine dosage interval and the decrement in the urinary 1MX to 1MU ratio fitted well by a sigmoid Emax model. Mean (+/-s.d.) values of the oxypurinol EC50(3.9 +/- 1.4 mg l-1), EC90(8.7 +/- 1.8 mgl-1) and the exponent, n (3.0 +/- 1.2) were similar to those obtained previously following either the direct administration of 1MX or the use of theophylline as a prodrug for 1MX. CONCLUSIONS: These data indicate that the use of caffeine as a source of 1MX could provide a simple and ethically acceptable method for monitoring oxypurinol effect in patients taking allopurinol for the treatment of gout.


Subject(s)
Caffeine/pharmacokinetics , Oxypurinol/pharmacology , Xanthines/pharmacokinetics , Adult , Female , Humans , Male , Molecular Probes , Oxypurinol/blood , Oxypurinol/chemistry , Oxypurinol/urine , Uric Acid/analogs & derivatives , Uric Acid/pharmacokinetics
8.
J Comput Aided Mol Des ; 10(6): 535-44, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9007687

ABSTRACT

Tautomerism of neutral xanthine and alloxanthine has been examined both in the gas phase and in aqueous solution. The tautomeric preference in the gas phase has been studied by means of semiempirical and ab initio quantum-mechanical computations with inclusion of correlation effects at the Møller-Plesset level, and from density-functional calculations. The influence of solvent on the relative stability between tautomers has been estimated from self-consistent reaction field calculations performed with different models. The results provide a detailed picture of tautomerism for these biologically relevant purine bases. The functional implications in the recognition by xanthine oxidase are analyzed from inspection of the interaction patterns of the most stable tautomeric forms. A model for the recognition of these purine derivatives in the enzyme binding site is discussed.


Subject(s)
Models, Chemical , Oxypurinol/chemistry , Xanthine Oxidase/metabolism , Xanthines/chemistry , Binding Sites , Drug Design , Gases , Molecular Structure , Solutions , Substrate Specificity , Thermodynamics , Water , Xanthine , Xanthine Oxidase/chemistry
9.
Proc Soc Exp Biol Med ; 211(4): 366-73, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8618943

ABSTRACT

Nitric oxide (NO), now almost synonymous with endothelium-derived relaxing factor (EDRF), reacts with superoxide anion radical (O2-) and forms a potentially toxic molecular species, peroxynitrite (ONOO-). Because xanthine oxidase (XO) seems to be a major O2- -producing enzyme in the vascular system, it is important to clarify the mechanism of XO regulation of NO/EDRF. We first characterized the inhibition of XO in vitro by three types of pyrazolopyrimidine derivatives. Kinetic studies indicated that 4-amino-6-hydroxpyrazolo[3,4-d]pyrimidine (AHPP) and allopurinol competitively inhibited the conversion of xanthine to uric acid catalyzed by XO, with apparent Ki values of 0.17 +/- 0.02 and 0.50 +/- 0.03 micro M respectively; alloxanthine inhibited this conversion in a noncompetitive manner with an apparent Ki value of 3.54 +/- 1.12 microM. O2- generation in the xanthine/XO system assayed by lucigenin-dependent chemiluminescence was suppressed most strongly by AHPP in a dose-dependent fashion; allopurinol itself appears to reduce the enzyme by transfer of an electron to O2, thus generating O(2-). AHPP significantly augmented EDRF-mediated relaxation of aortic rings from both rabbits and spontaneously hypertensive rats (SHR) in a dose-dependent manner, whereas allopurinol did not affect the relaxation and only marginal potentiation of the vasorelaxation was observed with alloxanthine. Finally, iv injection of AHPP (50.4 mg/kg; 100 micromol/300 g rat) reduced the blood pressure of SHR rats to 70% of the initial pressure; this pressure is almost the blood pressure of normal rats. Allopurinol (100 micromol/300 g rat; iv) showed transient decrease in blood pressure and moderate reduction of hypertension of SHR (10%) was observed with iv injection of alloxanthine (100 mumol/300 g rat). On the basis of these results, it seems that XO regulates EDRF/NO via production of O2-.


Subject(s)
Enzyme Inhibitors/toxicity , Nitric Oxide/pharmacology , Vasodilation/drug effects , Xanthine Oxidase/antagonists & inhibitors , Allopurinol/chemistry , Allopurinol/toxicity , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Blood Pressure/drug effects , Drug Interactions , Female , In Vitro Techniques , Luminescent Measurements , Male , Oxypurinol/chemistry , Oxypurinol/toxicity , Propylamines/chemistry , Propylamines/toxicity , Rabbits , Rats , Rats, Inbred SHR
10.
Eur J Clin Pharmacol ; 49(3): 215-20, 1995.
Article in English | MEDLINE | ID: mdl-8665998

ABSTRACT

We have investigated the disposition and plasma uric acid lowering effect of oxipurinol in ten healthy individuals following oral administration of three different formulations of oxipurinol and of allopurinol in equimolar doses. The reduction of plasma uric acid was clearcut up to 48 h. As estimated from plasma AUC0-infinity, Cmax, tmax, tlag, and urinary drug excretion, a conventional rapid release preparation of oxipurinol sodium was clearly superior to oxipurinol as free acid and to enteric coated microtablets of oxipurinol sodium. Plasma oxipurinol concentrations following a single dose of the conventional formulation of oxipurinol sodium were approximately 25% lower than those observed after an equimolar dose (300 mg) of allopurinol, but mean Cmax reached the value reported to be necessary for 90% inhibition of xanthine oxidase. Since prolonged administration will result in accumulation of oxipurinol because of its slow elimination, this type of oxipurinol formulation can be expected to meet the therapeutic requirements for a drug to lower plasma uric acid.


Subject(s)
Allopurinol/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , Gout Suppressants/pharmacokinetics , Oxypurinol/pharmacokinetics , Uric Acid/blood , Administration, Oral , Adult , Allopurinol/administration & dosage , Allopurinol/blood , Allopurinol/chemistry , Chemistry, Pharmaceutical , Cross-Over Studies , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/blood , Enzyme Inhibitors/chemistry , Gout Suppressants/administration & dosage , Gout Suppressants/blood , Gout Suppressants/chemistry , Humans , Male , Oxypurinol/administration & dosage , Oxypurinol/blood , Oxypurinol/chemistry
11.
J Biol Chem ; 266(9): 5608-15, 1991 Mar 25.
Article in English | MEDLINE | ID: mdl-2005100

ABSTRACT

Electron transfer within milk xanthine oxidase has been examined by the technique of pulse radiolysis. Radiolytically generated N-methylnicotinamide radical or 5-deazalumiflavin radical has been used to rapidly and selectively introduce reducing equivalents into the enzyme so that subsequent equilibration among the four redox-active centers of the enzyme (a molybdenum center, two iron-sulfur centers, and FAD) could be monitored spectrophotometrically. Experiments have been performed at pH 6 and 8.5, and a comprehensive scheme describing electron equilibration within the enzyme at both pH values has been developed. All rate constants ascribed to equilibration between specific pairs of centers in the enzyme are found to be rapid relative to enzyme turnover under the same conditions. Electron equilibration between the molybdenum center and one of the iron-sulfur centers of the enzyme (tentatively assigned Fe/S I) is particularly rapid, with a pH-independent first-order rate constant of approximately 8.5 x 10(3) s-1. The results unambiguously demonstrate the role of the iron-sulfur centers of xanthine oxidase in mediating electron transfer between the molybdenum and flavin centers of the enzyme.


Subject(s)
Electron Transport , Milk/enzymology , Xanthine Oxidase/chemistry , Alkylation , Animals , Hydrogen-Ion Concentration , Kinetics , Nicotinamide Mononucleotide/chemistry , Oxidation-Reduction , Oxypurinol/chemistry , Pulse Radiolysis , Spectrophotometry, Ultraviolet
SELECTION OF CITATIONS
SEARCH DETAIL
...