Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
J Vet Pharmacol Ther ; 44(1): 36-46, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32757313

ABSTRACT

The in vivo metabolism and pharmacokinetics of flunixin meglumine and phenylbutazone have been extensively characterized; however, there are no published reports describing the in vitro metabolism, specifically the enzymes responsible for the biotransformation of these compounds in horses. Due to their widespread use and, therefore, increased potential for drug-drug interactions and widespread differences in drug disposition, this study aims to build on the limited current knowledge regarding P450-mediated metabolism in horses. Drugs were incubated with equine liver microsomes and a panel of recombinant equine P450s. Incubation of phenylbutazone in microsomes generated oxyphenbutazone and gamma-hydroxy phenylbutazone. Microsomal incubations with flunixin meglumine generated 5-OH flunixin, with a kinetic profile suggestive of substrate inhibition. In recombinant P450 assays, equine CYP3A97 was the only enzyme capable of generating oxyphenbutazone while several members of the equine CYP3A family and CYP1A1 were capable of catalyzing the biotransformation of flunixin to 5-OH flunixin. Flunixin meglumine metabolism by CYP1A1 and CYP3A93 showed a profile characteristic of biphasic kinetics, suggesting two substrate binding sites. The current study identifies specific enzymes responsible for the metabolism of two NSAIDs in horses and provides the basis for future study of drug-drug interactions and identification of reasons for varying pharmacokinetics between horses.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Clonixin/analogs & derivatives , Cytochrome P-450 Enzyme System/metabolism , Horses/metabolism , Phenylbutazone/pharmacokinetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Clonixin/chemistry , Clonixin/metabolism , Clonixin/pharmacokinetics , DNA, Complementary/genetics , DNA, Complementary/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Microsomes, Liver/metabolism , Molecular Structure , Phenylbutazone/chemistry , Phenylbutazone/metabolism
2.
Methods ; 138-139: 69-75, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29471063

ABSTRACT

The nuclear Overhauser effect (NOE) is a primary means to characterize intermolecular interactions using modern NMR spectroscopy. Multiple experiments measured using different mixing time can be used for quantifying NOE buildup and measuring cross-relaxation rates. However, this approach using conventional multi-dimensional NMR is time consuming. Hyperpolarization by dissolution dynamic nuclear polarization (D-DNP) can generate deviations from equilibrium spin polarization by orders of magnitude, thereby enhancing signals and allowing to characterize NOE build up in real-time. Since most small molecules can be hyperpolarized using D-DNP, this method is applicable to the study of intermolecular interactions between small molecules and macromolecules. This application is demonstrated using a model system for host-guest interactions including the third generation polyamidoamine dendrimer (G3 PAMAM) and the pharmaceutical phenylbutazone (PBZ). After mixing 1H hyperpolarized PBZ with PAMAM, the NOE build up is directly observed at different sites of the dendrimer in series of one-dimensional NMR spectra. Cross-relaxation rates specific to individual source and target spins are determined from the build up curves. Further, the polarization enhancement is shown to be sufficiently large to allow identification of cross-peaks not observed in a conventional 2D-NOESY spectrum. The improved signal-to-noise ratio provided by hyperpolarization allows for characterizing the intermolecular interaction in an almost instantaneous measurement, opening an application to macromolecular and biomacromolecular NMR.


Subject(s)
Dendrimers/chemistry , Magnetic Resonance Spectroscopy , Phenylbutazone/chemistry , Polyamines/chemistry , Dendrimers/analysis
3.
Molecules ; 24(1)2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30609687

ABSTRACT

A drug design for safer phenylbutazone was been explored by reactivity and docking studies involving single electron transfer mechanism, as well as toxicological predictions. Several approaches about its structural properties were performed through quantum chemistry calculations at the B3LYP level of theory, together with the 6-31+G(d,p) basis sets. Molecular orbital and ionization potential were associated to electron donation capacity. The spin densities contribution showed a preferential hydroxylation at the para-positions of phenyl ring when compared to other positions. In addition, on electron abstractions the aromatic hydroxylation has more impact than alkyl hydroxylation. Docking studies indicate that six structures 1, 7, 8 and 13⁻15 have potential for inhibiting human as well as murine COX-2, due to regions showing similar intermolecular interactions to the observed for the control compounds (indomethacin and refecoxib). Toxicity can be related to aromatic hydroxylation. In accordance to our calculations, the derivatives here proposed are potentially more active as well safer than phenylbutazone and only structures 8 and 13⁻15 were the most promising. Such results can explain the biological properties of phenylbutazone and support the design of potentially safer candidates.


Subject(s)
Phenylbutazone/chemistry , Phenylbutazone/pharmacology , Drug Discovery/methods , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Conformation , Molecular Structure , Phenylbutazone/adverse effects , Phenylbutazone/toxicity , Structure-Activity Relationship
4.
J Mol Recognit ; 31(8): e2716, 2018 08.
Article in English | MEDLINE | ID: mdl-29659061

ABSTRACT

The intermolecular interaction of fosinopril, an angiotensin converting enzyme inhibitor with bovine serum albumin (BSA), has been investigated in physiological buffer (pH 7.4) by multi-spectroscopic methods and molecular docking technique. The results obtained from fluorescence and UV absorption spectroscopy revealed that the fluorescence quenching mechanism of BSA induced by fosinopril was mediated by the combined dynamic and static quenching, and the static quenching was dominant in this system. The binding constant, Kb , value was found to lie between 2.69 × 103 and 9.55 × 103  M-1 at experimental temperatures (293, 298, 303, and 308 K), implying the low or intermediate binding affinity between fosinopril and BSA. Competitive binding experiments with site markers (phenylbutazone and diazepam) suggested that fosinopril preferentially bound to the site I in sub-domain IIA on BSA, as evidenced by molecular docking analysis. The negative sign for enthalpy change (ΔH0 ) and entropy change (ΔS0 ) indicated that van der Waals force and hydrogen bonds played important roles in the fosinopril-BSA interaction, and 8-anilino-1-naphthalenesulfonate binding assay experiments offered evidence of the involvements of hydrophobic interactions. Moreover, spectroscopic results (synchronous fluorescence, 3-dimensional fluorescence, and Fourier transform infrared spectroscopy) indicated a slight conformational change in BSA upon fosinopril interaction.


Subject(s)
Fosinopril/chemistry , Protein Binding , Serum Albumin, Bovine/chemistry , Animals , Binding Sites/drug effects , Binding, Competitive/drug effects , Cattle , Diazepam/chemistry , Fosinopril/pharmacology , Hydrogen-Ion Concentration , Molecular Docking Simulation , Phenylbutazone/chemistry , Serum Albumin, Bovine/drug effects , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared , Temperature
5.
Int J Mol Sci ; 19(10)2018 Sep 21.
Article in English | MEDLINE | ID: mdl-30241420

ABSTRACT

Human serum albumin (HSA) is a target for reactive oxygen species (ROS), and alterations of its physiological functions caused by oxidation is a current issue. In this work, the amino-acid residues Trp-214 and Lys-199, which are located at site I of HSA, were experimentally and computationally oxidized, and the effect on the binding constant with phenylbutazone was measured. HSA was submitted to two mild oxidizing reagents, taurine monochloramine (Tau-NHCl) and taurine dibromamine (Tau-NBr2). The oxidation of Trp-214 provoked spectroscopic alterations in the protein which were consistent with the formation of N'-formylkynurenine. It was found that the oxidation of HSA by Tau-NBr2, but not by Tau-NHCl, provoked a significant increase in the association constant with phenylbutazone. The alterations of Trp-214 and Lys-199 were modeled and simulated by changing these residues using the putative oxidation products. Based on the Amber score function, the interaction energy was measured, and it showed that, while native HSA presented an interaction energy of -21.3 kJ/mol, HSA with Trp-214 altered to N'-formylkynurenine resulted in an energy of -28.4 kJ/mol, and HSA with Lys-199 altered to its carbonylated form resulted in an energy of -33.9 kJ/mol. In summary, these experimental and theoretical findings show that oxidative alterations of amino-acid residues at site I of HSA affect its binding efficacy.


Subject(s)
Lysine/chemistry , Models, Theoretical , Phenylbutazone/metabolism , Serum Albumin, Human/metabolism , Tryptophan/chemistry , Binding Sites , Humans , Oxidation-Reduction , Phenylbutazone/chemistry , Protein Binding , Serum Albumin, Human/chemistry
6.
Chem Res Toxicol ; 28(7): 1476-83, 2015 Jul 20.
Article in English | MEDLINE | ID: mdl-26090772

ABSTRACT

We investigated the effect of Cu,Zn-superoxide dismutase (Cu,Zn-SOD)-peroxidase activity on the oxidation of the nonsteroidal anti-inflammatory drug phenylbutazone (PBZ). We utilized electron paramagnetic resonance (EPR) spectroscopy to detect free radical intermediates of PBZ, UV-vis spectrophotometry to monitor PBZ oxidation, oxygen analysis to determine the involvement of C-centered radicals, and LC/MS to determine the resulting metabolites. Using EPR spectroscopy and spin-trapping with 5,5-dimethyl-1-pyrroline-N-oxide (DMPO), we found that the spin adduct of CO3(•-) (DMPO/(•)OH) was attenuated with increasing PBZ concentrations. The resulting PBZ radical, which was assigned as a carbon-centered radical based on computer simulation of hyperfine splitting constants, was trapped by both DMPO and MNP spin traps. Similar to Cu,Zn-SOD-peroxidase activity, an identical PBZ carbon-centered radical was also detected with the presence of both myeloperoxidase (MPO/H2O2) and horseradish peroxidase (HRP/H2O2). Oxygen analysis revealed depletion in oxygen levels when PBZ was oxidized by SOD peroxidase-activity, further supporting carbon radical formation. In addition, UV-vis spectra showed that the λmax for PBZ (λ = 260 nm) declined in intensity and shifted to a new peak that was similar to the spectrum for 4-hydroxy-PBZ when oxidized by Cu,Zn-SOD-peroxidase activity. LC/MS evidence supported the formation of 4-hydroxy-PBZ when compared to that of a standard, and 4-hydroperoxy-PBZ was also detected in significant yield. These findings together indicate that the carbonate radical, a product of SOD peroxidase activity, appears to play a role in PBZ metabolism. Interestingly, these results are similar to findings from heme peroxidase enzymes, and the context of this metabolic pathway is discussed in terms of a mechanism for PBZ-induced toxicity.


Subject(s)
Phenylbutazone/metabolism , Superoxide Dismutase/metabolism , Chromatography, High Pressure Liquid , Cyclic N-Oxides/chemistry , Electron Spin Resonance Spectroscopy , Free Radicals/chemistry , Mass Spectrometry , Oxidation-Reduction , Oxygen/analysis , Oxygen/chemistry , Phenylbutazone/chemistry , Spectrophotometry, Ultraviolet
7.
J Phys Chem B ; 113(24): 8339-46, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19469491

ABSTRACT

The interactions between dendrimers and surfactants led to the formation of aggregates dispersed in aqueous solutions. The potential of the resulting dendrimer-surfactant aggregates as new drug formulations was evaluated. The size, morphology, and stability of the aggregates and the localization of drugs in them were determined by dynamic laser light scattering, atomic force microscopy, agarose gel electrophoresis, and nuclear magnetic resonance studies. The drug-loaded aggregates have a spherical shape and an average size of 40 nm. The drug-loading efficiency of dendrimers is significantly influenced in the presence of surfactants. The release rate of the drugs from the dendrimer-surfactant aggregates can be modulated by varying the amount of surfactant in the aggregates. The dendrimer-surfactant aggregates are promising carriers for hydrophobic drugs in transdermal administration routes.


Subject(s)
Dendrimers/chemistry , Drug Carriers/chemistry , Drug Design , Methotrexate/chemistry , Phenylbutazone/chemistry , Sulfamethoxazole/chemistry , Surface-Active Agents/chemistry , Cell Survival/drug effects , Chemistry, Pharmaceutical , Dendrimers/pharmacology , HeLa Cells , Humans , Methotrexate/pharmacology , Molecular Structure , Phenylbutazone/pharmacology , Solubility , Sulfamethoxazole/pharmacology , Surface-Active Agents/pharmacology
8.
Int J Biol Macromol ; 134: 645-652, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31100401

ABSTRACT

Human serum albumin (HSA) is the binding cargo in blood plasma. The binding of drugs to HSA determines the pharmacokinetics and pharmacodynamics of the drugs. There are 67 natural genetic variants of HSA were reported in literature. Studying the effect of albumin modifications on drug binding helps to treat the patients with proper medication. In the present study, we have aimed to understand the effect of two natural variants of HSA, such as Herborn (K240E) and Milano Slow (D375H) on the binding of phenylbutazone and ibuprofen. For this, we have generated K240E and D375H mutants and also double mutant (K240E/D375H) of HSA using site directed mutagenesis. The recombinant HSA and its variants were expressed in Pichia pastoris. The interaction of HSA and its variants to phenylbutazone and ibuprofen was studied using fluorescence spectroscopy. Our results showed that there is no significant effect of K240E and D375H mutations on phenylbutazone and ibuprofen binding. But the effect is significant when both the mutations were there in a single protein (K240E/D375H). Further, the CD spectroscopy data showed that there is no effect of phenylbutazone and ibuprofen binding on the conformation of protein, except in case of D375H, where there is a conformational change in the binding pocket with the ibuprofen binding.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Ibuprofen/chemistry , Mutant Proteins , Phenylbutazone/chemistry , Serum Albumin, Human/chemistry , Serum Albumin, Human/genetics , Alleles , Amino Acid Substitution , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Circular Dichroism , Fluorescent Antibody Technique , Humans , Ibuprofen/metabolism , Mutagenesis, Site-Directed , Phenylbutazone/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Recombinant Proteins , Spectrometry, Fluorescence , Structure-Activity Relationship
9.
Arch Biochem Biophys ; 478(1): 75-80, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18602884

ABSTRACT

Phenylbutazone (PB) is known to be biotransformed to its O- and C-glucuronide. Recently, we reported that PB C-glucuronide formation is catalyzed by UGT1A9. Interestingly, despite UGT1A8 sharing high amino acid sequence identity with UGT1A9, UGT1A8 had no PB C-glucuronidating activity. In the present study, we constructed eight UGT1A9/UGT1A8 chimeras and evaluated which region is important for PB C-glucuronide formation. All of the chimeras and UGT1A8 and UGT1A9 had 7-hydroxy-(4-trifluoromethyl)coumarin (HFC) O-glucuronidating activity. The K(m) values for HFC glucuronidation of UGT1A8, UGT1A9 and their chimeras were divided into two types, UGT1A8 type (high K(m)) and UGT1A9 type (low K(m)), and these types were determined according to whether their amino acids at positions 69-132 were those of UGT1A8 or UGT1A9. Likewise, PB O-glucuronidating activity was also detected by all of the chimeras, and their K(m) values were divided into two types. On the contrary, PB C-glucuronidating activity was detected by UGT1A9((1-132))/1A8((133-286)), UGT1A9((1-212))/1A8((213-286)), UGT1A8((1-68))/1A9((69-286)), and UGT1A8((1-68))/1A9((69-132))/1A8((133-286)) chimeras. The region 1A9((69-132)) was common among chimeras having PB C-glucuronidating activity. Of interest is that UGT1A9((1-68))/1A8((69-132))/1A9((133-286)) had lost PB C-glucuronidation activity, but retained activities of PB and HFC O-glucuronidation. These results strongly suggested that amino acid positions 69-132 of UGT1A9 are responsible for chemoselectivity for PB and affinity to substrates such as PB and HFC.


Subject(s)
Amino Acids/chemistry , Glucuronosyltransferase/chemistry , Phenylbutazone/chemistry , Catalysis , Coumarins/pharmacology , DNA Primers/chemistry , Glucuronosyltransferase/metabolism , Humans , Kinetics , Models, Biological , Phenylbutazone/pharmacology , Protein Isoforms , Recombinant Fusion Proteins/chemistry , UDP-Glucuronosyltransferase 1A9
10.
J Biomol Struct Dyn ; 35(10): 2280-2292, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27453381

ABSTRACT

In this study, molecular binding affinity was investigated for Nefopam analogues (NFs), a functionalized benzoxazocine, with human serum albumin (HSA), a major transport protein in the blood. Its binding affinity and concomitant changes in its conformation, binding site and simulations were also studied. Fluorescence data revealed that the fluorescence quenching of HSA upon binding of NFs analogues is based on a static mechanism. The three analogues of NFs binding constants (KA) are in the order of NF3 > NF2 > NF1 with values of 1.53 ± .057 × 104, 2.16 ± .071 × 104 and 3.6 ± .102 × 105 M-1, respectively. Concurrently, thermodynamic parameters indicate that the binding process was spontaneous, and the complexes were stabilized mostly by hydrophobic interactions, except for NF2 has one hydrogen bond stabilizes it along with hydrophobic interactions. Circular dichroism (CD) studies revealed that there is a decrease in α-helix with an increase in ß-sheets and random coils signifying partial unfolding of the protein upon binding of NFs, which might be due to the formation of NFs-HSA complexes. Further, molecular docking studies showed that NF1, NF2 and NF3 bound to subdomains IIIA, IB and IIA through hydrophobic interactions. However, NF1 have additionally formed a single hydrogen bond with LYS 413. Furthermore, molecular simulations unveiled that NFs binding was in support with the structural perturbation observed in CD, which is evident from the root mean square deviation and Rg fluctuations. We hope our insights will provide ample scope for engineering new drugs based on the resemblances with NFs for enhanced efficacy with HSA.


Subject(s)
Analgesics, Non-Narcotic/chemistry , Ibuprofen/chemistry , Lidocaine/chemistry , Nefopam/chemistry , Phenylbutazone/chemistry , Serum Albumin, Human/chemistry , Binding Sites , Binding, Competitive , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Kinetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Nefopam/analogs & derivatives , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Stability , Solutions , Thermodynamics
11.
Basic Clin Pharmacol Toxicol ; 99(3): 261-6, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16930300

ABSTRACT

To clarify the action of a side-effect of phenylbutazone, we investigated the inactivation of alpha(1)-antiproteinase induced by phenylbutazone in the presence of horseradish peroxidase (HRP) and H(2)O(2) (HRP-H(2)O(2)). The activity of alpha(1)-antiproteinase was rapidly lost during the interaction of phenylbutazone with HRP-H(2)O(2) under aerobic conditions. Phenylbutazone showed a marked spectral change under aerobic conditions but not under anaerobic conditions. Spin trap agents were very effective in inhibiting alpha(1)-antiproteinase inactivation induced by phenylbutazone. Oxidation of phenylbutazone was stopped by catalase, but the inactivation reaction of alpha(1)-antiproteinase proceeded even after removal of H(2)O(2) in the reaction mixture. Formation of the peroxidative product from phenylbutazone was detected by iodometric assay. These results indicate that both peroxyl radicals and the peroxidative product of phenylbutazone participated in the inactivation of alpha(1)-antiproteinase. Other anti-inflammatory drugs did not inactivate alpha(1)-antiproteinase during interaction with HRP-H(2)O(2). Inactivation of alpha(1)-antiproteinase may contribute to serious side effects of phenylbutazone.


Subject(s)
Free Radicals/metabolism , Hydrogen Peroxide/metabolism , Phenylbutazone/pharmacology , Serine Proteinase Inhibitors/pharmacology , alpha 1-Antitrypsin/metabolism , Amino Acids/metabolism , Free Radical Scavengers , Humans , Molecular Structure , Phenylbutazone/chemistry , Phenylbutazone/metabolism , Time Factors
12.
Chem Biol Drug Des ; 87(6): 936-45, 2016 06.
Article in English | MEDLINE | ID: mdl-26808199

ABSTRACT

As a NPY-2 receptor agonist, PYY24-36- Leu31 is reported to suppress appetite and has a potential in obesity treatment, but its short half-life limits the clinical application. The use of chemical modification to improve interactions with human serum albumin (HSA) is an effective strategy for prolonging the half-lives of peptide analogues. So based on the characteristics that phenylbutazone has a good combination with HSA, we selected a proper linker to link with PYY24-36 -Leu31 to create long-acting and highly biologically active PYY24-36 -Leu31 conjugates, and successfully find a novel, long-acting PYY24-36 -Leu31 conjugate 8 that, when dosed every other day in diet induce obese (DIO) mice for 2 weeks, results in a significant reduction in food intake and body weight and improvement in blood parameter and hepatic steatosis.


Subject(s)
Drug Carriers , Fatty Liver/drug therapy , Phenylbutazone , Receptors, Neuropeptide Y/agonists , Serum Albumin , Animals , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Fatty Liver/blood , Humans , Male , Mice , Mice, Inbred ICR , Mice, Obese , Phenylbutazone/chemistry , Phenylbutazone/pharmacokinetics , Phenylbutazone/pharmacology , Receptors, Neuropeptide Y/metabolism , Serum Albumin/chemistry , Serum Albumin/pharmacokinetics , Serum Albumin/pharmacology
13.
Int J Pharm ; 489(1-2): 45-57, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25888803

ABSTRACT

The robustness of the polymer strip film platform to successfully deliver a variety of BCS Class II drug nanoparticles without the need for surfactant while retaining positive characteristics such as nanoparticle redispersibility and fast dissolution is demonstrated. Fenofibrate (FNB), griseofulvin (GF), naproxen (NPX), phenylbutazone (PB), and azodicarbonamide (AZD) were considered as model poorly water-soluble drugs. Their aqueous nanosuspensions, produced via wet stirred media milling, were mixed with hydroxypropyl methylcellulose solution containing glycerin as plasticizer, followed by casting and drying to form films. For the purpose of comparison, sodium dodecyl sulfate (SDS) was used as surfactant, but was found to be unnecessary for achieving fast dissolution (t80 between 18 and 28 min) for all five drugs. Interestingly, SDS was required for the full recovery of nanoparticles for PB, yet lack of it did not impact the dissolution. Interactions between drug and polymer were investigated with FTIR spectroscopy whereas drug crystallinity within the film was investigated via Raman spectroscopy. Films for all drugs, even for very small samples, exhibited excellent content uniformity (RSD <4%) regardless of use of surfactant. Overall, these results demonstrate the novelty and robustness of the polymer strip film platform for fast release of poorly water-soluble drugs without requiring any surfactants.


Subject(s)
Drug Delivery Systems , Hypromellose Derivatives/chemistry , Nanoparticles/chemistry , Azo Compounds/chemistry , Fenofibrate/chemistry , Glycerol/chemistry , Griseofulvin/chemistry , Naproxen/chemistry , Phenylbutazone/chemistry , Solubility
14.
Biochem Pharmacol ; 43(5): 931-5, 1992 Mar 03.
Article in English | MEDLINE | ID: mdl-1554390

ABSTRACT

The binding of phenylbutazone (PB) to human serum albumin (HSA) at different pH and in the presence of different NaSCN and urea concentrations that alter the conformation of the protein was examined qualitatively on the basis of extrinsic elliptical strength at 288 nm by means of circular dichroism (CD). The values of the binding index expressed as a ratio of [theta]max/[theta]pH7.4(288) at each extrinsic rotational strength in the presence of various concentrations of NaSCN, urea and hydrogen ion were directly proportional to the alpha-helix content based on the peptide backbone alteration of HSA by NaSCN, urea and hydrogen ion except for the pH range of 5.0 to 10.0. The values in the pH range of 7.4 to 10.0 depended on the concentration of hydrogen ion and not on the alpha-helix content, showing a significant effect of the hydrogen ion on the tertiary conformation with respect to the binding sites of the amino acid chain rather than the peptide backbone of HSA. The increases in the binding index observed in the pH range of 7.4 to 10.0 were not observed at all in the case of NaSCN and urea at the concentrations studied. It was demonstrated that the binding of PB to HSA increased with the change in the tertiary conformation caused by hydrogen ions but decreased with that in the secondary conformation caused by a concentration change of NaSCN and urea. Thus, the binding was closely associated with skeletal conformational alterations as well as changes in the binding sites of the amino acid chains of the protein.


Subject(s)
Phenylbutazone/chemistry , Serum Albumin/chemistry , Circular Dichroism , Humans , Hydrogen-Ion Concentration , Protein Binding/drug effects , Protein Conformation/drug effects , Thiocyanates/pharmacology , Urea/pharmacology
15.
Biochem Pharmacol ; 44(5): 873-9, 1992 Sep 01.
Article in English | MEDLINE | ID: mdl-1382424

ABSTRACT

The fluorescent probes warfarin and dansylsarcosine are known to selectively interact with binding sites I and II, respectively, on human albumin. This paper investigates whether similar binding sites exist on bovine, dog, horse, sheep and rat albumins. Binding sites on albumins were studied by: (1) displacement of warfarin and dansylsarcosine by site I (phenylbutazone) and site II (diazepam) selective ligands; (2) the effects of non-esterified fatty acids (carbon chain lengths: C5-C20) and changes in pH (6-9) on the fluorescence of warfarin and dansylsarcosine; and (3) the ability of site selective ligands to inhibit hydrolysis of 4-nitrophenyl acetate. For bovine, dog, horse, human and sheep albumins the fluorescence of bound warfarin and dansylsarcosine was selectively decreased by phenylbutazone and diazepam, respectively. For these albumins medium chain fatty acids (C1-C12) reduced the fluorescence of dansylsarcosine (maximum inhibition with C9) whereas long chain acids (C12-C20) enhanced the fluorescence of warfarin (maximum increases with C12). In addition, changes in pH from 6 to 9 increased the fluorescence of warfarin and although site I ligands (warfarin/phenylbutazone) had no pronounced effects on 4-nitrophenyl acetate hydrolysis, site II ligands (dansylsarcosine/diazepam) significantly inhibited this reaction. Rat albumin behaved differently from the other albumins studied in that the C12-C20 fatty acids and changes in pH did not enhance the fluorescence of warfarin. Moreover, the differential effects of site I and site II ligands on the fluorescence of warfarin/dansylsarcosine and hydrolysis of 4-nitrophenyl acetate were less apparent with rat albumin. The results suggest bovine, dog, horse and sheep albumins have binding sites for warfarin and dansylsarcosine with similar properties to sites I and II on human albumin. By contrast, the warfarin binding site and to a lesser degree the dansylsarcosine site, of rat albumin have different characteristics from these sites on the other albumins studied.


Subject(s)
Albumins/chemistry , Animals , Binding Sites , Binding, Competitive , Cattle , Dansyl Compounds/chemistry , Diazepam/chemistry , Diazepam/pharmacology , Dogs , Fatty Acids, Nonesterified/pharmacology , Fluorescence , Horses , Humans , Hydrogen-Ion Concentration , Kinetics , Nitrophenols/chemistry , Phenylbutazone/chemistry , Phenylbutazone/pharmacology , Rats , Sarcosine/analogs & derivatives , Sarcosine/chemistry , Sheep , Warfarin/chemistry
16.
J Pharm Sci ; 80(4): 357-62, 1991 Apr.
Article in English | MEDLINE | ID: mdl-1865336

ABSTRACT

The solid-state 13C NMR spectra of mofebutazone, phenylbutazone, and oxyphenbutazone monohydrate and anhydrate are presented. The crystal structures of these pyrazolidinedione derivatives, obtained by single-crystal X-ray analysis, were previously reported, revealing distinct differences in crystal structure. In this report, the chemical shift values observed for the solid-state 13C spectra are related to the chemical environment of the various carbon atoms and compared with the crystallographic data. Results indicate that solid-state NMR spectroscopy is potentially useful in the study of drugs in the solid state.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Oxyphenbutazone/chemistry , Phenylbutazone/analogs & derivatives , Phenylbutazone/chemistry , Carbon Isotopes , Computer Simulation , Magnetic Resonance Spectroscopy/methods , Models, Molecular , Molecular Conformation , X-Ray Diffraction
17.
J Pharm Sci ; 84(6): 737-41, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7562415

ABSTRACT

Simultaneous binding of two drugs to human serum albumin (HSA) was investigated by difference circular dichroism (delta CD) spectroscopy. Phenylbutazone and diazepam were chosen as specific markers for binding areas I (cumarines) and II (indoles), respectively, and their stereospecific interactions with protein were selectively characterized. Displacers were drugs known to specifically bind to areas I (salicylate) and II (racemic ibuprofen). The results indicate two different interaction mechanisms: a direct competition one (diazepam-ibuprofen and phenylbutazone-salicylate) and an indirect competition one (diazepam-salicylate and phenylbutazone-ibuprofen). The two major binding areas on HSA are distinct, but not independent, entities. Finally, the dissociation constants of marker ligands and competitors complexed to HSA were determined by quantitative analysis of CD data.


Subject(s)
Serum Albumin/chemistry , Binding, Competitive , Circular Dichroism , Diazepam/chemistry , Humans , Ibuprofen/chemistry , Ligands , Phenylbutazone/chemistry , Protein Binding , Salicylates/chemistry , Stereoisomerism
18.
J Pharm Sci ; 83(12): 1712-6, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7891299

ABSTRACT

Binding sites on human serum albumin (HSA) for anionic drugs and fatty acids have been thermodynamically characterized by microcalorimetry. The binding and the thermodynamic parameters were directly computed from the calorimetric titration data at 37 degrees C in a phosphate buffer (pH 7.4) using one- and two-class binding models. From compensation analyses plotting the molar enthalpy change (delta Hm,i) versus those of the molar free energy (delta Gm,i) and molar entropy (delta Sm,i) for each class of binding sites, HSA binding sites were classified into groups S1, S2, and S3. Group S1 included high-affinity binding sites for site II-bound drugs, such as ibuprofen, flufenamic acid, and ethacrynic acid, and short- or medium-length alkyl-chain fatty acids; group S2 included low-affinity binding sites of site II-bound drugs and long-length alkyl-chain fatty acids; and group S3 contained the high-affinity binding sites for site I-bound drugs, such as phenylbutazone, oxphenbutazone, and warfarin, and long-length alkyl-chain fatty acids. High- and low-affinity bindings sites for salicylic acid and acetylaslicylic acid agreed with the regions of groups S3 and S2, respectively. Groups S1 and S2 were characterized by large negative values of delta Hm,i and delta Sm,i, reflecting van der Waals interaction and hydrogen-bonding formation in low dielectric media, and the main force to stabilize the binding complex in group S3 was a hydrophobic interaction, characterized by a small negative delta Hm,i and minor or positive values of delta Sm,i (entropy-driven).


Subject(s)
Pharmaceutical Preparations/metabolism , Serum Albumin/metabolism , Anions , Binding Sites , Calorimetry/methods , Fatty Acids/metabolism , Humans , Pharmaceutical Preparations/chemistry , Phenylbutazone/analogs & derivatives , Phenylbutazone/chemistry , Phenylbutazone/metabolism , Protein Binding , Serum Albumin/chemistry , Thermodynamics , Warfarin/chemistry , Warfarin/metabolism
19.
Int J Pharm ; 195(1-2): 1-6, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10675674

ABSTRACT

Surface modification and solid dispersion formulations using hydrophilic excipients can significantly alter the dissolution behaviour of hydrophobic drug materials. The effect of these techniques used individually and in combination on the dissolution properties of the hydrophobic drug, phenylbutazone (PB), are compared. PB was treated with a poloxamer, Synperonic((R)) F127 by an adsorption method. Solid dispersions (10 and 20% w/w) were prepared with untreated PB or PB previously modified with Synperonic((R)) F127 (PBT) in molten F127. Dissolution tests of capsule formulations of PB, PBT and solid dispersion formulations, in pH 6.4 buffer at 37+/-0.5 degrees C demonstrated that after 140 min, release of PB was 16.7%, but 71.4% from the solid dispersion, whereas from the PBT formulation 85.6% was released. The Synperonic((R)) F127 content of PBT was only 0.05% of that in the solid dispersion formulation which suggests that it is the nature of the drug polymer contact rather than the amount of polymer which is more critical in influencing dissolution behaviour. Comparison of PBT and the 10% w/w solid dispersion of PBT in F127 showed similar amounts of drug in solution after 140 min. However there was a significantly higher release rate for PBT. Both formulation techniques offer significant improvements in drug release over untreated PB, and a combination of techniques changes the rate but not the extent of release in comparison with the surface modification technique alone.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Chemistry, Pharmaceutical , Phenylbutazone/chemistry , Poloxamer/pharmacology , Surface-Active Agents/pharmacology , Adsorption , Capsules , Surface Properties
20.
J Pharm Biomed Anal ; 16(2): 287-93, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9408846

ABSTRACT

A direct injection HPLC method has been developed for the determination of phenylbutazone and its active metabolite oxyphenylbutazone in serum using a semipermeable surface (SPS) column. The method is easy to perform and requires 20 microliters of a filtered serum sample. The chromatographic time is less than 13 min using a mobile phase of 15:85 v/v acetonitrile-0.05M phosphate buffer pH 7.5. The method was linear in the range 0.5-20 micrograms ml-1 (r > 0.99, n = 6) with R.S.D. less than 6%. Interday and intraday variability were found to be less than 8.3%. The limit of quantitation and detection were 0.5 and 0.25 microgram ml-1 (s/n > 3), respectively, for both drug and metabolite.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Chromatography, High Pressure Liquid/methods , Oxyphenbutazone/isolation & purification , Phenylbutazone/isolation & purification , Animals , Anti-Inflammatory Agents, Non-Steroidal/blood , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Horses , Oxyphenbutazone/blood , Oxyphenbutazone/chemistry , Phenylbutazone/blood , Phenylbutazone/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL