Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 74(10): 1859-1869, 2017 05.
Article in English | MEDLINE | ID: mdl-28083596

ABSTRACT

Cytochrome P450 2U1 (CYP2U1) exhibits several distinctive characteristics among the 57 human CYPs, such as its presence in almost all living organisms with a highly conserved sequence, its particular gene organization with only five exons, its major location in thymus and brain, and its protein sequence involving an unusually long N-terminal region containing 8 proline residues and an insert of about 20 amino acids containing 5 arginine residues after the transmembrane helix. Few substrates, including fatty acids, N-arachidonoylserotonin (AS), and some drugs, have been reported so far. However, its biological roles remain largely unknown, even though CYP2U1 mutations have been involved in some pathological situations, such as complicated forms of hereditary spastic paraplegia. These data together with its ability to hydroxylate some fatty acids and AS suggest its possible role in lipid metabolism.


Subject(s)
Cytochrome P450 Family 2/analysis , Cytochrome P450 Family 2/metabolism , Amino Acid Sequence , Animals , Arachidonic Acids/metabolism , Brain/metabolism , Cytochrome P450 Family 2/genetics , Fatty Acids/metabolism , Humans , Hydroxylation , Molecular Docking Simulation , Mutation , Sequence Alignment , Serotonin/analogs & derivatives , Serotonin/metabolism , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism , Substrate Specificity , Thymus Gland/metabolism
2.
Ann Pharm Fr ; 69(1): 62-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21296219

ABSTRACT

The present brief overview of the history of the development of our knowledge on cytochromes P450 (P450s) illustrates the spectacular progress that have been made on P450 mechanisms and structures especially during these last 20 years. Recently published structures of mammalian P450-substrate complexes have shown the great diversity of size, shape, and binding modes that are offered by the conformationally flexible substrate binding sites of xenobiotic-metabolizing P450s. They have also shown that these binding sites can adapt themselves to the great structural diversity of xenobiotics, to facilitate their oxidation and elimination. Our present detailed knowledge of the mechanisms and chemistry of P450s allows us to understand, at the molecular level, the origin of the various consequences of P450-dependent metabolism of drugs in pharmacology and toxicology. This is here illustrated by recent data on the detailed mechanism of bioactivation of the anti-thrombotic prodrugs ticlopidine, clopidogrel, and prasugrel.


Subject(s)
Aerobiosis/physiology , Biotransformation/physiology , Cytochrome P-450 Enzyme System/history , Cytochrome P-450 Enzyme System/metabolism , Prodrugs/metabolism , Adaptation, Physiological , Animals , Environment , Fibrinolytic Agents/metabolism , History, 20th Century , History, 21st Century , Humans
3.
Chem Res Toxicol ; 16(12): 1547-54, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14680368

ABSTRACT

Nitric oxide synthases (NOSs) are flavohemeproteins that catalyze the oxidation of l-arginine to l-citrulline with formation of the widespread signal molecule NO. Beside their fundamental role in NO biosynthesis, these enzymes are also involved in the formation of reactive oxygen species and in the interactions with some xenobiotic compounds. Nilutamide is a nonsteroidal antiandrogen that behaves as a competitive antagonist of the androgen receptors and is proposed in the treatment of metastatic prostatic carcinoma. However, therapeutic effects of nilutamide are overshadowed by the occurrence of several adverse reactions mediated by toxic mechanism(s), which remain(s) poorly investigated. Here, we studied the interaction of NOSs with nilutamide. Our results show that the purified recombinant neuronal NOS reduced the nitroaromatic nilutamide to the corresponding hydroxylamine. The reduction of nilutamide catalyzed by neuronal NOS proceeded with intermediate formation of a nitro anion free radical easily observed by EPR, was insensitive to the addition of the usual heme ligands and l-arginine analogues, but strongly inhibited by O(2) and a flavin/NADPH binding inhibitor. Involvement of the reductase domain of nNOS in the reduction of nilutamide was confirmed by (i) the ability of the isolated reductase domain of nNOS to catalyze the reaction and (ii) the stimulating effect of Ca(2+)/calmodulin on the accumulation of hydroxylamine and nitro anion radical. In a similar manner, the recombinant inducible and endothelial NOS isoforms also displayed nitroreductase activity, albeit with lower yields. The selective reduction of nilutamide to its hydroxylamino derivative by the NOSs could explain some of the toxic effects of this drug.


Subject(s)
Androgen Antagonists/metabolism , Imidazoles/metabolism , Imidazolidines , Nitric Oxide Synthase/metabolism , Amines/chemistry , Amines/metabolism , Anaerobiosis , Androgen Antagonists/adverse effects , Androgen Antagonists/chemistry , Animals , Cattle , Electron Spin Resonance Spectroscopy , Free Radicals/chemistry , Free Radicals/metabolism , Imidazoles/adverse effects , Imidazoles/chemistry , Mice , NADP/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Oxidation-Reduction , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Time Factors
4.
Biochemistry ; 40(40): 12112-22, 2001 Oct 09.
Article in English | MEDLINE | ID: mdl-11580286

ABSTRACT

Experiments using recombinant yeast-expressed human liver cytochromes P450 confirmed previous literature data indicating that ticlopidine is an inhibitor of CYP 2C19. The present studies demonstrated that ticlopidine is selective for CYP 2C19 within the CYP 2C subfamily. UV-visible studies on the interaction of a series of ticlopidine derivatives with CYP 2C19 showed that ticlopidine binds to the CYP 2C19 active site with a K(s) value of 2.8 +/- 1 microM. Derivatives that do not involve either the o-chlorophenyl substituent, the free tertiary amine function, or the thiophene ring of ticlopidine did not lead to such spectral interactions and failed to inhibit CYP 2C19. Ticlopidine is oxidized by CYP 2C19 with formation of two major metabolites, the keto tautomer of 2-hydroxyticlopidine (1) and the dimers of ticlopidine S-oxide (TSOD) (V(max) = 13 +/- 2 and 0.4 +/- 0.1 min(-1)). During this oxidation, CYP 2C19 was inactivated; the rate of its inactivation was time and ticlopidine concentration dependent. This process meets the chemical and kinetic criteria generally accepted for mechanism-based enzyme inactivation. It occurs in parralel with CYP 2C19-catalyzed oxidation of ticlopidine, is inhibited by an alternative well-known substrate of CYP 2C19, omeprazole, and correlates with the covalent binding of ticlopidine metabolite(s) to proteins. Moreover, CYP 2C19 inactivation is not inhibited by the presence of 5 mM glutathione, suggesting that it is due to an alkylation occurring inside the CYP 2C19 active site. The effects of ticlopidine on CYP 2C19 are very analogous with those previously described for the inactivation of CYP 2C9 by tienilic acid. This suggests that a similar electrophilic intermediate, possibly a thiophene S-oxide, is involved in the inactivation of CYP 2C19 and CYP 2C9 by ticlopidine and tienilic acid, respectively. The kinetic parameters calculated for ticlopidine-dependent inactivation of CYP 2C19, i.e., t(1/2max) = 3.4 min, k(inact) = 3.2 10(-3) s(-1), K(I) = 87 microM, k(inact)/K(I) = 37 L.mol(-1).s(-1), and r (partition ratio) = 26 (in relation with formation of 1 + TSOD), classify ticlopidine as an efficient mechanism-based inhibitor although somewhat less efficient than tienilic acid for CYP 2C9. Importantly, ticlopidine is the first selective mechanism-based inhibitor of human liver CYP 2C19 and should be a new interesting tool for studying the topology of the active site of CYP 2C19.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Mixed Function Oxygenases/antagonists & inhibitors , Ticlopidine/pharmacology , Alkylation , Binding Sites , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2C19 , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/chemistry , Glutathione/pharmacology , Humans , Kinetics , Liver/enzymology , Mixed Function Oxygenases/metabolism , Molecular Structure , Omeprazole/pharmacology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Spectrum Analysis , Ticlopidine/antagonists & inhibitors , Ticlopidine/chemistry
5.
Arch Biochem Biophys ; 394(2): 189-200, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11594733

ABSTRACT

A series of new derivatives of sulfaphenazole (SPA), in which the NH(2) and phenyl substituents of SPA are replaced by various groups or in which the sulfonamide function of SPA is N-alkylated, were synthesized in order to further explore CYP 2C9 active site and to determine the structural factors explaining the selectivity of SPA for CYP 2C9 within the human P450 2C subfamily. Compounds in which the NH(2) group of SPA was replaced with R(1) = CH(3), Br, CH = CH(2), CH(2)CH = CH(2), and CH(2)CH(2)OH exhibited a high affinity for CYP 2C9, as shown by the dissociation constant of their CYP 2C9 complexes, K(s), which was determined by difference visible spectroscopy (K(s) between 0.1 and 0.4 microM) and their constant of CYP 2C9 inhibition (K(i) between 0.3 and 0.6 microM). This indicates that the CYP 2C9-iron(III)-NH(2)R bond previously described to exist in the CYP 2C9-SPA complex does not play a key role in the high affinity of SPA for CYP 2C9. Compounds in which the phenyl group of SPA was replaced with various aryl or alkyl R(2) substituents only exhibited a high affinity for CYP 2C9 if R(2) is a freely rotating and sufficiently electron-rich aryl substituent. Finally, compounds resulting from a N-alkylation of the SPA sulfonamide function (R(3) = CH(3), C(2)H(5), or C(3)H(7)) did not retain the selective inhibitory properties of SPA toward CYP 2C9. However, they are reasonably good inhibitors of CYP 2C8 and CYP 2C18 (IC(50) approximately 20 microM). These data allow one to better understand the structural factors that are important for selective binding in the CYP 2C9 active site. They also provide us with clues towards new selective inhibitors of CYP 2C8 and CYP 2C18.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Sulfaphenazole/chemistry , Sulfaphenazole/metabolism , Binding Sites/physiology , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Microsomes/enzymology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Spectrophotometry , Spectrophotometry, Ultraviolet , Structure-Activity Relationship , Sulfaphenazole/analogs & derivatives , Sulfaphenazole/pharmacology , Transfection , Yeasts/chemistry , Yeasts/metabolism
6.
Drug Metab Dispos ; 29(11): 1366-76, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11602511

ABSTRACT

This article is a report on a symposium sponsored by the American Society for Pharmacology and Experimental Therapeutics and held at the Experimental Biology 01 meeting in Orlando, FL. The presentations addressed the mechanisms of inhibition and regulation of cytochrome P450 and flavin monooxygenase enzymes by nitric oxide. They also highlighted the consequences of these effects on metabolism of drugs and volatile amines as well as on important physiological parameters, such as control of blood pressure, renal ion transport, and steroidogenesis. This is achieved via regulation of P450-dependent prostacyclin, hydroxyeicosatetraenoic acid, and epoxyeicosatrienoic acid formation. Conversely, the mechanisms and relative importance of nitric oxide synthases and P450 enzymes in NO production from endogenous and synthetic substrates were also addressed.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Nitric Oxide/metabolism , Oxygenases/metabolism , Animals , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Hepatitis/enzymology , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , NG-Nitroarginine Methyl Ester/pharmacology , Nitrates/metabolism , Nitrates/pharmacology , Nitric Oxide Synthase/metabolism , Oxygenases/antagonists & inhibitors , Rats , Zona Glomerulosa/drug effects , Zona Glomerulosa/enzymology
7.
J Med Chem ; 44(22): 3622-31, 2001 Oct 25.
Article in English | MEDLINE | ID: mdl-11606127

ABSTRACT

Twenty-three new derivatives of sulfaphenazole (SPA) were synthesized to further explore the topology of the active sites of human liver cytochromes P450 of the 2C subfamily and to find new selective inhibitors of these cytochromes. These compounds are derived from SPA by replacement of the NH(2) and H (of the SO(2)NH function) substituents of SPA with various R(1) and R(2) groups, respectively. Their inhibitory effects were studied on recombinant CYP 2C8, 2C9, 2C18, and 2C19 expressed in yeast. High affinities for CYP 2C9 (IC(50) < 1 microM) were only observed for SPA derivatives having the SO(2)NH function and a relatively small R(1) substituent (R(1) = NH(2), CH(3)). Any increase in the size of R(1) led to a moderate decrease of the affinity, and the N-alkylation of the SO(2)NH function of SPA to a greater decrease of this affinity. The same structural changes led to opposite effects on molecular recognition by CYP 2C8 and 2C18, which generally exhibited similar behaviors. Thus, contrary to CYP 2C9, CYP 2C8 and 2C18 generally prefer neutral compounds with relatively large R(1) and R(2) substituents. CYP 2C19 showed an even lower affinity for anionic compounds than CYP 2C8 and 2C18. However, as CYP 2C8 and 2C18, CYP 2C19 showed a much better affinity for neutral compounds derived from N-alkylation of SPA and for anionic compounds bearing a larger R(1) substituent. One of the new compounds (R(1) = methyl, R(2) = propyl) inhibited all human CYP 2Cs with IC(50) values between 10 and 20 microM, while another one (R(1) = allyl, R(2) = methyl) inhibited all CYP 2Cs except CYP 2C9, and a third one (R(1) = R(2) = methyl) inhibited all CYP 2Cs except CYP 2C8. Only 2 compounds of the 25 tested derivatives were highly selective toward one human CYP 2C; these are SPA and compound 1 (R(1) = CH(3), R(2) = H), which acted as selective CYP 2C9 inhibitors. However, some SPA derivatives selectively inhibited CYP 2C8 and 2C18. Since CYP 2C18 is hardly detectable in human liver, these derivatives could be interesting molecules to selectively inhibit CYP 2C8 in human liver microsomes. Thus, compound 11 (R(1) = NH(2), R(2) = (CH(2))(2)CH(CH(3))(2)) appears to be particularly interesting for that purpose as its IC(50) value for CYP 2C8 is low (3 microM) and 20-fold smaller than those found for CYP 2C9 and 2C19.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/chemical synthesis , Liver/enzymology , Steroid 16-alpha-Hydroxylase , Sulfaphenazole/analogs & derivatives , Sulfaphenazole/chemical synthesis , Sulfonamides/chemical synthesis , Binding Sites , Cytochrome P-450 CYP2C19 , Cytochrome P-450 Enzyme System , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Microsomes/enzymology , Mixed Function Oxygenases/antagonists & inhibitors , Recombinant Proteins/antagonists & inhibitors , Steroid Hydroxylases/antagonists & inhibitors , Structure-Activity Relationship , Sulfaphenazole/chemistry , Sulfaphenazole/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Yeasts/enzymology
9.
Biochemistry ; 40(33): 9909-17, 2001 Aug 21.
Article in English | MEDLINE | ID: mdl-11502185

ABSTRACT

Interaction between microperoxidase-8 (MP8), a water-soluble hemeprotein model, and a wide range of N-aryl and N-alkyl N'-hydroxyguanidines and related compounds has been investigated using UV-visible, EPR, and resonance Raman spectroscopies. All the N-hydroxyguanidines studied bind to the ferric form of MP8 with formation of stable low-spin iron(III) complexes characterized by absorption maxima at 405, 535, and 560 nm. The complex obtained with N-(4-methoxyphenyl) N'-hydroxyguanidine exhibits EPR g-values at 2.55, 2.26, and 1.86. The resonance Raman (RR) spectrum of this complex is also in agreement with an hexacoordinated low-spin iron(III) structure. The dissociation constants (K(s)) of the MP8 complexes with mono- and disubstituted N-hydroxyguanidines vary between 15 and 160 microM at pH 7.4. Amidoximes also form low-spin iron(III) complexes of MP8, although with much larger dissociation constants. Under the same conditions, ketoximes, aldoximes, methoxyguanidines, and guanidines completely fail to form such complexes with MP8. The K(s) values of the MP8-N-hydroxyguanidine complexes decrease as the pH of the solution is increased, and the affinity of the N-hydroxyguanidines toward MP8 increases with the pK(a) of these ligands. Altogether these results show that compounds involving a -C(NHR)=NOH moiety act as good ligands of MP8-Fe(III) with an affinity that depends on the electron-richness of this moiety. The analysis of the EPR spectrum of the MP8-N-hydroxyguanidine complexes according to Taylor's equations shows a strong axial distortion of the iron, typical of those observed for hexacoordinated heme-Fe(III) complexes with at least one pi donor axial ligand (HO(-), RO(-), or RS(-)). These data strongly suggest that N-hydroxyguanidines bind to MP8 iron via their oxygen atom after deprotonation or weakening of their O-H bond. It thus seems that N-hydroxyguanidines could constitute a new class of strong ligands for hemeproteins and iron(III)-porphyrins.


Subject(s)
Electron Spin Resonance Spectroscopy/methods , Guanidines/chemistry , Heme/chemistry , Peroxidases/chemistry , Peroxidases/metabolism , Spectrophotometry/methods , Spectrum Analysis, Raman/methods , Animals , Horses , Hydroxylamines , Iron , Kinetics , Ligands , Models, Chemical , Myocardium/metabolism , Protein Binding , Ultraviolet Rays
10.
J Biochem ; 130(2): 227-33, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11481039

ABSTRACT

There is growing evidence in the literature emphasizing the significance of the post-translational modification of cysteine thiols to sulfenic acids (SOH), which have been found in a number of proteins. Crystallographic and mass spectrometric evidence has shown the presence of this group in an inactive form of the industrially important enzyme nitrile hydratase (NHase). This oxidized cysteine is unique in that it forms part of the coordination sphere of the low-spin iron III at the active site of the enzyme. The presence of this unstable sulfenic group in the active form of NHase is the subject of some controversy. To try to detect this function in NHase, we have studied the inhibitory effect on nitrile hydration of reagents known to react with sulfenic acids. Two NHases were studied, namely, Rhodococcus rhodochrous R312 NHase and Comamonas testosteroni NI1 NHase, and the reagents used were meta-chlorocarbonyldicyano-phenylhydrazone (m-ClCP), 7-chloro-4-nitrobenzo-2-oxa-1,3-diazole (NBD-Cl), and 2-nitro-5-thiocyanato-benzoic acid (NTBA). Following this approach we report three novel inhibitors of NHases. In addition, we report thiocyanate reagents that can be used to monitor NHase activity spectroscopically.


Subject(s)
4-Chloro-7-nitrobenzofurazan/pharmacology , Enzyme Inhibitors/pharmacology , Hydro-Lyases/antagonists & inhibitors , Iron/metabolism , Sulfenic Acids/metabolism , Thiocyanates/pharmacology , 4-Chloro-7-nitrobenzofurazan/metabolism , Catalytic Domain , Enzyme Inhibitors/metabolism , Hydrazones/metabolism , Hydrazones/pharmacology , Hydro-Lyases/chemistry , Hydro-Lyases/metabolism , Molecular Structure , Oxidation-Reduction , Spectrum Analysis , Thiocyanates/metabolism
11.
Eur J Biochem ; 268(13): 3783-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11432746

ABSTRACT

Microperoxidase 8 (MP8) is a heme octapeptide obtained by hydrolytic digestion of horse heart cytochrome c. At pH below 9, the heme iron is axially coordinated to the imidazole side chain of His18 and to a water molecule. Replacement of this weak ligand by H2O2 allows the formation of high-valent iron-oxo species which are responsible for both peroxidase-like and cytochrome P450-like activities of MP8. This paper shows that MP8 is able to catalyze the nitration of phenol by nitrite. The reaction requires H2O2 and is inhibited by ligands having a high affinity for the iron, catalase and radical scavengers. This suggests that the nitrating species could be NO2* radicals formed by the oxidation of nitrite by high-valent iron-oxo species. This new activity of MP8 opens a new access to nitro-aromatic compounds under mild conditions and validates the use of this minienzyme to mimick heme peroxidases, especially in the reactions of NO-derived species with biomolecules under oxidative stress conditions.


Subject(s)
Cytochrome c Group/chemistry , Cytochrome c Group/metabolism , Peroxidases/metabolism , Phenol/chemistry , Animals , Catalysis , Free Radicals , Horses , Hydrogen Peroxide , Hydrogen-Ion Concentration , Kinetics , Models, Chemical , Myocardium/metabolism , Nitrates/metabolism , Nitrogen Dioxide , Sodium Nitrite/chemistry
12.
Biochem Biophys Res Commun ; 283(2): 487-92, 2001 May 04.
Article in English | MEDLINE | ID: mdl-11327727

ABSTRACT

Interaction of rat and human cystathionine-beta-synthase (CBS) with various potential ligands has been studied by visible and EPR spectroscopy in order to explore the coordination chemistry of this atypical hemeprotein. Ferric CBS did not react with any classical hemeprotein ligands, such as various imidazole and pyridine derivatives, N(-)(3) and isonitriles RNC. Ferrous CBS also failed to bind these nitrogenous ligands or nitrosoalkanes. However, it reacts with various isonitriles RNC, leading to complexes characterized by a Soret peak at 433 +/- 2 nm. Binding of isonitriles to ferrous CBS is a relatively slow process; its rate markedly depends on the nature of R. It thus seems that the only exogenous ligands able to bind CBS iron are carbon-centered, very strong heme-Fe(II) ligands such as CNR, CO, and CN(-), presumably after dissociation of the CBS-iron(II)-cysteinate bond. Isonitriles appear as interesting tools for further studies on the topology of CBS active site.


Subject(s)
Cystathionine beta-Synthase/chemistry , Animals , Catalytic Domain , Cystathionine beta-Synthase/metabolism , Electron Spin Resonance Spectroscopy , Heme/chemistry , Humans , In Vitro Techniques , Iron/chemistry , Kinetics , Ligands , Nitriles/chemistry , Rats , Spectrophotometry , Spectrophotometry, Ultraviolet
13.
J Inorg Biochem ; 84(3-4): 207-13, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11374583

ABSTRACT

A new [Co(N2(SO2)2)(CNtBu)2](Et4N) complex 6 was prepared from N,N'-(3-mercapto-3-methyl-butyryl)-o-phenylenediamine and completely characterized. While the starting square planar complex [Co(N2S2)](Et4N) 4 was destroyed by dioxirane, the Co ligated thiolates of the six-coordinate intermediate [Co(N2S2)(CNtBu)2](Et4N) complex 5 was readily oxidized to sulfinates with a stoichiometric amount of this oxidant. The resulting complex 6 crystallizes with an octahedral structure. The SO bonds of the SO2 groups are almost equivalent (approximately 1.483 and approximately 1.453 A). The isonitrile is linearly bonded to the cobalt with a Co-C-N angle of 177.5 degrees and a very short C-N(tBu) distance of 1.13 A, which has a triple bond character. As expected for six-coordinate CoIII complexes, 5 and 6 are diamagnetic in agreement with their 1H and 13C NMR spectra. The SO2 IR bands are located at 1210 cm(-1) (v(as)SO2) and 1070 cm(-1) (v(s)SO2), while the CN vibration of the isonitrile is observed at 2170 cm(-1) in 5 and 2210 cm(-1) in 6. Very recently, it has been reported in the literature that oxidation of the coordinated thiolates was required for activity of both Fe and Co nitrile hydratases. Complex 6, with two oxidized thiolates trans to two deprotonated carboxamido nitrogens, is the first to have an in-plane closely related to that of the Co-NHase active site.


Subject(s)
Cobalt , Sulfinic Acids/chemical synthesis , Catalytic Domain , Hydro-Lyases , Molecular Structure , Organometallic Compounds/chemistry , Oxidation-Reduction , Spectrophotometry, Infrared
14.
Biochemistry ; 40(9): 2689-701, 2001 Mar 06.
Article in English | MEDLINE | ID: mdl-11258880

ABSTRACT

Arginase is a binuclear Mn(2+) metalloenzyme that catalyzes the hydrolysis of L-arginine to L-ornithine and urea. X-ray crystal structures of arginase complexed to substrate analogues N(omega)-hydroxy-L-arginine and N(omega)-hydroxy-nor-L-arginine, as well as the products L-ornithine and urea, complete a set of structural "snapshots" along the reaction coordinate of arginase catalysis when interpreted along with the X-ray crystal structure of the arginase-transition-state analogue complex described in Kim et al. [Kim, N. N., Cox, J. D., Baggio, R. F., Emig, F. A., Mistry, S., Harper, S. L., Speicher, D. W., Morris, Jr., S. M., Ash, D. E., Traish, A. M., and Christianson, D. W. (2001) Biochemistry 40, 2678-2688]. Taken together, these structures render important insight on the structural determinants of tight binding inhibitors. Furthermore, we demonstrate for the first time the structural mechanistic link between arginase and NO synthase through their respective complexes with N(omega)-hydroxy-L-arginine. That N(omega)-hydroxy-L-arginine is a catalytic intermediate for NO synthase and an inhibitor of arginase reflects the reciprocal metabolic relationship between these two critical enzymes of L-arginine catabolism.


Subject(s)
Arginase/chemistry , Arginase/metabolism , Arginine/analogs & derivatives , Amino Acid Substitution/genetics , Animals , Arginase/antagonists & inhibitors , Arginase/genetics , Arginine/chemistry , Arginine/metabolism , Binding, Competitive/genetics , Catalysis , Crystallography, X-Ray , Cysteine/genetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Histidine/genetics , Macromolecular Substances , Models, Molecular , Mutagenesis, Site-Directed , Ornithine/chemistry , Ornithine/metabolism , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structure-Activity Relationship , Substrate Specificity/genetics , Urea/chemistry , Urea/metabolism
15.
Chem Res Toxicol ; 14(2): 202-10, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11258969

ABSTRACT

Oxidations of L-arginine 2, homo-L-arginine 1, their N(omega)-hydroxy derivatives 4 and 3 (NOHA and homo-NOHA, respectively), and four N-hydroxyguanidines, N(omega)-hydroxynor-L-arginine 5 (nor-NOHA), N(omega)-hydroxydinor-L-arginine 6 (dinor-NOHA), N-(4-chlorophenyl)-N'-hydroxyguanidine (8), and N-hydroxyguanidine (7) itself, by either NOS II or (6R)-5,6,7,8-tetrahydro-L-biopterin (BH4)-free NOS II, have been studied in a comparative manner. Recombinant BH4-free NOS II catalyzes the oxidation of all N-hydroxyguanidines by NADPH and O2, with formation of NO2(-) and NO3(-) at rates between 20 and 80 nmol min(-1) (mg of protein)(-1). In the case of compound 8, formation of the corresponding urea and cyanamide was also detected besides that of NO2(-) and NO3(-). These BH4-free NOS II-dependent reactions are inhibited by modulators of electron transfer in NOS such as thiocitrulline (TC) or imidazole (ImH), but not by Arg, and are completely suppressed by superoxide dismutase (SOD). They exhibit characteristics very similar to those previously reported for microsomal cytochrome P450-catalyzed oxidation of N-hydroxyguanidines. Both P450 and BH4-free NOS II reactions appear to be mainly performed by O2(.-) derived from the oxidase function of those heme proteins. In the presence of increasing concentrations of BH4, these nonselective oxidations progressively disappear while a much more selective monooxygenation takes place only with the N-hydroxyguanidines that are recognized well by NOS II, NOHA, homo-NOHA, and 8. These monooxygenations are much more chemoselective (8 being selectively transformed into the corresponding urea and NO) and are inhibited by Arg but not by SOD, as expected for reactions performed by the NOS Fe(II)-O2 species. Altogether, these results provide a further clear illustration of the key role of BH4 in regulating the monooxygenase/oxidase ratio in NOS. They also suggest a possible implication of NOSs in the oxidative metabolism of certain classes of xenobiotics such as N-hydroxyguanidines, not only via their monooxygenase function but also via their oxidase function.


Subject(s)
Antioxidants/metabolism , Arginine/analogs & derivatives , Arginine/metabolism , Biopterins/analogs & derivatives , Biopterins/metabolism , Guanidines/metabolism , Nitric Oxide Synthase/metabolism , Hydroxylamines , NADP/metabolism , Nitric Oxide Synthase Type II , Nitrites/metabolism , Oxidation-Reduction , Substrate Specificity , Superoxide Dismutase/metabolism
16.
Chem Commun (Camb) ; (18): 1718-9, 2001 Sep 21.
Article in English | MEDLINE | ID: mdl-12240281

ABSTRACT

A series of iron porphyrins bearing one to eight beta-nitro substituents were synthesized and evalauted as catalysts for hydrocarbon oxidation with H2O2; iron porphyrins bearing five or six beta-nitro groups were the best catalysts for cyclooctene epoxidation and adamantane or anisole hydroxylation without need of a cocatalyst, and led to very different regioselectivities with either H2O2 or PhIO as oxidants, as shown by an unusual ortho-hydroxylation of alkoxybenzenes highly favored in the H2O2-dependent oxidations.

20.
Biochem Biophys Res Commun ; 278(1): 217-23, 2000 Nov 11.
Article in English | MEDLINE | ID: mdl-11071875

ABSTRACT

Microperoxidase 8 (MP8) is a heme octapeptide, obtained by enzymatic hydrolysis of heart cytochrome c, in which a histidine is axially coordinated to the heme iron, and acts as its fifth ligand. It exhibits two kinds of activities: a peroxidase-like activity and a cytochrome P450-like activity. We here show that MP8 is not only able to oxidize various aliphatic and aromatic hydroxylamines with the formation of MP8-Fe(II)-nitrosoalkane or -arene complexes absorbing around 414 nm, but also that these complexes can be obtained by reduction of nitroalkanes. This is the first example of fully characterized iron(II)-metabolite complexes of MP8. Such complexes constitute good models for those obtained upon oxidation of amphetamine or macrolids by cytochromes P450. In addition, this is a new catalytic activity of MP8, which validates the use of this mini-enzyme as a convenient model for hemoproteins of interest in toxicology and pharmacology such as cytochromes P450 and peroxidases.


Subject(s)
Alkanes/metabolism , Amines/metabolism , Iron/metabolism , Peroxidases/metabolism , Propane/analogs & derivatives , Alkanes/chemistry , Amines/chemistry , Animals , Catalysis , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , Cytochrome c Group/metabolism , Heme/chemistry , Heme/metabolism , Horses , Hydroxylamines/chemistry , Iron/chemistry , Ligands , Models, Chemical , Myocardium/chemistry , Nitroparaffins/pharmacology , Oxygen/metabolism , Peroxidases/chemistry , Propane/pharmacology , Spectrophotometry, Ultraviolet , Time Factors , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...