Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Biochem Soc Trans ; 31(Pt 3): 506-9, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12773145

ABSTRACT

Galactose oxidase (GO; EC 1.1.3.9) is a monomeric 68 kDa enzyme that contains a single copper and an amino acid-derived cofactor. The mechanism of this radical enzyme has been widely studied by structural, spectroscopic, kinetic and mutational approaches and there is a reasonable understanding of the catalytic mechanism and activation by oxidation to generate the radical cofactor that resides on Tyr-272, one of the copper ligands. Biogenesis of this cofactor involves the post-translational, autocatalytic formation of a thioether cross-link between the active-site residues Cys-228 and Tyr-272. This process is closely linked to a peptide bond cleavage event that releases the N-terminal 17-amino-acid pro-peptide. We have shown using pro-enzyme purified in copper-free conditions that mature oxidized GO can be formed by an autocatalytic process upon addition of copper and oxygen. Structural comparison of pro-GO (GO with the prosequence present) with mature GO reveals overall structural similarity, but with some regions showing significant local differences in main chain position and some active-site-residue side chains differing significantly from their mature enzyme positions. These structural effects of the pro-peptide suggest that it may act as an intramolecular chaperone to provide an open active-site structure conducive to copper binding and chemistry associated with cofactor formation. Various models can be proposed to account for the formation of the thioether bond and oxidation to the radical state; however, the mechanism of prosequence cleavage remains unclear.


Subject(s)
Galactose Oxidase/metabolism , Binding Sites , Coenzymes/metabolism , Copper/analysis , Enzyme Precursors/metabolism , Fusarium/enzymology , Galactose Oxidase/chemistry , Galactose Oxidase/genetics , Oxidation-Reduction , Protein Processing, Post-Translational
2.
J Am Chem Soc ; 124(41): 12135-43, 2002 Oct 16.
Article in English | MEDLINE | ID: mdl-12371853

ABSTRACT

We previously reported that 3-pyrroline and 3-phenyl-3-pyrroline effect a time-dependent inactivation of the copper-containing quinone-dependent amine oxidase from bovine plasma (BPAO) (Lee et al. J. Am. Chem. Soc. 1996, 118, 7241-7242). Quinone cofactor model studies suggested a mechanism involving stoichiometric turnover to a stable pyrrolylated cofactor. Full details of the model studies are now reported along with data on the inhibition of BPAO by a family of 3-aryl-3-pyrrolines (aryl = substituted phenyl, 1-naphthyl, 2-naphthyl), with the 4-methoxy-3-nitrophenyl analogue being the most potent. At the same time, the parent 3-phenyl analogue is a pure substrate for the flavin-dependent mitochondrial monoamine oxidase B from bovine liver. Spectroscopic studies (including resonance Raman) on BPAO inactivated by the 4-methoxy-3-nitrophenyl analogue are consistent with covalent derivatization of the 2,4,5-trihydroxyphenylalanine quinone (TPQ) cofactor. The distinction of a class of compounds acting as an inactivator of one amine oxidase family and a pure substrate of another amine oxidase family represents a unique lead to the development of selective inhibitors of the mammalian copper-containing amine oxidases.


Subject(s)
Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Pyrroles/pharmacology , Amine Oxidase (Copper-Containing)/metabolism , Animals , Cattle , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Kinetics , Mitochondria, Liver/enzymology , Monoamine Oxidase/chemistry , Pyrroles/chemistry , Pyrroles/metabolism , Pyrrolidines/chemistry , Pyrrolidines/metabolism , Spectrophotometry , Spectrum Analysis, Raman , Substrate Specificity
3.
Proc Natl Acad Sci U S A ; 98(23): 12932-7, 2001 Nov 06.
Article in English | MEDLINE | ID: mdl-11698678

ABSTRACT

Galactose oxidase (EC ) is a monomeric enzyme that contains a single copper ion and catalyses the stereospecific oxidation of primary alcohols to their corresponding aldehydes. The protein contains an unusual covalent thioether bond between a tyrosine, which acts as a radical center during the two-electron reaction, and a cysteine. The enzyme is produced in a precursor form lacking the thioether bond and also possessing an additional 17-aa pro-sequence at the N terminus. Previous work has shown that the aerobic addition of Cu(2+) to the precursor is sufficient to generate fully processed mature enzyme. The structure of the precursor protein has been determined to 1.4 A, revealing the location of the pro-sequence and identifying structural differences between the precursor and the mature protein. Structural alignment of the precursor and mature forms of galactose oxidase shows that five regions of main chain and some key residues of the active site differ significantly between the two forms. The precursor structure provides a starting point for modeling the chemistry of thioether bond formation and pro-sequence cleavage.


Subject(s)
Enzyme Precursors/chemistry , Galactose Oxidase/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Molecular Sequence Data , Protein Conformation , Protein Processing, Post-Translational
5.
Protein Expr Purif ; 22(3): 455-61, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11483008

ABSTRACT

The copper-containing amine oxidase from Arthrobacter globiformis has been expressed and purified as a fusion protein with a C-terminal Strep-tag II peptide. This tag facilitates the rapid purification of the enzyme on a large scale using the StrepTactin POROS medium. For example, we have demonstrated that 50 mg of protein can be obtained in 2 days from 2 L of Escherichia coli. The purified fusion protein displays turnover and spectroscopic properties that are essentially identical to those of the wild-type enzyme. Given the location of the C-terminus in four amine oxidase crystal structures, this strategy should be quite general for the rapid purification of amine oxidases from multiple sources.


Subject(s)
Amine Oxidase (Copper-Containing)/genetics , Arthrobacter/enzymology , Oligopeptides , Amine Oxidase (Copper-Containing)/isolation & purification , Amine Oxidase (Copper-Containing)/metabolism , Circular Dichroism , Cloning, Molecular , Escherichia coli/genetics , Kinetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism
6.
J Am Chem Soc ; 123(4): 576-87, 2001 Jan 31.
Article in English | MEDLINE | ID: mdl-11456570

ABSTRACT

Nitrous oxide reductase (N(2)OR) from Pseudomonas stutzeri, a dimeric enzyme with a canonical metal ion content of at least six Cu ions per subunit, contains two types of multinuclear copper sites: Cu(A) and Cu(Z). An electron-transfer role for the dinuclear Cu(A) site is indicated based on its similarity to the Cu(A) site in cytochrome c oxidase (CcO), a dicysteinate-bridged, mixed-valence cluster. The Cu(Z) site is the catalytic site, which had long been thought to have novel spectroscopic properties. However, the low-energy electronic transitions and resonance Raman features attributable to Cu(Z) have been difficult to reconcile with a lack of conserved cysteine residues in standard alignments of N(2)OR sequences, other than those associated with the Cu(A) site. Recent evidence indicates that nitrous oxide reductase contains acid-labile sulfide and that this sulfide is a constituent of the Cu(Z) site (Rasmussen, T.; Berks, B. C.; Sanders-Loehr, J.; Dooley, D. M.; Zumft, W. G.; Thomson, A. J. Biochemistry 2000, 39, 12753-12756). We have used resonance Raman (RR) spectroscopy to selectively probe the Cu(A) and Cu(Z) sites of N(2)OR in three oxidation states (oxidized, semireduced, and reduced) as well as Cu(A)-only and Cu(Z)-only variants. The Cu(A) (mixed-valence, also designated as A(mv)) RR spectrum exhibits 10 vibrational modes between 220 and 410 cm(-1), with >1-cm(-1) (34)S isotope shifts that sum to -16.6 cm(-1). Many of these modes are also sensitive to (65)Cu and (15)N(His) and, thus, can be assigned to coupling of the Cu-S stretch, nu(Cu-S), with cysteine and histidine vibrations of the Cu(2)Cys(2)His(2) core. The RR spectrum of the Cu(Z) site (Z(ox)) reveals a novel Cu-sulfur chromophore with four S isotope-sensitive modes at 293, 347, 352, and 408 cm(-1), with a total (34)S shift of -19.9 cm(-)(1). The magnitude of the S isotope shifts and wide spread of perturbed frequencies are similar to those observed in Cu(A) and therefore suggest a sulfur-bridged cluster in Z(ox). The Z(ox) site has its nu(Cu-S)-containing modes at higher energy and exhibits less mixing with ligand deformations, compared to Cu(A). Reduction by dithionite produces a mixed-valence Cu(Z) site (Z(mv)) with six S isotope-sensitive RR modes between 282 and 382 cm(-1) and a total (34)S-shift of -16.9 cm(-1). The observation of a nearly identical RR spectrum in the C622D variant of N(2)OR, which lacks one of the conserved Cu(A) Cys residues, establishes that Cu-S vibrations observed in this variant arise from the Z(mv) site. Furthermore, none of the features assigned to Cu(Z) are detected in a second variant that contains only Cu(A). Therefore the resonance Raman spectra reported here provide compelling evidence for a unique Cu-S cluster in the catalytic site of nitrous oxide reductase.


Subject(s)
Oxidoreductases/chemistry , Bacterial Proteins/chemistry , Catalytic Domain , Copper/chemistry , Histidine/chemistry , Ligands , Models, Molecular , Oxidation-Reduction , Pseudomonas/enzymology , Soil Microbiology , Spectrum Analysis, Raman , Sulfur/chemistry
7.
J Biol Inorg Chem ; 6(2): 189-95, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11293413

ABSTRACT

NosL, one of the accessory proteins of the nos (nitrous oxide reductase) gene cluster, has been heterologously expressed, purified, and characterized. NosL is a monomeric protein of 18,540 MW that specifically and stoichiometrically binds Cu(I). The copper ion in NosL is ligated by a Cys residue, and one Met and one His are thought to serve as the other ligands. While it is possible to oxidize Cu(I)-NosL with ferricyanide, the Cu(II) ion thus formed appears to dissociate from the protein. The function of Cu(I)NosL is not yet known, but the data indicate that NosL does not act as an electron transfer partner to nitrous oxide reductase. NosL is encoded on the same transcript as three other gene products (NosD, NosF, and NosY). These have been shown to be required for assembly of the active site in nitrous oxide reductase, which is thought to be a copper cluster. Accordingly, it is possible that NosL is a copper chaperone involved in metallocenter assembly.


Subject(s)
Alcaligenes/chemistry , Bacterial Outer Membrane Proteins/chemistry , Copper/chemistry , Lipoproteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Binding Sites , Escherichia coli , Gene Expression Regulation , Lipoproteins/genetics , Lipoproteins/metabolism , Metalloproteins/chemistry , Metalloproteins/genetics , Metalloproteins/metabolism , Molecular Chaperones , Molecular Structure , Oxidoreductases , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrometry, X-Ray Emission
8.
J Inorg Biochem ; 83(2-3): 193-204, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11237259

ABSTRACT

Lysyl oxidase from Pichia pastoris has been successfully overexpressed. EPR and resonance Raman experiments have shown that copper and TPQ are present, respectively. Lysyl oxidase from P. pastoris has a similar substrate specificity to the mammalian enzyme (both have been shown to oxidize peptidyl lysine residues) and is 30% identical to the human kidney diamine oxidase (the highest of any non-mammalian source). This enzyme also has a relatively broad substrate specificity compared to other amine oxidases. Molecular modeling data suggest that the substrate channel in lysyl oxidase from P. pastoris permits greater active site access than observed in structurally-characterized amine oxidases. This larger channel may account for the diversity of substrates that are turned over by this enzyme.


Subject(s)
Copper/chemistry , Pichia/enzymology , Protein-Lysine 6-Oxidase/chemistry , Amino Acid Sequence , Animals , Binding Sites , Circular Dichroism , Cloning, Molecular , Crystallography, X-Ray , Electron Spin Resonance Spectroscopy , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein-Lysine 6-Oxidase/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Spectrum Analysis, Raman
9.
Biochemistry ; 40(3): 822-9, 2001 Jan 23.
Article in English | MEDLINE | ID: mdl-11170400

ABSTRACT

Incubation of bovine plasma amine oxidase (BPAO) with benzylamine and various p-substituted analogues results in a time-dependent inactivation that is attributable to buildup of the H(2)O(2)-turnover product on the basis of protection afforded by coincubation with catalase. The mechanism of inactivation is distinct from that effected by H(2)O(2) itself, which requires higher concentrations. Solution studies using models for the 2,4,5-trihydroxyphenylalanine quinone (TPQ) cofactor reveal a loss of catalytic activity arising from oxidation of the dihydrobenzoxazole tautomer of the product Schiff base, that competes with hydrolytic release of benzaldehyde product. The resulting stable benzoxazole exhibits a characteristic absorption depending on the nature of the benzylamine p-substituent. For benzylamine itself, the model benzoxazole absorbs at 313 nm, in an area of strong absorption by the enzyme, whereas for 4-nitrobenzylamine, the absorption of the model benzoxazole is sufficiently red-shifted (at 365 nm) to be discerned above the background enzyme absorption. Inactivation of BPAO by 4-nitrobenzylamine is accompanied by loss of the resting TPQ anion absorption at 480 nm concomitant with generation of a new absorption near 360 nm. Resonance Raman spectra of the inactivated enzyme show a close correspondence with those for the model 4-nitrobenzylamine-derived benzoxazole. Substrate-dependent inactivation is also observed for the other two mammalian enzymes examined, equine plasma amine oxidase and human kidney amine oxidase. Catalase provides complete protection in these instances as well. Benzoxazole formation may constitute a common mechanism of inactivation of quinone-dependent amine oxidases by normal substrates in vitro if the product H(2)O(2) is permitted to accumulate. More importantly, the results suggest that the benzoxazole inactivation pathway may be important physiologically and may have influenced the distribution of amine oxidases and catalase in cells.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Benzoquinones/chemistry , Benzoxazoles/chemistry , Benzylamines/chemistry , Enzyme Inhibitors/chemistry , Hydrogen Peroxide/chemistry , Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Amine Oxidase (Copper-Containing)/blood , Amine Oxidase (Copper-Containing)/chemistry , Animals , Benzoquinones/metabolism , Benzoxazoles/metabolism , Benzylamines/metabolism , Benzylamines/pharmacology , Catalysis , Cattle , Enzyme Activation , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Hydrogen Peroxide/metabolism , Spectrophotometry , Spectrum Analysis, Raman , Substrate Specificity
10.
Biochemistry ; 39(42): 12753-6, 2000 Oct 24.
Article in English | MEDLINE | ID: mdl-11041839

ABSTRACT

The crystal structure of nitrous oxide reductase, the enzyme catalyzing the final step of bacterial denitrification in which nitrous oxide is reduced to dinitrogen, exhibits a novel catalytic site, called Cu(Z). This comprises a cluster of four copper ions bound by seven histidines and three other ligands modeled in the X-ray structure as OH(-) or H(2)O. However, elemental analyses and resonance Raman spectroscopy of isotopically labeled enzyme conclusively demonstrate that Cu(Z) has one acid-labile sulfur ligand. Thus, nitrous oxide reductase contains the first reported biological copper-sulfide cluster.


Subject(s)
Catalytic Domain , Copper/chemistry , Oxidoreductases/chemistry , Sulfides/chemistry , Acids , Circular Dichroism , Crystallization , Dimerization , Electron Spin Resonance Spectroscopy , Ligands , Magnetics , Pseudomonas/enzymology , Spectrum Analysis , Sulfur/chemistry , X-Rays
11.
J Biol Inorg Chem ; 4(1): 1-11, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10499097

ABSTRACT

The structure of a new biological redox cofactor-topaquinone (TPQ), the quinone of 2,4,5-trihydroxyphenylalanine-was elucidated in 1990. TPQ is the cofactor in most copper-containing amine oxidases. It is produced by post-translational modification of a strictly conserved active-site tyrosine residue. Recent work has established that TPQ biogenesis proceeds via a novel self-processing pathway requiring only the protein, copper, and molecular oxygen. The oxidation of tyrosine to TPQ by dioxygen is a six-electron process, which has intriguing mechanistic implications because copper is a one-electron redox agent, and dioxygen can function as either a two-electron or four-electron oxidant. This review adopts an historical perspective in discussing the structure and reactivity of TPQ in amine oxidases, and then assesses what is currently understood about the mechanism of the oxidation of tyrosine to produce TPQ. Aspects of the structures and chemistry of related cofactors, such as the Tyr-Cys radical in galactose oxidase and the lysine tyrosylquinone of lysyl oxidase, are also discussed.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Copper/metabolism , Dihydroxyphenylalanine/analogs & derivatives , Amine Oxidase (Copper-Containing)/chemistry , Binding Sites , Dihydroxyphenylalanine/chemistry , Dihydroxyphenylalanine/metabolism , Galactose Oxidase/metabolism , Lysine/analogs & derivatives , Lysine/chemistry , Lysine/metabolism , Mechanics , Models, Molecular , Oxidation-Reduction , Quinones/chemistry , Quinones/metabolism , Tyrosine/chemistry , Tyrosine/metabolism
12.
Biochemistry ; 38(34): 11164-71, 1999 Aug 24.
Article in English | MEDLINE | ID: mdl-10460173

ABSTRACT

1H NMR spectra of the CuA center of N2OR from Pseudomonas stutzeri, and a mutant enzyme that contains only CuA, were recorded in both H2O- and D2O-buffered solution at pH 7.5. Several sharp, well-resolved hyperfine-shifted 1H NMR signals were observed in the 60 to -10 ppm chemical shift range. Comparison of the native and mutant N2OR spectra recorded in H2O-buffered solutions indicated that several additional signals are present in the native protein spectrum. These signals are attributed to a dinuclear copperII center. At least two of the observed hyperfine-shifted signals associated with the dinuclear center, those at 23.0 and 13.2 ppm, are lost upon replacement of H2O buffer with D2O buffer. These data indicate that at least two histidine residues are ligands of a dinuclear CuII center. Comparison of the mutant N2OR 1H NMR spectra recorded in H2O and D2O indicates that three signals, c (27.5 ppm), e (23.6 ppm), and i (12.4 ppm), are solvent exchangeable. The two most strongly downfield-shifted signals (c and e) are assigned to the two N epsilon 2H (N-H) protons of the coordinated histidine residues, while the remaining exchangeable signal is assigned to a backbone N-H proton in close proximity to the CuA cluster. Signal e was found to decrease in intensity as the temperature was increased, indicating that proton e resides on a more solvent-exposed histidine residue. One-dimensional nOe studies at pH 7.5 allowed the histidine ring protons to be definitively assigned, while the remaining signals were assigned by comparison to previously reported spectra from CuA centers. The temperature dependence of the observed hyperfine-shifted 1H NMR signals of mutant N2OR were recorded over the temperature range of 276-315 K. Both Curie and anti-Curie temperature dependencies are observed for sets of hyperfine-shifted protons. Signals a and h (cysteine protons) follow anti-Curie behavior (contact shift increases with increasing temperatures), while signals b-g, i, and j (histidine protons) follow Curie behavior (contact shift decreases with increasing temperatures). Fits of the temperature dependence of the observed hyperfine-shifted signals provided the energy separation (Delta EL) between the ground (2B3u) and excited (2B2u) states. The temperature data obtained for all of the observed hyperfine-shifted histidine ligand protons provided a Delta EL value of 62 +/- 35 cm-1. The temperature dependence of the observed cysteine C beta H and C alpha H protons (a and h) were fit in a separate experiment providing a Delta EL value of 585 +/- 125 cm-1. The differences between the Delta EL values determined by 1H NMR spectroscopy and those determined by EPR or MCD likely arise from coupling between relatively low-frequency vibrational states and the ground and excited electronic states.


Subject(s)
Copper/chemistry , Oxidoreductases/chemistry , Pseudomonas/enzymology , Copper/metabolism , Electron Transport , Hydrogen-Ion Concentration , Models, Molecular , Mutation , Nuclear Magnetic Resonance, Biomolecular , Oxidoreductases/genetics , Oxidoreductases/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Structure, Tertiary , Protons , Pseudomonas/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Temperature
13.
Curr Opin Chem Biol ; 3(2): 138-44, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10226045

ABSTRACT

Major advances have been made during 1997 and 1998 toward understanding the structure/function relationships of the active sites in copper-containing oxidases. Central to this progress has been the elucidation of crystal structures for many of these enzymes. For example, studies of the mechanisms of biogenesis and/or catalysis of amine oxidase and galactose oxidase have been both stimulated and directed by the availability of structures for these proteins. Similarly, it is anticipated that the recently published crystal structures of peptidylglycine alpha-hydroxylating monooxygenase and laccase will contribute greatly toward understanding the roles of copper in these two proteins.


Subject(s)
Copper/chemistry , Oxidoreductases/metabolism , Binding Sites , Models, Molecular , Oxidoreductases/chemistry , Protein Conformation
14.
Biochemistry ; 38(10): 2892-8, 1999 Mar 09.
Article in English | MEDLINE | ID: mdl-10074341

ABSTRACT

The stoichiometry of the topa quinone biogenesis reaction in phenylethylamine oxidase from Arthrobacter globiformis (AGAO) has been determined. We have shown that the 6e- oxidation of tyrosine to topa quinone (TPQ) consumes 2 mol of O2 and produces 1 mol of H2O2/mol of TPQ formed. The rate of H2O2 production is first-order (kobs = 1.0 +/- 0.2 min-1), a rate only slightly lower than the rate of TPQ formation directly determined previously (kobs = 1.5 +/- 0.2 min-1). This gives the following net reaction stoichiometry for TPQ biogenesis: E-Tyr + 2O2 --> E-TPQ + H2O2. This stoichiometry is in agreement with recently proposed mechanisms for TPQ biogenesis, and rules out several possible alternatives.


Subject(s)
Amine Oxidase (Copper-Containing)/chemistry , Amine Oxidase (Copper-Containing)/metabolism , Dihydroxyphenylalanine/analogs & derivatives , Arthrobacter/enzymology , Dihydroxyphenylalanine/chemistry , Dihydroxyphenylalanine/metabolism , Dimerization , Hydrogen Peroxide/metabolism , Oxidation-Reduction , Oxygen/metabolism , Oxygen Consumption , Spectrophotometry, Ultraviolet
15.
J Inorg Biochem ; 70(3-4): 155-69, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9720302

ABSTRACT

The nitrous oxide (N2O) reductase (nos) gene cluster from Achromobacter cycloclastes has been cloned and sequenced. Seven protein coding regions corresponding to nosR, nosZ (structural N2O reductase gene), nosD, nosF, nosY, nosL, and nosX are detected, indicating a genetic organization similar to that of Rhizobium meliloti. To aid homology studies, nosR from R. meliloti has also been sequenced. Comparison of the deduced amino acid sequences with corresponding sequences from other organisms has also allowed structural and functional inferences to be made. The heterologous expression of NosD, NosZ (N2O reductase), and NosL is also reported. A model of the CuA site in N2O reductase, based on the crystal structure of this site in bovine heart cytochrome c oxidase, is presented. The model suggests that a His residue of the CuA domain may be a ligand to the catalytic CuZ site. In addition, the origin of the spectroscopically-observed Cys coordination to CuZ is discussed in terms of the sequence alignment of seven N2O reductases.


Subject(s)
Alcaligenes/enzymology , Alcaligenes/genetics , Multigene Family , Oxidoreductases/genetics , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Cattle , Cloning, Molecular , Electron Transport Complex IV/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Periplasm/genetics , Periplasm/metabolism , Protein Conformation , RNA, Ribosomal, 16S , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Analysis , Sequence Homology, Amino Acid , Sinorhizobium meliloti/enzymology , Sinorhizobium meliloti/genetics
16.
Biochemistry ; 36(8): 1953-9, 1997 Feb 25.
Article in English | MEDLINE | ID: mdl-9047291

ABSTRACT

An alternative purification for apophenylethylamine oxidase from Arthrobacter globiformis has been developed, which avoids the use of possible contaminants that may interfere with the topa quinone (TPQ) self-processing reaction. The binding of Cu(II) and the kinetics of TPQ formation in these enzyme preparations have been reinvestigated. Our results show that Cu(II) is not significantly reduced when added to the apoprotein under anaerobic conditions. The Cu(II) EPR and circular dichroism spectra of the initially formed complex are different from the spectra of the mature Cu(II)/TPQ-containing protein, indicating that the active site structure must be altered during TPQ formation. The kinetics we observe are cleanly first-order in protein [measured subsequent to Cu(II) binding] when dioxygen is present in pseudo-first-order excess (k(obs) = 1.5 min(-1)). We found no rate dependence on copper, so long as one copper per subunit was present. This indicates that tyrosine oxidation to give TPQ depends only on the copper that is bound in the active site. These results differ from those originally reported; an alternative mechanism, which involves attack of an activated copper-oxygen species on a tyrosine radical intermediate, is proposed for TPQ formation.


Subject(s)
Amine Oxidase (Copper-Containing)/isolation & purification , Arthrobacter/enzymology , Dihydroxyphenylalanine/analogs & derivatives , Amine Oxidase (Copper-Containing)/metabolism , Copper/metabolism , Dihydroxyphenylalanine/isolation & purification , Dihydroxyphenylalanine/metabolism , Kinetics
17.
Biochemistry ; 36(51): 16116-33, 1997 Dec 23.
Article in English | MEDLINE | ID: mdl-9405045

ABSTRACT

The crystal structures of the copper enzyme phenylethylamine oxidase from the Gram-positive bacterium Arthrobacter globiformis (AGAO) have been determined and refined for three forms of the enzyme: the holoenzyme in its active form (at 2.2 A resolution), the holoenzyme in an inactive form (at 2.8 A resolution), and the apoenzyme (at 2.2 A resolution). The holoenzyme has a topaquinone (TPQ) cofactor formed from the apoenzyme by the post-translational modification of a tyrosine residue in the presence of Cu2+. Significant differences between the three forms of AGAO are limited to the active site. The polypeptide fold is closely similar to those of the amine oxidases from Escherichia coli [Parsons, M. R., et al. (1995) Structure 3, 1171-1184] and pea seedlings [Kumar, V., et al. (1996) Structure 4, 943-955]. In the active form of holo-AGAO, the active-site Cu atom is coordinated by three His residues and two water molecules in an approximately square-pyramidal arrangement. In the inactive form, the Cu atom is coordinated by the same three His residues and by the phenolic oxygen of the TPQ, the geometry being quasi-trigonal-pyramidal. There is evidence of disorder in the crystals of both forms of holo-AGAO. As a result, only the position of the aromatic group of the TPQ cofactor, but not its orientation about the Cbeta-Cgamma bond, is determined unequivocally. In apo-AGAO, electron density consistent with an unmodified Tyr occurs at a position close to that of the TPQ in the inactive holo-AGAO. This observation has implications for the biogenesis of TPQ. Two features which have not been described previously in amine oxidase structures are a channel from the molecular surface to the active site and a solvent-filled cavity at the major interface between the two subunits of the dimer.


Subject(s)
Amine Oxidase (Copper-Containing)/chemistry , Arthrobacter/enzymology , Dihydroxyphenylalanine/analogs & derivatives , Protein Conformation , Amine Oxidase (Copper-Containing)/metabolism , Amino Acid Sequence , Apoenzymes/chemistry , Binding Sites , Copper/chemistry , Copper/metabolism , Crystallography, X-Ray , Dihydroxyphenylalanine/biosynthesis , Dimerization , Hydrogen Bonding , Metalloproteins/chemistry , Molecular Sequence Data , Protein Processing, Post-Translational/physiology , Sequence Alignment , Substrate Specificity , Tyrosine/chemistry , Tyrosine/metabolism
18.
FEBS Lett ; 398(2-3): 231-4, 1996 Dec 02.
Article in English | MEDLINE | ID: mdl-8977113

ABSTRACT

A copper amine oxidase from Pichia pastoris is the only known non-mammalian lysyl oxidase [Tur, S.S. and Lerch, K. (1988) FEBS Lett. 238, 74-76]. Recently, the cofactor in mammalian lysyl oxidase has been identified as a novel lysine tyrosylquinone moiety [Wang, S.X., Mure, M., Medzihradszky, K.F., Burlingame, A.L., Brown, D.E., Dooley, D.M., Smith, A.J., Kagan, H.M. and Klinman, J.P. (1996) Science 273, 1078-1084]. In order to identify the cofactor in P. pastoris lysyl oxidase, we have isolated the phenylhydrazone-derivative of the active-site peptide. This peptide has the active-site sequence conserved among topa quinone containing amine oxidases. The resonance Raman spectra of the phenylhydrazone derivatives of the enzyme, active-site peptide, and a topa quinone model compound are essentially identical. Collectively, these results establish that P. pastoris lysyl oxidase is a topa quinone enzyme.


Subject(s)
Dihydroxyphenylalanine/analogs & derivatives , Pichia/enzymology , Protein-Lysine 6-Oxidase/chemistry , Amino Acid Sequence , Binding Sites , Chromatography, High Pressure Liquid , Dihydroxyphenylalanine/analysis , Hydrazones , Protein-Lysine 6-Oxidase/isolation & purification , Spectrum Analysis, Raman
19.
Structure ; 4(8): 943-55, 1996 Aug 15.
Article in English | MEDLINE | ID: mdl-8805580

ABSTRACT

BACKGROUND: Copper-containing amine oxidases catalyze the oxidative deamination of primary amines to aldehydes, in a reaction that requires free radicals. These enzymes are important in many biological processes, including cell differentiation and growth, would healing, detoxification and signalling. The catalytic reaction requires a redox cofactor, topa quinone (TPQ), which is derived by post-translational modification of an invariant tyrosine residue. Both the biogenesis of the TPQ cofactor and the reaction catalyzed by the enzyme require the presence of a copper atom at the active site. The crystal structure of a prokaryotic copper amine oxidase from E. coli (ECAO) has recently been reported. RESULTS: The first structure of a eukaryotic (pea seedling) amine oxidase (PSAO) has been solved and refined at 2.2 A resolution. The crystallographic phases were derived from a single phosphotungstic acid derivative. The positions of the tungsten atoms in the W12 clusters were obtained by molecular replacement using E. coli amine oxidase as a search model. The methodology avoided bias from the search model, and provides an essentially independent view of a eukaryotic amine oxidase. The PSAO molecule is a homodimer; each subunit has three domains. The active site of each subunit lies near an edge of the beta-sandwich of the largest domain, but is not accessible from the solvent. The essential active-site copper atom is coordinated by three histidine side chains and two water molecules in an approximately square-pyramidal arrangement. All the atoms of the TPQ cofactor are unambiguously defined, the shortest distance to the copper atom being approximately 6 A. CONCLUSIONS: There is considerable structural homology between PSAO and ECAO. A combination of evidence from both structures indicates that the TPQ side chain is sufficiently flexible to permit the aromatic grouf to rotate about the Cbeta-Cgamma bond, and to move between bonding and non-bonding positions with respect to the Cu atom. Conformational flexibility is also required at the surface of the molecule to allow the substrates access to the active site, which is inaccessible to solvent, as expected for an enzyme that uses radical chemistry.


Subject(s)
Amine Oxidase (Copper-Containing) , Oxidoreductases Acting on CH-NH Group Donors/chemistry , Pisum sativum/enzymology , Plant Proteins/chemistry , Protein Conformation , Amino Acid Sequence , Binding Sites , Copper/chemistry , Crystallography, X-Ray , Cystine/chemistry , Dihydroxyphenylalanine/analogs & derivatives , Dihydroxyphenylalanine/chemistry , Dimerization , Glycosylation , Models, Molecular , Molecular Sequence Data , Protein Processing, Post-Translational , Seeds/enzymology , Sequence Alignment , Sequence Homology, Amino Acid
20.
Science ; 273(5278): 1078-84, 1996 Aug 23.
Article in English | MEDLINE | ID: mdl-8688089

ABSTRACT

A previously unknown redox cofactor has been identified in the active site of lysyl oxidase from the bovine aorta. Edman sequencing, mass spectrometry, ultraviolet-visible spectra, and resonance Raman studies showed that this cofactor is a quinone. Its structure is derived from the crosslinking of the epsilon-amino group of a peptidyl lysine with the modified side chain of a tyrosyl residue, and it has been designated lysine tyrosylquinone. This quinone appears to be the only example of a mammalian cofactor formed from the crosslinking of two amino acid side chains. This discovery expands the range of known quino-cofactor structures and has implications for the mechanism of their biogenesis.


Subject(s)
Lysine/analogs & derivatives , Protein-Lysine 6-Oxidase/chemistry , Quinones/chemistry , Amino Acid Sequence , Animals , Aorta/enzymology , Binding Sites , Cattle , Chromatography, High Pressure Liquid , Lysine/chemistry , Lysine/metabolism , Mass Spectrometry , Molecular Sequence Data , Molecular Weight , Mutagenesis, Site-Directed , Oxidation-Reduction , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/isolation & purification , Protein-Lysine 6-Oxidase/metabolism , Quinones/metabolism , Spectrophotometry, Ultraviolet , Spectrum Analysis, Raman
SELECTION OF CITATIONS
SEARCH DETAIL
...