Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
FEBS J ; 279(24): 4501-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23083473

ABSTRACT

Indoleamine 2,3-dioxygenase and tryptophan 2,3-dioxygenase catalyze the O(2) -dependent oxidation of l-tryptophan to N-formylkynurenine. Both are heme-containing enzymes, with a proximal histidine ligand, as found in the globins and peroxidases. From the structural information available so far, the distal heme pockets of these enzymes can contain a histidine residue (in tryptophan 2,3-dioxygenases), an arginine residue and numerous hydrophobic residues that line the pocket. We have examined the functional role of each of these residues in both human indoleamine 2,3-dioxygenase and human tryptophan 2,3-dioxygenase. We found that the distal histidine does not play an essential catalytic role, although substrate binding can be affected by removing the distal arginine and reducing the hydrophobic nature of the binding pocket. We collate the information obtained in the present study with that reported in the available literature to draw comparisons across the family and to provide a more coherent picture of how the heme pocket is optimized for tryptophan binding.


Subject(s)
Hemeproteins/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Tryptophan Oxygenase/metabolism , Tryptophan/metabolism , Catalytic Domain , Oxidation-Reduction , Protein Binding , Substrate Specificity
2.
J Am Chem Soc ; 134(6): 3034-41, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22299628

ABSTRACT

Indoleamine 2,3-dioxygenase catalyzes the O(2)-dependent oxidation of L-tryptophan (L-Trp) to N-formylkynurenine (NFK) as part of the kynurenine pathway. Inhibition of enzyme activity at high L-Trp concentrations was first noted more than 30 years ago, but the mechanism of inhibition has not been established. Using a combination of kinetic and reduction potential measurements, we present evidence showing that inhibition of enzyme activity in human indoleamine 2,3-dioxygenase (hIDO) and a number of site-directed variants during turnover with L-tryptophan (L-Trp) can be accounted for by the sequential, ordered binding of O(2) and L-Trp. Analysis of the data shows that at low concentrations of L-Trp, O(2) binds first followed by the binding of L-Trp; at higher concentrations of L-Trp, the order of binding is reversed. In addition, we show that the heme reduction potential (E(m)(0)) has a regulatory role in controlling the overall rate of catalysis (and hence the extent of inhibition) because there is a quantifiable correlation between E(m)(0) (that increases in the presence of L-Trp) and the rate constant for O(2) binding. This means that the initial formation of ferric superoxide (Fe(3+)-O(2)(•-)) from Fe(2+)-O(2) becomes thermodynamically less favorable as substrate binds, and we propose that it is the slowing down of this oxidation step at higher concentrations of substrate that is the origin of the inhibition. In contrast, we show that regeneration of the ferrous enzyme (and formation of NFK) in the final step of the mechanism, which formally requires reduction of the heme, is facilitated by the higher reduction potential in the substrate-bound enzyme and the two constants (k(cat) and E(m)(0)) are shown also to be correlated. Thus, the overall catalytic activity is balanced between the equal and opposite dependencies of the initial and final steps of the mechanism on the heme reduction potential. This tuning of the reduction potential provides a simple mechanism for regulation of the reactivity, which may be used more widely across this family of enzymes.


Subject(s)
Biochemistry/methods , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Catalysis , Chemistry, Pharmaceutical/methods , Heme/chemistry , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kinetics , Kynurenine/analogs & derivatives , Kynurenine/chemistry , Mutagenesis, Site-Directed , Oxygen/chemistry , Protein Binding , Substrate Specificity , Thermodynamics , Tryptophan/chemistry
3.
J Am Chem Soc ; 133(40): 16251-7, 2011 Oct 12.
Article in English | MEDLINE | ID: mdl-21892828

ABSTRACT

Heme dioxygenases catalyze the oxidation of L-tryptophan to N-formylkynurenine (NFK), the first and rate-limiting step in tryptophan catabolism. Although recent progress has been made on early stages in the mechanism, there is currently no experimental data on the mechanism of product (NFK) formation. In this work, we have used mass spectrometry to examine product formation in a number of dioxygenases. In addition to NFK formation (m/z = 237), the data identify a species (m/z = 221) that is consistent with insertion of a single atom of oxygen into the substrate during O(2)-driven turnover. The fragmentation pattern for this m/z = 221 species is consistent with a cyclic amino acetal structure; independent chemical synthesis of the 3a-hydroxypyrroloindole-2-carboxylic acid compound is in agreement with this assignment. Labeling experiments with (18)O(2) confirm the origin of the oxygen atom as arising from O(2)-dependent turnover. These data suggest that the dioxygenases use a ring-opening mechanism during NFK formation, rather than Criegee or dioxetane mechanisms as previously proposed.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenine/analogs & derivatives , Tryptophan Oxygenase/metabolism , Heme/metabolism , Humans , Kynurenine/metabolism , Mass Spectrometry , Oxygen/metabolism , Xanthomonas campestris/enzymology
4.
J Am Chem Soc ; 132(15): 5494-500, 2010 Apr 21.
Article in English | MEDLINE | ID: mdl-20353179

ABSTRACT

We have applied cryoreduction/EPR/ENDOR techniques to characterize the active-site structure of the ferrous-oxy complexes of human (hIDO) and Shewanella oneidensis (sIDO) indoleamine 2,3-dioxygenases, Xanthomonas campestris (XcTDO) tryptophan 2,3-dioxygenase, and the H55S variant of XcTDO in the absence and in the presence of the substrate L-Trp and a substrate analogue, L-Me-Trp. The results reveal the presence of multiple conformations of the binary ferrous-oxy species of the IDOs. In more populated conformers, most likely a water molecule is within hydrogen-bonding distance of the bound ligand, which favors protonation of a cryogenerated ferric peroxy species at 77 K. In contrast to the binary complexes, cryoreduction of all of the studied ternary [enzyme-O(2)-Trp] dioxygenase complexes generates a ferric peroxy heme species with very similar EPR and (1)H ENDOR spectra in which protonation of the basic peroxy ligand does not occur at 77 K. Parallel studies with L-Me-Trp, in which the proton of the indole nitrogen is replaced with a methyl group, eliminate the possibility that the indole NH group of the substrate acts as a hydrogen bond donor to the bound O(2), and we suggest instead that the ammonium group of the substrate hydrogen-bonds to the dioxygen ligand. The present data show that substrate binding, primarily through this H-bond, causes the bound dioxygen to adopt a new conformation, which presumably is oriented for insertion of O(2) into the C(2)-C(3) double bond of the substrate. This substrate interaction further helps control the reactivity of the heme-bound dioxygen by "shielding" it from water.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Tryptophan Oxygenase/chemistry , Catalytic Domain , Electron Spin Resonance Spectroscopy/methods , Ferrous Compounds/metabolism , Humans , Hydrogen Bonding , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Oxygen/chemistry , Tryptophan/metabolism , Tryptophan Oxygenase/metabolism , Xanthomonas campestris/enzymology
5.
Biochem Soc Trans ; 37(Pt 2): 408-12, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19290871

ABSTRACT

The family of haem dioxygenases catalyse the initial oxidative cleavage of L-tryptophan to N-formylkynurenine, which is the first, rate-limiting, step in the L-kynurenine pathway. In the present paper, we discuss and compare structure and function across the family of haem dioxygenases by focusing on TDO (tryptophan 2,3-dioxygenase) and IDO (indoleamine 2,3-dioxygenase), including a review of recent structural information for both enzymes. The present paper describes how the recent development of recombinant expression systems has informed our more detailed understanding of the substrate binding, catalytic activity and mechanistic properties of these haem dioxygenases.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Tryptophan Oxygenase/metabolism , Tryptophan/metabolism , Binding Sites , Catalysis , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Oxidation-Reduction , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tryptophan Oxygenase/chemistry
6.
J Am Chem Soc ; 131(12): 4186-7, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19275153

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) are heme enzymes that catalyze the O(2)-dependent oxidation of L-tryptophan to N-formyl-kynurenine. Previous proposals for the mechanism of this reaction have suggested that deprotonation of the indole NH group, either by an active-site base or by oxygen bound to the heme iron, as the initial step. In this work, we have examined the activity of 1-Me-L-Trp with three different heme dioxygenases and their site-directed variants. We find, in contrast to previous work, that 1-Me-L-Trp is a substrate for the heme dioxygenase enzymes. These observations suggest that deprotonation of the indole N(1) is not essential for catalysis, and an alternative reaction mechanism, based on the known chemistry of indoles, is presented.


Subject(s)
Chemistry, Organic/methods , Dioxygenases/chemistry , Heme/chemistry , Catalysis , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoles/chemistry , Kinetics , Kynurenine/chemistry , Models, Chemical , Mutagenesis, Site-Directed , Oxygen/chemistry , Protons , Tryptophan/chemistry , Tryptophan Oxygenase/chemistry
7.
Biochemistry ; 47(16): 4752-60, 2008 Apr 22.
Article in English | MEDLINE | ID: mdl-18370401

ABSTRACT

The family of heme dioxygenases, as exemplified by indoleamine 2,3-dioxygenase and tryptophan 2,3-dioxygenase, catalyzes the oxidative cleavage of L-tryptophan to N-formylkynurenine. Here, we describe a bacterial expression system for human tryptophan 2,3-dioxygenase (rhTDO) together with spectroscopic, kinetic, and redox analyses. We find unexpected differences between human tryptophan 2,3-dioxygenase and human indoleamine 2,3-dioxygenase [Chauhan et al. (2008) Biochemistry 47, 4761-4769 ]. Thus, in contrast to indoleamine 2,3-dioxygenase, the catalytic ferrous-oxy complex of rhTDO is not observed, nor does the enzyme discriminate against substrate binding to the ferric derivative. In addition, we show that the rhTDO is also catalytically active in the ferric form. These new findings illustrate that significant mechanistic differences exist across the heme dioxygenase family, and the data are discussed within this broader framework.


Subject(s)
Tryptophan Oxygenase/chemistry , Tryptophan Oxygenase/metabolism , Electrons , Gene Expression , Humans , Iron/metabolism , Kinetics , Ligands , Molecular Structure , Oxidation-Reduction , Oxygen/metabolism , Potentiometry , Protein Binding , Spectrophotometry , Tryptophan/chemistry , Tryptophan/metabolism , Tryptophan Oxygenase/genetics , Tryptophan Oxygenase/isolation & purification
8.
Biochemistry ; 47(16): 4761-9, 2008 Apr 22.
Article in English | MEDLINE | ID: mdl-18370410

ABSTRACT

The initial step in the l-kynurenine pathway is oxidation of l-tryptophan to N-formylkynurenine and is catalyzed by one of two heme enzymes, tryptophan 2,3-dioxygenase (TDO) or indoleamine 2,3-dioxygenase (IDO). Here, we address the role of the conserved active site Ser167 residue in human IDO (S167A and S167H variants), which is replaced with a histidine in other mammalian and bacterial TDO enzymes. Our kinetic and spectroscopic data for S167A indicate that this residue is not essential for O 2 or substrate binding, and we propose that hydrogen bond stabilization of the catalytic ferrous-oxy complex involves active site water molecules in IDO. The data for S167H show that the ferrous-oxy complex is dramatically destabilized in this variant, which is similar to the behavior observed in human TDO [Basran et al. (2008) Biochemistry 47, 4752-4760], and that this destabilization essentially destroys catalytic activity. New kinetic data for the wild-type enzyme also identify the ternary [enzyme-O 2-substrate] complex. The data reveal significant differences between the IDO and TDO enzymes, and the implications of these results are discussed in terms of our current understanding of IDO and TDO catalysis.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Tryptophan Oxygenase/chemistry , Tryptophan Oxygenase/metabolism , Binding Sites , Catalysis , Cyanides/chemistry , Cyanides/metabolism , Electron Spin Resonance Spectroscopy , Ferrous Compounds/metabolism , Humans , Hydrogen Bonding , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Models, Molecular , Molecular Structure , Mutation/genetics , Oxidation-Reduction , Protein Binding , Serine/genetics , Serine/metabolism , Spectrophotometry , Substrate Specificity , Thermodynamics , Tryptophan/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...