Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem J ; 469(2): 211-21, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26171830

ABSTRACT

Mammalian sulfite oxidase (SO) is a dimeric enzyme consisting of a molybdenum cofactor- (Moco) and haem-containing domain and catalyses the oxidation of toxic sulfite to sulfate. Following sulfite oxidation, electrons are passed from Moco via the haem cofactor to cytochrome c, the terminal electron acceptor. In contrast, plant SO (PSO) lacks the haem domain and electrons shuttle from Moco to molecular oxygen. Given the high similarity between plant and mammalian SO Moco domains, factors that determine the reactivity of PSO towards oxygen, remained unknown. In the present study, we generated mammalian haem-deficient and truncated SO variants and demonstrated their oxygen reactivity by hydrogen peroxide formation and oxygen-consumption studies. We found that intramolecular electron transfer between Moco and haem showed an inverse correlation to SO oxygen reactivity. Haem-deficient SO variants exhibited oxygen-dependent sulfite oxidation similar to PSO, which was confirmed further using haem-deficient human SO in a cell-based assay. This finding suggests the possibility to use oxygen-reactive SO variants in sulfite detoxification, as the loss of SO activity is causing severe neurodegeneration. Therefore we evaluated the potential use of PEG attachment (PEGylation) as a modification method for future enzyme substitution therapies using oxygen-reactive SO variants, which might use blood-dissolved oxygen as the electron acceptor. PEGylation has been shown to increase the half-life of other therapeutic proteins. PEGylation resulted in the modification of up to eight surface-exposed lysine residues of SO, an increased conformational stability and similar kinetic properties compared with wild-type SO.


Subject(s)
Amino Acid Metabolism, Inborn Errors/drug therapy , Enzyme Replacement Therapy , Oxidoreductases Acting on Sulfur Group Donors , Oxygen , Sulfite Oxidase/deficiency , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Animals , Enzyme Stability/genetics , HEK293 Cells , Heme/chemistry , Heme/genetics , Heme/metabolism , Humans , Hydrogen Peroxide , Mice , Oxidoreductases Acting on Sulfur Group Donors/chemistry , Oxidoreductases Acting on Sulfur Group Donors/genetics , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Oxidoreductases Acting on Sulfur Group Donors/therapeutic use , Oxygen/chemistry , Oxygen/metabolism , Oxygen Consumption/drug effects , Polyethylene Glycols/chemistry , Sulfite Oxidase/genetics , Sulfite Oxidase/metabolism
2.
Antioxid Redox Signal ; 23(4): 283-94, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25314640

ABSTRACT

AIMS: Recent studies suggest that the molybdenum enzymes xanthine oxidase, aldehyde oxidase, and mARC exhibit nitrite reductase activity at low oxygen pressures. However, inhibition studies of xanthine oxidase in humans have failed to block nitrite-dependent changes in blood flow, leading to continued exploration for other candidate nitrite reductases. Another physiologically important molybdenum enzyme­sulfite oxidase (SO)­has not been extensively studied. RESULTS: Using gas-phase nitric oxide (NO) detection and physiological concentrations of nitrite, SO functions as nitrite reductase in the presence of a one-electron donor, exhibiting redox coupling of substrate oxidation and nitrite reduction to form NO. With sulfite, the physiological substrate, SO only facilitates one turnover of nitrite reduction. Studies with recombinant heme and molybdenum domains of SO indicate that nitrite reduction occurs at the molybdenum center via coupled oxidation of Mo(IV) to Mo(V). Reaction rates of nitrite to NO decreased in the presence of a functional heme domain, mediated by steric and redox effects of this domain. Using knockdown of all molybdopterin enzymes and SO in fibroblasts isolated from patients with genetic deficiencies of molybdenum cofactor and SO, respectively, SO was found to significantly contribute to hypoxic nitrite signaling as demonstrated by activation of the canonical NO-sGC-cGMP pathway. INNOVATION: Nitrite binds to and is reduced at the molybdenum site of mammalian SO, which may be allosterically regulated by heme and molybdenum domain interactions, and contributes to the mammalian nitrate-nitrite-NO signaling pathway in human fibroblasts. CONCLUSION: SO is a putative mammalian nitrite reductase, catalyzing nitrite reduction at the Mo(IV) center.


Subject(s)
Coenzymes/chemistry , Metalloproteins/chemistry , Nitric Oxide/chemistry , Nitrites/chemistry , Pteridines/chemistry , Sulfite Oxidase/chemistry , Electron Transport , Fibroblasts/enzymology , Fibroblasts/metabolism , Heme/chemistry , Humans , Molybdenum/chemistry , Molybdenum Cofactors , Nitric Oxide/metabolism , Nitrites/metabolism , Oxidation-Reduction , Protein Structure, Tertiary , Signal Transduction , Sulfite Oxidase/metabolism
3.
Biochemistry ; 49(37): 8177-86, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20690630

ABSTRACT

Eukaryotic assimilatory nitrate reductase (NR) is a dimeric multidomain molybdo-heme-flavo protein that catalyzes the first and rate-limiting step in the nitrate assimilation of plants, algae, and fungi. Nitrate reduction takes place at the N-terminal molybdenum cofactor-containing domain. Reducing equivalents are derived from NADH, which reduce the C-terminal FAD domain followed by single-electron transfer steps via the middle heme domain to the molybdenum center. In plants, nitrate reduction is post-translationally inhibited by phosphorylation and subsequent binding of 14-3-3 protein to a conserved phosphoserine located in the surface-exposed hinge between the catalytic and heme domain. Here we investigated Arabidopsis thaliana NR activity upon phosphorylation and 14-3-3 binding by using a fully defined in vitro system with purified proteins. We demonstrate that among different calcium-dependent protein kinases (CPKs), CPK-17 efficiently phosphorylates Ser534 in NR. Out of eight purified Arabidopsis 14-3-3 proteins, isoforms ω, κ, and λ exhibited the strongest inhibition of NR. The kinetic parameters of noninhibited, phosphorylated NR (pNR) and pNR in a complex with 14-3-3 were investigated. An 18-fold reduction in k(cat) and a decrease in the apparent K(M)(nitrate) (from 280 to 141 µM) were observed upon binding of 14-3-3 to pNR, suggesting a noncompetitive inhibition with a preferential binding to the substrate-bound state of the enzyme. Recording partial activities of NR demonstrated that the transfer of electrons to the heme is not affected by 14-3-3 binding. The Ser534Ala variant of NR was not inhibited by 14-3-3 proteins. We propose that 14-3-3 binding to Ser534 blocks the transfer of electrons from heme to nitrate by arresting the domain movement via hinge 1.


Subject(s)
14-3-3 Proteins/metabolism , Arabidopsis/enzymology , Nitrate Reductase (NADH)/metabolism , Arabidopsis/metabolism , Catalysis , Coenzymes , Eukaryota , Heme/metabolism , Kinetics , Metalloproteins , Molybdenum/metabolism , Molybdenum Cofactors , NAD , Nitrate Reductase/metabolism , Nitrate Reductases/chemistry , Nitrate Reductases/metabolism , Oxidation-Reduction , Phosphorylation , Protein Kinases , Pteridines
SELECTION OF CITATIONS
SEARCH DETAIL
...