Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 205
Filter
1.
Nitric Oxide ; 119: 41-49, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34942379

ABSTRACT

Nitric oxide synthase (NOS) catalyzes NO formation from the substrate l-arginine (Arg). Previously, NOS with distinct biochemical properties were characterized from two photosynthetic microorganisms, the unicellular algae Ostreococcus tauri (OtNOS) and the cyanobacteria Synechococcus PCC 7335 (SyNOS). In this work we studied the effect of recombinant OtNOS and SyNOS expressed under IPTG-induced promoter in E. coli, a bacterium that lacks NOS. Results show that OtNOS and SyNOS expression promote E. coli growth in a nutrient replete medium and allow to better metabolize Arg as N source. In LB medium, OtNOS induces the expression of the NO dioxygenase hmp in E. coli, in accordance with high NO levels visualized with the probe DAF-FM DA. In contrast, SyNOS expression does not induce hmp and show a slight increase of NO production compared to OtNOS. NOS expression reduces ROS production and increases viability of E. coli cultures growing in LB. A strong nitrosative stress provoked by the addition of 1 mM of the NO donors sodium nitroprusside (SNP) and nitrosoglutathione (GSNO) inhibits bacterial growth rate. Under these conditions, the expression of OtNOS or SyNOS counteracts NO donor toxicity restoring bacterial growth. Finally, using bioinformatic tools and ligand docking analyses, we postulate that tetrahydromonapterin (MH4), an endogenous pterin found in E. coli, could act as cofactor required for NOS catalytic activity. Our findings could be useful for the development of biotechnological applications using NOS expression to improve growth in NOS-lacking bacteria.


Subject(s)
Biopterins/analogs & derivatives , Coenzymes/metabolism , Escherichia coli/growth & development , Nitric Oxide Synthase/metabolism , Nitrosative Stress/physiology , Algal Proteins/chemistry , Algal Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Biopterins/chemistry , Biopterins/metabolism , Chlorophyta/enzymology , Coenzymes/chemistry , Escherichia coli/metabolism , Molecular Docking Simulation , Nitric Oxide/metabolism , Nitric Oxide Synthase/chemistry , Protein Binding , Reactive Oxygen Species/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Synechococcus/enzymology
2.
Biochem Soc Trans ; 48(5): 2253-2259, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33125483

ABSTRACT

Lipid peroxidation has been associated with a wide array of (patho)physiological conditions. Remarkably, in the last few years, a novel cell death modality termed ferroptosis was recognized as a process initiated by iron-dependent oxidation of lipids. The sensitivity to ferroptosis is determined by the activity of antioxidant systems working on the repair of oxidized phospholipids and also metabolic pathways controlling the availability of substrates susceptible to lipid peroxidation. Non-enzymatic antioxidants such as vitamin E, which has long been acknowledged as an efficient inhibitor of lipid peroxidation, play an important and often neglected role in subverting ferroptosis. Recent works dissecting the mechanisms that determine ferroptosis sensitivity have provided further insights into the contribution of alternative metabolic pathways able to suppress lipid peroxidation. Specifically, the role of ubiquinone and tetrahydrobiopterin (BH4) has been brought forth, with the identification of specific enzymatic systems responsible for their regeneration, as critical factors suppressing ferroptosis. Therefore, in the present manuscript, we address these emerging concepts and propose that the characterization of these antioxidant repair mechanisms will not only open a new understanding of disease conditions where ferroptosis plays a role but also offer opportunities to identify and sensitize cells to ferroptosis in the context of cancer treatment.


Subject(s)
Antioxidants/chemistry , Ferroptosis , Gene Expression Regulation , Oxygen/chemistry , Animals , Biopterins/analogs & derivatives , Biopterins/chemistry , Cell Death/drug effects , Humans , Iron , Lipid Peroxidation , Lipids/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Ubiquinone/chemistry , Vitamin E/metabolism
3.
J Nat Prod ; 83(10): 3156-3165, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33030894

ABSTRACT

Fourteen aromatic metabolites (6-19) were isolated from an aqueous extract of the solitary tunicate Cnemidocarpa irene collected in Hokkaido, Japan. The structures of the metabolites were determined based on the spectroscopic interpretations, including one- and two-dimensional NMR, mass spectra, UV, and circular dichroism data. The biopterin analogue 10 modulated the behavior of mice after intracerebroventricular injection and showed a weak affinity to ionotropic glutamate receptor subtypes. Analyses of fluorescent coelomic fluid of the tunicate revealed that pterin 12 was responsible for the fluorescence of the blood cells, while ß-carbolines 1 and 3 were fluorescent compounds in the serum. The metabolic profiles in adults, juveniles, larvae, and eggs of the animal differed substantially, suggesting that the metabolism of the animal, especially biosynthesis of aromatic secondary metabolites, changes over different life stages.


Subject(s)
Hydrocarbons, Aromatic/metabolism , Urochordata/chemistry , Urochordata/metabolism , Animals , Behavior, Animal/drug effects , Biopterins/analogs & derivatives , Biopterins/chemistry , Biopterins/pharmacology , Carbolines/chemistry , Carbolines/pharmacology , Cholinesterase Inhibitors/pharmacology , Circular Dichroism , HeLa Cells/drug effects , Humans , Injections, Intraventricular , Larva , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Molecular Structure , Nucleosides/chemistry , Nucleosides/pharmacology , Ovum/metabolism , Pterins/chemistry , Pterins/isolation & purification , Pterins/pharmacology , Receptors, Ionotropic Glutamate/drug effects , Spectrophotometry, Ultraviolet , Tyramine/chemistry , Tyramine/pharmacology , Urochordata/growth & development
4.
Genes (Basel) ; 10(6)2019 06 15.
Article in English | MEDLINE | ID: mdl-31208052

ABSTRACT

The molecular genetics of well-characterized inherited diseases, such as phenylketonuria (PKU) and hyperphenylalaninemia (HPA) predominantly caused by mutations in the phenylalanine hydroxylase (PAH) gene, is often complicated by the identification of many novel variants, often with no obvious impact on the associated disorder. To date, more than 1100 PAH variants have been identified of which a substantial portion have unknown clinical significance. In this work, we study the functionality of seven yet uncharacterized PAH missense variants p.Asn167Tyr, p.Thr200Asn, p.Asp229Gly, p.Gly239Ala, p.Phe263Ser, p.Ala342Pro, and p.Ile406Met first identified in the Czech PKU/HPA patients. From all tested variants, three of them, namely p.Asn167Tyr, p.Thr200Asn, and p.Ile406Met, exerted residual enzymatic activity in vitro similar to wild type (WT) PAH, however, when expressed in HepG2 cells, their protein level reached a maximum of 72.1% ± 4.9%, 11.2% ± 4.2%, and 36.6% ± 7.3% compared to WT PAH, respectively. Remaining variants were null with no enzyme activity and decreased protein levels in HepG2 cells. The chaperone-like effect of applied BH4 precursor increased protein level significantly for p.Asn167Tyr, p.Asp229Gly, p.Ala342Pro, and p.Ile406Met. Taken together, our results of functional characterization in combination with in silico prediction suggest that while p.Asn167Tyr, p.Thr200Asn, and p.Ile406Met PAH variants have a mild impact on the protein, p.Asp229Gly, p.Gly239Ala, p.Phe263Ser, and p.Ala342Pro severely affect protein structure and function.


Subject(s)
Biopterins/analogs & derivatives , Mutation, Missense/genetics , Phenylalanine Hydroxylase/chemistry , Phenylketonurias/genetics , Biopterins/chemistry , Biopterins/genetics , Computer Simulation , Genotype , Hep G2 Cells , Humans , Phenylalanine Hydroxylase/genetics , Phenylketonurias/metabolism , Phenylketonurias/pathology , Structure-Activity Relationship
5.
J Biol Chem ; 294(27): 10708-10719, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31113865

ABSTRACT

Nitric oxide synthases (NOSs) are heme-based monooxygenases that convert l-Arg to l-citrulline and nitric oxide (NO), a key signaling molecule and cytotoxic agent in mammals. Bacteria also contain NOS proteins, but the role of NO production within these organisms, where understood, differs considerably from that of mammals. For example, a NOS protein in the marine cyanobacterium Synechococcus sp. PCC 7335 (syNOS) has recently been proposed to function in nitrogen assimilation from l-Arg. syNOS retains the oxygenase (NOSox) and reductase (NOSred) domains present in mammalian NOS enzymes (mNOSs), but also contains an N-terminal globin domain (NOSg) homologous to bacterial flavohemoglobin proteins. Herein, we show that syNOS functions as a dimer and produces NO from l-Arg and NADPH in a tetrahydrobiopterin (H4B)-dependent manner at levels similar to those produced by other NOSs but does not require Ca2+-calmodulin, which regulates NOSred-mediated NOSox reduction in mNOSs. Unlike other bacterial NOSs, syNOS cannot function with tetrahydrofolate and requires high Ca2+ levels (>200 µm) for its activation. NOSg converts NO to NO3- in the presence of O2 and NADPH; however, NOSg did not protect Escherichia coli strains against nitrosative stress, even in a mutant devoid of NO-protective flavohemoglobin. We also found that syNOS does not have NOS activity in E. coli (which lacks H4B) and that the recombinant protein does not confer growth advantages on l-Arg as a nitrogen source. Our findings indicate that syNOS has both NOS and NO oxygenase activities, requires H4B, and may play a role in Ca2+-mediated signaling.


Subject(s)
Arginine/metabolism , Bacterial Proteins/metabolism , NADP/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Synechococcus/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biopterins/analogs & derivatives , Biopterins/chemistry , Biopterins/metabolism , Calcium/chemistry , Calcium/metabolism , Dimerization , Escherichia coli/metabolism , Kinetics , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/genetics , Protein Domains , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
6.
Proc Natl Acad Sci U S A ; 116(23): 11229-11234, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31118288

ABSTRACT

Phenylalanine hydroxylase (PAH) is a key enzyme in the catabolism of phenylalanine, and mutations in this enzyme cause phenylketonuria (PKU), a genetic disorder that leads to brain damage and mental retardation if untreated. Some patients benefit from supplementation with a synthetic formulation of the cofactor tetrahydrobiopterin (BH4) that partly acts as a pharmacological chaperone. Here we present structures of full-length human PAH (hPAH) both unbound and complexed with BH4 in the precatalytic state. Crystal structures, solved at 3.18-Å resolution, show the interactions between the cofactor and PAH, explaining the negative regulation exerted by BH4 BH4 forms several H-bonds with the N-terminal autoregulatory tail but is far from the catalytic FeII Upon BH4 binding a polar and salt-bridge interaction network links the three PAH domains, explaining the stability conferred by BH4 Importantly, BH4 binding modulates the interaction between subunits, providing information about PAH allostery. Moreover, we also show that the cryo-EM structure of hPAH in absence of BH4 reveals a highly dynamic conformation for the tetramers. Structural analyses of the hPAH:BH4 subunits revealed that the substrate-induced movement of Tyr138 into the active site could be coupled to the displacement of BH4 from the precatalytic toward the active conformation, a molecular mechanism that was supported by site-directed mutagenesis and targeted molecular dynamics simulations. Finally, comparison of the rat and human PAH structures show that hPAH is more dynamic, which is related to amino acid substitutions that enhance the flexibility of hPAH and may increase the susceptibility to PKU-associated mutations.


Subject(s)
Biopterins/analogs & derivatives , Phenylalanine Hydroxylase/chemistry , Biopterins/chemistry , Biopterins/genetics , Humans , Models, Molecular , Molecular Dynamics Simulation , Mutagenesis, Site-Directed/methods , Mutation/genetics , Phenylalanine Hydroxylase/genetics , Phenylketonurias/genetics
7.
Nitric Oxide ; 89: 14-21, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31022534

ABSTRACT

Nitric oxide synthase (NOS) catalyzes the transformation of l-arginine, molecular oxygen (O2), and NADPH-derived electrons to nitric oxide (NO) and l-citrulline. Under some conditions, however, NOS catalyzes the reduction of O2 to superoxide (O2-) instead, a phenomenon that is generally referred to as uncoupling. In principle, both the heme in the oxygenase domain and the flavins in the reductase domain could catalyze O2- formation. In the former case the oxyferrous (Fe(II)O2) complex that is formed as an intermediate during catalysis would dissociate to heme and O2-; in the latter case the reduced flavins would reduce O2 to O2-. The NOS cofactor tetrahydrobiopterin (BH4) is indispensable for coupled catalysis. In the case of uncoupling at the heme this is explained by the essential role of BH4 as an electron donor to the oxyferrous complex; in the case of uncoupling at the flavins it is assumed that the absence of BH4 results in NOS monomerization, with the monomers incapable to sustain NO synthesis but still able to support uncoupled catalysis. In spite of little supporting evidence, uncoupling at the reductase after NOS monomerization appears to be the predominant hypothesis at present. To set the record straight we extended prior studies by determining under which conditions uncoupling of the neuronal and endothelial isoforms (nNOS and eNOS) occurred and if a correlation exists between uncoupling and the monomer/dimer equilibrium. We determined the rates of coupled/uncoupled catalysis by measuring NADPH oxidation spectrophotometrically at 340 nm and citrulline synthesis as the formation of [3H]-citrulline from [3H]-Arg. The monomer/dimer equilibrium was determined by FPLC and, for comparison, by low-temperature polyacrylamide gel electrophoresis. Uncoupling occurred in the absence of Arg and/or BH4, but not in the absence of Ca2+ or calmodulin (CaM). Since omission of Ca2+/CaM will completely block heme reduction while still allowing substantial FMN reduction, this argues against uncoupling by the reductase domain. In the presence of heme-directed NOS inhibitors uncoupling occurred to the extent that these compound allowed heme reduction, again arguing in favor of uncoupling at the heme. The monomer/dimer equilibrium showed no correlation with uncoupling. We conclude that uncoupling by BH4 deficiency takes place exclusively at the heme, with virtually no contribution from the flavins and no role for NOS monomerization.


Subject(s)
Nitric Oxide Synthase/chemistry , Biopterins/analogs & derivatives , Biopterins/chemistry , Citrulline/chemistry , Enzyme Inhibitors/chemistry , Heme/chemistry , Humans , Imidazoles/chemistry , NADP/chemistry , Nitric Oxide Synthase/antagonists & inhibitors , Nitroarginine/chemistry , Oxygen/chemistry , Pichia/genetics , Protein Multimerization
8.
Hum Mol Genet ; 27(10): 1732-1742, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29514280

ABSTRACT

Metabolic control of phenylalanine concentrations in body fluids is essential for cognitive development and executive function. The hepatic phenylalanine hydroxylating system is regulated by the ratio of l-phenylalanine, which is substrate of phenylalanine hydroxylase (PAH), to the PAH cofactor tetrahydrobiopterin (BH4). Physiologically, phenylalanine availability is governed by nutrient intake, whereas liver BH4 is kept at constant level. In phenylketonuria, PAH deficiency leads to elevated blood phenylalanine and is often caused by PAH protein misfolding with loss of function. Here, we report secondary hepatic BH4 deficiency in Pah-deficient mice. Alterations in de novo synthesis and turnover of BH4 were ruled out as molecular causes. We demonstrate that kinetically instable and aggregation-prone variant Pah proteins trap BH4, shifting the pool of free BH4 towards bound BH4. Interference of PAH protein misfolding with metabolite-based control of l-phenylalanine turnover suggests a mechanistic link between perturbation of protein homeostasis and disturbed regulation of metabolic pathways.


Subject(s)
Biopterins/analogs & derivatives , Phenylalanine Hydroxylase/genetics , Phenylalanine/metabolism , Phenylketonurias/genetics , Animals , Biopterins/chemistry , Biopterins/genetics , Biopterins/metabolism , Disease Models, Animal , Humans , Inactivation, Metabolic/genetics , Kinetics , Liver/enzymology , Mice , Phenylalanine/chemistry , Phenylalanine/genetics , Phenylalanine Hydroxylase/chemistry , Phenylalanine Hydroxylase/metabolism , Phenylketonurias/metabolism , Phenylketonurias/pathology , Protein Folding , Proteostasis/genetics
9.
J Inorg Biochem ; 181: 28-40, 2018 04.
Article in English | MEDLINE | ID: mdl-29407906

ABSTRACT

Nitric oxide (NO·) is a messenger molecule with diverse physiological roles including host defense, neurotransmission and vascular function. The synthesis of NO· from l-arginine is catalyzed by NO-synthases and occurs in two steps through the intermediary Nω-hydroxy-l-arginine (NHA). In both steps the P450-like reaction cycle is coupled with the redox cycle of the cofactor tetrahydrobiopterin (H4B). The mechanism of the second step is studied by Density Functional Theory calculations to ascertain the canonical sequence of proton and electron transfer (PT and ET) events. The proposed mechanism is controlled by the interplay of two electron donors, H4B and NHA. Consistent with experimental data, the catalytic cycle proceeds through the ferric-hydroperoxide complex (Cpd 0) and the following aqua-ferriheme resting state, and involves interim partial oxidation of H4B. The mechanism starts with formation of Cpd 0 from the ferrous-dioxy reactant complex by PT from the C-ring heme propionate coupled with hole transfer to H4B through the highest occupied π-orbital of NHA as a bridge. This enables PT from NHA+· to the proximal oxygen leading to the shallow ferriheme-H2O2 oxidant. Subsequent Fenton-like peroxide bond cleavage triggered by ET from the NHA-derived iminoxy-radical leads to the protonated Cpd II diradicaloid singlet stabilized by spin delocalization in H4B, and the closed-shell coordination complex of HO- with iminoxy-cation. The complex is converted to the transient C-adduct, which releases intended products upon PT to the ferriheme-HO- complex coupled with ET to the H4B+·. Deferred ET from the substrate or undue ET from/to the cofactor leads to side products.


Subject(s)
Arginine/analogs & derivatives , Biopterins/analogs & derivatives , Models, Molecular , NADP/metabolism , Nitric Oxide Synthase Type II/metabolism , Animals , Arginine/chemistry , Arginine/metabolism , Biocatalysis , Biopterins/chemistry , Biopterins/metabolism , Catalytic Domain , Citrulline/chemistry , Citrulline/metabolism , Conserved Sequence , Databases, Protein , Electron Transport , Humans , Hydrogen Bonding , NADP/chemistry , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/chemistry , Oxidation-Reduction , Protons , Quantum Theory , Thermodynamics
10.
Proc Natl Acad Sci U S A ; 114(27): E5317-E5324, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28630292

ABSTRACT

Monoamine neurotransmitters are among the hundreds of signaling small molecules whose target interactions are switched "on" and "off" via transfer of the sulfuryl-moiety (-SO3) from PAPS (3'-phosphoadenosine 5'-phosphosulfate) to the hydroxyls and amines of their scaffolds. These transfer reactions are catalyzed by a small family of broad-specificity enzymes-the human cytosolic sulfotransferases (SULTs). The first structure of a SULT allosteric-binding site (that of SULT1A1) has recently come to light. The site is conserved among SULT1 family members and is promiscuous-it binds catechins, a naturally occurring family of flavanols. Here, the catechin-binding site of SULT1A3, which sulfonates monoamine neurotransmitters, is modeled on that of 1A1 and used to screen in silico for endogenous metabolite 1A3 allosteres. Screening predicted a single high-affinity allostere, tetrahydrobiopterin (THB), an essential cofactor in monoamine neurotransmitter biosynthesis. THB is shown to bind and inhibit SULT1A3 with high affinity, 23 (±2) nM, and to bind weakly, if at all, to the four other major SULTs found in brain and liver. The structure of the THB-bound binding site is determined and confirms that THB binds the catechin site. A structural comparison of SULT1A3 with SULT1A1 (its immediate evolutionary progenitor) reveals how SULT1A3 acquired high affinity for THB and that the majority of residue changes needed to transform 1A1 into 1A3 are clustered at the allosteric and active sites. Finally, sequence records reveal that the coevolution of these sites played an essential role in the evolution of simian neurotransmitter metabolism.


Subject(s)
Amines/chemistry , Biopterins/analogs & derivatives , Neurotransmitter Agents/chemistry , Allosteric Site , Arylsulfotransferase/chemistry , Binding Sites , Biopterins/chemistry , Escherichia coli/metabolism , Genetic Vectors , Humans , Isoenzymes/chemistry , Kinetics , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Protein Binding , Software , Sulfur/chemistry
11.
Biochemistry ; 56(5): 748-756, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28074650

ABSTRACT

Nitric oxide is produced in mammals by the nitric oxide synthase (NOS) isoforms at a catalytic site comprising a heme associated with a biopterin cofactor. Through genome sequencing, proteins that are highly homologous to the oxygenase domain of NOSs have been identified, in particular in bacteria. The active site is highly conserved except for a valine residue in the distal pocket that is replaced with an isoleucine in bacteria. This switch was previously reported to influence the kinetics of the reaction. We have used the V346I mutant of the mouse inducible NOS (iNOS) as well as the I224V mutant of the NOS from Bacillus subtilis (bsNOS) to study their spectroscopic signatures in solution and look for potential structural differences compared to their respective wild types. Both mutants seem destabilized in the absence of substrate and cofactor. When both substrate and cofactor are present, small differences can be detected with Nω-hydroxy-l-arginine compared to arginine, which is likely due to the differences in the hydrogen bonding network of the distal pocket. Stopped-flow experiments evidence significant changes in the kinetics of the reaction due to the mutation as was already known. We found these effects particularly marked for iNOS. On the basis of these results, we performed rapid freeze-quench experiments to trap the biopterin radical and found the same results that we had obtained for the wild types. Despite differences in kinetics, a radical could be trapped in both steps for the iNOS mutant but only for the first step in the mutant of bsNOS. This strengthens the hypothesis that mammalian and bacterial NOSs may have a different mechanism during the second catalytic step.


Subject(s)
Bacterial Proteins/chemistry , Isoleucine/chemistry , Mutation , Nitric Oxide Synthase Type II/chemistry , Nitric Oxide Synthase/chemistry , Valine/chemistry , Amino Acid Substitution , Animals , Arginine/analogs & derivatives , Arginine/chemistry , Arginine/metabolism , Bacillus subtilis/enzymology , Bacillus subtilis/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biocatalysis , Biopterins/chemistry , Biopterins/metabolism , Catalytic Domain , Coenzymes/chemistry , Coenzymes/metabolism , Conserved Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Hydrogen Bonding , Isoleucine/metabolism , Kinetics , Mice , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxidation-Reduction , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Species Specificity , Valine/metabolism
12.
Free Radic Biol Med ; 101: 367-377, 2016 12.
Article in English | MEDLINE | ID: mdl-27989753

ABSTRACT

Reaction intermediates trapped during the single-turnover reaction of the neuronal ferrous nitric oxide synthase oxygenase domain (Fe(II)nNOSOX) show four EPR spectra of free radicals. Fully-coupled nNOSOX with cofactor (tetrahydrobiopterin, BH4) and substrate (l-arginine) forms the typical BH4 cation radical with an EPR spectrum ~4.0mT wide and hyperfine tensors similar to reports for a biopterin cation radical in inducible NOSOX (iNOSOX). With excess thiol, nNOSox lacking BH4 and l-arg is known to produce superoxide. In contrast, we find that nNOSOX with BH4 but no l-arg forms two radicals with rather different, fast (~250µs at 5K) and slower (~500µs at 20K), electron spin relaxation rates and a combined ~7.0mT wide EPR spectrum. Rapid freeze-quench CW- and pulsed-EPR measurements are used to identify these radicals and their origin. These two species are the same radical with identical nuclear hyperfine couplings, but with spin-spin couplings to high-spin (4.0mT component) or low-spin (7.0mT component) Fe(III) heme. Uncoupled reactions of nNOS leave the enzyme in states that can be chemically reduced to sustain unregulated production of NO and reactive oxygen species in ischemia-reperfusion injury. The broad EPR signal is a convenient indicator of uncoupled nNOS reactions producing low-spin Fe(III) heme.


Subject(s)
Biopterins/analogs & derivatives , Heme/chemistry , Nitric Oxide Synthase Type I/chemistry , Recombinant Proteins/chemistry , Animals , Biopterins/chemistry , Brain Chemistry , Electron Spin Resonance Spectroscopy , Free Radicals/chemistry , Gene Expression , Nitric Oxide Synthase Type I/genetics , Oxidation-Reduction , Protein Domains , Rats , Recombinant Proteins/genetics , Solutions , Temperature
13.
PLoS One ; 11(10): e0164305, 2016.
Article in English | MEDLINE | ID: mdl-27711248

ABSTRACT

Tetrahydrobiopterin (BH4) is an essential cofactor of nitric oxide synthase (NOS) and aromatic amino acid hydroxylases. BH4 and 7,8-dihydrobiopterin (BH2) are metabolically interchangeable at the expense of NADPH. Exogenously administered BH4 can be metabolized by the body, similar to vitamins. At present, synthetic BH4 is used as an orphan drug for patients with inherited diseases requiring BH4 supplementation. BH4 supplementation has also drawn attention as a means of treating certain cardiovascular symptoms, however, its application in human patients remains limited. Here, we tracked biopterin (BP) distribution in blood, bile, urine, liver, kidney and brain after BH4 administration (5 mg/kg rat, i.v.) with or without prior treatment with probenecid, a potent inhibitor of uptake transporters particularly including organic anion transporter families such as OTA1 and OAT3. The rapid excretion of BP in urine was driven by elevated blood concentrations and its elimination reached about 90% within 120 min. In the very early period, BP was taken up by the liver and kidney and gradually released back to the blood. BH4 administration caused a considerable decrease in the BH4% in blood BP as an inevitable compensatory process. Probenecid treatment slowed down the decrease in blood BP and simultaneously inhibited its initial rapid excretion in the kidney. At the same time, the BH4% was further lowered, suggesting that the probenecid-sensitive BP uptake played a crucial role in BH2 scavenging in vivo. This suggested that the overproduced BH2 was taken up by organs by means of the probenecid-sensitive process, and was then scavenged by counter-conversion to BH4 via the BH4 salvage pathway. Taken together, BH4 administration was effective at raising BP levels in organs over the course of hours but with extremely low efficiency. Since a high BH2 relative to BH4 causes NOS dysfunction, the lowering of the BH4% must be avoided in practice, otherwise the desired effect of the supplementation in ameliorating NOS dysfunction would be spoiled.


Subject(s)
Biopterins/analogs & derivatives , Biopterins/analysis , Kidney/drug effects , Liver/drug effects , Probenecid/pharmacology , Animals , Bile/chemistry , Bile/drug effects , Bile/metabolism , Biopterins/blood , Biopterins/chemistry , Biopterins/metabolism , Biopterins/pharmacology , Biopterins/urine , Brain/drug effects , Brain/metabolism , Cyclosporine/pharmacology , Erythrocytes/chemistry , Erythrocytes/drug effects , Erythrocytes/metabolism , Female , Kidney/chemistry , Kidney/metabolism , Liver/chemistry , Liver/metabolism , Male , Nitric Oxide Synthase/metabolism , Rats
14.
FEBS J ; 283(24): 4491-4501, 2016 12.
Article in English | MEDLINE | ID: mdl-27760279

ABSTRACT

The nitric oxide synthases (NOS) catalyze a two-step oxidation of l-arginine (Arg) to generate NO. In the first step, O2 activation involves one electron being provided to the heme by an enzyme-bound 6R-tetrahydro-l-biopterin cofactor (H4 B), and the H4 B radical must be reduced back to H4 B in order for NOS to continue catalysis. Although an NADPH-derived electron is used to reduce the H4 B radical, how this occurs is unknown. We hypothesized that the NOS flavoprotein domain might reduce the H4 B radical by utilizing the NOS heme porphyrin as a conduit to deliver the electron. This model predicts that factors influencing NOS heme reduction should also influence the extent and rate of H4 B radical reduction in kind. To test this, we utilized single catalytic turnover and stop-freeze methods, along with electron paramagnetic resonance spectroscopy, to measure the rate and extent of reduction of the 5-methyl-H4 B radical formed in neuronal NOS (nNOS) during Arg hydroxylation. We used several nNOS variants that supported either a slower or faster than normal rate of ferric heme reduction. We found that the rates and extents of nNOS heme reduction correlated well with the rates and extents of 5-methyl-H4 B radical reduction among the various nNOS enzymes. This supports a model where the heme porphyrin transfers an electron from the NOS flavoprotein to the H4 B radical formed during catalysis, revealing that the heme plays a dual role in catalyzing O2 activation or electron transfer at distinct points in the reaction cycle.


Subject(s)
Biopterins/analogs & derivatives , Electrons , Heme/metabolism , Nitric Oxide Synthase Type I/metabolism , Animals , Arginine/chemistry , Arginine/metabolism , Biocatalysis , Biopterins/chemistry , Biopterins/metabolism , Electron Spin Resonance Spectroscopy , Electron Transport , Free Radicals/chemistry , Free Radicals/metabolism , Heme/chemistry , Kinetics , Models, Molecular , Molecular Structure , Mutation , Nitric Oxide Synthase Type I/chemistry , Nitric Oxide Synthase Type I/genetics , Oxidation-Reduction , Protein Binding , Protein Domains , Rats
15.
Phytochemistry ; 123: 69-74, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26804212

ABSTRACT

Five previously undescribed biopterin glycosides, microcystbiopterin A-E, were isolated from the extracts of two bloom materials of Microcystis spp. collected from a fishpond (IL-337) and Lake Kinneret (IL-347), Israel. The structure of the pterins was established by interpretation of their UV, CD, 1D and 2D NMR spectra and HR mass measurements. Microcystbiopterin D is the first heptose containing pterin glycoside to be reported in the literature. Their antimicrobial and cytotoxic properties were evaluated but all were found not active in both assays.


Subject(s)
Biopterins/isolation & purification , Glycosides/isolation & purification , Microcystis/chemistry , Biomass , Biopterins/analogs & derivatives , Biopterins/chemistry , Biopterins/pharmacology , Glycosides/chemistry , Glycosides/pharmacology , Israel , Molecular Structure , Ponds
16.
Mol Genet Metab ; 117(1): 5-11, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26653793

ABSTRACT

Central nervous system (CNS) deficiencies of the monoamine neurotransmitters, dopamine and serotonin, have been implicated in the pathophysiology of neuropsychiatric dysfunction in phenylketonuria (PKU). Increased brain phenylalanine concentration likely competitively inhibits the activities of tyrosine hydroxylase (TH) and tryptophan hydroxylase (TPH), the rate limiting steps in dopamine and serotonin synthesis respectively. Tetrahydrobiopterin (BH4) is a required cofactor for TH and TPH activity. Our hypothesis was that treatment of hyperphenylalaninemic Pah(enu2/enu2) mice, a model of human PKU, with sapropterin dihydrochloride, a synthetic form of BH4, would stimulate TH and TPH activities leading to improved dopamine and serotonin synthesis despite persistently elevated brain phenylalanine. Sapropterin (20, 40, or 100mg/kg body weight in 1% ascorbic acid) was administered daily for 4 days by oral gavage to Pah(enu2/enu2) mice followed by measurement of brain biopterin, phenylalanine, tyrosine, tryptophan and monoamine neurotransmitter content. A significant increase in brain biopterin content was detected only in mice that had received the highest sapropterin dose, 100mg/kg. Blood and brain phenylalanine concentrations were unchanged by sapropterin therapy. Sapropterin therapy also did not alter the absolute amounts of dopamine and serotonin in brain but was associated with increased homovanillic acid (HVA) and 5-hydroxyindoleacetic acid (5-HIAA), dopamine and serotonin metabolites respectively, in both wild type and Pah(enu2/enu2) mice. Oral sapropterin therapy likely does not directly affect central nervous system monoamine synthesis in either wild type or hyperphenylalaninemic mice but may stimulate synaptic neurotransmitter release and subsequent metabolism.


Subject(s)
Biopterins/analogs & derivatives , Brain/metabolism , Neurotransmitter Agents/metabolism , Phenylketonurias/drug therapy , Phenylketonurias/metabolism , Administration, Oral , Animals , Biopterins/administration & dosage , Biopterins/chemistry , Biopterins/therapeutic use , Disease Models, Animal , Dopamine/metabolism , Genotype , Homovanillic Acid/metabolism , Humans , Indoles/metabolism , Mice , Phenylalanine/blood , Serotonin/metabolism , Tryptophan Hydroxylase/metabolism , Tyrosine 3-Monooxygenase/metabolism
17.
J Biol Chem ; 291(2): 652-7, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26565027

ABSTRACT

Tryptophan metabolites in the kynurenine pathway are up-regulated by pro-inflammatory cytokines or glucocorticoids, and are linked to anti-inflammatory and immunosuppressive activities. In addition, they are up-regulated in pathologies such as cancer, autoimmune diseases, and psychiatric disorders. The molecular mechanisms of how kynurenine pathway metabolites cause these effects are incompletely understood. On the other hand, pro-inflammatory cytokines also up-regulate the amounts of tetrahydrobiopterin (BH4), an enzyme cofactor essential for the synthesis of several neurotransmitter and nitric oxide species. Here we show that xanthurenic acid is a potent inhibitor of sepiapterin reductase (SPR), the final enzyme in de novo BH4 synthesis. The crystal structure of xanthurenic acid bound to the active site of SPR reveals why among all kynurenine pathway metabolites xanthurenic acid is the most potent SPR inhibitor. Our findings suggest that increased xanthurenic acid levels resulting from up-regulation of the kynurenine pathway could attenuate BH4 biosynthesis and BH4-dependent enzymatic reactions, linking two major metabolic pathways known to be highly up-regulated in inflammation.


Subject(s)
Biopterins/analogs & derivatives , Kynurenine/metabolism , Metabolic Networks and Pathways , Xanthurenates/metabolism , Animals , Biopterins/biosynthesis , Biopterins/chemistry , Calorimetry , Crystallography, X-Ray , Humans , Inhibitory Concentration 50 , Kynurenine/chemistry , Mice , Models, Molecular , Rats , Surface Plasmon Resonance , Thermodynamics
18.
PLoS One ; 10(11): e0142854, 2015.
Article in English | MEDLINE | ID: mdl-26560496

ABSTRACT

Endothelial nitric oxide synthase (eNOS) is responsible for maintaining systemic blood pressure, vascular remodeling and angiogenesis. In addition to producing NO, eNOS can also generate superoxide (O2-.) in the absence of the cofactor tetrahydrobiopterin (BH4). Previous studies have shown that bovine eNOS serine 1179 (Serine 1177/human) phosphorylation critically modulates NO synthesis. However, the effect of serine 1179 phosphorylation on eNOS superoxide generation is unknown. Here, we used the phosphomimetic form of eNOS (S1179D) to determine the effect of S1179 phosphorylation on superoxide generating activity, and its sensitivity to regulation by BH4, Ca2+, and calmodulin (CAM). S1179D eNOS exhibited significantly increased superoxide generating activity and NADPH consumption compared to wild-type eNOS (WT eNOS). The superoxide generating activities of S1179D eNOS and WT eNOS did not differ significantly in their sensitivity to regulation by either Ca2+ or CaM. The sensitivity of the superoxide generating activity of S1179D eNOS to inhibition by BH4 was significantly reduced compared to WT eNOS. In eNOS-overexpressing 293 cells, BH4 depletion with 10mM DAHP for 48 hours followed by 50ng/ml VEGF for 30 min to phosphorylate eNOS S1179 increased ROS accumulation compared to DAHP-only treated cells. Meanwhile, MTT assay indicated that overexpression of eNOS in HEK293 cells decreased cellular viability compared to control cells at BH4 depletion condition (P<0.01). VEGF-mediated Serine 1179 phosphorylation further decreased the cellular viability in eNOS-overexpressing 293 cells (P<0.01). Our data demonstrate that eNOS serine 1179 phosphorylation, in addition to enhancing NO production, also profoundly affects superoxide generation: S1179 phosphorylation increases superoxide production while decreasing sensitivity to the inhibitory effect of BH4 on this activity.


Subject(s)
Nitric Oxide Synthase Type III/chemistry , Serine/chemistry , Superoxides/chemistry , Animals , Arginine/chemistry , Biopterins/analogs & derivatives , Biopterins/chemistry , Calcium/chemistry , Calmodulin/chemistry , Cattle , Cell Survival , Citrulline/chemistry , Electron Spin Resonance Spectroscopy , Endothelium, Vascular/metabolism , HEK293 Cells , Humans , Mutation , NADP/chemistry , Oxygen/chemistry , Phosphorylation , Reactive Oxygen Species/metabolism , Recombinant Proteins/chemistry , Spin Trapping , Vascular Endothelial Growth Factor A/metabolism
19.
Proc Natl Acad Sci U S A ; 112(37): 11648-53, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26297248

ABSTRACT

In the postischemic heart, coronary vasodilation is impaired due to loss of endothelial nitric oxide synthase (eNOS) function. Although the eNOS cofactor tetrahydrobiopterin (BH4) is depleted, its repletion only partially restores eNOS-mediated coronary vasodilation, indicating that other critical factors trigger endothelial dysfunction. Therefore, studies were performed to characterize the unidentified factor(s) that trigger endothelial dysfunction in the postischemic heart. We observed that depletion of the eNOS substrate NADPH occurs in the postischemic heart with near total depletion from the endothelium, triggering impaired eNOS function and limiting BH4 rescue through NADPH-dependent salvage pathways. In isolated rat hearts subjected to 30 min of ischemia and reperfusion (I/R), depletion of the NADP(H) pool occurred and was most marked in the endothelium, with >85% depletion. Repletion of NADPH after I/R increased NOS-dependent coronary flow well above that with BH4 alone. With combined NADPH and BH4 repletion, full restoration of NOS-dependent coronary flow occurred. Profound endothelial NADPH depletion was identified to be due to marked activation of the NAD(P)ase-activity of CD38 and could be prevented by inhibition or specific knockdown of this protein. Depletion of the NADPH precursor, NADP(+), coincided with formation of 2'-phospho-ADP ribose, a CD38-derived signaling molecule. Inhibition of CD38 prevented NADP(H) depletion and preserved endothelium-dependent relaxation and NO generation with increased recovery of contractile function and decreased infarction in the postischemic heart. Thus, CD38 activation is an important cause of postischemic endothelial dysfunction and presents a novel therapeutic target for prevention of this dysfunction in unstable coronary syndromes.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Endothelium, Vascular/metabolism , Ischemia/pathology , NADP/metabolism , Animals , Biopterins/analogs & derivatives , Biopterins/chemistry , Coronary Artery Disease/pathology , Electron Spin Resonance Spectroscopy , Endothelium, Vascular/pathology , Heart/physiology , Hypoxia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/chemistry , Nitric Oxide Synthase Type III/metabolism , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury
20.
Mol Endocrinol ; 29(8): 1123-33, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26121233

ABSTRACT

We have recently shown that angiotensin II-mediated uncoupling of endothelial nitric oxide synthase (eNOS) contributes to endothelial dysfunction in streptozotocin-induced type 1 diabetes mellitus. However, it has remained unclear whether and how eNOS uncoupling occurs in type 2 diabetes mellitus (T2DM) and the consequences of such in regulating vascular function. Here we investigated a role of bone morphogenic protein (BMP)-4 in mediating eNOS uncoupling, endothelial dysfunction, and inflammation in db/db mice. Circulating levels of BMP4 were markedly elevated in db/db mice but not in mice with type 1 diabetes mellitus, in which angiotensin II levels were significantly increased. Infusion of BMP4 antagonist noggin into db/db mice (15 µg/kg/day, 4 weeks) abolished eNOS uncoupling activity while restoring tetrahydrobiopterin (H(4)B) bioavailability. The impaired endothelium-dependent vasorelaxation in db/db aortas was significantly improved by noggin infusion. Exposure of aortic endothelial cells to BMP4 (50 ng/mL, 24 hours) resulted in eNOS uncoupling, which was attenuated by H(4)B precursor sepiapterin or small interfering RNA silencing nicotinamide adenine dinucleotide phosphate oxidase isoform 1 (NOX1). Interestingly, BMP4-dependent NOX1 up-regulation was abrogated by sepiapterin, implicating a NOX1-uncoupled eNOS-NOX1 feed-forward loop. BMP4 induction of cyclooxygenase 2 (COX2) expression and vascular cell adhesion protein 1 was found in db/db mice. Consistently, COX2 was up-regulated by BMP4 in endothelial cells, which was attenuated by sepiapterin, implicating an upstream role of eNOS uncoupling in COX2-mediated inflammatory activation. Taken together, our data for the first time reveal a novel role of BMP4 in inducing NOX1-dependent eNOS uncoupling in T2DM, which may promote development of novel therapeutics restoring endothelial function in T2DM.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Cyclooxygenase 2/metabolism , Diabetes Mellitus, Type 2/metabolism , Endothelium, Vascular/metabolism , NADH, NADPH Oxidoreductases/metabolism , Nitric Oxide Synthase Type III/metabolism , Angiotensin II/metabolism , Animals , Aorta/cytology , Aorta/pathology , Biopterins/analogs & derivatives , Biopterins/chemistry , Blood Glucose/chemistry , Carrier Proteins/metabolism , Cattle , Cells, Cultured , Cholesterol/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Inflammation/metabolism , Male , Mice , NADPH Oxidase 1 , RNA, Small Interfering/metabolism , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL