Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
J Biol Chem ; 300(6): 107381, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38762175

RESUMO

Enzymes are potent catalysts that increase biochemical reaction rates by several orders of magnitude. Flavoproteins are a class of enzymes whose classification relies on their ability to react with molecular oxygen (O2) during catalysis using ionizable active site residues. Pseudomonas aeruginosa D-arginine dehydrogenase (PaDADH) is a flavoprotein that oxidizes D-arginine for P. aeruginosa survival and biofilm formation. The crystal structure of PaDADH reveals the interaction of the glutamate 246 (E246) side chain with the substrate and at least three other active site residues, establishing a hydrogen bond network in the active site. Additionally, E246 likely ionizes to facilitate substrate binding during PaDADH catalysis. This study aimed to investigate how replacing the E246 residue with leucine affects PaDADH catalysis and its ability to react with O2 using steady-state kinetics coupled with pH profile studies. The data reveal a gain of O2 reactivity in the E246L variant, resulting in a reduced flavin semiquinone species and superoxide (O2•-) during substrate oxidation. The O2•- reacts with active site protons, resulting in an observed nonstoichiometric slope of 1.5 in the enzyme's log (kcat/Km) pH profile with D-arginine. Adding superoxide dismutase results in an observed correction of the slope to 1.0. This study demonstrates how O2•- can alter the slopes of limbs in the pH profiles of flavin-dependent enzymes and serves as a model for correcting nonstoichiometric slopes in elucidating reaction mechanisms of flavoproteins.


Assuntos
Aminoácido Oxirredutases , Domínio Catalítico , Oxigênio , Pseudomonas aeruginosa , Superóxidos , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/genética , Oxigênio/metabolismo , Oxigênio/química , Superóxidos/metabolismo , Superóxidos/química , Aminoácido Oxirredutases/química , Aminoácido Oxirredutases/metabolismo , Aminoácido Oxirredutases/genética , Prótons , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Cinética , Oxirredução , Mutação , Substituição de Aminoácidos , Arginina/química , Arginina/metabolismo
2.
J Biol Chem ; 299(3): 103007, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36775126

RESUMO

Pseudomonas aeruginosa couples the oxidation of d-2-hydroxyglutarate (D2HG) to l-serine biosynthesis for survival, using d-2-hydroxyglutarate dehydrogenase from P. aeruginosa (PaD2HGDH). Knockout of PaD2HGDH impedes P. aeruginosa growth, making PaD2HGDH a potential target for therapeutics. Previous studies showed that the enzyme's activity increased with Zn2+, Co2+, or Mn2+ but did not establish the enzyme's metal composition and whether the metal is an activator or a required cofactor for the enzyme, which we addressed in this study. Comparable to the human enzyme, PaD2HGDH showed only 15% flavin reduction with D2HG or d-malate. Upon purifying PaD2HGDH with 1 mM Zn2+, the Zn2+:protein stoichiometry was 2:1, yielding an enzyme with ∼40 s-1kcat for d-malate. Treatment with 1 mM EDTA decreased the Zn2+:protein ratio to 1:1 without changing the kinetic parameters with d-malate. We observed complete enzyme inactivation for the metalloapoenzyme with 100 mM EDTA treatment, suggesting that Zn2+ is essential for PaD2HGDH activity. The presence of Zn2+ increased the flavin N3 atom pKa value to 11.9, decreased the flavin ε450 at pH 7.4 from 13.5 to 11.8 mM-1 cm-1, and yielded a charged transfer complex with a broad absorbance band >550 nm, consistent with a Zn2+-hydrate species altering the electronic properties of the enzyme-bound FAD. The exogenous addition of Zn2+, Co2+, Cd2+, Mn2+, or Ni2+ to the metalloapoenzyme reactivated the enzyme in a sigmoidal pattern, consistent with an induced fit rapid-rearrangement mechanism. Collectively, our data demonstrate that PaD2HGDH is a Zn2+-dependent metallo flavoprotein, which requires Zn2+ as an essential cofactor for enzyme activity.


Assuntos
Malatos , Pseudomonas aeruginosa , Humanos , Pseudomonas aeruginosa/metabolismo , Ácido Edético , Oxirredução , Flavinas/metabolismo , Zinco , Cinética , Flavina-Adenina Dinucleotídeo/metabolismo
3.
J Biol Chem ; 299(3): 103008, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36775127

RESUMO

Pseudomonas aeruginosa PAO1 d-2-hydroxyglutarate (D2HG) dehydrogenase (PaD2HGDH) oxidizes D2HG to 2-ketoglutarate during the vital l-serine biosynthesis and is a potential therapeutic target against P. aeruginosa. PaD2HGDH, which oxidizes d-malate as an alternative substrate, has been demonstrated to be a metallo flavoprotein that requires Zn2+ for activity. However, the role of Zn2+ in the enzyme has not been elucidated, making it difficult to rationalize why nature employs both a redox center and a metal ion for catalysis in PaD2HGDH and other metallo flavoenzymes. In this study, recombinant His-tagged PaD2HGDH was purified to high levels in the presence of Zn2+ or Co2+ to investigate the metal's role in catalysis. We found that the flavin reduction step was reversible and partially rate limiting for the enzyme's turnover at pH 7.4 with either D2HG or d-malate with similar rate constants for both substrates, irrespective of whether Zn2+ or Co2+ was bound to the enzyme. The steady-state pL profiles of the kcat and kcat/Km values with d-malate demonstrate that Zn2+ mediates the activation of water coordinated to the metal. Our data are consistent with a dual role for the metal, which orients the hydroxy acid substrate in the enzyme's active site and rapidly deprotonates the substrate to yield an alkoxide species for hydride transfer to the flavin. Thus, we propose a catalytic mechanism for PaD2HGDH oxidation that establishes Zn2+ as a cofactor required for substrate orientation and activation during enzymatic turnover.


Assuntos
Malatos , Pseudomonas aeruginosa , Pseudomonas aeruginosa/metabolismo , Malatos/metabolismo , Oxirredução , Catálise , Flavoproteínas/metabolismo , Flavinas/metabolismo , Zinco/metabolismo , Cinética , Especificidade por Substrato
4.
J Biol Chem ; 299(4): 103044, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36803963

RESUMO

Enzymes require flexible regions to adopt multiple conformations during catalysis. The mobile regions of enzymes include gates that modulate the passage of molecules in and out of the enzyme's active site. The enzyme PA1024 from Pseudomonas aeruginosa PA01 is a recently discovered flavin-dependent NADH:quinone oxidoreductase (NQO, EC 1.6.5.9). Q80 in loop 3 (residues 75-86) of NQO is ∼15 Å away from the flavin and creates a gate that seals the active site through a hydrogen bond with Y261 upon NADH binding. In this study, we mutated Q80 to glycine, leucine, or glutamate to investigate the mechanistic significance of distal residue Q80 in NADH binding in the active site of NQO. The UV-visible absorption spectrum reveals that the mutation of Q80 minimally affects the protein microenvironment surrounding the flavin. The anaerobic reductive half-reaction of the NQO-mutants yields a ≥25-fold increase in the Kd value for NADH compared to the WT enzyme. However, we determined that the kred value was similar in the Q80G, Q80L, and wildtype enzymes and only ∼25% smaller in the Q80E enzyme. Steady-state kinetics with NQO-mutants and NQO-WT at varying concentrations of NADH and 1,4-benzoquinone establish a ≤5-fold decrease in the kcat/KNADH value. Moreover, there is no significant difference in the kcat/KBQ (∼1 × 106 M-1s-1) and kcat (∼24 s-1) values in NQO-mutants and NQO-WT. These results are consistent with the distal residue Q80 being mechanistically essential for NADH binding to NQO with minimal effect on the quinone binding to the enzyme and hydride transfer from NADH to flavin.


Assuntos
NAD(P)H Desidrogenase (Quinona) , NAD , Pseudomonas aeruginosa , Flavinas/metabolismo , Cinética , Mutação , NAD/metabolismo , Oxirredução , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/genética , NAD(P)H Desidrogenase (Quinona)/genética
5.
Biochemistry ; 62(5): 1070-1081, 2023 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-36795942

RESUMO

Numerous studies demonstrate that enzymes undergo multiple conformational changes during catalysis. The malleability of enzymes forms the basis for allosteric regulation: residues located far from the active site can exert long-range dynamical effects on the active site residues to modulate catalysis. The structure of Pseudomonas aeruginosa d-arginine dehydrogenase (PaDADH) shows four loops (L1, L2, L3, and L4) that span the substrate and the FAD-binding domains. Loop L4 comprises residues 329-336, spanning over the flavin cofactor. The I335 residue on loop L4 is ∼10 Šaway from the active site and ∼3.8 Šfrom N(1)-C(2)═O atoms of the flavin. In this study, we used molecular dynamics and biochemical techniques to investigate the effect of the mutation of I335 to histidine on the catalytic function of PaDADH. Molecular dynamics showed that the conformational dynamics of PaDADH are shifted to a more closed conformation in the I335H variant. In agreement with an enzyme that samples more in a closed conformation, the kinetic data of the I335H variant showed a 40-fold decrease in the rate constant of substrate association (k1), a 340-fold reduction in the rate constant of substrate dissociation from the enzyme-substrate complex (k2), and a 24-fold decrease in the rate constant of product release (k5), compared to that of the wild-type. Surprisingly, the kinetic data are consistent with the mutation having a negligible effect on the reactivity of the flavin. Altogether, the data indicate that the residue at position 335 has a long-range dynamical effect on the catalytic function in PaDADH.


Assuntos
Aminoácido Oxirredutases , Simulação de Dinâmica Molecular , Aminoácido Oxirredutases/química , Domínio Catalítico , Catálise , Flavinas/metabolismo , Cinética , Especificidade por Substrato , Sítios de Ligação , Conformação Proteica
6.
Arch Biochem Biophys ; 727: 109342, 2022 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-35777523

RESUMO

NAD(P)H:quinone oxidoreductases (NQOs) play an essential protective role as antioxidants in the detoxification of quinones in both Prokaryotes and Eukaryotes. NQO from Pseudomonas aeruginosa PAO1 uses FMN to catalyze the two-electron reduction of various quinones with NADH. In this study, steady-state kinetics, kinetic solvent viscosity effects, and rapid reaction kinetics were used to determine which kinetic steps control the overall turnover of the enzyme with benzoquinone or juglone. The rate constant for flavin reduction (kred) at pH 6.0 was 12.9 ± 0.3 s-1, and the Kd for NADH was at least an order of magnitude lower than 90 µM. With benzoquinone, the kcat value was 11.7 ± 0.3 s-1, consistent with flavin reduction being almost entirely rate-limiting for overall turnover. With juglone, a kcat value of 10.0 ± 0.5 s-1 was recorded. The normalized plot of the relative solvent viscosity effects on the kcat values established that hydride transfer from NADH to the FMN and quinol product release, with a calculated rate constant (kP-rel) of 52 s-1, are partially rate-limiting for the overall turnover of NQO. Kinetic solvent viscosity effects with glucose or sucrose revealed a hyperbolic dependence on the kcat and kcat/Km values with benzoquinone or juglone, respectively, consistent with the presence of a solvent-sensitive internal isomerization of the enzyme-substrate complex (ES). The data demonstrate opposing effects of benzoquinone and juglone on the equilibrium of the NQO ES isomerization with glucose or sucrose. Thus, our study demonstrates how quinol substrate properties alter the equilibrium of NQO ES isomerization.


Assuntos
NAD , Pseudomonas aeruginosa , Benzoquinonas , Mononucleotídeo de Flavina , Flavinas/metabolismo , Glucose , Hidroquinonas , Isomerismo , Cinética , NAD/metabolismo , Oxirredução , Quinonas , Solventes , Sacarose , Viscosidade
7.
Arch Biochem Biophys ; 715: 109100, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34864048

RESUMO

d-Arginine dehydrogenase from Pseudomonas aeruginosa (PaDADH) catalyzes the flavin-dependent oxidation of d-arginine and other d-amino acids. Here, we report the crystal structure at 1.29 Å resolution for PaDADH-Y249F expressed and co-crystallized with d-arginine. The overall structure of PaDADH-Y249F resembled PaDADH-WT, but the electron density for the flavin cofactor was ambiguous, suggesting the presence of modified flavins. Electron density maps and mass spectrometric analysis confirmed the presence of both N5-(4-guanidino-oxobutyl)-FAD and 6-OH-FAD in a single crystal of PaDADH-Y249F and helped with the further refinement of the X-ray crystal structure. The versatility of the reduced flavin is apparent in the PaDADH-Y249F structure and is evidenced by the multiple functions it can perform in the same active site.


Assuntos
Aminoácido Oxirredutases/química , Proteínas de Bactérias/química , Flavina-Adenina Dinucleotídeo/análogos & derivados , Guanidinas/química , Aminoácido Oxirredutases/genética , Aminoácido Oxirredutases/metabolismo , Arginina/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Flavina-Adenina Dinucleotídeo/química , Flavina-Adenina Dinucleotídeo/metabolismo , Guanidinas/metabolismo , Ligação de Hidrogênio , Mutação , Ligação Proteica , Pseudomonas aeruginosa/enzimologia , Eletricidade Estática
8.
Biochemistry ; 60(9): 711-724, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33630571

RESUMO

Proteins are inherently dynamic, and proper enzyme function relies on conformational flexibility. In this study, we demonstrated how an active site residue changes an enzyme's reactivity by modulating fluctuations between conformational states. Replacement of tyrosine 249 (Y249) with phenylalanine in the active site of the flavin-dependent d-arginine dehydrogenase yielded an enzyme with both an active yellow FAD (Y249F-y) and an inactive chemically modified green FAD, identified as 6-OH-FAD (Y249F-g) through various spectroscopic techniques. Structural investigation of Y249F-g and Y249F-y variants by comparison to the wild-type enzyme showed no differences in the overall protein structure and fold. A closer observation of the active site of the Y249F-y enzyme revealed an alternative conformation for some active site residues and the flavin cofactor. Molecular dynamics simulations probed the alternate conformations observed in the Y249F-y enzyme structure and showed that the enzyme variant with FAD samples a metastable conformational state, not available to the wild-type enzyme. Hybrid quantum/molecular mechanical calculations identified differences in flavin electronics between the wild type and the alternate conformation of the Y249F-y enzyme. The computational studies further indicated that the alternate conformation in the Y249F-y enzyme is responsible for the higher spin density at the C6 atom of flavin, which is consistent with the formation of 6-OH-FAD in the variant enzyme. The observations in this study are consistent with an alternate conformational space that results in fine-tuning the microenvironment around a versatile cofactor playing a critical role in enzyme function.


Assuntos
Aminoácido Oxirredutases/química , Aminoácido Oxirredutases/metabolismo , Flavinas/metabolismo , Fenilalanina/química , Mutação Puntual , Pseudomonas aeruginosa/enzimologia , Tirosina/química , Aminoácido Oxirredutases/genética , Sítios de Ligação , Catálise , Domínio Catalítico , Cinética , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Fenilalanina/genética , Fenilalanina/metabolismo , Conformação Proteica , Tirosina/genética , Tirosina/metabolismo
9.
Angew Chem Int Ed Engl ; 60(43): 23289-23298, 2021 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-34436811

RESUMO

Multi-scale calcium (Ca2+ ) dynamics, exhibiting wide-ranging temporal kinetics, constitutes a ubiquitous mode of signal transduction. We report a novel endoplasmic-reticulum (ER)-targeted Ca2+ indicator, R-CatchER, which showed superior kinetics in vitro (koff ≥2×103  s-1 , kon ≥7×106  M-1 s-1 ) and in multiple cell types. R-CatchER captured spatiotemporal ER Ca2+ dynamics in neurons and hotspots at dendritic branchpoints, enabled the first report of ER Ca2+ oscillations mediated by calcium sensing receptors (CaSRs), and revealed ER Ca2+ -based functional cooperativity of CaSR. We elucidate the mechanism of R-CatchER and propose a principle to rationally design genetically encoded Ca2+ indicators with a single Ca2+ -binding site and fast kinetics by tuning rapid fluorescent-protein dynamics and the electrostatic potential around the chromophore. The design principle is supported by the development of G-CatchER2, an upgrade of our previous (G-)CatchER with improved dynamic range. Our work may facilitate protein design, visualizing Ca2+ dynamics, and drug discovery.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/análise , Retículo Endoplasmático/metabolismo , Proteínas Luminescentes/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Proteínas de Ligação ao Cálcio/química , Células HEK293 , Células HeLa , Humanos , Proteínas Luminescentes/química , Camundongos , Simulação de Dinâmica Molecular , Ligação Proteica , Engenharia de Proteínas , Espectrometria de Fluorescência
10.
Biochemistry ; 59(51): 4833-4844, 2020 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-33301690

RESUMO

d-2-Hydroxyglutarate dehydrogenase from Pseudomonas aeruginosa PAO1 (PaD2HGDH) catalyzes the oxidation of d-2-hydroxyglutarate to 2-ketoglutarate, which is a necessary step in the serine biosynthetic pathway. The dependence of P. aeruginosa on PaD2HGDH makes the enzyme a potential therapeutic target against P. aeruginosa. In this study, recombinant His-tagged PaD2HGDH was expressed and purified to high levels from gene PA0317, which was previously annotated as an FAD-binding PCMH-type domain-containing protein. The enzyme cofactor was identified as FAD with fluorescence emission after phosphodiesterase treatment and with mass spectrometry analysis. PaD2HGDH had a kcat value of 11 s-1 and a Km value of 60 µM with d-2-hydroxyglutarate at pH 7.4 and 25 °C. The enzyme was also active with d-malate but did not react with molecular oxygen. Steady-state kinetics with d-malate and phenazine methosulfate as an electron acceptor established a mechanism that was consistent with ping-pong bi-bi steady-state kinetics at pH 7.4. A comparison of the kcat/Km values with d-2-hydroxyglutarate and d-malate suggested that the C5 carboxylate of d-2-hydroxyglutarate is important for the substrate specificity of the enzyme. Other homologues of the enzyme have been previously grouped in the VAO/PMCH family of flavoproteins. PaD2HGDH shares fully conserved residues with other α-hydroxy acid oxidizing enzymes, and these conserved residues are found in the active site of the PaD2HDGH homology model. An Enzyme Function Initiative-Enzyme Similarity Tool Sequence Similarity Network analysis suggests a functional difference between PaD2HGDH and human D2HGDH, and no relationship with VAO. A phylogenetic tree analysis of PaD2HGDH, VAO, and human D2HGDH establishes genetic diversity among these enzymes.


Assuntos
Oxirredutases do Álcool/química , Proteínas de Bactérias/química , Pseudomonas aeruginosa/enzimologia , Oxirredutases do Álcool/isolamento & purificação , Sequência de Aminoácidos , Proteínas de Bactérias/isolamento & purificação , Domínio Catalítico , Biologia Computacional , Flavina-Adenina Dinucleotídeo/química , Glutaratos/química , Cinética , Filogenia , Alinhamento de Sequência
11.
Arch Biochem Biophys ; 695: 108625, 2020 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-33038312

RESUMO

Nitronate monooxygenase from Neurospora crassa (NcNMO) is an FMN-dependent enzyme that oxidizes nitronates. The enzyme belongs to Group H of flavin monooxygenases. Previous biochemical and mechanistic studies on the enzyme showed that NcNMO oxidizes both anionic nitronates and neutral nitroalkanes, a feature distinguishing NcNMO from bacterial and other fungal NMOs which are active exclusively on nitronates. Recently, NMOs have been shown to oxidize propionate 3-nitronate (P3N), a toxic nitro acid present in legumes, fungi, and leaf beetles; P3N is an irreversible inhibitor of succinate dehydrogenase causing anywhere from neurological disorders to death in livestock and humans. In this study, we report the first kinetic investigation of NcNMO with P3N and its conjugated acid 3-nitropropionic acid (3NPA), and a mechanistic investigation with 3NPA using kinetic solvent viscosity effects. The kcat value with P3N (300 s-1) was 7-times larger than with 3NPA and the kcat/KP3N value (1,700,000 M-1s-1) was ~500-times larger than the kcat/K3NPA value, consistent with P3N being a faster and better substrate than 3NPA. The normalized kcat and kcat/K3NPA values showed inverse hyperbolic dependences on the relative solvent viscosity, consistent with an internal isomerization of the enzyme-substrate complex. A similar inverse hyperbolic pattern of the normalized kred value for the rate constant of flavin reduction determined anaerobically in a stopped-flow spectrophotometer suggested that the internal enzyme isomerization occurs before the flavin reduction step in the reductive half-reaction.


Assuntos
Oxigenases de Função Mista/química , Neurospora crassa/enzimologia , Nitrocompostos/química , Propionatos/química , Proteínas de Protozoários/química , Isoenzimas , Oxirredução , Solventes , Viscosidade
12.
Chem Rev ; 118(4): 1742-1769, 2018 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-29323892

RESUMO

Over time, organisms have evolved strategies to cope with the abundance of dioxygen on Earth. Oxygen-utilizing enzymes tightly control the reactions involving O2 mostly by modulating the reactivity of their cofactors. Flavins are extremely versatile cofactors that are capable of undergoing redox reactions by accepting either one electron or two electrons, alternating between the oxidized and the reduced states. The physical and chemical principles of flavin-based chemistry have been investigated widely. In the following pages we summarize the state of the art on a key area of research in flavin enzymology: the molecular basis for the activation of O2 by flavin-dependent oxidases and monooxygenases. In general terms, oxidases use O2 as an electron acceptor to produce H2O2, while monooxygenases activate O2 by forming a flavin intermediate and insert an oxygen atom into the substrate. First, we analyze how O2 reaches the flavin cofactor embedded in the protein matrix through dedicated access pathways. Then we approach O2 activation from the perspective of the monooxygenases, their preferred intermediate, the C(4a)-(hydro)peroxyflavin, and the cases in which other intermediates have been described. Finally, we focus on understanding how the architectures developed in the active sites of oxidases promote O2 activation and which other factors operate in its reactivity.


Assuntos
Enzimas/metabolismo , Flavinas/metabolismo , Oxigênio/metabolismo , Peróxido de Hidrogênio/metabolismo , Oxigenases de Função Mista/metabolismo , Especificidade por Substrato
13.
Biochemistry ; 58(22): 2594-2607, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31075192

RESUMO

PA0660 from Pseudomonas aeruginosa PAO1 is currently classified as a hypothetical nitronate monooxygenase (NMO), but no evidence at the transcript or protein level has been presented. In this study, PA0660 was purified and its biochemical and kinetic properties were characterized. Absorption spectroscopy and mass spectrometry demonstrated a tightly, noncovalently bound FMN in the active site of the enzyme. Analytical ultracentrifugation showed that the enzyme exists as a dimer in solution. Despite its annotation, PA0660 did not exhibit nitronate monooxygenase activity. The enzyme could be reduced with NADPH or NADH with a marked preference for NADPH, as indicated by ∼30-fold larger kcat/ Km and kred/ Kd values. Turnover could be sustained with NAD(P)H and quinones, DCPIP, and to a lesser extent molecular oxygen. However, PA0660 did not turn over with methyl red, consistent with a lack of azoreductase activity. The enzyme turned over through a ping-pong bi-bi steady-state kinetic mechanism with NADPH and 1,4-benzoquinone showing a kcat value of 90 s-1. The rate constant for flavin reduction with saturating NADPH was 360 s-1, whereas that for flavin oxidation with 1,4-benzoquinone was 270 s-1, consistent with both hydride transfers from the pyridine nucleotide to the flavin and from the flavin to 1,4-benzoquinone being partially rate-limiting for enzyme turnover. A BlastP search and a multiple-sequence alignment analysis of PA0660 highlighted the presence of six conserved motifs in >1000 open reading frames currently annotated as hypothetical NMOs. Our results suggest that PA0660 should be classified as an NAD(P)H:quinone reductase and serve as a paradigm enzyme for a new class of enzymes.


Assuntos
Flavoproteínas/química , Pseudomonas aeruginosa/enzimologia , Quinona Redutases/química , Sequência de Aminoácidos , Ensaios Enzimáticos , Escherichia coli/genética , Mononucleotídeo de Flavina/química , Flavoproteínas/genética , Flavoproteínas/isolamento & purificação , Cinética , NADP/química , Naftoquinonas/química , Oxirredução , Quinona Redutases/genética , Quinona Redutases/isolamento & purificação , Alinhamento de Sequência
14.
Chembiochem ; 20(13): 1646-1652, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30748074

RESUMO

Fluorescent cofactors like flavins can be exploited to probe their local environment with spatial and temporal resolution. Although the fluorescence properties of the oxidized and two-electron-reduced states of flavins have been studied extensively, this is not the case for the one-electron-reduced state. Both the neutral and anionic semiquinones have proven particularly challenging to examine, as they are unstable in solution and are transient, short-lived species in many catalytic cycles. Here, we report that the nitronate monooxygenase (NMO) from Pseudomonas aeruginosa PAO1 is capable of stabilizing both semiquinone forms anaerobically for hours, thus enabling us to study their spectroscopy in a constant protein environment. We found that in the active site of NMO, the anionic semiquinone exhibits no fluorescence, whereas the neutral semiquinone radical shows a relatively strong fluorescence, with a behavior that violates the Kasha-Vavilov rule. These fluorescence properties are discussed in the context of time-dependent density functional theory calculations, which reveal low-lying dark states in both systems.


Assuntos
Mononucleotídeo de Flavina/química , Flavina-Adenina Dinucleotídeo/análogos & derivados , Radicais Livres/química , Oxigenases de Função Mista/química , Teoria da Densidade Funcional , Flavina-Adenina Dinucleotídeo/química , Fluorescência , Modelos Químicos , Oxirredução , Pseudomonas aeruginosa/enzimologia
15.
Arch Biochem Biophys ; 672: 108058, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31356775

RESUMO

Propionate 3-nitronate (P3N) is a natural toxin that irreversibly inhibits mitochondrial succinate dehydrogenase. P3N poisoning leads to a variety of neurological disorders and even death. Nitronate monooxygenase (NMO) from Pseudomonas aeruginosa PAO1 was the first NMO characterized in bacteria and serves as a paradigm for Class I NMO. Here, we hypothesized that the carboxylate group of P3N might form a hydrogen bond with one or more of the four tyrosine or a lysine residues that are conserved in the active site of the enzyme. In the wild-type enzyme, the kcat value was pH independent between pH 6.0 and 11.0, while the kcat/KP3N value decreased at high pH, suggesting that a protonated group with a pKa value of 9.5 is required for binding the anionic substrate. A pH titration of the UV-visible absorption spectrum of the enzyme showed an increased absorbance at 297 nm with increasing pH, defining a pKa value of 9.5 and a Δε297 nm of 2.4 M-1cm-1, consistent with a tyrosine being important for substrate binding. The N3 atom of the oxidized flavin, instead, did not ionize likely because its pKa was perturbed by the ionization of a tyrosine in the active site of the enzyme. The Y109F, Y254F, Y299F, Y303F, and K307 M, substitutions had small effects (i.e., <3.5-fold) on the steady-state kinetic parameters of the enzyme. With all mutated enzymes, the kcat/KP3N value was less than 2.5-fold different from the wild-type enzyme, suggesting that none of the residues is solely essential for substrate binding.


Assuntos
Proteínas de Bactérias/química , Oxigenases de Função Mista/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Domínio Catalítico/genética , Ensaios Enzimáticos , Escherichia coli/genética , Concentração de Íons de Hidrogênio , Cinética , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Mutagênese Sítio-Dirigida , Nitrocompostos/metabolismo , Propionatos/metabolismo , Ligação Proteica , Pseudomonas aeruginosa/enzimologia , Tirosina/química
16.
Biochemistry ; 57(25): 3445-3453, 2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29874467

RESUMO

The study of enzyme reaction mechanisms is fundamentally important to our understanding of biochemistry, cellular metabolism, and drug development. This Perspective focuses on the use of kinetic solvent viscosity effects (KSVEs) to study enzyme reactions. This technique is easily implemented and uses steady-state kinetic analyses to probe whether substrate binding is diffusion-controlled and whether product release is the rate-limiting step in the catalytic cycle. In addition, KSVEs can identify isomerization steps that are important for catalysis. The use of KSVEs in combination with other techniques, such as kinetic isotope effects, pH effects, and site-directed mutagenesis, can provide a detailed view of the mechanism of enzyme action. We present the basic theory, important experimental considerations, and potential outcomes and briefly discuss some examples from the literature. The derivation of the equations that are important for data analysis is also presented.


Assuntos
Ensaios Enzimáticos/métodos , Algoritmos , Animais , Biocatálise , Difusão , Humanos , Isomerismo , Cinética , Modelos Moleculares , Solventes/química , Especificidade por Substrato , Viscosidade
17.
Biochemistry ; 57(21): 3050-3058, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29715013

RESUMO

The pa1225 gene of Pseudomonas aeruginosa strain PAO1 was cloned, and the resulting enzyme (PA1225) was purified and revealed to be an NADPH:quinone reductase. By using kinetics, fluorescence, and mass spectrometric analyses, PA1225 was shown to utilize FAD to transfer a hydride ion from NADPH to quinones. The enzyme could also use NADH, but with an efficiency that was 40-fold lower than that of NADPH as suggested by the kcat/ Km values at pH 6.0. Similar initial rates of reaction were determined with 1,4-benzoquinone and 2,6-dimethoxy-1,4-benzoquinone in the range between 25 and 200 µM, suggesting a low Km value for the quinone-oxidizing substrate. The lack of inhibition by NADP+ versus NADPH at saturating concentrations of 1,4-benzoquinone was consistent with a ping-pong bi-bi mechanism. The reductive half-reaction at pH 6.0 had Kd values of 0.07 mM with NADPH and 1.8 mM with NADH; the kred for flavin reduction was independent of pH with values of ∼10 s-1 with NADPH and ∼5 s-1 with NADH. Thus, the enzyme specificity for the reducing substrate arises primarily from a tighter binding of NADPH than of NADH. At pH 6.0, the kcat value with NADPH and 1,4-benzoquinone was 10.1 s-1, consistent with the hydride transfer from NADPH to FAD being fully rate limiting for the overall turnover of the enzyme. The enzyme showed negligible NADPH oxidase and azoreductase activities. This study enables annotation of the pa1225 gene as NADPH:quinone reductase, elucidates the enzymatic function of PA1225 in P. aeruginosa PAO1, and establishes that PA1225 is not an azoreductase as previously proposed.


Assuntos
NAD(P)H Desidrogenase (Quinona)/metabolismo , Pseudomonas aeruginosa/metabolismo , Benzoquinonas , Flavinas/metabolismo , Cinética , NAD/metabolismo , NAD(P)H Desidrogenase (Quinona)/química , NADP/metabolismo , Oxirredução , Oxirredutases/química , Oxirredutases/metabolismo , Pseudomonas aeruginosa/química , Quinonas/metabolismo
18.
Biochemistry ; 57(11): 1790-1797, 2018 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-29485859

RESUMO

The mechanism of dioxygen reduction by the flavoenzyme aryl-alcohol oxidase was investigated with kinetic isotope, viscosity, and pL (pH/pD) effects in rapid kinetics experiments by stopped-flow spectrophotometry of the oxidative half-reaction of the enzyme. Double mixing of the enzyme in a stopped-flow spectrophotometer with [α-2H2]- p-methoxybenzyl alcohol and oxygen at varying aging times established a slow rate constant of 0.0023 s-1 for the wash-out of the D atom from the N5 atom of the reduced flavin. Thus, the deuterated substrate could be used to probe the cleavage of the N-H bond of the reduced flavin in the oxidative half-reaction. A significant and pH-independent substrate kinetic isotope effect (KIE) of 1.5 between pH 5.0 and 8.0 demonstrated that H transfer is partially limiting the oxidative half-reaction of the enzyme; a negligible solvent KIE of 1.0 between pD 5.0 and 8.0 proved a fast H+ transfer reaction that does not contribute to determining the flavin oxidation rates. Thus, a mechanism for dioxygen reduction in which the H atom originating from the reduced flavin and a H+ from a solvent exchangeable site are transferred in separate kinetic steps is proposed. The spectroscopic and kinetic data presented also showed a lack of stabilization of transient flavin intermediates. The substantial differences in the mechanistic details of O2 reduction by aryl-alcohol oxidase with respect to other alcohol oxidases like choline oxidase, pyranose 2-oxidase, and glucose oxidase further demonstrate the high level of versatility of the flavin cofactor in flavoenzymes.


Assuntos
Oxirredutases do Álcool/química , Proteínas Fúngicas/química , Oxigênio/química , Pleurotus/enzimologia , Prótons , Ligação de Hidrogênio , Cinética , Oxirredução
19.
Biochemistry ; 57(40): 5818-5826, 2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30226367

RESUMO

Formate oxidase (FOX) was previously shown to contain a noncovalently bound 8-formyl FAD (8-fFAD) cofactor. However, both the absorption spectra and the kinetic parameters previously reported for FOX are inconsistent with more recent reports. The ultraviolet-visible (UV-vis) absorption spectrum reported in early studies closely resembles the spectra observed for protein-bound 8-formyl flavin semiquinone species, thus suggesting FOX may be photosensitive. Therefore, the properties of dark and light-exposed FOX were investigated using steady-state kinetics and site-directed mutagenesis analysis along with inductively coupled plasma optical emission spectroscopy, UV-vis absorption spectroscopy, circular dichroism spectroscopy, liquid chromatography and mass spectrometry, and electron paramagnetic resonance (EPR) spectroscopy. Surprisingly, these experimental results demonstrate that FOX is deactivated in the presence of light through generation of an oxygen stable, anionic (red) 8-fFAD semiquinone radical capable of persisting either in an aerobic environment for multiple weeks or in the presence of a strong reducing agent like sodium dithionite. Herein, we study the photoinduced formation of the 8-fFAD semiquinone radical in FOX and report the first EPR spectrum of this radical species. The stability of the 8-fFAD semiquinone radical suggests FOX to be a model enzyme for probing the structural and mechanistic features involved in stabilizing flavin semiquinone radicals. It is likely that the photoinduced formation of a stable 8-fFAD semiquinone radical is a defining characteristic of 8-formyl flavin-dependent enzymes. Additionally, a better understanding of the radical stabilization process may yield a FOX enzyme with more robust activity and broader industrial usefulness.


Assuntos
Aspergillus/enzimologia , Benzoquinonas/química , Flavina-Adenina Dinucleotídeo/análogos & derivados , Proteínas Fúngicas/química , Oxirredutases/química , Raios Ultravioleta , Aspergillus/genética , Flavina-Adenina Dinucleotídeo/química , Proteínas Fúngicas/genética , Mutagênese Sítio-Dirigida
20.
Proteins ; 86(5): 599-605, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29383742

RESUMO

Nitronate monooxygenase (NMO) is an FMN-dependent enzyme that oxidizes the neurotoxin propionate 3-nitronate (P3N) and represents the best-known system for P3N detoxification in different organisms. The crystal structure of the first eukaryotic Class I NMO from Cyberlindnera saturnus (CsNMO) has been solved at 1.65 Å resolution and refined to an R-factor of 14.0%. The three-dimensional structures of yeast CsNMO and bacterial PaNMO are highly conserved with the exception of three additional loops on the surface in the CsNMO enzyme and differences in four active sites residues. A PEG molecule was identified in the structure and formed extensive interactions with CsNMO, suggesting a specific binding site; however, 8% PEG showed no significant effect on the enzyme activity. This new crystal structure of a eukaryotic NMO provides insight into the function of this class of enzymes.


Assuntos
Proteínas Fúngicas/química , Oxigenases de Função Mista/química , Modelos Moleculares , Nitrocompostos/química , Propionatos/química , Saccharomycetales/enzimologia , Aminoácidos/química , Domínio Catalítico , Cinética , Oxirredução , Ligação Proteica , Estrutura Secundária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA