Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Nitric Oxide ; 89: 14-21, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31022534

RESUMEN

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.


Asunto(s)
Óxido Nítrico Sintasa/química , Biopterinas/análogos & derivados , Biopterinas/química , Citrulina/química , Inhibidores Enzimáticos/química , Hemo/química , Humanos , Imidazoles/química , NADP/química , Óxido Nítrico Sintasa/antagonistas & inhibidores , Nitroarginina/química , Oxígeno/química , Pichia/genética , Multimerización de Proteína
2.
Mol Pharmacol ; 93(2): 73-78, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29138269

RESUMEN

Belonging to the class of so-called soluble guanylate cyclase (sGC) activators, cinaciguat and BAY 60-2770 are interesting therapeutic tools for the treatment of various cardiovascular pathologies. The drugs are supposed to preferentially stimulate oxidized or heme-depleted, but not native sGC. Since this concept has been challenged by studies demonstrating complete relaxation of nondiseased vessels, this study was designed to reinvestigate the mode of action in greater detail. To this purpose, the effect of cinaciguat was studied on vessel tone of porcine coronary arteries and rat thoracic aortas. Organ bath studies showed that the compound caused time- and concentration-dependent relaxation of precontracted vessels with a maximal effect observed at 90 minutes. The dilatory response was not affected by extensive washout of the drug. Cinaciguat-induced vasodilation was associated with a time- and concentration-dependent increase of cGMP levels. Experiments with purified sGC in the presence of Tween 20 showed that cinaciguat activates the heme-free enzyme in a concentration-dependent manner with an EC50 value of ∼0.2 µM and maximal cGMP formation at 10 µM. By contrast, the effect of cinaciguat on 1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one-oxidized (ferric) sGC was moderate, reaching ∼10%-15% of maximal activity. Dilution experiments of cinaciguat/Tween 20-preincubated sGC revealed the irreversible character of the drug. Assuming a sensitive balance between heme-free, ferric, and nitric oxide-sensitive ferrous sGC in cells and tissues, we propose that cinaciguat by virtue of its irreversible mode of action is capable of shifting this equilibrium toward the heme-free apo-sGC species.


Asunto(s)
Benzoatos/farmacología , Inhibidores Enzimáticos/farmacología , Imitación Molecular , Protoporfirinas/metabolismo , Guanilil Ciclasa Soluble/antagonistas & inhibidores , Vasodilatación/efectos de los fármacos , Animales , Aorta Torácica/fisiología , Bovinos , Vasos Coronarios/metabolismo , GMP Cíclico/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Activación Enzimática , Estabilidad de Enzimas , Pulmón/efectos de los fármacos , Pulmón/enzimología , Protoporfirinas/química , Ratas Sprague-Dawley , Guanilil Ciclasa Soluble/metabolismo , Porcinos , Vasodilatadores/farmacología
3.
Mol Pharmacol ; 93(4): 335-343, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29358221

RESUMEN

According to current views, oxidation of aldehyde dehydrogenase-2 (ALDH2) during glyceryltrinitrate (GTN) biotransformation is essentially involved in vascular nitrate tolerance and explains the dependence of this reaction on added thiols. Using a novel fluorescent intracellular nitric oxide (NO) probe expressed in vascular smooth muscle cells (VSMCs), we observed ALDH2-catalyzed formation of NO from GTN in the presence of exogenously added dithiothreitol (DTT), whereas only a short burst of NO, corresponding to a single turnover of ALDH2, occurred in the absence of DTT. This short burst of NO associated with oxidation of the reactive C302 residue in the active site was followed by formation of low-nanomolar NO, even without added DTT, indicating slow recovery of ALDH2 activity by an endogenous reductant. In addition to the thiol-reversible oxidation of ALDH2, thiol-refractive inactivation was observed, particularly under high-turnover conditions. Organ bath experiments with rat aortas showed that relaxation by GTN lasted longer than that caused by the NO donor diethylamine/NONOate, in line with the long-lasting nanomolar NO generation from GTN observed in VSMCs. Our results suggest that an endogenous reductant with low efficiency allows sustained generation of GTN-derived NO in the low-nanomolar range that is sufficient for vascular relaxation. On a longer time scale, mechanism-based, thiol-refractive irreversible inactivation of ALDH2, and possibly depletion of the endogenous reductant, will render blood vessels tolerant to GTN. Accordingly, full reactivation of oxidized ALDH2 may not occur in vivo and may not be necessary to explain GTN-induced vasodilation.


Asunto(s)
Aldehído Deshidrogenasa Mitocondrial/metabolismo , Tolerancia a Medicamentos/fisiología , Músculo Liso Vascular/metabolismo , Nitratos/metabolismo , Óxido Nítrico/metabolismo , Nitroglicerina/metabolismo , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Línea Celular Transformada , Línea Celular Tumoral , Ditiotreitol/farmacología , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Nitratos/farmacología , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley
4.
J Biol Chem ; 290(41): 24932-44, 2015 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-26296888

RESUMEN

Citrulline formation by both human neuronal nitric-oxide synthase (nNOS) and mouse macrophage inducible NOS was inhibited by the hydrogen sulfide (H2S) donor Na2S with IC50 values of ∼2.4·10(-5) and ∼7.9·10(-5) m, respectively, whereas human endothelial NOS was hardly affected at all. Inhibition of nNOS was not affected by the concentrations of l-arginine (Arg), NADPH, FAD, FMN, tetrahydrobiopterin (BH4), and calmodulin, indicating that H2S does not interfere with substrate or cofactor binding. The IC50 decreased to ∼1.5·10(-5) m at pH 6.0 and increased to ∼8.3·10(-5) m at pH 8.0. Preincubation of concentrated nNOS with H2S under turnover conditions decreased activity after dilution by ∼70%, suggesting irreversible inhibition. However, when calmodulin was omitted during preincubation, activity was not affected, suggesting that irreversible inhibition requires both H2S and NO. Likewise, NADPH oxidation was inhibited with an IC50 of ∼1.9·10(-5) m in the presence of Arg and BH4 but exhibited much higher IC50 values (∼1.0-6.1·10(-4) m) when Arg and/or BH4 was omitted. Moreover, the relatively weak inhibition of nNOS by Na2S in the absence of Arg and/or BH4 was markedly potentiated by the NO donor 1-(hydroxy-NNO-azoxy)-l-proline, disodium salt (IC50 ∼ 1.3-2.0·10(-5) m). These results suggest that nNOS and inducible NOS but not endothelial NOS are irreversibly inhibited by H2S/NO at modest concentrations of H2S in a reaction that may allow feedback inhibition of NO production under conditions of excessive NO/H2S formation.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Sulfuro de Hidrógeno/farmacología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Óxido Nítrico/farmacología , Animales , Citrulina/biosíntesis , Interacciones Farmacológicas , Escherichia coli/citología , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Glutatión/farmacología , Humanos , Ratones , NADP/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Oxidación-Reducción/efectos de los fármacos , Compuestos de Sulfhidrilo/farmacología
5.
Biochemistry ; 53(8): 1284-95, 2014 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-24512289

RESUMEN

Recombinant neuronal nitric-oxide synthase (nNOS) expressed in baculovirus-infected Sf9 cells contains approximately 1 equiv of tightly bound tetrahydrobiopterin (BH4) per dimer and binds a second equivalent with a dissociation constant in the 10(-7)-10(-6) M range. Less is known about the pterin-binding properties of nNOS originating from expression systems such as Escherichia coli that do not produce BH4. We determined the binding properties of E. coli-expressed nNOS for BH4 and several inhibitory pterins by monitoring their effects on enzyme activity. E. coli-expressed nNOS as isolated was activated by BH4 monophasically with EC50 ≈ 2 × 10(-7) M, demonstrating a lack of tight pterin binding. However, overnight incubation with BH4 resulted in tight binding of one BH4 per dimer, yielding an enzyme that resembled Sf9-expressed nNOS. Tight pterin binding was also induced by preincubation with 4-amino-tetrahydrobiopterin, but not by 7,8-dihydrobiopterin or 4-amino-dihydrobiopterin, suggesting that tight-binding site formation requires preincubation with a fully reduced pteridine. Kinetic experiments showed that tight-binding site formation takes approximately 10 min with 1 µM BH4 (2 min with 1 µM 4-amino-BH4) at 4 °C. Anaerobic preincubation experiments demonstrated that O2 is not involved in the process. Gel electrophoretic studies suggest that tight-binding site formation is accompanied by an increase in the strength of the NOS dimer. We propose that incubation of pterin-free nNOS with BH4 creates one tight pterin-binding site per dimer, leaving the other site unaffected, in a reaction that involves redox chemistry.


Asunto(s)
Biopterinas/análogos & derivados , Óxido Nítrico Sintasa de Tipo I/metabolismo , Animales , Sitios de Unión , Biopterinas/metabolismo , Biopterinas/farmacología , Estabilidad de Enzimas , Cinética , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo I/química , Oxígeno/metabolismo , Unión Proteica/efectos de los fármacos , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Ratas , Células Sf9 , Spodoptera
6.
Mol Pharmacol ; 84(3): 407-14, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23793290

RESUMEN

Aldehyde dehydrogenase-2 (ALDH2) catalyzes vascular bioactivation of the antianginal drug nitroglycerin (GTN) to yield nitric oxide (NO) or a related species that activates soluble guanylate cyclase (sGC), resulting in cGMP-mediated vasodilation. Accordingly, established ALDH2 inhibitors attenuate GTN-induced vasorelaxation in vitro and in vivo. However, the ALDH2 hypothesis has not been reconciled with early studies demonstrating potent inhibition of the GTN response by diphenyleneiodonium (DPI), a widely used inhibitor of flavoproteins, in particular NADPH oxidases. We addressed this issue and investigated the effects of DPI on GTN-induced relaxation of rat aortic rings and the function of purified ALDH2. DPI (0.3 µM) inhibited the high affinity component of aortic relaxation to GTN without affecting the response to NO, indicating that the drug interfered with GTN bioactivation. Denitration and bioactivation of 1-2 µM GTN, assayed as 1,2-glycerol dinitrate formation and activation of purified sGC, respectively, were inhibited by DPI with a half-maximally active concentration of about 0.2 µM in a GTN-competitive manner. Molecular modeling indicated that DPI binds to the catalytic site of ALDH2, and this was confirmed by experiments showing substrate-competitive inhibition of the dehydrogenase and esterase activities of the enzyme. Our data identify ALDH2 as highly sensitive target of DPI and explain inhibition of GTN-induced relaxation by this drug observed previously. In addition, the data provide new evidence for the essential role of ALDH2 in GTN bioactivation and may have implications to other fields of ALDH2 research, such as hepatic ethanol metabolism and cardiac ischemia/reperfusion injury.


Asunto(s)
Aldehído Deshidrogenasa/antagonistas & inhibidores , Proteínas Mitocondriales/antagonistas & inhibidores , Nitroglicerina/metabolismo , Compuestos Onio/farmacología , Vasodilatadores/metabolismo , Aldehído Deshidrogenasa/química , Aldehído Deshidrogenasa Mitocondrial , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiología , Dominio Catalítico , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Femenino , Humanos , Técnicas In Vitro , Masculino , Simulación del Acoplamiento Molecular , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Unión Proteica , Ratas , Ratas Sprague-Dawley , Porcinos , Vasodilatación/efectos de los fármacos
7.
J Biol Chem ; 287(45): 38124-34, 2012 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-22988236

RESUMEN

Aldehyde dehydrogenase-2 (ALDH2) catalyzes the bioactivation of nitroglycerin (glyceryl trinitrate, GTN) in blood vessels, resulting in vasodilation by nitric oxide (NO) or a related species. Because the mechanism of this reaction is still unclear we determined the three-dimensional structures of wild-type (WT) ALDH2 and of a triple mutant of the protein that exhibits low denitration activity (E268Q/C301S/C303S) in complex with GTN. The structure of the triple mutant showed that GTN binds to the active site via polar contacts to the oxyanion hole and to residues 268 and 301 as well as by van der Waals interactions to hydrophobic residues of the catalytic pocket. The structure of the GTN-soaked wild-type protein revealed a thionitrate adduct to Cys-302 as the first reaction intermediate, which was also found by mass spectrometry (MS) experiments. In addition, the MS data identified sulfinic acid as the irreversibly inactivated enzyme species. Assuming that the structures of the triple mutant and wild-type ALDH2 reflect binding of GTN to the catalytic site and the first reaction step, respectively, superposition of the two structures indicates that denitration of GTN is initiated by nucleophilic attack of Cys-302 at one of the terminal nitrate groups, resulting in formation of the observed thionitrate intermediate and release of 1,2-glyceryl dinitrate. Our results shed light on the molecular mechanism of the GTN denitration reaction and provide useful information on the structural requirements for high affinity binding of organic nitrates to the catalytic site of ALDH2.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Vasos Sanguíneos/metabolismo , Proteínas Mutantes/metabolismo , Nitroglicerina/metabolismo , Aldehído Deshidrogenasa/química , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa Mitocondrial , Sustitución de Aminoácidos , Biocatálisis/efectos de los fármacos , Dominio Catalítico , Cristalografía por Rayos X , Electroforesis en Gel de Poliacrilamida , Humanos , Isoflavonas/química , Isoflavonas/farmacología , Espectrometría de Masas , Modelos Moleculares , Estructura Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Mutación , Nitratos/química , Nitratos/metabolismo , Nitroglicerina/química , Unión Proteica , Estructura Terciaria de Proteína , Compuestos de Azufre/química , Compuestos de Azufre/metabolismo
8.
Mol Pharmacol ; 82(3): 420-7, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22648973

RESUMEN

Tetrahydrobiopterin (BH4) is a major endogenous vasoprotective agent that improves endothelial function by increasing nitric oxide (NO) synthesis and scavenging of superoxide and peroxynitrite. Therefore, administration of BH4 is considered a promising therapy for cardiovascular diseases associated with endothelial dysfunction and oxidative stress. Here we report on a novel function of BH4 that might contribute to the beneficial vascular effects of the pteridine. Treatment of cultured porcine aortic endothelial cells with nitroglycerin (GTN) or 1H-[1,2,4]-oxadiazolo[4,3-a]quinoxaline-1-one (ODQ) resulted in heme oxidation of soluble guanylate cyclase (sGC), as evident from diminished NO-induced cGMP accumulation that was paralleled by increased cGMP response to a heme- and NO-independent activator of soluble guanylate cyclase [4-([(4-carboxybutyl)[2-(5-fluoro-2-([4'-(trifluoromethyl)biphenyl-4-yl]methoxy)phenyl)ethyl]amino]methyl)benzoic acid (BAY 60-2770)]. Whereas scavenging of superoxide and/or peroxynitrite with superoxide dismutase, tiron, Mn(III)tetrakis(4-benzoic acid)porphyrin, and urate had no protective effects, supplementation of the cells with BH4, either by application of BH4 directly or of its precursors dihydrobiopterin or sepiapterin, completely prevented the inhibition of NO-induced cGMP accumulation by GTN and ODQ. Tetrahydroneopterin had the same effect, and virtually identical results were obtained with RFL-6 fibroblasts, suggesting that our observation reflects a general feature of tetrahydropteridines that is unrelated to NO synthase function and not limited to endothelial cells. Protection of sGC against oxidative inactivation may contribute to the known beneficial effects of BH4 in cardiovascular disorders associated with oxidative stress.


Asunto(s)
Biopterinas/análogos & derivados , Guanilato Ciclasa/metabolismo , Estrés Oxidativo/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Biopterinas/farmacología , Enfermedades Cardiovasculares/metabolismo , Células Cultivadas , GMP Cíclico/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Hemo/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Nitroglicerina/farmacología , Oxidación-Reducción/efectos de los fármacos , Ácido Peroxinitroso/metabolismo , Pterinas/farmacología , Guanilil Ciclasa Soluble , Superóxidos/metabolismo , Porcinos
9.
Mol Pharmacol ; 80(2): 258-66, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21536753

RESUMEN

To elucidate the mechanism underlying reduction of nitroglycerin (GTN) to nitric oxide (NO) by mitochondrial aldehyde dehydrogenase (ALDH2), we generated mutants of the enzyme lacking the cysteines adjacent to reactive Cys302 (C301S and C303S), the glutamate that participates as a general base in aldehyde oxidation (E268Q) or combinations of these residues. The mutants were characterized regarding acetaldehyde dehydrogenation, GTN-triggered enzyme inactivation, GTN denitration, NO formation, and soluble guanylate cyclase activation. Lack of the cysteines did not affect dehydrogenase activity but impeded GTN denitration, aggravated GTN-induced enzyme inactivation, and increased NO formation. A triple mutant lacking the cysteines and Glu268 catalyzed sustained formation of superstoichiometric amounts of NO and exhibited slower rates of inactivation. These results suggest three alternative pathways for the reaction of ALDH2 with GTN, all involving formation of a thionitrate/sulfenyl nitrite intermediate at Cys302 as the initial step. In the first pathway, which predominates in the wild-type enzyme and reflects clearance-based GTN denitration, the thionitrate apparently reacts with one of the adjacent cysteine residues to yield nitrite and a protein disulfide. The predominant reaction catalyzed by the single and double cysteine mutants requires Glu268 and results in irreversible enzyme inactivation. Finally, combined lack of the cysteines and Glu268 shifts the reaction toward formation of the free NO radical, presumably through homolytic cleavage of the sulfenyl nitrite intermediate. Although the latter reaction accounts for less than 10% of total turnover of GTN metabolism catalyzed by wild-type ALDH2, it is most likely essential for vascular GTN bioactivation.


Asunto(s)
Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa/metabolismo , Mutagénesis Sitio-Dirigida/métodos , Nitroglicerina/metabolismo , Transducción de Señal/genética , Aldehído Deshidrogenasa/antagonistas & inhibidores , Aldehído Deshidrogenasa Mitocondrial , Animales , Biotransformación/genética , Bovinos , Silenciador del Gen , Humanos , Nitroglicerina/química , Nitrosación
10.
Mol Pharmacol ; 79(3): 541-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21156756

RESUMEN

Mitochondrial aldehyde dehydrogenase (ALDH2) contributes to vascular bioactivation of the antianginal drugs nitroglycerin (GTN) and pentaerythrityl tetranitrate (PETN), resulting in cGMP-mediated vasodilation. Although continuous treatment with GTN results in the loss of efficacy that is presumably caused by inactivation of ALDH2, PETN does not induce vascular tolerance. To clarify the mechanisms underlying the distinct pharmacological profiles of GTN and PETN, bioactivation of the nitrates was studied with aortas isolated from ALDH2-deficient and nitrate-tolerant mice, isolated mitochondria, and purified ALDH2. Pharmacological inhibition or gene deletion of ALDH2 attenuated vasodilation to both GTN and PETN to virtually the same degree as long-term treatment with GTN, whereas treatment with PETN did not cause tolerance. Purified ALDH2 catalyzed bioactivation of PETN, assayed as activation of soluble guanylate cyclase (sGC) and formation of nitric oxide (NO). The EC(50) value of PETN for sGC activation was 2.2 ± 0.5 µM. Denitration of PETN to pentaerythrityl trinitrate was catalyzed by ALDH2 with a specific activity of 9.6 ± 0.8 nmol · min(-1) · mg(-1) and a very low apparent affinity of 94.7 ± 7.4 µM. In contrast to GTN, PETN did not cause significant inactivation of ALDH2. Our data suggest that ALDH2 catalyzes bioconversion of PETN in two distinct reactions. Besides the major denitration pathway, which occurs only at high PETN concentrations, a minor high-affinity pathway may reflect vascular bioactivation of the nitrate yielding NO. The very low rate of ALDH2 inactivation, presumably as a result of low affinity of the denitration pathway, may at least partially explain why PETN does not induce vascular tolerance.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Proteínas Mitocondriales/metabolismo , Tetranitrato de Pentaeritritol/análogos & derivados , Aldehído Deshidrogenasa Mitocondrial , Animales , Aorta/efectos de los fármacos , Aorta/enzimología , Aorta/metabolismo , Relación Dosis-Respuesta a Droga , Guanilato Ciclasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/biosíntesis , Nitroglicerina/metabolismo , Nitroglicerina/farmacología , Tetranitrato de Pentaeritritol/metabolismo , Tetranitrato de Pentaeritritol/farmacología , Ratas , Ratas Sprague-Dawley , Vasodilatación/efectos de los fármacos
11.
J Biol Chem ; 285(2): 943-52, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-19906643

RESUMEN

The East Asian variant of mitochondrial aldehyde dehydrogenase (ALDH2) exhibits significantly reduced dehydrogenase, esterase, and nitroglycerin (GTN) denitrating activities. The small molecule Alda-1 was reported to partly restore low acetaldehyde dehydrogenase activity of this variant. In the present study we compared the wild type enzyme (ALDH2*1) with the Asian variant (ALDH2*2) regarding GTN bioactivation and the effects of Alda-1. Alda-1 increased acetaldehyde oxidation by ALDH2*1 and ALDH2*2 approximately 1.5- and 6-fold, respectively, and stimulated the esterase activities of both enzymes to similar extent as the coenzyme NAD. The effect of NAD was biphasic with pronounced inhibition occurring at > or = 5 mM. In the presence of 1 mM NAD, Alda-1 stimulated ALDH2*2-catalyzed ester hydrolysis 73-fold, whereas the NAD-stimulated activity of ALDH2*1 was inhibited because of 20-fold increased inhibitory potency of NAD in the presence of the drug. Although ALDH2*2 exhibited 7-fold lower GTN denitrating activity and GTN affinity than ALDH2*1, the rate of nitric oxide formation was only reduced 2-fold, and soluble guanylate cyclase (sGC) activation was more pronounced than with wild type ALDH2 at saturating GTN. Alda-1 caused slight inhibition of GTN denitration and did not increase GTN-induced sGC activation in the presence of either variant. The present results indicate that Alda-1 stimulates established ALDH2 activities by improving NAD binding but does not improve the GTN binding affinity of the Asian variant. In addition, our data revealed an unexpected discrepancy between GTN reductase activity and sGC activation, suggesting that GTN denitration and bioactivation may reflect independent pathways of ALDH2-catalyzed GTN biotransformation.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Benzamidas/farmacología , Benzodioxoles/farmacología , Variación Genética , Proteínas Mitocondriales/metabolismo , Nitroglicerina/farmacología , Vasodilatadores/farmacología , Acetaldehído/metabolismo , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa Mitocondrial , Animales , Benzamidas/metabolismo , Benzodioxoles/metabolismo , Bovinos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Asia Oriental , Guanilato Ciclasa/metabolismo , Humanos , Proteínas Mitocondriales/genética , NAD/metabolismo , Óxido Nítrico/biosíntesis , Nitroglicerina/metabolismo , Oxidación-Reducción/efectos de los fármacos , Vasodilatadores/metabolismo
12.
Sci Rep ; 10(1): 9846, 2020 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-32528086

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

13.
Biochim Biophys Acta ; 1784(5): 806-10, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18371313

RESUMEN

Isothermal titration calorimetry has been used to determine thermodynamic parameters of substrate binding to the oxygenase domain of neuronal nitric oxide synthase (nNOS(oxy)) in the presence of the cofactor tetrahydrobiopterin. The intermediate N(omega)-hydroxy-L-arginine (NHA) has a larger affinity than L-Arginine (L-Arg) for nNOS(oxy), with K(d)=0.4+/-0.1 microM and 1.7+/-0.3 microM at 25 degrees C, respectively. nNOS(oxy) binds NHA and L-Arg with DeltaH -4.1+/-0.2 and -1.0+/-0.1 kcal/mol and DeltaS=15 and 23 cal/Kmol respectively. NHA binding is more exothermic probably due to formation of an extra hydrogen bond in the active site compared to L-Arg. The changes in heat capacity (DeltaC(p)) are relatively small for binding of both NHA and L-Arg (-53+/-18 and -95+/-23 cal/L mol, respectively), which indicates that hydrophobic interactions contribute little to binding.


Asunto(s)
Arginina/análogos & derivados , Arginina/metabolismo , Entropía , Óxido Nítrico Sintasa/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Temperatura
14.
Sci Rep ; 9(1): 15403, 2019 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-31659183

RESUMEN

Murine 3T3-L1 adipocytes share many similarities with primary fat cells and represent a reliable in vitro model of adipogenesis. The aim of this study was to probe the effect of S-nitrosoglutathione (GSNO) on adipocyte differentiation. Adipogenesis was induced with a mixture of insulin, dexamethasone, and 3-isobutyl-1-methylxanthine in the absence and presence of increasing GSNO concentrations. Biochemical analysis after 7 days of differentiation showed a prominent anti-adipogenic effect of GSNO which was evident as reduced cellular triglycerides and total protein content as well as decreased mRNA and protein expression of late transcription factors (e.g. peroxisome proliferator activated receptor γ) and markers of terminal differentiation (e.g. leptin). By contrast, the nitrosothiol did not affect mRNA and protein expression of CCAAT/enhancer-binding protein ß (C/EBPß), which represents a pivotal early transcription factor of the adipogenic cascade. Differentiation was also inhibited by the NO donor (Z)-1-[2-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate. Biotin switch experiments showed significantly increased S-nitrosation of C/EBPß variants indicating that posttranslational S-nitrosative modification of this transcription factor accounts for the observed anti-adipogenic effect of NO. Our results suggest that S-nitrosation might represent an important physiological regulatory mechanism of fat cell maturation.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipogénesis , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Donantes de Óxido Nítrico/farmacología , S-Nitrosoglutatión/farmacología , 1-Metil-3-Isobutilxantina/farmacología , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Dexametasona/farmacología , Insulina/farmacología , Leptina/genética , Leptina/metabolismo , Ratones , PPAR gamma/genética , PPAR gamma/metabolismo
15.
Biochim Biophys Acta ; 1770(3): 432-45, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17014963

RESUMEN

Nitric-oxide synthase (NOS), the enzyme responsible for mammalian NO generation, is no cytochrome P450, but there are striking similarities between both enzymes. First and foremost, both are heme-thiolate proteins, employing the same prosthetic group to perform similar chemistry. Moreover, they share the same redox partner, a diflavoprotein reductase, which in the case of NOS is incorporated with the oxygenase in one polypeptide chain. There are, however, also conspicuous differences, such as the presence in NOS of the additional cofactor tetrahydrobiopterin, which is applied as an auxiliary electron donor to prevent decay of the oxyferrous complex to ferric heme and superoxide. In this review similarities and differences between NOS and cytochrome P450 are analyzed in an attempt to explain why NOS requires BH4 and why NO synthesis is not catalyzed by a member of the cytochrome P450 family.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Óxido Nítrico Sintasa/metabolismo , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Transporte Biológico , Sistema Enzimático del Citocromo P-450/química , Humanos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/química , Oxidación-Reducción , Protones
16.
Biochem Pharmacol ; 156: 168-176, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30099008

RESUMEN

Soluble guanylyl cyclase (sGC, EC 4.6.1.2) is a key enzyme in the regulation of vascular tone. In view of the therapeutic interest of the NO/cGMP pathway, drugs were developed that either increase the NO sensitivity of the enzyme or activate heme-free apo-sGC. However, modulation of sGC activity by endogenous agents is poorly understood. In the present study we show that the maximal activity of NO-stimulated purified sGC is significantly increased by cytosolic preparations of porcine coronary arteries. Purification of the active principle by several chromatographic steps resulted in a protein mixture consisting of 100, 70, and 40 kDa bands on SDS polyacrylamide gel electrophoresis. The respective proteins were identified by LC-MS/MS as gelsolin, annexin A6, and actin, respectively. Further purification resulted in loss of activity, indicating an interaction of sGC with a protein complex rather than a single protein. The partially purified preparation had no effect on basal sGC activity or enzyme activation by the heme mimetic BAY 60-2770, suggesting a specific effect on the conformation of the NO-bound heterodimeric holoenzyme. Since the three proteins identified are all related to contractile elements of smooth muscle, our data suggest that regulation of vascular tone involves a modulatory interaction of sGC with the cytoskeleton.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Músculo Liso Vascular/metabolismo , Óxido Nítrico/farmacología , Guanilil Ciclasa Soluble/metabolismo , Animales , Vasos Coronarios , Proteínas del Citoesqueleto/genética , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Guanilil Ciclasa Soluble/genética , Porcinos
17.
Biochim Biophys Acta ; 1764(3): 578-85, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16376159

RESUMEN

Nitric-oxide synthase (NOS) generates nitric oxide from l-arginine in two reaction cycles with N(omega)-hydroxy-l-arginine as an obligate intermediate. Although much progress has been made in recent years in the elucidation of the reaction mechanism of NOS, many questions remain to be answered. The use of low temperature has been instrumental in the revelation of the mechanism of NO synthesis, particularly regarding the role of the cofactor 5,6,7,8-tetrahydrobopterin (BH4). High-pressure studies may be expected to be similarly useful, but have been very few so far. In this short review, we depict the present state of knowledge about the reaction mechanism of NO synthesis, and the role(s) BH4 plays in it. This exposition is followed by a summary of the results obtained thus far in high-pressure studies and of the conclusions that can be drawn from them.


Asunto(s)
Biopterinas/análogos & derivados , Óxido Nítrico Sintasa/química , Óxido Nítrico/biosíntesis , Animales , Arginina/química , Biopterinas/química , Catálisis , Humanos , Presión , Temperatura
18.
Free Radic Biol Med ; 41(3): 455-63, 2006 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16843826

RESUMEN

Uncoupling of nitric-oxide synthase (NOS) by deficiency of the substrate L-arginine or the cofactor (6R)-5,6,7,8-tetrahydrobiopterin (BH4) is known to generate the reactive oxygen species H2O2 and superoxide. Discrimination between these two compounds is usually achieved by spin trapping of superoxide. We measured superoxide formation by uncoupled rat neuronal NOS, which contained one equivalent of tightly bound BH4 per dimer, using 5-(diethoxyphosphoryl)-5-methyl-1-pyrroline-N-oxide (DEPMPO) as a spin trap. As expected, the Ca2+-stimulated enzyme exhibited reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity that was accompanied by generation of superoxide and H2O2 in the absence of added L-arginine and BH4. Addition of BH4 (10 microM) did not significantly affect the rate of H2O2 formation but almost completely inhibited the apparent formation of superoxide, suggesting direct formation of H2O2. Although L-arginine (0.1 mM) increased the rate of NADPH oxidation about two-fold, the substrate largely attenuated apparent formation of both superoxide and H2O2, indicating that the spin trap did not efficiently outcompete the reaction between NO and superoxide. The efficiency of DEPMPO to scavenge superoxide in the presence of NO was studied by measuring free NO with a Clark-type electrode under conditions of NO/superoxide cogeneration. Neuronal NOS half-saturated with BH4 and the donor compound 3-morpholinosydnonimine (SIN-1) were used as enzymatic and nonenzymatic sources of NO/superoxide, respectively. Neither of the two systems gave rise to considerable NO signals in the presence of 50-100 mM DEPMPO, and even at 400 mM the spin trap uncovered less than 50% of the NO release that was detectable in the presence of 5000 U/ml superoxide dismutase. These results indicate that DEPMPO and all other currently available superoxide spin traps do not efficiently outcompete the reaction with NO. In addition, the similar behavior of nNOS and SIN-1 provides further evidence for NO as initial product of the NOS reaction.


Asunto(s)
Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Superóxidos/química , Superóxidos/metabolismo , Animales , Arginina , Biopterinas/análogos & derivados , Línea Celular , Electroquímica , Espectroscopía de Resonancia por Spin del Electrón , Unión Proteica , Pirroles , Ratas , Detección de Spin , Spodoptera , Superóxido Dismutasa/metabolismo
19.
Biochem J ; 390(Pt 2): 625-31, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15907191

RESUMEN

GTN (nitroglycerin; glycerol trinitrate) causes dilation of blood vessels via activation of nitric oxide (NO)-sensitive sGC (soluble guanylate cyclase), a heterodimeric haem protein that catalyses the conversion of GTP into cGMP. Activation of sGC by GTN requires enzymatic or non-enzymatic bioactivation of the nitrate. Based on insufficient NO release and lack of spectroscopic evidence for formation of NO-sGC, the cysteine (Cys)-dependent activation of sGC by GTN was proposed to occur in an NO-independent manner. This extraordinary claim is questioned by the present findings. First, the effect of GTN/Cys was blocked by the NO scavenger oxyhaemoglobin, the superoxide-generating compound flavin mononucleotide and the haem-site sGC inhibitor ODQ (1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one). Secondly, at equi-effective concentrations, GTN/Cys and the NO donor 2,2-diethyl-1-nitroso-oxyhydrazine released identical amounts of NO. Finally, at sufficiently high rates of NO release, activation of sGC by GTN/Cys was accompanied by a shift of the Soret band from 431 to 399 nm, indicating formation of NO-sGC. In the absence of Cys, GTN caused haem oxidation, apparent as a shift of the Soret band to 392 nm, which was accompanied by inactivation of the NO-stimulated enzyme. These results suggest that the effect of GTN/Cys is the result of an activation/inactivation equilibrium that is controlled by the rate of NO release and haem oxidation.


Asunto(s)
Cisteína/farmacología , Hemo/metabolismo , Óxido Nítrico/metabolismo , Nitroglicerina/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Bovinos , Cisteína/metabolismo , Activación Enzimática/efectos de los fármacos , Guanilato Ciclasa , Hidrazinas/farmacología , Luz , Pulmón/enzimología , Óxidos de Nitrógeno/farmacología , Nitroglicerina/metabolismo , Oxidación-Reducción/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Guanilil Ciclasa Soluble
20.
Methods Enzymol ; 396: 456-66, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16291253

RESUMEN

The role of tetrahydrobiopterin (BH4) as a cofactor in nitric oxide synthase (NOS) has been the object of intense research in the last few years. It was found that in addition to its established effects on the NOS heme spin state, substrate affinity, and enzyme dimerization, BH4 is required as a one-electron donor to oxyferrous [Fe(II).O2] heme that is formed as an intermediate in the catalytic cycle. Cryogenic spectroscopic techniques proved particularly useful in the identification of this role of BH4 in NO synthesis. With these methods, the mechanism of fast reactions, such as the reaction of ferrous NOS with O2, can be unraveled by lowering the reaction temperature to subzero values. This may not only reduce the rate to such an extent that the reaction can be followed on a time scale from seconds to minutes, but intermediates may be observed that do not accumulate at higher temperatures. Cryogenic ultraviolet-visible (UV-vis) and electron paramagnetic resonance spectroscopy have been applied to clarify why the BH4 analogue 4-amino-tetrahydrobiopterin (4-amino-BH4) is unable to support NO synthesis. In the course of these studies, evidence was gathered supporting a role for BH4 as an obligate proton and electron donor. It is believed that the inhibitory action of 4-amino-BH4 derives from an inability to serve as a proton donor, even though it is perfectly able to serve as an electron donor. In this chapter, the suitability, drawbacks, and advantages of cryogenic methods are discussed.


Asunto(s)
Biopterinas/análogos & derivados , Espectroscopía de Resonancia por Spin del Electrón/métodos , Óxido Nítrico/fisiología , Espectrofotometría Ultravioleta/métodos , Biopterinas/fisiología , Electrones , Cinética , Protones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA