Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Org Biomol Chem ; 22(32): 6550-6560, 2024 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-39081262

RESUMEN

Kynurenine 3-monooxygenase (KMO) regulates the levels of important physiological intermediates in the kynurenine pathway [Guillemin, et al., Journal of Neuroscience, 2007, 27, 12884], which is the major route for L-tryptophan catabolism. Its catalytic activity (hydroxylation) is dependent on the formation of a short-lived intermediate that forms after the reduction of the coenzyme FAD. The reduction takes place fast when the substrate binds to KMO. Crystal structures of the apo form and in complex with an effector inhibitor, which prevents the hydroxylation of the substrate but also stimulates KMO like the substrate, and a competitive inhibitor, which suppresses the substrate hydroxylation, are available for the resting in conformation only. The active out conformational state that enables the reduction of FAD at an exposed location of KMO after its stimulation by an effector, however, was implicated but not resolved experimentally and has remained elusive so far. Molecular dynamics simulations of apo KMO and the inhibitor-KMO complexes are carried out using extensive multi-dimensional umbrella sampling to explore the free-energy surface of the coenzyme FAD's conformational conversion from the in state (buried within the active site) to the out state. This allows a discussion and comparison with the experimental results, which showed a significant increase in the rate of reduction of FAD in the presence of an effector inhibitor and absence of enzymatic function in the presence of a competitive inhibitor [Kim, et al., Cell Chemical Biology, 2018, 25, 426]. The free-energy barriers associated with those conformational changes and structural models for the active out conformation are obtained. The interactions during the conformational changes are determined to identify the influence of the effector.


Asunto(s)
Quinurenina 3-Monooxigenasa , Simulación de Dinámica Molecular , Conformación Proteica , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Quinurenina 3-Monooxigenasa/metabolismo , Quinurenina 3-Monooxigenasa/química , Modelos Moleculares , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos
2.
J Cell Physiol ; 237(12): 4339-4355, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36088660

RESUMEN

Kynurenine-3-monooxygenase (KMO) is a mitochondrial enzyme involved in the eukaryotic kynurenine pathway (KP), which is the major catabolic route of tryptophan. KMO can convert the substrate kynurenine into the neurotoxin 3-hydroxykynurenine and quinolinic acid, which promote the production of toxic metabolites and formation of free radical in the blood, while decrease the neuroprotective metabolite kynurenic acid. As a result of branch point, KMO is predicted as an attractive drug target for several diseases, especially neurodegenerative diseases, psychosis, and cancer. This review mainly pays attention to KMO structure and the research of mechanisms and functions, with a particular emphasis on the roles of KMO in the pathogenesis of various conditions. Furthermore, we also summarized important KMO inhibitors to supporting their effects on these diseases, indicating the prospect to find novel KMO inhibitors for diseases therapy.


Asunto(s)
Quinurenina 3-Monooxigenasa , Enfermedades Neurodegenerativas , Humanos , Progresión de la Enfermedad , Ácido Quinurénico/metabolismo , Quinurenina/metabolismo , Quinurenina 3-Monooxigenasa/química , Quinurenina 3-Monooxigenasa/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Triptófano/metabolismo
3.
Molecules ; 27(1)2022 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-35011505

RESUMEN

Under normal physiological conditions, the kynurenine pathway (KP) plays a critical role in generating cellular energy and catabolizing tryptophan. Under inflammatory conditions, however, there is an upregulation of the KP enzymes, particularly kynurenine 3-monooxygenase (KMO). KMO has garnered much attention due to its production of toxic metabolites that have been implicated in many diseases and disorders. With many of these illnesses having an inadequate or modest treatment, there exists a need to develop KMO inhibitors that reduce the production of these toxic metabolites. Though prior efforts to find an appropriate KMO inhibitor were unpromising, the development of a KMO crystal structure has provided the opportunity for a rational structure-based design in the development of inhibitors. Therefore, the purpose of this review is to describe the kynurenine pathway, the kynurenine 3-monooxygenase enzyme, and KMO inhibitors and their potential candidacy for clinical use.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Quinurenina 3-Monooxigenasa , Quinurenina , Animales , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/uso terapéutico , Humanos , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Quinurenina/química , Quinurenina/metabolismo , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Quinurenina 3-Monooxigenasa/biosíntesis , Quinurenina 3-Monooxigenasa/química , Relación Estructura-Actividad
4.
J Phys Chem A ; 125(43): 9459-9477, 2021 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-34676771

RESUMEN

Recently, two new mechanistic proposals for the kynurenine 3-monooxygenase (KMO) catalyzed hydroxylation reaction of l-Kynurenine (l-Kyn) have been proposed. According to the first proposal, instead of the distal oxygen, the proximal oxygen of the hydroperoxide intermediate of flavin adenine dinucleotide (FAD) is transferred to the substrate ring. The second study proposes that l-Kyn participates in its base form in the reaction. To address these proposals, the reaction was reconsidered with a 386 atom quantum cluster model that is based on a recent X-ray structure (PDB id: 6FOX). The computations were carried out at the UB3LYP/6-311+G(2d,2p)//UB3LYP/6-31G(d,p) level with solvation (polarizable continuum model) and dispersion (DFT-D3(BJ)) corrections. To supplement the results of the density functional theory (DFT) calculations, molecular dynamics (MD) simulations of the protein-substrate complex were employed. The comparison of a proximal oxygen transfer mechanism to the distal oxygen transfer mechanism revealed that the former requires too high of a barrier energy while the latter validated our previous results. According to the MD simulations, the hydroperoxy moiety does not favor an alignment that might promote the proximal oxygen transfer mechanism. In the second part of the study, hydroxylation reaction with the base form of l-Kyn was sought. Although DFT calculations confirmed a much more facile reaction with the base form of l-Kyn, a mechanism which would allow the deprotonation of the l-Kyn before the oxygen transfer could not be determined with the X-ray-based positions. A concerted mechanism with both the oxygen transfer and the deprotonation required a high barrier energy. A stepwise mechanism involving the deprotonation of l-Kyn was found, starting from an MD frame. The overall barrier of the oxygen transfer step of this model was found to be in the range of that of with neutral l-Kyn. MD simulations supported the idea of ineffectiveness of the nearby shell surrounding the utilized active site core on the deprotonation of l-Kyn.


Asunto(s)
Quinurenina 3-Monooxigenasa/química , Quinurenina 3-Monooxigenasa/metabolismo , Simulación de Dinámica Molecular , Dominio Catalítico , Hidroxilación , Quinurenina/química , Quinurenina/metabolismo , Modelos Moleculares , Estructura Molecular , Oxidación-Reducción , Conformación Proteica
5.
Molecules ; 26(11)2021 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-34073016

RESUMEN

The pathogenesis of several neurodegenerative diseases such as Alzheimer's or Huntington's disease has been associated with metabolic dysfunctions caused by imbalances in the brain and cerebral spinal fluid levels of neuroactive metabolites. Kynurenine monooxygenase (KMO) is considered an ideal therapeutic target for the regulation of neuroactive tryptophan metabolites. Despite significant efforts, the known KMO inhibitors lack blood-brain barrier (BBB) permeability and upon the mimicking of the substrate binding mode, are subject to produce reactive oxygen species as a side reaction. The computational drug design is further complicated by the absence of complete crystal structure information for human KMO (hKMO). In the current work, we performed virtual screening of readily available compounds using several protein-ligand complex pharmacophores. Each of the pharmacophores accounts for one of three distinct reported KMO protein-inhibitor binding conformations. As a result, six novel KMO inhibitors were discovered based on an in vitro fluorescence assay. Compounds VS1 and VS6 were predicted to be BBB permeable and avoid the hydrogen peroxide production dilemma, making them valuable, novel hit compounds for further drug property optimization and advancement in the drug design pipeline.


Asunto(s)
Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Quinurenina 3-Monooxigenasa/metabolismo , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Biología Computacional/métodos , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/farmacología , Humanos , Quinurenina/metabolismo , Quinurenina 3-Monooxigenasa/química , Simulación del Acoplamiento Molecular/métodos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Conformación Proteica
6.
Arch Biochem Biophys ; 690: 108474, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32687799

RESUMEN

Kynurenine 3-monoxygenase (KMO) catalyzes the conversion of l-kynurenine (L-Kyn) to 3-hydroxykynurenine (3-OHKyn) in the pathway for tryptophan catabolism. We have investigated the effects of pH and deuterium substitution on the oxidative half-reaction of KMO from P. fluorescens (PfKMO). The three phases observed during the oxidative half reaction are formation of the hydroperoxyflavin, hydroxylation and product release. The measured rate constants for these phases proved largely unchanging with pH, suggesting that the KMO active site is insulated from exchange with solvent during catalysis. A solvent inventory study indicated that a solvent isotope effect of 2-3 is observed for the hydroxylation phase and that two or more protons are in flight during this step. An inverse isotope effect of 0.84 ± 0.01 on the rate constant for the hydroxylation step with ring perdeutero-L-Kyn as a substrate indicates a shift from sp2 to sp3 hybridization in the transition state leading to the formation of a non-aromatic intermediate. The pH dependence of transient state data collected for the substrate analog meta-nitrobenzoylalanine indicate that groups proximal to the hydroperoxyflavin are titrated in the range pH 5-8.5 and can be described by a pKa of 8.8. That higher pH values do not slow the rate of hydroxylation precludes that the pKa measured pertains to the proton of the hydroperoxflavin. Together, these observations indicate that the C4a-hydroperoxyflavin has a pKa ≫ 8.5, that a non-aromatic species is the immediate product of hydroxylation and that at least two solvent derived protons are in-flight during oxygen insertion to the substrate aromatic ring. A unifying mechanistic proposal for these observations is proposed.


Asunto(s)
Hidrógeno/química , Quinurenina 3-Monooxigenasa/química , Quinurenina 3-Monooxigenasa/metabolismo , Quinurenina/química , Pseudomonas fluorescens/química , Catálisis , Dominio Catalítico , Deuterio/química , Dinitrocresoles/metabolismo , Flavinas/química , Concentración de Iones de Hidrógeno , Hidroxilación , Cinética , Oxigenasas de Función Mixta/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Oxígeno/química , Protones , Solventes/química
7.
FASEB J ; 32(4): 2036-2045, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29208702

RESUMEN

The human kynurenine 3-monooxygenase (hKMO) is a potential therapeutic target for neurodegenerative and neurologic disorders. Inhibition of KMO by Ro 61-8048, a potent, selective, and the most widely used inhibitor of KMO, was shown effective in various models of neurodegenerative or neurologic disorders. However, the molecular basis of hKMO inhibition by Ro 61-8048 is not clearly understood. Here, we report biochemistry studies on hKMO and crystal structures of an hKMO homolog, pfKMO from Pseudomonas fluorescens, in complex with the substrate l-kynurenine and Ro 61-8048. We found that the C-terminal ∼110 aa are essential for the enzymatic activity of hKMO and the homologous C-terminal region of pfKMO folds into a distinct, all-α-helical domain, which associates with the N-terminal catalytic domain to form a unique tunnel in proximity to the substrate-binding pocket. The tunnel binds the Ro 61-8048 molecule, which fills most of the tunnel, and Ro 61-8048 is hydrogen bonded with several completely conserved residues, including an essential catalytic residue. Modification of Ro 61-8048 and biochemical studies of the modified Ro 61-8048 derivatives suggested that Ro 61-8048 inhibits the enzyme in an allosteric manner by affecting the conformation of the essential catalytic residue and by blocking entry of the substrate or product release. The unique binding sites distinguish Ro 61-8048 as a noncompetitive and highly selective inhibitor from other competitive inhibitors, which should facilitate further optimization of Ro 61-8048 and the development of new inhibitory drugs to hKMO.-Gao, J., Yao, L., Xia, T., Liao, X., Zhu, D., Xiang, Y. Biochemistry and structural studies of kynurenine 3-monooxygenase reveal allosteric inhibition by Ro 61-8048.


Asunto(s)
Sitio Alostérico , Proteínas Bacterianas/química , Inhibidores Enzimáticos/farmacología , Quinurenina 3-Monooxigenasa/química , Sulfonamidas/farmacología , Tiazoles/farmacología , Regulación Alostérica , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/metabolismo , Inhibidores Enzimáticos/química , Células HEK293 , Humanos , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Quinurenina 3-Monooxigenasa/metabolismo , Unión Proteica , Pseudomonas fluorescens/enzimología , Sulfonamidas/química , Tiazoles/química
8.
J Phys Chem A ; 123(14): 3149-3159, 2019 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-30888816

RESUMEN

The mechanism of the hydroxylation reaction between l-Kyn and model flavin adenine dinucleotide (FAD)-hydroperoxide was investigated via density functional theory (DFT) calculations in the absence and in the presence of the kynurenine 3-monooxygenase (KMO) enzyme by considering possible pathways that can lead to the product 3-hydroxykynurenine (3-HK). Crystal structure (pdb code: 5NAK )-based calculations involved a quantum cluster model in which the active site of the enzyme with the substrate l-Kyn was represented with 348 atoms. According to the deduced mechanism, KMO-catalyzed hydroxylation reaction takes place with four transformations. In the initial transition state, FAD delivers its peroxy hydroxyl to the l-Kyn ring, creating an sp3-hybridized carbon center. Then, the hydrogen on the hydroxyl moiety is immediately transferred back to the proximal oxygen that remained on FAD. These consequent transformations are in line with the somersault rearrangement previously described for similar enzymatic systems. The second step corresponds to a hydride shift from the sp3-hybridized carbon of the substrate ring to its adjacent carbon, producing the keto form of 3-HK. Then, keto-3-HK is transformed into its enol form (3-HK) with a water-assisted tautomerization. Lastly, FAD is oxidized with a water-assisted dehydration, which also involves 3-HK as a catalyst. In the proposed pathway, Asn54, Pro318, and a crystal water molecule were seen to play significant roles in the proton relays. The energies obtained via the cluster approach were calculated at the B3LYP/6-311+G(2d,2p)//B3LYP/6-31G(d,p) level with solvation (polarizable continuum model) and dispersion (DFT-D3(BJ)) corrections.


Asunto(s)
Biocatálisis , Teoría Funcional de la Densidad , Quinurenina 3-Monooxigenasa/química , Quinurenina 3-Monooxigenasa/metabolismo , Dominio Catalítico , Hidroxilación , Modelos Moleculares
9.
Nature ; 496(7445): 382-5, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23575632

RESUMEN

Inhibition of kynurenine 3-monooxygenase (KMO), an enzyme in the eukaryotic tryptophan catabolic pathway (that is, kynurenine pathway), leads to amelioration of Huntington's-disease-relevant phenotypes in yeast, fruitfly and mouse models, as well as in a mouse model of Alzheimer's disease. KMO is a flavin adenine dinucleotide (FAD)-dependent monooxygenase and is located in the outer mitochondrial membrane where it converts l-kynurenine to 3-hydroxykynurenine. Perturbations in the levels of kynurenine pathway metabolites have been linked to the pathogenesis of a spectrum of brain disorders, as well as cancer and several peripheral inflammatory conditions. Despite the importance of KMO as a target for neurodegenerative disease, the molecular basis of KMO inhibition by available lead compounds has remained unknown. Here we report the first crystal structure of Saccharomyces cerevisiae KMO, in the free form and in complex with the tight-binding inhibitor UPF 648. UPF 648 binds close to the FAD cofactor and perturbs the local active-site structure, preventing productive binding of the substrate l-kynurenine. Functional assays and targeted mutagenesis reveal that the active-site architecture and UPF 648 binding are essentially identical in human KMO, validating the yeast KMO-UPF 648 structure as a template for structure-based drug design. This will inform the search for new KMO inhibitors that are able to cross the blood-brain barrier in targeted therapies against neurodegenerative diseases such as Huntington's, Alzheimer's and Parkinson's diseases.


Asunto(s)
Ciclopropanos/química , Ciclopropanos/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Quinurenina 3-Monooxigenasa/química , Saccharomyces cerevisiae/enzimología , Arginina/metabolismo , Barrera Hematoencefálica/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/enzimología , Quinurenina/metabolismo , Quinurenina 3-Monooxigenasa/metabolismo , Modelos Moleculares , Terapia Molecular Dirigida , Conformación Proteica , Reproducibilidad de los Resultados , Relación Estructura-Actividad
10.
Protein Expr Purif ; 95: 96-103, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24316190

RESUMEN

Kynurenine 3-monooxygenase (KMO) is an enzyme central to the kynurenine pathway of tryptophan metabolism. KMO has been implicated as a therapeutic target in several disease states, including Huntington's disease. Recombinant human KMO protein production is challenging due to the presence of transmembrane domains, which localise KMO to the outer mitochondrial membrane and render KMO insoluble in many in vitro expression systems. Efficient bacterial expression of human KMO would accelerate drug development of KMO inhibitors but until now this has not been achieved. Here we report the first successful bacterial (Escherichia coli) expression of active FLAG™-tagged human KMO enzyme expressed in the soluble fraction and progress towards its purification.


Asunto(s)
Quinurenina 3-Monooxigenasa/aislamiento & purificación , Quinurenina 3-Monooxigenasa/metabolismo , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Escherichia coli/genética , Histidina , Humanos , Cinética , Quinurenina 3-Monooxigenasa/química , Quinurenina 3-Monooxigenasa/genética , Redes y Vías Metabólicas , Oligopéptidos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Solubilidad
11.
Commun Biol ; 4(1): 159, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33542467

RESUMEN

The structural mechanisms of single-pass transmembrane enzymes remain elusive. Kynurenine 3-monooxygenase (KMO) is a mitochondrial protein involved in the eukaryotic tryptophan catabolic pathway and is linked to various diseases. Here, we report the mammalian full-length structure of KMO in its membrane-embedded form, complexed with compound 3 (identified internally) and compound 4 (identified via DNA-encoded chemical library screening) at 3.0 Å resolution. Despite predictions suggesting that KMO has two transmembrane domains, we show that KMO is actually a single-pass transmembrane protein, with the other transmembrane domain lying laterally along the membrane, where it forms part of the ligand-binding pocket. Further exploration of compound 3 led to identification of the brain-penetrant compound, 5. We show that KMO is dimeric, and that mutations at the dimeric interface abolish its activity. These results will provide insight for the drug discovery of additional blood-brain-barrier molecules, and help illuminate the complex biology behind single-pass transmembrane enzymes.


Asunto(s)
Membrana Celular/enzimología , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Quinurenina 3-Monooxigenasa/metabolismo , Animales , Sitios de Unión , Inhibidores Enzimáticos/química , Células HEK293 , Humanos , Quinurenina 3-Monooxigenasa/química , Quinurenina 3-Monooxigenasa/genética , Ligandos , Simulación del Acoplamiento Molecular , Mutación , Unión Proteica , Dominios Proteicos , Ratas , Relación Estructura-Actividad
12.
J Neurochem ; 109(2): 316-25, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19226371

RESUMEN

In the mammalian brain, kynurenine aminotransferase II (KAT II) and kynurenine 3-monooxygenase (KMO), key enzymes of the kynurenine pathway (KP) of tryptophan degradation, form the neuroactive metabolites kynurenic acid (KYNA) and 3-hydroxykynurenine (3-HK), respectively. Although physically segregated, both enzymes use the pivotal KP metabolite l-kynurenine as a substrate. We studied the functional consequences of this cellular compartmentalization in vivo using two specific tools, the KAT II inhibitor BFF 122 and the KMO inhibitor UPF 648. The acute effects of selective KAT II or KMO inhibition were studied using a radiotracing method in which the de novo synthesis of KYNA, and of 3-HK and its downstream metabolite quinolinic acid (QUIN), is monitored following an intrastriatal injection of (3)H-kynurenine. In naïve rats, intrastriatal BFF 122 decreased newly formed KYNA by 66%, without influencing 3-HK or QUIN production. Conversely, UPF 648 reduced 3-HK synthesis (by 64%) without affecting KYNA formation. Similar, selective effects of KAT II and KMO inhibition were observed when the inhibitors were applied acutely together with the excitotoxin QUIN, which impairs local KP metabolism. Somewhat different effects of KMO (but not KAT II) inhibition were obtained in rats that had received an intrastriatal QUIN injection 7 days earlier. In these neuron-depleted striata, UPF 648 not only decreased both 3-HK and QUIN production (by 77% and 66%, respectively) but also moderately raised KYNA synthesis (by 27%). These results indicate a remarkable functional segregation of the two pathway branches in the brain, boding well for the development of selective KAT II or KMO inhibitors for cognitive enhancement and neuroprotection, respectively.


Asunto(s)
Encéfalo/metabolismo , Quinurenina 3-Monooxigenasa/química , Quinurenina/metabolismo , Transducción de Señal/fisiología , Transaminasas/química , Animales , Encéfalo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Quinurenina/química , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Quinurenina 3-Monooxigenasa/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transaminasas/antagonistas & inhibidores , Transaminasas/fisiología
13.
Cell Chem Biol ; 25(4): 426-438.e4, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29429898

RESUMEN

Kynurenine 3-monooxygenase (KMO) inhibitors have been developed for the treatment of neurodegenerative disorders. The mechanisms of flavin reduction and hydrogen peroxide production by KMO inhibitors are unknown. Herein, we report the structure of human KMO and crystal structures of Saccharomyces cerevisiae (sc) and Pseudomonas fluorescens (pf) KMO with Ro 61-8048. Proton transfer in the hydrogen bond network triggers flavin reduction in p-hydroxybenzoate hydroxylase, but the mechanism triggering flavin reduction in KMO is different. Conformational changes via π-π interactions between the loop above the flavin and substrate or non-substrate effectors lead to disorder of the C-terminal α helix in scKMO and shifts of domain III in pfKMO, stimulating flavin reduction. Interestingly, Ro 61-8048 has two different binding modes. It acts as a competitive inhibitor in scKMO and as a non-substrate effector in pfKMO. These findings provide understanding of the catalytic cycle of KMO and insight for structure-based drug design of KMO inhibitors.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Peróxido de Hidrógeno/metabolismo , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Quinurenina 3-Monooxigenasa/metabolismo , Pseudomonas fluorescens/enzimología , Saccharomyces cerevisiae/enzimología , Sulfonamidas/farmacología , Tiazoles/farmacología , Secuencia de Aminoácidos , Animales , Flavinas/metabolismo , Humanos , Quinurenina 3-Monooxigenasa/química , Simulación del Acoplamiento Molecular , Oxidación-Reducción/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Pseudomonas fluorescens/química , Saccharomyces cerevisiae/química , Alineación de Secuencia
14.
Nat Commun ; 8: 15827, 2017 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-28604669

RESUMEN

Kynurenine-3-monooxygenase (KMO) is a key FAD-dependent enzyme of tryptophan metabolism. In animal models, KMO inhibition has shown benefit in neurodegenerative diseases such as Huntington's and Alzheimer's. Most recently it has been identified as a target for acute pancreatitis multiple organ dysfunction syndrome (AP-MODS); a devastating inflammatory condition with a mortality rate in excess of 20%. Here we report and dissect the molecular mechanism of action of three classes of KMO inhibitors with differentiated binding modes and kinetics. Two novel inhibitor classes trap the catalytic flavin in a previously unobserved tilting conformation. This correlates with picomolar affinities, increased residence times and an absence of the peroxide production seen with previous substrate site inhibitors. These structural and mechanistic insights culminated in GSK065(C1) and GSK366(C2), molecules suitable for preclinical evaluation. Moreover, revising the repertoire of flavin dynamics in this enzyme class offers exciting new opportunities for inhibitor design.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Insuficiencia Multiorgánica/metabolismo , Pancreatitis/metabolismo , Animales , Inhibidores Enzimáticos/química , Escherichia coli/genética , Humanos , Peróxido de Hidrógeno/metabolismo , Quinurenina 3-Monooxigenasa/química , Quinurenina 3-Monooxigenasa/metabolismo , Modelos Moleculares , Dominios Proteicos , Células Sf9
15.
Drug Discov Today ; 21(2): 315-24, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26589832

RESUMEN

Kynurenine monooxygenase (KMO) is an enzyme of the kynurenine (Kyn) pathway (KP), which is the major catabolic route of tryptophan. Kyn represents a branch point of the KP, being converted into the neurotoxin 3-hydroxykynurenine via KMO, neuroprotectant kynurenic acid, and anthranilic acid. As a result of this branch point, KMO is an attractive drug target for several neurodegenerative and/or neuroinflammatory diseases, especially Huntington's (HD), Alzheimer's (AD), and Parkinson's (PD) diseases. Although a neurological target, administration of KMO inhibitors in the periphery has demonstrated promising pharmacological results. In light of a recent crystal structure release and reports of preclinical candidates, here we provide a concise yet comprehensive update on the current state of research into the enzymology of KMO and related drug discovery efforts, highlighting areas where further work is required.


Asunto(s)
Quinurenina 3-Monooxigenasa , Animales , Humanos , Inflamación/metabolismo , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Quinurenina 3-Monooxigenasa/química , Quinurenina 3-Monooxigenasa/metabolismo , Estructura Molecular , Enfermedades Neurodegenerativas/metabolismo , Saccharomyces cerevisiae/enzimología
16.
J Biomol Screen ; 19(4): 508-15, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24381207

RESUMEN

Kynurenine 3-monooxygenase (KMO) is a therapeutically important target on the eukaryotic tryptophan catabolic pathway, where it converts L-kynurenine (Kyn) to 3-hydroxykynurenine (3-HK). We have cloned and expressed the human form of this membrane protein as a full-length GST-fusion in a recombinant baculovirus expression system. An enriched membrane preparation was used for a directed screen of approximately 78,000 compounds using a RapidFire mass spectrometry (RF-MS) assay. The RapidFire platform provides an automated solid-phase extraction system that gives a throughput of approximately 7 s per well to the mass spectrometer, where direct measurement of both the substrate and product allowed substrate conversion to be determined. The RF-MS methodology is insensitive to assay interference, other than where compounds have the same nominal mass as Kyn or 3-HK and produce the same mass transition on fragmentation. These instances could be identified by comparison with the product-only data. The screen ran with excellent performance (average Z' value 0.8) and provided several tractable hit series for further investigation.


Asunto(s)
Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Espectrometría de Masas , Animales , Dominio Catalítico , Línea Celular , Evaluación Preclínica de Medicamentos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/química , Humanos , Cinética , Quinurenina 3-Monooxigenasa/química , Quinurenina 3-Monooxigenasa/metabolismo , Espectrometría de Masas/métodos , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
17.
J Biomol Screen ; 18(8): 879-89, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23690293

RESUMEN

Kynurenine monooxygenase (KMO) catalyzes the conversion of kynurenine to 3-hydroxykynurenine. Modulation of KMO activity has been implicated in several neurodegenerative diseases, including Huntington disease. Our goal is to develop potent and selective small-molecule KMO inhibitors with suitable pharmacokinetic characteristics for in vivo proof-of-concept studies and subsequent clinical development. We developed a comprehensive panel of biochemical and cell-based assays that use liquid chromatography/tandem mass spectrometry to quantify unlabeled kynurenine and 3-hydroxykynurenine. We describe assays to measure KMO inhibition in cell and tissue extracts, as well as cellular assays including heterologous cell lines and primary rat microglia and human peripheral blood mononuclear cells.


Asunto(s)
Pruebas de Enzimas/métodos , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Quinurenina 3-Monooxigenasa/metabolismo , Animales , Células CHO , Línea Celular , Cromatografía Liquida/métodos , Cricetulus , Perros , Descubrimiento de Drogas , Células HEK293 , Humanos , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/metabolismo , Quinurenina/análogos & derivados , Quinurenina/biosíntesis , Quinurenina/metabolismo , Quinurenina 3-Monooxigenasa/química , Leucocitos Mononucleares/efectos de los fármacos , Macaca fascicularis , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Wistar , Espectrometría de Masas en Tándem/métodos
18.
Curr Biol ; 21(11): 961-6, 2011 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-21636279

RESUMEN

Neuroactive metabolites of the kynurenine pathway (KP) of tryptophan degradation have been implicated in the pathophysiology of neurodegenerative disorders, including Huntington's disease (HD) [1]. A central hallmark of HD is neurodegeneration caused by a polyglutamine expansion in the huntingtin (htt) protein [2]. Here we exploit a transgenic Drosophila melanogaster model of HD to interrogate the therapeutic potential of KP manipulation. We observe that genetic and pharmacological inhibition of kynurenine 3-monooxygenase (KMO) increases levels of the neuroprotective metabolite kynurenic acid (KYNA) relative to the neurotoxic metabolite 3-hydroxykynurenine (3-HK) and ameliorates neurodegeneration. We also find that genetic inhibition of tryptophan 2,3-dioxygenase (TDO), the first and rate-limiting step in the pathway, leads to a similar neuroprotective shift toward KYNA synthesis. Importantly, we demonstrate that the feeding of KYNA and 3-HK to HD model flies directly modulates neurodegeneration, underscoring the causative nature of these metabolites. This study provides the first genetic evidence that inhibition of KMO and TDO activity protects against neurodegenerative disease in an animal model, indicating that strategies targeted at two key points within the KP may have therapeutic relevance in HD, and possibly other neurodegenerative disorders.


Asunto(s)
Drosophila melanogaster/metabolismo , Enfermedad de Huntington/patología , Quinurenina/metabolismo , Degeneración Nerviosa/metabolismo , Animales , Animales Modificados Genéticamente/metabolismo , Modelos Animales de Enfermedad , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Ácido Quinurénico/química , Ácido Quinurénico/metabolismo , Ácido Quinurénico/uso terapéutico , Quinurenina/análogos & derivados , Quinurenina/química , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Quinurenina 3-Monooxigenasa/química , Degeneración Nerviosa/tratamiento farmacológico , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/metabolismo , Triptófano/química , Triptófano/metabolismo , Triptófano Oxigenasa/antagonistas & inhibidores , Triptófano Oxigenasa/química , Triptófano Oxigenasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA