Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 223
Filtrar
1.
J Clin Invest ; 133(19)2023 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-37561584

RESUMEN

Biological aging can be described as accumulative, prolonged metabolic stress and is the major risk factor for cognitive decline and Alzheimer's disease (AD). Recently, we identified and described a quinone reductase 2 (QR2) pathway in the brain, in which QR2 acts as a removable memory constraint and metabolic buffer within neurons. QR2 becomes overexpressed with age, and it is possibly a novel contributing factor to age-related metabolic stress and cognitive deficit. We found that, in human cells, genetic removal of QR2 produced a shift in the proteome opposing that found in AD brains while simultaneously reducing oxidative stress. We therefore created highly specific QR2 inhibitors (QR2is) to enable evaluation of chronic QR2 inhibition as a means to reduce biological age-related metabolic stress and cognitive decline. QR2is replicated results obtained by genetic removal of QR2, while local QR2i microinjection improved hippocampal and cortical-dependent learning in rats and mice. Continuous consumption of QR2is in drinking water improved cognition and reduced pathology in the brains of AD-model mice (5xFAD), with a noticeable between-sex effect on treatment duration. These results demonstrate the importance of QR2 activity and pathway function in the healthy and neurodegenerative brain and what we believe to be the great therapeutic potential of QR2is as first-in-class drugs.


Asunto(s)
Enfermedad de Alzheimer , Quinona Reductasas , Animales , Humanos , Ratones , Ratas , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Hipocampo/metabolismo , Estrés Oxidativo , Quinona Reductasas/antagonistas & inhibidores , Quinona Reductasas/genética , Quinona Reductasas/metabolismo , Estrés Fisiológico
2.
Int J Mol Sci ; 22(23)2021 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-34884863

RESUMEN

The ability of NQO2 to increase the production of free radicals under enhanced generation of quinone derivatives of catecholamines is considered to be a component of neurodegenerative disease pathogenesis. The present study aimed to investigate the neuroprotective mechanisms of original NQO2 inhibitor M-11 (2-[2-(3-oxomorpholin-4-il)-ethylthio]-5-ethoxybenzimidazole hydrochloride) in a cellular damage model using NQO2 endogenous substrate adrenochrome (125 µM) and co-substrate BNAH (100 µM). The effects of M-11 (10-100 µM) on the reactive oxygen species (ROS) generation, apoptosis and lesion of nuclear DNA were evaluated using flow cytometry and single-cell gel electrophoresis assay (comet assay). Results were compared with S29434, the reference inhibitor of NQO2. It was found that treatment of HT-22 cells with M-11 results in a decline of ROS production triggered by incubation of cells with NQO2 substrate and co-substrate. Pre-incubation of HT-22 cells with compounds M-11 or S29434 results in a decrease of DNA damage and late apoptotic cell percentage reduction. The obtained results provide a rationale for further development of the M-11 compound as a potential neuroprotective agent.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Quinona Reductasas/antagonistas & inhibidores , Adrenocromo/metabolismo , Animales , Apoptosis/efectos de los fármacos , Bencimidazoles/química , Línea Celular , Daño del ADN , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/química , Hipocampo/citología , Masculino , Ratones Endogámicos ICR , Neuronas/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/química , Piridinas/farmacología , Alcaloides de Pirrolicidina/farmacología , Quinona Reductasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
3.
ACS Chem Biol ; 16(9): 1770-1778, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34427427

RESUMEN

The utility of in vitro human disease models is mainly dependent on the availability and functional maturity of tissue-specific cell types. We have previously screened for and identified small molecules that can enhance hepatocyte function in vitro. Here, we characterize the functional effects of one of the hits, FH1, on primary human hepatocytes in vitro, and also in vivo on primary hepatocytes in a zebrafish model. Furthermore, we conducted an analogue screen to establish the structure-activity relationship of FH1. We performed affinity-purification proteomics that identified NQO2 to be a potential binding target for this small molecule, revealing a possible link between inflammatory signaling and hepatocellular function in zebrafish and human hepatocyte model systems.


Asunto(s)
Biomarcadores/metabolismo , Inhibidores Enzimáticos/química , Hepatocitos/metabolismo , Quinona Reductasas/antagonistas & inhibidores , Animales , Inhibidores Enzimáticos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Humanos , Interleucina-6/genética , Hígado , Simulación del Acoplamiento Molecular , Unión Proteica , Factor de Transcripción STAT3/genética , Transducción de Señal , Relación Estructura-Actividad , Factores de Necrosis Tumoral/genética , Pez Cebra
4.
Toxicon ; 188: 117-121, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33122156

RESUMEN

Consumption of Cassia occidentalis (CO) seeds, a ubiquitously distributed weed plant, is responsible for a pathological condition known as hepato-myo-encephalopathy (HME). The toxicity of CO seeds is largely attributed to the presence of anthraquinones (AQs). Here, we report that Emodin, a CO anthraquinone, inhibits the enzymatic activity of NADPH-Quinone reductase, which is an intracellular enzyme fundamentally involved in the detoxification of quinone containing compounds. Emodin binds to the active site of the enzyme and acts as a competitive inhibitor with respect to 2, 6-Dichlorophenolindophenol, a known substrate of NADPH-Quinone reductase. Moreover, our in-vitro study further revealed that Emodin was cytotoxic to primary rat hepatocytes.


Asunto(s)
Emodina/toxicidad , Hepatocitos/efectos de los fármacos , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Senna , Animales , Hepatocitos/fisiología , NAD(P)H Deshidrogenasa (Quinona)/antagonistas & inhibidores , NADP , Intoxicación por Plantas , Quinona Reductasas/antagonistas & inhibidores , Ratas
5.
J Agric Food Chem ; 68(43): 11975-11986, 2020 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-33054205

RESUMEN

Garcinia mangostana L. (mangosteen) is a famous tropical fruit that contains a large number of xanthones. Regular consumption of mangosteen may confer health benefits and prevent some diseases, such as malaria. Quinone reductase 2 (QR-2) is a cytosolic enzyme found in human red blood cells, and it is becoming a target for chemoprevention because it is involved in the mechanisms of several diseases, including malaria. To understand whether the xanthones present in mangosteen might inhibit the activity of QR-2, blood samples were collected from rat following the oral administration of mangosteen extract and then incubated with QR-2 followed by UF-HPLC-QTOF/MS analysis to rapidly screen for and identify the QR-2-inhibiting xanthones. A total of 16 xanthones were identified, and six of these (α-mangostin, γ-mangostin, 8-deoxyartanin, 1,3,7-trihydroxy-2,8-di(3-methylbut-2-enyl)xanthone, garcinone E, and 9-hydroxycalabaxanthone) were subjected to QR-2 inhibition assay. γ-Mangostin exhibited the strongest inhibition, achieving an IC50 value of 3.82 ± 0.51 µM. Its interaction with QR-2 was found to involve hydrogen bond and arene-arene interaction as revealed by molecular docking. The present study could provide new insight into the potential application of mangosteen as functional food ingredients for inhibiting the activity of QR-2. However, the extent of daily intake of mangosteen required and the exact contribution of mangosteen to the prevention and treatment of malaria remain subjects of further study.


Asunto(s)
Inhibidores Enzimáticos/farmacocinética , Garcinia mangostana/química , Extractos Vegetales/farmacocinética , Quinona Reductasas/antagonistas & inhibidores , Administración Oral , Animales , Cromatografía Liquida , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/química , Frutas/química , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Quinona Reductasas/química , Quinona Reductasas/metabolismo , Ratas , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem , Xantonas/administración & dosificación , Xantonas/química , Xantonas/farmacocinética
6.
Eur J Med Chem ; 182: 111649, 2019 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-31514018

RESUMEN

(NRH):quinone oxidoreductase 2 (NQO2) is associated with various processes involved in cancer initiation and progression probably via the production of ROS during quinone metabolism. Thus, there is a need to develop inhibitors of NQO2 that are active in vitro and in vivo. As part of a strategy to achieve this we have used the 4-aminoquinoline backbone as a starting point and synthesized 21 novel analogues. The syntheses utilised p-anisidine with Meldrum's acid and trimethyl orthoacetate or trimethyl orthobenzoate to give the 4-hydrazin-quinoline scaffold, which was derivatised with aldehydes or acid chlorides to give hydrazone or hydrazide analogues, respectively. The hydrazones were the most potent inhibitors of NQO2 in cell free systems, some with low nano-molar IC50 values. Structure-activity analysis highlighted the importance of a small substituent at the 2-position of the 4-aminoquinoline ring, to reduce steric hindrance and improve engagement of the scaffold within the NQO2 active site. Cytotoxicity and NQO2-inhibitory activity in vitro was evaluated using ovarian cancer SKOV-3 and TOV-112 cells (expressing high and low levels of NQO2, respectively). Generally, the hydrazones were more toxic than hydrazide analogues and further, toxicity is unrelated to cellular NQO2 activity. Pharmacological inhibition of NQO2 in cells was measured using the toxicity of CB1954 as a surrogate end-point. Both the hydrazone and hydrazide derivatives are functionally active as inhibitors of NQO2 in the cells, but at different inhibitory potency levels. In particular, 4-((2-(6-methoxy-2-methylquinolin-4-yl)hydrazono)methyl)phenol has the greatest potency of any compound yet evaluated (53 nM), which is 50-fold lower than its toxicity IC50. This compound and some of its analogues could serve as useful pharmacological probes to determine the functional role of NQO2 in cancer development and response to therapy.


Asunto(s)
Aminoquinolinas/farmacología , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Hidrazonas/farmacología , Quinona Reductasas/antagonistas & inhibidores , Aminoquinolinas/síntesis química , Aminoquinolinas/química , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Hidrazonas/síntesis química , Hidrazonas/química , Modelos Moleculares , Estructura Molecular , Quinona Reductasas/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
7.
Biosci Rep ; 39(9)2019 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-31431515

RESUMEN

Human NAD(P)H quinone oxidoreductase (DT-diaphorase, NQO1) exhibits negative cooperativity towards its potent inhibitor, dicoumarol. Here, we addressed the hypothesis that the effects of the two cancer-associated polymorphisms (p.R139W and p.P187S) may be partly mediated by their effects on inhibitor binding and negative cooperativity. Dicoumarol stabilized both variants and bound with much higher affinity for p.R139W than p.P187S. Both variants exhibited negative cooperativity towards dicoumarol; in both cases, the Hill coefficient (h) was approximately 0.5 and similar to that observed with the wild-type protein. NQO1 was also inhibited by resveratrol and by nicotinamide. Inhibition of NQO1 by resveratrol was approximately 10,000-fold less strong than that observed with the structurally similar enzyme, NRH quinine oxidoreductase 2 (NQO2). The enzyme exhibited non-cooperative behaviour towards nicotinamide, whereas resveratrol induced modest negative cooperativity (h = 0.85). Nicotinamide stabilized wild-type NQO1 and p.R139W towards thermal denaturation but had no detectable effect on p.P187S. Resveratrol destabilized the wild-type enzyme and both cancer-associated variants. Our data suggest that neither polymorphism exerts its effect by changing the enzyme's ability to exhibit negative cooperativity towards inhibitors. However, it does demonstrate that resveratrol can inhibit NQO1 in addition to this compound's well-documented effects on NQO2. The implications of these findings for molecular pathology are discussed.


Asunto(s)
Estabilidad de Enzimas/efectos de los fármacos , NAD(P)H Deshidrogenasa (Quinona)/genética , Neoplasias/genética , Quinona Reductasas/genética , Dicumarol/química , Dicumarol/farmacología , Humanos , Cinética , NAD(P)H Deshidrogenasa (Quinona)/antagonistas & inhibidores , NAD(P)H Deshidrogenasa (Quinona)/química , Neoplasias/química , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Niacinamida/química , Niacinamida/farmacología , Polimorfismo Genético , Unión Proteica , Quinona Reductasas/antagonistas & inhibidores , Quinona Reductasas/química
8.
J Biol Chem ; 294(32): 12077-12090, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31213529

RESUMEN

Unlike most other tissues, the colon epithelium is exposed to high levels of H2S derived from gut microbial metabolism. H2S is a signaling molecule that modulates various physiological effects. It is also a respiratory toxin that inhibits complex IV in the electron transfer chain (ETC). Colon epithelial cells are adapted to high environmental H2S exposure as they harbor an efficient mitochondrial H2S oxidation pathway, which is dedicated to its disposal. Herein, we report that the sulfide oxidation pathway enzymes are apically localized in human colonic crypts at the host-microbiome interface, but that the normal apical-to-crypt gradient is lost in colorectal cancer epithelium. We found that sulfide quinone oxidoreductase (SQR), which catalyzes the committing step in the mitochondrial sulfide oxidation pathway and couples to complex III, is a critical respiratory shield against H2S poisoning. H2S at concentrations ≤20 µm stimulated the oxygen consumption rate in colon epithelial cells, but, when SQR expression was ablated, H2S concentrations as low as 5 µm poisoned cells. Mitochondrial H2S oxidation altered cellular bioenergetics, inducing a reductive shift in the NAD+/NADH redox couple. The consequent electron acceptor insufficiency caused uridine and aspartate deficiency and enhanced glutamine-dependent reductive carboxylation. The metabolomic signature of this H2S-induced stress response mapped, in part, to redox-sensitive nodes in central carbon metabolism. Colorectal cancer tissues and cell lines appeared to counter the growth-restricting effects of H2S by overexpressing sulfide oxidation pathway enzymes. Our findings reveal an alternative mechanism for H2S signaling, arising from alterations in mitochondrial bioenergetics that drive metabolic reprogramming.


Asunto(s)
Metabolismo Energético , Sulfuro de Hidrógeno/metabolismo , Mitocondrias/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Colon/citología , Colon/metabolismo , Colon/patología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Cisteína/química , Cisteína/metabolismo , Metabolismo Energético/efectos de los fármacos , Humanos , Sulfuro de Hidrógeno/química , Sulfuro de Hidrógeno/farmacología , NAD/química , Oxidación-Reducción , Consumo de Oxígeno/efectos de los fármacos , Quinona Reductasas/antagonistas & inhibidores , Quinona Reductasas/genética , Quinona Reductasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
9.
J Bacteriol ; 201(11)2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30858300

RESUMEN

Korormicin is an antibiotic produced by some pseudoalteromonads which selectively kills Gram-negative bacteria that express the Na+-pumping NADH:quinone oxidoreductase (Na+-NQR.) We show that although korormicin is an inhibitor of Na+-NQR, the antibiotic action is not a direct result of inhibiting enzyme activity. Instead, perturbation of electron transfer inside the enzyme promotes a reaction between O2 and one or more redox cofactors in the enzyme (likely the flavin adenine dinucleotide [FAD] and 2Fe-2S center), leading to the production of reactive oxygen species (ROS). All Pseudoalteromonas contain the nqr operon in their genomes, including Pseudoalteromonas strain J010, which produces korormicin. We present activity data indicating that this strain expresses an active Na+-NQR and that this enzyme is not susceptible to korormicin inhibition. On the basis of our DNA sequence data, we show that the Na+-NQR of Pseudoalteromonas J010 carries an amino acid substitution (NqrB-G141A; Vibrio cholerae numbering) that in other Na+-NQRs confers resistance against korormicin. This is likely the reason that a functional Na+-NQR is able to exist in a bacterium that produces a compound that typically inhibits this enzyme and causes cell death. Korormicin is an effective antibiotic against such pathogens as Vibrio cholerae, Aliivibrio fischeri, and Pseudomonas aeruginosa but has no effect on Bacteroides fragilis and Bacteroides thetaiotaomicron, microorganisms that are important members of the human intestinal microflora.IMPORTANCE As multidrug antibiotic resistance in pathogenic bacteria continues to rise, there is a critical need for novel antimicrobial agents. An essential requirement for a useful antibiotic is that it selectively targets bacteria without significant effects on the eukaryotic hosts. Korormicin is an excellent candidate in this respect because it targets a unique respiratory enzyme found only in prokaryotes, the Na+-pumping NADH:quinone oxidoreductase (Na+-NQR). Korormicin is synthesized by some species of the marine bacterium Pseudoalteromonas and is a potent and specific inhibitor of Na+-NQR, an enzyme that is essential for the survival and proliferation of many Gram-negative human pathogens, including Vibrio cholerae and Pseudomonas aeruginosa, among others. Here, we identified how korormicin selectively kills these bacteria. The binding of korormicin to Na+-NQR promotes the formation of reactive oxygen species generated by the reaction of the FAD and the 2Fe-2S center cofactors with O2.


Asunto(s)
Antibacterianos/farmacología , Antibiosis , Pseudoalteromonas/metabolismo , Especies Reactivas de Oxígeno/agonistas , Aliivibrio fischeri/efectos de los fármacos , Aliivibrio fischeri/enzimología , Aliivibrio fischeri/crecimiento & desarrollo , Aliivibrio fischeri/patogenicidad , Antibacterianos/metabolismo , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bacteroides fragilis/efectos de los fármacos , Bacteroides fragilis/enzimología , Bacteroides fragilis/crecimiento & desarrollo , Bacteroides thetaiotaomicron/efectos de los fármacos , Bacteroides thetaiotaomicron/enzimología , Bacteroides thetaiotaomicron/crecimiento & desarrollo , Ácidos Grasos Insaturados/biosíntesis , Ácidos Grasos Insaturados/farmacología , Flavina-Adenina Dinucleótido/metabolismo , Expresión Génica , Lactonas/farmacología , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Operón , Oxidación-Reducción , Estructura Secundaria de Proteína , Pseudoalteromonas/genética , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/enzimología , Pseudomonas aeruginosa/crecimiento & desarrollo , Pseudomonas aeruginosa/patogenicidad , Quinona Reductasas/antagonistas & inhibidores , Quinona Reductasas/genética , Quinona Reductasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Vibrio cholerae/efectos de los fármacos , Vibrio cholerae/enzimología , Vibrio cholerae/crecimiento & desarrollo , Vibrio cholerae/patogenicidad
10.
Mol Pharmacol ; 95(3): 269-285, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30567956

RESUMEN

Quinone reductase 2 (QR2, E.C. 1.10.5.1) is an enzyme with a feature that has attracted attention for several decades: in standard conditions, instead of recognizing NAD(P)H as an electron donor, it recognizes putative metabolites of NADH, such as N-methyl- and N-ribosyl-dihydronicotinamide. QR2 has been particularly associated with reactive oxygen species and memory, strongly suggesting a link among QR2 (as a possible key element in pro-oxidation), autophagy, and neurodegeneration. In molecular and cellular pharmacology, understanding physiopathological associations can be difficult because of a lack of specific and powerful tools. Here, we present a thorough description of the potent, nanomolar inhibitor [2-(2-methoxy-5H-1,4b,9-triaza(indeno[2,1-a]inden-10-yl)ethyl]-2-furamide (S29434 or NMDPEF; IC50 = 5-16 nM) of QR2 at different organizational levels. We provide full detailed syntheses, describe its cocrystallization with and behavior at QR2 on a millisecond timeline, show that it penetrates cell membranes and inhibits QR2-mediated reactive oxygen species (ROS) production within the 100 nM range, and describe its actions in several in vivo models and lack of actions in various ROS-producing systems. The inhibitor is fairly stable in vivo, penetrates cells, specifically inhibits QR2, and shows activities that suggest a key role for this enzyme in different pathologic conditions, including neurodegenerative diseases.


Asunto(s)
Piridinas/farmacología , Alcaloides de Pirrolicidina/farmacología , Quinona Reductasas/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Hep G2 , Humanos , Masculino , Ratones , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
11.
Biotechnol Appl Biochem ; 65(6): 857-864, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30063071

RESUMEN

NADH:quinone oxidoreductases (NQOs) act as the electron entry sites in bacterial respiration and oxidize intracellular NADH that is essential for the synthesis of numerous molecules. Klebsiella pneumoniae contains three NQOs (NDH-1, NDH-2, and NQR). The effects of inactivating these NQOs, separately and together, on cell metabolism were investigated under different culture conditions. Defective growth was evident in NDH-1-NDH-2 double and NDH-1-NDH-2-NQR triple deficient mutants, which was probably due to damage to the respiratory chain. The results also showed that K. pneumoniae can flexibly use NQOs to maintain normal growth in single NQO-deficient mutants. And more interestingly, under aerobic conditions, inactivating NDH-1 resulted in a high intracellular NADH:NAD+ ratio, which was proven to be beneficial for 2,3-butanediol production. Compared with the parent strain, 2,3-butanediol production by the NDH-1-deficient mutant was increased by 46% and 62% in glycerol- and glucose-based media, respectively. Thus, our findings provide a practical strategy for metabolic engineering of respiratory chains to promote the biosynthesis of 2,3-butanediol in K. pneumoniae.


Asunto(s)
Klebsiella pneumoniae/crecimiento & desarrollo , Klebsiella pneumoniae/metabolismo , Ingeniería Metabólica , Quinona Reductasas/antagonistas & inhibidores , Quinona Reductasas/deficiencia , Butileno Glicoles/química , Butileno Glicoles/metabolismo , Klebsiella pneumoniae/citología , Klebsiella pneumoniae/enzimología , Quinona Reductasas/genética , Quinona Reductasas/metabolismo
12.
J Antibiot (Tokyo) ; 71(8): 731-740, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29691485

RESUMEN

Biotransformation of wortmannilactone F (3) using the marine-derived fungus DL1103 generated wortmannilactone M (1), a novel analog of wortmannilactone, which was a reduction product of 3 at the C-3 carbonyl group. The in vitro inhibitory activities of 10 wortmannilactones, including 1, against electron transport enzymes indicated that all the wortmannilactones were selective inhibitors of NADH-fumarate reductase and NADH-rhodoquinone reductase. The structure-activity relationship analysis showed that the relative configuration of C1" and C5", the positions of double bonds, the oxygen atoms in the dihydropyran moiety, and the keto-carbonyl group in the oxabicyclo-[2.2.1]-heptane moiety were important to the inhibitory activity of wortmannilactones. In vivo studies indicated that 3 significantly decreased the number and size of adult worms in Trichinella spiralis-infected mice in a dose-dependent manner. Notable changes in the cuticle and microvilli of T. spiralis were also observed. Our data provided useful information in the research and development of polyketides with dihydropyran and oxabicyclo [2.2.1] heptane moieties as antihelminthics.


Asunto(s)
Antihelmínticos/farmacología , Proteínas del Complejo de Cadena de Transporte de Electrón/antagonistas & inhibidores , Macrólidos/farmacología , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/antagonistas & inhibidores , Quinona Reductasas/antagonistas & inhibidores , Trichinella spiralis/efectos de los fármacos , Triquinelosis/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Transporte de Electrón/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/metabolismo , Relación Estructura-Actividad
13.
Biochim Biophys Acta Bioenerg ; 1859(7): 482-490, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29621505

RESUMEN

Type II NADH:quinone oxidoreductase (NDH-2) is a proposed drug-target of major pathogenic microorganisms such as Mycobacterium tuberculosis and Plasmodium falciparum. Many NDH-2 inhibitors have been identified, but rational drug development is impeded by the lack of information regarding their mode of action and associated inhibitor-bound NDH-2 structure. We have determined the crystal structure of NDH-2 complexed with a quinolone inhibitor 2-heptyl-4-hydroxyquinoline-N-oxide (HQNO). HQNO is nested into the slot-shaped tunnel of the Q-site, in which the quinone-head group is clamped by Q317 and I379 residues, and hydrogen-bonds to FAD. The interaction of HQNO with bacterial NDH-2 is very similar to the native substrate ubiquinone (UQ1) interactions in the yeast Ndi1-UQ1 complex structure, suggesting a conserved mechanism for quinone binding. Further, the structural analysis provided insight how modifications of quinolone scaffolds improve potency (e.g. quinolinyl pyrimidine derivatives) and suggests unexplored target space for the rational design of new NDH-2 inhibitors.


Asunto(s)
Quinolonas/química , Quinona Reductasas/antagonistas & inhibidores , Quinona Reductasas/química , Bacterias/enzimología , Sitios de Unión , Cristalografía , Diseño de Fármacos , Enlace de Hidrógeno , Ubiquinona/química
14.
Bioorg Med Chem Lett ; 28(8): 1292-1297, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29567345

RESUMEN

Inhibitors of the enzyme NQO2 (NRH: quinone oxidoreductase 2) are of potential use in cancer chemotherapy and malaria. We have previously reported that non-symmetrical furan amidines are potent inhibitors of NQO2 and here novel analogues are evaluated. The furan ring has been changed to other heterocycles (imidazole, N-methylimidazole, oxazole, thiophene) and the amidine group has been replaced with imidate, reversed amidine, N-arylamide and amidoxime to probe NQO2 activity, improve solubility and decrease basicity of the lead furan amidine. All compounds were fully characterised spectroscopically and the structure of the unexpected product N-hydroxy-4-(5-methyl-4-phenylfuran-2-yl)benzamidine was established by X-ray crystallography. The analogues were evaluated for inhibition of NQO2, which showed lower activity than the lead furan amidine. The observed structure-activity relationship for the furan-amidine series with NQO2 was rationalized by preliminary molecular docking and binding mode analysis. In addition, the oxazole-amidine analogue inhibited the growth of Plasmodium falciparum with an IC50 value of 0.3 µM.


Asunto(s)
Amidinas/farmacología , Inhibidores Enzimáticos/farmacología , Furanos/farmacología , Quinona Reductasas/antagonistas & inhibidores , Amidinas/síntesis química , Amidinas/química , Antimaláricos/síntesis química , Antimaláricos/química , Antimaláricos/farmacología , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Furanos/síntesis química , Furanos/química , Imidazoles/síntesis química , Imidazoles/química , Imidazoles/farmacología , Estructura Molecular , Oxazoles/síntesis química , Oxazoles/química , Oxazoles/farmacología , Oximas/síntesis química , Oximas/química , Oximas/farmacología , Plasmodium falciparum/efectos de los fármacos , Relación Estructura-Actividad , Tiofenos/síntesis química , Tiofenos/química , Tiofenos/farmacología
15.
Chem Res Toxicol ; 31(2): 116-126, 2018 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-29281794

RESUMEN

Detoxicating enzymes NAD(P)H:quinone oxidoreductase 1 (NQO1) and NRH:quinone oxidoreductase 2 (NQO2) catalyze the two-electron reduction of quinone-like compounds. The protective role of the polymorphic NQO1 and NQO2 enzymes is especially of interest in the liver as the major site of drug bioactivation to chemically reactive drug metabolites. In the current study, we quantified the concentrations of NQO1 and NQO2 in 20 human liver donors and NQO1 and NQO2 activities with quinone-like drug metabolites. Hepatic NQO1 concentrations ranged from 8 to 213 nM. Using recombinant NQO1, we showed that low nM concentrations of NQO1 are sufficient to reduce synthetic amodiaquine and carbamazepine quinone-like metabolites in vitro. Hepatic NQO2 concentrations ranged from 2 to 31 µM. NQO2 catalyzed the reduction of quinone-like metabolites derived from acetaminophen, clozapine, 4'-hydroxydiclofenac, mefenamic acid, amodiaquine, and carbamazepine. The reduction of the clozapine nitrenium ion supports association studies showing that NQO2 is a genetic risk factor for clozapine-induced agranulocytosis. The 5-hydroxydiclofenac quinone imine, which was previously shown to be reduced by NQO1, was not reduced by NQO2. Tacrine was identified as a potent NQO2 inhibitor and was applied to further confirm the catalytic activity of NQO2 in these assays. While the in vivo relevance of NQO2-catalyzed reduction of quinone-like metabolites remains to be established by identification of the physiologically relevant co-substrates, our results suggest an additional protective role of the NQO2 protein by non-enzymatic scavenging of quinone-like metabolites. Hepatic NQO1 activity in detoxication of quinone-like metabolites becomes especially important when other detoxication pathways are exhausted and NQO1 levels are induced.


Asunto(s)
Iminas/farmacología , NAD(P)H Deshidrogenasa (Quinona)/antagonistas & inhibidores , Quinona Reductasas/antagonistas & inhibidores , Quinonas/farmacología , Biocatálisis , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Iminas/síntesis química , Iminas/química , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/metabolismo , Estructura Molecular , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Oxidación-Reducción , Quinona Reductasas/metabolismo , Quinonas/síntesis química , Quinonas/química , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad
16.
Biol Reprod ; 97(4): 598-611, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-29025057

RESUMEN

NRH: quinone oxidoreductase 2 (NQO2) is a cytosolic and ubiquitously expressed flavoprotein that catalyzes the two-electron reduction of quinone to hydroquinones. Herein, we assessed the protein expression, subcellular localization, and possible functions of NQO2 in mouse oocyte meiotic maturation and embryo development. Western blot analysis detected high and stable protein expression of NQO2 in mouse oocytes during meiotic progression. Immunofluorescence illustrated NQO2 distribution on nuclear membrane, chromosomes, and meiotic spindles. Microtubule poisons treatment (nocodazole and taxol) showed that filamentous assembly of NQO2 and its co-localization with microtubules require microtubule integrity and normal dynamics. Increased levels of NQO2, reactive oxygen species (ROS), malondialdehyde (MDA), and autophagy protein Beclin1 expression were detected in oocytes cultured with ROS stimulator vitamin K3 (VK3), combined with decreased antioxidant glutathione (GSH). These oocytes were arrested at metaphase I with abnormal spindle structure and chromosome configuration. However, this impact was counteracted by melatonin or NQO2 inhibitor S29434, and the spindle configuration and first polar body extrusion were restored. Similarly, morpholino oligo-induced NQO2 knockdown suppressed ROS, MDA, and Beclin1, instead increased GSH in oocytes under VK3. Supplementary S29434 or melatonin limited changes in NQO2, ROS, MDA, Beclin1, and GSH during in vitro aging of ovulated oocytes, thereby maintaining spindle structure, as well as ordered chromosome separation and embryo development potential after parthenogenetic activation with SrCl2. Taken together, NQO2 is involved in ROS generation and subsequent cytotoxicity in oocytes, and its inhibition can restore oocyte maturation and embryo development, suggesting NQO2 as a pharmacological target for infertility cure.


Asunto(s)
Oocitos/fisiología , Transporte de Proteínas/fisiología , Quinona Reductasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Antifibrinolíticos/farmacología , Beclina-1/metabolismo , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Malondialdehído/metabolismo , Meiosis , Melatonina/farmacología , Ratones , Nocodazol/farmacología , Oocitos/efectos de los fármacos , Oocitos/enzimología , Paclitaxel/farmacología , Partenogénesis , Piridinas/farmacología , Alcaloides de Pirrolicidina/farmacología , Quinona Reductasas/antagonistas & inhibidores , Quinona Reductasas/genética , Moduladores de Tubulina/farmacología , Vitamina K 3/farmacología
17.
Bioorg Med Chem Lett ; 27(16): 3787-3793, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28687205

RESUMEN

The study represents the new findings at the crossroads of chemistry and medicine, particularly between medicinal and organic chemistry and ophthalmology. In this work we describe how the chemical reactivity of indolinone scaffold may be used to create small molecule ligands with strong biological response comparable with and larger than that of endogenous hormone. The synthesis of oxindole-based melatonin and 5-methoxycarbonylamino-N-acetyltryptamine (5-MCA-NAT) analogues was proposed and their ability to influence intraocular pressure (IOP) was studied in vivo. Time-dependent study revealed the prolonged effect (more than 6h) of the lead-compound. This effect in combination with high IOP reducing effect (41±6%) in low concentrations of the active compound (0.1wt%) and with high water solubility represents a great potential of low-cost oxindole derivatives as potent antiglaucoma agents.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Presión Intraocular/efectos de los fármacos , Quinona Reductasas/antagonistas & inhibidores , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Indoles/síntesis química , Indoles/química , Ligandos , Modelos Moleculares , Estructura Molecular , Oxindoles , Quinona Reductasas/metabolismo , Relación Estructura-Actividad , Factores de Tiempo
18.
Biochem Cell Biol ; 94(2): 159-66, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26914540

RESUMEN

Sulfide:quinone oxidoreductase (SQR) is a peripheral membrane enzyme that catalyzes the oxidation of sulfide and the reduction of ubiquinone. Ubiquinone binds to a conserved hydrophobic domain and shuttles electrons from a noncovalent flavin adenine dinucleotide cofactor to the membrane-bound quinone pool. Utilizing the structure of decylubiquinone bound to Acidithiobacillus ferrooxidans SQR, we combined site-directed mutagenesis and kinetic approaches to analyze quinone binding. SQR can reduce both benzoquinones and naphthoquinones. The alkyl side-chain of ubiquinone derivatives enhances binding to SQR but limits the enzyme turnover. Pentachlorophenol and 2-n-heptyl-4-hydroxyquinoline-N-oxide are potent inhibitors of SQR with apparent inhibition constants (Ki) of 0.46 µmol·L(-1) and 0.58 µmol·L(-1), respectively. The highly conserved amino acids surrounding the quinone binding site play an important role in quinone reduction. The phenyl side-chains of Phe357 and Phe391 sandwich the benzoquinone head group and are critical for quinone binding. Importantly, conserved amino acids that define the ubiquinone-binding site also play an important role in sulfide oxidation/flavin reduction.


Asunto(s)
Acidithiobacillus/química , Benzoquinonas/metabolismo , Quinona Reductasas/metabolismo , Sulfuros/metabolismo , Benzoquinonas/química , Sitios de Unión , Oxidación-Reducción , Quinona Reductasas/antagonistas & inhibidores , Quinona Reductasas/química , Sulfuros/química
19.
Expert Opin Ther Targets ; 20(3): 303-17, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26465080

RESUMEN

INTRODUCTION: The neurohormone melatonin is secreted by the pineal gland and shows a circadian rhythm. It has been used in pathological conditions to evaluate its protective capacity against disease, often in connection to oxidative stress. The abundance of cases reported suggests that the mechanisms of action through which melatonin confers this protection may be linked to other molecular targets besides its G-protein-coupled receptors or its intrinsic scavenging capacity. AREAS COVERED: In this review, 300 melatonin-induced protection cases are collated together with basic facts about melatonin. Some of the experimental results lead us to believe that, at high 'pharmacological' concentrations (500 µM and beyond), melatonin inhibits the 'newly' discovered target, quinone reductase 2 (QR2). It is possible that QR2 acts through its specific capacity to reduce orthoquinone to particularly unstable chemical species, which cycle spontaneously back to orthoquinone. This process generates high quantities of radical oxygen species, which are associated with many diseases states, such as inflammation. Therefore, inhibition of QR2 could explain some of the protective activities of melatonin. EXPERT OPINION: The tools have been generated to further study this particular and singular mechanism of action of melatonin. The property of QR2 inhibition could unify the many aspects of melatonin action reported in the literature.


Asunto(s)
Antioxidantes/farmacología , Melatonina/farmacología , Quinona Reductasas/antagonistas & inhibidores , Animales , Antioxidantes/metabolismo , Humanos , Melatonina/metabolismo , Terapia Molecular Dirigida , Estrés Oxidativo/efectos de los fármacos , Quinona Reductasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
20.
J Mol Graph Model ; 62: 235-244, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26521207

RESUMEN

Primaquine is a traditional antimalarial drug with low parasitic resistance and generally good acceptance at higher doses, which has been used for over 60 years in malaria treatment. However, several limitations related to its hematotoxicity have been reported. It is believed that this toxicity comes from the hydroxylation of the C-5 and C-6 positions of its 8-aminoquinoline ring before binding to the molecular target: the quinone reductase II (NQO2) human protein. In this study we propose primaquine derivatives, with substitution at position C-6 of the 8-aminoquinoline ring, planned to have better binding to NQO2, compared to primaquine, but with a reduced toxicity related to the C-5 position being possible to be oxidized. On this sense the proposed analogues were suggested in order to reduce or inhibit hydroxylation and further oxidation to hemotoxic metabolites. Five C-6 substituted primaquine analogues were selected by de novo design and further submitted to docking and molecular dynamics simulations. Our results suggest that all analogues bind better to NQO2 than primaquine and may become better antimalarials. However, the analogues 3 and 4 are predicted to have a better activity/toxicity balance.


Asunto(s)
Inhibidores Enzimáticos/química , Primaquina/análogos & derivados , Primaquina/química , Quinona Reductasas/química , Dominio Catalítico , Humanos , Enlace de Hidrógeno , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Estructura Secundaria de Proteína , Quinona Reductasas/antagonistas & inhibidores , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...