Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem J ; 477(8): 1499-1513, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32239172

RESUMEN

Hereditary hemochromatosis (HH) is mostly caused by mutations in the iron-regulatory gene HFE. The disease is associated with iron overload, resulting in liver cirrhosis/cancer, cardiomegaly, kidney dysfunction, diabetes, and arthritis. Fe2+-induced oxidative damage is suspected in the etiology of these symptoms. Here we examined, using Hfe-/- mice, whether disruption of uric acid (UA) homeostasis plays any role in HH-associated arthritis. We detected elevated levels of UA in serum and intestine in Hfe-/- mice compared with controls. Though the expression of xanthine oxidase, which generates UA, was not different in liver and intestine between wild type and Hfe-/- mice, the enzymatic activity was higher in Hfe-/- mice. We then examined various transporters involved in UA absorption/excretion. Glut9 expression did not change; however, there was an increase in Mrp4 and a decrease in Abcg2 in Hfe-/- mice. As ABCG2 mediates intestinal excretion of UA and mutations in ABCG2 cause hyperuricemia, we examined the potential connection between iron and ABCG2. We found p53-responsive elements in hABCG2 promoter and confirmed with chromatin immunoprecipitation that p53 binds to this promoter. p53 protein was reduced in Hfe-/- mouse intestine. p53 is a heme-binding protein and p53-heme complex is subjected to proteasomal degradation. We conclude that iron/heme overload in HH increases xanthine oxidase activity and also promotes p53 degradation resulting in decreased ABCG2 expression. As a result, systemic UA production is increased and intestinal excretion of UA via ABCG2 is decreased, causing serum and tissue accumulation of UA, a potential factor in the etiology of HH-associated arthritis.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Hemocromatosis/metabolismo , Hiperuricemia/enzimología , Ácido Úrico/metabolismo , Xantina Oxidasa/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Animales , Femenino , Hemocromatosis/complicaciones , Hemocromatosis/congénito , Hemocromatosis/enzimología , Proteína de la Hemocromatosis/genética , Proteína de la Hemocromatosis/metabolismo , Homeostasis , Humanos , Hiperuricemia/etiología , Hiperuricemia/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Xantina Oxidasa/genética
2.
J Sci Food Agric ; 101(4): 1349-1354, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-32820534

RESUMEN

BACKGROUND: There has been growing interest in the use of xanthine oxidase (XO) as a therapeutic agent to prevent gout and hyperuricemia. In the present study, XO inhibitory peptides were identified from tuna protein by virtual screening, and molecular docking was used to elicit the interaction mechanism between XO and peptides. RESULTS: A novel tetrapeptide, EEAK, exhibited high XO inhibitory activity with an IC50 of 173.00 ± 0.06 µM. Molecular docking analysis revealed that EEAK bound with the pivotal residues of XO's active sites (i.e., Glu802, Arg880, Glu1261) through two conventional hydrogen bond interactions, two attractive charge interactions, and one salt bridge. EEAK could also bind with the residues Phe649, Leu648, Lys771, Ser876, Phe914, and Thr1010 of XO. CONCLUSION: This study suggested that conventional hydrogen bond interactions and electrostatic interactions play an important role in XO inhibition. The novel XO inhibitory peptide EEAK from tuna protein could be used as potential candidate for controlling gout and hyperuricemia. © 2020 Society of Chemical Industry.


Asunto(s)
Inhibidores Enzimáticos/química , Proteínas de Peces/química , Péptidos/química , Xantina Oxidasa/antagonistas & inhibidores , Animales , Dominio Catalítico , Inhibidores Enzimáticos/farmacología , Proteínas de Peces/farmacología , Gota/tratamiento farmacológico , Gota/enzimología , Humanos , Enlace de Hidrógeno , Hiperuricemia/tratamiento farmacológico , Hiperuricemia/enzimología , Cinética , Simulación del Acoplamiento Molecular , Péptidos/farmacología , Atún , Xantina Oxidasa/química , Xantina Oxidasa/metabolismo
3.
Mar Drugs ; 18(12)2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33271765

RESUMEN

This study was designed to investigate the effects and underlying mechanisms of Astaxanthin (AST) on high-fructose-induced hyperuricemia (HUA) from the perspectives of the uric acid (UA) synthesis and excretion in rat models. Following six weeks of a 10% fructose diet, the level of serum UA effectively decreased in the AST groups as compared to the model group. The enzymatic activities of xanthine oxidase (XOD) and adenosine deaminase (ADA) were significantly inhibited, and the mRNA expression levels of XOD and ADA significantly decreased after the AST administration. These results suggested that the AST reduced UA synthesis by inhibiting the mRNA expressions and enzyme activities of XOD and ADA, thereby contributing to HUA improvement. On the hand, the relative expressions of the mRNA and protein of kidney reabsorption transport proteins (GLUT9 and URAT1) were significantly down-regulated by AST, while that of the kidney secretion proteins (OAT1, OAT3 and ABCG2) were significantly up-regulated by AST. These results indicated that the AST promoted UA excretion by regulating the urate transport proteins, and thus alleviated HUA. This study suggested that the AST could serve as an effective alternative to traditional medicinal drugs for the prevention of fructose-induced HUA.


Asunto(s)
Inhibidores de la Adenosina Desaminasa/farmacología , Adenosina Desaminasa/metabolismo , Hiperuricemia/prevención & control , Proteínas de Transporte de Membrana/efectos de los fármacos , Ácido Úrico/sangre , Xantina Oxidasa/antagonistas & inhibidores , Adenosina Desaminasa/genética , Animales , Biomarcadores/sangre , Biomarcadores/orina , Modelos Animales de Enfermedad , Fructosa , Hiperuricemia/inducido químicamente , Hiperuricemia/enzimología , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratas Sprague-Dawley , Reabsorción Renal/efectos de los fármacos , Ácido Úrico/orina , Xantina Oxidasa/genética , Xantina Oxidasa/metabolismo , Xantófilas/farmacología
4.
Molecules ; 26(1)2020 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-33383880

RESUMEN

Dryopteris crassirhizoma rhizomes are used as a traditional medicine in Asia. The EtOAc extract of these roots has shown potent xanthine oxidase (XO) inhibitory activity. However, the main phloroglucinols in D. crassirhizoma rhizomes have not been analyzed. Thus, we investigated the major constituents responsible for this effect. Bioassay-guided purification isolated four compounds: flavaspidic acid AP (1), flavaspidic acid AB (2), flavaspidic acid PB (3), and flavaspidic acid BB (4). Among these, 1 showed the most potent inhibitory activity with a half-maximal inhibitory concentration (IC50) value of 6.3 µM, similar to that of allopurinol (IC50 = 5.7 µM) and better than that of oxypurinol (IC50 = 43.1 µM), which are XO inhibitors. A comparative activity screen indicated that the acetyl group at C3 and C3' is crucial for XO inhibition. For example, 1 showed nearly 4-fold higher efficacy than 4 (IC50 = 20.9 µM). Representative inhibitors (1-4) in the rhizomes of D. crassirhizoma showed reversible and noncompetitive inhibition toward XO. Furthermore, the potent inhibitors were shown to be present in high quantities in the rhizomes by a UPLC-QTOF-MS analysis. Therefore, the rhizomes of D. crassirhizoma could be used to develop nutraceuticals and medicines for the treatment of gout.


Asunto(s)
Dryopteris/química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Floroglucinol/análogos & derivados , Floroglucinol/farmacología , Xantina Oxidasa/antagonistas & inhibidores , Butirofenonas/química , Butirofenonas/farmacología , Humanos , Hiperuricemia/tratamiento farmacológico , Hiperuricemia/enzimología , Rizoma/química , Xantina Oxidasa/metabolismo
5.
FASEB J ; 32(2): 1007-1016, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29042453

RESUMEN

The prevalence of hyperuricemia has increased rapidly over the past decades. Bisphenol A (BPA) is an environmental endocrine disruptor. We investigated the effects of BPA on uric acid metabolism and its potential mechanisms. Experiments were performed in different animal models, cell cultures, and humans. In 3 different animal models, BPA exposure increased serum and hepatic uric acid with enhanced activity of xanthine oxidase (XO) in liver, whereas the excretion of uric acid was unchanged. Both in vivo and in vitro, BPA-induced uric acid production was decreased after treatment with allopurinol, which is a XO inhibitor. XO led to the accumulation of uric acid after xanthine was added, with the enzyme-catalyzed reaction, which was enhanced by BPA. Altered secondary structures of XO were found by circular dichroism analysis in the conditions of different BPA concentrations. Molecular docking portrayed Asp360 and Lys422 of XO to be the preferred binding sites for BPA. Mutation of both sites significantly blocked the effect of BPA on XO activity. In humans, patients with hyperuricemia exhibited higher levels of serum BPA than subjects without hyperuricemia. These findings demonstrate BPA promotes hyperuricemia by increasing hepatic uric acid synthesis via the activation of XO, probably through direct binding.-Ma, L., Hu, J., Li, J., Yang, Y., Zhang, L., Zou, L., Gao, R., Peng, C., Wang, Y., Luo, T., Xiang, X., Qing, H., Xiao, X., Wu, C., Wang, Z., He, J. C., Li, Q., Yang, S. Bisphenol A promotes hyperuricemia via activating xanthine oxidase.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Hiperuricemia , Hígado/enzimología , Simulación del Acoplamiento Molecular , Fenoles/toxicidad , Xantina Oxidasa , Animales , Sitios de Unión , Inducción Enzimática/efectos de los fármacos , Hiperuricemia/inducido químicamente , Hiperuricemia/enzimología , Masculino , Ratones , Xantina Oxidasa/biosíntesis , Xantina Oxidasa/química
6.
Nutr Metab Cardiovasc Dis ; 29(10): 1011-1022, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31378626

RESUMEN

BACKGROUND AND AIMS: Systemic reviews and meta-analyses suggest hyperuricemia is a cardiovascular risk factor. The effects of xanthine oxidase inhibitors on cardiac outcomes remain unclear. We assessed the effects of febuxostat and allopurinol on mortality and adverse reactions in adult patients with hyperuricemia. METHODS AND RESULTS: PubMed and EMBASE were searched to retrieve randomized controlled trials of febuxostat and allopurinol from January 2005 to July 2018. The meta-analysis consisted of 13 randomized controlled trials with a combined sample size of 13,539 patients. Febuxostat vs. allopurinol was not associated with an increased risk of cardiac-related mortality in the overall population (OR: 0.72, 95% CI: 0.24-2.13, P = 0.55). Regarding adverse skin reactions, the patients receiving febuxostat had significantly fewer adverse skin reactions than those receiving allopurinol treatment (OR: 0.50, 95% CI: 0.30-085, P = 0.01). Compared with allopurinol, febuxostat was associated with an improved safety outcome of cardiac-related mortality and adverse skin reactions (OR: 0.72, 95% CI: 0.55-0.96, P = 0.02). The net clinical outcome, composite of incident gout and the safety outcome, was not different significantly in the patients receiving febuxostat or allopurinol (OR: 1.04, 95% CI: 0.76-0.1.42, P = 0.79). In sensitivity analyses, a borderline significance was found in the patients randomized to febuxostat vs. allopurinol regarding cardiac-related mortality (OR: 1.29, 95% CI: 1.00-1.67, P = 0.05) after the CARES study was included. CONCLUSION: Febuxostat vs. allopurinol was associated with the improved safety outcome and have comparable mortality and net clinical outcome in patients with hyperuricemia. REGISTRATION NUMBER: PROSPERO(CRD42018091657).


Asunto(s)
Alopurinol/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Febuxostat/uso terapéutico , Supresores de la Gota/uso terapéutico , Gota/tratamiento farmacológico , Hiperuricemia/tratamiento farmacológico , Ácido Úrico/sangre , Anciano , Alopurinol/efectos adversos , Enfermedades Asintomáticas , Biomarcadores/sangre , Inhibidores Enzimáticos/efectos adversos , Febuxostat/efectos adversos , Femenino , Gota/sangre , Gota/enzimología , Gota/mortalidad , Supresores de la Gota/efectos adversos , Humanos , Hiperuricemia/sangre , Hiperuricemia/enzimología , Hiperuricemia/mortalidad , Masculino , Persona de Mediana Edad , Ensayos Clínicos Controlados Aleatorios como Asunto , Medición de Riesgo , Factores de Riesgo , Resultado del Tratamiento , Xantina Oxidasa/antagonistas & inhibidores
7.
Kidney Int ; 93(1): 69-80, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28729031

RESUMEN

The urate oxidase (Uox) gene encodes uricase that in the rodent liver degrades uric acid into allantoin, forming an obstacle for establishing stable mouse models of hyperuricemia. The loss of uricase in humans during primate evolution causes their vulnerability to hyperuricemia. Thus, we generated a Uox-knockout mouse model on a pure C57BL/6J background using the transcription activator-like effector nuclease (TALEN) technique. These Uox-knockout mice spontaneously developed hyperuricemia (over 420 µmol/l) with about 40% survival up to 62 weeks. Renal dysfunction (elevated serum creatinine and blood urea nitrogen) and glomerular/tubular lesions were observed in these Uox-knockout mice. Male Uox-knockout mice developed glycol-metabolic disorders associated with compromised insulin secretion and elevated vulnerability to streptozotocin-induced diabetes, whereas female mice developed hypertension accompanied by aberrant lipo-metabolism. Urate-lowering drugs reduced serum uric acid and improved hyperuricemia-induced disorders. Thus, uricase knockout provides a suitable mouse model to investigate hyperuricemia and associated disorders mimicking the human condition, suggesting that hyperuricemia has a causal role in the development of metabolic disorders and hypertension.


Asunto(s)
Hiperuricemia/enzimología , Riñón/metabolismo , Hígado/enzimología , Urato Oxidasa/deficiencia , Ácido Úrico/sangre , Animales , Biomarcadores/sangre , Glucemia/metabolismo , Presión Sanguínea , Nitrógeno de la Urea Sanguínea , Creatinina/sangre , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Dislipidemias/sangre , Dislipidemias/enzimología , Dislipidemias/genética , Femenino , Predisposición Genética a la Enfermedad , Supresores de la Gota/farmacología , Hipertensión/enzimología , Hipertensión/genética , Hipertensión/fisiopatología , Hiperuricemia/sangre , Hiperuricemia/tratamiento farmacológico , Hiperuricemia/genética , Insulina/sangre , Riñón/patología , Riñón/fisiopatología , Lípidos/sangre , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Factores de Tiempo , Urato Oxidasa/genética
8.
Int J Mol Sci ; 19(10)2018 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-30340390

RESUMEN

Inonotus obliquus is an edible mushroom and also a remedy against various diseases, especially metabolic syndrome. In this paper we report the actions of an ethanol extract of I. obliquus (IOE) against hyperuricemia in hyperuricemic mice, and the screen of bioactives. The extract (IOE) was prepared by extracting I. obliquus at 65 °C with ethanol, and characterized by HPLC. IOE at low, middle, and high doses reduced serum uric acid (SUA) of hyperuricemic mice (353 µmol/L) to 215, 174, and 152 µmol/L (p < 0.01), respectively, showing similar hypouricemic effectiveness to the positive controls. IOE showed a non-toxic impact on kidney and liver functions. Of note, IOE suppressed xanthine oxidase (XOD) activity in serum and liver, and also down-regulated renal uric acid transporter 1 (URAT1). Four compounds hit highly against XOD in molecular docking. Overall, the four compounds all occupied the active tunnel, which may inhibit the substrate from entering. The IC50 of betulin was assayed at 121.10 ± 4.57 µM, which was near to that of allopurinol (148.10 ± 5.27 µM). Betulin may be one of the anti-hyperuricemia bioactives in I. obliquus.


Asunto(s)
Basidiomycota/química , Productos Biológicos/química , Productos Biológicos/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Hiperuricemia/enzimología , Modelos Moleculares , Xantina Oxidasa/química , Animales , Descubrimiento de Drogas , Hiperuricemia/sangre , Hiperuricemia/tratamiento farmacológico , Ratones , Relación Estructura-Actividad Cuantitativa , Xantina Oxidasa/antagonistas & inhibidores
9.
Med Sci Monit ; 23: 1129-1140, 2017 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-28258276

RESUMEN

BACKGROUND The aim of this study was to study the effects of gypenosides (GPS) on lowering uric acid (UA) levels in hyperuricemic rats induced by lipid emulsion (LE) and the related mechanisms. GPS are natural saponins extracted from Gynostemma pentaphyllum. MATERIAL AND METHODS Forty-eight male SD rats were randomly divided into six groups: normal, model, two positive controls, and two GPS treated groups (two different doses of GPS). The normal group rats were fed a basic diet, and the other rats were orally pretreated with LE. Urine and blood were collected at regular intervals. Full automatic biochemical analyzer was used to detect the concentration levels of serum UA (SUA), serum creatinine (SCr), BUN, and urine UA (UUA), and urine creatinine (UCr) and fractional excretion of UA (FEUA). ELISA kits were used to detect enzymes activities: xanthine oxidase (XOD), adenosime deaminase (ADA), guanine deaminase (GDA), and xanthine dehydrogenase (XDH). Immunohistochemistry was used to observe kidney changes and protein (URAT1, GLUT9, and OAT1) expression levels. RT-PCR was used to detect the relevant mRNA expression levels. RESULTS Treatment with GPS significantly reduced the SUA, prevented abnormal weight loss caused by LE, and improved kidney pathomorphology. Treatment with GPS also decreased the levels of XOD, ADA, and XDH expression, increased the kidney index and FEUA, downregulated URAT1 and GLUT9 expression and upregulated OAT1 expression in the kidney. CONCLUSIONS GPS may be an effective treatment for hyperuricemia via a decrease in xanthine oxidoreductase through the XOD/XDH system; and via an increase in urate excretion through regulating URAT1, GLUT9, and OAT1 transporters.


Asunto(s)
Hiperuricemia/tratamiento farmacológico , Xantina Deshidrogenasa/antagonistas & inhibidores , Animales , Colesterol/administración & dosificación , Colesterol/metabolismo , Dieta Alta en Grasa , Gynostemma , Hipercolesterolemia/metabolismo , Hiperuricemia/sangre , Hiperuricemia/enzimología , Hiperuricemia/orina , Riñón/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Extractos Vegetales/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Ácido Úrico/metabolismo , Xantina Deshidrogenasa/metabolismo
10.
Pharm Biol ; 55(1): 33-42, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27650786

RESUMEN

CONTEXT: Boswellia dalzielii Hutch. (Burseraceae) is an aromatic plant. The leaves are used for beverage flavouring. OBJECTIVE: This study investigates the chemical composition and biological activities of various extracts. MATERIALS AND METHODS: The essential oil was prepared via hydrodistillation. Identification and quantification were realized via GC-MS and GC-FID. Consecutive extractions (cyclohexane, dichloromethane, ethyl acetate and methanol) were carried out and various chemical groups (phenolics, flavonoids, tannins, antocyanins and sugar) were quantified. The volatile compounds of organic extracts were identified before and after derivatization. Antioxidant, antihyperuricemia, anti-Alzheimer, anti-inflammatory and anticancer activities were evaluated. RESULTS: In the essential oil, 50 compounds were identified, including 3-carene (27.72%) and α-pinene (15.18%). 2,5-Dihydroxy acetophenone and ß-d-xylopyranose were identified in the methanol extract. Higher phenolic (315.97 g GAE/kg dry mass) and flavonoid (37.19 g QE/kg dry mass) contents were observed in the methanol extract. The methanol extract has presented remarkable IC50 = 6.10 mg/L for antiDPPH, 35.10 mg/L for antixanthine oxidase and 28.01 mg/L for anti-5-lipoxygenase. For acetylcholinesterase inhibition, the best IC50 (76.20 and 67.10 mg/L) were observed, respectively, with an ethyl acetate extract and the essential oil. At 50 mg/L, the dichloromethane extract inhibited OVCAR-3 cell lines by 65.10%, while cyclohexane extract inhibited IGROV-1 cell lines by 92.60%. DISCUSSION AND CONCLUSION: Biological activities were fully correlated with the chemical groups of the extracts. The ethyl acetate and methanol extracts could be considered as potential alternatives for use in dietary supplements for the prevention or treatment of diseases because of these extracts natural antioxidant, antihyperuricemic and anti-inflammatory activities.


Asunto(s)
Antiinflamatorios/farmacología , Antineoplásicos/farmacología , Antioxidantes/farmacología , Boswellia/química , Supresores de la Gota/farmacología , Aceites Volátiles/farmacología , Extractos Vegetales/farmacología , Hojas de la Planta/química , Aceites de Plantas/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/enzimología , Antiinflamatorios/aislamiento & purificación , Antineoplásicos/aislamiento & purificación , Antioxidantes/aislamiento & purificación , Compuestos de Bifenilo/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidores de la Colinesterasa/aislamiento & purificación , Inhibidores de la Colinesterasa/farmacología , Femenino , Ionización de Llama , Cromatografía de Gases y Espectrometría de Masas , Supresores de la Gota/aislamiento & purificación , Humanos , Hiperuricemia/tratamiento farmacológico , Hiperuricemia/enzimología , Inhibidores de la Lipooxigenasa/aislamiento & purificación , Inhibidores de la Lipooxigenasa/farmacología , Aceites Volátiles/aislamiento & purificación , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Fitoterapia , Picratos/química , Extractos Vegetales/aislamiento & purificación , Aceites de Plantas/aislamiento & purificación , Plantas Medicinales , Solventes/química , Xantina Oxidasa/antagonistas & inhibidores , Xantina Oxidasa/metabolismo
11.
Med Sci Monit ; 22: 2501-12, 2016 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-27423335

RESUMEN

Uric acid is the final oxidation product of purine metabolism in humans. Xanthine oxidoreductase (XOR) catalyzes oxidative hydroxylation of hypoxanthine to xanthine to uric acid, accompanying the production of reactive oxygen species (ROS). Uric acid usually forms ions and salts known as urates and acid urates in serum. Clinically, overproduction or under-excretion of uric acid results in the elevated level of serum uric acid (SUA), termed hyperuricemia, which has long been established as the major etiologic factor in gout. Accordingly, urate-lowering drugs such as allopurinol, an XOR-inhibitor, are extensively used for the treatment of gout. In recent years, the prevalence of hyperuricemia has significantly increased and more clinical investigations have confirmed that hyperuricemia is an independent risk factor for cardiovascular disease, hypertension, diabetes, and many other diseases. Urate-lowering therapy may also play a critical role in the management of these diseases. However, current XOR-inhibitor drugs such as allopurinol and febuxostat may have significant adverse effects. Therefore, there has been great effort to develop new XOR-inhibitor drugs with less or no toxicity for the long-term treatment or prevention of these hyperuricemia-related diseases. In this review, we discuss the mechanism of uric acid homeostasis and alterations, updated prevalence, therapeutic outcomes, and molecular pathophysiology of hyperuricemia-related diseases. We also summarize current discoveries in the development of new XOR inhibitors.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Hiperuricemia/enzimología , Hiperuricemia/terapia , Xantina Deshidrogenasa/antagonistas & inhibidores , Xantina Deshidrogenasa/metabolismo , Alopurinol/farmacología , Animales , Inhibidores Enzimáticos/uso terapéutico , Febuxostat/farmacología , Humanos , Especies Reactivas de Oxígeno/metabolismo , Factores de Riesgo
12.
Pharm Biol ; 54(9): 1680-6, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26916555

RESUMEN

Context Mangiferin has been reported to possess a potential hypouricaemic effect. However, the pharmacokinetic studies in rats showed that its oral bioavailability was only 1.2%, suggesting that mangiferin metabolites might exert the action. Objective The hypouricaemic effect and the xanthine oxidase inhibition of mangiferin and norathyriol, a mangiferin metabolite, were investigated. Inhibition of norathyriol analogues (compounds 3-9) toward xanthine oxidase was also evaluated. Materials and methods For a dose-dependent study, mangiferin (1.5-6.0 mg/kg) and norathyriol (0.92-3.7 mg/kg) were administered intragastrically to mice twice daily for five times. For a time-course study, mice received mangiferin and norathyriol both at a single dose of 7.1 µmol/kg. In vitro, inhibition of test compounds (2.4-2.4 mM) against xanthine oxidase activity was evaluated by the spectrophotometrical method. The inhibition type was identified from Lineweaver-Burk plots. Results Norathyriol (0.92, 1.85 and 3.7 mg/kg) dose dependently decreased the serum urate levels by 27.0, 33.6 and 37.4%, respectively. The action was more potent than that of mangiferin at the low dose, but was equivalent at the higher doses. Additionally, the hypouricaemic action of them exhibited a time dependence. In vitro, norathyriol markedly inhibited the xanthine oxidase activities, with the IC50 value of 44.6 µM, but mangiferin did not. The kinetic studies showed that norathyriol was an uncompetitive inhibitor by Lineweaver-Burk plots. The structure-activity relationships exhibited that three hydroxyl groups in norathyriol at the C-1, C-3 and C-6 positions were essential for maintaining xanthine oxidase inhibition. Discussion and conclusion Norathyriol was responsible for the hypouricaemic effect of mangiferin via inhibiting xanthine oxidase activity.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Supresores de la Gota/farmacología , Hiperuricemia/tratamiento farmacológico , Ácido Úrico/sangre , Xantenos/farmacología , Xantina Oxidasa/antagonistas & inhibidores , Xantonas/farmacología , Administración Oral , Animales , Biomarcadores/sangre , Biotransformación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/metabolismo , Supresores de la Gota/administración & dosificación , Supresores de la Gota/metabolismo , Hiperuricemia/sangre , Hiperuricemia/inducido químicamente , Hiperuricemia/enzimología , Cinética , Ratones , Estructura Molecular , Ácido Oxónico , Relación Estructura-Actividad , Xantenos/administración & dosificación , Xantenos/metabolismo , Xantina Oxidasa/metabolismo , Xantonas/administración & dosificación , Xantonas/metabolismo
13.
J Hepatol ; 62(6): 1412-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25623823

RESUMEN

BACKGROUND & AIMS: Hyperuricemia is a common feature of patients with non-alcoholic fatty liver disease (NAFLD). This study aimed to explore the causal relationship and underlying mechanisms between NAFLD and hyperuricemia. METHODS: We evaluated the impact of NAFLD on the development of hyperuricemia in a cohort of 5541 baseline hyperuricemia-free individuals. We further analyzed xanthine oxidase (XO), a rate-limiting enzyme that catalyzes uric acid production, as a candidate to link NAFLD and hyperuricemia. RESULTS: In the first study, a 7-year prospective analysis found that NAFLD was strongly associated with subsequent development of hyperuricemia. Cox proportional hazards regression analyses showed that age, gender, and body mass index adjusted hazard ratio (95% confidence interval) for incident hyperuricemia was 1.609 (1.129-2.294) in individuals with NAFLD, as compared with those without NAFLD at baseline. In the second study, we observed that expression and activity of XO were significantly increased in cellular and mouse models of NAFLD. Knocking down XO expression or inhibiting XO activity significantly decreases uric acid production and attenuates free fatty acids-induced fat accumulation in HepG2 cells. Inhibiting XO activity also significantly prevents the development of and ameliorates established hepatic steatosis induced by a high-fat diet in mice. Further experiments indicated that XO regulates activation of the NLRP3 inflammasome, which may be essential for the regulatory effect of XO on NAFLD. CONCLUSIONS: NAFLD significantly increases the risk of incident hyperuricemia. XO is a mediator of the relationship between NAFLD and hyperuricemia, and may serve as a novel therapeutic target for the two linked diseases.


Asunto(s)
Hiperuricemia/enzimología , Hiperuricemia/etiología , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/enzimología , Xantina Oxidasa/metabolismo , Adulto , Animales , Proteínas Portadoras/metabolismo , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Células Hep G2 , Humanos , Inflamasomas/metabolismo , Metabolismo de los Lípidos , Masculino , Ratones , Persona de Mediana Edad , Proteína con Dominio Pirina 3 de la Familia NLR , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Estudios Prospectivos , Factores de Riesgo , Ácido Úrico/metabolismo , Xantina Oxidasa/antagonistas & inhibidores , Xantina Oxidasa/genética
14.
Am J Physiol Gastrointest Liver Physiol ; 309(1): G42-51, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25999428

RESUMEN

Xanthine oxidase (XO) is an enzyme involved in the production of uric acid (UA) from purine nucleotides. Numerous recent studies have revealed the likelihood of metabolic syndrome including nonalcoholic fatty liver disease (NAFLD) or steatohepatitis (NASH) to be related to hyperuricemia. However, it remains unclear whether elevated serum UA during the development of NAFLD or NASH is a cause or a consequence of these diseases. In this study, the XO inhibitor febuxostat was administered to two types of NASH model mice. Febuxostat exerted a strong protective effect against NASH development induced by a high-fat diet containing trans fatty acid (HFDT). In contrast, methionine choline-deficient-diet-induced NASH development not accompanied by hyperuricemia showed no UA normalization, suggesting that the ameliorating effect of febuxostat occurs via the normalization of hyperuricemia itself and/or accompanying molecular mechanism(s) such as oxidative stress. In the HFDT-fed mice, hyperuricemia, elevated alanine aminotransferase, and increased Tunnel-positive cells in the liver were normalized by febuxostat administration. In addition, upregulation of fatty acid oxidation-related genes, fibrotic change, and increases in collagen deposition, inflammatory cytokine expressions, and lipid peroxidation in the HFDT-fed mice were also normalized by febuxostat administration. Taken together, these observations indicate that administration of febuxostat has a protective effect against HFDT-induced NASH development, suggesting the importance of XO in its pathogenesis. Thus XO inhibitors are potentially potent therapies for patients with NASH, particularly that associated with hyperuricemia.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Supresores de la Gota/farmacología , Hiperuricemia/tratamiento farmacológico , Hígado/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Tiazoles/farmacología , Xantina Oxidasa/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Deficiencia de Colina/complicaciones , Citoprotección , Dieta Alta en Grasa , Febuxostat , Hiperuricemia/sangre , Hiperuricemia/enzimología , Hiperuricemia/etiología , Hiperuricemia/patología , Hígado/enzimología , Hígado/patología , Cirrosis Hepática Experimental/enzimología , Cirrosis Hepática Experimental/patología , Cirrosis Hepática Experimental/prevención & control , Metionina/deficiencia , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/enzimología , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/patología , Ácido Úrico/sangre , Xantina Oxidasa/metabolismo
15.
J Biol Inorg Chem ; 20(2): 195-207, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25501928

RESUMEN

Xanthine oxidoreductase (XOR), which is widely distributed from humans to bacteria, has a key role in purine catabolism, catalyzing two steps of sequential hydroxylation from hypoxanthine to xanthine and from xanthine to urate at its molybdenum cofactor (Moco). Human XOR is considered to be a target of drugs not only for therapy of hyperuricemia and gout, but also potentially for a wide variety of other diseases. In this review, we focus on studies of XOR inhibitors and their implications for understanding the chemical nature and reaction mechanism of the Moco active site of XOR. We also discuss further experimental or clinical studies that would be helpful to clarify remaining issues.


Asunto(s)
Coenzimas/química , Gota/enzimología , Hiperuricemia/enzimología , Metaloproteínas/química , Pteridinas/química , Xantina Deshidrogenasa/química , Dominio Catalítico , Coenzimas/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Gota/tratamiento farmacológico , Gota/patología , Humanos , Hiperuricemia/tratamiento farmacológico , Hiperuricemia/patología , Metaloproteínas/metabolismo , Cofactores de Molibdeno , Pteridinas/metabolismo , Xantina/química , Xantina Deshidrogenasa/antagonistas & inhibidores , Xantina Deshidrogenasa/metabolismo
16.
Cell Physiol Biochem ; 33(2): 479-90, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24556878

RESUMEN

BACKGROUND: The exact etiology of hyperuricemia-induced endothelial injury remains ill-defined. To elucidate the mechanism that leads to endothelial injury in hyperuricemia, we investigated proteins expressed in human umbilical vein endothelial cells (HUVECs) cultured with high concentrations of uric acid (HUA) in vitro. METHODS: We used stable isotope labeling with amino acids in cell culture (SILAC) combined with LC-MS/MS analysis to compare proteins expressed in HUVECs cultured in media with or without HUA. The results were confirmed by Western blotting. Reactive oxygen species (ROS) were detected using a confocal microscope. RESULTS: Thirty-nine proteins with various cellular functions were differentially expressed. Among them, aldose reductase (ALDR) protein expression was enhanced significantly, indicating increased aldehyde reductase and oxidoreductase activities. ROS levels decreased when ALDR protein activity was inhibited by siALDR. CONCLUSIONS: ALDR protein may play an important role in endothelial injury induced by hyperuricemia, and activity of the ALDR protein is associated with oxidative stress.


Asunto(s)
Aldehído Reductasa/biosíntesis , Regulación Enzimológica de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/enzimología , Hiperuricemia/enzimología , Estrés Oxidativo , Antioxidantes/efectos adversos , Antioxidantes/farmacología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Hiperuricemia/patología , Marcaje Isotópico , Ácido Úrico/efectos adversos , Ácido Úrico/farmacología
17.
Biosci Biotechnol Biochem ; 78(4): 655-61, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25036964

RESUMEN

We applied Chrysanthemum flower oil (CFO) to a hyperuricemia model by feeding rats a hyperuricemia-inducing diet (HID) and investigated its effect on serum uric acid (SUA) levels and its mode of action. CFO is the oily fraction that contains polyphenols derived from chrysanthemum flowers. Oral administration of CFO to HID-fed rats significantly decreased their SUA levels. It also inhibited xanthine oxidase activities in the liver and increased urine uric acid levels. The effects of CFO on the renal gene expressions that accompanied the induction of hyperuricemia were comprehensively confirmed by DNA microarray analysis. The analysis showed up-regulation of those genes for uric acid excretion by CFO administration. These results suggest that CFO suppresses the increase in SUA levels via two mechanisms: suppression of uric acid production by inhibition of xanthine oxidase in the liver and acceleration of its excretion by up-regulation of uric acid transporter genes in the kidney.


Asunto(s)
Chrysanthemum/química , Flores/química , Hiperuricemia/tratamiento farmacológico , Hiperuricemia/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Aceites de Plantas/farmacología , Animales , Bovinos , Hiperuricemia/sangre , Hiperuricemia/enzimología , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Aceites de Plantas/administración & dosificación , Aceites de Plantas/uso terapéutico , Ratas , Ácido Úrico/sangre , Xantina Oxidasa/metabolismo
18.
J Dairy Sci ; 97(7): 4029-42, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24835972

RESUMEN

Hyperuricemia is a biochemical hallmark of gout, renal urate lithiasis, and inherited purine disorders, and may be a result of enormous ATP breakdown or purine release as a result of cardiovascular disease, hypertension, kidney disease, eclampsia, obesity, metabolic syndrome, psoriasis, tumor lysis syndrome, or intense physical training. The beneficial role of dairy products on hyperuricemia management and prevention is well documented in the literature. The primary aim of our experimental study was to examine the effect of milk dietary regimen (commercial 1.5% fat UHT milk or patented depurinized milk) compared with allopurinol therapy on experimental hyperuricemia induced by oxonic acid in rats. Principal component analysis was applied on a data set consisting of 11 variables for 8 different experimental groups. Among the 11 parameters measured (plasma uric acid and the liver parameters NFκB-p65, Akt kinase/phospho-Akt kinase, ERK kinase/phospho-ERK kinase, IRAK kinase/phospho IRAK kinase, p38/phospho-p38, and DNase), Akt/phospho Akt and ERK/phospho-ERK signaling were extracted as the most discriminating. We also compared the content of various potentially toxic compounds (sulfur compounds, ketones, aldehydes, alcohols, esters, carboxylic acids, and phthalates) in untreated commercial milk and depurinized milk. Of all the compounds investigated in this study that were observed in commercial milk (24 volatile organic compounds and 4 phthalates), 6 volatile organic compounds were not detected in depurinized milk. For almost all of the other compounds, significant decreases in concentration were observed in depurinized milk compared with commercial milk. In conclusion, a depurinized milk diet may be recommended in nutritional treatment of primary and secondary hyperuricemia to avoid uric acid and other volatile, potentially toxic compounds that may slow down liver regeneration and may induce chronic liver diseases.


Asunto(s)
Alopurinol/farmacología , Alopurinol/uso terapéutico , Endonucleasas/metabolismo , Hiperuricemia/dietoterapia , Hígado/enzimología , Leche/metabolismo , FN-kappa B/metabolismo , Alimentación Animal/análisis , Animales , Dieta , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Hiperuricemia/inducido químicamente , Hiperuricemia/tratamiento farmacológico , Hiperuricemia/enzimología , Hígado/efectos de los fármacos , Masculino , Leche/química , Ácido Oxónico/toxicidad , Distribución Aleatoria , Ratas , Ratas Wistar
19.
J Biol Chem ; 286(15): 12901-11, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21266577

RESUMEN

Urate and myeloperoxidase (MPO) are associated with adverse outcomes in cardiovascular disease. In this study, we assessed whether urate is a likely physiological substrate for MPO and if the products of their interaction have the potential to exacerbate inflammation. Urate was readily oxidized by MPO and hydrogen peroxide to 5-hydroxyisourate, which decayed to predominantly allantoin. The redox intermediates of MPO were reduced by urate with rate constants of 4.6 × 10(5) M(-1) s(-1) for compound I and 1.7 × 10(4) M(-1) s(-1) for compound II. Urate competed with chloride for oxidation by MPO and at hyperuricemic levels is expected to be a substantive substrate for the enzyme. Oxidation of urate promoted super-stoichiometric consumption of glutathione, which indicates that it is converted to a free radical intermediate. In combination with superoxide and hydrogen peroxide, MPO oxidized urate to a reactive hydroperoxide. This would form by addition of superoxide to the urate radical. Urate also enhanced MPO-dependent consumption of nitric oxide. In human plasma, stimulated neutrophils produced allantoin in a reaction dependent on the NADPH oxidase, MPO and superoxide. We propose that urate is a physiological substrate for MPO that is oxidized to the urate radical. The reactions of this radical with superoxide and nitric oxide provide a plausible link between urate and MPO in cardiovascular disease.


Asunto(s)
Peróxido de Hidrógeno/metabolismo , Hiperuricemia/enzimología , Neutrófilos/enzimología , Peroxidasa/metabolismo , Superóxidos/metabolismo , Alantoína/biosíntesis , Alantoína/química , Enfermedades Cardiovasculares/enzimología , Humanos , Peróxido de Hidrógeno/química , Inflamación , NADPH Oxidasas/química , NADPH Oxidasas/metabolismo , Oxidación-Reducción , Peroxidasa/química , Especificidad por Sustrato , Superóxidos/química , Ácido Úrico
20.
Horm Metab Res ; 44(13): 966-74, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22821008

RESUMEN

We conducted a longitudinal study to investigate whether increased serum gamma-glutamyltransferase independently predicts subsequent development of hyperuricemia. The study participants included 3,310 Japanese men without hyperuricemia, aged 20-54 years. The participants had annual heath examinations for 6 years to assess incident hyperuricemia (defined as serum uric acid>416.4 µmol/l and/or taking medication for hyperuricemia). The risk of incident hyperuricemia was compared in participants grouped according to their baseline serum gamma-glutamyltransferase level. During follow-up, there were 529 incident cases of hyperuricemia. A positive, dose-response relationship was observed between serum gamma-glutamyltransferase and the risk of incident hyperuricemia. The hazard ratios (95% confidence intervals) for hyperuricemia, compared with a serum gamma-glutamyltransferase level ≤19 U/l, were 1.32 (1.05-1.67) for 20-39 U/l, 1.28 (0.90-1.83) for 40-59 U/l, 1.56 (0.98-2.47) for 60-79 U/l, and 1.57 (1.02-2.41) for ≥80 U/l after adjustment for baseline serum uric acid, creatinine, total cholesterol, and glycated hemoglobin levels, ln(serum alanine aminotransferase), age, systolic blood pressure, medications for hypertension, hypercholesterolemia, and diabetes, body mass index, and smoking and exercise habits. A similar positive relationship was observed regardless of the presence or absence of alcohol drinking, obesity, metabolic disorders (any combination of hypertension, hypercholesterolemia and/or diabetes), or clinically high serum aminotransferases, without evidence of a significant interaction between increased serum gamma-glutamyltransferase and risk factors for incident hyperuricemia. These findings indicate that increased serum gamma-glutamyltransferase is an independent predictor of subsequent development of hyperuricemia.


Asunto(s)
Hiperuricemia/etiología , gamma-Glutamiltransferasa/sangre , Adulto , Humanos , Hiperuricemia/enzimología , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Riesgo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA