Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 819
Filtrer
1.
Biomed Pharmacother ; 178: 117223, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39094541

RÉSUMÉ

Hyperuricemia is with growing incidence and of high risk to develop into gout and other metabolic diseases. The key enzyme catalyzing uric acid synthesis, xanthine oxidoreductase (XOR) is a vital target for anti-hyperuricemic drugs, while XOR inhibitors characterized as both potent and safe are currently in urgent need. In this study, a novel small molecule compound, CC15009, was identified as a specific XOR inhibitor. CC15009 exerted strongest in vitro XOR inhibitory activity among current XOR inhibitors. It also showed favorable dose-dependent uric acid-lowering effects in two different XOR substrate-induced hyperuricemic mouse models, which was significantly superior than the current first-line drug, allopurinol. Mechanically, the direct binding of CC15009 against XOR was confirmed by molecular docking and SPR analysis. The inhibition mode was competitive and reversible. Besides, the potential antioxidant activity of CC15009 was indicated by its strong inhibitory activity against the oxidized isoform of XOR, which reduced ROS generation as the byproduct. Regarding the safety concerns of current XOR inhibitors, especially in cardiovascular risks, the safety of CC15009 was comprehensively evaluated. No significant abnormality was observed in the acute, subacute toxicity tests and mini-AMES test. Notably, there was no obvious inhibition of CC15009 against cardiac ion channels, including hERG, Nav1.5, Cav1.2 at the concentration of 30 µM, indicating its lower cardiovascular risk. Taken together, our results supported CC15009 as a candidate of high efficacy and safety profile to treat hyperuricemia through direct XOR inhibition.


Sujet(s)
Antienzymes , Hyperuricémie , Acide urique , Xanthine dehydrogenase , Animaux , Humains , Mâle , Souris , Allopurinol/pharmacologie , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Antienzymes/pharmacologie , Antienzymes/composition chimique , Antienzymes/effets indésirables , Antienzymes/usage thérapeutique , Hyperuricémie/traitement médicamenteux , Souris de lignée C57BL , Simulation de docking moléculaire , Acide urique/sang , Xanthine dehydrogenase/antagonistes et inhibiteurs , Xanthine dehydrogenase/métabolisme
2.
Eur J Pharmacol ; 982: 176915, 2024 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-39154822

RÉSUMÉ

Reactive oxygen species production might be prevented by xanthine oxidoreductase (XOR) inhibitors, which can cause glomerulosclerosis. We aimed to investigate whether topiroxostat, an XOR inhibitor, prevents diabetic kidney disease development in mice. Six-week-old control Institute of Cancer Research (ICR) mice and type 2 diabetic Nagoya Shibata Yasuda (NSY) mice were divided into the ICR group (ICR mice which received a lard-containing high-fat diet [HFD] based on the AIN-93G diet), NSY control group (NSY mice which received the same aforementioned diet), and NSY + topiroxostat group (NSY mice which received the same aforementioned diet with addition of 0.0012% topiroxostat). After 20 weeks, plasma biomarkers, XOR activity and oxidative stress levels, which were assessed using malondialdehyde (MDA), were measured through enzyme-linked immunosorbent assay or enzymatic methods. Renal pathology was evaluated using periodic acid-Schiff staining. Redox gene and protein expression were determined using RT-qPCR and western blotting, respectively. Plasma XOR activity was lower in NSY mice treated with topiroxostat than those without. Plasma cystatin C and creatinine levels did not differ between the ICR and NSY control groups or between the NSY control and NSY + topiroxostat groups. The NSY + topiroxostat group showed a smaller mesangial area than the NSY control group. The mRNA expression of Sod3, Prdx1, Gpx2, and Gpx3 was higher in the NSY + topiroxostat group than in the NSY control group. Renal MDA levels were lower in the NSY + topiroxostat group than in the NSY control group. Topiroxostat can reduce glomerulosclerosis, and the reduction is associated with renal oxidative markers.


Sujet(s)
Diabète de type 2 , Néphropathies diabétiques , Animaux , Néphropathies diabétiques/traitement médicamenteux , Néphropathies diabétiques/anatomopathologie , Néphropathies diabétiques/métabolisme , Souris , Diabète de type 2/traitement médicamenteux , Diabète de type 2/complications , Mâle , Stress oxydatif/effets des médicaments et des substances chimiques , Xanthine dehydrogenase/métabolisme , Xanthine dehydrogenase/antagonistes et inhibiteurs , Xanthine dehydrogenase/génétique , Souris de lignée ICR , Pyridines/pharmacologie , Pyridines/usage thérapeutique , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Rein/métabolisme , Marqueurs biologiques/sang , Nitriles
3.
J Biol Chem ; 300(8): 107524, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38960035

RÉSUMÉ

Previous studies suggest that uric acid or reactive oxygen species, products of xanthine oxidoreductase (XOR), may associate with neurodegenerative diseases. However, neither relationship has ever been firmly established. Here, we analyzed human brain samples, obtained under protocols approved by research ethics committees, and found no expression of XOR and only low levels of uric acid in various regions of the brain. In the absence of XOR, hypoxanthine will be preserved and available for incorporation into the purine salvage pathway. To clarify the importance of salvage in the brain, we tested using human-induced pluripotent stem cell-derived neuronal cells. Stable isotope analyses showed that the purine salvage pathway was more effective for ATP synthesis than purine de novo synthesis. Blood uric acid levels were related to the intracellular adenylate pool (ATP + ADP + AMP), and reduced levels of this pool result in lower uric acid levels. XOR inhibitors are related to extracellular hypoxanthine levels available for uptake into the purine salvage pathway by inhibiting the oxidation of hypoxanthine to xanthine and uric acid in various organs where XOR is present and can prevent further decreases in the intracellular adenylate pool under stress. Furthermore, adding precursors of the pentose phosphate pathway enhanced hypoxanthine uptake, indicating that purine salvage is activated by phosphoribosyl pyrophosphate replenishment. These findings resolve previous contradictions regarding XOR products and provide new insights into clinical studies. It is suggested that therapeutic strategies maximizing maintenance of intracellular adenylate levels may effectively treat pathological conditions associated with ischemia and energy depletion.


Sujet(s)
Encéphale , Purines , Acide urique , Xanthine dehydrogenase , Humains , Purines/métabolisme , Encéphale/métabolisme , Xanthine dehydrogenase/métabolisme , Acide urique/métabolisme , Hypoxanthine/métabolisme , Mâle , Neurones/métabolisme , Femelle , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Voie des pentoses phosphates , Adulte d'âge moyen , Adénosine triphosphate/métabolisme , Sujet âgé , Adulte
4.
Yakugaku Zasshi ; 144(6): 659-674, 2024.
Article de Japonais | MEDLINE | ID: mdl-38825475

RÉSUMÉ

Serum urate levels are determined by the balance between uric acid production and uric acid excretion capacity from the kidneys and intestinal tract. Dysuricemia, including hyperuricemia and hypouricemia, develops when the balance shifts towards an increase or a decrease in the uric acid pool. Hyperuricemia is mostly a multifactorial genetic disorder involving several disease susceptibility genes and environmental factors. Hypouricemia, on the other hand, is caused by genetic abnormalities. The main genes involved in dysuricemia are xanthine oxidoreductase, an enzyme that produces uric acid, and the urate transporters urate transporter 1/solute carrier family 22 member 12 (URAT1/SLC22A12), glucose transporter 9/solute carrier family 2 member 9 (GLUT9/SLC2A9) and ATP binding cassette subfamily G member 2 (ABCG2). Deficiency of xanthine oxidoreductase results in xanthinuria, a rare disease with marked hypouricemia. Xanthinuria can be due to a single deficiency of xanthine oxidoreductase or in combination with aldehyde oxidase deficiency as well. The latter is caused by a deficiency in molybdenum cofactor sulfurase, which is responsible for adding sulphur atoms to the molybdenum cofactor required for xanthine oxidoreductase and aldehyde oxidase to exert their action. URAT1/SLC22A12 and GLUT9/SLC2A9 are involved in urate reabsorption and their deficiency leads to renal hypouricemia, a condition that is common in Japanese due to URAT1/SLC22A12 deficiency. On the other hand, ABCG2 is involved in the secretion of urate, and many Japanese have single nucleotide polymorphisms that result in its reduced function, leading to hyperuricemia. In particular, severe dysfunction of ABCG2 leads to hyperuricemia with reduced extrarenal excretion.


Sujet(s)
Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP , Transporteurs de glucose par diffusion facilitée , Hyperuricémie , Protéines tumorales , Transporteurs d'anions organiques , Acide urique , Xanthine dehydrogenase , Humains , Hyperuricémie/étiologie , Hyperuricémie/métabolisme , Hyperuricémie/génétique , Acide urique/métabolisme , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/génétique , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/métabolisme , Transporteurs d'anions organiques/métabolisme , Transporteurs d'anions organiques/génétique , Transporteurs de glucose par diffusion facilitée/métabolisme , Transporteurs de glucose par diffusion facilitée/génétique , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Xanthine dehydrogenase/métabolisme , Xanthine dehydrogenase/génétique , Xanthine dehydrogenase/déficit , Animaux , Transporteurs de cations organiques/génétique , Transporteurs de cations organiques/métabolisme , Erreurs innées du transport tubulaire rénal/génétique , Erreurs innées du transport tubulaire rénal/étiologie , Erreurs innées du transport tubulaire rénal/métabolisme , Calculs urinaires/étiologie , Calculs urinaires/métabolisme , Calculs urinaires/génétique , Erreurs innées du métabolisme
5.
Sci Rep ; 14(1): 11167, 2024 05 15.
Article de Anglais | MEDLINE | ID: mdl-38750091

RÉSUMÉ

Xanthine oxidoreductase (XOR) contributes to reactive oxygen species production. We investigated the cytoprotective mechanisms of XOR inhibition against high glucose (HG)-induced glomerular endothelial injury, which involves activation of the AMP-activated protein kinase (AMPK). Human glomerular endothelial cells (GECs) exposed to HG were subjected to febuxostat treatment for 48 h and the expressions of AMPK and its associated signaling pathways were evaluated. HG-treated GECs were increased xanthine oxidase/xanthine dehydrogenase levels and decreased intracellular AMP/ATP ratio, and these effects were reversed by febuxostat treatment. Febuxostat enhanced the phosphorylation of AMPK, the activation of peroxisome proliferator-activated receptor (PPAR)-gamma coactivator (PGC)-1α and PPAR-α and suppressed the phosphorylation of forkhead box O (FoxO)3a in HG-treated GECs. Febuxostat also decreased nicotinamide adenine dinucleotide phosphate oxidase (Nox)1, Nox2, and Nox4 expressions; enhanced superoxide dismutase activity; and decreased malondialdehyde levels in HG-treated GECs. The knockdown of AMPK inhibited PGC-1α-FoxO3a signaling and negated the antioxidant effects of febuxostat in HG-treated GECs. Despite febuxostat administration, the knockdown of hypoxanthine phosphoribosyl transferase 1 (HPRT1) also inhibited AMPK-PGC-1α-FoxO3a in HG-treated GECs. XOR inhibition alleviates oxidative stress by activating AMPK-PGC-1α-FoxO3a signaling through the HPRT1-dependent purine salvage pathway in GECs exposed to HG conditions.


Sujet(s)
AMP-Activated Protein Kinases , Atteinte rénale aigüe , Cellules endothéliales , Glucose , Purines , Xanthine dehydrogenase , Humains , AMP-Activated Protein Kinases/effets des médicaments et des substances chimiques , AMP-Activated Protein Kinases/métabolisme , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Fébuxostat/pharmacologie , Glucose/métabolisme , Glomérule rénal/métabolisme , Glomérule rénal/anatomopathologie , Glomérule rénal/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Purines/pharmacologie , Espèces réactives de l'oxygène/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Xanthine dehydrogenase/antagonistes et inhibiteurs , Xanthine dehydrogenase/métabolisme , Atteinte rénale aigüe/induit chimiquement , Atteinte rénale aigüe/métabolisme
6.
Eur J Med Chem ; 271: 116407, 2024 May 05.
Article de Anglais | MEDLINE | ID: mdl-38663283

RÉSUMÉ

Xanthine oxidoreductase (XOR) and uric acid transporter 1 (URAT1) are two most widely studied targets involved in production and reabsorption of uric acid, respectively. Marketed drugs almost target XOR or URAT1, but sometimes, single agents might not achieve aim of lowering uric acid to ideal value in clinic. Thus, therapeutic strategies of combining XOR inhibitors with uricosuric drugs were proposed and implemented. Based on our initial work of virtual screening, A and B were potential hits for dual-targeted inhibitors on XOR/URAT1. By docking A/B with XOR/URAT1 respectively, compounds I1-7 were designed to get different degree of inhibition effect on XOR and URAT1, and I7 showed the best inhibitory effect on XOR (IC50 = 0.037 ± 0.001 µM) and URAT1 (IC50 = 546.70 ± 32.60 µM). Further docking research on I7 with XOR/URAT1 led to the design of compounds II with the significantly improved inhibitory activity on XOR and URAT1, such as II11 and II15. Especially, for II15, the IC50 of XOR is 0.006 ± 0.000 µM, superior to that of febuxostat (IC50 = 0.008 ± 0.000 µM), IC50 of URAT1 is 12.90 ± 2.30 µM, superior to that of benzbromarone (IC50 = 27.04 ± 2.55 µM). In acute hyperuricemia mouse model, II15 showed significant uric acid lowering effect. The results suggest that II15 had good inhibitory effect on XOR/URAT1, with the possibility for further investigation in in-vivo models of hyperuricemia.


Sujet(s)
Conception de médicament , Antienzymes , Transporteurs d'anions organiques , Transporteurs de cations organiques , Pyridines , Animaux , Pyridines/pharmacologie , Pyridines/composition chimique , Pyridines/synthèse chimique , Souris , Humains , Relation structure-activité , Transporteurs d'anions organiques/antagonistes et inhibiteurs , Transporteurs d'anions organiques/métabolisme , Transporteurs de cations organiques/antagonistes et inhibiteurs , Transporteurs de cations organiques/métabolisme , Antienzymes/pharmacologie , Antienzymes/synthèse chimique , Antienzymes/composition chimique , Structure moléculaire , Simulation de docking moléculaire , Xanthine dehydrogenase/antagonistes et inhibiteurs , Xanthine dehydrogenase/métabolisme , Relation dose-effet des médicaments , Hyperuricémie/traitement médicamenteux , Hyperuricémie/métabolisme , Mâle , Acide urique/métabolisme
7.
Int J Mol Sci ; 25(5)2024 Mar 03.
Article de Anglais | MEDLINE | ID: mdl-38474193

RÉSUMÉ

Upregulation of free radical-generating NADPH oxidases (NOX), xanthine oxidoreductase (XOR), and neutrophil infiltration-induced, NOX2-mediated respiratory burst contribute to renal ischemia-reperfusion injury (IRI), but their roles may depend on the severity of IRI. We investigated the role of NOX, XOR, and neutrophils in developing IRI of various severities. C57BL/6 and Mcl-1ΔMyelo neutrophil-deficient mice were used. Oxidases were silenced by RNA interference (RNAi) or pharmacologically inhibited. Kidney function, morphology, immunohistochemistry and mRNA expression were assessed. After reperfusion, the expression of NOX enzymes and XOR increased until 6 h and from 15 h, respectively, while neutrophil infiltration was prominent from 3 h. NOX4 and XOR silencing or pharmacological XOR inhibition did not protect the kidney from IRI. Attenuation of NOX enzyme-induced oxidative stress by apocynin and neutrophil deficiency improved kidney function and ameliorated morphological damage after mild but not moderate/severe IRI. The IR-induced postischemic renal functional impairment (BUN, Lcn-2), tubular necrosis score, inflammation (TNF-α, F4/80), and decreases in the antioxidant enzyme (GPx3) mRNA expression were attenuated by both apocynin and neutrophil deficiency. Inhibition of NOX enzyme-induced oxidative stress or the lack of infiltration by NOX2-expressing neutrophils can attenuate reperfusion injury after mild but not moderate/severe renal IR.


Sujet(s)
Acétophénones , Atteinte rénale aigüe , Lésion d'ischémie-reperfusion , Souris , Animaux , NADPH oxidase/métabolisme , Granulocytes neutrophiles/métabolisme , Souris de lignée C57BL , Rein/métabolisme , Lésion d'ischémie-reperfusion/génétique , Xanthine dehydrogenase/métabolisme , ARN messager
8.
Clin Sci (Lond) ; 138(5): 269-288, 2024 03 06.
Article de Anglais | MEDLINE | ID: mdl-38358003

RÉSUMÉ

The development of the kidney involves essential cellular processes, such as cell proliferation and differentiation, which are led by interactions between multiple signaling pathways. Xanthine dehydrogenase (XDH) catalyzes the reaction producing uric acid in the purine catabolism, which plays a multifaceted role in cellular metabolism. Our previous study revealed that the genetic ablation of the Xdh gene in rats leads to smaller kidneys, kidney damage, decline of renal functions, and failure to thrive. Rats, unlike humans, continue their kidney development postnatally. Therefore, we explored whether XDH plays a critical role in kidney development using SS-/- rats during postnatal development phase. XDH expression was significantly increased from postnatal day 5 to 15 in wild-type but not homozygote rat kidneys. The transcriptomic profile of renal tissue revealed several dysregulated pathways due to the lack of Xdh expression with the remodeling in inflammasome, purinergic signaling, and redox homeostasis. Further analysis suggested that lack of Xdh affects kidney development, likely via dysregulation of epidermal growth factor and its downstream STAT3 signaling. The present study showed that Xdh is essential for kidney maturation. Our data, alongside the previous research, suggests that loss of Xdh function leads to developmental issues, rendering them vulnerable to kidney diseases in adulthood.


Sujet(s)
Rein , Xanthine dehydrogenase , Humains , Rats , Animaux , Xanthine dehydrogenase/génétique , Xanthine dehydrogenase/métabolisme , Rein/métabolisme , Acide urique
9.
Curr Hypertens Rev ; 20(1): 10-22, 2024.
Article de Anglais | MEDLINE | ID: mdl-38318826

RÉSUMÉ

Xanthine oxidoreductase (XOR) is a rate-limiting enzyme in the formation of uric acid (UA) and is involved in the generation of reactive oxygen species (ROS). Overproduction of ROS has been linked to the pathogenesis of hypertension, atherosclerosis, and cardiovascular disease, with multiple studies over the last 30 years demonstrating that XOR inhibition is beneficial. The involvement of XOR and its constituents in the advancement of chronic inflammation and ROS, which are responsible for endothelial dysfunction, is the focus of this evidence-based review. An overabundance of XOR products and ROS appears to drive the inflammatory response, resulting in significant endothelium damage. It has also been demonstrated that XOR activity and ED are connected. Diabetes, hypertension, and cardiovascular disease are all associated with endothelial dysfunction. ROS mainly modifies the activity of vascular cells and can be important in normal vascular physiology as well as the development of vascular disease. Suppressing XOR activity appears to decrease endothelial dysfunction, probably because it lessens the generation of reactive oxygen species and the oxidative stress brought on by XOR. Although there has long been a link between higher vascular XOR activity and worse clinical outcomes, new research suggests a different picture in which positive results are mediated by XOR enzymatic activity. Here in this study, we aimed to review the association between XOR and vascular endothelial dysfunction. The prevention and treatment approaches against vascular endothelial dysfunction in atherosclerotic disease.


Sujet(s)
Endothélium vasculaire , Stress oxydatif , Espèces réactives de l'oxygène , Xanthine dehydrogenase , Humains , Xanthine dehydrogenase/métabolisme , Endothélium vasculaire/physiopathologie , Endothélium vasculaire/métabolisme , Endothélium vasculaire/enzymologie , Espèces réactives de l'oxygène/métabolisme , Animaux , Hypertension artérielle/physiopathologie , Hypertension artérielle/enzymologie , Hypertension artérielle/métabolisme , Maladies cardiovasculaires/physiopathologie , Maladies cardiovasculaires/enzymologie , Maladies cardiovasculaires/métabolisme , Acide urique/métabolisme , Acide urique/sang , Antienzymes/pharmacologie
10.
Inflammation ; 47(4): 1142-1155, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38206514

RÉSUMÉ

Xanthine oxidoreductase (XOR) serves as the primary source of hydrogen peroxide and superoxide anions in the intestinal mucosa. However, its specific contribution to the progression of colonic disease remains unclear. In this study, we investigated the role of XOR in ulcerative colitis (UC) and attempted to identify the underlying mechanisms. We used the dextran sulfate sodium (DSS)-induced mouse model to mimic UC and observed that XOR inhibitors, allopurinol and diphenyleneiodonium sulfate (DPI), significantly alleviated UC in mice. In addition, treatment with cobalt chloride (CoCl2) and 1% O2 increased the expression of XOR and induced DNA oxidative damage in colonic epithelial cells. Furthermore, we identified that XOR accumulation in the nucleus may directly cause DNA oxidative damage and regulates HIF1α protein levels. In addition, allopurinol effectively protected colon epithelial cells from CoCl2-induced DNA damage. Altogether, our data provided evidence that XOR could induce DNA damage under hypoxic conditions, indicating a significant role of XOR in the initiation and early development of colitis-associated colorectal cancer (CAC).


Sujet(s)
Côlon , Altération de l'ADN , Cellules épithéliales , Régulation positive , Xanthine dehydrogenase , Xanthine dehydrogenase/métabolisme , Xanthine dehydrogenase/antagonistes et inhibiteurs , Animaux , Souris , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Côlon/anatomopathologie , Côlon/métabolisme , Côlon/effets des médicaments et des substances chimiques , Cobalt/toxicité , Cobalt/pharmacologie , Humains , Colite/induit chimiquement , Colite/métabolisme , Colite/anatomopathologie , Rectocolite hémorragique/induit chimiquement , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/anatomopathologie , Allopurinol/pharmacologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Sulfate dextran/toxicité , Hypoxie cellulaire/physiologie , Souris de lignée C57BL , Hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme
11.
FEBS J ; 291(3): 527-546, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-37899720

RÉSUMÉ

Xanthine oxidoreductase (XOR) catalyzes the oxidation of purines (hypoxanthine and xanthine) to uric acid. XOR is widely used in various therapeutic and biotechnological applications. In this study, we characterized the biophysical and mechanistic properties of a novel bacterial XOR from Sulfobacillus acidophilus TPY (SaXOR). Our results showed that SaXOR is a heterotrimer consisting of three subunits, namely XoA, XoB, and XoC, which denote the molybdenum cofactor (Moco), 2Fe-2S, and FAD-binding domains, respectively. XoC was found to be stable when co-expressed with XoB, forming an XoBC complex. Furthermore, we prepared a fusion of XoB and XoC via a flexible linker (fusXoBC) and evaluated its function in comparison to that of XoBC. Spectroscopic analysis revealed that XoB harbors two 2Fe-2S clusters, whereas XoC bears a single-bound FAD cofactor. Electron transfer from reduced forms of XoC, XoBC, and fusXoBC to molecular oxygen (O2 ) during oxidative half-reaction yielded no flavin semiquinones, implying ultrafast single-electron transfer from 2Fe-2Sred to FAD. In the presence of XoA, XoBC and fusXoBC exhibited comparable XoA affinity and exploited a shared overall mechanism. Nonetheless, the linkage may accelerate the two-step, single-electron transfer cascade from 2Fe-2Sred to FAD while augmenting protein stability. Collectively, our findings provide novel insights into SaXOR properties and oxidation mechanisms divergent from prior mammalian and bacterial XOR paradigms.


Sujet(s)
Clostridiales , Ferrosulfoprotéines , Xanthine dehydrogenase , Animaux , Xanthine dehydrogenase/génétique , Xanthine dehydrogenase/métabolisme , Fer/métabolisme , Oxydoréduction , Flavines/métabolisme , Soufre/métabolisme , Ferrosulfoprotéines/métabolisme , Mammifères/métabolisme
12.
Redox Biol ; 67: 102864, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37713777

RÉSUMÉ

Several rare genetic variations of human XDH have been shown to alter xanthine oxidoreductase (XOR) activity leading to impaired purine catabolism. However, XOR is a multi-functional enzyme that depending upon the environmental conditions also expresses oxidase activity leading to both O2·- and H2O2 and nitrite (NO2-) reductase activity leading to nitric oxide (·NO). Since these products express important, and often diametrically opposite, biological activity, consideration of the impact of XOR mutations in the context of each aspect of the biochemical activity of the enzyme is needed to determine the potential full impact of these variants. Herein, we show that known naturally occurring hXDH mutations do not have a uniform impact upon the biochemical activity of the enzyme in terms of uric acid (UA), reactive oxygen species (ROS) and nitric oxide ·NO formation. We show that the His1221Arg mutant, in the presence of xanthine, increases UA, O2·- and NO generation compared to the WT, whilst the Ile703Val increases UA and ·NO formation, but not O2·-. We speculate that this change in the balance of activity of the enzyme is likely to endow those carrying these mutations with a harmful or protective influence over health that may explain the current equipoise underlying the perceived importance of XDH mutations. We also show that, in presence of inorganic NO2-, XOR-driven O2·- production is substantially reduced. We suggest that targeting enzyme activity to enhance the NO2--reductase profile in those carrying such mutations may provide novel therapeutic options, particularly in cardiovascular disease.


Sujet(s)
Nitrites , Xanthine dehydrogenase , Humains , Xanthine dehydrogenase/génétique , Xanthine dehydrogenase/métabolisme , Nitrites/métabolisme , Monoxyde d'azote/métabolisme , Oxidoreductases/métabolisme , Dioxyde d'azote , Peroxyde d'hydrogène , Oxydoréduction , Acide urique/métabolisme , Mutation , Xanthine oxidase/métabolisme
13.
Redox Biol ; 67: 102866, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37703667

RÉSUMÉ

We recently reported a previously unknown salutary role for xanthine oxidoreductase (XOR) in intravascular heme overload whereby hepatocellular export of XOR to the circulation was identified as a seminal step in affording protection. However, the cellular signaling and export mechanisms underpinning this process were not identified. Here, we present novel data showing hepatocytes upregulate XOR expression/protein abundance and actively release it to the extracellular compartment following exposure to hemopexin-bound hemin, hemin or free iron. For example, murine (AML-12 cells) hepatocytes treated with hemin (10 µM) exported XOR to the medium in the absence of cell death or loss of membrane integrity (2.0 ± 1.0 vs 16 ± 9 µU/mL p < 0.0001). The path of exocytosis was found to be noncanonical as pretreatment of the hepatocytes with Vaculin-1, a lysosomal trafficking inhibitor, and not Brefeldin A inhibited XOR release and promoted intracellular XOR accumulation (84 ± 17 vs 24 ± 8 hemin vs 5 ± 3 control µU/mg). Interestingly, free iron (Fe2+ and Fe3+) induced similar upregulation and release of XOR compared to hemin. Conversely, concomitant treatment with hemin and the classic transition metal chelator DTPA (20 µM) or uric acid completely blocked XOR release (p < 0.01). Our previously published time course showed XOR release from hepatocytes likely required transcriptional upregulation. As such, we determined that both Sp1 and NF-kB were acutely activated by hemin treatment (∼2-fold > controls for both, p < 0.05) and that silencing either or TLR4 with siRNA prevented hemin-induced XOR upregulation (p < 0.01). Finally, to confirm direct action of these transcription factors on the Xdh gene, chromatin immunoprecipitation was performed indicating that hemin significantly enriched (∼5-fold) both Sp1 and NF-kB near the transcription start site. In summary, our study identified a previously unknown pathway by which XOR is upregulated via SP1/NF-kB and subsequently exported to the extracellular environment. This is, to our knowledge, the very first study to demonstrate mechanistically that XOR can be specifically targeted for export as the seminal step in a compensatory response to heme/Fe overload.


Sujet(s)
Hémine , Xanthine dehydrogenase , Animaux , Souris , Xanthine dehydrogenase/génétique , Xanthine dehydrogenase/métabolisme , Hémine/pharmacologie , Fer , Facteur de transcription NF-kappa B , Hème , Hépatocytes/métabolisme
14.
Microbiol Spectr ; 11(4): e0481422, 2023 08 17.
Article de Anglais | MEDLINE | ID: mdl-37458582

RÉSUMÉ

The xanthine oxidoreductase (XOR) family are metal-containing enzymes that use the molybdenum cofactor (Moco), 2Fe-2S clusters, and flavin adenine dinucleotide (FAD) for their catalytic activity. This large molybdoenzyme family includes xanthine, aldehyde, and CO dehydrogenases. XORs are widely distributed from bacteria to humans due to their key roles in the catabolism of purines, aldehydes, drugs, and xenobiotics, as well as interconversions between CO and CO2. Assessing the effect of excess metals on the Rubrivivax gelatinosus bacterium, we found that exposure to copper (Cu) or cadmium (Cd) caused a dramatic decrease in the activity of a high-molecular-weight soluble complex exhibiting nitroblue tetrazolium reductase activity. Mass spectrometry and genetic analyses showed that the complex corresponds to a putative CO dehydrogenase (pCOD). Using mutants that accumulate either Cu+ or Cd2+ in the cytoplasm, we show that Cu+ or Cd2+ is a potent inhibitor of XORs (pCOD and the xanthine dehydrogenase [XDH]) in vivo. This is the first in vivo demonstration that Cu+ affects Moco-containing enzymes. The specific inhibitory effect of these compounds on the XOR activity is further supported in vitro by direct addition of competing metals to protein extracts. Moreover, emphasis is given on the inhibitory effect of Cu on bovine XOR, showing that the XOR family could be a common target of Cu. Given the conservation of XOR structure and function across the tree of life, we anticipate that our findings could be transferable to other XORs and organisms. IMPORTANCE The high toxicity of Cu, Cd, Pb, As, and other metals arises from their ability to cross membranes and target metalloenzymes in the cytoplasm. Identifying these targets provides insights into the toxicity mechanisms. The vulnerability of metalloenzymes arises from the accessibility of their cofactors to ions. Accordingly, many enzymes whose cofactors are solvent exposed are likely to be targets of competing metals. Here, we describe for the first time, with in vivo and in vitro experiments, a direct effect of excess Cu on the xanthine oxidoreductase family (XOR/XDH/pCOD). We show that toxic metal affects these Moco enzymes, and we suggest that access to the Moco center by Cu ions could explain the Cu inhibition of XORs in living organisms. Human XOR activity is associated with hyperuricemia, xanthinuria, gout arthritis, and other diseases. Our findings in vivo highlight XOR as a Cu target and thus support the potential use of Cu in metal-based therapeutics against these diseases.


Sujet(s)
Métalloprotéines , Xanthine dehydrogenase , Animaux , Bovins , Humains , Xanthine dehydrogenase/composition chimique , Xanthine dehydrogenase/génétique , Xanthine dehydrogenase/métabolisme , Cadmium/toxicité , Métaux
15.
Molecules ; 28(11)2023 May 30.
Article de Anglais | MEDLINE | ID: mdl-37298917

RÉSUMÉ

The author will outline the research history of the main issues addressed in this paper. The author has worked on this research himself. XDH, which is responsible for purine degradation, is present in various organisms. However, conversion to XO only occurs in mammals. The molecular mechanism of this conversion was elucidated in this study. The physiological and pathological significance of this conversion is presented. Finally, enzyme inhibitors were successfully developed, two of which are used as therapeutic agents for gout. Their wide application potential is also discussed.


Sujet(s)
Xanthine dehydrogenase , Xanthine oxidase , Animaux , Xanthine oxidase/métabolisme , Xanthine dehydrogenase/métabolisme , Antienzymes/pharmacologie , Découverte de médicament , Mammifères/métabolisme
16.
J Chem Inf Model ; 63(13): 4190-4206, 2023 07 10.
Article de Anglais | MEDLINE | ID: mdl-37319436

RÉSUMÉ

Xanthine oxidoreductase (XOR) is an enzyme found in various organisms. It converts hypoxanthine to xanthine and urate, which are crucial steps in purine elimination in humans. Elevated uric acid levels can lead to conditions like gout and hyperuricemia. Therefore, there is significant interest in developing drugs that target XOR for treating these conditions and other diseases. Oxipurinol, an analogue of xanthine, is a well-known inhibitor of XOR. Crystallographic studies have revealed that oxipurinol directly binds to the molybdenum cofactor (MoCo) in XOR. However, the precise details of the inhibition mechanism are still unclear, which would be valuable for designing more effective drugs with similar inhibitory functions. In this study, molecular dynamics and quantum mechanics/molecular mechanics calculations are employed to investigate the inhibition mechanism of XOR by oxipurinol. The study examines the structural and dynamic effects of oxipurinol on the pre-catalytic structure of the metabolite-bound system. Our results provide insights on the reaction mechanism catalyzed by the MoCo center in the active site, which aligns well with experimental findings. Furthermore, the results provide insights into the residues surrounding the active site and propose an alternative mechanism for developing alternative covalent inhibitors.


Sujet(s)
Métalloprotéines , Oxipurinol , Humains , Xanthine dehydrogenase/composition chimique , Xanthine dehydrogenase/métabolisme , Xanthine/métabolisme , Acide urique/métabolisme , Coenzymes/métabolisme , Métalloprotéines/composition chimique
17.
Hypertens Res ; 46(10): 2293-2301, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37258622

RÉSUMÉ

Serum uric acid (UA) level is associated with the high cumulative incidence or prevalence of coronary artery disease (CAD), and hyperuricemia is considered as an independent risk marker for CAD. Sleep-disordered breathing (SDB) is also associated with an increased risk of CAD. Several studies have shown that SDB is associated with hyperuricemia, but the mechanisms are unclear. We measured serum levels of UA and xanthine oxidoreductase (XOR) activity and urinary levels of 8-hydroxy-2'-deoxyguanosine (8-OHdG), all of which were assessed at 6 p.m. and the following 6 a.m. in males with CAD. In addition, nocturnal pulse oximetry was performed for the night. Overall 32 eligible patients with CAD were enrolled. Serum UA levels significantly increased overnight. (5.32 ± 0.98 mg/dl to 5.46 ± 1.02 mg/dl, p < 0.001) Moreover, XOR activity and urinary 8-OHdG levels significantly increased from 6 p.m. to 6 a.m. Furthermore, 3% Oxygen desaturation index (ODI) was correlated with the overnight changes in XOR activity (r = 0.36, P = 0.047) and urinary 8-OHdG levels (r = 0.41, P = 0.02). In addition, 3%ODI was independently correlated with the changes in XOR activity (correlation coefficient, 0.36; P = 0.047) and 8-OHdG (partial correlation coefficient, 0.63; P = 0.004) in multivariable analyses. SDB severity was associated with the overnight changes in XOR activity and urinary 8-OHdG, suggesting that SDB may be associated with oxidative stress via UA production. This trial is registered at University Hospital Medical Information Network (UMIN), number: UMIN000021624.


Sujet(s)
Maladie des artères coronaires , Hyperuricémie , Syndromes d'apnées du sommeil , Mâle , Humains , Maladie des artères coronaires/complications , Acide urique , Xanthine dehydrogenase/métabolisme , Hyperuricémie/complications , Syndromes d'apnées du sommeil/complications , Stress oxydatif
18.
Cells ; 12(8)2023 04 15.
Article de Anglais | MEDLINE | ID: mdl-37190079

RÉSUMÉ

Oxidative stress is a contributing factor to Parkinson's disease (PD). Considering the prevalence of sporadic PD, environmental exposures are postulated to increase reactive oxygen species and either incite or exacerbate neurodegeneration. We previously determined that exposure to the common soil bacterium, Streptomyces venezuelae (S. ven), enhanced oxidative stress and mitochondrial dysfunction in Caenorhabditis elegans, leading to dopaminergic (DA) neurodegeneration. Here, S. ven metabolite exposure in C. elegans was followed by RNA-Seq analysis. Half of the differentially identified genes (DEGs) were associated with the transcription factor DAF-16 (FOXO), which is a key node in regulating stress response. Our DEGs were enriched for Phase I (CYP) and Phase II (UGT) detoxification genes and non-CYP Phase I enzymes associated with oxidative metabolism, including the downregulated xanthine dehydrogenase gene, xdh-1. The XDH-1 enzyme exhibits reversible interconversion to xanthine oxidase (XO) in response to calcium. S. ven metabolite exposure enhanced XO activity in C. elegans. The chelation of calcium diminishes the conversion of XDH-1 to XO and results in neuroprotection from S. ven exposure, whereas CaCl2 supplementation enhanced neurodegeneration. These results suggest a defense mechanism that delimits the pool of XDH-1 available for interconversion to XO, and associated ROS production, in response to metabolite exposure.


Sujet(s)
Caenorhabditis elegans , Xanthine dehydrogenase , Animaux , Xanthine dehydrogenase/métabolisme , Caenorhabditis elegans/métabolisme , Calcium/métabolisme , Xanthine oxidase/métabolisme , Espèces réactives de l'oxygène/métabolisme
19.
Int J Mol Sci ; 24(8)2023 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-37108190

RÉSUMÉ

Purines, such as adenine and guanine, perform several important functions in the cell. They are found in nucleic acids; are structural components of some coenzymes, including NADH and coenzyme A; and have a crucial role in the modulation of energy metabolism and signal transduction. Moreover, purines have been shown to play an important role in the physiology of platelets, muscles, and neurotransmission. All cells require a balanced number of purines for growth, proliferation, and survival. Under physiological conditions, enzymes involved in purines metabolism maintain a balanced ratio between their synthesis and degradation in the cell. In humans, the final product of purine catabolism is uric acid, while most other mammals possess the enzyme uricase that converts uric acid to allantoin, which can be easily eliminated with urine. During the last decades, hyperuricemia has been associated with a number of human extra-articular diseases (in particular, the cardiovascular ones) and their clinical severity. In this review, we go through the methods of investigation of purine metabolism dysfunctions, looking at the functionality of xanthine oxidoreductase and the formation of catabolites in urine and saliva. Finally, we discuss how these molecules can be used as markers of oxidative stress.


Sujet(s)
Purines , Acide urique , Animaux , Humains , Acide urique/métabolisme , Purines/métabolisme , Adénine , Guanine/métabolisme , Xanthine dehydrogenase/métabolisme , Mammifères/métabolisme
20.
Nat Metab ; 5(4): 607-625, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-37024752

RÉSUMÉ

The lifetime risk of kidney disease in people with diabetes is 10-30%, implicating genetic predisposition in the cause of diabetic kidney disease (DKD). Here we identify an expression quantitative trait loci (QTLs) in the cis-acting regulatory region of the xanthine dehydrogenase, or xanthine oxidoreductase (Xor), a binding site for C/EBPß, to be associated with diabetes-induced podocyte loss in DKD in male mice. We examine mouse inbred strains that are susceptible (DBA/2J) and resistant (C57BL/6J) to DKD, as well as a panel of recombinant inbred BXD mice, to map QTLs. We also uncover promoter XOR orthologue variants in humans associated with high risk of DKD. We introduced the risk variant into the 5'-regulatory region of XOR in DKD-resistant mice, which resulted in increased Xor activity associated with podocyte depletion, albuminuria, oxidative stress and damage restricted to the glomerular endothelium, which increase further with type 1 diabetes, high-fat diet and ageing. Therefore, differential regulation of Xor contributes to phenotypic consequences with diabetes and ageing.


Sujet(s)
Diabète , Néphropathies diabétiques , Humains , Mâle , Souris , Animaux , Néphropathies diabétiques/génétique , Xanthine dehydrogenase/génétique , Xanthine dehydrogenase/métabolisme , Prédisposition génétique à une maladie , Souris de lignée DBA , Souris de lignée C57BL
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE