Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 182
Filter
1.
Eur J Pharmacol ; 982: 176915, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39154822

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Animals , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Diabetic Nephropathies/metabolism , Mice , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/complications , Male , Oxidative Stress/drug effects , Xanthine Dehydrogenase/metabolism , Xanthine Dehydrogenase/antagonists & inhibitors , Xanthine Dehydrogenase/genetics , Mice, Inbred ICR , Pyridines/pharmacology , Pyridines/therapeutic use , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Biomarkers/blood , Nitriles
2.
Biomed Pharmacother ; 178: 117223, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39094541

ABSTRACT

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.


Subject(s)
Enzyme Inhibitors , Hyperuricemia , Uric Acid , Xanthine Dehydrogenase , Animals , Humans , Male , Mice , Allopurinol/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/therapeutic use , Hyperuricemia/drug therapy , Mice, Inbred C57BL , Molecular Docking Simulation , Uric Acid/blood , Xanthine Dehydrogenase/antagonists & inhibitors , Xanthine Dehydrogenase/metabolism
3.
J Diabetes Investig ; 15(10): 1500-1509, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39078414

ABSTRACT

AIM/INTRODUCTION: Xanthine oxidoreductase (XOR) inhibitor treatment, which reduces reactive oxygen species (ROS) production and increases adenosine triphosphate (ATP) synthesis, has been reported to improve glycemic control. The possible protective effects of XOR inhibitor treatment on insulin secretory capacity were investigated in patients with type 2 diabetes. MATERIALS AND METHODS: This retrospective cross-sectional study included 428 patients with type 2 diabetes. Insulin secretory capacity was assessed based on fasting serum C-peptide concentration (CPR) and C-peptide index (CPI) in all subjects, while insulin resistance in non-insulin users (n = 312) was determined using the homeostasis model assessment of insulin resistance (HOMA-IR) index. RESULTS: Median values for CPR and CPI in all subjects were 2.4 ng/mL and 1.5, respectively, while that for HOMA-IR in non-insulin users was 3.2. The XOR inhibitor users (n = 72) had significantly (P < 0.001) higher CPR and CPI levels than non-users (n = 356). Multivariable regression analyses showed XOR inhibitor use was positively associated with CPR (ß = 0.153, P = 0.001) and CPI (ß = 0.144, P = 0.001). Similar results were observed in propensity score analyses. In subgroup analyses of patients with a preserved estimated glomerular filtration rate (≥60 mL/min/1.73 m2) and non-insulin users, these associations remained significant. Furthermore, the associations were significant in patients with lower (≤6.0 mg/dL) but not with higher (>6.0 mg/dL) uric acid levels (P for interaction <0.05). On the other hand, XOR inhibitor use showed no significant association with HOMA-IR. CONCLUSIONS: The results of XOR inhibitor treatment, especially a sufficient reduction in serum uric acid level, may provide protective effects on insulin secretory capacity in patients with type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Insulin Secretion , Xanthine Dehydrogenase , Humans , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Male , Female , Xanthine Dehydrogenase/antagonists & inhibitors , Retrospective Studies , Cross-Sectional Studies , Middle Aged , Insulin Secretion/drug effects , Insulin/therapeutic use , Aged , Enzyme Inhibitors/therapeutic use , C-Peptide/blood , Blood Glucose/analysis , Biomarkers/blood
4.
Sci Rep ; 14(1): 11167, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38750091

ABSTRACT

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.


Subject(s)
AMP-Activated Protein Kinases , Acute Kidney Injury , Endothelial Cells , Glucose , Purines , Xanthine Dehydrogenase , Humans , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Febuxostat/pharmacology , Glucose/metabolism , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Kidney Glomerulus/drug effects , Oxidative Stress/drug effects , Purines/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Xanthine Dehydrogenase/antagonists & inhibitors , Xanthine Dehydrogenase/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism
5.
Eur J Med Chem ; 271: 116407, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38663283

ABSTRACT

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.


Subject(s)
Drug Design , Enzyme Inhibitors , Organic Anion Transporters , Organic Cation Transport Proteins , Pyridines , Animals , Pyridines/pharmacology , Pyridines/chemistry , Pyridines/chemical synthesis , Mice , Humans , Structure-Activity Relationship , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/antagonists & inhibitors , Organic Cation Transport Proteins/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Molecular Structure , Molecular Docking Simulation , Xanthine Dehydrogenase/antagonists & inhibitors , Xanthine Dehydrogenase/metabolism , Dose-Response Relationship, Drug , Hyperuricemia/drug therapy , Hyperuricemia/metabolism , Male , Uric Acid/metabolism
6.
Inflammation ; 47(4): 1142-1155, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38206514

ABSTRACT

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).


Subject(s)
Colon , DNA Damage , Epithelial Cells , Up-Regulation , Xanthine Dehydrogenase , Xanthine Dehydrogenase/metabolism , Xanthine Dehydrogenase/antagonists & inhibitors , Animals , Mice , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Colon/pathology , Colon/metabolism , Colon/drug effects , Cobalt/toxicity , Cobalt/pharmacology , Humans , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Allopurinol/pharmacology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/drug effects , Dextran Sulfate/toxicity , Cell Hypoxia/physiology , Mice, Inbred C57BL , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
7.
J Am Soc Nephrol ; 33(2): 326-341, 2022 02.
Article in English | MEDLINE | ID: mdl-34799437

ABSTRACT

BACKGROUND: Hereditary renal hypouricemia type 1 (RHUC1) is caused by URAT1/SLC22A12 dysfunction, resulting in urolithiasis and exercise-induced AKI (EIAKI). However, because there is no useful experimental RHUC1 animal model, the precise pathophysiologic mechanisms underlying EIAKI have yet to be elucidated. We established a high HPRT activity Urat1-Uox double knockout (DKO) mouse as a novel RHUC1 animal model for investigating the cause of EIAKI and the potential therapeutic effect of xanthine oxidoreductase inhibitors (XOIs). METHODS: The novel Urat1-Uox DKO mice were used in a forced swimming test as loading exercise to explore the onset mechanism of EIAKI and evaluate related purine metabolism and renal injury parameters. RESULTS: Urat1-Uox DKO mice had uricosuric effects and elevated levels of plasma creatinine and BUN as renal injury markers, and decreased creatinine clearance observed in a forced swimming test. In addition, Urat1-Uox DKO mice had increased NLRP3 inflammasome activity and downregulated levels of Na+-K+-ATPase protein in the kidney, as Western blot analysis showed. Finally, we demonstrated that topiroxostat and allopurinol, XOIs, improved renal injury and functional parameters of EIAKI. CONCLUSIONS: Urat1-Uox DKO mice are a useful experimental animal model for human RHUC1. The pathogenic mechanism of EIAKI was found to be due to increased levels of IL-1ß via NLRP3 inflammasome signaling and Na+-K+-ATPase dysfunction associated with excessive urinary urate excretion. In addition, XOIs appear to be a promising therapeutic agent for the treatment of EIAKI.


Subject(s)
Acute Kidney Injury/drug therapy , Hypoxanthine Phosphoribosyltransferase/metabolism , Organic Anion Transporters/deficiency , Urate Oxidase/deficiency , Xanthine Dehydrogenase/antagonists & inhibitors , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Allopurinol/pharmacology , Animals , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Hypoxanthine Phosphoribosyltransferase/genetics , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nitriles/pharmacology , Organic Anion Transporters/genetics , Physical Exertion , Pyridines/pharmacology , Renal Tubular Transport, Inborn Errors/drug therapy , Renal Tubular Transport, Inborn Errors/etiology , Renal Tubular Transport, Inborn Errors/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Urate Oxidase/genetics , Urinary Calculi/drug therapy , Urinary Calculi/etiology , Urinary Calculi/metabolism
8.
Oxid Med Cell Longev ; 2021: 1470380, 2021.
Article in English | MEDLINE | ID: mdl-33854690

ABSTRACT

Uric acid is the end product of purine metabolism in humans. Hyperuricemia is a metabolic disease caused by the increased formation or reduced excretion of serum uric acid (SUA). Alterations in SUA homeostasis have been linked to a number of diseases, and hyperuricemia is the major etiologic factor of gout and has been correlated with metabolic syndrome, cardiovascular disease, diabetes, hypertension, and renal disease. Oxidative stress is usually defined as an imbalance between free radicals and antioxidants in our body and is considered to be one of the main causes of cell damage and the development of disease. Studies have demonstrated that hyperuricemia is closely related to the generation of reactive oxygen species (ROS). In the human body, xanthine oxidoreductase (XOR) catalyzes the oxidative hydroxylation of hypoxanthine to xanthine to uric acid, with the accompanying production of ROS. Therefore, XOR is considered a drug target for the treatment of hyperuricemia and gout. In this review, we discuss the mechanisms of uric acid transport and the development of hyperuricemia, emphasizing the role of oxidative stress in the occurrence and development of hyperuricemia. We also summarize recent advances and new discoveries in XOR inhibitors.


Subject(s)
Xanthine Dehydrogenase/antagonists & inhibitors , Humans , Hyperuricemia , Oxidative Stress
9.
Nutr Diabetes ; 11(1): 12, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33850106

ABSTRACT

Plasma xanthine oxidoreductase (XOR) activity is high in metabolic disorders such as diabetic mellitus, obesity, or overweight. Thus, this study investigated whether the XOR inhibitor, topiroxostat, affected body weight. Male db/db mice were fed standard diets with or without topiroxostat for 4 weeks. Body weight and food intake were constantly monitored, along with monitoring plasma biochemical markers, including insulin and XOR activity. Additionally, hepatic hypoxanthine and XOR activity were also documented. Single regression analysis was performed to determine the mechanism. Topiroxostat treatment suppressed weight gain relative to the vehicle without any impact on food intake. However, the weight of fat pads and hepatic and muscle triglyceride content did not change. Topiroxostat decreased the plasma uric acid and increased hepatic hypoxanthine in response to the inhibition of XOR activity. Plasma ketone body and free fatty acid were also increased. Moreover, fat weight was weakly associated with plasma XOR activity in the diabetic state and was negatively associated with ketone body by topiroxostat. These results suggested that topiroxostat amplified the burning of lipids and the salvage pathway, resulting in predisposing the body toward catabolism. The inhibition of plasma XOR activity may contribute to weight loss.


Subject(s)
Body Weight/drug effects , Diabetes Mellitus, Experimental/drug therapy , Enzyme Inhibitors/pharmacology , Nitriles/pharmacology , Obesity/drug therapy , Pyridines/pharmacology , Xanthine Dehydrogenase/antagonists & inhibitors , Animals , Diabetes Mellitus, Experimental/blood , Fatty Acids, Nonesterified/blood , Humans , Hypoxanthine/analysis , Insulin/blood , Liver/metabolism , Male , Mice , Mice, Obese , Obesity/blood , Triglycerides/analysis , Uric Acid/blood , Weight Gain/drug effects , Xanthine Dehydrogenase/blood
10.
Int J Mol Sci ; 22(7)2021 Mar 29.
Article in English | MEDLINE | ID: mdl-33805516

ABSTRACT

Reactive oxygen species (ROS) metabolism is regulated by the oxygen-mediated enzyme reaction and antioxidant mechanism within cells under physiological conditions. Xanthine oxidoreductase (XOR) exhibits two inter-convertible forms (xanthine oxidase (XO) and xanthine dehydrogenase (XDH)), depending on the substrates. XO uses oxygen as a substrate and generates superoxide (O2•-) in the catalytic pathway of hypoxanthine. We previously showed that superoxide dismutase 1 (SOD1) loss induced various aging-like pathologies via oxidative damage due to the accumulation of O2•- in mice. However, the pathological contribution of XO-derived O2•- production to aging-like tissue damage induced by SOD1 loss remains unclear. To investigate the pathological significance of O2•- derived from XOR in Sod1-/- mice, we generated Sod1-null and XO-type- or XDH-type-knock-in (KI) double-mutant mice. Neither XO-type- nor XDH-type KI mutants altered aging-like phenotypes, such as anemia, fatty liver, muscle atrophy, and bone loss, in Sod1-/- mice. Furthermore, allopurinol, an XO inhibitor, or apocynin, a nicotinamide adenine dinucleotide phosphate oxidase (NOX) inhibitor, failed to improve aging-like tissue degeneration and ROS accumulation in Sod1-/- mice. These results showed that XOR-mediated O2•- production is relatively uninvolved in the age-related pathologies in Sod1-/- mice.


Subject(s)
Aging/physiology , Superoxide Dismutase-1/genetics , Superoxides/metabolism , Xanthine Dehydrogenase/metabolism , Acetophenones/pharmacology , Aging/drug effects , Allopurinol/pharmacology , Anemia/genetics , Animals , Fatty Liver/genetics , Mice, Mutant Strains , Muscular Atrophy/genetics , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Superoxide Dismutase-1/metabolism , Xanthine Dehydrogenase/antagonists & inhibitors , Xanthine Dehydrogenase/genetics
11.
Biol Pharm Bull ; 43(11): 1792-1798, 2020.
Article in English | MEDLINE | ID: mdl-33132325

ABSTRACT

Xanthine and hypoxanthine are intermediate metabolites of uric acid and a source of reactive oxidative species (ROS) by xanthine oxidoreductase (XOR), suggesting that facilitating their elimination is beneficial. Since they are reabsorbed in renal proximal tubules, we investigated their reabsorption mechanism by focusing on the renal uric acid transporters URAT1 and GLUT9, and examined the effect of clinically used URAT1 inhibitor on their renal clearance when their plasma concentration is increased by XOR inhibitor. Uptake study for [3H]xanthine and [3H]hypoxanthine was performed using URAT1- and GLUT9-expressing Xenopus oocytes. Transcellular transport study for [3H]xanthine was carried out using Madin-Darby canine kidney (MDCK)II cells co-expressing URAT1 and GLUT9. In in vivo pharmacokinetic study, renal clearance of xanthine was estimated based on plasma concentration and urinary recovery. Uptake by URAT1- and GLUT9-expressing oocytes demonstrated that xanthine is a substrate of URAT1 and GLUT9, while hypoxanthine is not. Transcellular transport of xanthine in MDCKII cells co-expressing URAT1 and GLUT9 was significantly higher than those in mock cells and cells expressing URAT1 or GLUT9 alone. Furthermore, dotinurad, a URAT1 inhibitor, increased renal clearance of xanthine in rats treated with topiroxostat to inhibit XOR. It was suggested that xanthine is reabsorbed in the same manner as uric acid through URAT1 and GLUT9, while hypoxanthine is not. Accordingly, it is expected that treatment with XOR and URAT1 inhibitors will effectively decrease purine pools in the body and prevent cell injury due to ROS generated during XOR-mediated reactions.


Subject(s)
Anion Transport Proteins/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Monosaccharide Transport Proteins/metabolism , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/metabolism , Xanthine/pharmacokinetics , Animals , Anion Transport Proteins/antagonists & inhibitors , Benzothiazoles/administration & dosage , Dogs , Glucose Transport Proteins, Facilitative/genetics , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Madin Darby Canine Kidney Cells , Models, Animal , Nitriles/administration & dosage , Oocytes , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/genetics , Pyridines/administration & dosage , Rats , Rats, Wistar , Reactive Oxygen Species , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Renal Elimination/drug effects , Uric Acid/metabolism , Xanthine/blood , Xanthine/metabolism , Xanthine/urine , Xanthine Dehydrogenase/antagonists & inhibitors , Xanthine Dehydrogenase/metabolism , Xenopus laevis
12.
Medicine (Baltimore) ; 99(35): e21576, 2020 Aug 28.
Article in English | MEDLINE | ID: mdl-32871874

ABSTRACT

BACKGROUND: Cellular energetics play an important role in Parkinsons disease etiology, but no treatments directly address this deficiency. Our past research showed that treatment with febuxostat and inosine increased blood hypoxanthine and ATP in healthy adults, and a preliminary trial in 3 Parkinson's disease patients suggested some symptomatic improvements with no adverse effects. METHODS: To examine the efficacy on symptoms and safety in a larger group of Parkinsons disease patients, we conducted a single-arm, open-label trial at 5 Japanese neurology clinics and enrolled thirty patients (nmales = 11; nfemales = 19); 26 patients completed the study (nmales = 10; nfemales = 16). Each patient was administered febuxostat 20 mg and inosine 500 mg twice-per-day (after breakfast and dinner) for 8 weeks. The primary endpoint was the difference of MDS-UPDRS Part III score immediately before and after 57 days of treatment. RESULTS: Serum hypoxanthine concentrations were raised significantly after treatment (Pre = 11.4 µM; Post = 38.1 µM; P < .0001). MDS-UPDRS Part III score was significantly lower after treatment (Pre = 28.1 ±â€Š9.3; Post = 24.7 ±â€Š10.8; mean ±â€ŠSD; P = .0146). Sixteen adverse events occurred in 13/29 (44.8%) patients, including 1 serious adverse event (fracture of the second lumbar vertebra) that was considered not related to the treatment. CONCLUSIONS: The results of this study suggest that co-administration of febuxostat and inosine is relatively safe and effective for improving symptoms of Parkinsons disease patients. Further controlled trials need to be performed to confirm the symptomatic improvement and to examine the disease-modifying effect in long-term trials.


Subject(s)
Febuxostat/therapeutic use , Gout Suppressants/therapeutic use , Inosine/therapeutic use , Parkinson Disease/drug therapy , Adenosine Triphosphate/blood , Administration, Oral , Aged , Case-Control Studies , Drug Therapy, Combination , Febuxostat/administration & dosage , Febuxostat/adverse effects , Female , Gout Suppressants/administration & dosage , Gout Suppressants/adverse effects , Humans , Hypoxanthine/blood , Inosine/administration & dosage , Inosine/adverse effects , Japan/epidemiology , Male , Middle Aged , Nervous System Diseases/epidemiology , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Safety , Treatment Outcome , Xanthine Dehydrogenase/antagonists & inhibitors
13.
Clin Drug Investig ; 40(9): 847-859, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32621143

ABSTRACT

BACKGROUND AND OBJECTIVES: Topiroxostat, a selective xanthine oxidoreductase inhibitor, is used for the management of hyperuricemic patients with or without gout in Japan. Accumulating evidence has demonstrated the efficacy of topiroxostat for the treatment of hyperuricemia with or without gout. However, the safety and efficacy of topiroxostat in the clinical setting remain unclear, and there is little large-scale clinical evidence. We conducted a post-marketing observational study over 54 weeks. PATIENTS AND METHODS: Patients were centrally enrolled, and case report forms of 4491 patients were collected between April 2014 and March 2019 from 825 medical sites. RESULTS: Overall, 4329 patients were assessed for safety and 4253 patients for effectiveness. The overall incidence of adverse drug reactions was 6.95%, and the incidence rates of adverse drug reactions of gouty arthritis, hepatic dysfunction, and skin disorders, which are of special interest in this study, were 0.79%, 1.73%, and 0.95%, respectively. No case of serious gouty arthritis was observed. Serum urate levels decreased stably over time and showed a significant reduction rate at 54 weeks (21.19% ± 22.07%) and on the final visit (19.91% ± 23.35%) compared to the baseline. The rates for subjects who achieved serum uric acid levels ≤ 6.0 mg/dL at 18 and 54 weeks after administration were 43.80% and 48.28%, respectively. CONCLUSIONS: This study suggests that there is no particular concern about adverse drug reactions or the efficacy of topiroxostat for hyperuricemic patients with or without gout in a post-marketing setting in Japan.


Subject(s)
Gout Suppressants/therapeutic use , Gout/drug therapy , Hyperuricemia/drug therapy , Nitriles/therapeutic use , Product Surveillance, Postmarketing , Pyridines/therapeutic use , Xanthine Dehydrogenase/antagonists & inhibitors , Adult , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/therapeutic use , Female , Gout Suppressants/adverse effects , Humans , Incidence , Japan/epidemiology , Male , Middle Aged , Nitriles/adverse effects , Pyridines/adverse effects , Treatment Outcome , Uric Acid/blood
14.
Biomed Chromatogr ; 34(2): e4727, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31657461

ABSTRACT

A potent xanthine oxidoreductase inhibitor (LS087) was recently proved to exhibit a similar hypouricemic potency to febuxostat. A hyperuricemia model induced by potassium oxonate and hypoxanthine was proposed in specific pathogen-free male Kunming mice, and the serum urea nitrogen, creatinine and uric acid levels were measured after oral administration of LS087. Furthermore, renal histopathology was conducted by staining with hematoxylin and eosin, periodic acid-Schiff and Masson's trichrome stains, respectively. The results showed that the levels of serum urea nitrogen and uric acid significantly decreased compared with the model group, but the level of creatinine showed no significant changes. The pathological abnormalities in kidney tubules were improved after LS087 administration. Ten metabolites (M1-M10) of LS087 were identified after a single oral dosing of 10 mg/kg in rats. M6 was the primary LS087 metabolite in vivo with a pathway of methylation. The toxicity and potential risks of LS087 and its metabolites were predicted using the ProTox-II software. LS087 and the major metabolites (M2, M3, M5, M6, M7 and M8) were predicted to have no potential hepatotoxicity, but some metabolites with a total rate of <1% (M1, M4, M9, and M10) showed potential hepatotoxicity. M1 and M8 showed potential carcinogenicity. The LS087 biotransformation pathway in rat was well characterized.


Subject(s)
Enzyme Inhibitors , Hyperuricemia/metabolism , Xanthine Dehydrogenase/antagonists & inhibitors , Animals , Biotransformation , Creatinine/blood , Disease Models, Animal , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Kidney/drug effects , Kidney/metabolism , Male , Mice , Rats , Rats, Sprague-Dawley
15.
Eur J Med Chem ; 186: 111883, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31761385

ABSTRACT

As part of a continuing study, we designed and synthesized four series of 1-phenylimidazole-4-carboxylic acid derivatives as xanthine oxidoreductase (XOR) inhibitors, evaluated their in vitro inhibitory potencies against XOR and hypouricemic effects in mice, and determined their structure-activity relationships (SARs). Most of the compounds exhibited in vitro XOR inhibition at the nanomolar level. In comparison to febuxostat (half-maximal inhibitory concentration [IC50] value of 7.0 nM), compounds Ie and IVa exhibited the most promising XOR inhibitory effects with IC50 values of 8.0 and 7.2 nM, respectively. In the potassium oxonate/hypoxanthine-induced acute and long-term hyperuricemia mouse models, compounds Ie and IVa displayed significant hypouricemic potencies (P < 0.05), that were slightly weaker than and similar to febuxostat, respectively. More interestingly, both compounds showed a capacity to improve kidney damage by decreasing creatinine and urea nitrogen levels compared to the long-term hyperuricemia mouse group (P < 0.05), while febuxostat showed no significant effect.


Subject(s)
Carboxylic Acids/pharmacology , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Xanthine Dehydrogenase/antagonists & inhibitors , Animals , Carboxylic Acids/chemical synthesis , Carboxylic Acids/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Hyperuricemia/chemically induced , Hyperuricemia/drug therapy , Hyperuricemia/metabolism , Imidazoles/chemical synthesis , Imidazoles/chemistry , Mice , Mice, Inbred ICR , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Xanthine Dehydrogenase/metabolism
16.
Sci Rep ; 9(1): 17314, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31754153

ABSTRACT

The nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome mediates caspase-1 activation and IL-1ß processing and is implicated in autoinflammatory as well as other chronic inflammatory diseases. Recent studies have demonstrated that xanthine oxidoreductase (XOR) inhibition attenuated IL-1ß secretion in activated macrophages, but the detailed mechanism of inhibition remains unclear. In this study, we report that febuxostat, an inhibitor of XOR, suppressed NLRP3 inflammasome-mediated IL-1ß secretion and cell death by two mechanisms: in a mitochondrial ROS (mitoROS)-dependent and mitoROS-independent manner. MitoROS-independent effects of febuxostat were mediated by an increase of intracellular ATP and improved mitochondrial energetics via the activation of purine salvage pathway. Our findings suggest that cellular bioenergetics are important in regulating NLRP3 activation, and XOR inhibition may be clinically relevant in NLRP3-related inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Febuxostat/pharmacology , Inflammation/drug therapy , Macrophages/drug effects , Xanthine Dehydrogenase/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/therapeutic use , Cells, Cultured , Energy Metabolism/drug effects , Energy Metabolism/immunology , Febuxostat/therapeutic use , Humans , Inflammasomes/drug effects , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Primary Cell Culture , Purines/metabolism , Reactive Oxygen Species/metabolism , Xanthine Dehydrogenase/metabolism
17.
J Clin Hypertens (Greenwich) ; 21(11): 1713-1720, 2019 11.
Article in English | MEDLINE | ID: mdl-31556223

ABSTRACT

Xanthine oxidoreductase (XOR) inhibitors, such as allopurinol and febuxostat, inhibit the catalysis of serum uric acid (SUA) synthesis. In doing so, they are thought to improve vascular endothelial function in patients with hyperuricemia and cardiovascular risk by reducing increases in SUA and reactive oxygen species levels. We performed a retrospective cohort study to evaluate the effects of topiroxostat, a novel XOR inhibitor, on vascular function measured by flow-mediated dilation (FMD) on ultrasonography. In total, 23 patients with hyperuricemia were enrolled. After approximately 8 weeks, topiroxostat was associated with a significant increase in the peak percentage change in diameter (∆FMD) from 4.53% ± 2.09% to 5.54% ± 3.08% (P = .045). It also significantly reduced the SUA levels from 7.31 ± 1.43 to 5.44 ± 1.11 mg/dL (P < .001). Although further studies are needed to validate these results, it appears that topiroxostat improves vascular endothelial function in patients with hyperuricemia.


Subject(s)
Hyperuricemia , Nitriles , Pyridines , Uric Acid/blood , Vasodilation/drug effects , Xanthine Dehydrogenase/antagonists & inhibitors , Biological Availability , Blood Flow Velocity/drug effects , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Hyperuricemia/blood , Hyperuricemia/drug therapy , Hyperuricemia/physiopathology , Male , Middle Aged , Nitriles/administration & dosage , Nitriles/pharmacokinetics , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Reproducibility of Results , Retrospective Studies , Ultrasonography, Doppler/methods
18.
Mol Med ; 25(1): 40, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31438839

ABSTRACT

BACKGROUND: Clinically applied as anti-gout drugs, xanthine oxidoreductase (XOR) inhibitors, especially the potent, selective, non-purine-analog XOR inhibitors febuxostat and topiroxostat, exert organ-protective effects. We tested the hypothesis that preservation of tissue concentrations of high-energy phosphates, such as ATP and ADP, contributes to organ-protective effects through CE-TOFMS metabolomics. METHODS: Rats were subjected to 30 min of renal ischemia-reperfusion (I/R) injury 60 min after oral administration of 10 mg/kg febuxostat, 10 mg/kg topiroxostat, 50 mg/kg allopurinol, or vehicle. RESULTS: In non-purine-analog XOR inhibitor-treated groups, renal concentrations of high-energy phosphates were greater before and after I/R injury, and renal adenine compounds were less depleted by I/R injury than in the vehicle and allopurinol groups. These findings were well in accordance with the proposed hypothesis that the recomposition of high-energy phosphates is promoted by non-purine-analog XOR inhibitors via the salvage pathway through blockade of hypoxanthine catabolism, whereas non-specific inhibitory effects of allopurinol on purine/pyrimidine enzymes impede this re-synthesis process. CONCLUSIONS: This metabolic approach shed light on the physiology of the organ-protective effects of XOR inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacology , Metabolome , Purines/metabolism , Pyrimidines/metabolism , Reperfusion Injury/metabolism , Xanthine Dehydrogenase/antagonists & inhibitors , Animals , Disease Models, Animal , Male , Metabolome/drug effects , Metabolome/physiology , Metabolomics , Rats , Rats, Sprague-Dawley , Xanthine Dehydrogenase/metabolism
19.
Adv Exp Med Biol ; 1127: 59-65, 2019.
Article in English | MEDLINE | ID: mdl-31140171

ABSTRACT

Inhibition of xanthine oxidoreductase (XOR) has proven beneficial in a plethora of inflammatory disease processes due to a net reduction in pro-inflammatory oxidants and secondary nitrating species. Electrophilic nitrated fatty acid derivatives, such as nitro-oleic acid (OA-NO2) are also noted to display a broad spectrum of anti-inflammatory effects via interaction with critical signaling pathways. An alternative process in which nitrated fatty acids may extend anti-inflammatory actions is via inactivation of XOR, a process that is more effective than allo/oxypurinol-mediated inhibition. Herein, we describe the molecular aspects of nitrated fatty acid-associated inactivation of XOR, identify specificity via structure function relationships and discuss XOR as a crucial component of the anti-inflammatory portfolio of nitrated fatty acids.


Subject(s)
Fatty Acids/pharmacology , Inflammation , Nitrates/chemistry , Oxidants/chemistry , Xanthine Dehydrogenase/antagonists & inhibitors , Humans
20.
Diabetes ; 68(6): 1221-1229, 2019 06.
Article in English | MEDLINE | ID: mdl-30936145

ABSTRACT

Systemic hyperuricemia (HyUA) in obesity/type 2 diabetes facilitated by elevated activity of xanthine oxidoreductase (XOR), which is the sole source of uric acid (UA) in mammals, has been proposed to contribute to the pathogenesis of insulin resistance/dyslipidemia in obesity. Here, the effects of hepatocyte-specific ablation of Xdh, the gene encoding XOR (HXO), and whole-body pharmacologic inhibition of XOR (febuxostat) on obesity-induced insulin resistance/dyslipidemia were assessed. Deletion of hepatocyte Xdh substantially lowered liver and plasma UA concentration. When exposed to an obesogenic diet, HXO and control floxed (FLX) mice became equally obese, but systemic HyUA was absent in HXO mice. Despite this, obese HXO mice became as insulin resistant and dyslipidemic as obese FLX mice. Similarly, febuxostat dramatically lowered plasma and tissue UA and XOR activity in obese wild-type mice without altering obesity-associated insulin resistance/dyslipidemia. These data demonstrate that hepatocyte XOR activity is a critical determinant of systemic UA homeostasis, that deletion of hepatocyte Xdh is sufficient to prevent systemic HyUA of obesity, and that neither prevention nor correction of HyUA improves insulin resistance/dyslipidemia in obesity. Thus, systemic HyUA, although clearly a biomarker of the metabolic abnormalities of obesity, does not appear to be causative.


Subject(s)
Glucose/metabolism , Hepatocytes/metabolism , Hyperuricemia/genetics , Lipid Metabolism , Obesity/metabolism , Uric Acid/metabolism , Xanthine Dehydrogenase/genetics , Animals , Diet, High-Fat , Fatty Acids, Nonesterified/metabolism , Febuxostat/pharmacology , Glucose Tolerance Test , Hepatocytes/drug effects , Hyperuricemia/metabolism , Mice , Triglycerides/metabolism , Xanthine Dehydrogenase/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL