Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 187
Filter
1.
Am J Physiol Renal Physiol ; 326(6): F1004-F1015, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38634129

ABSTRACT

Humans are predisposed to gout because they lack uricase that converts uric acid to allantoin. Rodents have uricase, resulting in low basal serum uric acid. A uricase inhibitor raises serum uric acid in rodents. There were two aims of the study in polycystic kidney disease (PKD): 1) to determine whether increasing serum uric acid with the uricase inhibitor, oxonic acid, resulted in faster cyst growth and 2) to determine whether treatment with the xanthine oxidase inhibitor, oxypurinol, reduced the cyst growth caused by oxonic acid. Orthologous models of human PKD were used: PCK rats, a polycystic kidney and hepatic disease 1 (Pkhd1) gene model of autosomal recessive PKD (ARPKD) and Pkd1RC/RC mice, a hypomorphic Pkd1 gene model. In PCK rats and Pkd1RC/RC mice, oxonic acid resulted in a significant increase in serum uric acid, kidney weight, and cyst index. Mechanisms of increased cyst growth that were investigated were proinflammatory cytokines, the inflammasome, and crystal deposition in the kidney. Oxonic acid resulted in an increase in proinflammatory cytokines in the serum and kidney in Pkd1RC/RC mice. Oxonic acid did not cause activation of the inflammasome or uric acid crystal deposition in the kidney. In Pkd1RC/RC male and female mice analyzed together, oxypurinol decreased the oxonic acid-induced increase in cyst index. In summary, increasing serum uric acid by inhibiting uricase with oxonic acid results in an increase in kidney weight and cyst index in PCK rats and Pkd1RC/RC mice. The effect is independent of inflammasome activation or crystal deposition in the kidney.NEW & NOTEWORTHY This is the first reported study of uric acid measurements and xanthine oxidase inhibition in polycystic kidney disease (PKD) rodents. Raising serum uric acid with a uricase inhibitor resulted in increased kidney weight and cyst index in Pkd1RC/RC mice and PCK rats, elevated levels of proinflammatory cytokines in the serum and kidney in Pkd1RC/RC mice, and no uric acid crystal deposition or activation of the caspase-1 inflammasome in the kidney.


Subject(s)
Disease Models, Animal , Kidney , Polycystic Kidney Diseases , Urate Oxidase , Uric Acid , Animals , Uric Acid/blood , Polycystic Kidney Diseases/pathology , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/drug therapy , Kidney/pathology , Kidney/drug effects , Kidney/metabolism , Male , Oxypurinol/pharmacology , Oxonic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Rats , Female , Inflammasomes/metabolism , Cytokines/metabolism , Cytokines/blood , Mice , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism , Xanthine Oxidase/antagonists & inhibitors , Xanthine Oxidase/metabolism , Rats, Sprague-Dawley , Mice, Inbred C57BL
2.
BMC Cancer ; 23(1): 1044, 2023 Oct 30.
Article in English | MEDLINE | ID: mdl-37904102

ABSTRACT

BACKGROUND: Pancreatic cancer is a highly lethal disease with no effective treatments. Lactobacillus casei (L. casei) and Lactobacillus reuteri (L. reuteri) exhibited therapeutic effects on several cancers, but their roles in pancreatic cancer are unknown. This study aims to explore how L. casei & L. reuteri influence pancreatic cancer and the underlying mechanisms. METHODS: Pancreatic cancer cells were treated with L. casei & L. reuteri and co-cultured with macrophages in a transwell system in vitro. Pancreatic cancer xenograft model was established and L. casei & L. reuteri was used to treat mice in vivo. MTT, CCK-8 assay or immunohistochemical staining were used to determine the proliferation of pancreatic cancer cells or tumor tissues. Transwell assay was applied to test the migration and invasion of pancreatic cells. RT-qPCR was utilized to assess TLR4 and MyD88 expressions in pancreatic cells or tumor tissues. WB, immunofluorescence staining, or flow cytometry was used to evaluate the M1/M2 polarization of macrophages. Besides, the composition of gut microbiota of tumor-bearing mice was determined by 16 S rRNA sequencing, and ultra-high performance liquid chromatography-mass spectrometry (UPLC-MS) untargeted metabolomics was used to evaluate the metabolic profiles of feces. RESULTS: L. casei & L. reuteri inhibited the proliferation, migration, invasion of pancreatic cancer cells and pancreatic cancer cell-induced M2 polarization of macrophages by suppressing TLR4. Meanwhile, L. casei & L. reuteri repressed pancreatic cancer growth and promoted M1 macrophage polarization. Besides, L. casei & L. reuteri reduced fecal Alloprevotella and increased fecal azelate and glutamate in nude mice, while TLR4 inhibitor TAK-242 increased Clostridia UCG-014, azelate, uridine, methionine sulfoxide, oxypurinol, and decreased glyceryl monoester in the feces of pancreatic tumor-bearing mice. Fecal oxypurinol and glyceryl monoester levels were positively or negatively associated with gut Clostridia UCG-014 abundance, respectively. CONCLUSION: L. casei & L. reuteri alleviate pancreatic cancer by inhibiting TLR4 to promote macrophage M1 polarization and regulate gut microbial homeostasis.


Subject(s)
Gastrointestinal Microbiome , Lacticaseibacillus casei , Limosilactobacillus reuteri , Pancreatic Neoplasms , Mice , Humans , Animals , Toll-Like Receptor 4/metabolism , Mice, Nude , Chromatography, Liquid , Oxypurinol/metabolism , Oxypurinol/pharmacology , Tandem Mass Spectrometry , Macrophages/metabolism , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms
3.
Allergy ; 78(11): 2980-2993, 2023 11.
Article in English | MEDLINE | ID: mdl-37452515

ABSTRACT

Allopurinol (ALP) is a successful drug used in the treatment of gout. However, this drug has been implicated in hypersensitivity reactions that can cause severe to life-threatening reactions such as Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN). Individuals who carry the human leukocyte antigen (HLA)-B*58:01 allotype are at higher risk of experiencing a hypersensitivity reaction (odds ratios ranging from 5.62 to 580.3 for mild to severe reactions, respectively). In addition to the parent drug, the metabolite oxypurinol (OXP) is implicated in triggering T cell-mediated immunopathology via a labile interaction with HLA-B*58:01. To date, there has been limited information regarding the T-cell receptor (TCR) repertoire usage of reactive T cells in patients with ALP-induced SJS or TEN and, in particular, there are no reports examining paired αßTCRs. Here, using in vitro drug-treated PBMCs isolated from both resolved ALP-induced SJS/TEN cases and drug-naïve healthy donors, we show that OXP is the driver of CD8+ T cell-mediated responses and that drug-exposed memory T cells can exhibit a proinflammatory immunophenotype similar to T cells described during active disease. Furthermore, this response supported the pharmacological interaction with immune receptors (p-i) concept by showcasing (i) the labile metabolite interaction with peptide/HLA complexes, (ii) immunogenic complex formation at the cell surface, and (iii) lack of requirement for antigen processing to elicit drug-induced T cell responsiveness. Examination of paired OXP-induced αßTCR repertoires highlighted an oligoclonal and private clonotypic profile in both resolved ALP-induced SJS/TEN cases and drug-naïve healthy donors.


Subject(s)
Allopurinol , Stevens-Johnson Syndrome , Humans , Allopurinol/adverse effects , Oxypurinol/pharmacology , Stevens-Johnson Syndrome/genetics , CD8-Positive T-Lymphocytes , HLA-B Antigens/genetics
4.
Clin Pharmacokinet ; 61(11): 1545-1558, 2022 11.
Article in English | MEDLINE | ID: mdl-36040612

ABSTRACT

BACKGROUND AND OBJECTIVE: Previously, we developed a pharmacokinetic-pharmacodynamic model of allopurinol, oxypurinol, and biomarkers, hypoxanthine, xanthine, and uric acid, in neonates with hypoxic-ischemic encephalopathy, in which high initial biomarker levels were observed suggesting an impact of hypoxia. However, the full pharmacodynamics could not be elucidated in our previous study. The current study included additional data from the ALBINO study (NCT03162653) placebo group, aiming to characterize the dynamics of hypoxanthine, xanthine, and uric acid in neonates with hypoxic-ischemic encephalopathy. METHODS: Neonates from the ALBINO study who received allopurinol or placebo mannitol were included. An extended population pharmacokinetic-pharmacodynamic model was developed based on the mechanism of purine metabolism, where synthesis, salvage, and degradation via xanthine oxidoreductase pathways were described. The initial level of the biomarkers was a combination of endogenous turnover and high disease-related amounts. Model development was accomplished by nonlinear mixed-effects modeling (NONMEM®, version 7.5). RESULTS: In total, 20 neonates treated with allopurinol and 17 neonates treated with mannitol were included in this analysis. Endogenous synthesis of the biomarkers reduced with 0.43% per hour because of precursor exhaustion. Hypoxanthine was readily salvaged or degraded to xanthine with rate constants of 0.5 1/h (95% confidence interval 0.33-0.77) and 0.2 1/h (95% confidence interval 0.09-0.31), respectively. A greater salvage was found in the allopurinol treatment group consistent with its mechanism of action. High hypoxia-induced initial levels of biomarkers were quantified, and were 1.2-fold to 2.9-fold higher in neonates with moderate-to-severe hypoxic-ischemic encephalopathy compared with those with mild hypoxic-ischemic encephalopathy. Half-maximal xanthine oxidoreductase inhibition was achieved with a combined allopurinol and oxypurinol concentration of 0.68 mg/L (95% confidence interval 0.48-0.92), suggesting full xanthine oxidoreductase inhibition during the period studied. CONCLUSIONS: This extended pharmacokinetic-pharmacodynamic model provided an adequate description of the complex hypoxanthine, xanthine, and uric acid metabolism in neonates with hypoxic-ischemic encephalopathy, suggesting a positive allopurinol effect on these biomarkers. The impact of hypoxia on their dynamics was characterized, underlining higher hypoxia-related initial exposure with a more severe hypoxic-ischemic encephalopathy status.


Subject(s)
Hypoxia-Ischemia, Brain , Oxypurinol , Humans , Infant, Newborn , Allopurinol/pharmacology , Allopurinol/therapeutic use , Hypoxanthine , Hypoxia/drug therapy , Hypoxia-Ischemia, Brain/drug therapy , Mannitol , Oxypurinol/pharmacology , Uric Acid , Xanthine , Xanthine Dehydrogenase , Clinical Studies as Topic
5.
Clin Pharmacol Ther ; 106(3): 623-631, 2019 09.
Article in English | MEDLINE | ID: mdl-30924126

ABSTRACT

Allopurinol, which lowers uric acid (UA) concentration, is increasingly being recognized for its benefits in cardiovascular and renal disease. However, response to allopurinol is variable. We gathered samples from 4,446 multiethnic subjects for a genome-wide association study of allopurinol response. Consistent with previous studies, we observed that the Q141K variant in ABCG2 (rs2231142), which encodes the efflux pump breast cancer resistance protein (BCRP), associated with worse response to allopurinol. However, for the first time this association reached genome-wide level significance (P = 8.06 × 10-11 ). Additionally, we identified a novel association with a variant in GREM2 (rs1934341, P = 3.22 × 10-6 ). In vitro studies identified oxypurinol, the active metabolite of allopurinol, as an inhibitor of the UA transporter GLUT9, suggesting that oxypurinol may modulate UA reabsorption. These results provide strong evidence for a role of BCRP Q141K in allopurinol response, and suggest that allopurinol may have additional hypouricemic effects beyond xanthine oxidase inhibition.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Allopurinol/pharmacology , Neoplasm Proteins/genetics , Uric Acid/metabolism , Aged , Aged, 80 and over , Cytokines/genetics , Ethnicity , Female , Genome-Wide Association Study , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Humans , Male , Middle Aged , Oxypurinol/pharmacology , Prognosis
6.
Free Radic Biol Med ; 129: 364-371, 2018 12.
Article in English | MEDLINE | ID: mdl-30312761

ABSTRACT

Generation of superoxide by xanthine oxidase can be stimulated under ischemic and aberrant calcium homeostasis. Because patients and mice with Duchenne muscular dystrophy (DMD) suffer from ischemia and excessive calcium influx, we tested the hypothesis that xanthine oxidase activity is elevated and contributes to disease pathology. Xanthine oxidase activity was measured by urinary isoxanthopterin in DMD patients at rest and in response to exercise. Urinary isoxanthopterin/creatinine was elevated compared to age-matched controls and Becker muscular dystrophy (BMD) patients. Concentrations were also increased after a six minute walk test in ambulatory patients. We also measured urinary isoxanthopterin in wildtype mice and a number of dystrophic mouse models; the DMD mouse model (mdx), mdx mice overexpressing a variety of transgenic miniaturized and chimeric skeletal muscle-specific dystrophins and utrophin and the ß-sarcoglycan deficient (Scgb-/-) mouse which represents type 2E human limb-girdle muscular dystrophy. Mdx and Scgb-/-mice had greater urinary isoxanthopterin/creatinine than wildtype mice while mdx mice expressing dystrophin or utrophin linking the extracellular matrix to the actin cytoskeleton were not different than wildtype. We also measured higher levels of urinary ortho-tyrosine in humans and mice deficient for dystrophin to confirm elevated oxidative stress. Surprisingly, mdx had lower xanthine oxidase protein levels and higher mRNA in gastrocnemius muscle compared to wildtype mice, however, the enzymatic activity of skeletal muscle xanthine oxidase was elevated above wildtype and a transgenic rescued mdx mouse (DysΔMTB-mdx). Downhill treadmill running also caused significant increases in mdx urinary isoxanthopterin that was prevented with the xanthine oxidase inhibitor allopurinol. Similarly, in vitro eccentric contraction-induced force drop of mdx muscle was attenuated by the allopurinol metabolite, oxypurinol. Together, our data suggests hyper-activity of xanthine oxidase in DMD, identifies xanthine oxidase activity as a contributing factor in eccentric contraction-induced force drop of dystrophin-deficient skeletal muscle and highlights the potential of isoxanthopterin as a noninvasive biomarker in DMD.


Subject(s)
Dystrophin/deficiency , Muscular Dystrophy, Animal/enzymology , Muscular Dystrophy, Duchenne/enzymology , Xanthine Oxidase/urine , Xanthopterin/urine , Adolescent , Allopurinol/pharmacology , Animals , Biomarkers/urine , Case-Control Studies , Creatinine/urine , Dystrophin/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred mdx , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/physiopathology , Oxypurinol/pharmacology , Sarcoglycans/deficiency , Sarcoglycans/genetics , Tyrosine/urine , Utrophin/deficiency , Utrophin/genetics , Xanthine Oxidase/genetics , Young Adult
7.
Anal Bioanal Chem ; 408(30): 8755-8760, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27392750

ABSTRACT

Xanthine oxidase (XO) is an important enzyme, expressed at high levels in the vasculature in endothelial cells, that catalyzes the hydroxylation of hypoxanthine to xanthine and xanthine to uric acid. Excessive production of uric acid results in hyperuricemia linked to gout and cardiovascular diseases. Testing inhibition of XO is important for detection of potentially effective drugs or natural products that could be used to treat diseases caused by increased XO activity. In the present study, for the first time, we developed an in vitro chemiluminescent bioassay to determine XO activity in living endothelial cells and the IC50 value of oxypurinol, the active metabolite of the inhibitor drug allopurinol. Intracellular XO activity was measured in less than 20 min with a luminol/catalyst-based chemiluminescence assay able to measure XO with a limit of 0.4 µU/mL. Oxypurinol addition to 5 × 103 cells (ranging from 5.0 to 0.0 µM) caused a linear decrease in XO activity, with an IC50 of 1.0 ± 0.5 µM. The detection system developed was low-cost, rapid, reproducible, and easily miniaturizable so suitable to be used on small quantities of cells.


Subject(s)
Biological Assay , Luminescent Measurements/methods , Uric Acid/antagonists & inhibitors , Xanthine Oxidase/antagonists & inhibitors , Allopurinol/chemistry , Allopurinol/pharmacology , Cytoplasm/enzymology , Enzyme Inhibitors/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Kinetics , Limit of Detection , Luminol/chemistry , Oxidation-Reduction , Oxypurinol/pharmacology , Uric Acid/metabolism , Xanthine Oxidase/metabolism
8.
Br J Dermatol ; 175(5): 994-1002, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27106261

ABSTRACT

BACKGROUND: Allopurinol is a frequent cause of severe cutaneous adverse reactions (SCARs), such as drug reaction with eosinophilia and systemic symptoms (DRESS), Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN). The reactions can potentially be fatal. As drug rechallenge in patients with a history of drug-induced SCARs is contraindicated, in vitro testing may have a diagnostic role as a confirmation test. OBJECTIVES: To study the diagnostic value of interferon (IFN)-γ enzyme-linked immunospot (ELISpot) assay as a confirmatory test in patients with a history of allopurinol-induced SCARs. METHODS: Peripheral blood mononuclear cells (PBMCs) from 24 patients with a history of allopurinol-induced SCAR (13 DRESS, 11 SJS/TEN) and 21 control subjects were incubated with allopurinol or oxypurinol in the presence or absence of antiprogrammed death ligand 1 antibody (anti-PD-L1). The numbers of IFN-γ-releasing cells after stimulation in each group were subsequently measured with ELISpot. RESULTS: The numbers of IFN-γ-releasing cells in allopurinol-allergic subjects were significantly higher than in control subjects when stimulating PBMCs with oxypurinol 100 µg mL-1 , especially when adding anti-PD-L1 supplementation. According to the receiver operating characteristic curve results, the optimal discriminatory power of IFN-γ ELISpot in confirming diagnosis of allopurinol-induced SCARs can be obtained using 16 spot-forming cells per 106 PBMCs as a cut-off value upon oxypurinol/anti-PD-L1 stimulation (79·2% sensitivity and 95·2% specificity). CONCLUSIONS: The measurement of oxypurinol/anti-PD-L1-inducing IFN-γ-releasing cells yields a high diagnostic value in distinguishing between allopurinol-allergic and control subjects. This technique is beneficial in confirming diagnosis of allopurinol-induced SCARs in patients whose reaction develops while taking multiple drugs.


Subject(s)
Allopurinol/adverse effects , Stevens-Johnson Syndrome/diagnosis , Antibodies/metabolism , Area Under Curve , B7-H1 Antigen/immunology , Case-Control Studies , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunospot Assay/methods , Enzyme-Linked Immunospot Assay/standards , Female , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Oxypurinol/pharmacology , Stevens-Johnson Syndrome/etiology
9.
Exp Cell Res ; 342(2): 166-74, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26968635

ABSTRACT

Allopurinol and its active metabolite, oxypurinol are widely used in the treatment of gout and hyperuricemia. They inhibit xanthine oxidase (XO) an enzyme in the purine degradation pathway that converts xanthine to uric acid. This investigation examined the effect of allopurinol and oxypurinol on bone formation, cell number and viability, gene expression and enzyme activity in differentiating and mature, bone-forming osteoblasts. Although mRNA expression remained relatively constant, XO activity decreased over time with mature osteoblasts displaying reduced levels of uric acid (20% decrease). Treatment with allopurinol and oxypurinol (0.1-1 µM) reduced XO activity by up to 30%. At these concentrations, allopurinol and oxypurinol increased bone formation by osteoblasts ~4-fold and ~3-fold, respectively. Cell number and viability were unaffected. Both drugs increased tissue non-specific alkaline phosphatase (TNAP) activity up to 65%. Osteocalcin and TNAP mRNA expression was increased, 5-fold and 2-fold, respectively. Expression of NPP1, the enzyme responsible for generating the mineralisation inhibitor, pyrophosphate, was decreased 5-fold. Col1α1 mRNA expression and soluble collagen levels were unchanged. Osteoclast formation and resorptive activity were not affected by treatment with allopurinol or oxypurinol. Our data suggest that inhibition of XO activity promotes osteoblast differentiation, leading to increased bone formation in vitro.


Subject(s)
Allopurinol/pharmacology , Bone Density Conservation Agents/pharmacology , Cell Differentiation/drug effects , Osteoblasts/physiology , Animals , Cell Survival , Cells, Cultured , Collagen/metabolism , Drug Evaluation, Preclinical , Gene Expression , Mice , Osteoblasts/drug effects , Osteogenesis , Oxypurinol/pharmacology , Rats , Rats, Sprague-Dawley , Xanthine Oxidase/antagonists & inhibitors , Xanthine Oxidase/genetics , Xanthine Oxidase/metabolism
10.
J Invest Dermatol ; 135(9): 2237-2248, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25946710

ABSTRACT

Allopurinol, a first-line drug for treating gout and hyperuricemia, is one of the leading causes of severe cutaneous adverse reactions (SCARs). To investigate the molecular mechanism of allopurinol-induced SCAR, we enrolled 21 patients (13 Stevens-Johnson syndrome (SJS)/toxic epidermal necrolysis (TEN) and 8 drug reaction with eosinophilia and systemic symptoms (DRESS)), 11 tolerant controls, and 23 healthy donors. We performed in vitro T-cell activation assays by culturing peripheral blood mononuclear cells (PBMCs) with allopurinol, oxypurinol, or febuxostat and measuring the expression of granulysin and IFN-γ in the supernatants of cultures. TCR repertoire was investigated by next-generation sequencing. Oxypurinol stimulation resulted in a significant increase in granulysin in the cultures of blood samples from SCAR patients (n=14) but not tolerant controls (n=11) or healthy donors (n=23). Oxypurinol induced T-cell response in a concentration- and time-dependent manner, whereas allopurinol or febuxostat did not. T cells from patients with allopurinol-SCAR showed no crossreactivity with febuxostat. Preferential TCR-V-ß usage and clonal expansion of specific CDR3 (third complementarity-determining region) were found in the blister cells from skin lesions (n=8) and oxypurinol-activated T-cell cultures (n=4) from patients with allopurinol-SCAR. These data suggest that, in addition to HLA-B*58:01, clonotype-specific T cells expressing granulysin upon oxypurinol induction participate in the pathogenesis of allopurinol-induced SCAR.


Subject(s)
Allopurinol/adverse effects , Antigens, Differentiation, T-Lymphocyte/metabolism , Leukocytes, Mononuclear/drug effects , Lymphocyte Activation/drug effects , Oxypurinol/adverse effects , Adult , Aged , Aged, 80 and over , Allopurinol/pharmacology , Case-Control Studies , Cells, Cultured/immunology , Cross Reactions , Drug Eruptions/etiology , Drug Eruptions/immunology , Enzyme-Linked Immunosorbent Assay , Febuxostat , Female , Humans , Interferon-gamma/drug effects , Interferon-gamma/immunology , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Oxypurinol/pharmacology , Reference Values , Stevens-Johnson Syndrome/etiology , Stevens-Johnson Syndrome/immunology , Thiazoles/adverse effects , Thiazoles/pharmacology
11.
Int J Mol Sci ; 15(11): 20079-100, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25375191

ABSTRACT

BACKGROUND: Ventricular tachycardia or fibrillation (VT/VF) of focal origin due to triggered activity (TA) from delayed afterdepolarizations (DADs) is reproducibly inducible after anterior coronary artery occlusion. Both VT/VF and TA can be blocked by reducing reactive oxygen species (ROS). We tested the hypothesis that inhibition of NADPH oxidase and xanthine oxidase would block VT/VF. METHODS: 69 dogs received apocynin (APO), 4 mg/kg intraveneously (IV), oxypurinol (OXY), 4 mg/kg IV, or both APO and OXY (BOTH) agents, or saline 3 h after coronary occlusion. Endocardium from ischemic sites (3-D mapping) was sampled for Rac1 (GTP-binding protein in membrane NADPH oxidase) activation or standard microelectrode techniques. Results (mean±SE, * p<0.05): VT/VF originating from ischemic zones was blocked by APO in 6/10 *, OXY in 4/9 *, BOTH in 5/8 * or saline in 1/27; 11/16 VT/VFs blocked were focal. In isolated myocardium, TA was blocked by APO (10(-6) M) or OXY (10(-8) M). Rac1 levels in ischemic endocardium were decreased by APO or OXY. CONCLUSION: APO and OXY suppressed focal VT/VF due to DADs, but the combination of the drugs was not more effective than either alone. Both drugs inhibited ischemic Rac1 with inhibition by OXY suggesting ROS-induced ROS. The inability to totally prevent VT/VF suggests that other mechanisms also contribute to ischemic VT.


Subject(s)
Myocardial Ischemia/enzymology , NADPH Oxidases/metabolism , Tachycardia, Ventricular/enzymology , Ventricular Fibrillation/enzymology , Xanthine Oxidase/metabolism , Acetophenones/pharmacology , Acetophenones/therapeutic use , Action Potentials/drug effects , Animals , Blotting, Western , Disease Models, Animal , Dogs , Female , Male , Myocardial Ischemia/diagnostic imaging , Myocardial Ischemia/drug therapy , Myocardial Ischemia/physiopathology , NADPH Oxidases/antagonists & inhibitors , Oxypurinol/pharmacology , Oxypurinol/therapeutic use , Tachycardia, Ventricular/complications , Tachycardia, Ventricular/drug therapy , Tachycardia, Ventricular/physiopathology , Ultrasonography , Ventricular Fibrillation/complications , Ventricular Fibrillation/drug therapy , Ventricular Fibrillation/physiopathology , Xanthine Oxidase/antagonists & inhibitors , rac1 GTP-Binding Protein/metabolism
12.
J Immunol ; 192(7): 2984-93, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24591375

ABSTRACT

Allopurinol (ALP) hypersensitivity is a major cause of severe cutaneous adverse reactions and is strongly associated with the HLA-B*58:01 allele. However, it can occur in the absence of this allele with identical clinical manifestations. The immune mechanism of ALP-induced severe cutaneous adverse reactions is poorly understood, and the T cell-reactivity pattern in patients with or without the HLA-B*58:01 allele is not known. To understand the interactions among the drug, HLA, and TCR, we generated T cell lines that react to ALP or its metabolite oxypurinol (OXP) from HLA-B*58:01(+) and HLA-B*58:01(-) donors and assessed their reactivity. ALP/OXP-specific T cells reacted immediately to the addition of the drugs and bypassed intracellular Ag processing, which is consistent with the "pharmacological interaction with immune receptors" (p-i) concept. This direct activation occurred regardless of HLA-B*58:01 status. Although most OXP-specific T cells from HLA-B*58:01(+) donors were restricted by the HLA-B*58:01 molecule for drug recognition, ALP-specific T cells also were restricted to other MHC class I molecules. This can be explained by in silico docking data that suggest that OXP binds to the peptide-binding groove of HLA-B*58:01 with higher affinity. The ensuing T cell responses elicited by ALP or OXP were not limited to particular TCR Vß repertoires. We conclude that the drug-specific T cells are activated by OXP bound to HLA-B*58:01 through the p-i mechanism.


Subject(s)
HLA-B Antigens/immunology , Lymphocyte Activation/immunology , Oxypurinol/immunology , T-Lymphocytes/immunology , Allopurinol/chemistry , Allopurinol/immunology , Allopurinol/pharmacology , Binding, Competitive/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Calcium/immunology , Calcium/metabolism , Cells, Cultured , Flow Cytometry , HLA-B Antigens/chemistry , HLA-B Antigens/genetics , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lysosomal-Associated Membrane Protein 1/immunology , Lysosomal-Associated Membrane Protein 1/metabolism , Models, Molecular , Molecular Structure , Oxypurinol/chemistry , Oxypurinol/pharmacology , Protein Binding/immunology , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
13.
J Enzyme Inhib Med Chem ; 29(6): 796-803, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24506204

ABSTRACT

Abstract Retinoic acid is regarded as the retinol metabolite that controls proliferation and differentiation of epithelial cells. In the present study, we investigated the potential role of xanthine dehydrogenase (XDH) in retinoic acid biosynthesis in human thyroid glandular cells (HTGC). In particular, we observed that cellular retinoids binding proteins (CRBPs) are also implicated in the biosynthetic pathway leading to retinoic acid formation in primary cultures of HTGC, as we have already reported for human mammary epithelial cells (HMEC). After partial protein purification, the enzyme responsible for retinoic acid biosynthesis was identified and quantified as XDH by immunoassay, by its ability to oxidize xanthine to uric acid and its sensitivity to the inhibitory effect of oxypurinol. The evidence of XDH-driven formation of retinoic acid in HTGC cultures further corroborates the potential role of XDH in retinoic acid biosynthesis in the epithelia.


Subject(s)
Epithelial Cells/enzymology , Thyroid Gland/enzymology , Tretinoin/metabolism , Vitamin A/metabolism , Xanthine Dehydrogenase/metabolism , Adult , Enzyme Assays , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Humans , Immunoassay , Male , Middle Aged , Oxidation-Reduction , Oxypurinol/pharmacology , Primary Cell Culture , Retinol-Binding Proteins, Cellular/metabolism , Thyroid Gland/cytology , Thyroid Gland/drug effects , Uric Acid/metabolism , Xanthine/metabolism , Xanthine Dehydrogenase/chemistry , Xanthine Dehydrogenase/isolation & purification
14.
Drug Metab Dispos ; 42(4): 695-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24430612

ABSTRACT

When investigating the potential for xanthine oxidase (XO)-mediated metabolism of a new chemical entity in vitro, selective chemical inhibition experiments are typically used. Most commonly, these inhibition experiments are performed using the inhibitor allopurinol (AP) and commercially prepared human liver cytosol (HLC) as the enzyme source. For reasons detailed herein, it is also a common practice to perfuse livers with solutions containing AP prior to liver harvest. The exposure to AP in HLC preparations could obviously pose a problem for measuring in vitro XO activity. To investigate this potential problem, an HPLC-MS/MS assay was developed to determine whether AP and its primary metabolite, oxypurinol, are retained within the cytosol for livers that were treated with AP during liver harvest. Differences in enzymatic activity for XO and aldehyde oxidase (AO) in human cytosol that can be ascribed to AP exposure were also evaluated. The results confirmed the presence of residual AP (some) and oxypurinol (all) human liver cytosol preparations that had been perfused with an AP-containing solution. In every case where oxypurinol was detected, XO activity was not observed. In contrast, the presence of AP and oxypurinol did not appear to have an impact on AO activity. Pooled HLC that was purchased from a commercial source also contained residual oxypurinol and did not show any XO activity. In the future, it is recommended that each HLC batch is screened for oxypurinol and/or XO activity prior to testing for XO-mediated metabolism of a new chemical entity.


Subject(s)
Allopurinol/pharmacology , Cytosol/enzymology , Enzyme Inhibitors/pharmacology , Liver/enzymology , Oxypurinol/pharmacology , Xanthine Oxidase/metabolism , Aldehyde Oxidase/metabolism , Allopurinol/analysis , Allopurinol/metabolism , Chromatography, High Pressure Liquid , Cytosol/drug effects , Enzyme Inhibitors/analysis , Enzyme Inhibitors/metabolism , Female , Humans , Limit of Detection , Liver/drug effects , Male , Oxypurinol/analysis , Oxypurinol/metabolism , Perfusion , Tandem Mass Spectrometry , Tissue Culture Techniques/methods , Xanthine Oxidase/antagonists & inhibitors
15.
Biochem Pharmacol ; 86(4): 539-47, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23770457

ABSTRACT

Up to 1/5 of patients with wildtype thiopurine-S-methyltransferase (TPMT) activity prescribed azathioprine (AZA) or mercaptopurine (MP) demonstrate a skewed drug metabolism in which MP is preferentially methylated to yield methylmercaptopurine (MeMP). This is known as thiopurine hypermethylation and is associated with drug toxicity and treatment non-response. Co-prescription of allopurinol with low dose AZA/MP (25-33%) circumvents this phenotype and leads to a dramatic reduction in methylated metabolites; however, the biochemical mechanism remains unclear. Using intact and lysate red cell models we propose a novel pathway of allopurinol mediated TPMT inhibition, through the production of thioxanthine (TX, 2-hydroxymercaptopurine). In red blood cells pre-incubated with 250 µM MP for 2h prior to the addition of 250 µM TX or an equivalent volume of Earle's balanced salt solution, there was a significant reduction in the concentration of MeMP detected at 4h and 6h in cells exposed to TX (4 h, 1.68, p=0.0005, t-test). TX acts as a direct TPMT inhibitor with an apparent Ki of 0.329 mM. In addition we have confirmed that the mechanism is relevant to in vivo metabolism by demonstrating raised urinary TX levels in patients receiving combination therapy. We conclude that the formation of TX in patients receiving combination therapy with AZA/MP and allopurinol, likely explains the significant reduction of methylated metabolites due to direct TPMT inhibition.


Subject(s)
Allopurinol/pharmacology , Methyltransferases/antagonists & inhibitors , Adult , Allopurinol/pharmacokinetics , Allopurinol/therapeutic use , Azathioprine/pharmacokinetics , Azathioprine/therapeutic use , Case-Control Studies , Drug Therapy, Combination , Erythrocytes/drug effects , Erythrocytes/metabolism , Female , Humans , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/urine , Male , Mercaptopurine/analogs & derivatives , Mercaptopurine/blood , Mercaptopurine/pharmacokinetics , Mercaptopurine/pharmacology , Methyltransferases/metabolism , Oxypurinol/pharmacology , Oxypurinol/urine , Prospective Studies , Xanthines/blood , Xanthines/pharmacology , Xanthines/urine
16.
Int J Cardiol ; 168(4): 3527-31, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-23751350

ABSTRACT

BACKGROUND: Cachexia is a common complication of cancer and may be responsible for 22% of all cancer-related deaths. The exact cause of death in cancer cachexia patients is unknown. Recently, atrophy of the heart has been described in cancer cachexia animal models, which resulted in impaired cardiac function and is likely to contribute to mortality. In cancer patients hyperuricaemia independent of tumour lysis syndrome is often associated with a worse prognosis. Xanthine oxidase (XO) metabolizes purines to uric acid and its inhibition has been shown to improve clinical outcome in patients with chronic heart failure. METHODS: The rat Yoshida AH-130 hepatoma cancer cachexia model was used in this study. Rats were treated with 4 or 40 mg/kg/d oxypurinol or placebo starting one day after tumour-inoculation for maximal 15 days. Cardiac function was analyzed by echocardiography on day 11. RESULTS: Here we show that inhibition of XO by oxypurinol significantly reduces wasting of the heart and preserves cardiac function. LVEF was higher in tumour-bearing rats treated with 4 mg/kg/d (61±4%) or 40 mg/kg/d (64±5%) oxypurinol vs placebo (51±3%, both p<0.05). Fractional shortening was improved by 4 mg/kg/d (43±3%) oxypurinol vs placebo (30±2, p<0.05), while 40 mg/kg/d oxypurinol (41±5%) did not reach statistical significance. Cardiac output was increased in the 4 mg/kg/d dose only (71±11 mL/min vs placebo 38±4 mL/min, p<0.01). CONCLUSION: Inhibition of XO with oxypurinol has beneficial effects on cardiac mass and function in a rat model of severe cancer cachexia, suggesting that XO might be a viable drug target in cancer cachexia.


Subject(s)
Cachexia/drug therapy , Cardiomyopathies/drug therapy , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms/drug therapy , Oxypurinol/therapeutic use , Xanthine Oxidase/antagonists & inhibitors , Animals , Cachexia/complications , Cachexia/enzymology , Cardiomyopathies/enzymology , Cardiomyopathies/etiology , Liver Neoplasms/complications , Liver Neoplasms/enzymology , Liver Neoplasms, Experimental/complications , Liver Neoplasms, Experimental/enzymology , Male , Oxypurinol/pharmacology , Rats , Rats, Wistar
17.
PLoS One ; 7(9): e45900, 2012.
Article in English | MEDLINE | ID: mdl-23029301

ABSTRACT

Cancer cachexia is a wasting condition, driven by systemic inflammation and oxidative stress. This study investigated eicosapentaenoic acid (EPA) in combination with oxypurinol as a treatment in a mouse model of cancer cachexia. Mice with cancer cachexia were randomized into 4 treatment groups (EPA (0.4 g/kg/day), oxypurinol (1 mmol/L ad-lib), combination, or control), and euthanized after 29 days. Analysis of oxidative damage to DNA, mRNA analysis of pro-oxidant, antioxidant and proteolytic pathway components, along with enzyme activity of pro- and antioxidants were completed on gastrocnemius muscle. The control group displayed earlier onset of tumor compared to EPA and oxypurinol groups (P<0.001). The EPA group maintained body weight for an extended duration (20 days) compared to the oxypurinol (5 days) and combination (8 days) groups (P<0.05). EPA (18.2±3.2 pg/ml) and combination (18.4±3.7 pg/ml) groups had significantly higher 8-OH-dG levels than the control group (12.9±1.4 pg/ml, P≤0.05) indicating increased oxidative damage to DNA. mRNA levels of GPx1, MURF1 and MAFbx were higher following EPA treatment compared to control (P≤0.05). Whereas oxypurinol was associated with higher GPx1, MnSOD, CAT, XDH, MURF1, MAFbx and UbB mRNA compared to control (P≤0.05). Activity of total SOD was higher in the oxypurinol group (32.2±1.5 U/ml) compared to control (27.0±1.3 U/ml, P<0.01), GPx activity was lower in the EPA group (8.76±2.0 U/ml) compared to control (14.0±1.9 U/ml, P<0.05), and catalase activity was lower in the combination group (14.4±2.8 U/ml) compared to control (20.9±2.0 U/ml, P<0.01). There was no change in XO activity. The increased rate of weight decline in mice treated with oxypurinol indicates that XO may play a protective role during the progression of cancer cachexia, and its inhibition is detrimental to outcomes. In combination with EPA, there was little significant improvement from control, indicating oxypurinol is unlikely to be a viable treatment compound in cancer cachexia.


Subject(s)
Adenocarcinoma/complications , Cachexia/drug therapy , Eicosapentaenoic Acid/therapeutic use , Enzyme Inhibitors/therapeutic use , Muscular Atrophy/drug therapy , Neoplasms, Experimental/complications , Oxypurinol/therapeutic use , Adenocarcinoma/drug therapy , Animals , Cachexia/etiology , Catalase/metabolism , Cell Line, Tumor , Drug Evaluation, Preclinical , Drug Therapy, Combination , Eicosapentaenoic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Female , Gene Expression/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/etiology , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Organ Size/drug effects , Oxidative Stress , Oxypurinol/pharmacology , Superoxide Dismutase/metabolism , Tumor Burden , Weight Loss/drug effects , Xanthine Oxidase/antagonists & inhibitors , Xanthine Oxidase/metabolism
18.
Cell Mol Life Sci ; 69(23): 4017-27, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22773120

ABSTRACT

MicroRNAs (miRNAs) constitute a novel class of small, non-coding RNAs that act as post-transcriptional regulators of gene expression. Remarkably, it has been shown that these small molecules can coordinately regulate multiple genes coding for proteins with related cellular functions. Previously, we reported that brain-specific miR-338 modulates the axonal expression of cytochrome c oxidase IV (COXIV), a nuclear-encoded mitochondrial protein that plays a key role in oxidative phosphorylation and axonal function. Here, we report that ATP synthase (ATP5G1), like COXIV mRNA, contains a putative miR-338 binding site, and that modulation of miR-338 levels in the axon results in alterations in both COXIV and ATP5G1 expression. Importantly, miR-338 modulation of local COXIV and ATP5G1 expression has a marked effect on axonal ROS levels, as well as axonal growth. These findings point to a mechanism by which miR-338 modulates local energy metabolism through the coordinate regulation of the expression of multiple nuclear-encoded mitochondrial mRNAs in the axon.


Subject(s)
Axons/metabolism , Electron Transport Complex IV/genetics , MicroRNAs/genetics , Mitochondrial Proton-Translocating ATPases/genetics , Oxidative Phosphorylation , 3' Untranslated Regions/genetics , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Base Sequence , Blotting, Western , Cells, Cultured , Electron Transport Complex IV/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Masoprocol/pharmacology , MicroRNAs/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Oxypurinol/pharmacology , RNA/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Mitochondrial , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Superior Cervical Ganglion/cytology
19.
Clin Sci (Lond) ; 123(8): 509-18, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22568671

ABSTRACT

Oestrogen protects cardiovascular health partially via an up-regulation of NO• (NO radical) production. Its synthetic analogue DES (diethylstilbestrol), used as a potent androgen deprivation therapy for patients with prostate cancer, is however associated with high incidence of thromboembolic events. Exposure of BAECs (bovine aortic endothelial cells) to pharmacologically relevant dosage (12.5 µmol/l, 24 h) of DES resulted in a marked reduction in endothelial NO• bioavailability determined by ESR (electron spin resonance), while 17ß-oestradiol instead increased NO• production as expected. Intriguingly, endothelial O(2)•- (superoxide anion) production was up-regulated by DES in vitro and in vivo, which was, however, attenuated by the ER (oestrogen receptor) antagonist ICI 182780, the XO (xanthine oxidase) inhibitor oxypurinol or the NOX (NADPH oxidase) inhibitor NSC23766. These agents also restored NO• production. DES alone in a cell-free system did not produce any ESR-sound O(2)•- signal. Of note, eNOS (endothelial NO synthase) mRNA and protein remained unchanged in response to DES. These results suggest that receptor-dependent activation of XO or NOX, and subsequent production of O(2)•-, mediate DES-induced NO• deficiency. This could represent a previously unrecognized mechanism that is responsible for cardiovascular complications of DES administration. Importantly, DES-induced suppression of LNCaP cell invasion and apoptosis were not affected by XO or NOX inhibitor. Therefore combinatorial therapy of DES and XO/NOX inhibitor may prove to be an innovative and useful therapeutic option in eliminating cardiovascular complications of DES, while preserving its anti-cancer effects, benefiting patients with advanced cancer who do not respond well to any other treatments but DES.


Subject(s)
Diethylstilbestrol/pharmacology , Endothelium, Vascular/drug effects , NADPH Oxidases/antagonists & inhibitors , Nitric Oxide/deficiency , Prostatic Neoplasms/pathology , Xanthine Oxidase/antagonists & inhibitors , Aminoquinolines/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cattle , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Electron Spin Resonance Spectroscopy/methods , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Hydrogen Peroxide/metabolism , Male , Mice , Mice, Inbred C57BL , NADPH Oxidases/physiology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , Oxypurinol/pharmacology , Pyrimidines/pharmacology , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Tumor Cells, Cultured , Up-Regulation/drug effects , Xanthine Oxidase/physiology
20.
J Cardiol ; 59(3): 235-42, 2012 May.
Article in English | MEDLINE | ID: mdl-22398104

ABSTRACT

Uric acid is the end product of purine metabolism. Its immediate precursor, xanthine, is converted to uric acid by an enzymatic reaction involving xanthine oxidoreductase. Uric acid has been formerly considered a major antioxidant in human plasma with possible beneficial anti-atherosclerotic effects. In contrast, studies in the past two decades have reported associations between elevated serum uric acid levels and cardiovascular events, suggesting a potential role for uric acid as a risk factor for atherosclerosis and related diseases. In this paper, the molecular pattern of uric acid formation, its possible deleterious effects, as well as the involvement of xanthine oxidoreductase in reactive oxygen species generation are critically discussed. Reactive oxygen species contribute to vascular oxidative stress and endothelial dysfunction, which are associated with the risk of atherosclerosis. Recent studies have renewed attention to the xanthine oxidoreductase system, since xanthine oxidoreductase inhibitors, such as allopurinol and oxypurinol, would be capable of preventing atherosclerosis progression by reducing endothelial dysfunction. Also, beneficial effects could be obtained in patients with congestive heart failure. The simultaneous reduction in uric acid levels might contribute to these effects, or be a mere epiphenomenon of the drug action. The molecular mechanisms involved are discussed.


Subject(s)
Cardiovascular Diseases/etiology , Endothelium, Vascular/physiopathology , Hyperuricemia/complications , Uric Acid/adverse effects , Xanthine Dehydrogenase/antagonists & inhibitors , Xanthine Dehydrogenase/physiology , Allopurinol/pharmacology , Allopurinol/therapeutic use , Antioxidants , Atherosclerosis/drug therapy , Atherosclerosis/etiology , Atherosclerosis/prevention & control , Cardiovascular Diseases/drug therapy , Humans , Molecular Targeted Therapy , Oxidative Stress , Oxypurinol/pharmacology , Oxypurinol/therapeutic use , Reactive Oxygen Species/metabolism , Risk Factors , Uric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...