Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
1.
Sci Rep ; 14(1): 1338, 2024 01 16.
Article in English | MEDLINE | ID: mdl-38228698

ABSTRACT

Although uric acid-lowering agents such as xanthine oxidase inhibitors have potential cardioprotective effects, studies on their use in preventing cardiovascular diseases are lacking. We investigated the genetically proxied effects of reducing uric acid on ischemic cardiovascular diseases in a lipid-level-stratified population. We performed drug-target Mendelian randomization (MR) analyses using UK Biobank data to select genetic instruments within a uric acid-lowering gene, xanthine dehydrogenase (XDH), and construct genetic scores. For nonlinear MR analyses, individuals were stratified by lipid level. Outcomes included acute myocardial infarction (AMI), ischemic heart disease, cerebral infarction, transient cerebral ischemic attack, overall ischemic disease, and gout. We included 474,983 non-gout individuals with XDH-associated single-nucleotide polymorphisms. The XDH-variant-induced uric acid reduction was associated with reduced risk of gout (odds ratio [OR], 0.85; 95% confidence interval [CI], 0.78-0.93; P < 0.001), cerebral infarction (OR, 0.86; 95% CI, 0.75-0.98; P = 0.023), AMI (OR, 0.79; 95% CI, 0.66-0.94; P = 0.010) in individuals with triglycerides ≥ 188.00 mg/dL, and cerebral infarction in individuals with low-density lipoprotein cholesterol (LDL-C) ≤ 112.30 mg/dL (OR, 0.76; 95% CI, 0.61-0.96; P = 0.020) or LDL-C of 136.90-157.40 mg/dL (OR, 0.67; 95% CI, 0.49-0.92; P = 0.012). XDH-variant-induced uric acid reduction lowers the risk of gout, AMI for individuals with high triglycerides, and cerebral infarction except for individuals with high LDL-C, highlighting the potential heterogeneity in the protective effects of xanthine oxidase inhibitors for treating AMI and cerebral infarction depending on the lipid profiles.


Subject(s)
Gout , Myocardial Infarction , Humans , Uric Acid , Xanthine Oxidase/genetics , Mendelian Randomization Analysis , Cholesterol, LDL/genetics , Gout/drug therapy , Gout/genetics , Cerebral Infarction/drug therapy , Cerebral Infarction/genetics , Triglycerides/genetics , Genome-Wide Association Study , Polymorphism, Single Nucleotide
2.
Drug Des Devel Ther ; 17: 1371-1386, 2023.
Article in English | MEDLINE | ID: mdl-37181826

ABSTRACT

Purpose: This study aimed to investigate the underlying treatment mechanism of Radix Astragali (RA) in hyperuricemia from the perspective of microbiota and metabolomics. Methods: We used potassium oxyazinate (PO) to induce hyperuricemia mice, and we determined serum alanine aminotransferase/aspartate aminotransferase (ALT/AST), xanthine oxidase (XOD), creatinine (CRE), uric acid (UA), blood urea nitrogen (BUN) levels, liver XOD levels and assessed the kidney tissue histopathology. The therapeutic mechanism of RA in hyperuricemic mice was studied by 16S rRNA, metagenomic sequencing and metabolomics. Results: Our research showed that RA has therapeutic effect in hyperuricemia mice, such as slow the weight loss, repair kidney damage, and downregulate serum UA, XOD, CRE, ALT/AST, BUN, and liver XOD levels. RA restored the disturbance structure of the microbiota in hyperuricemia mice by increasing the relative abundances of beneficial bacteria (Lactobacillaceae and Lactobacillus murine) but decreasing the relative abundances of pathogenic bacteria (Prevotellaceae, Rikenellaceae and Bacteroidaceae). Meanwhile, we found that RA directly regulated the metabolic pathway (such as linoleic acid metabolism and glycerophospholipid metabolism) and indirectly regulated bile acid metabolism by mediating microbiota to ameliorate metabolic disorders. Subsequently, there was a robust correlation between specific microbiota, metabolites and the disease index. Conclusion: The ability of RA to protect mice against hyperuricemia is strongly linked to the microbiome-metabolite axis, which would provide evidence for RA as a medicine to prevent or treat hyperuricemia.


Subject(s)
Drugs, Chinese Herbal , Hyperuricemia , Mice , Animals , Hyperuricemia/drug therapy , RNA, Ribosomal, 16S , Metagenomics , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Xanthine Oxidase/genetics , Xanthine Oxidase/metabolism
3.
Redox Biol ; 59: 102573, 2023 02.
Article in English | MEDLINE | ID: mdl-36525890

ABSTRACT

The conversion of xanthine dehydrogenase (XDH) to xanthine oxidase (XO) occurs only in mammalian species. In fresh bovine milk, the enzyme exists predominantly as the oxidase form, in contrast to various normal organs where it is found primarily as the dehydrogenase: the mechanism of conversion to the oxidase in milk remains obscure. A systematic search for the essential factors for conversion from XDH to XO has been performed within fresh bovine milk using the highly purified dehydrogenase form after removal endogenous oxidase form by fractionation analysis. We find that conversion to the oxidase form requires four components under air: lactoperoxidase (LPO), XDH, SCN-, and substrate hypoxanthine or xanthine; the contribution of sulfhydryl oxidase appears to be minor. Disulfide bond formation between Cys-535 and Cys-995 is principally involved in the conversion, consistent with the result obtained from previous work with transgenic mice. In vitro reconstitution of LPO and SCN- results in synergistic conversion of the dehydrogenase to the oxidase the presence of xanthine, indicating the conversion is autocatalytic. Milk from an LPO knockout mouse contains a significantly greater proportion of the dehydrogenase form of the enzyme, although some oxidase form is also present, indicating that LPO contributes principally to the conversion, but that sulfhydryl oxidase (SO) may also be involved to a minor extent. All the components XDH/LPO/SCN- are necessary to inhibit bacterial growth in the presence of xanthine through disulfide bond formation in bacterial protein(s) required for replication, as part of an innate immunity system in mammals. Human GTEx Data suggest that mRNA of XDH and LPO are highly co-expressed in the salivary gland, mammary gland, mucosa of the airway and lung alveoli, and we have confirmed these human GTEx Data experimentally in mice. We discuss the possible roles of these components in the propagation of SARS-CoV-2 in these human cell types.


Subject(s)
COVID-19 , Xanthine Dehydrogenase , Mice , Animals , Humans , Xanthine Dehydrogenase/genetics , Xanthine Dehydrogenase/chemistry , Xanthine Oxidase/genetics , SARS-CoV-2/metabolism , Xanthines , Mammals/metabolism , Disulfides/chemistry
4.
Metab Syndr Relat Disord ; 20(8): 473-479, 2022 10.
Article in English | MEDLINE | ID: mdl-35796694

ABSTRACT

Background: Hyperuricemia (HU) is a metabolic disease characterized by high uric acid levels in the blood. HU is a risk factor for diabetes, cardiovascular complications, metabolic syndrome, and chronic kidney disease. Purpose: The present study was performed to determine the effect of experimental HU on xanthine oxidase (XO), tumor necrosis factor-alpha (TNF-α), nuclear factor-kappa B (NF-κB), interleukin-17 (IL-17), cytochrome C, glutathione peroxidase (GPx), caspase-3, and 8-hydroxydeoxyguanosine (8-OHdG) levels in liver tissues of rats. Study Design: Thirty-five, male, Wistar albino-type rats were used for this study. Experimental groups were formed as follows: Group 1: control group; Group 2: potassium oxonate (PO) group; group 3: PO+NAR (naringenin; 2 weeks) group; and Group 4: PO (2 weeks)+NAR (2 weeks) group (total of 4 weeks). Methods: The first group was not given anything other than normal rat food and drinking water. In the second group, a 250 mg/kg intraperitoneal dose of PO was administered for 2 weeks. In the third group, 250 mg/kg intraperitoneal PO (application for 2 weeks) and 100 mg/kg NAR intraperitoneally 1 hr after each application were administered. In the fourth group, intraperitoneal PO administration was applied for 2 weeks, followed by intraperitoneal administration of NAR for 2 weeks (4 weeks in total). At the end of the experimental period, XO, TNF-α, NF-κB, IL-17, cytochrome C, GPx, caspase-3, and 8-OHdG levels were determined in liver tissues. Results: HU increased XO, TNF-α, NF-κB, IL-17, cytochrome C, caspase-3, and 8-OHdG levels in liver tissues. However, both 2 and 4 weeks of NAR supplementation decreased these values, and also NAR supplementation led to an increase in GPx levels in tissues. Conclusions: The results of the study show that increased inflammation, apoptosis, and DNA damage in experimental HU can be prevented by administration of NAR due to inhibition of cytochrome C, NF-κB, caspase-3, and 8-OHdG.


Subject(s)
Drinking Water , Hyperuricemia , Male , Rats , Animals , NF-kappa B/genetics , NF-kappa B/metabolism , NF-kappa B/pharmacology , Caspase 3/genetics , Caspase 3/metabolism , Caspase 3/pharmacology , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukin-17/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Cytochromes c/genetics , Cytochromes c/metabolism , Cytochromes c/pharmacology , 8-Hydroxy-2'-Deoxyguanosine , Xanthine Oxidase/genetics , Xanthine Oxidase/metabolism , Xanthine Oxidase/pharmacology , Uric Acid , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Glutathione Peroxidase/pharmacology , Drinking Water/adverse effects , Drinking Water/metabolism , Rats, Wistar , Apoptosis , Inflammation/metabolism , Liver/metabolism , DNA Damage
5.
Infect Genet Evol ; 93: 104991, 2021 09.
Article in English | MEDLINE | ID: mdl-34229066

ABSTRACT

This study was designed to investigate the association of the xanthine oxidase (XO) polymorphisms and susceptibility to anti-tuberculosis drug-induced liver injury (ATDILI) in Chinese population. A total of 183 tuberculosis patients were enrolled. Patients with ATDILI were classified as cases and those without ATDILI were classified as controls. Genotyping for XO polymorphisms was determined by polymerase chain reaction and direct sequencing. The allele frequencies and genotype distribution was analyzed using the Chi square test to analyze the association between the gene polymorphisms and ATDILI. Binary logistic regression analysis was performed to assess the risk factors of ATDILI. A total of 21 patients were developed liver injury during anti-tuberculosis treatment in this study, with an incidence of 11.48%. In genotype analysis, no significant difference was observed in the alleles and genotypes frequencies of the six SNPs between two groups (P > 0.05). In haplotype analysis, carriers with GGGATA (rs1884725- rs2295475 -rs45523133- rs206812- rs206813- rs7575607) haplotype had a significantly higher risk of ATDILI compared with other haplotypes (OR = 2.445, 95%CI: 1.058-5.652, P < 0.05). This study suggested that the haplotype GGGATA constructed with rs206812 and rs7575607 mutant alleles might contribute to ATDILI susceptibility in a Chinese population.


Subject(s)
Antitubercular Agents/adverse effects , Chemical and Drug Induced Liver Injury/genetics , Polymorphism, Genetic , Tuberculosis/prevention & control , Xanthine Oxidase/genetics , Adult , Aged , Chemical and Drug Induced Liver Injury/epidemiology , Chemical and Drug Induced Liver Injury/etiology , China/epidemiology , Female , Humans , Incidence , Male , Middle Aged , Polymorphism, Single Nucleotide
6.
J Physiol Biochem ; 77(2): 295-304, 2021 May.
Article in English | MEDLINE | ID: mdl-33595776

ABSTRACT

Redox balance and methylation are crucial to homeostasis and are linked by the methionine-homocysteine cycle. We examined whether differences in methylation potential, measured as plasma levels of S-adenosyl methionine (SAM) and S-adenosyl homocysteine (SAH), occur at baseline and during anti-oxidant therapy with the xanthine oxidase inhibitor allopurinol in patients with heart failure with reduced ejection fraction. We analyzed plasma samples collected at baseline and 24 weeks in the Xanthine Oxidase Inhibition for Hyperuricemic Heart Failure Patients (EXACT-HF) study, which randomized patients with heart failure with reduced ejection fraction to allopurinol or placebo. Associations between plasma levels of SAM, SAH, SAM/SAH ratio, and outcomes, including laboratory markers and clinical events, were assessed. Despite randomization, median SAM levels were significantly lower at baseline in the allopurinol group. SAH levels at 24 weeks, and change in SAM from baseline to week 24, were significantly higher in the group of patients randomized to allopurinol compared to the placebo group. A significant correlation was observed between change in SAH levels and change in plasma uric acid (baseline to 24-week changes) in the allopurinol group. There were no significant associations between levels of SAM, SAH, and SAM/SAH ratio and clinical outcomes. Our results demonstrate significant biological variability in SAM and SAH levels at baseline and during treatment with an anti-oxidant and suggest a potential mechanism for the lack of efficacy observed in trials of anti-oxidant therapy. These data also highlight the need to explore personalized therapy for heart failure.


Subject(s)
Allopurinol/therapeutic use , Free Radical Scavengers/therapeutic use , Heart Failure/drug therapy , Hyperuricemia/drug therapy , S-Adenosylhomocysteine/blood , S-Adenosylmethionine/blood , Aged , Female , Heart Failure/blood , Heart Failure/physiopathology , Humans , Hyperuricemia/blood , Hyperuricemia/physiopathology , Male , Methylation/drug effects , Middle Aged , Oxidation-Reduction/drug effects , Precision Medicine , Stroke Volume/drug effects , Treatment Outcome , Uric Acid/blood , Xanthine Oxidase/blood , Xanthine Oxidase/genetics
7.
Arterioscler Thromb Vasc Biol ; 41(2): 769-782, 2021 02.
Article in English | MEDLINE | ID: mdl-33267657

ABSTRACT

OBJECTIVE: Chronic hemolysis is a hallmark of sickle cell disease (SCD) and a driver of vasculopathy; however, the mechanisms contributing to hemolysis remain incompletely understood. Although XO (xanthine oxidase) activity has been shown to be elevated in SCD, its role remains unknown. XO binds endothelium and generates oxidants as a byproduct of hypoxanthine and xanthine catabolism. We hypothesized that XO inhibition decreases oxidant production leading to less hemolysis. Approach and Results: Wild-type mice were bone marrow transplanted with control (AA) or sickle (SS) Townes bone marrow. After 12 weeks, mice were treated with 10 mg/kg per day of febuxostat (Uloric), Food and Drug Administration-approved XO inhibitor, for 10 weeks. Hematologic analysis demonstrated increased hematocrit, cellular hemoglobin, and red blood cells, with no change in reticulocyte percentage. Significant decreases in cell-free hemoglobin and increases in haptoglobin suggest XO inhibition decreased hemolysis. Myographic studies demonstrated improved pulmonary vascular dilation and blunted constriction, indicating improved pulmonary vasoreactivity, whereas pulmonary pressure and cardiac function were unaffected. The role of hepatic XO in SCD was evaluated by bone marrow transplanting hepatocyte-specific XO knockout mice with SS Townes bone marrow. However, hepatocyte-specific XO knockout, which results in >50% diminution in circulating XO, did not affect hemolysis levels or vascular function, suggesting hepatocyte-derived elevation of circulating XO is not the driver of hemolysis in SCD. CONCLUSIONS: Ten weeks of febuxostat treatment significantly decreased hemolysis and improved pulmonary vasoreactivity in a mouse model of SCD. Although hepatic XO accounts for >50% of circulating XO, it is not the source of XO driving hemolysis in SCD.


Subject(s)
Anemia, Sickle Cell/drug therapy , Enzyme Inhibitors/pharmacology , Erythrocytes/drug effects , Febuxostat/pharmacology , Hemodynamics/drug effects , Hemolysis/drug effects , Pulmonary Artery/drug effects , Xanthine Oxidase/antagonists & inhibitors , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/enzymology , Anemia, Sickle Cell/physiopathology , Animals , Disease Models, Animal , Erythrocytes/enzymology , Liver/enzymology , Male , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Artery/enzymology , Pulmonary Artery/physiopathology , Ventricular Function/drug effects , Xanthine Oxidase/genetics , Xanthine Oxidase/metabolism
8.
J Appl Microbiol ; 130(6): 2132-2140, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33090589

ABSTRACT

AIMS: Purine-degrading enzymes are favourable as medications and diagnostic tools for hyperuricemia. This study aimed to characterize enzymes isolated from micro-organisms, which may be useful for developing a new prophylaxis for hyperuricemia. METHODS AND RESULTS: Cellulosimicrobium funkei A153 was found to be a good catalyst for hypoxanthine degradation and could oxidize hypoxanthine to xanthine and further to uric acid. The enzyme catalysing this oxidation was purified, and its partial amino acid sequences were examined. Based on this information and genome sequencing results, this xanthine dehydrogenase family protein was cloned and expressed in Rhodococcus erythropolis L88. The recombinant enzyme with a His-tag was characterized. The enzyme was a xanthine oxidase as it could utilize molecular oxygen as an electron acceptor. It was stable under 50°C and exhibited maximum activity at pH 7·0. The kcat , Km and kcat /Km values for xanthine were 1·4 s-1 , 0·22 mmol l-1 and 6·4 s-1  mmol-1  l, respectively. CONCLUSIONS: Xanthine oxidase is favourable for hyperuricemia medication because it oxidizes hypoxanthine, an easily adsorbed purine, to xanthine and further to uric acid, which are hardly adsorbed purines. SIGNIFICANCE AND IMPACT OF THE STUDY: The enzyme is useful for decreasing serum uric acid levels via conversion of easily absorbed purines to hardly absorbed purines in the intestine. Enzymes from micro-organisms may be used as a novel prophylaxis for hyperuricemia.


Subject(s)
Actinobacteria/enzymology , Hypoxanthine/metabolism , Purines/metabolism , Rhodococcus/metabolism , Xanthine Oxidase/chemistry , Actinobacteria/genetics , Amino Acid Sequence , Bacterial Proteins , DNA, Bacterial , Oxidation-Reduction , Recombinant Proteins/metabolism , Rhodococcus/genetics , Uric Acid/metabolism , Whole Genome Sequencing , Xanthine/metabolism , Xanthine Dehydrogenase/metabolism , Xanthine Oxidase/genetics
9.
Mar Drugs ; 18(12)2020 Dec 01.
Article in English | MEDLINE | ID: mdl-33271765

ABSTRACT

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


Subject(s)
Adenosine Deaminase Inhibitors/pharmacology , Adenosine Deaminase/metabolism , Hyperuricemia/prevention & control , Membrane Transport Proteins/drug effects , Uric Acid/blood , Xanthine Oxidase/antagonists & inhibitors , Adenosine Deaminase/genetics , Animals , Biomarkers/blood , Biomarkers/urine , Disease Models, Animal , Fructose , Hyperuricemia/chemically induced , Hyperuricemia/enzymology , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/enzymology , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Rats, Sprague-Dawley , Renal Reabsorption/drug effects , Uric Acid/urine , Xanthine Oxidase/genetics , Xanthine Oxidase/metabolism , Xanthophylls/pharmacology
10.
Int J Mol Sci ; 21(20)2020 Oct 09.
Article in English | MEDLINE | ID: mdl-33050202

ABSTRACT

In the present study, we investigated the effects of xanthine oxidase (XO) inhibition on cholesterol-induced renal dysfunction in chronic kidney disease (CKD) mice, and in low-density lipoprotein (LDL)-treated human kidney proximal tubule epithelial (HK-2) cells. ApoE knockout (KO) mice underwent uninephrectomy to induce CKD, and were fed a normal diet or high-cholesterol (HC) diet along with the XO inhibitor topiroxostat (1 mg/kg/day). HK-2 cells were treated with LDL (200 µg/mL) and topiroxostat (5 µM) or small interfering RNA against xanthine dehydrogenase (siXDH; 20 nM). In uninephrectomized ApoE KO mice, the HC diet increased cholesterol accumulation, oxidative stress, XO activity, and kidney damage, while topiroxostat attenuated the hypercholesterolemia-associated renal dysfunction. The HC diet induced cholesterol accumulation by regulating the expressions of genes involved in cholesterol efflux (Nr1h3 and Abca1) and synthesis (Srebf2 and Hmgcr), which was reversed by topiroxostat. Topiroxostat suppressed the expressions of genes related to hypercholesterolemia-associated inflammation and fibrosis in the unilateral kidney. LDL stimulation evoked changes in the cholesterol metabolism, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, and NF-κB pathways in HK-2 cells, which were mitigated by XO inhibition with topiroxostat or siXDH. These findings suggest that XO inhibition exerts renoprotective effects against hypercholesterolemia-associated kidney injury. XO could be a novel therapeutic target for hypercholesterolemia-associated kidney injury in uninephrectomized patients.


Subject(s)
Hypercholesterolemia/complications , Hypercholesterolemia/metabolism , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , Xanthine Oxidase/metabolism , Cholesterol/metabolism , Disease Progression , Disease Susceptibility , Fibrosis , Humans , Lipid Metabolism , Lipoproteins, LDL/metabolism , Oxidative Stress , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/pathology , Signal Transduction , Xanthine Oxidase/genetics
11.
Toxicol Appl Pharmacol ; 402: 115122, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32628957

ABSTRACT

Benign prostatic hyperplasia (BPH) is a widespread disorder in elderly men. Cinnamaldehyde, which is a major constituent in the essential oil of cinnamon, has been previously reported to reduce xanthine oxidase activity, in addition to its anti-inflammatory, anti-oxidant, and anti-proliferative activities. Our study was designed to investigate the potential modulatory effects of cinnamaldehyde on testosterone model of BPH in rats through reduction of uric acid level, and suppression of IL-6/JAK1/STAT3 signaling pathway. Cinnamaldehyde (40 and 75 mg/kg) was orally administered to male Wistar rats for 3 weeks, and concurrently with testosterone (3 mg/kg, s.c.) from the second week. Cinnamaldehyde ameliorated the elevation in prostatic weight and index compared to rats treated with testosterone only, that was also confirmed by alleviation of histopathological changes in prostate architecture. The protective mechanisms of cinnamaldehyde were elucidated through inhibition of xanthine oxidase activity and reduced uric acid level. That was accompanied by reduction of the pro-inflammatory cytokines; interleukin-6 (IL-6), IL-1ß, tumor necrosis factor-alpha (TNF-α), and the nuclear translocation of the transcription factor NF-κB p65, that could be attributed also to the enhanced anti-oxidant defense by cinnamaldehyde. The protein expression of JAK1, which is IL-6 receptor linked protein, was reduced with subsequently reduced activation of STAT3 protein. That eventually suppressed the formation of the proliferation protein cyclin D1, while elevated Bax/Bcl2 ratio. It can be concluded that reducing uric acid level through xanthine oxidase inhibition and suppression of the inflammatory signaling cascade; IL-6/JAK1/STAT3; by cinnamaldehyde could be a novel and promising therapeutic approach against BPH.


Subject(s)
Acrolein/analogs & derivatives , Interleukin-6/metabolism , Janus Kinase 1/metabolism , Prostatic Hyperplasia/prevention & control , STAT3 Transcription Factor/metabolism , Uric Acid/blood , Acrolein/pharmacology , Animals , Biomarkers/blood , Cell Proliferation/physiology , Cyclin D1/genetics , Cyclin D1/metabolism , Gene Expression Regulation/drug effects , Immunohistochemistry , Interleukin-6/genetics , Janus Kinase 1/genetics , Male , Prostate/drug effects , Prostate/enzymology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , STAT3 Transcription Factor/genetics , Xanthine Oxidase/genetics , Xanthine Oxidase/metabolism
12.
Biochim Biophys Acta Mol Basis Dis ; 1866(8): 165802, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32311453

ABSTRACT

INTRODUCTION: Combined pulmonary fibrosis and emphysema (CPFE) is a relatively new entity within the spectrum of cigarette smoke induced lung disorders. Currently there is no consensus about its treatment. We hypothesized that caveolin-1 critically determines the parenchymal and vascular remodeling leading to the development of CPFE. We assessed the effect of therapeutic targeting of caveolin-1 in mesenchymal and endothelial cells by the phosphodiesterase-5 inhibitor, sildenafil. METHODS: Male Wistar rats (n = 168) were exposed to; room air (control); bleomycin (7 U/kg), bleomycin+sildenafil (50 mg/kg/day P.O.), cigarette smoke (CS) (4 Gold Flake 69 mm/day), CS + sildenafil, CS + bleomycin, CS + bleomycin+sildenafil. Animals were euthanized at 8, 9, 11, 12 weeks and lung histopathological changes, collagen deposition, ROS, Xanthine oxidase, caveolin-1 determined. RESULTS: Cigarette smoke causes progressive ROS accumulation, caveolin-1 up-regulation in alveolar epithelial cells, alveolar macrophages, peribronchiolar fibroblasts, endothelial and vascular smooth muscle cells, interstitial inflammation and emphysema. Sildenafil reduces oxidative stress, parenchymal caveolin-1 and attenuates emphysema caused by CS. Bleomycin increases lung ROS and downregulates caveolin-1 leading to fibroblast proliferation and fibrosis. Combined cigarette smoke and bleomycin exposure, results in differential caveolin-1 expression and heterogeneous parenchymal remodeling with alternating areas of emphysema and fibrosis. Increased caveolin-1 induces premature senescence of lung fibroblasts and emphysema. Decreased caveolin-1 is associated with propagation of EMT and fibrosis. Sildenafil attenuates the parenchymal remodeling however it is not effective in reducing VSMC hypertrophy in combined group. CONCLUSION: CPFE is characterized by heterogenous parenchymal remodeling and differential caveolin-1 expression. Sildenafil therapy attenuates parenchymal pathologies in CPFE. Additional therapy is however needed for attenuating VSMC remodeling.


Subject(s)
Caveolin 1/genetics , Cigarette Smoking/adverse effects , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Phosphodiesterase 5 Inhibitors/pharmacology , Pulmonary Emphysema/genetics , Pulmonary Fibrosis/genetics , Sildenafil Citrate/pharmacology , Animals , Bleomycin/administration & dosage , Caveolin 1/metabolism , Collagen/genetics , Collagen/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Pulmonary Emphysema/etiology , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Rats , Rats, Wistar , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Xanthine Oxidase/genetics , Xanthine Oxidase/metabolism
13.
J Biol Inorg Chem ; 25(4): 547-569, 2020 06.
Article in English | MEDLINE | ID: mdl-32279136

ABSTRACT

Mononuclear molybdenum enzymes catalyze a variety of reactions that are essential in the cycling of nitrogen, carbon, arsenic, and sulfur. For decades, the structure and function of these crucial enzymes have been investigated to develop a fundamental knowledge for this vast family of enzymes and the chemistries they carry out. Therefore, obtaining abundant quantities of active enzyme is necessary for exploring this family's biochemical capability. This mini-review summarizes the methods for overexpressing mononuclear molybdenum enzymes in the context of the challenges encountered in the process. Effective methods for molybdenum cofactor synthesis and incorporation, optimization of expression conditions, improving isolation of active vs. inactive enzyme, incorporation of additional prosthetic groups, and inclusion of redox enzyme maturation protein chaperones are discussed in relation to the current molybdenum enzyme literature. This article summarizes the heterologous and homologous expression studies providing underlying patterns and potential future directions.


Subject(s)
Iron-Sulfur Proteins/metabolism , Metalloproteins/metabolism , Molybdenum/metabolism , Oxidoreductases/metabolism , Sulfite Oxidase/metabolism , Xanthine Oxidase/metabolism , Cloning, Molecular , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Metalloproteins/chemistry , Metalloproteins/genetics , Molecular Structure , Molybdenum/chemistry , Oxidoreductases/chemistry , Oxidoreductases/genetics , Sulfite Oxidase/chemistry , Sulfite Oxidase/genetics , Xanthine Oxidase/chemistry , Xanthine Oxidase/genetics
14.
Biochem J ; 477(8): 1499-1513, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32239172

ABSTRACT

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


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Hemochromatosis/metabolism , Hyperuricemia/enzymology , Uric Acid/metabolism , Xanthine Oxidase/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Animals , Female , Hemochromatosis/complications , Hemochromatosis/congenital , Hemochromatosis/enzymology , Hemochromatosis Protein/genetics , Hemochromatosis Protein/metabolism , Homeostasis , Humans , Hyperuricemia/etiology , Hyperuricemia/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Xanthine Oxidase/genetics
15.
Int J Mol Sci ; 21(5)2020 Feb 25.
Article in English | MEDLINE | ID: mdl-32106617

ABSTRACT

Blueberries are rich in antioxidant anthocyanins. The hypotensive effects of blueberry anthocyanins in endothelial cells was investigated here. Pretreatment with blueberry anthocyanin extract, malvidin, malvidin-3-glucoside, and malvidin-3-galactoside significantly ameliorated high-glucose-induced damage by enhancing endogenous antioxidant superoxide dismutase (SOD) and heme oxygenase-1 (HO-1), lowering reactive oxygen species (ROS) generation and NADPH oxidase isoform 4 (NOX4) expression, and increasing the cell vitalities. They also effectively induced a vasodilatory effect by increasing the vasodilator nitric oxide (NO) and its promoters endothelial NO synthase (eNOS) and peroxisome proliferator-activated receptor-γ (PPARγ) levels as well as by decreasing the vasoconstrictor angiotensin-converting enzyme (ACE), xanthine oxidase-1 (XO-1), and low-density lipoprotein (LDL) levels. The activation of phosphoinositide 3-kinase (PI3K)/Akt signaling pathway and the breakdown of protein kinase C zeta (PKCζ) pathway were involved in the bioactivities. The results indicated blueberry anthocyanins protected endothelial function against high-glucose (HG) injury via antioxidant and vasodilatory mechanisms, which could be promising molecules as a hypotensive nutraceutical for diabetes patients.


Subject(s)
Anthocyanins/pharmacology , Antioxidants/pharmacology , Blueberry Plants/chemistry , Human Umbilical Vein Endothelial Cells/drug effects , Signal Transduction , Vasodilation , Anthocyanins/chemistry , Antioxidants/chemistry , Glucose/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lipoproteins, LDL/metabolism , Nitric Oxide/metabolism , Oxidative Stress , PPAR gamma/genetics , PPAR gamma/metabolism , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Xanthine Oxidase/genetics , Xanthine Oxidase/metabolism
16.
Food Funct ; 10(12): 8042-8055, 2019 Dec 11.
Article in English | MEDLINE | ID: mdl-31746890

ABSTRACT

This study was aimed at evaluating the prospect of edible chrysanthemum extract as a potential substance for the prevention and treatment of hyperuricemia. Chrysanthemum morifolium Ramat. 'Boju' extract (CBE), which had the strongest xanthine oxidase inhibitory activity, showed a significant hypouricemic effect on potassium oxonate-induced hyperuricemic rats through inhibiting serum xanthine oxidase activity, regulating renal uric acid transport-related protein (ABCG2, URAT1 and GLUT9) expression and blood lipid levels, and protecting renal function. Serum metabolomics based on UPLC-ESI-QTOF/MS was used to illustrate mechanisms underlying the amelioration effect of CBE on hyperuricemia. A total of 35 potential biomarkers were identified. CBE prevented the pathological process of hyperuricemia by regulating 16/17 biomarkers associated with tryptophan, sphingolipid, glycerophospholipid and arachidonic acid metabolisms. CBE could alleviate hyperuricemia-related diseases including chronic kidney disease, hyperlipidemia and inflammation via reducing indoxyl sulfate, lysophosphatidylcholines and arachidonic acid levels, exhibiting its applicability and superiority in the treatment of hyperuricemia.


Subject(s)
Chrysanthemum/chemistry , Drugs, Chinese Herbal/administration & dosage , Hyperuricemia/drug therapy , ATP Binding Cassette Transporter, Subfamily G, Member 2/blood , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Animals , Chromatography, High Pressure Liquid , Glucose Transport Proteins, Facilitative/blood , Glucose Transport Proteins, Facilitative/genetics , Humans , Hyperuricemia/blood , Hyperuricemia/genetics , Male , Mass Spectrometry , Metabolomics , Organic Anion Transporters/blood , Organic Anion Transporters/genetics , Rats , Serum/chemistry , Serum/metabolism , Xanthine Oxidase/blood , Xanthine Oxidase/genetics
17.
Nat Commun ; 10(1): 4904, 2019 10 28.
Article in English | MEDLINE | ID: mdl-31659168

ABSTRACT

Xanthine oxidoreductase has been implicated in cancer. Nonetheless, the role played by its two convertible forms, xanthine dehydrogenase (XDH) and oxidase (XO) during tumorigenesis is not understood. Here we produce XDH-stable and XO-locked knock-in (ki) mice to address this question. After tumor transfer, XO ki mice show strongly increased tumor growth compared to wild type (WT) and XDH ki mice. Hematopoietic XO expression is responsible for this effect. After macrophage depletion, tumor growth is reduced. Adoptive transfer of XO-ki macrophages in WT mice increases tumor growth. In vitro, XO ki macrophages produce higher levels of reactive oxygen species (ROS) responsible for the increased Tregs observed in the tumors. Blocking ROS in vivo slows down tumor growth. Collectively, these results indicate that the balance of XO/XDH plays an important role in immune surveillance of tumor development. Strategies that inhibit the XO form specifically may be valuable in controlling cancer growth.


Subject(s)
Neoplasms/enzymology , Xanthine Dehydrogenase/genetics , Xanthine Oxidase/genetics , Animals , Cell Proliferation , Female , Gene Knock-In Techniques , Humans , Macrophages/enzymology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/physiopathology , Reactive Oxygen Species/metabolism , Xanthine Dehydrogenase/metabolism , Xanthine Oxidase/metabolism
18.
J Nutr ; 149(11): 1911-1919, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31373370

ABSTRACT

BACKGROUND: Few studies have addressed the risk of nutritional iron overexposure in infancy. We previously found that excess dietary iron in nursing piglets resulted in iron overload in the liver and hippocampus and diminished socialization with novel conspecifics in a test for social novelty preference. OBJECTIVES: This experiment aimed to identify metabolites and metabolic pathways affected by iron overload in the liver and hippocampus of nursing piglets. METHODS: Liver and hippocampal tissues collected from 22-d-old piglets (Hampshire × Yorkshire crossbreed; 5.28 ± 0.53 kg body weight; 50% male) that received orally 0 (NI group) or 50 mg iron/(d · kg body weight) (HI group) from postnatal day (PD) 2 to PD21 were analyzed for mRNA and protein expression and enzyme activity of xanthine oxidase (XO). Untargeted metabolomics was performed using GC-MS. Expression of myelin basic protein (MBP) in the hippocampus was determined using western blot. RESULTS: There were 108 and 126 metabolites identified in the hippocampus and liver, respectively. Compared with NI, HI altered 15 metabolites (P < 0.05, q < 0.2) in the hippocampus, including a reduction in myo-inositol (0.86-fold) and N-acetylaspartic acid (0.84-fold), 2 metabolites important for neuronal function and myelination. Seven metabolites involved in purine and pyrimidine metabolism (e.g., hypoxanthine, xanthine, and ß-alanine) were coordinately changed in the hippocampus (P < 0.05, q < 0.2), suggesting that iron excess enhanced purine catabolism. The mRNA expression (2.3-fold) (P < 0.05) and activity of XO, a rate-limiting enzyme in purine degradation, was increased. Excess iron increased hippocampal lipid peroxidation by 74% (P < 0.05) and decreased MBP by 44% (P = 0.053). The hepatic metabolome was unaffected. CONCLUSIONS: In nursing piglets, excess iron enhances hippocampal purine degradation through activation of XO, which may induce oxidative stress and alter energy metabolism in the developing brain.


Subject(s)
Hippocampus/metabolism , Iron Overload/metabolism , Purines/metabolism , Xanthine Oxidase/metabolism , Animals , Disease Models, Animal , Enzyme Activation , Female , Gene Expression , Hippocampus/growth & development , Humans , Infant , Iron Overload/genetics , Iron, Dietary/administration & dosage , Iron, Dietary/adverse effects , Lipid Peroxidation , Liver/metabolism , Male , Metabolic Networks and Pathways , Metabolome , Metabolomics , Myelin Basic Protein/metabolism , Myelin Sheath/physiology , Oxidative Stress , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sus scrofa , Uric Acid/blood , Uric Acid/metabolism , Xanthine Oxidase/genetics
19.
Biosci Rep ; 39(7)2019 07 31.
Article in English | MEDLINE | ID: mdl-31273057

ABSTRACT

Silicosis is a lethal pneumoconiosis disease characterized by chronic lung inflammation and fibrosis. The present study was to explore the effect of against crystalline silica (CS)-induced pulmonary fibrosis. A total of 138 wild-type C57BL/6J mice were divided into control and experimental groups, and killed on month 0, 1, 2, 3, 4, and 5. Different doses of N-acetylcysteine (NAC) were gavaged to the mice after CS instillation to observe the effect of NAC on CS induced pulmonary fibrosis and inflammation. The pulmonary injury was evaluated with Hematoxylin and eosin/Masson staining. Reactive oxygen species level was analyzed by DCFH-DA labeling. Commercial ELISA kits were used to determine antioxidant activity (T-AOC, glutathione peroxidase (GSH-Px), and superoxide dismutase (SOD) and cytokines (TNF-α, IL-1ß, IL-4, and IL-6). The expression of oxidising enzymes (NOX2, iNOS, SOD2, and XO) were detected by real time PCR. Immunohistochemistry (IHC) staining was performed to examine epithelial-mesenchymal transition-related markers. The mice treated with NAC presented markedly reduced CS-induced pulmonary injury and ameliorated CS-induced pulmonary fibrosis and inflammation. The level of malondialdehyde was reduced, while the activities of GSH-PX, SOD, and T-AOC were markedly enhanced by NAC. We also found the down-regulation of oxidising enzymes (NOX2, iNOS, SOD2, and XO) after NAC treatment. Moreover, E-cadherin expression was increased while vimentin and Cytochrome C expressions were decreased by NAC. These encouraging findings suggest that NAC exerts pulmonary protective effects in CS-induced pulmonary fibrosis and might be considered as a promising agent for the treatment of silicosis.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Pulmonary Fibrosis/prevention & control , Reactive Oxygen Species/antagonists & inhibitors , Silicosis/prevention & control , Administration, Oral , Animals , Crystallization , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Reactive Oxygen Species/metabolism , Silicon Dioxide/administration & dosage , Silicosis/etiology , Silicosis/genetics , Silicosis/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Xanthine Oxidase/genetics , Xanthine Oxidase/metabolism
20.
Phytomedicine ; 62: 152975, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31181404

ABSTRACT

BACKGROUND: Alpinia oxyphylla is a well-known traditional medicine used in China and Korea to treat intestinal disorders, urosis, diuresis, and chronic glomerulonephritis. PURPOSE: We investigated the anti-hyperuricemic effects of Alpinia oxyphylla seed extract (AE), and the underlying mechanisms of action through in vitro and in vivo studies. METHODS: We evaluated levels of uric acid in the serum and urine, the expression of renal urate transport proteins, and levels of inflammatory cytokines in potassium oxonate (PO)-induced hyperuricemic rats. Xanthine oxidase activity was analyzed in vitro, while cellular uric acid uptake was assessed in oocytes expressing the human urate transporter 1 (hURAT1). Moreover, the main components of AE were analyzed using UPLC. RESULTS: In PO-induced hyperuricemic rats, 200 and 400 mg/kg of AE significantly decreased levels of uric acid in serum, while 400 mg/kg of AE increased uric acid levels in urine. AE did not inhibit xanthine oxidase in vitro; however, 1, 10, and 100 µg/ml of AE significantly decreased uric acid uptake into oocytes expressing hURAT1. Furthermore, 400 mg/kg of AE increased levels of organic anion transporter (OAT) 1 protein, while 200 and 400 mg/kg of AE decreased the protein content of urate transporter, URAT1 and inflammatory cytokines in the kidneys. Nootkatone was identified as one the main chemical components in AE from UPLC analysis. CONCLUSIONS: These findings suggest that AE exerts anti-hyperuricemic and uricosuric effects, which are related to the promotion of uric acid excretion via enhanced secretion and inhibition of uric acid reabsorption in the kidneys. Thus, AE may be a potential treatment for hyperuricemia and gout.


Subject(s)
Alpinia/chemistry , Hyperuricemia/drug therapy , Plant Extracts/administration & dosage , Uric Acid/urine , Xanthine Oxidase/metabolism , Animals , China/epidemiology , Gout , Humans , Kidney/drug effects , Kidney/metabolism , Male , Organic Anion Transport Protein 1/drug effects , Organic Anion Transporters/drug effects , Organic Anion Transporters/metabolism , Oxonic Acid , Plant Extracts/chemistry , Rats , Republic of Korea/epidemiology , Xanthine Oxidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...