Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
2.
Biol Pharm Bull ; 42(2): 173-178, 2019.
Article En | MEDLINE | ID: mdl-30713249

Optic neuritis is characterized by optic nerve inflammation, demyelination and axonal loss. Intravenous immunoglobulin (IVIg) has been reported to be effective for steroid-resistant patients. However, there is no report investigating the histopathological efficacy of IVIg in optic neuritis models. In this study, we examined the effects of IVIg on optic neuritis of experimental autoimmune encephalomyelitis (EAE) and experimental autoimmune optic neuritis (EAON). Inflammation, demyelination and axonal loss were assessed in the optic nerve sections. IVIg showed dose-dependent prevention of clinical symptoms in EAON. IVIg provided an anti-inflammatory effect in both EAE and EAON, associated with improved demyelination. Axonal loss in EAE was also significantly attenuated. These results suggest that IVIg has neuroprotective properties in experimental optic neuritis, and is a promising new treatment for optic neuritis.


Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunoglobulins, Intravenous/pharmacology , Optic Neuritis/drug therapy , Animals , Astrocytes/pathology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Freund's Adjuvant/pharmacology , Mice , Mice, Inbred C57BL , Microglia/pathology , Oligodendroglia/pathology , Optic Neuritis/chemically induced , Optic Neuritis/pathology , Optic Neuritis/prevention & control
3.
J Diabetes Investig ; 10(1): 94-103, 2019 Jan.
Article En | MEDLINE | ID: mdl-29862667

AIMS/INTRODUCTION: There is controversy as to whether hyperuricemia is an independent risk factor for cardiometabolic diseases. The serum level of uric acid is affected by a wide variety of factors involved in its production and excretion. In contrast, evidence has accumulated that locally- and systemically-activated xanthine oxidase (XO), a rate-limiting enzyme for production of uric acid, is linked to metabolic derangement in humans and rodents. We therefore explored the clinical implication of plasma XO activity in patients with type 2 diabetes mellitus and metabolic syndrome (MetS). MATERIALS AND METHODS: We enrolled 60 patients with type 2 diabetes mellitus and MetS. MetS was defined according to the 2005 International Diabetes Federation guidelines. Plasma XO activity was measured by highly-sensitive fluorometric assay measuring the conversion of pterin to isoxanthopterin, and explored associations between the value of plasma XO activity and metabolic parameters. RESULTS: The value of plasma XO activity was correlated with indices of insulin resistance and the level of circulating liver transaminases. In contrast, the level of serum uric acid was not correlated with indices of insulin resistance. The value of plasma XO activity was not correlated with the serum uric acid level. CONCLUSIONS: Plasma XO activity correlates with indices of insulin resistance and liver dysfunction in Japanese patients with type 2 diabetes mellitus and MetS. Through assessing the plasma XO activity, patients showing normal levels of serum uric acid with higher activity of XO can be screened, thereby possibly providing a clue to uncovering metabolic risks in type 2 diabetes mellitus and MetS patients.


Diabetes Mellitus, Type 2/blood , Insulin Resistance , Liver Diseases/blood , Metabolic Syndrome/blood , Xanthine Oxidase/blood , Adult , Aged , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Female , Humans , Liver Diseases/complications , Male , Metabolic Syndrome/complications , Middle Aged , Pilot Projects , Risk Factors , Young Adult
4.
PLoS One ; 12(11): e0187215, 2017.
Article En | MEDLINE | ID: mdl-29107957

Oxidative stress and mitochondrial dysfunction are important determinants of neurodegeneration in secondary progressive multiple sclerosis (SPMS). We previously showed that febuxostat, a xanthine oxidase inhibitor, ameliorated both relapsing-remitting and secondary progressive experimental autoimmune encephalomyelitis (EAE) by preventing neurodegeneration in mice. In this study, we investigated how febuxostat protects neuron in secondary progressive EAE. A DNA microarray analysis revealed that febuxostat treatment increased the CNS expression of several mitochondria-related genes in EAE mice, most notably including GOT2, which encodes glutamate oxaloacetate transaminase 2 (GOT2). GOT2 is a mitochondrial enzyme that oxidizes glutamate to produce α-ketoglutarate for the Krebs cycle, eventually leading to the production of adenosine triphosphate (ATP). Whereas GOT2 expression was decreased in the spinal cord during the chronic progressive phase of EAE, febuxostat-treated EAE mice showed increased GOT2 expression. Moreover, febuxostat treatment of Neuro2a cells in vitro ameliorated ATP exhaustion induced by rotenone application. The ability of febuxostat to preserve ATP production in the presence of rotenone was significantly reduced by GOT2 siRNA. GOT2-mediated ATP synthesis may be a pivotal mechanism underlying the protective effect of febuxostat against neurodegeneration in EAE. Accordingly, febuxostat may also have clinical utility as a disease-modifying drug in SPMS.


Aspartate Aminotransferase, Mitochondrial/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Febuxostat/therapeutic use , Mitochondria/drug effects , Adenosine Triphosphate/metabolism , Animals , Aspartate Aminotransferase, Mitochondrial/genetics , Cell Line , Encephalomyelitis, Autoimmune, Experimental/enzymology , Energy Metabolism , Febuxostat/pharmacology , Humans , Mice , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Rotenone/pharmacology , Xanthine Oxidase/antagonists & inhibitors
5.
Toxins (Basel) ; 9(2)2017 01 26.
Article En | MEDLINE | ID: mdl-28134751

Enterohemorrhagic Escherichia coli (EHEC) is the most common cause of hemorrhagic colitis and hemolytic uremic syndrome in human patients, with brain damage and dysfunction the main cause of acute death. We evaluated the efficacy of urtoxazumab (TMA-15, Teijin Pharma Limited), a humanized monoclonal antibody against Shiga toxin (Stx) 2 for the prevention of brain damage, dysfunction, and death in a piglet EHEC infection model. Forty-five neonatal gnotobiotic piglets were inoculated orally with 3 × 108 colony-forming units of EHEC O157:H7 strain EDL933 (Stx1⁺, Stx2⁺) when 22-24 h old. At 24 h post-inoculation, piglets were intraperitoneally administered placebo or TMA-15 (0.3, 1.0 or 3.0 mg/kg body weight). Compared to placebo (n = 10), TMA-15 (n = 35) yielded a significantly greater probability of survival, length of survival, and weight gain (p <0.05). The efficacy of TMA-15 against brain lesions and death was 62.9% (p = 0.0004) and 71.4% (p = 0.0004), respectively. These results suggest that TMA-15 may potentially prevent or reduce vascular necrosis and infarction of the brain attributable to Stx2 in human patients acutely infected with EHEC. However, we do not infer that TMA-15 treatment will completely protect human patients infected with EHEC O157:H7 strains that produce both Stx1 and Stx2.


Antibodies, Monoclonal, Humanized/pharmacology , Brain Infarction/prevention & control , Brain/drug effects , Escherichia coli O157/drug effects , Hemolytic-Uremic Syndrome/prevention & control , Meningitis, Escherichia coli/prevention & control , Shiga Toxin 2/antagonists & inhibitors , Animals , Animals, Newborn , Brain/immunology , Brain/microbiology , Brain/pathology , Brain Infarction/immunology , Brain Infarction/microbiology , Diarrhea/drug therapy , Diarrhea/immunology , Diarrhea/microbiology , Disease Models, Animal , Escherichia coli O157/immunology , Escherichia coli O157/pathogenicity , Germ-Free Life , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/microbiology , Meningitis, Escherichia coli/immunology , Meningitis, Escherichia coli/microbiology , Necrosis , Severity of Illness Index , Shiga Toxin 2/immunology , Sus scrofa , Time Factors
6.
Naunyn Schmiedebergs Arch Pharmacol ; 389(8): 831-8, 2016 Aug.
Article En | MEDLINE | ID: mdl-27198514

Xanthine oxidase (XO) is an enzyme responsible for the production of uric acid. XO produces considerable amount of oxidative stress throughout the body. To date, however, its pathophysiologic role in hypertension and endothelial dysfunction still remains controversial. To explore the possible involvement of XO-derived oxidative stress in the pathophysiology of vascular dysfunction, by use of a selective XO inhibitor, febuxostat, we investigated the impact of pharmacological inhibition of XO on hypertension and vascular endothelial dysfunction in spontaneously hypertensive rats (SHRs). Sixteen-week-old SHR and normotensive Wistar-Kyoto (WKY) rats were treated with tap water (control) or water containing febuxostat (3 mg/kg/day) for 6 weeks. Systolic blood pressure (SBP) in febuxostat-treated SHR (220 ± 3 mmHg) was significantly (P < 0.05) decreased compared with the control SHR (236 ± 4 mmHg) while SBP in febuxostat-treated WKY was constant. Acetylcholine-induced endothelium-dependent relaxation in aortas from febuxostat-treated SHR was significantly (P < 0.05) improved compared with the control SHR, whereas relaxation in response to sodium nitroprusside was not changed. Vascular XO activity and tissue nitrotyrosine level, a representative indicator of local oxidative stress, were considerably elevated in the control SHR compared with the control WKY, and this increment was abolished by febuxostat. Our results suggest that exaggerated XO activity and resultant increase in oxidative stress in this experimental model contribute to the hypertension and endothelial dysfunction, thereby supporting a notion that pharmacological inhibition of XO is valuable not only for hyperuricemia but also for treating hypertension and related endothelial dysfunction in human clinics.


Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Febuxostat/pharmacology , Hypertension/drug therapy , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Xanthine Oxidase/antagonists & inhibitors , Animals , Biomarkers/metabolism , Disease Models, Animal , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Hypertension/enzymology , Hypertension/physiopathology , Male , Oxidative Stress/drug effects , Rats, Inbred SHR , Rats, Inbred WKY , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Xanthine Oxidase/metabolism
7.
J Immunol ; 195(12): 5718-24, 2015 Dec 15.
Article En | MEDLINE | ID: mdl-26546608

Activation of the nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome initiates an inflammatory response, which is associated with host defense against pathogens and the progression of chronic inflammatory diseases such as gout and atherosclerosis. The NLRP3 inflammasome mediates caspase-1 activation and subsequent IL-1ß processing in response to various stimuli, including extracellular ATP, although the roles of intracellular ATP (iATP) in NLRP3 activation remain unclear. In this study, we found that in activated macrophages artificial reduction of iATP by 2-deoxyglucose, a glycolysis inhibitor, caused mitochondrial membrane depolarization, leading to IL-1ß secretion via NLRP3 and caspase-1 activation. Additionally, the NLRP3 activators nigericin and monosodium urate crystals lowered iATP through K(+)- and Ca(2+)-mediated mitochondrial dysfunction, suggesting a feedback loop between iATP loss and lowering of mitochondrial membrane potential. These results demonstrate the fundamental roles of iATP in the maintenance of mitochondrial function and regulation of IL-1ß secretion, and they suggest that maintenance of the intracellular ATP pools could be a strategy for countering NLRP3-mediated inflammation.


Adenosine Triphosphate/metabolism , Inflammation/immunology , Interleukin-1beta/metabolism , Macrophages/immunology , Nerve Tissue Proteins/metabolism , Nigericin/metabolism , Uric Acid/metabolism , Animals , Caspase 1/metabolism , Cells, Cultured , Deoxyglucose/metabolism , Humans , Inflammasomes/immunology , Intracellular Space , Membrane Glycoproteins , Mice, Inbred C57BL
8.
Sci Rep ; 4: 4554, 2014 Apr 01.
Article En | MEDLINE | ID: mdl-24686534

Atherosclerosis is a chronic inflammatory disease due to lipid deposition in the arterial wall. Multiple mechanisms participate in the inflammatory process, including oxidative stress. Xanthine oxidase (XO) is a major source of reactive oxygen species (ROS) and has been linked to the pathogenesis of atherosclerosis, but the underlying mechanisms remain unclear. Here, we show enhanced XO expression in macrophages in the atherosclerotic plaque and in aortic endothelial cells in ApoE(-/-) mice, and that febuxostat, a highly potent XO inhibitor, suppressed plaque formation, reduced arterial ROS levels and improved endothelial dysfunction in ApoE(-/-) mice without affecting plasma cholesterol levels. In vitro, febuxostat inhibited cholesterol crystal-induced ROS formation and inflammatory cytokine release in murine macrophages. These results demonstrate that in the atherosclerotic plaque, XO-mediated ROS formation is pro-inflammatory and XO-inhibition by febuxostat is a potential therapy for atherosclerosis.


Atherosclerosis/drug therapy , Plaque, Atherosclerotic/pathology , Reactive Oxygen Species/metabolism , Thiazoles/pharmacology , Xanthine Oxidase/antagonists & inhibitors , Animals , Aorta/cytology , Aorta/metabolism , Apolipoproteins E/genetics , Atherosclerosis/pathology , Body Weight , Cholesterol/blood , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Febuxostat , Gout Suppressants/pharmacology , Inflammation/drug therapy , L-Lactate Dehydrogenase/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/drug effects , Xanthine Oxidase/biosynthesis
9.
PLoS One ; 8(9): e75527, 2013.
Article En | MEDLINE | ID: mdl-24086554

Excess reactive oxygen species (ROS) formation can trigger various pathological conditions such as inflammation, in which xanthine oxidase (XO) is one major enzymatic source of ROS. Although XO has been reported to play essential roles in inflammatory conditions, the molecular mechanisms underlying the involvement of XO in inflammatory pathways remain unclear. Febuxostat, a selective and potent inhibitor of XO, effectively inhibits not only the generation of uric acid but also the formation of ROS. In this study, therefore, we examined the effects of febuxostat on lipopolysaccharide (LPS)-mediated inflammatory responses. Here we show that febuxostat suppresses LPS-induced MCP-1 production and mRNA expression via activating MAPK phosphatase-1 (MKP-1) which, in turn, leads to dephosphorylation and inactivation of JNK in macrophages. Moreover, these effects of febuxostat are mediated by inhibiting XO-mediated intracellular ROS production. Taken together, our data suggest that XO mediates LPS-induced phosphorylation of JNK through ROS production and MKP-1 inactivation, leading to MCP-1 production in macrophages. These studies may bring new insights into the novel role of XO in regulating inflammatory process through MAPK phosphatase, and demonstrate the potential use of XO inhibitor in modulating the inflammatory processes.


Chemokine CCL2/metabolism , Dual Specificity Phosphatase 1/metabolism , Lipopolysaccharides/pharmacology , MAP Kinase Kinase 4/metabolism , Thiazoles/pharmacology , Xanthine Oxidase/antagonists & inhibitors , Animals , Cell Line, Tumor , Chemokine CCL2/genetics , Dual Specificity Phosphatase 1/genetics , Febuxostat , Female , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , MAP Kinase Kinase 4/genetics , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Phosphorylation/genetics , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Xanthine Oxidase/metabolism
10.
J Biol Chem ; 288(38): 27138-27149, 2013 Sep 20.
Article En | MEDLINE | ID: mdl-23913681

Obesity is often accompanied by hyperuricemia. However, purine metabolism in various tissues, especially regarding uric acid production, has not been fully elucidated. Here we report, using mouse models, that adipose tissue could produce and secrete uric acid through xanthine oxidoreductase (XOR) and that the production was enhanced in obesity. Plasma uric acid was elevated in obese mice and attenuated by administration of the XOR inhibitor febuxostat. Adipose tissue was one of major organs that had abundant expression and activities of XOR, and adipose tissues in obese mice had higher XOR activities than those in control mice. 3T3-L1 and mouse primary mature adipocytes produced and secreted uric acid into culture medium. The secretion was inhibited by febuxostat in a dose-dependent manner or by gene knockdown of XOR. Surgical ischemia in adipose tissue increased local uric acid production and secretion via XOR, with a subsequent increase in circulating uric acid levels. Uric acid secretion from whole adipose tissue was increased in obese mice, and uric acid secretion from 3T3-L1 adipocytes was increased under hypoxia. Our results suggest that purine catabolism in adipose tissue could be enhanced in obesity.


Adipocytes/metabolism , Adipose Tissue/metabolism , Obesity/metabolism , Uric Acid/metabolism , Xanthine Dehydrogenase/metabolism , 3T3-L1 Cells , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Cell Hypoxia , Gene Knockdown Techniques , Male , Mice , Mice, Obese , Obesity/genetics , Obesity/pathology , Xanthine Dehydrogenase/genetics
11.
Neurochem Int ; 63(2): 93-100, 2013 Aug.
Article En | MEDLINE | ID: mdl-23727061

Amphotericin B (AmB) is a polyene antibiotic and reported to be one of a few reagents having therapeutic effects on prion diseases, such as the delay in the appearing of the clinical signs and the prolongation of the survival time. In prion diseases, glial cells have been suggested to play important roles by proliferating and producing various factors such as nitric oxide, proinflammatory cytokines, and neurotrophic factors. However, the therapeutic mechanism of AmB on prion diseases remains elusive. We have previously reported that AmB changed the expression of neurotoxic and neurotrophic factors in microglia (Motoyoshi et al., 2008, Neurochem. Int. 52, 1290-1296). In the present study, we examined the effects of AmB on cellular functions of rat cultured astrocytes. We found that AmB could activate astrocytes to produce nitric oxide via inducible nitric oxide synthase induction. AmB also induced mRNA expression of interleukin-1ß and tumor necrosis factor-α, and productions of their proteins in astrocytes. Moreover, AmB changed levels of neurotrophic factor mRNAs and proteins. Among three neurotrophic factors examined here, neurotrophin-3 mRNA expression and its protein production in the cells were down-regulated by AmB stimulation. On the other hand, AmB significantly enhanced the amounts of glial cell line-derived neurotrophic factor and brain-derived neurotrophic factor proteins in the cells and the medium. These results suggest that AmB might show therapeutic effects on prion diseases by controlling the expression and production of such mediators in astrocytes.


Amphotericin B/pharmacology , Astrocytes/drug effects , Cytokines/biosynthesis , Nerve Growth Factors/biosynthesis , Nitric Oxide/biosynthesis , Animals , Astrocytes/cytology , Astrocytes/metabolism , Base Sequence , Cells, Cultured , DNA Primers , Rats , Rats, Wistar
12.
Histochem Cell Biol ; 131(4): 483-90, 2009 Apr.
Article En | MEDLINE | ID: mdl-19159945

Adenine phosphoribosyltransferase deficiency in mice or an excessive oral intake of adenine leads to the accumulation of 2,8-dihydroxyadenine (DHA) in renal tubules and that causes progressive renal dysfunction accompanied by interstitial fibrosis. However, the precise mechanism responsible for DHA-induced progressive fibrosis is not fully understood. The present study investigates the possible involvement of monocytes/macrophages in the progressive fibrosis induced by feeding adenine to mice. Urinary calculi were deposited in tubules on day 7 after the initiation of adenine feeding. Elevation of the serum creatinine level and loss of body weight were observed in a time-dependent manner, suggesting the development of typical renal dysfunction induced by the adenine feeding. In renal tissue, mRNA expression of MCP-1, MIP-1alpha, RANTES, IL-1beta, CCR2, TGF-beta, alpha-smooth muscle actin (alpha-SMA) and collagen 1a1 was increased in parallel. Along with the increased expression of these genes, a remarkable infiltration of macrophages into the tubulointerstitial area was observed in a time-dependent manner. In addition, in the tubulointerstitial area, alpha-SMA positive fibroblasts were increased in parallel with collagen deposition. These results suggest that the excessive consumption of adenine leads to progressive renal dysfunction in mice. We speculate that the accumulation of DHA in tubules might stimulate epithelium to produce MCP-1 and that profibrogenic TGF-beta produced by infiltrated macrophages might stimulate interstitial fibroblasts to produce collagen. These results indicate that macrophage infiltration is one of the triggers that initiates interstitial fibroblast activation and collagen deposition followed by renal dysfunction.


Adenine/pharmacology , Chemotaxis/physiology , Kidney Tubules/pathology , Macrophages/physiology , Nephritis, Interstitial/metabolism , Actins/metabolism , Adenine/analogs & derivatives , Adenine/metabolism , Animals , Chemokines/metabolism , Collagen/metabolism , Cytokines/metabolism , Disease Models, Animal , Fibrosis/chemically induced , Fibrosis/metabolism , Fibrosis/pathology , Kidney Tubules/metabolism , Male , Mice , Mice, Inbred C57BL , Nephritis, Interstitial/chemically induced , Nephritis, Interstitial/pathology
...