Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters











Publication year range
1.
Life Sci ; 304: 120694, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-35679914

ABSTRACT

AIMS: Acetaminophen (APAP) is a relatively safe analgesic drug, but overdosing can cause acute liver failure. Ingested APAP is detoxified by metabolic conversion through conjugation reactions with glucuronate, sulfate, or glutathione (GSH). The consumption of GSH through conjugation as well as mitochondrial dysfunction is considered to be responsible for the increased susceptibility to APAP-induced hepatotoxicity. Compared to wild-type (WT) mice, Akr1a-knockout (KO) mice are vulnerable to developing hepatotoxicity due to the fact that ascorbate synthesis is attenuated. We used such KO mice to investigate how these conjugation reactions are involved in the hepatotoxicity caused by an overdose of APAP under ascorbate-deficient conditions. MAIN METHODS: APAP (400 mg/kg) was intraperitoneally administered to WT mice and KO mice. In addition to histological and blood biochemical analyses, metabolites in the liver, blood plasma, and urine were measured at several time points by liquid chromatography-mass spectrometry. KEY FINDINGS: Liver damage occurred earlier in the KO mice than in the WT mice. The levels of APAP-Cys, a final metabolite of GSH-conjugated APAP, as well as glucuronidated APAP and sulfated APAP were all higher in the KO mice compared to the WT mice. Treatment of the APAP-administered KO mice with N-acetylcysteine or supplementation of ascorbate suppressed the conjugation reactions at 6 h after APAP had been administrated, which mitigated the degree of liver damage. SIGNIFICANCE: An ascorbate deficiency coordinately stimulates conjugation reactions of APAP, which, combined with the mitochondrial damage caused by APAP metabolites, collectively results in the aggravation of the acute liver failure.


Subject(s)
Acetaminophen , Aldehyde Reductase , Chemical and Drug Induced Liver Injury , Acetaminophen/pharmacokinetics , Acetaminophen/toxicity , Aldehyde Reductase/deficiency , Aldehyde Reductase/metabolism , Animals , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Glutathione/metabolism , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Nature ; 565(7737): 96-100, 2019 01.
Article in English | MEDLINE | ID: mdl-30487609

ABSTRACT

Endothelial nitric oxide synthase (eNOS) is protective against kidney injury, but the molecular mechanisms of this protection are poorly understood1,2. Nitric oxide-based cellular signalling is generally mediated by protein S-nitrosylation, the oxidative modification of Cys residues to form S-nitrosothiols (SNOs). S-nitrosylation regulates proteins in all functional classes, and is controlled by enzymatic machinery that includes S-nitrosylases and denitrosylases, which add and remove SNO from proteins, respectively3,4. In Saccharomyces cerevisiae, the classic metabolic intermediate co-enzyme A (CoA) serves as an endogenous source of SNOs through its conjugation with nitric oxide to form S-nitroso-CoA (SNO-CoA), and S-nitrosylation of proteins by SNO-CoA is governed by its cognate denitrosylase, SNO-CoA reductase (SCoR)5. Mammals possess a functional homologue of yeast SCoR, an aldo-keto reductase family member (AKR1A1)5 with an unknown physiological role. Here we report that the SNO-CoA-AKR1A1 system is highly expressed in renal proximal tubules, where it transduces the activity of eNOS in reprogramming intermediary metabolism, thereby protecting kidneys against acute kidney injury. Specifically, deletion of Akr1a1 in mice to reduce SCoR activity increased protein S-nitrosylation, protected against acute kidney injury and improved survival, whereas this protection was lost when Enos (also known as Nos3) was also deleted. Metabolic profiling coupled with unbiased mass spectrometry-based SNO-protein identification revealed that protection by the SNO-CoA-SCoR system is mediated by inhibitory S-nitrosylation of pyruvate kinase M2 (PKM2) through a novel locus of regulation, thereby balancing fuel utilization (through glycolysis) with redox protection (through the pentose phosphate shunt). Targeted deletion of PKM2 from mouse proximal tubules recapitulated precisely the protective and mechanistic effects of S-nitrosylation in Akr1a1-/- mice, whereas Cys-mutant PKM2, which is refractory to S-nitrosylation, negated SNO-CoA bioactivity. Our results identify a physiological function of the SNO-CoA-SCoR system in mammals, describe new regulation of renal metabolism and of PKM2 in differentiated tissues, and offer a novel perspective on kidney injury with therapeutic implications.


Subject(s)
Acute Kidney Injury/enzymology , Acute Kidney Injury/prevention & control , Coenzyme A/metabolism , Metabolic Engineering , Oxidoreductases/metabolism , Aldehyde Reductase/deficiency , Aldehyde Reductase/genetics , Aldehyde Reductase/metabolism , Animals , Cell Line , Female , Glycolysis , HEK293 Cells , Humans , Kidney Tubules, Proximal/enzymology , Male , Mice , Mutation , Nitric Oxide Synthase Type III/metabolism , Oxidation-Reduction , Pentose Phosphate Pathway , Protein Multimerization , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/deficiency , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism
3.
Bioengineered ; 9(1): 233-241, 2018.
Article in English | MEDLINE | ID: mdl-29865993

ABSTRACT

Ethylene glycol (EG) is an important chemical used as antifreeze and a raw material in polyester synthesis. The EG biosynthetic pathway from D-xylose with D-xylonate as key intermediate has some advantages, but showed low EG production. Here, we reconstructed and optimized this pathway in Escherichia coli. In view of the greater intracellular prevalence of NADH, an aldehyde reductase FucO using NADH was employed to convert glycoaldehyde into EG, in replacement of NADPH-dependent reductase YqhD. To suppress the accumulation of by-products acetate and glycolate, two genes arcA and aldA were knocked out. The resultant strain Q2843 produced 72 g/L EG under fed-batch fermentation conditions, with the yield of 0.40 g/g D-xylose and EG productivity of 1.38 g/L/h. The use of NADH-dependent enzyme FucO and by-product elimination significantly improved the performance of EG producing strain, which represented the highest titer, yield and productivity of EG reported so far.


Subject(s)
Aldehyde Reductase/metabolism , Escherichia coli/metabolism , Ethylene Glycol/metabolism , Metabolic Engineering/methods , Xylose/metabolism , Acetic Acid/metabolism , Aldehyde Reductase/deficiency , Aldehyde Reductase/genetics , Bacterial Outer Membrane Proteins/genetics , Batch Cell Culture Techniques , Biosynthetic Pathways/genetics , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Fermentation , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glycolates/metabolism , Kinetics , NAD/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/deficiency , Repressor Proteins/genetics
4.
J Mol Cell Cardiol ; 118: 183-192, 2018 05.
Article in English | MEDLINE | ID: mdl-29627295

ABSTRACT

Pathological cardiac hypertrophy is associated with the accumulation of lipid peroxidation-derived aldehydes such as 4-hydroxy-trans-2-nonenal (HNE) and acrolein in the heart. These aldehydes are metabolized via several pathways, of which aldose reductase (AR) represents a broad-specificity route for their elimination. We tested the hypothesis that by preventing aldehyde removal, AR deficiency accentuates the pathological effects of transverse aortic constriction (TAC). We found that the levels of AR in the heart were increased in mice subjected to TAC for 2 weeks. In comparison with wild-type (WT), AR-null mice showed lower ejection fraction, which was exacerbated 2 weeks after TAC. Levels of atrial natriuretic peptide and myosin heavy chain were higher in AR-null than in WT TAC hearts. Deficiency of AR decreased urinary levels of the acrolein metabolite, 3-hydroxypropylmercapturic acid. Deletion of AR did not affect the levels of the other aldehyde-metabolizing enzyme - aldehyde dehydrogenase 2 in the heart, or its urinary product - (N-Acetyl-S-(2-carboxyethyl)-l-cystiene). AR-null hearts subjected to TAC showed increased accumulation of HNE- and acrolein-modified proteins, as well as increased AMPK phosphorylation and autophagy. Superfusion with HNE led to a greater increase in p62, LC3II formation, and GFP-LC3-II punctae formation in AR-null than WT cardiac myocytes. Pharmacological inactivation of JNK decreased HNE-induced autophagy in AR-null cardiac myocytes. Collectively, these results suggest that during hypertrophy the accumulation of lipid peroxidation derived aldehydes promotes pathological remodeling via excessive autophagy, and that metabolic detoxification of these aldehydes by AR may be essential for maintaining cardiac function during early stages of pressure overload.


Subject(s)
Aldehyde Reductase/deficiency , Autophagy , Heart/physiopathology , Pressure , Aldehyde Reductase/metabolism , Aldehydes/metabolism , Animals , Aorta/pathology , Cardiomegaly/diagnostic imaging , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Constriction, Pathologic , Gene Deletion , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice, Inbred C57BL , Myocardial Contraction , Myocardium/enzymology , Sequestosome-1 Protein/metabolism
5.
Oncotarget ; 8(5): 7357-7369, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28060768

ABSTRACT

The AKR1A1 protein is a member of the aldo-keto reductase superfamily that is responsible for the conversion of D-glucuronate to L-gulonate in the ascorbic acid (vitamin C) synthesis pathway. In a pCAG-eGFP transgenic mouse line that was produced by pronuclear microinjection, the integration of the transgene resulted in a 30-kb genomic DNA deletion, including the Akr1A1 gene, and thus caused the knockout (KO) of the Akr1A1 gene and targeting of the eGFP gene. The Akr1A1 KO mice (Akr1A1eGFP/eGFP) exhibited insufficient serum ascorbic acid levels, abnormal bone development and osteoporosis. Using micro-CT analysis, the results showed that the microarchitecture of the 12-week-old Akr1A1eGFP/eGFP mouse femur was shorter in length and exhibited less cortical bone thickness, enlargement of the bone marrow cavity and a complete loss of the trabecular bone in the distal femur. The femoral head and neck of the proximal femur also showed a severe loss of bone mass. Based on the decreased levels of serum osteocalcin and osteoblast activity in the Akr1A1eGFP/eGFP mice, the osteoporosis might be caused by impaired bone formation. In addition, administration of ascorbic acid to the Akr1A1eGFP/eGFP mice significantly prevented the condition of osteoporotic femurs and increased bone formation. Therefore, through ascorbic acid administration, the Akr1A1 KO mice exhibited controllable osteoporosis and may serve as a novel model for osteoporotic research.


Subject(s)
Aldehyde Reductase/genetics , Ascorbic Acid Deficiency/genetics , Femur/pathology , Gene Knockout Techniques , Osteogenesis , Osteoporosis/genetics , Aldehyde Reductase/deficiency , Animals , Ascorbic Acid/administration & dosage , Ascorbic Acid/blood , Ascorbic Acid Deficiency/enzymology , Ascorbic Acid Deficiency/pathology , Ascorbic Acid Deficiency/prevention & control , Disease Models, Animal , Female , Femur/diagnostic imaging , Femur/enzymology , Genetic Predisposition to Disease , Mice, Knockout , Osteoblasts/enzymology , Osteoblasts/pathology , Osteocalcin/blood , Osteoporosis/enzymology , Osteoporosis/pathology , Osteoporosis/prevention & control , Phenotype , Time Factors , X-Ray Microtomography
6.
J Biosci Bioeng ; 123(3): 333-341, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27865643

ABSTRACT

4-Hydroxy-2(or 5)-ethyl-5(or 2)-methyl-3(2H)-furanone (HEMF) is considered a key flavor compound in soy sauce. The compound has a caramel-like aroma and several important physiological activities, such as strong antioxidant activity. Here, we report the identification and characterization of an enzyme involved in the biosynthesis of HEMF in yeast. We fractionated yeast cell-free extract from Saccharomyces cerevisiae using column chromatography and partially purified a fraction with HEMF-forming activity. Peptide mass fingerprinting analysis showed that the partially purified fraction contains aldehyde reductase encoded by YNL134C. This reductase shares low sequence identity with enone oxidoreductase, which is responsible for the formation of 4-hydroxy-2,5-dimethyl-3(2H)-furanone (HDMF) and HEMF in plants. YNL134C was expressed heterologously in Escherichia coli, and the purified protein catalyzed the formation of HEMF from the mixture of Maillard reaction products, acetaldehydes, and NADPH. Multicopy expression in S. cerevisiae resulted in increased HEMF productivity, and gene knockout of YNL134C in S. cerevisiae resulted in decreased HEMF productivity. These data suggest that the translation product of YNL134C is the HEMF-producing enzyme in yeast. Detailed analyses of an intermediate in the enzymatic reaction mixture revealed that HEMF is synthesized from (2E)-2-ethylidene-4-hydroxy-5-methyl-3(2H)-furanone, which formed via Knoevenagel condensation between the acetaldehyde and 4-hydroxy-5-methyl-3(2H)-furanone derived from the Maillard reaction based on ribose and glycine, by YNL134Cp in an NADPH dependent manner. Overall, this study shed light on the molecular basis for the improvement of soy sauce flavor and the biotechnological production of HEMF.


Subject(s)
Furans/metabolism , Oxidoreductases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/metabolism , Acetaldehyde/metabolism , Aldehyde Reductase/deficiency , Aldehyde Reductase/genetics , Aldehyde Reductase/isolation & purification , Aldehyde Reductase/metabolism , Cell Extracts , Escherichia coli/genetics , Flavoring Agents/chemistry , Glycine/metabolism , NADP/metabolism , Oxidoreductases/deficiency , Oxidoreductases/genetics , Oxidoreductases/isolation & purification , Ribose/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/isolation & purification , Soy Foods
7.
Neurobiol Aging ; 50: 119-133, 2017 02.
Article in English | MEDLINE | ID: mdl-27960106

ABSTRACT

Fungicide exposure causes degeneration of dopaminergic neurons and contributes to Parkinson's disease (PD). Benomyl inhibits enzymes responsible for detoxifying the reactive dopamine metabolite 3,4-dihydroxyphenylacetaldehyde. Aldose reductase (AR) is known as tetrahydrobiopterin (BH4) reductase that generates BH4, a cofactor for tyrosine hydroxylase (TH) involved in dopamine synthesis. AR also acts as an aldehyde reductase involved in detoxifying 3,4-dihydroxyphenylacetaldehyde. In PD patients, the level of AR is significantly lower in the cerebellum. To determine if AR deficiency contributes to PD, AR wild-type (AR+/+) and knockout (AR-/-) mice were administrated with 1-methyl-4-phenyl -1,2,3,6- tetrahydropyridine (MPTP). The MPTP-treated AR-/- mice showed more severe behavioral deficits and brain damage than that of AR+/+ mice. Contrary to expectation, under normal or MPTP-treated condition, AR-/- mice showed a significant elevation of BH4 and dopamine in the midbrain, suggesting that either AR does not contribute to BH4 production, or other BH4 synthetic pathways are induced. The AR-/- brain showed upregulation of peroxynitrite, inducible nitric oxide synthase and downregulation of antioxidant enzymes, Cu/Zn superoxide dismutase (SOD) and peroxiredoxin 2 (Prx2), which indicate an increase in oxidative stress. In line with the animal data, pretreating the SH-SY5Y cells with AR inhibitors (Fidarestat or Epalrestat) before MPP+ treatment, increased severe cell death and mitochondrial fragmentation with downregulation of SOD were observed when compared to the MPP+ treatment alone. Cycloxygenase 2 (COX2), which can lead to the oxidation of dopamine, was upregulated in AR-/- brains. Autophagic proteins, beclin-1 and LC3B were also downregulated. The loss of dopaminergic neurons was associated with activation of p-ERK1/2. These findings suggest that AR plays an important role in protecting dopaminergic neuron against neurotoxic metabolites in PD.


Subject(s)
Aldehyde Reductase/deficiency , Autophagy , Dopaminergic Neurons/pathology , Oxidative Stress/physiology , Parkinson Disease/etiology , Parkinson Disease/pathology , Animals , Cells, Cultured , Disease Models, Animal , Male , Mice, Inbred C57BL , Mice, Transgenic , Parkinson Disease/physiopathology
8.
Biochem Biophys Res Commun ; 478(2): 765-71, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27501753

ABSTRACT

Aldehyde reductase (Akr1a) has been reported to be involved in the biosynthesis of ascorbic acid (AsA) in the mouse liver. Because Akr1a is expressed at high levels in the liver, we aimed to investigate the role of Akr1a in liver homeostasis by employing a carbon tetrachloride (CCl4)-induced hepatotoxicity model. Akr1a-deficient (Akr1a(-/-)) and wild-type (WT) mice were injected intraperitoneally with CCl4 and the extent of hepatic injury in the acute phase was assessed. Liver damage was heavier in the Akr1a(-/-) mice than in the WT mice. Furthermore, severe hepatic steatosis was observed in the livers of Akr1a(-/-) mice compared to WT mice and was restored to the levels in WT mice by AsA supplementation. Since the presence or absence of AsA had no effect on the decrease in CYP2E1 activity after the CCl4 treatment, it appears that AsA plays a role in the process after the bioactivation of CCl4. Biomarkers for oxidative stress and ER stress were markedly increased in the livers of Akr1a(-/-) mice and were effectively suppressed by AsA supplementation. Based on these collective results, we conclude that Akr1a exerts a protective effect against CCl4-induced hepatic steatosis by replenishing AsA via its antioxidative properties.


Subject(s)
Aldehyde Reductase/deficiency , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Chemical and Drug Induced Liver Injury/genetics , Endoplasmic Reticulum Stress/genetics , Fatty Liver/genetics , Oxidative Stress/genetics , Aldehyde Reductase/genetics , Animals , Antioxidants/metabolism , Ascorbic Acid/metabolism , Biomarkers/metabolism , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Endoplasmic Reticulum Chaperone BiP , Fatty Liver/chemically induced , Fatty Liver/enzymology , Fatty Liver/prevention & control , Gene Expression , Glutathione/agonists , Glutathione/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Lipid Peroxidation/drug effects , Liver/enzymology , Liver/pathology , Mice , Mice, Knockout , Perilipin-2/genetics , Perilipin-2/metabolism , Peroxiredoxins/genetics , Peroxiredoxins/metabolism
9.
Mol Neurobiol ; 53(1): 662-676, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25520004

ABSTRACT

Inflammatory reactions are the most critical pathological processes occurring after spinal cord injury (SCI). Activated microglia/macrophages have either detrimental or beneficial effects on neural regeneration based on their functional polarized M1/M2 subsets. However, the mechanism of microglia/macrophage polarization to M1/M2 at the injured spinal cord environment remains unknown. In this study, wild-type (WT) or aldose reductase (AR)-knockout (KO) mice were subjected to SCI by a spinal crush injury model. The expression pattern of AR, behavior tests for locomotor activity, and lesion size were assessed at between 4 h and 28 days after SCI. We found that the expression of AR is upregulated in microglia/macrophages after SCI in WT mice. In AR KO mice, SCI led to smaller injury lesion areas compared to WT. AR deficiency-induced microglia/macrophages induce the M2 rather than the M1 response and promote locomotion recovery after SCI in mice. In the in vitro experiments, microglia cell lines (N9 or BV2) were treated with the AR inhibitor (ARI) fidarestat. AR inhibition caused 4-hydroxynonenal (HNE) accumulation, which induced the phosphorylation of the cAMP response element-binding protein (CREB) to promote Arg1 expression. KG501, the specific inhibitor of phosphorylated CREB, could cancel the upregulation of Arg1 by ARI or HNE stimulation. Our results suggest that AR works as a switch which can regulate microglia by polarizing cells to either the M1 or the M2 phenotype under M1 stimulation based on its states of activity. We suggest that inhibiting AR may be a promising therapeutic method for SCI in the future.


Subject(s)
Aldehyde Reductase/biosynthesis , Cell Polarity/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Macrophages/metabolism , Microglia/metabolism , Spinal Cord Injuries/metabolism , Aldehyde Reductase/deficiency , Animals , Cell Line , Cell Polarity/drug effects , Cells, Cultured , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Tumor Necrosis Factor-alpha/pharmacology
10.
Graefes Arch Clin Exp Ophthalmol ; 253(9): 1503-13, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25921391

ABSTRACT

Retinopathy of prematurity (ROP) is a leading cause of childhood blindness where vascular abnormality and retinal dysfunction are reported. We showed earlier that genetic deletion of aldose reductase (AR), the rate-limiting enzyme in the polyol pathway, reduced the neovascularization through attenuating oxidative stress induction in the mouse oxygen-induced retinopathy (OIR) modeling ROP. In this study, we further investigated the effects of AR deficiency on retinal neurons in the mouse OIR. Seven-day-old wild-type and AR-deficient mice were exposed to 75 % oxygen for 5 days and then returned to room air. Electroretinography was used to assess the neuronal function at postnatal day (P) 30. On P17 and P30, retinal cytoarchitecture was examined by morphometric analysis and immunohistochemistry for calbindin, protein kinase C alpha, calretinin, Tuj1, and glial fibrillary acidic protein. In OIR, attenuated amplitudes and delayed implicit time of a-wave, b-wave, and oscillatory potentials were observed in wild-type mice, but they were not significantly changed in AR-deficient mice. The morphological changes of horizontal, rod bipolar, and amacrine cells were shown in wild-type mice and these changes were partly preserved with AR deficiency. AR deficiency attenuated the Müller cell gliosis induced in OIR. Our observations demonstrated AR deficiency preserved retinal functions in OIR and AR deficiency could partly reduce the extent of retinal neuronal histopathology. These findings suggested a therapeutic potential of AR inhibition in ROP treatment with beneficial effects on the retinal neurons.


Subject(s)
Aldehyde Reductase/deficiency , Disease Models, Animal , Gliosis/prevention & control , Retinal Neurons/enzymology , Retinopathy of Prematurity/prevention & control , Animals , Animals, Newborn , Calbindin 2/metabolism , Calbindins/metabolism , Electroretinography , Glial Fibrillary Acidic Protein , Gliosis/enzymology , Immunohistochemistry , Mice , Nerve Tissue Proteins/metabolism , Protein Kinase C-alpha/metabolism , Retina/physiopathology , Retinopathy of Prematurity/enzymology , Tubulin/metabolism
11.
Invest Ophthalmol Vis Sci ; 55(5): 2853-61, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24677107

ABSTRACT

PURPOSE: Retinal microglia become activated in diabetes and produce pro-inflammatory molecules associated with changes in retinal vasculature and increased apoptosis of retinal neurons and glial cells. We sought to determine if the action of aldose reductase (AR), an enzyme linked to the pathogenesis of diabetic retinopathy, contributes to activation of microglial cells. METHODS: Involvement of AR in the activation process was studied using primary cultures of retinal microglia (RMG) isolated from wild-type and AR-null mice, or in mouse macrophage cultures treated with either AR inhibitors or small interfering RNA (siRNA) directed to AR. Inflammatory cytokines were measured by ELISA. Cell migration was measured using a transwell assay. Gelatin zymography was used to detect active matrix metalloproteinase (MMP)-9, while RMG-induced apoptosis of adult retinal pigment epithelium (ARPE-19) cells was studied in a cell coculture system. RESULTS: Aldose reductase inhibition or genetic deficiency substantially reduced lipopolysacharide (LPS)-induced cytokine secretion from macrophages and RMG. Aldose reductase inhibition or deficiency also reduced the activation of MMP-9 and attenuated LPS-induced cell migration. Additionally, blockade of AR by sorbinil or through genetic means caused a reduction in the ability of activated RMG to induce apoptosis of ARPE-19 cells. CONCLUSIONS: These results demonstrate that the action of AR contributes to the activation of RMG. Inhibition of AR may be a therapeutic strategy to reduce inflammation associated with activation of RMG in disease.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Aldehyde Reductase/physiology , Endotoxins/pharmacology , Microglia/enzymology , Retina/enzymology , Retinal Diseases/enzymology , Aldehyde Reductase/deficiency , Analysis of Variance , Animals , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Western , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Imidazolidines/pharmacology , Macrophages/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Retina/cytology , Retinal Diseases/physiopathology
12.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 29(3): 273-6, 2013 Mar.
Article in Chinese | MEDLINE | ID: mdl-23643085

ABSTRACT

OBJECTIVE: To investigate the role of human aldo-keto reductase 1A1 (AKR1A1) in the resistance to oxidative stress and the metabolism of toxic aldehyde in astrocytoma cells. METHODS: The siRNA was transfected into 1321N1 astrocytoma cells using Lipofectamine(TM); RNAiMax. Western blotting and qRT-PCR were applied to evaluate the knock-down efficiency of AKR1A1. MTT assay was used to examine the cell viability after H2;O2; and 4-hydroxynonenal treatment in AKR1A1 knock-down cells. In addition, the effect of knocking down AKR1A1 on cellular reactive oxygen species (ROS) level in the presence of H2;O2; was measured using 2', 7'-dichlorofluorescein (DCFH-DA). RESULTS: Western blotting and qRT-PCR showed that the AKR1A1-specific siRNA inhibited AKR1A1 gene expression by about 70% in 1321N1 cells. Cells with knock-down of AKR1A1 were more sensitive to H2;O2; and 4-hydroxynonenal-induced cytotoxicity. Furthermore, cellular ROS level in the cells with knock-down of AKR1A1 was much higher than that in the control cells in the presence of H2;O2;. CONCLUSION: The specific siRNA could efficiently inhibit AKR1A1 expression in 1321N1 cells. AKR1A1 could be involved in the metabolism of 4-hydroxynonenal and play a role in the resistance to oxidative stress.


Subject(s)
Aldehyde Reductase/deficiency , Aldehyde Reductase/genetics , Aldehydes/toxicity , Astrocytoma/pathology , Gene Knockdown Techniques , Hydrogen Peroxide/toxicity , Aldehyde Reductase/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Hydrogen Peroxide/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
Vis Neurosci ; 29(6): 267-74, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23101909

ABSTRACT

Streptozotocin (STZ)-induced diabetes is associated with reductions in the electrical response of the outer retina and retinal pigment epithelium (RPE) to light. Aldose reductase (AR) is the first enzyme required in the polyol-mediated metabolism of glucose, and AR inhibitors have been shown to improve diabetes-induced electroretinogram (ERG) defects. Here, we used control and AR -/- mice to determine if genetic inactivation of this enzyme likewise inhibits retinal electrophysiological defects observed in a mouse model of type 1 diabetes. STZ was used to induce hyperglycemia and type 1 diabetes. Diabetic and age-matched nondiabetic controls of each genotype were maintained for 22 weeks, after which ERGs were used to measure the light-evoked components of the RPE (dc-ERG) and the neural retina (a-wave, b-wave). In comparison to their nondiabetic controls, wildtype (WT) and AR -/- diabetic mice displayed significant decreases in the c-wave, fast oscillation, and off response components of the dc-ERG but not in the light peak response. Nondiabetic AR -/- mice displayed larger ERG component amplitudes than did nondiabetic WT mice; however, the amplitude of dc-ERG components in diabetic AR -/- animals were similar to WT diabetics. ERG a-wave amplitudes were not reduced in either diabetic group, but b-wave amplitudes were lower in WT and AR -/-diabetic mice. These findings demonstrate that the light-induced responses of the RPE and outer retina are disrupted in diabetic mice, but these defects are not due to photoreceptor dysfunction, nor are they ameliorated by deletion of AR. This latter finding suggests that benefits observed in other studies utilizing pharmacological inhibitors of AR might have been secondary to off-target effects of the drugs.


Subject(s)
Aldehyde Reductase , Diabetes Mellitus, Experimental , Diabetic Retinopathy , Retinal Pigment Epithelium , Aldehyde Reductase/deficiency , Aldehyde Reductase/genetics , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/genetics , Diabetic Retinopathy/enzymology , Diabetic Retinopathy/genetics , Diabetic Retinopathy/pathology , Disease Models, Animal , Electroretinography , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Retinal Pigment Epithelium/enzymology , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/physiopathology , Streptozocin/adverse effects
14.
Invest Ophthalmol Vis Sci ; 53(9): 5698-712, 2012 Aug 20.
Article in English | MEDLINE | ID: mdl-22836764

ABSTRACT

PURPOSE: Retinal neovascularization is the major pathologic process in many ocular diseases and is associated with oxidative stress. Deficiency of aldose reductase (AR), the first enzyme in the polyol pathway for glucose metabolism, has been shown to reduce oxidative stress and blood vessel leakage. The present study aimed to investigate the effect of AR deficiency on retinal neovascularization in a murine oxygen-induced retinopathy (OIR) model. METHODS: Seven-day-old wild-type (WT) and AR-deficient (AR(-/-)) mice were exposed to 75% oxygen for 5 days and then returned to room air. Vascular obliteration, neovascularization, and blood vessel leakage were analyzed and compared. Immunohistochemistry for AR, nitrotyrosine (NT), poly(ADP-ribose) (PAR), glial fibrillary acidic protein (GFAP), and Iba-1, as well as Western blots for vascular endothelial growth factor (VEGF), phospho-Erk (p-Erk), phospho-Akt (p-Akt), and phospho-IκB (p-IκB) were performed. RESULTS: Compared with WT OIR retinae, AR(-/-) OIR retinae displayed significantly smaller central retinal vaso-obliterated area, less neovascularization, and reduced blood vessel leakage. Significantly reduced oxidative stress and glial responses were also observed in AR(-/-) OIR retinae. Moreover, reduced microglial response in the avascular area but increased microglial responses in the neovascular area were found with AR deficiency. Furthermore, expression levels of VEGF, p-Erk, p-Akt, and p-IκB were significantly reduced in AR(-/-) OIR retinae. CONCLUSIONS: Our observations indicated that AR deficiency reduced retinal vascular changes in the mouse model of OIR, indicating that AR can be a potential therapeutic target in ischemia-induced retinopathy.


Subject(s)
Aldehyde Reductase/deficiency , Disease Models, Animal , Retinal Neovascularization/prevention & control , Retinopathy of Prematurity/prevention & control , Animals , Animals, Newborn , Blotting, Western , Calcium-Binding Proteins/metabolism , Capillary Permeability , Cell Movement , Cell Proliferation , Glial Fibrillary Acidic Protein , Humans , I-kappa B Proteins/metabolism , Immunohistochemistry , Infant, Newborn , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Oxidative Stress , Oxygen/toxicity , Poly Adenosine Diphosphate Ribose/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Retinal Neovascularization/enzymology , Retinal Neovascularization/pathology , Retinal Vessels/metabolism , Retinopathy of Prematurity/enzymology , Retinopathy of Prematurity/pathology , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vascular Endothelial Growth Factor A/metabolism
15.
Am J Physiol Heart Circ Physiol ; 303(3): H297-308, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22661511

ABSTRACT

Earlier studies have demonstrated that aldose reductase (AR) plays a key role in mediating ischemia-reperfusion (I/R) injury. Our objective was to investigate if AR mediates I/R injury by influencing phosphorylation of glycogen synthase kinase-3ß (p-GSK3ß). To investigate this issue, we used three separate models to study the effects of stress injury on the heart. Hearts isolated from wild-type (WT), human expressing AR transgenic (ARTg), and AR knockout (ARKO) mice were perfused with/without GSK3ß inhibitors (SB-216763 and LiCl) and subjected to I/R. Ad-human AR (Ad-hAR)-expressing HL-1 cardiac cells were exposed to hypoxia (0.5% O(2)) and reoxygenation (20.9% O(2)) conditions. I/R in a murine model of transient occlusion and reperfusion of the left anterior descending coronary artery (LAD) was used to study if p-GSK3ß was affected through increased AR flux. Lactate dehydrogenase (LDH) release and left ventricular developed pressure (LVDP) were measured. LVDP was decreased in hearts from ARTg mice compared with WT and ARKO after I/R, whereas LDH release and apoptotic markers were increased (P < 0.05). p-GSK3ß was decreased in ARTg hearts compared with WT and ARKO (P < 0.05). In ARKO, p-GSK3ß and apoptotic markers were decreased compared with WT (P < 0.05). WT and ARTg hearts perfused with GSK3ß inhibitors improved p-GSK3ß expression and LVDP and exhibited decreased LDH release, apoptosis, and mitochondrial pore opening (P < 0.05). Ad-hAR-expressing HL-1 cardiac cells, exposed to hypoxia (0.5% O(2)) and reoxygenation (20.9% O(2)), had greater LDH release compared with control HL-1 cells (P < 0.05). p-GSK3ß was decreased and correlated with increased apoptotic markers in Ad-hAR HL-1 cells (P < 0.05). Treatment with phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) inhibitor increased injury demonstrated by increased LDH release in ARTg, WT, and ARKO hearts and in Ad-hAR-expressing HL-1 cells. Cells treated with protein kinase C (PKC) α/ß inhibitor displayed significant increases in p-Akt and p-GSK3ß expression, and resulted in decreased LDH release. In summary, AR mediates changes in p-GSK3ß, in part, via PKCα/ß and Akt during I/R.


Subject(s)
Aldehyde Reductase/metabolism , Glycogen Synthase Kinase 3/metabolism , Myocardial Reperfusion Injury/enzymology , Myocytes, Cardiac/enzymology , Aldehyde Reductase/deficiency , Aldehyde Reductase/genetics , Animals , Apoptosis , Cell Line , Disease Models, Animal , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase C beta , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Recovery of Function , Transfection , Ventricular Function, Left , Ventricular Pressure
16.
Exp Diabetes Res ; 2012: 789730, 2012.
Article in English | MEDLINE | ID: mdl-22110479

ABSTRACT

We previously demonstrated in streptozotocin-induced diabetic mice that deficiency or inhibition of aldose reductase (AR) caused significant dephosphorylation of hepatic transcriptional factor PPARα, leading to its activation and significant reductions in serum lipid levels. Herein, we report that inhibition of AR by zopolrestat or by a short-hairpin RNA (shRNA) against AR caused a significant reduction in serum and hepatic triglycerides levels in 10-week old diabetic db/db mice. Meanwhile, hyperglycemia-induced phosphorylation of hepatic ERK1/2 and PPARα was significantly attenuated in db/db mice treated with zopolrestat or AR shRNA. Further, in comparison with the untreated db/db mice, the hepatic mRNA expression of Aco and ApoA5, two target genes for PPARα, was increased by 93% (P < 0.05) and 73% (P < 0.05) in zopolrestat-treated mice, respectively. Together, these data indicate that inhibition of AR might lead to significant amelioration in hyperglycemia-induced dyslipidemia and nonalcoholic fatty liver disease.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus/drug therapy , Enzyme Inhibitors/administration & dosage , Fatty Liver/prevention & control , PPAR alpha/physiology , Aldehyde Reductase/deficiency , Animals , Benzothiazoles/administration & dosage , Diabetes Mellitus, Type 2/complications , Hyperglycemia/metabolism , Liver/chemistry , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease , Phosphorylation , Phthalazines/administration & dosage , RNA, Small Interfering/administration & dosage , Triglycerides/analysis , Triglycerides/blood
17.
Respir Res ; 12: 145, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-22054012

ABSTRACT

BACKGROUND: Childhood hospitalization related to asthma remains at historically high levels, and its incidence is on the rise world-wide. Previously, we have demonstrated that aldose reductase (AR), a regulatory enzyme of polyol pathway, is a major mediator of allergen-induced asthma pathogenesis in mouse models. Here, using AR null (AR-/-) mice we have investigated the effect of AR deficiency on the pathogenesis of ragweed pollen extract (RWE)-induced allergic asthma in mice and also examined the efficacy of enteral administration of highly specific AR inhibitor, fidarestat. METHODS: The wild type (WT) and AR-/- mice were sensitized and challenged with RWE to induce allergic asthma. AR inhibitor, fidarestat was administered orally. Airway hyper-responsiveness was measured in unrestrained animals using whole body plethysmography. Mucin levels and Th2 cytokine in broncho-alveolar lavage (BAL) were determined using mouse anti-Muc5A/C ELISA kit and multiplex cytokine array, respectively. Eosinophils infiltration and goblet cells were assessed by H&E and periodic acid Schiff (PAS)-staining of formalin-fixed, paraffin-embedded lung sections. T regulatory cells were assessed in spleen derived CD4+CD25+ T cells population. RESULTS: Deficiency of AR in mice led to significantly decreased PENH, a marker of airway hyper-responsiveness, metaplasia of airway epithelial cells and mucus hyper-secretion following RWE-challenge. This was accompanied by a dramatic decrease in infiltration of eosinophils into sub-epithelium of lung as well as in BAL and release of Th2 cytokines in response to RWE-challenge of AR-/- mice. Further, enteral administration of fidarestat significantly prevented eosinophils infiltration, airway hyper-responsiveness and also markedly increased population of T regulatory (CD4+CD25+FoxP3+) cells as compared to RWE-sensitized and challenged mice not treated with fidarestat. CONCLUSION: Our results using AR-/- mice strongly suggest the role of AR in allergic asthma pathogenesis and effectiveness of oral administration of AR inhibitor in RWE-induced asthma in mice supports the use of AR inhibitors in the treatment of allergic asthma.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Asthma/enzymology , Asthma/prevention & control , Imidazolidines/administration & dosage , Pollen , Rhinitis, Allergic, Seasonal/enzymology , Rhinitis, Allergic, Seasonal/prevention & control , Aldehyde Reductase/deficiency , Aldehyde Reductase/metabolism , Ambrosia/chemistry , Animals , Mice , Mice, Knockout , Plant Extracts , Treatment Outcome
18.
Free Radic Biol Med ; 51(9): 1686-96, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21856412

ABSTRACT

The bacterial endotoxin lipopolysaccharide (LPS) is known to induce release of arachidonic acid (AA) and its metabolic products, which play important roles in the inflammatory process. We have shown earlier that LPS-induced signals in macrophages are mediated by aldose reductase (AR). Here we have investigated the role of AR in LPS-induced release of AA metabolites and their modulation using a potent pharmacological inhibitor, fidarestat, and AR siRNA ablation in RAW264.7 macrophages and AR-knockout mouse peritoneal macrophages and heart tissue. Inhibition or genetic ablation of AR prevented the LPS-induced synthesis and release of AA metabolites such as PGE2, TXB, PGI2, and LTBs in macrophages. LPS-induced activation of cPLA2 was also prevented by AR inhibition. Similarly, AR inhibition also prevented the calcium ionophore A23187-induced cPLA2 and LTB4 in macrophages. Further, AR inhibition by fidarestat prevented the expression of AA-metabolizing enzymes such as COX-2 and LOX-5 in RAW264.7 cells and AR-knockout mouse-derived peritoneal macrophages. LPS-induced expression of AA-metabolizing enzymes and their catalyzed metabolic products was significantly lower in peritoneal macrophages and heart tissue from AR-knockout mice. LPS-induced activation of redox-sensitive signaling intermediates such as MAPKs, transcription factor NF-κB, and EGR-1, a transcriptional regulator of mPGES-1, which in collaboration with COX-2 leads to the production of PGE2, was also significantly prevented by AR inhibition. Taken together, our results indicate that AR mediates LPS-induced inflammation by regulating the AA-metabolic pathway and thus provide a novel role for AR inhibition in preventing inflammatory complications such as sepsis.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Arachidonic Acid/metabolism , Endotoxins/toxicity , Indomethacin/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Quinolines/pharmacology , Aldehyde Reductase/deficiency , Aldehyde Reductase/metabolism , Animals , Cells, Cultured , Inflammation , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Structure-Activity Relationship
19.
Chem Biol Interact ; 191(1-3): 346-50, 2011 May 30.
Article in English | MEDLINE | ID: mdl-21376710

ABSTRACT

Aldose reductase (AKR1B1), which catalyzes the reduction of glucose to sorbitol and lipid aldehydes to lipid alcohols, has been shown to be involved in secondary diabetic complications including cataractogenesis. Rats have high levels of AKR1B1 in lenses and readily develop diabetic cataracts, whereas mice have very low levels of AKR1B1 in their lenses and are not susceptible to hyperglycemic cataracts. Studies with transgenic mice that over-express AKR1B1 indicate that it is the key protein for the development of diabetic complications including diabetic cataract. However, no such studies were performed in genetically altered AKR1B1 rats. Hence, we developed siRNA-based AKR1B1 knockdown rats (ARKO) using the AKR1B1-siRNA-pSuper vector construct. Genotyping analysis suggested that more than 90% of AKR1B1 was knocked down in the littermates. Interestingly, all the male animals were born dead and only 3 female rats survived. Furthermore, all 3 female animals were not able to give birth to F1 generation. Hence, we could not establish an AKR1B1 rat knockdown colony. However, we examined the effect of AKR1B1 knockdown on sugar-induced lens opacification in ex vivo. Our results indicate that rat lenses obtained from AKR1B1 knockdown rats were resistant to high glucose-induced lens opacification as compared to wild-type (WT) rat lenses. Biochemical analysis of lens homogenates showed that the AKR1B1 activity and sorbitol levels were significantly lower in sugar-treated AKR1B1 knockdown rat lenses as compared to WT rat lenses treated with 50mM glucose. Our results thus confirmed the significance of AKR1B1 in the mediation of sugar-induced lens opacification and indicate the use of AKR1B1 inhibitors in the prevention of cataractogenesis.


Subject(s)
Aldehyde Reductase/deficiency , Cataract/chemically induced , Cataract/enzymology , Glucose/pharmacology , Aldehyde Reductase/genetics , Animals , Base Sequence , Cataract/etiology , Cataract/prevention & control , Female , Gene Knockdown Techniques , Hyperglycemia/complications , In Vitro Techniques , Lens, Crystalline/drug effects , Lens, Crystalline/enzymology , Male , Molecular Sequence Data , RNA, Small Interfering/genetics , Rats
20.
Chem Biol Interact ; 191(1-3): 339-45, 2011 May 30.
Article in English | MEDLINE | ID: mdl-21334316

ABSTRACT

Airway inflammation induced by reactive oxygen species (ROS)-mediated activation of redox-sensitive transcription factors is the hallmark of asthma, a prevalent chronic respiratory disease. In various cellular and animal models, we have recently demonstrated that, in response to multiple stimuli, aldose reductase (AKR1B1) regulates the inflammatory signals via NF-kappa B activation. Since NF-κB activation is implicated in asthma pathogenesis, we investigated whether AKR1B1 inhibition could prevent ovalbumin (Ova)- and ragweed pollen extract (RWE)-induced airway inflammation and hyper-responsiveness in mice models and tumor necrosis factor-alpha (TNF-α)-, lipopolysachharide (LPS)- and RWE-induced cytotoxic and inflammatory signals in primary human small airway epithelial cells (SAEC). Sensitization and challenge with Ova or RWE caused airway inflammation and production of inflammatory cytokines, accumulation of eosinophils in airways and sub-epithelial regions, mucin production in the bronchoalveolar lavage fluid, airway hyperresponsiveness, elevated IgE levels and release of Th2 cytokines in the airway and treatment with AKR1B1 inhibitors markedly reduced these pathological changes in mice. In SAEC, treatment with TNF-α, LPS or RWE induced apoptosis, reactive oxygen species generation, synthesis of inflammatory markers IL-6, IL-8, and PGE2 and activation of NF-κB and AP-1. Pharmacological inhibition prevented these changes suggesting that AKR1B1 mediates ROS induced inflammation in small airway epithelial cells. Our results indicate that AKR1B1 inhibitors may offer a novel therapeutic approach to treat inflammatory airway diseases such as asthma.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Aldehyde Reductase/genetics , Asthma/enzymology , Enzyme Inhibitors/pharmacology , RNA, Small Interfering/genetics , Aldehyde Reductase/deficiency , Ambrosia/immunology , Animals , Apoptosis/drug effects , Apoptosis/immunology , Asthma/immunology , Asthma/metabolism , Asthma/pathology , Endotoxins/immunology , Eosinophils/drug effects , Eosinophils/immunology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/immunology , Epithelial Cells/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Gene Knockdown Techniques , Humans , Inflammation/enzymology , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/immunology , Mice , NF-kappa B/metabolism , Ovalbumin/immunology , Pollen/immunology , Reactive Oxygen Species/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL