Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 191
Filter
Add more filters

Publication year range
1.
PLoS Biol ; 16(5): e2006203, 2018 05.
Article in English | MEDLINE | ID: mdl-29795546

ABSTRACT

Ferroptosis is a cell death process driven by damage to cell membranes and linked to numerous human diseases. Ferroptosis is caused by loss of activity of the key enzyme that is tasked with repairing oxidative damage to cell membranes-glutathione peroxidase 4 (GPX4). GPX4 normally removes the dangerous products of iron-dependent lipid peroxidation, protecting cell membranes from this type of damage; when GPX4 fails, ferroptosis ensues. Ferroptosis is distinct from apoptosis, necroptosis, necrosis, and other modes of cell death. Several key mysteries regarding how cells die during ferroptosis remain unsolved. First, the drivers of lipid peroxidation are not yet clear. Second, the subcellular location of lethal lipid peroxides remains an outstanding question. Finally, how exactly lipid peroxidation leads to cell death is an unsolved mystery. Answers to these questions will provide insights into the mechanisms of ferroptotic cell death and associated human diseases, as well as new therapeutic strategies for such diseases.


Subject(s)
Cell Death , Glutathione Peroxidase/deficiency , Lipid Peroxidation , Humans , Lipoxygenases/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase
2.
Development ; 144(9): 1687-1697, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28302747

ABSTRACT

The Wnt/ß-catenin signaling pathway plays pivotal roles in axis formation during embryogenesis and in adult tissue homeostasis. Glutathione peroxidase 4 (GPX4) is a selenoenzyme and participates in the reduction of peroxides. Its synthesis depends on the availability of the element selenium. However, the roles of GPX4 in vertebrate embryonic development and underlying mechanisms are largely unknown. Here, we show that maternal loss of zebrafish gpx4b promotes embryonic dorsal organizer formation, whereas overexpression of gpx4b inhibits the development of the dorsal organizer. Depletion of human GPX4 and zebrafish gpx4b (GPX4/gpx4b) increases, while GPX4/gpx4b overexpression decreases, Wnt/ß-catenin signaling in vivo and in vitro Functional and epistatic studies showed that GPX4 functions at the Tcf/Lef level, independently of selenocysteine activation. Mechanistically, GPX4 interacts with Tcf/Lefs and inhibits Wnt activity by preventing the binding of Tcf/Lefs to the promoters of Wnt target genes, resulting in inhibitory action in the presence of Wnt/ß-catenin signaling. Our findings unravel GPX4 as a suppressor of Wnt/ß-catenin signals, suggesting a possible relationship between the Wnt/ß-catenin pathway and selenium via the association of Tcf/Lef family proteins with GPX4.


Subject(s)
Embryo, Nonmammalian/enzymology , Glutathione Peroxidase/metabolism , Organizers, Embryonic/enzymology , Wnt Signaling Pathway , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Amino Acid Sequence , Animals , Base Sequence , CRISPR-Cas Systems/genetics , Embryo, Nonmammalian/cytology , Evolution, Molecular , Gene Expression Regulation, Developmental , Glutathione Peroxidase/chemistry , Glutathione Peroxidase/deficiency , HEK293 Cells , Humans , Phenotype , Phospholipid Hydroperoxide Glutathione Peroxidase , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Selenium/metabolism , Signal Transduction/genetics , Transcription, Genetic , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics , Zygote/metabolism
3.
Amino Acids ; 52(3): 371-385, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31902007

ABSTRACT

The aim of this study was to investigate the effect of the chronic administration of methionine (Met) and/or its metabolite, methionine sulfoxide (MetO), on the behavior and neurochemical parameters of young rats. Rats were treated with saline (control), Met (0.2-0.4 g/kg), MetO (0.05-0.1 g/kg), and/or a combination of Met + MetO, subcutaneously twice a day from postnatal day 6 (P6) to P28. The results showed that Met, MetO, and Met + MetO impaired short-term and spatial memories (P < 0.05), reduced rearing and grooming (P < 0.05), but did not alter locomotor activity (P > 0.05). Acetylcholinesterase activity was increased in the cerebral cortex, hippocampus, and striatum following Met and/or MetO (P < 0.05) treatment, while Na+, K+-ATPase activity was reduced in the hippocampus (P < 0.05). There was an increase in the level of thiobarbituric acid reactive substances (TBARS) in the cerebral cortex in Met-, MetO-, and Met + MetO-treated rats (P < 0.05). Met and/or MetO treatment reduced superoxide dismutase, catalase, and glutathione peroxidase activity, total thiol content, and nitrite levels, and increased reactive oxygen species and TBARS levels in the hippocampus and striatum (P < 0.05). Hippocampal brain-derived neurotrophic factor was reduced by MetO and Met + MetO compared with the control group. The number of NeuN-positive cells was decreased in the CA3 in Met + MetO group and in the dentate gyrus in the Met, MetO, and Met + MetO groups compared to control group (P < 0.05). Taken together, these findings further increase our understanding of changes in the brain in hypermethioninemia by elucidating behavioral alterations, biological mechanisms, and the vulnerability of brain function to high concentrations of Met and MetO.


Subject(s)
Amino Acid Metabolism, Inborn Errors/complications , Glycine N-Methyltransferase/deficiency , Hippocampus/pathology , Memory Disorders/etiology , Memory Disorders/pathology , Methionine/analogs & derivatives , Reactive Oxygen Species/metabolism , Acetylcholinesterase/metabolism , Amino Acid Metabolism, Inborn Errors/chemically induced , Amino Acid Metabolism, Inborn Errors/metabolism , Animals , Catalase/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Female , Glutathione Peroxidase/deficiency , Glycine N-Methyltransferase/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Memory Disorders/metabolism , Memory, Short-Term/drug effects , Methionine/metabolism , Methionine/toxicity , Rats , Rats, Wistar , Spatial Memory/drug effects , Superoxide Dismutase/deficiency , Thiobarbituric Acid Reactive Substances/metabolism
4.
Neurochem Res ; 45(12): 2991-3002, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33064252

ABSTRACT

A growing body evidence suggests that selenium (Se) deficiency is associated with an increased risk of developing Alzheimer's disease (AD). Se-dependent glutathione peroxidase-1 (GPx-1) of a major antioxidant enzyme, and the most abundant isoform of GPx in the brain. In the present study, we investigated whether GPx-1 is protective against memory impairments induced by beta-amyloid (Aß) (1-42) in mice. As the alteration of protein kinase C (PKC)-mediated ERK activation was recognized in the early stage of AD, we examined whether the GPx-1 gene modulates Aß (1-42)-induced changes in PKC and ERK levels. We observed that Aß (1-42) treatment (400 pmol, i.c.v.) significantly decreased PKC ßII expression in the hippocampus of mice. Aß (1-42)-induced neurotoxic changes [i.e., oxidative stress (i.e., reactive oxygen species, 4-hydroxy-2-noneal, and protein carbonyl), reduced PKC ßII and phospho-ERK expressions, and memory impairment under Y-maze and passive avoidance test] were more pronounced in GPx-1 knockout than in wild type mice. Importantly, exposure to a GPx-1 gene-encoded adenovirus vector (Adv-GPx-1) significantly increased GPx-1 mRNA and GPx activity in the hippocampus of GPx-1 knockout mice. Adv-GPx-1 exposure also significantly blocked the neurotoxic changes induced by Aß (1-42) in GPx-1 knockout mice. Treatment with ERK inhibitor U0126 did not significantly change Adv-GPx-1-mediated attenuation in PKC ßII expression. In contrast, treatment with PKC inhibitor chelerythrine (CHE) reversed Adv-GPx-1-mediated attenuation in ERK phosphorylation, suggesting that PKC ßII-mediated ERK signaling is important for Adv-GPx-1-mediated potentials against Aß (1-42) insult. Our results suggest that treatment with the antioxidant gene GPx-1 rescues Aß (1-42)-induced memory impairment via activating PKC ßII-mediated ERK signaling.


Subject(s)
Glutathione Peroxidase/deficiency , Glutathione Peroxidase/pharmacology , MAP Kinase Signaling System/drug effects , Memory Disorders/enzymology , Memory/drug effects , Protein Kinase C beta/metabolism , Adenoviridae/genetics , Amyloid beta-Peptides , Animals , Gene Expression/drug effects , Genetic Therapy , Glutathione Peroxidase/genetics , Hippocampus/enzymology , Hippocampus/metabolism , Male , Memory Disorders/chemically induced , Memory Disorders/genetics , Memory Disorders/therapy , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments , Glutathione Peroxidase GPX1
5.
Nat Chem Biol ; 13(1): 91-98, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27842070

ABSTRACT

Ferroptosis is a form of regulated necrotic cell death controlled by glutathione peroxidase 4 (GPX4). At present, mechanisms that could predict sensitivity and/or resistance and that may be exploited to modulate ferroptosis are needed. We applied two independent approaches-a genome-wide CRISPR-based genetic screen and microarray analysis of ferroptosis-resistant cell lines-to uncover acyl-CoA synthetase long-chain family member 4 (ACSL4) as an essential component for ferroptosis execution. Specifically, Gpx4-Acsl4 double-knockout cells showed marked resistance to ferroptosis. Mechanistically, ACSL4 enriched cellular membranes with long polyunsaturated ω6 fatty acids. Moreover, ACSL4 was preferentially expressed in a panel of basal-like breast cancer cell lines and predicted their sensitivity to ferroptosis. Pharmacological targeting of ACSL4 with thiazolidinediones, a class of antidiabetic compound, ameliorated tissue demise in a mouse model of ferroptosis, suggesting that ACSL4 inhibition is a viable therapeutic approach to preventing ferroptosis-related diseases.


Subject(s)
Apoptosis , Coenzyme A Ligases/metabolism , Glutathione Peroxidase/metabolism , Mammary Neoplasms, Experimental/metabolism , Animals , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Coenzyme A Ligases/antagonists & inhibitors , Coenzyme A Ligases/deficiency , Female , Glutathione Peroxidase/deficiency , Humans , Hypoglycemic Agents/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Necrosis , Phospholipid Hydroperoxide Glutathione Peroxidase , Thiazolidinediones/pharmacology
6.
Clin Exp Pharmacol Physiol ; 46(9): 791-797, 2019 09.
Article in English | MEDLINE | ID: mdl-31332816

ABSTRACT

The aim of this study was to investigate whether the glutathione peroxidase-1 gene (GPx-1) affects cocaine-induced conditioned place preference (CPP) using a mouse model. Cocaine-induced CPP was accompanied by an increase in the level of σ-1 receptor in the nucleus accumbens (NAc). This phenomenon was more pronounced in the GPx-1 gene knockout (GPx-1 KO) than in wild type (WT) mice. In contrast, the CPP and expression of σ-1 receptor were much less pronounced in GPx-1-overexpressing transgenic (GPx-1 TG) mice than non-transgenic (non-TG) mice. Treatment of the mice with BD1047, a σ-1 receptor antagonist, significantly attenuated both cocaine-induced CPP and c-Fos-immunoreactivity (c-Fos-IR) in WT and GPx-1 KO mice, although the effects were more evident in the latter group. Despite the protective effects of BD1047 on cocaine-induced CPP and c-Fos in non-TG mice, there were no additional protective effects in cocaine-treated GPx-1 TG mice, indicating that the σ-1 receptor is a critical target for GPx-1-mediated psychoprotective activity. Overall, our results suggest that GPx-1 attenuates cocaine-induced CPP via inhibition of σ-1 receptor expression.


Subject(s)
Behavior, Animal/drug effects , Cocaine/pharmacology , Conditioning, Psychological/drug effects , Gene Expression Regulation/drug effects , Glutathione Peroxidase/genetics , Receptors, sigma/genetics , Animals , Gene Knockout Techniques , Glutathione Peroxidase/deficiency , Mice , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Glutathione Peroxidase GPX1 , Sigma-1 Receptor
7.
Diabetologia ; 59(12): 2632-2644, 2016 12.
Article in English | MEDLINE | ID: mdl-27628106

ABSTRACT

AIMS/HYPOTHESIS: In obesity oxidative stress is thought to contribute to the development of insulin resistance, non-alcoholic fatty liver disease and the progression to non-alcoholic steatohepatitis. Our aim was to examine the precise contributions of hepatocyte-derived H2O2 to liver pathophysiology. METHODS: Glutathione peroxidase (GPX) 1 is an antioxidant enzyme that is abundant in the liver and converts H2O2 to water. We generated Gpx1 lox/lox mice to conditionally delete Gpx1 in hepatocytes (Alb-Cre;Gpx1 lox/lox) and characterised mice fed chow, high-fat or choline-deficient amino-acid-defined (CDAA) diets. RESULTS: Chow-fed Alb-Cre;Gpx1 lox/lox mice did not exhibit any alterations in body composition or energy expenditure, but had improved insulin sensitivity and reduced fasting blood glucose. This was accompanied by decreased gluconeogenic and increased glycolytic gene expression as well as increased hepatic glycogen. Hepatic insulin receptor Y1163/Y1163 phosphorylation and Akt Ser-473 phosphorylation were increased in fasted chow-fed Alb-Cre;Gpx1 lox/lox mice, associated with increased H2O2 production and insulin signalling in isolated hepatocytes. The enhanced insulin signalling was accompanied by the increased oxidation of hepatic protein tyrosine phosphatases previously implicated in the attenuation of insulin signalling. High-fat-fed Alb-Cre;Gpx1 lox/lox mice did not exhibit alterations in weight gain or hepatosteatosis, but exhibited decreased hepatic inflammation, decreased gluconeogenic gene expression and increased insulin signalling in the liver. Alb-Cre;Gpx1 lox/lox mice fed a CDAA diet that promotes non-alcoholic steatohepatitis exhibited decreased hepatic lymphocytic infiltrates, inflammation and liver fibrosis. CONCLUSIONS/INTERPRETATION: Increased hepatocyte-derived H2O2 enhances hepatic insulin signalling, improves glucose control and protects mice from the development of non-alcoholic steatohepatitis.


Subject(s)
Fatty Liver/metabolism , Glucose/metabolism , Glutathione Peroxidase/deficiency , Glutathione Peroxidase/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/metabolism , Alleles , Animals , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/metabolism , Hepatocytes/metabolism , Hydrogen Peroxide/metabolism , Insulin Resistance/physiology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Reactive Oxygen Species/metabolism , Glutathione Peroxidase GPX1
8.
Circ Res ; 113(4): 408-17, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23770613

ABSTRACT

RATIONALE: Growing evidence indicates that oxidative stress contributes markedly to endothelial dysfunction. The selenoenzyme glutathione peroxidase 4 (Gpx4) is an intracellular antioxidant enzyme important for the protection of membranes by its unique activity to reduce complex hydroperoxides in membrane bilayers and lipoprotein particles. Yet a role of Gpx4 in endothelial cell function has remained enigmatic. OBJECTIVE: To investigate the role of Gpx4 ablation and subsequent lipid peroxidation in the vascular compartment in vivo. METHODS AND RESULTS: Endothelium-specific deletion of Gpx4 had no obvious impact on normal vascular homeostasis, nor did it impair tumor-derived angiogenesis in mice maintained on a normal diet. In stark contrast, aortic explants from endothelium-specific Gpx4 knockout mice showed a markedly reduced number of endothelial branches in sprouting assays. To shed light onto this apparent discrepancy between the in vivo and ex vivo results, we depleted mice of a second antioxidant, vitamin E, which is normally absent under ex vivo conditions. Therefore, mice were fed a vitamin E-depleted diet for 6 weeks before endothelial deletion of Gpx4 was induced by 4-hydroxytamoxifen. Surprisingly, ≈80% of the knockout mice died. Histopathological analysis revealed detachment of endothelial cells from the basement membrane and endothelial cell death in multiple organs, which triggered thrombus formation. Thromboembolic events were the likely cause of various clinical pathologies, including heart failure, renal and splenic microinfarctions, and paraplegia. CONCLUSIONS: Here, we show for the first time that in the absence of Gpx4, sufficient vitamin E supplementation is crucial for endothelial viability.


Subject(s)
Glutathione Peroxidase/deficiency , Glutathione Peroxidase/genetics , Thrombosis/etiology , Thrombosis/mortality , Vitamin E Deficiency/complications , Vitamin E/genetics , Animals , Apoptosis/physiology , Blood Pressure/physiology , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Glutathione Peroxidase/metabolism , Heart Rate/physiology , Lipid Peroxidation/physiology , Male , Mice , Mice, Knockout , Mice, Transgenic , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/physiopathology , Oxidative Stress/physiology , Phospholipid Hydroperoxide Glutathione Peroxidase , Thrombosis/physiopathology , Vitamin E/metabolism , Vitamin E Deficiency/metabolism , Vitamin E Deficiency/physiopathology
9.
J Cell Sci ; 125(Pt 24): 6147-56, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23132926

ABSTRACT

Protection of satellite cells from cytotoxic damages is crucial to ensure efficient adult skeletal muscle regeneration and to improve therapeutic efficacy of cell transplantation in degenerative skeletal muscle diseases. It is therefore important to identify and characterize molecules and their target genes that control the viability of muscle stem cells. Recently, we demonstrated that high aldehyde dehydrogenase activity is associated with increased viability of human myoblasts. In addition to its detoxifying activity, aldehyde dehydrogenase can also catalyze the irreversible oxidation of vitamin A to retinoic acid; therefore, we examined whether retinoic acid is important for myoblast viability. We showed that when exposed to oxidative stress induced by hydrogen peroxide, adherent human myoblasts entered apoptosis and lost their capacity for adhesion. Pre-treatment with retinoic acid reduced the cytotoxic damage ex vivo and enhanced myoblast survival in transplantation assays. The effects of retinoic acid were maintained in dystrophic myoblasts derived from facioscapulohumeral patients. RT-qPCR analysis of antioxidant gene expression revealed glutathione peroxidase 3 (Gpx3), a gene encoding an antioxidant enzyme, as a potential retinoic acid target gene in human myoblasts. Knockdown of Gpx3 using short interfering RNA induced elevation in reactive oxygen species and cell death. The anti-cytotoxic effects of retinoic acid were impaired in GPx3-inactivated myoblasts, which indicates that GPx3 regulates the antioxidative effects of retinoic acid. Therefore, retinoid status and GPx3 levels may have important implications for the viability of human muscle stem cells.


Subject(s)
Glutathione Peroxidase/genetics , Myoblasts/cytology , Myoblasts/enzymology , Adult , Animals , Antioxidants/pharmacology , Apoptosis , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Gene Knockdown Techniques , Glutathione Peroxidase/deficiency , Glutathione Peroxidase/metabolism , Humans , Mice , Mice, SCID , Myoblasts/drug effects , Reactive Oxygen Species/metabolism , Tretinoin/pharmacology
10.
Am J Respir Cell Mol Biol ; 48(1): 17-26, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23002098

ABSTRACT

Oxidative stress caused by excessive reactive oxygen species production is implicated in influenza A virus-induced lung disease. Glutathione peroxidase (GPx)-1 is an antioxidant enzyme that may protect lungs from such damage. The objective of this study was to determine if GPx-1 protects the lung against influenza A virus-induced lung inflammation in vivo. Male wild-type (WT) or GPx-1(-/-) mice were inoculated with HKx31 (H3N2, 1 × 10(4) plaque-forming units), and bronchoalveolar lavage fluid (BALF)/lung compartments were analyzed on Days 3 and 7 after infection for inflammatory marker expression, histology, and viral titer. WT mice infected with HKx31 had significantly more BALF total cells, macrophages, neutrophils, and lymphocytes at Days 3 and 7 compared with naive WT animals (n = 5-8; P < 0.05). However, infected GPx-1(-/-) mice had significantly more BALF inflammation, which included more total cells, macrophages, and neutrophils, compared with WT mice, and this was abolished by treatment with the GPx mimetic ebselen. BALF inflammation persisted in GPx-1(-/-) mice on Day 10 after infection, and GPx-1(-/-) mice had significantly more influenza-specific CD8(+) T cells in spleen compared with WT mice (n = 3-4; P < 0.05). Infected GPx-1(-/-) mice had greater peribronchial and parenchymal inflammation than WT mice, and viral titer was significantly reduced in GPx-1(-/-) mice at Day 3 (n = 5; P < 0.05). Gene expression analysis revealed that infected GPx-1(-/-) mice had higher whole lung TNF-α, macrophage inflammatory protein (MIP)-1α, MIP-2, KC, and matrix metalloproteinase (MMP)-12 mRNA compared with infected WT mice. GPx-1(-/-) mice had more MIP-2 protein in BALF at Day 3 and more active MMP-9 protease in BALF at Days 3 and 7 than WT mice. These data indicate that GPx-1 reduces influenza A virus-induced lung inflammation.


Subject(s)
Glutathione Peroxidase/physiology , Influenza A Virus, H3N2 Subtype , Orthomyxoviridae Infections/enzymology , Pneumonia/enzymology , Pneumonia/prevention & control , Adaptive Immunity , Animals , Azoles/pharmacology , Bronchoalveolar Lavage Fluid/cytology , CD8-Positive T-Lymphocytes/immunology , Chemokines/genetics , Cytokines/genetics , Glutathione Peroxidase/deficiency , Glutathione Peroxidase/genetics , Influenza A Virus, H3N2 Subtype/immunology , Isoindoles , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organoselenium Compounds/pharmacology , Orthomyxoviridae Infections/etiology , Orthomyxoviridae Infections/pathology , Peptide Hydrolases/genetics , Pneumonia/etiology , Pneumonia/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species , Viral Load , Glutathione Peroxidase GPX1
11.
Am J Respir Cell Mol Biol ; 49(5): 721-30, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23590304

ABSTRACT

Protein phosphatase-2A (PP2A) is a primary serine-threonine phosphatase that modulates inflammatory responses in asthma and chronic obstructive pulmonary disease (COPD). Despite its importance, the mechanisms that regulate lung PP2A activity remain to be determined. The redox-sensitive enzyme protein tyrosine phosphatase-1B (PTP1B) activates PP2A by dephosphorylating the catalytic subunit of the protein at tyrosine 307. This study aimed to identify how the interaction between the intracellular antioxidant glutathione peroxidase-1 (GPx-1) and PTP1B affected lung PP2A activity and airway inflammation. Experiments using gene silencing techniques in mouse lung or human small airway epithelial cells determined that knocking down PTP1B expression blocked GPx-1's activation of PP2A and negated the anti-inflammatory effects of GPx-1 protein in the lung. Similarly, the expression of human GPx-1 in transgenic mice significantly increased PP2A and PTP1B activities and prevented chronic cigarette smoke-induced airway inflammation and alveolar destruction. GPx-1 knockout mice, however, exhibited an exaggerated emphysema phenotype, correlating with a nonresponsive PP2A pathway. Importantly, GPx-1-PTP1B-PP2A signaling becomes inactivated in advanced lung disease. Indeed, PTP1B protein was oxidized in the lungs of subjects with advanced emphysema, and cigarette smoke did not increase GPx-1 or PTP1B activity within epithelial cells isolated from subjects with COPD, unlike samples of healthy lung epithelial cells. In conclusion, these findings establish that the GPx-1-PTP1B-PP2A axis plays a critical role in countering the inflammatory and proteolytic responses that result in lung-tissue destruction in response to cigarette smoke exposure.


Subject(s)
Glutathione Peroxidase/metabolism , Pneumonia/enzymology , Protein Phosphatase 2/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Pulmonary Alveoli/enzymology , Respiratory Mucosa/enzymology , Signal Transduction , Animals , Case-Control Studies , Cell Line , Enzyme Activation , Gene Knockdown Techniques , Glutathione Peroxidase/deficiency , Glutathione Peroxidase/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Oxidation-Reduction , Oxidative Stress , Phosphorylation , Pneumonia/etiology , Pneumonia/genetics , Pneumonia/pathology , Pneumonia/prevention & control , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Pulmonary Alveoli/pathology , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Emphysema/enzymology , Pulmonary Emphysema/pathology , RNA Interference , Respiratory Mucosa/pathology , Smoking/adverse effects , Transfection , Glutathione Peroxidase GPX1
12.
Circ Res ; 109(7): 739-49, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21835911

ABSTRACT

RATIONALE: Peroxiredoxin 2 (Prdx2), a thiol-specific peroxidase, has been reported to regulate proinflammatory responses, vascular remodeling, and global oxidative stress. OBJECTIVE: Although Prdx2 has been proposed to retard atherosclerosis development, no direct evidence and mechanisms have been reported. METHODS AND RESULTS: We show that Prdx2 is highly expressed in endothelial and immune cells in atherosclerotic lesions and blocked the increase of endogenous H(2)O(2) by atherogenic stimulation. Deficiency of Prdx2 in apolipoprotein E-deficient (ApoE(-/-)) mice accelerated plaque formation with enhanced activation of p65, c-Jun, JNKs, and p38 mitogen-activated protein kinase; and these proatherogenic effects of Prdx2 deficiency were rescued by administration of the antioxidant ebselen. In bone marrow transplantation experiments, we found that Prdx2 has a major role in inhibiting atherogenic responses in both vascular and immune cells. Prdx2 deficiency resulted in increased expression of vascular adhesion molecule-1, intercellular adhesion molecule-1, and monocyte chemotactic protein-1, which led to increased immune cell adhesion and infiltration into the aortic intima. Compared with deficiency of glutathione peroxidase 1 or catalase, Prdx2 deficiency showed a severe predisposition to develop atherosclerosis. CONCLUSIONS: Prdx2 is a specific peroxidase that inhibits atherogenic responses in vascular and inflammatory cells, and specific activation of Prdx2 may be an effective means of antiatherogenic therapy.


Subject(s)
Aorta/enzymology , Apolipoproteins E/deficiency , Atherosclerosis/enzymology , Peroxiredoxins/deficiency , Animals , Antioxidants/pharmacology , Aorta/drug effects , Aorta/immunology , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Azoles/pharmacology , Bone Marrow Cells/enzymology , Bone Marrow Transplantation , Catalase/genetics , Catalase/metabolism , Chemokine CCL2/metabolism , Disease Models, Animal , Endothelial Cells/enzymology , Glutathione Peroxidase/deficiency , Glutathione Peroxidase/genetics , Hydrogen Peroxide/metabolism , Intercellular Adhesion Molecule-1/metabolism , Isoindoles , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Organoselenium Compounds/pharmacology , Peroxiredoxins/genetics , Severity of Illness Index , Signal Transduction , Time Factors , Transcription Factor RelA/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Glutathione Peroxidase GPX1
13.
Biosci Biotechnol Biochem ; 77(5): 917-22, 2013.
Article in English | MEDLINE | ID: mdl-23649250

ABSTRACT

To remove nitrogen efficiently from high-concentration organic wastewater, we studied breeding methods using Saccharomyces cerevisiae as a model yeast with improved nitrogen accumulation ability. By DNA microarray analysis under various nitrogen concentrations with two nitrogen sources (peptone and L-asparagine), we obtained 295 commonly overexpressed (over 2-fold) genes and 283 commonly underexpressed (under one-half) genes under nitrogen-starvation conditions. We speculated that overexpression or underexpression recombination of some of these genes might enhance nitrogen uptake. Because a complete collection of nonessential gene deletion strains had been created, we investigated the nitrogen accumulation profiles of underexpressed gene deletion strains. From 256 nonessential gene deletion strains, three (URE2, SNO1, and AVT3) were selected. Strain SUD2 (ure2Δ::kanMX4) improved by 1.2-fold total nitrogen per cell (TN/OD660) as compared to the parent strain, S288c. Positive selection of methylamine-resistant mutants to obtain URE2 mutants was useful for improving nitrogen accumulation ability without recombinant techniques.


Subject(s)
Genetic Engineering/methods , Genomics , Nitrogen/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Biological Transport , Gene Deletion , Glutathione Peroxidase/deficiency , Glutathione Peroxidase/genetics , Nitrogen/isolation & purification , Prions/genetics , Saccharomyces cerevisiae Proteins/genetics , Waste Management , Wastewater/microbiology
14.
J Cell Physiol ; 227(4): 1420-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21618532

ABSTRACT

The nuclear isoform of the selenoprotein Phospholipid Hydroperoxide Glutathione Peroxidase (nGPx4) is expressed in haploid male germ cells, contains several cysteines and is able to oxidize protein thiols, besides glutathione. In this study we have investigated the subnuclear localization of this isoform in isolated mouse male germ cells at different steps of maturation. Immunoblotting and confocal microscopy analyses of subnuclear fractions showed that nGPx4 is localized to the nuclear matrix together with well known markers of this subnuclear compartment like lamin B and topoisomerase IIß at all stages of germ cell differentiation. The peculiar nGPx4 distribution was confirmed by both biochemical and morphological analyses of COS-1 cells overexpressing Flag-tagged nGPx4. To test the functional role of nGPx4 in the process of chromatin assembly, sperm isolated from the caput and the cauda epididymides of wild-type (WT) and genetically deficient in nGPx4 (nGPx4-KO) mice were analyzed in an in vitro chromatin decondensation assay. Results showed that sperm from nGPx4-KO mice were more prone to decondense than those from WT mice at all stages of epididymal maturation, providing conclusive evidence that nGPx4 is required for a correct sperm chromatin compaction. We next addressed the issue of whether the lack of nGPx4 impacts on early events occurring at fertilization. Indeed, in vitro fertilization experiments showed an acceleration of sperm chromatin dispersion in oocytes fertilized by nGpx4-KO sperm compared with control. Overall these data indicate that the absence of nGPx4 leads to sperm nuclear matrix/chromatin instability that may negatively affect the embryo development.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Fertilization/physiology , Glutathione Peroxidase/metabolism , Spermatozoa/enzymology , Animals , COS Cells , Chlorocebus aethiops , Chromosomal Instability/physiology , Embryonic Development/physiology , Epididymis/cytology , Epididymis/enzymology , Female , Fertilization in Vitro , Glutathione Peroxidase/deficiency , Glutathione Peroxidase/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Matrix/enzymology , Oocytes/physiology , Phospholipid Hydroperoxide Glutathione Peroxidase , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spermatogenesis/physiology , Spermatozoa/physiology , Transfection
15.
J Neuroinflammation ; 9: 123, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22691312

ABSTRACT

BACKGROUND: Gold nanoparticles (AuNPs) are finding increased use in therapeutics and imaging. However, their toxic effects still remain to be elucidated. Therefore this study was undertaken to study the biochemical effects of AuNPs on rat brain and identify potential biomarkers of AuNP toxicity. METHODS: Male Wister rats weighing 150-200 g were injected with 20 µg/kg body weight of 20-nm gold nanoparticles for 3 days through the intraperitoneal route. The rats were killed by carbon dioxide asphyxiation 24 h after the last dose of gold nanoparticle injection. The parameters studied included lipid peroxidation, glutathione peroxidase, 8- hydroxydeoxyguanosine, caspase-3, heat shock protein70, serotonin, dopamine, gamma amino-butyric acid and interferon-γ. RESULTS: In this study AuNPs caused generation of oxidative stress and a decrease of antioxidant enzyme, viz., glutathione peroxidase activity in rat brain. This was accompanied by an increase in 8-hydroxydeoxyguanosine, caspase-3 and heat shock protein70, which might lead to DNA damage and cell death. Gold nanoparticles also caused a significant decrease in the levels of neurotransmitters like dopamine and serotonin, indicating a possible change in the behavior of the treated animals. There was a significant increase in the cerebral levels of IFN-γ in treated animals. CONCLUSION: This study concludes that AuNPs cause generation of oxidative stress and an impairment of the antioxidant enzyme glutathione peroxidase in rat brain. AuNPs also cause generation of 8-hydroxydeoxyguanosine (8OHdG), caspase-3 and heat shock protein70 (Hsp70), and IFN-γ, which may lead to inflammation and DNA damage/cell death.


Subject(s)
Brain Chemistry/drug effects , Gold/toxicity , Metal Nanoparticles/toxicity , Animals , Antioxidants/chemistry , Antioxidants/physiology , Biomarkers/chemistry , Brain Chemistry/physiology , Contraindications , Glutathione Peroxidase/antagonists & inhibitors , Glutathione Peroxidase/biosynthesis , Glutathione Peroxidase/deficiency , Gold/chemistry , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Male , Metal Nanoparticles/chemistry , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Wistar
16.
FASEB J ; 24(7): 2525-32, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20219985

ABSTRACT

CD14 contributes to LPS signaling in leukocytes through formation of toll-like receptor 4/CD14 receptor complexes; however, a specific role for endogenous cell-surface CD14 in endothelial cells is unclear. We have found that suppression of glutathione peroxidase-1 (GPx-1) in human microvascular endothelial cells increases CD14 gene expression compared to untreated or siControl (siCtrl)-treated conditions. Following LPS treatment, GPx-1 deficiency augmented LPS-induced intracellular reactive oxygen species accumulation, CD14 expression, and intercellular adhesion molecule-1 (ICAM-1) mRNA and protein expression compared to LPS-treated control cells. GPx-1 deficiency also transiently augmented LPS-induced vascular cell adhesion molecule-1 (VCAM-1) expression. Adenoviral overexpression of GPx-1 significantly diminished LPS-mediated responses in adhesion molecule expression. Consistent with these findings, LPS responses were also greater in endothelial cells derived from GPx-1-knockout mice, whereas adhesion molecule expression was decreased in cells from GPx-1-overexpressing transgenic mice. Knockdown of CD14 attenuated LPS-mediated up-regulation of ICAM-1 and VCAM-1 mRNA and protein, and it mitigated the effects of GPx-1 deficiency on LPS-induced adhesion molecule expression. Taken together, these data suggest that GPx-1 modulates the endothelial cell response to LPS, in part, by altering CD14-mediated effects.


Subject(s)
Cell Adhesion Molecules/genetics , Endothelial Cells/metabolism , Glutathione Peroxidase/physiology , Lipopolysaccharide Receptors/genetics , Lipopolysaccharides/pharmacology , Transcriptional Activation/drug effects , Cell Adhesion Molecules/analysis , Cells, Cultured , Endothelium, Vascular/cytology , Glutathione Peroxidase/deficiency , Humans , Intercellular Adhesion Molecule-1/analysis , Intercellular Adhesion Molecule-1/genetics , RNA, Messenger/analysis , Reactive Oxygen Species/metabolism , Vascular Cell Adhesion Molecule-1/analysis , Vascular Cell Adhesion Molecule-1/genetics , Glutathione Peroxidase GPX1
17.
Pflugers Arch ; 460(1): 55-68, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20306076

ABSTRACT

Glutathione peroxidase 1 (GPx1) plays an important role in preventing cardiac dysfunction following ischemia-reperfusion injury. However, its role in protecting cardiac mitochondria against reoxygenation-induced reactive oxygen species (ROS) generation in vivo is unclear. We examined the role of GPx1 in protecting cardiac mitochondria against hypoxia-reoxygenation (HR) damage by testing for alterations in cardiac mitochondrial function. We used a two-dimensional gel electrophoresis proteomics analysis to examine the effects of reoxygenation on cardiac protein in wild-type (GPx1(+/+)) and GPx1 knockout (GPx1(-/-)) mouse hearts. We identified 42 protein spots showing differential expression in the two groups. Sixteen of the proteins identified were located in mitochondria and were involved in a number of key metabolic pathways. To verify our proteomics findings functionally, we performed NADH autofluorescence measurements and ATP production assays. The reduced expression of oxidative phosphorylation proteins in GPx1(-/-) mice following HR treatment resulted in loss of the mitochondrial membrane potential and decreased mitochondrial respiration. Mitochondrial ROS production and oxidative mtDNA damage were increased markedly during reoxygenation in GPx1(-/-) hearts. We also found morphological abnormalities in cardiac mitochondria and myocytes in HR-treated GPx1(-/-). This is the first report of the role of GPx1 in protecting cardiac mitochondria against reoxygenation damage in vivo. These findings will help clarify the mechanisms of HR injury and will aid in the development of antioxidant therapies to prevent cardiac mitochondrial dysfunction associated with reoxygenation.


Subject(s)
Glutathione Peroxidase/metabolism , Mitochondria, Muscle/enzymology , Myocardial Reperfusion Injury/prevention & control , Myocardium/enzymology , Reactive Oxygen Species/metabolism , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , Cell Hypoxia , Cytoprotection , DNA Damage , DNA, Mitochondrial/metabolism , Electrophoresis, Gel, Two-Dimensional , Glutathione Peroxidase/deficiency , Glutathione Peroxidase/genetics , Hydrogen Peroxide/metabolism , Male , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Muscle/pathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , NAD/metabolism , Oxygen Consumption , Perfusion , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Proteomics/methods , Superoxides/metabolism , Time Factors , Glutathione Peroxidase GPX1
18.
Am J Physiol Renal Physiol ; 298(5): F1244-53, 2010 May.
Article in English | MEDLINE | ID: mdl-20015939

ABSTRACT

Glutathione peroxidase-3 (Gpx3), also known as plasma or extracellular glutathione peroxidase, is a selenoprotein secreted primarily by kidney proximal convoluted tubule cells. In this study Gpx3(-/-) mice have been produced and immunocytochemical techniques have been developed to investigate Gpx3 metabolism. Gpx3(-/-) mice maintained the same whole-body content and urinary excretion of selenium as did Gpx3(+/+) mice. They tolerated selenium deficiency without observable ill effects. The simultaneous knockout of Gpx3 and selenoprotein P revealed that these two selenoproteins account for >97% of plasma selenium. Immunocytochemistry experiments demonstrated that Gpx3 binds selectively, both in vivo and in vitro, to basement membranes of renal cortical proximal and distal convoluted tubules. Based on calculations using selenium content, the kidney pool of Gpx3 is over twice as large as the plasma pool. These data indicate that Gpx3 does not serve in the regulation of selenium metabolism. The specific binding of a large pool of Gpx3 to basement membranes in the kidney cortex strongly suggests a need for glutathione peroxidase activity in the cortical peritubular space.


Subject(s)
Basement Membrane/metabolism , Glutathione Peroxidase/metabolism , Kidney Cortex/cytology , Kidney Cortex/metabolism , Animals , Female , Glutathione Peroxidase/deficiency , Glutathione Peroxidase/genetics , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Selenium/metabolism , Selenoprotein P/deficiency , Selenoprotein P/genetics , Selenoprotein P/metabolism
19.
Am J Physiol Lung Cell Mol Physiol ; 299(3): L425-33, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20511341

ABSTRACT

Reactive oxygen species (ROS) produced from cigarette smoke cause oxidative lung damage including protein denaturation, lipid peroxidation, and DNA damage. Glutathione peroxidase-1 (gpx-1) is a detoxifying enzyme that may protect lungs from such damage. The aim of this study was to determine whether gpx-1 protects the lung against oxidative stress-induced lung inflammation in vivo. Male wild-type (WT) or gpx-1(-/-) mice were exposed to cigarette smoke generated from nine cigarettes per day for 4 days to induce oxidative stress and lung inflammation. The effect of the gpx mimetic ebselen on cigarette smoke-induced lung inflammation was evaluated when given prophylactically and therapeutically, i.e., during established inflammation. Mice were killed, and the lungs were lavaged with PBS and then harvested for genomic and proteomic analysis. Gpx-1(-/-) mice exposed to cigarette smoke had enhanced BALF neutrophils, macrophages, proteolytic burden, whole lung IL-17A, and MIP1alpha mRNA compared with WT mice. The gpx mimetic ebselen (10 and 100 microM) inhibited cigarette smoke extract-induced oxidation of MH-S cells in vitro and inhibited cigarette smoke-induced increases in BALF macrophages, neutrophils, proteolytic burden, and macrophage and neutrophil chemotactic factor gene expression when administered prophylactically. In addition, ebselen inhibited established BALF inflammation when administered therapeutically. These data show that gpx-1 protects against cigarette smoke-induced lung inflammation, and agents that mimic the actions of gpx-1 may have therapeutic utility in inflammatory lung diseases where cigarette smoke plays a role.


Subject(s)
Glutathione Peroxidase/metabolism , Pneumonia/etiology , Pneumonia/prevention & control , Smoking , Animals , Antioxidants/pharmacology , Azoles/pharmacology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Chemotactic Factors/metabolism , Gene Expression/drug effects , Glutathione Peroxidase/deficiency , Intercellular Signaling Peptides and Proteins/blood , Isoindoles , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Neutrophils/pathology , Organoselenium Compounds/pharmacology , Oxidation-Reduction/drug effects , Peptide Hydrolases/metabolism , Pneumonia/pathology , Proteins/metabolism , Glutathione Peroxidase GPX1
20.
FASEB J ; 23(9): 3233-42, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19417079

ABSTRACT

Selenium is linked to male fertility. Glutathione peroxidase 4 (GPx4), first described as an antioxidant enzyme, is the predominant selenoenzyme in testis and has been suspected of being vital for spermatogenesis. Cytosolic, mitochondrial, and nuclear isoforms are all encoded by the same gene. While disruption of entire GPx4 causes early embryonic lethality in mice, inactivation of nuclear GPx4 does not impair embryonic development or fertility. Here, we show that deletion of mitochondrial GPx4 (mGPx4) allows both normal embryogenesis and postnatal development, but causes male infertility. Infertility was associated with impaired sperm quality and severe structural abnormalities in the midpiece of spermatozoa. Knockout sperm display higher protein thiol content and recapitulate features typical of severe selenodeficiency. Interestingly, male infertility induced by mGPx4 depletion could be bypassed by intracytoplasmic sperm injection. We also show for the first time that mGPx4 is the prevailing GPx4 product in male germ cells and that mGPx4 disruption has no effect on proliferation or apoptosis of germinal or somatic tissue. Our study finally establishes that mitochondrial GPx4 confers the vital role of selenium in mammalian male fertility and identifies cytosolic GPx4 as the only GPx4 isoform being essential for embryonic development and apoptosis regulation.


Subject(s)
Glutathione Peroxidase/physiology , Infertility, Male/etiology , Mitochondrial Proteins/physiology , Animals , Apoptosis , Embryonic Development , Glutathione Peroxidase/deficiency , Male , Mice , Phospholipid Hydroperoxide Glutathione Peroxidase , Selenium/physiology , Spermatozoa/pathology
SELECTION OF CITATIONS
SEARCH DETAIL