Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell Microbiol ; 21(2): e12987, 2019 02.
Article in English | MEDLINE | ID: mdl-30489694

ABSTRACT

The relapsing fever spirochete Borrelia turicatae possesses a complex life cycle in its soft-bodied tick vector, Ornithodoros turicata. Spirochetes enter the tick midgut during a blood meal, and, during the following weeks, spirochetes disseminate throughout O. turicata. A population persists in the salivary glands allowing for rapid transmission to the mammalian hosts during tick feeding. Little is known about the physiological environment within the salivary glands acini in which B. turicatae persists. In this study, we examined the salivary gland transcriptome of O. turicata ticks and detected the expression of 57 genes involved in oxidant metabolism or antioxidant defences. We confirmed the expression of five of the most highly expressed genes, including glutathione peroxidase (gpx), thioredoxin peroxidase (tpx), manganese superoxide dismutase (sod-1), copper-zinc superoxide dismutase (sod-2), and catalase (cat) by reverse-transcriptase droplet digital polymerase chain reaction (RT-ddPCR). We also found distinct differences in the expression of these genes when comparing the salivary glands and midguts of unfed O. turicata ticks. Our results indicate that the salivary glands of unfed O. turicata nymphs are highly oxidative environments where reactive oxygen species (ROS) predominate, whereas midgut tissues comprise a primarily nitrosative environment where nitric oxide synthase is highly expressed. Additionally, B. turicatae was found to be hyperresistant to ROS compared with the Lyme disease spirochete Borrelia burgdorferi, suggesting it is uniquely adapted to the highly oxidative environment of O. turicata salivary gland acini.


Subject(s)
Borrelia/growth & development , Borrelia/physiology , Ornithodoros/microbiology , Relapsing Fever/transmission , Salivary Glands/metabolism , Animals , Catalase/biosynthesis , Catalase/genetics , Gene Expression Regulation/genetics , Glutathione Peroxidase/biosynthesis , Glutathione Peroxidase/genetics , Oxidative Stress/physiology , Peroxiredoxins/biosynthesis , Peroxiredoxins/genetics , Reactive Oxygen Species/metabolism , Relapsing Fever/microbiology , Salivary Glands/microbiology , Superoxide Dismutase-1/biosynthesis , Superoxide Dismutase-1/genetics
2.
Mol Cell ; 48(5): 713-22, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23122414

ABSTRACT

Protein mistranslation causes growth arrest in bacteria, mitochondrial dysfunction in yeast, and neurodegeneration in mammals. It remains poorly understood how mistranslated proteins cause such cellular defects. Here we demonstrate that streptomycin, a bactericidal aminoglycoside that increases ribosomal mistranslation, induces transient protein aggregation in wild-type Escherichia coli. We further determined the aggregated proteome using label-free quantitative mass spectrometry. To identify genes that reduce cellular mistranslation toxicity, we selected from an overexpression library protein products that increased resistance against streptomycin and kanamycin. The selected proteins were significantly enriched in members of the oxidation-reduction pathway. Overexpressing one of these proteins, alkyl hydroperoxide reductase subunit F (a protein defending bacteria against hydrogen peroxide), but not its inactive mutant suppressed aggregated protein formation upon streptomycin treatment and increased aminoglycoside resistance. This work provides in-depth analyses of an aggregated proteome caused by streptomycin and suggests that cellular defense against hydrogen peroxide lowers the toxicity of mistranslation.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Escherichia coli Proteins/biosynthesis , Escherichia coli/drug effects , Free Radical Scavengers/pharmacology , Hydrogen Peroxide/metabolism , Oxidative Stress/drug effects , Protein Biosynthesis/drug effects , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Kanamycin/pharmacology , Mass Spectrometry , Microbial Viability/drug effects , Mutation , Oxidative Stress/genetics , Peroxiredoxins/biosynthesis , Peroxiredoxins/genetics , Protein Folding , Proteomics/methods , Ribosomes/drug effects , Ribosomes/metabolism , Streptomycin/pharmacology , Time Factors
3.
Int J Neurosci ; 130(1): 1-8, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31518514

ABSTRACT

Aim: Sevoflurane could induce apoptosis of rat hippocampal neurons, while theaflavins (TFs) have antioxidant and anti-inflammatory properties. This study aims to explore whether TFs could alleviate sevoflurane-induced neuronal cell injury.Materials and methods: Cells were treated by concentration gradient of sevoflurane and TFs. Cell viability, level of reactive oxygen species (ROS) and apoptosis rate were determined by cell counting kit-8 (CCK-8) and flow cytometry, respectively. Quantitative PCR (qPCR) and western blot were performed to determine mRNA and protein expressions.Results: TFs promoted viability of cells under the treatment of sevoflurane, while it suppressed apoptosis and down-regulated ROS level in a concentration-dependent manner. TFs could also down-regulate expression levels of caspase-3 and caspase-9 and cytosol and intranuclear nuclear factor E2-related factor 2 (Nrf2) in rat hippocampal nerve cells, while it up-regulated those of heme oxygenase 1 (HO-1), NADPH quinine oxidoreductase 1 (NQO1), glutamate cysteine ligase (GCL) and peroxiredoxin 1 (Prx1).Conclusions: Our study suggests that TFs exert protective effects on sevoflurane-induced neurocytotoxicity and therefore could be used as a potential drug for treatment of neuronal injury.


Subject(s)
Biflavonoids/pharmacology , Catechin/pharmacology , Hippocampus/drug effects , NF-E2-Related Factor 2/physiology , Neurons/metabolism , Neurotoxicity Syndromes/prevention & control , Sevoflurane/toxicity , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caspase 3/biosynthesis , Caspase 9/biosynthesis , Cell Survival/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Glutamate-Cysteine Ligase/biosynthesis , Heme Oxygenase-1/biosynthesis , Hippocampus/metabolism , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , NF-E2-Related Factor 2/biosynthesis , Neurons/physiology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/physiopathology , Peroxiredoxins/biosynthesis , Primary Cell Culture , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Up-Regulation/drug effects
4.
BMC Pulm Med ; 19(1): 265, 2019 Dec 30.
Article in English | MEDLINE | ID: mdl-31888585

ABSTRACT

BACKGROUND: Acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF) is life-threatening. Several serum biomarkers, such as Krebs von den Lungen-6 (KL-6) and surfactant protein D (SP-D), are clinically used for evaluating AE-IPF, but these biomarkers are not adequate for establishing an early and accurate diagnosis of AE-IPF. Recently, the protective roles of the members of the peroxiredoxin (PRDX) family have been reported in IPF; however, the role of PRDX4 in AE-IPF is unclear. METHODS: Serum levels of PRDX4 protein, KL-6, SP-D and lactate dehydrogenase (LDH) in 51 patients with stable IPF (S-IPF), 38 patients with AE-IPF and 15 healthy volunteers were retrospectively assessed using enzyme-linked immunosorbent assay. Moreover, as an animal model of pulmonary fibrosis, wild-type (WT) and PRDX4-transgenic (Tg) mice were intratracheally administered with bleomycin (BLM, 2 mg/kg), and fibrotic and inflammatory changes in lungs were evaluated 3 weeks after the intratracheal administration. RESULTS: Serum levels of PRDX4 protein, KL-6, SP-D and LDH in patients with S-IPF and AE-IPF were significantly higher than those in healthy volunteers, and those in AE-IPF patients were the highest among the three groups. Using receiver operating characteristic curves, area under the curve values of serum PRDX4 protein, KL-6, SP-D, and LDH for detecting AE-IPF were 0.873, 0.698, 0.675, and 0.906, respectively. BLM-treated Tg mice demonstrated aggravated histopathological findings and poor prognosis compared with BLM-treated WT mice. Moreover, PRDX4 expression was observed in alveolar macrophages and lung epithelial cells of BLM-treated Tg mice. CONCLUSIONS: PRDX4 is associated with the aggravation of inflammatory changes and fibrosis in the pathogenesis of IPF, and serum PRDX4 may be useful in clinical practice of IPF patients.


Subject(s)
Disease Progression , Idiopathic Pulmonary Fibrosis/blood , Idiopathic Pulmonary Fibrosis/etiology , Peroxiredoxins/biosynthesis , Adult , Aged , Animals , Biomarkers/blood , Disease Models, Animal , Female , Humans , Male , Mice , Middle Aged , Retrospective Studies
5.
Clin Sci (Lond) ; 132(13): 1471-1485, 2018 07 18.
Article in English | MEDLINE | ID: mdl-29674526

ABSTRACT

Galectin-3 (Gal-3) is increased in heart failure (HF) and promotes cardiac fibrosis and inflammation. We investigated whether Gal-3 modulates oxidative stress in human cardiac fibroblasts, in experimental animal models and in human aortic stenosis (AS). Using proteomics and immunodetection approaches, we have identified that Gal-3 down-regulated the antioxidant peroxiredoxin-4 (Prx-4) in cardiac fibroblasts. In parallel, Gal-3 increased peroxide, nitrotyrosine, malondialdehyde, and N-carboxymethyl-lysine levels and decreased total antioxidant capacity. Gal-3 decreased prohibitin-2 expression without modifying other mitochondrial proteins. Prx-4 silencing increased oxidative stress markers. In Gal-3-silenced cells and in heart from Gal-3 knockout mice, Prx-4 was increased and oxidative stress markers were decreased. Pharmacological inhibition of Gal-3 with modified citrus pectin restored cardiac Prx-4 as well as prohibitin-2 levels and improved oxidative status in spontaneously hypertensive rats. In serum from 87 patients with AS, Gal-3 negatively correlated with total antioxidant capacity and positively correlated with peroxide. In myocardial biopsies from 26 AS patients, Gal-3 up-regulation paralleled a decrease in Prx-4 and in prohibitin-2. Cardiac Gal-3 inversely correlated with Prx-4 levels in myocardial biopsies. These data suggest that Gal-3 decreased Prx-4 antioxidant system in cardiac fibroblasts, increasing oxidative stress. In pathological models presenting enhanced cardiac Gal-3, the decrease in Prx-4 expression paralleled increased oxidative stress. Gal-3 blockade restored Prx-4 expression and improved oxidative stress status. In AS, circulating levels of Gal-3 could reflect oxidative stress. The alteration of the balance between antioxidant systems and reactive oxygen species production could be a new pathogenic mechanism by which Gal-3 induces cardiac damage in HF.


Subject(s)
Down-Regulation/drug effects , Galectin 3/pharmacology , Heart/drug effects , Peroxiredoxins/biosynthesis , Aged , Aged, 80 and over , Animals , Antioxidants/metabolism , Aortic Valve Stenosis/blood , Aortic Valve Stenosis/pathology , Aortic Valve Stenosis/physiopathology , Biopsy , Blood Proteins , Cells, Cultured , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Galectin 3/blood , Galectin 3/deficiency , Galectins , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocardium/metabolism , Myocardium/pathology , Oxidative Stress/drug effects , Peroxiredoxins/genetics , Prospective Studies , Proteomics/methods
6.
Exp Mol Pathol ; 105(3): 334-344, 2018 12.
Article in English | MEDLINE | ID: mdl-30290159

ABSTRACT

Rapamycin (sirolimus) is employed as an immunosuppressant following liver transplant, to inhibit the re-growth of cancer cells following liver resection for hepatocellular carcinoma (HCC), and for the treatment of advanced HCC. Rapamycin also induces the expression of antioxidant enzymes in the liver, suggesting that pretreatment with the drug could provide a potential strategy to reduce ischemia reperfusion injury following liver surgery. The aim of this study was to further investigate the actions of rapamycin in inducing expression of the antioxidant enzymes heme oxygenase-1 (HO-1) and peroxiredoxin-1 (Prx-1) in normal liver and in tumorigenic liver cells. A rat model of segmental hepatic ischemia and reperfusion, cultured freshly-isolated rat hepatocytes, and tumorigenic H4IIE rat liver cells in culture were employed. Expression of HO-1 and Prx-1 was measured using quantitative PCR and western blot. Rapamycin pre-treatment of normal liver in vivo or normal hepatocytes in vitro led to a substantial induction of mRNA encoding HO-1 and Prx-1. The dose-response curve for the action of rapamycin on mRNA expression was biphasic, showing an increase in expression at 0 - 0.1 µM rapamycin but a decrease from maximum at concentrations greater than 0.1 µM. By contrast, in H4IIE cells, rapamycin inhibited the expression of HO-1 and Prx-1 mRNA. Oltipraz, an established activator of transcription factor Nrf2, caused a large induction of HO-1 and Prx-1 mRNA. The dose response curve for the inhibition by rapamycin of HO-1 and Prx-4 mRNA expression, determined in the presence of oltipraz, was monophasic with half maximal inhibition at about 0.01 µM. It is concluded that, at concentrations comparable to those used clinically, pre-treatment of the liver with rapamycin induces the expression of HO-1 and Prx-1. However, the actions of rapamycin on the expression of these two antioxidant enzymes in normal hepatocytes are complex and, in tumorigenic liver cells, differ from those in normal hepatocytes. Further studies are warranted to evaluate preconditioning the livers of patients subject to liver resection or liver transplant with rapamycin as a viable strategy to reduce IR injury following liver surgery.


Subject(s)
Heme Oxygenase-1/biosynthesis , Hepatocytes/drug effects , Immunosuppressive Agents/pharmacology , Liver Neoplasms/metabolism , Peroxiredoxins/biosynthesis , Sirolimus/pharmacology , Animals , Cell Line, Tumor , Hepatocytes/metabolism , Male , Rats , Rats, Zucker
7.
Med Sci Monit ; 24: 4113-4120, 2018 Jun 16.
Article in English | MEDLINE | ID: mdl-29908016

ABSTRACT

BACKGROUND Osteosarcoma and fibrosarcoma are malignant tumors with poor prognosis. Peroxiredoxin 1 (PRDX1) is considered to prevent tumors in many malignances. However, few studies have focused on the functions of PRDX1 in osteosarcoma and fibrosarcoma. MATERIAL AND METHODS PRDX1 mRNA in tumors and adjacent tissues of 32 osteosarcoma patients and 16 fibrosarcoma patients was extracted and measured. Proliferation and invasion of MG63 and HT1080 cell lines after silencing or overexpressing PRDX1 were used to detect the role of PRDX1 in metastasis of osteosarcoma and fibrosarcoma. RESULTS PRDX1 mRNA level was lower in tumor tissues than in adjacent tissues of osteosarcoma (F=50.105) and fibrosarcoma (F=28.472) patients, both significantly (P<0.05). Silencing PRDX1 promoted proliferation of MG63 and HT1080 cells, while overexpressing PRDX1 suppressed proliferation after 24 h, 48 h, and 72 h, compared to the control group, both significantly (P<0.05). Silencing PRDX1 increased invasive cells of MG63 (F=246.218) and HT1080 (F=245.602), while overexpressing PRDX1 decreased invasive cells of both, compared to the control, and the difference was significant (P<0.05). CONCLUSIONS PRDX1 expression is low in osteosarcoma and fibrosarcoma tumors. PRDX1 suppressed the progression and metastasis of osteosarcoma and fibrosarcoma cells.


Subject(s)
Bone Neoplasms/genetics , Fibrosarcoma/genetics , Osteosarcoma/genetics , Peroxiredoxins/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Disease Progression , Female , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Humans , Male , Neoplasm Invasiveness , Neoplasm Metastasis , Osteosarcoma/metabolism , Osteosarcoma/pathology , Peroxiredoxins/biosynthesis , Peroxiredoxins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Med Sci Monit ; 24: 2259-2270, 2018 Apr 15.
Article in English | MEDLINE | ID: mdl-29656298

ABSTRACT

BACKGROUND Recent studies show that peroxiredoxin 1 (Prdx1) contributes to the progression and poor prognosis of carcinoma through multiple mechanisms. However, there is little information on its expression and prognostic value in gastric cancer. This study investigated the expression of Prdx1 in gastric cancer, along with evaluating its clinical-pathological and prognostic importance. MATERIAL AND METHODS A total of 189 pairs of gastric cancer and paracarcinomatous tissues were assessed for Prdx1 expression and its association with clinical characteristics. The molecular mechanism was further investigated through in vitro experimentation. RESULTS The mRNA and protein levels of Prdx1 in the GC tissues were higher than in the peri-tumor tissues. We also found that high Prdx1 expression was positively correlated with the lymph node invasion and poor prognosis. It also served as an autonomous prognostic factor for patients with gastric cancer. Moreover, Prdx1 regulates the invasion and metastasis of GC cell lines through inhibiting E-Ca expression. CONCLUSIONS Prdx1 can promote epithelial-mesenchymal transition and gastric cancer progression. Therefore, it might be a therapeutic target and prognostic indicator for gastric cancer patients.


Subject(s)
Peroxiredoxins/biosynthesis , Stomach Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Antigens, CD , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor/metabolism , Cell Movement/physiology , Cell Proliferation/physiology , Epithelial-Mesenchymal Transition/physiology , Female , Humans , Immunohistochemistry , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness , Peroxiredoxins/genetics , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
9.
Mol Cell Proteomics ; 15(2): 426-44, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26419955

ABSTRACT

Many biological processes involve the mechanistic/mammalian target of rapamycin complex 1 (mTORC1). Thus, the challenge of deciphering mTORC1-mediated functions during normal and pathological states in the central nervous system is challenging. Because mTORC1 is at the core of translation, we have investigated mTORC1 function in global and regional protein expression. Activation of mTORC1 has been generally regarded to promote translation. Few but recent works have shown that suppression of mTORC1 can also promote local protein synthesis. Moreover, excessive mTORC1 activation during diseased states represses basal and activity-induced protein synthesis. To determine the role of mTORC1 activation in protein expression, we have used an unbiased, large-scale proteomic approach. We provide evidence that a brief repression of mTORC1 activity in vivo by rapamycin has little effect globally, yet leads to a significant remodeling of synaptic proteins, in particular those proteins that reside in the postsynaptic density. We have also found that curtailing the activity of mTORC1 bidirectionally alters the expression of proteins associated with epilepsy, Alzheimer's disease, and autism spectrum disorder-neurological disorders that exhibit elevated mTORC1 activity. Through a protein-protein interaction network analysis, we have identified common proteins shared among these mTORC1-related diseases. One such protein is Parkinson protein 7, which has been implicated in Parkinson's disease, yet not associated with epilepsy, Alzheimers disease, or autism spectrum disorder. To verify our finding, we provide evidence that the protein expression of Parkinson protein 7, including new protein synthesis, is sensitive to mTORC1 inhibition. Using a mouse model of tuberous sclerosis complex, a disease that displays both epilepsy and autism spectrum disorder phenotypes and has overactive mTORC1 signaling, we show that Parkinson protein 7 protein is elevated in the dendrites and colocalizes with the postsynaptic marker postsynaptic density-95. Our work offers a comprehensive view of mTORC1 and its role in regulating regional protein expression in normal and diseased states.


Subject(s)
Autism Spectrum Disorder/genetics , Epilepsy/genetics , Multiprotein Complexes/genetics , Oncogene Proteins/biosynthesis , Parkinson Disease/genetics , Peroxiredoxins/biosynthesis , Protein Biosynthesis/genetics , TOR Serine-Threonine Kinases/genetics , Tuberous Sclerosis/genetics , Animals , Autism Spectrum Disorder/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Dendrites/genetics , Dendrites/pathology , Disease Models, Animal , Epilepsy/pathology , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/antagonists & inhibitors , Oncogene Proteins/genetics , Parkinson Disease/pathology , Peroxiredoxins/genetics , Protein Deglycase DJ-1 , Proteomics/methods , Signal Transduction/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tuberous Sclerosis/pathology
10.
Pediatr Hematol Oncol ; 35(2): 131-144, 2018 Mar.
Article in English | MEDLINE | ID: mdl-30020823

ABSTRACT

The aim of the study was to investigate the expression and methylation status of seven distinctive genes with tumor suppressing properties in childhood and adolescent lymphomas. A total of 96 patients with Hodgkin Lymphoma (HL, n = 41), Non-Hodgkin Lymphoma (NHL, n = 15), and reactive lymphoid hyperplasia (RLH, n = 40, as controls) are included in the research. The expression status of CDKN2A, SPI1, PRDX2, DLEC1, FOXO1, KLF4 and DAPK1 genes were measured with QPCR method after the RNA isolation from paraffin blocks of tumor tissue and cDNA conversion. DNA isolation was performed from samples with low gene expression followed by methylation PCR study specific to promoter regions of these genes. We found that SPI1, PRDX2, DLEC1, KLF4, and DAPK1 genes are significantly less expressed in patient than the control group (p = 0.0001). However, expression of CDKNA2 and FOXO1 genes in the patient and control groups were not statistically different. The methylation ratios of all genes excluding the CDKN2A and FOXO1 were significantly higher in the HL and NHL groups than the controls (p = 0.0001). We showed that SPI1, PRDX2, DLEC1, KLF4 and DAPK1 genes are epigenetically silenced via hypermethylation in the tumor tissues of children with HL and NHL. As CDKN2A gene was not expressed in both patient and control groups, we conclude that it is not specific to malignancy. As FOXO1 gene was similarly expressed in both groups, its relationship with malignancy could not be established. The epigenetically silenced genes may be candidates for biomarkers or therapeutic targets in childhood and adolescent lymphomas.


Subject(s)
Death-Associated Protein Kinases/biosynthesis , Gene Expression Regulation, Neoplastic , Gene Silencing , Kruppel-Like Transcription Factors/biosynthesis , Lymphoma/metabolism , Peroxiredoxins/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Trans-Activators/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Adolescent , Child , Female , Humans , Kruppel-Like Factor 4 , Lymphoma/pathology , Male
11.
J Biol Chem ; 291(31): 16038-47, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27268058

ABSTRACT

The bacterial transcriptional regulator OxyR is known to function as a two-state redox switch. OxyR senses cellular levels of H2O2 via a "sensing cysteine" that switches from the reduced to a disulfide state upon H2O2 exposure, inducing the expression of antioxidant genes. The reduced and disulfide states of OxyR, respectively, bind to extended and compact regions of DNA, where the reduced state blocks and the oxidized state allows transcription and further induces target gene expression by interacting with RNA polymerase. Vibrio vulnificus OxyR2 senses H2O2 with high sensitivity and induces the gene encoding the antioxidant Prx2. In this study, we used mass spectrometry to identify a third redox state of OxyR2, in which the sensing cysteine was overoxidized to S-sulfonated cysteine (Cys-SO3H) by high H2O2 in vitro and in vivo, where the modification deterred the transcription of prx2 The DNA binding preferences of OxyR25CA-C206D, which mimics overoxidized OxyR2, suggested that overoxidized OxyR2 binds to the extended DNA site, masking the -35 region of the prx2 promoter. These combined results demonstrate that OxyR2 functions as a three-state redox switch to tightly regulate the expression of prx2, preventing futile production of Prx2 in cells exposed to high levels of H2O2 sufficient to inactivate Prx2. We further provide evidence that another OxyR homolog, OxyR1, displays similar three-state behavior, inviting further exploration of this phenomenon as a potentially general regulatory mechanism.


Subject(s)
Bacterial Proteins , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic/physiology , Peroxiredoxins , Promoter Regions, Genetic/physiology , Transcription Factors , Vibrio vulnificus , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cysteine/genetics , Cysteine/metabolism , Hydrogen Peroxide/metabolism , Oxidation-Reduction , Peroxiredoxins/biosynthesis , Peroxiredoxins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Vibrio vulnificus/genetics , Vibrio vulnificus/metabolism
12.
Microbiology (Reading) ; 163(3): 343-354, 2017 03.
Article in English | MEDLINE | ID: mdl-28073401

ABSTRACT

Rhodococcus jostii RHA1 is able to degrade toxic compounds and accumulate high amounts of triacylglycerols (TAG) upon nitrogen starvation. These NADPH-dependent processes are essential for the adaptation of rhodococci to fluctuating environmental conditions. In this study, we used an MS-based, label-free and quantitative proteomic approach to better understand the integral response of R. jostii RHA1 to the presence of methyl viologen (MV) in relation to the synthesis and accumulation of TAG. The addition of MV promoted a decrease of TAG accumulation in comparison to cells cultivated under nitrogen-limiting conditions in the absence of this pro-oxidant. Proteomic analyses revealed that the abundance of key proteins of fatty acid biosynthesis, the Kennedy pathway, glyceroneogenesis and methylmalonyl-CoA pathway, among others, decreased in the presence of MV. In contrast, some proteins involved in lipolysis and ß-oxidation of fatty acids were upregulated. Some metabolic pathways linked to the synthesis of NADPH remained activated during oxidative stress as well as under nitrogen starvation conditions. Additionally, exposure to MV resulted in the activation of complete antioxidant machinery comprising superoxide dismutases, catalases, mycothiol biosynthesis, mycothione reductase and alkyl hydroperoxide reductases, among others. Our study suggests that oxidative stress response affects TAG accumulation under nitrogen-limiting conditions through programmed molecular mechanisms when both stresses occur simultaneously.


Subject(s)
Nitrogen/deficiency , Oxidative Stress/physiology , Paraquat/metabolism , Rhodococcus/metabolism , Triglycerides/biosynthesis , Acyl Coenzyme A/metabolism , Adaptation, Physiological , Catalase/metabolism , Cysteine/biosynthesis , Fatty Acids/biosynthesis , Glycopeptides/biosynthesis , Inositol/biosynthesis , NADP/metabolism , Nitrogen/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Oxidoreductases/biosynthesis , Peroxiredoxins/biosynthesis , Proteome , Rhodococcus/growth & development , Superoxide Dismutase/metabolism
13.
Microvasc Res ; 103: 1-10, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26409120

ABSTRACT

H2O2 mediates autocrine and paracrine signaling in the vasculature and can propagate endothelial dysfunction. However, it is not clear how endothelial cells withstand H2O2 exposure and promote H2O2-induced vascular remodeling. To understand the innate ability of endothelial cells for sustaining excess H2O2 exposure, we investigated the genotypic and functional regulation of redox systems in primary HUVECs following an H2O2 treatment. Primary HUVECs were exposed to transient H2O2 exposure and consistent H2O2 exposure. Following H2O2 treatments for 24, 48 and 72 h, we measured O2(-) production, mitochondrial membrane polarization (MMP), and gene expressions of pro-oxidative enzymes, peroxidase enzymes, and cytoprotective intermediates. Our results showed that the 24 h H2O2 exposure significantly increased O2(-) levels, hyperpolarized MMP, and downregulated CAT, GPX1, TXNRD1, NFE2L2, ASK1, and ATF2 gene expression in HUVECs. At 72 h, HUVECs in both treatment conditions were shown to adapt to reduce O2(-) levels and normalize MMP. An upregulation of GPX1, TXNRD1, and HMOX1 gene expression and a recovery of NFE2L2 and PRDX1 gene expression to control levels were observed in both consistent and transient treatments at 48 and 72 h. The response of endothelial cells to excess levels of H2O2 involves a complex interaction amongst O2(-) levels, mitochondrial membrane polarization and anti- and pro-oxidant gene regulation. As a part of this response, HUVECs induce cytoprotective mechanisms including the expression of peroxidase and antioxidant enzymes along with the downregulation of pro-apoptotic genes. This adaptation assists HUVECs to withstand subsequent exposures to H2O2.


Subject(s)
Human Umbilical Vein Endothelial Cells/drug effects , Hydrogen Peroxide/toxicity , Oxidants/toxicity , Oxidative Stress/drug effects , Peroxidases/biosynthesis , Adaptation, Physiological , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Induction , Glutathione Peroxidase/biosynthesis , Heme Oxygenase-1/biosynthesis , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Membrane Potential, Mitochondrial/drug effects , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Peroxidases/genetics , Peroxiredoxins/biosynthesis , Superoxides/metabolism , Thioredoxin Reductase 1/biosynthesis , Time Factors , Glutathione Peroxidase GPX1
14.
Exp Parasitol ; 163: 8-15, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26821294

ABSTRACT

Cryptosporidium parvum is one of the most radioresistant organisms identified to date. In a previous study, we found that thioredoxin peroxidase (CpTPx) was significantly upregulated in this species following exposure to high dose (10 kGy) of γ-irradiation. To assess the potential of CpTPx to confer radioprotection in mammalian cells, it was expressed in COS-7 African green monkey kidney cells (CpTPx-COS7). For comparison, the thioredoxin peroxidase of Cryptosporidium muris (CmTPx) was also expressed in these cells (CmTPx-COS7 cells), which has been confirmed to have lesser antioxidant activity than CpTPx in the previous study. Notably, the survival rates of CpTPx-COS7 cells were significantly higher (12-22%) at 72 h after 8 Gy irradiation than CmTPx-COS7 or non-transfected COS-7 (ntCOS-7) counterparts. In addition, CpTPx revealed a 50% of ROS reduction in irradiated CpTPx-COS7 cells, while γ-H2AX DNA damage marker expression was not significantly changed. Furthermore, the amount of apoptosis only increased to about 120% after 2-8 Gy irradiation compared to 200-300% increase observed in ntCOS-7 cells. CmTPx was shown to have antioxidant and DNA damage protection activities; however, these activities were always lower than those of CpTPx. These results suggest that the potent antioxidant and protective activities of CpTPx are well conserved in this cell-based system and that CpTPx contributed to the radioprotection of mammalian cells through its exceptional antioxidant activity.


Subject(s)
Antioxidants/metabolism , COS Cells/enzymology , Cryptosporidium parvum/enzymology , Gamma Rays , Peroxiredoxins/biosynthesis , Animals , COS Cells/parasitology , COS Cells/radiation effects , Chlorocebus aethiops , Cryptosporidium parvum/radiation effects , Gene Expression Regulation, Enzymologic , Microscopy, Confocal , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Reactive Oxygen Species/metabolism , Transfection
15.
Mol Biol (Mosk) ; 50(2): 336-46, 2016.
Article in Russian | MEDLINE | ID: mdl-27239855

ABSTRACT

Reactive oxygen species (ROS) are produced via catabolic and anabolic processes during normal embryonic development, and ROS content in the cell is maintained at a certain level. Peroxiredoxins are a family of selenium-independent peroxidases and play a key role in maintaining redox homeostasis of the cell. In addition to regulating the ROS level, peroxiredoxins are involved in intracellular and intercellular signaling, cell differentiation, and tissue development. The time course of peroxiredoxin gene (prx1-6) expression was studied in Xenopus laevis during early ontogeny (Nieuwkoop and Faber stages 10-63). The highest expression level was observed for prx1 at these developmental stages. The prx1, prx3, and prx4 expression level changed most dramatically in response to oxidative stress artificially induced in X. laevis embryos. In X. laevis adults, prx1-6 were all intensely expressed in all organs examined, the prx1 expression level being the highest. The X. laevis prx1-6 genes were cloned and expressed in Escherichia coli, and physico-chemical characteristics were compared for the recombinant enzymes. The highest peroxidase activity and thermal stability were observed for Prx1 and Prx2. It was assumed that Prx1 plays a leading role in X. laevis early development.


Subject(s)
Homeodomain Proteins/genetics , Oxidative Stress/genetics , Peroxidases/genetics , Peroxiredoxins/genetics , Xenopus Proteins/genetics , Xenopus laevis/growth & development , Animals , Cytoplasm/genetics , Embryo, Nonmammalian , Embryonic Development/genetics , Enzyme Stability , Gene Expression Regulation, Developmental , Peroxidases/biosynthesis , Peroxidases/chemistry , Peroxiredoxins/biosynthesis , Peroxiredoxins/chemistry , Reactive Oxygen Species/metabolism , Xenopus laevis/genetics
16.
J Biol Chem ; 289(39): 26904-26913, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25122762

ABSTRACT

Oxidative folding of (pro)insulin is crucial for its assembly and biological function. This process takes place in the endoplasmic reticulum (ER) and is accomplished by protein disulfide isomerase and ER oxidoreductin 1ß, generating stoichiometric amounts of hydrogen peroxide (H2O2) as byproduct. During insulin resistance in the prediabetic state, increased insulin biosynthesis can overwhelm the ER antioxidative and folding capacity, causing an imbalance in the ER redox homeostasis and oxidative stress. Peroxiredoxin 4 (Prdx4), an ER-specific antioxidative peroxidase can utilize luminal H2O2 as driving force for reoxidizing protein disulfide isomerase family members, thus efficiently contributing to disulfide bond formation. Here, we examined the functional significance of Prdx4 on ß-cell function with emphasis on insulin content and secretion during stimulation with nutrient secretagogues. Overexpression of Prdx4 in glucose-responsive insulin-secreting INS-1E cells significantly metabolized luminal H2O2 and improved the glucose-induced insulin secretion, which was accompanied by the enhanced proinsulin mRNA transcription and insulin content. This ß-cell beneficial effect was also observed upon stimulation with the nutrient insulin secretagogue combination of leucine plus glutamine, indicating that the effect is not restricted to glucose. However, knockdown of Prdx4 had no impact on H2O2 metabolism or ß-cell function due to the fact that Prdx4 expression is negligibly low in pancreatic ß-cells. Moreover, we provide evidence that the constitutively low expression of Prdx4 is highly susceptible to hyperoxidation in the presence of high glucose. Overall, these data suggest an important role of Prdx4 in maintaining insulin levels and improving the ER folding capacity also under conditions of a high insulin requirement.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Glucose/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Peroxiredoxins/biosynthesis , Sweetening Agents/pharmacology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Gene Expression Regulation, Enzymologic/physiology , Gene Knockdown Techniques , Glucose/metabolism , Hep G2 Cells , Humans , Hydrogen Peroxide/metabolism , Insulin/genetics , Insulin Secretion , Insulin-Secreting Cells/cytology , Oxidation-Reduction/drug effects , Peroxiredoxins/genetics , Protein Folding/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sweetening Agents/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
17.
Immunology ; 146(1): 173-83, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26094816

ABSTRACT

Natural killer (NK) cells are considered critical components of the innate and adaptive immune responses. Deficiencies in NK cell activity are common, such as those that occur in cancer patients, and they can be responsible for dysfunctional immune surveillance. Persistent oxidative stress is intrinsic to many malignant tumours, and numerous studies have focused on the effects of reactive oxygen species on the anti-tumour activity of NK cells. Indeed, investigations in animal models have suggested that one of the most important thiol-dependent antioxidant enzymes, peroxiredoxin 1 (PRDX1), is essential for NK cell function. In this work, our analysis of the transcriptomic expression pattern of antioxidant enzymes in human NK cells has identified PRDX1 as the most prominently induced transcript out of the 18 transcripts evaluated in activated NK cells. The change in PRDX1 expression was followed by increased expression of two other enzymes from the PRDX-related antioxidant chain: thioredoxin and thioredoxin reductase. To study the role of thiol-dependent antioxidants in more detail, we applied a novel compound, adenanthin, to induce an abrupt dysfunction of the PRDX-related antioxidant chain in NK cells. In human primary NK cells, we observed profound alterations in spontaneous and antibody-dependent NK cell cytotoxicity against cancer cells, impaired degranulation, and a decreased expression of activation markers under these conditions. Collectively, our study pinpoints the unique role for the antioxidant activity of the PRDX-related enzymatic chain in human NK cell functions. Further understanding this phenomenon will prospectively lead to fine-tuning of the novel NK-targeted therapeutic approaches to human disease.


Subject(s)
Diterpenes, Kaurane/pharmacology , Enzyme Inhibitors/pharmacology , Killer Cells, Natural/immunology , Neoplasms/immunology , Peroxiredoxins/antagonists & inhibitors , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Antioxidants , Cell Degranulation/drug effects , Cell Degranulation/immunology , Cell Line, Tumor , Glutathione/analysis , Humans , Oxidative Stress/drug effects , Peroxiredoxins/biosynthesis , Reactive Oxygen Species/metabolism , Thioredoxin Reductase 1/biosynthesis , Thioredoxins/biosynthesis
18.
Tumour Biol ; 36(12): 9829-37, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26159854

ABSTRACT

Despite the recent advances in diagnostic and therapeutic strategies, oral squamous cell carcinoma (OSCC) remains a major health burden. Protein biomarker discovery for early detection will help to improve patient survival rate in OSCC. Mass spectrometry-based proteomics has emerged as an excellent approach for detection of protein biomarkers in various types of cancers. In the current study, we have used 4-Plex isobaric tags for relative and absolute quantitation (iTRAQ)-based shotgun quantitative proteomic approach to identify proteins that are differentially expressed in cancerous tissues compared to normal tissues. The high-resolution mass spectrometric analysis resulted in identifying 2,074 proteins, among which 288 proteins were differentially expressed. Further, it was noticed that 162 proteins were upregulated, while 125 proteins were downregulated in OSCC-derived cancer tissue samples as compared to the adjacent normal tissues. We identified some of the known molecules which were reported earlier in OSCC such as MMP-9 (8.4-fold), ZNF142 (5.6-fold), and S100A7 (3.5-fold). Apart from this, we have also identified some novel signature proteins which have not been reported earlier in OSCC including ras-related protein Rab-2A isoform, RAB2A (4.6-fold), and peroxiredoxin-1, PRDX1 (2.2-fold). The immunohistochemistry-based validation using tissue microarray slides in OSCC revealed overexpression of the RAB2A and PRDX1 gene in 80 and 68 % of the tested clinical cases, respectively. This study will not only serve as a resource of candidate biomarkers but will contribute towards the existing knowledge on the role of the candidate molecules towards disease progression and therapeutic potential.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/genetics , Mouth Neoplasms/genetics , Peroxiredoxins/biosynthesis , rab GTP-Binding Proteins/biosynthesis , Aged , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Mouth Neoplasms/pathology , Peroxiredoxins/genetics , Proteome/genetics , Proteomics , Tandem Mass Spectrometry , rab GTP-Binding Proteins/genetics
19.
Exp Eye Res ; 139: 22-36, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26215528

ABSTRACT

DJ-1/PARK7 mutations or deletions cause autosomal recessive early onset Parkinson's disease (PD). Thus, DJ-1 protein has been extensively studied in brain and neurons. PD patients display visual symptoms; however, the visual symptoms specifically attributed to PD patients carrying DJ-1/PARK7 mutations are not known. In this study, we analyzed the structure and physiology of retinas of 3- and 6-month-old DJ-1 knockout (KO) mice to determine how loss of function of DJ-1 specifically contributes to the phenotypes observed in PD patients. As compared to controls, the DJ-1 KO mice displayed an increase in the amplitude of the scotopic ERG b-wave and cone ERG, while the amplitude of a subset of the dc-ERG components was decreased. The main structural changes in the DJ-1 KO retinas were found in the outer plexiform layer (OPL), photoreceptors and retinal pigment epithelium (RPE), which were observed at 3 months and progressively increased at 6 months. RPE thinning and structural changes within the OPL were observed in the retinas in DJ-1 KO mice. DJ-1 KO retinas also exhibited disorganized outer segments, central decrease in red/green cone opsin staining, decreased labeling of ezrin, broader distribution of ribeye labeling, decreased tyrosine hydroxylase in dopaminergic neurons, and increased 7,8-dihydro-8-oxoguanine-labeled DNA oxidation. Accelerated outer retinal atrophy was observed in DJ-1 KO mice after selective oxidative damage induced by a single tail vein injection of NaIO3, exposing increased susceptibility to oxidative stress. Our data indicate that DJ-1-deficient retinas exhibit signs of morphological abnormalities and physiological dysfunction in association with increased oxidative stress. Degeneration of RPE cells in association with oxidative stress is a key hallmark of age-related macular degeneration (AMD). Therefore, in addition to detailing the visual defects that occur as a result of the absence of DJ-1, our data is also relevant to AMD pathogenesis.


Subject(s)
DNA/genetics , Mutation , Oncogene Proteins/genetics , Peroxiredoxins/genetics , Retinal Degeneration/genetics , Retinal Pigment Epithelium/metabolism , Animals , Blotting, Western , DNA Mutational Analysis , Disease Models, Animal , Electroretinography , Female , Genotype , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Oncogene Proteins/biosynthesis , Oxidative Stress , Peroxiredoxins/biosynthesis , Polymerase Chain Reaction , Protein Deglycase DJ-1 , Retinal Degeneration/pathology , Retinal Degeneration/physiopathology , Retinal Pigment Epithelium/physiopathology , Retinal Pigment Epithelium/ultrastructure , Signal Transduction
20.
Cell Mol Neurobiol ; 35(8): 1217-26, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26003307

ABSTRACT

Reactive astrogliosis and microgliosis after spinal cord injury (SCI) contribute to glial scar formation that impedes axonal regeneration. The mechanisms underlying reactive astrocyte and microglia proliferation upon injury remain partially understood. Peroxiredoxin 1 (PRDX1) is an antioxidant participating in cell proliferation, differentiation, and apoptosis. However, PRDX1 functions in SCI-induced astrocyte and microglia proliferation are unknown. In this study, we established an acute spinal cord contusion injury model in adult rats to investigate the potential role of PRDX1 during the pathological process of SCI. We found the palpable expression increase of PRDX1 after SCI by western blot and immunohistochemistry staining. Double immunofluorescence staining showed that PRDX1 expression mainly increased in astrocytes and microglia. In addition, PRDX1/proliferating cell nuclear antigen (PCNA) colocalized in astrocytes and microglia. Furthermore, PCNA expression also elevated after SCI, as well as was positively correlated with PRDX1 expression. In vitro, PRDX1 expression in primary rat spinal cord astrocytes and microglia changed in a concentration- and time-dependent manner according to LPS treatment. In addition, PRDX1 knockdown in astrocytes and microglia resulted in the decrease of PCNA expression after LPS stimulation, showing that PRDX1 promoted astrocyte and microglia proliferation after inflammation. Our results suggested that PRDX1 might play a crucial role in astrocyte and microglia proliferation after SCI.


Subject(s)
Peroxiredoxins/biosynthesis , Spinal Cord Injuries/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cell Proliferation/physiology , Gene Expression Regulation , Male , Microglia/metabolism , Microglia/pathology , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology
SELECTION OF CITATIONS
SEARCH DETAIL