Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Redox Biol ; 47: 102153, 2021 11.
Article in English | MEDLINE | ID: mdl-34610554

ABSTRACT

Protein cysteine residues are essential for protein folding, participate in enzymatic catalysis, and coordinate the binding of metal ions to proteins. Enzymatically catalyzed and redox-dependent post-translational modifications of cysteine residues are also critical for signal transduction and regulation of protein function and localization. S-nitrosylation, the addition of a nitric oxide equivalent to a cysteine residue, is a redox-dependent modification. In this study, we curated and analyzed four different studies that employed various chemoselective platforms coupled to mass spectrometry to precisely identify S-nitrosocysteine residues in mouse heart proteins. Collectively 1974 S-nitrosocysteine residues in 761 proteins were identified and 33.4% were identified in two or more studies. A core of 75 S-nitrosocysteine residues in 44 proteins were identified in all four studies. Bioinformatic analysis of each study indicated a significant enrichment of mitochondrial proteins participating in metabolism. Regulatory proteins in glycolysis, TCA cycle, oxidative phosphorylation and ATP production, long chain fatty acid ß-oxidation, and ketone and amino acid metabolism constitute the major functional pathways impacted by protein S-nitrosylation. In the cardiovascular system, nitric oxide signaling regulates vasodilation and cardiac muscle contractility. The meta-analysis of the proteomic data supports the hypothesis that nitric oxide signaling via protein S-nitrosylation is also a regulator of cardiomyocyte metabolism that coordinates fuel utilization to maximize ATP production. As such, protein cysteine S-nitrosylation represents a third functional dimension of nitric oxide signaling in the cardiovascular system to ensure optimal cardiac function.


Subject(s)
Proteomics , S-Nitrosothiols , Animals , Cysteine/analogs & derivatives , Cysteine/metabolism , Metabolic Networks and Pathways , Mice , Nitric Oxide/metabolism , Protein Processing, Post-Translational
2.
Biomed J ; 44(6): 663-670, 2021 12.
Article in English | MEDLINE | ID: mdl-34478892

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a multifactorial life-threatening lung injury, characterized by diffuse lung inflammation and increased alveolocapillary barrier permeability. The different stages of ARDS have distinctive biochemical and clinical profiles. Despite the progress of our understanding on ARDS pathobiology, the mechanisms underlying its pathogenesis are still obscure. Herein, we review the existing literature about the implications of phospholipases 2 (PLA2s), a large family of enzymes that catalyze the hydrolysis of fatty acids at the sn-2 position of glycerophospholipids, in ARDS-related pathology. We emphasize on the versatile way of participation of different PLA2s isoforms in the distinct ARDS subgroup phenotypes by either potentiating lung inflammation and damage or by preserving the normal lung. Current research supports that PLA2s are associated with the progression and the outcome of ARDS. We herein discuss the transcellular communication of PLA2s through secreted extracellular vesicles and suggest it as a new mechanism of PLA2s involvement in ARDS. Thus, the elucidation of the spatiotemporal features of PLA2s expression may give new insights and provide valuable information about the risk of an individual to develop ARDS or advance to more severe stages, and potentially identify PLA2 isoforms as biomarkers and target for pharmacological intervention.


Subject(s)
Extracellular Vesicles , Respiratory Distress Syndrome , Biomarkers , Extracellular Vesicles/metabolism , Humans , Inflammation , Phospholipases A2/metabolism , Respiratory Distress Syndrome/diagnosis
3.
Nitric Oxide ; 117: 1-6, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34536587

ABSTRACT

Cysteine S-nitrosation mediates NO signaling and protein function under pathophysiological conditions. Herein, we provide a detailed protocol regarding the organic mercury chemoselective enrichment of S-nitrosated proteins and peptides. We discuss key aspects of the enrichment strategy and provide technical tips for the best performance of the experimental protocol.


Subject(s)
Mercury/chemistry , Nitrates , Proteins , Proteomics/methods , Chromatography , Cysteine/analysis , Cysteine/isolation & purification , Cysteine/metabolism , Nitrates/analysis , Nitrates/isolation & purification , Nitrates/metabolism , Nitric Oxide/metabolism , Nitrosation , Peptides/analysis , Peptides/isolation & purification , Peptides/metabolism , Proteins/analysis , Proteins/isolation & purification , Proteins/metabolism
4.
F1000Res ; 92020.
Article in English | MEDLINE | ID: mdl-33042519

ABSTRACT

Nitric oxide is an endogenously formed gas that acts as a signaling molecule in the human body. The signaling functions of nitric oxide are accomplished through two primer mechanisms: cGMP-mediated phosphorylation and the formation of S-nitrosocysteine on proteins. This review presents and discusses previous and more recent findings documenting that nitric oxide signaling regulates metabolic activity. These discussions primarily focus on endothelial nitric oxide synthase (eNOS) as the source of nitric oxide.


Subject(s)
Nitric Oxide Synthase Type III/physiology , Nitric Oxide/physiology , Signal Transduction , Humans
5.
JCI Insight ; 3(16)2018 08 23.
Article in English | MEDLINE | ID: mdl-30135317

ABSTRACT

Alterations in the synthesis and bioavailability of NO are central to the pathogenesis of cardiovascular and metabolic disorders. Although endothelial NO synthase-derived (eNOS-derived) NO affects mitochondrial long-chain fatty acid ß-oxidation, the pathophysiological significance of this regulation remains unclear. Accordingly, we determined the contributions of eNOS/NO signaling in the adaptive metabolic responses to fasting and in age-induced metabolic dysfunction. Four-month-old eNOS-/- mice are glucose intolerant and exhibit serum dyslipidemia and decreased capacity to oxidize fatty acids. However, during fasting, eNOS-/- mice redirect acetyl-CoA to ketogenesis to elevate circulating levels of ß-hydroxybutyrate similar to wild-type mice. Treatment of 4-month-old eNOS-/- mice with nitrite for 10 days corrected the hypertension and serum hyperlipidemia and normalized the rate of fatty acid oxidation. Fourteen-month-old eNOS-/- mice exhibited metabolic derangements, resulting in reduced utilization of fat to generate energy, lower resting metabolic activity, and diminished physical activity. Seven-month administration of nitrite to eNOS-/- mice reversed the age-dependent metabolic derangements and restored physical activity. While the eNOS/NO signaling is not essential for the metabolic adaptation to fasting, it is critical for regulating systemic metabolic homeostasis in aging. The development of age-dependent metabolic disorder is prevented by low-dose replenishment of bioactive NO.


Subject(s)
Aging/metabolism , Homeostasis/drug effects , Nitric Oxide Synthase Type III/deficiency , Sodium Nitrite/administration & dosage , Administration, Oral , Aging/drug effects , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Fasting/metabolism , Humans , Hyperlipidemias/drug therapy , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Hypertension/drug therapy , Hypertension/genetics , Hypertension/metabolism , Male , Mice , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Signal Transduction/drug effects , Time Factors , Treatment Outcome
6.
J Biol Chem ; 290(16): 10486-94, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25737446

ABSTRACT

Very long acyl-CoA dehydrogenase (VLCAD) deficiency is a genetic pediatric disorder presenting with a spectrum of phenotypes that remains for the most part untreatable. Here, we present a novel strategy for the correction of VLCAD deficiency by increasing mutant VLCAD enzymatic activity. Treatment of VLCAD-deficient fibroblasts, which express distinct mutant VLCAD protein and exhibit deficient fatty acid ß-oxidation, with S-nitroso-N-acetylcysteine induced site-specific S-nitrosylation of VLCAD mutants at cysteine residue 237. Cysteine 237 S-nitrosylation was associated with an 8-17-fold increase in VLCAD-specific activity and concomitant correction of acylcarnitine profile and ß-oxidation capacity, two hallmarks of the disorder. Overall, this study provides biochemical evidence for a potential therapeutic modality to correct ß-oxidation deficiencies.


Subject(s)
Acetylcysteine/analogs & derivatives , Acyl-CoA Dehydrogenase/metabolism , Carnitine/analogs & derivatives , Fibroblasts/drug effects , Acetylcysteine/pharmacology , Acyl-CoA Dehydrogenase/chemistry , Acyl-CoA Dehydrogenase/genetics , Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Amino Acid Sequence , Carnitine/metabolism , Congenital Bone Marrow Failure Syndromes , Cysteine/metabolism , Dose-Response Relationship, Drug , Fatty Acids/metabolism , Fibroblasts/enzymology , Fibroblasts/pathology , Genetic Therapy/methods , Humans , Kinetics , Lipid Metabolism, Inborn Errors/drug therapy , Lipid Metabolism, Inborn Errors/enzymology , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Molecular Sequence Data , Muscular Diseases/drug therapy , Muscular Diseases/enzymology , Muscular Diseases/genetics , Muscular Diseases/pathology , Mutation , Oxidation-Reduction , Primary Cell Culture , Skin/drug effects , Skin/enzymology , Skin/pathology
7.
J Biol Chem ; 288(37): 26473-9, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23861393

ABSTRACT

NO is a versatile free radical that mediates numerous biological functions within every major organ system. A molecular pathway by which NO accomplishes functional diversity is the selective modification of protein cysteine residues to form S-nitrosocysteine. This post-translational modification, S-nitrosylation, impacts protein function, stability, and location. Despite considerable advances with individual proteins, the in vivo biological chemistry, the structural elements that govern the selective S-nitrosylation of cysteine residues, and the potential overlap with other redox modifications are unknown. In this minireview, we explore the functional features of S-nitrosylation at the proteome level and the structural diversity of endogenously modified residues, and we discuss the potential overlap and complementation that may exist with other cysteine modifications.


Subject(s)
Cysteine/analogs & derivatives , Gene Expression Regulation , Proteins/chemistry , S-Nitrosothiols/chemistry , Animals , Cysteine/chemistry , Humans , Mice , Myocardium/metabolism , Nitric Oxide/chemistry , Nitric Oxide Synthase/metabolism , Nitrogen/chemistry , Oxidation-Reduction , Protein Processing, Post-Translational , Proteomics/methods , Signal Transduction
8.
J Proteomics ; 92: 195-203, 2013 Oct 30.
Article in English | MEDLINE | ID: mdl-23748021

ABSTRACT

Cysteine S-nitrosylation is a post-translational modification regulating protein function and nitric oxide signaling. Herein the selectivity, reproducibility, and sensitivity of a mass spectrometry-based proteomic method for the identification of endogenous S-nitrosylated proteins are outlined. The method enriches for either S-nitrosylated proteins or peptides through covalent binding of the cysteine sulfur with phenylmercury at pH=6.0. Phenylmercury reacts selectively and efficiently with S-nitrosocysteine since no reactivity can be documented for disulfides, sulfinic or sulfonic acids, S-glutathionylated, S-alkylated or S-sulfhydrylated cysteine residues. A specificity of 97±1% for the identification of S-nitrosocysteine peptides in mouse liver tissue is achieved by the inclusion of negative controls. The method enables the detection of 36 S-nitrosocysteine peptides starting with 5pmolS-nitrosocysteine/mg of total tissue protein. Both the percentage of protein molecules modified as well as the occupancy by S-nitrosylation can be determined. Overall, selective, sensitive and reproducible enrichment of S-nitrosylated proteins and peptides is achieved by the use of phenylmercury. The inclusion of appropriate negative controls secures the precise identification of endogenous S-nitrosylated sites and proteins in biological samples. BIOLOGICAL SIGNIFICANCE: The current study describes a selective, sensitive and reproducible method for the acquisition of endogenously S-nitrosylated proteins and peptides. The acquisition of endogenous S-nitrosoproteomes provides robust data that is necessary for investigating the mechanism(s) of S-nitrosylation in vivo, the factors that govern its selectivity, the dependency of the modification on different isoforms of nitric oxide synthases (NOS), as well as the physiological functions of this protein modification. This article is part of a Special Issue entitled: Posttranslational Protein modifications in biology and Medicine.


Subject(s)
Cysteine/analogs & derivatives , Mass Spectrometry/methods , Nitric Oxide Synthase/metabolism , Protein Processing, Post-Translational , Proteome/metabolism , S-Nitrosothiols/metabolism , Animals , Cattle , Cysteine/chemistry , Cysteine/metabolism , Mice , Nitric Oxide Synthase/chemistry , Phenylmercury Compounds/chemistry , Proteome/chemistry , Rabbits , S-Nitrosothiols/chemistry , Sensitivity and Specificity
9.
Sci Signal ; 6(256): rs1, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23281369

ABSTRACT

Cysteine S-nitrosylation is a posttranslational modification by which nitric oxide regulates protein function and signaling. Studies of individual proteins have elucidated specific functional roles for S-nitrosylation, but knowledge of the extent of endogenous S-nitrosylation, the sites that are nitrosylated, and the regulatory consequences of S-nitrosylation remains limited. We used mass spectrometry-based methodologies to identify 1011 S-nitrosocysteine residues in 647 proteins in various mouse tissues. We uncovered selective S-nitrosylation of enzymes participating in glycolysis, gluconeogenesis, tricarboxylic acid cycle, and oxidative phosphorylation, indicating that this posttranslational modification may regulate metabolism and mitochondrial bioenergetics. S-nitrosylation of the liver enzyme VLCAD [very long chain acyl-coenzyme A (CoA) dehydrogenase] at Cys(238), which was absent in mice lacking endothelial nitric oxide synthase, improved its catalytic efficiency. These data implicate protein S-nitrosylation in the regulation of ß-oxidation of fatty acids in mitochondria.


Subject(s)
Cysteine/analogs & derivatives , Energy Metabolism/physiology , Fatty Acids/metabolism , Mitochondria/metabolism , Nitric Oxide/metabolism , Protein Processing, Post-Translational/physiology , S-Nitrosothiols/metabolism , Signal Transduction/physiology , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Analysis of Variance , Animals , Cysteine/metabolism , DNA Primers/genetics , Liver/anatomy & histology , Liver/metabolism , Mass Spectrometry , Mice , Mutagenesis, Site-Directed , Oxidation-Reduction , Protein Processing, Post-Translational/genetics , Proteomics , Signal Transduction/genetics
10.
Methods ; 62(2): 165-70, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23116708

ABSTRACT

Protein S-nitrosylation is considered as one of the molecular mechanisms by which nitric oxide regulates signaling events and protein function. The present review presents an updated method which allows for the site-specific detection of S-nitrosylated proteins in vivo. The method is based on enrichment of S-nitrosylated proteins or peptides using organomercury compounds followed by LC-MS/MS detection. Technical aspects for determining the reaction and binding efficiency of the mercury resin that assists enrichment of S-nitrosylated proteins are presented and discussed. In addition, emphasis is given to the specificity of the method by providing technical details for the generation of four chemically distinct negative controls. Finally it is provided an overview of the key steps for generation and evaluation of mass spectrometry derived data.


Subject(s)
Cysteine/analogs & derivatives , Proteome/isolation & purification , S-Nitrosothiols/isolation & purification , Animals , Chromatography, Affinity , Cysteine/chemistry , Cysteine/isolation & purification , Cysteine/metabolism , Humans , Muramidase/chemistry , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Protein Binding , Protein Processing, Post-Translational , Proteome/chemistry , Proteome/metabolism , S-Nitrosothiols/chemistry , S-Nitrosothiols/metabolism , Tandem Mass Spectrometry
11.
Circulation ; 126(20): 2392-401, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23081989

ABSTRACT

BACKGROUND: Several lines of evidence support a pathophysiological role of immunity in atherosclerosis. Tyrosine-nitrated proteins, a footprint of oxygen- and nitrogen-derived oxidants generated by cells of the immune system, are enriched in atheromatous lesions and in circulation of patients with coronary artery disease (CAD). However, the consequences of possible immune reactions triggered by the presence of nitrated proteins in subjects with clinically documented atherosclerosis have not been explored. METHODS AND RESULTS: Specific immunoglobulins that recognize 3-nitrotyrosine epitopes were identified in human lesions, as well as in circulation of patients with CAD. The levels of circulating immunoglobulins against 3-nitrotyrosine epitopes were quantified in patients with CAD (n=374) and subjects without CAD (non-CAD controls, n=313). A 10-fold increase in the mean level of circulating immunoglobulins against protein-bound 3-nitrotyrosine was documented in patients with CAD (3.75±1.8 µg antibody Eq/mL plasma versus 0.36±0.8 µg antibody Eq/mL plasma), and was strongly associated with angiographic evidence of significant CAD. CONCLUSIONS: The results of this cross-sectional study suggest that posttranslational modification of proteins via nitration within atherosclerotic plaque-laden arteries and in circulation serve as neo-epitopes for the elaboration of immunoglobulins, thereby providing an association between oxidant production and the activation of the immune system in CAD.


Subject(s)
Coronary Artery Disease/immunology , Epitopes/immunology , Immunoglobulins/blood , Tyrosine/analogs & derivatives , Aged , Case-Control Studies , Coronary Angiography , Coronary Artery Disease/blood , Coronary Artery Disease/diagnostic imaging , Cross-Sectional Studies , Female , Humans , Immune System/physiopathology , Male , Middle Aged , Tyrosine/immunology
12.
Biochim Biophys Acta ; 1820(6): 684-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21651963

ABSTRACT

BACKGROUND: A biochemical pathway by which nitric oxide accomplishes functional diversity is the specific modification of protein cysteine residues to form S-nitrosocysteine. This post-translational modification, S-nitrosylation, impacts protein function, interactions and location. However, comprehensive studies exploring protein signaling pathways or interrelated protein clusters that are regulated by S-nitrosylation have not been performed on a global scale. SCOPE OF REVIEW: To provide insights to these important biological questions, sensitive, validated and quantitative proteomic approaches are required. This review summarizes current approaches for the global identification of S-nitrosylated proteins. MAJOR CONCLUSIONS: The application of novel methods for identifying S-nitrosylated proteins, especially when combined with mass-spectrometry based proteomics to provide site-specific identification of the modified cysteine residues, promises to deliver critical clues for the regulatory role of this dynamic posttranslational modification in cellular processes. GENERAL SIGNIFICANCE: Though several studies have established S-nitrosylation as a regulator of protein function in individual proteins, the biological chemistry and the structural elements that govern the specificity of this modification in vivo are vastly unknown. Additionally, a gap in knowledge exists concerning the potential global regulatory role(s) this modification may play in cellular physiology. By further studying S-nitrosylation at a global scale, a greater appreciation of nitric oxide and protein S-nitrosylation in cellular function can be achieved. This article is part of a Special Issue entitled Regulation of Cellular Processes by S-nitrosylation.


Subject(s)
Cysteine/analogs & derivatives , Cysteine/metabolism , Proteins/metabolism , Cysteine/biosynthesis , Mass Spectrometry , Nitric Oxide/metabolism , Nitrosation/physiology , Protein Processing, Post-Translational , Proteins/chemistry , Proteomics , S-Nitrosothiols , Signal Transduction
13.
Proc Natl Acad Sci U S A ; 107(39): 16958-63, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20837516

ABSTRACT

S-nitrosylation, the selective posttranslational modification of protein cysteine residues to form S-nitrosocysteine, is one of the molecular mechanisms by which nitric oxide influences diverse biological functions. In this study, unique MS-based proteomic approaches precisely pinpointed the site of S-nitrosylation in 328 peptides in 192 proteins endogenously modified in WT mouse liver. Structural analyses revealed that S-nitrosylated cysteine residues were equally distributed in hydrophobic and hydrophilic areas of proteins with an average predicted pK(a) of 10.01 ± 2.1. S-nitrosylation sites were over-represented in α-helices and under-represented in coils as compared with unmodified cysteine residues in the same proteins (χ(2) test, P < 0.02). A quantile-quantile probability plot indicated that the distribution of S-nitrosocysteine residues was skewed toward larger surface accessible areas compared with the unmodified cysteine residues in the same proteins. Seventy percent of the S-nitrosylated cysteine residues were surrounded by negatively or positively charged amino acids within a 6-Å distance. The location of cysteine residues in α-helices and coils in highly accessible surfaces bordered by charged amino acids implies site directed S-nitrosylation mediated by protein-protein or small molecule interactions. Moreover, 13 modified cysteine residues were coordinated with metals and 15 metalloproteins were endogenously modified supporting metal-catalyzed S-nitrosylation mechanisms. Collectively, the endogenous S-nitrosoproteome in the liver has structural features that accommodate multiple mechanisms for selective site-directed S-nitrosylation.


Subject(s)
Cysteine/analogs & derivatives , Liver/metabolism , Protein Processing, Post-Translational , Proteins/metabolism , S-Nitrosothiols/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cysteine/analysis , Cysteine/metabolism , Hydrophobic and Hydrophilic Interactions , Mice , Molecular Sequence Data , Proteins/chemistry , Proteome , Proteomics , S-Nitrosothiols/analysis
14.
Free Radic Biol Med ; 42(4): 567-77, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17275689

ABSTRACT

Heat shock protein-70 (Hsp70) is the main heat-inducible member of the 70-kDa family of chaperones that assist cells in maintaining proteins functional under stressful conditions. In the present investigation, the role of Hsp70 in the molecular mechanism of hydrogen peroxide-induced DNA damage to HeLa cells in culture was examined. Stably transfected HeLa cell lines, overexpressing or lacking Hsp70, were created by utilizing constitutive expression of plasmids containing the functional hsp70 gene or hsp70-siRNA, respectively. Compared to control cells, the Hsp70-overexpressing ones were significantly resistant to hydrogen peroxide-induced DNA damage, while Hsp70-depleted cells showed an enhanced sensitivity. In addition, the "intracellular calcein-chelatable iron pool" was determined in the presence or absence of Hsp70 and found to be related to the sensitivity of nuclear DNA to H(2)O(2). It seems likely that the main action of Hsp70, at least in this system, is exerted at the lysosomal level, by protecting the membranes of these organelles against oxidative stress-induced destabilization. Apart from shedding additional light on the mechanistic details behind the action of Hsp70 during oxidative stress, our results indicate that modulation of cellular Hsp70 may represent a way to make cancer cells more sensitive to normal host defense mechanisms or chemotherapeutic drug treatment.


Subject(s)
DNA Damage , HSP70 Heat-Shock Proteins/physiology , Hydrogen Peroxide/pharmacology , Base Sequence , Blotting, Western , DNA Primers , Fluorescent Antibody Technique , HeLa Cells , Humans , Reverse Transcriptase Polymerase Chain Reaction
15.
Biochem J ; 403(2): 261-6, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17233627

ABSTRACT

The calcein-AM (calcein-acetoxymethyl ester) method is a widely used technique that is supposed to assay the intracellular 'labile iron pool' (LIP). When cells in culture are exposed to this ester, it passes the plasma membrane and reacts with cytosolic unspecific esterases. One of the reaction products, calcein, is a fluorochrome and a hydrophilic alcohol to which membranes are non-permeable and which, consequently, is retained within the cytosol of cells. Calcein fluorescence is quenched following chelation of low-mass labile iron, and the degree of quenching gives an estimate of the amounts of chelatable iron. However, a requirement for the assay to be able to demonstrate cellular LIP in total is that such iron be localized in the cytosol and not in a membrane-limited compartment. For some time it has been known that a major part of cellular, redox-active, labile, low-mass iron is temporarily localized in the lysosomal compartment as a result of the autophagic degradation of ferruginous materials, such as mitochondrial complexes and ferritin. Even if some calcein-AM may escape cytosolic esterases and enter lysosomes to be cleaved by lysosomal acidic esterases, the resulting calcein does not significantly chelate iron at

Subject(s)
Esters/analysis , Fluoresceins/analysis , Iron Chelating Agents/analysis , Iron/analysis , Iron/metabolism , Cell Membrane Permeability , Cytosol/enzymology , Esterases/metabolism , Esters/metabolism , Fluoresceins/metabolism , HeLa Cells , Humans , Hydrogen-Ion Concentration , Iron Chelating Agents/metabolism , Kinetics , Lysosomes/metabolism , Oxidative Stress , Phagocytosis , Substrate Specificity
16.
Biochem J ; 387(Pt 3): 703-10, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15579135

ABSTRACT

Jurkat cells in culture were exposed to oxidative stress in the form of continuously generated hydrogen peroxide, obtained by the addition of glucose oxidase to the medium. This treatment induced a rapid, dose-dependent increase in the ICIP (intracellular calcein-chelatable iron pool). Early destabilization of lysosomal membranes and subsequent nuclear DNA strand breaks were also observed, as evaluated by the Acridine Orange relocation test and the comet assay respectively. Somewhat later, these effects were followed by a lowered mitochondrial membrane potential, with release of cytochrome c and apoptosis-inducing factor. These events were all prevented if cells were pretreated with the potent iron chelator DFO (desferrioxamine) for a period of time (2-3 h) long enough to allow the drug to reach the lysosomal compartment following fluid-phase endocytosis. The hydrophilic calcein, a cleavage product of calcein acetoxymethyl ester following the action of cytosolic esterases, obviously does not penetrate intact lysosomal membranes, thus explaining why ICIP increased dramatically following lysosomal rupture. The rapid decrease in ICIP after addition of DFO to the medium suggests draining of cytosolic iron to the medium, rather than penetration of DFO through the plasma membrane. Most importantly, these observations directly connect oxidative stress and resultant DNA damage with lysosomal rupture and the release of redox-active iron into the cytosol and, apparently, the nucleus.


Subject(s)
Apoptosis/physiology , DNA Damage/physiology , Hydrogen Peroxide/pharmacology , Iron Chelating Agents/pharmacology , Iron/physiology , Cell Membrane/physiology , Deferoxamine/pharmacology , Fluoresceins/pharmacology , Humans , Jurkat Cells , Lysosomes/physiology , Membrane Potentials/physiology , Mitochondria/physiology , Oxidation-Reduction , Oxidative Stress , Temperature , Time
17.
Free Radic Biol Med ; 33(5): 691-702, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12208356

ABSTRACT

Aspects of the molecular mechanism(s) of hydrogen peroxide-induced DNA damage and cell death were studied in the present investigation. Jurkat T-cells in culture were exposed either to low rates of continuously generated H(2)O(2) by the action of glucose oxidase or to a bolus addition of the same agent. In the first case, steady state conditions were prevailing, while in the latter, H(2)O(2) was removed by the cellular defense systems following first order kinetics. By using single-cell gel electrophoresis (also called comet assay), an initial increase in the formation of DNA single-strand breaks was observed in cells exposed to a bolus of 150 microM H(2)O(2). As the H(2)O(2) was exhausted, a gradual decrease in DNA damage was apparent, indicating the existence of an effective repair of single-strand breaks. Addition of 10 ng glucose oxidase in 100 microl growth medium (containing 1.5 x 10(5) cells) generated 2.0 +/- 0.2 microM H(2)O(2) per min. This treatment induced an increase in the level of single-strand breaks reaching the upper limit of detection by the methodology used and continued to be high for the following 6 h. However, when a variety of markers for apoptotic cell death (DNA cell content, DNA laddering, activation of caspases, PARP cleavage) were examined, only bolus additions of H(2)O(2) were able to induce apoptosis, while the continuous presence of this agent inhibited the execution of the apoptotic process no matter whether the inducer was H(2)O(2) itself or an anti-Fas antibody. These observations stress that, apart from the apparent genotoxic and proapoptotic effects of H(2)O(2), it can also exert antiapoptotic actions when present, even at low concentrations, during the execution of apoptosis.


Subject(s)
Apoptosis , DNA Damage , Hydrogen Peroxide/pharmacology , Blotting, Western , Caspases/metabolism , Cell Survival , Coloring Agents/pharmacology , Comet Assay , DNA Fragmentation , Enzyme Activation , Flow Cytometry , Humans , Jurkat Cells , Poly(ADP-ribose) Polymerases , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...