Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Margarita Tenopoulou is active.

Publication


Featured researches published by Margarita Tenopoulou.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Structural profiling of endogenous S-nitrosocysteine residues reveals unique features that accommodate diverse mechanisms for protein S-nitrosylation

Paschalis-Thomas Doulias; Jennifer L. Greene; Todd M. Greco; Margarita Tenopoulou; Steve H. Seeholzer; Roland L. Dunbrack; Harry Ischiropoulos

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 pKa 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 (χ2 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.


Free Radical Biology and Medicine | 2002

DNA damage and apoptosis in hydrogen peroxide-exposed Jurkat cells: bolus addition versus continuous generation of H2O2

Alexandra Barbouti; Paschalis-Thomas Doulias; Lambros Nousis; Margarita Tenopoulou; Dimitrios Galaris

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 H2O2 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, H2O2 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 μM H2O2. As the H2O2 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 μl growth medium (containing 1.5 × 105 cells) generated 2.0 ± 0.2 μM H2O2 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 H2O2 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 H2O2 itself or an anti-Fas antibody. These observations stress that, apart from the apparent genotoxic and proapoptotic effects of H2O2, it can also exert antiapoptotic actions when present, even at low concentrations, during the execution of apoptosis.


Journal of Biological Chemistry | 2013

Regulation of Protein Function and Signaling by Reversible Cysteine S-Nitrosylation

Neal S. Gould; Paschalis-Thomas Doulias; Margarita Tenopoulou; Karthik Raju; Harry Ischiropoulos

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.


Science Signaling | 2013

Nitric Oxide Regulates Mitochondrial Fatty Acid Metabolism Through Reversible Protein S-Nitrosylation

Paschalis-Thomas Doulias; Margarita Tenopoulou; Jennifer L. Greene; Karthik Raju; Harry Ischiropoulos

S-nitrosylation increases the catalytic efficiency of an enzyme critical to fatty acid β-oxidation. Altering Metabolism Through S-Nitrosylation Derivatives of the gasotransmitter nitric oxide can be covalently linked to cysteine residues through a process called S-nitrosylation. Using mass spectrometry, Doulias et al. identified S-nitrosylated proteins in multiple mouse tissues, including the liver. Many of the S-nitrosylated proteins had roles in metabolic processes that occur in mitochondria, such as β-oxidation of fatty acids. S-nitrosylation of VLCAD [very long chain acyl–coenzyme A (CoA) dehydrogenase], an enzyme in the liver that catalyzes the first committed step in fatty acid β-oxidation, improved its catalytic efficiency in vitro, which would be expected to increase fatty acid metabolism and decrease hepatic steatosis, or the accumulation of fat and triglycerides in the liver. Mice used as a model for obesity (ob/ob mice) spontaneously develop hepatic steatosis, and treatment of ob/ob mice with GSNO, a compound that releases nitric oxide, reduced liver triglyceride concentrations and fatty deposits in the liver. Thus, these results implicate S-nitrosylation in the regulation of the activity of several metabolic enzymes. 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 Cys238, which was absent in mice lacking endothelial nitric oxide synthase, improved its catalytic efficiency. These data implicate protein S-nitrosylation in the regulation of β-oxidation of fatty acids in mitochondria.


Biochemical Journal | 2005

Role of compartmentalized redox-active iron in hydrogen peroxide-induced DNA damage and apoptosis

Margarita Tenopoulou; Paschalis-Thomas Doulias; Alexandra Barbouti; Ulf T. Brunk; Dimitrios Galaris

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.


Biochimica et Biophysica Acta | 2012

Strategies and tools to explore protein S-nitrosylation

Karthik Raju; Paschalis-Thomas Doulias; Margarita Tenopoulou; Jennifer L. Greene; Harry Ischiropoulos

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.


Circulation | 2012

Immunoglobulins against Tyrosine Nitrated Epitopes in Coronary Artery Disease

Leonor Thomson; Margarita Tenopoulou; Richard Lightfoot; Epida Tsika; Ioannis Parastatidis; Marissa Martinez; Todd M. Greco; Paschalis-Thomas Doulias; W.H. Wilson Tang; Stanley L. Hazen; Harry Ischiropoulos

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 &mgr;g antibody Eq/mL plasma versus 0.36±0.8 &mgr;g 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.


Journal of Proteomics | 2013

Site specific identification of endogenous S-nitrosocysteine proteomes

Paschalis-Thomas Doulias; Margarita Tenopoulou; Karthik Raju; Lynn A. Spruce; Steven H. Seeholzer; Harry Ischiropoulos

UNLABELLED 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.


Methods | 2013

Mass spectrometry-based identification of S-nitrosocysteine in vivo using organic mercury assisted enrichment

Paschalis-Thomas Doulias; Karthik Raju; Jennifer L. Greene; Margarita Tenopoulou; Harry Ischiropoulos

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.


Journal of Biological Chemistry | 2015

Strategies for Correcting Very Long Chain Acyl-CoA Dehydrogenase Deficiency

Margarita Tenopoulou; Jie Chen; Jean Bastin; Michael Bennett; Harry Ischiropoulos; Paschalis-Thomas Doulias

Background: VLCAD deficiency is a mitochondrial fatty acid β-oxidation disorder. Results: S-Nitrosylation of Cys-237 in VLCAD increased enzymatic activity and normalized β-oxidation capacity and acylcarnitine levels in VLCAD-deficient cells. Conclusion: Correction of VLCAD deficiency alleviates disease-associated metabolic derangement and biomarker accumulation. Significance: Data provide proof-of-concept for a potential therapeutic approach that may significantly impact the lives of children and adults with β-oxidation deficiencies. 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 β-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 β-oxidation capacity, two hallmarks of the disorder. Overall, this study provides biochemical evidence for a potential therapeutic modality to correct β-oxidation deficiencies.

Collaboration


Dive into the Margarita Tenopoulou's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karthik Raju

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Epida Tsika

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Ioannis Parastatidis

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Leonor Thomson

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge