Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
Add more filters










Publication year range
2.
Free Radic Biol Med ; 163: 268-280, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33359261

ABSTRACT

Chagas disease caused by Trypanosoma cruzi parasite is an endemic infection in America. It is well known that T. cruzi causes a strong immunosuppression during the acute phase of infection. However, it is not clear whether T. cruzi infection is related to metabolic alterations in CD4 T cells that prevent downstream effector function. Here, we evaluated the CD4 T cell metabolic and mitochondrial profiles from non-infected (NI), acute phase (AP) and chronic phase (CP) T. cruzi infected mice. CD4 T cells from all groups showed increased glucose uptake after stimulation. Moreover, the bioenergetic analysis revealed a rise in glycolysis and a higher oxidative metabolism in CD4 T cells from the AP. These cells showed increased proton leak and uncoupling protein 3 (UCP3) expression that correlated with mitochondrial ROS (mROS) accumulation, mitochondrial membrane potential (MMP) depolarization and expression of PD-1. In addition, CD4 T cells with mitochondrial alteration displayed an activated phenotype, and were less functional and more prone to apoptosis. In contrast, mitochondrial alterations were not observed during in vivo activation of CD4 T cells in a model of OVA-immunization. The Mn-superoxide dismutase (SOD2) expression, which is involved in mROS detoxification, was increased during the AP and CP of infection. Remarkably, the apoptosis observed in CD4 T cells with MMP depolarization was prevented by incubation with N-acetyl cysteine (NAC). Thus, our results showed that infection triggered an exacerbated metabolism together with mROS production in CD4 T cells from the AP of infection. However, antioxidant availability may not be sufficient to avoid mitochondrial alterations rendering these cells more susceptible to apoptosis. Our investigation is the first to demonstrate an association between a disturbed metabolism and an impaired CD4 T cell response during T. cruzi infection.


Subject(s)
Chagas Disease , Trypanosoma cruzi , Animals , Apoptosis , CD4-Positive T-Lymphocytes , Chagas Disease/genetics , Mice , Reactive Oxygen Species
3.
J Chem Inf Model ; 60(2): 843-853, 2020 02 24.
Article in English | MEDLINE | ID: mdl-31718175

ABSTRACT

In this work, we employ a multiscale quantum-classical mechanics (QM/MM) scheme to investigate the chemical reactivity of sulfenic acids toward hydrogen peroxide, both in aqueous solution and in the protein environment of the peroxiredoxin alkyl hydroperoxide reductase E from Mycobacterium tuberculosis (MtAhpE). The reaction of oxidation of cysteine with hydrogen peroxides, catalyzed by peroxiredoxins, is usually accelerated several orders of magnitude in comparison with the analogous reaction in solution. The resulting cysteine sulfenic acid is then reduced in other steps of the catalytic cycle, recovering the original thiol. However, under some conditions, the sulfenic acid can react with another equivalent of oxidant to form a sulfinic acid. This process is called overoxidation and has been associated with redox signaling. Herein, we employed a multiscale scheme based on density function theory calculations coupled to the classical AMBER force field, developed in our group, to establish the molecular basis of thiol overoxidation by hydrogen peroxide. Our results suggest that residues that play key catalytic roles in the oxidation of MtAhpE are not relevant in the overoxidation process. Indeed, the calculations propose that the process is unfavored by this particular enzyme microenvironment.


Subject(s)
Hydrogen Peroxide/metabolism , Molecular Dynamics Simulation , Peroxiredoxins/metabolism , Sulfhydryl Compounds/metabolism , Mycobacterium tuberculosis/enzymology , Oxidation-Reduction , Peroxiredoxins/chemistry , Protein Conformation , Thermodynamics
4.
J Crit Care ; 51: 204-212, 2019 06.
Article in English | MEDLINE | ID: mdl-30903936

ABSTRACT

PURPOSE: During sepsis and mechanical ventilation oxidative stress is generated by endothelial and inflammatory lung cells. Our main objective was to study pulmonary NO (nitric oxide) production and nitroxidative stress in mechanically-ventilated septic patients. METHODS: We study 69 mechanically ventilated patients, 36 with sepsis and 33 without sepsis within the first 48 h of ICU admission compared with 33 mechanically ventilated patients without sepsis (MV) plus eight operating room patients without lung disease served as control healthy group (ORCG). Nitrite plus nitrate (NOx-), 3-nitrotyrosine and malondialdehyde (MDA) in bronchoalveolar lavage fluid (BALF) were analyzed. RESULTS: BALF NOx-, BALF 3-nitrotyrosine, BALF MDA, and plasma NOx- were higher in the Sepsis than in MV patients (all p < .05). Both SG and MV patients had higher BALF NOx- than the healthy control group (p < .001). In the Sepsis patients, the ICU non-survivors had higher levels of BALF NOx- than ICU survivors 80(70-127) µM versus 31(15-47) µM, p < .001. CONCLUSIONS: We conclude that during early phases of sepsis there is an enhanced lung nitroxidative stress due to an increase of NO production leading to secondary NO-derived oxidants, which promote protein nitration and lipid peroxidation.


Subject(s)
Nitric Oxide/metabolism , Oxidative Stress/physiology , Respiration, Artificial/adverse effects , Respiratory Insufficiency , Sepsis/complications , Adult , Aged , Bronchoalveolar Lavage Fluid , Case-Control Studies , Female , Humans , Male , Malondialdehyde/metabolism , Middle Aged , Pilot Projects , Respiratory Insufficiency/metabolism , Respiratory Insufficiency/therapy , Sepsis/metabolism
5.
Free Radic Res ; 49(2): 122-32, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25373783

ABSTRACT

Increased production of reactive nitrogen (RNS) and oxygen (ROS) species and its detrimental effect to mitochondria are associated with endothelial dysfunction. This study was designed to determine the effect of a peroxynitrite flux, promoted by 1,3-morpholinosydnonimine (SIN-1), in mitochondrial function and some redox homeostasis parameters in bovine aortic endothelial cells (BAEC). Moreover, the effect of diphenyl diselenide (PhSe)2, a simple organic selenium compound, in preventing peroxynitrite-mediated cytotoxicity was also investigated. Our results showed that overnight exposure to SIN-1 (250 µM) caused a profound impairment of oxygen consumption, energy generation and reserve capacity in mitochondria of BAEC. Mitochondrial dysfunction resulted in an additional intracellular production of peroxynitrite, amplifying the phenomenon and leading to changes in redox homeostasis. Moreover, we observed an extensive decline in mitochondrial membrane potential (ΔΨm) induced by peroxynitrite and this event was associated with apoptotic-type cell death. Alternatively, the pretreatment of BAEC with (PhSe)2, hindered peroxynitrite-mediated cell damage by preserving mitochondrial and endothelial function and consequently preventing apoptosis. The protective effect of (PhSe)2 was related to its ability to improve the intracellular redox state by increasing the expression of different isoforms of peroxiredoxins (Prx-1-3), efficient enzymes in peroxynitrite detoxification.


Subject(s)
Benzene Derivatives/pharmacology , Endothelial Cells/drug effects , Energy Metabolism/drug effects , Mitochondria/drug effects , Molsidomine/analogs & derivatives , Organoselenium Compounds/pharmacology , Peroxiredoxins/metabolism , Peroxynitrous Acid/metabolism , Animals , Aorta/cytology , Cattle , Endothelial Cells/enzymology , Homeostasis/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/enzymology , Molsidomine/chemistry , Oxidation-Reduction , Peroxynitrous Acid/chemistry , Peroxynitrous Acid/toxicity
6.
Free Radic Res ; 48(6): 684-93, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24601712

ABSTRACT

The Fe-S cluster of mitochondrial aconitase is rapidly and selectively inactivated by oxidants, yielding an inactive enzyme that can be reactivated by reductants and iron in vivo. In order to elucidate the metabolic impact of oxidant-dependent aconitase inhibition over the citric acid cycle, the respiratory chain reactions, and reactive species formation, we performed a metabolic analysis using isolated mitochondria from different rat tissues. Titrations with fluorocitrate showed IC50 for aconitase inhibition ranging from 7 to 24 µM. The aconitase inhibition threshold in mitochondrial oxygen consumption was determined to range from 63 to 98%. Of the tissues examined, brain and heart exhibited the highest values in the flux control coefficient (> 0.95). Aconitase-specific activity varied widely among tissues examined from ~60 mU/mg in liver to 321 mU/mg in kidney at 21% O2. In brain and heart, aconitase-specific activity increased by 42 and 12%, respectively, at 2% O2 reflecting aconitase inactivation by oxygen-derived oxidants at 21% O2. Both mitochondrial membrane potential and hydrogen peroxide production significantly decreased upon aconitase inhibition in heart and brain mitochondria. These results indicate that aconitase can exert control over respiration (with tissue specificity) and support the hypothesis that inactivation of aconitase may provide a control mechanism to prevent O2(●-) and H2O2 formation by the respiratory chain.


Subject(s)
Aconitate Hydratase/biosynthesis , Hydrogen Peroxide/metabolism , Mitochondria/enzymology , Oxygen Consumption/physiology , Superoxides/metabolism , Aconitate Hydratase/antagonists & inhibitors , Animals , Brain/metabolism , Citric Acid Cycle/physiology , Electron Transport/physiology , Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Myocardium/metabolism , Oxidative Stress , Rats , Rats, Wistar
7.
Curr Pharm Des ; 17(35): 3905-32, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21933142

ABSTRACT

Nitric oxide participates in a wide array of physiological processes, ranging from neurotransmission to precursor of cytotoxic effector molecules of the immune system. Although nitric oxide is a mildly reactive intermediary, it can act as a precursor of strong oxidants under pathological conditions associated with oxidative stress including cardiovascular, inflammatory and neurodegenerative disorders. Peroxynitrite, the reaction product of nitric oxide with superoxide radicals, emerges as one of the principal players of nitric oxidederived toxicity due to its facile formation and ability to react with several critical cellular targets including, thiols, proteins, lipids and DNA. The extent of "nitroxidative stress" is determined by several factors, including the concentration and exposure time to this reactive species or its derived radicals and by the ability of the cell to face the oxidative challenge by means of its antioxidant defenses. The inflicted biomolecular damage can result on minimal and reversible changes to cell and tissue physiology, to alteration in bioenergetics, disruption of DNA integrity, mitochondrial dysfunction and even cell death. Although dissecting the free radical chemistry pathways responsible of cell/tissue disturbance of oxidative signaling and promotion of oxidative damage arising from nitric oxide-derived oxidants in a biological context is a vast endeavor, is an ineludible task in order to generate a rational therapeutic approach to modulate nitroxidative stress. Several redox-based pharmacological strategies with a collection of compounds with varying mechanisms of action have been tested at the cellular, preclinical and even clinical levels, and some novel and promising developments are underway. This review deals with key kinetic and biochemical aspects of nitric oxide-derived oxidant formation and reactions in biological systems, emphasizing the current evidence at the biochemical, cell/tissue and animal/human levels that support a pathophysiological role for peroxynitrite and related species in human pathology. In addition, a selection of available pharmacological tools will be discussed as an effort to rationalize antioxidant and/or redox-based therapeutic interventions in disease models.


Subject(s)
Antioxidants/therapeutic use , Molecular Targeted Therapy , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Peroxynitrous Acid/metabolism , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Necrosis/drug therapy , Necrosis/metabolism , Nitric Oxide/antagonists & inhibitors , Oxidants/antagonists & inhibitors , Oxidants/metabolism , Peroxynitrous Acid/antagonists & inhibitors
8.
Int J Parasitol ; 39(13): 1455-64, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19505468

ABSTRACT

Virulence of Trypanosoma cruzi depends on a variety of genetic and biochemical factors. It has been proposed that components of the parasites' antioxidant system may play a key part in this process by pre-adapting the pathogen to the oxidative environment encountered during host cell invasion. Using several isolates (10 strains) belonging to the two major phylogenetic lineages (T. cruzi-I and T. cruzi-II), we investigated whether there was an association between virulence (ranging from highly aggressive to attenuated isolates at the parasitemia and histopathological level) and the antioxidant enzyme content. Antibodies raised against trypanothione synthetase (TcTS), ascorbate peroxidase (TcAPX), mitochondrial and cytosolic tryparedoxin peroxidases (TcMPX and TcCPX) and trypanothione reductase (TcTR) were used to evaluate the antioxidant enzyme levels in epimastigote and metacyclic trypomastigote forms in the T. cruzi strains. Levels of TcCPX, TcMPX and TcTS were shown to increase during differentiation from the non-infective epimastigote to the infective metacyclic trypomastigote stage in all parasite strains examined. Peroxiredoxins were found to be present at higher levels in the metacyclic infective forms of the virulent isolates compared with the attenuated strains. Additionally, an increased resistance of epimastigotes from virulent T. cruzi populations to hydrogen peroxide and peroxynitrite challenge was observed. In mouse infection models, a direct correlation was found between protein levels of TcCPX, TcMPX and TcTS, and the parasitemia elicited by the different isolates studied (Pearson's coefficient: 0.617, 0.771, 0.499; respectively, P<0.01). No correlation with parasitemia was found for TcAPX and TcTR proteins in any of the strains analyzed. Our data support that enzymes of the parasite antioxidant armamentarium at the onset of infection represent new virulence factors involved in the establishment of disease.


Subject(s)
Antioxidants/metabolism , Chagas Disease/enzymology , Trypanosoma cruzi/enzymology , Trypanosoma cruzi/pathogenicity , Virulence Factors/metabolism , Virulence , Animals , Disease Models, Animal , Male , Mice
9.
Braz. j. med. biol. res ; 42(4): 305-311, Apr. 2009. ilus, tab
Article in English | LILACS | ID: lil-509174

ABSTRACT

Human serum albumin (HSA) is the most abundant protein in the intravascular compartment. It possesses a single thiol, Cys34, which constitutes ~80 percent of the total thiols in plasma. This thiol is able to scavenge plasma oxidants. A central intermediate in this potential antioxidant activity of human serum albumin is sulfenic acid (HSA-SOH). Work from our laboratories has demonstrated the formation of a relatively stable sulfenic acid in albumin through complementary spectrophotometric and mass spectrometric approaches. Recently, we have been able to obtain quantitative data that allowed us to measure the rate constants of sulfenic acid reactions with molecules of analytical and biological interest. Kinetic considerations led us to conclude that the most likely fate for sulfenic acid formed in the plasma environment is the reaction with low molecular weight thiols to form mixed disulfides, a reversible modification that is actually observed in ~25 percent of circulating albumin. Another possible fate for sulfenic acid is further oxidation to sulfinic and sulfonic acids. These irreversible modifications are also detected in the circulation. Oxidized forms of albumin are increased in different pathophysiological conditions and sulfenic acid lies in a mechanistic junction, relating oxidizing species to final thiol oxidation products.


Subject(s)
Humans , Serum Albumin/chemistry , Serum Albumin/metabolism , Sulfenic Acids/metabolism , Sulfhydryl Compounds/metabolism , Oxidation-Reduction , Protein Conformation , Sulfenic Acids/isolation & purification
10.
Braz J Med Biol Res ; 42(4): 305-11, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19330257

ABSTRACT

Human serum albumin (HSA) is the most abundant protein in the intravascular compartment. It possesses a single thiol, Cys34, which constitutes ~80% of the total thiols in plasma. This thiol is able to scavenge plasma oxidants. A central intermediate in this potential antioxidant activity of human serum albumin is sulfenic acid (HSA-SOH). Work from our laboratories has demonstrated the formation of a relatively stable sulfenic acid in albumin through complementary spectrophotometric and mass spectrometric approaches. Recently, we have been able to obtain quantitative data that allowed us to measure the rate constants of sulfenic acid reactions with molecules of analytical and biological interest. Kinetic considerations led us to conclude that the most likely fate for sulfenic acid formed in the plasma environment is the reaction with low molecular weight thiols to form mixed disulfides, a reversible modification that is actually observed in ~25% of circulating albumin. Another possible fate for sulfenic acid is further oxidation to sulfinic and sulfonic acids. These irreversible modifications are also detected in the circulation. Oxidized forms of albumin are increased in different pathophysiological conditions and sulfenic acid lies in a mechanistic junction, relating oxidizing species to final thiol oxidation products.


Subject(s)
Serum Albumin/chemistry , Serum Albumin/metabolism , Sulfenic Acids/metabolism , Sulfhydryl Compounds/metabolism , Humans , Oxidation-Reduction , Protein Conformation , Sulfenic Acids/isolation & purification
11.
Amino Acids ; 32(4): 543-51, 2007.
Article in English | MEDLINE | ID: mdl-17061035

ABSTRACT

Sulfenic acid (RSOH) is a central intermediate in both the reversible and irreversible redox modulation by reactive species of an increasing number of proteins involved in signal transduction and enzymatic pathways. In this paper we focus on human serum albumin (HSA), the most abundant plasma protein, proposed to serve antioxidant functions in the vascular compartment. Sulfenic acid in HSA has been previously detected using different methods after oxidation of its single free thiol Cys34 through one- or two-electron mechanisms. Since recent evidence suggests that sulfenic acid in HSA is stabilized within the protein environment, this derivative represents an appropriate model to examine protein sulfenic acid biochemistry, structure and reactivity. Sulfenic acid in HSA could be involved in mixed disufide formation, supporting a role of HSA-Cys34 as an important redox regulator in extracellular compartments.


Subject(s)
Free Radicals/metabolism , Serum Albumin/chemistry , Serum Albumin/metabolism , Sulfenic Acids/metabolism , Humans , Models, Molecular , Oxidants , Oxidation-Reduction , Protein Conformation , Sulfenic Acids/isolation & purification
12.
Amino Acids ; 32(4): 501-15, 2007.
Article in English | MEDLINE | ID: mdl-17077966

ABSTRACT

In this review we address current concepts on the biological occurrence, levels and consequences of protein tyrosine nitration in biological systems. We focused on mechanistic aspects, emphasizing on the free radical mechanisms of protein 3-nitrotyrosine formation and critically analyzed the restrictions for obtaining large tyrosine nitration yields in vivo, mainly due to the presence of strong reducing systems (e.g. glutathione) that can potently inhibit at different levels the nitration process. Evidence is provided to show that the existence of metal-catalyzed processes, the assistance of nitric oxide-dependent nitration steps and the facilitation by hydrophobic environments, provide individually and/or in combination, feasible scenarios for nitration in complex biological milieux. Recent studies using hydrophobic tyrosine analogs and tyrosine-containing peptides have revealed that factors controlling nitration in hydrophobic environments such as biomembranes and lipoproteins can differ to those in aqueous compartments. In particular, exclusion of key soluble reductants from the lipid phase will more easily allow nitration and lipid-derived radicals are suggested as important mediators of the one-electron oxidation of tyrosine to tyrosyl radical in proteins associated to hydrophobic environments. Development and testing of hydrophilic and hydrophobic probes that can compete with endogenous constituents for the nitrating intermediates provide tools to unravel nitration mechanisms in vitro and in vivo; additionally, they could also serve to play cellular and tissue protective functions against the toxic effects of protein tyrosine nitration.


Subject(s)
Cell Membrane/metabolism , Free Radicals/metabolism , Peptides/metabolism , Proteins/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Dimerization , Hydrophobic and Hydrophilic Interactions , Tyrosine/chemistry
13.
Braz. j. med. biol. res ; 38(12): 1825-1834, Dec. 2005. ilus
Article in English | LILACS | ID: lil-417191

ABSTRACT

Nitric oxide (ÀNO) is a diffusible messenger implicated in Trypanosoma cruzi resistance. Excess production of ÀNO and oxidants leads to the generation of nitrogen dioxide (ÀNO2), a strong nitrating agent. Tyrosine nitration is a post-translational modification resulting from the addition of a nitro (-NO2) group to the ortho-position of tyrosine residues. Detection of protein 3-nitrotyrosine is regarded as a marker of nitro-oxidative stress and is observed in inflammatory processes. The formation and role of nitrating species in the control and myocardiopathy of T. cruzi infection remain to be studied. We investigated the levels of ÀNO and protein 3-nitrotyrosine in the plasma of C3H and BALB/c mice and pharmacologically modulated their production during the acute phase of T. cruzi infection. We also looked for protein 3-nitrotyrosine in the hearts of infected animals. Our results demonstrated that C3H animals produced higher amounts of ÀNO than BALB/c mice, but their generation of peroxynitrite was not proportionally enhanced and they had higher parasitemias. While N G-nitro-arginine methyl ester treatment abolished ÀNO production and drastically augmented the parasitism, mercaptoethylguanidine and guanido-ethyl disulfide, at doses that moderately reduced the ÀNO and 3-nitrotyrosine levels, paradoxically diminished the parasitemia in both strains. Nitrated proteins were also demonstrated in myocardial cells of infected mice. These data suggest that the control of T. cruzi infection depends not only on the capacity to produce ÀNO, but also on its metabolic fate, including the generation of nitrating species that may constitute an important element in parasite resistance and collateral myocardial damage.


Subject(s)
Animals , Mice , Chagas Cardiomyopathy/metabolism , Nitric Oxide/biosynthesis , Tyrosine/analogs & derivatives , Acute Disease , Chagas Cardiomyopathy/blood , Chagas Cardiomyopathy/pathology , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Mice, Inbred BALB C , Biomarkers/blood , Nitric Oxide/blood , Parasitemia/etiology , Tyrosine/biosynthesis , Tyrosine/blood
14.
Braz J Med Biol Res ; 38(12): 1825-34, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16302097

ABSTRACT

Nitric oxide (.NO) is a diffusible messenger implicated in Trypanosoma cruzi resistance. Excess production of .NO and oxidants leads to the generation of nitrogen dioxide (.NO2), a strong nitrating agent. Tyrosine nitration is a post-translational modification resulting from the addition of a nitro (-NO2) group to the ortho-position of tyrosine residues. Detection of protein 3-nitrotyrosine is regarded as a marker of nitro-oxidative stress and is observed in inflammatory processes. The formation and role of nitrating species in the control and myocardiopathy of T. cruzi infection remain to be studied. We investigated the levels of .NO and protein 3-nitrotyrosine in the plasma of C3H and BALB/c mice and pharmacologically modulated their production during the acute phase of T. cruzi infection. We also looked for protein 3-nitrotyrosine in the hearts of infected animals. Our results demonstrated that C3H animals produced higher amounts of .NO than BALB/c mice, but their generation of peroxynitrite was not proportionally enhanced and they had higher parasitemias. While N G-nitro-arginine methyl ester treatment abolished .NO production and drastically augmented the parasitism, mercaptoethylguanidine and guanido-ethyl disulfide, at doses that moderately reduced the .NO and 3-nitrotyrosine levels, paradoxically diminished the parasitemia in both strains. Nitrated proteins were also demonstrated in myocardial cells of infected mice. These data suggest that the control of T. cruzi infection depends not only on the capacity to produce .NO, but also on its metabolic fate, including the generation of nitrating species that may constitute an important element in parasite resistance and collateral myocardial damage.


Subject(s)
Chagas Cardiomyopathy/metabolism , Nitric Oxide/biosynthesis , Tyrosine/analogs & derivatives , Acute Disease , Animals , Biomarkers/blood , Chagas Cardiomyopathy/blood , Chagas Cardiomyopathy/pathology , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Nitric Oxide/blood , Parasitemia/etiology , Tyrosine/biosynthesis , Tyrosine/blood
15.
Neuroscience ; 135(1): 111-20, 2005.
Article in English | MEDLINE | ID: mdl-16111821

ABSTRACT

3-Hydroxyglutaric acid (3HGA) accumulates in the inherited neurometabolic disorder known as glutaryl-CoA dehydrogenase deficiency. The disease is clinically characterized by severe neurological symptoms, frontotemporal atrophy and striatum degeneration. Because of the pathophysiology of the brain damage in glutaryl-CoA dehydrogenase deficiency is not completed clear, we investigated the in vitro effect of 3HGA (0.01-5.0mM) on critical enzyme activities of energy metabolism, including the respiratory chain complexes I-V, creatine kinase isoforms and Na(+),K(+)-ATPase in cerebral cortex and striatum from 30-day-old rats. Complex II activity was also studied in rat C6-glioma cells exposed to 3HGA. The effect of 3HGA was further investigated on the rate of oxygen consumption in mitochondria from rat cerebrum. We observed that 1.0mM 3HGA significantly inhibited complex II in cerebral cortex and C6 cells but not the other activities of the respiratory chain complexes. Creatine kinase isoforms and Na(+),K(+)-ATPase were also not affected by the acid. Furthermore, no inhibition of complex II activity occurred when mitochondrial preparations from cerebral cortex or striatum homogenates were used. In addition, 3HGA significantly lowered the respiratory control ratio in the presence of glutamate/malate and succinate under stressful conditions or when mitochondria were permeabilized with digitonin. Since 3HGA stimulated oxygen consumption in state IV and compromised ATP formation, it can be presumed that this organic acid might act as an endogenous uncoupler of mitochondria respiration. Finally, we observed that 3HGA changed C6 cell morphology from a round flat to a spindle-differentiated shape, but did not alter cell viability neither induced apoptosis. The data provide evidence that 3HGA provokes a moderate impairment of brain energy metabolism and do not support the view that 3HGA-induced energy failure would solely explain the characteristic brain degeneration observed in glutaryl-CoA dehydrogenase deficiency patients.


Subject(s)
Brain Chemistry/drug effects , Energy Metabolism/drug effects , Glutarates/pharmacology , Animals , Ca(2+) Mg(2+)-ATPase/metabolism , Cell Line, Tumor/metabolism , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cell Survival/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Creatine Kinase/metabolism , Cytosol/enzymology , Electron Transport/drug effects , Glioma/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Neostriatum/drug effects , Neostriatum/metabolism , Nerve Tissue Proteins/biosynthesis , Neurons/drug effects , Neurons/ultrastructure , Oxygen Consumption/drug effects , Rats , Sodium-Potassium-Exchanging ATPase/metabolism
16.
Amino Acids ; 25(3-4): 295-311, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14661092

ABSTRACT

Peroxynitrite, the product of the fast reaction between nitric oxide ((*)NO) and superoxide O(2)(*-) radicals, is an oxidizing and nitrating agent which is able to traverse biological membranes. The reaction of peroxynitrite with proteins occurs through three possible pathways. First, peroxynitrite reacts directly with cysteine, methionine and tryptophan residues. Second, peroxynitrite reacts fast with transition metal centers and selenium-containing amino acids. Third, secondary free radicals arising from peroxynitrite homolysis such as hydroxyl and nitrogen dioxide, and the carbonate radical formed in the presence of carbon dioxide, react with protein moieties too. Nitration of tyrosine residues is being recognized as a marker of the contribution of nitric oxide to oxidative damage. Peroxynitrite-dependent tyrosine nitration is likely to occur through the initial reaction of peroxynitrite with carbon dioxide or metal centers leading to secondary nitrating species. The preferential protein targets of peroxynitrite and the role of proteins in peroxynitrite detoxifying pathways are discussed.


Subject(s)
Amino Acids/chemistry , Peroxynitrous Acid/chemistry , Proteins/chemistry , Amino Acids/metabolism , Animals , Antioxidants/chemistry , Antioxidants/metabolism , Cytosol/metabolism , Free Radicals/metabolism , Humans , Models, Chemical , Molecular Structure , Nitric Oxide/metabolism , Oxidation-Reduction , Peroxynitrous Acid/metabolism , Proteins/metabolism , Superoxides/metabolism
17.
Arch Biochem Biophys ; 395(2): 225-32, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11697860

ABSTRACT

Peroxynitrite (PN), the product of the diffusion-limited reaction between nitric oxide (*NO) and superoxide (O*-(2)), represents a relevant mediator of oxidative modifications in low-density lipoprotein (LDL). This work shows for the first time the simultaneous action of low-controlled fluxes of PN and *NO on LDL oxidation in terms of lipid and protein modifications as well as oxidized lipid-protein adduct formation. Fluxes of PN (e.g., 1 microM min(-1)) initiated lipid oxidation in LDL as measured by conjugated dienes and cholesteryl ester hydroperoxides formation. Oxidized-LDL exhibited a characteristic fluorescent emission spectra (lambda(exc) = 365 nm, lambda(max) = 417 nm) in parallel with changes in both the free amino groups content and the relative electrophoretic mobility of the particle. Physiologically relevant fluxes of *NO (80-300 nM min(-1)) potently inhibited these PN-dependent oxidative processes. These results are consistent with PN-induced adduct formation between lipid oxidation products and free amino groups of LDL in a process prevented by the simultaneous presence of *NO. The balance between rates of PN and *NO production in the vascular wall will critically determine the final extent of LDL oxidative modifications leading or not to scavenger receptor-mediated LDL uptake and foam cell formation.


Subject(s)
Lipid Metabolism , Lipoproteins, LDL/metabolism , Nitric Oxide/metabolism , Peroxynitrous Acid/metabolism , Proteins/metabolism , Humans , Liposomes/metabolism , Models, Chemical , Oxygen/metabolism , Protein Binding , Time Factors
18.
J Biol Chem ; 276(49): 46017-23, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11590168

ABSTRACT

Tyrosine hydroxylase (TH) is modified by nitration after exposure of mice to 1-methyl-4-phenyl-1,2,3,6-tetrahydrophenylpyridine. The temporal association of tyrosine nitration with inactivation of TH activity in vitro suggests that this covalent post-translational modification is responsible for the in vivo loss of TH function (Ara, J., Przedborski, S., Naini, A. B., Jackson-Lewis, V., Trifiletti, R. R., Horwitz, J., and Ischiropoulos, H. (1998) Proc. Natl. Acad. Sci. U. S. A. 95, 7659-7663). Recent data showed that cysteine oxidation rather than tyrosine nitration is responsible for TH inactivation after peroxynitrite exposure in vitro (Kuhn, D. M., Aretha, C. W., and Geddes, T. J. (1999) J. Neurosci. 19, 10289-10294). However, re-examination of the reaction of peroxynitrite with purified TH failed to produce cysteine oxidation but resulted in a concentration-dependent increase in tyrosine nitration and inactivation. Cysteine oxidation is only observed after partial unfolding of the protein. Tyrosine residue 423 and to lesser extent tyrosine residues 428 and 432 are modified by nitration. Mutation of Tyr(423) to Phe resulted in decreased nitration as compared with wild type protein without loss of activity. Stopped-flow experiments reveal a second order rate constant of (3.8 +/- 0.9) x 10(3) m(-1) s(-1) at pH 7.4 and 25 degrees C for the reaction of peroxynitrite with TH. Collectively, the data indicate that peroxynitrite reacts with the metal center of the protein and results primarily in the nitration of tyrosine residue 423, which is responsible for the inactivation of TH.


Subject(s)
Enzyme Inhibitors/pharmacology , Nitrates/metabolism , Peroxynitrous Acid/pharmacology , Tyrosine 3-Monooxygenase/antagonists & inhibitors , Tyrosine 3-Monooxygenase/metabolism , Base Sequence , Circular Dichroism , DNA Primers , Kinetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism
19.
Proc Natl Acad Sci U S A ; 98(13): 7301-6, 2001 Jun 19.
Article in English | MEDLINE | ID: mdl-11404465

ABSTRACT

Until recently, a capacity for apoptosis and synthesis of nitric oxide *NO) were viewed as exclusive to multicellular organisms. The existence of these processes in unicellular parasites was recently described, with their biological significance remaining to be elucidated. We have evaluated L-arginine metabolism in Trypanosoma cruzi in the context of human serum-induced apoptotic death. Apoptosis was evidenced by the induction of DNA fragmentation and the inhibition of [3H]thymidine incorporation, which were inhibited by the caspase inhibitor Ac-Asp-Glu-Val-aspartic acid aldehyde (DEVD-CHO). In T. cruzi exposed to death stimuli, supplementation with L-arginine inhibited DNA fragmentation, restored [3H]thymidine incorporation, and augmented parasite *NO production. These effects were inhibited by the *NO synthase inhibitor N(omega)-nitroarginine methyl ester (L-NAME). Exogenous *NO limited DNA fragmentation but did not restore proliferation rates. Because L-arginine is also a substrate for arginine decarboxylase (ADC), and its product agmatine is a precursor for polyamine synthesis, we evaluated the contribution of polyamines to limiting apoptosis. Addition of agmatine, putrescine, and the polyamines spermine and spermidine to T. cruzi sustained parasite proliferation and inhibited DNA fragmentation. Also, the ADC inhibitor difluoromethylarginine inhibited L-arginine-dependent restoration of parasite replication rates, while the protection from DNA fragmentation persisted. In aggregate, these results indicate that T. cruzi epimastigotes can undergo programmed cell death that can be inhibited by L-arginine by means of (i) a *NO synthase-dependent *NO production that suppresses apoptosis and (ii) an ADC-dependent production of polyamines that support parasite proliferation.


Subject(s)
Apoptosis/physiology , Arginine/metabolism , Nitric Oxide/metabolism , Polyamines/metabolism , Polyamines/pharmacology , Trypanosoma cruzi/physiology , Agmatine/pharmacology , Animals , Apoptosis/drug effects , Arginine/pharmacology , Cell Division/drug effects , Cysteine Proteinase Inhibitors/pharmacology , DNA Fragmentation , DNA, Protozoan/biosynthesis , Humans , NG-Nitroarginine Methyl Ester/pharmacology , Oligopeptides/pharmacology , Putrescine/pharmacology , Spermidine/pharmacology , Spermine/pharmacology , Thymidine/metabolism , Trypanosoma cruzi/cytology , Trypanosoma cruzi/drug effects
20.
Free Radic Res ; 34(5): 467-75, 2001 May.
Article in English | MEDLINE | ID: mdl-11378530

ABSTRACT

We have reported previously that the apparent rate of peroxynitrite (ONOO(-) ) decay, as followed from its absorbance at 302 nm, decreases in the presence of hydrogen peroxide, mannitol and ethanol (Alvarez et al., 1995, Chem. Res. Toxicol. 8:859-864; Alvarez et al., 1998, Free Radic. Biol. Med. 24:1331-1337). Recently, two papers confirmed the observation and proposed that this slowing effect was due to the formation of absorbing peroxynitrate (O(2) NOO(-) ) as intermediate (Goldstein and Czapski, 1998, J. Am. Chem. Soc. 120:3458-3463; Hodges and Ingold, 1999, J. Am. Chem. Soc. 121:10695-10701). Peroxynitrate would be formed from the reaction of peroxynitrite-derived nitrogen dioxide with superoxide. Superoxide, in turn, would arise from the one-electron oxidation of hydrogen peroxide, or from the reaction of reductive radicals derived from mannitol and ethanol with dioxygen. In agreement with this concept, we show herein that under the conditions of our previous work, the slowing effect is prevented by superoxide dismutase and, in the case of mannitol and ethanol, by reducing the dioxygen concentration of the reaction solutions. Thus, superoxide formation is necessary for the decrease in the rate of absorbance decay. In addition, by simulations using known rate constants and absorption coefficients, we show that the slowing effect can be quantitatively accounted for by the formation of peroxynitrate.


Subject(s)
Ethanol/chemistry , Hydrogen Peroxide/chemistry , Mannitol/chemistry , Nitrates/chemistry , Biophysics/methods , Ethanol/metabolism , Mannitol/metabolism , Nitrates/metabolism , Superoxide Dismutase/chemistry , Superoxide Dismutase/metabolism , Superoxides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL