Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(18): e2221047120, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-37098065

RESUMO

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) contains an active site Cys and is one of the most sensitive cellular enzymes to oxidative inactivation and redox regulation. Here, we show that inactivation by hydrogen peroxide is strongly enhanced in the presence of carbon dioxide/bicarbonate. Inactivation of isolated mammalian GAPDH by H2O2 increased with increasing bicarbonate concentration and was sevenfold faster in 25 mM (physiological) bicarbonate compared with bicarbonate-free buffer of the same pH. H2O2 reacts reversibly with CO2 to form a more reactive oxidant, peroxymonocarbonate (HCO4-), which is most likely responsible for the enhanced inactivation. However, to account for the extent of enhancement, we propose that GAPDH must facilitate formation and/or targeting of HCO4- to promote its own inactivation. Inactivation of intracellular GAPDH was also strongly enhanced by bicarbonate: treatment of Jurkat cells with 20 µM H2O2 in 25 mM bicarbonate buffer for 5 min caused almost complete GAPDH inactivation, but no loss of activity when bicarbonate was not present. H2O2-dependent GAPDH inhibition in bicarbonate buffer was observed even in the presence of reduced peroxiredoxin 2 and there was a significant increase in cellular glyceraldehyde-3-phosphate/dihydroxyacetone phosphate. Our results identify an unrecognized role for bicarbonate in enabling H2O2 to influence inactivation of GAPDH and potentially reroute glucose metabolism from glycolysis to the pentose phosphate pathway and NAPDH production. They also demonstrate what could be wider interplay between CO2 and H2O2 in redox biology and the potential for variations in CO2 metabolism to influence oxidative responses and redox signaling.


Assuntos
Dióxido de Carbono , Peróxido de Hidrogênio , Humanos , Animais , Peróxido de Hidrogênio/química , Dióxido de Carbono/química , Bicarbonatos , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Peroxirredoxinas/metabolismo , Oxirredução , Mamíferos/metabolismo
2.
J Biol Chem ; 296: 100494, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33667550

RESUMO

Peroxiredoxin 2 (Prdx2) is a thiol peroxidase with an active site Cys (C52) that reacts rapidly with H2O2 and other peroxides. The sulfenic acid product condenses with the resolving Cys (C172) to form a disulfide which is recycled by thioredoxin or GSH via mixed disulfide intermediates or undergoes hyperoxidation to the sulfinic acid. C172 lies near the C terminus, outside the active site. It is not established whether structural changes in this region, such as mixed disulfide formation, affect H2O2 reactivity. To investigate, we designed mutants to cause minimal (C172S) or substantial (C172D and C172W) structural disruption. Stopped flow kinetics and mass spectrometry showed that mutation to Ser had minimal effect on rates of oxidation and hyperoxidation, whereas Asp and Trp decreased both by ∼100-fold. To relate to structural changes, we solved the crystal structures of reduced WT and C172S Prdx2. The WT structure is highly similar to that of the published hyperoxidized form. C172S is closely related but more flexible and as demonstrated by size exclusion chromatography and analytical ultracentrifugation, a weaker decamer. Size exclusion chromatography and analytical ultracentrifugation showed that the C172D and C172W mutants are also weaker decamers than WT, and small-angle X-ray scattering analysis indicated greater flexibility with partially unstructured regions consistent with C-terminal unfolding. We propose that these structural changes around C172 negatively impact the active site geometry to decrease reactivity with H2O2. This is relevant for Prdx turnover as intermediate mixed disulfides with C172 would also be disruptive and could potentially react with peroxides before resolution is complete.


Assuntos
Cisteína/química , Cisteína/metabolismo , Peróxido de Hidrogênio/metabolismo , Peroxirredoxinas/química , Peroxirredoxinas/metabolismo , Sequência de Aminoácidos , Domínio Catalítico , Cristalografia por Raios X , Humanos , Peróxido de Hidrogênio/química , Mutação , Oxidantes/química , Oxidantes/metabolismo , Oxirredução , Relação Estrutura-Atividade
3.
Biochemistry (Mosc) ; 86(1): 84-91, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33705284

RESUMO

2-Cys peroxiredoxins are abundant thiol proteins that react efficiently with a wide range of peroxides. Unlike other enzymes, their exceptionally high reactivity does not rely on cofactors. The mechanism of oxidation and reduction of peroxiredoxins places them in a good position to act as antioxidants as well as key players in redox signaling. Understanding of the intimate details of peroxiredoxin functioning is important for translational research.


Assuntos
Peroxirredoxinas/metabolismo , Animais , Humanos , Oxirredução , Transdução de Sinais
4.
Rapid Commun Mass Spectrom ; 34(11): e8774, 2020 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-32119756

RESUMO

RATIONALE: Oxidative stress is an imbalance between reactive free radical oxygen species and antioxidant defenses. Its consequences can lead to numerous pathologies. Regulating oxidative stress is the complex interplay between antioxidant recycling and thiol-containing regulatory proteins. Understanding these regulatory mechanisms is important for preventing onset of oxidative stress. The aim of this study was to investigae S-thiol protein chemistry associated with oxidized vitamin C (dehydroascorbate, DHA), homocysteine (HcySH) and glutathione (GSH) using mass spectrometry. METHODS: Glutaredoxin-1 (Grx-1) was incubated with DHA, with and without GSH and HcySH. Disulfide formation was followed by electrospray ionization mass spectrometry (ESI-MS) of intact proteins and by LC/ESI-MS/MS of peptides from protein tryptic digestions. The mechanism of DHA-mediated S-thiolation was investigated using two synthetic peptides: AcFHACAAK and AcFHACE. Three proteins, i.e. human hemoglobin (HHb), recombinant peroxiredoxin 2 (Prdx2) and Grx-1, were S-homocysteinylated followed by S-transthiolyation with GSH and investigated by ESI-MS and ESI-MS/MS. RESULTS: ESI-MS analysis reveals that DHA mediates disulfide formation and S-thiolation by HcySH as well as GSH of Grx-1. LC/ESI-MS/MS analysis allows identification of Grx-1 S-thiolated cysteine adducts. The mechanism by which DHA mediates S-thiolation of heptapeptide AcFHACAAK is shown to be via initial formation of a thiohemiketal adduct. In addition, ESI-MS of intact proteins shows that GSH can S-transthiolate S-homocysteinylated Grx-1_ HHb and Prdx2. The GS-S-protein adducts over time dominate the ESI-MS spectrum profile. CONCLUSIONS: Mass spectrometry is a unique analytical technique for probing complex reaction mechanisms associated with oxidative stress. Using model proteins, ESI-MS reveals the mechanism of DHA-facilitated S-thiolation, which consists of thiohemiketal formation, disulfide formation or S-thiolation. Furthermore, protein S-thiolation by HcySH can be reversed by reversible GSH thiol exchange. The use of mass spectrometry with in vitro models of protein S-thiolation in oxidative stress may provide significant insight into possible mechanisms of action occurring in vivo.


Assuntos
Ácido Desidroascórbico , Glutationa , Homocisteína , Espectrometria de Massas por Ionização por Electrospray/métodos , Compostos de Sulfidrila/análise , Ácido Desidroascórbico/análise , Ácido Desidroascórbico/química , Ácido Desidroascórbico/metabolismo , Glutationa/análise , Glutationa/química , Glutationa/metabolismo , Homocisteína/análise , Homocisteína/química , Homocisteína/metabolismo , Humanos , Estresse Oxidativo/fisiologia , Proteínas/análise , Proteínas/química , Proteínas/metabolismo , Compostos de Sulfidrila/química , Compostos de Sulfidrila/metabolismo , Espectrometria de Massas em Tandem/métodos
5.
Biochem Biophys Res Commun ; 497(2): 558-563, 2018 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-29438714

RESUMO

Peroxiredoxins are abundant peroxidase enzymes that are key regulators of the cellular redox environment. A major subgroup of these proteins, the typical 2-Cys peroxiredoxins, can switch between dimers and decameric or dodecameric rings, during the catalytic cycle. The necessity of this change in quaternary structure for function as a peroxidase is not fully understood. In order to explore this, human peroxiredoxin 3 (Prx3) protein was engineered to form both obligate dimers (S75E Prx3) and stabilised dodecameric rings (S78C Prx3), uncoupling structural transformations from the catalytic cycle. The obligate dimer, S75E Prx3, retained catalytic activity towards hydrogen peroxide, albeit significantly lower than the wildtype and S78C proteins, suggesting an evolutionary advantage of having higher order self-assemblies.


Assuntos
Peroxirredoxina III/química , Cristalografia por Raios X , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Estabilidade Enzimática , Humanos , Modelos Moleculares , Mutação , Peroxirredoxina III/genética , Peroxirredoxina III/metabolismo , Conformação Proteica , Multimerização Proteica
6.
J Biol Chem ; 291(6): 3053-62, 2016 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-26601956

RESUMO

Peroxiredoxin 2 (Prx2) is a thiol protein that functions as an antioxidant, regulator of cellular peroxide concentrations, and sensor of redox signals. Its redox cycle is widely accepted to involve oxidation by a peroxide and reduction by thioredoxin/thioredoxin reductase. Interactions of Prx2 with other thiols are not well characterized. Here we show that the active site Cys residues of Prx2 form stable mixed disulfides with glutathione (GSH). Glutathionylation was reversed by glutaredoxin 1 (Grx1), and GSH plus Grx1 was able to support the peroxidase activity of Prx2. Prx2 became glutathionylated when its disulfide was incubated with GSH and when the reduced protein was treated with H2O2 and GSH. The latter reaction occurred via the sulfenic acid, which reacted sufficiently rapidly (k = 500 m(-1) s(-1)) for physiological concentrations of GSH to inhibit Prx disulfide formation and protect against hyperoxidation to the sulfinic acid. Glutathionylated Prx2 was detected in erythrocytes from Grx1 knock-out mice after peroxide challenge. We conclude that Prx2 glutathionylation is a favorable reaction that can occur in cells under oxidative stress and may have a role in redox signaling. GSH/Grx1 provide an alternative mechanism to thioredoxin and thioredoxin reductase for Prx2 recycling.


Assuntos
Glutarredoxinas , Glutationa , Peroxirredoxinas , Processamento de Proteína Pós-Traducional/fisiologia , Animais , Domínio Catalítico , Linhagem Celular , Cisteína , Glutarredoxinas/química , Glutarredoxinas/genética , Glutarredoxinas/metabolismo , Glutationa/química , Glutationa/genética , Glutationa/metabolismo , Humanos , Peróxido de Hidrogênio/química , Camundongos , Camundongos Knockout , Estresse Oxidativo/efeitos dos fármacos , Peroxirredoxinas/química , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Tiorredoxina Dissulfeto Redutase/química , Tiorredoxina Dissulfeto Redutase/genética , Tiorredoxina Dissulfeto Redutase/metabolismo , Tiorredoxinas/química , Tiorredoxinas/genética , Tiorredoxinas/metabolismo
7.
Biochem J ; 473(4): 411-21, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26614766

RESUMO

Mammalian 2-cysteine peroxiredoxins (Prxs) are susceptible to hyperoxidation by excess H2O2. The cytoplasmic family member Prx2 hyperoxidizes more readily than mitochondrial Prx3 due to slower dimerization of the sulfenic acid (SpOH) intermediate. Four variant amino acids near the C-terminus have been shown to contribute to this difference. We have performed kinetic analysis of the relationship between hyperoxidation and disulfide formation, using whole-protein MS and comparing wild-type (WT) Prx2 and Prx3 with tail-swap mutants in which the four amino acids were reversed. These changes make Prx3 more sensitive and Prx2 less sensitive to hyperoxidation and accounted for ∼70% of the difference between the two proteins. The tail swap mutant of Prx3 was also more susceptible when expressed in the mitochondria of HeLa cells. The hyperoxidized product at lower excesses of H2O2 was a semi-hyperoxidized dimer with one active site disulfide and the other a sulfinic acid. For Prx2, increasing the H2O2 concentration resulted in complete hyperoxidation. In contrast, only approximately half the Prx3 active sites underwent hyperoxidation and, even with high H2O2, the predominant product was the hyperoxidized dimer. Size exclusion chromatography (SEC) showed that the oligomeric forms of all redox states of Prx3 dissociated more readily into dimeric units than their Prx2 counterparts. Notably the species with one disulfide and one hyperoxidized active site was decameric for Prx2 and dimeric for Prx3. Reduction and re-oxidation of the hyperoxidized dimer of Prx3 produced hyperoxidized monomers, implying dissociation and rearrangement of the subunits of the functional homodimer.


Assuntos
Peroxirredoxina III/metabolismo , Peroxirredoxinas/metabolismo , Sequência de Aminoácidos , Células HeLa , Humanos , Cinética , Dados de Sequência Molecular , Mutação , Oxirredução , Peroxirredoxina III/química , Peroxirredoxina III/genética , Peroxirredoxinas/química , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos
8.
FASEB J ; 28(7): 3205-10, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24636884

RESUMO

Erythrocytes require glucose-6-phosphate dehydrogenase (G6PD) to generate NADPH and protect themselves against hemolytic anemia induced by oxidative stress. Peroxiredoxin 2 (Prx2) is a major antioxidant enzyme that requires NADPH to recycle its oxidized (disulfide-bonded) form. Our aims were to determine whether Prx2 is more highly oxidized in G6PD-deficient erythrocytes and whether these cells are able to recycle oxidized Prx2 after oxidant challenge. Blood was obtained from 61 Malaysian neonates with G6PD deficiency (average 33% normal activity) and 86 controls. Prx2 redox state was analyzed by Western blotting under nonreducing conditions. Prx2 in freshly isolated blood was predominantly reduced in both groups, but the median level of oxidation was significantly higher (8 vs 3%) and the range greater for the G6PD-deficient population. When treated with reagent H2O2, the G6PD-deficient erythrocytes were severely compromised in their ability to recycle oxidized Prx2, with only 27 or 4% reduction after 1 h treatment with 0.1 or 1 mM H2O2 respectively, compared with >97% reduction in control erythrocytes. The accumulation of oxidized Prx2 in oxidatively stressed erythrocytes with common G6PD variants suggests that impaired antioxidant activity of Prx2 could contribute to the hemolysis and other complications associated with the condition.-Cheah, F.-C., Peskin, A. V., Wong, F.-L., Ithnin, A., Othman, A., Winterbourn, C. C. Increased basal oxidation of peroxiredoxin 2 and limited peroxiredoxin recycling in glucose-6-phosphate dehydrogenase deficient erythrocytes from newborn infants.


Assuntos
Eritrócitos/metabolismo , Glucose-6-Fosfato/metabolismo , Deficiência de Glucosefosfato Desidrogenase/metabolismo , Proteínas de Homeodomínio/metabolismo , Peroxirredoxinas/metabolismo , Antioxidantes/metabolismo , Eritrócitos/efeitos dos fármacos , Feminino , Humanos , Peróxido de Hidrogênio/farmacologia , Lactente , Recém-Nascido , Masculino , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos
9.
J Biol Chem ; 288(20): 14170-14177, 2013 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-23543738

RESUMO

Typical 2-Cys peroxiredoxins (Prxs) react rapidly with H2O2 to form a sulfenic acid, which then condenses with the resolving cysteine of the adjacent Prx in the homodimer or reacts with another H2O2 to become hyperoxidized. Hyperoxidation inactivates the Prx and is implicated in cell signaling. Prxs vary in susceptibility to hyperoxidation. We determined rate constants for disulfide formation and hyperoxidation for human recombinant Prx2 and Prx3 by analyzing the relative proportions of hyperoxidized and dimeric products using mass spectrometry as a function of H2O2 concentration (in the absence of reductive cycling) and in competition with catalase at a fixed concentration of H2O2. This gave a second order rate constant for hyperoxidation of 12,000 M(-1) s(-1) and a rate constant for disulfide formation of 2 s(-1) for Prx2. A similar hyperoxidation rate constant for Prx3 was measured, but its rate of disulfide formation was ~10-fold higher, making it is more resistant than Prx2 to hyperoxidation. There are two active sites within the homodimer, and at low H2O2 concentrations one site was hyperoxidized and the other present as a disulfide. Prx with two hyperoxidized sites formed progressively at higher H2O2 concentrations. Although the sulfenic acid forms of Prx2 and Prx3 are ~1000-fold less reactive with H2O2 than their active site thiols, they react several orders of magnitude faster than most reduced thiol proteins. This observation has important implications for understanding the mechanism of peroxide sensing in cells.


Assuntos
Cisteína/química , Regulação da Expressão Gênica , Peroxirredoxina III/química , Peroxirredoxinas/química , Ácidos Sulfênicos/química , Dissulfetos/química , Humanos , Peróxido de Hidrogênio/química , Modelos Químicos , Oxirredução , Oxigênio/química , Peróxidos/química , Dobramento de Proteína , Transdução de Sinais , Compostos de Sulfidrila/química
10.
Biochem J ; 453(3): 475-85, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23713588

RESUMO

Prx (peroxiredoxin) 2 protects cells from deleterious oxidative damage. It catalyses the breakdown of hydroperoxides through a highly reactive cysteine residue and has been linked to chaperone activity that promotes cell survival under conditions of oxidative stress. It may also be involved in redox signalling by binding to other proteins. In the present study we have searched for binding partners of Prx2 in H2O2-treated Jurkat and human umbilical vein endothelial cells and discovered that the hyperoxidized form selectively co-precipitated with the protein disulfide-isomerase ERp46. Mutant analyses revealed that loss of the peroxidative cysteine residue of Prx2 also facilitated complex formation with ERp46, even without H2O2 treatment, whereas the resolving cysteine residue of Prx2 was indispensible for the interaction to occur. The complex involved a stable non-covalent interaction that was disassociated by the reduction of intramolecular disulfides in ERp46, or by disruption of the decameric structure of hyperoxidized Prx2. This is the first example of a protein interaction dependent on the hyperoxidized status of a Prx.


Assuntos
Peroxirredoxinas/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , Immunoblotting , Imunoprecipitação , Células Jurkat/efeitos dos fármacos , Células Jurkat/metabolismo , Microscopia de Fluorescência , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
11.
J Biol Chem ; 286(20): 18048-55, 2011 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-21385867

RESUMO

Peroxiredoxins (Prx) are thiol peroxidases that exhibit exceptionally high reactivity toward peroxides, but the chemical basis for this is not well understood. We present strong experimental evidence that two highly conserved arginine residues play a vital role in this activity of human Prx2 and Prx3. Point mutation of either ArgI or ArgII (in Prx3 Arg-123 and Arg-146, which are ∼3-4 Šor ∼6-7 Šaway from the active site peroxidative cysteine (C(p)), respectively) in each case resulted in a 5 orders of magnitude loss in reactivity. A further 2 orders of magnitude decrease in the second-order rate constant was observed for the double arginine mutants of both isoforms, suggesting a cooperative function for these residues. Detailed ab initio theoretical calculations carried out with the high level G4 procedure suggest strong catalytic effects of H-bond-donating functional groups to the C(p) sulfur and the reactive and leaving oxygens of the peroxide in a cooperative manner. Using a guanidinium cation in the calculations to mimic the functional group of arginine, we were able to locate two transition structures that indicate rate enhancements consistent with our experimentally observed rate constants. Our results provide strong evidence for a vital role of ArgI in activating the peroxide that also involves H-bonding to ArgII. This mechanism could explain the exceptional reactivity of peroxiredoxins toward H(2)O(2) and may have wider implications for protein thiol reactivity toward peroxides.


Assuntos
Simulação por Computador , Peróxido de Hidrogênio/química , Modelos Químicos , Peroxirredoxinas/química , Substituição de Aminoácidos , Catálise , Humanos , Peróxido de Hidrogênio/metabolismo , Cinética , Peroxirredoxina III , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Mutação Puntual
12.
Chem Res Toxicol ; 25(11): 2322-32, 2012 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-23009681

RESUMO

Serotonin (5-hydroxytryptamine; 5HT) is a favorable substrate for myeloperoxidase and is likely to be oxidized by this heme enzyme during inflammation. In this study, we have investigated how serotonin becomes conjugated to amino acid residues and proteins when it is oxidized by myeloperoxidase. 5HT formed three adducts with N-acetylcysteine (NAC) when it was incubated with myeloperoxidase, xanthine oxidase, and acetaldehyde. One of the adducts was identified as 5HT-NAC, and the others were conjugates of NAC and tryptamine-4,5-dione (TD). There was no evidence for coupling of oxidized serotonin to amine residues. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was exposed to 5HT with the enzymatic system or synthetic TD. Both caused a loss of thiols on GAPDH and covalent attachment of quinones derived from TD to the protein. Biotin-labeled 5HT was used instead of 5HT to confirm the conjugation of 5HT to GAPDH. It was incorporated into the GAPDH when oxidized by myeloperoxidase. Analysis of tryptic peptides of human GAPDH by liquid chromatography with mass spectrometry revealed that an adduct of TD was formed with the peptide containing Cys(152) and Cys(156). Our results indicate that myeloperoxidase can oxidize serotonin to species that form adducts with low molecular weight thiols and cysteine residues in proteins. Low molecular weight conjugates will redox cycle and fuel oxidative stress. Conjugation of serotonin to proteins will affect their function and may provide useful biomarkers of serotonin oxidation during inflammatory events.


Assuntos
Indolquinonas/metabolismo , Peroxidase/metabolismo , Serotonina/metabolismo , Compostos de Sulfidrila/metabolismo , Triptaminas/metabolismo , Biocatálise , Radicais Livres/química , Radicais Livres/metabolismo , Humanos , Indolquinonas/química , Oxirredução , Serotonina/química , Compostos de Sulfidrila/química , Fatores de Tempo , Triptaminas/química
13.
Biochem J ; 432(2): 313-21, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20840079

RESUMO

Prxs (peroxiredoxins) are a ubiquitous family of cysteine-dependent peroxidases that react rapidly with H2O2 and alkyl hydroperoxides and provide defence against these reactive oxidants. Hydroperoxides are also formed on amino acids and proteins during oxidative stress, and they too are a potential cause of biological damage. We have investigated whether Prxs react with amino acid, peptide and protein hydroperoxides, and whether the reactions are sufficiently rapid for these enzymes to provide antioxidant protection against these oxidants. Isolated Prx2, which is a cytosolic protein, and Prx3, which resides within mitochondria, were reacted with a selection of hydroperoxides generated by γ-radiolysis or singlet oxygen, on free amino acids, peptides and proteins. Reactions were followed by measuring the accumulation of disulfide-linked Prx dimers, via non-reducing SDS/PAGE, or the loss of the corresponding hydroperoxide, using quench-flow and LC (liquid chromatography)/MS. All the hydroperoxides induced rapid oxidation, with little difference in reactivity between Prx2 and Prx3. N-acetyl leucine hydroperoxides reacted with Prx2 with a rate constant of 4 × 10(4) M-1 · s-1. Hydroperoxides present on leucine, isoleucine or tyrosine reacted at a comparable rate, whereas histidine hydroperoxides were ~10-fold less reactive. Hydroperoxides present on lysozyme and BSA reacted with rate constants of ~100 M-1 · s-1. Addition of an uncharged derivative of leucine hydroperoxide to intact erythrocytes caused Prx2 oxidation with no concomitant loss in GSH, as did BSA hydroperoxide when added to concentrated erythrocyte lysate. Prxs are therefore favoured intracellular targets for peptide/protein hydroperoxides and have the potential to detoxify these species in vivo.


Assuntos
Peroxirredoxinas/sangue , Aminoácidos/metabolismo , Dipeptídeos/metabolismo , Dipeptídeos/efeitos da radiação , Eritrócitos/enzimologia , Raios gama , Glutationa/sangue , Humanos , Peróxido de Hidrogênio/metabolismo , Isoleucina/metabolismo , Isoleucina/efeitos da radiação , Leucina/metabolismo , Leucina/efeitos da radiação , Estresse Oxidativo , Peptídeos/metabolismo , Peróxidos/metabolismo , Peroxirredoxina III , Peroxirredoxinas/isolamento & purificação , Peroxirredoxinas/metabolismo , Oxigênio Singlete/metabolismo
14.
Free Radic Biol Med ; 158: 115-125, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32702382

RESUMO

Peroxiredoxin 2 (Prdx2) and other typical 2-Cys Prdxs function as homodimers in which hydrogen peroxide oxidizes each active site cysteine to a sulfenic acid which then condenses with the resolving cysteine on the alternate chain. Previous kinetic studies have considered both sites as equally reactive. Here we have studied Prdx2 using a combination of non-reducing SDS-PAGE to separate reduced monomers and dimers with one and two disulfide bonds, and stopped flow analysis of tryptophan fluorescence, to investigate whether there is cooperativity between the sites. We have observed positive cooperativity when H2O2 is added as a bolus and oxidation of the second site occurs while the first site is present as a sulfenic acid. Modelling of this reaction showed that the second site reacts 2.2 ± 0.1 times faster. In contrast, when H2O2 was generated slowly and the first active site condensed to a disulfide before the second site reacted, no cooperativity was evident. Conversion of the sulfenic acid to the disulfide showed negative cooperativity, with modelling of the exponential rise in tryptophan fluorescence yielding a rate constant of 0.75 ± 0.08 s-1 when the alternate active site was present as a sulfenic acid and 2.29 ± 0.08-fold lower when it was a disulfide. No difference in the rate of hyperoxidation at the two sites was detected. Our findings imply that oxidation of one active site affects the conformation of the second site and influences which intermediate forms of the protein are favored under different cellular conditions.


Assuntos
Cisteína , Peroxirredoxinas , Domínio Catalítico , Cisteína/metabolismo , Peróxido de Hidrogênio , Cinética , Oxirredução , Peroxirredoxinas/metabolismo
15.
Biochemistry ; 48(42): 10175-82, 2009 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-19775156

RESUMO

During infection and inflammation, neutrophils and eosinophils produce hypochlorous acid, hypobromous acid, chloramines, and bromamines. These reactive halogen species preferentially oxidize methionine and thiols. It is commonly assumed that they convert methionine to methionine sulfoxide. However, iodine and organic chloramines are known to convert methionine to dehydromethionine, which is a cyclic azasulfonium salt. The potential for this reaction to occur in biologically relevant situations has so far been neglected. Therefore, we investigated the oxidation of methionine and N-terminal methionine residues by biologically relevant reactive halogen species and neutrophils. When hypochlorous acid reacted with methionine, two major products in addition to methionine sulfoxide were formed. They both had molecular masses two mass units lower than that of methionine and were identified as the diastereomers of dehydromethionine. Hypochlorous acid and chloramines converted methionine to a mixture of approximately 25% dehydromethionine and 75% methionine sulfoxide. Hypobromous acid and bromamines produced upward of 50% dehydromethionine. When methionine was present on the N-termini of peptides, reactive halogen species oxidized them to dehydromethionine with yields as high as 80%. Formylated N-terminal methionines and non-N-terminal methionine residues gave stoichiometric production of the corresponding sulfoxides only. Purified myeloperoxidase used hydrogen peroxide and chloride to catalyze the oxidation of N-terminal methionines to dehydromethionine. Neutrophils oxidized extracellular methionine to 30% dehydromethionine and 70% methionine sulfoxide. They also oxidized their intracellular methionine to dehydromethionine as well as methionine sulfoxide. We propose that reactive halogen species will produce dehydromethionine and form azasulfonium cations on the N-termini of peptides and proteins during inflammatory events.


Assuntos
Halogênios/química , Metionina/química , Neutrófilos/metabolismo , Tiazóis/química , Halogênios/metabolismo , Humanos , Metionina/metabolismo , Oxirredução , Peptídeos/química , Peptídeos/metabolismo , Peroxidase/metabolismo , Tiazóis/metabolismo
16.
Biochemistry ; 48(27): 6495-501, 2009 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-19462976

RESUMO

Peroxiredoxins (Prxs) are a ubiquitous family of thiol peroxidases that protect cells from peroxides and have a putative role in redox signaling. In this study, we investigated the redox properties of human Prx 3, a typical 2-Cys Prx that is localized to the mitochondrial matrix. We found that Prx 3 displayed strong reactivity with H(2)O(2), with a competitive kinetic approach generating a second order rate constant of 2 x 10(7) M(-1) s(-1). This is considerably higher than typical thiols and similar to values for other mammalian 2-Cys Prxs. In contrast, Prx 3 reacted very slowly with the thiol alkylating agents iodoacetamide and N-ethylmaleimide. Using dithiothreitol redox buffers, we measured the redox potential of Prx 3 of -290 mV. This is similar to the redox potential of mitochondrial thioredoxin 2 and is consistent with optimal operation of Prx 3 in the mitochondrial matrix.


Assuntos
Peróxidos/química , Peroxirredoxinas/química , Alquilação , Eletroforese em Gel de Poliacrilamida , Humanos , Cinética , Oxirredução , Peroxirredoxina III , Compostos de Sulfidrila/química
17.
Free Radic Biol Med ; 145: 1-7, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31521665

RESUMO

Hydrogen peroxide undergoes an equilibrium reaction with bicarbonate/CO2 to produce peroxymonocarbonate (HCO4-). Peroxymonocarbonate is more reactive with thiols than H2O2 but it makes up only a small fraction of the H2O2 in physiological bicarbonate buffers so the increase in rate of oxidation of low molecular weight thiols is modest. However, for some thiol proteins such as protein tyrosine phosphatases, the rate enhancement is very much greater. We have investigated the effect of bicarbonate/CO2 on the oxidation of peroxiredoxins (Prdxs) 2 and 3. Using an assay in which reduced Prdx2 inhibits oxidation of horseradish peroxidase by H2O2, we saw no difference between phosphate and bicarbonate buffers (pH 7.4). However, hyperoxidation of both Prdxs in bicarbonate was considerably enhanced. Hyperoxidation involves the reaction of the sulfenic acid formed at the active site with a second H2O2, and prevents its condensation to a disulfide. Using LC/MS analysis, we determined that the presence of 25 mM bicarbonate/CO2 increased the ratio of hyperoxidation compared with condensation 6-fold for Prdx2 and 11-fold for Prdx3. These results imply that Prdx hyperoxidation will occur more readily under physiological conditions than appreciated from in vitro experiments, which seldom use bicarbonate buffers. They also raise the possibility that variations in bicarbonate concentration could provide a mechanism for regulating the cellular level of active Prdxs.


Assuntos
Dióxido de Carbono/química , Peróxido de Hidrogênio/química , Peroxirredoxina III/química , Peroxirredoxinas/química , Bicarbonatos , Carbonatos , Dissulfetos/química , Peroxidase do Rábano Silvestre/química , Humanos , Cinética , Oxirredução , Peroxirredoxina III/genética , Peroxirredoxinas/genética , Ácidos Sulfênicos/química , Compostos de Sulfidrila/química
18.
Free Radic Biol Med ; 129: 383-393, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30315937

RESUMO

Hydrogen peroxide (H2O2) acts as a signaling molecule in cells by oxidising cysteine residues in regulatory proteins such as phosphatases, kinases and transcription factors. It is unclear exactly how many of these proteins are specifically targeted by H2O2 because they appear too unreactive to be directly oxidised. One proposal is that peroxiredoxins (Prxs) initially react with H2O2 and then oxidise adjacent proteins via a thiol relay mechanism. The aim of this study was to identify constitutive interaction partners of Prx2 in Jurkat T-lymphoma cells, in which thiol protein oxidation occurs at low micromolar concentrations of H2O2. Immunoprecipitation and proximity ligation assays identified a physical interaction between collapsin response mediator protein 2 (CRMP2) and cytoplasmic Prx2. CRMP2 regulates microtubule structure during lymphocyte migration and neuronal development. Exposure of Jurkat cells to low micromolar levels of H2O2 caused rapid and reversible oxidation of CRMP2, in parallel with Prx2 oxidation, despite purified recombinant CRMP2 protein reacting slowly with H2O2 (k~1 M-1s-1). Lowering Prx expression should inhibit oxidation of proteins oxidised by a relay mechanism, however knockout of Prx2 had no effect on CRMP2 oxidation. CRMP2 also interacted with Prx1, suggesting redundancy in single knockout cells. Prx 1 and 2 double knockout Jurkat cells were not viable. An interaction between Prx2 and CRMP2 was also detected in other human and rodent cells, including primary neurons. However, low concentrations of H2O2 did not cause CRMP2 oxidation in these cells. This indicates a cell-type specific mechanism for promoting CRMP2 oxidation in Jurkat cells, with insufficient evidence to attribute oxidation to a Prx-dependent redox relay.


Assuntos
Citoesqueleto de Actina/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Peróxido de Hidrogênio/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas do Tecido Nervoso/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestrutura , Animais , Movimento Celular/efeitos dos fármacos , Regulação da Expressão Gênica , Células HEK293 , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células Jurkat , Camundongos , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Oxirredução , Células PC12 , Cultura Primária de Células , Ligação Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Transdução de Sinais
19.
Free Radic Biol Med ; 103: 188-191, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28017897

RESUMO

One of the most convenient methods for measuring superoxide dismutase activity is a plate assay using xanthine oxidase and the water soluble tetrazolium WST-1. For reliable results with WST-1, certain aspects of the procedure need to be adhered to. This article describes an appropriate protocol that minimizes sources of variability.


Assuntos
Ensaios Enzimáticos/métodos , Superóxido Dismutase/química , Sais de Tetrazólio/química , Animais , Bovinos , Ensaios Enzimáticos/instrumentação , Cinética , Oxirredução , Reprodutibilidade dos Testes , Soluções
20.
Free Radic Biol Med ; 40(1): 45-53, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16337878

RESUMO

Hypochlorous acid (HOCl) and chloramines are produced by the neutrophil enzyme, myeloperoxidase. Both react readily with thiols, although chloramines differ from HOCl in discriminating between low molecular weight thiols on the basis of their pKa. Here, we have compared the reactivity of HOCl and taurine chloramine with thiol proteins by examining inactivation of creatine kinase (CK) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH). With both enzymes, loss of activity paralleled thiol loss. For CK both were complete at a 1:1 taurine chloramine:thiol mole ratio. For GAPDH each chloramine oxidized two thiols. Three times more HOCl than taurine chloramine was required for inactivation, indicating that HOCl is less thiol specific. Competition studies showed that thiols of CK were 4 times more reactive with taurine chloramine than thiols of GAPDH (rate constants of 1200 and 300 M-1s-1 respectively). These compare with 205 M-1s-1 for cysteine and are consistent with their lower pKa's. Both enzymes were equally susceptible to HOCl. GSH competed directly with the enzyme thiols for taurine chloramine and protected against oxidative inactivation. At lower GSH concentrations, mixed disulfides were formed. We propose that chloramines should preferentially attack proteins with low pKa thiols and this could be important in regulatory processes.


Assuntos
Creatina Quinase/antagonistas & inibidores , Cisteína/metabolismo , Inibidores Enzimáticos/farmacologia , Gliceraldeído-3-Fosfato Desidrogenases/antagonistas & inibidores , Ácido Hipocloroso/farmacologia , Oxidantes/farmacologia , Taurina/análogos & derivados , Animais , Ligação Competitiva , Creatina Quinase/metabolismo , Dissulfetos/metabolismo , Glutationa/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Metionina/metabolismo , Músculos/enzimologia , Oxirredução , Coelhos , Compostos de Sulfidrila/metabolismo , Taurina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA