Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Antioxidants (Basel) ; 10(11)2021 Oct 20.
Article in English | MEDLINE | ID: mdl-34829517

ABSTRACT

Lactoperoxidase (LPO, FeIII in its resting state in the absence of substrates)-an enzyme secreted from human mammary, salivary, and other mucosal glands-catalyzes the oxidation of thiocyanate (SCN-) by hydrogen peroxide (H2O2) to produce hypothiocyanite (OSCN-), which functions as an antimicrobial agent. The accepted catalytic mechanism, called the halogen cycle, comprises a two-electron oxidation of LPO by H2O2 to produce oxoiron(IV) radicals, followed by O-atom transfer to SCN-. However, the mechanism does not explain biphasic kinetics and inhibition by H2O2 at low concentration of reducing substrate, conditions that may be biologically relevant. We propose an ordered sequential mechanism in which the order of substrate binding is reversed, first SCN- and then H2O2. The sequence of substrate binding that is described by the halogen cycle mechanism is actually inhibitory.

2.
Antioxidants (Basel) ; 5(4)2016 Nov 22.
Article in English | MEDLINE | ID: mdl-27879667

ABSTRACT

There are no two main-group elements that exhibit more similar physical and chemical properties than sulfur and selenium. Nonetheless, Nature has deemed both essential for life and has found a way to exploit the subtle unique properties of selenium to include it in biochemistry despite its congener sulfur being 10,000 times more abundant. Selenium is more easily oxidized and it is kinetically more labile, so all selenium compounds could be considered to be "Reactive Selenium Compounds" relative to their sulfur analogues. What is furthermore remarkable is that one of the most reactive forms of selenium, hydrogen selenide (HSe- at physiologic pH), is proposed to be the starting point for the biosynthesis of selenium-containing molecules. This review contrasts the chemical properties of sulfur and selenium and critically assesses the role of hydrogen selenide in biological chemistry.

3.
Arch Biochem Biophys ; 564: 1-11, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25172223

ABSTRACT

Human defensive peroxidases, including lactoperoxidase (LPO) and myeloperoxidase (MPO), are capable of catalyzing the oxidation of halides (X(-)) by H2O2 to give hypohalous acids (HOX) for the purpose of cellular defense. Substrate selectivity depends upon the relative abundance of the halides, but the pseudo-halide thiocyanate (SCN(-)) is a major substrate, and sometimes the exclusive substrate, of all defensive peroxidases in most physiologic fluids. The resulting hypothiocyanous acid (HOSCN) has been implicated in cellular damage via thiol oxidation. While thiols are believed to be the primary target of HOSCN in vivo, Trp residues have also been implicated as targets for HOSCN. However, the mechanism involved in HOSCN-mediated Trp oxidation was not established. Trp residues in proteins appeared to be susceptible to oxidation by HOSCN, whereas free Trp and Trp residues in small peptides were found to be unreactive. We show that HOSCN-induced Trp oxidation is dependent on pH, with oxidation of free Trp, and Trp-containing peptides observed when the pH is below 2. These conditions mimic those employed previously to precipitate proteins after treatment with HOSCN, which accounts for the discrepancy in the results reported for proteins versus free Trp and small peptides. The reactant in these cases may be thiocyanogen ((SCN)2), which is produced by comproportionation of HOSCN and SCN(-) at low pH. Reaction of thiocyanate-derived oxidants with protein Trp residues at low pH results in the formation of a number of oxidation products, including mono- and di-oxygenated derivatives, which are also formed with other hypohalous acids. Our data suggest that significant modification of Trp by HOSCN in vivo is likely to have limited biological relevance.


Subject(s)
Oxidants/chemistry , Peptides/chemistry , Thiocyanates/chemistry , Tryptophan/chemistry , Humans , Hydrogen-Ion Concentration , Oxidants/metabolism , Oxidation-Reduction , Peptides/metabolism , Thiocyanates/metabolism , Tryptophan/metabolism
4.
Org Lett ; 14(13): 3248-51, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22690827

ABSTRACT

Host-guest complexation between the dicationic viologen 1-tri(ethylene glycol)-1'-methyl-m-xylyl-4,4'-bipyridinium and cucurbit[7]uril (CB7) was studied at pH = 4.5 in water. The stability constants of the mono- and bis-CB7 adducts were determined at 25 °C by UV-vis spectroscopy. Stopped-flow kinetic experiments were performed to measure the formation and dissociation rate constants of the monoadduct: k(1) = (6.01 ± 0.03) × 10(6) M(-1)s(-1) and k(-1) = 52.7 ± 0.4 s(-1), respectively. Possible mechanisms of complexation are discussed in view of the kinetic results.


Subject(s)
Bridged-Ring Compounds/chemistry , Imidazoles/chemistry , Viologens/chemistry , Kinetics , Models, Molecular , Molecular Structure
5.
Free Radic Biol Med ; 53(1): 172-81, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22580125

ABSTRACT

The free-radical-operated mechanism of death of activated macrophages at sites of inflammation is unclear, but it is important to define it in order to find targets to prevent further tissue dysfunction. A well-defined model of macrophage activation at sites of inflammation is the treatment of RAW 264.7 cells with lipopolysaccharide (LPS), with the resulting production of reactive oxygen species (ROS). ROS and other free radicals can be trapped with the nitrone spin trap 5,5-dimethyl-1-pyrroline N-oxide (DMPO), a cell-permeable probe with antioxidant properties, which thus interferes with free-radical-operated oxidation processes. Here we have used immuno-spin trapping to investigate the role of free-radical-operated protein oxidation in LPS-induced cytotoxicity in macrophages. Treatment of RAW 264.7 cells with LPS resulted in increased ROS production, oxidation of proteins, cell morphological changes and cytotoxicity. DMPO was found to trap protein radicals to form protein-DMPO nitrone adducts, to reduce protein carbonyls, and to block LPS-induced cell death. N-Acetylcysteine (a source of reduced glutathione), diphenyleneiodonium (an inhibitor of NADPH oxidase), and 2,2'-dipyridyl (a chelator of Fe(2+)) prevented LPS-induced oxidative stress and cell death and reduced DMPO-nitrone adduct formation, suggesting a critical role of ROS, metals, and protein-radical formation in LPS-induced cell cytotoxicity. We also determined the subcellular localization of protein-DMPO nitrone adducts and identified some candidate proteins for DMPO attachment by LC-MS/MS. The LC-MS/MS data are consistent with glyceraldehyde-3-phosphate dehydrogenase, one of the most abundant, sensitive, and ubiquitous proteins in the cell, becoming labeled with DMPO when the cell is primed with LPS. This information will help find strategies to treat inflammation-associated tissue dysfunction by focusing on preventing free radical-operated proteotoxic stress and death of macrophages.


Subject(s)
Free Radicals/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/pathology , Oxidative Stress/drug effects , Protein Carbonylation/drug effects , Proteins/chemistry , Acetylcysteine/pharmacology , Animals , Blotting, Western , Cell Death/drug effects , Cells, Cultured , Cyclic N-Oxides/pharmacology , Free Radical Scavengers/pharmacology , Immunoenzyme Techniques , Immunoprecipitation , Inflammation/chemically induced , Inflammation/prevention & control , Macrophages/metabolism , Mice , Nitrogen Oxides/metabolism , Oxidation-Reduction , Proteins/metabolism , Reactive Oxygen Species/metabolism , Spin Labels , Spin Trapping , Tandem Mass Spectrometry
6.
J Am Chem Soc ; 133(49): 19911-21, 2011 Dec 14.
Article in English | MEDLINE | ID: mdl-22031955

ABSTRACT

Relatively little is known about the reaction chemistry of the human defense factor hypothiocyanite (OSCN(-)) and its conjugate acid hypothiocyanous acid (HOSCN), in part because of their instability in aqueous solutions. Herein we report that HOSCN/OSCN(-) can engage in a cascade of pH- and concentration-dependent comproportionation, disproportionation, and hydrolysis reactions that control its stability in water. On the basis of reaction kinetic, spectroscopic, and chromatographic methods, a detailed mechanism is proposed for the decomposition of HOSCN/OSCN(-) in the range of pH 4-7 to eventually give simple inorganic anions including CN(-), OCN(-), SCN(-), SO(3)(2-), and SO(4)(2-). Thiocyanogen ((SCN)(2)) is proposed to be a key intermediate in the hydrolysis; and the facile reaction of (SCN)(2) with OSCN(-) to give NCS(═O)SCN, a previously unknown reactive sulfur species, has been independently investigated. The mechanism of the aqueous decomposition of (SCN)(2) around pH 4 is also reported. The resulting mechanistic models for the decomposition of HOSCN and (SCN)(2) address previous empirical observations, including the facts that the presence of SCN(-) and/or (SCN)(2) decreases the stability of HOSCN/OSCN(-), that radioisotopic labeling provided evidence that under physiological conditions decomposing OSCN(-) is not in equilibrium with (SCN)(2) and SCN(-), and that the hydrolysis of (SCN)(2) near neutral pH does not produce OSCN(-). Accordingly, we demonstrate that, during the human peroxidase-catalyzed oxidation of SCN(-), (SCN)(2) cannot be the precursor of the OSCN(-) that is produced.


Subject(s)
Thiocyanates/chemistry , Anions/chemistry , Humans , Hydrogen-Ion Concentration , Hydrolysis , Spectrum Analysis , Water/chemistry
7.
J Biol Chem ; 285(26): 20062-71, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20406811

ABSTRACT

Myeloperoxidase (MPO) released by activated neutrophils can initiate and promote carcinogenesis. MPO produces hypochlorous acid (HOCl) that oxidizes the genomic DNA in inflammatory cells as well as in surrounding epithelial cells. DNA-centered radicals are early intermediates formed during DNA oxidation. Once formed, DNA-centered radicals decay by mechanisms that are not completely understood, producing a number of oxidation products that are studied as markers of DNA oxidation. In this study we employed the 5,5-dimethyl-1-pyrroline N-oxide-based immuno-spin trapping technique to investigate the MPO-triggered formation of DNA-centered radicals in inflammatory and epithelial cells and to test whether resveratrol blocks HOCl-induced DNA-centered radical formation in these cells. We found that HOCl added exogenously or generated intracellularly by MPO that has been taken up by the cell or by MPO newly synthesized produces DNA-centered radicals inside cells. We also found that resveratrol passed across cell membranes and scavenged HOCl before it reacted with the genomic DNA, thus blocking DNA-centered radical formation. Taken together our results indicate that the formation of DNA-centered radicals by intracellular MPO may be a useful point of therapeutic intervention in inflammation-induced carcinogenesis.


Subject(s)
DNA Adducts/chemistry , DNA/chemistry , Free Radicals/chemistry , Peroxidase/metabolism , Animals , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Cattle , Cell Line , Cell Line, Tumor , Coculture Techniques , Cyclic N-Oxides/chemistry , Cyclic N-Oxides/metabolism , DNA/genetics , DNA/metabolism , DNA Adducts/metabolism , Free Radicals/metabolism , Glutathione/pharmacology , HL-60 Cells , Halogenation/drug effects , Humans , Hydrogen Peroxide/pharmacology , Hypochlorous Acid/chemistry , Hypochlorous Acid/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Oxidants/pharmacology , Oxidation-Reduction/drug effects , Resveratrol , Stilbenes/pharmacology
8.
Biochemistry ; 49(9): 2068-74, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20085320

ABSTRACT

Thiocyanate reacts noncatalytically with myeloperoxidase-derived HOCl to produce hypothiocyanite (OSCN(-)), thereby potentially limiting the propensity of HOCl to inflict host tissue damage that can lead to inflammatory diseases. However, the efficiency with which SCN(-) captures HOCl in vivo depends on the concentration of SCN(-) relative to other chemical targets. In blood plasma, where the concentration of SCN(-) is relatively low, proteins may be the principal initial targets of HOCl, and chloramines are a significant product. Chloramines eventually decompose to irreversibly damage proteins. In the present study, we demonstrate that SCN(-) reacts efficiently with chloramines in small molecules, in proteins, and in Escherichia coli cells to give OSCN(-) and the parent amine. Remarkably, OSCN(-) reacts faster than SCN(-) with chloramines. These reactions of SCN(-) and OSCN(-) with chloramines may repair some of the damage that is inflicted on protein amines by HOCl. Our observations are further evidence for the importance of secondary reactions during the redox cascades that are associated with oxidative stress by hypohalous acids.


Subject(s)
Anti-Infective Agents/pharmacology , Chloramines/metabolism , Macromolecular Substances/metabolism , Thiocyanates/metabolism , Anti-Infective Agents/metabolism , Chloramines/chemistry , Chlorine , Disinfection , Escherichia coli/drug effects , Escherichia coli/growth & development , Humans , Hypochlorous Acid/metabolism , Oxidation-Reduction , Spectrophotometry, Ultraviolet , Taurine/analogs & derivatives , Taurine/metabolism , Thiocyanates/chemistry , Thiocyanates/pharmacology , Trinitrobenzenesulfonic Acid , Ubiquitin/metabolism
9.
J Phys Chem A ; 114(4): 1670-6, 2010 Feb 04.
Article in English | MEDLINE | ID: mdl-20039719

ABSTRACT

The mechanism of oxidation of ArSO(2)(-) (PhSO(2)(-) and 5-sulfinato-2-nitrobenzoic acid = TNBO(2)(1-/2-)) with HOCl/OCl(-) has been investigated using the kinetic method. In contrast to previous reports for PhSO(2)(-) (for which it was suggested that OCl(-) and not HOCl was the reactant), the reaction proceeds through a conventional pathway: nucleophilic attack by ArSO(2)(-) on HOCl with concomitant Cl(+) transfer to give a sulfonyl chloride intermediate (ArSO(2)Cl), which we have identified spectrophotometrically. Remarkably, the rate constant for the reaction of HOCl with ArSO(2)(-) is on the order of 10(9) M(-1) s(-1), larger than the rate constants for corresponding thiolates, and is nearly diffusion-controlled. In contrast, the rate constant for the reaction of OCl(-) with ArSO(2)(-) is approximately 7 orders of magnitude smaller.

10.
Biochemistry ; 48(46): 11142-8, 2009 Nov 24.
Article in English | MEDLINE | ID: mdl-19839600

ABSTRACT

Electrophilic halogenating agents, including hypohalous acids and haloamines, oxidize free methionine and the N-terminal methionines of peptides and proteins (e.g., Met-1 of anti-inflammatory peptide 1 and ubiquitin) to produce dehydromethionine (a five-membered isothiazolidinium heterocycle). Amide derivatives of methionine are oxidized to the corresponding sulfoxide derivatives under the same reaction conditions (e.g., Met-3 of anti-inflammatory peptide 1). Other biological oxidants, including hydrogen peroxide and peroxynitrite, also produce only the corresponding sulfoxides. Hypothiocyanite does not react with methionine residues. We suggest that dehydromethionine may be a useful biomarker for the myeloperoxidase-induced oxidative stress associated with many inflammatory diseases.


Subject(s)
Hypochlorous Acid/chemistry , Methionine/chemistry , Neutrophils/metabolism , Oxidative Stress , Proteins/chemistry , Thiazoles/chemistry , Biomarkers/analysis , Biomarkers/chemistry , Glutathione/analogs & derivatives , Glutathione/chemistry , Glutathione Disulfide/chemistry , Hydrogen Peroxide/chemistry , Magnetic Resonance Spectroscopy , Methionine/analogs & derivatives , Models, Biological , Models, Chemical , Neutrophils/enzymology , Oxidants/chemistry , Oxidation-Reduction , Peptide Fragments/chemistry , Peroxidase/metabolism , Spectrometry, Mass, Electrospray Ionization , Sulfones/chemistry , Thiazoles/analysis , Ubiquitin/chemistry , Uteroglobin/chemistry
11.
Microbiology (Reading) ; 155(Pt 11): 3691-3700, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19684069

ABSTRACT

Streptococcus is a dominant genus in the human oral cavity, making up about 20 % of the more than 800 species of bacteria that have been identified, and about 80 % of the early biofilm colonizers. Oral streptococci include both health-compatible (e.g. Streptococcus gordonii and Streptococcus sanguinis) and pathogenic strains (e.g. the cariogenic Streptococcus mutans). Because the streptococci have similar metabolic requirements, they have developed defence strategies that lead to antagonism (also known as bacterial interference). S. mutans expresses bacteriocins that are cytotoxic toward S. gordonii and S. sanguinis, whereas S. gordonii and S. sanguinis differentially produce H(2)O(2) (under aerobic growth conditions), which is relatively toxic toward S. mutans. Superimposed on the inter-bacterial combat are the effects of the host defensive mechanisms. We report here on the multifarious effects of bovine lactoperoxidase (bLPO) on the antagonism between S. gordonii and S. sanguinis versus S. mutans. Some of the effects are apparently counterproductive with respect to maintaining a health-compatible population of streptococci. For example, the bLPO system (comprised of bLPO+SCN(-)+H(2)O(2)) destroys H(2)O(2), thereby abolishing the ability of S. gordonii and S. sanguinis to inhibit the growth of S. mutans. Furthermore, bLPO protein (with or without its substrate) inhibits bacterial growth in a biofilm assay, but sucrose negates the inhibitory effects of the bLPO protein, thereby facilitating adherence of S. mutans in lieu of S. gordonii and S. sanguinis. Our findings may be relevant to environmental pressures that select early supragingival colonizers.


Subject(s)
Antibiosis , Lactoperoxidase/metabolism , Streptococcus gordonii/growth & development , Streptococcus mutans/growth & development , Streptococcus sanguis/growth & development , Animals , Biofilms/growth & development , Catalase/metabolism , Cattle , Hydrogen Peroxide/metabolism , Mouth/microbiology , Thiocyanates/metabolism
12.
Chem Res Toxicol ; 22(9): 1622-8, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19705807

ABSTRACT

The chief sources of cyanide (CN(-)) in humans are tobacco and occupationally derived smoke, inflammation [vis-a-vis myeloperoxidase (MPO)-induced chlorination of glycine], and microbial cyanogenesis (including Pseudomonas aeruginosa infection of the cystic fibrosis lung). The human mucosae of healthy individuals are usually protected from infection by innate defense mechanisms that include the defensive peroxidase systems. In the oral cavity, salivary peroxidase and MPO catalyze the oxidation of the pseudohalide thiocyanate (SCN(-)) by hydrogen peroxide to produce the antimicrobial hypothiocyanite (OSCN(-)). Lactoperoxidase carries out the same reaction in the human lung (as does MPO during inflammatory response). In the present study, we show that OSCN(-) and CN(-) react with pH-dependent kinetics to produce SCN(-) and cyanate (OCN(-)) via dicyanosulfide (NCSCN), with the maximum rate occurring near neutral, physiological pH. In addition to presenting a detailed chemical mechanism, we discuss unresolved issues, including the possible biological relevance of the NCSCN intermediate.


Subject(s)
Cyanides/chemistry , Nitriles/chemistry , Sulfides/chemistry , Sulfur/metabolism , Thiocyanates/chemistry , Water/chemistry , Glutathione/chemistry , Humans , Hydrogen-Ion Concentration , Kinetics , Lung/enzymology , Nitriles/metabolism , Peroxidase/metabolism , Smoking , Sulfides/metabolism , Sulfur/chemistry
13.
Chem Res Toxicol ; 21(11): 2120-6, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18942795

ABSTRACT

Hypothiocyanite (OSCN(-)) is a putative antimicrobial that is produced by defensive human peroxidases, including salivary peroxidase, lactoperoxidase, eosinophil peroxidase, and myeloperoxidase. The reaction of OSCN(-) with cysteine-derived sulfhydryl groups is believed to be involved in the antimicrobial mechanism of action. Hypothiocyanite decomposes via an unknown mechanism that involves multiple redox and hydrolytic steps to eventually yield mixtures of CN(-)/OCN(-) and SO(3)(2-)/SO(4)(2-). Until recently, no information was available regarding the chemical nature of the intermediate(s) that are produced during the chemical cascade, but we have shown that OSCN(-) undergoes hydrolysis to give the new compound thiocarbamate-S-oxide, H(2)NC(=O)SO(-). In the present paper, we demonstrate that H(2)NC(=O)SO(-) reacts with cysteine (CySH) via a two-step mechanism that is analogous to that of OSCN(-) with cysteine: HOZ + CySH --> CySZ + H(2)O and CySZ + CySH --> CySSCy + HZ, where Z = SCN and SC(=O)NH(2), respectively. The kinetics and mechanism of both steps of the reaction of H(2)NC(=O)SO(-) with CySH have been investigated as a function of pH.


Subject(s)
Cysteine/chemistry , Thiocarbamates/chemistry , Hydrogen-Ion Concentration , Kinetics , Oxides/chemistry , Thiocyanates/chemistry
14.
Chem Res Toxicol ; 21(5): 1011-6, 2008 May.
Article in English | MEDLINE | ID: mdl-18447396

ABSTRACT

Reduced glutathione (GSH) is a cornerstone of the antioxidant stratagem for eukaryotes and some prokaryotes. Hypochlorous acid (HOCl), which is produced by neutrophilic myeloperoxidase, reacts rapidly with excess GSH to yield mainly oxidized glutathione (GSSG). GSSG can be further oxidized to give first N-chloro derivatives and, later, higher oxidation states at the S centers. Under certain conditions, another major species that is observed during the oxidation of GSH by HOCl (and a minor species for other oxidants) exhibits a molecular mass that is 30 mass units heavier than GSH. This GSH+2O-2H species, which has been employed as a biomarker for oxidative stress, has been previously proposed to be a sulfonamide. Employing NMR spectroscopy and mass spectrometry, we demonstrate that the GSH+2O-2H species is indeed a nine-membered cyclic sulfonamide. Alternative formulations, including six-membered 1,2,5-oxathiazine heterocycles, have been ruled out. Remarkably, the sulfonamide exists as a 2:1 equilibrium mixture of two diastereomers. Isotope tracer studies have demonstrated that it is the Glu C alpha center that has undergone racemization. It is proposed that the racemization takes place via an acyclic imine-sulfinic acid intermediate. The glutathione sulfonamides are stable products of GSH that have been detected in physiological systems. Elucidation of the structures of the glutathione sulfonamides provides further impetus to explore their potential as biomarkers of hypochlorous acid formation.


Subject(s)
Glutathione/chemistry , Hypochlorous Acid/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Molecular , Molecular Structure , Oxidation-Reduction/drug effects , Oxidative Stress
15.
J Org Chem ; 73(8): 3017-23, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18351774

ABSTRACT

The kinetics and mechanism of the hydrolysis of cysteine sulfenyl thiocyanate (CySSCN) to give cysteine thiosulfinate ester (CyS(=O)SCy) have been investigated between pH 0 and 4. The reaction is reversible. The hydrolysis of CySSCN is second-order in [CySSCN] and inverse first-order in [H+] and [SCN-]. The following mechanism is proposed for the hydrolysis of CySSCN (where the charge depends upon the pH): CySSCN0/+ + H2O <==>CySOH0/+ + SCN- + H+, CySOH0/+ + CySSCN0/+ --> CyS(=O)SCy0/+/2+ + SCN- + H+; k1 = 3.36 +/- 0.01 x 10-3 s-1, K1k2 = 0.13 +/- 0.05 Ms-1 (which yields k2/k-1 = 39 M). The observed rate law rules out alternative mechanisms for 1 0.4 M). The following mechanism is proposed: CyS(=O)SCy2+ + H+ <==> CyS(OH)=SCy3+, Ka; CyS(OH)SCy3+ + SCN- --> CySOH+ + CySSCN+, k-2 = 0.239 +/- 0.007 M-2s-1/Ka M-1. Since cysteine sulfenic acids are known to play an important function in many enzymes, and SCN- exists in abundance in physiologic fluids, we discuss the possible role of sulfenyl thiocyanates in vivo.


Subject(s)
Acids/chemistry , Cystine/chemistry , Esters/chemistry , Sulfur/chemistry , Thiocyanates/chemistry , Thiosulfonic Acids/chemistry , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Mercaptoethanol/chemistry , Solutions
16.
J Am Chem Soc ; 129(51): 15756-7, 2007 Dec 26.
Article in English | MEDLINE | ID: mdl-18052381

ABSTRACT

Hypothiocyanite (OSCN-) hydrolyzes under alkaline conditions to give thiocarbamate-S-oxide (H2NC(=O)SO-, the conjugate base of carbamothioperoxoic acid) via a mechanism that involves rate-limiting nucleophilic attack of OH- on OSCN-, followed by fast protonation (with no net consumption of H+/OH- at pH 11.7). Thiocarbamate-S-oxide has been characterized by 13C NMR, 15N NMR, UV spectroscopy, and ion chromatography. It has also been independently synthesized by the reaction of thiocarbamate (H2NC(=O)S-) and hypochlorite (OCl-). The properties of thiocarbamate-S-oxide that is produced by hydrolysis of OSCN- and by oxidation of H2NC(=O)S- are the same. The possible relevance of thiocarbamate-S-oxide in human peroxidase defense mechanisms remains to be explored.


Subject(s)
Sulfur/chemistry , Thiocarbamates/chemistry , Thiocyanates/chemistry , Chromatography, Liquid , Hydrolysis , Magnetic Resonance Spectroscopy , Spectrophotometry, Ultraviolet
17.
J Am Chem Soc ; 129(45): 14082-91, 2007 Nov 14.
Article in English | MEDLINE | ID: mdl-17939659

ABSTRACT

Cysteine sulfenic acid has been generated in alkaline aqueous solution by oxidation of cysteine with hypohalous acid (HOX, X = Cl or Br). The kinetics and mechanisms of the oxidation reaction and the subsequent reactions of cysteine sulfenic acid have been studied by stopped-flow spectrophotometry between pH 10 and 14. Two reaction pathways were observed: (1) below pH 12, the condensation of two sulfenic acids to give cysteine thiosulfinate ester followed by the nucleophilic attack of cysteinate on cysteine thiosulfinate ester and (2) above pH 10, a pH-dependent fast equilibrium protonation of cysteine sulfenate that is followed by rate-limiting comproportionation of cysteine sulfenic acid with cysteinate to give cystine. The observation of the first reaction suggests that the condensation of cysteine sulfenic acid to give cysteine thiosulfinate ester can be competitive with the reaction of cysteine sulfenic acid with cysteine.


Subject(s)
Bromates/chemistry , Cysteine/analogs & derivatives , Cysteine/chemistry , Hypochlorous Acid/chemistry , Sulfenic Acids/chemical synthesis , Cysteine/chemical synthesis , Hydrogen-Ion Concentration , Kinetics , Oxidation-Reduction , Sulfenic Acids/chemistry , Time Factors
18.
J Org Chem ; 72(23): 8838-46, 2007 Nov 09.
Article in English | MEDLINE | ID: mdl-17941676

ABSTRACT

The kinetics and mechanisms of the reaction of cysteine with cysteine thiosulfinate ester in aqueous solution have been studied by stopped-flow spectrophotometry between pH 6 and 14. Two reaction pathways were observed for pH > 12: (1) an essentially pH-independent nucleophilic attack of cysteinate on cysteine thiosulfinate ester, and (2) a pH-dependent fast equilibrium protonation of cysteine sulfenate that is followed by rate-limiting comproportionation of cysteine sulfenic acid with cysteinate to give cystine. For 6 < pH < 12, the rate-determining reaction between cysteinate and cysteine thiosulfinate ester becomes pH-dependent due to the protonation of their amine groups. Hydrolysis of cysteine thiosulfinate ester does not play a role in the aforementioned mechanisms because the rate-determining nucleophilic attack by hydroxide is relatively slow.


Subject(s)
Cysteine/analogs & derivatives , Cysteine/chemistry , Esters/chemistry , Sulfinic Acids/chemistry , Cysteine/chemical synthesis , Hydrogen-Ion Concentration , Kinetics , Molecular Structure
19.
Chem Res Toxicol ; 20(9): 1364-72, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17764150

ABSTRACT

The kinetics and mechanisms of the hydrolysis of cysteine thiosulfinate ester (CyS(O)SCy ( x- ), x = 0-2) have been investigated by stopped-flow spectrophotometry between pH 6 and pH 14. The rate-limiting reaction of hydroxide is observed for pH < 13. More complicated kinetics are observed above pH 13, where the hydrolysis of CyS(O)SCy (2-) can be fast relative to subsequent reactions. The eventual products of hydrolysis are a 1:1 molar ratio of cystine (CySSCy) and cysteine sulfinic acid (CySO 2H) under all reaction conditions. The rate of hydrolysis is dependent upon the proton state of CyS(O)SCy ( x- ). Furthermore, cysteine thiosulfonate ester (CyS(O) 2SCy) was observed as an intermediate during the hydrolysis of CyS(O)SCy ( x- ) at lower pH. CyS(O) 2SCy eventually hydrolyzes to give stoichiometric amounts of CySSCy and CySO 2H. However, CySO 2H is observed under some conditions for which hydrolysis of CyS(O) 2SCy is relatively slow, thus suggesting multiple hydrolysis pathways for CyS(O)SCy ( x- ). The mechanism up to the rate-limiting step is proposed to be as follows: CyS(O)SCy (0) = H (+) + CyS(O)SCy (-), p K a3 = 7.32; CyS(O)SCy (-) = H (+) + CyS(O)SCy (2-), p K a4 = 7.92; CyS(O)SCy (0) + OH (-) --> products, P 0 k 0 = (5.0 +/- 0.01) x 10 (3) M (-1) s (-1); CyS(O)SCy (-) + OH (-) --> products, P 1 k 1 = 60 +/- 18 M (-1) s (-1); and CyS(O)SCy (2-) + OH (-) --> products, P 2 k 2 = 0.36 +/- 0.01 M (-1) s (-1), where P x is a constant (1

Subject(s)
Cysteine/chemistry , Sulfur/chemistry , Thiosulfonic Acids/chemistry , Cysteine/chemical synthesis , Disulfides , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Lactams/chemistry , Macrolides/chemistry , Molecular Structure , Oxidation-Reduction , Sulfinic Acids/chemistry , Thiazoles/chemistry , Thiones/chemistry
20.
Inorg Chem ; 46(1): 285-92, 2007 Jan 08.
Article in English | MEDLINE | ID: mdl-17198438

ABSTRACT

The kinetics of comproportionation of hypothiocyanous acid (HOSCN) and thiocyanate (SCN-) to give thiocyanogen ((SCN)2) in acidic aqueous solutions have been determined by double-mixing stopped-flow UV spectroscopy. Hypothiocyanite (OSCN-) was generated at pH 13 by oxidation of excess SCN- with hypobromite (OBr-), followed by a pH jump to acidic conditions ([H+] = 0.20-0.46 M). The observed pseudo-first-order rate constants exhibit first-order dependencies on [H+] and [SCN-] with overall third-order kinetics. The corresponding kinetics of hydrolysis of (SCN)2 have also been examined. Under conditions of high (and constant) [H+] and [SCN-], the kinetics exhibit second-order behavior with respect to [(SCN)2] and complex inverse dependences on [H+] and [SCN-]. Under conditions of low [H+] and [SCN-], the kinetics exhibit first-order behavior with respect to [(SCN)2] and independence with respect to [H+] and [SCN-]. We attribute this behavior to a shift in the rate-limiting step from disproportionation of HOSCN (second-order dependency on [(SCN)2]) to rate-limiting hydrolysis (first-order dependency on [(SCN)2]). Thus, we have determined the following equilibrium constant by the kinetic method: (SCN)2 + H2O HOSCN + SCN- + H+; Khyd = [HOSCN][SCN-][H+]/[(SCN)2] = khyd/kcomp = 19.8(+/-0.7) s-1/ 5.14(+/-0.07) x 103 M-2 s-1 = 3.9 x 10-3 M2.


Subject(s)
Thiocyanates/chemistry , Water/chemistry , Hydrogen-Ion Concentration , Kinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...