Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
ACS Chem Biol ; 16(8): 1413-1424, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34374506

ABSTRACT

This report characterizes and quantifies endogenous hydrogen sulfide (H2S) and small oxoacids of sulfur (SOS = HOSH, HOSOH) in a panel of cell lines including human cancer (A375 melanoma cells, HeLa cervical cells) and noncancer (HEK293 embryonic kidney cells), as well as E. coli DH5α and S. cerevisiae S288C. The methodology used is a translation of well-studied nucleophilic and electrophilic traps for cysteine and oxidized cysteines residues to target small molecular weight sulfur species; mass spectrometric analysis allows for species quantification. The observed intracellular concentrations of H2S and SOS vary in different cell types, from nanomolar to femtomolar, typically with H2S > HOSOH > HOSH. We propose the term sulfome, a subset of the metabolome, describing the nonproteinaceous metabolites of H2S; the sulfomic index is as a measure of the S-oxide redox status, which gives a profile of endogenous sulfur at different oxidation states. An important observation is that H2S and SOS were found to be continuously extruded into surrounding media against a concentration gradient, implying an active efflux process. Small molecule inhibition of several H2S generating enzymes suggest that SOS are not derived solely from H2S oxidation. Even after successful inhibition of H2S production, cells maintain constant efflux and repopulate H2S and SOS over time. This work proves that these small sulfur oxoacids are generated in cells of all types, and their efflux implies that they play a role in cell signaling and possibly other vascular physiology attributed to H2S.


Subject(s)
Hydrogen Sulfide/metabolism , Sulfenic Acids/metabolism , Cell Line, Tumor , Escherichia coli/physiology , HEK293 Cells , Humans , Hydrogen Sulfide/analysis , Metabolome/physiology , Saccharomyces cerevisiae/physiology , Sulfenic Acids/analysis
2.
Drug Metab Dispos ; 48(8): 698-707, 2020 08.
Article in English | MEDLINE | ID: mdl-32499340

ABSTRACT

The mass balance, excretion, and metabolism of LY3202626 were determined in healthy subjects after oral administration of a single dose of 10 mg of (approximately 100 µCi) [14C]LY3202626. Excretion of radioactivity was slow and incomplete, with approximately 75% of the dose recovered after 504 hours of sample collection. The mean total recovery of the radioactive dose was 31% and 44% in the feces and urine, respectively. Because of low plasma total radioactivity, plasma metabolite profiling was conducted by accelerator mass spectrometry. Metabolism of LY3202626 occurred primarily via O-demethylation (M2) and amide hydrolysis (M1, M3, M4, and M5). Overall, parent drug, M1, M2, and M4 were the largest circulating components in plasma, and M2 and M4 were the predominant excretory metabolites. The slow elimination of total radioactivity was proposed to result from an unusual enterohepatic recirculation pathway involving microbial reduction of metabolite M2 to M16 in the gut and reabsorption of M16, followed by hepatic oxidation of M16 back to M2. Supporting in vitro experiments showed that M2 is reduced to M16 anaerobically in fecal homogenate and that M16 is oxidized in the liver by aldehyde oxidase to M2. LY3202626 also showed a potential to form a reactive sulfenic acid intermediate. A portion of plasma radioactivity was unextractable and presumably bound covalently to plasma proteins. In vitro incubation of LY3202626 in human liver microsomes in the presence of NADPH with dimedone as a trapping agent implicated the formation of the proposed sulfenic acid intermediate. SIGNIFICANCE STATEMENT: The excretion of radioactivity in humans after oral administration of a single dose of 10 mg of [14C]LY3202626 was very slow. The results from in vitro experiments suggested that an interplay between microbial reduction, reabsorption, and aldehyde oxidase oxidation (M2 → M16 → M2) could be a reason for extended radioactivity excretion profile. In vitro metabolism also showed that LY3202626 has the potential to form a reactive sulfenic acid intermediate that could potentially covalently bind to plasma protein and result in the observed unextractable radioactivity from plasma.


Subject(s)
Aldehyde Oxidase/metabolism , Gastrointestinal Microbiome/physiology , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Pyrazines/pharmacokinetics , Pyrroles/pharmacokinetics , Administration, Oral , Adult , Animals , Carbon Radioisotopes/analysis , Healthy Volunteers , Heterocyclic Compounds, 2-Ring/administration & dosage , Heterocyclic Compounds, 2-Ring/analysis , Heterocyclic Compounds, 2-Ring/chemistry , Humans , Intestinal Elimination , Intestinal Mucosa/enzymology , Intestinal Mucosa/microbiology , Intestinal Reabsorption , Male , Middle Aged , Models, Animal , Oxidation-Reduction , Pyrazines/administration & dosage , Pyrazines/analysis , Pyrazines/chemistry , Pyrroles/administration & dosage , Pyrroles/analysis , Pyrroles/chemistry , Radiometry , Rats , Sulfenic Acids/analysis , Sulfenic Acids/metabolism
3.
J Biol Chem ; 294(16): 6522-6530, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30850396

ABSTRACT

Oxidative modifications of cysteine residues are an important component in signaling pathways, enzymatic regulation, and redox homeostasis. Current direct and indirect methods detect specific modifications and a general binary population of "free" or "oxidized" cysteines, respectively. In an effort to combine both direct and indirect detection strategies, here we developed a method that we designate isotopic tagging of oxidized and reduced cysteines (iTORC). This method uses synthetic molecules for rapid isotopic coding of sulfenic acids, reduced cysteines, and disulfides in cells. Our approach utilizes isotopically distinct benzothiazine and halogenated benzothiazine probes to sequentially alkylate sulfenic acids and then free thiols and, finally, after a reduction step, cysteines oxidized to disulfides or other phosphine-reducible states. We ascertained that the iodinated benzothiazine probe has reduced cross-reactivity toward primary amines and is highly reactive with the cysteine of GSH, with a calculated rate constant of 2 × 105 m-1 s-1 (pH 8.0, 23 °C) (i.e. 10-20 times faster than N-ethylmaleimide). We applied iTORC to a mouse hepatocyte lysate to identify known sulfenylated and disulfide-bonded proteins, including elongation factor 1-α1 and mouse serum albumin, and found that iTORC reliably detected their expected oxidation status. This method can be easily employed to study the effects of oxidants on recombinant proteins and cell and tissue extracts, and the efficiencies of the alkylating agents enable completion of all three labeling steps within 2 h. In summary, we demonstrate here that halogenated benzothiazine-based alkylating agents can be utilized to rapidly measure the cellular thiol status in cells.


Subject(s)
Benzothiadiazines/chemistry , Cysteine/metabolism , Hepatocytes/metabolism , Isotope Labeling/methods , Sulfenic Acids/metabolism , Animals , Benzothiadiazines/pharmacology , Cysteine/analysis , Male , Mice , Oxidation-Reduction , Sulfenic Acids/analysis
4.
ACS Chem Biol ; 14(4): 594-598, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30893551

ABSTRACT

Norbornene derivatives were validated as probes for cysteine sulfenic acid on proteins and in live cells. Trapping sulfenic acids with norbornene probes is highly selective and revealed a different reactivity profile than the traditional dimedone reagent. The norbornene probe also revealed a superior chemoselectivity when compared to a commonly used dimedone probe. Together, these results advance the study of cysteine oxidation in biological systems.


Subject(s)
Cysteine/analogs & derivatives , Molecular Probes/chemistry , Norbornanes/chemistry , Sulfenic Acids/analysis , Cysteine/analysis , Oxidation-Reduction
5.
Chem Res Toxicol ; 32(3): 526-534, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30784263

ABSTRACT

Redox-mediated protein modifications control numerous processes in both normal and disease metabolism. Protein sulfenic acids, formed from the oxidation of protein cysteine residues, play a critical role in thiol-based redox signaling. The reactivity of protein sulfenic acids requires their identification through chemical trapping, and this paper describes the use of the triphenylphosphonium (TPP) ion to direct known sulfenic acid traps to the mitochondria, a verified source of cellular reactive oxygen species. Coupling of the TPP group with the 2,4-(dioxocyclohexyl)propoxy (DCP) unit and the bicyclo[6.1.0]nonyne (BCN) group produces two new probes, DCP-TPP and BCN-TPP. DCP-TPP and BCN-TPP react with C165A AhpC-SOH, a model protein sulfenic acid, to form the expected adducts with second-order rate constants of k = 1.1 M-1 s-1 and k = 5.99 M-1 s-1, respectively, as determined by electrospray ionization time-of-flight mass spectrometry. The TPP group does not alter the rate of DCP-TPP reaction with protein sulfenic acid compared to dimedone but slows the rate of BCN-TPP reaction compared to a non-TPP-containing BCN-OH control by 4.6-fold. The hydrophobic TPP group may interact with the protein, preventing an optimal reaction orientation for BCN-TPP. Unlike BCN-OH, BCN-TPP does not react with the protein persulfide, C165A AhpC-SSH. Extracellular flux measurements using A549 cells show that DCP-TPP and BCN-TPP influence mitochondrial energetics, with BCN-TPP producing a drastic decrease in basal respiration, perhaps due to its faster reaction kinetics with sulfenylated proteins. Further control experiments with BCN-OH, TPP-COOH, and dimedone provide strong evidence for mitochondrial localization and accumulation of DCP-TPP and BCN-TPP. These results reveal the compatibility of the TPP group with reactive sulfenic acid probes as a mitochondrial director and support the use of the TPP group in the design of sulfenic acid traps.


Subject(s)
Mitochondria/drug effects , Organophosphorus Compounds/chemical synthesis , Organophosphorus Compounds/pharmacology , Proteins/chemistry , Sulfenic Acids/analysis , A549 Cells , Humans , Mitochondria/metabolism , Molecular Probes/chemistry , Molecular Structure , Organophosphorus Compounds/chemistry
6.
ACS Nano ; 11(1): 358-367, 2017 01 24.
Article in English | MEDLINE | ID: mdl-27997794

ABSTRACT

Protein sulfenic acids play a key role in oxidative signal transduction of many biological and pathological processes; however, current chemical tools rely on visible fluorescence signals, limiting their utility to in vitro assays. We herein report reaction-based semiconducting polymer nanoprobes (rSPNs) with near-infrared absorption for in vivo photoacoustic (PA) imaging of protein sulfenic acids. rSPNs comprise an optically active semiconducting polymer as the core shielded with inert silica and poly(ethylene glycol) corona. The sulfenic acid reactive group (1,3-cyclohexanedione) is efficiently conjugated to the surface of nanoparticles via click chemistry. Such a nanostructure enables the specific recognition reaction between rSPNs and protein sulfenic acids without compromising the fluorescence and PA properties. In addition to in vitro tracking of the production of protein sulfenic acids in cancer cells under oxidative stress, rSPNs permit real-time PA and fluorescence imaging of protein sulfenic acids in tumors of living mice. This study thus not only demonstrates the first reaction-based PA probes with submolecular level recognition ability but also highlights the opportunities provided by hybrid nanoparticles for advanced molecular imaging.


Subject(s)
Fluorescent Dyes/chemistry , Nanoparticles/chemistry , Neoplasm Proteins/chemistry , Optical Imaging , Photoacoustic Techniques , Polyethylene Glycols/chemistry , Sulfenic Acids/analysis , Animals , Cell Survival/drug effects , Dose-Response Relationship, Drug , Fluorescent Dyes/pharmacology , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Oxidative Stress , Particle Size , Polyethylene Glycols/pharmacology , Semiconductors , Structure-Activity Relationship , Sulfenic Acids/metabolism , Surface Properties , Tumor Cells, Cultured
7.
J Proteome Res ; 15(12): 4221-4233, 2016 12 02.
Article in English | MEDLINE | ID: mdl-27690452

ABSTRACT

Physiological stimuli such as thrombin, or pathological stimuli such as lysophosphatidic acid (LPA), activate platelets. The activated platelets bind to monocytes through P-selectin-PSGL-1 interactions but also release the contents of their granules, commonly called "platelet releasate". It is known that monocytes in contact with platelet releasate produce reactive oxygen species (ROS). Reversible cysteine oxidation by ROS is considered to be a potential regulator of protein function. In a previous study, we used THP-1 monocytic cells exposed to LPA- or thrombin-induced platelet releasate and a modified biotin switch assay to unravel the biological processes that are influenced by reversible cysteine oxidation. To gain a better understanding of the redox regulation of monocytes in atherosclerosis, we have now altered the modified biotin switch to selectively quantify protein sulfenic acid, a subpopulation of reversible cysteine oxidation. Using arsenite as reducing agent in the modified biotin switch assay, we were able to quantify 1161 proteins, in which more than 100 sulfenic acid sites were identified. Bioinformatics analysis of the quantified sulfenic acid sites highlighted the relevant, previously missed biological process of monocyte transendothelial migration, which included integrin ß2. Flow cytometry validated the activation of LFA-1 (αLß2) and Mac-1 (αMß2), two subfamilies of integrin ß2 complexes, on human primary monocytes following platelet releasate treatment. The activation of LFA-1 was mediated by ROS from NADPH oxidase (NOX) activation. Production of ROS and activation of LFA-1 in human primary monocytes were independent of P-selectin-PSGL-1 interaction. Our results proved the modified biotin switch assay to be a powerful tool with the ability to reveal new regulatory mechanisms and identify new therapeutic targets.


Subject(s)
Blood Platelets/metabolism , CD18 Antigens/metabolism , Monocytes/metabolism , NADPH Oxidases/metabolism , Sulfenic Acids/analysis , Arsenites , Computational Biology , Humans , Lymphocyte Function-Associated Antigen-1/metabolism , Macrophage-1 Antigen/metabolism , Oxidation-Reduction , Proteins/analysis , Reactive Oxygen Species/metabolism , Secretory Vesicles , Transendothelial and Transepithelial Migration
8.
ACS Chem Biol ; 11(12): 3300-3304, 2016 12 16.
Article in English | MEDLINE | ID: mdl-27792307

ABSTRACT

Reactive oxygen species act as important second messengers in cell signaling and homeostasis through the oxidation of protein thiols. However, the dynamic nature of protein oxidation and the lack of sensitivity of existing molecular probes have hindered our understanding of such reactions; therefore, new tools are required to address these challenges. We designed a bifunctional variant of the strained bicyclo[6.1.0]nonyne (BCN-E-BCN) that enables the tagging of intracellular protein sulfenic acids for biorthogonal copper-free click chemistry. In validation studies, BCN-E-BCN binds the sulfenylated form of the actin-severing protein cofilin, while mutation of the cognate cysteine residues abrogates its binding. BCN-E-BCN is cell permeable and reacts rapidly with cysteine sulfenic acids in cultured cells. Using different azide-tagged conjugates, we demonstrate that BCN-E-BCN can be used in various applications for the detection of sulfenylated proteins. Remarkably, cycloaddition of an azide-tagged fluorophore to BCN-E-BCN labeled proteins produced in vivo can be visualized by fluorescence microscopy to reveal their localization. These findings demonstrate a novel and multifaceted approach to the detection and trapping of sulfenic acids.


Subject(s)
Azides/chemistry , Bridged Bicyclo Compounds/chemistry , Proteins/chemistry , Sulfenic Acids/analysis , Actin Depolymerizing Factors/chemistry , Blotting, Western , Cell Line, Tumor , Click Chemistry , Humans , Indicators and Reagents/chemistry , Microscopy, Fluorescence , Molecular Probes/chemistry
9.
Bioconjug Chem ; 27(5): 1411-8, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27123991

ABSTRACT

The comparative reaction efficiencies of currently used nucleophilic and electrophilic probes toward cysteine sulfenic acid have been thoroughly evaluated in two different settings-(i) a small molecule dipeptide based model and (ii) a recombinant protein model. We further evaluated the stability of corresponding thioether and sulfoxide adducts under reducing conditions which are commonly encountered during proteomic protocols and in cell analysis. Powered by the development of new cyclic and linear C-nucleophiles, the unsurpassed efficiency in the capture of sulfenic acid under competitive conditions is achieved and thus holds great promise as highly potent tools for activity-based sulfenome profiling.


Subject(s)
Sulfenic Acids/analysis , Sulfenic Acids/chemistry , Electron Transport , Models, Molecular , Protein Conformation
10.
Free Radic Res ; 50(2): 172-94, 2016.
Article in English | MEDLINE | ID: mdl-26340608

ABSTRACT

Selective modification of proteins at cysteine residues by reactive oxygen, nitrogen or sulfur species formed under physiological and pathological states is emerging as a critical regulator of protein activity impacting cellular function. This review focuses primarily on protein sulfenylation (-SOH), a metastable reversible modification connecting reduced cysteine thiols to many products of cysteine oxidation. An overview is first provided on the chemistry principles underlining synthesis, stability and reactivity of sulfenic acids in model compounds and proteins, followed by a brief description of analytical methods currently employed to characterize these oxidative species. The following chapters present a selection of redox-regulated proteins for which the -SOH formation was experimentally confirmed and linked to protein function. These chapters are organized based on the participation of these proteins in the regulation of signaling, metabolism and epigenetics. The last chapter discusses the therapeutic implications of altered redox microenvironment and protein oxidation in disease.


Subject(s)
Proteins/metabolism , Sulfenic Acids/metabolism , Sulfhydryl Compounds/metabolism , Animals , Cysteine , Epigenomics , Humans , Oxidation-Reduction , Proteins/analysis , Signal Transduction , Sulfenic Acids/analysis
11.
J Am Chem Soc ; 136(17): 6167-70, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24724926

ABSTRACT

Protein sulfenic acids are formed by the reaction of biologically relevant reactive oxygen species with protein thiols. Sulfenic acid formation modulates the function of enzymes and transcription factors either directly or through the subsequent formation of protein disulfide bonds. Identifying the site, timing, and conditions of protein sulfenic acid formation remains crucial to understanding cellular redox regulation. Current methods for trapping and analyzing sulfenic acids involve the use of dimedone and other nucleophilic 1,3-dicarbonyl probes that form covalent adducts with cysteine-derived protein sulfenic acids. As a mechanistic alternative, the present study describes highly strained bicyclo[6.1.0]nonyne (BCN) derivatives as concerted traps of sulfenic acids. These strained cycloalkynes react efficiently with sulfenic acids in proteins and small molecules yielding stable alkenyl sulfoxide products at rates more than 100× greater than 1,3-dicarbonyl reagents enabling kinetic competition with physiological sulfur chemistry. Similar to the 1,3-dicarbonyl reagents, the BCN compounds distinguish the sulfenic acid oxoform from the thiol, disulfide, sulfinic acid, and S-nitrosated forms of cysteine while displaying an acceptable cell toxicity profile. The enhanced rates demonstrated by these strained alkynes identify them as new bioorthogonal probes that should facilitate the discovery of previously unknown sulfenic acid sites and their parent proteins.


Subject(s)
Bridged Bicyclo Compounds/chemistry , Cycloparaffins/chemistry , Cysteine/analogs & derivatives , Proteins/chemistry , Sulfenic Acids/analysis , Cell Line , Cysteine/analysis , Humans , Models, Molecular , Oxidation-Reduction
12.
Free Radic Biol Med ; 71: 49-60, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24657741

ABSTRACT

Lysophosphatidic acid (LPA) is a growth factor for many cells including prostate and ovarian cancer-derived cell lines. LPA stimulates H2O2 production which is required for growth. However, there are significant gaps in our understanding of the spatial and temporal regulation of H2O2-dependent signaling and the way in which signals are transmitted following receptor activation. Herein, we describe the use of two reagents, DCP-Bio1 and DCP-Rho1, to evaluate the localization of active protein oxidation after LPA stimulation by detection of nascent protein sulfenic acids. We found that LPA stimulation causes internalization of LPA receptors into early endosomes that contain NADPH oxidase components and are sites of H2O2 generation. DCP-Rho1 allowed visualization of sulfenic acid formation, indicative of active protein oxidation, which was stimulated by LPA and decreased by an LPA receptor antagonist. Protein oxidation sites colocalized with LPAR1 and the endosomal marker EEA1. Concurrent with the generation of these redox signaling-active endosomes (redoxosomes) is the H2O2- and NADPH oxidase-dependent oxidation of Akt2 and PTP1B detected using DCP-Bio1. These new approaches therefore enable detection of active, H2O2-dependent protein oxidation linked to cell signaling processes. DCP-Rho1 may be a particularly useful protein oxidation imaging agent enabling spatial resolution due to the transient nature of the sulfenic acid intermediate it detects.


Subject(s)
Cysteine/analogs & derivatives , Gene Expression Regulation , Hydrogen Peroxide/metabolism , Lysophospholipids/pharmacology , Benzamides/chemistry , Cell Line, Tumor , Cysteine/analysis , Cysteine/biosynthesis , Endosomes/drug effects , Endosomes/metabolism , Female , Humans , Lysophospholipids/metabolism , Male , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Oxidation-Reduction , Phenylpropionates/chemistry , Protein Transport , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction , Sulfenic Acids/analysis , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
13.
Anal Chem ; 86(4): 1943-8, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24437714

ABSTRACT

A robust ICPMS-based method is introduced to obtain relative and absolute quantification of sulfenic acid (SA) in peptides and proteins. A new metal-containing reagent (Ln-DOTA-Dimedone) devised to react specifically with SA has been developed. The lanthanide-containing metal-coded affinity tag (Ln-MeCAT) was used to quantify thiol residues. We presented two approaches which allow the parallel and consecutive determination of SA and thiols in peptide and protein samples. The high sensitivity, structure-independent signal, and multiplexing capabilities of ICPMS together with the specificity of Ln-DOTA-Dimedone and Ln-MeCAT toward sulfenic acid and thiol residues, respectively, allow the characterization of various biological states and offer closer insight onto thiol-sulphenic acid equilibria which are involved in intracellular redox-mediated events altering structure and function of proteins in important diseases.


Subject(s)
Mass Spectrometry/methods , Peptides/analysis , Proteins/analysis , Sulfenic Acids/analysis , Peptides/chemistry , Protein Structure, Secondary , Proteins/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Spectrophotometry, Atomic/methods
14.
Biochim Biophys Acta ; 1840(2): 847-75, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23748139

ABSTRACT

BACKGROUND: Reactive oxygen species-mediated cysteine sulfenic acid modification has emerged as an important regulatory mechanism in cell signaling. The stability of sulfenic acid in proteins is dictated by the local microenvironment and ability of antioxidants to reduce this modification. Several techniques for detecting this cysteine modification have been developed, including direct and in situ methods. SCOPE OF REVIEW: This review presents a historical discussion of sulfenic acid chemistry and highlights key examples of this modification in proteins. A comprehensive survey of available detection techniques with advantages and limitations is discussed. Finally, issues pertaining to rates of sulfenic acid formation, reduction, and chemical trapping methods are also covered. MAJOR CONCLUSIONS: Early chemical models of sulfenic acid yielded important insights into the unique reactivity of this species. Subsequent pioneering studies led to the characterization of sulfenic acid formation in proteins. In parallel, the discovery of oxidant-mediated cell signaling pathways and pathological oxidative stress has led to significant interest in methods to detect these modifications. Advanced methods allow for direct chemical trapping of protein sulfenic acids directly in cells and tissues. At the same time, many sulfenic acids are short-lived and the reactivity of current probes must be improved to sample these species, while at the same time, preserving their chemical selectivity. Inhibitors with binding scaffolds can be rationally designed to target sulfenic acid modifications in specific proteins. GENERAL SIGNIFICANCE: Ever increasing roles for protein sulfenic acids have been uncovered in physiology and pathology. A more complete understanding of sulfenic acid-mediated regulatory mechanisms will continue to require rigorous and new chemical insights. This article is part of a Special Issue entitled Current methods to study reactive oxygen species - pros and cons and biophysics of membrane proteins. Guest Editor: Christine Winterbourn.


Subject(s)
Cell Physiological Phenomena , Proteins/chemistry , Sulfenic Acids/analysis , Sulfenic Acids/chemistry , Animals , Humans , Reactive Oxygen Species/metabolism , Signal Transduction
15.
Mass Spectrom Rev ; 33(2): 126-46, 2014.
Article in English | MEDLINE | ID: mdl-24105931

ABSTRACT

Orchestration of many processes relying on intracellular signal transduction is recognized to require the generation of hydrogen peroxide as a second messenger, yet relatively few molecular details of how this oxidant acts to regulate protein function are currently understood. This review describes emerging chemical tools and approaches that can be applied to study protein oxidation in biological systems, with a particular emphasis on a key player in protein redox regulation, cysteine sulfenic acid. While sulfenic acids (within purified proteins or simple mixtures) are detectable by physical approaches like X-ray crystallography, nuclear magnetic resonance and mass spectrometry, the propensity of these moieties to undergo further modification in complex biological systems has necessitated the development of chemical probes, reporter groups and analytical approaches to allow for their selective detection and quantification. Provided is an overview of techniques that are currently available for the study of sulfenic acids, and some of the biologically meaningful data that have been collected using such approaches.


Subject(s)
Cysteine/analogs & derivatives , Mass Spectrometry/methods , Proteins/chemistry , Sulfenic Acids/analysis , Animals , Cysteine/analysis , Humans , Oxidation-Reduction
16.
Org Biomol Chem ; 11(43): 7566-73, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24097070

ABSTRACT

Sulfenylation is one of the reversible post-translational modifications, playing significant roles in cellular redox homeostasis and signaling systems. Herein, small fluorescent probe (CPD and CPDDM) based live-cell labelling technology for the visualization of protein sulfenylation responses in living cells has been developed. This approach enables the detection of protein sulfenylation without the need for cell lysis, fixation or purification, and permits the noninvasive study of protein sulfenylation in live cells through the direct fluorescent readout. This technology also can realize dynamic tracking of protein sulfenylation in situ with minimal perturbation to sulfenylated proteins and less interference with cellular function. Information on the global distribution and dynamic changes of endogenous protein sulfenylation has been obtained.


Subject(s)
Cyclohexanones/chemistry , Fluorescent Dyes/chemistry , Hepatocytes/chemistry , Proteins/analysis , Sulfenic Acids/analysis , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cyclohexanones/metabolism , Dose-Response Relationship, Drug , Fluorescent Dyes/metabolism , Fluorescent Dyes/pharmacology , HL-60 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Molecular Structure , Protein Processing, Post-Translational , Proteins/metabolism , Spectrometry, Fluorescence , Structure-Activity Relationship , Sulfenic Acids/metabolism
17.
Obesity (Silver Spring) ; 21(2): 297-305, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23404913

ABSTRACT

UNLABELLED: A number of clinical and biochemical studies demonstrate that obesity and insulin resistance are associated with increases in oxidative stress and inflammation. Paradoxically, insulin sensitivity can be enhanced by oxidative inactivation of cysteine residues of phosphatases, and inflammation can be reduced by S-glutathionylation with formation of protein-glutathione mixed disulfides (PSSG). Although oxidation of protein-bound thiols (PSH) is increased in multiple diseases, it is not known whether there are changes in PSH oxidation species in obesity. OBJECTIVE: In this work, the hypothesis that obesity is associated with decreased levels of proteins containing oxidized protein thiols was tested. DESIGN AND METHODS: The tissue levels of protein sulfenic acids (PSOH) and PSSG in liver, visceral adipose tissue, and skeletal muscle derived from glucose intolerant, obese-prone Sprague-Dawley rats were examined. RESULTS: The data in this study indicate that decreases in PSSG content occurred in liver (44%) and adipose (26%) but not skeletal muscle in obese rats that were fed a 45% fat-calorie diet versus lean rats that were fed a 10% fat-calorie diet. PSOH content did not change in the tissue between the two groups. The activity of the enzyme glutaredoxin (GLRX) responsible for reversal of PSSG formation did not change in muscle and liver between the two groups. However, levels of GLRX1 were elevated 70% in the adipose tissue of the obese, 45% fat calorie-fed rats. CONCLUSION: These are the first data to link changes in S-glutathionylation and GLRX1 to adipose tissue in the obese and demonstrate that redox changes in thiol status occur in adipose tissue as a result of obesity.


Subject(s)
Intra-Abdominal Fat/metabolism , Liver/metabolism , Obesity/metabolism , Animals , Disulfides/analysis , Energy Intake , Glucose Intolerance , Glutaredoxins/genetics , Glutaredoxins/metabolism , Insulin Resistance , Magnetic Resonance Spectroscopy , Male , Muscle, Skeletal/metabolism , Oxidative Stress , Rats , Rats, Sprague-Dawley , Sulfenic Acids/analysis , Sulfhydryl Compounds/metabolism
18.
Yao Xue Xue Bao ; 47(3): 280-90, 2012 Mar.
Article in Chinese | MEDLINE | ID: mdl-22645750

ABSTRACT

Redox signal transduction, especially the oxidative modification of proein thiols, correlates with many diseases and becomes an expanding research area. However, there was rare method for quick and specific detection of protein thiols and their oxidative modification in living cells. In this article, we review the current chemical strategies for the detection and quantification of protein thiols and related cysteine oxidation. We also look into the future of the development of fluorescent probes for protein thiols and their potential application in the research of reactive cysteine proteomes and early detection of redox-related diseases.


Subject(s)
Proteins/chemistry , Sulfenic Acids/analysis , Sulfhydryl Compounds , Animals , Cysteine/metabolism , Fluorescent Dyes , Humans , Nitrosation , Oxidation-Reduction , Proteins/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/analysis , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism
19.
SELECTION OF CITATIONS
SEARCH DETAIL
...