Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
J Extracell Biol ; 1(7)2022 Jul.
Article in English | MEDLINE | ID: mdl-36590238

ABSTRACT

Quiescin sulfhydryl oxidase 1 (QSOX1) is a disulfide bond generating catalyst that is overexpressed in solid tumors. Expression of QSOX1 is linked to cancer cell invasion, tumor grade, and extracellular matrix (ECM) protein deposition. While the secreted version of QSOX1 is known to be present in various fluids and secretory tissues, its presence in the ECM of cancer is less understood. To characterize secreted QSOX1, we separated conditioned media based on size and density. We discovered that the majority of secreted QSOX1 resides in the EV-depleted fraction and in the soluble protein fraction. Very little QSOX1 could be detected in the EVP fraction. We used immunofluorescence to image subpopulations of EVs and found QSOX1 in Golgi-derived vesicles and medium/large vesicles, but in general, most extracellular QSOX1 was not attributed to these vesicles. Next, we quantified QSOX1 co-localization with the EV marker Alix. For the medium/large EVs, ~98% contained QSOX1 when fibronectin was used as a coating. However, on collagen coatings, only ~60% of these vesicles contained QSOX1, suggesting differences in EV cargo based on ECM coated surfaces. About 10% of small EVs co-localized with QSOX1 on every ECM protein surface except for collagen (0.64%). We next investigated adhesion of QSOX1 to ECM proteins in vitro and in situ and found that QSOX1 preferentially adheres to fibronectin, laminins, and Matrigel compared to gelatin and collagen. This mechanism was found to be, in part, mediated by the formation of mixed disulfides between QSOX1 and cysteine-rich ECM proteins. In summary, we found that QSOX1 (1) is in subpopulations of medium/large EVs, (2) seems to interact with small Alix+ EVs, and (3) adheres to cysteine-rich ECM proteins, potentially through the formation of intermediate disulfides. These observations offer significant insight into how enzymes, such as QSOX1, can facilitate matrix remodeling events in solid tumor progression.

2.
J Am Chem Soc ; 143(28): 10793-10803, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34250803

ABSTRACT

Chromophores that absorb in the tissue-penetrant far-red/near-infrared window have long served as photocatalysts to generate singlet oxygen for photodynamic therapy. However, the cytotoxicity and side reactions associated with singlet oxygen sensitization have posed a problem for using long-wavelength photocatalysis to initiate other types of chemical reactions in biological environments. Herein, silicon-Rhodamine compounds (SiRs) are described as photocatalysts for inducing rapid bioorthogonal chemistry using 660 nm light through the oxidation of a dihydrotetrazine to a tetrazine in the presence of trans-cyclooctene dienophiles. SiRs have been commonly used as fluorophores for bioimaging but have not been applied to catalyze chemical reactions. A series of SiR derivatives were evaluated, and the Janelia Fluor-SiR dyes were found to be especially effective in catalyzing photooxidation (typically 3%). A dihydrotetrazine/tetrazine pair is described that displays high stability in both oxidation states. A protein that was site-selectively modified by trans-cyclooctene was quantitatively conjugated upon exposure to 660 nm light and a dihydrotetrazine. By contrast, a previously described methylene blue catalyst was found to rapidly degrade the protein. SiR-red light photocatalysis was used to cross-link hyaluronic acid derivatives functionalized by dihydrotetrazine and trans-cyclooctenes, enabling 3D culture of human prostate cancer cells. Photoinducible hydrogel formation could also be carried out in live mice through subcutaneous injection of a Cy7-labeled hydrogel precursor solution, followed by brief irradiation to produce a stable hydrogel. This cytocompatible method for using red light photocatalysis to activate bioorthogonal chemistry is anticipated to find broad applications where spatiotemporal control is needed in biological environments.


Subject(s)
Cyclooctanes/chemistry , Fluorescent Dyes/chemistry , Rhodamines/chemistry , Silicon/chemistry , Tetrazoles/chemical synthesis , Animals , Catalysis , Humans , Infrared Rays , Mice , Molecular Structure , Photochemical Processes , Tetrazoles/chemistry , Tumor Cells, Cultured
3.
Protein Sci ; 28(1): 5-7, 2019 01.
Article in English | MEDLINE | ID: mdl-30461094
4.
Protein Sci ; 27(8): 1509-1517, 2018 08.
Article in English | MEDLINE | ID: mdl-29696739

ABSTRACT

Gaussia princeps luciferase (GLuc) generates an intense burst of blue light when exposed to coelenterazine in the absence of ATP. Here we show that this 5-disulfide containing enzyme can be used as a facile and convenient substrate for studies of oxidative protein folding. Reduced GLuc (rGLuc), with 10 free cysteine residues, is completely inactive as a luciferase but >60% bioluminescence activity, compared to controls, can be recovered using a range of oxidizing regimens in the absence of the exogenous shuffling activity of protein disulfide isomerase (PDI). The sulfhydryl oxidase QSOX1 can be assayed using rGLuc in a simple bioluminescence plate reader format. Similarly, low concentrations of rGLuc can be oxidized by millimolar levels of dehydroascorbate, hydrogen peroxide or much lower concentrations of sodium tetrathionate. The oxidative refolding of rGLuc in the presence of a range of glutathione redox buffers is only marginally accelerated by micromolar levels of PDI. This modest rate enhancement probably results from a relatively simple disulfide connectivity in native GLuc; reflecting two homologous domains each carrying two disulfide bonds with a single interdomain disulfide. When GLuc is reoxidized under denaturing conditions the resulting scrambled protein (sGLuc) can be used in a sensitive bioluminescence assay for reduced PDI in the absence of added exogenous thiols. Finally, the general facility by which rGLuc can recover bioluminescent activity in vitro provides a sensitive method for the assessment of inhibitors of oxidative protein folding.


Subject(s)
Copepoda/enzymology , Enzyme Assays/methods , Luciferases/chemistry , Luciferases/metabolism , Oxidoreductases/analysis , Oxidoreductases/metabolism , Animals , Disulfides/chemistry , Oxidation-Reduction , Protein Folding
5.
Biochemistry ; 57(7): 1178-1189, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29341594

ABSTRACT

The development of genetically encoded fluorescent probes for analyte-specific imaging has revolutionized our understanding of intracellular processes. Current classes of intracellular probes depend on the selection of binding domains that either undergo conformational changes on analyte binding or can be linked to thiol redox chemistry. Here we have designed novel probes by fusing a flavoenzyme, whose fluorescence is quenched on reduction by the analyte of interest, with a GFP domain to allow for rapid and specific ratiometric sensing. Two flavoproteins, Escherichia coli thioredoxin reductase and Saccharomyces cerevisiae lipoamide dehydrogenase, were successfully developed into thioredoxin and NAD+/NADH specific probes, respectively, and their performance was evaluated in vitro and in vivo. A flow cell format, which allowed dynamic measurements, was utilized in both bacterial and mammalian systems. In E. coli the first reported intracellular steady-state of the cytoplasmic thioredoxin pool was measured. In HEK293T mammalian cells, the steady-state cytosolic ratio of NAD+/NADH induced by glucose was determined. These genetically encoded fluorescent constructs represent a modular approach to intracellular probe design that should extend the range of metabolites that can be quantitated in live cells.


Subject(s)
Escherichia coli/enzymology , Flavoproteins/metabolism , Luminescent Agents/metabolism , Optical Imaging/methods , Saccharomyces cerevisiae/enzymology , Dihydrolipoamide Dehydrogenase/analysis , Dihydrolipoamide Dehydrogenase/metabolism , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Models, Molecular , NADP/analysis , NADP/metabolism , Oxidation-Reduction , Recombinant Fusion Proteins/metabolism , Thioredoxin-Disulfide Reductase/analysis , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxins/analysis , Thioredoxins/metabolism
6.
Chem Rev ; 118(3): 1169-1198, 2018 02 14.
Article in English | MEDLINE | ID: mdl-28699750

ABSTRACT

Cysteine thiols are among the most reactive functional groups in proteins, and their pairing in disulfide linkages is a common post-translational modification in proteins entering the secretory pathway. This modest amino acid alteration, the mere removal of a pair of hydrogen atoms from juxtaposed cysteine residues, contrasts with the substantial changes that characterize most other post-translational reactions. However, the wide variety of proteins that contain disulfides, the profound impact of cross-linking on the behavior of the protein polymer, the numerous and diverse players in intracellular pathways for disulfide formation, and the distinct biological settings in which disulfide bond formation can take place belie the simplicity of the process. Here we lay the groundwork for appreciating the mechanisms and consequences of disulfide bond formation in vivo by reviewing chemical principles underlying cysteine pairing and oxidation. We then show how enzymes tune redox-active cofactors and recruit oxidants to improve the specificity and efficiency of disulfide formation. Finally, we discuss disulfide bond formation in a cellular context and identify important principles that contribute to productive thiol oxidation in complex, crowded, dynamic environments.


Subject(s)
Disulfides/chemistry , Proteins/metabolism , Sulfhydryl Compounds/chemistry , Bacteria/enzymology , Bacteria/metabolism , Glutathione Peroxidase/chemistry , Glutathione Peroxidase/metabolism , Periplasm/metabolism , Peroxiredoxins/chemistry , Peroxiredoxins/metabolism , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/metabolism , Protein Folding , Protein Processing, Post-Translational , Proteins/chemistry
7.
Free Radic Biol Med ; 108: 741-749, 2017 07.
Article in English | MEDLINE | ID: mdl-28465261

ABSTRACT

This paper addresses how to evaluate the efficacy of the growing inventory of thiol-reactive inhibitors of mammalian protein disulfide Isomerase (PDI) enzymes under realistic concentrations of potentially competing thiol-containing peptides and proteins. For this purpose, we introduce a variant of the widely-used reductase assay by using a commercially-available cysteine derivative (BODIPY FL L-Cystine; BD-SS) that yields a 55-fold increase in fluorescence (excitation/emission; 490/513nm) on scission of the disulfide bond. This plate reader-compatible method detects human PDI down to 5-10nM, can utilize a range of thiol substrates (including 5µM dithiothreitol, 10µM reduced RNase thiols, and 5mM glutathione; GSH), and can operate from pH 6-9.5 in a variety of buffers. PDI assays often employ low micromolar levels of substrates leading to ambiguities when thiol-directed inhibitors are evaluated. The present work utilizes 5mM GSH for both pre-incubation and assay phases to more realistically reflect the high concentration of thiols that an inhibitor would encounter intracellularly. Extracellular PDI faces a much lower concentration of potentially competing thiols; to assess reductase activity under these conditions, the pre-reduced PDI is treated with inhibitor and then fluorescence increase upon reduction of BD-SS is followed in the absence of additional competing thiols. Both assay modes were tested with four mechanistically diverse PDI inhibitors. Two reversible reagents, 3,4-methylenedioxy-ß-nitrostyrene (MNS) and the arsenical APAO, were found to be strong inhibitors of PDI in the absence of competing thiols, but were ineffective in the presence of 5mM GSH. A further examination of the nitrostyrene showed that MNS not only forms facile Michael adducts with GSH, but also with the thiols of unfolded proteins (Kd values of 7 and <0.1µM, respectively) suggesting the existence of multiple potential intracellular targets for this membrane-permeant reagent. The inhibition of PDI by the irreversible alkylating agent, the chloroacetamide 16F16, was found to be only modestly attenuated by 5mM GSH. Finally, the thiol-independent flavonoid inhibitor quercetin-3-O-rutinoside was found to show equal efficacy in reoxidation and turnover assay types. This work provides a framework to evaluate inhibitors that may target the CxxC motifs of PDI and addresses some of the complexities in the interpretation of the behavior of thiol-directed reagents in vivo.


Subject(s)
Arsenicals/metabolism , Boron Compounds/metabolism , Cystine/analogs & derivatives , Protein Disulfide-Isomerases/metabolism , Sulfhydryl Compounds/chemistry , Animals , Arsenicals/chemistry , Binding Sites , Boron Compounds/chemistry , Cell-Free System , Cysteine/metabolism , Cystine/chemistry , Cystine/metabolism , Dioxolanes/metabolism , Glucosides/metabolism , Glutathione/chemistry , Glutathione/metabolism , Humans , Mammals , Oxidation-Reduction , Protein Disulfide-Isomerases/antagonists & inhibitors , Quercetin/analogs & derivatives , Quercetin/metabolism , Spectrometry, Fluorescence
8.
J Am Chem Soc ; 138(18): 5978-83, 2016 05 11.
Article in English | MEDLINE | ID: mdl-27078610

ABSTRACT

Rapid bioorthogonal reactivity can be induced by controllable, catalytic stimuli using air as the oxidant. Methylene blue (4 µM) irradiated with red light (660 nm) catalyzes the rapid oxidation of a dihydrotetrazine to a tetrazine thereby turning on reactivity toward trans-cyclooctene dienophiles. Alternately, the aerial oxidation of dihydrotetrazines can be efficiently catalyzed by nanomolar levels of horseradish peroxidase under peroxide-free conditions. Selection of dihydrotetrazine/tetrazine pairs of sufficient kinetic stability in aerobic aqueous solutions is key to the success of these approaches. In this work, polymer fibers carrying latent dihydrotetrazines were catalytically activated and covalently modified by trans-cyclooctene conjugates of small molecules, peptides, and proteins. In addition to visualization with fluorophores, fibers conjugated to a cell adhesive peptide exhibited a dramatically increased ability to mediate contact guidance of cells.


Subject(s)
Heterocyclic Compounds, 1-Ring/chemistry , Adhesives , Catalysis , Enzymes/chemistry , Horseradish Peroxidase/chemistry , Kinetics , Light , Methylene Blue/chemistry , Oxidation-Reduction , Photochemical Processes , Spectrophotometry, Ultraviolet
9.
Arch Biochem Biophys ; 579: 1-7, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26014136

ABSTRACT

Mia40 participates in oxidative protein folding within the mitochondrial intermembrane space (IMS) by mediating the transfer of reducing equivalents from client proteins to FAD-linked oxidoreductases of the Erv1 family (lfALR in mammals). Here we investigate the specificity of the human Mia40/lfALR system towards non-cognate unfolded protein substrates to assess whether the efficient introduction of disulfides requires a particular amino acid sequence context or the presence of an IMS targeting signal. Reduced pancreatic ribonuclease A (rRNase), avian lysozyme, and riboflavin binding protein are all competent substrates of the Mia40/lfALR system, although they lack those sequence features previously thought to direct disulfide bond formation in cognate IMS substrates. The oxidation of rRNase by Mia40 does not limit overall turnover of unfolded substrate by the Mia40/lfALR system. Mia40 is an ineffective protein disulfide isomerase when its ability to restore enzymatic activity from scrambled RNase is compared to that of protein disulfide isomerase. Mia40's ability to bind amphipathic peptides is evident by avid binding to the isolated B-chain during the insulin reductase assay. In aggregate these data suggest that the Mia40/lfALR system has a broad sequence specificity and that potential substrates may be protected from adventitious oxidation by kinetic sequestration within the mitochondrial IMS.


Subject(s)
Cytochrome Reductases/chemistry , Cytochrome Reductases/ultrastructure , Isomerases/chemistry , Isomerases/ultrastructure , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/ultrastructure , Amino Acid Sequence , Binding Sites , Computer Simulation , Enzyme Activation , Humans , Mitochondrial Precursor Protein Import Complex Proteins , Models, Chemical , Models, Molecular , Molecular Sequence Data , Oxidants/chemistry , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors , Protein Binding , Protein Conformation , Protein Folding , Structure-Activity Relationship
10.
Free Radic Biol Med ; 80: 171-82, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25091901

ABSTRACT

This review examines oxidative protein folding within the mammalian endoplasmic reticulum (ER) from an enzymological perspective. In protein disulfide isomerase-first (PDI-first) pathways of oxidative protein folding, PDI is the immediate oxidant of reduced client proteins and then addresses disulfide mispairings in a second isomerization phase. In PDI-second pathways the initial oxidation is PDI-independent. Evidence for the rapid reduction of PDI by reduced glutathione is presented in the context of PDI-first pathways. Strategies and challenges are discussed for determination of the concentrations of reduced and oxidized glutathione and of the ratios of PDI(red):PDI(ox). The preponderance of evidence suggests that the mammalian ER is more reducing than first envisaged. The average redox state of major PDI-family members is largely to almost totally reduced. These observations are consistent with model studies showing that oxidative protein folding proceeds most efficiently at a reducing redox poise consistent with a stoichiometric insertion of disulfides into client proteins. After a discussion of the use of natively encoded fluorescent probes to report the glutathione redox poise of the ER, this review concludes with an elaboration of a complementary strategy to discontinuously survey the redox state of as many redox-active disulfides as can be identified by ratiometric LC-MS-MS methods. Consortia of oxidoreductases that are in redox equilibrium can then be identified and compared to the glutathione redox poise of the ER to gain a more detailed understanding of the factors that influence oxidative protein folding within the secretory compartment.


Subject(s)
Disulfides/chemistry , Endoplasmic Reticulum/metabolism , Oxidoreductases Acting on Sulfur Group Donors/chemistry , Protein Disulfide-Isomerases/chemistry , Protozoan Proteins/chemistry , Sulfhydryl Compounds/chemistry , Animals , Disulfides/metabolism , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/ultrastructure , Eukaryotic Cells/cytology , Eukaryotic Cells/enzymology , Glutathione/chemistry , Glutathione/metabolism , Glutathione Disulfide/chemistry , Glutathione Disulfide/metabolism , Humans , Kinetics , Models, Molecular , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Protein Disulfide-Isomerases/metabolism , Protein Folding , Protozoan Proteins/metabolism , Sulfhydryl Compounds/metabolism , Trypanosoma brucei brucei/cytology , Trypanosoma brucei brucei/enzymology
11.
Biochemistry ; 54(2): 612-21, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25506675

ABSTRACT

The renewed use of arsenicals as chemotherapeutics has rekindled interest in the biochemistry of As(III) species. In this work, simple bis- and tris-arsenical derivatives were synthesized with the aim of exploiting the chelate effect in the inhibition of thiol-disulfide oxidoreductases (here, Quiescin sulfhydryl oxidase, QSOX, and protein disulfide isomerase, PDI) that utilize two or more CxxC motifs in the catalysis of oxidative protein folding. Coupling 4-aminophenylarsenoxide (APAO) to acid chloride or anhydride derivatives yielded two bis-arsenical prototypes, BA-1 and BA-2, and a tris-arsenical, TA-1. Unlike the monoarsenical, APAO, these new reagents proved to be strong inhibitors of oxidative protein folding in the presence of a realistic intracellular concentration of competing monothiol (here, 5 mM reduced glutathione, GSH). However, this inhibition does not reflect direct inactivation of QSOX or PDI, but avid binding of MVAs to the reduced unfolded protein substrates themselves. Titrations of reduced riboflavin-binding protein with MVAs show that all 18 protein -SH groups can be captured by these arsenicals. With reduced RNase, addition of substoichiometric levels of MVAs is accompanied by the formation of Congo Red- and Thioflavin T-positive fibrillar aggregates. Even with Kd values of ∼50 nM, MVAs are ineffective inhibitors of PDI in the presence of millimolar levels of competing GSH. These results underscore the difficulties of designing effective and specific arsenical inhibitors for folded enzymes and proteins. Some of the cellular effects of arsenicals likely reflect their propensity to associate very tightly and nonspecifically to conformationally mobile cysteine-rich regions of proteins, thereby interfering with folding and/or function.


Subject(s)
Arsenicals/pharmacology , Enzyme Inhibitors/pharmacology , Oxidoreductases Acting on Sulfur Group Donors/chemistry , Protein Disulfide-Isomerases/chemistry , Protein Folding/drug effects , Protozoan Proteins/chemistry , Trypanosoma brucei brucei/enzymology , Amino Acid Motifs , Arsenic , Arsenicals/chemistry , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Oxidation-Reduction/drug effects , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Protein Disulfide-Isomerases/metabolism , Protozoan Proteins/metabolism
12.
Arch Biochem Biophys ; 548: 60-5, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24582598

ABSTRACT

Augmenter of liver regeneration (sfALR) is a small disulfide-bridged homodimeric flavoprotein with sulfhydryl oxidase activity. Here, we investigate the catalytic and spectroscopic consequences of selectively replacing C145 by a selenocysteine to complement earlier studies in which random substitution of ∼90% of the 6 cysteine residues per sfALR monomer was achieved growing Escherichia coli on selenite. A selenocysteine insertion sequence (SECIS) element was installed within the gene for human sfALR. SecALR2 showed a spectrum comparable to that of wild-type sfALR. The catalytic efficiency of SecALR2 towards dithiothreitol was 6.8-fold lower than a corresponding construct in which position 145 was returned to a cysteine residue while retaining the additional mutations introduced with the SECIS element. This all-cysteine control enzyme formed a mixed disulfide between C142 and ß-mercaptoethanol releasing C145 to form a thiolate-flavin charge transfer absorbance band at ∼530nm. In contrast, SecALR2 showed a prominent long-wavelength absorbance at 585 nm consistent with the expectation that a selenolate would be a better charge-transfer donor to the isoalloxazine ring. These data show the robustness of the ALR protein fold towards the multiple mutations required to insert the SECIS element and provide the first example of a selenolate to flavin charge-transfer complex.


Subject(s)
Flavoproteins/genetics , Flavoproteins/metabolism , Proteins/genetics , Proteins/metabolism , Selenocysteine/genetics , Selenocysteine/metabolism , Amino Acid Sequence , Base Sequence , Catalytic Domain , Cloning, Molecular , Escherichia coli/genetics , Flavins/metabolism , Flavoproteins/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Insertional , Oxidation-Reduction , Proteins/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Selenocysteine/chemistry
13.
Free Radic Biol Med ; 69: 129-35, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24468475

ABSTRACT

A sensitive new plate-reader assay has been developed showing that adult mammalian blood serum contains circulating soluble sulfhydryl oxidase activity that can introduce disulfide bonds into reduced proteins with the reduction of oxygen to hydrogen peroxide. The activity was purified 5000-fold to >90% homogeneity from bovine serum and found by mass spectrometry to be consistent with the short isoform of quiescin-sulfhydryl oxidase 1 (QSOX1). This FAD-dependent enzyme is present at comparable activity levels in fetal and adult commercial bovine sera. Thus cell culture media that are routinely supplemented with either fetal or adult bovine sera will contain this facile catalyst of protein thiol oxidation. QSOX1 is present at approximately 25 nM in pooled normal adult human serum. Examination of the unusual kinetics of QSOX1 toward cysteine and glutathione at low micromolar concentrations suggests that circulating QSOX1 is unlikely to significantly contribute to the oxidation of these monothiols in plasma. However, the ability of QSOX1 to rapidly oxidize conformationally mobile protein thiols suggests a possible contribution to the redox status of exofacial and soluble proteins in blood plasma. Recent proteomic studies showing that plasma QSOX1 can be utilized in the diagnosis of pancreatic cancer and acute decompensated heart failure, together with the overexpression of this secreted enzyme in a number of solid tumors, suggest that the robust QSOX assay developed here may be useful in the quantitation of enzyme levels in a wide range of biological fluids.


Subject(s)
Disulfides/blood , Oxidoreductases Acting on Sulfur Group Donors/isolation & purification , Protein Isoforms/isolation & purification , Amino Acid Motifs , Amino Acid Sequence , Animals , Catalysis , Cattle , Humans , Kinetics , Oxidoreductases Acting on Sulfur Group Donors/blood , Oxidoreductases Acting on Sulfur Group Donors/chemistry , Protein Conformation , Protein Isoforms/blood , Protein Isoforms/chemistry , Proteomics , Rats
14.
J Biol Chem ; 289(8): 5274-84, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24379406

ABSTRACT

The quiescin sulfhydryl oxidase (QSOX) family of enzymes generates disulfide bonds in peptides and proteins with the reduction of oxygen to hydrogen peroxide. Determination of the potentials of the redox centers in Trypanosoma brucei QSOX provides a context for understanding catalysis by this facile oxidant of protein thiols. The CXXC motif of the thioredoxin domain is comparatively oxidizing (E'0 of -144 mV), consistent with an ability to transfer disulfide bonds to a broad range of thiol substrates. In contrast, the proximal CXXC disulfide in the ERV (essential for respiration and vegetative growth) domain of TbQSOX is strongly reducing (E'0 of -273 mV), representing a major apparent thermodynamic barrier to overall catalysis. Reduction of the oxidizing FAD cofactor (E'0 of -153 mV) is followed by the strongly favorable reduction of molecular oxygen. The role of a mixed disulfide intermediate between thioredoxin and ERV domains was highlighted by rapid reaction studies in which the wild-type CGAC motif in the thioredoxin domain of TbQSOX was replaced by the more oxidizing CPHC or more reducing CGPC sequence. Mixed disulfide bond formation is accompanied by the generation of a charge transfer complex with the flavin cofactor. This provides thermodynamic coupling among the three redox centers of QSOX and avoids the strongly uphill mismatch between the formal potentials of the thioredoxin and ERV disulfides. This work identifies intriguing mechanistic parallels between the eukaryotic QSOX enzymes and the DsbA/B system catalyzing disulfide bond generation in the bacterial periplasm and suggests that the strategy of linked disulfide exchanges may be exploited in other catalysts of oxidative protein folding.


Subject(s)
Biocatalysis , Disulfides/metabolism , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Protozoan Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Anaerobiosis , Crystallography, X-Ray , Dithiothreitol/metabolism , Flavin-Adenine Dinucleotide/metabolism , Kinetics , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation/genetics , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors/chemistry , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Spectrophotometry , Thermodynamics , Thioredoxins/chemistry
15.
Biochim Biophys Acta ; 1840(2): 838-46, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23567800

ABSTRACT

BACKGROUND: Disulfide bond formation is a key posttranslational modification, with implications for structure, function and stability of numerous proteins. While disulfide bond formation is a necessary and essential process for many proteins, it is deleterious and disruptive for others. Cells go to great lengths to regulate thiol-disulfide bond homeostasis, typically with several, apparently redundant, systems working in parallel. Dissecting the extent of oxidation and reduction of disulfides is an ongoing challenge due, in part, to the facility of thiol/disulfide exchange reactions. SCOPE OF REVIEW: In the present account, we briefly survey the toolbox available to the experimentalist for the chemical determination of thiols and disulfides. We have chosen to focus on the key chemical aspects of current methodology, together with identifying potential difficulties inherent in their experimental implementation. MAJOR CONCLUSIONS: While many reagents have been described for the measurement and manipulation of the redox status of thiols and disulfides, a number of these methods remain underutilized. The ability to effectively quantify changes in redox conditions in living cells presents a continuing challenge. GENERAL SIGNIFICANCE: Many unresolved questions in the metabolic interconversion of thiols and disulfides remain. For example, while pool sizes of redox pairs and their intracellular distribution are being uncovered, very little is known about the flux in thiol-disulfide exchange pathways. New tools are needed to address this important aspect of cellular metabolism. 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)
Disulfides/analysis , Proteins/chemistry , Sulfhydryl Compounds/analysis , Animals , Disulfides/chemistry , Humans , Oxidation-Reduction , Sulfhydryl Compounds/chemistry
16.
Biochemistry ; 52(46): 8323-32, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24147449

ABSTRACT

Augmenter of liver regeneration is a member of the ERV family of small flavin-dependent sulfhydryl oxidases that contain a redox-active CxxC disulfide bond in redox communication with the isoalloxazine ring of bound FAD. These enzymes catalyze the oxidation of thiol substrates with the reduction of molecular oxygen to hydrogen peroxide. This work studies the catalytic mechanism of the short, cytokine form of augmenter of liver regeneration (sfALR) using model thiol substrates of the enzyme. The redox potential of the proximal disulfide in sfALR was found to be approximately 57 mV more reducing than the flavin chromophore, in agreement with titration experiments. Rapid reaction studies show that dithiothreitol (DTT) generates a transient mixed disulfide intermediate with sfALR signaled by a weak charge-transfer interaction between the thiolate of C145 and the oxidized flavin. The subsequent transfer of reducing equivalents to the flavin ring is relatively slow, with a limiting apparent rate constant of 12.4 s(-1). However, reoxidation of the reduced flavin by molecular oxygen is even slower (2.3 s(-1) at air saturation) and thus largely limits turnover at 5 mM DTT. The nature of the charge-transfer complexes observed with DTT was explored using a range of simple monothiols to mimic the initial nucleophilic attack on the proximal disulfide. While ß-mercaptoethanol is a very poor substrate of sfALR (∼0.3 min(-1) at 100 mM thiol), it rapidly generates a mixed disulfide intermediate allowing the thiolate of C145 to form a strong charge-transfer complex with the flavin. Unlike the other monothiols tested, glutathione is unable to form charge-transfer complexes and is an undetectable substrate of the oxidase. These data are rationalized on the basis of the stringent steric requirements for thiol-disulfide exchange reactions. The inability of the relatively bulky glutathione to attain the in-line geometry required for efficient disulfide exchange in sfALR may be physiologically important in preventing the oxidase from catalyzing the potentially harmful oxidation of intracellular glutathione.


Subject(s)
Cytochrome Reductases/metabolism , Oxidoreductases/metabolism , Catalysis , Cytochrome Reductases/genetics , Disulfides/chemistry , Flavins/chemistry , Flavins/metabolism , Humans , Mercaptoethanol/metabolism , Models, Molecular , Oxidation-Reduction , Oxidoreductases/genetics , Oxidoreductases Acting on Sulfur Group Donors , Oxygen/chemistry
17.
J Am Chem Soc ; 135(7): 2415-8, 2013 Feb 20.
Article in English | MEDLINE | ID: mdl-23384038

ABSTRACT

The finding that arsenic trioxide is an effective treatment for acute promyelocytic leukemia has renewed interest in the pharmacological uses of inorganic and organic arsenicals. Here we synthesized and characterized the reactivity of an arsenical-maleimide (As-Mal) that can be efficiently conjugated to exposed cysteine residues in peptides and proteins with the ultimate goal of directing these As(III) species to vicinal thiols in susceptible targets within cells and tissues. As-Mal conjugated to a surface cysteine in thioredoxin provides a more potent inhibitor for Escherichia coli thioredoxin reductase than comparable simple inorganic or organic arsenicals. As-Mal can be coupled to all of the eight cysteine residues of reduced unfolded ribonuclease A or to site-specific locations using appropriate cysteine mutations. We observed particularly strong binding to the two CxxC motifs of protein disulfide isomerase using a mutant RNase in which As-Mal was specifically incorporated at residues 26 and 110. As-Mal will serve as a facile reagent for the incorporation of As(III) species into a wide range of thiol-containing proteins, biomaterials, and surfaces.


Subject(s)
Arsenicals/chemical synthesis , Maleimides/chemical synthesis , Arsenicals/chemistry , Maleimides/chemistry , Models, Molecular , Protein Folding , Thioredoxin-Disulfide Reductase/antagonists & inhibitors
18.
J Mol Biol ; 425(2): 222-31, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23159557

ABSTRACT

Sulfur, a key contributor to biological reactivity, is not amendable to investigations by biological NMR spectroscopy. To utilize selenium as a surrogate, we have developed a generally applicable (77)Se isotopic enrichment method for heterologous proteins expressed in Escherichia coli. We demonstrate (77)Se NMR spectroscopy of multiple selenocysteine and selenomethionine residues in the sulfhydryl oxidase augmenter of liver regeneration (ALR). The resonances of the active-site residues were assigned by comparing the NMR spectra of ALR bound to oxidized and reduced flavin adenine dinucleotide. An additional resonance appears only in the presence of the reducing agent and disappears readily upon exposure to air and subsequent reoxidation of the flavin. Hence, (77)Se NMR spectroscopy can be used to report the local electronic environment of reactive and structural sulfur sites, as well as changes taking place in those locations during catalysis.


Subject(s)
Cysteine/metabolism , Cytochrome Reductases/chemistry , Flavins/metabolism , Magnetic Resonance Spectroscopy , Selenocysteine/metabolism , Selenomethionine/metabolism , Catalysis , Crystallography, X-Ray , Cysteine/chemistry , Cysteine/genetics , Cytochrome Reductases/genetics , Cytochrome Reductases/metabolism , Humans , Mutation/genetics , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors , Protein Conformation , Protein Folding , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
Nature ; 488(7411): 414-8, 2012 Aug 16.
Article in English | MEDLINE | ID: mdl-22801504

ABSTRACT

Protein stability, assembly, localization and regulation often depend on the formation of disulphide crosslinks between cysteine side chains. Enzymes known as sulphydryl oxidases catalyse de novo disulphide formation and initiate intra- and intermolecular dithiol/disulphide relays to deliver the disulphides to substrate proteins. Quiescin sulphydryl oxidase (QSOX) is a unique, multi-domain disulphide catalyst that is localized primarily to the Golgi apparatus and secreted fluids and has attracted attention owing to its overproduction in tumours. In addition to its physiological importance, QSOX is a mechanistically intriguing enzyme, encompassing functions typically carried out by a series of proteins in other disulphide-formation pathways. How disulphides are relayed through the multiple redox-active sites of QSOX and whether there is a functional benefit to concatenating these sites on a single polypeptide are open questions. Here we present the first crystal structure of an intact QSOX enzyme, derived from a trypanosome parasite. Notably, sequential sites in the disulphide relay were found more than 40 Å apart in this structure, too far for direct disulphide transfer. To resolve this puzzle, we trapped and crystallized an intermediate in the disulphide hand-off, which showed a 165° domain rotation relative to the original structure, bringing the two active sites within disulphide-bonding distance. The comparable structure of a mammalian QSOX enzyme, also presented here, shows further biochemical features that facilitate disulphide transfer in metazoan orthologues. Finally, we quantified the contribution of concatenation to QSOX activity, providing general lessons for the understanding of multi-domain enzymes and the design of new catalytic relays.


Subject(s)
Disulfides/metabolism , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Trypanosoma brucei brucei/enzymology , Amino Acid Motifs , Animals , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Humans , Mice , Models, Molecular , Oxidation-Reduction , Protein Conformation , Rotation
20.
Biochemistry ; 51(20): 4226-35, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22582951

ABSTRACT

This work explores the substrate specificity of the quiescin sulfhydryl oxidase (QSOX) family of disulfide-generating flavoenzymes to provide enzymological context for investigation of the physiological roles of these facile catalysts of oxidative protein folding. QSOX enzymes are generally unable to form disulfide bonds within well-structured proteins. Use of a temperature-sensitive mutant of ubiquitin-conjugating enzyme 4 (Ubc4') as a model substrate shows that QSOX activity correlates with the unfolding of Ubc4' monitored by circular dichroism. Fusion of Ubc4' with the more stable glutathione-S-transferase domain demonstrates that QSOX can selectively introduce disulfides into the less stable domain of the fusion protein. In terms of intermolecular disulfide bond generation, QSOX is unable to cross-link well-folded globular proteins via their surface thiols. However, the construction of a septuple mutant of RNase A, retaining a single cysteine residue, demonstrates that flexible protein monomers can be directly coupled by the oxidase. Steady- and pre-steady-state kinetic experiments, combined with static fluorescence approaches, indicate that while QSOX is an efficient catalyst for disulfide bond formation between mobile elements of structure, it does not appear to have a significant binding site for unfolded proteins. These aspects of protein substrate discrimination by QSOX family members are rationalized in terms of the stringent steric requirements for disulfide exchange reactions.


Subject(s)
Oxidoreductases Acting on Sulfur Group Donors/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Birds/metabolism , Circular Dichroism , Cysteine/chemistry , Disulfides/chemistry , Glutathione Transferase/chemistry , Humans , Molecular Sequence Data , Oxidoreductases Acting on Sulfur Group Donors/genetics , Protein Conformation , Protein Folding , Ribonuclease, Pancreatic/chemistry , Substrate Specificity , Ubiquitin-Conjugating Enzymes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL