Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
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
4.
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
SELECTION OF CITATIONS
SEARCH DETAIL