Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Anal Biochem ; 544: 57-63, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29273238

ABSTRACT

Recent developments in microfluidic and nanofluidic technologies have resulted in development of new chip-based microfluidic calorimeters with potential use in different fields. One application would be the accurate high-throughput measurement of enzyme activity. Calorimetry is a generic way to measure activity of enzymes, but unlike conventional calorimeters, chip-based calorimeters can be easily automated and implemented in high-throughput screening platforms. However, application of chip-based microfluidic calorimeters to measure enzyme activity has been limited due to problems associated with miniaturization such as incomplete mixing and a decrease in volumetric heat generated. To address these problems we introduced a calibration method and devised a convenient protocol for using a chip-based microfluidic calorimeter. Using the new calibration method, the progress curve of alkaline phosphatase, which has product inhibition for phosphate, measured by the calorimeter was the same as that recorded by UV-visible spectroscopy. Our results may enable use of current chip-based microfluidic calorimeters in a simple manner as a tool for high-throughput screening of enzyme activity with potential applications in drug discovery and enzyme engineering.


Subject(s)
Alkaline Phosphatase/analysis , Calorimetry , Microfluidic Analytical Techniques , Alkaline Phosphatase/metabolism , Animals , Calibration , Cattle , Diffusion , High-Throughput Screening Assays , Protein Engineering
2.
Mol Biosyst ; 12(12): 3576-3588, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27722502

ABSTRACT

Ferritin is a nanocage protein made of 24 subunits. Its major role is to manage intracellular concentrations of free Fe(ii) and Fe(iii) ions, which is pivotal for iron homeostasis across all domains of life. This function of the protein is regulated by a conserved di-iron catalytic center and has been the subject of extensive studies over the past 50 years. Yet, it has not been fully understood how Fe(ii) is oxidized in the di-iron catalytic center and it is not known why eukaryotic and microbial ferritins oxidize Fe(ii) with different kinetics. In an attempt to obtain a new insight into the mechanism of Fe(ii) oxidation and understand the origin of the observed differences in the catalysis of Fe(ii) oxidation among ferritins we studied and compared the mechanism of Fe(ii) oxidation in the eukaryotic human H-type ferritin (HuHF) and the archaeal ferritin from Pyrococcus furiosus (PfFtn). The results show that the spectroscopic characteristics of the intermediate of Fe(ii) oxidation and the Fe(iii)-products are the same in these two ferritins supporting the proposal of unity in the mechanism of Fe(ii) oxidation among eukaryotic and microbial ferritins. Moreover, we observed that a site in the di-iron catalytic center controls the distribution of Fe(ii) among subunits of HuHF and PfFtn differently. This observation explains the reported differences between HuHF and PfFtn in the kinetics of Fe(ii) oxidation and the amount of O2 consumed per Fe(ii) oxidized. These results provide a fresh understanding of the mechanism of Fe(ii) oxidation by ferritins.


Subject(s)
Catalytic Domain , Ferritins/chemistry , Ferrous Compounds/chemistry , Oxidation-Reduction , Binding Sites , Catalysis , Ceruloplasmin/chemistry , Humans , Kinetics , Models, Molecular , Molecular Conformation , Protein Binding , Protein Interaction Domains and Motifs , Pyrococcus furiosus/metabolism , Spectrum Analysis
3.
Sci Rep ; 5: 16380, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26574737

ABSTRACT

Accurate label-free methods or assays to obtain the initial reaction rates have significant importance in fundamental studies of enzymes and in application-oriented high throughput screening of enzyme activity. Here we introduce a label-free approach for obtaining initial rates of enzyme activity from heat measurements, which we name initial rate calorimetry (IrCal). This approach is based on our new finding that the data recorded by isothermal titration calorimetry for the early stages of a reaction, which have been widely ignored, are correlated to the initial rates. Application of the IrCal approach to various enzymes led to accurate enzyme kinetics parameters as compared to spectroscopic methods and enabled enzyme kinetic studies with natural substrate, e.g. proteases with protein substrates. Because heat is a label-free property of almost all reactions, the IrCal approach holds promise in fundamental studies of various enzymes and in use of calorimetry for high throughput screening of enzyme activity.


Subject(s)
Hot Temperature , Models, Theoretical , Kinetics
4.
J Biol Chem ; 290(44): 26801-10, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26370076

ABSTRACT

Fe(III) storage by ferritin is an essential process of the iron homeostasis machinery. It begins by translocation of Fe(II) from outside the hollow spherical shape structure of the protein, which is formed as the result of self-assembly of 24 subunits, to a di-iron binding site, the ferroxidase center, buried in the middle of each active subunit. The pathway of Fe(II) to the ferroxidase center has remained elusive, and the importance of self-assembly for the functioning of the ferroxidase center has not been investigated. Here we report spectroscopic and metal ion binding studies with a mutant of ferritin from Pyrococcus furiosus (PfFtn) in which self-assembly was abolished by a single amino acid substitution. We show that in this mutant metal ion binding to the ferroxidase center and Fe(II) oxidation at this site was obliterated. However, metal ion binding to a conserved third site (site C), which is located in the inner surface of each subunit in the vicinity of the ferroxidase center and is believed to be the path for Fe(II) to the ferroxidase center, was not disrupted. These results are the basis of a new model for Fe(II) translocation to the ferroxidase center: self-assembly creates channels that guide the Fe(II) ions toward the ferroxidase center directly through the protein shell and not via the internal cavity and site C. The results may be of significance for understanding the molecular basis of ferritin-related disorders such as neuroferritinopathy in which the 24-meric structure with 432 symmetry is distorted.


Subject(s)
Archaeal Proteins/chemistry , Ceruloplasmin/chemistry , Ferritins/chemistry , Iron/chemistry , Protein Subunits/chemistry , Amino Acid Substitution , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Biocatalysis , Catalytic Domain , Cations, Divalent , Ceruloplasmin/genetics , Ceruloplasmin/metabolism , Ferritins/genetics , Ferritins/metabolism , Gene Expression , Kinetics , Models, Molecular , Mutation , Oxidation-Reduction , Protein Binding , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/genetics , Protein Subunits/metabolism , Pyrococcus furiosus/chemistry , Pyrococcus furiosus/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thermodynamics
5.
Chembiochem ; 14(9): 1123-33, 2013 Jun 17.
Article in English | MEDLINE | ID: mdl-23737293

ABSTRACT

A highly conserved tyrosine residue of unknown function is present in the vicinity of the di-iron catalytic center of the ubiquitous iron-storage protein ferritin. The di-iron center with a gateway FeII/FeIII-binding site nearby provides the vital iron-storage mechanism of the protein. It is believed that, in eukaryotic ferritin, this center catalyzes simultaneous oxidation of two FeII ions, whereas in microbial ferritin it catalyzes simultaneous oxidation of three FeII ions. To understand the role of the conserved tyrosine, we studied the intermediates and products that are formed during catalysis of FeII oxidation in the di-iron catalytic centers of the hyperthermophilic archaeal Pyrococcus furiosus ferritin and of eukaryotic human H ferritin. Based on our spectroscopic studies and modeling, we propose a merger of the models for eukaryotic and bacterial ferritin into a common mechanism of FeII oxidation in which the conserved tyrosine acts as a single-electron molecular capacitor to facilitate oxidation of FeII.


Subject(s)
Ferritins/chemistry , Tyrosine/chemistry , Catalytic Domain , Ferritins/metabolism , Ferrous Compounds/chemistry , Kinetics , Oxidation-Reduction , Pyrococcus/metabolism
6.
PLoS One ; 7(8): e40287, 2012.
Article in English | MEDLINE | ID: mdl-22916096

ABSTRACT

The ß-amyloid precursor protein (APP), which is a key player in Alzheimer's disease, was recently reported to possess an Fe(II) binding site within its E2 domain which exhibits ferroxidase activity [Duce et al. 2010, Cell 142: 857]. The putative ligands of this site were compared to those in the ferroxidase site of ferritin. The activity was indirectly measured using transferrin, which scavenges the Fe(III) product of the reaction. A 22-residue synthetic peptide, named FD1, with the putative ferroxidase site of APP, and the E2 domain of APP were each reported to exhibit 40% of the ferroxidase activity of APP and of ceruloplasmin. It was also claimed that the ferroxidase activity of APP is inhibited by Zn(II) just as in ferritin. We measured the ferroxidase activity indirectly (i) by the incorporation of the Fe(III) product of the ferroxidase reaction into transferrin and directly (ii) by monitoring consumption of the substrate molecular oxygen. The results with the FD1 peptide were compared to the established ferroxidase activities of human H-chain ferritin and of ceruloplasmin. For FD1 we observed no activity above the background of non-enzymatic Fe(II) oxidation by molecular oxygen. Zn(II) binds to transferrin and diminishes its Fe(III) incorporation capacity and rate but it does not specifically bind to a putative ferroxidase site of FD1. Based on these results, and on comparison of the putative ligands of the ferroxidase site of APP with those of ferritin, we conclude that the previously reported results for ferroxidase activity of FD1 and - by implication - of APP should be re-evaluated.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Iron/metabolism , Peptides/metabolism , Binding Sites , Biocatalysis , Humans , Oxidation-Reduction , Oxygen/metabolism , Thermodynamics , Zinc/metabolism
7.
J Biol Inorg Chem ; 17(6): 975-85, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22739810

ABSTRACT

Storage of iron in a nontoxic and bioavailable form is essential for many forms of life. Three subfamilies of the ferritin-like superfamily, namely, ferritin, bacterioferritin, and Dps (DNA-binding proteins from starved cells), are able to store iron. Although the function of these iron-storage proteins is constitutive to many organisms to sustain life, the genome of some organisms appears not to encode any of these proteins. In an attempt to identify new iron-storage systems, we have found and characterized a new member of the ferritin-like superfamily of proteins, which unlike the multimeric storage system of ferritin, bacterioferritin, and Dps is monomeric in the absence of iron. Monomers catalyze oxidation of Fe(II) and they store the Fe(III) product as they assemble to form structures comparable to those of 24-meric ferritin. We propose that this mechanism is an alternative method of iron storage by the ferritin-like superfamily of proteins in organisms that lack the regular preassociated 24-meric/12-meric ferritins.


Subject(s)
Ferritins/metabolism , Pyrococcus furiosus/metabolism , Amino Acid Sequence , Ferritins/chemistry , Ferritins/genetics , Molecular Sequence Data , Polymerase Chain Reaction , Pyrococcus furiosus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...