Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 334
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 142(6): 857-67, 2010 Sep 17.
Article in English | MEDLINE | ID: mdl-20817278

ABSTRACT

Alzheimer's Disease (AD) is complicated by pro-oxidant intraneuronal Fe(2+) elevation as well as extracellular Zn(2+) accumulation within amyloid plaque. We found that the AD ß-amyloid protein precursor (APP) possesses ferroxidase activity mediated by a conserved H-ferritin-like active site, which is inhibited specifically by Zn(2+). Like ceruloplasmin, APP catalytically oxidizes Fe(2+), loads Fe(3+) into transferrin, and has a major interaction with ferroportin in HEK293T cells (that lack ceruloplasmin) and in human cortical tissue. Ablation of APP in HEK293T cells and primary neurons induces marked iron retention, whereas increasing APP695 promotes iron export. Unlike normal mice, APP(-/-) mice are vulnerable to dietary iron exposure, which causes Fe(2+) accumulation and oxidative stress in cortical neurons. Paralleling iron accumulation, APP ferroxidase activity in AD postmortem neocortex is inhibited by endogenous Zn(2+), which we demonstrate can originate from Zn(2+)-laden amyloid aggregates and correlates with Aß burden. Abnormal exchange of cortical zinc may link amyloid pathology with neuronal iron accumulation in AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/antagonists & inhibitors , Amyloid beta-Protein Precursor/metabolism , Ceruloplasmin/antagonists & inhibitors , Zinc/metabolism , Alzheimer Disease/metabolism , Amino Acid Sequence , Amyloid beta-Protein Precursor/chemistry , Animals , Cell Line , Ceruloplasmin/chemistry , Ceruloplasmin/metabolism , Humans , Iron/metabolism , Mice , Sequence Alignment
2.
Inorg Chem ; 63(7): 3359-3365, 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38315811

ABSTRACT

The Fe(II) oxidation mechanism in the ferroxidase center of heavy chain ferritin has been studied extensively. However, the actual production of H2O2 was found to be substantially lower than expected at low flux of Fe(II) to ferritin subunits. Here, we demonstrated that H2O2 could interact with the di-iron nuclear center, leading to the production of hydroxyl radicals and oxygen. Two reaction intermediates were captured in the ferroxidase center by using the time-lapse crystallographic techniques in a shellfish ferritin. The crystal structures revealed the binding of H2O2 as a µ -1,2-peroxo-diferric species and the binding of O2 to the diferric structure. This investigation sheds light on the reaction between the di-iron nuclear center and H2O2 and provides insights for the exploitation of metalloenzymes.


Subject(s)
Ferritins , Iron , Iron/chemistry , Ferritins/chemistry , Hydrogen Peroxide/chemistry , Ceruloplasmin/chemistry , Oxidation-Reduction , Ferrous Compounds/chemistry
3.
Angew Chem Int Ed Engl ; 63(16): e202401379, 2024 04 15.
Article in English | MEDLINE | ID: mdl-38407997

ABSTRACT

Ferritins are multimeric cage-forming proteins that play a crucial role in cellular iron homeostasis. All H-chain-type ferritins harbour a diiron site, the ferroxidase centre, at the centre of a 4 α-helical bundle, but bacterioferritins are unique in also binding 12 hemes per 24 meric assembly. The ferroxidase centre is known to be required for the rapid oxidation of Fe2+ during deposition of an immobilised ferric mineral core within the protein's hollow interior. In contrast, the heme of bacterioferritin is required for the efficient reduction of the mineral core during iron release, but has little effect on the rate of either oxidation or mineralisation of iron. Thus, the current view is that these two cofactors function in iron uptake and release, respectively, with no functional overlap. However, rapid electron transfer between the heme and ferroxidase centre of bacterioferritin from Escherichia coli was recently demonstrated, suggesting that the two cofactors may be functionally connected. Here we report absorbance and (magnetic) circular dichroism spectroscopies, together with in vitro assays of iron-release kinetics, which demonstrate that the ferroxidase centre plays an important role in the reductive mobilisation of the bacterioferritin mineral core, which is dependent on the heme-ferroxidase centre electron transfer pathway.


Subject(s)
Ceruloplasmin , Iron , Iron/chemistry , Ceruloplasmin/chemistry , Escherichia coli/metabolism , Ferritins/chemistry , Bacterial Proteins/metabolism , Cytochrome b Group/chemistry , Minerals , Oxidation-Reduction , Heme/metabolism
4.
J Am Chem Soc ; 145(24): 13284-13301, 2023 06 21.
Article in English | MEDLINE | ID: mdl-37294874

ABSTRACT

In multicopper oxidases (MCOs), the type 1 (T1) Cu accepts electrons from the substrate and transfers these to the trinuclear Cu cluster (TNC) where O2 is reduced to H2O. The T1 potential in MCOs varies from 340 to 780 mV, a range not explained by the existing literature. This study focused on the ∼350 mV difference in potential of the T1 center in Fet3p and Trametes versicolor laccase (TvL) that have the same 2His1Cys ligand set. A range of spectroscopies performed on the oxidized and reduced T1 sites in these MCOs shows that they have equivalent geometric and electronic structures. However, the two His ligands of the T1 Cu in Fet3p are H-bonded to carboxylate residues, while in TvL they are H-bonded to noncharged groups. Electron spin echo envelope modulation spectroscopy shows that there are significant differences in the second-sphere H-bonding interactions in the two T1 centers. Redox titrations on type 2-depleted derivatives of Fet3p and its D409A and E185A variants reveal that the two carboxylates (D409 and E185) lower the T1 potential by 110 and 255-285 mV, respectively. Density functional theory calculations uncouple the effects of the charge of the carboxylates and their difference in H-bonding interactions with the His ligands on the T1 potential, indicating 90-150 mV for anionic charge and ∼100 mV for a strong H-bond. Finally, this study provides an explanation for the generally low potentials of metallooxidases relative to the wide range of potentials of the organic oxidases in terms of different oxidized states of their TNCs involved in catalytic turnover.


Subject(s)
Ceruloplasmin , Histidine , Ceruloplasmin/chemistry , Ligands , Copper/chemistry , Trametes , Static Electricity , Laccase/metabolism
5.
Inorg Chem ; 62(1): 178-191, 2023 Jan 09.
Article in English | MEDLINE | ID: mdl-36525578

ABSTRACT

The self-assembled ferritin nanocages, nature's solution to iron toxicity and its low solubility, scavenge free iron to synthesize hydrated ferric oxyhydroxide mineral inside their central cavity by protein-mediated ferroxidase and hydrolytic/nucleation reactions. These complex processes in ferritin commence with the rapid influx of Fe2+ ions via the inter-subunit contact points (i.e., pores/channels). Investigation of these pores as Fe2+ uptake routes in ferritins remains a subject of intense research, in iron metabolism, toxicity, and bacterial pathogenesis, which are yet to be established in the bacterioferritin (BfrA) from Mycobacterium tuberculosis (Mtb). The electrostatic properties of this protein indicate that the 4-fold and B-pores might serve as potential Fe2+ entry routes. Therefore, in the current work, electrostatics at/along these pores was altered by site-directed mutagenesis to establish their role in Fe2+ uptake/oxidation (ferroxidase activity) in Mtb BfrA. Despite forming self-assembled protein nanocompartment, these 4-fold and B-pore variants exhibited partial loss of ferroxidase activity and lower accumulation of transient species, which not only indicated their role in Fe2+ entry but also suggested the existence of multiple pathways. Although the B-pore variants inhibited the rapid ferroxidase activity to a larger extent, they had minimal impact on their cage stability. The current work revealed the relative contribution of these pores toward rapid Fe2+ uptake/oxidation and cage stability, possibly as consequences of their differential symmetry, number of modified residues (at each pore), and heme content. Therefore, these findings may help to understand the role of these pores in iron acquisition and Mtb proliferation under iron-limiting conditions to control its pathogenesis.


Subject(s)
Mycobacterium tuberculosis , Ceruloplasmin/chemistry , Ferritins/chemistry , Iron/chemistry , Mycobacterium tuberculosis/metabolism , Oxidation-Reduction , Oxidoreductases/metabolism
6.
Proc Natl Acad Sci U S A ; 116(6): 2058-2067, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30659147

ABSTRACT

The gene encoding the cyanobacterial ferritin SynFtn is up-regulated in response to copper stress. Here, we show that, while SynFtn does not interact directly with copper, it is highly unusual in several ways. First, its catalytic diiron ferroxidase center is unlike those of all other characterized prokaryotic ferritins and instead resembles an animal H-chain ferritin center. Second, as demonstrated by kinetic, spectroscopic, and high-resolution X-ray crystallographic data, reaction of O2 with the di-Fe2+ center results in a direct, one-electron oxidation to a mixed-valent Fe2+/Fe3+ form. Iron-O2 chemistry of this type is currently unknown among the growing family of proteins that bind a diiron site within a four α-helical bundle in general and ferritins in particular. The mixed-valent form, which slowly oxidized to the more usual di-Fe3+ form, is an intermediate that is continually generated during mineralization. Peroxide, rather than superoxide, is shown to be the product of O2 reduction, implying that ferroxidase centers function in pairs via long-range electron transfer through the protein resulting in reduction of O2 bound at only one of the centers. We show that electron transfer is mediated by the transient formation of a radical on Tyr40, which lies ∼4 Å from the diiron center. As well as demonstrating an expansion of the iron-O2 chemistry known to occur in nature, these data are also highly relevant to the question of whether all ferritins mineralize iron via a common mechanism, providing unequivocal proof that they do not.


Subject(s)
Ferric Compounds/chemistry , Ferrous Compounds/chemistry , Oxygen/chemistry , Peroxides/chemistry , Proteins/chemistry , Ceruloplasmin/chemistry , Electron Transport , Ferritins/chemistry , Iron/chemistry , Models, Molecular , Molecular Conformation , Oxidation-Reduction , Structure-Activity Relationship
7.
J Biol Chem ; 295(46): 15511-15526, 2020 11 13.
Article in English | MEDLINE | ID: mdl-32878987

ABSTRACT

Encapsulated ferritins belong to the universally distributed ferritin superfamily, whose members function as iron detoxification and storage systems. Encapsulated ferritins have a distinct annular structure and must associate with an encapsulin nanocage to form a competent iron store that is capable of holding significantly more iron than classical ferritins. The catalytic mechanism of iron oxidation in the ferritin family is still an open question because of the differences in organization of the ferroxidase catalytic site and neighboring secondary metal-binding sites. We have previously identified a putative metal-binding site on the inner surface of the Rhodospirillum rubrum encapsulated ferritin at the interface between the two-helix subunits and proximal to the ferroxidase center. Here we present a comprehensive structural and functional study to investigate the functional relevance of this putative iron-entry site by means of enzymatic assays, MS, and X-ray crystallography. We show that catalysis occurs in the ferroxidase center and suggest a dual role for the secondary site, which both serves to attract metal ions to the ferroxidase center and acts as a flow-restricting valve to limit the activity of the ferroxidase center. Moreover, confinement of encapsulated ferritins within the encapsulin nanocage, although enhancing the ability of the encapsulated ferritin to undergo catalysis, does not influence the function of the secondary site. Our study demonstrates a novel molecular mechanism by which substrate flux to the ferroxidase center is controlled, potentially to ensure that iron oxidation is productively coupled to mineralization.


Subject(s)
Bacterial Proteins/metabolism , Ceruloplasmin/metabolism , Metals/metabolism , Rhodospirillum rubrum/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Catalytic Domain , Ceruloplasmin/chemistry , Ceruloplasmin/genetics , Crystallography, X-Ray , Iron/chemistry , Iron/metabolism , Metals/chemistry , Mutagenesis, Site-Directed , Oxidation-Reduction , Protein Conformation , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Zinc/chemistry , Zinc/metabolism
8.
Microbiology (Reading) ; 167(11)2021 11.
Article in English | MEDLINE | ID: mdl-34825885

ABSTRACT

Ferritins are proteins forming 24meric rhombic dodecahedral cages that play a key role in iron storage and detoxification in all cell types. Their function requires the transport of Fe2+ from the exterior of the protein to buried di-iron catalytic sites, known as ferroxidase centres, where Fe2+ is oxidized to form Fe3+-oxo precursors of the ferritin mineral core. The route of iron transit through animal ferritins is well understood: the Fe2+ substrate enters the protein via channels at the threefold axes and conserved carboxylates on the inner surface of the protein cage have been shown to contribute to transient binding sites that guide Fe2+ to the ferroxidase centres. The routes of iron transit through prokaryotic ferritins are less well studied but for some, at least, there is evidence that channels at the twofold axes are the major route for Fe2+ uptake. SynFtn, isolated from the cyanobacterium Synechococcus CC9311, is an atypical prokaryotic ferritin that was recently shown to take up Fe2+ via its threefold channels. However, the transfer site carboxylate residues conserved in animal ferritins are absent, meaning that the route taken from the site of iron entry into SynFtn to the catalytic centre is yet to be defined. Here, we report the use of a combination of site-directed mutagenesis, absorbance-monitored activity assays and protein crystallography to probe the effect of substitution of two residues potentially involved in this pathway. Both Glu141 and Asp65 play a role in guiding the Fe2+ substrate to the ferroxidase centre. In the absence of Asp65, routes for Fe2+ to, and Fe3+ exit from, the ferroxidase centre are affected resulting in inefficient formation of the mineral core. These observations further define the iron transit route in what may be the first characterized example of a new class of ferritins peculiar to cyanobacteria.


Subject(s)
Ferritins , Iron , Synechococcus , Animals , Catalytic Domain , Ceruloplasmin/chemistry , Ceruloplasmin/genetics , Ferritins/chemistry , Ferritins/genetics , Iron/metabolism , Minerals/chemistry , Oxidation-Reduction , Synechococcus/chemistry
9.
Inorg Chem ; 60(10): 7207-7216, 2021 May 17.
Article in English | MEDLINE | ID: mdl-33852289

ABSTRACT

Here, we present a 1.9 Å resolution crystal structure of Mycoplasma Penetrans ferritin, which reveals that its ferroxidase center is located on the inner surface of ferritin but not buried within the four-helix of each subunit. Such a ferroxidase center exhibits a lower iron oxidation activity as compared to the reported ferritin. More importantly, we found that Fe2+ enters into the center via the rarely reported B-channels rather than the normal 3- or 4-fold channels. All these findings may provide the structural bases to explore the new iron oxidation mechanism adopted by this special ferritin, which is beneficial for understanding the relationship between the structure and function of ferritin.


Subject(s)
Ceruloplasmin/metabolism , Ferritins/metabolism , Ferrous Compounds/metabolism , Mycoplasma/chemistry , Ceruloplasmin/chemistry , Ferritins/chemistry , Ferrous Compounds/chemistry , Molecular Dynamics Simulation , Mycoplasma/metabolism , Oxidation-Reduction
10.
Int J Mol Sci ; 22(24)2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34948458

ABSTRACT

Coppers play crucial roles in the maintenance homeostasis in living species. Approximately 20 enzyme families of eukaryotes and prokaryotes are known to utilize copper atoms for catalytic activities. However, small-molecule inhibitors directly targeting catalytic centers are rare, except for those that act against tyrosinase and dopamine-ß-hydroxylase (DBH). This study tested whether known tyrosinase inhibitors can inhibit the copper-containing enzymes, ceruloplasmin, DBH, and laccase. While most small molecules minimally reduced the activities of ceruloplasmin and DBH, aside from known inhibitors, 5 of 28 tested molecules significantly inhibited the function of laccase, with the Ki values in the range of 15 to 48 µM. Enzyme inhibitory kinetics classified the molecules as competitive inhibitors, whereas differential scanning fluorimetry and fluorescence quenching supported direct bindings. To the best of our knowledge, this is the first report on organic small-molecule inhibitors for laccase. Comparison of tyrosinase and DBH inhibitors using cheminformatics predicted that the presence of thione moiety would suffice to inhibit tyrosinase. Enzyme assays confirmed this prediction, leading to the discovery of two new dual tyrosinase and DBH inhibitors.


Subject(s)
Ceruloplasmin/metabolism , Copper/chemistry , Dopamine beta-Hydroxylase/metabolism , Fungi/enzymology , Laccase/metabolism , Small Molecule Libraries/pharmacology , Agaricales/enzymology , Biocatalysis , Catalytic Domain , Ceruloplasmin/chemistry , Cheminformatics , Dopamine beta-Hydroxylase/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Humans , Laccase/chemistry , Models, Molecular , Protein Conformation , Small Molecule Libraries/chemistry
11.
Int J Mol Sci ; 22(2)2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33440850

ABSTRACT

Neurodegenerative disorders can induce modifications of several proteins; one of which is ceruloplasmin (Cp), a ferroxidase enzyme found modified in the cerebrospinal fluid (CSF) of neurodegenerative diseases patients. Cp modifications are caused by the oxidation induced by the pathological environment and are usually associated with activity loss. Together with oxidation, deamidation of Cp was found in the CSF from Alzheimer's and Parkinson's disease patients. Protein deamidation is a process characterized by asparagine residues conversion in either aspartate or isoaspartate, depending on protein sequence/structure and cellular environment. Cp deamidation occurs at two Asparagine-Glycine-Arginine (NGR)-motifs which, once deamidated to isoAspartate-Glycine-Arginine (isoDGR), bind integrins, a family of receptors mediating cell adhesion. Therefore, on the one hand, Cp modifications lead to loss of enzymatic activity, while on the other hand, these alterations confer gain of function to Cp. In fact, deamidated Cp binds to integrins and triggers intracellular signaling on choroid plexus epithelial cells, changing cell functioning. Working in concert with the oxidative environment, Cp deamidation could reach different target cells in the brain, altering their physiology and causing detrimental effects, which might contribute to the pathological mechanism.


Subject(s)
Ceruloplasmin/genetics , Ceruloplasmin/metabolism , Disease Susceptibility , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Amino Acid Motifs , Amino Acids/metabolism , Animals , Brain/metabolism , Ceruloplasmin/chemistry , Gain of Function Mutation , Genetic Predisposition to Disease , Humans , Integrins/metabolism , Loss of Function Mutation , Oligopeptides/chemistry
12.
Angew Chem Int Ed Engl ; 60(15): 8376-8379, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33460502

ABSTRACT

The iron redox cycle in ferritins is not completely understood. Bacterioferritins are distinct from other ferritins in that they contain haem groups. It is acknowledged that the two iron motifs in bacterioferritins, the di-nuclear ferroxidase centre and the haem B group, play key roles in two opposing processes, iron sequestration and iron mobilisation, respectively, and the two redox processes are independent. Herein, we show that in Escherichia coli bacterioferritin, there is an electron transfer pathway from the haem to the ferroxidase centre suggesting a new role(s) haem might play in bacterioferritins.


Subject(s)
Bacterial Proteins/metabolism , Ceruloplasmin/metabolism , Cytochrome b Group/metabolism , Ferritins/metabolism , Heme/metabolism , Bacterial Proteins/chemistry , Ceruloplasmin/chemistry , Cytochrome b Group/chemistry , Electron Transport , Escherichia coli/chemistry , Escherichia coli/metabolism , Ferritins/chemistry , Heme/chemistry
13.
Angew Chem Int Ed Engl ; 60(15): 8361-8369, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33482043

ABSTRACT

Both O2 and H2 O2 can oxidize iron at the ferroxidase center (FC) of Escherichia coli bacterioferritin (EcBfr) but mechanistic details of the two reactions need clarification. UV/Vis, EPR, and Mössbauer spectroscopies have been used to follow the reactions when apo-EcBfr, pre-loaded anaerobically with Fe2+ , was exposed to O2 or H2 O2 . We show that O2 binds di-Fe2+ FC reversibly, two Fe2+ ions are oxidized in concert and a H2 O2 molecule is formed and released to the solution. This peroxide molecule further oxidizes another di-Fe2+ FC, at a rate circa 1000 faster than O2 , ensuring an overall 1:4 stoichiometry of iron oxidation by O2 . Initially formed Fe3+ can further react with H2 O2 (producing protein bound radicals) but relaxes within seconds to an H2 O2 -unreactive di-Fe3+ form. The data obtained suggest that the primary role of EcBfr in vivo may be to detoxify H2 O2 rather than sequester iron.


Subject(s)
Bacterial Proteins/metabolism , Ceruloplasmin/metabolism , Cytochrome b Group/metabolism , Escherichia coli/chemistry , Ferritins/metabolism , Hydrogen Peroxide/metabolism , Iron/metabolism , Oxygen/metabolism , Bacterial Proteins/chemistry , Ceruloplasmin/chemistry , Cytochrome b Group/chemistry , Escherichia coli/metabolism , Ferritins/chemistry , Hydrogen Peroxide/chemistry , Iron/chemistry , Models, Molecular , Oxidation-Reduction , Oxygen/chemistry
14.
Chemistry ; 26(22): 4884, 2020 Apr 16.
Article in English | MEDLINE | ID: mdl-32297426

ABSTRACT

Invited for the cover of this issue is the group of Ulrich Schwaneberg at the Institute of Biotechnology, RWTH-Aachen University and DWI Lebniz Institute of Interactive Materials. The picture calls for special attention to be paid to the extra Cu binding site of Copper efflux Oxidase (CueO), due to its predominant function in tuning the electrocatalytic kinetics towards oxygen reduction. Read the full text of the article at 10.1002/chem.201905598.


Subject(s)
Ceruloplasmin/metabolism , Laccase/chemistry , Oxidoreductases/chemistry , Binding Sites , Ceruloplasmin/chemistry , Electron Transport , Escherichia coli Proteins/chemistry , Kinetics , Oxidation-Reduction , Oxidoreductases/metabolism
15.
Chemistry ; 26(22): 4974-4979, 2020 Apr 16.
Article in English | MEDLINE | ID: mdl-31985091

ABSTRACT

Copper efflux oxidase (CueO) from Escherichia coli is a special bacterial laccase due to its fifth copper binding site. Herein, it is discovered that the fifth Cu occupancy plays a crucial and favorable role of electron relay in bioelectrocatalytic oxygen reduction. By substituting the residues at the four coordinated positions of the fifth Cu, 11 beneficial variants are identified with ≥2.5-fold increased currents at -250 mV (up to 6.13 mA cm-2 ). Detailed electrocatalytic characterization suggests the microenvironment of the fifth Cu binding site governs the electrocatalytic current of CueO. Additionally, further electron transfer analysis assisted by molecular dynamics (MD) simulation demonstrates that an increase in localized structural stability and a decrease of distance between the fifth Cu and the T1 Cu are two main factors contributing to the improved kinetics of CueO variants. It may guide a novel way to tailor laccases and perhaps other oxidoreductases for bioelectrocatalytic applications.


Subject(s)
Ceruloplasmin/metabolism , Laccase/chemistry , Oxidoreductases/chemistry , Binding Sites , Ceruloplasmin/chemistry , Electron Transport , Escherichia coli Proteins/chemistry , Kinetics , Oxidation-Reduction , Oxidoreductases/metabolism
16.
J Am Chem Soc ; 141(27): 10736-10743, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31203609

ABSTRACT

Unraveling the mechanism of ceruloplasmin (Cp) is fundamentally important toward understanding the pathogenesis of metal-mediated diseases and metal neurotoxicity. Here we report that Cl-, the most abundant anion in blood plasma, is a key component of Cp catalysis. Based on detailed spectroscopic analyses, Cl- preferentially interacts with the partially reduced trinuclear Cu cluster (TNC) in Cp under physiological conditions and shifts the electron equilibrium distribution among the two redox active type 1 (T1) Cu sites and the TNC. This shift in potential enables the intramolecular electron transfer (IET) from the T1 Cu to the native intermediate (NI) and accelerates the IET from the T1 Cu to the TNC, resulting in faster turnover in Cp catalysis.


Subject(s)
Ceruloplasmin/metabolism , Chlorides/metabolism , Catalytic Domain , Ceruloplasmin/chemistry , Crystallography, X-Ray , Electron Transport , Humans , Models, Molecular , Oxidation-Reduction , Oxygen/metabolism , Protein Conformation
17.
Glycoconj J ; 36(1): 13-26, 2019 02.
Article in English | MEDLINE | ID: mdl-30612270

ABSTRACT

Significant changes of glycan structures are observed in humans if diseases like cancer, arthritis or inflammation are present. Thus, interest in biomarkers based on glycan structures has rapidly emerged in recent years and monitoring disease specific changes of glycosylation and their quantification is of great interest. Mass spectrometry is most commonly used to characterize and quantify glycopeptides and glycans liberated from the glycoprotein of interest. However, ionization properties of glycopeptides can strongly depend on their composition and can therefore lead to intensities that do not reflect the actual proportions present in the intact glycoprotein. Here we show that an increase in the length of the peptide can lead to a more accurate determination and quantification of the glycans. The four glycosylation sites of human serum ceruloplasmin from 17 different individuals were analyzed using glycopeptides of varying peptide lengths, obtained by action of different proteases and by limited digestion. In most cases, highly sialylated compositions showed an increased relative abundance with increasing peptide length. We observed a relative increase of triantennary glycans of up to a factor of three and, even more, MS peaks corresponding to tetraantennary compositions on ceruloplasmin at glycosite 137N in all 17 samples, which we did not detect using a bottom up approach. The data presented here leads to the conclusion that a middle down - or when possible a top down - approach is favorable for qualitative and quantitative analysis of the glycosylation of glycoproteins.


Subject(s)
Ceruloplasmin/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Glycosylation , Humans , Peptides/chemistry , Polysaccharides/analysis
18.
Biometals ; 32(2): 195-210, 2019 04.
Article in English | MEDLINE | ID: mdl-30895493

ABSTRACT

Ceruloplasmin (CP) is a mammalian blood plasma ferroxidase. More than 95% of the copper found in plasma is carried by this protein, which is a member of the multicopper oxidase family. Proteins from this group are able to oxidize substrates through the transfer of four electrons to oxygen. The essential role of CP in iron metabolism in humans is particularly evident in the case of loss-of-function mutations in the CP gene resulting in a neurodegenerative syndrome known as aceruloplasminaemia. However, the functions of CP are not limited to the oxidation of ferrous iron to ferric iron, which allows loading of the ferric iron into transferrin and prevents the deleterious reactions of Fenton chemistry. In recent years, a number of novel CP functions have been reported, and many of these functions depend on the ability of CP to form stable complexes with a number of proteins.


Subject(s)
Ceruloplasmin/chemistry , Animals , Ceruloplasmin/deficiency , Ceruloplasmin/genetics , Ceruloplasmin/metabolism , Humans , Iron/metabolism , Iron Metabolism Disorders/metabolism , Neurodegenerative Diseases/metabolism , Protein Binding
19.
Int J Mol Sci ; 20(17)2019 Aug 26.
Article in English | MEDLINE | ID: mdl-31454948

ABSTRACT

The transmembrane (TM) proteins are gateways for molecular transport across the cell membrane that are often selected as potential targets for drug design. The bilitranslocase (BTL) protein facilitates the uptake of various anions, such as bilirubin, from the blood into the liver cells. As previously established, there are four hydrophobic transmembrane segments (TM1-TM4), which constitute the structure of the transmembrane channel of the BTL protein. In our previous studies, the 3D high-resolution structure of the TM2 and TM3 transmembrane fragments of the BTL in sodium dodecyl sulfate (SDS) micellar media were solved using Nuclear Magnetic Resonance (NMR) spectroscopy and molecular dynamics simulations (MD). The high-resolution 3D structure of the fourth transmembrane region (TM4) of the BTL was evaluated using NMR spectroscopy in two different micellar media, anionic SDS and zwitterionic DPC (dodecylphosphocholine). The presented experimental data revealed the existence of an α -helical conformation in the central part of the TM4 in both micellar media. In the case of SDS surfactant, the α -helical conformation is observed for the Pro258-Asn269 region. The use of the zwitterionic DPC micelle leads to the formation of an amphipathic α -helix, which is characterized by the extension of the central α -helix in the TM4 fragment to Phe257-Thr271. The complex character of the dynamic processes in the TM4 peptide within both surfactants was analyzed based on the relaxation data acquired on 15 N and 31 P isotopes. Contrary to previously published and present observations in the SDS micelle, the zwitterionic DPC environment leads to intensive low-frequency molecular dynamic processes in the TM4 fragment.


Subject(s)
Ceruloplasmin/chemistry , Membrane Proteins/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Conformation , Protein Interaction Domains and Motifs , Ceruloplasmin/metabolism , Magnetic Resonance Spectroscopy , Membrane Proteins/metabolism , Micelles , Peptides/chemistry , Peptides/metabolism , Structure-Activity Relationship
20.
Fish Shellfish Immunol ; 82: 460-468, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30144565

ABSTRACT

Ceruloplasmin is an ancient multicopper oxidase evolved to insure a safe handling of oxygen in some metabolic pathways of vertebrates. The current knowledge of its structure provides a glimpse of its plasticity, revealing a multitude of binding sites that point to an elaborate mechanism of multifunctional activity. Ceruloplasmin is highly conserved throughout the vertebrate evolution. Cupredoxin, a multi-cupper blue protein is believed to be the evolutionary precursor of ceruloplasmin with three trinuclear and three mononuclear copper binding sites. There are 20 copper-binding residues in ceruloplasmin gene out of which 16 residues are conserved in fish. This ceruloplasmin gene is being characterized in zebrafish (Danio rerio), rohu (Labeo rohita), Indian medaka (Oryzias melastigama), catfish (Ictalurus punctatus), icefish (Chionodraco rastrospinosus), goldfish (Carassius auratus) and yellow perch (Perca flaviscens). The complete coding sequence of fish ceruloplasmin gene is around 3.2 kb which codes for 1000 to 1100 amino acid residues. The size of ceruloplasmin gene sequence in fish ranges around 13 kb containing 20 exons and 19 introns. Liver is the major site of synthesis in fish. Increased expression of this gene during bacterial infection in channel catfish and rohu suggested its potential involvement in bacterial disease response in fish. It has been found to serve as an indirect marker for selection against Aeromonas hydrophila resistance in rohu carp. Ceruloplasmin expression is also evident during parasitic infection in few fish species. The role of this gene is well studied during inflammatory response to hormonal, drug and heavy metal mediated toxicity in fish. Overall, ceruloplasmin represents an example of a 'moonlighting' protein that overcomes the one gene-one structure-one function concept to follow the changes of the organism in its physiological and pathological conditions.


Subject(s)
Ceruloplasmin/genetics , Evolution, Molecular , Fish Proteins/genetics , Fishes/genetics , Amino Acid Sequence , Animals , Ceruloplasmin/chemistry , Ceruloplasmin/metabolism , Fish Proteins/chemistry , Fish Proteins/metabolism , Fishes/metabolism , Phylogeny , Sequence Alignment/veterinary
SELECTION OF CITATIONS
SEARCH DETAIL