Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Biomed Pharmacother ; 141: 111872, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34246187

ABSTRACT

Ferroptosis is a type of regulated cell death driven by iron dependent accumulation of cellular reactive oxygen species (ROS) when glutathione (GSH)-dependent lipid peroxidation repair systems are compromised. Nuclear receptor co-activator 4 (NCOA4)-mediated selective autophagy of ferritin, termed ferritinophagy, involves the regulation of ferroptosis. Emerging evidence has revealed that ferritinophagy and ferroptosis exert a significant role in the occurrence and development of cardiovascular disease. In the present review, we aimed to present a brief overview of ferritinophagy and ferroptosis focusing on the underlying mechanism and regulations involved. We summarize and discuss relevant research progress on the role of ferritinophagy and ferroptosis in cardiovascular diseases accompanied with potential applications of ferritinophagy and ferroptosis modulators in the treatment of ferroptosis-associated cardiovascular diseases.


Subject(s)
Autophagy/physiology , Cardiovascular Diseases/metabolism , Ferritins/metabolism , Ferroptosis/physiology , Animals , Autophagy/drug effects , Cardiovascular Agents/pharmacology , Cardiovascular Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/pathology , Ferritins/antagonists & inhibitors , Ferroptosis/drug effects , Humans , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Myocardium/metabolism , Myocardium/pathology , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism
2.
Metallomics ; 9(6): 646-659, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28318006

ABSTRACT

Iron is an essential nutrient for bacteria but the reactivity of Fe2+ and the insolubility of Fe3+ present significant challenges to bacterial cells. Iron storage proteins contribute to ameliorating these challenges by oxidizing Fe2+ using O2 and H2O2 as electron acceptors, and by compartmentalizing Fe3+. Two types of iron-storage proteins coexist in bacteria, the ferritins (Ftn) and the heme-containing bacterioferritins (Bfr), but the reasons for their coexistence are largely unknown. P. aeruginosa cells harbor two iron storage proteins (FtnA and BfrB), but nothing is known about their relative contributions to iron homeostasis. Prior studies in vitro have shown that iron mobilization from BfrB requires specific interactions with a ferredoxin (Bfd), but the relevance of the BfrB:Bfd interaction to iron homeostasis in P. aeruginosa is unknown. In this work we explore the repercussions of (i) deleting the bfrB gene, and (ii) perturbing the BfrB:Bfd interaction in P. aeruginosa cells by either deleting the bfd gene or by replacing the wild type bfrB gene with a L68A/E81A double mutant allele in the P. aeruginosa chromosome. The effects of the mutations were evaluated by following the accumulation of iron in BfrB, analyzing levels of free and total intracellular iron, and by characterizing the ensuing iron homeostasis dysregulation phenotypes. The results reveal that P. aeruginosa accumulates iron mainly in BfrB, and that the nutrient does not accumulate in FtnA to detectable levels, even after deletion of the bfrB gene. Perturbing the BfrB:Bfd interaction causes irreversible flow of iron into BfrB, which leads to the accumulation of unusable intracellular iron while severely depleting the levels of free intracellular iron, which drives the cells to an acute iron starvation response despite harboring "normal" levels of total intracellular iron. These results are discussed in the context of a dynamic equilibrium between free cytosolic Fe2+ and Fe3+ compartmentalized in BfrB, which functions as a buffer to oppose rapid changes of free cytosolic iron. Finally, we also show that P. aeruginosa cells utilize iron stored in BfrB for growth in iron-limiting conditions, and that the utilization of BfrB-iron requires a functional BfrB:Bfd interaction.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Cytochrome b Group/antagonists & inhibitors , Cytosol/metabolism , Ferredoxins/antagonists & inhibitors , Ferritins/antagonists & inhibitors , Gene Expression Regulation, Bacterial , Iron/metabolism , Pseudomonas aeruginosa/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , Cytochrome b Group/genetics , Cytochrome b Group/metabolism , Ferredoxins/genetics , Ferredoxins/metabolism , Ferritins/genetics , Ferritins/metabolism , Homeostasis , Models, Molecular , Mutation , Oxidation-Reduction , Protein Conformation , Protein Interaction Domains and Motifs , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/growth & development
3.
Redox Biol ; 6: 112-121, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26209813

ABSTRACT

We have recently shown that increased hydrogen peroxide (H2O2) generation is involved in hypoxia-ischemia (HI)-mediated neonatal brain injury. H2O2 can react with free iron to form the hydroxyl radical, through Fenton Chemistry. Thus, the objective of this study was to determine if there was a role for the hydroxyl radical in neonatal HI brain injury and to elucidate the underlying mechanisms. Our data demonstrate that HI increases the deposition of free iron and hydroxyl radical formation, in both P7 hippocampal slice cultures exposed to oxygen-glucose deprivation (OGD), and the neonatal rat exposed to HI. Both these processes were found to be nitric oxide (NO) dependent. Further analysis demonstrated that the NO-dependent increase in iron deposition was mediated through increased transferrin receptor expression and a decrease in ferritin expression. This was correlated with a reduction in aconitase activity. Both NO inhibition and iron scavenging, using deferoxamine administration, reduced hydroxyl radical levels and neuronal cell death. In conclusion, our results suggest that increased NO generation leads to neuronal cell death during neonatal HI, at least in part, by altering iron homeostasis and hydroxyl radical generation.


Subject(s)
Hippocampus/metabolism , Hypoxia-Ischemia, Brain/metabolism , Iron/metabolism , Neurons/metabolism , Nitric Oxide/pharmacology , Aconitate Hydratase/antagonists & inhibitors , Aconitate Hydratase/genetics , Aconitate Hydratase/metabolism , Animals , Animals, Newborn , Cell Death/drug effects , Cell Hypoxia , Culture Media/chemistry , Deferoxamine/pharmacology , Ferritins/antagonists & inhibitors , Ferritins/genetics , Ferritins/metabolism , Gene Expression Regulation , Glucose/deficiency , Hippocampus/drug effects , Hippocampus/pathology , Hydroxyl Radical/metabolism , Hypoxia-Ischemia, Brain/chemically induced , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/prevention & control , Microtomy , NG-Nitroarginine Methyl Ester/pharmacology , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , Receptors, Transferrin/agonists , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Tissue Culture Techniques
4.
Biometals ; 28(4): 679-85, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25860295

ABSTRACT

Both human and horse fibrinogen are heme-binding proteins, and horse fibrinogen also exhibits heme-mediated ferritin binding. This study found that bovine and human fibrinogen are heme-mediated ferritin-binding proteins and demonstrated direct binding of bovine ferritin to protoporphyrin (PPIX) and its derivatives or to Zn ions. Binding of bovine and human fibrinogen to bovine spleen ferritin coated on microtiter plate wells was detected using an anti-human fibrinogen antibody, and this binding was inhibited in a dose-dependent manner by hemin (iron-PPIX) and also inhibited by Zn-PPIX. PPIX showed less of an inhibitory effect on the binding of bovine and human fibrinogen to bovine ferritin. The inhibitory effect of Sn-PPIX was similar to that of PPIX, but with respect to human fibrinogen, PPIX did not inhibit the binding of human fibrinogen to ferritin. Bovine fibrinogen immobilized on CNBr-activated Sepharose 4B beads showed affinity for hemin, Sn-PPIX, Zn-PPIX, and iron-free PPIX in the order Sn-PPIX < iron-free PPIX < hemin < Zn-PPIX. The fibrinogen beads also directly bound to zinc ions. These results suggest that bovine fibrinogen is a heme- and zinc-binding protein and that binding of circulating mammalian fibrinogen to ferritin is heme mediated.


Subject(s)
Ferritins/chemistry , Fibrinogen/chemistry , Animals , Cattle , Dose-Response Relationship, Drug , Ferritins/antagonists & inhibitors , Fibrinogen/antagonists & inhibitors , Hemin/chemistry , Hemin/pharmacology , Humans , Metalloporphyrins/chemistry , Metalloporphyrins/pharmacology , Protein Binding/drug effects , Protoporphyrins/chemistry , Protoporphyrins/pharmacology , Structure-Activity Relationship
5.
PLoS One ; 9(1): e85271, 2014.
Article in English | MEDLINE | ID: mdl-24416376

ABSTRACT

It is well-known that individuals with increased iron levels are more prone to thrombotic diseases, mainly due to the presence of unliganded iron, and thereby the increased production of hydroxyl radicals. It is also known that erythrocytes (RBCs) may play an important role during thrombotic events. Therefore the purpose of the current study was to assess whether RBCs had an altered morphology in individuals with hereditary hemochromatosis (HH), as well as some who displayed hyperferritinemia (HF). Using scanning electron microscopy, we also assessed means by which the RBC and fibrin morphology might be normalized. An important objective was to test the hypothesis that the altered RBC morphology was due to the presence of excess unliganded iron by removing it through chelation. Very striking differences were observed, in that the erythrocytes from HH and HF individuals were distorted and had a much greater axial ratio compared to that accompanying the discoid appearance seen in the normal samples. The response to thrombin, and the appearance of a platelet-rich plasma smear, were also markedly different. These differences could largely be reversed by the iron chelator desferal and to some degree by the iron chelator clioquinol, or by the free radical trapping agents salicylate or selenite (that may themselves also be iron chelators). These findings are consistent with the view that the aberrant morphology of the HH and HF erythrocytes is caused, at least in part, by unliganded ('free') iron, whether derived directly via raised ferritin levels or otherwise, and that lowering it or affecting the consequences of its action may be of therapeutic benefit. The findings also bear on the question of the extent to which accepting blood donations from HH individuals may be desirable or otherwise.


Subject(s)
Erythrocytes/pathology , Ferritins/blood , Fibrin/ultrastructure , Hemochromatosis/drug therapy , Iron Chelating Agents/therapeutic use , Iron/metabolism , Adolescent , Adult , Child , Clioquinol/therapeutic use , Deferoxamine/therapeutic use , Erythrocytes/drug effects , Erythrocytes/metabolism , Female , Ferritins/antagonists & inhibitors , Fibrin/metabolism , Hemochromatosis/metabolism , Hemochromatosis/pathology , Humans , Hydroxyl Radical/antagonists & inhibitors , Hydroxyl Radical/metabolism , Male , Microscopy, Electron, Scanning , Middle Aged , Platelet-Rich Plasma/chemistry , Salicylic Acid/therapeutic use , Selenious Acid/therapeutic use , Thrombin/pharmacology , Up-Regulation
6.
Haematologica ; 98(7): 1045-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23585526

ABSTRACT

Reports are emerging of hematologic responses associated with iron chelation therapy; however, studies are limited in aplastic anemia patients. Deferasirox reduced iron overload in aplastic anemia patients enrolled in the EPIC (Evaluation of Patients' Iron Chelation with Exjade(®)) study (n=116). A post hoc analysis of hematologic responses was conducted on 72 patients with evaluable hematologic parameters (according to UK guideline criteria), 24 of whom received deferasirox without concomitant immunosuppressive treatment. Partial hematologic responses were observed in 11 of 24 (45.8%) patients; all became transfusion-independent. One patient had an additional platelet response and one patient had an additional platelet and hemoglobin response. Mean serum ferritin levels at end of study were significantly reduced in partial hematologic responders (n=11; -3948 ± 4998 ng/mL; baseline 6693 ± 7014 ng/mL; percentage change from baseline -45.7%; P=0.0029). In non-responders, the reduction in serum ferritin was less pronounced (n=13; -2021 ± 3242 ng/mL; baseline 4365 ± 3063 ng/mL; % change from baseline -27.6%; P=0.0171). Alongside reduction in iron overload, deferasirox may, therefore, improve hematologic parameters in a subset of aplastic anemia patients. Further investigation is required to elucidate the mechanisms involved.


Subject(s)
Anemia, Aplastic/blood , Anemia, Aplastic/drug therapy , Benzoates/therapeutic use , Iron Chelating Agents/therapeutic use , Triazoles/therapeutic use , Adolescent , Adult , Anemia, Aplastic/pathology , Blood Platelets/metabolism , Blood Platelets/pathology , Child , Deferasirox , Female , Ferritins/antagonists & inhibitors , Ferritins/blood , Hemoglobins/metabolism , Humans , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Neutrophils/metabolism , Neutrophils/pathology , Prospective Studies , Treatment Outcome , Young Adult
7.
J Chem Inf Model ; 52(8): 2079-88, 2012 Aug 27.
Article in English | MEDLINE | ID: mdl-22817270

ABSTRACT

We have performed a systematic study of the entropy term in the MM/GBSA (molecular mechanics combined with generalized Born and surface-area solvation) approach to calculate ligand-binding affinities. The entropies are calculated by a normal-mode analysis of harmonic frequencies from minimized snapshots of molecular dynamics simulations. For computational reasons, these calculations have normally been performed on truncated systems. We have studied the binding of eight inhibitors of blood clotting factor Xa, nine ligands of ferritin, and two ligands of HIV-1 protease and show that removing protein residues with distances larger than 8-16 Å to the ligand, including a 4 Å shell of fixed protein residues and water molecules, change the absolute entropies by 1-5 kJ/mol on average. However, the change is systematic, so relative entropies for different ligands change by only 0.7-1.6 kJ/mol on average. Consequently, entropies from truncated systems give relative binding affinities that are identical to those obtained for the whole protein within statistical uncertainty (1-2 kJ/mol). We have also tested to use a distance-dependent dielectric constant in the minimization and frequency calculation (ε = 4r), but it typically gives slightly different entropies and poorer binding affinities. Therefore, we recommend entropies calculated with the smallest truncation radius (8 Å) and ε =1. Such an approach also gives an improved precision for the calculated binding free energies.


Subject(s)
Entropy , Proteins/chemistry , Proteins/metabolism , Electric Impedance , Factor Xa/chemistry , Factor Xa/metabolism , Factor Xa Inhibitors , Ferritins/antagonists & inhibitors , Ferritins/chemistry , Ferritins/metabolism , HIV Protease/chemistry , HIV Protease/metabolism , HIV Protease Inhibitors/metabolism , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Proteins/antagonists & inhibitors
8.
Toxicol Appl Pharmacol ; 250(1): 69-77, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20950636

ABSTRACT

Arsenic trioxide (ATO) treatment is a useful therapy against human acute promyelocytic leukemia (APL), however, it concomitantly brings potential adverse consequences including serious side effect, human carcinogenicity and possible development of resistance. This investigation revealed that those problems might be relaxed by simultaneous application with (-)-epigallocatechin-3-gallate (EGCG), one of the major components from green tea. EGCG significantly lowered down the ATO concentration required for an effective control of APL cells, HL-60. The simultaneous treatment of ATO with EGCG induced a mitochondria-dependent apoptosis in HL-60 cells significantly, which accounted for more than 70% of the cell death in the treatment. The mechanism of apoptosis induction was elucidated. EGCG in HL-60 cells acted as a pro-oxidant enhancing intracellular hydrogen peroxide significantly. ATO, on the other hand, induced heme oxygenase-1 (HO-1) to catalyze heme degradation, thereby provided ferrous iron for EGCG-induced hydrogen peroxide to precede Fenton reaction, which in turn generated deleterious reactive oxygen species to damage cell. In addition, EGCG inhibited expression of ferritin, which supposedly to sequester harmful ferrous iron, thereby augmented the occurrence of Fenton reaction. This investigation also provided evidence that ATO, since mainly acted to induce HO-1 in simultaneous treatment with EGCG, could be replaced by other HO-1 inducer with much less human toxicity. Furthermore, several of our preliminary investigations revealed that the enhanced cytotoxicity induced by combining heme degradation and Fenton reaction is selectively toxic to malignant but not non-malignant cells.


Subject(s)
Antineoplastic Agents/toxicity , Catechin/analogs & derivatives , Cytotoxins/toxicity , Ferritins/antagonists & inhibitors , Oxides/toxicity , Apoptosis/drug effects , Arsenic Trioxide , Arsenicals , Caffeic Acids/toxicity , Catechin/toxicity , Cell Line, Tumor , Drug Synergism , Ferritins/metabolism , HL-60 Cells , Heme Oxygenase (Decyclizing)/metabolism , Humans , Neoplasms/enzymology , Neoplasms/metabolism , Neoplasms/pathology , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/toxicity
9.
BMB Rep ; 43(10): 683-7, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21034531

ABSTRACT

Previous studies have shown that one of the primary causes of increased iron content in the brain may be the release of excess iron from intracellular iron storage molecules such as ferritin. Free iron generates ROS that cause oxidative cell damage. Carnosine and related compounds such as endogenous histidine dipetides have antioxidant activities. We have investigated the protective effects of carnosine and homocarnosine against oxidative damage of DNA induced by reaction of ferritin with H(2)O(2). The results show that carnosine and homocarnosine prevented ferritin/H(2)O(2)-mediated DNA strand breakage. These compounds effectively inhibited ferritin/H(2)O(2)-mediated hydroxyl radical generation and decreased the mutagenicity of DNA induced by the ferritin÷H(2)O(2) reaction. Our results suggest that carnosine and related compounds might have antioxidant effects on DNA under pathophysiological conditions leading to degenerative damage such as neurodegenerative disorders.


Subject(s)
Carnosine/analogs & derivatives , Carnosine/pharmacology , DNA Damage/drug effects , Ferritins/antagonists & inhibitors , Hydrogen Peroxide/antagonists & inhibitors , Antioxidants/pharmacology , Ferritins/pharmacology , Hydrogen Peroxide/pharmacology , Hydroxyl Radical/metabolism , Hydroxyl Radical/pharmacokinetics , In Vitro Techniques , Lac Operon/drug effects , Mutagenesis/drug effects , Mutagenicity Tests , Oxidation-Reduction/drug effects , Plasmids/chemistry , Plasmids/drug effects , Plasmids/metabolism
10.
Vaccine ; 28(17): 2993-8, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20171306

ABSTRACT

Ixodes ricinus is one the most abundant tick species in Europe and these ticks transmit pathogens causing human and animal diseases. The cattle ticks, Rhipicephalus (Boophilus) spp., affect cattle production in tropical and subtropical regions of the world. Development of vaccines directed against tick proteins may reduce tick infestations and the transmission of tick-borne pathogens. However, a limiting step in tick vaccine development has been the identification of tick protective antigens. Herein, the tick iron metabolism pathway was targeted in an effort to identify new tick protective antigens. Recombinant I. ricinus (IrFER2) and Rhipicephalus microplus (RmFER2) ferritin 2 proteins were expressed in Escherichia coli and used to immunize rabbits and cattle, respectively. Vaccination with IrFER2 reduced I. ricinus tick numbers, weight and fertility in rabbits with an overall vaccine efficacy (E) of 98%. Control of cattle tick, R. microplus and Rhipicephalus annulatus infestations was obtained in vaccinated cattle with overall E of 64% and 72%, respectively. Notably, the efficacy of the RmFER2 vaccine was similar to that obtained with Bm86 against R. microplus. These collective results demonstrated the feasibility of using ferritin 2 to develop vaccines for the control of tick infestations.


Subject(s)
Cattle Diseases/prevention & control , Ferritins/immunology , Ferritins/isolation & purification , Insect Proteins/immunology , Insect Proteins/isolation & purification , Rabbits/parasitology , Tick Infestations/veterinary , Animals , Cattle , Cattle Diseases/parasitology , Escherichia coli/genetics , Ferritins/antagonists & inhibitors , Insect Proteins/antagonists & inhibitors , Ixodes/immunology , Rhipicephalus/immunology , Tick Infestations/prevention & control , Vaccines, Synthetic/immunology
11.
Anal Chem ; 81(24): 10097-105, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19908819

ABSTRACT

Immunolatex particles (LAmP-s), which were prepared by covalently co-immobilizing antiferritin and a mixture of pentaethylenehexamine-ended poly(ethylene glycol) (N6-PEG) of two different chain lengths onto the surface of polystyrene submicroparticles, were formulated with various antiferritin loads. Following the quantification of the bound antiferritin, as well as the differentiation of the physically adsorbed antiferritin from chemically bound antiferritin, using the copper reduction/bicinchoninic acid reaction (the Micro BCA method), dynamic light scattering (DLS) and electrophoretic mobility (mu(e)) measurements were performed to characterize the size, homogeneity, and surface charge of the complex. Compared to the control immunolatex particles, which were prepared similarly but traditionally using bovine serum albumin (BSA) as a blocking agent (LAB-s), the LAmP-s complex showed a difference only in the surface charge property, because of the altered surface treatment in the case of the LAmP-s (PEGylation) and LAB-s complexes (BSA covering). However, the LAmP-s complex was much more reactive than the LAB-s complex, not only in phosphate buffer (PB, 10 mM, pH 7.4) but also in 100% fetal bovine serum (FBS), as measured by the turbidimetric monitoring method. The electrical repulsion between the negatively charged LAB-s complex and the antigen was the primary obstacle in the former case, and the overwhelming nonspecific deposition of contaminants from FBS onto the LAB-s complex was the main reason in the latter case. Moreover, the PEGylation procedure allowed the LAmP-s complex to possess invariable size and reactivity for at least one month at 4 degrees C without salt, and colloidal stability at high salt concentrations up to 2.0 M for at least one week, obviously demonstrating that the PEG surface modification technique described in this paper is a promising method of constructing efficient immunoassay systems.


Subject(s)
Antibodies/chemistry , Ferritins/antagonists & inhibitors , Serum Albumin, Bovine/pharmacology , Antibodies/immunology , Antigen-Antibody Reactions , Ferritins/analysis , Ferritins/immunology , Immunoassay , Particle Size , Polyamines/chemistry , Polyethylene Glycols/chemistry , Polystyrenes/chemistry , Serum Albumin, Bovine/chemistry , Surface Properties
12.
J Biol Chem ; 281(33): 23579-88, 2006 Aug 18.
Article in English | MEDLINE | ID: mdl-16782706

ABSTRACT

Ferritins play an essential role in iron homeostasis by sequestering iron in a bioavailable and non-toxic form. In plants, ferritin mRNAs are highly and quickly accumulated in response to iron overload. Such accumulation leads to a subsequent ferritin protein synthesis and iron storage, thus avoiding oxidative stress to take place. By combining pharmacological and imaging approaches in an Arabidopsis cell culture system, we have identified several elements in the signal transduction pathway leading to the increase of AtFer1 transcript level after iron treatment. Nitric oxide quickly accumulates in the plastids after iron treatment. This compound acts downstream of iron and upstream of a PP2A-type phosphatase to promote an increase of AtFer1 mRNA level. The AtFer1 gene transcription has been previously shown to be repressed under low iron conditions with the involvement of the cis-acting element iron-dependent regulatory sequence identified within the AtFer1 promoter sequence. We show here that the repressor is unlikely a transcription factor directly bound to the iron-dependent regulatory sequence; such a repressor is ubiquitinated upon iron treatment and subsequently degraded through a 26 S proteasome-dependent pathway.


Subject(s)
Arabidopsis/genetics , Arabidopsis/metabolism , Ferritins/genetics , Ferritins/metabolism , Gene Expression Regulation, Plant , Iron/chemistry , Nitric Oxide/metabolism , Ubiquitin/physiology , Arabidopsis/chemistry , Ferritins/antagonists & inhibitors , Ferritins/biosynthesis , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Iron/physiology , Iron-Regulatory Proteins/metabolism , Plastids/metabolism , Proteasome Endopeptidase Complex/physiology , RNA, Messenger/genetics , Repressor Proteins/metabolism , Signal Transduction/physiology
13.
J Biol Inorg Chem ; 8(4): 489-97, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12679873

ABSTRACT

Zinc and terbium, inhibitors of iron incorporation in the ferritins, have been used for many years as probes of structure-function relationships in these proteins. Isothermal titration calorimetric and kinetic measurements of Zn(II) and Tb(III) binding and inhibition of Fe(II) oxidation were used to identify and characterize thermodynamically ( n, K, Delta H degrees, Delta S degrees, and Delta G degrees ) the functionally important binding sites for these metal ions in recombinant human H-chain, L-chain, and H-chain site-directed variant ferritins. The data reveal at least two classes of binding sites for both Zn(II) and Tb(III) in human H-chain ferritin: one strong, corresponding to binding of one metal ion in each of the eight three-fold channels, and the other weak, involving binding at the ferroxidase and nucleation sites of the protein as well as at other weak unidentified binding sites. Zn(II) and Tb(III) binding to recombinant L-chain ferritin showed similar stoichiometries for the strong binding sites within the channels, but fewer weaker binding sites when compared to the H-chain protein. The kinetics and binding data indicate that the binding of Zn(II) and Tb(III) in the three-fold channels, which is the main pathway of iron(II) entry in ferritin, blocks the access of most of the iron to the ferroxidase sites on the interior of the protein, accounting for the strong inhibition by these metal ions of the oxidative deposition of iron in ferritin.


Subject(s)
Ferritins/metabolism , Metals/metabolism , Recombinant Proteins/metabolism , Apoferritins , Calorimetry/methods , Ferritins/antagonists & inhibitors , Metals/chemistry , Recombinant Proteins/chemistry
14.
J Biol Inorg Chem ; 8(1-2): 195-205, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12459915

ABSTRACT

The ability of ferritin as an Fe(II) detoxifier and Fe(III) storage protein is limited by its ability to recognize and incorporate Fe(II), which is then oxidized and mineralized at internal protein sites. The Cr(III) amine complex [Cr(N(CH(2)CH(2)NH(2))(3)(H(2)O)(OH)](2+) [abbreviated as Cr(TREN)] is a kinetically inert inhibitor of iron incorporation and mineralization in ferritin. Unlike other inhibitors, Cr(TREN) can only exchange its two aqua/hydroxy ligands. Competition studies between Cr(TREN) and Tb(III) binding have been performed in horse spleen ferritin (HoSF) to probe uptake of Fe(II). From these studies, we propose that Cr(TREN) inhibits Fe(II) uptake by obstructing the routes of metal uptake and by disrupting the early recognition events at the protein surface that precede metal ion uptake. Using an improved luminescence approach to quantify Tb(III) binding to the protein, we demonstrate that Tb(III) cannot interfere with Cr(TREN) binding to ferritin, but that Cr(TREN) dramatically inhibits Tb(III) binding. We show that bound Tb(III) serves as a reliable reporter for Cr(TREN) binding, as the latter efficiently quenches the Tb(III) luminescence via inter-ion energy transfer. Two types of Cr(TREN) binding sites were successfully distinguished from these competition experiments. A common Tb(III)/Cr(TREN) site was identified with stoichiometry of approximately 0.6 equivalents of metal cation per ferritin subunit. We propose that the sites along the three-fold channels and the ferroxidase sites are common binding sites for Tb(III) and Cr(TREN). The remaining Cr(TREN) (2.4 equivalents of metal ions/subunit) does not compete with Tb(III) but rather blocks Tb(III) access into the cavity and decreases the protein's affinity for Tb(III).


Subject(s)
Ferritins/antagonists & inhibitors , Organometallic Compounds/chemistry , Spleen/chemistry , Terbium/chemistry , Animals , Binding Sites , Binding, Competitive , Ferritins/chemistry , Ferritins/metabolism , Ferrous Compounds/metabolism , Horses , Luminescent Measurements , Models, Molecular , Organometallic Compounds/metabolism , Organometallic Compounds/pharmacology , Protein Binding , Protein Subunits , Spectrometry, Fluorescence/methods , Terbium/metabolism , Terbium/pharmacology
15.
Anticancer Res ; 22(3): 1513-24, 2002.
Article in English | MEDLINE | ID: mdl-12168831

ABSTRACT

BACKGROUND: Ferritin is the major iron-storage protein which sequesters and detoxifies excess iron that is taken up by cells but is not utilized in normal metabolic processes. Human ferritin consists of various combinations of heavy (FerH, Mr 21,000) and light (FerL, Mr 19,000) chains and excess iron leads to an increase in the synthesis of both heavy and light chains. MATERIALS AND METHODS: In this study four pairs of antisense oligodeoxynucleotides (ODNs) were synthesized: FerH-A1 and FerL-A1 were complementary to the 24-base pair sequence overlapping the starting codons of the FerH and FerL genes, respectively, but the sequences of FerH-A2 and FerL-A2 only covered the coding sequences of the ferritin genes. The corresponding sense chain sequences (FerH-S1, FerH-S2, FerL-S1 and FerL-S2) were used as controls. RESULTS: Treatment with FerH-S1, FerH-A1, FerH-S2, FerH-A2, FerL-S1, FerL-A1, FerL-S2 and FerL-A2 at 40 microM, 25 microM, 30 microM, 17 microM, 45 microM, 18 microM, 40 microM and 26 microM, respectively, for 72 hours resulted in 50% inhibition of DNA synthesis (IC50) in MCF-7 breast carcinoma cells, as measured by [3H]-thymidine incorporation. FerH chain mRNA, FerL chain mRNA and total ferritin protein levels were significantly decreased by the IC50 concentrations of each of the antisense ODNs but were not inhibited by IC50 concentrations of sense ODNs, as measured by quantitative RT-PCR and microparticle enzyme immunoassay. However, antisense ferritin ODNs had no effect on the total iron concentration in MCF-7 cells. Incubation with IC50 concentrations of antisense ferritin ODNs caused reduction in cell volume, condensation of nuclear structures and lower levels of Bcl-2 mRNA and protein compared to control cells, but Bax mRNA and protein levels remained unchanged. CONCLUSION: This study demonstrates that antisense ODNs to ferritin genes are about two-fold more cytotoxic than sense ODNs, and that antisense ODNs are specific inhibitors of ferritin gene expression at both the transcriptional and the translational levels. Further, the antisense ferritin ODNs promote programmed cell death with low ratios of Bcl-2 to Bax mRNA and protein expression providing evidence that antisense ferritin ODNs specifically inhibit MCF-7 breast carcinoma cell growth through increased apoptosis. Finally, since the IC50 concentrations of FerH-A1 and FerH-A2, and FerL-A1 and FerL-A2 are very similar for inhibition of DNA synthesis and gene expression in human breast carcinoma MCF-7 cells, it does not seem necessary for the antisense ODNs to overlap the starting codons of ferritin gene to maximize inhibition.


Subject(s)
Breast Neoplasms/drug therapy , Ferritins/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division/drug effects , DNA, Neoplasm/biosynthesis , Ferritins/antagonists & inhibitors , Ferritins/biosynthesis , Growth Inhibitors/chemical synthesis , Growth Inhibitors/genetics , Growth Inhibitors/pharmacology , Humans , Iron/metabolism , Oligodeoxyribonucleotides, Antisense/chemical synthesis , Oligodeoxyribonucleotides, Antisense/genetics , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tumor Cells, Cultured
16.
J Cell Physiol ; 190(2): 218-26, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11807826

ABSTRACT

Iron regulatory proteins (IRPs), the cytosolic proteins involved in the maintenance of cellular iron homeostasis, bind to stem loop structures found in the mRNA of key proteins involved iron uptake, storage, and metabolism and regulate the expression of these proteins in response to changes in cellular iron needs. We have shown previously that HFE-expressing fWTHFE/tTA HeLa cells have slightly increased transferrin receptor levels and dramatically reduced ferritin levels when compared to the same clonal cell line without HFE (Gross et al., 1998, J Biol Chem 273:22068-22074). While HFE does not alter transferrin receptor trafficking or non-transferrin mediated iron uptake, it does specifically reduce (55)Fe uptake from transferrin (Roy et al., 1999, J Biol Chem 274:9022-9028). In this report, we show that IRP RNA binding activity is increased by up to 5-fold in HFE-expressing cells through the activation of both IRP isoforms. Calcein measurements show a 45% decrease in the intracellular labile iron pool in HFE-expressing cells, which is in keeping with the IRP activation. These results all point to the direct effect of the interaction of HFE with transferrin receptor in lowering the intracellular labile iron pool and establishing a new set point for iron regulation within the cell.


Subject(s)
HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Iron-Sulfur Proteins/metabolism , Iron/metabolism , Membrane Proteins , RNA-Binding Proteins/metabolism , RNA/metabolism , Ferritins/antagonists & inhibitors , Ferritins/biosynthesis , Fluoresceins , Fluorescent Dyes , HeLa Cells , Hemochromatosis Protein , Humans , Intracellular Membranes/metabolism , Iron Regulatory Protein 1 , Iron Regulatory Protein 2 , Iron-Regulatory Proteins
17.
Biochemistry ; 39(20): 6235-42, 2000 May 23.
Article in English | MEDLINE | ID: mdl-10821699

ABSTRACT

Iron-responsive elements (IREs), a natural group of mRNA-specific sequences, bind iron regulatory proteins (IRPs) differentially and fold into hairpins [with a hexaloop (HL) CAGUGX] with helical distortions: an internal loop/bulge (IL/B) (UGC/C) or C-bulge. C-bulge iso-IREs bind IRP2 more poorly, as oligomers (n = 28-30), and have a weaker signal response in vivo. Two trans-loop GC base pairs occur in the ferritin IRE (IL/B and HL) but only one in C-bulge iso-IREs (HL); metal ions and protons perturb the IL/B [Gdaniec et al. (1998) Biochemistry 37, 1505-1512]. IRE function (translation) and physical properties (T(m) and accessibility to nucleases) are now compared for IL/B and C-bulge IREs and for HL mutants. Conversion of the IL/B into a C-bulge by a single deletion in the IL/B or by substituting the HL CG base pair with UA both derepressed ferritin synthesis 4-fold in rabbit reticulocyte lysates (IRP1 + IRP2), confirming differences in IRP2 binding observed for the oligomers. Since the engineered C-bulge IRE was more helical near the IL/B [Cu(phen)(2) resistant] and more stable (T(m) increased) and the HL mutant was less helical near the IL/B (ribonuclease T1 sensitive) and less stable (T(m) decreased), both CG trans-loop base pairs contribute to maximum IRP2 binding and translational regulation. The (1)H NMR spectrum of the Mg-IRE complex revealed, in contrast to the localized IL/B effects of Co(III) hexaammine observed previously, perturbation of the IL/B plus HL and interloop helix. The lower stability and greater helix distortion in the ferritin IL/B-IRE compared to the C-bulge iso-IREs create a combinatorial set of RNA/protein interactions that control protein synthesis rates with a range of signal sensitivities.


Subject(s)
Ferritins/chemistry , Ferritins/genetics , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Nucleic Acid Conformation , Protein Biosynthesis , RNA, Messenger/chemistry , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Animals , Copper/chemistry , Ferritins/antagonists & inhibitors , Ferritins/biosynthesis , Hot Temperature , Iron Regulatory Protein 1 , Iron Regulatory Protein 2 , Iron-Regulatory Proteins , Iron-Sulfur Proteins/physiology , Lizards , Magnesium/chemistry , Multigene Family , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Phenanthrolines/chemistry , Protein Binding/genetics , Protein Denaturation , Protein Isoforms/chemistry , Protein Isoforms/genetics , RNA-Binding Proteins/physiology , Rabbits , Rana catesbeiana , Repressor Proteins/genetics , Repressor Proteins/physiology , Ribonuclease T1/chemistry
18.
Biochemistry ; 39(16): 4915-23, 2000 Apr 25.
Article in English | MEDLINE | ID: mdl-10769150

ABSTRACT

Bacterioferritins are members of a class of spherical shell-like iron storage proteins that catalyze the oxidation and hydrolysis of iron at specific sites inside the protein shell, resulting in formation of a mineral core of hydrated ferric oxide within the protein cavity. Electrode oximetry/pH stat was used to study iron oxidation and hydrolysis chemistry in E. coli bacterioferritin. Consistent with previous UV-visible absorbance measurements, three distinct kinetic phases were detected, and the stoichiometric equations corresponding to each have been determined. The rapid phase 1 reaction corresponds to pairwise binding of 2 Fe(2+) ions at a dinuclear site, called the ferroxidase site, located within each of the 24 subunits, viz., 2Fe(2+) + P(Z) --> [Fe(2)-P](Z) + 4H(+), where P(Z) is the apoprotein of net charge Z and [Fe(2)-P](Z) represents a diferrous ferroxidase complex. The slower phase 2 reaction corresponds to the oxidation of this complex by molecular oxygen according to the net equation: [Fe(2)-P](Z) + (1)/(2)O(2) --> [Fe(2)O-P](Z) where [Fe(2)O-P](Z) represents an oxidized diferric ferroxidase complex, probably a mu-oxo-bridged species as suggested by UV-visible and EPR spectrometric titration data. The third phase corresponds to mineral core formation according to the net reaction: 4Fe(2+) + O(2) + 6H(2)O --> 4FeO(OH)((core)) + 8H(+). Iron oxidation is inhibited by the presence of Zn(2+) ions. The patterns of phase 2 and phase 3 inhibition are different, though inhibition of both phases is complete at 48 Zn(2+)per 24mer, i.e., 2 Zn(2+) per ferroxidase center.


Subject(s)
Bacterial Proteins , Cytochrome b Group/metabolism , Escherichia coli/chemistry , Ferritins/metabolism , Iron/metabolism , Oxygen/metabolism , Amino Acid Substitution/genetics , Apoproteins/chemistry , Apoproteins/metabolism , Binding Sites , Catalase/metabolism , Ceruloplasmin/metabolism , Cytochrome b Group/antagonists & inhibitors , Cytochrome b Group/genetics , Electron Spin Resonance Spectroscopy , Ferric Compounds/metabolism , Ferritins/antagonists & inhibitors , Ferritins/genetics , Ferrous Compounds/metabolism , Humans , Hydrogen Peroxide/metabolism , Hydrogen-Ion Concentration , Hydrolysis/drug effects , Kinetics , Mutation/genetics , Oxidation-Reduction/drug effects , Protons , Spectrophotometry , Titrimetry , Zinc/pharmacology
19.
Biochemistry (Mosc) ; 62(12): 1415-23, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9481874

ABSTRACT

The thermal inactivation of horse spleen ferritin was studied over a range of temperatures (36-52 degrees C) in 0.1 M acetate buffer (pH 4.2) as a decrease of its peroxidase activity during tetramethylbenzidine (TMB) oxidation by hydrogen peroxide. The activation energy of this process was 163.3 kJ/mole. Thermodynamic activation parameters for the loss of peroxidase activity of ferritin were calculated. The influence of various detergents on ferritin-dependent oxidation of TMB, ortho-tolidine, and ortho-phenylenediamine (PDA) by hydrogen peroxide was studied in 0.1 M phosphate buffer (pH 6.0) at 20 degrees C. Relatively high concentrations of charged detergents (SDS and cetyltrimethylammonium bromide) decreased the peroxidase activity of ferritin with all three amines, whereas moderate concentrations of the nonionic detergent Triton X-100 did not influence oxidation of these substrates. Increase of dimethylformamide concentration in 0.02 M acetate buffer (pH 4.2) from 5 to 40% strongly decreased the rate of TMB and PDA oxidation by hydrogen peroxide or cumene hydroperoxide. Decrease in the activity of thermally inactivated ferritin with TMB as substrate, reduction of alpha-helical content of the protein at 40-50 degrees C, an inactivating effect of charged surfactants and organic co-solvent on the peroxidase activity of ferritin indicate a very important role of the apoprotein in peroxidase function. A possible mechanism of apoferritin participation in peroxidase catalysis is discussed.


Subject(s)
Apoproteins/metabolism , Ferritins/metabolism , Peroxidases/metabolism , Animals , Catalysis , Circular Dichroism , Detergents , Dimethylformamide/chemistry , Ferritins/antagonists & inhibitors , Ferritins/chemistry , Horses , Hot Temperature , Kinetics , Oxidation-Reduction , Spleen/metabolism , Thermodynamics
20.
Free Radic Biol Med ; 20(1): 63-73, 1996.
Article in English | MEDLINE | ID: mdl-8903680

ABSTRACT

Ferritin protects endothelial cells from the damaging effects of iron-catalyzed oxidative injury. Regulation of ferritin occurs through the formation of an iron-sulfur cluster within a cytoplasmic protein, the iron regulatory protein (IRP) that controls ferritin mRNA translation. Nitric oxide has been shown to inhibit iron-sulfur proteins and is present at vascular sites of inflammation; therefore, we undertook a study to examine the influence of nitric oxide on changes in endothelial cell ferritin content in response to iron exposure, and the subsequent effects on susceptibility to oxidative injury. Iron-loaded endothelial cells (EC) exposed to nitric oxide donors synthesize markedly less ferritin. Treatment of EC with a nitric oxide donor increases IRP affinity for ferritin mRNA concomitant with a loss of cytoplasmic aconitase activity in iron-laden EC. Iron-treated EC exposed to NO donors were resistant to oxidative injury despite their low ferritin content when examined 1 h after the treatment period. In contrast, 24 h later, these same cells become sensitive to oxidants, whereas iron-treated EC that are ferritin-rich continue to be resistant. In conclusion, NO inhibits the increase of EC ferritin after exposure to iron but provides short-term protection against oxidants; ferritin, in turn, provides durable cytoprotection by inactivating reactive iron.


Subject(s)
Endothelium/metabolism , Ferritins/metabolism , Iron-Sulfur Proteins/metabolism , Nitric Oxide/pharmacology , RNA-Binding Proteins/metabolism , Aconitate Hydratase/antagonists & inhibitors , Aconitate Hydratase/metabolism , Animals , Aorta/metabolism , Base Sequence , Blotting, Northern , Cells, Cultured , DNA Probes/chemistry , Ferritins/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/metabolism , Hemin/metabolism , Humans , Iron Compounds/metabolism , Iron Compounds/pharmacology , Iron-Regulatory Proteins , Molecular Sequence Data , Oxidative Stress , Penicillamine/analogs & derivatives , Penicillamine/metabolism , Polyamines/metabolism , RNA, Messenger/metabolism , S-Nitroso-N-Acetylpenicillamine , Swine/metabolism , Umbilical Veins/metabolism , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...