Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
J Neural Transm (Vienna) ; 127(2): 189-203, 2020 02.
Article in English | MEDLINE | ID: mdl-31912279

ABSTRACT

Focal iron accumulation associated with brain iron dyshomeostasis is a pathological hallmark of various neurodegenerative diseases (NDD). The application of iron-sensitive sequences in magnetic resonance imaging has provided a useful tool to identify the underlying NDD pathology. In the three major NDD, degeneration occurs in central nervous system (CNS) regions associated with memory (Alzheimer's disease, AD), automaticity (Parkinson's disease, PD) and motor function (amyotrophic lateral sclerosis, ALS), all of which require a high oxygen demand for harnessing neuronal energy. In PD, a progressive degeneration of the substantia nigra pars compacta (SNc) is associated with the appearance of siderotic foci, largely caused by increased labile iron levels resulting from an imbalance between cell iron import, storage and export. At a molecular level, α-synuclein regulates dopamine and iron transport with PD-associated mutations in this protein causing functional disruption to these processes. Equally, in ALS, an early iron accumulation is present in neurons of the cortico-spinal motor pathway before neuropathology and secondary iron accumulation in microglia. High serum ferritin is an indicator of poor prognosis in ALS and the application of iron-sensitive sequences in magnetic resonance imaging has become a useful tool in identifying pathology. The molecular pathways that cascade down from such dyshomeostasis still remain to be fully elucidated but strong inroads have been made in recent years. Far from being a simple cause or consequence, it has recently been discovered that these alterations can trigger susceptibility to an iron-dependent cell-death pathway with unique lipoperoxidation signatures called ferroptosis. In turn, this has now provided insight into some key modulators of this cell-death pathway that could be therapeutic targets for the NDD. Interestingly, iron accumulation and ferroptosis are highly sensitive to iron chelation. However, whilst chelators that strongly scavenge intracellular iron protect against oxidative neuronal damage in mammalian models and are proven to be effective in treating systemic siderosis, these compounds are not clinically suitable due to the high risk of developing iatrogenic iron depletion and ensuing anaemia. Instead, a moderate iron chelation modality that conserves systemic iron offers a novel therapeutic strategy for neuroprotection. As demonstrated with the prototype chelator deferiprone, iron can be scavenged from labile iron complexes in the brain and transferred (conservatively) either to higher affinity acceptors in cells or extracellular transferrin. Promising preclinical and clinical proof of concept trials has led to several current large randomized clinical trials that aim to demonstrate the efficacy and safety of conservative iron chelation for NDD, notably in a long-term treatment regimen.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Chelation Therapy , Deferiprone/pharmacology , Iron Chelating Agents/pharmacology , Iron/metabolism , Parkinson Disease/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Animals , Chelation Therapy/methods , Chelation Therapy/standards , Humans , Parkinson Disease/metabolism
3.
Antioxid Redox Signal ; 29(8): 742-748, 2018 09 10.
Article in English | MEDLINE | ID: mdl-29287521

ABSTRACT

Iron accumulation has been observed in mouse models and in both sporadic and familial forms of amyotrophic lateral sclerosis (ALS). Iron chelation could reduce iron accumulation and the related excess of oxidative stress in the motor pathways. However, classical iron chelation would induce systemic iron depletion. We assess the safety and efficacy of conservative iron chelation (i.e., chelation with low risk of iron depletion) in a murine preclinical model and pilot clinical trial. In Sod1G86R mice, deferiprone increased the mean life span compared with placebo. The safety was good, without anemia after 12 months of deferiprone in the 23 ALS patients enrolled in the clinical trial. The decreases in the ALS Functional Rating Scale and the body mass index were significantly smaller for the first 3 months of deferiprone treatment (30 mg/kg/day) than for the first treatment-free period. Iron levels in the cervical spinal cord, medulla oblongata, and motor cortex (according to magnetic resonance imaging), as well as cerebrospinal fluid levels of oxidative stress and neurofilament light chains were lower after deferiprone treatment. Our observation leads to the hypothesis that moderate iron chelation regimen that avoids changes in systemic iron levels may constitute a novel therapeutic modality of neuroprotection for ALS. Antioxid. Redox Signal. 29, 742-748.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Deferiprone/therapeutic use , Iron Chelating Agents/therapeutic use , Neuroprotective Agents/therapeutic use , Adult , Amyotrophic Lateral Sclerosis/metabolism , Animals , Deferiprone/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Iron Chelating Agents/administration & dosage , Male , Mice , Mice, Transgenic , Middle Aged , Neuroprotective Agents/administration & dosage , Oxidative Stress/drug effects , Young Adult
4.
Haematologica ; 101(1): 38-45, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26385212

ABSTRACT

Non-transferrin-bound iron and its labile (redox active) plasma iron component are thought to be potentially toxic forms of iron originally identified in the serum of patients with iron overload. We compared ten worldwide leading assays (6 for non-transferrin-bound iron and 4 for labile plasma iron) as part of an international inter-laboratory study. Serum samples from 60 patients with four different iron-overload disorders in various treatment phases were coded and sent in duplicate for analysis to five different laboratories worldwide. Some laboratories provided multiple assays. Overall, highest assay levels were observed for patients with untreated hereditary hemochromatosis and ß-thalassemia intermedia, patients with transfusion-dependent myelodysplastic syndromes and patients with transfusion-dependent and chelated ß-thalassemia major. Absolute levels differed considerably between assays and were lower for labile plasma iron than for non-transferrin-bound iron. Four assays also reported negative values. Assays were reproducible with high between-sample and low within-sample variation. Assays correlated and correlations were highest within the group of non-transferrin-bound iron assays and within that of labile plasma iron assays. Increased transferrin saturation, but not ferritin, was a good indicator of the presence of forms of circulating non-transferrin-bound iron. The possibility of using non-transferrin-bound iron and labile plasma iron measures as clinical indicators of overt iron overload and/or of treatment efficacy would largely depend on the rigorous validation and standardization of assays.


Subject(s)
Blood Transfusion , Hemochromatosis/blood , Iron/blood , Myelodysplastic Syndromes/blood , Transferrin/metabolism , beta-Thalassemia/blood , Adult , Aged , Female , Humans , Male , Middle Aged , Myelodysplastic Syndromes/therapy , beta-Thalassemia/therapy
5.
PLoS One ; 9(11): e112915, 2014.
Article in English | MEDLINE | ID: mdl-25390893

ABSTRACT

BACKGROUND: Iron overload cardiomyopathy that prevails in some forms of hemosiderosis is caused by excessive deposition of iron into the heart tissue and ensuing damage caused by a raise in labile cell iron. The underlying mechanisms of iron uptake into cardiomyocytes in iron overload condition are still under investigation. Both L-type calcium channels (LTCC) and T-type calcium channels (TTCC) have been proposed to be the main portals of non-transferrinic iron into heart cells, but controversies remain. Here, we investigated the roles of LTCC and TTCC as mediators of cardiac iron overload and cellular damage by using specific Calcium channel blockers as potential suppressors of labile Fe(II) and Fe(III) ingress in cultured cardiomyocytes and ensuing apoptosis. METHODS: Fe(II) and Fe(III) uptake was assessed by exposing HL-1 cardiomyocytes to iron sources and quantitative real-time fluorescence imaging of cytosolic labile iron with the fluorescent iron sensor calcein while iron-induced apoptosis was quantitatively measured by flow cytometry analysis with Annexin V. The role of calcium channels as routes of iron uptake was assessed by cell pretreatment with specific blockers of LTCC and TTCC. RESULTS: Iron entered HL-1 cardiomyocytes in a time- and dose-dependent manner and induced cardiac apoptosis via mitochondria-mediated caspase-3 dependent pathways. Blockade of LTCC but not of TTCC demonstrably inhibited the uptake of ferric but not of ferrous iron. However, neither channel blocker conferred cardiomyocytes with protection from iron-induced apoptosis. CONCLUSION: Our study implicates LTCC as major mediators of Fe(III) uptake into cardiomyocytes exposed to ferric salts but not necessarily as contributors to ensuing apoptosis. Thus, to the extent that apoptosis can be considered a biological indicator of damage, the etiopathology of cardiosiderotic damage that accompanies some forms of hemosiderosis would seem to be unrelated to LTCC or TTCC, but rather to other routes of iron ingress present in heart cells.


Subject(s)
Apoptosis/drug effects , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/metabolism , Iron Overload/metabolism , Iron/metabolism , Myocytes, Cardiac/drug effects , Animals , Calcium/metabolism , Cardiomyopathies/metabolism , Caspase 3/metabolism , Cell Line , Ferric Compounds/pharmacology , Heart/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Myocytes, Cardiac/metabolism
6.
Antioxid Redox Signal ; 21(2): 195-210, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24251381

ABSTRACT

AIMS: The pathophysiological role of iron in Parkinson's disease (PD) was assessed by a chelation strategy aimed at reducing oxidative damage associated with regional iron deposition without affecting circulating metals. Translational cell and animal models provided concept proofs and a delayed-start (DS) treatment paradigm, the basis for preliminary clinical assessments. RESULTS: For translational studies, we assessed the effect of oxidative insults in mice systemically prechelated with deferiprone (DFP) by following motor functions, striatal dopamine (HPLC and MRI-PET), and brain iron deposition (relaxation-R2*-MRI) aided by spectroscopic measurements of neuronal labile iron (with fluorescence-sensitive iron sensors) and oxidative damage by markers of protein, lipid, and DNA modification. DFP significantly reduced labile iron and biological damage in oxidation-stressed cells and animals, improving motor functions while raising striatal dopamine. For a pilot, double-blind, placebo-controlled randomized clinical trial, early-stage Parkinson's patients on stabilized dopamine regimens enrolled in a 12-month single-center study with DFP (30 mg/kg/day). Based on a 6-month DS paradigm, early-start patients (n=19) compared to DS patients (n=18) (37/40 completed) responded significantly earlier and sustainably to treatment in both substantia nigra iron deposits (R2* MRI) and Unified Parkinson's Disease Rating Scale motor indicators of disease progression (p<0.03 and p<0.04, respectively). Apart from three rapidly resolved neutropenia cases, safety was maintained throughout the trial. INNOVATION: A moderate iron chelation regimen that avoids changes in systemic iron levels may constitute a novel therapeutic modality for PD. CONCLUSIONS: The therapeutic features of a chelation modality established in translational models and in pilot clinical trials warrant comprehensive evaluation of symptomatic and/or disease-modifying potential of chelation in PD.


Subject(s)
Iron Chelating Agents/therapeutic use , Iron/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Pyridones/therapeutic use , Animals , Cell Line , Combined Modality Therapy , Deferiprone , Disease Models, Animal , Double-Blind Method , Humans , Iron Chelating Agents/administration & dosage , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Oxidative Stress/drug effects , Pilot Projects , Pyridones/administration & dosage , Pyridones/pharmacology
7.
Haematologica ; 97(5): 670-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22180428

ABSTRACT

BACKGROUND: In transfusional siderosis, the iron binding capacity of plasma transferrin is often surpassed, with concomitant generation of non-transferrin-bound iron. Although implicated in tissue siderosis, non-transferrin-bound iron modes of cell ingress remain undefined, largely because of its variable composition and association with macromolecules. Using fluorescent tracing of labile iron in endosomal vesicles and cytosol, we examined the hypothesis that non-transferrin-bound iron fractions detected in iron overloaded patients enter cells via bulk endocytosis. DESIGN AND METHODS: Fluorescence microscopy and flow cytometry served as analytical tools for tracing non-transferrin-bound iron entry into endosomes with the redox-reactive macromolecular probe Oxyburst-Green and into the cytosol with cell-laden calcein green and calcein blue. Non-transferrin-bound iron-containing media were from sera of polytransfused thalassemia major patients and model iron substances detected in thalassemia major sera; cell models were cultured macrophages, and cardiac myoblasts and myocytes. RESULTS: Exposure of cells to ferric citrate together with albumin, or to non-transferrin-bound iron-containing sera from thalassemia major patients caused an increase in labile iron content of endosomes and cytosol in macrophages and cardiac cells. This increase was more striking in macrophages, but in both cell types was largely reduced by co-exposure to non-transferrin-bound iron-containing media with non-penetrating iron chelators or apo-transferrin, or by treatment with inhibitors of endocytosis. Endosomal iron accumulation traced with calcein-green was proportional to input non-transferrin-bound iron levels (r(2) = 0.61) and also preventable by pre-chelation. CONCLUSIONS: Our studies indicate that macromolecule-associated non-transferrin-bound iron can initially gain access into various cells via endocytic pathways, followed by iron translocation to the cytosol. Endocytic uptake of plasma non-transferrin-bound iron is a possible mechanism that can contribute to iron loading of cell types engaged in bulk/adsorptive endocytosis, highlighting the importance of its prevention by iron chelation.


Subject(s)
Biomarkers/metabolism , Endocytosis/physiology , Endosomes/metabolism , Iron/metabolism , Transferrin/metabolism , beta-Thalassemia/metabolism , Adolescent , Adult , Biological Transport , Cells, Cultured , Cytosol/metabolism , Humans , Insulinoma/metabolism , Insulinoma/pathology , Iron/blood , Iron Chelating Agents/pharmacology , Macrophages/cytology , Macrophages/metabolism , Microscopy, Fluorescence , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Young Adult , beta-Thalassemia/pathology
8.
Am J Hematol ; 87(1): 55-61, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22125177

ABSTRACT

Non-transferrin bound iron (NTBI) is commonly detected in patients with systemic iron overload whose serum iron-binding capacity has been surpassed. It has been perceived as an indicator of iron overload, impending organ damage and a chelation target in poly-transfused thalassemia patients. However, NTBI is a heterogeneous entity comprising various iron complexes, including a significant redox-active and readily chelatable fraction, which we have designated as "labile plasma iron" (LPI). We found that LPI levels can be affected by plasma components such as citrate, uric acid, and albumin. However, the inclusion of a mild metal mobilizing agent in the LPI assay (designated here as "eLPI"), at concentrations that do not affect transferrin-bound iron, largely overcomes such effects and provides a measure of the full NTBI content. We analyzed three distinct groups of poly-transfused, iron overloaded thalassemia patients: non-chelated children (3-13 yrs, Gaza, Palestine), chelated adolescents-young adults (13-28 yrs, Israel), and chelated adults (27-61 yrs, Israel) for LPI and eLPI. The eLPI levels in all three groups were roughly commensurate (r(2) = 0.61-0.75) with deferrioxamine-detectable NTBI, i.e., DCI. In older chelated patients, eLPI levels approximated those of LPI, but in poly-transfused unchelated children eLPI was notably higher than LPI, a difference attributed to plasma properties affected by labile iron due to lack of chelation, possibly reflecting age-dependent attrition of plasma components. We propose that the two formats of NTBI measurement presented here are complementary and used together could provide more comprehensive information on the forms of NTBI in patients and their response to chelation.


Subject(s)
Iron/metabolism , Thalassemia/metabolism , Transferrin/metabolism , Adolescent , Adult , Chelating Agents/metabolism , Child , Child, Preschool , Humans , Iron/blood , Middle Aged , Nitrilotriacetic Acid/metabolism , Oxidation-Reduction , Protein Binding , Thalassemia/blood , Young Adult
9.
Br J Pharmacol ; 164(2b): 406-18, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21091647

ABSTRACT

BACKGROUND AND PURPOSE: Systemic iron deficiency concomitant with macrophage iron retention is characteristic of iron-refractory anaemias associated with chronic disease. The systemic misdistribution of iron, which is further exacerbated by parenteral iron supplementation, is mainly attributable to iron retention exerted on resident macrophages by hepcidin-mediated down-regulation of the iron exporter ferroportin. We aimed at developing an experimental macrophage-based cell model that recapitulates pathophysiological features of iron misdistribution found in chronic disorders and use it as a screening platform for identifying agents with the potential for relocating the accumulated metal and restoring affected functions. EXPERIMENTAL APPROACH: A raw macrophage subline was selected as cell model of iron retention based on their capacity to take up polymeric iron or aged erythrocytes excessively, resulting in a demonstrable increase of cell labile iron pools and oxidative damage that are aggravated by hepcidin. KEY RESULTS: This model provided a three-stage high throughput screening platform for identifying agents with the combined ability to: (i) scavenge cell iron and thereby rescue macrophage cells damaged by iron-overload; (ii) bypass the ferroportin blockade by conveying the scavenged iron to other iron-starved cells in co-culture via transferrin but (iii) without promoting utilization of the scavenged iron by intracellular pathogens. As test agents we used chelators in clinical practice and found the oral chelator deferiprone fulfilled essentially all of the three criteria. CONCLUSIONS AND IMPLICATIONS: We provide a proof of principle for conservative iron relocation as complementary therapeutic approach for correcting the misdistribution of iron associated with chronic disease and exacerbated by parenteral iron supplementation.


Subject(s)
Iron Overload/drug therapy , Iron Overload/metabolism , Iron/metabolism , Macrophages/metabolism , Anemia, Iron-Deficiency/drug therapy , Anemia, Iron-Deficiency/metabolism , Animals , Antimicrobial Cationic Peptides/metabolism , Biological Transport/physiology , Cation Transport Proteins/metabolism , Cells, Cultured , Coculture Techniques , Deferiprone , Endocytosis/physiology , Erythrocytes/metabolism , Hepcidins , Humans , Iron/blood , Iron Chelating Agents/pharmacology , Iron Overload/blood , K562 Cells , Macrophages/drug effects , Mice , Oxidative Stress/physiology , Pyridones/pharmacology , Transferrin/metabolism , Tumor Cells, Cultured
10.
Leuk Res ; 34(12): 1560-5, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20615548

ABSTRACT

We report the first prospective study evaluating the effects of deferasirox on liver iron concentration (LIC), labile plasma iron (LPI) and pharmacokinetics (PK) along with serum ferritin values in patients with IPSS Low- and Intermediate-1 risk myelodysplastic syndromes (MDS) and evidence of iron overload. Twenty-four heavily transfused MDS patients were enrolled in a planned 52 weeks of therapy. PK studies showed dose-proportional total drug exposure. Data demonstrated that deferasirox was well tolerated and effectively reduced LIC, LPI and serum ferritin in the iron-overloaded patients with MDS who completed 24 and 52 weeks of therapy despite ongoing receipt of red blood cell transfusions.


Subject(s)
Benzoates/administration & dosage , Iron Chelating Agents/administration & dosage , Iron Overload/drug therapy , Myelodysplastic Syndromes/therapy , Triazoles/administration & dosage , Aged , Aged, 80 and over , Benzoates/adverse effects , Deferasirox , Dose-Response Relationship, Drug , Erythrocyte Transfusion , Female , Ferritins/blood , Humans , Iron/blood , Iron Chelating Agents/adverse effects , Iron Overload/blood , Iron Overload/etiology , Liver/metabolism , Male , Middle Aged , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/complications , Prospective Studies , Triazoles/adverse effects
11.
Can J Physiol Pharmacol ; 88(3): 187-96, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20393584

ABSTRACT

Defective iron utilization leading to either systemic or regional misdistribution of the metal has been identified as a critical feature of several different disorders. Iron concentrations can rise to toxic levels in mitochondria of excitable cells, often leaving the cytosol iron-depleted, in some forms of neurodegeneration with brain accumulation (NBIA) or following mutations in genes associated with mitochondrial functions, such as ABCB7 in X-linked sideroblastic anemia with ataxia (XLSA/A) or the genes encoding frataxin in Friedreich's ataxia (FRDA). In anemia of chronic disease (ACD), iron is withheld by macrophages, while iron levels in extracellular fluids (e.g., plasma) are drastically reduced. One possible therapeutic approach to these diseases is iron chelation, which is known to effectively reduce multiorgan iron deposition in iron-overloaded patients. However, iron chelation is probably inappropriate for disorders associated with misdistribution of iron within selected tissues or cells. One chelator in clinical use for treating iron overload, deferiprone (DFP), has been identified as a reversed siderophore, that is, an agent with iron-relocating abilities in settings of regional iron accumulation. DFP was applied to a cell model of FRDA, a paradigm of a disorder etiologically associated with cellular iron misdistribution. The treatment reduced the mitochondrial levels of labile iron pools (LIP) that were increased by frataxin deficiency. DFP also conferred upon cells protection against oxidative damage and concomitantly mediated the restoration of various metabolic parameters, including aconitase activity. Administration of DFP to FRDA patients for 6 months resulted in selective and significant reduction in foci of brain iron accumulation (assessed by T2* MRI) and initial functional improvements, with only minor changes in net body iron stores. The prospects of drug-mediated iron relocation versus those of chelation are discussed in relation to other disorders involving iron misdistribution, such as ACD and XLSA/A.


Subject(s)
Iron Overload/blood , Iron Overload/therapy , Iron/blood , Animals , Friedreich Ataxia/blood , Friedreich Ataxia/therapy , Humans , Iron Chelating Agents/therapeutic use , Parkinson Disease/blood , Parkinson Disease/therapy , Siderophores/therapeutic use , Treatment Outcome
12.
Biochem J ; 429(1): 185-93, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20408812

ABSTRACT

In the present study we analysed the mechanism of intracellular routing of iron acquired by erythroid cells via receptor-mediated endocytosis of Tf-Fe [Tf (transferrin)-iron]. Using real-time fluorimetry and flow cytometry, in conjunction with targeted fluorescent metal sensors, we monitored concurrently the cytosolic and mitochondrial changes in labile iron evoked by endocytosed Tf-Fe. In K562 human erythroleukaemia cells, most of the Tf-Fe was found to be delivered to the cytosolic labile iron pool by a saturable mechanism [60-120 nM Km (app)] that was quantitatively dependent on: Tf receptor levels, endosomal acidification/reduction for dislodging iron from Tf and ensuing translocation of labile iron into the cytosolic compartment. The parallel ingress of iron to mitochondria was also saturable, but with a relatively lower Km (app) (26-42 nM) and a lower maximal ingress per cell than into the cytosol. The ingress of iron into the mitochondrial labile iron pool was blocked by cytosol-targeted iron chelators, implying that a substantial fraction of Tf-Fe delivered to these organelles passes through the cytosol in non-occluded forms that remain accessible to high-affinity ligands. The present paper is the first report describing intracellular iron routing measured in intact cells in real-time and in quantitative terms, opening the road for also exploring the process in mixed-cell populations of erythroid origin.


Subject(s)
Computer Systems , Cytosol/metabolism , Fluorometry , Iron/metabolism , Mitochondria/metabolism , Transferrin/metabolism , Cytosol/chemistry , Endocytosis/physiology , Fluorescence , Fluorometry/methods , Humans , Iron/analysis , K562 Cells , Mitochondria/chemistry , Transferrin/analysis
13.
Nat Med ; 16(2): 177-82, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20098432

ABSTRACT

Individuals with beta-thalassemia develop progressive systemic iron overload, resulting in high morbidity and mortality. These complications are caused by labile plasma iron, which is taken up by parenchymal cells in a dysregulated manner; in contrast, erythropoiesis depends on transferrin-bound iron uptake via the transferrin receptor. We hypothesized that the ineffective erythropoiesis and anemia observed in beta-thalassemia might be ameliorated by increasing the amount of circulating transferrin. We tested the ability of transferrin injections to modulate iron metabolism and erythropoiesis in Hbb(th1/th1) mice, an experimental model of beta-thalassemia. Injected transferrin reversed or markedly improved the thalassemia phenotype in these mice. Specifically, transferrin injections normalized labile plasma iron concentrations, increased hepcidin expression, normalized red blood cell survival and increased hemoglobin production; this treatment concomitantly decreased reticulocytosis, erythropoietin abundance and splenomegaly. These results indicate that transferrin is a limiting factor contributing to anemia in these mice and suggest that transferrin therapy might be beneficial in human beta-thalassemia.


Subject(s)
Transferrin/therapeutic use , beta-Thalassemia/drug therapy , Animals , Apoptosis , Cell Survival , Disease Models, Animal , Erythropoietin/blood , Hematopoiesis, Extramedullary , Mice , Splenomegaly/drug therapy , Transferrin/administration & dosage , beta-Thalassemia/blood
14.
Exp Hematol ; 37(2): 172-83, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19059700

ABSTRACT

OBJECTIVE: Beta-thalassemia results from beta-globin gene mutations that lead to ineffective erythropoiesis, shortened red cell survival, and anemia. Patients with beta-thalassemia develop iron overload, despite which, hepcidin levels are low. This suggests that hepcidin regulation in beta-thalassemia is more sensitive to factors unrelated to iron state. Our preliminary data demonstrates that Hbb(th1/th1) mice, a model of beta-thalassemia intermedia, have lower bone marrow iron levels while levels in the liver and spleen are increased; the later account for the increased systemic iron burden in beta-thalassemia intermedia. We hypothesized that exogenous iron would improve anemia in beta-thalassemia intermedia despite systemic iron overload and further suppress hepcidin secondary to progressive expansion of erythroid precursors. MATERIALS AND METHODS: We investigate parameters involved in red cell production, precursor apoptosis, parenchymal iron distribution, and hepcidin expression in iron treated Hbb(th1/th1) mice. RESULTS: Exogenous iron results in an expansion of erythroid precursors in the liver and spleen, leading to an increase in the number of red cells, reticulocytes, and hemoglobin production. A decrease in hepcidin expression is also observed. CONCLUSIONS: These findings demonstrate for the first time that iron results in expansion of extramedullary erythropoiesis, which improves anemia and suggests that expansion of extramedullary erythropoiesis itself results in hepcidin suppression in beta-thalassemia intermedia.


Subject(s)
Erythropoiesis/drug effects , Hematinics/pharmacology , Hematopoiesis, Extramedullary/drug effects , Hemoglobins/biosynthesis , Iron-Dextran Complex/pharmacology , beta-Thalassemia/metabolism , Animals , Antimicrobial Cationic Peptides/biosynthesis , Antimicrobial Cationic Peptides/genetics , Bone Marrow/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Disease Models, Animal , Erythroid Precursor Cells/metabolism , Erythropoiesis/genetics , Hematopoiesis, Extramedullary/genetics , Hemoglobins/genetics , Hepcidins , Iron/metabolism , Mice , Mice, Knockout , Mutation , beta-Thalassemia/drug therapy , beta-Thalassemia/genetics
15.
Blood ; 112(13): 5219-27, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18796625

ABSTRACT

Various human disorders are associated with misdistribution of iron within or across cells. Friedreich ataxia (FRDA), a deficiency in the mitochondrial iron-chaperone frataxin, results in defective use of iron and its misdistribution between mitochondria and cytosol. We assessed the possibility of functionally correcting the cellular properties affected by frataxin deficiency with a siderophore capable of relocating iron and facilitating its metabolic use. Adding the chelator deferiprone at clinical concentrations to inducibly frataxin-deficient HEK-293 cells resulted in chelation of mitochondrial labile iron involved in oxidative stress and in reactivation of iron-depleted aconitase. These led to (1) restoration of impaired mitochondrial membrane and redox potentials, (2) increased adenosine triphosphate production and oxygen consumption, and (3) attenuation of mitochondrial DNA damage and reversal of hypersensitivity to staurosporine-induced apoptosis. Permeant chelators of higher affinity than deferiprone were not as efficient in restoring affected functions. Thus, although iron chelation might protect cells from iron toxicity, rendering the chelated iron bioavailable might underlie the capacity of deferiprone to restore cell functions affected by frataxin deficiency, as also observed in FRDA patients. The siderophore-like properties of deferiprone provide a rational basis for treating diseases of iron misdistribution, such as FRDA, anemia of chronic disease, and X-linked sideroblastic anemia with ataxia.


Subject(s)
Iron Chelating Agents/pharmacology , Iron-Binding Proteins/physiology , Iron/metabolism , Pyridones/pharmacology , Adenosine Triphosphate/biosynthesis , Cell Line , DNA Damage/drug effects , DNA, Mitochondrial , Deferiprone , Friedreich Ataxia , Humans , Mitochondria/chemistry , Mitochondria/drug effects , Oxygen Consumption/drug effects , Frataxin
16.
Int J Biochem Cell Biol ; 40(3): 350-4, 2008.
Article in English | MEDLINE | ID: mdl-17451993

ABSTRACT

Cells maintain organellar pools of "labile iron" (LI), despite its propensity for catalyzing the formation of reactive oxygen species. These pools are identifiable by iron-chelating probes and accessible to pharmacological agents. Cytosolic LI has been assumed to have a dual function: providing a rapidly adjustable source of iron for immediate metabolic utilization, and for sensing by iron-regulatory proteins (IRPs) that regulate iron uptake and compartmentalization via transferrin receptors and ferritin. However, it now appears that IRPs may respond both to fluctuations in LI per se and to secondary signals associated with redox-active species. Recent information also indicates that iron can be delivered to mitochondria via pathways that circumvent cytosolic LI, suggesting possible alternative mechanisms of cell iron mobilization and trafficking. We discuss the changing views of intracellular LI pools in relation to iron homeostasis and cellular distribution in physiological and pathological states.


Subject(s)
Cytoplasm/physiology , Ferritins/metabolism , Iron-Regulatory Proteins/metabolism , Iron/metabolism , Transferrin/metabolism , Animals , Humans , Metabolic Networks and Pathways , Mitochondria/physiology , Reactive Oxygen Species/metabolism , Receptors, Transferrin/metabolism
17.
Blood ; 111(3): 1690-9, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17975016

ABSTRACT

Various pathologies are characterized by the accumulation of toxic iron in cell compartments. In anemia of chronic disease, iron is withheld by macrophages, leaving extracellular fluids iron-depleted. In Friedreich ataxia, iron levels rise in the mitochondria of excitable cells but decrease in the cytosol. We explored the possibility of using deferiprone, a membrane-permeant iron chelator in clinical use, to capture labile iron accumulated in specific organelles of cardiomyocytes and macrophages and convey it to other locations for physiologic reuse. Deferiprone's capacity for shuttling iron between cellular organelles was assessed with organelle-targeted fluorescent iron sensors in conjunction with time-lapse fluorescence microscopy imaging. Deferiprone facilitated transfer of iron from extracellular media into nuclei and mitochondria, from nuclei to mitochondria, from endosomes to nuclei, and from intracellular compartments to extracellular apotransferrin. Furthermore, it mobilized iron from iron-loaded cells and donated it to preerythroid cells for hemoglobin synthesis, both in the presence and in the absence of transferrin. These unique properties of deferiprone underlie mechanistically its capacity to alleviate iron accumulation in dentate nuclei of Friedreich ataxia patients and to donate tissue-chelated iron to plasma transferrin in thalassemia intermedia patients. Deferiprone's shuttling properties could be exploited clinically for treating diseases involving regional iron accumulation.


Subject(s)
Iron Chelating Agents/pharmacology , Iron/metabolism , Animals , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Endosomes/drug effects , Endosomes/metabolism , Hemoglobins/biosynthesis , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Protein Binding , Rats , Transferrin/metabolism
18.
Am J Physiol Cell Physiol ; 293(4): C1383-94, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17670894

ABSTRACT

Non-transferrin-bound iron, commonly found in the plasma of iron-overloaded individuals, permeates into cells via pathways independent of the transferrin receptor. This may lead to excessive cellular accumulation of labile iron followed by oxidative damage and eventually organ failure. Mitochondria are the principal destination of iron in cells and a primary site of prooxidant generation, yet their mode of acquisition of iron is poorly understood. Using fluorescent probes sensitive to iron or to reactive oxygen species, targeted to cytosol and/or to mitochondria, we traced the ingress of labile iron into these compartments by fluorescence microscopy and quantitative fluorimetry. We observed that 1) penetration of non-transferrin-bound iron into the cytosol and subsequently into mitochondria occurs with barely detectable delay and 2) loading of the cytosol with high-affinity iron-binding chelators does not abrogate iron uptake into mitochondria. Therefore, a fraction of non-transferrin-bound iron acquired by cells reaches the mitochondria in a nonlabile form. The physiological role of occluded iron transfer might be to confer cells with a "safe and efficient cytosolic iron corridor" to mitochondria. However, such a mechanism might be deleterious in iron-overload conditions, because it could lead to surplus accumulation of iron in these critical organelles.


Subject(s)
Iron Chelating Agents/metabolism , Iron/metabolism , Mitochondria/metabolism , Aldehydes/metabolism , Animals , Biological Transport/drug effects , Cell Line , Cytosol/drug effects , Cytosol/metabolism , Deferoxamine/metabolism , Deferoxamine/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/metabolism , Egtazic Acid/pharmacology , Fluoresceins/metabolism , Fluorescent Dyes/metabolism , Fluorometry , Hydrazones/metabolism , Iron Chelating Agents/pharmacology , Microscopy, Fluorescence , Models, Biological , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Protein Binding , Rhodamines/metabolism , Spectrometry, Fluorescence , Transferrin/metabolism
19.
Blood ; 110(1): 401-8, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17379741

ABSTRACT

Genetic disorders of iron metabolism and chronic inflammation often evoke local iron accumulation. In Friedreich ataxia, decreased iron-sulphur cluster and heme formation leads to mitochondrial iron accumulation and ensuing oxidative damage that primarily affects sensory neurons, the myocardium, and endocrine glands. We assessed the possibility of reducing brain iron accumulation in Friedreich ataxia patients with a membrane-permeant chelator capable of shuttling chelated iron from cells to transferrin, using regimens suitable for patients with no systemic iron overload. Brain magnetic resonance imaging (MRI) of Friedreich ataxia patients compared with age-matched controls revealed smaller and irregularly shaped dentate nuclei with significantly (P < .027) higher H-relaxation rates R2*, indicating regional iron accumulation. A 6-month treatment with 20 to 30 mg/kg/d deferiprone of 9 adolescent patients with no overt cardiomyopathy reduced R2* from 18.3 s(-1) (+/- 1.6 s(-1)) to 15.7 s(-1) (+/- 0.7 s(-1); P < .002), specifically in dentate nuclei and proportionally to the initial R2* (r = 0.90). Chelator treatment caused no apparent hematologic or neurologic side effects while reducing neuropathy and ataxic gait in the youngest patients. To our knowledge, this is the first clinical demonstration of chelation removing labile iron accumulated in a specific brain area implicated in a neurodegenerative disease. The use of moderate chelation for relocating iron from areas of deposition to areas of deprivation has clinical implications for various neurodegenerative and hematologic disorders.


Subject(s)
Brain Chemistry/drug effects , Brain/metabolism , Friedreich Ataxia/drug therapy , Iron Chelating Agents/administration & dosage , Pyridones/administration & dosage , Adolescent , Adult , Biological Transport , Case-Control Studies , Deferiprone , Female , Humans , Iron/metabolism , Iron Chelating Agents/pharmacokinetics , Iron Chelating Agents/therapeutic use , Magnetic Resonance Imaging , Male , Pyridones/pharmacokinetics , Pyridones/therapeutic use , Treatment Outcome
20.
Blood ; 109(11): 5027-35, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17299088

ABSTRACT

Progressive iron overload is the most salient and ultimately fatal complication of beta-thalassemia. However, little is known about the relationship among ineffective erythropoiesis (IE), the role of iron-regulatory genes, and tissue iron distribution in beta-thalassemia. We analyzed tissue iron content and iron-regulatory gene expression in the liver, duodenum, spleen, bone marrow, kidney, and heart of mice up to 1 year old that exhibit levels of iron overload and anemia consistent with both beta-thalassemia intermedia (th3/+) and major (th3/th3). Here we show, for the first time, that tissue and cellular iron distribution are abnormal and different in th3/+ and th3/th3 mice, and that transfusion therapy can rescue mice affected by beta-thalassemia major and modify both the absorption and distribution of iron. Our study reveals that the degree of IE dictates tissue iron distribution and that IE and iron content regulate hepcidin (Hamp1) and other iron-regulatory genes such as Hfe and Cebpa. In young th3/+ and th3/th3 mice, low Hamp1 levels are responsible for increased iron absorption. However, in 1-year-old th3/+ animals, Hamp1 levels rise and it is rather the increase of ferroportin (Fpn1) that sustains iron accumulation, thus revealing a fundamental role of this iron transporter in the iron overload of beta-thalassemia.


Subject(s)
Antimicrobial Cationic Peptides/biosynthesis , Cation Transport Proteins/biosynthesis , Down-Regulation , Erythropoiesis , Gene Expression Regulation , Iron/pharmacokinetics , Up-Regulation , beta-Thalassemia/blood , Animals , Blood Transfusion , Flow Cytometry , Hepcidins , Iron/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , beta-Thalassemia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...