Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Mol Biol Cell ; 24(13): 2076-87, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23637465

RESUMEN

Ubiquitin accumulation in amyloid plaques is a pathological marker observed in the vast majority of neurodegenerative diseases, yet ubiquitin function in these inclusions is controversial. It has been suggested that ubiquitylated proteins are directed to inclusion bodies under stress conditions, when both chaperone-mediated refolding and proteasomal degradation are compromised or overwhelmed. Alternatively, ubiquitin and chaperones may be recruited to preformed inclusions to promote their elimination. We address this issue using a yeast model system, based on expression of several mildly misfolded degradation substrates in cells with altered chaperone content. We find that the heat shock protein 70 (Hsp70) chaperone pair Ssa1/Ssa2 and the Hsp40 cochaperone Sis1 are essential for degradation. Substrate ubiquitylation is strictly dependent on Sis1, whereas Ssa1 and Ssa2 are dispensable. Remarkably, in Ssa1/Ssa2-depleted cells, ubiquitylated substrates are sequestered into detergent-insoluble, Hsp42-positive inclusion bodies. Unexpectedly, sequestration is abolished by preventing substrate ubiquitylation. We conclude that Hsp40 is required for the targeting of misfolded proteins to the ubiquitylation machinery, whereas the decision to degrade or sequester ubiquitylated proteins is mediated by the Hsp70s. Accordingly, diminished Hsp70 levels, as observed in aging or certain pathological conditions, might be sufficient to trigger ubiquitin-dependent sequestration of partially misfolded proteins into inclusion bodies.


Asunto(s)
Adenosina Trifosfatasas/genética , Regulación Fúngica de la Expresión Génica , Proteínas del Choque Térmico HSP40/genética , Proteínas HSP70 de Choque Térmico/genética , Complejo de la Endopetidasa Proteasomal/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Ubiquitina/genética , Adenosina Trifosfatasas/metabolismo , Citoplasma/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Cuerpos de Inclusión/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Pliegue de Proteína , Estabilidad Proteica , Proteolisis , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Ubiquitina/metabolismo
2.
Haematologica ; 97(5): 670-8, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22180428

RESUMEN

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.


Asunto(s)
Biomarcadores/metabolismo , Endocitosis/fisiología , Endosomas/metabolismo , Hierro/metabolismo , Transferrina/metabolismo , Talasemia beta/metabolismo , Adolescente , Adulto , Transporte Biológico , Células Cultivadas , Citosol/metabolismo , Humanos , Insulinoma/metabolismo , Insulinoma/patología , Hierro/sangre , Quelantes del Hierro/farmacología , Macrófagos/citología , Macrófagos/metabolismo , Microscopía Fluorescente , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Adulto Joven , Talasemia beta/patología
3.
Am J Hematol ; 87(1): 55-61, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22125177

RESUMEN

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.


Asunto(s)
Hierro/metabolismo , Talasemia/metabolismo , Transferrina/metabolismo , Adolescente , Adulto , Quelantes/metabolismo , Niño , Preescolar , Humanos , Hierro/sangre , Persona de Mediana Edad , Ácido Nitrilotriacético/metabolismo , Oxidación-Reducción , Unión Proteica , Talasemia/sangre , Adulto Joven
4.
Br J Pharmacol ; 164(2b): 406-18, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21091647

RESUMEN

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.


Asunto(s)
Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/metabolismo , Hierro/metabolismo , Macrófagos/metabolismo , Anemia Ferropénica/tratamiento farmacológico , Anemia Ferropénica/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Transporte Biológico/fisiología , Proteínas de Transporte de Catión/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Deferiprona , Endocitosis/fisiología , Eritrocitos/metabolismo , Hepcidinas , Humanos , Hierro/sangre , Quelantes del Hierro/farmacología , Sobrecarga de Hierro/sangre , Células K562 , Macrófagos/efectos de los fármacos , Ratones , Estrés Oxidativo/fisiología , Piridonas/farmacología , Transferrina/metabolismo , Células Tumorales Cultivadas
5.
Can J Physiol Pharmacol ; 88(3): 187-96, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20393584

RESUMEN

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.


Asunto(s)
Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/terapia , Hierro/sangre , Animales , Ataxia de Friedreich/sangre , Ataxia de Friedreich/terapia , Humanos , Quelantes del Hierro/uso terapéutico , Enfermedad de Parkinson/sangre , Enfermedad de Parkinson/terapia , Sideróforos/uso terapéutico , Resultado del Tratamiento
6.
Br J Haematol ; 147(5): 744-51, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19764989

RESUMEN

Labile plasma iron (LPI), a non-transferrin-bound component of plasma iron detected in iron overload disorders is a potential source of cellular iron accumulation and ensuing oxidative damage. Periodic monitoring of LPI over a 24 h time-span was used to compare the ability of chelation to control daily LPI levels in 40 Thalassaemia major patients (9-11/group) who had been receiving one of three different chelation protocols for more than a year: Group I. deferrioxamine overnight, Group II. deferiprone daily, Group III. deferrioxamine and deferiprone sequentially. An additional group (Group IV) was treated with desferasirox for up to 6 months. The patterns of daily LPI recrudescence showed significant individual variations, especially in patients treated with deferrioxamine or deferiprone, although these patterns were maintained over 6-9 months of treatment in all groups. Group data analysis showed that the proportion of patients whose daily LPI were maintained within the normal range (<0.45 micromol/l) varied with treatment: 6/10 with deferrioxamine, 5/11 with deferiprone, 9/10 with deferrioxamine + deferiprone and 8/10 at the onset and 10/10 after 6 months treatment with deferasirox. Although the clinical significance and therapeutic value of LPI remain to be established, monitoring of daily LPI level may provide an analytical basis for assessing chelation efficacy in preventing daily LPI recrudescence.


Asunto(s)
Quelantes del Hierro/uso terapéutico , Hierro/sangre , Talasemia beta/tratamiento farmacológico , Adulto , Biomarcadores/sangre , Ritmo Circadiano , Deferiprona , Deferoxamina/uso terapéutico , Monitoreo de Drogas/métodos , Quimioterapia Combinada , Femenino , Humanos , Masculino , Persona de Mediana Edad , Piridonas/uso terapéutico , Talasemia beta/sangre
7.
Exp Hematol ; 37(2): 172-83, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19059700

RESUMEN

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.


Asunto(s)
Eritropoyesis/efectos de los fármacos , Hematínicos/farmacología , Hematopoyesis Extramedular/efectos de los fármacos , Hemoglobinas/biosíntesis , Complejo Hierro-Dextran/farmacología , Talasemia beta/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/biosíntesis , Péptidos Catiónicos Antimicrobianos/genética , Médula Ósea/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Células Precursoras Eritroides/metabolismo , Eritropoyesis/genética , Hematopoyesis Extramedular/genética , Hemoglobinas/genética , Hepcidinas , Hierro/metabolismo , Ratones , Ratones Noqueados , Mutación , Talasemia beta/tratamiento farmacológico , Talasemia beta/genética
8.
Blood ; 112(13): 5219-27, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18796625

RESUMEN

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.


Asunto(s)
Quelantes del Hierro/farmacología , Proteínas de Unión a Hierro/fisiología , Hierro/metabolismo , Piridonas/farmacología , Adenosina Trifosfato/biosíntesis , Línea Celular , Daño del ADN/efectos de los fármacos , ADN Mitocondrial , Deferiprona , Ataxia de Friedreich , Humanos , Mitocondrias/química , Mitocondrias/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Frataxina
9.
Int J Biochem Cell Biol ; 40(3): 350-4, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17451993

RESUMEN

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.


Asunto(s)
Citoplasma/fisiología , Ferritinas/metabolismo , Proteínas Reguladoras del Hierro/metabolismo , Hierro/metabolismo , Transferrina/metabolismo , Animales , Humanos , Redes y Vías Metabólicas , Mitocondrias/fisiología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Transferrina/metabolismo
10.
Blood ; 111(3): 1690-9, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17975016

RESUMEN

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.


Asunto(s)
Quelantes del Hierro/farmacología , Hierro/metabolismo , Animales , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Hemoglobinas/biosíntesis , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Unión Proteica , Ratas , Transferrina/metabolismo
11.
Blood ; 109(11): 5027-35, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17299088

RESUMEN

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.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/biosíntesis , Proteínas de Transporte de Catión/biosíntesis , Regulación hacia Abajo , Eritropoyesis , Regulación de la Expresión Génica , Hierro/farmacocinética , Regulación hacia Arriba , Talasemia beta/sangre , Animales , Transfusión Sanguínea , Citometría de Flujo , Hepcidinas , Hierro/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Talasemia beta/metabolismo
12.
Br J Haematol ; 135(1): 129-38, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16939499

RESUMEN

Beta-thalassaemia represents a group of diseases, in which ineffective erythropoiesis is accompanied by iron overload. In a mouse model of beta-thalassaemia, we observed that the liver expressed relatively low levels of hepcidin, which is a key factor in the regulation of iron absorption by the gut and of iron recycling by the reticuloendothelial system. It was hypothesised that, despite the overt iron overload, a putative plasma factor found in beta-thalassaemia might suppress liver hepcidin expression. Sera from beta-thalassaemia and haemochromatosis (C282Y mutation) patients were compared with those of healthy individuals regarding their capacity to induce changes the expression of key genes of iron metabolism in human HepG2 hepatoma cells. Sera from beta-thalassaemia major patients induced a major decrease in hepcidin (HAMP) and lipocalin2 (oncogene 24p3) (LCN2) expression, as well as a moderate decrease in haemojuvelin (HFE2) expression, compared with sera from healthy individuals. A significant correlation was found between the degree of downregulation of HAMP and HFE2 induced by beta-thalassaemia major sera (r = 0.852, P < 0.0009). Decreased HAMP expression was also found in HepG2 cells treated with sera from beta-thalassaemia intermedia patients. In contrast, the majority of sera from hereditary haemochromatosis patients induced an increase in HAMP expression, which correlated with transferrin (Tf) saturation (r = 0.765, P < 0.0099). Our results suggest that, in beta-thalassaemia, serum factors might override the potential effect of iron overload on HAMP expression, thereby providing an explanation for the failure to arrest excessive intestinal iron absorption in these patients.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/biosíntesis , Regulación hacia Abajo , Hepatocitos/metabolismo , Proteínas de la Membrana/biosíntesis , Talasemia beta/sangre , Proteínas de Fase Aguda/biosíntesis , Proteínas de Fase Aguda/genética , Péptidos Catiónicos Antimicrobianos/genética , Transfusión Sanguínea , Línea Celular , Proteínas Ligadas a GPI , Hemocromatosis/sangre , Proteína de la Hemocromatosis , Hepcidinas , Humanos , Lipocalina 2 , Lipocalinas , Proteínas de la Membrana/genética , Reacción en Cadena de la Polimerasa/métodos , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Talasemia beta/terapia
13.
Blood ; 108(9): 3195-203, 2006 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16835377

RESUMEN

Labile iron in hemosiderotic plasma and tissue are sources of iron toxicity. We compared the iron chelators deferoxamine, deferiprone, and deferasirox as scavengers of labile iron in plasma and cardiomyocytes at therapeutic concentrations. This comprised chelation of labile plasma iron (LPI) in samples from thalassemia patients; extraction of total cellular iron; accessing labile iron accumulated in organelles and preventing formation of reactive-oxidant species; and restoring impaired cardiac contractility. Neonatal rat cardiomyocytes were used for monitoring chelator extraction of LCI (labile cell iron) as 59Fe; assessing in situ cell iron chelation by epifluorescence microscope imaging using novel fluorescent sensors for iron and reactive oxygen species (ROS) selectively targeted to organelles, and monitoring contractility by time-lapse microscopy. At plasma concentrations attained therapeutically, all 3 chelators eliminated LPI but the orally active chelators rapidly gained access to the LCI pools of cardiomyocytes, bound labile iron, attenuated ROS formation, extracted accumulated iron, and restored contractility impaired by iron overload. The effect of deferoxamine at therapeutically relevant concentrations was primarily by elimination of LPI. The rapid accessibility of the oral chelators deferasirox and deferiprone to intracellular labile iron compartments renders them potentially efficacious for protection from and possibly reversal of cardiac damage induced by iron overload.


Asunto(s)
Quelantes del Hierro/farmacología , Hierro/metabolismo , Miocardio/metabolismo , Animales , Animales Recién Nacidos , Fluoresceínas , Corazón/efectos de los fármacos , Hierro/sangre , Hierro/toxicidad , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/fisiología , Células Musculares/efectos de los fármacos , Células Musculares/fisiología , Ratas , Especies Reactivas de Oxígeno/metabolismo
14.
Circulation ; 113(16): 1942-9, 2006 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-16618820

RESUMEN

BACKGROUND: Epidemiological studies aimed at correlating coronary heart disease (CHD) with serum ferritin levels have thus far yielded inconsistent results. We hypothesized that a labile iron component associated with non-transferrin-bound iron (NTBI) that appears in individuals with overt or cryptic iron overload might be more suitable for establishing correlations with CHD. METHODS AND RESULTS: We investigated the relation of NTBI, serum iron, transferrin saturation, and serum ferritin with risk of CHD and acute myocardial infarction (AMI). The cohort used comprised a population-based sample of 11 471 postmenopausal women aged 49 to 70 years at enrollment in 1993 to 1997. During a median follow-up of 4.3 years (quartile limits Q1 to Q3: 3.3 to 5.4), 185 CHD events were identified, including 66 AMI events. We conducted a case-cohort study using all CHD cases and a random sample from the baseline cohort (n=1134). A weighted Cox proportional hazards model was used to estimate hazard ratios for tertiles of iron variables in relation to CHD and AMI. Adjusted hazard ratios of women in the highest NTBI tertile (range 0.38 to 3.51) compared with the lowest (range -2.06 to -0.32) were 0.84 (95% confidence interval 0.61 to 1.16) for CHD and 0.47 (95% confidence interval 0.31 to 0.71) for AMI. The results were similar for serum iron, transferrin saturation, and serum ferritin. CONCLUSIONS: Our results show no excess risk of CHD or AMI within the highest NTBI tertile compared with the lowest but rather seem to demonstrate a decreased risk. Additional studies are warranted to confirm our findings.


Asunto(s)
Enfermedad Coronaria/etiología , Hierro/sangre , Posmenopausia/sangre , Anciano , Proteína C-Reactiva/análisis , Estudios de Casos y Controles , Enfermedad Coronaria/sangre , Femenino , Estudios de Seguimiento , Humanos , Persona de Mediana Edad , Modelos de Riesgos Proporcionales , Transferrina/análisis
15.
Anal Biochem ; 341(2): 241-50, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15907869

RESUMEN

Non-transferrin-bound iron (NTBI) appears in the circulation of patients with iron overload. Various methods to measure NTBI were comparatively assessed as part of an international interlaboratory study. Six laboratories participated in the study, using methods based on iron mobilization and detection with iron chelators or on reactivity with bleomycin. Serum samples of 12 patients with hereditary (n=11) and secondary (n=1) hemochromatosis were measured during a 3-day analysis using 4 determinations per sample per day, making a total of 144 measurements per laboratory. Bland-Altman plots for repeated measurements are presented. The methods differed widely in mean serum NTBI level (range 0.12-4.32mumol/L), between-sample variation (SD range 0.20-2.13mumol/L and CV range 49.3-391.3%), and within-sample variation (SD range 0.02-0.45mumol/L and CV range 4.4-193.2%). The results obtained with methods based on chelators correlated significantly (R(2) range 0.86-0.99). On the other hand, NTBI values obtained by the various methods related differently from those of serum transferrin saturation (TS) when expressed in terms of both regression coefficients and NTBI levels at TS of 50%. Recent studies underscore the clinical relevance of NTBI in the management of iron-overloaded patients. However, before measurement of NTBI can be introduced into clinical practice, there is a need for more reproducible protocols as well as information on which method best represents the pathophysiological phenomenon and is most pertinent for diagnostic and therapeutic purposes.


Asunto(s)
Hemocromatosis/diagnóstico , Hierro/sangre , Bleomicina/química , Análisis Químico de la Sangre/normas , Quelantes/química , Humanos , Isoformas de Proteínas/sangre , Transferrina/análisis
16.
Best Pract Res Clin Haematol ; 18(2): 277-87, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15737890

RESUMEN

Labile plasma iron (LPI) represents a component of non-transferrin-bound iron (NTBI) that is both redox-active and chelatable, capable of permeating into organs and inducing tissue iron overload. It appears in various types of hemosiderosis (transfusional and non-transfusional) and in other iron-overload conditions. Sustained levels of LPI could over time compromise organ (e.g. heart) function and patient survival. With the advent of methods for measuring LPI in the clinical setting, it has become possible to assess the implications of LPI in the management of iron overload based on regimens of iron chelation. As LPI is detected primarily in patients with transfusional iron overload and other forms of hemosiderosis, we review here regimens of iron chelation with deferrioxamine and deferiprone (separately or combined) in terms of their efficacy in minimizing daily exposure to LPI in thalassemia major and thalassemia intermedia patients.


Asunto(s)
Sobrecarga de Hierro/metabolismo , Hierro/sangre , Talasemia/sangre , Deferiprona , Humanos , Hierro/metabolismo , Quelantes del Hierro/metabolismo , Quelantes del Hierro/uso terapéutico , Oxidación-Reducción , Piridonas/uso terapéutico , Talasemia/tratamiento farmacológico , Talasemia/etiología , Talasemia beta/sangre , Talasemia beta/tratamiento farmacológico
17.
Blood ; 105(11): 4527-31, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15671444

RESUMEN

Labile plasma iron (LPI) represents the redox active component of non-transferrin-bound iron (NTBI). Its presence in thalassemic patients has been recently reported. The aim of the present study was to quantify LPI in HFE genetic hemochromatosis (GH) and to characterize the mechanisms accounting for its appearance. We studied 159 subjects subdivided into the following groups: (1) 23 with iron overloaded GH; (2) 14 with iron-depleted GH; (3) 26 with dysmetabolic hepatosiderosis; (4) 33 with alcoholic cirrhosis; (5) 63 healthy controls. Both NTBI and LPI were substantially higher in patients with iron-overloaded GH than in those with iron-depleted GH or in healthy controls. LPI was significantly correlated with serum transaminase increase in this group. LPI was elevated in the alcoholic cirrhosis subgroup of severely affected patients. LPI was found essentially when transferrin saturation exceeded 75%, regardless of the etiologic condition. Transferrin saturation above 75% was related to iron overload in GH and to liver failure in alcoholic cirrhosis. LPI is present in C282Y/C282Y hemochromatosis and may be a marker of toxicity due to its potential for catalyzing the generation of reactive oxygen radicals in vivo.


Asunto(s)
Hemocromatosis/genética , Hierro/sangre , Mutación Missense , Adulto , Estudios de Casos y Controles , Genotipo , Hemocromatosis/sangre , Proteína de la Hemocromatosis , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Hierro/metabolismo , Deficiencias de Hierro , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/genética , Cirrosis Hepática Alcohólica/sangre , Cirrosis Hepática Alcohólica/genética , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Oxidación-Reducción , Estudios Prospectivos , Transaminasas/sangre , Transferrina/metabolismo
19.
Blood ; 104(5): 1504-10, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15155464

RESUMEN

Persistent levels of plasma nontransferrin bound iron (NTBI) have been associated with tissue iron overload and toxicity. We characterized NTBI's susceptibility to deferoxamine (directly chelatable iron [DCI]) and redox activity (labile plasma iron [LPI]) during the course of long-term, continuous L1 (deferiprone) treatment of patients with hemoglobin E disease and beta-thalassemia (n = 17). In 97% of serum samples (n = 267), the LPI levels were more than 0.4 microM (mean +/- SEM, 3.1 +/- 0.2 microM) and the percent transferrin (Tf) saturation more than 85 (111 +/- 6), whereas only in 4% of sera were the LPI levels more than 0.4 microM for Tf saturation less than 85%. Daily administration of L1 (50 mg/kg) for 13 to 17 months caused both LPI and DCI to decrease from respective initial 5.1 +/- 0.5 and 5.4 +/- 0.6 microM to steady mean levels of 2.18 +/- 0.24 and 2.81 +/- 0.14 microM. The steady lowest levels of LPI and DCI were attained after 6 to 8 months, with a half time (t(1/2)) of 2 to 3 months. Serum ferritin and red cell membrane-associated iron followed a similar course but attained steady basal levels only after 10 to 12 months of continuous treatment, with a t(1/2) of 5 to 7 months. These studies indicate that LPI and DCI can serve as early indicators of iron overload and as measures for the effectiveness of iron chelation in reducing potentially toxic iron in the plasma.


Asunto(s)
Hemoglobina E , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Hierro/sangre , Piridonas/uso terapéutico , Talasemia beta/tratamiento farmacológico , Biomarcadores , Deferiprona , Membrana Eritrocítica/metabolismo , Estudios de Seguimiento , Humanos , Sobrecarga de Hierro/sangre , Oxidación-Reducción , Valor Predictivo de las Pruebas , Transferrina/metabolismo , Talasemia beta/sangre
20.
Hematol J ; 5(1): 32-4, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14745427

RESUMEN

INTRODUCTION: The abnormalities in iron metabolism associated with megaloblastic anemia are rapidly reversed by B(12) therapy in pernicious anemia (PA). Although non-tranferrin-bound plasma iron (NTBI) was previously shown to be associated with severe iron overload, its origin is unknown. METHODS AND RESULTS: Four patients with PA were studied before and after B(12) treatment. NTBI was measured by a fluorescence-based one-step assay. All patients had very high transferrin saturation, NTBI values ranging from 1.1 to 2.6 micromol/l and normal serum ferritins. B(12) treatment resulted in the disappearance of NTBI and normalization of transferrin saturation within 22-42 h. CONCLUSIONS: The prompt disappearance of NTBI following B(12) therapy implicates catabolic iron derived from ineffective erythropoiesis as the major source of NTBI in untreated PA and possibly in thalassemia major and sideroblastic anemia. Our findings offer further insight into the pathogenesis of NTBI in diseases associated with abnormal erythropoiesis.


Asunto(s)
Anemia Megaloblástica/sangre , Anemia Megaloblástica/etiología , Hierro/sangre , Vitamina B 12/farmacología , Adulto , Anciano , Anemia Megaloblástica/tratamiento farmacológico , Eritropoyesis , Femenino , Ferritinas/sangre , Humanos , Hierro/metabolismo , Sobrecarga de Hierro/etiología , Masculino , Persona de Mediana Edad , Transferrina/análisis , Vitamina B 12/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA