Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters










Publication year range
1.
Semin Immunol ; 24(6): 393-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23375134

ABSTRACT

Iron is necessary for both mammalian cells and microorganisms, which fiercely compete for this essential nutrient. Accordingly, macrophages exploit the denial of iron from microbial pathogens as an important strategy to accomplish their key role in innate immunity and host defense. Macrophages employ multiple mechanisms to accumulate iron and thus contain microbial infections, but this may come at a price. In particular, at the systemic level iron sequestration in the reticuloendothelial cells can lead to the development of anemia of chronic disease. At the local level, iron sequestration in macrophages, which is targeted to extracellular invaders, can in turn favor intracellular pathogens. Moreover, iron accumulation can per se promote pro-inflammatory activation of macrophages and consequently contribute to maintain the process of inflammation, without resolution. Finally, the peculiar iron trafficking that characterizes alternatively polarized macrophages can influence neighboring cells in the microenvironment and impact on the resolution phase of inflammation. In this review, we describe the role of macrophages in iron metabolism in the context of host defense and iron balance.


Subject(s)
Immunity, Innate/immunology , Inflammation/immunology , Iron/metabolism , Macrophages/immunology , Macrophages/metabolism , Anemia/etiology , Animals , Homeostasis/immunology , Humans , Inflammation/metabolism , Macrophage Activation/immunology , Macrophages/cytology
2.
Br J Pharmacol ; 163(2): 299-312, 2011 May.
Article in English | MEDLINE | ID: mdl-21232037

ABSTRACT

BACKGROUND AND PURPOSE: Iron aggravates the cardiotoxicity of doxorubicin, a widely used anticancer anthracycline, and the iron chelator dexrazoxane is the only agent protecting against doxorubicin cardiotoxicity; however, the mechanisms underlying the role of iron in doxorubicin-mediated cardiotoxicity and the protective role of dexrazoxane remain to be established. As iron is required for the degradation of hypoxia-inducible factors (HIF), which control the expression of antiapoptotic and protective genes, we tested the hypothesis that dexrazoxane-dependent HIF activation may mediate the cardioprotective effect of dexrazoxane. EXPERIMENTAL APPROACH: Cell death, protein levels (by immunoblotting) and HIF-mediated transcription (using reporter constructs) were evaluated in the rat H9c2 cardiomyocyte cell line exposed to low doses of doxorubicin with or without dexrazoxane pretreatment. HIF levels were genetically manipulated by transfecting dominant-negative mutants or short hairpin RNA. KEY RESULTS: Treatment with dexrazoxane induced HIF-1α and HIF-2α protein levels and transactivation capacity in H9c2 cells. It also prevented the induction of cell death and apoptosis by exposure of H9c2 cells to clinically relevant concentrations of doxorubicin. Suppression of HIF activity strongly reduced the protective effect of dexrazoxane. Conversely, HIF-1α overexpression protected against doxorubicin-mediated cell death and apoptosis also in cells not exposed to the chelator. Exposure to dexrazoxane increased the expression of the HIF-regulated, antiapoptotic proteins survivin, Mcl1 and haem oxygenase. CONCLUSIONS AND IMPLICATIONS: Our results showing HIF-dependent prevention of doxorubicin toxicity in dexrazoxane-treated H9c2 cardiomyocytes suggest that HIF activation may be a mechanism contributing to the protective effect of dexrazoxane against anthracycline cardiotoxicity.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Basic Helix-Loop-Helix Transcription Factors/physiology , Doxorubicin/toxicity , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Iron Chelating Agents/pharmacology , Myocytes, Cardiac/drug effects , Razoxane/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Gene Expression Profiling , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Protein Binding , Rats , Transcriptional Activation
3.
Genes Nutr ; 1(1): 25-39, 2006 Mar.
Article in English | MEDLINE | ID: mdl-18850218

ABSTRACT

Iron is an important cofactor required for a number of essential cell functions and hence is a vital nutrient. However, iron can also be dangerous as a catalyst of free radical reactions. Accordingly, intracellular iron homeostasis and body iron balance are tightly regulated. In this review, we presented an overview of the remarkable new insights that over the last years have been gained into the multifaceted and sophisticated molecular mechanisms controlling iron acquisition, storage and release. We also reviewed the data about nutrition-related abnormalities of iron metabolism, such as iron overload and deficiency. Finally, we discussed how pathogenic microorganisms and host cells compete for iron, a battle whose outcome has a relevant role in infectious disease.

4.
Cancer Res ; 61(23): 8422-8, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11731422

ABSTRACT

Changes in iron homeostasis have been implicated in cardiotoxicity induced by the anticancer anthracycline doxorubicin (DOX). Certain products of DOX metabolism, like the secondary alcohol doxorubicinol (DOXol) or reactive oxygen species (ROS), may contribute to cardiotoxicity by inactivating iron regulatory proteins (IRP) that modulate the fate of mRNAs for transferrin receptor and ferritin. It is important to know whether DOXol and ROS act by independent or combined mechanisms. Therefore, we monitored IRP activities in H9c2 rat embryo cardiomyocytes exposed to DOX or to analogues which were selected to achieve a higher formation of secondary alcohol metabolite (daunorubicin), a concomitant increase of alcohol metabolite and decrease of ROS (5-iminodaunorubicin), or a defective conversion to alcohol metabolite (mitoxantrone). On the basis of such multiple comparisons, we characterized that DOXol was able to remove iron from the catalytic Fe-S cluster of cytoplasmic aconitase, making this enzyme switch to the cluster-free IRP-1. ROS were not involved in this step, but they converted the IRP-1 produced by DOXol into a null protein which did not bind to mRNA, nor was it able to switch back to aconitase. DOX was also shown to inactivate IRP-2, which does not assemble or disassemble a Fe-S cluster. Comparisons between DOX and the analogues revealed that IRP-2 was inactivated only by ROS. Thus, DOX can inactivate both IRP through a sequential action of DOXol and ROS on IRP-1 or an independent action of ROS on IRP-2. This information serves guidelines for designing anthracyclines that spare iron homeostasis and induce less severe cardiotoxicity.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , Heart Diseases/chemically induced , Heart/drug effects , Iron-Sulfur Proteins/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Animals , Antibiotics, Antineoplastic/metabolism , Cells, Cultured , Doxorubicin/metabolism , Heart Diseases/metabolism , Iron Regulatory Protein 1 , Iron Regulatory Protein 2 , Iron-Regulatory Proteins , Myocardium/cytology , Myocardium/metabolism , Rats , Reactive Oxygen Species/metabolism
5.
Br J Haematol ; 114(1): 226-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11472372

ABSTRACT

As ceruloplasmin (Cp) seems to be involved in iron mobilization, serum Cp levels were measured in 35 patients with hereditary haemochromatosis (HH), 12 with acquired iron overload (AIO) and 36 healthy subjects. Cp was lower in HH patients than in controls (P < 0.001); no difference was found between untreated HH patients and those on a phlebotomy programme (P = 0.07) and between the HH patients carrying the normal and the mutated alleles of the HFE gene (P = 0.8). Cp levels in AIO subjects were significantly higher than in HH patients (P < 0.004) and similar to those of controls (P = 0.2). No differences in albumin, alpha1 acid glycoprotein and copper serum levels were observed in the three groups.


Subject(s)
Ceruloplasmin/analysis , Hemochromatosis/blood , Hemochromatosis/genetics , Adult , Case-Control Studies , Female , Hemochromatosis/therapy , Humans , Iron Overload/blood , Male , Middle Aged , Phlebotomy , Statistics, Nonparametric
6.
Biochem J ; 352 Pt 2: 241-50, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11085915

ABSTRACT

The capacity of readily exchanging electrons makes iron not only essential for fundamental cell functions, but also a potential catalyst for chemical reactions involving free-radical formation and subsequent oxidative stress and cell damage. Cellular iron levels are therefore carefully regulated in order to maintain an adequate substrate while also minimizing the pool of potentially toxic 'free iron'. Iron homoeostasis is controlled through several genes, an increasing number of which have been found to contain non-coding sequences [i.e. the iron-responsive elements (IREs)] which are recognized at the mRNA level by two cytoplasmic iron-regulatory proteins (IRP-1 and IRP-2). The IRPs belong to the aconitase superfamily. By means of an Fe-S-cluster-dependent switch, IRP-1 can function as an mRNA-binding protein or as an enzyme that converts citrate into isocitrate. Although structurally and functionally similar to IRP-1, IRP-2 does not seem to assemble a cluster nor to possess aconitase activity; moreover, it has a distinct pattern of tissue expression and is modulated by means of proteasome-mediated degradation. In response to fluctuations in the level of the 'labile iron pool', IRPs act as key regulators of cellular iron homoeostasis as a result of the translational control of the expression of a number of iron metabolism-related genes. Conversely, various agents and conditions may affect IRP activity, thereby modulating iron and oxygen radical levels in different pathobiological settings. As the number of mRNAs regulated through IRE-IRP interactions keeps growing, the definition of IRPs as iron-regulatory proteins may in the near future become limiting as their role expands to other essential metabolic pathways.


Subject(s)
Iron-Sulfur Proteins/physiology , Iron/metabolism , RNA-Binding Proteins/physiology , Cell Division , Cell Hypoxia , Homeostasis , Iron Regulatory Protein 1 , Iron Regulatory Protein 2 , Iron-Regulatory Proteins , Oxidative Stress , Xenobiotics/pharmacology
7.
Lab Invest ; 80(12): 1781-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11140691

ABSTRACT

Hemozoin (malaria pigment), a polymer of hematin (ferri-protoporphyrin IX) derived from hemoglobin ingested by intraerythrocytic plasmodia, modulates cytokine production by phagocytes. Mouse peritoneal macrophages (PM) fed with synthetic beta-hematin (BH), structurally identical to native hemozoin, no longer produce tumor necrosis factor alpha (TNFalpha) and nitric oxide (NO) in response to lipopolysaccharide (LPS). Impairment of NO synthesis is due to inhibition of inducible nitric oxide synthase (iNOS) production. BH-mediated inhibition of PM functions cannot be ascribed to iron release from BH because neither prevention by iron chelators nor down-regulation of iron-regulatory protein activity was detected. Inhibition appears to be related to pigment-induced oxidative stress because (a) thiol compounds partially restored PM functions, (b) heme oxygenase (HO-1) and catalase mRNA levels were up-regulated, and (c) free radicals production increased in BH-treated cells. The antioxidant defenses of the cells determine the response to BH: microglia cells, which show a lower extent of induction of HO-1 and catalase mRNAs and lower accumulation of oxygen radicals, are less sensitive to the inhibitory effect of BH on cytokine production. Results indicate that BH is resistant to degradation by HO-1 and that heme-iron mediated oxidative stress may contribute to malaria-induced immunosuppression. This study may help correlate the different clinical manifestations of malaria, ranging from uncomplicated to severe disease, with dysregulation of phagocyte functions and promote better therapeutic strategies to counteract the effects of hemozoin accumulation.


Subject(s)
Hemeproteins/pharmacology , Macrophages, Peritoneal/physiology , Oxidative Stress , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Line , Cell Survival , Cells, Cultured , Enzyme Induction , Female , Free Radicals/metabolism , Heme Oxygenase (Decyclizing)/biosynthesis , Heme Oxygenase-1 , Hemin/pharmacology , Iron/pharmacology , Iron-Regulatory Proteins , Iron-Sulfur Proteins/analysis , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Membrane Proteins , Mice , Microglia/cytology , Microglia/physiology , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , Pigments, Biological/pharmacology , RNA-Binding Proteins/analysis , Recombinant Proteins/pharmacology
8.
Nucleic Acids Res ; 27(21): 4223-7, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10518614

ABSTRACT

Treatment with iron chelators mimics hypoxic induction of the hypoxia inducible factor (HIF-1) which activates transcription by binding to hypoxia responsive elements (HRE). We investigated whether HIF-1 is involved in transcriptional activation of the transferrin receptor (TfR), a membrane protein which mediates cellular iron uptake, in response to iron deprivation. The transcription rate of the TfR gene in isolated nuclei was up-regulated by treatment of Hep3B human hepatoma cells with the iron chelator desferrioxamine (DFO). The role of HIF-1 in the activation of TfR was indicated by the following observations: (i) DFO-dependent activation of a luciferase reporter gene in transfected Hep3B cells was mediated by a fragment of the human TfR promoter containing a putative HRE sequence; (ii) mutation of this sequence prevented stimulation of luciferase activity; (iii) binding to this sequence of HIF-1alpha, identified by competition experiments and supershift assays, was induced by DFO. Furthermore, in mouse hepatoma cells unable to assemble functional HIF-1, inducibility of TfR transcription by DFO was lost and TfR mRNA up-regulation was reduced. These results, which show the role of HIF-1 in the control of TfR gene expression in conditions of iron depletion, give insights into the mechanisms of transcriptional regulation which concur with the well-characterized post-transcriptional control of TfR expression to expand the extent of response to iron deficiency.


Subject(s)
DNA-Binding Proteins/metabolism , Iron Chelating Agents/pharmacology , Iron/metabolism , Nuclear Proteins/metabolism , Receptors, Transferrin/genetics , Transcription Factors , Transcriptional Activation/drug effects , Animals , Base Sequence , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Hypoxia/drug effects , Cobalt/pharmacology , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Deferoxamine/pharmacology , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Iron Deficiencies , Mice , Mutation/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , Receptors, Transferrin/metabolism , Response Elements/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Transfection , Tumor Cells, Cultured
9.
J Biol Chem ; 274(34): 24142-6, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10446187

ABSTRACT

The tight relationship between oxygen and iron prompted us to investigate whether the expression of transferrin receptor (TfR), which mediates cellular iron uptake, is regulated by hypoxia. In Hep3B human hepatoma cells incubated in 1% O(2) or treated with CoCl(2), which mimics hypoxia, we detected a 3-fold increase of TfR mRNA despite a decrease of iron regulatory proteins activity. Increased expression resulted from a 4-fold stimulation of the nuclear transcription rate of the TfR gene by both hypoxia and CoCl(2). A role for hypoxia-inducible factor (HIF-1), which activates transcription by binding to hypoxia-responsive elements in the activation of TfR, stems from the following observations. (a) Hypoxia and CoCl(2)-dependent expression of luciferase reporter gene in transiently transfected Hep3B cells was mediated by a fragment of the human TfR promoter containing a putative hypoxia-responsive element sequence, (b) mutation of this sequence prevented hypoxic stimulation of luciferase activity, (c) binding to this sequence of HIF-1alpha, identified by competition experiments and supershift assays, was induced in Hep3B cells by hypoxia and CoCl(2). In erythroid K562 cells, the same treatments did not affect iron regulatory proteins activity, thus resulting in a stimulation of TfR gene expression higher than in hepatoma cells.


Subject(s)
Cell Hypoxia , DNA-Binding Proteins/physiology , Gene Expression Regulation , Nuclear Proteins/physiology , Receptors, Transferrin/genetics , Transcription Factors , Transcriptional Activation , DNA/metabolism , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Iron-Regulatory Proteins , Iron-Sulfur Proteins/analysis , Promoter Regions, Genetic , RNA, Messenger/analysis , RNA-Binding Proteins/analysis , Tumor Cells, Cultured
10.
FASEB J ; 13(2): 199-212, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9973309

ABSTRACT

The clinical use of anticancer anthracyclines is limited by the development of a distinctive and life-threatening form of cardiomyopathy upon chronic treatment. Commonly accepted mechanistic hypotheses have assigned a pivotal role to iron, which would act as a catalyst for free radical reactions and oxidative stress. Although perhaps involved in acute aspects of anthracycline cardiotoxicity, the role of free radical-based mechanisms in long-term effects has been challenged on both experimental and clinical grounds, and alternative hypotheses independent of iron and free radicals have flourished. More recently, studies of the role of C-13 hydroxy metabolites of anthracyclines have provided new perspectives on the role of iron in the cardiotoxicity of these drugs, showing that such metabolites can impair intracellular iron handling and homeostasis. The present review applies a multisided approach to the critical evaluation of various hypotheses proposed over the last decade for the role of iron in anthracycline-induced cardiotoxicity. The main goal of the authors is to build a unifying pattern that would both account for hitherto unexplained experimental observations and help design novel and more rational strategies toward a much-needed improvement in the therapeutic index of anthracyclines.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Antineoplastic Agents/adverse effects , Cardiomyopathies/chemically induced , Cardiomyopathies/metabolism , Iron/metabolism , Animals , Humans
11.
Hepatology ; 29(2): 464-70, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9918923

ABSTRACT

Nitric oxide (NO) donors S-nitroso-N-acetylpenicillamine (SNAP) and sodium nitroprusside (SNP) modulate iron regulatory protein (IRP) activity and may, therefore, affect iron uptake through transferrin receptor expression. However, iron also enters the cell as nontransferrin-bound iron (NTBI), and the aim of this study was to evaluate the effects of NO donors on NTBI transport in HepG2 cells, a model of liver physiology. Incubation with SNP and SNAP led to a time- and concentration-dependent reduction in Fe3+ and Fe2+ uptake, thus indicating an effect on the transporter rather than on the reductase. In terms of Fe2+ uptake, no variations in the Michaelis-Menten constant (Km) and a reduction in maximum uptake (Vmax) (50, 33, and 16.6 fmol/microgram protein/min in control, SNP-, and SNAP-treated cells, respectively) were detected, which suggested a decrease in the number of putative NTBI transport protein(s). Gel shift assays showed that IRP activity was reduced by SNP and slightly increased by SNAP. Northern blot analysis of transferrin receptor messenger RNA (mRNA) levels showed variations similar to those observed for IRPs, but both NO donors increased L-ferritin mRNA levels and had no effect on the stimulator of Fe transport (SFT) mRNA. In conclusion, NO donors significantly reduce NTBI transport in HepG2 cells, an effect that seems to be IRP and SFT independent. Moreover, the reduction in NTBI uptake after NO treatment suggests that this form of iron may play a minor role in the increased hepatic iron stores observed in inflammation or that other liver cells are more involved in this pathological condition.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Iron/metabolism , Liver Neoplasms/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide/pharmacology , Transferrin/metabolism , Apoptosis , Biological Transport/drug effects , Ferric Compounds/metabolism , Ferritins/genetics , Ferrous Compounds/metabolism , Humans , Kinetics , Nitroprusside/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , RNA, Messenger/metabolism , Receptors, Transferrin/genetics , Tumor Cells, Cultured
12.
Eur J Biochem ; 259(1-2): 304-9, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9914507

ABSTRACT

Iron regulatory proteins (IRP)-1 and 2 are cytoplasmic mRNA-binding proteins that control intracellular iron homeostasis by regulating the translation of ferritin mRNA and stability of transferrin receptor mRNA in an iron-dependent fashion. Although structurally and functionally similar, the two IRP are different in their mode of regulation, pattern of tissue expression and modulation by multiple factors, such as bioradicals. In the present study RNA bandshift assays demonstrated that IRP-2, but not IRP-1, activity was higher in cultured cells than in tissues. Increased expression of IRP-2 in cell lines was not related to immortalization and differentiation but seemed associated to cell proliferation, although not closely dependent on cell growth rate. As a growing cell consumes more iron than its quiescent counterpart, we assessed the iron status of cell lines and found that ferritin content was lower than in tissues. Analysis of IRP activity in cell lines supplemented with heme or non-heme iron and in livers of iron-loaded and iron-deficient rats indicated that IRP-2 responds more promptly than IRP-1 to modulations of iron content. We propose that enhanced IRP-2 activity in cultured cells could be due to a proliferation-dependent, relative iron deficiency that is sensed first by IRP-2.


Subject(s)
Iron-Binding Proteins , Iron-Sulfur Proteins/metabolism , Iron/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Differentiation , Cell Division , Cell Line , Ferritins/biosynthesis , Iron Regulatory Protein 1 , Iron Regulatory Protein 2 , Iron-Regulatory Proteins , Male , Rats , Receptors, Cell Surface/metabolism , Tissue Distribution
13.
Life Sci ; 63(14): PL213-9, 1998.
Article in English | MEDLINE | ID: mdl-9771919

ABSTRACT

Iron Regulatory Proteins (IRPs), by modulating expression of ferritin, which stores excess iron in a non toxic form, and transferrin receptor, which controls iron uptake, are the main controller of cellular iron metabolism. During inflammation, modification of IRP activity may affect iron availability, free radical generation and cytokine gene response in inflammatory cells. In the present study we tested the effect of inflammatory stimuli on IRP function in a human monocytic-macrophagic cell line and the possibility of interfering with these pathways by using an antiinflammatory compound, diacerhein (DAR). IRP activity was enhanced by interferon gamma/lipopolysaccarhide (IFN/LPS), and this effect was consistently counteracted by increasing concentrations of DAR. No direct effect of DAR on IRP activity was found in vitro. However, in vivo, similar IRP activation was achieved by exposing cells to nitric oxide (NO) donors and the LPS/IFN-induced activation of IRP was reversed by NO inhibitors. Interestingly, NO-induced IRP activation was efficiently blocked by DAR. These data show for the first time that a clinically useful antiinflammatory compound, DAR, interferes with IRP activation by NO in inflammed human cells.


Subject(s)
Anthraquinones/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Inflammation/metabolism , Iron-Sulfur Proteins/antagonists & inhibitors , Monocytes/drug effects , RNA-Binding Proteins/antagonists & inhibitors , Cell Line , Humans , Iron-Regulatory Proteins , Nitric Oxide/physiology
14.
Ann N Y Acad Sci ; 851: 179-86, 1998 Jun 30.
Article in English | MEDLINE | ID: mdl-9668619

ABSTRACT

Iron may be important in catalyzing excessive production of reactive oxygen species (ROS). Cellular iron homeostasis is regulated by iron regulatory proteins (IRPs), which bind to iron-responsive elements (IRE) of mRNAs for ferritin and transferrin receptor (TfR) modulating iron uptake and sequestration, respectively. Although iron is the main regulator of IRP activity, IRP is also influenced by other factors, including the redox state. Therefore, IRP might be sensitive to pathophysiological alterations of redox state caused by ROS. However, previous studies have produced diverging evidence on the effect of oxidative injury on IRP. Results obtained in an animal model close to a pathophysiological condition, such as ischemia reperfusion of the liver as well as in a cell-free system involving an enzymatic source of O2 and H2O2, indicate that IRP is downregulated by oxidative stress. In fact, IRP activity is inhibited at early times of post-ischemic reperfusion. Moreover, the concerted action of O2 and H2O2 produced by xanthine oxidase in a cell-free system caused a remarkable inhibition of IRP activity. IRP seems a direct target of ROS; in fact, in vivo inhibition can be prevented by the antioxidant N-acetylcysteine and by interleukin-1 receptor antagonist. In addition, modulation of iron levels of the cell-free assay did not affect the downregulation imposed by xanthine oxidase. Conceivably, downregulation of IRP activity by O2 and H2O2 may facilitate iron sequestration into ferritin, thus limiting the pro-oxidant challenge of iron.


Subject(s)
Iron-Sulfur Proteins/metabolism , Iron/metabolism , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins/metabolism , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Animals , Down-Regulation/physiology , Ferritins/metabolism , Iron-Regulatory Proteins , Ischemia/physiopathology , Liver/physiopathology , Oxidation-Reduction , Oxidative Stress/physiology , RNA-Binding Proteins/physiology , Rats , Wnt2 Protein , Xanthine/metabolism , Xanthine Oxidase/metabolism
15.
Hepatology ; 28(1): 173-8, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9657110

ABSTRACT

Transferrin receptor (TfR) and ferritin, key proteins of cellular iron metabolism, are coordinately and divergently controlled by cytoplasmic proteins (iron regulatory proteins, IRP-1 and IRP-2) that bind to conserved mRNA motifs called iron-responsive elements (IRE). IRP, in response to specific stimuli (low iron levels, growth and stress signals) are activated and prevent TfR mRNA degradation and ferritin mRNA translation by hindering ferritin mRNA binding to polysomes. We previously found that, in regenerating liver, IRP activation was accompanied by increased TfR mRNA levels, but not by reduced ferritin expression. The basis for this unexpected behavior was investigated in the present study. Liver regeneration triggered by carbon tetrachloride (CCl4) stimulated by four- to fivefold the synthesis of both L and H ferritin chains. This increase was accompanied with a transcriptionally regulated twofold rise in the amount of ferritin mRNAs. Moreover, polysome-associated ferritin transcripts were fourfold higher in CCl4-treated animals than in control animals. Because RNA bandshift assays showed a fourfold increase in IRP-2 binding activity after CCl4 administration, activated IRP in regenerating liver seemed unable to prevent ferritin mRNAs binding to polysomes. This was confirmed by direct demonstration in the wheat germ translation system that the efficiency of IRP as a translational repressor of a mRNA bearing an IRE motif in front of a reporter transcript is impaired in CCl4-treated rats in spite of an enhanced IRE-binding capacity. In conclusion, we show for the first time that the paradigm of coordinate and opposite control of ferritin and TfR by IRP is contradicted in liver regeneration. Under these circumstances, growth-dependent signals may activate ferritin gene transcription and at the same time hamper the ability of activated IRP-2 to repress translation of ferritin mRNAs, thus preserving for growing liver cells an essential iron-storage compartment.


Subject(s)
Iron-Binding Proteins , Liver Regeneration/physiology , Liver/metabolism , Receptors, Cell Surface/metabolism , Receptors, Transferrin/metabolism , Animals , Carbon Tetrachloride/pharmacology , Ferritins/biosynthesis , Ferritins/genetics , Iron Regulatory Protein 1 , Iron Regulatory Protein 2 , Iron-Regulatory Proteins , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Liver/drug effects , Male , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rats , Rats, Wistar
16.
FASEB J ; 12(7): 541-52, 1998 May.
Article in English | MEDLINE | ID: mdl-9576481

ABSTRACT

Anticancer therapy with doxorubicin (DOX) is limited by severe cardiotoxicity, presumably reflecting the intramyocardial formation of drug metabolites that alter cell constituents and functions. In a previous study, we showed that NADPH-supplemented cytosolic fractions from human myocardial samples can enzymatically reduce a carbonyl group in the side chain of DOX, yielding a secondary alcohol metabolite called doxorubicinol (DOXol). Here we demonstrate that DOXol delocalizes low molecular weight Fe(II) from the [4Fe-4S] cluster of cytoplasmic aconitase. Iron delocalization proceeds through the reoxidation of DOXol to DOX and liberates DOX-Fe(II) complexes as ultimate by-products. Under physiologic conditions, cluster disassembly abolishes aconitase activity and forms an apoprotein that binds to mRNAs, coordinately increasing the synthesis of transferrin receptor but decreasing that of ferritin. Aconitase is thus converted into an iron regulatory protein-1 (IRP-1) that causes iron uptake to prevail over sequestration, forming a pool of free iron that is used for metabolic functions. Conversely, cluster reassembly converts IRP-1 back to aconitase, providing a regulatory mechanism to decrease free iron when it exceeds metabolic requirements. In contrast to these physiologic mechanisms, DOXol-dependent iron release and cluster disassembly not only abolish aconitase activity, but also affect irreversibly the ability of the apoprotein to function as IRP-1 or to reincorporate iron within new Fe-S motifs. This damage is mediated by DOX-Fe(II) complexes and reflects oxidative modifications of -SH residues having the dual role to coordinate cluster assembly and facilitate interactions of IRP-1 with mRNAs. Collectively, these findings describe a novel mechanism of cardiotoxicity, suggesting that intramyocardial formation of DOXol may perturb the homeostatic processes associated with cluster assembly or disassembly and the reversible switch between aconitase and IRP-1. These results may also provide a guideline to design new drugs that mitigate the cardiotoxicity of DOX.


Subject(s)
Aconitate Hydratase/antagonists & inhibitors , Aconitate Hydratase/biosynthesis , Doxorubicin/analogs & derivatives , Iron-Sulfur Proteins/antagonists & inhibitors , Iron-Sulfur Proteins/biosynthesis , Myocardium/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/biosynthesis , Citrates/pharmacology , Cytosol/metabolism , Doxorubicin/pharmacology , Female , Humans , Iron Regulatory Protein 1 , Iron-Regulatory Proteins , Kinetics , Male , Middle Aged , Myocardium/enzymology , Transcription, Genetic
17.
Blood ; 91(7): 2565-72, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9516158

ABSTRACT

In genetic hemochromatosis (GH), iron overload affects mainly parenchymal cells, whereas little iron is found in reticuloendothelial (RE) cells. We previously found that RE cells from GH patients had an inappropriately high activity of iron regulatory protein (IRP), the key regulator of intracellular iron homeostasis. Elevated IRP should reflect a reduction of the iron pool, possibly because of a failure to retain iron. A defect in iron handling by RE cells that results in a lack of feedback regulation of intestinal absorption might be the basic abnormality in GH. To further investigate the capacity of iron retention in RE cells of GH patients, we used inflammation as a model system as it is characterized by a block of iron release from macrophages. We analyzed the iron status of RE cells by assaying IRP activity and ferritin content after 4, 8, and 24 hours of incubation with lipopolysaccharide (LPS) and interferon-gamma (IFN-gamma). RNA-bandshift assays showed that in monocytes and macrophages from 16 control subjects, IRP activity was transiently elevated 4 hours after treatment with LPS and IFN-gamma but remarkably downregulated thereafter. Treatment with NO donors produced the same effects whereas an inducible Nitric Oxide Synthase (iNOS) inhibitor prevented them, which suggests that the NO pathway was involved. Decreased IRP activity was also found in monocytes from eight patients with inflammation. Interestingly, no late decrease of IRP activity was detected in cytokine-treated RE cells from 12 GH patients. Ferritin content was increased 24 hours after treatment in monocytes from normal subjects but not in monocytes from GH patients. The lack of downregulation of IRP activity under inflammatory conditions seems to confirm that the control of iron release from RE cells is defective in GH.


Subject(s)
Hemochromatosis/blood , Inflammation/metabolism , Iron-Sulfur Proteins/metabolism , Iron/metabolism , Monocytes/metabolism , Mononuclear Phagocyte System/metabolism , RNA-Binding Proteins/metabolism , Adult , Female , Hemochromatosis/immunology , Hemochromatosis/pathology , Humans , Iron-Regulatory Proteins , Macrophages/metabolism , Male , Middle Aged
18.
Blood ; 91(3): 1059-66, 1998 Feb 01.
Article in English | MEDLINE | ID: mdl-9446669

ABSTRACT

Cytokine-treated macrophages represent a useful model to unravel the molecular basis of reticuloendothelial (RE) iron retention in inflammatory conditions. In the present study, we showed that stimulation of murine macrophage J774 cells with interferon (IFN)-gamma/lipopolysaccharide (LPS) resulted in a nitric oxide-dependent modulation of the activity of iron regulatory proteins (IRP)-1 and 2, cytoplasmic proteins which, binding to RNA motifs called iron responsive elements (IRE), control ferritin translation. Stimulation with cytokines caused a small increase of IRP-1 activity and a strong reduction of IRP-2 activity accompanied by increased ferritin synthesis and accumulation. Cytokines induced only a minor increase of H chain ferritin mRNA, thus indicating that IRP-2-mediated posttranscriptional regulation plays a major role in the control of ferritin expression. This was confirmed by direct demonstration that the translational repression function of IRP was impaired in stimulated cells. In fact, translation in cell-free extracts of a reporter transcript under the control of an IRE sequence was repressed less efficiently by IRP-containing lysates from cytokine-treated cells than by lysates from control cells. Our findings throw light on the role of IRP-2 showing that: (1) this protein responds to a stimulus in opposite fashion to IRP-1; (2) when abundantly expressed, as in J774 cells, IRP-2 is sufficient to regulate intracellular iron metabolism in living cells; and (3) by allowing increased ferritin synthesis, IRP-2 may play a role in the regulation of iron homeostasis in RE cells during inflammation.


Subject(s)
Cytokines/pharmacology , Ferritins/biosynthesis , Iron-Sulfur Proteins/metabolism , Macrophages/metabolism , Nitric Oxide/pharmacology , RNA-Binding Proteins/metabolism , Animals , Cell Line , Deferoxamine/pharmacology , Enzyme Inhibitors/pharmacology , Ferritins/genetics , Interferon-gamma/pharmacology , Iron/pharmacology , Iron Chelating Agents/pharmacology , Iron Regulatory Protein 1 , Iron Regulatory Protein 2 , Iron-Regulatory Proteins , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mice , Nitric Oxide Synthase/antagonists & inhibitors , Protein Biosynthesis , RNA/metabolism , RNA, Messenger/metabolism , omega-N-Methylarginine/pharmacology
19.
Gastroenterology ; 113(3): 946-53, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9287988

ABSTRACT

BACKGROUND & AIMS: Iron may catalyze the production of reactive oxygen species (ROS) during postischemic reoxygenation. Ferritin, a cellular iron storage protein, can either represent a source of iron or perform a cytoprotective action against ROS. The aim of this study was to address the role of ferritin in postischemic reperfusion. METHODS: Transcriptional and posttranscriptional mechanisms controlling ferritin gene expression were studied in reperfused rat livers. RESULTS: Proteolysis reduced ferritin levels 2 hours after reperfusion, but a concomitant increase of synthesis, accompanied by enhanced transcription and accumulation of H and L ferritin subunit messenger RNAs (mRNAs), almost re-established normal ferritin content at 4 hours. Pretreatment with interleukin 1 receptor antagonist (IL-1RA) did not prevent the rise of ferritin mRNAs. RNA bandshift assays showed that the activity of the iron regulatory proteins (IRPs), which control ferritin mRNA translation, declined early after reperfusion and recovered progressively thereafter. Pretreatment with either the antioxidant N-acetyl cysteine or IL-1RA was sufficient to prevent almost completely down-regulation of IRP activity. CONCLUSIONS: Postischemic reperfusion causes degradation of ferritin, possibly increasing iron levels. However, induction of ferritin gene transcription, possibly mediated by ferritin-derived iron and ROS-mediated inactivation of IRP, which allows translation of ferritin mRNAs, counteracts this effect and concurs to reestablish the amount of ferritin, which may thus act to limit reperfusion damage.


Subject(s)
Ferritins/biosynthesis , Ischemia/metabolism , Liver Circulation , Liver/metabolism , Reperfusion , Animals , Electrophoresis, Polyacrylamide Gel , Ferritins/genetics , In Vitro Techniques , Iron-Regulatory Proteins , Iron-Sulfur Proteins/metabolism , Male , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Rats , Rats, Wistar , Receptors, Interleukin-1/antagonists & inhibitors
20.
Blood ; 90(2): 814-21, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9226182

ABSTRACT

Recent reports have described families in whom a combination of elevated serum ferritin not related to iron overload and congenital nuclear cataract is transmitted as an autosomal dominant trait. We have studied the molecular pathogenesis of hyperferritinemia in two families showing different phenotypic expression of this new genetic disorder. Serum ferritin levels ranged from 950 to 1,890 microg/L in affected individuals from family 1, and from 366 to 635 microg/L in those from family 2. Cataract was clinically manifested in family 1 and asymptomatic in family 2. By using monoclonal antibodies specific for the H and L ferritin subunits, serum ferritin was found to be essentially L type in both normal and affected individuals. The latter also showed normal amounts of H-type ferritin in circulating mononuclear cells; on the contrary, L-type ferritin contents were 13 times normal in family 1 and five times normal in family 2 on average. Serum ferritin was glycosylated in both normal and affected individuals. There was a close relationship between mononuclear cell L-type ferritin content and serum ferritin concentration (r = 0.95, P < .00001), suggesting that the excess production of ferritin in cells was directly responsible for the hyperferritinemia. The dysregulated L-subunit synthesis was found to result from different point mutations in a noncoding sequence of genomic L-subunit DNA, which behaves as an mRNA cis-acting element known as iron regulatory element (IRE). Affected individuals from family 1 were heterozygous for a point mutation (a single G to A change) in the highly conserved, three-nucleotide motif forming the IRE bulge. Affected members from family 2 were heterozygous for a double point mutation in the IRE lower stem. Using a gel retardation assay, the observed molecular lesions were shown to variably reduce the IRE affinity for an iron regulatory protein (IRP), which inhibits ferritin mRNA translation. The direct relationship between the degree of hyperferritinemia and severity of cataract suggests that this latter is the consequence of excessive ferritin production within the lens fibers. These findings provide strong evidence that serum ferritin is a byproduct of intracellular ferritin synthesis and that the L-subunit gene on chromosome 19 is the source of glycosylated serum ferritin. From a practical standpoint, this new genetic disorder should be taken into account by clinicians when facing a high serum ferritin in an apparently healthy person.


Subject(s)
Cataract/genetics , Ferritins/blood , Ferritins/genetics , Iron Metabolism Disorders/genetics , Iron/metabolism , Point Mutation , RNA, Messenger/genetics , Adult , Antibodies, Monoclonal , Base Sequence , Binding Sites , Cataract/blood , Child , Exons , Female , Genetic Carrier Screening , Glycosylation , Humans , Iron Metabolism Disorders/blood , Macromolecular Substances , Male , Molecular Sequence Data , Pedigree , Phenotype , Protein Biosynthesis , Reference Values , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...