Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Sci China Life Sci ; 66(8): 1841-1857, 2023 08.
Article in English | MEDLINE | ID: mdl-36929272

ABSTRACT

Iron is important for life, and iron deficiency impairs development, but whether the iron level regulates neural differentiation remains elusive. In this study, with iron-regulatory proteins (IRPs) knockout embryonic stem cells (ESCs) that showed severe iron deficiency, we found that the Pax6- and Sox2-positive neuronal precursor cells and Tuj1 fibers in IRP1-/-IRP2-/- ESCs were significantly decreased after inducing neural differentiation. Consistently, in vivo study showed that the knockdown of IRP1 in IRP2-/- fetal mice remarkably affected the differentiation of neuronal precursors and the migration of neurons. These findings suggest that low intracellular iron status significantly inhibits neurodifferentiation. When supplementing IRP1-/-IRP2-/- ESCs with iron, these ESCs could differentiate normally. Further investigations revealed that the underlying mechanism was associated with an increase in reactive oxygen species (ROS) production caused by the substantially low level of iron and the down-regulation of iron-sulfur cluster protein ISCU, which, in turn, affected the proliferation and differentiation of stem cells. Thus, the appropriate amount of iron is crucial for maintaining normal neural differentiation that is termed ferrodifferentiation.


Subject(s)
Iron Deficiencies , Iron-Sulfur Proteins , Reactive Oxygen Species , Animals , Mice , Iron/metabolism , Iron Regulatory Protein 1/metabolism , Iron Regulatory Protein 2/metabolism , Iron-Sulfur Proteins/metabolism , Reactive Oxygen Species/metabolism
2.
Int J Neurosci ; 133(1): 67-76, 2023 Jan.
Article in English | MEDLINE | ID: mdl-33535005

ABSTRACT

We propose that neural damage in Parkinson's disease (PD) is due to dysregulation of iron utilization rather than to high iron levels per se. Iron deposits are associated with neuronal cell death in substantia nigra (SN) resulting in PD where high levels of iron in SNs are due to dysregulation of iron utilization. Cytosolic aconitase (ACO1) upon losing an iron-sulfur cluster becomes iron regulatory protein 1 (IRP1). Rotenone increases levels of IRP1 and induces PD in rats. An increase in iron leads to inactivation of IRP1. We propose a novel treatment strategy to prevent PD. Specifically in rats given rotenone by subcutaneous injections, iron, from iron carbonyl from which iron is slowly absorbed, given three times a day by gavage will keep iron levels constant in the gut whereby iron levels and iron utilization systematically can be tightly regulated. Rotenone adversely affects complex 1 iron-sulfur proteins. Iron supplementation will increase iron-sulfur cluster formation switching IRP1 to ACO1. With IRP1 levels kept constantly low, iron utilization will systematically be tightly regulated stopping dysregulation of complex 1 and the neural damage done by rotenone preventing PD.


Subject(s)
Iron Regulatory Protein 1 , Parkinson Disease , Rats , Animals , Iron Regulatory Protein 1/metabolism , Parkinson Disease/etiology , Parkinson Disease/prevention & control , Rotenone , Aconitate Hydratase/metabolism , Iron/metabolism , Sulfur/metabolism
3.
Anim Sci J ; 90(10): 1377-1387, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31436009

ABSTRACT

The purpose of this study was to determine whether the enteric coating process affects growth performance, Fe bioavailability, and gene expression levels that maintain iron balance in the body. The test was divided into the control group, ferrous sulfate group, ferrous fumarate group, ferrous glycine chelate(1:1) (Fe-Gly(1:1)) group, ferrous glycine chelate(2:1) (Fe-Gly(2:1)) group, enteric-coated Fe-Gly(1:1) group, and enteric-coated Fe-Gly(2:1) group. The results showed that the growth performance of the rats in each iron supplement group was no significant difference among them. The results of serum biochemical indicators showed that the antioxidant capacity of the rats in the iron supplement group after enteric coating increased. The iron supplementation effect of Fe-Gly(1:1) and Fe-Gly(2:1) was better than that of ferrous sulfate, and the effect of Fe-Gly(1:1) after enteric coating was enhanced. The expression levels of IRP1 and IRP2 in the genes of enteric-coated Fe-Gly(1:1) and enteric-coated Fe-Gly(2:1) were significantly higher than those of ferrous sulfate. The expression levels of IRP1 and IRP2 in the protein of enteric-coated Fe-Gly(1:1) group were significantly higher than those in the Fe-Gly(1:1) group. The above results show that Fe-Gly can improve the bioavailability and antioxidant capacity of iron and reduce the iron output of feces after enteric coating.


Subject(s)
Ferrous Compounds/pharmacology , Iron/metabolism , Animals , Antioxidants , Biological Availability , Dietary Supplements/analysis , Ferrous Compounds/metabolism , Homeostasis/genetics , Homeostasis/physiology , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 2/genetics , Rats , Rats, Sprague-Dawley , Tablets, Enteric-Coated/analysis
4.
J Clin Invest ; 128(4): 1317-1325, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29480820

ABSTRACT

Chuvash polycythemia is an inherited disease caused by a homozygous germline VHLR200W mutation, which leads to impaired degradation of HIF2α, elevated levels of serum erythropoietin, and erythrocytosis/polycythemia. This phenotype is recapitulated by a mouse model bearing a homozygous VhlR200W mutation. We previously showed that iron-regulatory protein 1-knockout (Irp1-knockout) mice developed erythrocytosis/polycythemia through translational derepression of Hif2α, suggesting that IRP1 could be a therapeutic target to treat Chuvash polycythemia. Here, we fed VhlR200W mice supplemented with Tempol, a small, stable nitroxide molecule and observed that Tempol decreased erythropoietin production, corrected splenomegaly, normalized hematocrit levels, and increased the lifespans of these mice. We attribute the reversal of erythrocytosis/polycythemia to translational repression of Hif2α expression by Tempol-mediated increases in the IRE-binding activity of Irp1, as reversal of polycythemia was abrogated in VhlR200W mice in which Irp1 was genetically ablated. Thus, a new approach to the treatment of patients with Chuvash polycythemia may include dietary supplementation of Tempol, which decreased Hif2α expression and markedly reduced life-threatening erythrocytosis/polycythemia in the VhlR200W mice.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cyclic N-Oxides/pharmacology , Gene Expression Regulation/drug effects , Iron Regulatory Protein 1/metabolism , Polycythemia/drug therapy , Protein Biosynthesis/drug effects , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Humans , Iron Regulatory Protein 1/genetics , Mice , Mice, Mutant Strains , Polycythemia/genetics , Polycythemia/metabolism , Polycythemia/pathology , Spin Labels
5.
J Biol Chem ; 292(38): 15976-15989, 2017 09 22.
Article in English | MEDLINE | ID: mdl-28768766

ABSTRACT

Iron-regulatory protein 1 (IRP1) belongs to a family of RNA-binding proteins that modulate metazoan iron metabolism. Multiple mechanisms are employed to control the action of IRP1 in dictating changes in the uptake and metabolic fate of iron. Inactivation of IRP1 RNA binding by iron primarily involves insertion of a [4Fe-4S] cluster by the cytosolic iron-sulfur cluster assembly (CIA) system, converting it into cytosolic aconitase (c-acon), but can also involve iron-mediated degradation of IRP1 by the E3 ligase FBXL5 that also targets IRP2. How CIA and FBXL5 collaborate to maintain cellular iron homeostasis through IRP1 and other pathways is poorly understood. Because impaired Fe-S cluster biogenesis associates with human disease, we determined the importance of FBXL5 for regulating IRP1 when CIA is impaired. Suppression of FBXL5 expression coupled with induction of an IRP1 mutant (IRP13C>3S) that cannot insert the Fe-S cluster, or along with knockdown of the CIA factors NUBP2 or FAM96A, reduced cell viability. Iron supplementation reversed this growth defect and was associated with FBXL5-dependent polyubiquitination of IRP1. Phosphorylation of IRP1 at Ser-138 increased when CIA was inhibited and was required for iron rescue. Impaired CIA activity, as noted by reduced c-acon activity, was associated with enhanced FBXL5 expression and a concomitant reduction in IRP1 and IRP2 protein level and RNA-binding activity. Conversely, expression of either IRP induced FBXL5 protein level, demonstrating a negative feedback loop limiting excessive accumulation of iron-response element RNA-binding activity, whose disruption reduces cell growth. We conclude that a regulatory circuit involving FBXL5 and CIA acts through both IRPs to control iron metabolism and promote optimal cell growth.


Subject(s)
F-Box Proteins/metabolism , Iron Regulatory Protein 1/metabolism , Iron/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , F-Box Proteins/genetics , Ferritins/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , Iron Regulatory Protein 1/chemistry , Iron Regulatory Protein 2/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RNA/metabolism , Serine/metabolism , Sulfur/metabolism , Ubiquitin-Protein Ligase Complexes/deficiency , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitination
6.
Int J Hematol ; 104(4): 491-501, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27380194

ABSTRACT

Iron overload remains a concern in myelodysplastic syndrome (MDS) patients. Iron chelation therapy (ICT) thus plays an integral role in the management of these patients. Moreover, ICT has been shown to prolong leukemia-free survival in MDS patients; however, the mechanisms responsible for this effect are unclear. Iron is a key molecule for regulating cytosolic aconitase 1 (ACO1). Additionally, the mutation of isocitrate dehydrogenase (IDH), the enzyme downstream of ACO1 in the TCA cycle, is associated with epigenetic abnormalities secondary to 2-hydroxyglutarate (2-HG) and DNA methylation. However, epigenetic abnormalities observed in many MDS patients occur without IDH mutation. We hypothesized that iron itself activates the ACO1-IDH pathway, which may increase 2-HG and DNA methylation, and eventually contribute to leukemogenesis without IDH mutation. Using whole RNA sequencing of bone marrow cells in iron-overloaded mice, we observed that the enzymes, phosphoglucomutase 1, glycogen debranching enzyme, and isocitrate dehydrogenase 1 (Idh1), which are involved in glycogen and glucose metabolism, were increased. Digital PCR further showed that Idh1 and Aco1, enzymes involved in the TCA cycle, were also elevated. Additionally, enzymatic activities of TCA cycle and methylated DNA were increased. Iron chelation reversed these phenomena. In conclusion, iron activation of glucose metabolism causes an increase of 2-HG and DNA methylation.


Subject(s)
Bone Marrow/metabolism , DNA Methylation/drug effects , Iron Regulatory Protein 1/metabolism , Iron/pharmacology , Isocitrate Dehydrogenase/metabolism , Animals , Carcinogenesis/chemically induced , Glucose/metabolism , Glutarates/blood , Iron Regulatory Protein 1/drug effects , Isocitrate Dehydrogenase/drug effects , Mice
7.
Exp Parasitol ; 157: 1-11, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26115940

ABSTRACT

During its parasitic life stages, the marine ectoparasitic copepod Lepeophtheirus salmonis ingests large amounts of host blood, which contains high amounts of iron. Iron is an essential micronutrient, but also toxic in high dosages, and blood-feeding parasites like the salmon louse must thus possess an efficient system to handle the excess iron. Iron regulatory protein 1 and 2 (IRP1 and IRP2) are known to play crucial roles in this process, by regulating several proteins involved in iron transport and storage, depending on the cellular iron concentration. To gain knowledge about the regulation of the iron metabolism in salmon lice, two IRP homologues (LsIRP1A and LsIRP1B) were identified by sequence and predicted structure similarity to known IRPs in other species. In situ hybridisation revealed that LsIRP1A and LsIRP1B mRNAs were expressed in the ovaries, oviducts and vitellogenic oocytes of adult females. Transcription levels of LsIRP1A and LsIRP1B mRNAs did not differ significantly between the different developmental stages of the salmon louse. Adults in the absence of blood as a feed source had decreased levels of LsIRP1A, but not LsIRP1B mRNA. RNA binding experiments indicated the presence of functioning IRP in salmon lice. In order to explore the biological functions of LsIRP1A and LsIRP1B, the mRNAs of both proteins were knocked down by RNA interference (RNAi) in preadult females. The knockdown was confirmed by qRT-PCR. LsIRP1B knockdown lice produced less offspring than control lice due to slightly shorter egg strings and had decreased levels of transcripts involved in egg development. Knockdown of both LsIRP1A and LsIRP1B caused increased expression of a salmon louse Ferritin (LsFer). These results confirm that salmon lice have two IRP1 homologues, LsIRP1A and LsIRP1B, and might suggest a function in cellular iron regulation in the reproductive organs and eggs.


Subject(s)
Copepoda/chemistry , Ectoparasitic Infestations/veterinary , Fish Diseases/parasitology , Iron Regulatory Protein 1/physiology , Salmo salar/parasitology , Amino Acid Sequence , Animals , Copepoda/classification , Copepoda/metabolism , Ectoparasitic Infestations/parasitology , Female , Gene Expression Regulation , Humans , In Situ Hybridization , Iron/metabolism , Iron Regulatory Protein 1/chemistry , Iron Regulatory Protein 1/genetics , Male , Molecular Sequence Data , Phylogeny , RNA Interference , RNA, Messenger/analysis , RNA, Messenger/metabolism , Sequence Alignment
8.
PLoS One ; 10(2): e0116855, 2015.
Article in English | MEDLINE | ID: mdl-25647178

ABSTRACT

Increased serum ferritin associated with mild hepatic iron accumulation, despite preserved upregulation of the iron hormone hepcidin, is frequently observed in patients with dysmetabolic overload syndrome (DIOS). Genetic factors and Western diet represent predisposing conditions, but the mechanisms favoring iron accumulation in DIOS are still unclear. Aims of this study were to assess the effect a high-fat diet (HFD) on hepatic iron metabolism in an experimental model in rats, to further characterize the effect of free fatty acids on iron metabolism in HepG2 hepatocytes in vitro, and to assess the translational relevance in patients with fatty liver with and without iron accumulation. Despite decreased uptake of dietary iron, rats fed HFD accumulated more hepatic iron than those fed regular diet, which was associated with steatosis development. Hepatic iron accumulation was paralleled by induction of ferritin, in the presence of preserved upregulation of hepcidin, recapitulating the features of DIOS. HFD was associated with increased expression of the major iron uptake protein Transferrin receptor-1 (TfR-1), consistently with upregulation of the intracellular iron sensor Iron regulated protein-1 (IRP1). Supplementation with fatty acids induced TfR-1 and IRP1 in HepG2 hepatocytes, favoring intracellular iron accumulation following exposure to iron salts. IRP1 silencing completely abrogated TfR-1 induction and the facilitation of intracellular iron accumulation induced by fatty acids. Hepatic TfR-1 mRNA levels were upregulated in patients with fatty liver and DIOS, whereas they were not associated with liver fat nor with inflammation. In conclusion, increased exposure to fatty acids subverts hepatic iron metabolism, favoring the induction of an iron uptake program despite hepatocellular iron accumulation.


Subject(s)
Diet, High-Fat/adverse effects , Iron Overload/etiology , Iron Overload/metabolism , Iron/metabolism , Liver/drug effects , Liver/metabolism , Animals , Biological Transport/drug effects , Fatty Acids/metabolism , Fatty Acids/pharmacology , Female , Gene Silencing , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Iron Regulatory Protein 1/deficiency , Iron Regulatory Protein 1/genetics , Liver/pathology , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Up-Regulation/drug effects
9.
Metallomics ; 7(5): 766-75, 2015 May.
Article in English | MEDLINE | ID: mdl-25652229

ABSTRACT

The kidney plays an important role in iron homeostasis and actively reabsorbs citrate. The bifunctional iron-regulatory protein IRP-1 potentially regulates iron trafficking and participates in citrate metabolism as a cytosolic (c-) aconitase. We investigated the role of cellular iron status in determining the expression and dynamics of IRP-1 in two renal cell types, with the aim of identifying a role of the protein in cellular ROS levels, citrate metabolism and glutamate production. The effects of iron supplementation and chelation on IRP-1 protein and mRNA levels and protein turnover were compared in cultured primary rat mesangial cells and a porcine renal tubule cell line (LLC-PK1). Levels of ROS were measured in both cell types, and c-aconitase activity, glutamate, and glutathione were measured in LLC-PK1 cells, with and without IRP-1 silencing and in glutamine-supplemented or nominally glutamine-free medium. Iron supplementation decreased IRP-1 levels (e.g., approx. 40% in mesangial cells treated with 10 µg ml(-1) iron for 16 h) and increased ubiquitinated IRP-1 levels in both cells types, with iron chelation having the opposite effect. Although iron increased ROS levels (three-fold with 20 µg ml(-1) iron in mesangial cells and more modestly by about 30% with 50 µg ml(-1) in LLC-PK1 cells, both after 24 h), protein degradation was not ROS-dependent. In LLC-PK1 cells, 10 µg ml(-1) iron (24 h) increased both aconitase activity (30%) and secreted glutamate levels (65%). Silencing did not remove the glutamate response to iron but decreased the c-aconitase activity of the residual protein independent of iron loading (37% and 46% of control levels, without and with iron treatment, respectively). However, in glutamine-free medium, glutamate was still increased by iron, even in IRP-1-silenced cells, and did not correspond to c-aconitase. Silencing decreased the amount of ferritin measured in response to iron loading, decreased the affect of iron on total glutathione by 48%, and increased the response of ROS to iron loading by 38%. We conclude that iron increases turnover of IRP-1 in kidney cells, while increasing aconitase activity, suggesting that the apoprotein (aconitase-inactive) form is not exclusively responsible for turnover. Iron increases glutamate levels in tubule epithelial cells, but this appears to be independent of c-aconitase activity or the availability of extracellular glutamine. IRP-1 protein levels are not regulated by ROS, but IRP-1-dependent ferritin expression may decrease ROS and increase total glutathione levels, suggesting that ferritin levels are more important than citrate metabolism in protecting renal cells against iron.


Subject(s)
Iron Regulatory Protein 1/metabolism , Iron/metabolism , Kidney/metabolism , Animals , Cell Line , Cells, Cultured , Gene Silencing , Glutamic Acid/metabolism , Glutathione/metabolism , Iron Regulatory Protein 1/genetics , Kidney/cytology , LLC-PK1 Cells , Proteolysis , Rats , Reactive Oxygen Species/metabolism , Swine
10.
Cell Mol Life Sci ; 70(23): 4449-61, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23817740

ABSTRACT

Several dietary factors and their genetic modifiers play a role in neurological disease and affect the human brain. The structural and functional integrity of the living brain can be assessed using neuroimaging, enabling large-scale epidemiological studies to identify factors that help or harm the brain. Iron is one nutritional factor that comes entirely from our diet, and its storage and transport in the body are under strong genetic control. In this review, we discuss how neuroimaging can help to identify associations between brain integrity, genetic variations, and dietary factors such as iron. We also review iron's essential role in cognition, and we note some challenges and confounds involved in interpreting links between diet and brain health. Finally, we outline some recent discoveries regarding the genetics of iron and its effects on the brain, suggesting the promise of neuroimaging in revealing how dietary factors affect the brain.


Subject(s)
Brain/metabolism , Iron/metabolism , Neuroimaging/methods , Nutrigenomics , Nutritional Status , Dietary Supplements , Humans , Iron/administration & dosage , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 1/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Transferrin/genetics , Transferrin/metabolism
11.
Cell Microbiol ; 11(1): 83-94, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18823384

ABSTRACT

Intracellular pathogens employ several strategies for iron acquisition from host macrophages for survival and growth, whereas macrophage resists infection by actively sequestering iron. Here, we show that instead of allowing macrophage to sequester iron, protozoan parasite Leishmania donovani (LD) uses a novel strategy to manipulate iron uptake mechanisms of the host and utilizes the taken up iron for its intracellular growth. To do so, intracellular LD directly scavenges iron from labile iron pool of macrophages. Depleted labile iron pool activates iron sensors iron-regulatory proteins IRP1 and IRP2. IRPs then bind to iron-responsive elements present in the 3' UTR of iron uptake gene transferrin receptor 1 by a post-transcriptional mRNA stability mechanism. Increased iron-responsive element-IRP interaction and transferrin receptor 1 expressions in spleen-derived macrophages from LD-infected mice confirm that LD employs similar mechanism to acquire iron during infection into mammalian hosts. Increased intracellular LD growth by holo-transferrin supplementation and inhibited growth by iron chelator treatment confirm the significance of this modulated iron uptake pathway of host in favour of the parasite.


Subject(s)
Iron/metabolism , Leishmania donovani/growth & development , Leishmania donovani/metabolism , Macrophages/parasitology , 3' Untranslated Regions , Animals , Female , Iron Regulatory Protein 1/metabolism , Iron Regulatory Protein 2/metabolism , Mice , Mice, Inbred BALB C , Protein Binding , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism
12.
Proc Natl Acad Sci U S A ; 105(33): 12028-33, 2008 Aug 19.
Article in English | MEDLINE | ID: mdl-18685102

ABSTRACT

In mammals, two homologous cytosolic regulatory proteins, iron regulatory protein 1 (also known as IRP1 and Aco1) and iron regulatory protein 2 (also known as IRP2 and Ireb2), sense cytosolic iron levels and posttranscriptionally regulate iron metabolism genes, including transferrin receptor 1 (TfR1) and ferritin H and L subunits, by binding to iron-responsive elements (IREs) within target transcripts. Mice that lack IRP2 develop microcytic anemia and neurodegeneration associated with functional cellular iron depletion caused by low TfR1 and high ferritin expression. IRP1 knockout (IRP1(-/-)) animals do not significantly misregulate iron metabolism, partly because IRP1 is an iron-sulfur protein that functions mainly as a cytosolic aconitase in mammalian tissues and IRP2 activity increases to compensate for loss of the IRE binding form of IRP1. The neurodegenerative disease of IRP2(-/-) animals progresses slowly as the animals age. In this study, we fed IRP2(-/-) mice a diet supplemented with a stable nitroxide, Tempol, and showed that the progression of neuromuscular impairment was markedly attenuated. In cell lines derived from IRP2(-/-) animals, and in the cerebellum, brainstem, and forebrain of animals maintained on the Tempol diet, IRP1 was converted from a cytosolic aconitase to an IRE binding protein that stabilized the TfR1 transcript and repressed ferritin synthesis. We suggest that Tempol protected IRP2(-/-) mice by disassembling the cytosolic iron-sulfur cluster of IRP1 and activating IRE binding activity, which stabilized the TfR1 transcript, repressed ferritin synthesis, and partially restored normal cellular iron homeostasis in the brain.


Subject(s)
Iron Regulatory Protein 2/deficiency , Iron Regulatory Protein 2/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/prevention & control , Animals , Cell Line , Cyclic N-Oxides/chemistry , Cyclic N-Oxides/pharmacology , Disease Progression , Enzyme Activation , Humans , Iron Regulatory Protein 1/metabolism , Iron Regulatory Protein 2/genetics , Mice , Mice, Knockout , Molecular Structure , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Protein Binding , Receptors, Transferrin/metabolism , Spin Labels
13.
J Biol Chem ; 283(8): 5168-77, 2008 Feb 22.
Article in English | MEDLINE | ID: mdl-18073202

ABSTRACT

One consequence of zinc deficiency is an elevation in cell and tissue iron concentrations. To examine the mechanism(s) underlying this phenomenon, Swiss 3T3 cells were cultured in zinc-deficient (D, 0.5 microM zinc), zinc-supplemented (S, 50 microM zinc), or control (C, 4 microM zinc) media. After 24 h of culture, cells in the D group were characterized by a 50% decrease in intracellular zinc and a 35% increase in intracellular iron relative to cells in the S and C groups. The increase in cellular iron was associated with increased transferrin receptor 1 protein and mRNA levels and increased ferritin light chain expression. The divalent metal transporter 1(+)iron-responsive element isoform mRNA was decreased during zinc deficiency-induced iron accumulation. Examination of zinc-deficient cells revealed increased binding of iron regulatory protein 2 (IRP2) and decreased binding of IRP1 to a consensus iron-responsive element. The increased IRP2-binding activity in zinc-deficient cells coincided with an increased level of IRP2 protein. The accumulation of IRP2 protein was independent of zinc deficiency-induced intracellular nitric oxide production but was attenuated by the addition of the antioxidant N-acetylcysteine or ascorbate to the D medium. These data support the concept that zinc deficiency can result in alterations in iron transporter, storage, and regulatory proteins, which facilitate iron accumulation.


Subject(s)
Gene Expression Regulation/physiology , Iron/metabolism , Zinc/deficiency , 3T3 Cells , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Apoferritins/biosynthesis , Ascorbic Acid/pharmacology , Gene Expression Regulation/drug effects , Iron Regulatory Protein 1/biosynthesis , Iron Regulatory Protein 2/biosynthesis , Mice , Nitric Oxide/metabolism , RNA, Messenger/biosynthesis , Receptors, Transferrin/biosynthesis , Response Elements/physiology
14.
Free Radic Biol Med ; 40(7): 1152-60, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16545682

ABSTRACT

Curcumin is among the more successful chemopreventive compounds investigated in recent years, and is currently in human trials to prevent cancer. The mechanism of action of curcumin is complex and likely multifactorial. We have made the unexpected observation that curcumin strikingly modulates proteins of iron metabolism in cells and in tissues, suggesting that curcumin has properties of an iron chelator. Curcumin increased mRNA levels of ferritin and GSTalpha in cultured liver cells. Unexpectedly, however, although levels of GSTalpha protein increased in parallel with mRNA levels in response to curcumin, levels of ferritin protein declined. Since iron chelators repress ferritin translation, we considered that curcumin may act as an iron chelator. To test this hypothesis, we measured the effect of curcumin on transferrin receptor 1, a protein stabilized under conditions of iron limitation, as well as the ability of curcumin to activate iron regulatory proteins (IRPs). Both transferrin receptor 1 and activated IRP, indicators of iron depletion, increased in response to curcumin. Consistent with the hypothesis that curcumin acts as an iron chelator, mice that were fed diets supplemented with curcumin exhibited a decline in levels of ferritin protein in the liver. These results suggest that iron chelation may be an additional mode of action of curcumin.


Subject(s)
Curcumin/pharmacology , Iron Chelating Agents/pharmacology , Animals , Cells, Cultured , Diet , Female , Ferritins/biosynthesis , Glutathione Transferase/biosynthesis , Iron/pharmacology , Iron Regulatory Protein 1/biosynthesis , Liver/metabolism , Mice , Receptors, Transferrin/metabolism
15.
Biochemistry ; 44(23): 8470-8, 2005 Jun 14.
Article in English | MEDLINE | ID: mdl-15938636

ABSTRACT

Iron regulatory proteins (IRPs) regulate iron metabolism in mammalian cells. We used biophysical techniques to examine the solution properties of apo-IRP1 and apo-IRP2 and the interaction with their RNA ligand, the iron regulatory element (IRE). Sedimentation velocity and equilibrium experiments have shown that apo-IRP1 exists as an equilibrium mixture of monomers and dimers in solution, with an equilibrium dissociation constant in the low micromolar range and slow kinetic exchange between the two forms. However, only monomeric IRP1 is observed in complex with IRE. In contrast, IRP2 exists as monomer in both the apo-IRP2 form and in the IRP2/IRE complex. For both IRPs, sedimentation velocity and dynamic light-scattering experiments show a decrease of the Stokes radius upon binding of IRE. This conformational change was also observed by circular dichroism. Studies with an RNA molecule complementary to IRE indicate that, although specific base interactions can increase the stability of the protein/RNA complex, they are not essential for inducing this conformational change. The dynamic change of the IRP between different oligomeric and conformational states induced by interaction with IRE may play a role in the iron regulatory functions of IRPs.


Subject(s)
Iron Regulatory Protein 1/chemistry , Iron Regulatory Protein 1/metabolism , Iron Regulatory Protein 2/chemistry , Iron Regulatory Protein 2/metabolism , Picolines/chemistry , Picolines/metabolism , Response Elements , Apoproteins/chemistry , Apoproteins/metabolism , Centrifugation, Density Gradient , Circular Dichroism , Dimerization , Humans , Ligands , Light , Pichia/genetics , Protein Binding , Protein Conformation , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Scattering, Radiation , Solutions
16.
J Biol Chem ; 280(20): 20010-20, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15753094

ABSTRACT

Insulin modulates glucose homeostasis, but the role of insulin-responsive transcription factors in such actions is not well understood. Recently, we have identified the insulin-response element-binding protein-1 (IRE-BP1) as a transcription factor that appears to mediate insulin action on multiple target genes. To examine the possibility that IRE-BP1 is an insulin-responsive glucoregulatory factor involved in the metabolic actions of insulin, we investigated the effect of adenoviral overexpression of hepatic IRE-BP1 on the glycemic control of insulin-resistant diabetic rats. Adenoviral IRE-BP1 lowered both fasting and postprandial glucose levels, and microarray of hepatic RNA revealed modulation of the expression of genes involved in gluconeogenesis, lipogenesis, and fatty acid oxidation. The insulin mimetic effects of IRE-BP1 were also confirmed in L6 myocytes; stable constitutive expressions of IRE-BP1 enhanced glucose transporter expression, glucose uptake, and glycogen accumulation in these cells. These findings showed physiologic sufficiency of IRE-BP1 as the transcriptional mediator of the metabolic action of insulin. Understanding IRE-BP1 action should constitute a useful probe into the mechanisms of metabolic regulation and an important target to develop therapeutic agents that mimic or enhance insulin action.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Iron Regulatory Protein 1/metabolism , Animals , Base Sequence , Blood Glucose/metabolism , Cell Line , DNA, Complementary/genetics , Diabetes Mellitus, Experimental/genetics , Gene Expression Profiling , Glucose/metabolism , Glycogen/metabolism , Hyperglycemia/genetics , Hyperglycemia/metabolism , Insulin Resistance/genetics , Iron Regulatory Protein 1/genetics , Liver/metabolism , Male , Models, Biological , Monosaccharide Transport Proteins/metabolism , Rats , Rats, Sprague-Dawley , Rats, Zucker , Transfection
17.
J Biol Chem ; 278(48): 47365-9, 2003 Nov 28.
Article in English | MEDLINE | ID: mdl-12972424

ABSTRACT

Iron and oxygen are essential but potentially toxic constituents of most organisms, and their transport is meticulously regulated both at the cellular and systemic levels. Compartmentalization may be a homeostatic mechanism for isolating these biological reactants in cells. To investigate this hypothesis, we have undertaken a genetic analysis of the interaction between iron and oxygen metabolism in Drosophila. We show that Drosophila iron regulatory protein-1 (IRP1) registers cytosolic iron and oxidative stress through its labile iron sulfur cluster by switching between cytosolic aconitase and RNA-binding functions. IRP1 is strongly activated by silencing and genetic mutation of the cytosolic superoxide dismutase (Sod1), but is unaffected by silencing of mitochondrial Sod2. Conversely, mitochondrial aconitase activity is relatively insensitive to loss of Sod1 function, but drops dramatically if Sod2 activity is impaired. This strongly suggests that the mitochondrial boundary limits the range of superoxide reactivity in vivo. We also find that exposure of adults to paraquat converts cytosolic aconitase to IRP1 but has no affect on mitochondrial aconitase, indicating that paraquat generates superoxide in the cytosol but not in mitochondria. Accordingly, we find that transgene-mediated overexpression of Sod2 neither enhances paraquat resistance in Sod1+ flies nor compensates for lack of SOD1 activity in Sod1-null mutants. We conclude that in vivo, superoxide is confined to the subcellular compartment in which it is formed, and that the mitochondrial and cytosolic SODs provide independent protection to compartment-specific protein iron-sulfur clusters against attack by superoxide generated under oxidative stress within those compartments.


Subject(s)
Iron-Sulfur Proteins/chemistry , Superoxide Dismutase/chemistry , Aconitate Hydratase/chemistry , Animals , Cell Line , Cytosol/enzymology , Cytosol/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Drosophila , Herbicides , Iron/metabolism , Iron Regulatory Protein 1/metabolism , Mitochondria/enzymology , Mutation , Oxidative Stress , Oxygen/metabolism , Paraquat/pharmacology , Protein Binding , RNA/metabolism , RNA Interference , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Superoxides , Time Factors , Transgenes
18.
J Inorg Biochem ; 91(1): 9-18, 2002 Jul 25.
Article in English | MEDLINE | ID: mdl-12121757

ABSTRACT

Iron is an essential metal for almost all living organisms due to its involvement in a large number of iron-containing enzymes and proteins, yet it is also toxic. The mechanisms involved in iron absorption across the intestinal tract, its transport in serum and delivery to cells and iron storage within cells is briefly reviewed. Current views on cellular iron homeostasis involving the iron regulatory proteins IRP1 and IRP2 and their interactions with the iron regulatory elements, affecting either mRNA translation (ferritin and erythroid cell delta-aminolaevulinate synthase) or mRNA stability (transferrin receptor) are discussed. The potential of Fe(II) to catalyse hydroxyl radical formation via the Fenton reaction means that iron is potentially toxic. The toxicity of iron in specific tissues and cell types (liver, macrophages and brain) is illustrated by studies with appropriate cellular and animal models. In liver, the high levels of cyoprotective enzymes and antioxidants, means that to observe toxic effects substantial levels of iron loading are required. In reticuloendothelial cells, such as macrophages, relatively small increases in cellular iron (2-3-fold) can affect cellular signalling, as measured by NO production and activation of the nuclear transcription factor NF kappa B, as well as cellular function, as measured by the capacity of the cells to produce reactive oxygen species when stimulated. The situation in brain, where anti-oxidative defences are relatively low, is highly regionally specific, where iron accumulation in specific brain regions is associated with a number of neurodegenerative diseases. In the brains of animals treated with either trimethylhexanoylferrocene or aluminium gluconate, iron and aluminium accumulate, respectively. With the latter compound, iron also increases, which may reflect an effect of aluminium on the IRP2 protein. Chelation therapy can reduce brain aluminium levels significantly, while iron can also be removed, but with greater difficulty. The prospects for chelation therapy in the treatment and possible prevention of neurodegenerative diseases is reviewed.


Subject(s)
Homeostasis , Iron Overload/metabolism , Iron/metabolism , Iron/toxicity , Animals , Biological Transport/physiology , Brain/cytology , Brain/metabolism , Chelating Agents/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Hepatocytes/metabolism , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Iron Regulatory Protein 1 , Iron Regulatory Protein 2 , Iron-Regulatory Proteins , Iron-Sulfur Proteins/metabolism , Macrophages/metabolism , Oxidative Stress , RNA-Binding Proteins/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Transferrin/metabolism
19.
Insect Biochem Mol Biol ; 32(1): 85-96, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11719072

ABSTRACT

Manduca sexta IRP1 was cloned and sequenced. The deduced amino acid sequence of Manduca IRP1 shows high similarity to other IRP1 proteins. The Cys residues required as ligands for the iron sulfur cluster, as well as all residues necessary for aconitase activity are conserved in the insect protein. Purified recombinant Manduca IRP1 binds specifically to transcripts of the iron responsive element (IRE) of Manduca or human ferritin subunit mRNA. Binding activity of the recombinant protein was not influenced by the presence of beta-mercaptoethanol. However, IRP/IRE binding activity of cytoplasmic extracts from fat body was decreased by reducing agents in a dose-responsive manner. Fat body IRP1/IRE binding activity was reduced for Manduca sexta larvae injected with low doses of iron, while IRP1 mRNA and protein levels remained stable. At higher iron doses, binding activity increased and stabilized. Hemolymph ferritin levels showed an inverse relationship to IRP1/IRE binding activity. These data suggest that the Manduca IRP1 is likely involved in translational control of ferritin synthesis in a manner similar to that found in vertebrates. However, factors other than iron can influence IRP/IRE interaction and hemolymph ferritin levels in insects.


Subject(s)
Carrier Proteins/metabolism , Ferritins/genetics , Insect Proteins , Iron Regulatory Protein 1 , Iron/metabolism , Manduca/metabolism , Animals , Base Sequence , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/isolation & purification , DNA, Complementary , Humans , Manduca/genetics , Molecular Sequence Data , RNA/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Response Elements
20.
Blood ; 98(8): 2555-62, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11588054

ABSTRACT

This study cloned and sequenced the complementary DNA (cDNA) encoding of a putative malarial iron responsive element-binding protein (PfIRPa) and confirmed its identity to the previously identified iron-regulatory protein (IRP)-like cDNA from Plasmodium falciparum. Sequence alignment showed that the plasmodial sequence has 47% identity with human IRP1. Hemoglobin-free lysates obtained from erythrocyte-stage P falciparum contain a protein that binds a consensus mammalian iron-responsive element (IRE), indicating that a protein(s) with iron-regulatory activity was present in the lysates. IRE-binding activity was found to be iron regulated in the electrophoretic mobility shift assays. Western blot analysis showed a 2-fold increase in the level of PfIRPa in the desferrioxamine-treated cultures versus control or iron-supplemented cells. Malarial IRP was detected by anti-PfIRPa antibody in the IRE-protein complex from P falciparum lysates. Immunofluorescence studies confirmed the presence of PfIRPa in the infected red blood cells. These findings demonstrate that erythrocyte P falciparum contains an iron-regulated IRP that binds a mammalian consensus IRE sequence, raising the possibility that the malaria parasite expresses transcripts that contain IREs and are iron-dependently regulated.


Subject(s)
Erythrocytes/parasitology , Iron-Sulfur Proteins/metabolism , Plasmodium falciparum/physiology , Protozoan Proteins/metabolism , RNA-Binding Proteins/metabolism , Aconitate Hydratase/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Blotting, Western , Cloning, Molecular , Consensus Sequence , DNA Primers , Deferoxamine/pharmacology , Fluorescent Antibody Technique, Indirect , Hemoglobins/metabolism , Humans , Iron/metabolism , Iron Regulatory Protein 1 , Iron-Regulatory Proteins , Iron-Sulfur Proteins/genetics , Mammals , Molecular Sequence Data , Plasmodium falciparum/drug effects , RNA-Binding Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL