Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Carcinogenesis ; 41(8): 1113-1122, 2020 08 12.
Article in English | MEDLINE | ID: mdl-31734690

ABSTRACT

Precursor T-cell lymphoblastic neoplasms are aggressive malignancies in need for more effective and specific therapeutic treatments. A significant fraction of these neoplasms harbor deletions on the locus 9p21, targeting the tumor suppressor CDKN2A but also deleting the aconitase 1 (ACO1) gene, a neighboring housekeeping gene involved in cytoplasm and mitochondrial metabolism. Here we show that reducing the aconitase activity with fluorocitrate decreases the viability of T-cell lymphoblastic neoplasia cells in correlation to the differential aconitase expression. The consequences of the treatment were evidenced in vitro using T-cell lymphoblastic neoplasia cell lines exhibiting 9p21 deletions and variable levels of ACO1 expression or activity. Similar results were observed in melanoma cell lines, suggesting a true potential for fluorocitrate in different cancer types. Notably, ectopic expression of ACO1 alleviated the susceptibility of cell lines to fluorocitrate and, conversely, knockdown experiments increased susceptibility of resistant cell lines. These findings were confirmed in vivo on athymic nude mice by using tumor xenografts derived from two T-cell lines with different levels of ACO1. Taken together, our results indicate that the non-targeted ACO1 deficiency induced by common deletions exerts a collateral cellular lethality that can be used as a novel therapeutic strategy in the treatment of several types of cancer.


Subject(s)
Chromosomes, Human, Pair 9/genetics , Citrates/pharmacology , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/pharmacology , Gene Deletion , Iron Regulatory Protein 1/deficiency , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Cell Line, Tumor , Cell Survival/drug effects , Citrates/therapeutic use , Cyclin-Dependent Kinase Inhibitor p16/genetics , Enzyme Inhibitors/therapeutic use , Female , Heterografts , Humans , Iron Regulatory Protein 1/antagonists & inhibitors , Iron Regulatory Protein 1/genetics , Melanoma/genetics , Mice , Mice, Nude , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Skin Neoplasms/genetics
2.
Sci Rep ; 8(1): 5118, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29572489

ABSTRACT

Iron is essential for growth and proliferation of mammalian cells. The maintenance of cellular iron homeostasis is regulated by iron regulatory proteins (IRPs) through binding to the cognate iron-responsive elements in target mRNAs and thereby regulating the expression of target genes. Irp1 or Irp2-null mutation is known to reduce the cellular iron level by decreasing transferrin receptor 1 and increasing ferritin. Here, we report that Irp1 or Irp2-null mutation also causes downregulation of frataxin and IscU, two of the core components in the iron-sulfur cluster biogenesis machinery. Interestingly, while the activities of some of iron-sulfur cluster-containing enzymes including mitochondrial aconitase and cytosolic xanthine oxidase were not affected by the mutations, the activities of respiratory chain complexes were drastically diminished resulting in mitochondrial dysfunction. Overexpression of human ISCU and frataxin in Irp1 or Irp2-null cells was able to rescue the defects in iron-sulfur cluster biogenesis and mitochondrial quality. Our results strongly suggest that iron regulatory proteins regulate the part of iron sulfur cluster biogenesis tailored specifically for mitochondrial electron transport chain complexes.


Subject(s)
Electron Transport Chain Complex Proteins/metabolism , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Iron Regulatory Protein 1/deficiency , Iron Regulatory Protein 2/deficiency , Iron-Binding Proteins/biosynthesis , Animals , Embryo, Mammalian/pathology , Ferritins/metabolism , Fibroblasts/pathology , Humans , Mice , Mice, Knockout , Mitochondria/pathology , Mutation , Frataxin
3.
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
4.
Cell Metab ; 21(2): 311-323, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25651183

ABSTRACT

Mitochondrial iron accumulation is a hallmark of diseases associated with impaired iron-sulfur cluster (Fe-S) biogenesis, such as Friedreich ataxia linked to frataxin (FXN) deficiency. The pathophysiological relevance of the mitochondrial iron loading and the underlying mechanisms are unknown. Using a mouse model of hepatic FXN deficiency in combination with mice deficient for iron regulatory protein 1 (IRP1), a key regulator of cellular iron metabolism, we show that IRP1 activation in conditions of Fe-S deficiency increases the available cytosolic labile iron pool. Surprisingly, our data indicate that IRP1 activation sustains mitochondrial iron supply and function rather than driving detrimental iron overload. Mitochondrial iron accumulation is shown to depend on mitochondrial dysfunction and heme-dependent upregulation of the mitochondrial iron importer mitoferrin-2. Our results uncover an unexpected protective role of IRP1 in pathological conditions associated with altered Fe-S metabolism.


Subject(s)
Iron Regulatory Protein 1/metabolism , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Iron/metabolism , Mitochondria/metabolism , Animals , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Friedreich Ataxia/pathology , Iron Regulatory Protein 1/deficiency , Iron Regulatory Protein 1/genetics , Iron-Sulfur Proteins/deficiency , Iron-Sulfur Proteins/metabolism , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Frataxin
5.
Cell Metab ; 17(2): 271-81, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23395173

ABSTRACT

Iron regulatory proteins (Irps) 1 and 2 posttranscriptionally control the expression of transcripts that contain iron-responsive element (IRE) sequences, including ferritin, ferroportin, transferrin receptor, and hypoxia-inducible factor 2α (HIF2α). We report here that mice with targeted deletion of Irp1 developed pulmonary hypertension and polycythemia that was exacerbated by a low-iron diet. Hematocrits increased to 65% in iron-starved mice, and many polycythemic mice died of abdominal hemorrhages. Irp1 deletion enhanced HIF2α protein expression in kidneys of Irp1(-/-) mice, which led to increased erythropoietin (EPO) expression, polycythemia, and concomitant tissue iron deficiency. Increased HIF2α expression in pulmonary endothelial cells induced high expression of endothelin-1, likely contributing to the pulmonary hypertension of Irp1(-/-) mice. Our results reveal why anemia is an early physiological consequence of iron deficiency, highlight the physiological significance of Irp1 in regulating erythropoiesis and iron distribution, and provide important insights into the molecular pathogenesis of pulmonary hypertension.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Deletion , Hypertension, Pulmonary/complications , Iron Regulatory Protein 1/metabolism , Polycythemia/complications , Protein Biosynthesis , Animals , Diet , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelin-1/genetics , Endothelin-1/metabolism , Erythropoietin/blood , Gastrointestinal Hemorrhage/blood , Gastrointestinal Hemorrhage/complications , Gastrointestinal Hemorrhage/pathology , Hematopoiesis, Extramedullary/drug effects , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/pathology , Iron/pharmacology , Iron Regulatory Protein 1/deficiency , Iron Regulatory Protein 2/metabolism , Longevity , Mice , Models, Biological , Nerve Degeneration/blood , Nerve Degeneration/complications , Nerve Degeneration/pathology , Organ Specificity/drug effects , Polycythemia/blood , Polycythemia/pathology , Protein Biosynthesis/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
6.
Cell Metab ; 17(2): 282-90, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23395174

ABSTRACT

Red blood cell production is a finely tuned process that requires coordinated oxygen- and iron-dependent regulation of cell differentiation and iron metabolism. Here, we show that translational regulation of hypoxia-inducible factor 2α (HIF-2α) synthesis by iron regulatory protein 1 (IRP1) is critical for controlling erythrocyte number. IRP1-null (Irp1(-/-)) mice display a marked transient polycythemia. HIF-2α messenger RNA (mRNA) is derepressed in kidneys of Irp1(-/-) mice but not in kidneys of Irp2(-/-) mice, leading to increased renal erythropoietin (Epo) mRNA and inappropriately elevated serum Epo levels. Expression of the iron transport genes DCytb, Dmt1, and ferroportin, as well as other HIF-2α targets, is enhanced in Irp1(-/-) duodenum. Analysis of mRNA translation state in the liver revealed IRP1-dependent dysregulation of HIF-2α mRNA translation, whereas IRP2 deficiency derepressed translation of all other known 5' iron response element (IRE)-containing mRNAs expressed in the liver. These results uncover separable physiological roles of each IRP and identify IRP1 as a therapeutic target for manipulating HIF-2α action in hematologic, oncologic, and other disorders.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Erythropoiesis , Iron Regulatory Protein 1/metabolism , Iron/metabolism , Oxygen/metabolism , Signal Transduction , Absorption , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Duodenum/metabolism , Duodenum/pathology , Erythroid Cells/metabolism , Erythroid Precursor Cells/metabolism , Erythropoietin/blood , Gene Expression Regulation , Hematopoiesis, Extramedullary , Iron Regulatory Protein 1/deficiency , Mice , Polycythemia/blood , Polycythemia/pathology , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spleen/metabolism
7.
PLoS One ; 6(10): e25404, 2011.
Article in English | MEDLINE | ID: mdl-22003390

ABSTRACT

Genetic ablation of Iron Regulatory Protein 2 (Irp2, Ireb2), which post-transcriptionally regulates iron metabolism genes, causes a gait disorder in mice that progresses to hind-limb paralysis. Here we have demonstrated that misregulation of iron metabolism from loss of Irp2 causes lower motor neuronal degeneration with significant spinal cord axonopathy. Mitochondria in the lumbar spinal cord showed significantly decreased Complex I and II activities, and abnormal morphology. Lower motor neurons appeared to be the most adversely affected neurons, and we show that functional iron starvation due to misregulation of iron import and storage proteins, including transferrin receptor 1 and ferritin, may have a causal role in disease. We demonstrated that two therapeutic approaches were beneficial for motor neuron survival. First, we activated a homologous protein, IRP1, by oral Tempol treatment and found that axons were partially spared from degeneration. Secondly, we genetically decreased expression of the iron storage protein, ferritin, to diminish functional iron starvation. These data suggest that functional iron deficiency may constitute a previously unrecognized molecular basis for degeneration of motor neurons in mice.


Subject(s)
Gene Deletion , Iron Deficiencies , Iron Regulatory Protein 2/deficiency , Iron Regulatory Protein 2/genetics , Mitochondria/metabolism , Motor Neurons/metabolism , Motor Neurons/pathology , Animals , Apoferritins/biosynthesis , Atrophy/metabolism , Axons/drug effects , Axons/metabolism , Axons/pathology , Biomarkers/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Cyclic N-Oxides/pharmacology , Homeostasis/drug effects , Homeostasis/genetics , Iron/metabolism , Iron Regulatory Protein 1/deficiency , Iron Regulatory Protein 1/metabolism , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/pathology , Motor Neurons/drug effects , Oxidative Stress/drug effects , Oxidative Stress/genetics , Spin Labels , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology
8.
Cell Metab ; 12(2): 194-201, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20674864

ABSTRACT

Mitochondria supply cells with ATP, heme, and iron sulfur clusters (ISC), and mitochondrial energy metabolism involves both heme- and ISC-dependent enzymes. Here, we show that mitochondrial iron supply and function require iron regulatory proteins (IRP), cytosolic RNA-binding proteins that control mRNA translation and stability. Mice lacking both IRP1 and IRP2 in their hepatocytes suffer from mitochondrial iron deficiency and dysfunction associated with alterations of the ISC and heme biosynthetic pathways, leading to liver failure and death. These results uncover a major role of the IRPs in cell biology: to ensure adequate iron supply to the mitochondrion for proper function of this critical organelle.


Subject(s)
Iron-Regulatory Proteins/metabolism , Iron/metabolism , Mitochondria/metabolism , Animals , Energy Metabolism , Heme/biosynthesis , Iron Regulatory Protein 1/deficiency , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 1/metabolism , Iron Regulatory Protein 2/deficiency , Iron Regulatory Protein 2/genetics , Iron Regulatory Protein 2/metabolism , Iron-Regulatory Proteins/deficiency , Iron-Regulatory Proteins/genetics , Liver Failure/etiology , Liver Failure/metabolism , Liver Failure/pathology , Mice , Mice, Transgenic
9.
Brain Res ; 1337: 95-103, 2010 Jun 14.
Article in English | MEDLINE | ID: mdl-20399759

ABSTRACT

Iron is deposited in perihematomal tissue after an intracerebral hemorrhage (ICH), and may contribute to oxidative injury. Cell culture studies have demonstrated that enhancing ferritin expression by targeting iron regulatory protein (IRP) binding activity reduces cellular vulnerability to iron and hemoglobin. In order to assess the therapeutic potential of this approach after striatal ICH, the effect of IRP1 or IRP2 gene knockout on ferritin expression and injury was quantified. Striatal ferritin in IRP1 knockout mice was similar to that in wild-type controls 3 days after stereotactic injection of artificial CSF or autologous blood. Corresponding levels in IRP2 knockouts were increased by 11-fold and 8.4-fold, respectively, compared with wild-type. Protein carbonylation, a sensitive marker of hemoglobin neurotoxicity, was increased by 2.4-fold in blood-injected wild-type striata, was not altered by IRP1 knockout, but was reduced by approximately 60% by IRP2 knockout. Perihematomal cell viability in wild-type mice, assessed by MTT assay, was approximately half of that in contralateral striata at 3 days, and was significantly increased in IRP2 knockouts but not in IRP1 knockouts. Protection was also observed when hemorrhage was induced by collagenase injection. These results suggest that IRP2 binding activity reduces ferritin expression in the striatum after ICH, preventing an optimal response to elevated local iron concentrations. IRP2 binding activity may be a novel therapeutic target after hemorrhagic CNS injuries.


Subject(s)
Cerebral Hemorrhage , Ferritins/metabolism , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 2/genetics , Iron/metabolism , Animals , Cell Survival , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Collagenases/metabolism , Disease Models, Animal , Female , Hemoglobins/metabolism , Iron Regulatory Protein 1/deficiency , Iron Regulatory Protein 2/deficiency , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Oxidative Stress/genetics , Protein Carbonylation
10.
J Mol Med (Berl) ; 84(7): 551-60, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16770644

ABSTRACT

Iron aggravates the cardiotoxicity of doxorubicin (DOX), a widely used anticancer anthracycline. The amount of iron in the cell is regulated by the iron regulatory proteins (IRPs)-1 and -2 that control the posttranscriptional expression of key iron metabolism genes. In vitro and cell culture studies revealed the ability of DOX to modulate the activity of both IRPs. However, conflicting data were obtained from different cell types and experimental conditions. To investigate the connection between acute DOX cardiotoxicity and the IRPs in a mammalian organism, we analyzed IRP activity and the expression of IRP target genes in the heart of mice subjected to DOX treatment. DOX exposure elicits a differential modulation of the two IRPs with reduced IRP2 activity and unchanged IRP1 activity. IRP2 downmodulation is associated with the upregulation of the ferritin L and H genes and decreased expression of the transferrin receptor 1 (TfR1). To directly test the role of IRP1 in DOX cardiotoxicity, the DOX response was analyzed in mice lacking IRP1. DOX-mediated IRP2 downmodulation and regulation of ferritin and TfR1 expression is identical in Irp1 (-/-) mice compared to wild type, as is the degree of oxidative damage of the heart assessed by thioredoxin and thiobarbituric acid reactive substance levels and by brain natriuretic peptide mRNA expression. These data demonstrate that the alterations of cardiac iron homeostasis related to acute anthracycline cardiotoxicity occur independently of IRP1. The observed IRP2 downmodulation could serve as a means to counteract DOX cardiotoxicity by reducing the "free" cellular iron pool.


Subject(s)
Doxorubicin/pharmacology , Heart/drug effects , Iron/metabolism , Myocardium/metabolism , Animals , Ferritins/genetics , Ferritins/metabolism , Gene Deletion , Gene Expression Regulation , Iron Regulatory Protein 1/deficiency , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 1/metabolism , Mice , Mice, Knockout
11.
Blood Cells Mol Dis ; 36(2): 283-7, 2006.
Article in English | MEDLINE | ID: mdl-16480904

ABSTRACT

Iron regulatory proteins 1 and 2 (IRPs) are homologous mammalian cytosolic proteins that sense intracellular iron levels and post-transcriptionally regulate expression of ferritin, transferrin receptor, and other iron metabolism proteins. Adult mice with homozygous targeted deletion of IRP2 develop microcytic anemia, elevated red cell protoporphyrin IX levels, high serum ferritin, and adult-onset neurodegeneration. Mice with homozygous deletion of IRP1 develop no overt abnormalities, but mice that lack both copies of IRP2 and one copy of IRP1 develop a more severe anemia and neurodegeneration than mice with deletion of IRP2 alone. Here, we have demonstrated that IRP1-/- IRP2-/- embryos do not survive gestation, and that although IRP1-/- IRP2-/blastocysts can be genotyped and harvested, implanted embryos with the IRP1-/- IRP2-/genotype are undetectable at embryonic day 6.5 and beyond. Blastocysts derived from a cross in which 25% of the fertilized embryos were expected to have the IRP1-/- IRP2-/genotype often showed brown discoloration and abnormal morphology. These abnormal blastocysts likely have the IRP1-/- IRP2-/- genotype, and the brown discoloration may be attributable to ferritin overexpression and sequestration of ferric iron in ferritin, whereas abnormal morphology may be due to concomitant functional iron deficiency. These results demonstrate that IRPs are indispensable for regulation of mammalian iron homeostasis at the post-implantation stage of murine embryonic development.


Subject(s)
Embryonic Development/genetics , Iron Regulatory Protein 1/deficiency , Iron Regulatory Protein 2/deficiency , Animals , Blastocyst , Embryo, Mammalian , Ferritins/metabolism , Genotype , Homeostasis , Iron/metabolism , Mice , Survival Rate , Zygote
SELECTION OF CITATIONS
SEARCH DETAIL
...