Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38732071

ABSTRACT

Iron regulatory proteins (IRP1 and IRP2) are the master regulators of mammalian iron homeostasis. They bind to the iron-responsive elements (IREs) of the transcripts of iron-related genes to regulate their expression, thereby maintaining cellular iron availability. The primary method to measure the IRE-binding activity of IRPs is the electrophoresis mobility shift assay (EMSA). This method is particularly useful for evaluating IRP1 activity, since IRP1 is a bifunctional enzyme and its protein levels remain similar during conversion between the IRE-binding protein and cytosolic aconitase forms. Here, we exploited a method of using a biotinylated-IRE probe to separate IRE-binding IRPs followed by immunoblotting to analyze the IRE-binding activity. This method allows for the successful measurement of IRP activity in cultured cells and mouse tissues under various iron conditions. By separating IRE-binding IRPs from the rest of the lysates, this method increases the specificity of IRP antibodies and verifies whether a band represents an IRP, thereby revealing some previously unrecognized information about IRPs. With this method, we showed that the S711-phosphorylated IRP1 was found only in the IRE-binding form in PMA-treated Hep3B cells. Second, we found a truncated IRE-binding IRP2 isoform that is generated by proteolytic cleavage on sites in the 73aa insert region of the IRP2 protein. Third, we found that higher levels of SDS, compared to 1-2% SDS in regular loading buffer, could dramatically increase the band intensity of IRPs in immunoblots, especially in HL-60 cells. Fourth, we found that the addition of SDS or LDS to cell lysates activated protein degradation at 37 °C or room temperature, especially in HL-60 cell lysates. As this method is more practical, sensitive, and cost-effective, we believe that its application will enhance future research on iron regulation and metabolism.


Subject(s)
Iron Regulatory Protein 1 , Iron , Humans , Animals , Iron/metabolism , Iron Regulatory Protein 1/metabolism , Iron Regulatory Protein 1/genetics , Mice , Iron Regulatory Protein 2/metabolism , Iron Regulatory Protein 2/genetics , Biotinylation , Response Elements , Phosphorylation , Iron-Regulatory Proteins/metabolism , Iron-Regulatory Proteins/genetics , Protein Binding , Cell Line, Tumor
2.
Proc Natl Acad Sci U S A ; 120(33): e2303860120, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37552760

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, uses an RNA-dependent RNA polymerase along with several accessory factors to replicate its genome and transcribe its genes. Nonstructural protein (nsp) 13 is a helicase required for viral replication. Here, we found that nsp13 ligates iron, in addition to zinc, when purified anoxically. Using inductively coupled plasma mass spectrometry, UV-visible absorption, EPR, and Mössbauer spectroscopies, we characterized nsp13 as an iron-sulfur (Fe-S) protein that ligates an Fe4S4 cluster in the treble-clef metal-binding site of its zinc-binding domain. The Fe-S cluster in nsp13 modulates both its binding to the template RNA and its unwinding activity. Exposure of the protein to the stable nitroxide TEMPOL oxidizes and degrades the cluster and drastically diminishes unwinding activity. Thus, optimal function of nsp13 depends on a labile Fe-S cluster that is potentially targetable for COVID-19 treatment.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , COVID-19 Drug Treatment , DNA Helicases/metabolism , RNA , Sulfur , Viral Nonstructural Proteins/metabolism , RNA Helicases/genetics
3.
Int J Mol Sci ; 23(11)2022 Jun 04.
Article in English | MEDLINE | ID: mdl-35682986

ABSTRACT

In this study, a series of 4-[(quinolin-4-yl)amino]benzamide derivatives as the novel anti-influenza agents were designed and synthesized. Cytotoxicity assay, cytopathic effect assay and plaque inhibition assay were performed to evaluate the anti-influenza virus A/WSN/33 (H1N1) activity of the target compounds. The target compound G07 demonstrated significant anti-influenza virus A/WSN/33 (H1N1) activity both in cytopathic effect assay (EC50 = 11.38 ± 1.89 µM) and plaque inhibition assay (IC50 = 0.23 ± 0.15 µM). G07 also exhibited significant anti-influenza virus activities against other three different influenza virus strains A/PR/8 (H1N1), A/HK/68 (H3N2) and influenza B virus. According to the result of ribonucleoprotein reconstitution assay, G07 could interact well with ribonucleoprotein with an inhibition rate of 80.65% at 100 µM. Furthermore, G07 exhibited significant activity target PA-PB1 subunit of RNA polymerase according to the PA-PB1 inhibitory activity prediction by the best pharmacophore Hypo1. In addition, G07 was well drug-likeness based on the results of Lipinski's rule and ADMET prediction. All the results proved that 4-[(quinolin-4-yl)amino]benzamide derivatives could generate potential candidates in discovery of anti-influenza virus agents.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Antiviral Agents/pharmacology , Benzamides/pharmacology , DNA Viruses , Molecular Docking Simulation , Ribonucleoproteins , Virus Replication
4.
J Am Chem Soc ; 144(3): 1130-1137, 2022 01 26.
Article in English | MEDLINE | ID: mdl-35029378

ABSTRACT

Herein, we report the first Ni-catalyzed enantioselective deaminative alkylation of amino acid and peptide derivatives with unactivated olefins. Key for success was the discovery of a new sterically encumbered bis(oxazoline) ligand backbone, thus offering a de novo technology for accessing enantioenriched sp3-sp3 linkages via sp3 C-N functionalization. Our protocol is distinguished by its broad scope and generality across a wide number of counterparts, even in the context of late-stage functionalization. In addition, an enantioselective deaminative remote hydroalkylation reaction of unactivated internal olefins is within reach, thus providing a useful entry point for forging enantioenriched sp3-sp3 centers at remote sp3 C-H sites.


Subject(s)
Alkenes
5.
Semin Hematol ; 58(3): 161-174, 2021 07.
Article in English | MEDLINE | ID: mdl-34389108

ABSTRACT

To maintain an adequate iron supply for hemoglobin synthesis and essential metabolic functions while counteracting iron toxicity, humans and other vertebrates have evolved effective mechanisms to conserve and finely regulate iron concentration, storage, and distribution to tissues. At the systemic level, the iron-regulatory hormone hepcidin is secreted by the liver in response to serum iron levels and inflammation. Hepcidin regulates the expression of the sole known mammalian iron exporter, ferroportin, to control dietary absorption, storage and tissue distribution of iron. At the cellular level, iron regulatory proteins 1 and 2 (IRP1 and IRP2) register cytosolic iron concentrations and post-transcriptionally regulate the expression of iron metabolism genes to optimize iron availability for essential cellular processes, including heme biosynthesis and iron-sulfur cluster biogenesis. Genetic malfunctions affecting the iron sensing mechanisms or the main pathways that utilize iron in the cell cause a broad range of human diseases, some of which are characterized by mitochondrial iron accumulation. This review will discuss the mechanisms of systemic and cellular iron sensing with a focus on the main iron utilization pathways in the cell, and on human conditions that arise from compromised function of the regulatory axes that control iron homeostasis.


Subject(s)
Erythropoiesis , Iron , Animals , Homeostasis , Humans , Iron/metabolism , Mammals/metabolism
6.
Blood ; 137(18): 2509-2519, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33512384

ABSTRACT

Polycythemia and pulmonary hypertension are 2 human diseases for which better therapies are needed. Upregulation of hypoxia-inducible factor-2α (HIF-2α) and its target genes, erythropoietin (EPO) and endothelin-1, causes polycythemia and pulmonary hypertension in patients with Chuvash polycythemia who are homozygous for the R200W mutation in the von Hippel Lindau (VHL) gene and in a murine mouse model of Chuvash polycythemia that bears the same homozygous VhlR200W mutation. Moreover, the aged VhlR200W mice developed pulmonary fibrosis, most likely due to the increased expression of Cxcl-12, another Hif-2α target. Patients with mutations in iron regulatory protein 1 (IRP1) also develop polycythemia, and Irp1-knockout (Irp1-KO) mice exhibit polycythemia, pulmonary hypertension, and cardiac fibrosis attributable to translational derepression of Hif-2α, and the resultant high expression of the Hif-2α targets EPO, endothelin-1, and Cxcl-12. In this study, we inactivated Hif-2α with the second-generation allosteric HIF-2α inhibitor MK-6482 in VhlR200W, Irp1-KO, and double-mutant VhlR200W;Irp1-KO mice. MK-6482 treatment decreased EPO production and reversed polycythemia in all 3 mouse models. Drug treatment also decreased right ventricular pressure and mitigated pulmonary hypertension in VhlR200W, Irp1-KO, and VhlR200W;Irp1-KO mice to near normal wild-type levels and normalized the movement of the cardiac interventricular septum in VhlR200Wmice. MK-6482 treatment reduced the increased expression of Cxcl-12, which, in association with CXCR4, mediates fibrocyte influx into the lungs, potentially causing pulmonary fibrosis. Our results suggest that oral intake of MK-6482 could represent a new approach to treatment of patients with polycythemia, pulmonary hypertension, pulmonary fibrosis, and complications caused by elevated expression of HIF-2α.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Gene Expression Regulation/drug effects , Hypertension, Pulmonary/prevention & control , Iron Regulatory Protein 1/physiology , Polycythemia/prevention & control , Sulfones/pharmacology , Von Hippel-Lindau Tumor Suppressor Protein/physiology , Animals , Endothelin-1/antagonists & inhibitors , Endothelin-1/genetics , Endothelin-1/metabolism , Erythropoietin/antagonists & inhibitors , Erythropoietin/genetics , Erythropoietin/metabolism , Female , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polycythemia/etiology , Polycythemia/metabolism , Polycythemia/pathology
7.
PLoS One ; 13(10): e0204471, 2018.
Article in English | MEDLINE | ID: mdl-30321179

ABSTRACT

The exact route of iron through the kidney and its regulation during iron overload are not completely elucidated. Under physiologic conditions, non-transferrin and transferrin bound iron passes the glomerular filter and is reabsorbed through kidney epithelial cells, so that hardly any iron is found in the urine. To study the route of iron reabsorption through the kidney, we analyzed the location and regulation of iron metabolism related proteins in kidneys of mice with iron overload, elicited by iron dextran injections. Transferrin Receptor 1 was decreased as expected, following iron overload. In contrast, the multi-ligand hetero-dimeric receptor-complex megalin/cubilin, which also mediates the internalization of transferrin, was highly up-regulated. Moreover, with increasing iron, intracellular ferritin distribution shifted in renal epithelium from an apical location to a punctate distribution throughout the epithelial cells. In addition, in contrast to many other tissues, the iron exporter ferroportin was not reduced by iron overload in the kidney. Iron accumulated mainly in interstitial macrophages, and more prominently in the medulla than in the cortex. This suggests that despite the reduction of Transferrin Receptor 1, alternative pathways may effectively mediate re-absorption of iron that cycles through the kidney during parenterally induced iron-overload. The most iron consuming process of the body, erythropoiesis, is regulated by the renal erythropoietin producing cells in kidney interstitium. We propose, that the efficient re-absorption of iron by the kidney, also during iron overload enables these cells to sense systemic iron and regulate its usage based on the systemic iron state.


Subject(s)
Biological Transport/physiology , Iron Overload/metabolism , Iron/metabolism , Kidney/metabolism , Animals , Disease Models, Animal , Epithelial Cells/metabolism , Ferritins/metabolism , Intracellular Space/metabolism , Iron Overload/pathology , Iron-Dextran Complex , Kidney/pathology , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , RNA, Messenger/metabolism , Receptors, Cell Surface/metabolism , Receptors, Transferrin/metabolism , Spleen/metabolism , Spleen/pathology
8.
Blood Adv ; 2(20): 2732-2743, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30337301

ABSTRACT

Heme oxygenase 1 (HMOX1), the inducible enzyme that catabolizes the degradation of heme into biliverdin, iron, and carbon monoxide, plays an essential role in the clearance of senescent and damaged red blood cells, systemic iron homeostasis, erythropoiesis, vascular hemostasis, and oxidative and inflammatory stress responses. In humans, HMOX1 deficiency causes a rare and lethal disease, characterized by severe anemia, intravascular hemolysis, as well as vascular and tissue damage. Hmox1 knockout (KO) mice recapitulated the phenotypes of HMOX1-deficiency patients and could be rescued by bone marrow (BM) transplantation that engrafted donor's hematopoietic stem cells into the recipient animals after myeloablation. To find better therapy and elucidate the contribution of macrophages to the pathogenesis of HMOX1-deficiency disease, we infused wild-type (WT) macrophages into Hmox1 KO mice. Results showed that WT macrophages engrafted and proliferated in the livers of Hmox1 KO mice, which corrected the microcytic anemia, rescued the intravascular hemolysis, restored iron homeostasis, eliminated kidney iron overload and tissue damage, and provided long-term protection. These results showed that a single macrophage infusion delivered a long-term curative effect in Hmox1 KO mice, obviating the need for BM transplantation, and suggested that the HMOX1 disease stems mainly from the loss of viable reticuloendothelial macrophages. Our work provides new insights into the etiology of HMOX1 deficiency and demonstrates the potential of infusion of WT macrophages to prevent disease in patients with HMOX1 deficiency and potentially other macrophage-related diseases.


Subject(s)
Anemia, Hemolytic/complications , Anemia/genetics , Growth Disorders/complications , Heme Oxygenase-1/deficiency , Hemolysis/genetics , Iron Metabolism Disorders/complications , Liver/physiopathology , Macrophages/metabolism , Animals , Humans , Mice
9.
Blood ; 132(19): 2078-2087, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30213870

ABSTRACT

Ferroportin (FPN), the only known vertebrate iron exporter, transports iron from intestinal, splenic, and hepatic cells into the blood to provide iron to other tissues and cells in vivo. Most of the circulating iron is consumed by erythroid cells to synthesize hemoglobin. Here we found that erythroid cells not only consumed large amounts of iron, but also returned significant amounts of iron to the blood. Erythroblast-specific Fpn knockout (Fpn KO) mice developed lower serum iron levels in conjunction with tissue iron overload and increased FPN expression in spleen and liver without changing hepcidin levels. Our results also showed that Fpn KO mice, which suffer from mild hemolytic anemia, were sensitive to phenylhydrazine-induced oxidative stress but were able to tolerate iron deficiency upon exposure to a low-iron diet and phlebotomy, supporting that the anemia of Fpn KO mice resulted from erythrocytic iron overload and resulting oxidative injury rather than a red blood cell (RBC) production defect. Moreover, we found that the mean corpuscular volume (MCV) values of gain-of-function FPN mutation patients were positively associated with serum transferrin saturations, whereas MCVs of loss-of-function FPN mutation patients were not, supporting that erythroblasts donate iron to blood through FPN in response to serum iron levels. Our results indicate that FPN of erythroid cells plays an unexpectedly essential role in maintaining systemic iron homeostasis and protecting RBCs from oxidative stress, providing insight into the pathophysiology of FPN diseases.


Subject(s)
Anemia, Iron-Deficiency/genetics , Cation Transport Proteins/genetics , Erythroid Cells/pathology , Hemolysis , Iron/blood , Loss of Function Mutation , Oxidative Stress , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/pathology , Animals , Cation Transport Proteins/analysis , Erythroid Cells/metabolism , Gain of Function Mutation , Humans , Iron/analysis , Iron Deficiencies , Iron Overload/blood , Iron Overload/genetics , Iron Overload/pathology , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Spleen/metabolism , Spleen/pathology
10.
Blood Adv ; 2(10): 1146-1156, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29784770

ABSTRACT

Given the essential roles of iron-sulfur (Fe-S) cofactors in mediating electron transfer in the mitochondrial respiratory chain and supporting heme biosynthesis, mitochondrial dysfunction is a common feature in a growing list of human Fe-S cluster biogenesis disorders, including Friedreich ataxia and GLRX5-related sideroblastic anemia. Here, our studies showed that restriction of Fe-S cluster biogenesis not only compromised mitochondrial oxidative metabolism but also resulted in decreased overall histone acetylation and increased H3K9me3 levels in the nucleus and increased acetylation of α-tubulin in the cytosol by decreasing the lipoylation of the pyruvate dehydrogenase complex, decreasing levels of succinate dehydrogenase and the histone acetyltransferase ELP3, and increasing levels of the tubulin acetyltransferase MEC17. Previous studies have shown that the metabolic shift in Toll-like receptor (TLR)-activated myeloid cells involves rapid activation of glycolysis and subsequent mitochondrial respiratory failure due to nitric oxide (NO)-mediated damage to Fe-S proteins. Our studies indicated that TLR activation also actively suppresses many components of the Fe-S cluster biogenesis machinery, which exacerbates NO-mediated damage to Fe-S proteins by interfering with cluster recovery. These results reveal new regulatory pathways and novel roles of the Fe-S cluster biogenesis machinery in modifying the epigenome and acetylome and provide new insights into the etiology of Fe-S cluster biogenesis disorders.


Subject(s)
Histones/metabolism , Iron-Sulfur Proteins/metabolism , Toll-Like Receptors/metabolism , Tubulin/metabolism , Acetylation , Humans
11.
Science ; 359(6383): 1520-1523, 2018 03 30.
Article in English | MEDLINE | ID: mdl-29599243

ABSTRACT

Malaria parasites invade red blood cells (RBCs), consume copious amounts of hemoglobin, and severely disrupt iron regulation in humans. Anemia often accompanies malaria disease; however, iron supplementation therapy inexplicably exacerbates malarial infections. Here we found that the iron exporter ferroportin (FPN) was highly abundant in RBCs, and iron supplementation suppressed its activity. Conditional deletion of the Fpn gene in erythroid cells resulted in accumulation of excess intracellular iron, cellular damage, hemolysis, and increased fatality in malaria-infected mice. In humans, a prevalent FPN mutation, Q248H (glutamine to histidine at position 248), prevented hepcidin-induced degradation of FPN and protected against severe malaria disease. FPN Q248H appears to have been positively selected in African populations in response to the impact of malaria disease. Thus, FPN protects RBCs against oxidative stress and malaria infection.


Subject(s)
Cation Transport Proteins/metabolism , Erythrocytes/metabolism , Hemolysis , Iron/metabolism , Malaria/epidemiology , Amino Acid Substitution , Anemia/metabolism , Animals , Black People/genetics , Cation Transport Proteins/genetics , Child , Erythrocytes/drug effects , Female , Hepcidins/metabolism , Hepcidins/pharmacology , Humans , Iron/administration & dosage , Iron/pharmacology , Malaria/blood , Malaria/genetics , Male , Mice , Mice, Knockout , Mutation , Oxidative Stress , Risk , Selection, Genetic , Sequence Deletion , Zambia/epidemiology
12.
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
13.
Blood ; 131(8): 840-842, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29472371
14.
Neurobiol Dis ; 81: 66-75, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25771171

ABSTRACT

Iron regulatory proteins 1 and 2 (IRP1 and IRP2) are two cytosolic proteins that maintain cellular iron homeostasis by binding to RNA stem loops known as iron responsive elements (IREs) that are found in the untranslated regions of target mRNAs that encode proteins involved in iron metabolism. IRPs modify the expression of iron metabolism genes, and global and tissue-specific knockout mice have been made to evaluate the physiological significance of these iron regulatory proteins (Irps). Here, we will discuss the results of the studies that have been performed with mice engineered to lack the expression of one or both Irps and made in different strains using different methodologies. Both Irp1 and Irp2 knockout mice are viable, but the double knockout (Irp1(-/-)Irp2(-/-)) mice die before birth, indicating that these Irps play a crucial role in maintaining iron homeostasis. Irp1(-/-) mice develop polycythemia and pulmonary hypertension, and when these mice are challenged with a low iron diet, they die early of abdominal hemorrhages, suggesting that Irp1 plays an essential role in erythropoiesis and in the pulmonary and cardiovascular systems. Irp2(-/-) mice develop microcytic anemia, erythropoietic protoporphyria and a progressive neurological disorder, indicating that Irp2 has important functions in the nervous system and erythropoietic homeostasis. Several excellent review articles have recently been published on Irp knockout mice that mainly focus on Irp1(-/-) mice (referenced in the introduction). In this review, we will briefly describe the phenotypes and physiological implications of Irp1(-/-) mice and discuss the phenotypes observed for Irp2(-/-) mice in detail with a particular emphasis on the neurological problems of these mice.


Subject(s)
Iron Metabolism Disorders/etiology , Iron-Regulatory Proteins/deficiency , Neurodegenerative Diseases/complications , Neurodegenerative Diseases/metabolism , Animals , Disease Models, Animal , Humans , Iron Metabolism Disorders/genetics , Iron-Regulatory Proteins/genetics , Mice , Mice, Transgenic , Neurodegenerative Diseases/genetics
15.
Front Pharmacol ; 5: 124, 2014.
Article in English | MEDLINE | ID: mdl-24982634

ABSTRACT

Iron regulatory proteins (IRPs) regulate the expression of genes involved in iron metabolism by binding to RNA stem-loop structures known as iron responsive elements (IREs) in target mRNAs. IRP binding inhibits the translation of mRNAs that contain an IRE in the 5'untranslated region of the transcripts, and increases the stability of mRNAs that contain IREs in the 3'untranslated region of transcripts. By these mechanisms, IRPs increase cellular iron absorption and decrease storage and export of iron to maintain an optimal intracellular iron balance. There are two members of the mammalian IRP protein family, IRP1 and IRP2, and they have redundant functions as evidenced by the embryonic lethality of the mice that completely lack IRP expression (Irp1 (-/-)/Irp2(-/-) mice), which contrasts with the fact that Irp1 (-/-) and Irp2 (-/-) mice are viable. In addition, Irp2 (-/-) mice also display neurodegenerative symptoms and microcytic hypochromic anemia, suggesting that IRP2 function predominates in the nervous system and erythropoietic homeostasis. Though the physiological significance of IRP1 had been unclear since Irp1 (-/-) animals were first assessed in the early 1990s, recent studies indicate that IRP1 plays an essential function in orchestrating the balance between erythropoiesis and bodily iron homeostasis. Additionally, Irp1 (-/-) mice develop pulmonary hypertension, and they experience sudden death when maintained on an iron-deficient diet, indicating that IRP1 has a critical role in the pulmonary and cardiovascular systems. This review summarizes recent progress that has been made in understanding the physiological roles of IRP1 and IRP2, and further discusses the implications for clinical research on patients with idiopathic polycythemia, pulmonary hypertension, and neurodegeneration.

16.
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
17.
Blood ; 118(10): 2868-77, 2011 Sep 08.
Article in English | MEDLINE | ID: mdl-21700773

ABSTRACT

The iron-regulatory hormone, hepcidin, regulates systemic iron homeostasis by interacting with the iron export protein ferroportin (FPN1) to adjust iron absorption in enterocytes, iron recycling through reticuloendothelial macrophages, and iron release from storage in hepatocytes. We previously demonstrated that FPN1 was highly expressed in erythroblasts, a cell type that consumes most of the serum iron for use in hemoglobin synthesis. Herein, we have demonstrated that FPN1 localizes to the plasma membrane of erythroblasts, and hepcidin treatment leads to decreased expression of FPN1 and a subsequent increase in intracellular iron concentrations in both erythroblast cell lines and primary erythroblasts. Moreover, injection of exogenous hepcidin decreased FPN1 expression in BM erythroblasts in vivo, whereas iron depletion and associated hepcidin reduction led to increased FPN1 expression in erythroblasts. Taken together, hepcidin decreased FPN1 expression and increased intracellular iron availability of erythroblasts. We hypothesize that FPN1 expression in erythroblasts allows fine-tuning of systemic iron utilization to ensure that erythropoiesis is partially suppressed when nonerythropoietic tissues risk developing iron deficiency. Our results may explain why iron deficiency anemia is the most pronounced early manifestation of mammalian iron deficiency.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Cation Transport Proteins/metabolism , Erythroblasts/drug effects , Erythroblasts/metabolism , Iron/metabolism , Animals , Blotting, Western , Cation Transport Proteins/genetics , Cell Membrane/metabolism , Electrophoretic Mobility Shift Assay , Erythroblasts/cytology , Fetus/cytology , Fetus/drug effects , Fetus/metabolism , Hepcidins , Homeostasis , Immunoprecipitation , Iron, Dietary/administration & dosage , Liver/cytology , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
Mitochondrion ; 10(5): 497-509, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20580947

ABSTRACT

A homozygous mutation in the complex III chaperone BCS1L causes GRACILE syndrome (intrauterine growth restriction, aminoaciduria, cholestasis, hepatic iron overload, lactacidosis). In control and patient fibroblasts we localized BCS1L in inner mitochondrial membranes. In patient liver, kidney, and heart BCS1L and Rieske protein levels, as well as the amount and activity of complex III, were decreased. Major histopathology was found in kidney and liver with cirrhosis and iron deposition, but of iron-related proteins only ferritin levels were high. In placenta from a GRACILE fetus, the ferrooxidases ceruloplasmin and hephaestin were upregulated suggesting association between iron overload and placental dysfunction.


Subject(s)
Congenital Abnormalities/pathology , Electron Transport Complex III/metabolism , Liver Diseases , ATPases Associated with Diverse Cellular Activities , Ceruloplasmin/metabolism , Electron Transport Complex III/deficiency , Female , Ferritins/metabolism , Histocytochemistry , Humans , Infant , Infant, Newborn , Iron/metabolism , Kidney/pathology , Liver/pathology , Membrane Proteins/metabolism , Myocardium/pathology , Placenta/pathology , Pregnancy , Up-Regulation
19.
Blood ; 116(9): 1574-84, 2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20472835

ABSTRACT

The serum ferritin concentration is a clinical parameter measured widely for the differential diagnosis of anemia. Its levels increase with elevations of tissue iron stores and with inflammation, but studies on cellular sources of serum ferritin as well as its subunit composition, degree of iron loading and glycosylation have given rise to conflicting results. To gain further understanding of serum ferritin, we have used traditional and modern methodologies to characterize mouse serum ferritin. We find that both splenic macrophages and proximal tubule cells of the kidney are possible cellular sources for serum ferritin and that serum ferritin is secreted by cells rather than being the product of a cytosolic leak from damaged cells. Mouse serum ferritin is composed mostly of L-subunits, whereas it contains few H-subunits and iron content is low. L-subunits of serum ferritin are frequently truncated at the C-terminus, giving rise to a characteristic 17-kD band that has been previously observed in lysosomal ferritin. Taken together with the fact that mouse serum ferritin is not detectably glycosylated, we propose that mouse serum ferritin is secreted through the nonclassical lysosomal secretory pathway.


Subject(s)
Ferritins/blood , Iron/metabolism , Lysosomes/metabolism , Macrophages/metabolism , Secretory Pathway , Amino Acid Sequence , Animals , Enzyme-Linked Immunosorbent Assay , Glycosylation , Iron Overload/metabolism , Iron Overload/pathology , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Protein Subunits , Sequence Homology, Amino Acid
20.
Metab Brain Dis ; 24(4): 673-84, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19851851

ABSTRACT

Maintenance of appropriate iron homeostasis in the brain is important, but the mechanisms involved in brain iron uptake are incompletely understood. Here, we have analyzed where messenger RNAs that encode iron transport proteins are expressed in the brain, using the Allen Brain atlas, and we conclude that several important iron transporters are highly expressed in the choroid plexus. Based on recent estimates of the surface area of the choroid plexus and on MRI imaging studies of manganese uptake in the brain, we propose that the choroid plexus may have a much greater role than has been previously appreciated in brain iron transport.


Subject(s)
Brain/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Choroid Plexus/metabolism , Homeostasis/physiology , Iron/metabolism , Animals , Atlases as Topic , Brain/anatomy & histology , Brain Chemistry/physiology , Brain Mapping , Capillaries/cytology , Capillaries/metabolism , Cation Transport Proteins/metabolism , Ceruloplasmin/metabolism , Choroid Plexus/cytology , Epithelial Cells/metabolism , Ferritins/metabolism , Gene Expression/physiology , Magnetic Resonance Imaging , Mice , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...