Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
Blood ; 142(2): 185-196, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37146247

ABSTRACT

Pregnancy rates in ß-thalassemia are increasing but the risk of complications is higher; thus, better understanding of maternal and fetal iron homeostasis in this disorder is needed. HbbTh3/+ (Th3/+) mice model human ß-thalassemia. Both the murine and human diseases are characterized by low hepcidin, high iron absorption, and tissue iron overload, with concurrent anemia. We hypothesized that disordered iron metabolism in pregnant Th3/+ mice would negatively affect their unborn offspring. The experimental design included these groups: wild-type (WT) dams carrying WT fetuses (WT1); WT dams carrying WT and Th3/+ fetuses (WT2); Th3/+ dams carrying WT and Th3/+ fetuses (Th3/+); and age-matched, nonpregnant adult females. Serum hepcidin was low, and mobilization of splenic and hepatic storage iron was enhanced in all 3 groups of experimental dams. Intestinal 59Fe absorption was lower in Th3/+ dams (as compared with WT1/2 dams) but splenic 59Fe uptake was higher. Th3/+ dams had hyperferremia, which led to fetal and placenta iron loading, fetal growth restriction, and placentomegaly. Notably, Th3/+ dams loaded Th3/+ and WT fetuses, with the latter situation more closely mirroring human circumstances when mothers with thalassemia have relatively unaffected (thalassemia trait) offspring. Iron-related oxidative stress likely contributed to fetal growth impairment; enhanced placental erythropoiesis is a probable cause of placental enlargement. Moreover, high fetal liver iron transactivated Hamp; fetal hepcidin downregulated placental ferroportin expression, limiting placental iron flux and thus mitigating fetal iron loading. Whether gestational iron loading occurs in human thalassemic pregnancy, when blood transfusion can further elevate serum iron, is worth consideration.


Subject(s)
Hepcidins , beta-Thalassemia , Mice , Female , Humans , Pregnancy , Animals , beta-Thalassemia/metabolism , Placenta/metabolism , Iron/metabolism , Fetus/metabolism , Homeostasis
2.
Biometals ; 32(5): 745-755, 2019 10.
Article in English | MEDLINE | ID: mdl-31368012

ABSTRACT

Hinokitiol, a natural lipophilic chelator, appears capable of replacing several iron transporters after they have been genetically ablated. Divalent metal-ion transporter (DMT1) is the major iron importer in enterocytes and erythroblasts. We have compared DMT1 and hinokitiol in multiple fashions to learn if the smaller molecule is a suitable substitute using two HEK293 cell lines engineered to overexpress different isoforms of DMT1. Both the macromolecule and the lipophilic chelator enable import of ferrous ions into HEK293 cells. Hinokitiol also mediates ferric ion import but DMT1 cannot do so. While DMT1 can also import Mn2+ ions, hinokitiol lacks this ability. The Michaelis-Menten analysis for kinetics of macromolecular catalysis is also suitable for hinokitiol-supported iron import. To compare hinokitiol to DMT1 relative to other metal ions that DMT1 can transport, we employed an organic extraction procedure with which we initially matched the results obtained for Fe2+, Fe3+ and Mn2+, and then showed that multiple other cations were unlikely to enter via hinokitiol. The small chelator thus shares some functional properties with DMT1, but distinct difference were also noted.


Subject(s)
Ferrous Compounds/metabolism , Manganese/metabolism , Monoterpenes/metabolism , Transcription Factors/metabolism , Tropolone/analogs & derivatives , Genetic Therapy , HEK293 Cells , Humans , Iron/metabolism , Kinetics , Tropolone/metabolism
3.
Sci Rep ; 8(1): 211, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29317744

ABSTRACT

Much of iron and manganese metabolism occurs in mitochondria. Uptake of redox-active iron must be tightly controlled, but little is known about how metal ions enter mitochondria. Recently, we established that the divalent metal transporter 1 (DMT1) is present in the outer mitochondrial membrane (OMM). Therefore we asked if it mediates Fe2+ and Mn2+ influx. Mitochondria were isolated from HEK293 cells permanently transfected with inducible rat DMT1 isoform 1 A/+IRE (HEK293-rDMT1). Fe2+-induced quenching of the dye PhenGreen™SK (PGSK) occurred in two phases, one of which reflected OMM DMT1 with stronger Fe2+ uptake after DMT1 overexpression. DMT1-specific quenching showed an apparent affinity of ~1.5 µM for Fe2+and was blocked by the DMT1 inhibitor CISMBI. Fe2+ influx reflected an imposed proton gradient, a response that was also observed in purified rat kidney cortex (rKC) mitochondria. Non-heme Fe accumulation assayed by ICPOES and stable 57Fe isotope incorporation by ICPMS were increased in HEK293-rDMT1 mitochondria. HEK293-rDMT1 mitochondria displayed higher 59Fe2+ and 54Mn2+ uptake relative to controls with 54Mn2+ uptake blocked by the DMT1 inhibitor XEN602. Such transport was defective in rKC mitochondria with the Belgrade (G185R) mutation. Thus, these results support a role for DMT1 in mitochondrial Fe2+ and Mn2+ acquisition.


Subject(s)
Cation Transport Proteins/metabolism , Iron/metabolism , Manganese/metabolism , Mitochondria/metabolism , Animals , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/genetics , Cell Line, Tumor , Cells, Cultured , HEK293 Cells , Humans , Ion Transport , Mice , Mutation, Missense , Rats , Rats, Long-Evans
4.
Channels (Austin) ; 8(5): 458-66, 2014.
Article in English | MEDLINE | ID: mdl-25483589

ABSTRACT

The divalent metal transporter (DMT1) is well known for its roles in duodenal iron absorption across the apical enterocyte membrane, in iron efflux from the endosome during transferrin-dependent cellular iron acquisition, as well as in uptake of non-transferrin bound iron in many cells. Recently, using multiple approaches, we have obtained evidence that the mitochondrial outer membrane is another subcellular locale of DMT1 expression. While iron is of vital importance for mitochondrial energy metabolism, its delivery is likely to be tightly controlled due to iron's damaging redox properties. Here we provide additional support for a role of DMT1 in mitochondrial iron acquisition by immunofluorescence colocalization with mitochondrial markers in cells and isolated mitochondria, as well as flow cytometric quantification of DMT1-positive mitochondria from an inducible expression system. Physiological consequences of mitochondrial DMT1 expression are discussed also in consideration of other DMT1 substrates, such as manganese, relevant to mitochondrial antioxidant defense.


Subject(s)
Mitochondria/metabolism , Transcription Factors/metabolism , Animals , CHO Cells , Cricetulus , Humans
5.
FASEB J ; 28(5): 2134-45, 2014 May.
Article in English | MEDLINE | ID: mdl-24448823

ABSTRACT

In mammalian cells, mitochondria receive most incoming iron, yet no entry pathway for iron at the outer mitochondrial membrane (OMM) has been characterized. Our results show that the divalent metal transporter 1 (DMT1) occurs in the OMM. Immunoblots detected DMT1 in mitochondria from a pneumocyte cell model in their OMM. Using the split-ubiquitin yeast 2-hybrid system, we found that cytochrome c oxidase subunit II (COXII) and the translocase of OMM 6-kDa subunit (Tom6) homologue interact with DMT1. COXII coimmunoprecipitates with DMT1. There are 4 DMT1 isoforms that differ at the N and C termini. Using HEK293 cells that inducibly express all of the 4 ends of DMT1, we found all of them in the OMM, as detected by immunoblots after cell fractionation, and in isolated mitochondria, as detected by immunofluorescence. Immunoblot analysis of purified cell fractions from rat renal cortex confirmed and extended these results to the kidney, which expressed high levels of DMT1. Immunogold labeling detected DMT1 colocalization in mitochondria with the voltage-dependent anion-selective channel protein-1, which is expressed in the OMM. We suggest that DMT1 not only exports iron from endosomes, but also serves to import the metal into the mitochondria.


Subject(s)
Cation Transport Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Alveolar Epithelial Cells/cytology , Animals , Anions , Electron Transport Complex IV/metabolism , Endosomes/metabolism , HEK293 Cells , Humans , Kidney Cortex/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Plasmids/metabolism , Protein Isoforms/metabolism , Rats , Rats, Wistar , Saccharomyces cerevisiae Proteins/metabolism , Two-Hybrid System Techniques
6.
J Nutr ; 143(12): 1927-33, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24089420

ABSTRACT

Intracellular copper-binding proteins (metallothionein I/II) and a copper exporter (Menkes copper-transporting ATPase) are upregulated in duodenal enterocytes from iron-deficient rats, consistent with copper accumulation in the intestinal mucosa. How copper enters enterocytes during iron deficiency is, however, not clear. Divalent metal transporter 1 (Dmt1), the predominant iron importer in the mammalian duodenum, also transports other metal ions, possibly including copper. Given this possibility and that Dmt1 expression is upregulated by iron deprivation, we sought to test the hypothesis that Dmt1 transports copper during iron deficiency. Two model systems were utilized: the Belgrade (b) rat, expressing mutant Dmt1, and an inducible Dmt1-overexpression cell culture system. Mutant rats (b/b) were fed a semipurified, AIN93G-based control diet and phenotypically normal littermates (+/b) were fed control or iron-deficient diets for ~14 wk. An everted gut sleeve technique and a colorimetric copper quantification assay were utilized to assess duodenal copper transport. The control diet-fed +/b rats had normal hematological parameters, whereas iron-deprived +/b and b/b rats were iron deficient and Dmt1 mRNA and protein levels increased. Importantly, duodenal copper transport was similar in the control +/b and b/b rats; however, it significantly increased (~4-fold) in the iron-deprived +/b rats. Additional experiments in Dmt1 overexpressing HEK-293 cells showed that copper ((64)Cu) uptake was stimulated (∼3-fold) in the presence of an iron chelator. Dmt1 transcript stabilization due to a 3' iron-responsive element was also documented, likely contributing to increased transport activity. In summary, these studies suggest that Dmt1 enhances copper uptake into duodenal enterocytes during iron deprivation.


Subject(s)
Cation Transport Proteins/physiology , Copper/metabolism , Duodenum/metabolism , Iron/metabolism , Animals , Base Sequence , Biological Transport , Cation Transport Proteins/genetics , DNA Primers , Female , HEK293 Cells , Humans , Male , Polymerase Chain Reaction , RNA, Messenger/genetics , Rats
7.
Biometals ; 25(4): 787-93, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22310887

ABSTRACT

Divalent metal ion transporter (DMT1) is the major transporter for iron entrance into mammalian cells and iron exit from endosomes during the transferrin cycle. Four major mRNA isoforms correspond to four protein isoforms, differing at 5'/3' and N-/C-termini, respectively. Isoforms are designated 1A versus 1B reflecting where transcription starts or +iron responsive element (+IRE) versus -IRE reflecting the presence/absence of an IRE in the 3' end of the mRNA. These differences imply regulation at transcriptional and posttranscriptional levels. Many proteins are degraded by a ubiquitination-dependent mechanism. Two different ubiquitin ligases (E3s) appear to be involved in DMT1 ubiquitination: Parkin or neuronal precursor cell-expressed developmentally downregulated 4 (Nedd4) family E3s which often utilize Nedd4 family interacting protein-1 and -2 (Ndfip1 and 2) to ubiquitinate their substrate proteins. Prior data suggest that Parkin ubiquitinates 1B DMT1 but not 1A DMT1 while Nedd4/Ndfips ligate ubiquitin to DMT1 in the duodenum where 1A/+IRE DMT1 predominates. Our assay for whether these systems target DMT1 depends on two HEK293 cell lines that express permanently transfected 1A/+IRE DMT1 or 1B/-IRE DMT1 after induction by doxycycline. Transient transfection with a Parkin construct before induction diminishes 1B/-IRE DMT1 detected by immune-blots but not 1A/+IRE DMT1. Mutant Parkin serves as a control that does not affect DMT1 levels. Thus DMT1 regulation in an isoform specific fashion can occur by ubiquitination and the events involved have implications for DMT1 function and disease processes.


Subject(s)
Cation Transport Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Isoforms/metabolism , Carrier Proteins/metabolism , Cell Line , Endosomal Sorting Complexes Required for Transport/metabolism , Humans , Iron/metabolism , Membrane Proteins/metabolism , Nedd4 Ubiquitin Protein Ligases , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
9.
Biochim Biophys Acta ; 1790(5): 309-25, 2009 May.
Article in English | MEDLINE | ID: mdl-19344751

ABSTRACT

Iron has a split personality as an essential nutrient that also has the potential to generate reactive oxygen species. We discuss how different cell types within specific tissues manage this schizophrenia. The emphasis in enterocytes is on regulating the body's supply of iron by regulating transport into the blood stream. In developing red blood cells, adaptations in transport manage the body's highest flux of iron. Hepatocytes buffer the body's stock of iron. Macrophage recycle the iron from effete red cells among other iron management tasks. Pneumocytes provide a barrier to prevent illicit entry that, when at risk of breaching, leads to a need to handle the dangers in a fashion essentially shared with macrophage. We also discuss or introduce cell types including renal cells, neurons, other brain cells, and more where our ignorance, currently still vast, needs to be removed by future research.


Subject(s)
Cells/metabolism , Iron/metabolism , Animals , Biological Transport , Humans , Mitochondria/metabolism , Transferrin/metabolism
10.
Proc Natl Acad Sci U S A ; 105(47): 18578-83, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-19011085

ABSTRACT

Dopaminergic cell death in the substantia nigra (SN) is central to Parkinson's disease (PD), but the neurodegenerative mechanisms have not been completely elucidated. Iron accumulation in dopaminergic and glial cells in the SN of PD patients may contribute to the generation of oxidative stress, protein aggregation, and neuronal death. The mechanisms involved in iron accumulation also remain unclear. Here, we describe an increase in the expression of an isoform of the divalent metal transporter 1 (DMT1/Nramp2/Slc11a2) in the SN of PD patients. Using the PD animal model of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) intoxication in mice, we showed that DMT1 expression increases in the ventral mesencephalon of intoxicated animals, concomitant with iron accumulation, oxidative stress, and dopaminergic cell loss. In addition, we report that a mutation in DMT1 that impairs iron transport protects rodents against parkinsonism-inducing neurotoxins MPTP and 6-hydroxydopamine. This study supports a critical role for DMT1 in iron-mediated neurodegeneration in PD.


Subject(s)
Cation Transport Proteins/physiology , Parkinson Disease/pathology , Aged , Aged, 80 and over , Animals , Disease Models, Animal , Dopamine/metabolism , Humans , Iron/metabolism , Mice , Oxidative Stress , Parkinson Disease/metabolism , Parkinson Disease/physiopathology
11.
Am J Physiol Gastrointest Liver Physiol ; 294(4): G948-62, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18258795

ABSTRACT

To identify novel genes associated with iron metabolism, we performed gene chip studies in two models of iron deficiency: iron-deprived rats and rats deficient in the principal intestinal iron transporter, divalent metal transporter 1 (i.e., Belgrade rats). Affymetrix rat genome gene chips were utilized (RAE230) with cRNA samples derived from duodenum and jejunum of experimental and control animals. Computational analysis and statistical data reduction identified 29 candidate genes, which were induced in both models of iron deficiency. Gene ontology analysis showed enrichment for genes related to lipid homeostasis, and one gene related to this physiological process, a leukocyte type, arachidonate 12-lipoxygenase (Alox15), was selected for further examination. TaqMan real-time PCR studies demonstrated strong induction of Alox15 throughout the small and large intestine, and in the liver of iron-deficient rats. Polyclonal antibodies were developed and utilized to demonstrate that proteins levels are significantly increased in the intestinal epithelium of iron-deprived rats. HPLC analysis revealed altered intestinal lipid metabolism indicative of Alox15 activity, which resulted in the production of biologically active lipid molecules (12-HETE, 13-HODE, and 13-HOTE). The overall effect is a perturbation of intestinal lipid homeostasis, which results in the production of lipids essentially absent in the intestine of control rats. We have thus provided mechanistic insight into the alteration in lipid metabolism that occurs during iron deficiency, in that induction of Alox15 mRNA expression may be the primary event. The resulting lipid mediators may be related to documented alterations in villus structure and cell proliferation rates in iron deficiency, or to structural alterations in membrane lipid composition.


Subject(s)
Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 15-Lipoxygenase/biosynthesis , Duodenum/metabolism , Iron Deficiencies , Iron Metabolism Disorders/metabolism , Jejunum/metabolism , Lipid Metabolism/genetics , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Algorithms , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Blotting, Western , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Chromatography, High Pressure Liquid , Cluster Analysis , Disease Models, Animal , Duodenum/enzymology , Duodenum/pathology , Enzyme Induction , Gene Expression Profiling/methods , Immunohistochemistry , Iron Metabolism Disorders/enzymology , Iron Metabolism Disorders/genetics , Iron Metabolism Disorders/pathology , Jejunum/enzymology , Jejunum/pathology , Linoleic Acids/metabolism , Liver/metabolism , Male , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
12.
Am J Physiol Lung Cell Mol Physiol ; 292(1): L134-43, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16905637

ABSTRACT

We tested the hypothesis that oxidative stress and biological effect after ozone (O3) exposure are dependent on changes in iron homeostasis. After O3 exposure, healthy volunteers demonstrated increased lavage concentrations of iron, transferrin, lactoferrin, and ferritin. In normal rats, alterations of iron metabolism after O3 exposure were immediate and preceded the inflammatory influx. To test for participation of this disruption in iron homeostasis in lung injury following O3 inhalation, we exposed Belgrade rats, which are functionally deficient in divalent metal transporter 1 (DMT1) as a means of iron uptake, and controls to O3. Iron homeostasis was disrupted to a greater extent and the extent of injury was greater in Belgrade rats than in control rats. Nonheme iron and ferritin concentrations were higher in human bronchial epithelial (HBE) cells exposed to O3 than in HBE cells exposed to filtered air. Aldehyde generation and IL-8 release by the HBE cells was also elevated following O3 exposure. Human embryonic kidney (HEK 293) cells with elevated expression of a DMT1 construct were exposed to filtered air and O3. With exposure to O3, elevated DMT1 expression diminished oxidative stress (i.e., aldehyde generation) and IL-8 release. We conclude that iron participates critically in the oxidative stress and biological effects after O3 exposure.


Subject(s)
Iron/metabolism , Lung Injury , Lung/drug effects , Ozone/toxicity , Adolescent , Adult , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cation Transport Proteins/deficiency , Cell Line , Disease Models, Animal , Ferritins/metabolism , Homeostasis , Humans , Lactoferrin/metabolism , Lung/metabolism , Lung/pathology , Male , Oxidative Stress , Ozone/administration & dosage , Rats , Rats, Inbred F344 , Rats, Mutant Strains , Rats, Sprague-Dawley , Receptors, Transferrin/metabolism , Time Factors , Transferrin/metabolism
13.
Biochim Biophys Acta ; 1773(2): 105-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17087999

ABSTRACT

The hbd (hemoglobin deficit) mutation affects iron trafficking in murine reticulocytes. It is due to a deletion that eliminates exon 8 of Sec15l1, the homolog of a gene that encodes an exocyst component in yeast. We tested the hypothesis that the mutation causes defective slow or rapid receptor recycling by measuring endocytosis and exocytosis of transferrin by hbd reticulocytes. Endocytosis and initial iron incorporation were relatively unaffected, but exocytosis was unexpectedly slowed. These data indicate that rapid transferrin recycling is defective after pSec15l1 has mutated.


Subject(s)
Endocytosis , Hemoglobins/deficiency , Membrane Proteins/genetics , Mutation/genetics , Receptors, Transferrin/metabolism , Animals , Exocytosis , Iodine Radioisotopes , Iron/metabolism , Mice , Mice, Mutant Strains , Reticulocytes/metabolism , Time Factors
14.
Biochem J ; 398(3): 539-46, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16737442

ABSTRACT

DMT1 (divalent metal transporter; also known as SLC11A2, DCT1 or Nramp2) is responsible for ferrous iron uptake in the duodenum, iron exit from endosomes during the transferrin cycle and some transferrin-independent iron uptake in many cells. Four protein isoforms differ by starting in exon 1A or 2 and ending with alternative peptides encoded by mRNA that contains or lacks an IRE (iron responsive element; +/-IRE). We have compared 1A/+IRE and 2/-IRE DMT1 during regulated ectopic expression. HEK-293-F (human embryonic kidney-293-fast growing variant) cells were stably transfected with each construct expressed from a tetracycline-regulated CMV promoter. Reverse transcriptase-PCR analysis showed that construct expression responded to doxycycline. Immunofluorescence staining of cells, using antibodies specific for DMT1 isoforms, confirmed an increase in expression in the plasma membrane and cytosolic vesicles after doxycycline treatment, but with isoform specific distributions. Immunoblotting also revealed stimulation of expression. Nevertheless, both DMT1 isoforms performed similarly in assays for functional properties based on 54Mn2+ and 59Fe2+ uptake. Mn incorporation after doxycycline treatment was approximately 10-fold greater than that of untreated cells, while expression in the untreated cells was approximately 5-fold greater than in the untransfected cells. Uptake of Mn depended on addition of doxycycline, with half maximal response at approximately 1 nM doxycycline. Doxycycline-stimulated Mn and Fe uptake was linear with time for 10 min but not over longer periods. Transport exhibited a pH optimum at approximately 5.5 and dependence on incubation temperature and Mn or Fe concentration. The new cell lines should prove useful for research on metal homoeostasis, toxicological studies and efforts to identify distinctive properties of the isoforms.


Subject(s)
Cation Transport Proteins/metabolism , Doxycycline/pharmacology , Gene Expression Regulation/drug effects , Iron-Binding Proteins/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Cation Transport Proteins/genetics , Cell Line , Humans , Hydrogen-Ion Concentration , Iron/metabolism , Iron-Binding Proteins/genetics , Manganese/metabolism , Mice , Protein Isoforms , Rats , Time Factors
15.
Biol Res ; 39(1): 67-77, 2006.
Article in English | MEDLINE | ID: mdl-16629166

ABSTRACT

Iron is essential for many aspects of cellular function. However, it also can generate oxygen-based free radicals that result in injury to biological molecules. For this reason, iron acquisition and distribution are tightly regulated. Constant exposure to the atmosphere results in significant exposure of the lungs to catalytically active iron. The lungs have a mechanism for detoxification to prevent associated generation of oxidative stress. Those same proteins that participate in iron uptake in the gut are also employed in the lung, to transport iron intracellularly and sequester it in an inactive form within ferritin. The release of metal is expedited (as transferrin and ferritin) from lung tissue to the respiratory lining fluid for clearance by the mucocilliary pathway or to the reticuloendothelial system for long-term storage. This pathway is likely to be the major method for the control of oxidative stress presented to the respiratory tract.


Subject(s)
Epithelial Cells/metabolism , Homeostasis/physiology , Iron/metabolism , Lung/metabolism , Oxidative Stress , Ferritins/metabolism , Humans , Iron-Binding Proteins/metabolism , Lung/cytology , Macrophages/metabolism , Neutrophils/metabolism
16.
Biol Res ; 39(1): 79-85, 2006.
Article in English | MEDLINE | ID: mdl-16629167

ABSTRACT

DMT1-Divalent Metal (Ion) Transporter 1 or SLC11A2/DCT1/Nramp2 - transports Fe2+ into the duodenum and out of the endosome during the transferrin cycle. DMTI also is important in non-transferrin bound iron uptake. It plays similar roles in Mn2+ trafficking. Voltage clamping showed that six other metals evoked currents, but it is unclear if these metals are substrates for DMT1. This report summarizes progress on which metals DMT1 transports, focusing on results from the authors' labs. We recently cloned 1A/+IRE and 2/-IRE DMT1 isoforms to generate HEK293 cell lines that express them in a tetracycline-inducible fashion, then compared induced expression to uninduced expression and to endogenous DMT1 expression. Induced expression increases approximately 50x over endogenous expression and approximately 10x over uninduced levels. Fe2+, Mn2+, Ni2+ and Cu1+ or Cu2+ are transported. We also explored competition between metal ions using this system because incorporation essentially represents DMT1 transport and find this order for transport affinity: Mn>?Cd>?Fe>Pb-Co-Ni>Zn. The effects of decreased DMT1 also could be examined. The Belgrade rat has diminished DMT1 function and thus provides ways of testing. A series of DNA constructs that generate siRNAs specific for DMT1 or certain DMT1 isoforms yield another way to test DMT1-based transport.


Subject(s)
Cation Transport Proteins/metabolism , Metals/metabolism , RNA, Small Interfering , Animals , Biological Transport , Caco-2 Cells , Cation Transport Proteins/genetics , Humans , Rats
17.
Biol. Res ; 39(1): 67-77, 2006. ilus, tab
Article in English | LILACS | ID: lil-430699

ABSTRACT

Iron is essential for many aspects of cellular function. However, it also can generate oxygen-based free radicals that result in injury to biological molecules. For this reason, iron acquisition and distribution are tightly regulated. Constant exposure to the atmosphere results in significant exposure of the lungs to catalytically active iron. The lungs have a mechanism for detoxification to prevent associated generation of oxidative stress. Those same proteins that participate in iron uptake in the gut are also employed in the lung to transport iron intracellularly and sequester it in an inactive form within ferritin. The release of metal is expedited (as transferrin and ferritin) from lung tissue to the respiratory lining fluid for clearance by the mucocilliary pathway or to the reticuloendothelial system for long-term storage. This pathway is likely to be the major method for the control of oxidative stress presented to the respiratory tract.


Subject(s)
Humans , Epithelial Cells/metabolism , Homeostasis/physiology , Iron/metabolism , Lung/metabolism , Oxidative Stress , Ferritins/metabolism , Iron-Binding Proteins/metabolism , Lung/cytology , Macrophages/metabolism , Neutrophils/metabolism
18.
Biol. Res ; 39(1): 79-85, 2006. ilus, tab
Article in English | LILACS | ID: lil-430700

ABSTRACT

DMT1 _ Divalent Metal (Ion) Transporter 1 or SLC11A2/DCT1/Nramp2 _ transports Fe2+ into the duodenum and out of the endosome during the transferrin cycle. DMT1 also is important in non-transferrin bound iron uptake. It plays similar roles in Mn2+ trafficking. Voltage clamping showed that six other metals evoked currents, but it is unclear if these metals are substrates for DMT1. This report summarizes progress on which metals DMT1 transports, focusing on results from the authors' labs. We recently cloned 1A/+IRE and 2/-IRE DMT1 isoforms to generate HEK293 cell lines that express them in a tetracycline-inducible fashion, then compared induced expression to uninduced expression and to endogenous DMT1 expression. Induced expression increases about 50x over endogenous expression and about 10x over uninduced levels. Fe2+, Mn2+, Ni2+ and Cu1+ or Cu2+ are transported. We also explored competition between metal ions using this system because incorporation essentially represents DMT1 transport and find this order for transport affinity: Mn>?Cd>?Fe>Pb Co Ni>Zn. The effects of decreased DMT1 also could be examined. The Belgrade rat has diminished DMT1 function and thus provides ways of testing. A series of DNA constructs that generate siRNAs specific for DMT1 or certain DMT1 isoforms yield another way to test DMT1-based transport.


Subject(s)
Animals , Humans , Rats , Cation Transport Proteins/metabolism , Metals/metabolism , RNA, Small Interfering , Biological Transport , Cation Transport Proteins/genetics
19.
Am J Physiol Lung Cell Mol Physiol ; 289(3): L460-7, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15908475

ABSTRACT

Exposure to airborne particulates makes the detoxification of metals a continuous challenge for the lungs. Based on the fate of iron in airway epithelial cells, we postulated that divalent metal transporter-1 (DMT1) participates in detoxification of metal associated with air pollution particles. Homozygous Belgrade rats, which are functionally deficient in DMT1, exhibited diminished metal transport from the lower respiratory tract and greater lung injury than control littermates when exposed to oil fly ash. Preexposure of normal rats to iron in vivo increased expression of the isoform of DMT1 protein that lacked an iron-response element (-IRE), accelerated metal transport out of the lung, and decreased injury after particle exposure. In contrast, normal rats preexposed to vanadium showed less expression of the -IRE isoform of DMT1, decreased metal transport, and greater pulmonary injury after particle instillation. Respiratory epithelial cells in culture gave similar results. Also, DMT1 mRNA and protein expression for the -IRE isoform increased or decreased in these cells when exposed to iron or vanadium, respectively. These results thus demonstrate for the first time a primary role for DMT1 in lung metal transport and detoxification.


Subject(s)
Cation Transport Proteins/physiology , Iron-Binding Proteins/physiology , Lung Diseases/chemically induced , Lung Diseases/prevention & control , Metals , Animals , Biological Transport/drug effects , Blotting, Western , Cation Transport Proteins/deficiency , Cation Transport Proteins/metabolism , Cell Line, Transformed , Ferric Compounds/pharmacology , Immunohistochemistry , Iron/pharmacokinetics , Iron-Binding Proteins/metabolism , Metals/metabolism , Oxidative Stress , Protein Isoforms/metabolism , Quaternary Ammonium Compounds/pharmacology , Rats , Rats, Inbred Strains , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Vanadium/pharmacokinetics , Vanadium Compounds/pharmacology
20.
Kidney Int ; 64(5): 1755-64, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14531808

ABSTRACT

BACKGROUND: We have previously shown that the rat kidney reabsorbs metabolically significant amounts of iron and that it expresses the divalent metal transporter 1, DMT1. The Belgrade (b) rat carries a mutation in DMT1 gene, which causes hypochromic, microcytic anemia due to impaired intestinal iron absorption and transport of iron out of the transferrin cycle endosome. In the duodenum of b/b rats, expression of DMT1 mRNA and protein is increased, suggesting a feedback regulation by iron stores. The aim of this study was to investigate iron handling and DMT1 expression in the kidneys of Belgrade rats. METHODS: Animals were maintained for 3 weeks on a synthetic diet containing 185 mg/kg iron (FeSO4), after which functional and molecular parameters were analyzed in male heterozygous (+/b) and homozygous (b/b) rats (N = 4 to 6 for each group). RESULTS: Serum iron concentration was significantly higher in b/b compared to +/b rats while urinary iron excretion rates were unchanged in b/b compared to +/b rats. Northern analysis using a rat DMT1 probe showed comparable mRNA levels between +/b and b/b animals. Western analysis and immunofluorescence microscopy performed using a polyclonal antibody against rat DMT1 showed that DMT1-specific immunoreactivity was almost absent in the kidneys of b/b rats compared to that seen in +/b animals. CONCLUSION: Our results indicate that the G185R mutation of DMT1 causes protein instability in the kidneys of b/b rats. Given that +/b and b/b rats excrete comparable amounts of iron, the lack of DMT1 protein is compensated by an alternative, yet to be identified, mechanism.


Subject(s)
Anemia/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Iron/blood , Iron/urine , Kidney/metabolism , Anemia/pathology , Animals , Drinking , Eating , Feces , Female , Kidney/pathology , Magnesium/blood , Male , Point Mutation , Potassium/blood , RNA, Messenger/analysis , Rats , Rats, Mutant Strains , Urine
SELECTION OF CITATIONS
SEARCH DETAIL