Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Mamm Genome ; 24(11-12): 427-38, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24121729

ABSTRACT

Disorders of iron metabolism are among the most common acquired and constitutive diseases. Hemochromatosis has a solid genetic basis and in Northern European populations it is usually associated with homozygosity for the C282Y mutation in the HFE protein. However, the penetrance of this mutation is incomplete and the clinical presentation is highly variable. The rare and common variants identified so far as genetic modifiers of HFE-related hemochromatosis are unable to account for the phenotypic heterogeneity of this disorder. There are wide variations in the basal iron status of common inbred mouse strains, and this diversity may reflect the genetic background of the phenotypic diversity under pathological conditions. We therefore examined the genetic basis of iron homeostasis using quantitative trait loci mapping applied to the HcB-15 recombinant congenic strains for tissue and serum iron indices. Two highly significant QTL containing either the N374S Mon1a mutation or the Ferroportin locus were found to be major determinants in spleen and liver iron loading. Interestingly, when considering possible epistatic interactions, the effects of Mon1a on macrophage iron export are conditioned by the genotype at the Slc40a1 locus. Only mice that are C57BL/10ScSnA homozygous at both loci display a lower spleen iron burden. Furthermore, the liver-iron lowering effect of the N374S Mon1a mutation is observed only in mice that display a nonsense mutation in the Ceruloplasmin (Cp) gene. This study highlights the existence of genetic interactions between Cp, Mon1a, and the Slc40a1 locus in iron metabolism, suggesting that epistasis may be a crucial determinant of the variable biological and clinical presentations in iron disorders.


Subject(s)
Carrier Proteins/genetics , Cation Transport Proteins/genetics , Ceruloplasmin/genetics , Epistasis, Genetic , Hemochromatosis/veterinary , Iron/metabolism , Mice/genetics , Rodent Diseases/genetics , Animals , Female , Hemochromatosis/genetics , Hemochromatosis/metabolism , Liver/metabolism , Male , Mice/metabolism , Mice, Inbred C3H , Mice, Inbred C57BL , Quantitative Trait Loci , Rodent Diseases/metabolism , Spleen/metabolism
2.
Haematologica ; 94(3): 335-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19176363

ABSTRACT

BACKGROUND: Elevated serum ferritin levels are frequently encountered in clinical situations and once iron overload or inflammation has been ruled out, many cases remain unexplained. Genetic causes of hyperferritinemia associated to early cataract include mutations in the iron responsive element in the 5' untranslated region of the L ferritin mRNA, responsible for the hereditary hyperferritinemia cataract syndrome. DESIGN AND METHODS: We studied 91 probands with hyperferritinemia comprising 25 family cases belonging to families with at least two cases of unexplained hyperferritinemia, and 66 isolated cases. In the families, we also analyzed 30 relatives. Hyperferritinemia was considered as unexplained when transferrin saturation was below 45% and/or serum iron below 25 mumol/L and/or no tissue iron excess was detected, when inflammation had been ruled out and when iron responsive element mutation was absent. We carried out sequencing analysis of the FTL gene coding the L ferritin. RESULTS: A novel heterozygous p.Thr30Ile mutation in the NH2 terminus of L ferritin subunit was identified in 17 probands out of the cohort. The mutation was shown to cosegregate with hyperferritinemia in all the 10 families studied. No obvious clinical symptom was found associated with the presence of the mutation. This unique mutation is associated with an unusually high percentage of ferritin glycosylation. CONCLUSIONS: This missense mutation of FTL represents a new cause of genetic hyperferritinemia without iron overload. We hypothesized that the mutation increases the efficacy of L ferritin secretion by increasing the hydrophobicity of the N terminal "A" alpha helix.


Subject(s)
Ferritins/blood , Iron Overload/blood , Mutation, Missense , Adolescent , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Apoferritins , Child , Family Health , Female , Ferritins/chemistry , Ferritins/genetics , Ferritins/metabolism , Glycosylation , Heterozygote , Humans , Iron Overload/genetics , Male , Middle Aged , Models, Molecular , Molecular Sequence Data , Pedigree , Protein Structure, Secondary , Young Adult
3.
Crit Care Med ; 36(8): 2388-94, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18664788

ABSTRACT

OBJECTIVE: Anemia in critically ill patients is frequent and consists of chronic disease associated with blood losses. These two mechanisms have opposite effects on iron homeostasis, especially on the expression of the iron regulatory hormone hepcidin. We developed a mouse model mimicking the intensive care anemia to explore iron homeostasis. DESIGN: Experimental study. SETTING: University-based research laboratory. SUBJECTS: C57BL/6 mice. INTERVENTIONS: Mice received either a single intraperitoneal injection of lipopolysaccharide followed 1 week later by zymosan, or were subjected to repeated phlebotomies by retro-orbital punctures, or both. Several subsets of mice were analyzed over a 14-day period to describe the mouse model of intensive care anemia. Additional mice received erythropoietin injections with or without the zymosan treatment and were killed at day 5, to perform a more detailed analysis. MEASUREMENTS AND MAIN RESULTS: We observed anemia as soon as 5 days after zymosan injection, together with increased messenger RNA (mRNA) levels for interleukin-6 and hepcidin. Phlebotomies alone fully suppressed hepcidin mRNA expression. Interestingly, in mice treated with zymosan and phlebotomies, hepcidin expression was suppressed, despite the persistent increase in interleukin-6. Stimulation of erythropoiesis by erythropoietin injections also led to a decrease in hepcidin mRNA in zymosan-treated mice. In these situations combining inflammation and erythropoiesis stimulation, there was no change in ferroportin, the membrane iron exporter, at the mRNA level, whereas ferroportin protein increased. Macrophage iron stores (assessed by histology using diaminobenzidine staining, or by quantification of nonheme iron and ferritin concentrations) were depleted in the spleen. CONCLUSIONS: These results suggest that the erythroid factor dominates over inflammation for hepcidin regulation, and that iron could be mobilized in these situations combining inflammation and erythropoiesis stimulation.


Subject(s)
Anemia/drug therapy , Antimicrobial Cationic Peptides/physiology , Cation Transport Proteins/metabolism , Erythropoietin/therapeutic use , Homeostasis/drug effects , Iron/metabolism , Microsomes, Liver/drug effects , Anemia/etiology , Anemia/metabolism , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Cation Transport Proteins/genetics , Cell Membrane/drug effects , Disease Models, Animal , Hepcidins , Interleukin-6/genetics , Male , Mice , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Microsomes, Liver/pathology , Phlebotomy , RNA, Messenger/metabolism
4.
Gastroenterology ; 132(2): 679-86, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17258727

ABSTRACT

BACKGROUND & AIMS: Genetic hemochromatosis is one of the most common genetic disorders, with progressive tissue iron overload leading to severe clinical complications. In Northern European populations, genetic hemochromatosis is usually caused by homozygosity for the C282Y mutation in the HFE protein. However, penetrance of this mutation is incomplete, suggesting that other genetic and environmental factors contribute to its differential biologic or clinical expression. METHODS: To identify genes modifying iron homeostasis, we screened the 27 recombinant congenic strains of the C3H/DiSnA-C57BL/10ScSnA/Dem series for tissue and serum iron indices and genotyped 18 microsatellite markers in (C3H/DiSnA x HcB-2) F2 hybrid mice. RESULTS: We identified 1 locus encompassing the Ceruloplasmin (Cp) gene with a strong linkage with liver iron, serum iron, and transferrin levels but not with spleen iron. Sequencing of Cp showed an R435X nonsense mutation in exon 7 in C3H/DiSnA mice. To evaluate whether Cp might act as a modifier gene of genetic hemochromatosis, we intercrossed C3H Hfe(-/-) and C3HDiSnA Cp(R435X/R435X) mice. As expected, we found that double-mutant mice deposited more iron in the liver than mice defective for either one or both genes. In contrast, Hfe(-/-) x Cp(R435/R435X) or Cp(R435X/R435X) x Hfe(+/-) showed 30% decrease in liver iron when compared with single mutant mice. CONCLUSIONS: This study highlights the existence of complex interactions between Cp and HFE and represents the first example of a modifier gene with a protective effect, in which heterozygosity reduces the iron load in the context of HFE deficiency.


Subject(s)
Ceruloplasmin/genetics , Genetic Linkage , Hemochromatosis/genetics , Hemochromatosis/metabolism , Iron/metabolism , Animals , Codon, Nonsense , Crosses, Genetic , Disease Models, Animal , Erythrocytes/metabolism , Genotype , Hemochromatosis/blood , Hemochromatosis/physiopathology , Hemochromatosis Protein , Histocompatibility Antigens Class I/genetics , Homeostasis/genetics , Iron/blood , Liver/metabolism , Lod Score , Macrophages/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Congenic , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Microsatellite Repeats/genetics , Molecular Sequence Data , Penetrance , Phagocytosis , Phenotype , Quantitative Trait Loci , Spleen/metabolism , Transferrin/metabolism
5.
Biochem J ; 400(2): 367-75, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16886906

ABSTRACT

RNA-binding activity of IRP1 (iron regulatory protein 1) is regulated by the insertion/extrusion of a [4Fe-4S] cluster into/from the IRP1 molecule. NO (nitic oxide), whose ability to activate IRP1 by removing its [4Fe-4S] cluster is well known, has also been shown to down-regulate expression of the IRP1 gene. In the present study, we examine whether this regulation occurs at the transcriptional level. Analysis of the mouse IRP1 promoter sequence revealed two conserved putative binding sites for transcription factor(s) regulated by NO and/or changes in intracellular iron level: Sp1 (promoter-selective transcription factor 1) and MTF1 (metal transcription factor 1), plus GAS (interferon-gamma-activated sequence), a binding site for STAT (signal transducer and activator of transcription) proteins. In order to define the functional activity of these sequences, reporter constructs were generated through the insertion of overlapping fragments of the mouse IRP1 promoter upstream of the luciferase gene. Transient expression assays following transfection of HuH7 cells with these plasmids revealed that while both the Sp1 and GAS sequences are involved in basal transcriptional activity of the IRP1 promoter, the role of the latter is predominant. Analysis of protein binding to these sequences in EMSAs (electrophoretic mobility-shift assays) using nuclear extracts from mouse RAW 264.7 macrophages stimulated to synthesize NO showed a significant decrease in the formation of Sp1-DNA and STAT-DNA complexes, compared with controls. We have also demonstrated that the GAS sequence is involved in NO-dependent down-regulation of IRP1 transcription. Further analysis revealed that levels of STAT5a and STAT5b in the nucleus and cytosol of NO-producing macrophages are substantially lower than in control cells. These findings provide evidence that STAT5 proteins play a role in NO-mediated down-regulation of IRP1 gene expression.


Subject(s)
Iron Regulatory Protein 1/biosynthesis , Nitric Oxide/physiology , STAT5 Transcription Factor/metabolism , Animals , Binding Sites , Cell Line , Cell Nucleus/metabolism , Cytosol/metabolism , DNA/genetics , DNA/metabolism , Down-Regulation/physiology , Humans , Interferon-gamma/pharmacology , Iron/metabolism , Iron Regulatory Protein 1/genetics , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Macrophages/physiology , Mice , Nitric Oxide/metabolism , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/biosynthesis , STAT5 Transcription Factor/genetics , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/metabolism , Transfection
6.
Biochem J ; 396(2): 265-75, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16457665

ABSTRACT

Ferroportin [FPN; Slc40a1 (solute carrier family 40, member 1)] is a transmembrane iron export protein expressed in macrophages and duodenal enterocytes. Heterozygous mutations in the FPN gene result in an autosomal dominant form of iron overload disorder, type-4 haemochromatosis. FPN mutants either have a normal iron export activity but have lost their ability to bind hepcidin, or are defective in their iron export function. The mutant protein has been suggested to act as a dominant negative over the wt (wild-type) protein by multimer formation. Using transiently transfected human epithelial cell lines expressing mouse FPN modified by the addition of a haemagglutinin or c-Myc epitope at the C-terminus, we show that the wtFPN is found at the plasma membrane and in Rab5-containing endosomes, as are the D157G and Q182H mutants. However, the delV162 mutant is mostly intracellular in HK2 cells (human kidney-2 cells) and partially addressed at the cell surface in HEK-293 cells (human embryonic kidney 293 cells). In both cell types, it is partially associated with the endoplasmic reticulum and with Rab5-positive vesicles. However, this mutant is complex-glycosylated like the wt protein. D157G and G323V mutants have a defective iron export capacity as judged by their inability to deplete the intracellular ferritin content, whereas Q182H and delV162 have normal iron export function and probably have lost their capacity to bind hepcidin. In co-transfection experiments, the delV162 mutant does not co-localize with the wtFPN, does not prevent its normal targeting to the plasma membrane and cannot be immunoprecipitated in the same complex, arguing against the formation of FPN hetero-oligomers.


Subject(s)
Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Epithelial Cells/metabolism , Mutation, Missense , Animals , Antibodies/metabolism , Biological Transport , Cell Membrane/metabolism , Cells, Cultured , Ferritins/metabolism , Fluorescent Antibody Technique , Glycosylation , Humans , Intracellular Membranes/metabolism , Iron/metabolism , Kidney/cytology , Kidney/metabolism , Mice , Models, Biological , Transfection
7.
Biochem Pharmacol ; 67(2): 213-25, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14698034

ABSTRACT

Fenofibrate, a peroxisome proliferator-activated receptor alpha (PPARalpha) activator, increases the expression of the cytosolic aspartate aminotransferase (cAspAT) gene in human liver cells, which may partially explain the increase of this enzyme in the serum of individuals undergoing fenofibrate treatment. Conversely, in rodents, fenofibrate represses the expression of the cAspAT gene. We compared the mechanisms of fenofibrate action in human and rat hepatoma cells. Transfection assays of the wild-type and mutated rat promoters in Fao and H4IIEC3 cells established a critical role for sequences similar to nuclear receptor responsive elements in the -404/-366 bp region. Nuclear proteins bound to these sequences and the amounts of protein bound were decreased by fenofibrate treatment, probably accounting for the decreased gene expression. Pharmacological studies confirmed the involvement of PPARalpha. However, this receptor did not bind directly to these sequences. The human promoter was cloned and the regulatory region localized between -2663/-706 bp. Co-transfection assays suggested that, in humans, the PPARalpha was also involved in the increase in expression of the cAspAT gene due to fibrates, without the presence of a canonical PPAR responsive element.


Subject(s)
Aspartate Aminotransferases/metabolism , Fenofibrate/pharmacology , Gene Expression Regulation/drug effects , Hypolipidemic Agents/pharmacology , Promoter Regions, Genetic/genetics , Animals , Aspartate Aminotransferases/genetics , Base Sequence , DNA/analysis , Genome , Humans , Molecular Sequence Data , Promoter Regions, Genetic/drug effects , Rats , Sequence Homology, Nucleic Acid , Species Specificity , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...