Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
J Am Soc Nephrol ; 28(12): 3605-3615, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28784700

ABSTRACT

Renal transplants remain a medical challenge, because the parameters governing allograft outcome are incompletely identified. Here, we investigated the role of serum iron in the sterile inflammation that follows kidney ischemia-reperfusion injury. In a retrospective cohort study of renal allograft recipients (n=169), increased baseline levels of serum ferritin reliably predicted a positive outcome for allografts, particularly in elderly patients. In mice, systemic iron overload protected against renal ischemia-reperfusion injury-associated sterile inflammation. Furthermore, chronic iron injection in mice prevented macrophage recruitment after inflammatory stimuli. Macrophages cultured in high-iron conditions had reduced responses to Toll-like receptor-2, -3, and -4 agonists, which associated with decreased reactive oxygen species production, increased nuclear localization of the NRF2 transcription factor, increased expression of the NRF2-related antioxidant response genes, and limited NF-κB and proinflammatory signaling. In macrophage-depleted animals, the infusion of macrophages cultured in high-iron conditions did not reconstitute AKI after ischemia-reperfusion, whereas macrophages cultured in physiologic iron conditions did. These findings identify serum iron as a critical protective factor in renal allograft outcome. Increasing serum iron levels in patients may thus improve prognosis of renal transplants.


Subject(s)
Iron/blood , Kidney/pathology , Reperfusion Injury/prevention & control , Adult , Allografts , Animals , Antioxidants/metabolism , Female , Ferritins/blood , Glomerular Filtration Rate , Humans , Inflammation , Iron/chemistry , Kidney/metabolism , Kidney Transplantation , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Monocytes/cytology , NF-E2-Related Factor 2/metabolism , Peritonitis/metabolism , Prognosis , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Signal Transduction
2.
Haematologica ; 102(2): 260-270, 2017 02.
Article in English | MEDLINE | ID: mdl-28143953

ABSTRACT

Hemolysis occurring in hematologic diseases is often associated with an iron loading anemia. This iron overload is the result of a massive outflow of hemoglobin into the bloodstream, but the mechanism of hemoglobin handling has not been fully elucidated. Here, in a congenital erythropoietic porphyria mouse model, we evaluate the impact of hemolysis and regenerative anemia on hepcidin synthesis and iron metabolism. Hemolysis was confirmed by a complete drop in haptoglobin, hemopexin and increased plasma lactate dehydrogenase, an increased red blood cell distribution width and osmotic fragility, a reduced half-life of red blood cells, and increased expression of heme oxygenase 1. The erythropoiesis-induced Fam132b was increased, hepcidin mRNA repressed, and transepithelial iron transport in isolated duodenal loops increased. Iron was mostly accumulated in liver and spleen macrophages but transferrin saturation remained within the normal range. The expression levels of hemoglobin-haptoglobin receptor CD163 and hemopexin receptor CD91 were drastically reduced in both liver and spleen, resulting in heme- and hemoglobin-derived iron elimination in urine. In the kidney, the megalin/cubilin endocytic complex, heme oxygenase 1 and the iron exporter ferroportin were induced, which is reminiscent of significant renal handling of hemoglobin-derived iron. Our results highlight ironbound hemoglobin urinary clearance mechanism and strongly suggest that, in addition to the sequestration of iron in macrophages, kidney may play a major role in protecting hepatocytes from iron overload in chronic hemolysis.


Subject(s)
Anemia, Hemolytic/metabolism , Hepatocytes/metabolism , Hepcidins/metabolism , Iron/metabolism , Anemia, Hemolytic/blood , Anemia, Hemolytic/complications , Anemia, Hemolytic/genetics , Animals , Apoptosis , Biological Transport , Biomarkers , Disease Models, Animal , Erythrocytes/metabolism , Erythropoiesis , Gene Expression , Heme/metabolism , Hepcidins/blood , Hepcidins/genetics , Humans , Iron/urine , Iron Overload/etiology , Iron Overload/metabolism , Macrophages , Mice , Mice, Knockout , Mice, Transgenic , Spleen/physiology , Stress, Physiological
3.
Gastroenterology ; 151(4): 771-2, 2016 10.
Article in English | MEDLINE | ID: mdl-27623323
4.
Gastroenterology ; 150(3): 672-683.e4, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26582087

ABSTRACT

BACKGROUND & AIMS: Hereditary hemochromatosis is a heterogeneous group of genetic disorders characterized by parenchymal iron overload. It is caused by defective expression of liver hepcidin, the main regulator of iron homeostasis. Iron stimulates the gene encoding hepcidin (HAMP) via the bone morphogenetic protein (BMP)6 signaling to SMAD. Although several genetic factors have been found to cause late-onset hemochromatosis, many patients have unexplained signs of iron overload. We investigated BMP6 function in these individuals. METHODS: We sequenced the BMP6 gene in 70 consecutive patients with a moderate increase in serum ferritin and liver iron levels who did not carry genetic variants associated with hemochromatosis. We searched for BMP6 mutations in relatives of 5 probands and in 200 healthy individuals (controls), as well as in 2 other independent cohorts of hyperferritinemia patients. We measured serum levels of hepcidin by liquid chromatography-tandem mass spectrometry and analyzed BMP6 in liver biopsy specimens from patients by immunohistochemistry. The functions of mutant and normal BMP6 were assessed in transfected cells using immunofluorescence, real-time quantitative polymerase chain reaction, and immunoblot analyses. RESULTS: We identified 3 heterozygous missense mutations in BMP6 (p.Pro95Ser, p.Leu96Pro, and p.Gln113Glu) in 6 unrelated patients with unexplained iron overload (9% of our cohort). These mutations were detected in less than 1% of controls. p.Leu96Pro also was found in 2 patients from the additional cohorts. Family studies indicated dominant transmission. Serum levels of hepcidin were inappropriately low in patients. A low level of BMP6, compared with controls, was found in a biopsy specimen from 1 patient. In cell lines, the mutated residues in the BMP6 propeptide resulted in defective secretion of BMP6; reduced signaling via SMAD1, SMAD5, and SMAD8; and loss of hepcidin production. CONCLUSIONS: We identified 3 heterozygous missense mutations in BMP6 in patients with unexplained iron overload. These mutations lead to loss of signaling to SMAD proteins and reduced hepcidin production. These mutations might increase susceptibility to mild-to-moderate late-onset iron overload.


Subject(s)
Bone Morphogenetic Protein 6/genetics , Hemochromatosis/genetics , Hemochromatosis/metabolism , Hepcidins/biosynthesis , Heterozygote , Iron/metabolism , Liver/metabolism , Mutation, Missense , Aged , Animals , Biopsy , Bone Morphogenetic Protein 6/metabolism , Case-Control Studies , Cell Line , Chromatography, Liquid , DNA Mutational Analysis , Female , Ferritins/blood , Genetic Association Studies , Genetic Predisposition to Disease , Hemochromatosis/blood , Hepcidins/blood , Humans , Immunohistochemistry , Male , Middle Aged , Opossums , Phenotype , Smad Proteins, Receptor-Regulated/metabolism , Tandem Mass Spectrometry , Transfection
5.
J Am Soc Nephrol ; 27(3): 835-46, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26293821

ABSTRACT

The iron-regulatory peptide hepcidin exhibits antimicrobial activity. Having previously shown hepcidin expression in the kidney, we addressed its role in urinary tract infection (UTI), which remains largely unknown. Experimental UTI was induced in wild-type (WT) and hepcidin-knockout (Hepc-/-) mice using the uropathogenic Escherichia coli CFT073 strain. Compared with infected WT mice, infected Hepc-/- mice showed a dramatic increase in renal bacterial load. Moreover, bacterial invasion was significantly dampened by the pretreatment of WT mice with hepcidin. Infected Hepc-/- mice exhibited decreased iron accumulation in the renal medulla and significant attenuation of the renal inflammatory response. Notably, we demonstrated in vitro bacteriostatic activity of hepcidin against CFT073. Furthermore, CFT073 repressed renal hepcidin, both in vivo and in cultured renal cells, and reduced phosphorylation of SMAD kinase in vivo, suggesting a bacterial strategy to escape the antimicrobial activities of hepcidin. In conclusion, we provide new mechanisms by which hepcidin contributes to renal host defense and suggest that targeting hepcidin offers a strategy to prevent bacterial invasion.


Subject(s)
Anti-Infective Agents/pharmacology , Escherichia coli Infections/metabolism , Escherichia coli/drug effects , Hepcidins/metabolism , Hepcidins/pharmacology , Urinary Tract Infections/metabolism , Animals , Anti-Infective Agents/metabolism , Bacterial Load/genetics , Cells, Cultured , Colony Count, Microbial , Cytokines/metabolism , Escherichia coli Infections/microbiology , Female , Hepcidins/genetics , Iron/metabolism , Kidney Medulla/cytology , Kidney Medulla/metabolism , Kidney Medulla/microbiology , Mice , Mice, Inbred CBA , Mice, Knockout , Nephritis/metabolism , Nephritis/microbiology , Nephritis/pathology , Neutrophils , Phosphorylation , RNA, Messenger/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Smad Proteins/metabolism , Urinary Tract Infections/microbiology
6.
Am J Pathol ; 185(11): 3039-52, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26343328

ABSTRACT

Tissue pantetheinase, encoded by the VNN1 gene, regulates response to stress, and previous studies have shown that VNN genes contribute to the susceptibility to malaria. Herein, we evaluated the role of pantetheinase on erythrocyte homeostasis and on the development of malaria in patients and in a new mouse model of pantetheinase insufficiency. Patients with cerebral malaria have significantly reduced levels of serum pantetheinase activity (PA). In mouse, we show that a reduction in serum PA predisposes to severe malaria, including cerebral malaria and severe anemia. Therefore, scoring pantetheinase in serum may serve as a severity marker in malaria infection. This disease triggers an acute stress in erythrocytes, which enhances cytoadherence and hemolysis. We speculated that serum pantetheinase might contribute to erythrocyte resistance to stress under homeostatic conditions. We show that mutant mice with a reduced serum PA are anemic and prone to phenylhydrazine-induced anemia. A cytofluorometric and spectroscopic analysis documented an increased frequency of erythrocytes with an autofluorescent aging phenotype. This is associated with an enhanced oxidative stress and shear stress-induced hemolysis. Red blood cell transfer and bone marrow chimera experiments show that the aging phenotype is not cell intrinsic but conferred by the environment, leading to a shortening of red blood cell half-life. Therefore, serum pantetheinase level regulates erythrocyte life span and modulates the risk of developing complicated malaria.


Subject(s)
Amidohydrolases/blood , Erythrocytes/physiology , Malaria/physiopathology , Adolescent , Adult , Amidohydrolases/metabolism , Anemia , Animals , Child , Child, Preschool , Disease Models, Animal , Disease Susceptibility , Female , GPI-Linked Proteins/blood , GPI-Linked Proteins/metabolism , Homeostasis , Humans , Infant , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , Young Adult
7.
Clin Chem Lab Med ; 53(10): 1557-67, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-25781546

ABSTRACT

BACKGROUND: The peptide hepcidin plays a central role in regulating dietary iron absorption and body iron distribution. This 25-amino acid hormone is produced and secreted predominantly by hepatocytes. Hepcidin has been suggested as a promising diagnostic marker for iron-related disorders. However, its accurate quantification for clinical use remains so far challenging. In this report we describe a highly specific and quantitative serum hepcidin method using liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). MATERIAL: The analytical validation included the determination of the limit of detection, of quantification, repeatability, reproducibility and linearity. This assay was developed for human and mouse hepcidin. The human assay was performed on serum patients with unexplained microcytic anemia. We applied our LC-MS/MS method for quantifying hepcidin-1 in mouse in various conditions: inflammation, hemolytic anemia, Hamp-1, Hjv and Hfe KO mice. RESULTS: We show that the LC-MS/MS is suitable for accurate determination of hepcidin-25 in clinical samples, thereby representing a useful tool for the clinical diagnosis and follow-up of iron-related diseases. In mouse, a strong correlation between hepatic Hamp-1 mRNA expression and serum hepcidin-1 levels was found (r=0.88; p=0.0002) and the expected variations in mouse models of iron disorders were observed. CONCLUSIONS: Therefore, we propose this adaptive LC-MS/MS method as a suitable method for accurate determination of hepcidin-25 in clinical samples and as a major tool contributing to the clinical diagnosis, follow-up and management of iron-related disorders. It also opens new avenues to measure hepcidin in animal models without interspecies antigenic limitations.


Subject(s)
Anemia, Iron-Deficiency/blood , Chromatography, Liquid/methods , Hepcidins/blood , Tandem Mass Spectrometry/methods , Adult , Amino Acid Sequence , Animals , Female , Humans , Iron/blood , Male , Mice , Middle Aged , Models, Animal , Molecular Sequence Data , Reproducibility of Results , Young Adult
8.
Hum Mutat ; 35(11): 1321-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25156943

ABSTRACT

Iron-refractory iron-deficiency anemia (IRIDA) is a rare autosomal-recessive disorder characterized by hypochromic microcytic anemia, low transferrin saturation, and inappropriate high levels of the iron hormone hepcidin. The disease is caused by variants in the transmembrane protease serine 6 (TMPRSS6) gene that encodes the type II serine protease matriptase-2, a negative regulator of hepcidin transcription. Sequencing analysis of the TMPRSS6 gene in 21 new IRIDA patients from 16 families with different ethnic origin reveal 17 novel mutations, including the most frequent mutation in Southern Italy (p.W590R). Eight missense mutations were analyzed in vitro. All but the p.T287N variant impair matriptase-2 autoproteotylic activation, decrease the ability to cleave membrane HJV and inhibit the HJV-dependent hepcidin activation. Genotype-phenotype studies in IRIDA patients have been so far limited due to the relatively low number of described patients. Our genotype-phenotype correlation analysis demonstrates that patients carrying two nonsense mutations present a more severe anemia and microcytosis and higher hepcidin levels than the other patients. We confirm that TMPRSS6 mutations are spread along the gene and that mechanistically they fully or partially abrogate hepcidin inhibition. Genotyping IRIDA patients help in predicting IRIDA severity and may be useful for predicting response to iron treatment.


Subject(s)
Anemia, Iron-Deficiency/diagnosis , Anemia, Iron-Deficiency/genetics , Genetic Association Studies , Genetic Variation , Genotype , Membrane Proteins/genetics , Phenotype , Serine Endopeptidases/genetics , Adolescent , Adult , Anemia, Iron-Deficiency/therapy , Child , Child, Preschool , Female , Gene Frequency , Gene Order , Genetic Loci , Humans , Infant , Male , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mutation , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Young Adult
9.
Am J Hum Genet ; 94(4): 611-7, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24680888

ABSTRACT

In 90% of people with erythropoietic protoporphyria (EPP), the disease results from the inheritance of a common hypomorphic FECH allele, encoding ferrochelatase, in trans to a private deleterious FECH mutation. The activity of the resulting FECH enzyme falls below the critical threshold of 35%, leading to the accumulation of free protoporphyrin IX (PPIX) in bone marrow erythroblasts and in red cells. The mechanism of low expression involves a biallelic polymorphism (c.315-48T>C) localized in intron 3. The 315-48C allele increases usage of the 3' cryptic splice site between exons 3 and 4, resulting in the transcription of an unstable mRNA with a premature stop codon, reducing the abundance of wild-type FECH mRNA, and finally reducing FECH activity. Through a candidate-sequence approach and an antisense-oligonucleotide-tiling method, we identified a sequence that, when targeted by an antisense oligonucleotide (ASO-V1), prevented usage of the cryptic splice site. In lymphoblastoid cell lines derived from symptomatic EPP subjects, transfection of ASO-V1 reduced the usage of the cryptic splice site and efficiently redirected the splicing of intron 3 toward the physiological acceptor site, thereby increasing the amount of functional FECH mRNA. Moreover, the administration of ASO-V1 into developing human erythroblasts from an overtly EPP subject markedly increased the production of WT FECH mRNA and reduced the accumulation of PPIX to a level similar to that measured in asymptomatic EPP subjects. Thus, EPP is a paradigmatic Mendelian disease in which the in vivo correction of a common single splicing defect would improve the condition of most affected individuals.


Subject(s)
Ferrochelatase/genetics , Oligonucleotides, Antisense/therapeutic use , Protoporphyria, Erythropoietic/therapy , Cell Line , Female , Humans , Male , Pedigree , Polymorphism, Genetic , Protoporphyrins/metabolism , RNA Splicing , RNA, Messenger/genetics
10.
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
11.
Haematologica ; 98(6): 845-53, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23729726

ABSTRACT

Iron refractory iron deficiency anemia is a hereditary recessive anemia due to a defect in the TMPRSS6 gene encoding Matriptase-2. This protein is a transmembrane serine protease that plays an essential role in down-regulating hepcidin, the key regulator of iron homeostasis. Hallmarks of this disease are microcytic hypochromic anemia, low transferrin saturation and normal/high serum hepcidin values. The anemia appears in the post-natal period, although in some cases it is only diagnosed in adulthood. The disease is refractory to oral iron treatment but shows a slow response to intravenous iron injections and partial correction of the anemia. To date, 40 different Matriptase-2 mutations have been reported, affecting all the functional domains of the large ectodomain of the protein. In vitro experiments on transfected cells suggest that Matriptase-2 cleaves Hemojuvelin, a major regulator of hepcidin expression and that this function is altered in this genetic form of anemia. In contrast to the low/undetectable hepcidin levels observed in acquired iron deficiency, in patients with Matriptase-2 deficiency, serum hepcidin is inappropriately high for the low iron status and accounts for the absent/delayed response to oral iron treatment. A challenge for the clinicians and pediatricians is the recognition of the disorder among iron deficiency and other microcytic anemias commonly found in pediatric patients. The current treatment of iron refractory iron deficiency anemia is based on parenteral iron administration; in the future, manipulation of the hepcidin pathway with the aim of suppressing it might become an alternative therapeutic approach.


Subject(s)
Anemia, Iron-Deficiency/etiology , Anemia, Iron-Deficiency/diagnosis , Anemia, Iron-Deficiency/therapy , Genes, Recessive , Humans , Membrane Proteins/deficiency , Phenotype , Serine Endopeptidases/deficiency
12.
Kidney Int ; 84(4): 756-66, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23615502

ABSTRACT

Hepcidin, the key regulatory hormone of iron homeostasis, and iron carriers such as transferrin receptor1 (TFR1), divalent metal transporter1 (DMT1), and ferroportin (FPN) are expressed in kidney. Whether hepcidin plays an intrinsic role in the regulation of renal iron transport is unknown. Here, we analyzed the renal handling of iron in hemochromatosis Hepc(-/-) and Hjv(-/-) mouse models, as well as in phenylhydrazine (PHZ)-treated mice. We found a marked medullary iron deposition in the kidneys of Hepc(-/-) mice, and iron leak in the urine. The kidneys of Hepc(-/-) mice exhibited a concomitant decrease in TFR1 and increase in ferritin and FPN expression. Increased FPN abundance was restricted to the thick ascending limb (TAL). DMT1 protein remained unaffected despite a significant decrease of its mRNA level, suggesting that DMT1 protein is stabilized in the absence of hepcidin. Treatment of kidney sections from Hepc(-/-) mice with hepcidin decreased DMT1 protein, an effect confirmed in renal cell lines where hepcidin markedly decreased (55)Fe transport. In the kidneys of Hjv(-/-) mice exhibiting low hepcidin expression, the iron overload was similar to that in the kidneys of Hepc(-/-) mice. However, in PHZ mice, iron accumulation resulting from hemoglobin leak was detected in the proximal tubule. Thus, kidneys exhibit a tissue-specific handling of iron that depends on the extra iron source. Hepcidin may control the expression of iron transporters to prevent renal iron overload.


Subject(s)
Hemochromatosis/metabolism , Hepcidins/metabolism , Homeostasis/physiology , Iron/metabolism , Kidney Tubules, Distal/metabolism , Animals , Cation Transport Proteins/metabolism , Cell Line , Cells, Cultured , Disease Models, Animal , GPI-Linked Proteins , Hemochromatosis/chemically induced , Hemochromatosis/genetics , Hemochromatosis Protein , Hepcidins/deficiency , Hepcidins/genetics , In Vitro Techniques , Kidney Tubules, Distal/pathology , Loop of Henle/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Opossums , Phenylhydrazines/adverse effects , Receptors, Transferrin/metabolism
13.
Ann Biol Clin (Paris) ; 70(4): 377-86, 2012.
Article in French | MEDLINE | ID: mdl-22796609

ABSTRACT

The characterization of hepcidin and of its role in iron metabolism in 2001 has entirely modified our understanding of the pathogenesis of iron-linked disorders. Clinical applications related to hepcidin are numerous, and involve iron-overload diseases, anemia, renal disease, chronic inflammation or cancer. Thus, new avenues have emerged from the discovery of this 25 amino acid peptide and numerous diagnostic and therapeutic tools have been developed and described in the past 10 years. In particular, various methods for accurate quantification of hepcidin in urine and serum have been published but development of a reliable assay is quite a challenging task because, in particular, of inherent properties of the peptide's structure and its low immunogenicity. Thus, the different methods described so far show high variability, in terms of quantitative value of hepcidin measured, specificity and sensitivity. Comparison of these methods has been recently described in a "Round Robin" study, in order to harmonise hepcidin assays and to improve and standardise the available detection/quantification tests. This article focuses on the molecular mechanisms of hepcidin regulation and reviews the various methods of hepcidin quantification, describing the advantages and limitations of each one of them.


Subject(s)
Antimicrobial Cationic Peptides/blood , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/urine , Chromatography, Liquid , Enzyme-Linked Immunosorbent Assay , Erythropoiesis , Gene Expression Regulation , Hepcidins , Humans , Inflammation/blood , Iron Metabolism Disorders/blood , Mass Spectrometry , Membrane Proteins/genetics , Radioimmunoassay , Serine Endopeptidases/genetics
14.
Hum Mutat ; 33(9): 1388-96, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22581667

ABSTRACT

Mutations of the TMPRSS6 gene, which encodes Matriptase-2, are responsible for iron-refractory iron-deficiency anemia. Matriptase-2 is a transmembrane protease that downregulates hepcidin expression. We report one frameshift (p.Ala605ProfsX8) and four novel missense mutations (p.Glu114Lys, p.Leu235Pro, p.Tyr418Cys, p.Pro765Ala) found in IRIDA patients. These mutations lead to changes in both the catalytic and noncatalytic domains of Matriptase-2. Analyses of the mutant proteins revealed a reduction of autoactivating cleavage and the loss of N-Boc-Gln-Ala-Arg-p-nitroanilide hydrolysis. This resulted either from a direct modification of the active site or from the lack of the autocatalytic cleavage that transforms the zymogen into an active protease. In a previously described transfection assay measuring the ability of Matriptase-2 to repress the hepcidin gene (HAMP) promoter, all mutants retained some, if not all, of their transcriptional repression activity. This suggests that caution is called for in interpreting the repression assay in assessing the functional relevance of Matriptase-2 substitutions. We propose that Matriptase-2 activity should be measured directly in the cell medium of transfected cells using the chromogenic substrate. This simple test can be used to determine whether a sequence variation leading to an amino acid substitution is functionally relevant or not.


Subject(s)
Anemia, Iron-Deficiency/enzymology , Antimicrobial Cationic Peptides/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Serine Proteases/metabolism , Transfection/methods , Adolescent , Adult , Amino Acid Sequence , Amino Acid Substitution , Anemia, Iron-Deficiency/genetics , Antimicrobial Cationic Peptides/blood , Antimicrobial Cationic Peptides/genetics , Catalytic Domain , Child , Child, Preschool , Chromogenic Compounds/metabolism , Culture Media/metabolism , Enzyme Activation , Enzyme Assays , Enzyme Precursors/metabolism , Frameshift Mutation , Gene Silencing , Genetic Testing , HeLa Cells , Hepcidins , Humans , Infant , Male , Membrane Proteins/genetics , Molecular Sequence Data , Mutation, Missense , Pedigree , Repressor Proteins/genetics , Repressor Proteins/metabolism , Serine Endopeptidases/genetics , Transcription, Genetic
15.
Crit Care Med ; 40(7): 2141-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22564959

ABSTRACT

OBJECTIVE: Anemia is common in critically ill patients, due to inflammation and blood loss. Anemia can be associated with iron deficiency and low serum hepcidin levels. However, iron administration in this setting remains controversial because of its potential toxicity, including oxidative stress induction and sepsis facilitation. The objective of this work was to determine the efficacy and toxicity of iron administration using a mouse model mimicking critical care anemia as well as a model of acute septicemia. DESIGN: Prospective, randomized, open label controlled animal study. SETTING: University-based research laboratory. SUBJECTS: C57BL/6 and OF1 mice. INTERVENTIONS: Intraperitoneal injection of zymosan inducing generalized inflammation in C57BL/6 mice, followed in our full model by repeated phlebotomies. A dose equivalent to 15 mg/kg of ferric carboxymaltose was injected intravenously on day 5. To assess the toxicity of iron in a septicemia model, OF1 mice were simultaneously injected with iron and different Escherichia coli strains. MEASUREMENTS AND MAIN RESULTS: To investigate the effect of iron on oxidative stress, we measured reactive oxygen species production in the blood using luminol-amplified chemiluminescence and superoxide dismutase 2 messenger RNA levels in the liver. These markers of oxidative stress were increased after iron administration in control mice but not in zymosan-treated mice. Liver catalase messenger RNA levels decreased in iron-treated control mice. Iron administration was not associated with increased mortality in the septicemia model or in the generalized inflammation model. Iron increased hemoglobin levels in mice fed with a low iron diet and subjected to phlebotomies and zymosan 2 wks after treatment administration. CONCLUSIONS: Adverse effects of intravenous iron supplementation by ferric carboxymaltose seem to be minimal in our animal models. Furthermore, iron appears to be effective in correcting anemia, despite inflammation. Studies of efficacy and safety of iron in critically ill patients are warranted.


Subject(s)
Anemia/drug therapy , Ferric Compounds/administration & dosage , Ferric Compounds/toxicity , Hematinics/administration & dosage , Hematinics/toxicity , Maltose/analogs & derivatives , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Catalase/genetics , Catalase/metabolism , Diet , Disease Models, Animal , Hemoglobins , Hepcidins , Inflammation/chemically induced , Injections, Intravenous , Iron/administration & dosage , Iron/analysis , Liver/chemistry , Liver/metabolism , Luminescence , Maltose/administration & dosage , Maltose/toxicity , Mice , Mice, Inbred C57BL , Phlebotomy , RNA, Messenger/metabolism , Random Allocation , Reactive Oxygen Species/blood , Sepsis/drug therapy , Spleen/chemistry , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Trace Elements/administration & dosage , Zymosan/pharmacology
16.
Ann Clin Biochem ; 49(Pt 3): 302-5, 2012 May.
Article in English | MEDLINE | ID: mdl-22535864

ABSTRACT

Investigating persistent hyperferritinaemia without apparent iron overload is challenging. Even when inflammation, cirrhosis, Still's disease, fatty liver and malignancy are excluded, there remains a group of patients with unexplained hyperferritinaemia for whom rare forms of haemochromatosis (ferroportin disease) are a consideration. Preliminary results suggest that abnormal percentage glycosylation of serum ferritin is seen in some cases of genetically determined hyperferritinaemia. Serum ferritin is normally 50-81% glycosylated, but low glycosylation (20-42%) prevails in hereditary hyperferritinaemia cataract syndrome. This contrasts with hyperglycosylation (>90%) associated with the benign hyperferritinaemia related to missense L ferritin (p.Thr30Ile) mutation. Here, we describe two novel missense L ferritin variants also associated with hyperglycosylation, p.Gln26Ile and p.Ala27Val. Ferritin glycosylation, a comparatively simple measurement, can identify patients for DNA sequencing as hyperglycosylation (>90%) is associated with benign hyperferritinaemia and mutant L ferritin chain.


Subject(s)
Apoferritins/genetics , Ferritins/genetics , Iron Metabolism Disorders/genetics , Mutation, Missense , Aged , Amino Acid Sequence , Apoferritins/blood , Ferritins/blood , Glycosylation , Humans , Iron Metabolism Disorders/blood , Iron Metabolism Disorders/diagnosis , Male , Middle Aged , Models, Molecular , Molecular Sequence Data , Open Reading Frames , Sequence Analysis, DNA
17.
Am J Clin Nutr ; 95(3): 548-54, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22301927

ABSTRACT

BACKGROUND: Only a few studies based on small cohorts have been carried out on iron status in anorexia nervosa (AN) patients. OBJECTIVE: The aim of this study was to evaluate the role of hepcidin in hyperferritinemia in AN adolescents. DESIGN: Twenty-seven adolescents hospitalized for AN in the pediatric inpatient unit of Ambroise Paré Academic Hospital were enrolled in the study. The control group comprised 11 patients. Hematologic variables and markers of iron status, including serum hepcidin, were measured before and after nutritional rehabilitation. RESULTS: The mean age of patients was 14.4 y. Except for 2 AN patients and 1 control patient, all patients presented normal hemoglobin, vitamin B-12, and folate concentrations. Markers of inflammation and cytokines were normal throughout the study. None of the muscular lysis markers were elevated. Most AN patients had normal serum iron concentrations on admission. Serum ferritin concentrations were significantly higher in patients than in control subjects (198 compared with 49 µg/L, respectively; P < 0.001). The median hepcidin concentration was significantly higher in AN patients than in the control group (186.5 compared with 39.5 µg/L, respectively; P = 0.002). There was a highly significant correlation between ferritinemia and serum hepcidin concentrations (P < 0.0001). After nutritional rehabilitation, a significant reduction was observed (P = 0.004) in serum ferritin. Serum hepcidin analyzed in a smaller number of patients also returned to within the normal range. CONCLUSIONS: Hepcidin and ferritin concentrations were higher in the serum of AN patients, without any evidence of iron overload or inflammation. These concentrations returned to normal after nutritional rehabilitation. These results suggest that nutritional stress induced by malnourishment in the hepatocyte could be yet another mechanism that regulates hepcidin.


Subject(s)
Anorexia Nervosa/physiopathology , Antimicrobial Cationic Peptides/blood , Iron, Dietary/metabolism , Adolescent , Anorexia Nervosa/complications , Anorexia Nervosa/metabolism , Biomarkers/blood , Case-Control Studies , Child , Cytokines/blood , Female , Ferritins/blood , Follow-Up Studies , Hepcidins , Hospitalization , Humans , Inflammation/metabolism , Inflammation/physiopathology , Iron Metabolism Disorders/complications , Iron Metabolism Disorders/physiopathology , Male
19.
Blood ; 118(25): 6660-6, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22031863

ABSTRACT

STEAP3/TSAP6 encodes a ferrireductase that is involved in the acquisition of iron by developing erythroblasts and steap3/tsap6 null-mice display severe microcytic anemia. We report a family in which 3 siblings born to healthy parents display transfusion-dependent hypochromic anemia. A nonsense STEAP3/TSAP6 was identified in the siblings at the heterozygous state. This mutation was inherited from their father while no mutation was found in their mother. A large variability of expression was found between normal alleles in a control population, confirming a previous report that STEAP3/TSAPS6 is an expressed quantitative trait locus (e-QTL). Determination of the relative allele expression showed that the "normal" allele was expressed at a significantly higher level in the father than in the affected siblings relative to the shared mutated allele. The blood level of STEAP3/TSAP6 mRNA was severely reduced in the siblings, while both parents were in the lower range of normal controls. The STEAP3/TSAP6 protein was also reduced in lymphocytic cell lines from the patients. Collectively, our data support the hypothesis that STEAP3/TSAP6 deficiency leads to severe anemia in the affected siblings and results from the combination of a mutated allele inherited from their father and a weakly expressed allele inherited from their mother.


Subject(s)
Anemia, Hypochromic/genetics , Codon, Nonsense , Oncogene Proteins/genetics , Adolescent , Anemia, Hypochromic/blood , Anemia, Hypochromic/congenital , Animals , Blotting, Western , Cell Cycle Proteins , Cell Line, Transformed , Cells, Cultured , Child , DNA Mutational Analysis , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Family Health , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Oncogene Proteins/metabolism , Oxidoreductases , Pedigree , RNA, Messenger/blood , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Young Adult
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...