Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
2.
Cell Host Microbe ; 18(2): 254-61, 2015 Aug 12.
Article in English | MEDLINE | ID: mdl-26190773

ABSTRACT

Macrophages are essential for systemic iron recycling, and also control iron availability to pathogens. Iron metabolism in mammalian cells is orchestrated posttranscriptionally by iron-regulatory proteins (IRP)-1 and -2. Here, we generated mice with selective and combined ablation of both IRPs in macrophages to investigate the role of IRPs in controlling iron availability. These animals are hyperferritinemic but otherwise display normal clinical iron parameters. However, mutant mice rapidly succumb to systemic infection with Salmonella Typhimurium, a pathogenic bacterium that multiplies within macrophages, with increased bacterial burdens in liver and spleen. Ex vivo infection experiments indicate that IRP function restricts bacterial access to iron via the EntC and Feo bacterial iron-acquisition systems. Further, IRPs contain Salmonella by promoting the induction of lipocalin 2, a host antimicrobial factor that inhibits bacterial uptake of iron-laden siderophores, and by suppressing the ferritin iron pool. This work reveals the importance of the IRPs in innate immunity.


Subject(s)
Disease Resistance , Iron Regulatory Protein 1/metabolism , Iron Regulatory Protein 2/metabolism , Salmonella Infections, Animal/immunology , Salmonella typhimurium/immunology , Animals , Bacterial Load , Disease Models, Animal , Gene Knockout Techniques , Iron/metabolism , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 2/genetics , Liver/microbiology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/microbiology
3.
Blood ; 123(23): 3646-50, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24646470

ABSTRACT

Hepcidin is a 25-amino-acid peptide demonstrated to be the iron regulatory hormone capable of blocking iron absorption from the duodenum and iron release from macrophages. Mutations affecting hepcidin regulators or the hepcidin gene itself cause hemochromatosis, a common genetic disorder. Hepcidin is produced mainly by the liver, but many cells and tissues express low levels of the hormone. To determine the contribution of these hepcidin-producing tissues in body iron homeostasis, we have developed a new mouse model in which the hepcidin gene can be conditionally inactivated. Here we compare a liver-specific knockout (KO) mouse model with total KO mice. We show that the liver-specific KO mice fully recapitulate the severe iron overload phenotype observed in the total KO mice, with increased plasma iron and massive parenchymal iron accumulation. This result demonstrates that the hepatocyte constitutes the predominant reservoir for systemic hepcidin and that the other tissues are unable to compensate.


Subject(s)
Hemochromatosis/genetics , Hepcidins/genetics , Liver/metabolism , Animals , Disease Models, Animal , Gene Targeting , Hemochromatosis/pathology , Hepcidins/metabolism , Iron/metabolism , Iron Overload/genetics , Iron Overload/pathology , Liver/pathology , Male , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Phenotype
6.
Biochimie ; 91(10): 1223-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19555735

ABSTRACT

Hepcidin, a peptide hormone produced by the liver, constitutes the master regulator of iron homeostasis in mammals allowing iron adaptation according to the body iron needs. In recent years there has been important breakthrough in our knowledge of hepcidin regulation that has also implications for understanding the physiopathology of human iron disorders. Different aspects of hepcidin regulation will be considered in this review, including regulation by the iron status and the BMP6/HJV/SMAD pathway. Hepcidin dysregulation in iron disorders will be also discussed. Although much can already be accomplished for treating iron disorders using the knowledge that has currently been developed, additional issues will be challenging for the coming years.


Subject(s)
Antimicrobial Cationic Peptides/physiology , Iron/metabolism , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , GPI-Linked Proteins , Hemochromatosis Protein , Hepcidins , Homeostasis/genetics , Homeostasis/physiology , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Biological , Signal Transduction/genetics , Signal Transduction/physiology , Smad Proteins/genetics , Smad Proteins/metabolism
7.
J Mol Med (Berl) ; 87(9): 913-21, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19533074

ABSTRACT

The maintenance of iron homeostasis is critical as both iron deficiency and iron excess are deleterious. In mammals, iron homeostasis is regulated systemically by the iron-hormone hepcidin, an acute-phase protein secreted by the liver which inhibits iron absorption and recycling. Cellularly, the interaction of iron regulatory proteins (IRP) 1 and 2 with iron-responsive elements controls the expression of target mRNAs encoding proteins of iron acquisition, storage, utilization, and export. These processes critically affect iron levels, which in turn impact on numerous aspects of inflammation. To explore the role of IRP1 and IRP2 in inflammation, IRP-deficient mice, i.e., mice with total and constitutive deficiency of either IRP, were subjected to acute aseptic local inflammation. Turpentine oil injection increases the expression of acute phase proteins in the liver and interleukin 6 levels in the serum of control mice. Both IRP-deficient mouse models mount the same responses, indicating that the treatment was efficient in all animals and that the acute phase response does not require expression of both IRPs. As expected, turpentine oil treatment enhances hepcidin mRNA expression in the liver of wild-type mice, associated with decreased serum iron levels. Importantly, Irp1 (-/-) and Irp2 (-/-) animals, respectively, display quantitatively similar hepcidin mRNA induction and the appropriate reduction of the serum iron values. Our data indicate that the response of Irp1 (-/-) and Irp2 (-/-) mice to acute local inflammation is largely preserved.


Subject(s)
Inflammation/etiology , Iron Regulatory Protein 1/physiology , Iron Regulatory Protein 2/physiology , Acute-Phase Proteins/genetics , Acute-Phase Reaction/chemically induced , Animals , Antimicrobial Cationic Peptides/genetics , Gene Expression Regulation , Hepcidins , Inflammation/chemically induced , Iron/blood , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 2/genetics , Mice , Mice, Knockout , RNA, Messenger/genetics , Turpentine/toxicity
10.
Blood ; 108(4): 1402-5, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16574947

ABSTRACT

We previously reported that mice made deficient for the transcriptional factor USF2 fail to express hepcidin 1 and hepcidin 2 genes as a consequence of targeted disruption of the Usf2 gene lying just upstream in the locus. These mice developed an iron overload phenotype with excess iron deposition in parenchymal cells and decreased reticuloendothelial iron. At that time, although the role of USF2 was still confounding, we proposed for the first time the role of hepcidin as a negative regulator of iron absorption and iron release from macrophages. Accordingly, we subsequently demonstrated that hyperexpression of hepcidin 1, but not hepcidin 2, resulted in a profound hyposideremic anemia. To analyze the consequences of hepcidin 1 deletion on iron metabolism without any disturbance due to USF2 deficiency, we disrupted the hepcidin 1 gene by targeting almost all the coding region. Confirming our prior results, Hepc1(-/-) mice developed early and severe multivisceral iron overload, with sparing of the spleen macrophages, and demonstrated increased serum iron and ferritin levels as compared with their controls.


Subject(s)
Antimicrobial Cationic Peptides/deficiency , Gene Deletion , Hemochromatosis/genetics , Open Reading Frames/genetics , Quantitative Trait Loci/genetics , Animals , Antimicrobial Cationic Peptides/metabolism , Ferritins/metabolism , Hemochromatosis/metabolism , Hemochromatosis/pathology , Hepcidins , Iron/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Spleen/metabolism , Spleen/pathology , Upstream Stimulatory Factors/deficiency , Upstream Stimulatory Factors/metabolism
11.
Blood ; 107(7): 2952-8, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16339398

ABSTRACT

We report the generation of a tetracycline-regulated (Tet ON) transgenic mouse model for acute and chronic expression of the iron regulatory peptide hepcidin in the liver. We demonstrate that short-term and long-term tetracycline-dependent activation of hepcidin in adult mice leads to hypoferremia and iron-limited erythropoiesis, respectively. This clearly establishes the key role of hepcidin in regulating the extracellular iron concentration. We previously demonstrated that, when expressed early in fetal development, constitutive transgenic hepcidin expression prevented iron accumulation in an Hfe-/- mouse model of hemochromatosis. We now explore the effect of chronic hepcidin expression in adult Hfe-/- mice that have already developed liver iron overload. We demonstrate that induction of chronic hepcidin expression in 2-month-old Hfe-/- mice alters their pattern of cellular iron accumulation, leading to increased iron in tissue macrophages and duodenal cells but less iron in hepatocytes. These hepcidin-induced changes in the pattern of cellular iron accumulation are associated with decreased expression of the iron exporter ferroportin in macrophages but no detectable alteration of ferroportin expression in the hepatocytes. We speculate that this change in iron homeostasis could offer a therapeutic advantage by protecting against damage to parenchymal cells.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Hemochromatosis/blood , Iron/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Doxycycline/therapeutic use , Hemochromatosis Protein , Hepcidins , Histocompatibility Antigens Class I/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Tetracycline
12.
J Clin Invest ; 115(8): 2079-82, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16075054

ABSTRACT

Recently, mutations causing juvenile hemochromatosis have been identified in a novel gene, hemojuvelin (HJV), located on chromosome 1. Mouse models of this disease have now been developed by 2 groups, Huang et al. and Niederkofler et al., through targeted disruption of the Hjv gene (see the related articles beginning on pages 2180 and 2187). These mutant mice will allow further investigation into the role of HJV in the regulation of iron homeostasis, a role that to date remains elusive.


Subject(s)
Hemochromatosis/genetics , Iron/metabolism , Membrane Proteins/genetics , Animals , Chromosomes, Human, Pair 1/genetics , Disease Models, Animal , GPI-Linked Proteins , Hemochromatosis/metabolism , Hemochromatosis Protein , Homeostasis/genetics , Humans , Membrane Proteins/metabolism , Mice , Mice, Knockout
13.
Hepatology ; 41(5): 1056-64, 2005 May.
Article in English | MEDLINE | ID: mdl-15793843

ABSTRACT

Hepcidin, a recently discovered iron regulatory peptide, is believed to inhibit the release of iron from absorptive enterocytes and macrophages. Liver hepcidin synthesis is induced in vivo by iron stores and inflammation. The molecular basis of the regulation of hepcidin gene expression by these effectors in hepatocytes is currently unknown, although there is strong evidence that indirect mechanisms are involved. The aims of this study were to gain insight into these mechanisms and to determine to what extent other liver cell types are responsible for transducing the signal by which hepcidin expression is regulated in mouse hepatocytes. For this, we depleted Kupffer cells by injection of liposome-encapsulated clodronate and then studied iron- and inflammation-induced hepcidin gene expression. In addition, we directly evaluated the role of the inflammatory cytokine interleukin 6 (IL-6) by using IL-6-deficient mice. Our results show that iron is able to induce hepcidin gene expression independently of Kupffer cells in the liver and circulating IL-6. In contrast, we show that hepcidin gene induction by inflammation is also independent of Kupffer cells, but involves, at least partly, IL-6. In conclusion, these results show that two independent regulatory pathways control hepcidin gene expression and suggest that hepatocytes play a key role in the regulation of hepcidin gene expression by sensing iron and inflammatory signals.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Hepatitis/metabolism , Hepatitis/physiopathology , Iron/metabolism , Kupffer Cells/metabolism , Animals , Antimetabolites/pharmacology , Clodronic Acid/pharmacology , Female , Gene Expression/drug effects , Gene Expression/physiology , Hepcidins , Interleukin-6/genetics , Iron/pharmacology , Liposomes/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Signal Transduction/physiology
14.
Blood ; 105(12): 4861-4, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15713792

ABSTRACT

Evidence is accumulating that hepcidin, a liver regulatory peptide, could be the common pathogenetic denominator of all forms of iron overload syndromes including HFE-related hemochromatosis, the most prevalent genetic disorder characterized by inappropriate iron absorption. To understand the mechanisms whereby hepcidin controls iron homeostasis in vivo, we have analyzed the level of iron-related proteins by Western blot and immunohistochemistry in hepcidin-deficient mice, a mouse model of severe hemochromatosis. These mice showed important increased levels of duodenal cytochrome b (Dcytb), divalent metal transporter 1 (DMT1), and ferroportin compared with control mice. Interestingly, the level of ferroportin was coordinately up-regulated in the duodenum, the spleen, and the liver (predominantly in the Kupffer cells). Finally, we also evidenced a decrease of ceruloplasmin in the liver of hepcidin-deficient mice. We hypothesized that the deregulation of these proteins might be central in the pathogenesis of iron overload, providing key therapeutic targets for iron disorders.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/physiology , Gene Expression Regulation , Iron/metabolism , Animals , Blotting, Western , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cytochromes b/genetics , Cytochromes b/metabolism , Disease Models, Animal , Duodenum/metabolism , Hemochromatosis , Hepcidins , Immunohistochemistry , Iron-Binding Proteins/genetics , Liver/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Spleen/metabolism , Transgenes , Up-Regulation
15.
Blood ; 103(7): 2816-21, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14604961

ABSTRACT

Hepcidin is a 25-amino acid peptide involved in iron homeostasis in mice and humans. It is produced in the liver from a larger precursor, and it is detectable in blood and urine. In contrast to the human genome, which contains only one copy of the gene, the mouse genome contains 2 highly similar hepcidin genes, hepc1 and hepc2, which are, however, considerably divergent at the level of the corresponding mature 25-amino acid peptide. This striking observation led us to ask whether hepc1 and hepc2 performed the same biologic activity with regard to iron metabolism in the mouse. We recently described the severe iron-deficient anemia phenotype in transgenic mice overexpressing hepc1 in the liver. Here we report that, in contrast to the hepc1-transgenic mice, none of the 7 founder hepc2-transgenic animals suffered from anemia. They all developed normally with hematologic parameters similar to the nontransgenic littermates. Hepc2 transgenic mRNA level was found to be very high for all lines compared with the level of hepc1 transgene mRNA necessary to produce severe anemia. These data provide evidence that hepc2 does not act on iron metabolism like hepc1 and give clues for the identification of amino acids important for the iron-regulatory action of the mature 25-amino acid peptide.


Subject(s)
Anemia/genetics , Antimicrobial Cationic Peptides/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Founder Effect , Genome , Hematologic Diseases/genetics , Hepcidins , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , RNA/genetics , RNA/isolation & purification , RNA, Messenger/genetics , Rats , Sequence Alignment , Sequence Homology, Amino Acid , Swine
16.
Nat Genet ; 34(1): 97-101, 2003 May.
Article in English | MEDLINE | ID: mdl-12704388

ABSTRACT

Hereditary hemochromatosis is a prevalent genetic disorder of iron hyperabsorption leading to hyperferremia, tissue iron deposition and complications including cirrhosis, hepatocarcinoma, cardiomyopathy and diabetes. Most individuals affected with hereditary hemochromatosis are homozygous with respect to a missense mutation that disrupts the conformation of HFE, an atypical HLA class I molecule (ref. 1; OMIM 235200). Mice lacking Hfe or producing a C282Y mutant Hfe protein develop hyperferremia and have high hepatic iron levels. In both humans and mice, hereditary hemochromatosis is associated with a paucity of iron in reticuloendothelial cells. It has been suggested that HFE modulates uptake of transferrin-bound iron by undifferentiated intestinal crypt cells, thereby programming the absorptive capacity of enterocytes derived from these cells; however, this model is unproven and controversial. Hepcidin, a peptide hormone (HAMP; OMIM 606464), seems to act in the same regulatory pathway as HFE. Although expression of mouse Hamp is normally greater during iron overload, Hfe-/- mice have inappropriately low expression of Hamp. We crossed Hfe-/- mice with transgenic mice overexpressing Hamp and found that Hamp inhibited the iron accumulation normally observed in the Hfe-/- mice. This argues against the crypt programming model and suggests that failure of Hamp induction contributes to the pathogenesis of hemochromatosis, providing a rationale for the use of HAMP in the treatment of this disease.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Hemochromatosis/genetics , Iron Overload/genetics , Animals , Crosses, Genetic , Gene Expression , Hemochromatosis/metabolism , Hemochromatosis Protein , Hepcidins , Histocompatibility Antigens Class I/genetics , Humans , Iron Overload/prevention & control , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mutation, Missense , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
J Clin Invest ; 110(7): 1037-44, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12370282

ABSTRACT

The present study was aimed at determining whether hepcidin, a recently identified peptide involved in iron metabolism, plays a role in conditions associated with both iron overload and iron deficiency. Hepcidin mRNA levels were assessed in two models of anemia, acute hemolysis provoked by phenylhydrazine and bleeding provoked by repeated phlebotomies. Hepcidin response to hypoxia was also studied, both ex vivo, in human hepatoma cells, and in vivo. Anemia and hypoxia were associated with a dramatic decrease in liver hepcidin gene expression, which may account for the increase in iron release from reticuloendothelial cells and increase in iron absorption frequently observed in these situations. A single injection of turpentine for 16 hours induced a sixfold increase in liver hepcidin mRNA levels and a twofold decrease in serum iron. The hyposideremic effect of turpentine was completely blunted in hepcidin-deficient mice, revealing hepcidin participation in anemia of inflammatory states. These modifications of hepcidin gene expression further suggest a key role for hepcidin in iron homeostasis under various pathophysiological conditions, which may support the pharmaceutical use of hepcidin agonists and antagonists in various iron homeostasis disorders.


Subject(s)
Anemia/metabolism , Antimicrobial Cationic Peptides/genetics , Gene Expression Regulation , Hypoxia/metabolism , Inflammation/metabolism , Acute Disease , Animals , Hepcidins , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
18.
Blood Cells Mol Dis ; 29(3): 327-35, 2002.
Article in English | MEDLINE | ID: mdl-12547223

ABSTRACT

Maintaining normal iron homeostasis is essential for the organism, as both iron deficiency and iron excess are associated with cellular dysfunction. Recently, several lines of evidence have suggested that hepcidin, a peptide mainly produced by the liver, plays a major role in the control of body iron homeostasis. The subject of this paper is to summarize the advances toward the understanding of function and regulation of hepcidin in iron metabolism and to provide new data on the regulation of hepcidin gene expression by erythropoietin, the major regulator of mammalian erythropoiesis.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Iron/metabolism , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides/genetics , Erythropoietin/metabolism , Gene Expression Regulation/physiology , Hepcidins , Mice , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...