Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
Cancer Lett ; 229(1): 107-14, 2005 Nov 08.
Article in English | MEDLINE | ID: mdl-16157222

ABSTRACT

We closely mimicked the in vivo setting in which sporadic hepatocarcinoma occurs by establishing a transgenic mouse model carrying regulatable SV40 early sequences under the control of the regulatory sequences of the human antithrombin III gene that confer hepatic expression. In this system, floxed dormant oncogenic sequences became functional after excision due to adenoviral expression of Cre recombinase or the stable transgenic expression in liver of a tamoxifen-inducible Cre. Hepatic oncogene expression was switched on by both methods, leading to the development of hepatocellular carcinoma. This model could be useful for investigating the key steps of the preneoplastic process, to identify suitable targets for the testing of new therapies.


Subject(s)
Carcinoma, Hepatocellular/virology , Disease Models, Animal , Liver Neoplasms/virology , Simian virus 40/genetics , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antithrombin III/genetics , Antithrombin III/physiology , Carcinoma, Hepatocellular/physiopathology , Carcinoma, Hepatocellular/veterinary , Enzyme Induction , Expressed Sequence Tags , Gene Expression Regulation, Neoplastic , Integrases/biosynthesis , Liver Neoplasms/physiopathology , Liver Neoplasms/veterinary , Mice , Mice, Transgenic , Tamoxifen/pharmacology , Viral Proteins/biosynthesis
4.
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
5.
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
6.
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
7.
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
8.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...