Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Blood ; 130(21): 2339-2343, 2017 11 23.
Article in English | MEDLINE | ID: mdl-29021231

ABSTRACT

Lack of either bone morphogenetic protein 6 (BMP6) or the BMP coreceptor hemojuvelin (HJV) in mice leads to a similar phenotype with hepcidin insufficiency, hepatic iron loading, and extrahepatic iron accumulation in males. This is consistent with the current views that HJV is a coreceptor for BMP6 in hepatocytes. To determine whether BMP6 and HJV may also signal to hepcidin independently of each other, we intercrossed Hjv-/- and Bmp6-/- mice and compared the phenotype of animals of the F2 progeny. Loss of Bmp6 further repressed Smad signaling and hepcidin expression in the liver of Hjv-/- mice of both sexes, and led to iron accumulation in the pancreas and the heart of females. These data suggest that, in Hjv-/- females, Bmp6 can provide a signal adequate to maintain hepcidin to a level sufficient to avoid extrahepatic iron loading. We also examined the impact of Bmp6 and/or Hjv deletion on the regulation of hepcidin by inflammation. Our data show that lack of 1 or both molecules does not prevent induction of hepcidin by lipopolysaccharide (LPS). However, BMP/Smad signaling in unchallenged animals is determinant for the level of hepcidin reached after stimulation, which is consistent with a synergy between interleukin 6/STAT3 and BMP/SMAD signaling in regulating hepcidin during inflammation.


Subject(s)
Bone Morphogenetic Protein 6/deficiency , Gene Deletion , Hepcidins/metabolism , Lipopolysaccharides/pharmacology , Membrane Proteins/deficiency , Animals , Bone Morphogenetic Protein 6/metabolism , Female , GPI-Linked Proteins , Hemochromatosis Protein , Male , Membrane Proteins/metabolism , Mice, Knockout , Phenotype , Signal Transduction/drug effects
2.
Blood ; 127(19): 2327-36, 2016 05 12.
Article in English | MEDLINE | ID: mdl-26755707

ABSTRACT

Hepcidin, the main regulator of iron homeostasis, is repressed when erythropoiesis is acutely stimulated by erythropoietin (EPO) to favor iron supply to maturing erythroblasts. Erythroferrone (ERFE) has been identified as the erythroid regulator that inhibits hepcidin in stress erythropoiesis. A powerful hepcidin inhibitor is the serine protease matriptase-2, encoded by TMPRSS6, whose mutations cause iron refractory iron deficiency anemia. Because this condition has inappropriately elevated hepcidin in the presence of high EPO levels, a role is suggested for matriptase-2 in EPO-mediated hepcidin repression. To investigate the relationship between EPO/ERFE and matriptase-2, we show that EPO injection induces Erfe messenger RNA expression but does not suppress hepcidin in Tmprss6 knockout (KO) mice. Similarly, wild-type (WT) animals, in which the bone morphogenetic protein-mothers against decapentaplegic homolog (Bmp-Smad) pathway is upregulated by iron treatment, fail to suppress hepcidin in response to EPO. To further investigate whether the high level of Bmp-Smad signaling of Tmprss6 KO mice counteracts hepcidin suppression by EPO, we generated double KO Bmp6-Tmprss6 KO mice. Despite having Bmp-Smad signaling and hepcidin levels that are similar to WT mice under basal conditions, double KO mice do not suppress hepcidin in response to EPO. However, pharmacologic downstream inhibition of the Bmp-Smad pathway by dorsomorphin, which targets the BMP receptors, improves the hepcidin responsiveness to EPO in Tmprss6 KO mice. We concluded that the function of matriptase-2 is dominant over that of ERFE and is essential in facilitating hepcidin suppression by attenuating the BMP-SMAD signaling.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Erythropoietin/pharmacology , Hepcidins/metabolism , Liver/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Signal Transduction/physiology , Smad Proteins/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Hepcidins/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Serine Endopeptidases/genetics , Smad Proteins/genetics
3.
Hepatology ; 63(1): 126-37, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26406355

ABSTRACT

UNLABELLED: Hereditary hemochromatosis, which is characterized by inappropriately low levels of hepcidin, increased dietary iron uptake, and systemic iron accumulation, has been associated with mutations in the HFE, transferrin receptor-2 (TfR2), and hemojuvelin (HJV) genes. However, it is still not clear whether these molecules intersect in vivo with bone morphogenetic protein 6 (BMP6)/mothers against decapentaplegic (SMAD) homolog signaling, the main pathway up-regulating hepcidin expression in response to elevated hepatic iron. To answer this question, we produced double knockout mice for Bmp6 and ß2-microglobulin (a surrogate for the loss of Hfe) and for Bmp6 and Tfr2, and we compared their phenotype (hepcidin expression, Bmp/Smad signaling, hepatic and extrahepatic tissue iron accumulation) with that of single Bmp6-deficient mice and that of mice deficient for Hjv, alone or in combination with Hfe or Tfr2. Whereas the phenotype of Hjv-deficient females was not affected by loss of Hfe or Tfr2, that of Bmp6-deficient females was considerably worsened, with decreased Smad5 phosphorylation, compared with single Bmp6-deficient mice, further repression of hepcidin gene expression, undetectable serum hepcidin, and massive iron accumulation not only in the liver but also in the pancreas, the heart, and the kidneys. CONCLUSION: These results show that (1) BMP6 does not require HJV to transduce signal to hepcidin in response to intracellular iron, even if the loss of HJV partly reduces this signal, (2) another BMP ligand can replace BMP6 and significantly induce hepcidin expression in response to extracellular iron, and (3) BMP6 alone is as efficient at inducing hepcidin as the other BMPs in association with the HJV/HFE/TfR2 complex; they provide an explanation for the compensatory effect of BMP6 treatment on the molecular defect underlying Hfe hemochromatosis in mice.


Subject(s)
Bone Morphogenetic Protein 6/genetics , Hemochromatosis/genetics , Histocompatibility Antigens Class I/genetics , Membrane Proteins/genetics , Receptors, Transferrin/genetics , Animals , Female , GPI-Linked Proteins , Gene Deletion , Gene Expression Regulation , Hemochromatosis Protein , Iron , Mice , Mice, Inbred C57BL , Phenotype
4.
Blood ; 123(10): 1564-73, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24398330

ABSTRACT

Hepcidin controls systemic iron availability, and its excess contributes to the anemia of chronic diseases, the most prevalent anemia in hospitalized patients. We previously reported that heparins are efficient hepcidin inhibitors both in vitro and in vivo, but their anticoagulant activity limits therapeutic use. We studied nonanticoagulant heparins produced by N-acetylation and oxidation/reduction (glycol-split) that lost antithrombin-binding affinity. Four nonanticoagulant heparins inhibited hepcidin expression in hepatic HepG2 cells and primary hepatocytes. The 2 most potent ones used in mice suppressed liver hepcidin expression and serum hepcidin in 6 hours, with a significant decrease of spleen iron. This occurred also in lipopolysaccharide (LPS)-treated animals that mimic inflammation, as well as after chronic 1-week treatments, without evident adverse effects on coagulation. Heparin injections increased iron mobilization and facilitated the recovery from the anemia induced by heat-killed Brucella abortus, a model of inflammatory anemia. The heparins were used also in Bmp6(-/-) mice. A single dose of heparin reduced the already low level of hepcidin of these mice and prevented its induction by LPS. These nonanticoagulant compounds impair bone morphogenetic protein /sons of mothers against decapentaplegic signaling with no evident adverse effect in vivo, even when administered chronically. They may offer a strategy for the treatment of diseases with high hepcidin levels.


Subject(s)
Gene Expression Regulation/drug effects , Heparin/analogs & derivatives , Hepcidins/genetics , Anemia/chemically induced , Anemia/drug therapy , Anemia/genetics , Animals , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , Cell Line , Dermatan Sulfate/pharmacology , Dose-Response Relationship, Drug , Female , Hep G2 Cells , Heparin/administration & dosage , Heparin/pharmacology , Hepcidins/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/metabolism , Inhibitor of Differentiation Protein 1/genetics , Iron/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Knockout , Promoter Regions, Genetic , Spleen/drug effects , Spleen/metabolism , Time Factors , Transcriptional Activation/drug effects
6.
Hepatology ; 59(2): 683-94, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23907767

ABSTRACT

UNLABELLED: Gender-related disparities in the regulation of iron metabolism may contribute to the differences exhibited by men and women in the progression of chronic liver diseases associated with reduced hepcidin expression, e.g., chronic hepatitis C, alcoholic liver disease, or hereditary hemochromatosis. However, their mechanisms remain poorly understood. In this study we took advantage of the major differences in hepcidin expression and tissue iron loading observed between Bmp6-deficient male and female mice to investigate the mechanisms underlying this sexual dimorphism. We found that testosterone robustly represses hepcidin transcription by enhancing Egfr signaling in the liver and that selective epidermal growth factor receptor (Egfr) inhibition by gefitinib (Iressa) in males markedly increases hepcidin expression. In males, where the suppressive effects of testosterone and Bmp6-deficiency on hepcidin expression are combined, hepcidin is more strongly repressed than in females and iron accumulates massively not only in the liver but also in the pancreas, heart, and kidneys. CONCLUSION: Testosterone-induced repression of hepcidin expression becomes functionally important during homeostatic stress from disorders that result in iron loading and/or reduced capacity for hepcidin synthesis. These findings suggest that novel therapeutic strategies targeting the testosterone/EGF/EGFR axis may be useful for inducing hepcidin expression in patients with iron overload and/or chronic liver diseases.


Subject(s)
ErbB Receptors/metabolism , Hepcidins/metabolism , Iron/metabolism , Liver/metabolism , Sex Factors , Signal Transduction/physiology , Testosterone/metabolism , Animals , Bone Morphogenetic Protein 6/deficiency , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , Cation Transport Proteins/metabolism , Female , Homeostasis/physiology , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Myocardium/metabolism , Pancreas/metabolism , Smad Proteins/metabolism
8.
Blood ; 120(2): 431-9, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22611157

ABSTRACT

Anemia is very common in patients suffering from infections or chronic inflammation and can add substantially to the morbidity of the underlying disease. It is mediated by excessive production of the iron-regulatory peptide hepcidin, but the signaling pathway responsible for hepcidin up-regulation in the inflammatory context is still not understood completely. In the present study, we show that activin B has an unexpected but crucial role in the induction of hepcidin by inflammation. There is a dramatic induction of Inhbb mRNA, encoding the activin ß(B)-subunit, in the livers of mice challenged with lipopolysaccharide, slightly preceding an increase in Smad1/5/8 phosphorylation and Hamp mRNA. Activin B also induces Smad1/5/8 phosphorylation in human hepatoma-derived cells and, synergistically with IL-6 and STAT-3 signaling, up-regulates hepcidin expression markedly, an observation confirmed in mouse primary hepatocytes. Pretreatment with a bone morphogenic protein type I receptor inhibitor showed that the effect of activin B on hepcidin expression is entirely attributable to its effect on bone morphogenetic protein signaling, most likely via activin receptor-like kinase 3. Activin B is therefore a novel and specific target for the treatment of anemia of inflammation.


Subject(s)
Activins/biosynthesis , Antimicrobial Cationic Peptides/genetics , Inflammation/metabolism , Smad Proteins/metabolism , Activins/genetics , Animals , Bone Morphogenetic Protein 6/deficiency , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepcidins , Humans , Inflammation/etiology , Interleukin-6/deficiency , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Up-Regulation/drug effects
10.
Blood Adv ; 8(8): 1898-1907, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38241484

ABSTRACT

ABSTRACT: Iron plays a major role in the deterioration of ß-thalassemia. Indeed, the high levels of transferrin saturation and iron delivered to erythroid progenitors are associated with production of α-globin precipitates that negatively affect erythropoiesis. Matriptase-2/TMPRSS6, a membrane-bound serine protease expressed in hepatocytes, negatively modulates hepcidin production and thus is a key target to prevent iron overload in ß-thalassemia. To address safety concerns raised by the suppression of Tmprss6 by antisense oligonucleotides or small interfering RNA, we tested a fully human anti-matriptase-2 antibody, RLYB331, which blocks the protease activity of matriptase-2. When administered weekly to Hbbth3/+ mice, RLYB331 induced hepcidin expression, reduced iron loading, prevented the formation of toxic α-chain/heme aggregates, reduced ros oxygen species formation, and improved reticulocytosis and splenomegaly. To increase the effectiveness of RLYB331 in ß-thalassemia treatment even further, we administered RLYB331 in combination with RAP-536L, a ligand-trapping protein that contains the extracellular domain of activin receptor type IIB and alleviates anemia by promoting differentiation of late-stage erythroid precursors. RAP-536L alone did not prevent iron overload but significantly reduced apoptosis in the erythroid populations of the bone marrow, normalized red blood cell counts, and improved hemoglobin and hematocrit levels. Interestingly, the association of RLYB331 with RAP-536L entirely reversed the ß-thalassemia phenotype in Hbbth3/+ mice and simultaneously corrected iron overload, ineffective erythropoiesis, splenomegaly, and hematological parameters, suggesting that a multifunctional molecule consisting of the fusion of RLYB331 with luspatercept (human version of RAP-536L) would allow administration of a single medication addressing simultaneously the different pathophysiological aspects of ß-thalassemia.


Subject(s)
Iron Overload , Membrane Proteins , Serine Endopeptidases , beta-Thalassemia , Mice , Humans , Animals , Hepcidins , beta-Thalassemia/genetics , Splenomegaly , Iron Overload/drug therapy , Iron/metabolism
12.
Blood ; 114(12): 2515-20, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19622835

ABSTRACT

Impaired regulation of hepcidin expression in response to iron loading appears to be the pathogenic mechanism for hereditary hemochromatosis. Iron normally induces expression of the BMP6 ligand, which, in turn, activates the BMP/Smad signaling cascade directing hepcidin expression. The molecular function of the HFE protein, involved in the most common form of hereditary hemochromatosis, is still unknown. We have used Hfe-deficient mice of different genetic backgrounds to test whether HFE has a role in the signaling cascade induced by BMP6. At 7 weeks of age, these mice have accumulated iron in their liver and have increased Bmp6 mRNA and protein. However, in contrast to mice with secondary iron overload, levels of phosphorylated Smads 1/5/8 and of Id1 mRNA, both indicators of BMP signaling, are not significantly higher in the liver of these mice than in wild-type livers. As a consequence, hepcidin mRNA levels in Hfe-deficient mice are similar or marginally reduced, compared with 7-week-old wild-type mice. The inappropriately low levels of Id1 and hepcidin mRNA observed at weaning further suggest that Hfe deficiency triggers iron overload by impairing hepatic Bmp/Smad signaling. HFE therefore appears to facilitate signal transduction induced by the BMP6 ligand.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Histocompatibility Antigens Class I/physiology , Membrane Proteins/physiology , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Blotting, Western , Bone Morphogenetic Protein 6/genetics , Hemochromatosis Protein , Hepcidins , Immunoenzyme Techniques , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Iron/administration & dosage , Iron Overload , Liver/cytology , Liver/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Smad1 Protein/genetics , Smad5 Protein/genetics , Smad8 Protein/genetics
13.
Haematologica ; 96(2): 199-203, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20952515

ABSTRACT

BACKGROUND: The bone morphogenetic protein BMP6 regulates hepcidin production by the liver. However, it is not yet known whether BMP6 derives from the liver itself or from other sources such as the small intestine, as has been recently suggested. This study was aimed at investigating the source of BMP6 further. DESIGN AND METHODS: We used three different strains of mice (C57BL/6, DBA/2, and 129/Sv) with iron overload induced either by an iron-enriched diet or by inactivation of the Hfe gene. We examined Bmp6 expression at both the mRNA (by quantitative PCR) and protein (by immunohistochemistry and Western blotting analyses) levels. RESULTS: We showed that iron overload induces Bmp6 mRNA expression in the liver but not in the duodenum of these mice. Bmp6 is also detected by immunohistochemistry in liver tissue sections of mice with iron overload induced either by an iron-enriched diet or by inactivation of the Hfe gene, but not in liver tissue sections from iron-loaded Bmp6-deficient mice. Bmp6 in the duodenum was below immunodetection threshold, thus confirming quantitative PCR data. Lack of specificity of available antibodies together with slight heterogeneity between 129 substrains may account for the differences with previously published data. CONCLUSIONS: Our data strongly support the importance of liver BMP6 for regulation of iron metabolism. Indeed, they demonstrate that intestinal Bmp6 expression is modulated by iron neither at the mRNA nor at the protein level.


Subject(s)
Bone Morphogenetic Protein 6/physiology , Duodenum/metabolism , Iron Overload , Iron/metabolism , Liver/metabolism , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Blotting, Western , Duodenum/cytology , Hemochromatosis Protein , Hepcidins , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Immunoenzyme Techniques , Intestinal Mucosa/metabolism , Intestines/cytology , Liver/cytology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL