Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Proc Natl Acad Sci U S A ; 118(40)2021 10 05.
Article in English | MEDLINE | ID: mdl-34593646

ABSTRACT

Iron is an essential biometal, but is toxic if it exists in excess. Therefore, iron content is tightly regulated at cellular and systemic levels to meet metabolic demands but to avoid toxicity. We have recently reported that adaptive thermogenesis, a critical metabolic pathway to maintain whole-body energy homeostasis, is an iron-demanding process for rapid biogenesis of mitochondria. However, little information is available on iron mobilization from storage sites to thermogenic fat. This study aimed to determine the iron-regulatory network that underlies beige adipogenesis. We hypothesized that thermogenic stimulus initiates the signaling interplay between adipocyte iron demands and systemic iron liberation, resulting in iron redistribution into beige fat. To test this hypothesis, we induced reversible activation of beige adipogenesis in C57BL/6 mice by administering a ß3-adrenoreceptor agonist CL 316,243 (CL). Our results revealed that CL stimulation induced the iron-regulatory protein-mediated iron import into adipocytes, suppressed hepcidin transcription, and mobilized iron from the spleen. Mechanistically, CL stimulation induced an acute activation of hypoxia-inducible factor 2-α (HIF2-α), erythropoietin production, and splenic erythroid maturation, leading to hepcidin suppression. Disruption of systemic iron homeostasis by pharmacological HIF2-α inhibitor PT2385 or exogenous administration of hepcidin-25 significantly impaired beige fat development. Our findings suggest that securing iron availability via coordinated interplay between renal hypoxia and hepcidin down-regulation is a fundamental mechanism to activate adaptive thermogenesis. It also provides an insight into the effects of adaptive thermogenesis on systemic iron mobilization and redistribution.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hepcidins/metabolism , Iron/metabolism , Thermogenesis/physiology , Adipocytes/metabolism , Adipocytes, Beige/metabolism , Adipogenesis/physiology , Adipose Tissue, Beige/metabolism , Animals , Down-Regulation/physiology , Erythropoietin/metabolism , Homeostasis/physiology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Signal Transduction/physiology
2.
J Neurochem ; 164(6): 847-857, 2023 03.
Article in English | MEDLINE | ID: mdl-36562685

ABSTRACT

Astrocytes are thought to play a crucial role in brain iron homeostasis. How they accomplish this regulation in vivo is unclear. In a recent transcriptomic analysis, we showed that polysomal Ftl1 and Fth1 mRNAs, encoding the ferritin light (Ftl) and heavy (Fth) chains that assemble into ferritin, a critical complex for iron storage and reduction, are enriched in perisynaptic astrocytic processes as compared to astrocytic soma. These data suggested that ferritin translation plays a specific role at the perisynaptic astrocytic interface and is tighly regulated by local translation. Here, we used our recently described AstroDot 3D in situ methodology to study the density and localization of ferritin mRNAs in astrocytes in the hippocampus in three different contexts in which local or systemic iron overload has been documented: aging, the hepcidin knock-out mouse model of hemochromatosis and the APP/PS1dE9 mouse model of Alzheimer's disease (AD). Our results showed that in wild type mice, Fth1 mRNA density was higher than Ftl1 and that both mRNAs were mostly distributed in astrocyte fine processes. Aging and absence of hepcidin caused an increased Fth1/Ftl1 ratio in astrocytes and in the case of aging, led to a redistribution of Fth1 mRNAs in astrocytic fine processes. In contrast, in AD mice, we observed a lower Fth1/Ftl1 ratio. Fth1 mRNAs became more somatic and Ftl1 mRNAs redistributed in large processes of astrocytes proximal to Amyloid beta (Aß) deposits. Hence, we propose that regulation of ferritin mRNA density and distribution in astrocytes contribute to iron homeostasis in physiology and pathophysiology.


Subject(s)
Alzheimer Disease , Ferritins , Mice , Animals , Ferritins/genetics , Ferritins/metabolism , Hepcidins , Astrocytes/metabolism , Amyloid beta-Peptides , RNA, Messenger , Iron/metabolism , Alzheimer Disease/pathology , Mice, Knockout , Hippocampus/metabolism
3.
J Pathol ; 241(1): 104-114, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27741349

ABSTRACT

Iron is both an essential and a potentially toxic element, and its systemic homeostasis is controlled by the iron hormone hepcidin. Hepcidin binds to the cellular iron exporter ferroportin, causes its degradation, and thereby diminishes iron uptake from the intestine and the release of iron from macrophages. Given that hepcidin-resistant ferroportin mutant mice show exocrine pancreas dysfunction, we analysed pancreata of aging hepcidin knockout (KO) mice. Hepcidin and Hfe KO mice were compared with wild-type (WT) mice kept on standard or iron-rich diets. Twelve-month-old hepcidin KO mice were subjected to daily minihepcidin PR73 treatment for 1 week. Six-month-old hepcidin KO mice showed cytoplasmic acinar iron overload and mild pancreatitis, together with elevated expression of the iron uptake mediators DMT1 and Zip14. Acinar atrophy, massive macrophage infiltration, fatty changes and pancreas fibrosis were noted in 1-year-old hepcidin KO mice. As an underlying mechanism, 6-month-old hepcidin KO mice showed increased pancreatic oxidative stress, with elevated DNA damage, apoptosis and activated nuclear factor-κB (NF-κB) signalling. Neither iron overload nor pancreatic damage was observed in WT mice fed iron-rich diet or in Hfe KO mice. Minihepcidin application to hepcidin KO mice led to an improvement in general health status and to iron redistribution from acinar cells to macrophages. It also resulted in decreased NF-κB activation and reduced DNA damage. In conclusion, loss of hepcidin signalling in mice leads to iron overload-induced chronic pancreatitis that is not seen in situations with less severe iron accumulation. The observed tissue injury can be reversed by hepcidin supplementation. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Acinar Cells/metabolism , Hepcidins/deficiency , Iron Overload/complications , Pancreatitis, Chronic/etiology , Animals , Apoptosis/physiology , Cytoplasm/metabolism , Disease Models, Animal , Hepcidins/genetics , Hepcidins/physiology , Iron Overload/metabolism , Iron Overload/pathology , Macrophages/pathology , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Oxidative Stress/physiology , Pancreas/ultrastructure , Pancreatitis, Chronic/metabolism , Pancreatitis, Chronic/pathology
4.
FASEB J ; 30(1): 252-61, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26370847

ABSTRACT

The amount of iron in the diet directly influences the composition of the microbiota. Inversely, the effects of the microbiota on iron homeostasis have been little studied. So, we investigate whether the microbiota itself may alter host iron sensing. Duodenal cytochrome b and divalent metal transporter 1, involved in apical iron uptake, are 8- and 10-fold, respectively, more abundant in the duodenum of germ-free (GF) mice than in mice colonized with a microbiota. In contrast, the luminal exporter ferroportin is 2-fold less abundant in GF. The overall signature of microbiota on iron-related proteins is similar in the colon. The colonization does not modify systemic parameters as plasma transferrin saturation (20%), plasma ferritin (150 ng/L), and liver (85 µg/g) iron load. Commensal organisms (Bacteroides thetaiotaomicron VPI-5482 and Faecalibacterium prausnitzii A2-165) and a probiotic strain (Streptococcus thermophilus LMD-9) led to up to 12-fold induction of ferritin in colon. Our data suggest that the intestinal cells of GF mice are depleted of iron and that following colonization, the epithelial cells favor iron storage. This study is the first to demonstrate that gut microbes induce a specific iron-related protein signature, highlighting new aspects of the crosstalk between the microbiota and the intestinal epithelium.


Subject(s)
Cation Transport Proteins/metabolism , Intestinal Mucosa/metabolism , Iron/metabolism , Microbiota , Animals , Cation Transport Proteins/genetics , Colon/metabolism , Colon/microbiology , Cytochromes b/genetics , Cytochromes b/metabolism , Duodenum/metabolism , Duodenum/microbiology , Ferritins/blood , Intestinal Mucosa/microbiology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL
5.
Blood ; 124(3): 441-4, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-24904115

ABSTRACT

Iron is an essential element required for development and survival of all living organisms. In fetuses, maternofetal iron transfer across the placenta is essential for growth and development. In neonates, efficient intestinal iron absorption is required to scavenge as much iron as possible from the low-iron-content milk. During these periods, efficient iron mobilization is ensured by the downregulation of the iron regulatory hormone hepcidin by as-yet uncharacterized molecular mechanisms. Here we demonstrate that the recently described hepcidin repressor-the serine protease matriptase-2 (encoded by Tmprss6)-is responsible for this repression throughout development, with its deficiency leading to increased hepcidin levels triggering iron deficiency and anemia starting in utero. This result might have implications for a better understanding of iron homeostasis during early development in iron-refractory iron deficiency anemia patients, who present with microcytic anemia caused by hyperhepcidinemia, and of questions about the role of matriptase-2 in human neonates.


Subject(s)
Hepcidins/metabolism , Iron/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Anemia, Iron-Deficiency/etiology , Animals , Bone Morphogenetic Protein 6/deficiency , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , Down-Regulation , Female , Fetus/metabolism , GPI-Linked Proteins , Hemochromatosis Protein , Homeostasis , Humans , Iron Deficiencies , Liver/metabolism , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Pregnancy , Serine Endopeptidases/deficiency , Serine Endopeptidases/genetics , Signal Transduction
6.
Protein Expr Purif ; 115: 11-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26169129

ABSTRACT

Hepcidin, a 25-amino-acid and highly disulfide bonded antimicrobial peptide, is the central regulator of iron homeostasis. This hormone is expressed in response to iron and inflammation and interacts with ferroportin1 (FPN1), the only known iron exporter in vertebrates, inducing its internalization and degradation. Thus, the export of iron from cells to plasma will be significantly diminished. Thereby, hepcidin has become the target of intense research studies due to its profound biomedical significance. This study describes the functional expression of recombinant camel hepcidin in Escherichia coli. Biologically active recombinant camel hepcidin was obtained thanks to the production of a hepcidin-thioredoxin fusion protein (TRX-HepcD) and a purified camel hepcidin, with an extra methionine at the N-terminus, was obtained after enterokinase cleavage of the fusion protein. Presence of the four disulfide bridges was verified using MALDI-ToF spectrometry. The recombinant camel hepcidin was compared to related synthetic bioactive peptides, including human hepcidin, and was found equally able to promote ferroportin degradation of mouse macrophages. Furthermore, camel hepcidins exhibits a high capacity to inhibit the growth of Leishmania major promastigotes. These results proved that production of functional camel hepcidin can be achieved in E. coli, this is a major interest for the production of cysteine rich peptides or proteins that can be purified under their functional form without the need of a refolding process.


Subject(s)
Cation Transport Proteins/metabolism , Hepcidins/isolation & purification , Hepcidins/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Amino Acid Sequence , Animals , Camelus/genetics , Cation Transport Proteins/chemistry , Disulfides/chemistry , Escherichia coli/genetics , Hepcidins/chemistry , Hepcidins/genetics , Humans , Macrophages/chemistry , Macrophages/metabolism , Mice , Molecular Sequence Data , Plasmids , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
7.
J Hepatol ; 61(3): 633-41, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24816174

ABSTRACT

BACKGROUND & AIMS: Hepcidin is the central regulator of iron homeostasis and altered hepcidin signalling results in both hereditary and acquired iron overload. While the association between iron overload and development of end-stage liver disease is well established, the underlying mechanisms are largely unknown. To improve that, we analysed hepcidin knockout (KO) mice as a model of iron overload-associated liver disease. METHODS: Hepcidin wild type (WT) and KO mice fed with 3% carbonyl iron-containing diet starting at one month of age were compared to age-matched animals kept on standard chow. Liver histology and serum parameters were used to assess the extent of liver injury and fibrosis. Iron distribution was determined by subcellular fractionation and electron microscopy. RESULTS: Among mice kept on iron-rich diet, 6 months old hepcidin KO mice (vs. WT) displayed profound hepatic iron overload (3,186 ± 411 vs. 1,045 ± 159 µg/mg tissue, p<0.005), elevated liver enzymes (ALT: KO 128 ± 6, WT 56 ± 5 IU/L, p<0.05), mild hepatic inflammation and hepatocellular apoptosis. Twelve, but not six months old KO mice fed with iron-rich diet developed moderate liver fibrosis. The liver injury was accompanied by a marked lysosomal iron overload and lysosomal fragility with release of cathepsin B into the cytoplasm. Increased p62 levels and autofluorescent iron complexes suggested impaired protein degradation. As a mechanism leading to lysosomal iron overload, the autophagy (lysosomal influx) was increased. CONCLUSIONS: Hepcidin KO mice represent a novel model of iron overload-related liver diseases and implicate lysosomal injury as a crucial event in iron toxicity.


Subject(s)
Hepcidins/deficiency , Iron, Dietary/adverse effects , Iron/metabolism , Liver Cirrhosis/etiology , Lung Injury/etiology , Lysosomes/metabolism , Animals , Apoptosis/physiology , Disease Models, Animal , Hepatic Stellate Cells/pathology , Hepatic Stellate Cells/physiology , Hepcidins/genetics , Hepcidins/physiology , Homeostasis/physiology , Liver/enzymology , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/physiopathology , Lung Injury/metabolism , Lung Injury/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Time Factors
8.
Blood ; 119(2): 587-90, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22128145

ABSTRACT

Hereditary hemochromatosis (HH) is a highly prevalent genetic disorder characterized by excessive parenchymal iron accumulation leading to liver cirrhosis, diabetes, and in some cases hepatocellular carcinoma. HH is caused by mutations in the genes encoding upstream regulators of hepcidin or more rarely in the hepcidin gene itself. A deficit in hepcidin results in intestinal iron hyperabsorption; however, the local effectors mediating the up-regulation of iron absorption genes are unknown. We hypothesized that HIF-2 could mediate high iron absorption rates in HH. We generated Hepc(-/-) mice (a murine model of hemochromatosis) lacking HIF-2 in the intestine and showed that duodenal HIF-2 was essential for the up-regulation of genes involved in intestinal iron import and the consequent iron accumulation in the liver and pancreas. This study highlights a role of HIF-2 in the dysregulation of iron absorption and chronic iron accumulation, as observed in patients with hemochromatosis.


Subject(s)
Antimicrobial Cationic Peptides/physiology , Basic Helix-Loop-Helix Transcription Factors/physiology , Enterocytes/metabolism , Intestinal Mucosa/metabolism , Iron Overload/prevention & control , Animals , Blotting, Western , Duodenum/metabolism , Duodenum/pathology , Enterocytes/pathology , Female , Hemochromatosis/etiology , Hepcidins , Immunoenzyme Techniques , Integrases/metabolism , Intestines/pathology , Iron Overload/etiology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction
9.
Blood ; 117(2): 647-50, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-20940420

ABSTRACT

Hepcidin is the master regulator of iron homeostasis. In the liver, iron-dependent hepcidin activation is regulated through Bmp6 and its membrane receptor hemojuvelin (Hjv), whereas, in response to iron deficiency, hepcidin repression seems to be controlled by a pathway involving the serine protease matriptase-2 (encoded by Tmprss6). To determine the relationship between Bmp6 and matriptase-2 pathways, Tmprss6(-/-) mice (characterized by increased hepcidin levels and anemia) and Bmp6(-/-) mice (exhibiting severe iron overload because of hepcidin deficiency) were intercrossed. We showed that loss of Bmp6 decreased hepcidin levels; increased hepatic iron; and, importantly, corrected hematologic abnormalities in Tmprss6(-/-) mice. This finding suggests that elevated hepcidin levels in patients with familial iron-refractory, iron-deficiency anemia are the result of excess signaling through the Bmp6/Hjv pathway.


Subject(s)
Anemia, Iron-Deficiency/metabolism , Bone Morphogenetic Protein 6/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Signal Transduction/physiology , Animals , Antimicrobial Cationic Peptides/metabolism , Female , Hepcidins , Iron/metabolism , Iron, Dietary/metabolism , Liver/metabolism , Mice , Mice, Knockout
10.
Sci Rep ; 13(1): 12794, 2023 08 07.
Article in English | MEDLINE | ID: mdl-37550331

ABSTRACT

The role of iron in the two major sites of adaptive thermogenesis, namely the beige inguinal (iWAT) and brown adipose tissues (BAT) has not been fully understood yet. Body iron levels and distribution is controlled by the iron regulatory peptide hepcidin. Here, we explored iron homeostasis and thermogenic activity in brown and beige fat in wild-type and iron loaded Hepcidin KO mice. Hepcidin-deficient mice displayed iron overload in both iWAT and BAT, and preferential accumulation of ferritin in stromal cells compared to mature adipocytes. In contrast to BAT, the iWAT of Hepcidin KO animals featured with defective thermogenesis evidenced by an altered beige signature, including reduced UCP1 levels and decreased mitochondrial respiration. This thermogenic modification appeared cell autonomous and persisted after a 48 h-cold challenge, a potent trigger of thermogenesis, suggesting compromised de novo adipogenesis. Given that WAT browning occurs in both mice and humans, our results provide physiological results to interrogate the thermogenic capacity of patients with iron overload disorders.


Subject(s)
Adipogenesis , Hepcidins , Animals , Mice , Adipose Tissue, Brown , Adipose Tissue, White , Hepcidins/genetics , Iron , Mice, Inbred C57BL , Thermogenesis , Uncoupling Protein 1/genetics
11.
J Nutr Biochem ; 111: 109175, 2023 01.
Article in English | MEDLINE | ID: mdl-36223834

ABSTRACT

Increased body iron stores and inflammation in adipose tissue have been implicated in the pathogenesis of insulin resistance (IR) and type 2 diabetes mellitus. However, the underlying basis of these associations is unclear. To attempt to investigate this, we studied the development of IR and associated inflammation in adipose tissue in the presence of increased body iron stores. Male hepcidin knock-out (Hamp1-/-) mice, which have increased body iron stores, and wild-type (WT) mice were fed a high-fat diet (HFD) for 12 and 24 weeks. Development of IR and metabolic parameters linked to this, insulin signaling in various tissues, and inflammation and iron-related parameters in visceral adipose tissue were studied in these animals. HFD-feeding resulted in impaired glucose tolerance in both genotypes of mice. In response to the HFD for 24 weeks, Hamp1-/- mice gained less body weight and developed less systemic IR than corresponding WT mice. This was associated with less lipid accumulation in the liver and decreased inflammation and lipolysis in the adipose tissue in the knock-out mice, than in the WT animals. Fewer macrophages infiltrated the adipose tissue in the knockout mice than in wild-type mice, with these macrophages exhibiting a predominantly anti-inflammatory (M2-like) phenotype and indirect evidence of a possible lowered intracellular iron content. The absence of hepcidin was thus associated with attenuated inflammation in the adipose tissue and increased whole-body insulin sensitivity, suggesting a role for it in these processes.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Male , Mice , Animals , Insulin Resistance/physiology , Diet, High-Fat/adverse effects , Hepcidins/metabolism , Diabetes Mellitus, Type 2/metabolism , Mice, Inbred C57BL , Adipose Tissue/metabolism , Inflammation/metabolism , Insulin/metabolism , Mice, Knockout , Iron/metabolism
12.
Blood Cells Mol Dis ; 47(1): 46-9, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21514188

ABSTRACT

Based on a microarray study, which demonstrated the upregulation of Sp6 transcriptional factor in iron deficient rat duodenum, we reasoned that SP6 could regulate iron absorption by controlling the expression of iron absorption genes (Collins,2006). For that, we generated Sp6 specific intestinal knockout mice. Our data suggest a lack of transcriptional upregulation of Sp6 in mice in conditions where iron absorption is promoted (phlebotomy, iron deficiency, hpx mouse), and an absence of iron-related phenotype in Sp6 intestinal knockout model. We propose that other Sp6 orthologues may be involved in the genetic response to increase iron absorption, possibly in co-operation with hypoxia inducible factor 2 alpha (HIF-2α)-a newly discovered regulator of iron absorption.


Subject(s)
Intestinal Mucosa/metabolism , Iron/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Phenotype , Anemia, Iron-Deficiency/genetics , Anemia, Iron-Deficiency/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cation Transport Proteins/genetics , Gene Expression Regulation , Iron Deficiencies , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , Transcriptional Activation/genetics
13.
FASEB J ; 24(6): 2093-103, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20124431

ABSTRACT

Erythropoietic activity is known to affect iron homeostasis through regulation of the liver iron regulatory hormone hepcidin. To identify new factors secreted by the erythroblasts that could influence hepcidin synthesis, we set up a coculture model. HuH7 hepatoma cells cocultured with primary human erythroblasts or erythroleukemic UT7 cells presented a 20- to 35-fold increase of hepcidin gene expression. This induction was fully blunted in the presence of a neutralizing oncostatin M antibody, demonstrating that this cytokine, belonging to the IL-6 family of cytokines, was responsible for increased levels of hepcidin expression. We further demonstrated that recombinant oncostatin M induced a dramatic transcriptional increase of hepcidin in HuH7 cells through specific activation of the STAT pathway. Hepcidin induction by oncostatin M was also observed in hepatocytes in primary culture and is believed to be cell specific since no induction was found in isolated bone marrow cells, macrophagic, stromal, and lymphoma-derived cell lines, nor in erythroblasts. Finally, we show that oncostatin M administration in vivo increases hepcidin expression and leads to significantly decreased serum iron levels. This work identifies a new potent inducer of hepcidin expression in the liver and supports a role for modulators of oncostatin M signaling pathway in treating iron disorders.


Subject(s)
Anti-Bacterial Agents/metabolism , Antimicrobial Cationic Peptides/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Erythroblasts/metabolism , Iron/metabolism , Liver Neoplasms/metabolism , Oncostatin M/pharmacology , Animals , Antineoplastic Agents/antagonists & inhibitors , Antineoplastic Agents/immunology , Blotting, Western , Carcinoma, Hepatocellular/pathology , Cells, Cultured , Coculture Techniques , Hepcidins , Humans , Interleukin-6/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oncostatin M/antagonists & inhibitors , Oncostatin M/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT Transcription Factors/metabolism
14.
Haematologica ; 95(3): 501-4, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19773263

ABSTRACT

Hepcidin, a circulating regulatory hormone peptide produced by hepatocytes, functions as the master regulator of cellular iron export by controlling the amount of ferroportin, an iron exporter present on the basolateral surface of intestinal enterocytes and macrophages. Hepcidin binding to ferroportin induces its internalization and degradation, resulting in cellular iron retention and decreased iron export. Whether hepatocytes express ferroportin that could be targeted by hepcidin has remained a subject of debate. Here, we describe a hepatocyte culture system expressing high levels of ferroportin, and demonstrate that both endogenously secreted and synthetic hepcidin are fully active in down-regulating membrane-associated ferroportin. In agreement with this result, ferroportin is stabilized in liver hepatocytes of hepcidin-deficient mice and accumulates in periportal areas, supporting the centrolobular iron deposition observed in these mice. In conclusion, we show that hepcidin can trigger ferroportin degradation in hepatocytes, which must be taken into account when considering hepcidin therapeutics.


Subject(s)
Antimicrobial Cationic Peptides/physiology , Cation Transport Proteins/metabolism , Hepatocytes/metabolism , Iron/metabolism , Animals , Blotting, Western , Cation Transport Proteins/genetics , Female , Hepatocytes/cytology , Hepcidins , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
Blood Adv ; 4(16): 3853-3863, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32810223

ABSTRACT

Iron is required for the oxidative response of neutrophils to allow the production of reactive oxygen species (ROS). However, neutrophil function may be severely altered in conditions of iron overload, as observed in chronically transfused patients. Therefore, a tight regulation of neutrophil iron homeostasis seems to be critical for avoiding iron toxicity. Hepcidin is the key iron regulator in organisms; however, no studies have investigated its role in maintaining neutrophil iron homeostasis or characterized neutrophil function in patients with hereditary hemochromatosis (HH), a common iron overload genetic disorder that results from a defect in hepcidin production. To explore these issues, we studied 2 mouse models of iron overload: an experimentally induced iron overload model (EIO), in which hepcidin is increased, and a genetic HH model of iron overload with a deletion of hepatic hepcidin. We found that iron-dependent increase of hepatic hepcidin results in neutrophil intracellular iron trapping and consecutive defects in oxidative burst activity. In contrast, in both HH mouse models and HH patients, the lack of hepcidin expression protects neutrophils from toxic iron accumulation. Moreover, systemic iron overload correlated with a surprising neutrophil priming and resulted in a more powerful oxidative burst. Indeed, important factors in neutrophil priming and activation, such as tumor necrosis factor α (TNF-α), VCAM-1, and ICAM-1 are increased in the plasma of HH patients and are associated with an increase in HH neutrophil phagocytosis capacity and a decrease in L-selectin surface expression. This is the first study to characterize neutrophil iron homeostasis and associated functions in patients with HH.


Subject(s)
Hemochromatosis , Iron Overload , Animals , Hemochromatosis/genetics , Hepcidins/genetics , Humans , Iron , Mice , Neutrophils
16.
Haematologica ; 94(6): 765-72, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19454495

ABSTRACT

BACKGROUND: The circulating hormone hepcidin plays a central role in iron homeostasis. Our goal was to establish an ex vivo iron-sensing model and to characterize the molecular mechanisms linking iron to hepcidin. DESIGN AND METHODS: Murine hepatocytes were isolated by the collagenase method, either from wild type or HFE knockout mice, and cultured 42 h without serum before treatments. RESULTS: After 42 h of serum-free culture, hepcidin gene expression was undetectable in the hepatocytes. Hepcidin gene expression could, however, be re-activated by an additional 24 h of incubation with 10% serum. Interestingly, addition of 30 microM holotransferrin consistently increased serum-dependent hepcidin levels 3- to 5-fold. The effects of serum and serum+holotransferrin were direct, transcriptional, independent of de novo protein synthesis and required the presence of bone morphogenetic protein. Transferrin receptor-2 activation by its ligand holotransferrin led to extracellular signal regulated kinase (ERK)/mitogen activated protein kinase pathway stimulation and the ERK specific inhibitor U0-126 blunted holotransferrin-mediated induction of hepcidin. ERK activation by holotransferrin provoked increased levels of phospho-Smad1/5/8 highlighting cross-talk between the bone morphogenetic protein/hemojuvelin and ERK1/2 pathways. Finally, we demonstrated, using hepatocytes isolated from Hfe(-/-) mice, that HFE was not critical for the hepcidin response to holotransferrin but important for basal hepcidin expression. CONCLUSIONS: We demonstrate that hepatocytes are liver iron-sensor cells and that transferrin receptor-2, by signaling through the ERK1/2 pathway, and bone morphogenetic protein/hemojuvelin, by signaling through the Smad pathways, coordinately regulate the iron-sensing machinery linking holotransferrin to hepcidin.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Bone Morphogenetic Proteins/genetics , Hepatocytes/drug effects , Membrane Proteins/genetics , Mitogen-Activated Protein Kinases/metabolism , Transferrin/pharmacology , Animals , Blotting, Northern , Bone Morphogenetic Proteins/metabolism , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Female , GPI-Linked Proteins , Gene Expression Regulation/drug effects , Hemochromatosis Protein , Hepatocytes/cytology , Hepatocytes/metabolism , Hepcidins , Humans , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Transferrin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors
17.
FEBS J ; 275(15): 3793-803, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18557934

ABSTRACT

Hepcidin is a liver produced cysteine-rich peptide hormone that acts as the central regulator of body iron metabolism. Hepcidin is synthesized under the form of a precursor, prohepcidin, which is processed to produce the biologically active mature 25 amino acid peptide. This peptide is secreted and acts by controlling the concentration of the membrane iron exporter ferroportin on intestinal enterocytes and macrophages. Hepcidin binds to ferroportin, inducing its internalization and degradation, thus regulating the export of iron from cells to plasma. The aim of the present study was to develop a novel method to produce human and mouse recombinant hepcidins, and to compare their biological activity towards their natural receptor ferroportin. Hepcidins were expressed in Escherichia coli as thioredoxin fusion proteins. The corresponding peptides, purified after cleavage from thioredoxin, were properly folded and contained the expected four-disulfide bridges without the need of any renaturation or oxidation steps. Human and mouse hepcidins were found to be biologically active, promoting ferroportin degradation in macrophages. Importantly, biologically inactive aggregated forms of hepcidin were observed depending on purification and storage conditions, but such forms were unrelated to disulfide bridge formation.


Subject(s)
Antimicrobial Cationic Peptides/biosynthesis , Iron-Regulatory Proteins/biosynthesis , Animals , Antimicrobial Cationic Peptides/isolation & purification , Antimicrobial Cationic Peptides/physiology , Base Sequence , Chromatography, High Pressure Liquid , DNA Primers , Electrophoresis, Polyacrylamide Gel , Hepcidins , Humans , Iron-Regulatory Proteins/isolation & purification , Iron-Regulatory Proteins/physiology , Mass Spectrometry/methods , Mice , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
19.
Sci Rep ; 7(1): 12679, 2017 10 04.
Article in English | MEDLINE | ID: mdl-28978947

ABSTRACT

The biguanide metformin is used for its antidiabetic effect for many years but how metformin acts remains poorly understood and controversial. AMP-activated protein kinase (AMPK), a protein kinase that plays a key role in maintaining energy homeostasis, is assumed to be one of the prime targets of metformin. However, since our demonstration that AMPK is not required for the beneficial effects of metformin on the control of glycemia, the list of AMPK-independent actions of metformin is rapidly increasing. Given the conflicting results on the effects of metformin we sought, using our genetic mouse models deficient in the catalytic subunits of AMPK, to determine whether this kinase is involved in the effects of metformin on the expression of the iron-regulatory hormone hepcidin, as recently proposed. Here we demonstrate, using different approaches, either isolated hepatocytes that lack AMPK, or direct AMPK activators, that, AMPK activation is not necessary for metformin to inhibit BMP6-induced hepcidin gene expression. These results may shed new lights on the increasingly recognized AMPK-independent metformin's molecular action, an important area of current research.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Bone Morphogenetic Protein 6/pharmacology , Gene Expression Regulation/drug effects , Hepatocytes/metabolism , Hepcidins/genetics , Metformin/pharmacology , Animals , Cells, Cultured , Hepatocytes/drug effects , Hepcidins/metabolism , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism
20.
Front Physiol ; 8: 804, 2017.
Article in English | MEDLINE | ID: mdl-29089902

ABSTRACT

Pulmonary iron excess is deleterious and contributes to a range of chronic and acute inflammatory diseases. Optimal lung iron concentration is maintained through dynamic regulation of iron transport and storage proteins. The iron-regulatory hormone hepcidin is also expressed in the lung. In order to better understand the interactions between iron-associated molecules and the hepcidin-ferroportin axis in lung iron balance, we examined lung physiology and inflammatory responses in two murine models of systemic iron-loading, either hepcidin knock-out (Hepc KO) or liver-specific hepcidin KO mice (Hepc KOliv), which do (Hepc KOliv) or do not (Hepc KO) express lung hepcidin. We have found that increased plasma iron in Hepc KO mice is associated with increased pulmonary iron levels, consistent with increased cellular iron uptake by pulmonary epithelial cells, together with an increase at the apical membrane of the cells of the iron exporter ferroportin, consistent with increased iron export in the alveoli. Subsequently, alveolar macrophages (AM) accumulate iron in a non-toxic form and this is associated with elevated production of ferritin. The accumulation of iron in the lung macrophages of hepcidin KO mice contrasts with splenic and hepatic macrophages which contain low iron levels as we have previously reported. Hepc KOliv mice with liver-specific hepcidin deficiency demonstrated same pulmonary iron overload profile as the Hepc KO mice, suggesting that pulmonary hepcidin is not critical in maintaining local iron homeostasis. In addition, the high iron load in the lung of Hepc KO mice does not appear to enhance acute lung inflammation or injury. Lastly, we have shown that intraperitoneal LPS injection is not associated with pulmonary hepcidin induction, despite high levels of inflammatory cytokines. However, intranasal LPS injection stimulates a hepcidin response, likely derived from AM, and alters pulmonary iron content in Hepc KO mice.

SELECTION OF CITATIONS
SEARCH DETAIL