Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biosci Rep ; 41(7)2021 07 30.
Article in English | MEDLINE | ID: mdl-34156073

ABSTRACT

The flavonol rutin has been shown to possess antioxidant and iron chelating properties in vitro and in vivo. These dual properties are beneficial as therapeutic options to reduce iron accumulation and the generation of reactive oxygen species (ROS) resultant from excess free iron. The effect of rutin on iron metabolism has been limited to studies performed in wildtype mice either injected or fed high-iron diets. The effect of rutin on iron overload caused by genetic dysregulation of iron homoeostasis has not yet been investigated. In the present study we examined the effect of rutin treatment on tissue iron loading in a genetic mouse model of iron overload, which mirrors the iron loading associated with Type 3 hereditary haemochromatosis patients who have a defect in Transferrin Receptor 2 (TFR2). Male TFR2 knockout (KO) mice were administered rutin via oral gavage for 21 continuous days. Following treatment, iron levels in serum, liver, duodenum and spleen were assessed. In addition, hepatic ferritin protein levels were determined by Western blotting, and expression of iron homoeostasis genes by quantitative real-time PCR. Rutin treatment resulted in a significant reduction in hepatic ferritin protein expression and serum transferrin saturation. In addition, trends towards decreased iron levels in the liver and serum, and increased serum unsaturated iron binding capacity were observed. This is the first study to explore the utility of rutin as a potential iron chelator and therapeutic in an animal model of genetic iron overload.


Subject(s)
Hemochromatosis/drug therapy , Iron/blood , Liver/drug effects , Receptors, Transferrin/deficiency , Rutin/pharmacology , Animals , Biomarkers/blood , Disease Models, Animal , Ferritins/metabolism , Hemochromatosis/blood , Hemochromatosis/genetics , Liver/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, Transferrin/blood , Receptors, Transferrin/genetics , Transferrin/metabolism
2.
Biosci Rep ; 40(8)2020 08 28.
Article in English | MEDLINE | ID: mdl-32766721

ABSTRACT

Exome sequencing has identified the glyceronephosphate O-acyltransferase (GNPAT) gene as a genetic modifier of iron overload in hereditary hemochromatosis (HH). Subjects with HFE (Homeostatic Iron Regulator) p.C282Y mutations and the GNPAT p.D519G variant had more iron loading compared with subjects without the GNPAT variant. In response to an oral iron challenge, women with GNPAT polymorphisms loaded more iron as compared with women without polymorphisms, reinforcing a role for GNPAT in iron homeostasis. The aim of the present study was to develop and characterize an animal model of disease to further our understanding of genetic modifiers, and in particular the role of GNPAT in iron homeostasis. We generated an Hfe/Gnpat mouse model reminiscent of the patients previously studied and studied these mice for up to 26 weeks. We also examined the effect of dietary iron loading on mice with reduced Gnpat expression. Gnpat heterozygosity in Hfe knockout mice does not play a role in systemic iron homeostasis; Gnpat+/- mice fed a high-iron diet, however, had lower hepatic hepcidin (HAMP) mRNA expression, whereas they have significantly higher serum iron levels and transferrin saturation compared with wildtype (WT) littermates on a similar diet. These results reinforce an independent role of GNPAT in systemic iron homeostasis, reproducing in an animal model, the observations in women with GNPAT polymorphisms subjected to an iron tolerance test.


Subject(s)
Acyltransferases/deficiency , Hemochromatosis/enzymology , Hepcidins/metabolism , Iron, Dietary/metabolism , Liver/metabolism , Acyltransferases/genetics , Animals , Disease Models, Animal , Hemochromatosis/blood , Hemochromatosis/genetics , Hemochromatosis Protein/deficiency , Hemochromatosis Protein/genetics , Hepcidins/genetics , Homeostasis , Iron, Dietary/blood , Male , Mice, Inbred C57BL , Mice, Knockout , Sex Factors , Transferrin/metabolism
3.
Blood Cells Mol Dis ; 85: 102463, 2020 11.
Article in English | MEDLINE | ID: mdl-32652459

ABSTRACT

Glyceronephosphate O-acyltransferase (GNPAT) p.D519G (rs11558492) was identified as a genetic modifier correlated with more severe iron overload in hemochromatosis through whole-exome sequencing of HFE p.C282Y homozygotes with extreme iron phenotypes. We studied the prevalence of p.D519G in HFE p.C282Y/p.H63D compound heterozygotes, a genotype associated with iron overload in some patients. Cases were Australian participants with elevated serum ferritin (SF) levels ≥300µg/L (males) and ≥200µg/L (females); subjects whose SF levels were below these cut-offs were designated as controls. Samples were genotyped for GNPAT p.D519G. We compared the allele frequency of the present subjects, with/without elevated SF, to p.D519G frequency in public datasets. GNPAT p.D519G was more prevalent in our cohort of p.C282Y/p.H63D compound heterozygotes with elevated SF (37%) than European public datasets: 1000G 21%, gnomAD 20% and ESP 21%. We conclude that GNPAT p.D519G is associated with elevated SF in Australian HFE p.C282Y/p.H63D compound heterozygotes.


Subject(s)
Acyltransferases/genetics , Hemochromatosis Protein/genetics , Hemochromatosis/genetics , Point Mutation , Adult , Female , Ferritins/blood , Hemochromatosis/blood , Heterozygote , Humans , Male , Middle Aged
4.
Biochim Biophys Acta Mol Basis Dis ; 1866(10): 165882, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32565019

ABSTRACT

Peroxisomes are organelles, abundant in the liver, involved in a variety of cellular functions, including fatty acid metabolism, plasmalogen synthesis and metabolism of reactive oxygen species. Several inherited disorders are associated with peroxisomal dysfunction; increasingly many are associated with hepatic pathologies. The liver plays a principal role in regulation of iron metabolism. In this study we examined the possibility of a relationship between iron homeostasis and peroxisomal integrity. We examined the effect of deleting Pex13 in mouse liver on systemic iron homeostasis. We also used siRNA-mediated knock-down of PEX13 in a human hepatoma cell line (HepG2/C3A) to elucidate the mechanisms of PEX13-mediated regulation of hepcidin. We demonstrate that transgenic mice lacking hepatocyte Pex13 have defects in systemic iron homeostasis. The ablation of Pex13 expression in hepatocytes leads to a significant reduction in hepatic hepcidin levels. Our results also demonstrate that a deficiency of PEX13 gene expression in HepG2/C3A cells leads to decreased hepcidin expression, which is mediated through an increase in the signalling protein SMAD7, and endoplasmic reticulum (ER) stress. This study identifies a novel role for a protein involved in maintaining peroxisomal integrity and function in iron homeostasis. Loss of Pex13, a protein important for peroxisomal function, in hepatocytes leads to a significant increase in ER stress, which if unresolved, can affect liver function. The results from this study have implications for the management of patients with peroxisomal disorders and the liver-related complications they may develop.


Subject(s)
Hepatocytes/metabolism , Iron/metabolism , Membrane Proteins/deficiency , Peroxisomes/pathology , Animals , Bone Morphogenetic Proteins/metabolism , Cell Membrane/pathology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress , Female , Gene Knockdown Techniques , Hep G2 Cells , Hepcidins/metabolism , Humans , Iron/blood , Liver/cytology , Liver/metabolism , Liver/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Models, Animal , Peroxisomal Disorders/pathology , Peroxisomes/metabolism , RNA, Small Interfering/metabolism , Smad7 Protein/metabolism
5.
Biosci Rep ; 40(5)2020 05 29.
Article in English | MEDLINE | ID: mdl-32301493

ABSTRACT

Mutations in the only known iron exporter ferroportin (FPN) in humans are associated with the autosomal dominantly inherited iron overload disorder ferroportin disease or type IV hereditary hemochromatosis (HH). While our knowledge of the central role of FPN in iron homeostasis has grown in the last 20 years, there exist some questions surrounding the structure and membrane topology of FPN with conflicting data on whether this receptor acts as a monomer or a multimer. To investigate and determine if FPN dimerization occurs in cells, we used novel tools including a variety of different FPN constructs expressing different tagged versions of the protein, a novel antibody that only detects cell surface FPN and proximity ligation assays. The results of the present study suggest that both the carboxy- and amino-termini of the FPN protein are intracellular. We also show that exogenously transfected FPN forms dimers; these dimers can be formed between the wild-type and mutant FPN proteins. This is the first study to examine the intracellular dimerization of FPN protein. Using proximity ligation assays, we show intracellular localization of FPN dimers and the interaction between FPN and hepcidin proteins as well. These results have important implications in the field of iron metabolism and add to our knowledge about FPN membrane topology and physiology of iron transport. This will be of importance in understanding the clinical implications of FPN mutations and of interest to future research aimed at targeting FPN expression to modulate iron homeostasis.


Subject(s)
Cation Transport Proteins/metabolism , Hepatocytes/metabolism , Iron/metabolism , Cation Transport Proteins/genetics , Cell Line, Tumor , Cell Membrane/metabolism , Hemochromatosis/genetics , Hepatocytes/cytology , Hepcidins/metabolism , Humans , Mutation , Protein Multimerization
6.
Biochim Biophys Acta Mol Basis Dis ; 1864(8): 2550-2556, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29752985

ABSTRACT

Our knowledge of iron homeostasis has increased steadily over the last two decades; much of this has been made possible through the study of animal models of iron-related disease. Analysis of transgenic mice with deletions or perturbations in genes known to be involved in systemic or local regulation of iron metabolism has been particularly informative. The effect of these genes on iron accumulation and hepcidin regulation is traditionally compared with wildtype mice fed a high iron diet, most often a 2% carbonyl iron diet. Recent studies have indicated that a very high iron diet could be detrimental to the health of the mice and could potentially affect homeostasis of other metals, for example zinc and copper. We analyzed mice fed a diet containing either 0.25%, 0.5%, 1% or 2% carbonyl iron for two weeks and compared them with mice on a control diet. Our results indicate that a 0.25% carbonyl iron diet is sufficient to induce maximal hepatic hepcidin response. Importantly these results also demonstrate that in a chronic setting of iron administration, the amount of excess hepatic iron may not further influence hepcidin regulation and that expression of hepcidin plateaus at lower hepatic iron levels. These studies provide further insights into the regulation of this important hormone.


Subject(s)
Hemochromatosis/metabolism , Hepcidins/metabolism , Iron , Liver/metabolism , Animals , Copper/metabolism , Disease Models, Animal , Hemochromatosis/pathology , Iron/metabolism , Iron/pharmacology , Liver/pathology , Male , Mice , Zinc/metabolism
7.
Hum Genomics ; 12(1): 23, 2018 04 25.
Article in English | MEDLINE | ID: mdl-29695288

ABSTRACT

BACKGROUND: Atypical iron overload without variation in the five clinically associated hereditary hemochromatosis genes is now recognized; however, their etiology remains unknown. Since the identification of iron overload in the bone morphogenetic protein 6 (Bmp6) knockout mouse, the search has been on for clinically pathogenic variants in the BMP6 gene. A recent report proposes that variants in the pro-peptide region of BMP6 are the underlying cause of several cases of iron overload. We performed targeted next-generation sequencing on three cases of atypical iron overload with Asian ethnicity and identified a p.Q118dup (aka p.E112indelEQ, p.Q115dup, p.Q118_L119insQ) variant in BMP6. The purpose of this study was to characterize the molecular function of the identified BMP6 variant. Molecular characterization by immunofluorescence microscopy and Western blotting of transfected cells, bioinformatics, and population analyses was performed. RESULTS: In contrast to reports for other BMP6 pro-peptide variants in this region, our data indicates that this variant does not affect the function of the mature BMP6 protein. CONCLUSIONS: Our data suggest that assignment of disease causation in clinical cases of iron overload to pro-peptide variants in BMP6 should thus be treated with caution and requires biological characterization.


Subject(s)
Bone Morphogenetic Protein 6/genetics , Genetic Predisposition to Disease , Hemochromatosis/genetics , Iron Overload/genetics , Animals , Hemochromatosis/metabolism , Hemochromatosis/pathology , High-Throughput Nucleotide Sequencing , Humans , Iron Overload/metabolism , Iron Overload/pathology , Mice , Mice, Knockout , Mutation , Peptides/genetics
8.
Cell Mol Gastroenterol Hepatol ; 5(3): 319-331, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29552621

ABSTRACT

BACKGROUND & AIMS: Iron has an increasingly recognized role in the regulation of adipose tissue function, including the expression of adipokines involved in the pathogenesis of nonalcoholic fatty liver disease. The cellular iron exporter, ferroportin, has been proposed as being a key determinant of adipocyte iron homeostasis. METHODS: We studied an adipocyte-specific ferroportin (Fpn1) knockout mouse model, using an Adipoq-Cre recombinase driven Fpn1 deletion and fed mice according to the fast food diet model of nonalcoholic steatohepatitis. RESULTS: We showed successful selective deletion of Fpn1 in adipocytes, but found that this did not lead to increased adipocyte iron stores as measured by atomic absorption spectroscopy or histologically quantified iron granules after staining with 3,3'-diaminobenzidine-enhanced Perls' stain. Mice with adipocyte-specific Fpn1 deletion did not show dysregulation of adiponectin, leptin, resistin, or retinol-binding protein-4 expression. Similarly, adipocyte-specific Fpn1 deletion did not affect insulin sensitivity during hyperinsulinemic-euglycemic clamp studies or lead to histologic evidence of increased liver injury. We have shown, however, that the fast food diet model of nonalcoholic steatohepatitis generates an increase in adipose tissue macrophage infiltration with crown-like structures, as seen in human beings, further validating the utility of this model. CONCLUSIONS: Ferroportin may not be a key determinant of adipocyte iron homeostasis in this knockout model. Further studies are needed to determine the mechanisms of iron metabolism in adipocytes and adipose tissue.

9.
Am J Hematol ; 91(8): 812-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27169626

ABSTRACT

Iron metabolism and erythropoiesis are inherently interlinked physiological processes. Regulation of iron metabolism is mediated by the iron-regulatory hormone hepcidin. Hepcidin limits the amount of iron released into the blood by binding to and causing the internalization of the iron exporter, ferroportin. A number of molecules and physiological stimuli, including erythropoiesis, are known to regulate hepcidin. An increase in erythropoietic demand decreases hepcidin, resulting in increased bioavailable iron in the blood. Transferrin receptor 2 (TFR2) is involved in the systemic regulation of iron metabolism. Patients and mice with mutations in TFR2 develop hemochromatosis due to inappropriate hepcidin levels relative to body iron. Recent studies from our laboratory and others have suggested an additional role for TFR2 in response to iron-restricted erythropoiesis. These studies used mouse models with perturbed systemic iron metabolism: anemic mice lacking matriptase-2 and Tfr2, or bone marrow transplants from iron-loaded Tfr2 null mice. We developed a novel transgenic mouse model which lacks Tfr2 in the hematopoietic compartment, enabling the delineation of the role of Tfr2 in erythroid development without interfering with its role in systemic iron metabolism. We show that in the absence of hematopoietic Tfr2 immature polychromatic erythroblasts accumulate with a concordant reduction in the percentage of mature erythroid cells in the spleen and bone marrow of anemic mice. These results demonstrate that erythroid Tfr2 is essential for an appropriate erythropoietic response in iron-deficient anemia. These findings may be of relevance in clinical situations in which an immediate and efficient erythropoietic response is required. Am. J. Hematol. 91:812-818, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Anemia, Iron-Deficiency/pathology , Erythropoiesis , Gene Deletion , Receptors, Transferrin/genetics , Anemia, Iron-Deficiency/etiology , Animals , Bone Marrow/pathology , Cell Differentiation , Erythroid Cells/pathology , Mice , Mice, Transgenic , Receptors, Transferrin/physiology , Spleen/pathology
10.
Am J Physiol Gastrointest Liver Physiol ; 310(3): G171-80, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26608187

ABSTRACT

Iron is an essential element, since it is a component of many macromolecules involved in diverse physiological and cellular functions, including oxygen transport, cellular growth, and metabolism. Systemic iron homeostasis is predominantly regulated by the liver through the iron regulatory hormone hepcidin. Hepcidin expression is itself regulated by a number of proteins, including transferrin receptor 2 (TFR2). TFR2 has been shown to be expressed in the liver, bone marrow, macrophages, and peripheral blood mononuclear cells. Studies from our laboratory have shown that mice with a hepatocyte-specific deletion of Tfr2 recapitulate the hemochromatosis phenotype of the global Tfr2 knockout mice, suggesting that the hepatic expression of TFR2 is important in systemic iron homeostasis. It is unclear how TFR2 in macrophages contributes to the regulation of iron metabolism. We examined the role of TFR2 in macrophages by analysis of transgenic mice lacking Tfr2 in macrophages by crossing Tfr2(f/f) mice with LysM-Cre mice. Mice were fed an iron-rich diet or injected with lipopolysaccharide to examine the role of macrophage Tfr2 in iron- or inflammation-mediated regulation of hepcidin. Body iron homeostasis was unaffected in the knockout mice, suggesting that macrophage TFR2 is not required for the regulation of systemic iron metabolism. However, peritoneal macrophages of knockout mice had significantly lower levels of ferroportin mRNA and protein, suggesting that TFR2 may be involved in regulating ferroportin levels in macrophages. These studies further elucidate the role of TFR2 in the regulation of iron homeostasis and its role in regulation of ferroportin and thus macrophage iron homeostasis.


Subject(s)
Homeostasis/genetics , Iron/metabolism , Macrophages, Peritoneal/metabolism , Receptors, Transferrin/genetics , Animals , Cation Transport Proteins/metabolism , Hepcidins/genetics , Hepcidins/metabolism , Humans , Iron/blood , Iron, Dietary/pharmacology , Lipopolysaccharides/pharmacology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
11.
Br J Haematol ; 168(6): 891-901, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25403101

ABSTRACT

Effective erythropoiesis requires an appropriate supply of iron and mechanisms regulating iron homeostasis and erythropoiesis are intrinsically linked. Iron dysregulation, typified by iron-deficiency anaemia and iron overload, is common in many clinical conditions and impacts the health of up to 30% of the world's population. The proteins transmembrane protease, serine 6 (TMPRSS6; also termed matriptase-2), HFE and transferrin receptor 2 (TFR2) play important and opposing roles in systemic iron homeostasis, by regulating expression of the iron regulatory hormone hepcidin. We have performed a systematic analysis of mice deficient in these three proteins and show that TMPRSS6 predominates over HFE and TFR2 in hepcidin regulation. The phenotype of mice lacking TMPRSS6 and TFR2 is characterized by severe anaemia and extramedullary haematopoiesis in the spleen. Stress erythropoiesis in these mice results in increased expression of the newly identified erythroid iron regulator erythroferrone, which does not appear to overcome the hepcidin overproduction mediated by loss of TMPRSS6. Extended analysis reveals that TFR2 plays an important role in erythroid cells, where it is involved in terminal erythroblast differentiation and the regulation of erythropoietin. In conclusion, we have identified an essential role for TFR2 in erythropoiesis that may provide new targets for the treatment of anaemia.


Subject(s)
Anemia, Iron-Deficiency/blood , Erythropoiesis/physiology , Receptors, Transferrin/physiology , Anemia, Iron-Deficiency/metabolism , Animals , Cell Differentiation/physiology , Erythroid Cells/pathology , Erythropoietin/biosynthesis , Hematopoiesis, Extramedullary/physiology , Hemochromatosis Protein , Hepcidins/metabolism , Histocompatibility Antigens Class I/blood , Histocompatibility Antigens Class I/physiology , Kidney/metabolism , Liver/metabolism , Male , Membrane Proteins/blood , Membrane Proteins/deficiency , Membrane Proteins/physiology , Mice , Mice, Knockout , Receptors, Erythropoietin/metabolism , Receptors, Transferrin/blood , Receptors, Transferrin/deficiency , Serine Endopeptidases/blood , Serine Endopeptidases/deficiency , Serine Endopeptidases/physiology , Splenomegaly/blood
SELECTION OF CITATIONS
SEARCH DETAIL