Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
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.
Am J Hematol ; 96(2): 251-257, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33180328

ABSTRACT

ß-thalassemias result from mutations in ß-globin, causing ineffective erythropoiesis and secondary iron overload due to inappropriately low levels of the iron regulatory hormone hepcidin. Mutations in transferrin receptor 2 (TFR2) lead to hereditary hemochromatosis (HH) as a result of inappropriately increased iron uptake from the diet, also due to improperly regulated hepcidin. TFR2 is also thought to be required for efficient erythropoiesis through its interaction with the erythropoietin receptor in erythroid progenitors. Transmembrane serine protease 6 (TMPRSS6), a membrane serine protease expressed selectively in the liver, participates in regulating hepcidin production in response to iron stores by cleaving hemojuvelin (HJV). We have previously demonstrated that inhibiting TMPRSS6 expression with a hepatocyte-specific siRNA formulation, induces hepcidin, mitigates anemia, and reduces iron overload in murine models of ß-thalassemia intermedia and HH. Here, we demonstrate that Tmprss6 siRNA treatment of double mutant Tfr2Y245X/Y245X HH Hbbth3/+ thalassemic mice induces hepcidin and diminishes tissue and serum iron levels. Importantly, treated double mutant animals produce more mature red blood cells and have a nearly 50% increase in hemoglobin compared to untreated ß-thalassemic mice. Furthermore, we also show that treatment of Tfr2Y245X/Y245X HH mice leads to increased hepcidin expression and reduced total body iron burden. These data indicate that siRNA suppression of Tmprss6, in conjunction with the targeting of TFR2, may be superior to inhibiting Tmprss6 alone in the treatment of the anemia and secondary iron loading in ß-thalassemia intermedia and may be useful as a method of suppressing the primary iron overload in TFR2-related (type 3) hereditary hemochromatosis.


Subject(s)
Hemochromatosis/metabolism , Iron Deficiencies , Receptors, Transferrin/deficiency , beta-Thalassemia/metabolism , Amino Acid Substitution , Animals , Disease Models, Animal , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Hemochromatosis/genetics , Hemochromatosis/pathology , Hemochromatosis Protein/genetics , Hemochromatosis Protein/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mutation, Missense , Receptors, Transferrin/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , beta-Thalassemia/genetics , beta-Thalassemia/pathology
3.
J Allergy Clin Immunol Pract ; 9(2): 753-759.e2, 2021 02.
Article in English | MEDLINE | ID: mdl-33096268

ABSTRACT

BACKGROUND: Iron uptake mediated by transferrin receptor 1 (TfR1), encoded by the TFRC gene, is essential for lymphocyte development and proliferation. Autosomal-recessive mutations in the human TFRC gene cause a combined immunodeficiency characterized by defective T- and B-cell proliferation as well as impaired class-switching. Clinical presentations have been severe in all reported cases, with symptoms including recurrent sinopulmonary infections, hypogammaglobulinemia, chronic diarrhea, and intermittent cytopenias. OBJECTIVE: To describe outcomes of allogeneic hematopoietic stem cell transplantation (HSCT) in patients with TFRC deficiency. METHODS: Retrospective chart review study of 5 patients with TFRC deficiency who underwent allogeneic HSCT between July 2011 and May 2018 at Boston Children's Hospital. RESULTS: Intermittent thrombocytopenia and neutropenia were a predominant feature of the clinical presentation in our cohort, and 3 patients who underwent bone marrow evaluation before HSCT were found to have signs of dysmyelopoiesis and dysplasia. One patient, who had a transplant at age 11 years, developed a clonal cytogenetic abnormality concerning for myelodysplastic syndrome. All 5 patients tolerated myeloablative conditioning regimens and had robust donor cell engraftment with resolution of cytopenias and independence from intravenous immunoglobulin substitution. All 5 patients were alive at a median follow-up of 47.1 months posttransplant (range, 15.7-85.4) and none had developed acute or chronic graft-versus-host disease. CONCLUSIONS: Allogeneic HSCT is curative for TFRC deficiency and rescues all known disease manifestations. Patients with TFRC deficiency may have a predisposition to malignant transformation of hematopoietic cells and may benefit from HSCT earlier in their disease course.


Subject(s)
Hematopoietic Stem Cell Transplantation , Receptors, Transferrin/deficiency , Transplantation Conditioning , Boston , Child , Humans , Receptors, Transferrin/genetics , Retrospective Studies , Transplantation, Homologous
4.
J Biol Chem ; 295(12): 3906-3917, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32054685

ABSTRACT

Transferrin receptor 2 (TFR2) is a transmembrane protein expressed mainly in hepatocytes and in developing erythroid cells and is an important focal point in systemic iron regulation. Loss of TFR2 function results in a rare form of the iron-overload disease hereditary hemochromatosis. Although TFR2 in the liver has been shown to be important for regulating iron homeostasis in the body, TFR2's function in erythroid progenitors remains controversial. In this report, we analyzed TFR2-deficient mice in the presence or absence of iron overload to distinguish between the effects caused by a high iron load and those caused by loss of TFR2 function. Analysis of bone marrow from TFR2-deficient mice revealed a reduction in the early burst-forming unit-erythroid and an expansion of late-stage erythroblasts that was independent of iron overload. Spleens of TFR2-deficient mice displayed an increase in colony-forming unit-erythroid progenitors and in all erythroblast populations regardless of iron overload. This expansion of the erythroid compartment coincided with increased erythroferrone (ERFE) expression and serum erythropoietin (EPO) levels. Rescue of hepatic TFR2 expression normalized hepcidin expression and the total cell count of the bone marrow and spleen, but it had no effect on erythroid progenitor frequency. On the basis of these results, we propose a model of TFR2's function in murine erythropoiesis, indicating that deficiency in this receptor is associated with increased erythroid development and expression of EPO and ERFE in extrahepatic tissues independent of TFR's role in the liver.


Subject(s)
Erythropoiesis , Iron Overload/pathology , Receptors, Transferrin/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cytokines/metabolism , Erythropoietin/blood , Hepcidins/metabolism , Iron Overload/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/metabolism , Receptors, Transferrin/deficiency , Spleen/pathology , Stem Cells/cytology , Stem Cells/metabolism
6.
Blood ; 133(4): 344-355, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30538134

ABSTRACT

Transferrin receptor 1 (Tfr1) mediates uptake of circulating transferrin-bound iron to developing erythroid cells and other cell types. Its critical physiological function is highlighted by the embryonic lethal phenotype of Tfr1-knockout (Tfrc-/-) mice and the pathologies of several tissue-specific knockouts. We generated TfrcAlb-Cre mice bearing hepatocyte-specific ablation of Tfr1 to explore implications in hepatocellular and systemic iron homeostasis. TfrcAlb-Cre mice are viable and do not display any apparent liver pathology. Nevertheless, their liver iron content (LIC) is lower compared with that of control Tfrcfl/fl littermates as a result of the reduced capacity of Tfr1-deficient hepatocytes to internalize iron from transferrin. Even though liver Hamp messenger RNA (mRNA) and serum hepcidin levels do not differ between TfrcAlb-Cre and Tfrcfl/fl mice, Hamp/LIC and hepcidin/LIC ratios are significantly higher in the former. Importantly, this is accompanied by modest hypoferremia and microcytosis, and it predisposes TfrcAlb-Cre mice to iron-deficiency anemia. TfrcAlb-Cre mice appropriately regulate Hamp expression following dietary iron manipulations or holo-transferrin injection. Holo-transferrin also triggers proper induction of Hamp mRNA, ferritin, and Tfr2 in primary TfrcAlb-Cre hepatocytes. We further show that these cells can acquire 59Fe from 59Fe-transferrin, presumably via Tfr2. We conclude that Tfr1 is redundant for basal hepatocellular iron supply but essential for fine-tuning hepcidin responses according to the iron load of hepatocytes. Our data are consistent with an inhibitory function of Tfr1 on iron signaling to hepcidin via its interaction with Hfe. Moreover, they highlight hepatocellular Tfr1 as a link between cellular and systemic iron-regulatory pathways.


Subject(s)
Antigens, CD/metabolism , Hepatocytes/metabolism , Hepcidins/metabolism , Homeostasis , Iron/metabolism , Receptors, Transferrin/metabolism , Anemia, Iron-Deficiency/pathology , Animals , Ferritins/metabolism , Gene Deletion , Gene Expression Regulation/drug effects , Gene Targeting , Hepatocytes/drug effects , Hepcidins/genetics , Homeostasis/drug effects , Integrases/metabolism , Iron, Dietary/pharmacology , Mice, Inbred C57BL , Receptors, Transferrin/deficiency , Transferrin/metabolism
7.
Immunity ; 49(1): 80-92.e7, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29958803

ABSTRACT

Iron deposition is frequently observed in human autoinflammatory diseases, but its functional significance is largely unknown. Here we showed that iron promoted proinflammatory cytokine expression in T cells, including GM-CSF and IL-2, via regulating the stability of an RNA-binding protein PCBP1. Iron depletion or Pcbp1 deficiency in T cells inhibited GM-CSF production by attenuating Csf2 3' untranslated region (UTR) activity and messenger RNA stability. Pcbp1 deficiency or iron uptake blockade in autoreactive T cells abolished their capacity to induce experimental autoimmune encephalomyelitis, an animal model for multiple sclerosis. Mechanistically, intracellular iron protected PCBP1 protein from caspase-mediated proteolysis, and PCBP1 promoted messenger RNA stability of Csf2 and Il2 by recognizing UC-rich elements in the 3' UTRs. Our study suggests that iron accumulation can precipitate autoimmune diseases by promoting proinflammatory cytokine production. RNA-binding protein-mediated iron sensing may represent a simple yet effective means to adjust the inflammatory response to tissue homeostatic alterations.


Subject(s)
Carrier Proteins/metabolism , Cytokines/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/metabolism , Iron/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology , 3' Untranslated Regions , Animals , Binding Sites , Cell Line , Cytokines/genetics , DNA-Binding Proteins , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Iron/agonists , Iron Deficiencies , Mice , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , RNA Processing, Post-Transcriptional , RNA Stability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , RNA-Binding Proteins , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Transferrin/deficiency , T-Lymphocytes, Helper-Inducer/transplantation
8.
ACS Chem Biol ; 13(8): 2143-2152, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29897731

ABSTRACT

Hereditary hemochromatosis (HH) is one of the most common genetic disorders in Caucasian populations, with no viable therapeutic options except phlebotomy. We describe a zebrafish model of human HH (HH) created by targeted mutagenesis of the gene encoding transferrin receptor 2 ( tfr2). TFR2 mutations in humans lead to HH Type 3, a rare but severe form of the disease. The tfr2 mutant model in zebrafish recapitulates the defining features of HH3: iron overload and suppression of hepcidin, the iron regulatory hormone. Using in vivo chemical screens in zebrafish embryos, we identify a new small molecule inducer of hepcidin: SC-514, a specific chemical inhibitor of NFkB signaling. Using independent small molecule inhibitors of the NFkB pathway, we demonstrate that inhibition of NFkB signaling causes induction of hepcidin transcription and reduction of iron overload in the HH3 model. This first successful chemical intervention for hereditary hemochromatosis may also have relevance in treatment of other very prevalent iron regulatory iron overload disorders such as thalassemia.


Subject(s)
Hemochromatosis/drug therapy , NF-kappa B/antagonists & inhibitors , Receptors, Transferrin/deficiency , Thiophenes/therapeutic use , Animals , Disease Models, Animal , Gene Knockdown Techniques , Hemochromatosis/metabolism , Hepcidins/genetics , Hepcidins/metabolism , Iron/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Signal Transduction/drug effects , Thiophenes/pharmacology , Up-Regulation/drug effects , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/physiology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
9.
Biomed Res Int ; 2017: 2408941, 2017.
Article in English | MEDLINE | ID: mdl-28540293

ABSTRACT

Type 3 haemochromatosis (HFE3) is a rare genetic iron overload disease which ultimately lead to compromised organs functioning. HFE3 is caused by mutations in transferrin receptor 2 (TFR2) gene that codes for two main isoforms (Tfr2α and Tfr2ß). Tfr2α is one of the hepatic regulators of iron inhibitor hepcidin. Tfr2ß is an intracellular isoform of the protein involved in the regulation of iron levels in reticuloendothelial cells. It has been recently demonstrated that Tfr2 is also involved in erythropoiesis. This study aims to further investigate Tfr2 erythropoietic role by evaluating the erythropoiesis of two Tfr2 murine models wherein either one or both of Tfr2 isoforms have been selectively silenced (Tfr2 KI and Tfr2 KO). The evaluations were performed in bone marrow and spleen, in 14 days' and 10 weeks' old mice, to assess erythropoiesis in young versus adult animals. The lack of Tfr2α leads to macrocytosis with low reticulocyte number and increased hemoglobin values, together with an anticipation of adult BM erythropoiesis and an increased splenic erythropoiesis. On the other hand, lack of Tfr2ß (Tfr2 KI mice) causes an increased and immature splenic erythropoiesis. Taken together, these data confirm the role of Tfr2α in modulation of erythropoiesis and of Tfr2ß in favoring iron availability for erythropoiesis.


Subject(s)
Hemochromatosis/genetics , Iron/metabolism , Protein Isoforms/genetics , Receptors, Transferrin/deficiency , Animals , Disease Models, Animal , Erythropoiesis/genetics , Hemochromatosis/pathology , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Mononuclear Phagocyte System/metabolism , Mononuclear Phagocyte System/pathology , Receptors, Transferrin/genetics , Spleen/metabolism , Spleen/pathology
10.
Neth J Med ; 75(2): 56-64, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28276324

ABSTRACT

BACKGROUND: Type 3 hereditary haemochromatosis (HH) is a rare iron overload disorder caused by variants in the transferrin 2 receptor (TFR2) gene. We aim to present characteristics of patients diagnosed with TFR2-HH in the Netherlands, in order to increase knowledge and awareness of this disease. METHODS: We collected clinical, biochemical and genetic data from four patients from three families diagnosed with HH type 3 in the Netherlands between 2009 and 2016. RESULTS: Three women and one man diagnosed with HH type 3 presented with arthralgia and elevated ferritin levels and transferrin saturation (TSAT) at ages 25-41 years. The hepcidin/ferritin ratio as measured in three patients was low. Liver iron content in two patients as assessed by MRI or liver biopsy was highly increased (250 and 362.7 µmol iron/g dry weight, respectively, reference < 35 µmol/g). DNA analysis revealed four different TFR2 pathogenic variants: one nonsense, one splicing and two missense variants, of which three are novel. Phlebotomy decreased the serum iron parameters but did not relieve the arthralgia. CONCLUSION: In patients with a combination of elevated TSAT and ferritin in the absence of anaemia, and after exclusion of HFE-related HH, rare forms of HH should be considered. In these cases, presentation with arthralgia in young adulthood, low hepcidin/ferritin ratio and/or liver iron content > 100 µmol/g form an indication for analysis of the TFR2 gene. Although type 3 HH is extremely rare, awareness of the disease among physicians is important in order to achieve an early diagnosis and prevent complications, such as liver damage.


Subject(s)
Arthralgia/genetics , Hemochromatosis/genetics , Receptors, Transferrin/deficiency , Adult , Arthralgia/blood , Female , Ferritins/blood , Genotype , Hemochromatosis/blood , Hepcidins/blood , Humans , Male , Netherlands , Receptors, Transferrin/blood , Receptors, Transferrin/genetics , Transferrin/analysis
11.
Proc Natl Acad Sci U S A ; 113(13): 3428-35, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26929359

ABSTRACT

Disrupted brain iron homeostasis is a common feature of neurodegenerative disease. To begin to understand how neuronal iron handling might be involved, we focused on dopaminergic neurons and asked how inactivation of transport proteins affected iron homeostasis in vivo in mice. Loss of the cellular iron exporter, ferroportin, had no apparent consequences. However, loss of transferrin receptor 1, involved in iron uptake, caused neuronal iron deficiency, age-progressive degeneration of a subset of dopaminergic neurons, and motor deficits. There was gradual depletion of dopaminergic projections in the striatum followed by death of dopaminergic neurons in the substantia nigra. Damaged mitochondria accumulated, and gene expression signatures indicated attempted axonal regeneration, a metabolic switch to glycolysis, oxidative stress, and the unfolded protein response. We demonstrate that loss of transferrin receptor 1, but not loss of ferroportin, can cause neurodegeneration in a subset of dopaminergic neurons in mice.


Subject(s)
Dopaminergic Neurons/metabolism , Iron/metabolism , Nerve Degeneration/etiology , Nerve Degeneration/metabolism , Animals , Brain/metabolism , Brain/pathology , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Dopaminergic Neurons/pathology , Female , Homeostasis , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/pathology , Receptors, Transferrin/deficiency , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism
12.
Int J Clin Exp Pathol ; 8(7): 8613-8, 2015.
Article in English | MEDLINE | ID: mdl-26339443

ABSTRACT

A case study of a female patient, diagnosed with iron deficiency anemia, was unresponsive to oral iron treatment and only partially responsive to parenteral iron therapy, a clinical profile resembling the iron-refractory iron deficiency anemia (IRIDA) disorder. However, the patient failed to exhibit microcytic phenotype, one of the IRIDA hallmarks. Biochemical assays revealed that serum iron, hepcidin, interluekin 6, and transferrin saturation were within the normal range of references or were comparable to her non-anemic offspring. Iron contents in serum and red blood cells and hemoglobin levels were measured, which confirmed the partial improvement of anemia after parenteral iron therapy. Strikingly, serum transferrin receptor in patient was almost undetectable, reflecting the very low activity of bone-marrow erythropoiesis. Our data demonstrate that this is not a case of systemic iron deficiency, but rather cellular iron deficit due to the low level of transferrin receptor, particularly in erythroid tissue.


Subject(s)
Anemia, Iron-Deficiency/blood , Iron/blood , Receptors, Transferrin/blood , Administration, Intravenous , Administration, Oral , Anemia, Iron-Deficiency/diagnosis , Anemia, Iron-Deficiency/drug therapy , Biomarkers/blood , Down-Regulation , Female , Humans , Iron/administration & dosage , Receptors, Transferrin/deficiency , Treatment Outcome
13.
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
14.
EBioMedicine ; 2(11): 1705-17, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26870796

ABSTRACT

Transferrin receptor (Tfr1) is ubiquitously expressed, but its roles in non-hematopoietic cells are incompletely understood. We used a tissue-specific conditional knockout strategy to ask whether skeletal muscle required Tfr1 for iron uptake. We found that iron assimilation via Tfr1 was critical for skeletal muscle metabolism, and that iron deficiency in muscle led to dramatic changes, not only in muscle, but also in adipose tissue and liver. Inactivation of Tfr1 incapacitated normal energy production in muscle, leading to growth arrest and a muted attempt to switch to fatty acid ß oxidation, using up fat stores. Starvation signals stimulated gluconeogenesis in the liver, but amino acid substrates became limiting and hypoglycemia ensued. Surprisingly, the liver was also iron deficient, and production of the iron regulatory hormone hepcidin was depressed. Our observations reveal a complex interaction between iron homeostasis and metabolism that has implications for metabolic and iron disorders.


Subject(s)
Muscles/metabolism , Receptors, Transferrin/deficiency , Animals , Cluster Analysis , Gene Expression Regulation , Genes, Lethal , Iron Deficiencies , Iron Metabolism Disorders/genetics , Iron Metabolism Disorders/metabolism , Iron Metabolism Disorders/pathology , Liver/metabolism , Metabolome , Metabolomics/methods , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscles/pathology , Oxidative Phosphorylation , Phenotype , Receptors, Transferrin/genetics
15.
J Nutr ; 144(6): 979-87, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24744314

ABSTRACT

Subclinical micronutrient deficiencies remain a hidden aspect of malnutrition for which comprehensive data are lacking in school-aged children. We assessed the micronutrient status of Nepalese children, aged 6 to 8 y, born to mothers who participated in a community-based antenatal micronutrient supplementation trial from 1999 to 2001. Of 3305 participants, plasma indicators were assessed in a random sample of 1000 children. Results revealed deficiencies of vitamins A (retinol <0.70 µmol/L, 8.5%), D (25-hydroxyvitamin D <50 nmol/L, 17.2%), E (α-tocopherol <9.3 µmol/L, 17.9%), K (decarboxy prothombin >2 µg/L, 20%), B-12 (cobalamin <150 pmol/L, 18.1%), B-6 [pyridoxal-5'-phosphate (PLP) <20 nmol/L, 43.1%], and ß-carotene (41.5% <0.09 µmol/L), with little folate deficiency (6.2% <13.6 nmol/L). Deficiencies of iron [ferritin <15 µg/L, 10.7%; transferrin receptor (TfR) >8.3 mg/L, 40.1%; TfR:ferritin >500 µg/µg, 14.3%], iodine (thyroglobulin >40 µg/L, 11.4%), and selenium (plasma selenium <0.89 µmol/L, 59.0%) were observed, whereas copper deficiency was nearly absent (plasma copper <11.8 µmol/L, 0.7%). Hemoglobin was not assessed. Among all children, 91.7% experienced at least 1 micronutrient deficiency, and 64.7% experienced multiple deficiencies. Inflammation (α-1 acid glycoprotein >1 g/L, C-reactive protein >5 mg/L, or both) was present in 31.6% of children, affecting the prevalence of deficiency as assessed by retinol, ß-carotene, PLP, ferritin, TfR, selenium, copper, or having any or multiple deficiencies. For any nutrient, population deficiency prevalence estimates were altered by ≤5.4% by the presence of inflammation, suggesting that the majority of deficiencies exist regardless of inflammation. Multiple micronutrient deficiencies coexist in school-aged children in rural Nepal, meriting more comprehensive strategies for their assessment and prevention.


Subject(s)
Anemia, Iron-Deficiency/epidemiology , Folic Acid Deficiency/epidemiology , Inflammation/epidemiology , Micronutrients/blood , Micronutrients/deficiency , Rural Population , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/drug therapy , C-Reactive Protein/metabolism , Child , Copper/administration & dosage , Copper/blood , Copper/deficiency , Cross-Sectional Studies , Dietary Supplements , Female , Ferritins/blood , Folic Acid Deficiency/blood , Folic Acid Deficiency/drug therapy , Hemoglobins/metabolism , Humans , Inflammation/blood , Logistic Models , Male , Micronutrients/administration & dosage , Nepal/epidemiology , Nutritional Status , Prevalence , Receptors, Transferrin/blood , Receptors, Transferrin/deficiency , Selenium/administration & dosage , Selenium/blood , Socioeconomic Factors , Vitamin A/administration & dosage , Vitamin A/blood , Vitamin D/administration & dosage , Vitamin D/analogs & derivatives , Vitamin D/blood , alpha-Tocopherol/administration & dosage , alpha-Tocopherol/blood
16.
Blood Cells Mol Dis ; 52(2-3): 83-7, 2014.
Article in English | MEDLINE | ID: mdl-24055163

ABSTRACT

OBJECTIVE: Hereditary hemochromatosis (HH) is a common Mendelian disorder of iron metabolism. Eighty percent of northern Europeans descendant HH patients carry the same mutation (p.C282Y) in the HFE gene. Simultaneously, due to a founder effect, its frequency varies considerably between different populations. In Central-Southern Italy the prevalence of p.C282Y mutation is low and in several patients the disease has different causes. Four additional rarer forms have been described. Type 3 HH has been reported in about 50 families and no more than 30 TFR2 pathogenic mutations have been globally identified. The aim of this study is to assess the TFR2 role in non-HFE HH pathogenesis. STUDY DESIGN AND SETTING: TFR2 sequence analysis was performed on 45 Italian patients without HFE mutations. RESULTS: This study revealed TFR2 biallelic pathogenic mutations in 7/45 (15.6%) individuals. Moreover monoallelic TFR2 deleterious defects (18%) or polymorphisms with unclear meaning (36%) were identified. Besides, we recognized 10 novel variants and 9 described changes. CONCLUSION: We believe this to be the largest series of type 3 HH patients described so far. Present findings support the hypothesis of a main role of the TFR2 gene in HH pathogenesis in those regions, such as Central-Southern Italy, where the p.C282Y frequency is low.


Subject(s)
Hemochromatosis/genetics , Receptors, Transferrin/deficiency , Alleles , Base Sequence , DNA Mutational Analysis , Hemochromatosis/metabolism , Hemochromatosis/pathology , Humans , Iron/metabolism , Iron Overload/genetics , Iron Overload/metabolism , Italy , Mutation , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism
17.
Am J Clin Nutr ; 97(6): 1364-74, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23615827

ABSTRACT

BACKGROUND: Iron deficiency (ID) is a major cause of anemia, along with other nutritional, parasitic, and genetic factors. Accurate biomarkers are needed to estimate the relative contribution of ID to anemia. Soluble transferrin receptor (sTfR) is thought to be unaffected by inflammation. OBJECTIVES: The objectives were to determine the difference in sTfR and plasma ferritin (PF) concentrations among infants (6-23 mo of age), school-age children (6-8 y of age), and women (15-25 y of age) with and without inflammation and with and without Plasmodium falciparum infection and to assess the effect of adjusting sTfR and PF for inflammation or for P. falciparum infection on the estimated prevalence of ID. DESIGN: The data were derived from a 14-mo prospective longitudinal survey on anemia, which was conducted in the Taabo area, south-central Côte d'Ivoire. RESULTS: At baseline, sTfR concentration was significantly higher in infants and school-age children with either inflammation or P. falciparum infection than in control individuals without inflammation or without P. falciparum infection. Individuals with inflammation had significantly higher PF concentrations than did subjects without inflammation. Adjustments in sTfR concentrations for inflammation or P. falciparum infection in infants and school-age children resulted in significantly lower ID prevalence. Adjustment of PF for inflammation and Plasmodium infection resulted in a higher ID prevalence in infants and women. CONCLUSIONS: In Ivorian infants and school-age children, ID prevalence was considerably lower after adjustment of sTfR for inflammation. However, as the prevalence estimates for ID differed widely if based on sTfR or PF, caution is still needed when estimating ID prevalence in areas with a high prevalence of inflammation or malaria. This trial was registered at controlled-trials.com as ISRCTN02181959.


Subject(s)
Ferritins/blood , Inflammation/epidemiology , Malaria/epidemiology , Receptors, Transferrin/blood , Adolescent , Adult , Age Factors , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/epidemiology , Anemia, Iron-Deficiency/metabolism , Biomarkers/blood , Child , Cote d'Ivoire/epidemiology , Cross-Sectional Studies , Female , Ferritins/deficiency , Humans , Infant , Inflammation/complications , Longitudinal Studies , Malaria/complications , Male , Plasmodium falciparum , Prevalence , Prospective Studies , Receptors, Transferrin/deficiency , Young Adult
19.
Biochim Biophys Acta ; 1832(1): 76-84, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22960056

ABSTRACT

The inhibitory Smad7 acts as a critical suppressor of hepcidin, the major regulator of systemic iron homeostasis. In this study we define the mRNA expression of the two functionally related Smad proteins, Smad6 and Smad7, within pathways known to regulate hepcidin levels. Using mouse models for hereditary hemochromatosis (Hfe-, TfR2-, Hfe/TfR2-, Hjv- and hepcidin1-deficient mice) we show that hepcidin, Smad6 and Smad7 mRNA expression is coordinated in such a way that it correlates with the activity of the Bmp/Smad signaling pathway rather than with liver iron levels. This regulatory circuitry is disconnected by iron treatment of Hfe-/- and Hfe/TfR2 mice that significantly increases hepatic iron levels as well as hepcidin, Smad6 and Smad7 mRNA expression but fails to augment pSmad1/5/8 levels. This suggests that additional pathways contribute to the regulation of hepcidin, Smad6 and Smad7 under these conditions which do not require Hfe.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Hemochromatosis/genetics , Smad6 Protein/genetics , Smad7 Protein/genetics , Animals , Antimicrobial Cationic Peptides/metabolism , Disease Models, Animal , Female , Hemochromatosis/metabolism , Hemochromatosis Protein , Hepcidins , Histocompatibility Antigens Class I/genetics , Humans , Iron/metabolism , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Transferrin/deficiency , Receptors, Transferrin/genetics , Smad6 Protein/metabolism , Smad7 Protein/metabolism
20.
Biochem J ; 449(1): 275-84, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23016877

ABSTRACT

The IKK [inhibitor of NF-κB (nuclear factor κB) kinase] complex has an essential role in the activation of the family of NF-κB transcription factors in response to a variety of stimuli. To identify novel IKK-interacting proteins, we performed an unbiased proteomics screen where we identified TfR1 (transferrin receptor 1). TfR1 is required for transferrin binding and internalization and ultimately for iron homoeostasis. TfR1 depletion does not lead to changes in IKK subunit protein levels; however, it does reduce the formation of the IKK complex, and inhibits TNFα (tumour necrosis factor α)-induced NF-κB-dependent transcription. We find that, in the absence of TfR1, NF-κB does not translocate to the nucleus efficiently, and there is a reduction in the binding to target gene promoters and consequentially less target gene activation. Significantly, depletion of TfR1 results in an increase in apoptosis in response to TNFα treatment, which is rescued by elevating the levels of RelA/NF-κB. Taken together, these results indicate a new function for TfR1 in the control of IKK and NF-κB. Our data indicate that IKK-NF-κB responds to changes in iron within the cell.


Subject(s)
Antigens, CD/metabolism , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Receptors, Transferrin/metabolism , Signal Transduction/physiology , Cell Line, Tumor , HEK293 Cells , Humans , I-kappa B Kinase/physiology , Iron/chemistry , Iron/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Protein Binding/physiology , Receptors, Transferrin/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...