Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
PLoS Biol ; 21(1): e3001936, 2023 01.
Article in English | MEDLINE | ID: mdl-36649314

ABSTRACT

Ferroportin (Fpn) is the only known iron exporter in humans and is essential for maintaining iron homeostasis. Fpn activity is suppressed by hepcidin, an endogenous peptide hormone, which inhibits iron export and promotes endocytosis of Fpn. Hepcidin deficiency leads to hemochromatosis and iron-loading anemia. Previous studies have shown that small peptides that mimic the first few residues of hepcidin, i.e., minihepcidins, are more potent than hepcidin. However, the mechanism of enhanced inhibition by minihepcidins remains unclear. Here, we report the structure of human ferroportin in complex with a minihepcidin, PR73 that mimics the first 9 residues of hepcidin, at 2.7 Å overall resolution. The structure reveals novel interactions that were not present between Fpn and hepcidin. We validate PR73-Fpn interactions through binding and transport assays. These results provide insights into how minihepcidins increase inhibition potency and will guide future development of Fpn inhibitors.


Subject(s)
Cation Transport Proteins , Hemochromatosis , Humans , Hepcidins/metabolism , Hepcidins/pharmacology , Iron/metabolism , Cation Transport Proteins/metabolism
2.
Blood ; 139(16): 2460-2470, 2022 04 21.
Article in English | MEDLINE | ID: mdl-34932791

ABSTRACT

The erythroid marrow and circulating red blood cells (RBCs) are the key components of the human erythron. Abnormalities of the erythron that are responsible for anemia can be separated into 3 major categories: erythroid hypoproliferation, ineffective erythropoiesis, and peripheral hemolysis. Ineffective erythropoiesis is characterized by erythropoietin-driven expansion of early-stage erythroid precursors, associated with apoptosis of late-stage precursors. This mechanism is primarily responsible for anemia in inherited disorders like ß-thalassemia, inherited sideroblastic anemias, and congenital dyserythropoietic anemias, as well as in acquired conditions like some subtypes of myelodysplastic syndrome (MDS). The inherited anemias that are due to ineffective erythropoiesis are also defined as iron-loading anemias because of the associated parenchymal iron loading caused by the release of erythroid factors that suppress hepcidin production. Novel treatments specifically targeting ineffective erythropoiesis are being developed. Iron restriction through enhancement of hepcidin activity or inhibition of ferroportin function has been shown to reduce ineffective erythropoiesis in murine models of ß-thalassemia. Luspatercept is a transforming growth factor-ß ligand trap that inhibits SMAD2/3 signaling. Based on preclinical and clinical studies, this compound is now approved for the treatment of anemia in adult patients with ß-thalassemia who require regular RBC transfusions. Luspatercept is also approved for the treatment of transfusion-dependent anemia in patients with MDS with ring sideroblasts, most of whom carry a somatic SF3B1 mutation. While the long-term effectiveness and safety of luspatercept need to be evaluated in ß-thalassemia and MDS, defining the molecular mechanisms of ineffective erythropoiesis in different disorders might allow the discovery of new effective compounds.


Subject(s)
Anemia, Sideroblastic , Myelodysplastic Syndromes , beta-Thalassemia , Adult , Humans , Mice , Animals , Hepcidins/pharmacology , Erythropoiesis/genetics , beta-Thalassemia/complications , beta-Thalassemia/genetics , beta-Thalassemia/therapy , Iron , Anemia, Sideroblastic/genetics , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics
3.
Int J Mol Sci ; 24(11)2023 May 25.
Article in English | MEDLINE | ID: mdl-37298202

ABSTRACT

Hepcidin, a cysteine-rich antimicrobial peptide, has a highly conserved gene structure in teleosts, and it plays an essential role in host immune response against various pathogenic bacteria. Nonetheless, few studies on the antibacterial mechanism of hepcidin in golden pompano (Trachinotus ovatus) have been reported. In this study, we synthesized a derived peptide, TroHepc2-22, from the mature peptide of T. ovatus hepcidin2. Our results showed that TroHepc2-22 has superior antibacterial abilities against both Gram-negative (Vibrio harveyi and Edwardsiella piscicida) and Gram-positive (Staphylococcus aureus and Streptococcus agalactiae) bacteria. Based on the results of a bacterial membrane depolarization assay and propidium iodide (PI) staining assay in vitro, TroHepc2-22 displayed antimicrobial activity by inducing the bacterial membrane depolarization and changing the bacterial membrane permeability. Scanning electron microscopy (SEM) visualization illustrated that TroHepc2-22 brought about membrane rupturing and the leakage of the cytoplasm for the bacteria. In addition, TroHepc2-22 was verified to have hydrolytic activity on bacterial genomic DNA in view of the results of the gel retardation assay. In terms of the in vivo assay, the bacterial loads of V. harveyi in the tested immune tissues (liver, spleen, and head kidney) were significantly reduced in T. ovatus, revealing that TroHepc2-22 significantly enhanced the resistance against V. harveyi infection. Furthermore, the expressions of immune-related genes, including tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), interleukin 1-ß (IL-1ß), IL-6, Toll-like receptor 1 (TLR1), and myeloid differentiation factor 88 (MyD88) were significantly increased, indicating that TroHepc2-22 might regulate inflammatory cytokines and activate immune-related signaling pathways. To summarize, TroHepc2-22 possesses appreciable antimicrobial activity and plays a vital role in resisting bacterial infection. The observation of our present study unveils the excellent application prospect of hepcidin as a substitute for antibiotics to resist pathogenic microorganisms in teleosts.


Subject(s)
Anti-Infective Agents , Fish Diseases , Perciformes , Vibrio Infections , Animals , Hepcidins/genetics , Hepcidins/pharmacology , Immunity, Innate/genetics , Perciformes/genetics , Fishes/metabolism , Peptides , Fish Proteins/genetics , Fish Proteins/pharmacology , Fish Proteins/chemistry
4.
Blood ; 136(19): 2206-2216, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32584957

ABSTRACT

Iron disorders are associated with adverse pregnancy outcomes, yet iron homeostatic mechanisms during pregnancy are poorly understood. In humans and rodents, the iron-regulatory hormone hepcidin is profoundly decreased in pregnant mothers, which is thought to ensure adequate iron availability for transfer across placenta. However, the fetal liver also produces hepcidin, which may regulate fetal iron endowment by controlling placental iron export. To determine the relative contribution of maternal vs embryo hepcidin to the control of embryo iron endowment in iron-sufficient or iron-overloaded mice, we generated combinations of mothers and embryos that had or lacked hepcidin. We found that maternal, but not embryonic, hepcidin determined embryo and placental iron endowment in a healthy pregnancy. We further determined that inflammation can counteract pregnancy-dependent suppression of maternal hepcidin. To establish how essential maternal hepcidin suppression is for embryo iron homeostasis, we mimicked the range of maternal hepcidin activity by administering a hepcidin peptide mimetic to pregnant mice. This also allowed us to determine the effect of isolated maternal hepcidin excess on pregnancy, in the absence of other confounding effects of inflammation. Higher doses of hepcidin agonist caused maternal iron restriction and anemia, lower placenta and embryo weight, embryo anemia, and increased embryo mortality. Low agonist doses did not cause maternal anemia but still adversely affected the embryo, causing anemia, tissue iron deficiency (including in the brain), and decreased weight. Our studies demonstrate that suppression of maternal hepcidin during pregnancy is essential for maternal and embryo iron homeostasis and health.


Subject(s)
Embryo, Mammalian/metabolism , Fetus/metabolism , Hepcidins/pharmacology , Homeostasis , Iron/metabolism , Maternal Nutritional Physiological Phenomena/drug effects , Placenta/drug effects , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Female , Fetus/drug effects , Maternal-Fetal Exchange , Mice , Mice, Inbred C57BL , Mothers , Placenta/metabolism , Pregnancy , Receptors, Transferrin/metabolism
5.
Int J Vitam Nutr Res ; 92(3-4): 223-230, 2022 Jul.
Article in English | MEDLINE | ID: mdl-32672503

ABSTRACT

The effects of an adequate supply of vitamin A and iron, in comparison with diets low or absent in vitamin A and low in iron, on the mRNA expression of some biomarkers of iron homeostasis as hepcidin (Hamp), transferrin receptor-1 (Tfrc), iron regulatory protein-2 (Ireb2) and ferritin (Fth1) in rats were investigated. 35 male Wistar rats were randomly divided into 5 dietary groups: control, sufficient in iron and insufficient in vitamin A (FesvAi), sufficient in iron and depleted in vitamin A (FesvAd), insufficient in iron and sufficient in vitamin A (FeivAs) and insufficient in both iron and vitamin A (FeivAi). After 6 weeks rats showed no significant effects of variations in vitamin A on the expression of Hamp relative to the control group (FesvAi: 1.37-fold; FesvAd: 1.22-fold); however, iron deficiency showed significant reduction on it relative to the control group (FeivAs: 71.4-fold, P = 0.0004; FeivAi: 16.1-fold, P = 0.0008). Vitamin A deficiency (FesvAd) affects expression of Fth1 independent of low dietary iron in spleen (0.29-fold, P = 0.002) and duodenum (5.15-fold, P = 0.02). Variations of dietary iron and vitamin A showed significant effects relative to the control group for expression of Tfrc in spleen (FesvAd: 0.18-fold, P = 0.01; FeivAs: 0.24-fold, P < 0.0001; FeivAi: 0.42-fold, P = 0.014), Ireb2 in spleen (FeivAs: 3.7-fold, P < 0.0001; FeivAi: 2.9-fold, P < 0.0001) and Ireb2 in duodenum (FeivAs: 2.68-fold, P = 0.012; FeivAi: 2.60-fold, P = 0.014). These results show that vitamin A and iron must be supplied together to regulate some of the main biomarkers of iron metabolism as a strategy to reduce prevalence of iron deficiency anemia.


Subject(s)
Anemia, Iron-Deficiency , Hepcidins , Animals , Biomarkers , Hepcidins/genetics , Hepcidins/metabolism , Hepcidins/pharmacology , Homeostasis , Hormones/pharmacology , Iron/metabolism , Iron, Dietary , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Vitamin A/pharmacology
6.
Med Res Rev ; 40(2): 633-653, 2020 03.
Article in English | MEDLINE | ID: mdl-31471929

ABSTRACT

Abnormally high brain iron, resulting from the disrupted expression or function of proteins involved in iron metabolism in the brain, is an initial cause of neuronal death in neuroferritinopathy and aceruloplasminemia, and also plays a causative role in at least some of the other neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, and Friedreich's ataxia. As such, iron is believed to be a novel target for pharmacological intervention in these disorders. Reducing iron toward normal levels or hampering the increases in iron associated with age in the brain is a promising therapeutic strategy for all iron-related neurodegenerative disorders. Hepcidin is a crucial regulator of iron homeostasis in the brain. Recent studies have suggested that upregulating brain hepcidin levels can significantly reduce brain iron content through the regulation of iron transport protein expression in the blood-brain barrier and in neurons and astrocytes. In this review, we focus on the discussion of the therapeutic potential of hepcidin in iron-associated neurodegenerative diseases and also provide a systematic overview of recent research progress on how misregulated brain iron metabolism is involved in the development of multiple neurodegenerative disorders.


Subject(s)
Hepcidins/therapeutic use , Neurodegenerative Diseases/drug therapy , Animals , Brain/drug effects , Brain/metabolism , Ferroptosis/drug effects , Hepcidins/pharmacology , Humans , Iron/metabolism , Models, Biological
7.
Fish Shellfish Immunol ; 104: 55-61, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32473358

ABSTRACT

Hepcidins, a group of antimicrobial peptides (AMPs), play a key role in the innate immune system of fishes and act against different pathogens. In this study, antimicrobial and immune-inflammatory activity of a synthetic EC-hepcidin1, previously identified from orange-spotted grouper, were evaluated. EC-hepcidin1 showed weak activity against the zoonotic fish pathogen Streptococcus iniae (MIC 100 µg mL-1 and MBC 150 µg mL-1). To study the effect of AMPs in general, and EC-hepcidin1 in particular, a primary cell culture (SC) from the fin tissue of the Caspian Trout (Salmo trutta caspius) was established. The neutral Red method on SC cells revealed that EC-hepcidin1 has no or very low cytotoxic properties. Treatment of cells with either EC-hepcidin1 (150 µg mL-1) or fish pathogen Streptococcus iniae (MOI = 10) and a mixture of both resulted in the up-regulation of gene expression of MHC-UBA, IL-6, and TNFα indicating the modulatory function on inflammatory processes. These findings indicate that EC-hepcidin1 might act as a candidate for modulation of the innate immune system in S. iniae-based infection.


Subject(s)
Gene Expression/drug effects , Hepcidins/pharmacology , Immunologic Factors/pharmacology , Trout/immunology , Animal Fins , Animals , Fish Proteins/pharmacology , Gene Expression/immunology , Immunomodulation/immunology , Primary Cell Culture
8.
Fish Physiol Biochem ; 45(1): 365-374, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30361820

ABSTRACT

Iron overload increases the risk of osteoporosis, which leads to an increase in the incidences of bone fracture after menopause. In vitro studies have demonstrated that excess iron can inhibit osteoblast activity. Hepcidin, a central regulator of iron homeostasis, prevents iron overload, and thus, it is considered to have anti-osteoporosis effects. In this study, a zebrafish model was employed to investigate the therapeutic role of hepcidin in iron overload-induced inhibition of bone formation. Our results show that ferric ammonium citrate (FAC) treatment decreased osteoblast-specific gene expression (runx2a, runx2b, and bglap) and bone mineralization in the zebrafish embryo, accompanied with increased whole-body iron levels and oxidative stress. Bone mineralization and osteoblast-specific gene expression increased with the microinjection of hepcidin-flag Capped-mRNA into zebrafish embryos. Moreover, the whole-body iron content and oxidative stress in the iron-overloaded zebrafish embryos decreased when microinjection of hepcidin preceded the FAC treatment. Therefore, our study suggests that hepcidin could prevent and rescue reduced bone formation caused by FAC treatment by preventing iron absorption.


Subject(s)
Bone Development/drug effects , Ferric Compounds/adverse effects , Hepcidins/pharmacology , Iron Overload/chemically induced , Iron Overload/prevention & control , Quaternary Ammonium Compounds/adverse effects , Animals , Anti-Infective Agents/pharmacology , Dose-Response Relationship, Drug , Down-Regulation , Ferric Compounds/administration & dosage , Ferric Compounds/pharmacology , Gene Expression Regulation/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Quaternary Ammonium Compounds/administration & dosage , Quaternary Ammonium Compounds/pharmacology , Zebrafish
9.
Am J Physiol Gastrointest Liver Physiol ; 315(3): G351-G363, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29792530

ABSTRACT

Manganese (Mn) toxicity arises from nutritional problems, community and occupational exposures, and genetic risks. Mn blood levels are controlled by hepatobiliary clearance. The goals of this study were to determine the cellular distribution of Mn transporters in polarized hepatocytes, to establish an in vitro assay for hepatocyte Mn efflux, and to examine possible roles the Mn transporters would play in metal import and export. For these experiments, hepatocytoma WIF-B cells were grown for 12-14 days to achieve maximal polarity. Immunoblots showed that Mn transporters ZIP8, ZnT10, ferroportin (Fpn), and ZIP14 were present. Indirect immunofluorescence microscopy localized Fpn and ZIP14 to WIF-B cell basolateral domains whereas ZnT10 and ZIP8 associated with intracellular vesicular compartments. ZIP8-positive structures were distributed uniformly throughout the cytoplasm, but ZnT10-positive vesicles were adjacent to apical bile compartments. WIF-B cells were sensitive to Mn toxicity, showing decreased viability after 16 h exposure to >250 µM MnCl2. However, the hepatocytes were resistant to 4-h exposures of up to 500 µM MnCl2 despite 50-fold increased Mn content. Washout experiments showed time-dependent efflux with 80% Mn released after a 4 h chase period. Hepcidin reduced levels of Fpn in WIF-B cells, clearing Fpn from the cell surface, but Mn efflux was unaffected. The secretory inhibitor, brefeldin A, did block release of Mn from WIF-B cells, suggesting vesicle fusion may be involved in export. These results point to a possible role of ZnT10 to import Mn into vesicles that subsequently fuse with the apical membrane and empty their contents into bile. NEW & NOTEWORTHY Polarized WIF-B hepatocytes express manganese (Mn) transporters ZIP8, ZnT10, ferroportin (Fpn), and ZIP14. Fpn and ZIP14 localize to basolateral domains. ZnT10-positive vesicles were adjacent to apical bile compartments, and ZIP8-positive vesicles were distributed uniformly throughout the cytoplasm. WIF-B hepatocyte Mn export was resistant to hepcidin but inhibited by brefeldin A, pointing to an efflux mechanism involving ZnT10-mediated uptake of Mn into vesicles that subsequently fuse with and empty their contents across the apical bile canalicular membrane.


Subject(s)
Biological Transport/physiology , Brefeldin A , Cation Transport Proteins/metabolism , Hepatocytes , Hepcidins , Manganese , Animals , Brefeldin A/metabolism , Brefeldin A/pharmacology , Cell Line , Cell Membrane/metabolism , Cell Polarity , Cytoplasmic Vesicles/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepcidins/metabolism , Hepcidins/pharmacology , Humans , Manganese/metabolism , Manganese/toxicity , Protein Synthesis Inhibitors/pharmacology
10.
Blood ; 128(2): 265-76, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27154187

ABSTRACT

In ß-thalassemia and polycythemia vera (PV), disordered erythropoiesis triggers severe pathophysiological manifestations. ß-Thalassemia is characterized by ineffective erythropoiesis, reduced production of erythrocytes, anemia, and iron overload and PV by erythrocytosis and thrombosis. Minihepcidins are hepcidin agonists that have been previously shown to prevent iron overload in murine models of hemochromatosis and induce iron-restricted erythropoiesis at higher doses. Here, we show that in young Hbb(th3/+) mice, which serve as a model of untransfused ß-thalassemia, minihepcidin ameliorates ineffective erythropoiesis, anemia, and iron overload. In older mice with untransfused ß-thalassemia, minihepcidin improves erythropoiesis and does not alter the beneficial effect of the iron chelator deferiprone on iron overload. In PV mice that express the orthologous JAK2 mutation causing human PV, administration of minihepcidin significantly reduces splenomegaly and normalizes hematocrit levels. These studies indicate that drug-like minihepcidins have a potential as future therapeutics for untransfused ß-thalassemia and PV.


Subject(s)
Erythropoiesis , Hepcidins/pharmacology , Peptides/pharmacology , Polycythemia Vera/metabolism , beta-Thalassemia/metabolism , Amino Acid Substitution , Animals , Hepcidins/genetics , Hepcidins/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mice , Mice, Mutant Strains , Mutation, Missense , Peptides/genetics , Peptides/metabolism , Polycythemia Vera/genetics , beta-Thalassemia/genetics
11.
J Neurochem ; 142(1): 140-152, 2017 07.
Article in English | MEDLINE | ID: mdl-28266714

ABSTRACT

Alzheimer's disease (AD) is characterized by extracellular senile plaques, intracellular neurofibrillary tangles, and neuronal death. Aggregated amyloid-ß (Aß) induces inflammation and oxidative stress, which have pivotal roles in the pathogenesis of AD. Hepcidin is a key regulator of systemic iron homeostasis. Recently, an anti-inflammatory response to hepcidin was reported in macrophages. Under the hypothesis that hepcidin mediates anti-inflammatory response in the brain, in this study, we evaluated the putative anti-inflammatory role of hepcidin on Aß-activated astrocytes and microglia. Primary culture of astrocytes and microglia were treated with Aß, with or without hepcidin, and cytokine levels were then evaluated. In addition, the toxicity of Aß-treated astrocyte- or microglia-conditioned media was tested on neurons, evaluating cellular death and oxidative stress generation. Finally, mice were injected in the right lateral ventricle with Aß, with or without hepcidin, and hippocampus glial activation and oxidative stress were evaluated. Pre-treatment with hepcidin reduced the expression and secretion of TNF-α and IL-6 in astrocytes and microglia treated with Aß. Hepcidin also reduced neurotoxicity and oxidative damage triggered by conditioned media obtained from astrocytes and microglia treated with Aß. Stereotaxic intracerebral injection of hepcidin reduced glial activation and oxidative damage triggered by Aß injection in mice. Overall, these results are consistent with the hypothesis that in astrocytes and microglia hepcidin down-regulates the inflammatory and pro-oxidant processes induced by Aß, thus protecting neighboring neurons. This is a newly described property of hepcidin in the central nervous system, which may be relevant for the development of strategies to prevent the neurodegenerative process associated with AD.


Subject(s)
Amyloid beta-Peptides/toxicity , Antioxidants/pharmacology , Astrocytes/drug effects , Hepcidins/pharmacology , Inflammation/chemically induced , Inflammation/prevention & control , Microglia/drug effects , Peptide Fragments/toxicity , Animals , Culture Media, Conditioned , Hippocampus/pathology , Inflammation/metabolism , Injections, Intraventricular , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Primary Cell Culture , Tumor Necrosis Factor-alpha/metabolism
12.
Article in English | MEDLINE | ID: mdl-28760901

ABSTRACT

Hepcidin, an antimicrobial peptide, was discovered to integrate diverse signals from iron status and an infection threat and orchestrate a series of host-protective responses. Several studies have investigated the antimicrobial role of hepcidin, but the results have been controversial. Here, we aimed to examine the role of hepcidin in bacterial adherence and invasion in vitro We found that porcine hepcidin could decrease the amount of the extracellular pathogen enterotoxigenic Escherichia coli (ETEC) K88 that adhered to cells because it caused the aggregation of the bacteria. However, addition of hepcidin to macrophages infected with the intracellular pathogen Salmonella enterica serovar Typhimurium enhanced the intracellular growth of the pathogen through the degradation of ferroportin, an iron export protein, and then the sequestration of intracellular iron. Intracellular iron was unavailable by use of the iron chelator deferiprone (DFO), which reduced intracellular bacterial growth. These results demonstrate that hepcidin exhibits different functions in extracellular and intracellular bacterial infections, which suggests that different defense strategies should be taken to prevent bacterial infection.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Enterotoxigenic Escherichia coli/drug effects , Escherichia coli Infections/drug therapy , Hepcidins/pharmacology , Salmonella Infections/drug therapy , Salmonella typhimurium/drug effects , Animals , Caco-2 Cells , Cation Transport Proteins/metabolism , Cell Line, Tumor , Deferiprone , Enterotoxigenic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Humans , Iron/metabolism , Macrophages/microbiology , Pyridones/metabolism , Salmonella Infections/microbiology , Salmonella typhimurium/growth & development , Salmonella typhimurium/pathogenicity , Swine
13.
Small ; 13(15)2017 04.
Article in English | MEDLINE | ID: mdl-28195425

ABSTRACT

Although numerous toxicological studies have been performed on carbon nanotubes (CNTs), a few studies have investigated their secondary and indirect effects beyond the primary target tissues/organs. Here, a cascade of events are investigated: the initiating event and the subsequent key events necessary for the development of phenotypes, namely CNT-induced pro-inflammatory effects on iron homeostasis and red blood cell formation, which are linked to anemia of inflammation (AI). A panel of CNTs are prepared including pristine multiwall CNTs (P-MWCNTs), aminated MWCNTs (MWCNTs-NH2 ), polyethylene glycol MWCNTs (MWCNTs-PEG), polyethyleneimine MWCNTs (MWCNTs-PEI), and carboxylated MWCNTs (MWCNTs-COOH). It has been demonstrated that all CNT materials provoke inflammatory cytokine interleukin-6 (IL-6) production and stimulate hepcidin induction, associated with disordered iron homeostasis, irrespective of exposure routes including intratracheal, intravenous, and intraperitoneal administration. Meanwhile, PEG and COOH modifications can ameliorate the activation of IL-6-hepcidin signaling. Long-term exposure of MWCNTs results in AI and extramedullary erythropoiesis. Thus, an adverse outcome pathway is identified: MWCNT exposure leads to inflammation, hepatic hepcidin induction, and disordered iron metabolism. Together, the combined data depict the hazardous secondary toxicity of CNTs in incurring anemia through inflammatory pathway. This study will also open a new avenue for future investigations on CNT-induced indirect and secondary adverse effects.


Subject(s)
Anemia/chemically induced , Homeostasis , Inflammation/chemically induced , Iron/metabolism , Nanotubes, Carbon/adverse effects , Anemia/pathology , Animals , Erythrocytes/drug effects , Erythrocytes/metabolism , Hematopoiesis, Extramedullary , Hepcidins/pharmacology , Homeostasis/drug effects , Inflammation/pathology , Inhalation Exposure , Interleukin-6/metabolism , Liver/pathology , Lung/pathology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice , Nanotubes, Carbon/ultrastructure , Spleen/pathology , Splenomegaly/pathology
14.
J Mol Recognit ; 30(1)2017 01.
Article in English | MEDLINE | ID: mdl-27507710

ABSTRACT

Hepcidin, a liver-expressed antimicrobial peptide, has been demonstrated to act as an iron regulatory hormone as well as to exert a wide spectrum of antimicrobial activity. The aim of this work was the expression, as secreted peptide, purification, and characterization of a new recombinant polyHis-tagged camel hepcidin (HepcD-His) in yeast Pichia pastoris. The use of this eukaryotic expression system, for the production of HepcD-His, having 6 histidine residues at its C terminus, was simpler and more efficient compared with the use of the prokaryotic system Escherichia coli. Indeed, a single purification step was required to isolate the soluble hepcidin with purity estimated more that 94% and a yield of 2.8 against 0.2 mg/L for the E coli system. Matrix-assisted laser desorption/ionization time-of-flight (TOF)/TOF mass spectrometry of the purified HepcD-His showed 2 major peaks at m/z 4524.64 and 4634.56 corresponding to camel hepcidin with 39 and 40 amino acids. Evaluation of disulfide bond connectivity with the Ellman method showed an absence of free thiol groups, testifying that the 8 cysteine residues in the peptide are displayed, forming 4 disulfide bridges. Circular dichroism spectroscopy showed that camel hepcidin structure was significantly modified at high temperature of 90°C and returns to its original structure when incubation temperature drops back to 20°C. Interestingly, this peptide showed also a greater bactericidal activity, at low concentration of 9.5µM, against E coli, than the synthetic analog DH3. Thus, the production, at a large scale, of the recombinant camel hepcidin, HepcD-His, may be helpful for future therapeutic applications including bacterial infection diseases.


Subject(s)
Hepcidins/chemistry , Hepcidins/isolation & purification , Histidine/chemistry , Pichia/genetics , Animals , Camelus , Circular Dichroism , Cloning, Molecular , Disulfides/chemistry , Escherichia coli/drug effects , Hepcidins/genetics , Hepcidins/pharmacology , Models, Molecular , Pichia/metabolism , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thermodynamics
15.
Blood ; 125(14): 2265-75, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25662334

ABSTRACT

Regulation of iron metabolism and innate immunity are tightly interlinked. The acute phase response to infection and inflammation induces alterations in iron homeostasis that reduce iron supplies to pathogens. The iron hormone hepcidin is activated by such stimuli causing degradation of the iron exporter ferroportin and reduced iron release from macrophages, suggesting that hepcidin is the crucial effector of inflammatory hypoferremia. Here, we report the discovery of an acute inflammatory condition that is mediated by Toll-like receptors 2 and 6 (TLR2 and TLR6) and which induces hypoferremia in mice injected with TLR ligands. Stimulation of TLR2/TLR6 triggers profound decreases in ferroportin messenger RNA and protein expression in bone marrow-derived macrophages, liver, and spleen of mice without changing hepcidin expression. Furthermore, C326S ferroportin mutant mice with a disrupted hepcidin/ferroportin regulatory circuitry respond to injection of the TLR2/6 ligands FSL1 or PAM3CSK4 by ferroportin downregulation and a reduction of serum iron levels. Our findings challenge the prevailing role of hepcidin in hypoferremia and suggest that rapid hepcidin-independent ferroportin downregulation in the major sites of iron recycling may represent a first-line response to restrict iron access for numerous pathogens.


Subject(s)
Cation Transport Proteins/physiology , Hepcidins/pharmacology , Inflammation/physiopathology , Iron Deficiencies , Toll-Like Receptor 2/physiology , Toll-Like Receptor 6/physiology , Acute Disease , Animals , Anti-Infective Agents/pharmacology , Blotting, Western , Cells, Cultured , HeLa Cells , Humans , Lipopeptides/pharmacology , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 2/agonists , Toll-Like Receptor 6/agonists
16.
Mol Cell Biochem ; 427(1-2): 201-208, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27995414

ABSTRACT

Recent investigation has shown that the liver-derived iron-regulating hormone, hepcidin, can potentiate intestinal calcium absorption in hemizygous ß-globin knockout thalassemic (BKO) mice. Since the upregulation of Fe2+ and H+ cotransporter, divalent metal transporter (DMT)-1, has been shown to correlate with thalassemia-induced intestinal calcium absorption impairment, the inhibition of the apical Na+/H+ exchanger (NHE)-3 that is essential for cytoplasmic pH regulation and transepithelial sodium absorption was hypothesized to negatively affect hepcidin action. Herein, the positive effect of hepcidin on the duodenal calcium transport was evaluated using Ussing chamber technique. The results showed that BKO mice had lower absorptive surface area and duodenal calcium transport than wild-type mice. Besides, paracellular transport of zinc in BKO mice was compromised. Hepcidin administration completely restored calcium transport. Since this hepcidin action was totally abolished by inhibitors of the basolateral calcium transporters, Na+/Ca2+ exchanger (NCX1) and plasma membrane Ca2+-ATPase (PMCA1b), the enhanced calcium flux potentially occurred through the transcellular pathway rather than paracellular pathway. Interestingly, the selective NHE3 inhibitor, 100 nM tenapanor, markedly inhibited hepcidin-enhanced calcium transport. Accordingly, hepcidin is one of the promising therapeutic agents for calcium malabsorption in ß-thalassemia. It mainly stimulates the transcellular calcium transport across the duodenal epithelium in an NHE3-dependent manner.


Subject(s)
Calcium/metabolism , Duodenum/metabolism , Hepcidins/pharmacology , Isoquinolines/pharmacology , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sulfonamides/pharmacology , Thalassemia/metabolism , beta-Globins/metabolism , Animals , Duodenum/pathology , Female , Ion Transport/drug effects , Ion Transport/genetics , Mice , Mice, Knockout , Plasma Membrane Calcium-Transporting ATPases/genetics , Plasma Membrane Calcium-Transporting ATPases/metabolism , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Thalassemia/genetics , Thalassemia/pathology , beta-Globins/genetics
17.
Pharmacol Res ; 115: 242-254, 2017 01.
Article in English | MEDLINE | ID: mdl-27867027

ABSTRACT

Iron is an essential biogenic element for both prokaryotic and eukaryotic cells. In humans iron is present in hundreds of different metalloproteins. The peptide hormone hepcidin serves as a master regulator of iron homeostasis on the level of single cells and whole organism - by altering cell surface expression of cellular iron exporter - protein ferroportin. Altered levels of extracellular hepcidin lead to pathological conditions such as hemochromatosis and iron loading or, on the other side, iron restrictive anemias. Therapeutic modulation of hepcidin is a new and promising approach to treatment of these conditions. In this review, a summary of the current knowledge of hepcidin function, regulation and pathological involvements are provided, followed by a section covering the therapeutic potential of hepcidin and the current strategies how to modulate its levels and biological functions for therapeutic purposes.


Subject(s)
Hepcidins/pharmacology , Hepcidins/therapeutic use , Iron/metabolism , Animals , Cation Transport Proteins/metabolism , Homeostasis/drug effects , Humans
18.
Am J Hematol ; 92(10): 1052-1061, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28681497

ABSTRACT

The hepcidin-ferroportin axis underlies the pathophysiology of many iron-associated disorders and is a key target for the development of therapeutics for treating iron-associated disorders. The aims of this study were to investigate the dynamics of hepcidin-mediated ferroportin internalization and the consequences of a novel disease-causing mutation on ferroportin function. Specific reagents for ferroportin are limited; we developed and characterized antibodies against the largest extracellular loop of ferroportin and developed a novel cell-based assay for studying hepcidin-ferroportin function. We show that hepcidin-mediated ferroportin internalization is a rapid process and could be induced using low concentrations of hepcidin. Targeted next-generation sequencing utilizing an iron metabolism gene panel developed in our group identified a novel ferroportin p.D84E variant in a patient with iron overload. Wild-type and mutant ferroportin constructs were generated, transfected into HEK293 cells and analysed using an all-in-one flow-cytometry-based assay to study the effects on hepcidin-mediated internalization and iron transport. Consistent with the classical phenotype of ferroportin disease, the p.D84E mutation results in an inability to transport iron and hepcidin insensitivity. These results validate a recently proposed 3D-structural model of ferroportin and highlight the significance of this variant in the structure and function of ferroportin. Our novel ferroportin antibody and assay will be valuable tools for investigating the regulation of hepcidin/ferroportin function and the development of novel approaches for the therapeutic modulation of iron homeostasis.


Subject(s)
Cation Transport Proteins/metabolism , Hepcidins/metabolism , Iron Metabolism Disorders/genetics , Iron/metabolism , Mutation , Biological Assay , Cation Transport Proteins/blood , Cation Transport Proteins/genetics , Female , Flow Cytometry , HEK293 Cells , Hepcidins/pharmacology , Humans , Iron Metabolism Disorders/blood , Iron Metabolism Disorders/metabolism , Kinetics , Protein Transport , Receptors, Cell Surface/metabolism , Transfection
19.
J Am Soc Nephrol ; 27(3): 835-46, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26293821

ABSTRACT

The iron-regulatory peptide hepcidin exhibits antimicrobial activity. Having previously shown hepcidin expression in the kidney, we addressed its role in urinary tract infection (UTI), which remains largely unknown. Experimental UTI was induced in wild-type (WT) and hepcidin-knockout (Hepc-/-) mice using the uropathogenic Escherichia coli CFT073 strain. Compared with infected WT mice, infected Hepc-/- mice showed a dramatic increase in renal bacterial load. Moreover, bacterial invasion was significantly dampened by the pretreatment of WT mice with hepcidin. Infected Hepc-/- mice exhibited decreased iron accumulation in the renal medulla and significant attenuation of the renal inflammatory response. Notably, we demonstrated in vitro bacteriostatic activity of hepcidin against CFT073. Furthermore, CFT073 repressed renal hepcidin, both in vivo and in cultured renal cells, and reduced phosphorylation of SMAD kinase in vivo, suggesting a bacterial strategy to escape the antimicrobial activities of hepcidin. In conclusion, we provide new mechanisms by which hepcidin contributes to renal host defense and suggest that targeting hepcidin offers a strategy to prevent bacterial invasion.


Subject(s)
Anti-Infective Agents/pharmacology , Escherichia coli Infections/metabolism , Escherichia coli/drug effects , Hepcidins/metabolism , Hepcidins/pharmacology , Urinary Tract Infections/metabolism , Animals , Anti-Infective Agents/metabolism , Bacterial Load/genetics , Cells, Cultured , Colony Count, Microbial , Cytokines/metabolism , Escherichia coli Infections/microbiology , Female , Hepcidins/genetics , Iron/metabolism , Kidney Medulla/cytology , Kidney Medulla/metabolism , Kidney Medulla/microbiology , Mice , Mice, Inbred CBA , Mice, Knockout , Nephritis/metabolism , Nephritis/microbiology , Nephritis/pathology , Neutrophils , Phosphorylation , RNA, Messenger/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Smad Proteins/metabolism , Urinary Tract Infections/microbiology
20.
Am J Physiol Endocrinol Metab ; 311(1): E214-23, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27245334

ABSTRACT

Previously, ß-thalassemia, an inherited anemic disorder with iron overload caused by loss-of-function mutation of ß-globin gene, has been reported to induce osteopenia and impaired whole body calcium metabolism, but the pathogenesis of aberrant calcium homeostasis remains elusive. Herein, we investigated how ß-thalassemia impaired intestinal calcium absorption and whether it could be restored by administration of 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] or hepcidin, the latter of which was the liver-derived antagonist of intestinal iron absorption. The results showed that, in hemizygous ß-globin knockout (BKO) mice, the duodenal calcium transport was lower than that in wild-type littermates, and severity was especially pronounced in female mice. Both active and passive duodenal calcium fluxes in BKO mice were found to be less than those in normal mice. This impaired calcium transport could be restored by 7-day 1,25(OH)2D3 treatment. The 1,25(OH)2D3-induced calcium transport was diminished by inhibitors of calcium transporters, e.g., L-type calcium channel, NCX1, and PMCA1b, as well as vesicular transport inhibitors. Interestingly, the duodenal calcium transport exhibited an inverse correlation with transepithelial iron transport, which was markedly enhanced in thalassemic mice. Thus, 3-day subcutaneous hepcidin injection and acute direct hepcidin exposure in the Ussing chamber were capable of restoring the thalassemia-associated impairment of calcium transport; however, the positive effect of hepcidin on calcium transport was completely blocked by proteasome inhibitors MG132 and bortezomib. In conclusion, both 1,25(OH)2D3 and hepcidin could be used to alleviate the ß-thalassemia-associated impairment of calcium absorption. Therefore, our study has shed light on the development of a treatment strategy to rescue calcium dysregulation in ß-thalassemia.


Subject(s)
Calcitriol/pharmacology , Calcium/metabolism , Duodenum/drug effects , Hepcidins/pharmacology , Intestinal Absorption/drug effects , Iron/metabolism , beta-Thalassemia/metabolism , Animals , Bortezomib/pharmacology , Calcium Channel Blockers/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Duodenum/metabolism , Female , Hemizygote , Leupeptins/pharmacology , Male , Mice , Mice, Knockout , Plasma Membrane Calcium-Transporting ATPases/antagonists & inhibitors , Sodium-Calcium Exchanger/antagonists & inhibitors , Vesicular Transport Proteins/antagonists & inhibitors , beta-Globins/genetics , beta-Thalassemia/genetics
SELECTION OF CITATIONS
SEARCH DETAIL