Increased Expression Of NGF In Hepatocytes Is An Early Event In Iron Overloaded Mouse By Transcriptome Analysis

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2194-2194
Author(s):  
Masayo Yamamoto ◽  
Hiroki Tanaka ◽  
Lynda Addo ◽  
Satoshi Ito ◽  
Motohiro Shindo ◽  
...  

Abstract The liver plays a central role in iron metabolism by storing and sensing the amounts of iron in the body. The dietary iron from duodenum and recycled-iron by reticuloendothelial system are the main source of body iron. When excess iron enters the liver, hepatocytes secrete hepcidin, an anti-microbial peptide which negatively regulates iron excretion from enterocytes and macrophages, and stores the excess iron as ferritin-bound iron. A dysfunction of this regulatory system causes iron overload in the liver. Aberrant iron accumulation in the liver is found in hereditary hemochromatosis and chronic liver disease, and this is considered to be an exacerbating factor in liver cirrhosis and hepatocellular carcinoma. It is therefore important to understand the precise molecular events that take place as a result of iron accumulation during the early stages of iron overload. In the present study, we performed transcriptome analysis on the liver of dietary iron overloaded mice. Transcriptome analysis using a high throughput sequencer is capable of comprehensive analysis with high sensitivity. We hypothesized that this method will be suitable in detecting the changes in gene expression induced by iron overload, even in slightly expressed genes. C57B1/6 mice were fed a normal diet, and a 2.5% iron diet for 8 weeks. Serum and liver tissue samples were then collected, and histological analysis showed the features of early stage iron overload without significant hepatic damage in the iron-fed mice. From the results of the transcriptome analysis, we found that nerve growth factor (NGF) was significantly expressed in the slightly iron overloaded liver. This observation was also confirmed by real time RT-PCR, Western blotting and immunohistochemistry. Similarly, NGF upregulation was induced in mice primary hepatocytes cultured in conditioned iron overloaded medium (with high concentration of holo-transferrin or ferric ammonium citrate). Furthermore, immunohistochemical analysis showed that TrkA, a high affinity NGF receptor, was expressed in liver sinusoidal endothelial cells (LSECs). Using scanning electron microscopy, we sought to examine any morphological changes in the sinusoids of the iron overloaded liver and observed that although sieve plate structures (so-called ‘fenestrae’) were found in the LSECs of mice fed a normal diet, they were not visible in the iron-fed mice. The loss of fenestrae was also observed in the LSECs of mice that received intraperitoneal injections of NGF. In cultured isolated primary LSECs, treatment with NGF, or conditioned medium from iron overloaded primary hepatocytes reduced the fenestrae while the anti-NGF neutralization antibody or TrkA inhibitor K252a cancelled this effect. In addition, a fresh iron overloaded medium did not reduce the fenestrae in primary LSECs, indicating that iron itself has no direct effects on the fenestrae in LSECs. LSECs constitute the sinusoidal wall in the liver and can be regarded as unique capillaries which differ from other capillaries in the body due to the presence of fenestrae which lack a diaphragm, and are therefore open connections between the lumen of the sinusoid and the space of Disse. The fenestrae in LSECs therefore play an important role in the exchange of solutes between the lumen of the sinusoid and hepatocytes. The results of this study indicate that iron accumulation induces the expression of NGF in hepatoctyes, which in turn leads to the loss of fenestrae in LSECs via TrkA. This phenomenon may therefore contribute to the defensive machinery against iron accumulation in hepatocytes in the early stages of iron overload. These data further suggest a novel function of NGF in the regulation of iron transport. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4815-4815
Author(s):  
David T Tran ◽  
Charles O Noble ◽  
Mark E Hayes ◽  
Francis C Szoka

Abstract Introduction: Long-term red blood cell transfusions effectively sustains patients who have β-thalassemia, sickle cell anemia, and myelodysplastic syndromes but they also lead to excess iron accumulation in the body. Iron overload is a major cause of morbidity and mortality in transfusion dependent patients. Chelation therapy reverses iron accumulation but marketed chelators have drawbacks such as: long infusions of deferoxamine (DFO, Novartis), large oral tablets with adverse effects (Exjade, Novartis), or twice daily oral dosing (Ferriprox, ApoPharma). These attributes contribute to poor compliance and poor outcomes in iron overload patients. To overcome long infusions and high doses of current therapies we have devised a stable nanoliposome encapsulated DFO (LDFO) for the treatment of iron overload. Methods: LDFO composed of saturated soy phosphatidylcholine and cholesterol (3/2 molar ratio) is manufactured using a proprietary remote loading method that provides high encapsulation of DFO in 90 nm diameter liposomes. For pharmacokinetics and bioavailability studies, DFO and lipid concentrations in CF-1 mice plasma and tissues were analyzed by HPLC utilizing an in-house method. For iron removal efficacy studies, CF-1 mice were overloaded with iron dextran and after 10 days washout were treated with 100 mg/kg LDFO or unencapsulated DFO. Animals were sacrificed 5 days post treatment and tissue iron was measured by a ferrozine based spectroscopic assay. Results: The manufacturing method to prepare LDFO results in a 300 g DFO/mole lipid encapsulation ratio. The formulation has greater than 6 months stability at 4 ºC. LDFO is long circulating and the DFO is bioavailable. At 24 hr post I.V. injection, there is 30% ID DFO in plasma and 10% ID DFO/g in liver whereas unencapsulated DFO is not detectable. Preclinical single dose safety studies in CF-1 mice indicate that LDFO is well tolerated at 300 mg/kg I.V. and 1250 mg/kg I.P. In the iron dextran overload model, LDFO greatly reduces iron levels in the liver and spleen. The absolute efficiency of LDFO is greater than 50% on a mole LDFO injected /mole iron removed from the liver (P<0.0004, n=4) and spleen (P<0.01, n=4). This is corroborated by an elevated iron accumulation in urine and feces from LDFO. Conclusion: LDFO effectively removes iron from the liver and spleen with an overall molar efficiency > 50%. This high efficacy could lead to a dramatically improved treatment that increases compliance and provides substantially better management of iron overload than current treatments in patients suffering from iron overload conditions. Disclosures Tran: ZoneOne Pharma, Inc.: Employment. Noble:ZoneOne Pharma, Inc.: Employment, Equity Ownership. Hayes:ZoneOne Pharma, Inc.: Employment, Equity Ownership. Szoka:ZoneOne Pharma, Inc.: Consultancy, Equity Ownership.


2020 ◽  
Vol 19 (3) ◽  
pp. 158-163
Author(s):  
E. E. Nazarova ◽  
D. A. Kupriyanov ◽  
G. A. Novichkova ◽  
G. V. Tereshchenko

The assessment of iron accumulation in the body is important for the diagnosis of iron overload syndrome or planning and monitoring of the chelation therapy. Excessive iron accumulation in the organs leads to their toxic damage and dysfunction. Until recently iron estimation was performed either directly by liver iron concentration and/or indirectly by measuring of serum ferritin level. However, noninvasive iron assessment by Magnetic resonance imaging (MRI) is more accurate method unlike liver biopsy or serum ferritin level test. In this article, we demonstrate the outlines of non-invasive diagnostics of iron accumulation by MRI and its specifications.


Blood ◽  
1963 ◽  
Vol 22 (4) ◽  
pp. 406-415 ◽  
Author(s):  
MARCEL E. CONRAD ◽  
WILLIAM H. CROSBY ◽  
Betty Merrill

Abstract Radioautographic studies provide evidence to support a concept of the mechanism whereby the small intestine controls absorption of iron. Three different states of the body’s iron stores have been considered in this regard: iron excess, iron deficiency and normal iron repletion. As the columnar epithelial cells of the duodenal villi are formed they incorporate a portion of intrinsic iron from the body’s iron store, the amount depending upon the body’s requirement for new iron. It is predicated that with iron excess the iron-receptor mechanism in these cells is saturated with intrinsic iron; this then prevents the cell from accepting dietary iron. In the normal state of iron repletion the receptor mechanism remains partly unsaturated, allowing small amounts of dietary iron to enter the cell. Part of this proceeds into the body to satisfy any metabolic requirement for iron. Part is retained in the mucosal epithelial cells to complete the saturation of the iron-receptor mechanism. This bound iron is subsequently lost when the epithelial cells are sloughed at the end of their life cycle. In iron deficiency it is postulated that the receptor system is inactive or diminished so that entry of dietary iron into the body is relatively uninhibited.


2018 ◽  
Vol 1 (1) ◽  

Iron is physiologically essential to life, but biochemically it is harmful because of its evident -but unappreciated- oxidative and inflammatory tissue power when it accumulates, is dosed in excess, or is free; and that because, after entering the body, unlike any other metal, its elimination is almost non-existent in man; thus, metal is a powerful promoter of chronic degenerative diseases, from diabetes, neurodegeneration to cancer, through extensive coronary and cardio-cerebrovascular disease; modifying its clinical expressivity and accelerating its severity. Iron is a powerful oxidizing and inflammatory agent, and its accumulation causes and promotes the proliferation of cancer cells in particular, both in animals and in humans. Free and accumulated iron triggers a powerful uncontrolled Cell Proliferation, permanently feeding the survival of the neoplastic cell. After more than 50 years of experimental and preclinical studies, it is clearly demonstrating the carcinogenicity of iron; and this is also proven in humans, from breast cancer and endometrium, in women, to cancer of the colon-rectum, prostate, and pancreas in men. In Western men and women, the reductions in iron deposits have an important anti-tumor and preventive effect for the development of cancer or diabetes, two entities biologically interrelated by the states of Resistance to Insulin, an inflammatory state that favors the development of malignant neoplasms, and can accelerate its aggressiveness. It is the chronic excess of insulin or its Tissue Resistance, the biological event and the clinical syndrome that increases the cancerous power of excess iron, both silent epidemics in modern man. Moderate increases in body iron levels increase the risk of acquiring cancer, and raise the level of their mortality. And its deficiency or chelation in vivo decreases the Tumor growth (Wang F, Elliott RL, Head JF: Inhibitory effect of deferoxamine mesylate and low iron diet on the 13762NF rat mammary adenocarcinoma Anticancer Res. 1999 Jan-Feb; 19 (1A): 445-50). If excess iron mediates and increases the risk of cancer associated with Insulin resistance, any subject with this syndrome can minimize any associated health risks (and their increased risk of cancer), avoiding iron-rich diets and donating blood with regularity; Iron is the metal that causes “exponential” and punctual mutations and fusion of genes through chromosomal translocations, constituting the greatest risk factor for human carcinogenesis. Iron is physiologically essential for life but biochemically dangerous. Chronic accumulation of iron causes pantropic organ damage and excess body iron play an important role in carcinogenesis, coronary artery disease, neurodegenerative disease, stroke and inflammatory disorders. Iron is very slowly excreted from humans once it is absorbed into the body. The significance of iron excess has been markedly underestimated, despite the fact that iron overloading disorders are as common place in the US white population. Iron-overload and catalytic iron promotes activation of oxidative responsive transcription factors and pro-inflammatory cytokines that increase cancer extension and aggravate them. There is accumulative evidence for iron as a carcinogenic metal in epidemiological, clinical, animal, and cell culture studies. The role of iron in various cancers, such as colorectal and liver cancer was demonstrated. Recent advancements on the molecular mechanisms of iron carcinogenesis evolved the Insulin-resistance generation and promotion, fisiopatologic condition that is not only permissive, but may be generated cancer and promoting it. Unlike other nutritional metals, iron is highly conserved: toxicity due to excess iron can occur either acutely after a single dose or chronically due to excessive accumulation in the body from diet. In vivo studies have demonstrated that an iron deficiency induced by either feeding a low iron diet injecting the iron chelator deferoxamine mesylate decreases tumor growth (Wang F, Elliott RL, Head JF: Inhibitory effect of deferoxamine mesylate and low iron diet on the 13762NF rat mammary adenocarcinoma Anticancer Res. 1999 Jan-Feb;19(1A):445-50). Iron supplementation has at times proven ineffective and even detrimental to health. Thus, iron excess may mediate the increased cancer risk associated with insulin resistance and heme-rich diets, and subjects who are insulin resistant can minimize any health risk associated with iron overload by avoiding heme-rich flesh foods and donating blood regularly. The energy that sustains cancer cells derived preferentially from glycolysis depends on the gene p53 deficiency-iron induced. This nutrient is postulated to contribute to the initiation of cancer in vivo, but iron overload initiates and sustain cancer development if chronic infection or insulin resistance conditions are present. Cancer cells require considerably more iron than normal cells. Since iron catalytic can induce driver point mutation and create fusion genes through chromosomal translocations, iron overload is one of the most important risk factors in human carcinogenesis. Because free iron may play a catalytic role in “spontaneous” mutagenesis, moderately elevated iron stores increased overall risk for cáncer.


2018 ◽  
Vol 47 (1) ◽  
pp. 119-128 ◽  
Author(s):  
Ming-Yue Sun ◽  
Miao Zhang ◽  
Shui-Ling Chen ◽  
Shu-Ping Zhang ◽  
Chun-Yu Guo ◽  
...  

Background/Aims: Systemic iron homeostasis is strictly governed in mammals; however, disordered iron metabolism (such as excess iron burden) is recognized as a risk factor for various types of diseases including AS (Atherosclerosis). The hepcidin-ferroportin axis plays the key role in regulation of iron homeostasis and modulation of this signaling could be a potential therapeutic strategy in the treatment of these diseases. TMP (Tetramethylpyrazine) has been reported to have therapeutical effect on AS. Here, we aimed to investigate the effect of iron overload under hyperlipidemia condition on the endothelial injury, inflammation and oxidative stress by employing FPN1 Tek-cre mouse model with or without TMP intervention. Methods: Subjects for this study were 80 FPN1 Tek-cre mice and 40 C57BL/6 mice and we randomly divided them into six groups: Group N: C57BL/6 mice with normal diet, Group M: C57BL/6 mice with high-fat diet, Group FN: FPN1 Tek-cre mice with normal diet, Group FNT: FPN1 Tek-cre mice with normal diet and TMP injection, Group FM: FPN1 Tek-cre mice with high-fat diet, Group FMT: FPN1 Tek-cre mice with high-fat diet and TMP injection. After seven days of treatment, blood samples were obtained to detect the levels of blood lipids, Hepcidin, NO, ET-1, ROS, MDA, SOD, IL-1, IL-6 and TNF-α respectively. The liver and aorta were used for testing the lipid deposition by using hematoxylin and eosin(HE). Results: Hyperlipidemia could cause iron overload in the aorta and increased serum hepcidin level, particularly in FPN1 Tek-cre mice, and can be reversed by TMP intervention. Knockout of Fpn1 induced increase of serum hepcidin, exacerbated endothelial dysfunction, oxidative stress and inflammatory response, particularly under hyperlipidemia condition. TMP intervention attenuated these processes. Conclusions: Our study signifies the potential application of certain natural compounds to ameliorating iron disorders induced by hyperlipidemia and protecting on endothelial function through modulation of hepcidin-ferroportin signaling.


2019 ◽  
Vol 20 (3) ◽  
pp. 609 ◽  
Author(s):  
Giuseppina Camiolo ◽  
Daniele Tibullo ◽  
Cesarina Giallongo ◽  
Alessandra Romano ◽  
Nunziatina Parrinello ◽  
...  

Iron toxicity is associated with organ injury and has been reported in various clinical conditions, such as hemochromatosis, thalassemia major, and myelodysplastic syndromes. Therefore, iron chelation therapy represents a pivotal therapy for these patients during their lifetime. The aim of the present study was to assess the iron chelating properties of α-lipoic acid (ALA) and how such an effect impacts on iron overload mediated toxicity. Human mesenchymal stem cells (HS-5) and animals (zebrafish, n = 10 for each group) were treated for 24 h with ferric ammonium citrate (FAC, 120 µg/mL) in the presence or absence of ALA (20 µg/mL). Oxidative stress was evaluated by reduced glutathione content, reactive oxygen species formation, mitochondrial dysfunction, and gene expression of heme oxygenase-1b and mitochondrial superoxide dismutase; organ injury, iron accumulation, and autophagy were measured by microscopical, cytofluorimetric analyses, and inductively coupled plasma‒optical mission Spectrometer (ICP-OES). Our results showed that FAC results in a significant increase of tissue iron accumulation, oxidative stress, and autophagy and such detrimental effects were reversed by ALA treatment. In conclusion, ALA possesses excellent iron chelating properties that may be exploited in a clinical setting for organ preservation, as well as exhibiting a good safety profile and low cost for the national health system.


2020 ◽  
Vol 10 (1) ◽  
pp. e05-e05
Author(s):  
Bijan Keikhaei ◽  
Neda Farmani-Anooshe ◽  
Mohammad Bahadoram ◽  
Mohammad-Reza Mahmoudian-Sani ◽  
Kosar Alikhani ◽  
...  

Thalassemic syndromes are the most common genetic diseases in the world that are related to blood transfusion and iron overload in the body. In ß-thalassemia major multiple blood transfusions due to ineffective erythropoiesis lead to iron excess in the body. Iron chelating agent deferoxamine is used to treat chronic iron overload in patients with β-thalassemia in an attempt to reduce morbidity and mortality related to deposition of excess iron in body tissue. However, a very short half-time, the need of repetitive injections and non-specialized distribution in tissues can lead to side effects, such as ophthalmic and renal complications, neurological, skeletal changes and hearing loss, headaches, and infection too. Patients should be monitored periodically for complications. The risk of toxic effects in doses of more than 50 mg/kg/d is higher. Keeping deferoxamine therapeutic index can avoid drug overdose and side effects.


Antioxidants ◽  
2021 ◽  
Vol 10 (11) ◽  
pp. 1818
Author(s):  
Christine Fischer ◽  
Chiara Volani ◽  
Timea Komlódi ◽  
Markus Seifert ◽  
Egon Demetz ◽  
...  

Iron is an essential co-factor for many cellular metabolic processes, and mitochondria are main sites of utilization. Iron accumulation promotes production of reactive oxygen species (ROS) via the catalytic activity of iron species. Herein, we investigated the consequences of dietary and genetic iron overload on mitochondrial function. C57BL/6N wildtype and Hfe−/− mice, the latter a genetic hemochromatosis model, received either normal diet (ND) or high iron diet (HI) for two weeks. Liver mitochondrial respiration was measured using high-resolution respirometry along with analysis of expression of specific proteins and ROS production. HI promoted tissue iron accumulation and slightly affected mitochondrial function in wildtype mice. Hepatic mitochondrial function was impaired in Hfe−/− mice on ND and HI. Compared to wildtype mice, Hfe−/− mice on ND showed increased mitochondrial respiratory capacity. Hfe−/− mice on HI showed very high liver iron levels, decreased mitochondrial respiratory capacity and increased ROS production associated with reduced mitochondrial aconitase activity. Although Hfe−/− resulted in increased mitochondrial iron loading, the concentration of metabolically reactive cytoplasmic iron and mitochondrial density remained unchanged. Our data show multiple effects of dietary and genetic iron loading on mitochondrial function and linked metabolic pathways, providing an explanation for fatigue in iron-overloaded hemochromatosis patients, and suggests iron reduction therapy for improvement of mitochondrial function.


2010 ◽  
Vol 64 (4) ◽  
pp. 281-286 ◽  
Author(s):  
Manuel Muñoz ◽  
José Antonio García-Erce ◽  
Ángel Francisco Remacha

Iron functionsIron is an essential micronutrient, as it is required for satisfactory erythropoietic function, oxidative metabolism and cellular immune response.Iron physiologyAbsorption of dietary iron (1–2 mg/day) is tightly regulated and just balanced against iron loss because there are no active iron excretory mechanisms. Dietary iron is found in haem (10%) and non-haem (ionic, 90%) forms, and their absorption occurs at the apical surface of duodenal enterocytes via different mechanisms. Iron is exported by ferroportin 1 (the only putative iron exporter) across the basolateral membrane of the enterocyte into the circulation (absorbed iron), where it binds to transferrin and is transported to sites of use and storage. Transferrin-bound iron enters target cells—mainly erythroid cells, but also immune and hepatic cells—via receptor-mediated endocytosis. Senescent erythrocytes are phagocytosed by reticuloendothelial system macrophages, haem is metabolised by haem oxygenase, and the released iron is stored as ferritin. Iron will be later exported from macrophages to transferrin. This internal turnover of iron is essential to meet the requirements of erythropoiesis (20–30 mg/day). As transferrin becomes saturated in iron-overload states, excess iron is transported to the liver, the other main storage organ for iron, carrying the risk of free radical formation and tissue damage.Regulation of iron homoeostasisHepcidin, synthesised by hepatocytes in response to iron concentrations, inflammation, hypoxia and erythropoiesis, is the main iron-regulatory hormone. It binds ferroportin on enterocytes, macrophages and hepatocytes triggering its internalisation and lysosomal degradation. Inappropriate hepcidin secretion may lead to either iron deficiency or iron overload.


Sign in / Sign up

Export Citation Format

Share Document