scholarly journals The Impact of Iron Overload and Ferroptosis on Reproductive Disorders in Humans: Implications for Preeclampsia

2019 ◽  
Vol 20 (13) ◽  
pp. 3283 ◽  
Author(s):  
Shu-Wing Ng ◽  
Sam G. Norwitz ◽  
Errol R. Norwitz

Iron is an essential element for the survival of most organisms, including humans. Demand for iron increases significantly during pregnancy to support growth and development of the fetus. Paradoxically, epidemiologic studies have shown that excessive iron intake and/or high iron status can be detrimental to pregnancy and is associated with reproductive disorders ranging from endometriosis to preeclampsia. Reproductive complications resulting from iron deficiency have been reviewed elsewhere. Here, we focus on reproductive disorders associated with iron overload and the contribution of ferroptosis—programmed cell death mediated by iron-dependent lipid peroxidation within cell membranes—using preeclampsia as a model system. We propose that the clinical expressions of many reproductive disorders and pregnancy complications may be due to an underlying ferroptopathy (elemental iron-associated disease), characterized by a dysregulation in iron homeostasis leading to excessive ferroptosis.

2011 ◽  
Vol 47 (3) ◽  
pp. 151-160 ◽  
Author(s):  
Jennifer L. McCown ◽  
Andrew J. Specht

Iron is an essential element for nearly all living organisms and disruption of iron homeostasis can lead to a number of clinical manifestations. Iron is used in the formation of both hemoglobin and myoglobin, as well as numerous enzyme systems of the body. Disorders of iron in the body include iron deficiency anemia, anemia of inflammatory disease, and iron overload. This article reviews normal iron metabolism, disease syndromes of iron imbalance, diagnostic testing, and treatment of either iron deficiency or excess. Recent advances in diagnosing iron deficiency using reticulocyte indices are reviewed.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1281-1281
Author(s):  
Corentin Orvain ◽  
Lydie Da Costa ◽  
Richard van Wijk ◽  
Serge Pissard ◽  
Veronique Picard ◽  
...  

Abstract Background: Hereditary stomatocytosis is an inherited disorder of the erythrocyte membrane responsible of chronic hemolytic anemia. Recent advances in the understanding of this group of diseases came from the identification of the molecular basis of this disorder. Mutations in the SLC4A1, FAM38A, RHAG, and SLC2A1 genes have been shown to cause different subtypes of hereditary stomatocytosis. Dehydrated hereditary stomatocytosis (DHSt) is due to mutations in the FAM38A gene coding for the mechanotransduction protein PIEZO1 and to the newly discovered mutations in the KCNN4 gene encoding the Gardos channel. It is important to recognize this entity and differentiate it from hereditary spherocytosis as patients with HSt develop severe and sometimes lethal thromboembolic complications following splenectomy. Also, some patients develop progressive and severe iron overload (IO) despite well compensated hemolysis and no or little transfusion requirement. It is unclear why patients have such different clinical features regarding hemolytic anemia and IO. We describe herein the impact of inherited and acquired modifiers of iron status on the phenotypic expression of DHSt. Patients & Methods: We describe four patients (3 related and 1 unrelated) with proven DHSt due to FAM38A mutations, who displayed varying degrees of iron load. Results: The four reported patients were referred to our specialized outpatient consultation (center of expertise on rare iron overload) for investigation. Their clinical, laboratory and radiological features are summarized in the Table. It is noteworthy that both index cases were initially referred for investigation of hyperferritinemia. Iron levels closely correlated with the degree of hemolysis and with the severity of the clinical complications. One female patient with severe iron overload suffered from chronic anemia, acute hemolytic episodes, and symptomatic gallstones requiring cholecystectomy while one male patient with severe iron overload suffered from a thrombotic event. The two other female patients with no or moderate iron overload had no or mild hemolysis. Genetic modifiers increasing iron stores, such as the presence of the HFE C282Y mutation, and possibly the gender (male), were accompanied with higher liver iron concentration, increased hemolysis and clinical manifestations. On the opposite, females with normal or low iron stores (iron deficiency anaemia (ID) due to gynecologic bleedings) displayed no or mild hemolytic manifestations. It is noteworthy that in the female with ID no clinical or biological manifestations of hemolysis and of stomatocytosis were found initially (normal specialized phenotypic tests). The diagnosis was made by genetic analyses. Restoration of the iron stores resulted in the appearance of biological signs of hemolysis. Conclusion: Iron overload or iron deficiency dramatically alter the clinical presentation of DHST due to PIEZO1 defects. The search for genetic or acquired causes of iron overload (or deficiency) is an important step in the evaluation of the clinical prognosis and the modulation of iron store may help in the management of the patients. Table Clinical, biological, and radiological characteristics of the 4 patients N: normal value; NA: not available; wt: wild-type; ID: iron deficiency Table. Clinical, biological, and radiological characteristics of the 4 patients. / N: normal value; NA: not available; wt: wild-type; ID: iron deficiency Disclosures Cartron: Roche: Consultancy, Honoraria; Celgene: Honoraria; Gilead: Honoraria; Jansen: Honoraria.


2020 ◽  
Author(s):  
Hanae Pouillevet ◽  
Nicolas Soetart ◽  
Delphine Boucher ◽  
Rudy Wedlarski ◽  
Laetitia Jaillardon

AbstractIron Overload Disorder (IOD) is a syndrome developed by captive browsing rhinoceroses like black rhinoceroses (Diceros bicornis) in which hemosiderosis settles in vital organs while free iron accumulates in the body, potentially predisposing to various secondary diseases. Captive grazing species like white rhinoceroses (Ceratotherium simum) do not seem to be affected. The pro-oxidant and pro-inflammatory properties of iron, associated with the poor antioxidant capacities of black rhinoceroses, could enhance high levels of inflammation and oxidative stress leading to rapid ageing and promoting diseases. In this prospective study, 15 black (BR) and 29 white rhinoceroses (WR) originating from 22 European zoos were blood-sampled and compared for their iron status (serum iron), liver/muscle biochemical parameters (AST, GGT, cholesterol), inflammatory status (total proteins, protein electrophoresis) and oxidative stress markers (SOD, GPX, dROMs). Results showed higher serum iron and liver enzyme levels in black rhinoceroses (P<0.01), as well as higher GPX (P<0.05) and dROM (P<0.01) levels. The albumin/globulin ratio was lower in black rhinoceroses (P<0.05) due to higher α2-globulin levels (P<0.001). The present study suggests a higher inflammatory and oxidative profile in captive BR than in WR, possibly in relation to iron status. This could be either a consequence or a cause of iron accumulation, potentially explaining rapid ageing and various diseases. Further investigations are needed to assess the prognostic value of the inflammatory and oxidative markers in captive black rhinoceroses, particularly for evaluating the impact of reduced-iron and antioxidant-supplemented diets.


Blood ◽  
2003 ◽  
Vol 102 (1) ◽  
pp. 371-376 ◽  
Author(s):  
Sven G. Gehrke ◽  
Hasan Kulaksiz ◽  
Thomas Herrmann ◽  
Hans-Dieter Riedel ◽  
Karin Bents ◽  
...  

Abstract Experimental data suggest the antimicrobial peptide hepcidin as a central regulator in iron homeostasis. In this study, we characterized the expression of human hepcidin in experimental and clinical iron overload conditions, including hereditary hemochromatosis. Using quantitative reverse transcriptase-polymerase chain reaction (RT-PCR), we determined expression of hepcidin and the most relevant iron-related genes in liver biopsies from patients with hemochromatosis and iron-stain-negative control subjects. Regulation of hepcidin mRNA expression in response to transferrin-bound iron, non-transferrin-bound iron, and deferoxamine was analyzed in HepG2 cells. Hepcidin expression correlated significantly with serum ferritin levels in controls, whereas no significant up-regulation was observed in patients with hemochromatosis despite iron-overload conditions and high serum ferritin levels. However, patients with hemochromatosis showed an inverse correlation between hepcidin transcript levels and the serum transferrin saturation. Moreover, we found a significant correlation between hepatic transcript levels of hepcidin and transferrin receptor-2 irrespective of the iron status. In vitro data indicated that hepcidin expression is down-regulated in response to non-transferrin-bound iron. In conclusion, the presented data suggest a close relationship between the transferrin saturation and hepatic hepcidin expression in hereditary hemochromatosis. Although the causality is not yet clear, this interaction might result from a down-regulation of hepcidin expression in response to significant levels of non-transferrin-bound iron. (Blood. 2003;102:371-376)


BMJ Open ◽  
2021 ◽  
Vol 11 (5) ◽  
pp. e045609
Author(s):  
Doreen Montag ◽  
Carlos A Delgado ◽  
Consuelo Quispe ◽  
David Wareham ◽  
Valentina Gallo ◽  
...  

BackgroundPreventing infantile anaemia and ensuring optimal growth and development during early childhood, particularly in resource-constrained settings, represent an ongoing public health challenge. Current responses are aligned to treatment-based solutions, instead of determining the roles of its inter-related causes. This project aims to assess and understand the complex interplay of eco-bio-social-political factors that determine infantile anaemia to inform policy, research design and prevention practices.MethodsThis is a longitudinal birth cohort study including four components: (1) biological, will assess known blood markers of iron homeostasis and anaemia and stool microbiota to identify and genetically analyse the participants’ flora; (2) ecological, will assess and map pollutants in air, water and soil and evaluate features of nutrition and perceived food security; (3) social, which will use different qualitative research methodologies to explore key stakeholders and informants’ perceptions related to nutritional, environmental and anaemia topics, participant observations and a participatory approach and (4) a political analysis, to identify and assess the impact of policies, guidelines and programmes at all levels for infantile anaemia in the three regions. Finally, we will also explore the role of social determinants and demographic variables longitudinally for all study participants. This project aims to contribute to the evidence of the inter-related causal factors of infantile anaemia, addressing the complexity of influencing factors from diverse methodological angles. We will assess infantile anaemia in three regions of Peru, including newborns and their mothers as participants, from childbirth until their first year of age.Ethics and disseminationEthical approval was obtained from the Institutional Research Ethics Committee of the Instituto Nacional de Salud del Niño (Lima, Peru), CIEI-043-2019. An additional opinion has been granted by the Ethical Committee of Queen Mary University of London (London, UK). Dissemination across stakeholders is taking part as a continues part of the research process.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. SCI-27-SCI-27
Author(s):  
Tracey Rouault

Ferroportin (FPN), the only known vertebrate iron exporter, transports iron from intestinal, splenic, and hepatic cells into the blood to provide iron to other tissues and cells in vivo. Most of the circulating iron is consumed by erythroid cells to synthesize hemoglobin. Recently, we found that erythroid cells not only consume large amounts of iron, but also return significant amounts of iron to the blood. Erythroblast-specific Fpn knockout (Fpn KO) mice developed lower serum iron levels in conjunction with tissue iron overload and increased FPN expression in spleen and liver without changing hepcidin levels. Our results also showed that Fpn KO mice, which suffer from mild hemolytic anemia, were sensitive to phenylhydrazine-induced oxidative stress but were able to tolerate iron deficiency upon exposure to a low-iron diet and phlebotomy, supporting that the anemia of Fpn KO mice resulted from erythrocytic iron overload and resulting oxidative injury rather than a red blood cell (RBC) production defect. Moreover, we found that the mean corpuscular volume (MCV) values of gain-of-function FPN mutation patients were positively associated with serum transferrin saturations, whereas MCVs of loss-of-function FPN mutation patients were not, supporting that erythroblasts donate iron to blood through FPN in response to serum iron levels. Our results indicate that FPN of erythroid cells has an unexpectedly essential role in maintaining systemic iron homeostasis and protecting RBCs from oxidative stress, providing insight into the pathophysiology of FPN diseases. When malaria parasites invade red blood cells (RBCs), they consume copious amounts of hemoglobin, and severely disrupt iron regulation in humans. Anemia often accompanies malaria disease; however, iron supplementation therapy inexplicably exacerbates malarial infections. We recently found that the iron exporter ferroportin (FPN) was highly abundant in RBCs, and iron supplementation suppressed its activity. Conditional deletion of the Fpn gene in erythroid cells resulted in accumulation of excess intracellular iron, cellular damage, hemolysis, and increased fatality in malaria-infected mice. In humans, a prevalent FPN mutation,Q248H (glutamine to histidine at position 248), prevented hepcidin-induced degradation of FPN and protected against severe malaria disease. FPNQ248H appears to have been positively selected in African populations in response to the impact of malaria disease. Thus, FPN protects RBCs against oxidative stress and malaria infection. Zhang DL, Wu J, Shah BN et al. Erythrocytic ferroportin reduces intracellular iron accumulation, hemolysis, and malaria risk. Science. 2018;359 (6383):1520-1523. Zhang DL, Ghosh MC, Ollivierre H, Li Y, Rouault TA. Ferroportin deficiency in erythroid cells causes serum iron deficiency and promotes hemolysis due to oxidative stress. Blood. 2018;132 (19):2078-2087. Zhang DL, Rouault TA. How does hepcidin hinder ferroportin activity. Blood. 2018;131 (8):840-842. Disclosures No relevant conflicts of interest to declare.


Author(s):  
Xiaomin Wang ◽  
Ye Li ◽  
Li Han ◽  
Jie Li ◽  
Cun Liu ◽  
...  

Iron overload, a high risk factor for many diseases, is seen in almost all human chronic and common diseases. Iron chelating agents are often used for treatment but, at present, most of these have a narrow scope of application, obvious side effects, and other disadvantages. Recent studies have shown that flavonoids can affect iron status, reduce iron deposition, and inhibit the lipid peroxidation process caused by iron overload. Therefore, flavonoids with iron chelating and antioxidant activities may become potential complementary therapies. In this study, we not only reviewed the research progress of iron overload and the regulation mechanism of flavonoids, but also studied the structural basis and potential mechanism of their function. In addition, the advantages and disadvantages of flavonoids as plant iron chelating agents are discussed to provide a foundation for the prevention and treatment of iron homeostasis disorders using flavonoids.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3811-3811
Author(s):  
Paola Delbini ◽  
Lorena Duca ◽  
Isabella Nava ◽  
Anna Meo ◽  
Marina La Rosa ◽  
...  

Abstract Hepcidin is a peptide hormone produced in the liver that acts as negative regulator of iron absorption from the enterocytes and of iron release from macrophages. Iron overload and inflammation up-regulate hepcidin synthesis, while anaemia and hypoxia suppress hepcidin expression. Thalassaemia Major (TM) is a hereditary haemolytic anaemia requiring long-life blood transfusions treatment with consequent iron overload. In β-thalassemias is a disorder in which hepcidin is regulated by opposing influences of ineffective erythropoiesis and concomitant iron overload. In order to get further insights on iron regulation in thalassemias, we screened hepcidin and HFE genes in fourty-three TM regularly transfused patients and sixty control subjects. Blood from TM was taken at least 48 hours after chelation therapy and just before blood transfusion. DNA was prepared from peripheral blood, according to standard protocols. Hepcidin and HFE sequences were amplified with PCR using specific primers and PCR products were sequenced, after purification, in a automatic sequencer. Moreover in all patients serum pro-hepcidin was evaluated by ELISA competitive binding assay (DRG,Germany); iron status was evaluated by serum ferritin (SF), percentage of transferrin saturation (TS) by standard procedures and non transferrin bound iron (NTBI) in serum by HPLC after nitrilotriacetic acid (NTA) chelation. Serum IL-6 as inflammation marker was measured by lateral flow immunoassay (Milenia QuickLine, Germany). Molecular analysis detected an undescribed G→T mutation at position +23 of the 5′-untranslated region in two unrelated TM patients; no mutations were found in control subjects. The probands have been regularly transfused since the age of 1 year, receiving 2–3 units of packed red cells and treated with Deferoxamine 40 mg/Kg/day 6 days/week. The first patient, wild type for HFE mutation, was a 29-years-old compound heterozygous IVS II-745/IVS I-110 man. The SF was 1052 ng/ml, TS 94% and NTBI 1.77 μM. Serum pro-hepcidin value was in normal range (213 ng/ml). The proband’s father was heterozygous for the same hepcidin mutation and showed signs of mild iron overload (SF 491 ng/ml and NTBI 0.50 μM). The second patient was a 26-years-old homozygous β039 man with high levels of SF (4346 ng/ml), TS (169%) and NTBI (2.10 μM), while serum pro-hepcidin was 269 ng/ml. HFE analysis revealed a homozygous genotype for H63D mutation. The patient’s mother was heterozygous for hepcidin and H63D mutation and showed mild iron overload (SF 500 ng/ml; NTBI 0.22 μM) whereas the father, heterozygous only for H63D, had normal iron status. According to recent findings (Bridle et al, 2003) our results indicate that hepcidin mutation in association with H63D synergizes the effect on iron homeostasis and it could be responsible for the development of marked iron overload poorly responsive to chelation therapy in β-thalassemia patients.


Nutrients ◽  
2019 ◽  
Vol 11 (5) ◽  
pp. 1090 ◽  
Author(s):  
Jorge Moreno-Fernandez ◽  
Julio J. Ochoa ◽  
Gladys O. Latunde-Dada ◽  
Javier Diaz-Castro

Iron is an essential micronutrient that is involved in many functions in humans, as it plays a critical role in the growth and development of the central nervous system, among others. Premature and low birth weight infants have higher iron requirements due to increased postnatal growth compared to that of term infants and are, therefore, susceptible to a higher risk of developing iron deficiency or iron deficiency anemia. Notwithstanding, excess iron could affect organ development during the postnatal period, particularly in premature infants that have an immature and undeveloped antioxidant system. It is important, therefore, to perform a review and analyze the effects of iron status on the growth of premature infants. This is a transversal descriptive study of retrieved reports in the scientific literature by a systematic technique. PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines were adapted for the review strategy. The inclusion criteria for the studies were made using the PICO (population, intervention, comparison, outcome) model. Consequently, the systematic reviews that included studies published between 2008–2018 were evaluated based on the impact of iron status on parameters of growth and development in preterm infants.


2017 ◽  
Vol 398 (9) ◽  
pp. 995-1007 ◽  
Author(s):  
Stephanie Rockfield ◽  
Joseph Raffel ◽  
Radhe Mehta ◽  
Nabila Rehman ◽  
Meera Nanjundan

AbstractIron is an essential element required for many processes within the cell. Dysregulation in iron homeostasis due to iron overload is detrimental. This nutrient is postulated to contribute to the initiation of cancer; however, the mechanisms by which this occurs remain unclear. Defining how iron promotes the development of ovarian cancers from precursor lesions is essential for developing novel therapeutic strategies. In this review, we discuss (1) how iron overload conditions may initiate ovarian cancer development, (2) dysregulated iron metabolism in cancers, (3) the interplay between bacteria, iron, and cancer, and (4) chemotherapeutic strategies targeting iron metabolism in cancer patients.


Sign in / Sign up

Export Citation Format

Share Document