Next Article in Journal
Transcriptional Response to Standard AML Drugs Identifies Synergistic Combinations
Next Article in Special Issue
Supplementation with Vitamin D3 Protects against Mitochondrial Dysfunction and Loss of BDNF-Mediated Akt Activity in the Hippocampus during Long-Term Dexamethasone Treatment in Rats
Previous Article in Journal
The Light- and Jasmonic Acid-Induced AaMYB108-like Positive Regulates the Initiation of Glandular Secretory Trichome in Artemisia annua L.
Previous Article in Special Issue
Extended Exposure Topotecan Significantly Improves Long-Term Drug Sensitivity by Decreasing Malignant Cell Heterogeneity and by Preventing Epithelial–Mesenchymal Transition
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Iron Load Toxicity in Medicine: From Molecular and Cellular Aspects to Clinical Implications

by
George J. Kontoghiorghes
Postgraduate Research Institute of Science, Technology, Environment and Medicine, 3, Ammochostou Street, Limassol 3021, Cyprus
Int. J. Mol. Sci. 2023, 24(16), 12928; https://doi.org/10.3390/ijms241612928
Submission received: 10 July 2023 / Revised: 12 August 2023 / Accepted: 15 August 2023 / Published: 18 August 2023
(This article belongs to the Collection Feature Papers in Molecular Pharmacology)

Abstract

:
Iron is essential for all organisms and cells. Diseases of iron imbalance affect billions of patients, including those with iron overload and other forms of iron toxicity. Excess iron load is an adverse prognostic factor for all diseases and can cause serious organ damage and fatalities following chronic red blood cell transfusions in patients of many conditions, including hemoglobinopathies, myelodyspasia, and hematopoietic stem cell transplantation. Similar toxicity of excess body iron load but at a slower rate of disease progression is found in idiopathic haemochromatosis patients. Excess iron deposition in different regions of the brain with suspected toxicity has been identified by MRI T2* and similar methods in many neurodegenerative diseases, including Alzheimer’s disease and Parkinson’s disease. Based on its role as the major biological catalyst of free radical reactions and the Fenton reaction, iron has also been implicated in all diseases associated with free radical pathology and tissue damage. Furthermore, the recent discovery of ferroptosis, which is a cell death program based on free radical generation by iron and cell membrane lipid oxidation, sparked thousands of investigations and the association of iron with cardiac, kidney, liver, and many other diseases, including cancer and infections. The toxicity implications of iron in a labile, non-protein bound form and its complexes with dietary molecules such as vitamin C and drugs such as doxorubicin and other xenobiotic molecules in relation to carcinogenesis and other forms of toxicity are also discussed. In each case and form of iron toxicity, the mechanistic insights, diagnostic criteria, and molecular interactions are essential for the design of new and effective therapeutic interventions and of future targeted therapeutic strategies. In particular, this approach has been successful for the treatment of most iron loading conditions and especially for the transition of thalassemia from a fatal to a chronic disease due to new therapeutic protocols resulting in the complete elimination of iron overload and of iron toxicity.

1. Introduction

Iron is an essential metal found in all the cells of the body, playing an important role in many physiological and cellular processes. There are also many diseases associated with iron metabolism affecting many people. In particular, diseases of iron imbalance affect billions of patients, mostly with iron deficiency anemia. There are also many diseases associated with iron overload and other toxicity. In this context, the presence of excess iron is an adverse prognostic factor for all diseases. Especially in heavily iron loaded patients, iron can cause serious organ damage and fatalities.
Millions of patients with hemoglobinopathies and other refractory anemias worldwide, including thalassemia, sickle cell disease (SCD), myelodyspasia, and hematopoietic stem cell transplantation (HCT), receive regular red blood cell (RBC) transfusions for the treatment of their anemia [1,2,3,4,5].
A major side effect of chronic RBC transfusions is the build-up of excess, toxic deposits of iron in various organs, which results in tissue damage and organ malfunction [6,7,8]. Congestive cardiac failure is the main cause of mortality in heavily iron-loaded beta thalassemia major (TM) patients who have no access to chelation therapy for the removal of excess iron [9].
A gradual increase in body iron levels from chronic increases in dietary iron absorption and the cause of iron overload toxicity in the liver and other organs was also observed in idiopathic haemochromatosis, which is a genetic disease affecting one in ten people of the Caucasian population [10].
The assessment of excess iron levels in different categories of iron-loaded patients is determined mainly from the estimation of iron diagnostic parameters such as serum ferritin, iron concentration from liver biopsies, and magnetic resonance imaging (MRI T2*) measurements. Iron toxicity may affect the function of many other organs in addition to those observed in iron-loaded patients. For example, iron accumulation in parts of the brain was detected using MRI T2* in many neurodegenerative and other diseases [11].
The presence of excess iron deposition or labile forms of iron in any part of the body is a potential hazard and source of continuous toxicity [6,9]. In most cases, iron overload toxicity in moderate iron-loaded organs is reversible and the restoration of normal function can be accomplished by the removal or reduction of excess iron deposition. The sooner the removal of excess or labile forms of iron can be accomplished, the lower the level of permanent damage in the organs affected [12].
The rate of body iron intake from chronic RBC transfusions is in most cases the major assessment parameter for the selection of effective chelating drug protocols for achieving negative iron balance in chronically transfused patients. In this context, the efficacy in the removal of excess iron by chelating drugs requires their daily use for such categories of iron-loaded patients. Iron chelation therapy worldwide is carried out using deferoxamine (DF), deferiprone (L1) and deferasirox (DFRA) [7,13]. The combination of chelating drugs, and especially the combination of L1 and DF, is also widely used in TM patients in many countries [14,15,16].
The main aim of iron chelation therapy in TM and other chronically transfused patients is the design of effective chelating drug protocols for the achievement of negative iron balance, which can lead to the reduction of the excess iron load. The ultimate aim of chelation therapy is the achievement and maintenance of normal iron stores, in which case patients are devoid of all iron overload toxic side effects and consequently the overall reduction in morbidity and mortality in transfusional iron-loaded patients [17]. This aim, including the long-term prevention of iron overload, can be accomplished in most cases using effective and safe chelation protocols [17]. Furthermore, regular monitoring of the iron load and selected chelating drug dose protocols will also be needed to ensure that no prolonged iron deficiency or chelating drug toxicity can be caused in TM and other categories of chronically transfused patients that have reached the stage of normal iron store levels.
The introduction of effective therapies and prophylactic measures is also necessary in all other diseases of iron overload and or toxicity. In each case the causes, affected sites, and levels of iron toxicity have to be identified, analyzed, and monitored for introducing effective protocols, which can achieve complete therapy of the disease or optimal therapeutic outcomes. Such approaches will require the understanding of the mechanisms of toxicity from the molecular level to the whole organism. In this context, the underlying mechanisms of iron toxicity in primarily transfusional iron overload will mainly be discussed, since this is considered the most affected disease of metal intoxication with the highest morbidity and mortality rate worldwide.
Other forms of iron toxicity will also be discussed for many other diseases, including ferroptotic cell death, which is identified as one of the most common but also rare human diseases, the presence of focal iron deposits in non-iron-loaded conditions, especially in neurodegenerative diseases, and also of iron diversion in the reticuloendothelial system in the anemia of chronic disease categories. Further discussion includes different forms of labile iron toxicity in other clinical conditions associated with tissue damage and free radical pathology, toxic forms of carcinogenic iron complexes in food and environmental pollution, and also other forms of toxicity arising from molecular interactions of iron with dietary, biochemical, drug, and other xenobiotic molecules.

2. The Source of Iron Overload from Chronic Red Blood Cell Transfusions

The main source of iron overload in regularly RBC transfused patients with refractory anemia is the blood obtained from blood donors. The quantitative aspects related to body iron intake from RBC transfusions and the metabolic pathways of iron accumulation in different organs provide important information for assessing therapeutic strategies. In particular, the information on the properties and the role of RBC, as well as its components, is crucial for understanding the extent of the complications of iron overload.
In considering the qualitative and quantitative aspects of iron distribution in man, it is estimated that about 58% of the iron in the body is found in the form of heme in hemoglobin, which is the major constituent of RBC and the component that carries oxygen and gives the red color to blood (Figure 1). The role and function of RBC and hemoglobin are crucial in respiration and other functions. In particular, normal cellular, tissue, and bodily functions can only be ensured if a sufficient and continuous supply of oxygen is available to all parts of the body through blood circulation and also if sufficient normal RBC and hemoglobin levels are available to carry over the necessary amounts of oxygen.
The quantitative aspects in relation to oxygen transport by RBC and hemoglobin and the role of iron in heme, where oxygen is bound, is of primary importance for normal bodily functions and overall human survival. Hemoglobin is estimated to occupy 95% of the RBC volume and amounts to about 670 g of the 25 kg dry body weight (2.7%) of an average human adult individual [18,19,20]. Furthermore, it is estimated that approximately 25 trillion RBC circulate in the bloodstream, each one packed with about 260 million hemoglobin molecules. Considering that one adult hemoglobin molecule is composed of two alpha and two beta globin protein subunits, to each of which one molecule of heme is embedded, the total amount of iron in the ferrous state as heme in one RBC is estimated to be 1.04 billion iron molecules (Figure 1). In this context, each unit of RBC transfused contains about 200–250 mg of iron, which is added to the body iron burden.
There are many other unique characteristics of RBC in relation to heme iron, such as the high concentration of both iron and oxygen, which is estimated to reach about 16 mM when the RBC are fully oxygenated. It should be noted that no other cell contains such a high concentration of protein, in this case hemoglobin with iron in the ferrous state in heme and also oxygen. This high concentration of ferrous iron and oxygen can potentially be a highly reactive mixture. This can lead to reactive oxygen species and other free radicals resulting in oxidative stress toxicity, especially in hemolytic and other damaging effects involving RBC [19,20,21,22]. In most cases, strict metabolic controls and the presence of antioxidant enzymes such as superoxide dismutase and molecules such as glutathione ensures the antioxidant protection inside the RBC [23].
There are different stages in the production and development of RBC and their release in the blood circulation, where on average they have a life span of 120 days. Erythroblasts are the early-stage RBC progenitor cells containing a nucleus and produced in the bone marrow, which progressively lose their nucleus and organelles, before their release in the bloodstream initially as reticulocytes and later as matured RBC. Heme production takes place in mitochondria and the protein part of hemoglobin is produced in the cytoplasm of both erythroblasts and reticulocytes. In contrast, mature RBC cannot synthesize new proteins during their 120 day lifespan in the human bloodstream [19,20,24].
At the end of their lifespan aging RBC undergo progressive structural and denaturing changes such as vesiculation, a process leading to the formation of vesicles [24,25,26]. Furthermore, senescent RBC become more rigid and fragile than younger RBC, with the former being identified and readily removed from the bloodstream via phagocytosis by macrophages of the reticuloendothelial system primarily in the spleen and the liver. The degradation process begins inside the macrophages and usually involves only old and damaged RBC, where iron is released from heme and stored initially in the reticuloendothelial cells and then released in the bloodstream, taken by plasma iron transporter protein transferrin and distributed in many organs for storage and re-utilization [20,24,25,26].
In addition to the main source of iron overload, which is iron from senescent RBC, many other sources contributing to iron body burden and associated iron toxicity can also take place in hemolytic and other events, which are observed in many of the categories of patients with refractory anemia receiving RBC transfusions. In this context and under normal physiological conditions, several innate mechanisms are in operation for the control of iron metabolic pathways and protection against iron toxicity. For example, the removal of denatured hemoglobin or other aggregated species containing iron inside the RBC is accomplished by a vesiculation process [23,24,25,26]. Similarly, protection against oxidative and other damage caused by the release of heme and hemoglobin into the blood stream during hemolysis is undertaken by hemopexin and haptoglobin, respectively [27,28]. Hemopexin is a plasma protein expressed mainly in the liver and has a high affinity to heme binding. Similarly, haptoglobin in plasma binds free hemoglobin released from RBC forming a haptoglobin–hemoglobin complex, which is removed by the reticuloendothelial system in the spleen, where, after a degradation process, iron and other components are released to follow their corresponding metabolic pathways [27,28].
Several toxicity and various other factors leading to changes in the concentration levels of iron, oxygen, hemoglobin, and RBC can also cause many other abnormalities and side effects, including anemia and insufficient transport of oxygen to the tissues. In such cases, the general function of all the cells, tissues, and organs of the body is undermined, leading to reversible or irreversible damage and the need for therapeutic intervention.

3. Major Diseases of Transfusional Iron Overload

Millions of hematologic and other categories of patients worldwide suffer from different types of anemia, in addition to iron deficiency anemia. Among these, there are many conditions with abnormalities in the structure and function of RBC and hemoglobin, where RBC transfusions may be required to maintain normal bodily functions (Table 1). For example, hundreds of hemoglobin mutations have been reported in humans, where in many cases the changes in the production rate and structural characteristics of normal hemoglobin can lead to abnormal function, toxic side effects, and anemia [29].
The hemoglobinopathies are the most common group of genetic disorders affecting millions of people worldwide, mostly in the developing countries [29,30,31]. In this context, there are many abnormalities in the function of hemoglobin which are related to changes in the globin structure and heme function (Figure 1). For example, patients with thalassemia have a low or absent production of the alpha or beta or other globin chains of hemoglobin (Table 1). Most TM patients cannot produce the beta globin chain of hemoglobin and are therefore severely anemic, requiring RBC transfusions from normal blood donors in order to survive [1,29,30]. Another inherited hemoglobinopathy affecting millions of people is SCD, which is prevalent in Africa and many other countries with descendants from Africa. In this abnormal hemoglobin condition, a single amino acid change in the beta globin chain can cause hemoglobin polymerization, sickling of RBC, anemia, and painful side effects as a result of the sickling crisis [3,29,30]. Treatment with RBC transfusions improves anemia and alleviates some categories of the SCD patients from the painful side effects of the sickling crisis.
In the absence of effective excretory iron mechanism in humans, the build-up of body iron burden progressively increases with each RBC transfusion. Considering that each unit of transfused RBC contains about 200–250 mg of iron and that a total 4.5–5.0 g of iron is present in the body of a 70–75 kg normal adult man, it is estimated that for every 25 RBC units transfused, the total body iron burden is doubled [20]. Under these conditions, different amounts of excess iron accumulate progressively in the body, which depends on the rate of RBC transfusions, considering that there are different requirements in each patient category and for each individual patient (Table 1).
Contribution to body iron burden from increased absorption of dietary iron is observed in TM and other categories of regularly RBC transfused patients, especially at periods of low concentration of hemoglobin and increased anemia prior to transfusion. This process involves a body intake of much smaller amounts of less than 6 mg of iron per day, in comparison to that obtained from transfusions [32,33,34,35]. In some cases, such as thalassemia intermedia (TI) or non-transfusion dependent thalassemia and similar conditions, the frequency of transfusions, if any, is significantly reduced (Table 1) [34,36,37,38]. In this context, there is a large difference between the genotypes in thalassemia and RBC transfusion requirements to the extent that increased iron absorption in most cases of TI is the dominant route of body iron intake and loading in comparison to that of iron intake from transfusions [34,36,37,38].
Many other differences in hematologic disease characteristics and therapeutic requirements, including the initiation and rate of RBC transfusions, exist between the various categories of transfusional iron-loaded patients (Table 1) [1,2,3,4]. For example, the initiation of RBC transfusions ranges from the first few years to the rest of remaining life in TM, whereas in SCD and TI they may be intermittent at different patient ages [3,36,37]. In contrast, in the myelodysplastic syndromes the patients affected are mostly of middle to old age and transfusions are usually initiated later in life [4]. Similarly, the rate of RBC transfusions may also vary, with transfusion frequency every 1–4 weeks in TM and a much lower rate for many TI patient categories. Further variations on RBC transfusions and rate of iron loading may be related to the underlying disease, individual and other factors such as hypersplenism, hemolysis, and RBC antibodies [39].
Limitations regarding RBC transfusion management may also be caused by factors related to changes in the underlying disease and as a result of variable effects due to the poly-pharmacotherapy in each case. For example, HCT is a widely used form of treatment for many hematologic malignancies and genetic disorders. The number of diseases and the number/age range of patients undergoing HCT is rapidly increasing as a result of technological and therapeutic management improvements (Table 1) [2,40,41,42,43].
The level of iron overload caused from RBC transfusions is considered an independent adverse prognostic factor in all diseases associated with HCT and is one of the many complications in pre- and post-transplantation side effects, affecting the long-term survival of about 0.5 million HCT patients [2,40,41,42,43]. Iron overload mostly develops in the pre-transplantation period in many HCT patient categories, including about 4000 patients with TM and 1000 with SCD, as well as many more with myelodysplasia, leukemia, and other patients [2,40,41,42,43]. The excess iron burden in the transfusion-independent or reduced transfusion dependent post-allogeneic iron-loaded HCT patients always remains a potential source of toxicity and clinical complications unless it is removed.
In addition to the different rate of RBC transfusions between the various hematologic and other categories of iron-loaded patients, there are also many patient categories needing transfusions because of different causes of anemia, such as sideroblastic anemia, where iron is deposited in the mitochondria of sideroblasts [44,45,46]. In all RBC, transfusional iron-loaded categories and non-transfusional iron-loaded categories such as idiopathic hemochromatosis patients where venesection is contraindicated, iron chelation therapy needs to be introduced in order to reduce or eliminate the iron toxicity (Table 1) [34,47,48].
The major category of hematologic diseases in need for lifelong RBC transfusions with the highest number of patients and daily iron chelation therapy is TM, which has the highest number of metal-related mortality and morbidity rate worldwide as a result of iron overload toxicity [1,13,29,30,31]. Thalassemia major is found mainly in developing countries and in the Mediterranean, Middle East, and South-East Asia, where more than 90% of TM patients are born [1,29,30]. It is estimated that worldwide there are 100 million thalassemia heterozygote asymptomatic carriers, and more than 100,000 TM babies are born each year [29,30]. In India, for example, the annual birth rate of TM patients is estimated at 9000 [49].

4. Iron Metabolism and Iron Toxicity in Chronically Transfused Patients

Iron is essential for many biological processes, including respiration, hemopoiesis, growth, and development [20,50,51,52,53,54]. The build-up of excess iron in several organs of patients receiving regular RBC transfusions is due to the lack of efficient excretion mechanism of accumulated iron. This is in contrast to the presence of efficient physiologic mechanisms of dietary iron body intake and conservation [20,35,50,51,52,53,54].
Under normal physiologic conditions, the transport, storage, and utilization of iron is controlled by metabolic pathways involving proteins such as transferrin in plasma, transferrin receptors on the cell membrane for intracellular iron uptake through endocytosis, and the presence of intracellular ferritin, which can accommodate up to 4500 iron molecules and is used for iron storage [20,55,56]. Hemosiderin is a breakdown product of ferritin, which is the major intracellular iron storage protein found in iron-loaded tissues of patients with iron overload [6,20,57].
Iron toxicity has been implicated in almost all diseases of tissue damage, including those associated with the presence of redox active iron and iron overload in hematologic and other diseases [20,21,22,54,57,58,59,60,61,62,63,64,65]. The basic molecular mechanism of iron toxicity is related to the role or association of iron as the main biological catalyst in free radical production and related pathology [20,57,58,59,60,61,62,63,64,65].
The general mechanism of free radical production primarily includes the Fenton reaction, which involves Fe(II) and hydrogen peroxide and leads to the catalytic formation of the highly reactive and toxic hydroxyl radicals with a half-life of reactivity within micro- to nano-seconds. Similarly, the production of other free radicals such as superoxide and nitrous oxide, as well as oxygen-activated products such as hydrogen peroxide, can all be involved in oxidative chain reactions and cascades, leading to oxidative damage to virtually all known organic biomolecules, including DNA, proteins, sugars, and lipids [54,58,59,60,66,67]. Several other toxicity mechanisms involving different forms of iron ions such as Fe(IV) and reducing agents such as ascorbate and chelating molecules may also enhance free radical toxicity [54,58,59,60,63,64,66,67].
Within this context, the presence of excess iron, mainly in the form of intracellular hemosiderin and associated low molecular weight (LMWt) labile iron, can progressively cause molecular, cellular, and tissue damage, and a vicious circle leading to oxidative stress toxicity damage in cells (Figure 2) [57,65,66,67].
The iron-induced oxidative stress toxicity damage can cause abnormalities to cell structure and function, including damage to cell membrane mainly through lipid peroxidation and organelle damage, such as the disruption of lysosomes causing the release of proteolytic enzymes and resulting in the malfunction of the cellular processes, all of which can lead to cell death such as ferroptosis, apoptosis, autophagy, and necrosis [54,60,66,67].
Under normal conditions, cell death is an essential process for controlled development and the maintenance of homeostasis, as well as for the prevention of proliferative diseases. The importance of iron in cellular function processes including cell death has been recently highlighted by the discovery of ferroptosis, which is a newly described programmed cell death based on oxytosis, which is caused by free radicals arising from iron catalysis and involving transcription and many other factors [68,69,70]. Ferroptosis has been implicated as an important process not only in health but also in diseases, including cancer, COVID-19, hematologic, neurodegenerative, cardiac, kidney, infectious diseases, and many others [71,72,73,74,75,76,77,78,79,80]. Ferroptosis is morphologically, biochemically, and genetically distinct from apoptotic and necrotic cell death and is mainly characterized by the accumulation of lipid peroxides on cell membranes, decrease in glutathione production, inhibition of glutathione peroxidase (GPX4) activity, an increase in intracellular iron, the sustainable production of increased free radical reactions caused mainly by labile but also other forms of iron, ferritinophagy, and an iron-dependent autophagic cell death program (Figure 2) [68,69,70,81,82,83,84,85].
Further information on iron overload toxicity in TM at the cellular and ultrastructural level has been obtained from electron microscopy studies [6,57]. In such cases, more detailed information was received from biopsy samples obtained from iron-loaded TM patients containing cardiomyocytes and hepatocytes, where the presence of iron-loaded ferritin arrays, mainly in primary lysosomes and hemosiderin iron aggregates in secondary lysosomes, was observed [6,57,85,86,87,88,89]. Ultrastructural observations of cardiomyocytes have also shown the presence of iron-laden lysosomes, which rupture into the cell sap, causing intracellular damage including large cytoplasmic vacuoles, swollen mitochondria with no iron deposits but with loss of their cristae, substantial loss of myofilaments, an increase in the electron density of nuclei, and also increased amounts of heterochromatin (Figure 3b,c) [86].
Iron overload toxicity in iron-loaded patients can cause damaging effects in many organs, including malfunction of the heart, liver, spleen, and pancreas, and also different complications including congestive cardiac failure, liver fibrosis and cirrhosis, diabetes mellitus, and diminished growth and infertility in teenage TM patients [5,6]. Organ damage in iron-overloaded conditions is progressively developed and can reach different stages, which in general depends on the level of iron load. Early stages of damage in patients’ organs can be detectable when approximately 20–50 units of RBC have been transfused [9,29,90,91,92,93,94,95,96]. At these early stages the organ damage can be reversible provided effective iron chelation therapy protocols are applied. However, in the absence of effective chelation therapy and following repeated RBC transfusions, the organ damage caused by increasing iron load can progressively become irreversible, e.g., in cardiac failure and liver fibrosis.
The major cause of mortality in regularly transfused iron-loaded TM patients not receiving chelation therapy is iron overload toxicity-related cardiomyopathy (Figure 3d) [9]. In this context, MRI diagnostic studies of iron load deposition in the heart of TM patients have shown the existence of a correlation between cardiac damage and the level of cardiac iron load. In particular, there is an increased risk of congestive cardiac failure in TM patients with excess cardiac iron deposition levels [97,98,99,100].
Iron overload can also promote microbial infections, which is the second major cause of morbidity and mortality observed in iron-loaded TM patients [101,102,103]. Similar adverse effects from microbial infections can also be seen in regularly RBC transfused immuno-compromised iron-loaded patients, and especially in HCT patients.
Figure 3. Iron overload toxicity from molecular aspects to effects in organs. (a) A cartoon showing oxyhydroxide polynuclear iron complexes resembling the molecular composition of the iron core in the iron storage proteins ferritin and hemosiderin. This is the form of excess iron detected in iron-loaded cells, tissues, and organs of iron-loaded patients. Some water and phosphate molecules are also shown to be associated with the iron core in ferritin. (b) Electron micrograph at a ×30,000 magnification with subcellular details of a part of a myocyte, obtained from a cardiac biopsy of an iron-loaded thalassemia patient, who suffered a congestive cardiac failure. It shows extensive iron deposition in the cytoplasm mostly in primary (black arrows) and secondary (white arrowheads) lysosomes, loss/damaged myofilaments (F) and swollen/damaged mitochondria (M). (c) Another electron micrograph at a ×12,000 magnification with similar details as (b) and an electron density of myocyte nucleus (N) with increased amounts of heterochromatin. (d) Magnetic resonance imaging picture of an iron-loaded thalassemia patient, showing differential iron loading of the heart and liver. The top arrow shows the heavy iron deposition in the interventricular septum of the heart of MRI T2* = 6.32 ms (normal T2* = >19), with low signal intensity (dark). The bottom arrow shows the liver of the patient with no excess iron deposition (MRI T2* = 19.2 ms, normal). (e) A spleen section of an iron-loaded thalassemia patient, where iron deposits were stained with Pearl’s Prussian blue. There are hemosiderin deposits within cytoplasm and nucleus of macrophages (grade 3) in blue color. (f) A liver section of an iron-loaded thalassemia patient, where iron deposits are stained with Pearl’s Prussian blue. There are hemosiderin deposits in hepatocytes and Kupffer cells and within bile ducts (grade 4) in blue color (Adapted with permission from References [88,104]).
Figure 3. Iron overload toxicity from molecular aspects to effects in organs. (a) A cartoon showing oxyhydroxide polynuclear iron complexes resembling the molecular composition of the iron core in the iron storage proteins ferritin and hemosiderin. This is the form of excess iron detected in iron-loaded cells, tissues, and organs of iron-loaded patients. Some water and phosphate molecules are also shown to be associated with the iron core in ferritin. (b) Electron micrograph at a ×30,000 magnification with subcellular details of a part of a myocyte, obtained from a cardiac biopsy of an iron-loaded thalassemia patient, who suffered a congestive cardiac failure. It shows extensive iron deposition in the cytoplasm mostly in primary (black arrows) and secondary (white arrowheads) lysosomes, loss/damaged myofilaments (F) and swollen/damaged mitochondria (M). (c) Another electron micrograph at a ×12,000 magnification with similar details as (b) and an electron density of myocyte nucleus (N) with increased amounts of heterochromatin. (d) Magnetic resonance imaging picture of an iron-loaded thalassemia patient, showing differential iron loading of the heart and liver. The top arrow shows the heavy iron deposition in the interventricular septum of the heart of MRI T2* = 6.32 ms (normal T2* = >19), with low signal intensity (dark). The bottom arrow shows the liver of the patient with no excess iron deposition (MRI T2* = 19.2 ms, normal). (e) A spleen section of an iron-loaded thalassemia patient, where iron deposits were stained with Pearl’s Prussian blue. There are hemosiderin deposits within cytoplasm and nucleus of macrophages (grade 3) in blue color. (f) A liver section of an iron-loaded thalassemia patient, where iron deposits are stained with Pearl’s Prussian blue. There are hemosiderin deposits in hepatocytes and Kupffer cells and within bile ducts (grade 4) in blue color (Adapted with permission from References [88,104]).
Ijms 24 12928 g003

5. Diagnosis and Monitoring of Transfusional Iron Overload

The level of excess iron in the body of iron-loaded patients can be diagnosed and monitored using a number of diagnostic techniques, most of which are also generally used for the monitoring of other diseases of iron metabolic imbalance. In addition to monitoring iron level parameters, regular clinical and biochemical assessment is also widely used in order to evaluate organ function and the extent of damage caused by iron overload, as well as other complications of the underlying disease [1,39,89].
The general routinely-used method for estimating body iron stores is blood sample estimation of serum ferritin, serum iron, and transferrin iron saturation. The physiologic non-toxic levels of iron are usually characterized by the normal range of serum ferritin values (male 40–340 μg/L and female 14–150 μg/L), transferrin saturation (25–35%), and serum iron (10–40 μmol/L). In rare cases, urinary iron excretion in response to chelating drugs such as DF and L1 may also be used [39,89].
Another diagnostic method widely used for the estimation of the iron load in regularly transfused and other patients is the measurement of iron concentration in liver biopsy samples, which usually reflects, to some extent, the level of total body iron stores but not the distribution of excess iron (Figure 3f). Liver iron concentration levels, equivalent to not more than 2 mg iron per g liver dry weight, are considered as the maximum normal physiologic levels in the assessment of liver biopsies, whereas higher levels of iron concentration have been classified to various grades of liver iron load, with the higher grades being associated with liver cirrhosis and fibrosis [1,7,9,104,105]. Furthermore, the use of histochemical techniques of the liver biopsy samples can lead to the identification of the non-heme iron concentration and the level, as well as the pattern of iron deposition in hepatocytes and Kupffer cells [39,89,104,105]. However, a drawback of the iron load estimation by this method is that the distribution and concentration of iron in liver and spleen biopsy samples is not uniform (Figure 3e,f) [104]. Another drawback of this method, which was identified by MRI T2* signal intensity relaxation studies, is that liver iron levels and serum ferritin levels do not reflect the iron load or distribution in other organs, such as the heart [106,107,108,109].
In the last 30 years, the newly developed non-invasive techniques of iron estimation using the MRI T2 and T2* signal intensity relaxation times have become the most useful tools for assessing differential organ iron deposition and, in particular, cardiac iron load levels, which are considered the most critical tool in the prognosis of transfused iron-loaded patients [97,98,99,100,104,106,107,108,109]. The concentration of excess iron in the heart was, until recently, impossible to identify by using biopsies or other methods. Furthermore, the introduction of the MRI T2 and T2* techniques have successfully been used for estimating the level of iron loading not only in the heart but also in the liver, spleen, pancreas, and other organs (Figure 3d) [39,96,100,104,108,109,110].
There are different ranges and classifications of the level of iron overload using MRI in relation to signal intensity relaxation times in different organs. In particular, patients with cardiac MRI T2* relaxation times lower than about 8 ms, are considered to be in the heavy hemosiderosis range and to be in danger of cardiac failure. Similarly, moderate cardiac hemosiderosis is considered for patients with cardiac MRI T2* relaxation times in the range of about 8–12 ms, mild hemosiderosis in the range of about 12–20 ms, and above 20 ms to be in the normal range of cardiac iron levels (Figure 3d) [39,100,104,108,109,110].
Different classifications have been described for liver iron overload where MRI T2* relaxation times have been compared to the iron content of liver biopsy samples. Accordingly, MRI T2* signal intensity relaxation times lower than 1.4 ms are estimated to contain more than 10 mg of iron per g dry weight of the biopsy sample and considered to be in the severe hepatic hemosiderosis range. Moderate hepatic hemosiderosis is considered for MRI T2* signal intensity relaxation times in the range 1.4–2.7 ms, corresponding to 5–10 mg/g dry weight, mild in the range of 2.7–6.3 ms corresponding to 2–5 mg/g dry weight, and normal hepatic iron for higher than 6.3 ms corresponding to less than 2 mg/g dry weight (Figure 3d) [39,104,108,109,110]. Although most of the MRI T2 and T2* signal intensity relaxation time techniques were used for the monitoring of iron load in TM, the same methods could also be applied for all the different categories of iron-loaded patients, including SCD, HCT, and myelodysplasia [110,111,112].
The introduction of the MRI T2 and T2* signal intensity relaxation time techniques in combination with histochemistry of mainly liver biopsy samples and electron microscopy have contributed to the evaluation of the mechanisms of iron deposition and toxicity in various organs of iron-loaded patients (Figure 3d–f) [39,89,108,109,110,111,112]. The methods described above can be used periodically for assessing the prognosis of the patients in relation to iron toxicity, which is directly related to their total body or individual organ iron load and organ function, as well as for the adjustment of the dose protocols during iron chelation therapy (Figure 3) [39,108,109,110,111,112].
In developed countries, the frequency of iron load monitoring and the progress of the chelation therapy in TM and other categories of regularly transfused patients is usually carried out by estimating serum ferritin levels every three months, while liver iron and cardiac iron concentration estimation can be carried out annually by using the MRI T2 and T2* signal intensity relaxation time techniques. In the developing countries where MRI facilities may be absent, serum ferritin levels are mostly used for iron load estimations. It should be noted that in regularly transfused TM and other patients, liver iron appears in most cases to correlate well with serum ferritin levels, but cardiac iron load does not appear to correlate with either liver iron or serum ferritin levels [106,107,108,109].
In most cases, serum ferritin levels greater than 2500 μg/L and transferrin iron saturation of 100% suggest the presence of highly toxic iron levels in the tissues and the need for the use of more intensive chelation therapy protocols and higher chelating drug doses. Despite the overall improvements using the above-established methods of monitoring iron levels, there can be limitations in the prediction of either the total body iron load or the extent of iron toxicity because of many other variables. These may include differences in the total body and organ iron distribution of stored iron, in the iron assessment and detection methods, or in the specificity of iron pool and iron removal effects from various organs using different chelating drugs or chelation therapy protocols [100]. A number of other factors can also influence the iron metabolic processes, or the estimation of the iron load levels as a result of individual variations such as dietary habits, infection, inflammation, rate of iron absorption and excretion, transfusion frequency, erythropoietic activity, bleeding, drug or RBC reactions, etc. [39,113,114,115].
Overall, the factors contributing to iron overload and the diagnostic techniques used for its assessment have played a major role in defining the etiology and the level of iron toxicity in different organs (Figure 4). This information is also critical for monitoring changes in iron load during iron chelation therapy and the design of effective iron chelation therapies for all categories of RBC transfused iron-loaded patients.

6. Excess Iron Deposition and Toxicity in Non-Transfusional Iron Loading Conditions

There are many non-transfusion-dependent iron overloading conditions affecting millions of patients worldwide, including idiopathic hemochromatosis, thalassemia intermedia, and transfusion-independent post-allogeneic HCT patients, which was introduced in previous sections (Table 1) [10,42,43,47,116,117,118,119,120,121,122]. In all these and similar conditions, iron overload toxicity affects the function of many organs. In each case, the level and intensity of the toxicity is proportional to the level of iron overload, as shown in transfusional iron loading conditions.
In most cases of non-transfusion-dependent iron overloading conditions, iron toxicity in iron-loaded organs is reversible and the restoration of normal function can be accomplished by the removal of excess iron. The gold standard form of treatment in almost all categories of non-transfusion dependent iron overloading conditions is phlebotomy, whereas chelation therapy is an alternative option for some cases, especially where phlebotomy cannot be used [34,47,116,117,118,119,120,121,122]. The ultimate aim of treatment is similar to that proposed for transfused patients, namely the complete removal of excess iron and the achievement of normal iron store levels. As shown in transfused patients, non-transfusion-dependent iron-loaded patients achieving this stage are devoid of excess iron toxicity and associated complications.
The same standard diagnostic techniques, including MRI T2* and serum ferritin, are used for monitoring iron overload and the progress of therapeutic intervention, including phlebotomy and chelation therapy in idiopathic hemochromatosis, thalassemia intermedia, post-allogeneic HCT patients, and other categories of patients with non-transfusion-dependent iron overloading conditions (Table 1) [122,123,124,125,126,127].
Excess iron deposition in small parts of organs has also been realized in non-iron-loaded patients. The identification of small iron deposits in different parts of the body, which are not linked to iron overloading diseases nor to diseases with abnormal iron metabolism, began in the 1980s and continues until today [128,129,130,131,132,133,134]. In particular, many such findings were related to neurological and neurodegenerative diseases associated with excess iron and detected using earlier versions of MRI signal intensity relaxation time techniques [128,129,130,131,135,136,137,138,139].
The iron toxicity and tissue damage identified in many of the non-iron-loaded categories of patients concerns the presence of focal iron deposits, which are usually detected by MRI T2* and are of major significance in many clinical conditions. In particular, focal iron accumulation in the brain with increased MRI T2* signal intensity has been detected in many neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, Friedreich’s ataxia, and neurodegeneration with brain iron accumulation (NBIA) (Table 1) [140,141,142,143,144]. In the latter, at least fifteen diseases, including pantothenate kinase 2-associated neurodegeneration (PKAN) with NBIA, have been characterized due to iron deposition in different parts of the brain [144].
The re-trafficking of iron is also found in many categories of chronically ill patients with anemia of chronic disease or anemia of inflammation, where the retention of iron in reticuloendothelial cells causes iron-restricted erythropoiesis, low production of hemoglobin, and also anemia. These diseases include cancer and hematological malignancies, infections, inflammatory diseases, immune-mediated diseases, chronic kidney disease, and congestive heart failure (Table 1) [145,146,147,148,149,150,151,152,153]. The treatment of the anemia of chronic disease is very challenging for almost all categories of patients affected and is the subject of research investigations and therapeutic approaches, including, in many cases, the fine balance between iron deficiency level and iron toxicity of administered iron [154,155,156,157,158,159,160,161].
A similar diversion of iron to that observed in the anemia of chronic disease is associated with iron accumulation in the macrophages of cancer patients. In general, macrophages associated with tumors play an important role in both the immune system and tumor microenvironment, and interact with different pathways, including ferroptosis, which are involved in cancer initiation, proliferation, metastasis, in drug resistance, etc. [81,162,163,164,165,166]. In particular, during cancer proliferation, adjacent endothelial and other cells are utilized by cancer cells for energy supply and nutrients, including iron, most of which is released during ferritinophagy. Iron is subsequently effluxed by cancer cells and taken by macrophages, causing the formation of iron-laden macrophages, a reduction in serum iron, and iron deficiency [74,84,85,167,168,169,170]. A concomitant effect of the presence of iron-laden macrophages is the substantial increase in serum ferritin levels called hyperferritinemia, which is a negative prognostic factor for many diseases in addition to cancer, such as different forms of inflammation, autoimmune disorders, and infection [171,172,173,174]. Overall, the immune response in all these conditions appears to lead to activation of macrophages, iron uptake into macrophages, reduction of serum iron, and to increased synthesis and secretion of ferritin in plasma by macrophages [167,168,169,170,171,172,173,174,175].
Excess iron deposition has been identified in many other organs of different categories of patients which are not related to iron overload. These include ovarian endometriosis, multiple sclerosis, cognitive function, depression, liver lesions, and ageing [176,177,178,179,180,181]. The precise mechanisms and the level of contribution of iron toxicity regarding the causes or impact on each of these conditions have not yet been fully clarified.

7. Iron Toxicity from Labile Forms of Iron and Other Molecular Interactions

There are thousands of research findings and reports in the medical literature of naturally occurring or xenobiotic molecules, including drugs with metal binding potential, interactions with iron and other metals, and different forms of toxicity [20,182,183,184,185,186]. Each of these molecules and their iron or other metal complexes have diverse biological, pharmacological, and toxicological activity. In cancer, for example, the activity of such molecules may range from pro-oxidant and pro-carcinogenic to antioxidant and anti-carcinogenic effects and also many other effects, including the enhancement or inhibition of ferroptosis [187,188,189,190,191,192,193,194]. Similarly, many old and new carcinogens may contain or interact with iron, including food additives and coloring agents, many industrial and other chemicals, inhaled carcinogens from smoke, and other compounds from environmental pollution (Table 1) [195,196,197]. In particular, iron in heme and the nitroso heme complexes found mainly in processed meat have been implicated in the rise of colorectal and other cancers [198,199,200,201,202,203,204,205,206]. Carcinogenic toxicity is also suspected from other iron complexes present or formed in asbestos fibers, during cigarette smoking, and in barbecuing (Table 1) [207,208,209,210,211,212,213,214].
Changes in iron metabolism with iron toxicity implications are also observed during therapeutic interventions. For example, excess iron release causing increased transferrin saturation and formation of non-transferrin-bound-iron (NTBI) has been observed from tissue damage in iron overloading conditions and during cancer chemotherapy and radiotherapy treatments (Figure 5) [215,216,217,218,219,220,221,222,223,224,225]. Under these conditions, excess iron present in plasma can increase oxidative stress and potentially cause cellular and organ toxicity [226,227]. It can also facilitate microbial growth, mostly affecting immunocompromized cancer, transplanted, and other patients who are more susceptible to infections in comparison to normal individuals [103,228,229].
The presence of labile, catalytically active iron forms have been suggested in many other diseases, in addition to those considered that are associated with NTBI (Figure 5). In particular, catalytically active iron has been identified in the urine of different categories of patients, including acute and chronic kidney disease and cardiovascular disease [62,230,231,232].
Other diseases associated with abnormalities in iron metabolism and heterogeneous iron distribution with toxic side effects include those related to heme biosynthesis, mitochondrial iron metabolism, and rare cases of iron overload [21,44,233,234]. Among these are porphyria cutanea tarda, pyruvate kinase deficiency, congenital dyserythropoietic anaemia, congenital atransferrinaemia, etc. (Table 1) [235,236,237,238,239,240]. Many diseases have also been linked or associated to other abnormalities of mitochondrial iron metabolism and of different metabolic changes [241,242,243,244].
The molecular interactions of drugs and dietary molecules with iron and iron metabolic pathways are an area of continuous and thorough investigation regarding iron-related toxicity in the field of general pharmacology, toxicology, and nutrition. Thousands of such interactions of iron with drugs and dietary molecules have been reported in the scientific and medical literature emphasizing the prospect of free radical and other toxicity, especially affecting particular organs and characterized as a toxic side effect of the therapy (Figure 2). For example, anthracyclines, including doxorubicin, can cause acute and chronic cardiotoxicity in cancer patients, with the latter considered responsible for death in about one-third of patients during the long-term use of doxorubicin and long-term cardiotoxicity (Table 1). Labile iron and free radical production have been implicated as one of the mechanisms of anthracycline cardiotoxicity [245,246,247,248]. In this context, the EDTA iron chelator prodrug dexrazoxane has been widely used in cancer patients, including young children, for cardioprotection against doxorubicin and similar drug cardiotoxicity [249,250,251]. In contrast, the kidney is the major target organ of toxicity for cisplatin, an anticancer drug widely used for the treatment of about 50% of patients with different cancer categories [252,253]. In this case, labile iron and hemosiderin iron accumulation were implicated and suspected to play a role in the cisplatin-induced kidney injury [62,254,255,256]. The enhancement of iron-induced free radical toxicity has also been observed by the chelating drug EDTA, with some investigators questioning its safety in alternative medicine (Figure 2) [257,258].
Another area of iron-induced toxicity is related to the mode of siderophore action affecting some drugs with iron binding properties [259,260]. In particular, the iron chelating drug DF has been shown to promote the growth of some bacterial and fungal species with serious infectious toxicity implications, including fatalities [260,261]. In particular, among the established serious toxic side effects of DF are yersiniasis and mucormycosis, which mainly occur in iron-loaded and renal dialysis patients, respectively. In both cases DF appears to act as a siderophore and donates iron for the growth of Yersinia enterocolitica and Zygomyces, respectively [260,261,262]. The pharmacological and toxicological effects of many other categories of drugs with iron binding potential have not yet been fully investigated [20,259,263,264,265].
In addition to drugs, pharmacological and toxicological implications are expected from hundreds of dietary and biological molecules with iron-binding potential, as well as from their iron complexes (Table 1) [20,184]. For example, the interactions of iron and ascorbic acid (vitamin C), both of which are essential nutrients of nutritional, physiological, pharmacological, and toxicological interest [266]. Millions of people use these two nutrients found in food daily, and as pharmaceutical and nutraceutical preparations. In particular, one of the main functions and use of vitamin C is its antioxidant activity, with a wide range of applications in many diseases and clinical investigations, including cancer and ageing [266,267,268,269,270,271,272,273]. Vitamin C has iron and other metal-binding capacities and is also a reducing agent, forming a complex with iron (III) followed by reduction to iron (II) [266]. The biological and clinical activities of iron, ascorbate, and the ascorbate–iron complex can also be affected by many other nutrients and pharmaceutical preparations, which may reduce or potentiate iron toxicity [274,275,276,277,278]. Similar effects to those shown in the interactions of vitamin C and iron are expected from other biomolecules with iron-chelating potential such as folic acid, lipoic acid, and catecholamines, as well as metal ions competing with iron [59,184,185,279,280,281,282,283].
Abnormalities of iron metabolism with toxicity and other implications regarding the role of regulatory molecules, such as the apical divalent metal transported protein (DMT1), ferroportin, hepcidin, erythroferrone, erythropoietin, and related genomic, redoxomic, transcription, and other factors is currently under evaluation for determining their impact in many diseases and also for the design of new pharmaceuticals [284,285,286,287,288,289,290,291,292]. In particular, the interplay of regulatory molecules and other factors with pharmaceuticals on the regulation of diseases related to iron imbalance and toxicity could facilitate the design of optimal therapies and personalized medicine [293,294].
Despite the complexities and the many factors involved in diseases of iron metabolism and iron toxicity, the identification of the mechanisms involved, and the use of appropriate diagnostic criteria could provide efficient targeted therapies and the complete elimination of iron toxicity, as shown in the case of the transition of thalassaemia from a fatal to a chronic disease [39,88,295,296,297,298].

8. Conclusions

The spectrum of diseases associated with abnormalities of iron metabolism linked to iron toxicity and the number of patients affected worldwide is continuously increasing. New research developments, such as the identification of ferroptotic cell death in almost all common and rare diseases and the introduction of new diagnostic techniques such as the MRI T2* for the detection of iron deposits in major organs, have attracted the attention of many investigators working on many diseases and fields of medicine outside the field of iron metabolism and iron toxicity. In particular, the introduction of MRI T2* could not only be used in the monitoring of new chelation and other therapies in transfusional and other iron-loaded categories of patients but also therapies for other organs, such as those associated with the brain of non-iron-loaded categories of patients and many categories of several common neurodegenerative diseases, including Alzheimer’s disease and Parkinson’s disease. Furthermore, progress in the molecular interactions of iron and the prospect of increasing toxicity involving dietary, biochemical, xenobiotic molecules, and drugs have also attracted the interest of different specializations in medicine and the general public. In particular, the association of processed meat implicating heme and nitroso heme complexes in carcinogenesis and also mesothelioma caused by asbestos fibers containing iron, raised the alarm in relation to different forms of iron toxicity from dietary and environmental factors. Similarly, research interest is also increasing in relation to the implication of iron interactions in the toxic side effects of widely-used drugs such a doxorubicin and cisplatin or the decrease of efficacy in other widely-used drugs such as tetracyclines and hydroxyurea. Many other developments regarding the molecular and cellular effects of iron toxicity have also been identified and reported in a number of other fields of iron metabolism and diseases such as cancer and the anemia of chronic disease, which affects millions of patients.
Despite the improvements in identifying the mechanisms and in the methods of diagnosis of iron toxicity, the complexities and the involvement of many factors associated with different forms of iron toxicity have not yet led to improvements in many therapeutic fields for many diseases. However, there are exceptions, and progress in the therapeutic outcomes has been reported for some diseases, especially in the treatment of transfusional iron overload in TM, the disease with the highest rate of mortality and morbidity related to iron toxicity worldwide. In particular, the identification of the mechanisms of iron toxicity involved and the use of appropriate diagnostic criteria, including MRI T2*, helped in the introduction of effective targeted chelation therapies, which has overall resulted in the complete elimination of excess iron from the body of TM patients and also of iron toxicity, eventually causing the transition of thalassaemia from a fatal to a chronic disease.

Funding

No external funding was used.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The study was supported from internal funds of the Postgraduate Research Institute of Science, Technology, Environment and Medicine, a non-profit, charitable organization. We thank Christina N. Kontoghiorghe for the comments.

Conflicts of Interest

The author declares no conflict of interest.

Abbreviations

DFdeferoxamine
DFRAdeferasirox
DMT1divalent metal transported protein
EDTAethylenediaminetetraacetic acid
GPX4glutathione peroxidase
HCThematopoietic stem cell transplantation
Hbhemoglobin
HPFHhigh persistent fetal hemoglobin
L1deferiprone
LMWtlow molecular weight
MRImagnetic resonance imaging
NBIAneurodegeneration with brain iron accumulation
NTBInon-transferrin bound iron
PKANpantothenate kinase 2-associated neurodegeneration
RBCred blood cell
SCDsickle cell disease
TIthalassemia intermedia
TMbeta thalassemia major

References

  1. Hokland, P.; Daar, S.; Khair, W.; Sheth, S.; Taher, A.T.; Torti, L.; Hantaweepant, C.; Rund, D. Thalassaemia—A global view. Br. J. Haematol. 2023, 201, 199–214. [Google Scholar] [CrossRef]
  2. Cattoni, A.; Capitoli, G.; Casagranda, S.; Corti, P.; Adavastro, M.; Molinaro, A.; Di Gennaro, F.; Bonanomi, S.; Biondi, A.; Galimberti, S.; et al. Iron Overload Following Hematopoietic Stem Cell Transplantation: Prevalence, Severity, and Management in Children and Adolescents with Malignant and Nonmalignant Diseases. Transplant. Cell Ther. 2023, 29, 271.e1–271.e12. [Google Scholar] [CrossRef] [PubMed]
  3. Tang, A.Y.; Zhou, M.; Maillis, A.N.; Lai, K.W.; Lane, P.A.; Snyder, A.B. Trends in blood transfusion, hydroxyurea use, and iron overload among children with sickle cell disease enrolled in Medicaid, 2004–2019. Pediatr. Blood Cancer 2023, 70, e30152. [Google Scholar] [CrossRef] [PubMed]
  4. Gu, Y.; Estcourt, L.J.; Doree, C.; Hopewell, S.; Vyas, P. Comparison of a restrictive versus liberal red cell transfusion policy for patients with myelodysplasia, aplastic anaemia, and other congenital bone marrow failure disorders. Cochrane Database Syst. Rev. 2015, 2015, CD011577. [Google Scholar] [CrossRef] [PubMed]
  5. Styczyński, J.; Słomka, A.; Łęcka, M.; Albrecht, K.; Romiszewski, M.; Pogorzała, M.; Kubicka, M.; Kuryło-Rafińska, B.; Tejza, B.; Gadomska, G.; et al. Soluble Hemojuvelin and Ferritin: Potential Prognostic Markers in Pediatric Hematopoietic Cell Transplantation. Cancers 2023, 15, 1041. [Google Scholar] [CrossRef]
  6. Iancu, T.C. Iron overload. Mol. Aspects Med. 1983, 6, 1–100. [Google Scholar] [CrossRef]
  7. Bruzzese, A.; Martino, E.A.; Mendicino, F.; Lucia, E.; Olivito, V.; Bova, C.; Filippelli, G.; Capodanno, I.; Neri, A.; Morabito, F.; et al. Iron chelation therapy. Eur. J. Haematol. 2023, 110, 490–497. [Google Scholar] [CrossRef]
  8. Rai, P.; Okhomina, V.I.; Kang, G.; Martinez, H.R.; Hankins, J.S.; Joshi, V. Longitudinal effect of disease-modifying therapy on left ventricular diastolic function in children with sickle cell anemia. Am. J. Hematol. 2023, 98, 838–847. [Google Scholar] [CrossRef]
  9. Zurlo, M.; De Stefano, P.; Borgna-Pignatti, C.; Di Palma, A.; Melevendi, C.; Piga, A.; Di Gregorio, F.; Burattini, M.; Terzoli, S. Survival and causes of death in thalassaemia major. Lancet 1989, 2, 27–30. [Google Scholar] [CrossRef] [PubMed]
  10. Adams, P.C. Epidemiology and diagnostic testing for hemochromatosis and iron overload. Int. J. Lab. Hematol. 2015, 37, 25–30. [Google Scholar] [CrossRef]
  11. Lehéricy, S.; Roze, E.; Goizet, C.; Mochel, F. MRI of neurodegeneration with brain iron accumulation. Curr. Opin. Neurol. 2020, 33, 462–473. [Google Scholar] [CrossRef] [PubMed]
  12. Kolnagou, A.; Fessas, C.; Papatryphonas, A.; Economides, C.; Kontoghiorghes, G.J. Prophylactic use of deferiprone (L1) and magnetic resonance imaging T2* or T2 for preventing heart disease in thalassaemia. Br. J. Haematol. 2004, 127, 360–361. [Google Scholar] [CrossRef]
  13. Kontoghiorghes, G.J.; Kontoghiorghe, C.N. Efficacy and safety of iron-chelation therapy with deferoxamine, deferiprone, and deferasirox for the treatment of iron-loaded patients with non-transfusion-dependent thalassemia syndromes. Drug Des. Dev. Ther. 2016, 10, 465–481. [Google Scholar] [CrossRef]
  14. Telfer, P.; Coen, P.G.; Christou, S.; Hadjigavriel, M.; Kolnakou, A.; Pangalou, E.; Pavlides, N.; Psiloines, M.; Simamonian, K.; Skordos, G.; et al. Survival of medically treated thalassemia patients in Cyprus. Trends and risk factors over the period 1980–2004. Haematologica 2006, 91, 1187–1192. [Google Scholar]
  15. Au, W.Y.; Lee, V.; Lau, C.W.; Yau, J.; Chan, D.; Chan, E.Y.T.; Cheung, W.W.W.; Ha, S.Y.; Kho, B.; Lee, C.Y.; et al. A synopsis of current care of thalassaemia major patients in Hong Kong. Hong Kong Med. J. 2011, 17, 261–266. [Google Scholar] [PubMed]
  16. Maggio, A.; Filosa, A.; Vitrano, A.; Aloj, G.; Kattamis, A.; Ceci, A.; Fucharoen, S.; Cianciulli, P.; Grady, R.W.; Prossomariti, L.; et al. Iron chelation therapy in thalassemia major: A systematic review with meta-analyses of 1520 patients included on randomized clinical trials. Blood Cells Mol. Dis. 2011, 47, 166–175. [Google Scholar] [CrossRef]
  17. Kontoghiorghes, G.J. The aim of iron chelation therapy in thalassaemia. Eur. J. Haematol. 2017, 99, 465–466. [Google Scholar] [CrossRef] [PubMed]
  18. Cairo, G.; Bernuzzi, F.; Recalcati, S. A precious metal: Iron, an essential nutrient for all cells. Genes Nutr. 2006, 1, 25–39. [Google Scholar] [CrossRef] [PubMed]
  19. Yoshida, T.; Prudent, M.; D’Alessandro, A. Red blood cell storage lesion: Causes and potential clinical consequences. Blood Transfus. 2019, 17, 27–52. [Google Scholar] [CrossRef]
  20. Kontoghiorghes, G.J.; Kontoghiorghe, C.N. Iron and Chelation in Biochemistry and Medicine: New Approaches to Controlling Iron Metabolism and Treating Related Diseases. Cells 2020, 9, 1456. [Google Scholar] [CrossRef] [PubMed]
  21. Galaris, D.; Pantopoulos, K. Oxidative Stress and Iron Homeostasis: Mechanistic and Health Aspects. Crit. Rev. Clin. Lab. Sci. 2008, 45, 1–23. [Google Scholar] [CrossRef]
  22. Halliwell, B.; Gutteridge, J.M.C.; Cross, C.E. Free radicals, antioxidants and human disease: Where are we now? J. Lab. Clin. Med. 1992, 119, 598–620. [Google Scholar]
  23. Fibach, E. The Redox Balance and Membrane Shedding in RBC Production, Maturation, and Senescence. Front. Physiol. 2021, 12, 604738. [Google Scholar] [CrossRef]
  24. Asaro, R.J.; Zhu, Q.; Cabrales, P. Erythrocyte Aging, Protection via Vesiculation: An Analysis Methodology via Oscillatory Flow. Front. Physiol. 2018, 9, 1607. [Google Scholar] [CrossRef] [PubMed]
  25. Alaarg, A.; Schiffelers, R.M.; van Solinge, W.W.; van Wijk, R. Red blood cell vesiculation in hereditary hemolytic anemia. Front. Physiol. 2013, 4, 365. [Google Scholar] [CrossRef]
  26. Tzounakas, V.L.; Anastasiadi, A.T.; Lekka, M.E.; Papageorgiou, E.G.; Stamoulis, K.; Papassideri, I.S.; Kriebardis, A.G.; Antonelou, M.H. Deciphering the Relationship Between Free and Vesicular Hemoglobin in Stored Red Blood Cell Units. Front. Physiol. 2022, 13, 840995. [Google Scholar] [CrossRef] [PubMed]
  27. Tolosano, E.; Altruda, F. Hemopexin: Structure, Function, and Regulation. DNA Cell Boil. 2002, 21, 297–306. [Google Scholar] [CrossRef] [PubMed]
  28. Shih, A.W.; McFarlane, A.; Verhovsek, M. Haptoglobin testing in hemolysis: Measurement and interpretation. Am. J. Hematol. 2014, 89, 443–447. [Google Scholar] [CrossRef]
  29. Weatherall, D.J.; Clegg, J.B. Inherited haemoglobin disorders: An increasing global health problem. Bull. World Health Organ. 2001, 79, 704–712. [Google Scholar]
  30. Anonymous. Community control of hereditary anaemias: Memorandum from a WHO meeting. Bull. World Health Organ. 1983, 61, 63–80. [Google Scholar]
  31. Teawtrakul, N.; Chansung, K.; Sirijerachai, C.; Wanitpongpun, C.; Thepsuthammarat, K. The impact and disease burden of thalassemia in Thailand: A population-based study in 2010. J. Med. Assoc. Thail. 2012, 95, 95. [Google Scholar]
  32. Pippard, M.J.; Callender, S.T.; Warner, G.T.; Weatherall, D.J. Iron absorption and loading in beta-thalassaemia intermedia. Lancet 1979, 2, 819–821. [Google Scholar] [CrossRef]
  33. Pootrakul, P.; Kitcharoen, K.; Yansukon, P.; Wasi, P.; Fucharoen, S.; Charoenlarp, P.; Brittenham, G.; Pippard, M.J.; Finch, C.A. The effect of erythroid hyperplasia on iron balance. Blood 1988, 71, 1124–1129. [Google Scholar] [CrossRef]
  34. Kontoghiorghes, G.J.; Spyrou, A.; Kolnagou, A. Iron Chelation Therapy in Hereditary Hemochromatosis and Thalassemia Intermedia: Regulatory and Non Regulatory Mechanisms of Increased Iron Absorption. Hemoglobin 2010, 34, 251–264. [Google Scholar] [CrossRef]
  35. Kontoghiorghes, G.J.; Kolnagou, A. Molecular Factors and Mechanisms Affecting Iron and Other Metal Excretion or Absorption in Health and Disease. The Role of Natural and Synthetic Chelators. Curr. Med. Chem. 2005, 12, 2695–2709. [Google Scholar] [CrossRef] [PubMed]
  36. Galanello, R.; Cao, A. Relationship between Genotype and Phenotype: Thalassemia Intermediaa. Ann. N. Y. Acad. Sci. 1998, 850, 325–333. [Google Scholar] [CrossRef] [PubMed]
  37. Borgna-Pignatti, C. Modern treatment of thalassaemia intermedia. Br. J. Haematol. 2007, 138, 291–304. [Google Scholar] [CrossRef] [PubMed]
  38. Singer, S.T. Variable Clinical Phenotypes of α-Thalassemia Syndromes. Sci. World J. 2008, 9, 615–625. [Google Scholar] [CrossRef] [PubMed]
  39. Kolnagou, A.; Kleanthous, M.; Kontoghiorghes, G.J. Benefits and Risks in Polypathology and Polypharmacotherapy Challenges in the Era of the Transition of Thalassaemia from a Fatal to a Chronic or Curable Disease. Front. Biosci. 2022, 14, 18. [Google Scholar] [CrossRef]
  40. Inamoto, Y.; Lee, S.J. Late effects of blood and marrow transplantation. Haematologica 2017, 102, 614–625. [Google Scholar] [CrossRef] [PubMed]
  41. Gratwohl, A.; Pasquini, M.C.; Aljurf, M.; Atsuta, Y.; Baldomero, H.; Foeken, L.; Gratwohl, M.; Bouzas, L.F.; Confer, D.; Frauendorfer, K.; et al. Worldwide Network for Blood and Marrow Transplantation (WBMT). One million haemopoietic stem-cell transplants: A retrospective observational study. Lancet Haematol. 2015, 2, e91–e100. [Google Scholar] [CrossRef]
  42. Shenoy, S.; Angelucci, E.; Arnold, S.D.; Baker, K.S.; Bhatia, M.; Bresters, D.; Dietz, A.C.; De La Fuente, J.; Duncan, C.; Gaziev, J.; et al. Current Results and Future Research Priorities in Late Effects after Hematopoietic Stem Cell Transplantation for Children with Sickle Cell Disease and Thalassemia: A Consensus Statement from the Second Pediatric Blood and Marrow Transplant Consortium International Conference on Late Effects after Pediatric Hematopoietic Stem Cell Transplantation. Biol. Blood Marrow Transplant. 2017, 23, 552–561. [Google Scholar] [CrossRef]
  43. Germing, U.; Schroeder, T.; Kaivers, J.; Kündgen, A.; Kobbe, G.; Gattermann, N. Novel therapies in low- and high-risk myelodysplastic syndrome. Expert Rev. Hematol. 2019, 12, 893–908. [Google Scholar] [CrossRef]
  44. Ricci, A.; Di Betto, G.; Bergamini, E.; Buzzetti, E.; Corradini, E.; Ventura, P. Iron Metabolism in the Disorders of Heme Biosynthesis. Metabolites 2022, 12, 819. [Google Scholar] [CrossRef]
  45. Gao, J.; Zhou, Q.; Wu, D.; Chen, L. Mitochondrial iron metabolism and its role in diseases. Clin. Chim. Acta 2021, 513, 6–12. [Google Scholar] [CrossRef]
  46. Girard, S.; Genevieve, F.; Rault, E.; Fenneteau, O.; Lesesve, J.-F. When Ring Sideroblasts on Bone Marrow Smears Are Inconsistent with the Diagnosis of Myelodysplastic Neoplasms. Diagnostics 2022, 12, 1752. [Google Scholar] [CrossRef]
  47. Brissot, P.; Troadec, M.-B.; Loréal, O.; Brissot, E. Pathophysiology and classification of iron overload diseases; update 2018. Transfus. Clin. Biol. 2019, 26, 80–88. [Google Scholar] [CrossRef]
  48. Camaschella, C.; Nai, A. Ineffective erythropoiesis and regulation of iron status in iron loading anaemias. Br. J. Haematol. 2016, 172, 512–523. [Google Scholar] [CrossRef]
  49. Verma, I.C. Burden of genetic disorders in India. Indian J. Pediatr. 2000, 67, 893–898. [Google Scholar] [CrossRef]
  50. Gozzelino, R.; Arosio, P. Iron Homeostasis in Health and Disease. Int. J. Mol. Sci. 2016, 17, 130. [Google Scholar] [CrossRef]
  51. Anderson, G.J.; Frazer, D.M. Current understanding of iron homeostasis. Am. J. Clin. Nutr. 2017, 106 (Suppl. 6), 1559S–1566S. [Google Scholar] [CrossRef]
  52. Andrews, N.C. Closing the iron gate. N. Engl. J. Med. 2012, 366, 376–377. [Google Scholar] [CrossRef]
  53. Kim, A.; Nemeth, E. New insights into iron regulation and erythropoiesis. Curr. Opin. Hematol. 2015, 22, 199–205. [Google Scholar] [CrossRef]
  54. Galaris, D.; Barbouti, A.; Pantopoulos, K. Iron homeostasis and oxidative stress: An intimate relationship. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2019, 1866, 118535. [Google Scholar] [CrossRef]
  55. Sargent, P.J.; Farnaud, S.; Evans, R.W. Structure/Function Overview of Proteins Involved in Iron Storage and Transport. Curr. Med. Chem. 2005, 12, 2683–2693. [Google Scholar] [CrossRef]
  56. Theil, E.C. Ferritin: The Protein Nanocage and Iron Biomineral in Health and in Disease. Inorg. Chem. 2013, 52, 12223–12233. [Google Scholar] [CrossRef]
  57. Iancu, T.C. Ferritin and hemosiderin in pathological tissues. Electron Microsc. Rev. 1992, 5, 209–229. [Google Scholar] [CrossRef]
  58. Denisov, E.T.; Afanas’Ev, I.B. Oxidation and Antioxidants in Organic Chemistry and Biology; CRC Press: Boca Raton, FL, USA; Taylor and Francis group: Abingdon, UK, 2005. [Google Scholar] [CrossRef]
  59. Kontoghiorghes, G.J. Iron Chelation in Biochemistry and Medicine; Free radicals, oxidant stress and drug action; Rice-Evans, C., Ed.; Rechelieu Press: London, UK, 1987; pp. 277–303. [Google Scholar]
  60. Duračková, Z. Some current insights into oxidative stress. Physiol. Res. 2010, 59, 459–469. [Google Scholar] [CrossRef]
  61. Nakamura, T.; Naguro, I.; Ichijo, H. Iron homeostasis and iron-regulated ROS in cell death, senescence and human diseases. Biochim. Biophys. Acta (BBA) Gen. Subj. 2019, 1863, 1398–1409. [Google Scholar] [CrossRef]
  62. Shah, S.V.; Rajapurkar, M.M. The Role of Labile Iron in Kidney Disease and Treatment with Chelation. Hemoglobin 2009, 33, 378–385. [Google Scholar] [CrossRef]
  63. Reeder, B.J.; Wilson, M.T. Hemoglobin and Myoglobin Associated Oxidative Stress: From Molecular Mechanisms to Disease States. Curr. Med. Chem. 2005, 12, 2741–2751. [Google Scholar] [CrossRef]
  64. Reeder, B.J.; Strader, M.B.; Alayash, A.I.; Di Domenico, F.; Barone, E.; Perluigi, M.; Butterfield, D.A.; Feliers, D.; Lee, H.J.; Kasinath, B.S.; et al. Redox and Peroxidase Activities of the Hemoglobin Superfamily: Relevance to Health and Disease. Antioxidants Redox Signal. 2017, 26, 763–776. [Google Scholar] [CrossRef] [PubMed]
  65. Jacobs, A. An intracellular transit iron pool. Ciba. Found Symp. 1976, 51, 91–106. [Google Scholar]
  66. Kontoghiorghes, G.J.; Efstathiou, A.; Kleanthous, M.; Michaelides, Y.; Kolnagou, A. Risk/Benefit Assessment, Advantages Over Other Drugs and Targeting Methods in the Use of Deferiprone as a Pharmaceutical Antioxidant in Iron Loading and Non Iron Loading Conditions. Hemoglobin 2009, 33, 386–397. [Google Scholar] [CrossRef]
  67. Kontoghiorghes, G.J.; Kontoghiorghe, C.N. Prospects for the introduction of targeted antioxidant drugs for the prevention and treatment of diseases related to free radical pathology. Expert Opin. Investig. Drugs 2019, 28, 593–603. [Google Scholar] [CrossRef]
  68. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef] [PubMed]
  69. Xie, Y.; Hou, W.; Song, X.; Yu, Y.; Huang, J.; Sun, X.; Kang, R.; Tang, D. Ferroptosis: Process and function. Cell Death Differ. 2016, 23, 369–379. [Google Scholar] [CrossRef] [PubMed]
  70. Cao, J.Y.; Dixon, S.J. Mechanisms of ferroptosis. Cell Mol. Life Sci. 2016, 73, 2195–2209. [Google Scholar] [CrossRef]
  71. Lu, B.; Chen, X.B.; Ying, M.D.; He, Q.J.; Cao, J.; Yang, B. The Role of Ferroptosis in Cancer Development and Treatment Response. Front. Pharmacol. 2018, 8, 992. [Google Scholar] [CrossRef]
  72. Li, G.; Liang, Y.; Yang, H.; Zhang, W.; Xie, T. The Research Landscape of Ferroptosis in Cancer: A Bibliometric Analysis. Front. Cell Dev. Biol. 2022, 10, 841724. [Google Scholar] [CrossRef]
  73. Wang, D.; Tang, L.; Zhang, Y.; Ge, G.; Jiang, X.; Mo, Y.; Wu, P.; Deng, X.; Li, L.; Zuo, S.; et al. Regulatory pathways and drugs associated with ferroptosis in tumors. Cell Death Dis. 2022, 13, 544. [Google Scholar] [CrossRef]
  74. Liu, L.; Li, L.; Li, M.; Luo, Z. Autophagy-Dependent Ferroptosis as a Therapeutic Target in Cancer. Chemmedchem 2021, 16, 2942–2950. [Google Scholar] [CrossRef] [PubMed]
  75. Xiao, L.; Huang, H.; Fan, S.; Zheng, B.; Wu, J.; Zhang, J.; Pi, J.; Xu, J.-F. Ferroptosis: A mixed blessing for infectious diseases. Front. Pharmacol. 2022, 13, 992734. [Google Scholar] [CrossRef] [PubMed]
  76. Lin, Y.; Xu, W.; Hou, Y.; Wang, S.; Zhang, H.; Ran, M.; Huang, Y.; Wang, Y.; Yang, G. The multifaceted role of ferroptosis in kidney diseases. Chem. Interactions 2022, 365, 110107. [Google Scholar] [CrossRef] [PubMed]
  77. Wang, K.; Chen, X.-Z.; Wang, Y.-H.; Cheng, X.-L.; Zhao, Y.; Zhou, L.-Y.; Wang, K. Emerging roles of ferroptosis in cardiovascular diseases. Cell Death Discov. 2022, 8, 394. [Google Scholar] [CrossRef]
  78. Thapa, K.; Khan, H.; Kanojia, N.; Singh, T.G.; Kaur, A.; Kaur, G. Therapeutic Insights on Ferroptosis in Parkinson’s disease. Eur. J. Pharmacol. 2022, 930, 175133. [Google Scholar] [CrossRef]
  79. Jacobs, W.; Lammens, M.; Kerckhofs, A.; Voets, E.; Van San, E.; Van Coillie, S.; Peleman, C.; Mergeay, M.; Sirimsi, S.; Matheeussen, V.; et al. Fatal lymphocytic cardiac damage in coronavirus disease 2019 (COVID-19): Autopsy reveals a ferroptosis signature. ESC Heart Fail. 2020, 7, 3772–3781. [Google Scholar] [CrossRef]
  80. Kontoghiorghes, G.J. Deferiprone: A Forty-Year-Old Multi-Targeting Drug with Possible Activity against COVID-19 and Diseases of Similar Symptomatology. Int. J. Mol. Sci. 2022, 23, 6735. [Google Scholar] [CrossRef]
  81. Yang, Y.; Wang, Y.; Guo, L.; Gao, W.; Tang, T.-L.; Yan, M. Interaction between macrophages and ferroptosis. Cell Death Dis. 2022, 13, 355. [Google Scholar] [CrossRef]
  82. Gao, M.; Monian, P.; Pan, Q.; Zhang, W.; Xiang, J.; Jiang, X. Ferroptosis is an autophagic cell death process. Cell Res. 2016, 26, 1021–1032. [Google Scholar] [CrossRef]
  83. Wang, Y.; Chen, Q.; Song, H.; Zhang, Y.; Chen, H.; Liu, P.; Sun, T.; Jiang, C. A Triple Therapeutic Strategy with Antiexosomal Iron Efflux for Enhanced Ferroptosis Therapy and Immunotherapy. Small 2022, 18, e2201704. [Google Scholar] [CrossRef]
  84. Hou, W.; Xie, Y.; Song, X.; Sun, X.; Lotze, M.T.; Zeh, H.J., 3rd; Kang, R.; Tang, D. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy 2016, 12, 1425–1428. [Google Scholar] [CrossRef]
  85. Park, E.; Chung, S.W. ROS-mediated autophagy increases intracellular iron levels and ferroptosis by ferritin and transferrin receptor regulation. Cell Death Dis. 2019, 10, 822. [Google Scholar] [CrossRef] [PubMed]
  86. Kyriacou, K.; Michaelides, Y.; Senkus, R.; Simamonian, K.; Pavlides, N.; Antoniades, L.; Zambartas, C. Ultrastructural pathology of the heart in patients with beta-thalassaemia major. Ultrastruct. Pathol. 2000, 24, 75–81. [Google Scholar] [CrossRef]
  87. Iancu, T.C.; Neustein, H.B.; Landing, B.H. The Liver in Thalassaemia Major: Ultra-Structural Observations. Ciba Foundation Symposium 51-Iron Metabolism; John Wiley & Sons, Ltd.: Chichester, UK, 1977; pp. 293–316. [Google Scholar] [CrossRef]
  88. Kolnagou, A.; Michaelides, Y.; Kontos, C.; Kyriacou, K.; Kontoghiorghes, G.J. Myocyte Damage and Loss of Myofibers is the Potential Mechanism of Iron Overload Toxicity in Congestive Cardiac Failure in Thalassemia. Complete Reversal of the Cardiomyopathy and Normalization of Iron Load by Deferiprone. Hemoglobin 2008, 32, 17–28. [Google Scholar] [CrossRef] [PubMed]
  89. Kontoghiorghes, G.J.; Kolnagou, A.; Kontoghiorghe, C.N. New targeted therapies and diagnostic methods for iron overload diseases. Front. Biosci. 2018, 10, 1–20. [Google Scholar] [CrossRef] [PubMed]
  90. Aessopos, A.; Kati, M.; Farmakis, D. Heart disease in thalassemia intermedia: A review of the underlying pathophysiology. Haematologica 2007, 92, 658–665. [Google Scholar] [CrossRef]
  91. Sonakul, D.; Pacharee, P.; Wasi, P.; Fucharoen, S. Cardiac pathology in 47 patients with beta thalassaemia/haemoglobin E. Southeast Asian J. Trop. Med. Public Health 1984, 15, 554–563. [Google Scholar]
  92. Mancuso, A. Evidence-based medicine and Management of Hepatocellular Carcinoma in Thalassemia. BMC Gastroenterol. 2020, 20, 409. [Google Scholar] [CrossRef]
  93. Ricchi, P. Liver fibrosis in young patients with transfusion-dependent thalassaemia (TDT), understanding the role of steatosis. Br. J. Haematol. 2023, 200, 399–400. [Google Scholar] [CrossRef]
  94. Tolis, G.J.; Vlachopapadopoulou, E.; Karydis, I. Reproductive health in patients with beta-thalassemia. Curr. Opin. Pediatr. 1996, 8, 406–410. [Google Scholar] [CrossRef] [PubMed]
  95. Angelopoulos, N.G.; Zervas, A.; Livadas, S.; Adamopoulos, I.; Giannopoulos, D.; Goula, A.; Tolis, G. Reduced insulin secretion in normoglycaemic patients with β-thalassaemia major. Diabet. Med. 2006, 23, 1327–1331. [Google Scholar] [CrossRef] [PubMed]
  96. Economides, C.P.; Soteriades, E.S.; Hadjigavriel, M.; Seimenis, I.; Karantanas, A. Iron deposits in the knee joints of a thalassemic patient. Acta Radiol. Short Rep. 2013, 2, 1–5. [Google Scholar] [CrossRef]
  97. Mavrogeni, S.I.; Gotsis, E.D.; Markussis, V.; Tsekos, N.; Politis, C.; Vretou, E.; Kermastinos, D. T2 relaxation time study of iron overload in b-thalassemia. MAGMA 1998, 6, 7–12. [Google Scholar] [CrossRef]
  98. Anderson, L.; Holden, S.; Davis, B.; Prescott, E.; Charrier, C.; Bunce, N.; Firmin, D.; Wonke, B.; Porter, J.; Walker, J.; et al. Cardiovascular T2-star (T2*) magnetic resonance for the early diagnosis of myocardial iron overload. Eur. Heart J. 2001, 22, 2171–2179. [Google Scholar] [CrossRef] [PubMed]
  99. Pennell, D.J. T2* Magnetic Resonance and Myocardial Iron in Thalassemia. Ann. N. Y. Acad. Sci. 2005, 1054, 373–378. [Google Scholar] [CrossRef] [PubMed]
  100. Kolnagou, A.; Yazman, D.; Economides, C.; Eracleous, E.; Kontoghiorghes, G.J. Uses and Limitations of Serum Ferritin, Magnetic Resonance Imaging T2 and T2* in the Diagnosis of Iron Overload and in the Ferrikinetics of Normalization of the Iron Stores in Thalassemia Using the International Committee on Chelation Deferiprone/Deferoxamine Combination Protocol. Hemoglobin 2009, 33, 312–322. [Google Scholar] [CrossRef]
  101. Rahav, G.; Volach, V.; Shapiro, M.; Rund, D.; Rachmilewitz, E.A.; Goldfarb, A. Severe infections in thalassaemic patients: Prevalence and predisposing factors. Br. J. Haematol. 2006, 133, 667–674. [Google Scholar] [CrossRef]
  102. Weinberg, E.D. Iron availability and infection. Biochim. Biophys. Acta (BBA) Gen. Subj. 2009, 1790, 600–605. [Google Scholar] [CrossRef]
  103. Kontoghiorghes, G.J.; Kolnagou, A.; Skiada, A.; Petrikkos, G. The Role of Iron and Chelators on Infections in Iron Overload and Non Iron Loaded Conditions: Prospects for the Design of New Antimicrobial Therapies. Hemoglobin 2010, 34, 227–239. [Google Scholar] [CrossRef]
  104. Kolnagou, A.; Michaelides, Y.; Kontoghiorghe, C.N.; Kontoghiorghes, G.J. The importance of spleen, spleen iron, and splenectomy for determining total body iron load, ferrikinetics, and iron toxicity in thalassemia major patients. Toxicol. Mech. Methods 2013, 23, 34–41. [Google Scholar] [CrossRef] [PubMed]
  105. Skandalis, K.; Vlachos, C.; Pliakou, X.; Gaitanis, G.; Kapsali, E.; Bassukas, I.D. Higher Serum Ferritin Levels Correlate with an Increased Risk of Cutaneous Morbidity in Adult Patients with β-Thalassemia: A Single-Center Retrospective Study. Acta Haematol. 2016, 135, 124–130. [Google Scholar] [CrossRef] [PubMed]
  106. Anderson, L.J.; Westwood, M.A.; Prescott, E.; Walker, J.M.; Pennell, D.J.; Wonke, B. Development of Thalassaemic Iron Overload Cardiomyopathy despite Low Liver Iron Levels and Meticulous Compliance to Desferrioxamine. Acta Haematol. 2006, 115, 106–108. [Google Scholar] [CrossRef] [PubMed]
  107. Kolnagou, A.; Economides, C.; Eracleous, E.; Kontoghiorghes, G.J. Low Serum Ferritin Levels are Misleading for Detecting Cardiac Iron Overload and Increase the Risk of Cardiomyopathy in Thalassemia Patients. The Importance of Cardiac Iron Overload Monitoring Using Magnetic Resonance Imaging T2 and T2*. Hemoglobin 2006, 30, 219–227. [Google Scholar] [CrossRef] [PubMed]
  108. Papakonstantinou, O.; Alexopoulou, E.; Economopoulos, N.; Benekos, O.; Kattamis, A.; Kostaridou, S.; Ladis, V.; Efstathopoulos, E.; Gouliamos, A.; Kelekis, N.L. Assessment of iron distribution between liver, spleen, pancreas, bone marrow, and myocardium by means of R2 relaxometry with MRI in patients with beta-thalassemia major. J. Magn. Reson. Imaging 2009, 29, 853–859. [Google Scholar] [CrossRef] [PubMed]
  109. Kolnagou, A.; Natsiopoulos, K.; Kleanthous, M.; Ioannou, A.; Kontoghiorghes, G.J. Liver iron and serum ferritin levels are misleading for estimating cardiac, pancreatic, splenic and total body iron load in thalassemia patients: Factors influencing the heterogenic distribution of excess storage iron in organs as identified by MRI T2*. Toxicol. Mech. Methods 2013, 23, 48–56. [Google Scholar] [CrossRef]
  110. Wood, J.C. Diagnosis and management of transfusion iron overload: The role of imaging. Am. J. Hematol. 2007, 82 (Suppl. 12), 1132–1135. [Google Scholar] [CrossRef]
  111. Chacko, J.; Pennell, D.J.; Tanner, M.A.; Hamblin, T.J.; Wonke, B.; Levy, T.; Thomas, P.W.; Killick, S.B. Myocardial iron loading by magnetic resonance imaging T2* in good prognostic myelodysplastic syndrome patients on long-term blood transfusions. Br. J. Haematol. 2007, 138, 587–593. [Google Scholar] [CrossRef]
  112. Bs, C.J.B.; Coates, T.D.; Wood, J.C. Spleen R2 and R2* in iron-overloaded patients with sickle cell disease and thalassemia major. J. Magn. Reson. Imaging 2009, 29, 357–364. [Google Scholar] [CrossRef]
  113. Kontoghiorghe, C.N.; Kolnagou, A.; Kontoghiorghes, G.J. Dietary and pharmacological factors affecting iron absorption in mice and man (Comment for a Letter to the editor). Haematologica 2016, 101, e120–e121. [Google Scholar] [CrossRef]
  114. Kolnagou, A.; Kleanthous, M.; Kontoghiorghes, G.J. Efficacy, Compliance and Toxicity Factors Are Affecting the Rate of Normalization of Body Iron Stores in Thalassemia Patients Using The Deferiprone and Deferoxamine Combination Therapy. Hemoglobin 2011, 35, 186–198. [Google Scholar] [CrossRef] [PubMed]
  115. Kolnagou, A.; Kontoghiorghe, C.N.; Kontoghiorghes, G.J. Transfusion-Related Acute Lung Injury (TRALI) in two Thalassaemia Patients Caused by the Same Multiparous Blood Donor. Mediterr. J. Hematol. Infect. Dis. 2017, 9, e2017060. [Google Scholar] [PubMed]
  116. Barton, J.C.; Edwards, C.Q. (Eds.) Hemochromatosis: Genetics, Pathophysiology, Diagnosis and Treatment; Cambridge University Press: Cambridge, UK, 2000. [Google Scholar]
  117. Feder, J.N.; Gnirke, A.; Thomas, W.; Tsuchihashi, Z.; Ruddy, D.A.; Basava, A.; Dormishian, F.; Domingo, R., Jr.; Ellis, M.C.; Fullan, A.; et al. A novel MHC class I–like gene is mutated in patients with hereditary haemochromatosis. Nat. Genet. 1996, 13, 399–408. [Google Scholar] [CrossRef] [PubMed]
  118. Pietrangelo, A. Hereditary Hemochromatosis—A New Look at an Old Disease. N. Engl. J. Med. 2004, 350, 2383–2397. [Google Scholar] [CrossRef]
  119. Allen, K.J.; Gurrin, L.C.; Constantine, C.C.; Osborne, N.J.; Delatycki, M.B.; Nicoll, A.J.; McLaren, C.E.; Bahlo, M.; Nisselle, A.E.; Vulpe, C.D.; et al. Iron-overload-related disease in HFE hereditary hemochromatosis. N. Engl. J. Med. 2008, 358, 221–230. [Google Scholar] [CrossRef]
  120. Kontoghiorghes, G.J. The Vital Role Played by Deferiprone in the Transition of Thalassaemia from a Fatal to a Chronic Disease and Challenges in Its Repurposing for Use in Non-Iron-Loaded Diseases. Pharmaceuticals 2023, 16, 1016. [Google Scholar] [CrossRef]
  121. Tenneti, P.; Chojecki, A.; Knovich, M.A. Iron overload in the HCT patient: A review. Bone Marrow Transplant. 2021, 56, 1794–1804. [Google Scholar] [CrossRef]
  122. Kontoghiorghes, G.J. How to manage iron toxicity in post-allogeneic hematopoietic stem cell transplantation? Expert. Rev. Hematol. 2020, 13, 299–302. [Google Scholar] [CrossRef]
  123. Meloni, A.; Pistoia, L.; Gamberini, M.R.; Ricchi, P.; Cecinati, V.; Sorrentino, F.; Cuccia, L.; Allò, M.; Righi, R.; Fina, P.; et al. The Link of Pancreatic Iron with Glucose Metabolism and Cardiac Iron in Thalassemia Intermedia: A Large, Multicenter Observational Study. J. Clin. Med. 2021, 10, 5561. [Google Scholar] [CrossRef]
  124. Verberckmoes, B.; Dekeyzer, S.; Decaestecker, K. The Dark Pituitary: Hemochromatosis as a Lesser-Known Cause of Pituitary Dysfunction. J. Belg. Soc. Radiol. 2022, 106, 63. [Google Scholar] [CrossRef]
  125. Byrne, D.; Walsh, J.P.; Murphy, R.T.; McMorrow, J.; Fagan, A.J.; Kecler Poetrzyk, A.; Durand, M.E.; Norris, S.; McKiernan, S.; King, G.; et al. Cardiac Magnetic Resonance at 3.0 T in Patients with C282Y Homozygous Hereditary Hemochromatosis: Superiority of Radial and Circumferential Strain Over Cardiac T2* Measurements at Baseline and at Post Venesection Follow-up. J. Thorac. Imaging. 2022, 37, 300–306. [Google Scholar] [CrossRef]
  126. Zhou, Y.; Luo, J. Relationship between Iron deposition and T lymphocytes in children with β-thalassemia with haematopoietic stem cell transplantation. Front. Pediatr. 2022, 10, 939157. [Google Scholar] [CrossRef]
  127. Aboobacker, F.N.; Dixit, G.; Lakshmi, K.M.; Korula, A.; Abraham, A.; George, B.; Mathews, V.; Srivastava, A. Outcome of iron reduction therapy in ex-thalassemics. PLoS ONE 2021, 16, e0238793. [Google Scholar] [CrossRef]
  128. Drayer, B.P.; Olanow, W.; Burger, P.; Johnson, G.A.; Herfkens, R.; Riederer, S. Parkinson plus syndrome: Diagnosis using high field MR imaging of brain iron. Radiology 1986, 159, 493–498. [Google Scholar] [CrossRef] [PubMed]
  129. Norfray, J.F.; Chiaradonna, N.L.; Heiser, W.J.; Song, S.H.; Manyam, B.V.; Devleschoward, A.B.; Eastwood, L.M. Brain iron in patients with Parkinson disease: MR visualization using gradient modification. Am. J. Neuroradiol. 1988, 9, 237–240. [Google Scholar]
  130. Brooks, D.J.; Luthert, P.; Gadian, D.; Marsden, C.D. Does signal-attenuation on high-field T2-weighted MRI of the brain reflect regional cerebral iron deposition? Observations on the relationship between regional cerebral water proton T2 values and iron levels. J. Neurol. Neurosurg. Psychiatry 1989, 52, 108–111. [Google Scholar] [CrossRef] [PubMed]
  131. Bartzokis, G.; Sultzer, D.; Mintz, J.; Holt, L.E.; Marx, P.; Phelan, C.K.; Marder, S.R. In vivo evaluation of brain iron in Alzheimer’s disease and normal subjects using MRI. Biol. Psychiatry 1994, 35, 480–487. [Google Scholar] [CrossRef] [PubMed]
  132. Giordano, N.; Fioravanti, A.; Sancasciani, S.; Marcolongo, R.; Borghi, C. Increased storage of iron and anaemia in rheumatoid arthritis: Usefulness of desferrioxamine. BMJ 1984, 289, 961–962. [Google Scholar] [CrossRef]
  133. Zhou, J.Y.; Wong, J.H.; Berman, Z.T.; Lombardi, A.F.; Chang, E.Y.; von Drygalski, A. Bleeding with iron deposition and vascular remodelling in subchondral cysts: A newly discovered feature unique to haemophilic arthropathy. Haemophilia 2021, 27, e730–e738. [Google Scholar] [CrossRef]
  134. Badat, M.; Kaya, B.; Telfer, P. Combination-therapy with concurrent deferoxamine and deferiprone is effective in treating resistant cardiac iron-loading in aceruloplasminaemia. Br. J. Haematol. 2015, 171, 430–432. [Google Scholar] [CrossRef]
  135. Gold, R.; Lenox, R.H. Is there a rationale for iron supplementation in the treatment of akathisia? A review of the evidence. J. Clin. Psychiatry 1995, 56, 476–483. [Google Scholar]
  136. Imon, Y.; Yamaguchi, S.; Katayama, S.; Harada, A.; Yamamura, Y.; Nakamura, S. The Effect of Age and Disease on the MR Imaging T2 Low Signal Intensity Area in the Cerebral Cortex. Nippon. Ronen Igakkai Zasshi. Jpn. J. Geriatr. 1994, 31, 697–704. [Google Scholar] [CrossRef]
  137. Lodi, R.; Hart, P.E.; Rajagopalan, B.; Taylor, D.J.; Crilley, J.G.; Bradley, J.L.; Blamire, A.M.; Manners, D.; Styles, P.; Schapira, A.H.; et al. Antioxidant treatment improves in vivo cardiac and skeletal muscle bioenergetics in patients with Friedreich’s ataxia. Ann. Neurol. 2001, 49, 590–596. [Google Scholar] [CrossRef]
  138. Koeppen, A.H.; Dickson, A.C. Iron in the Hallervorden-Spatz syndrome. Pediatr. Neurol. 2001, 25, 148–155. [Google Scholar] [CrossRef] [PubMed]
  139. Salomão, R.P.A.; Pedroso, J.L.; Gama, M.T.D.; Dutra, L.A.; Maciel, R.H.; Godeiro-Junior, C.; Chien, H.F.; Teive, H.A.G.; Cardoso, F.; Barsottini, O.G.P. A diagnostic approach for neurodegeneration with brain iron accumulation: Clinical features, genetics and brain imaging. Arq. Neuro-Psiquiatr. 2016, 74, 587–596. [Google Scholar] [CrossRef]
  140. Umeno, A.; Biju, V.; Yoshida, Y. In vivo ROS production and use of oxidative stress-derived biomarkers to detect the onset of diseases such as Alzheimer’s disease, Parkinson’s disease, and diabetes. Free Radic. Res. 2017, 51, 413–427. [Google Scholar] [CrossRef]
  141. Brar, S.; Henderson, D.; Schenck, J.; Zimmerman, E.A. Iron Accumulation in the Substantia Nigra of Patients With Alzheimer Disease and Parkinsonism. Arch. Neurol. 2009, 66, 371–374. [Google Scholar] [CrossRef]
  142. Wallis, L.I.; Paley, M.N.; Graham, J.M.; Grünewald, R.A.; Wignall, E.L.; Joy, H.M.; Griffiths, P.D. MRI assessment of basal ganglia iron deposition in Parkinson’s disease. J. Magn. Reson. Imaging 2008, 28, 1061–1067. [Google Scholar] [CrossRef]
  143. Waldvogel, D.; van Gelderen, P.; Hallett, M. Increased iron in the dentate nucleus of patients with Friedrich’s ataxia. Ann Neurol. 1999, 46, 123–125. [Google Scholar] [CrossRef]
  144. Iankova, V.; Karin, I.; Klopstock, T.; Schneider, S.A. Emerging Disease-Modifying Therapies in Neurodegeneration With Brain Iron Accumulation (NBIA) Disorders. Front. Neurol. 2021, 12, 629414. [Google Scholar] [CrossRef]
  145. Weiss, G.; Ganz, T.; Goodnough, L.T. Anemia of inflammation. Blood 2019, 133, 40–50. [Google Scholar] [CrossRef]
  146. Ganz, T. Anemia of Inflammation. N. Engl. J. Med. 2019, 381, 1148–1157. [Google Scholar] [CrossRef]
  147. Marques, O.; Weiss, G.; Muckenthaler, M.U. The role of iron in chronic inflammatory diseases: From mechanisms to treatment options in anemia of inflammation. Blood 2022, 140, 2011–2023. [Google Scholar] [CrossRef]
  148. Cacoub, P.; Choukroun, G.; Cohen-Solal, A.; Luporsi, E.; Peyrin-Biroulet, L.; Peoc’H, K.; Andrieu, V.; Lasocki, S.; Puy, H.; Trochu, J. Iron deficiency screening is a key issue in chronic inflammatory diseases: A call to action. J. Intern. Med. 2022, 292, 542–556. [Google Scholar] [CrossRef]
  149. Buliga-Finis, O.N.; Ouatu, A.; Tanase, D.M.; Gosav, E.M.; Seritean Isac, P.N.; Richter, P.; Rezus, C. Managing Anemia: Point of Convergence for Heart Failure and Chronic Kidney Disease? Life 2023, 13, 1311. [Google Scholar] [CrossRef]
  150. Canny, S.P.; Orozco, S.L.; Thulin, N.K.; Hamerman, J.A. Immune Mechanisms in Inflammatory Anemia. Annu. Rev. Immunol. 2023, 41, 405–429. [Google Scholar] [CrossRef]
  151. Pépin, M.; Klimkowicz-Mrowiec, A.; Godefroy, O.; Delgado, P.; Carriazo, S.; Ferreira, A.C.; Golenia, A.; Malyszko, J.; Grodzicki, T.; Giannakou, K.; et al. Cognitive disorders in patients with chronic kidney disease: Approaches to prevention and treatment. Eur. J. Neurol. 2023, 30, 2899–2911. [Google Scholar] [CrossRef]
  152. Toplicanin, A.; Toncev, L.; Zaric, V.M.; Milutinovic, A.S. Autoimmune Hemolytic Anemia in Inflammatory Bowel Disease—Report of a Case and Review of the Literature. Life 2022, 12, 1784. [Google Scholar] [CrossRef]
  153. Rashidi, A.; Garimella, P.S.; Al-Asaad, A.; Kharadjian, T.; Torres, M.N.; Thakkar, J. Anemia Management in the Cancer Patient With CKD and End-Stage Kidney Disease. Adv. Chronic Kidney Dis. 2022, 29, 180–187.e1. [Google Scholar] [CrossRef]
  154. Heras-Benito, M. Renal anemia: Current treatments and emerging molecules. Rev. Clin. Esp. 2023, 223, 433–439. [Google Scholar] [CrossRef]
  155. Ku, E.; Del Vecchio, L.; Eckardt, K.-U.; Haase, V.H.; Johansen, K.L.; Nangaku, M.; Tangri, N.; Waikar, S.S.; Więcek, A.; Cheung, M.; et al. Novel anemia therapies in chronic kidney disease: Conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference. Kidney Int. 2023; in press. [Google Scholar] [CrossRef]
  156. Afsar, B.; Afsar, R.E. Hypoxia-inducible factors and essential hypertension: Narrative review of experimental and clinical data. Pharmacol. Rep. 2023, 75, 861–875. [Google Scholar] [CrossRef]
  157. Kobayashi, M.; Suhara, T.; Baba, Y.; Kawasaki, N.K.; Higa, J.K.; Matsui, T. Pathological Roles of Iron in Cardiovascular Disease. Curr. Drug Targets 2018, 19, 1068–1076. [Google Scholar] [CrossRef]
  158. Murawska, N.; Fabisiak, A.; Fichna, J. Anemia of Chronic Disease and Iron Deficiency Anemia in Inflammatory Bowel Diseases:Pathophysiology, Diagnosis, and Treatment. Inflamm. Bowel Dis. 2016, 22, 1198–1208. [Google Scholar] [CrossRef] [PubMed]
  159. Nataatmadja, M.S.; Francis, R. Recurrent severe hypophosphatemia following intravenous iron administration. Clin. Case Rep. 2020, 8, 243–246. [Google Scholar] [CrossRef]
  160. Kshirsagar, A.V.; Li, X. Long-Term Risks of Intravenous Iron in End-Stage Renal Disease Patients. Adv. Chronic Kidney Dis. 2019, 26, 292–297. [Google Scholar] [CrossRef] [PubMed]
  161. Basak, T.; Kanwar, R.K. Iron imbalance in cancer: Intersection of deficiency and overload. Cancer Med. 2022, 11, 3837–3853. [Google Scholar] [CrossRef]
  162. Vassiliou, E.; Farias-Pereira, R. Impact of Lipid Metabolism on Macrophage Polarization: Implications for Inflammation and Tumor Immunity. Int. J. Mol. Sci. 2023, 24, 12032. [Google Scholar] [CrossRef]
  163. Zhou, Z.; Xu, B.; Hu, N.; Guo, Z.; Bao, W.; Shao, B.; Yang, W. Targeting the Macrophage-Ferroptosis Crosstalk: A Novel Insight into Tumor Immunotherapy. Front. Biosci. 2022, 27, 203. [Google Scholar] [CrossRef] [PubMed]
  164. Zhou, Q.; Fang, T.; Wei, S.; Chai, S.; Yang, H.; Tao, M.; Cao, Y. Macrophages in melanoma: A double-edged sword and targeted therapy strategies (Review). Exp. Ther. Med. 2022, 24, 640. [Google Scholar] [CrossRef]
  165. Furgiuele, S.; Descamps, G.; Cascarano, L.; Boucq, A.; Dubois, C.; Journe, F.; Saussez, S. Dealing with Macrophage Plasticity to Address Therapeutic Challenges in Head and Neck Cancers. Int. J. Mol. Sci. 2022, 23, 6385. [Google Scholar] [CrossRef] [PubMed]
  166. Hetzel, M.; Ackermann, M.; Lachmann, N. Beyond “Big Eaters”: The Versatile Role of Alveolar Macrophages in Health and Disease. Int. J. Mol. Sci. 2021, 22, 3308. [Google Scholar] [CrossRef]
  167. Li, J.; Jia, Y.C.; Ding, Y.X.; Bai, J.; Cao, F.; Li, F. The crosstalk between ferroptosis and mitochondrial dynamic regulatory networks. Int. J. Biol. Sci. 2023, 19, 2756–2771. [Google Scholar] [CrossRef]
  168. Lee, S.; Hwang, N.; Seok, B.G.; Lee, S.; Lee, S.J.; Chung, S.W. Autophagy mediates an amplification loop during ferroptosis. Cell Death Dis. 2023, 14, 464. [Google Scholar] [CrossRef] [PubMed]
  169. Chen, F.; Kang, R.; Tang, D.; Liu, J. Monitoring Lysosome Function in Ferroptosis. Methods Mol. Biol. 2023, 2712, 91–102. [Google Scholar] [CrossRef] [PubMed]
  170. Denz, H.; Huber, P.; Landmann, R.; Orth, B.; Wachter, H.; Fuchs, D. Association between the activation of macrophages, changes of iron metabolism and the degree of anaemia in patients with malignant disorders. Eur. J. Haematol. 1992, 48, 244–248. [Google Scholar] [CrossRef]
  171. Cohen, L.A.; Gutierrez, L.; Weiss, A.; Leichtmann-Bardoogo, Y.; Zhang, D.L.; Crooks, D.R.; Sougrat, R.; Morgenstern, A.; Galy, B.; Hentze, M.W.; et al. Serum ferritin is derived primarily from macrophages through a non-classical secretory pathway. Blood 2010, 116, 1574–1584. [Google Scholar] [CrossRef]
  172. Wang, W.; Knovich, M.A.; Coffman, L.G.; Torti, F.M.; Torti, S.V. Serum ferritin: Past, present and future. Biochim. Biophys Acta 2010, 1800, 760–769. [Google Scholar] [CrossRef]
  173. Plays, M.; Müller, S.; Rodriguez, R. Chemistry and biology of ferritin. Metallomics 2021, 3, mfab021. [Google Scholar] [CrossRef]
  174. Yuan, X.M.; Li, W.; Baird, S.K.; Carlsson, M.; Melefors, Ö. Secretion of ferritin by iron-laden macrophages and influence of lipoproteins. Free Radic. Res. 2004, 38, 1133–1142. [Google Scholar] [CrossRef]
  175. Leftin, A.; Zhao, H.; Turkekul, M.; de Stanchina, E.; Manova, K.; Koutcher, J.A. Iron deposition is associated with differential macrophage infiltration and therapeutic response to iron chelation in prostate cancer. Sci. Rep. 2017, 7, 11632. [Google Scholar] [CrossRef]
  176. Yan, S.; Dong, X.; Ding, D.; Xue, J.; Wang, X.; Huang, Y.; Pan, Z.; Sun, H.; Ren, Q.; Dou, W.; et al. Iron deposition in ovarian endometriosis evaluated by magnetic resonance imaging R2* correlates with ovarian function. Reprod. Biomed. Online, 2023; in press. [Google Scholar] [CrossRef]
  177. Lattuada, D.; Uberti, F.; Colciaghi, B.; Morsanuto, V.; Maldi, E.; Squarzanti, D.F.; Molinari, C.; Boldorini, R.; Bulfoni, A.; Colombo, P.; et al. Fimbrial Cells Exposure to Catalytic Iron Mimics Carcinogenic Changes. Int. J. Gynecol. Cancer 2015, 25, 389–398. [Google Scholar] [CrossRef]
  178. Pontillo, G.; Petracca, M.; Monti, S.; Quarantelli, M.; Lanzillo, R.; Costabile, T.; Carotenuto, A.; Tortora, F.; Elefante, A.; Morra, V.B.; et al. Clinical correlates of R1 relaxometry and magnetic susceptibility changes in multiple sclerosis: A multi-parameter quantitative MRI study of brain iron and myelin. Eur Radiol. 2023, 33, 2185–2194. [Google Scholar] [CrossRef] [PubMed]
  179. Zachariou, V.; Bauer, C.E.; Pappas, C.; Gold, B.T. High cortical iron is associated with the disruption of white matter tracts supporting cognitive function in healthy older adults. Cereb. Cortex 2023, 33, 4815–4828. [Google Scholar] [CrossRef] [PubMed]
  180. Wang, F.; Zhang, M.; Li, Y.; Li, Y.; Gong, H.; Li, J.; Zhang, Y.; Zhang, C.; Yan, F.; Sun, B.; et al. Alterations in brain iron deposition with progression of late-life depression measured by magnetic resonance imaging (MRI)-based quantitative susceptibility mapping. Quant. Imaging Med. Surg. 2022, 12, 3873–3888. [Google Scholar] [CrossRef]
  181. Pecorelli, A.; Franceschi, P.; Braccischi, L.; Izzo, F.; Renzulli, M.; Golfieri, R. MRI Appearance of Focal Lesions in Liver Iron Overload. Diagnostics 2022, 12, 891. [Google Scholar] [CrossRef]
  182. Blanusa, M.; Varnai, V.M.; Piasek, M.; Kostial, K. Chelators as Antidotes of Metal Toxicity: Therapeutic and Experimental Aspects. Curr. Med. Chem. 2005, 12, 2771–2794. [Google Scholar] [CrossRef]
  183. Perron, N.R.; Brumaghim, J.L. A Review of the Antioxidant Mechanisms of Polyphenol Compounds Related to Iron Binding. Cell Biochem. Biophys. 2009, 53, 75–100. [Google Scholar] [CrossRef]
  184. Kontoghiorghe, C.N.; Kolnagou, A.; Kontoghiorghes, G.J. Phytochelators Intended for Clinical Use in Iron Overload, Other Diseases of Iron Imbalance and Free Radical Pathology. Molecules 2015, 20, 20841–20872. [Google Scholar] [CrossRef] [PubMed]
  185. Kontoghiorghes, G.J. Advances on Chelation and Chelator Metal Complexes in Medicine. Int. J. Mol. Sci. 2020, 21, 2499. [Google Scholar] [CrossRef]
  186. Gulani, V.; Calamante, F.; Shellock, F.G.; Kanal, E.; Reeder, S.B. Gadolinium deposition in the brain: Summary of evidence and recommendations. Lancet Neurol. 2017, 16, 564–570. [Google Scholar] [CrossRef] [PubMed]
  187. García-Rodríguez, M.D.C.; Hernández-Cortés, L.M.; Mendoza-Núñez, V.M.; Arenas-Huertero, F. Effects of green tea polyphenols against metal-induced genotoxic damage: Underlying mechanistic pathways. J. Toxicol. Environ. Health Part B 2023, 1–16, in press. [Google Scholar] [CrossRef]
  188. Teranishi, K. Near-infrared chemiluminescence imaging of superoxide anion production in kidneys with iron3+-nitrilotriacetate-induced acute renal oxidative stress in rats. J. Photochem. Photobiol. B Biol. 2022, 228, 112391. [Google Scholar] [CrossRef]
  189. Yamamoto, R.; Williams, G.; Frankel, H.; Weisburger, J. 8-Hydroxyquinoline: Chronic toxicity and inhibitory effect on the carcinogenicity of N-2-fluorenylacetamide. Toxicol. Appl. Pharmacol. 1971, 19, 687–698. [Google Scholar] [CrossRef] [PubMed]
  190. Konorev, D.; Yao, L.; Turesky, R.J. Multi-DNA Adduct and Abasic Site Quantitation In Vivo by Nano-Liquid Chromatography/High-Resolution Orbitrap Tandem Mass Spectrometry: Methodology for Biomonitoring Colorectal DNA Damage. Chem. Res. Toxicol. 2022, 35, 1519–1532. [Google Scholar] [CrossRef]
  191. Zhu, B.Z.; Xu, D.; Qin, L.; Huang, C.H.; Xie, L.N.; Mao, L.; Shao, J.; Kalyanaraman, B. An unexpected new pathway for nitroxide radical production via more reactve nitrogen-centered amidyl radical intermediate during detoxification of the carcinogenic halogenated quinones by N-alkyl hydroxamic acids. Free Radic. Biol. Med. 2020, 146, 150–159. [Google Scholar] [CrossRef] [PubMed]
  192. Kontoghiorghes, G.J. New Iron Metabolic Pathways and Chelation Targeting Strategies Affecting the Treatment of All Types and Stages of Cancer. Int. J. Mol. Sci. 2022, 23, 13990. [Google Scholar] [CrossRef]
  193. Williams, G.M.; Yamamoto, R.S. Absence of Stainable Iron From Preneoplastic and Neoplastic Lesions in Rat Liver With 8-Hydroxyquinoline-Induced Siderosis. Gynecol. Oncol. 1972, 49, 685–692. [Google Scholar] [CrossRef]
  194. Li, G.; Yang, J.; Zhao, G.; Shen, Z.; Yang, K.; Tian, L.; Zhou, Q.; Chen, Y.; Huang, Y. Dysregulation of ferroptosis may involve in the development of non-small-cell lung cancer in Xuanwei area. J. Cell Mol. Med. 2021, 25, 2872–2884, Erratum in: J. Cell Mol. Med. 2022, 26, 240. [Google Scholar] [CrossRef]
  195. Pease, C.; Rücker, T.; Birk, T. Review of the Evidence from Epidemiology, Toxicology, and Lung Bioavailability on the Carcinogenicity of Inhaled Iron Oxide Particulates. Chem. Res. Toxicol. 2016, 29, 237–254. [Google Scholar] [CrossRef]
  196. Lovera-Leroux, M.; Crobeddu, B.; Kassis, N.; Petit, P.X.; Janel, N.; Baeza-Squiban, A.; Andreau, K. The iron component of particulate matter is antiapoptotic: A clue to the development of lung cancer after exposure to atmospheric pollutants? Biochimie 2015, 118, 195–206. [Google Scholar] [CrossRef] [PubMed]
  197. Xie, G.; Zhou, L.; Gao, W.; Li, Y. Organic additives enhance Fenton treatment of nitrobenzene at near-neutral pH. Environ. Sci. Pollut. Res. 2015, 22, 7082–7092. [Google Scholar] [CrossRef]
  198. Bouvard, V.; Loomis, D.; Guyton, K.Z.; Grosse, Y.; El Ghissassi, F.; Benbrahim-Tallaa, L.; Guha, N.; Mattock, H.; Straif, K. Carcinogenicity of consumption of red and processed meat. Lancet Oncol. 2015, 16, 1599–1600. [Google Scholar] [CrossRef]
  199. Seiwert, N.; Wecklein, S.; Demuth, P.; Hasselwander, S.; Kemper, T.A.; Schwerdtle, T.; Brunner, T.; Fahrer, J. Heme oxygenase 1 protects human colonocytes against ROS formation, oxidative DNA damage and cytotoxicity induced by heme iron, but not inorganic iron. Cell Death Dis. 2020, 11, 787. [Google Scholar] [CrossRef]
  200. Rizzolo-Brime, L.; Orta-Ramirez, A.; Martin, Y.P.; Jakszyn, P. Nutritional Assessment of Plant-Based Meat Alternatives: A Comparison of Nutritional Information of Plant-Based Meat Alternatives in Spanish Supermarkets. Nutrients 2023, 15, 1325. [Google Scholar] [CrossRef]
  201. Niedermaier, T.; Gredner, T.; Hoffmeister, M.; Mons, U.; Brenner, H. Impact of Reducing Intake of Red and Processed Meat on Colorectal Cancer Incidence in Germany 2020 to 2050—A Simulation Study. Nutrients 2023, 15, 1020. [Google Scholar] [CrossRef] [PubMed]
  202. Martin, O.C.B.; Olier, M.; Ellero-Simatos, S.; Naud, N.; Dupuy, J.; Huc, L.; Taché, S.; Graillot, V.; Levêque, M.; Bézirard, V.; et al. Haem iron reshapes colonic luminal environment: Impact on mucosal homeostasis and microbiome through aldehyde formation. Microbiome 2019, 7, 72. [Google Scholar] [CrossRef]
  203. Sasso, A.; Latella, G. Role of Heme Iron in the Association Between Red Meat Consumption and Colorectal Cancer. Nutr. Cancer 2018, 70, 1173–1183. [Google Scholar] [CrossRef] [PubMed]
  204. Chiang, V.S.-C.; Quek, S.-Y. The relationship of red meat with cancer: Effects of thermal processing and related physiological mechanisms. Crit. Rev. Food Sci. Nutr. 2017, 57, 1153–1173. [Google Scholar] [CrossRef]
  205. Bastide, N.M.; Pierre, F.H.; Corpet, D.E. Heme Iron from Meat and Risk of Colorectal Cancer: A Meta-analysis and a Review of the Mechanisms Involved. Cancer Prev. Res. 2011, 4, 177–184. [Google Scholar] [CrossRef] [PubMed]
  206. Cross, A.J.; Freedman, N.D.; Ren, J.; Ward, M.H.; Hollenbeck, A.R.; Schatzkin, A.; Sinha, R.; Abnet, C.C. Meat Consumption and Risk of Esophageal and Gastric Cancer in a Large Prospective Study. Am. J. Gastroenterol. 2011, 106, 432–442. [Google Scholar] [CrossRef] [PubMed]
  207. Okazaki, Y. Asbestos-induced mesothelial injury and carcinogenesis: Involvement of iron and reactive oxygen species. Pathol. Int. 2022, 72, 83–95. [Google Scholar] [CrossRef]
  208. Ito, F.; Yanatori, I.; Maeda, Y.; Nimura, K.; Ito, S.; Hirayama, T.; Nagasawa, H.; Kohyama, N.; Okazaki, Y.; Akatsuka, S.; et al. Asbestos conceives Fe(II)-dependent mutagenic stromal milieu through ceaseless macrophage ferroptosis and β-catenin induction in mesothelium. Redox Biol. 2020, 36, 101616. [Google Scholar] [CrossRef] [PubMed]
  209. Chew, S.H.; Toyokuni, S. Malignant mesothelioma as an oxidative stress-induced cancer: An update. Free. Radic. Biol. Med. 2015, 86, 166–178. [Google Scholar] [CrossRef] [PubMed]
  210. E Duncan, K.; Cook, P.M.; Gavett, S.H.; A Dailey, L.; Mahoney, R.K.; Ghio, A.J.; Roggli, V.L.; Devlin, R.B. In vitro determinants of asbestos fiber toxicity: Effect on the relative toxicity of Libby amphibole in primary human airway epithelial cells. Part. Fibre Toxicol. 2014, 11, 2. [Google Scholar] [CrossRef] [PubMed]
  211. Urso, L.; Cavallari, I.; Sharova, E.; Ciccarese, F.; Pasello, G.; Ciminale, V. Metabolic rewiring and redox alterations in malignant pleural mesothelioma. Br. J. Cancer 2020, 122, 52–61. [Google Scholar] [CrossRef]
  212. Ou, Z.; Wen, Q.; Deng, Y.; Yu, Y.; Chen, Z.; Sun, L. Cigarette smoking is associated with high level of ferroptosis in seminal plasma and affects semen quality. Reprod. Biol. Endocrinol. 2020, 18, 55. [Google Scholar] [CrossRef]
  213. Zhang, W.Z.; Butler, J.J.; Cloonan, S.M. Smoking-induced iron dysregulation in the lung. Free. Radic. Biol. Med. 2019, 133, 238–247. [Google Scholar] [CrossRef]
  214. Takahashi, M.; Mizumura, K.; Gon, Y.; Shimizu, T.; Kozu, Y.; Shikano, S.; Iida, Y.; Hikichi, M.; Okamoto, S.; Tsuya, K.; et al. Iron-Dependent Mitochondrial Dysfunction Contributes to the Pathogenesis of Pulmonary Fibrosis. Front. Pharmacol. 2022, 12, 643980. [Google Scholar] [CrossRef]
  215. Fibach, E.; Rachmilewitz, E.A. Iron overload in hematological disorders. Presse Med. 2017, 46 Pt 2, e296–e305. [Google Scholar] [CrossRef] [PubMed]
  216. Leitch, H.A.; Fibach, E.; Rachmilewitz, E. Toxicity of iron overload and iron overload reduction in the setting of hematopoietic stem cell transplantation for hematologic malignancies. Crit. Rev. Oncol. 2017, 113, 156–170. [Google Scholar] [CrossRef] [PubMed]
  217. Harrison, P.; Marwah, S.S.; Hughes, R.T.; Bareford, D. Non-transferrin bound iron and neutropenia after cytotoxic chemotherapy. J. Clin. Pathol. 1994, 47, 350–352. [Google Scholar] [CrossRef]
  218. Motoori, S.; Hosokawa, Y.; Miyazawa, T.; Iwamoto, N.; Kase, Y.; Kitagawa, N.; Saisho, H.; Furuse, J. Two cases of colorectal cancer presenting with periodic, transient elevation of serum iron after chemotherapy including irinotecan. Gan Kagaku Ryoho Cancer Chemother 2010, 37, 2185–2188. [Google Scholar]
  219. Łęcka, M.; Słomka, A.; Albrecht, K.; Żekanowska, E.; Romiszewski, M.; Styczyński, J. Unbalance in Iron Metabolism in Childhood Leukemia Converges with Treatment Intensity: Biochemical and Clinical Analysis. Cancers 2021, 13, 3029. [Google Scholar] [CrossRef] [PubMed]
  220. Belotti, A.; Duca, L.; Borin, L.; Realini, S.; Renso, R.; Parma, M.; Pioltelli, P.; Pogliani, E.; Cappellini, M.D. Non transferrin bound iron (NTBI) in acute leukemias throughout conventional intensive chemotherapy: Kinetics of its appearance and potential predictive role in infectious complications. Leuk. Res. 2015, 39, 88–91. [Google Scholar] [CrossRef]
  221. Sahlstedt, L.; von Bonsdorff, L.; Ebeling, F.; Parkkinen, J.; Juvonen, E.; Ruutu, T. Non-transferrin-bound iron in haematological patients during chemotherapy and conditioning for autologous stem cell transplantation. Eur. J. Haematol. 2009, 83, 455–459. [Google Scholar] [CrossRef]
  222. von Bonsdorff, L.; Lindeberg, E.; Sahlstedt, L.; Lehto, J.; Parkkinen, J. Bleomycin-detectable Iron Assay for Non-Transferrin-bound Iron in Hematologic Malignancies. Clin. Chem. 2002, 48, 307–314. [Google Scholar] [CrossRef]
  223. Dürken, M.; Herrnring, C.; Finckh, B.; Nagel, S.; Nielsen, P.; Fischer, R.; Berger, H.M.; Moison, R.M.; Pichlmeier, U.; Kohlschütter, B.; et al. Impaired plasma antioxidative defense and increased nontransferrin-bound iron during high-dose chemotherapy and radiochemotherapy preceding bone marrow transplantation. Free Radic. Biol. Med. 2000, 28, 887–894. [Google Scholar] [CrossRef]
  224. Sahlstedt, L.; Ebeling, F.; Von Bonsdorff, L.; Parkkinen, J.; Ruutu, T. Non-transferrin-bound iron during allogeneic stem cell transplantation. Br. J. Haematol. 2001, 113, 836–838. [Google Scholar] [CrossRef]
  225. Miranda, J.L.; Mesquita, R.B.; Leite, A.; Silva, A.M.; Rangel, M.; Rangel, A.O. Non-transferrin-bound iron determination in blood serum using microsequential injection solid phase spectrometry—Proof of concept. Talanta 2023, 257, 124345. [Google Scholar] [CrossRef]
  226. Jóźwik, M.; Jóźwik, M.; Jóźwik, M.; Szczypka, M.; Gajewska, J.; Laskowska-Klita, T. Antioxidant defence of red blood cells and plasma in stored human blood. Clin. Chim. Acta 1997, 267, 129–142. [Google Scholar] [CrossRef]
  227. Ferrara, F.; Coppi, F.; Riva, R.; Ventura, P.; Ricci, A.; Mattioli, A.V.; Talarico, M.; Garuti, C.; Bevini, M.; Rochira, V.; et al. Labile plasma iron and echocardiographic parameters are associated with cardiac events in β-thalassemic patients. Eur. J. Clin. Investig. 2023, 53, e13954. [Google Scholar] [CrossRef] [PubMed]
  228. Kontoghiorghes, G.J. Iron Mobilization From Transferrin And Non-Transferrin-Bound-Iron by Deferiprone. Implications in the Treatment of Thalassemia, Anemia of Chronic Disease, Cancer and Other Conditions. Hemoglobin 2006, 30, 183–200. [Google Scholar] [CrossRef] [PubMed]
  229. Kontoghiorghes, G.J.; Weinberg, E.D. Iron: Mammalian defense systems, mechanisms of disease, and chelation therapy approaches. Blood Rev. 1995, 9, 33–45. [Google Scholar] [CrossRef] [PubMed]
  230. Shah, S.V.; Rajapurkar, M.M.; Baliga, R. The Role of Catalytic Iron in Acute Kidney Injury. Clin. J. Am. Soc. Nephrol. 2011, 6, 2329–2331. [Google Scholar] [CrossRef]
  231. Lele, S.S.; Mukhopadhyay, B.N.; Mardikar, M.M.; Patel, T.A.; Vasavada, A.K.; Banker, D.N.; Kapasi, K.D.; Chauhan, V.C.; Chawla, K.C.; Raju, S.R.; et al. Impact of catalytic iron on mortality in patients with acute coronary syndrome exposed to iodinated radiocontrast—The Iscom Study. Am. Heart J. 2013, 165, 744–751. [Google Scholar] [CrossRef]
  232. Rajapurkar, M.M.; Shah, S.V.; Lele, S.S.; Hegde, U.N.; Lensing, S.Y.; Gohel, K.; Mukhopadhyay, B.; Gang, S.; Eigenbrodt, M.L. Association of Catalytic Iron With Cardiovascular Disease. Am. J. Cardiol. 2012, 109, 438–442. [Google Scholar] [CrossRef]
  233. Wang, C.; Zhao, M.; Nie, Y.; Yang, Y.; Tan, Y.; Du, Z.; Gao, S.; Li, W. Impact of iron overload on poor graft function after allo-HSCT in a patient with transfusion-dependent low-risk MDS: A case report and literature review. Medicine 2022, 101, e32012. [Google Scholar] [CrossRef]
  234. Brissot, P.; Bernard, D.G.; Brissot, E.; Loréal, O.; Troadec, M.-B. Rare anemias due to genetic iron metabolism defects. Mutat. Res. Mol. Mech. Mutagen. 2018, 777, 52–63. [Google Scholar] [CrossRef]
  235. Baravelli, C.M.; Aarsand, A.K.; Sandberg, S.; Tollånes, M.C. Porphyria cutanea tarda and patterns of long-term sick leave and disability pension: A 24-year nationwide matched-cohort study. Orphanet. J. Rare Dis. 2022, 17, 72. [Google Scholar] [CrossRef]
  236. Adams, P.C.; Horgan-Bell, C.; Walsh, S.; Sadikovic, B. Porphyria cutanea tarda associated with elevated serum ferritin, iron overload, and a bone morphogenetic protein 6 genetic variant. Can. Liver J. 2020, 3, 232–234. [Google Scholar] [CrossRef] [PubMed]
  237. Grace, R.F.; van Beers, E.J.; Corrons, J.-L.V.; Glader, B.; Glenthøj, A.; Kanno, H.; Kuo, K.H.M.; Lander, C.; Layton, D.M.; Pospíŝilová, D.; et al. The Pyruvate Kinase Deficiency Global Longitudinal (Peak) Registry: Rationale and study design. BMJ Open 2023, 13, e063605. [Google Scholar] [CrossRef] [PubMed]
  238. Julián-Serrano, S.; Yuan, F.; Barrett, M.J.; Pfeiffer, R.M.; Stolzenberg-Solomon, R.Z. Hemochromatosis, Iron Overload–Related Diseases, and Pancreatic Cancer Risk in the Surveillance, Epidemiology, and End Results (SEER)-Medicare. Cancer Epidemiol. Biomark. Prev. 2021, 30, 2136–2139. [Google Scholar] [CrossRef] [PubMed]
  239. Asleh, M.; Levitas, A.; Daniel, S.; Abu-Quider, A.; Ben-Harosh, M.; Kapelushnik, J. Hepatic and cardiac iron load as determined by MRI T2* in patients with congenital dyserythropoietic anemia type I. Ann. Hematol. 2020, 99, 2507–2512. [Google Scholar] [CrossRef]
  240. Beutler, E.; Felitti, V.; Gelbart, T.; Ho, N. Genetics of iron storage and hemochromatosis. Drug Metab. Dispos. 2001, 29 Pt 2, 495–499. [Google Scholar]
  241. Duan, G.; Li, J.; Duan, Y.; Zheng, C.; Guo, Q.; Li, F.; Zheng, J.; Yu, J.; Zhang, P.; Wan, M.; et al. Mitochondrial Iron Metabolism: The Crucial Actors in Diseases. Molecules 2022, 28, 29. [Google Scholar] [CrossRef] [PubMed]
  242. Pagano, G.; Talamanca, A.A.; Castello, G.; Cordero, M.D.; d’Ischia, M.; Gadaleta, M.N.; Pallardó, F.V.; Petrović, S.; Tiano, L.; Zatterale, A. Oxidative stress and mitochondrial dysfunction across broad-ranging pathologies: Toward mitochondria-targeted clinical strategies. Oxid. Med. Cell. Longev. 2014, 2014, 541230. [Google Scholar] [CrossRef]
  243. Uda, K.; Yashiro, M. Idiopathic Pulmonary Hemosiderosis. N. Engl. J. Med. 2023, 388, 2179. [Google Scholar] [CrossRef]
  244. Guo, Q.; Qian, C.; Qian, Z.-M. Iron metabolism and atherosclerosis. Trends Endocrinol. Metab. 2023, 34, 404–413. [Google Scholar] [CrossRef]
  245. Dempke, W.C.M.; Zielinski, R.; Winkler, C.; Silberman, S.; Reuther, S.; Priebe, W. Anthracycline-induced cardiotoxicity—Are we about to clear this hurdle? Eur. J. Cancer 2023, 185, 94–104. [Google Scholar] [CrossRef]
  246. Jones, I.C.; Dass, C.R. Doxorubicin-induced cardiotoxicity: Causative factors and possible interventions. J. Pharm. Pharmacol. 2022, 74, 1677–1688. [Google Scholar] [CrossRef]
  247. Sheibani, M.; Azizi, Y.; Shayan, M.; Nezamoleslami, S.; Eslami, F.; Farjoo, M.H.; Dehpour, A.R. Doxorubicin-Induced Cardiotoxicity: An Overview on Pre-clinical Therapeutic Approaches. Cardiovasc. Toxicol. 2022, 22, 292–310. [Google Scholar] [CrossRef]
  248. Menon, A.V.; Kim, J. Iron Promotes Cardiac Doxorubicin Retention and Toxicity Through Downregulation of the Mitochondrial Exporter ABCB8. Front. Pharmacol. 2022, 13, 817951. [Google Scholar] [CrossRef] [PubMed]
  249. Mody, H.; Vaidya, T.R.; Ait-Oudhia, S. In vitro to clinical translational pharmacokinetic/pharmacodynamic modeling of doxorubicin (DOX) and dexrazoxane (DEX) interactions: Safety assessment and optimization. Sci. Rep. 2023, 13, 3100. [Google Scholar] [CrossRef]
  250. Rahimi, P.; Barootkoob, B.; ElHashash, A.; Nair, A. Efficacy of Dexrazoxane in Cardiac Protection in Pediatric Patients Treated With Anthracyclines. Cureus 2023, 15, e37308. [Google Scholar] [CrossRef] [PubMed]
  251. de Baat, E.C.; van Dalen, E.C.; Mulder, R.L.; Hudson, M.M.; Ehrhardt, M.J.; Engels, F.K.; Feijen, E.A.M.; Grotenhuis, H.B.; Leerink, J.M.; Kapusta, L.; et al. Primary cardioprotection with dexrazoxane in patients with childhood cancer who are expected to receive anthracyclines: Recommendations from the International Late Effects of Childhood Cancer Guideline Harmonization Group. Lancet Child Adolesc. Health 2022, 6, 885–894. [Google Scholar] [CrossRef] [PubMed]
  252. Johnstone, T.C.; Suntharalingam, K.; Lippard, S.J. The next generation of platinum drugs: Targeted Pt (II) agents, nanoparticle delivery, and Pt(IV) Prodrugs. Chem. Rev. 2016, 116, 3436–3486. [Google Scholar] [CrossRef]
  253. Peng, H.; Jin, H.; Zhuo, H.; Huang, H. Enhanced antitumor efficacy of cisplatin for treating ovarian cancer in vitro and in vivo via transferrin binding. Oncotarget 2017, 8, 45597–45611. [Google Scholar] [CrossRef]
  254. Aggarwal, A.; Dinda, A.K.; Mukhopadhyay, C.K. Effect of Cisplatin on Renal Iron Homeostasis Components: Implication in Nephropathy. ACS Omega 2022, 7, 27804–27817. [Google Scholar] [CrossRef]
  255. Wang, Y.; Lv, J.; Ma, X.; Wang, D.; Ma, H.; Chang, Y.; Nie, G.; Jia, L.; Duan, X.; Liang, X.-J. Specific Hemosiderin Deposition in Spleen Induced by a Low Dose of Cisplatin: Altered Iron Metabolism and Its Implication as an Acute Hemosiderin Formation Model. Curr. Drug Metab. 2010, 11, 507–515. [Google Scholar] [CrossRef] [PubMed]
  256. Makhdoumi, P.; Abnous, K.; Mehri, S.; Etemad, L.; Imenshahidi, M.; Karimi, G. Oral deferiprone administration ameliorates cisplatin-induced nephrotoxicity in rats. J. Pharm. Pharmacol. 2018, 70, 1357–1368. [Google Scholar] [CrossRef] [PubMed]
  257. Kontoghiorghes, G.J.; Jackson, M.J.; Lunec, J. In Vitro Screening of Iron Chelators Using Models of Free Radical Damage. Free. Radic. Res. Commun. 1986, 2, 115–124. [Google Scholar] [CrossRef]
  258. Crisponi, G.; Nurchi, V.M.; Lachowicz, J.I.; Crespo-Alonso, M.; Zoroddu, M.A.; Peana, M. Kill or cure: Misuse of chelation therapy for human diseases. Coord. Chem. Rev. 2015, 284, 278–285. [Google Scholar] [CrossRef]
  259. Choi, J.J.; McCarthy, M.W. Cefiderocol: A novel siderophore cephalosporin. Expert Opin. Investig. Drugs 2018, 27, 193–197. [Google Scholar] [CrossRef]
  260. Robins-Browne, R.; Prpic, J. Desferrioxamine and systemic yersiniosis. Lancet 1983, 322, 1372. [Google Scholar] [CrossRef]
  261. Boelaert, J.R.; Fenves, A.Z.; Coburn, J.W. Deferoxamine Therapy and Mucormycosis in Dialysis Patients: Report of an International Registry. Am. J. Kidney Dis. 1991, 18, 660–667. [Google Scholar] [CrossRef]
  262. Brock, J.H.; Licéaga, J.; Kontoghiorghes, G.J. The effect of synthetic iron chelators on bacterial growth in human serum. FEMS Microbiol. Immunol. 1988, 1, 55–60. [Google Scholar] [CrossRef]
  263. Djaldetti, M.; Fishman, P.; Notti, I.; Bessler, H. The effect of tetracycline administration on iron absorption in mice. Biomedicine 1981, 35, 150–152. [Google Scholar]
  264. Madaan, K.; Kaushik, D.; Verma, T. Hydroxyurea: A key player in cancer chemotherapy. Expert Rev. Anticancer. Ther. 2012, 2, 19–29. [Google Scholar] [CrossRef] [PubMed]
  265. Konstantinou, E.; Pashalidis, I.; Kolnagou, A.; Kontoghiorghes, G.J. Interactions Of Hydroxycarbamide (Hydroxyurea) With Iron And Copper: Implications On Toxicity and Therapeutic Strategies. Hemoglobin 2011, 35, 237–246. [Google Scholar] [CrossRef]
  266. Kontoghiorghes, G.J.; Kolnagou, A.; Kontoghiorghe, C.N.; Mourouzidis, L.; Timoshnikov, V.A.; Polyakov, N.E. Trying to Solve the Puzzle of the Interaction of Ascorbic Acid and Iron: Redox, Chelation and Therapeutic Implications. Medicines 2020, 7, 45. [Google Scholar] [CrossRef] [PubMed]
  267. Ohno, S.; Ohno, Y.; Suzuki, N.; Soma, G.-I.; Inoue, M. High-dose vitamin C (ascorbic acid) therapy in the treatment of patients with advanced cancer. Anticancer. Res. 2009, 29, 809–815. [Google Scholar] [PubMed]
  268. Nielsen, T.K.; Højgaard, M.; Andersen, J.T.; Poulsen, H.E.; Lykkesfeldt, J.; Mikines, K.J. Elimination of Ascorbic Acid After High-Dose Infusion in Prostate Cancer Patients: A Pharmacokinetic Evaluation. Basic Clin. Pharmacol. Toxicol. 2015, 116, 343–348. [Google Scholar] [CrossRef] [PubMed]
  269. Levine, M.; Conry-Cantilena, C.; Wang, Y.; Welch, R.W.; Washko, P.W.; Dhariwal, K.R.; Park, J.B.; Lazarev, A.; Graumlich, J.F.; King, J.; et al. Vitamin C pharmacokinetics in healthy volunteers: Evidence for a recommended dietary allowance. Proc. Natl. Acad. Sci. USA 1996, 93, 3704–3709. [Google Scholar] [CrossRef]
  270. Tarng, D.; Huang, T.; Wei, Y. Erythropoietin and iron: The role of ascorbic acid. Nephrol. Dial. Transplant. 2001, 16, 35–39. [Google Scholar] [CrossRef]
  271. Sultana, T.; DeVita, M.V.; Michelis, M.F. Oral vitamin C supplementation reduces erythropoietin requirement in hemodialysis patients with functional iron deficiency. Int. Urol. Nephrol. 2016, 48, 1519–1524. [Google Scholar] [CrossRef]
  272. Sourabh, S.; Bhatia, P.; Jain, R. Favourable improvement in haematological parameters in response to oral iron and vitamin C combination in children with Iron Refractory Iron Deficiency Anemia (IRIDA) phenotype. Blood Cells Mol. Dis. 2019, 75, 26–29. [Google Scholar] [CrossRef]
  273. Scheers, N.; Sandberg, A.-S. Iron Transport through Ferroportin Is Induced by Intracellular Ascorbate and Involves IRP2 and HIF2α. Nutrients 2014, 6, 249–260. [Google Scholar] [CrossRef]
  274. Timoshnikov, V.A.; Selyutina, O.Y.; Polyakov, N.E.; Didichenko, V.; Kontoghiorghes, G.J. Mechanistic Insights of Chelator Complexes with Essential Transition Metals: Antioxidant/Pro-Oxidant Activity and Applications in Medicine. Int. J. Mol. Sci. 2022, 23, 1247. [Google Scholar] [CrossRef]
  275. Timoshnikov, V.A.; Kobzeva, T.V.; Polyakov, N.E.; Kontoghiorghes, G.J. Redox Interactions of Vitamin C and Iron: Inhibition of the Pro-Oxidant Activity by Deferiprone. Int. J. Mol. Sci. 2020, 21, 3967. [Google Scholar] [CrossRef] [PubMed]
  276. Kontoghiorghes, G.J. Deferiprone and Iron-Maltol: Forty Years since Their Discovery and Insights into Their Drug Design, Development, Clinical Use and Future Prospects. Int. J. Mol. Sci. 2023, 24, 4970. [Google Scholar] [CrossRef] [PubMed]
  277. Ueda, K.; Kim, H.J.; Zhao, J.; Song, Y.; Dunaief, J.L.; Sparrow, J.R. Iron promotes oxidative cell death caused by bisretinoids of retina. Proc. Natl. Acad. Sci. USA 2018, 115, 4963–4968. [Google Scholar] [CrossRef] [PubMed]
  278. Selyutina, O.Y.; Timoshnikov, V.A.; Polyakov, N.E.; Kontoghiorghes, G.J. Metal Complexes of Omadine (N-Hydroxypyridine-2-thione): Differences of Antioxidant and Pro-Oxidant Behavior in Light and Dark Conditions with Possible Toxicity Implications. Molecules 2023, 28, 4210. [Google Scholar] [CrossRef]
  279. Kontoghiorghes, G.J. Chemical, Pharmacological, Toxicological and Therapeutic Advances of Deferiprone (L1) and Other Iron and Aluminium Chelators. Arch. Toxicol. Suppl. 1996, 18, 202–214. [Google Scholar] [CrossRef] [PubMed]
  280. Campbell, N.; Hasinoff, B. Iron supplements: A common cause of drug interactions. Br. J. Clin. Pharmacol. 1991, 31, 251–255. [Google Scholar] [CrossRef] [PubMed]
  281. Carota, G.; Distefano, A.; Spampinato, M.; Giallongo, C.; Broggi, G.; Longhitano, L.; Palumbo, G.A.; Parenti, R.; Caltabiano, R.; Giallongo, S.; et al. Neuroprotective Role of α-Lipoic Acid in Iron-Overload-Mediated Toxicity and Inflammation in In Vitro and In Vivo Models. Antioxidants 2022, 11, 1596. [Google Scholar] [CrossRef] [PubMed]
  282. Ott, D.B.; Hartwig, A.; Stillman, M.J. Competition between Al3+ and Fe3+ binding to human transferrin and toxicological implications: Structural investigations using ultra-high resolution ESI MS and CD spectroscopy. Metallomics 2019, 11, 968–981. [Google Scholar] [CrossRef] [PubMed]
  283. Crapper McLachlan, D.R.; Dalton, A.J.; Kruck, T.P.; Bell, M.Y.; Smith, W.L.; Kalow, W.; Andrews, D.F. Intramuscular desferrioxamine in patients with Alzheimer’s disease. Lancet 1991, 337, 1304–1308. [Google Scholar] [CrossRef]
  284. Nemeth, E.; Ganz, T. Hepcidin and Iron in Health and Disease. Annu. Rev. Med. 2023, 74, 261–277. [Google Scholar] [CrossRef]
  285. Ruchala, P.; Nemeth, E. The pathophysiology and pharmacology of hepcidin. Trends Pharmacol. Sci. 2014, 35, 155–161. [Google Scholar] [CrossRef] [PubMed]
  286. Arezes, J.; Nemeth, E. Hepcidin and iron disorders: New biology and clinical approaches. Int. J. Lab. Hematol. 2015, 37 (Suppl. 1), 92–98. [Google Scholar] [CrossRef] [PubMed]
  287. Hawula, Z.J.; Wallace, D.F.; Subramaniam, V.N.; Rishi, G. Therapeutic Advances in Regulating the Hepcidin/Ferroportin Axis. Pharmaceuticals 2019, 12, 170. [Google Scholar] [CrossRef]
  288. Rishi, G.; Subramaniam, V.N. Biology of the iron efflux transporter, ferroportin. Adv. Protein. Chem. Struct. Biol. 2021, 123, 1–16. [Google Scholar] [CrossRef] [PubMed]
  289. Ribeiro, S.; Belo, L.; Reis, F.; Santos-Silva, A. Iron therapy in chronic kidney disease: Recent changes, benefits and risks. Blood Rev. 2016, 30, 65–72. [Google Scholar] [CrossRef]
  290. Chekmarev, J.; Azad, M.G.; Richardson, D.R. The Oncogenic Signaling Disruptor, NDRG1: Molecular and Cellular Mechanisms of Activity. Cells 2021, 10, 2382. [Google Scholar] [CrossRef]
  291. Zhang, P.; Tchou-Wong, K.M.; Costa, M. Egr-1 mediates hypoxia-inducible transcription of the NDRG1 gene through an overlapping Egr-1/Sp1 binding site in the promoter. Cancer Res. 2007, 67, 9125–9133. [Google Scholar] [CrossRef]
  292. Orfanou, I.-M.; Argyros, O.; Papapetropoulos, A.; Tseleni-Balafouta, S.; Vougas, K.; Tamvakopoulos, C. Discovery and Pharmacological Evaluation of STEAP4 as a Novel Target for HER2 Overexpressing Breast Cancer. Front. Oncol. 2021, 11, 608201. [Google Scholar] [CrossRef]
  293. Wang, C.Y.; Canali, S.; Bayer, A.; Dev, S.; Agarwal, A.; Babitt, J.L. Iron, erythropoietin, and inflammation regulate hepcidin in Bmp2-deficient mice, but serum iron fails to induce hepcidin in Bmp6-deficient mice. Am. J. Hematol. 2019, 94, 240–248. [Google Scholar] [CrossRef]
  294. Camaschella, C.; Pagani, A.; Silvestri, L.; Nai, A. The mutual crosstalk between iron and erythropoiesis. Int. J. Hematol. 2022, 116, 182–191. [Google Scholar] [CrossRef]
  295. Kolnagou, A.; Kleanthous, M.; Kontoghiorghes, G.J. Reduction of body iron stores to normal range levels in thalassaemia by using a deferiprone/deferoxamine combination and their maintenance thereafter by deferiprone monotherapy. Eur. J. Haematol. 2010, 85, 430–438. [Google Scholar] [CrossRef] [PubMed]
  296. Farmaki, K.; Tzoumari, I.; Pappa, C.; Chouliaras, G.; Berdoukas, V. Normalisation of total body iron load with very intensive combined chelation reverses cardiac and endocrine complications of thalassaemia major. Br. J. Haematol. 2010, 148, 466–475. [Google Scholar] [CrossRef] [PubMed]
  297. Kolnagou, A.; Kontoghiorghe, C.N.; Kontoghiorghes, G.J. Prevention of Iron Overload and Long Term Maintenance of Normal Iron Stores in Thalassaemia Major Patients using Deferiprone or Deferiprone Deferoxamine Combination. Drug Res. 2017, 67, 404–411. [Google Scholar] [CrossRef] [PubMed]
  298. Kontoghiorghes, G.J.; Kleanthous, M.; Kontoghiorghe, C.N. The History of Deferiprone (L1) and the Paradigm of the Complete Treatment of Iron Overload in Thalassaemia. Mediterr. J. Hematol. Infect. Dis. 2020, 12, e2020011. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The important role of iron in heme, hemoglobin, and red blood cells. A cartoon representation of the essential role of iron (red circle, (A)), which is responsible for the red color of blood. Iron is embedded in a protoporphyrin ring forming heme (B), which is the prosthetic group in each of two alpha and two beta globin chain components of the protein hemoglobin (C). Each hemoglobin molecule (C) contains four molecules of iron, which are responsible for binding and carrying oxygen. The protein hemoglobin is the main constituent (95%) of red blood cells, as shown in the picture in (D).
Figure 1. The important role of iron in heme, hemoglobin, and red blood cells. A cartoon representation of the essential role of iron (red circle, (A)), which is responsible for the red color of blood. Iron is embedded in a protoporphyrin ring forming heme (B), which is the prosthetic group in each of two alpha and two beta globin chain components of the protein hemoglobin (C). Each hemoglobin molecule (C) contains four molecules of iron, which are responsible for binding and carrying oxygen. The protein hemoglobin is the main constituent (95%) of red blood cells, as shown in the picture in (D).
Ijms 24 12928 g001
Figure 2. The catalytic role of iron in free radical pathology and ferroptosis. Iron catalyzes the formation of free radicals, mainly the hydroxyl radical (OH), through the Fenton reaction, causing oxidative stress and leading to molecular, subcellular, cellular, and tissue damage (A). A similar pathway is followed in ferroptosis, where iron mostly from ferritinophagy catalyzes the formation of free radical cascades, causing lipid peroxidation on the cell membrane and cell death (B). Cell and tissue damage and ferroptotic cell death cause the release of more iron, resulting in a vicious circle of free radical production (C).
Figure 2. The catalytic role of iron in free radical pathology and ferroptosis. Iron catalyzes the formation of free radicals, mainly the hydroxyl radical (OH), through the Fenton reaction, causing oxidative stress and leading to molecular, subcellular, cellular, and tissue damage (A). A similar pathway is followed in ferroptosis, where iron mostly from ferritinophagy catalyzes the formation of free radical cascades, causing lipid peroxidation on the cell membrane and cell death (B). Cell and tissue damage and ferroptotic cell death cause the release of more iron, resulting in a vicious circle of free radical production (C).
Ijms 24 12928 g002
Figure 4. Toxicity aspects of transfusional iron overload from molecular to organ damage. Iron overload toxicity is implicated at different levels and involves progressively molecular, cellular and organ damaging changes, which can lead to fatalities. In each case the impact of toxicity depends on the concentration of excess iron. Congestive cardiac failure is the main cause of mortality in thalassemia major in the absence of effective chelation therapy. Excess iron is also implicated in some fatal cases of infections, liver fibrosis, and hepatocellular carcinoma (NTBI: Non-transferrin bound iron; LMWt: Low molecular weight).
Figure 4. Toxicity aspects of transfusional iron overload from molecular to organ damage. Iron overload toxicity is implicated at different levels and involves progressively molecular, cellular and organ damaging changes, which can lead to fatalities. In each case the impact of toxicity depends on the concentration of excess iron. Congestive cardiac failure is the main cause of mortality in thalassemia major in the absence of effective chelation therapy. Excess iron is also implicated in some fatal cases of infections, liver fibrosis, and hepatocellular carcinoma (NTBI: Non-transferrin bound iron; LMWt: Low molecular weight).
Ijms 24 12928 g004
Figure 5. The iron pools and metabolic pathways in iron overload. A cartoon displaying a cell, e.g., hepatocyte, where intracellular iron uptake is mostly accomplished by diferric transferrin (Tr-Fe2) through transferrin receptors (TrR), release of iron in a low molecular weight iron pool (LMWtFe) at low pH, and utilization of iron for storage in ferritin and hemosiderin and, to a smaller extent, for the turnover of other iron proteins. Intracellular iron intake in iron overload is also accomplished via low molecular weight (LMWt) non-transferrin bound iron (NTBI). The black dots represent iron molecules.
Figure 5. The iron pools and metabolic pathways in iron overload. A cartoon displaying a cell, e.g., hepatocyte, where intracellular iron uptake is mostly accomplished by diferric transferrin (Tr-Fe2) through transferrin receptors (TrR), release of iron in a low molecular weight iron pool (LMWtFe) at low pH, and utilization of iron for storage in ferritin and hemosiderin and, to a smaller extent, for the turnover of other iron proteins. Intracellular iron intake in iron overload is also accomplished via low molecular weight (LMWt) non-transferrin bound iron (NTBI). The black dots represent iron molecules.
Ijms 24 12928 g005
Table 1. Iron overload and other categories of iron toxicity.
Table 1. Iron overload and other categories of iron toxicity.
A. Transfusional and other iron-loaded categories of patients
(a) Thalassemia
β-thalassemia major
β-thalassaemia intermedia: Heterozygous mild β+/severe β+. Homozygous β (o) or β+ with other genetic factors leading to increased γ chain production. Homozygous normal Hb A2 β thalassemia (type I). Heterozygous β (o) or β+/Normal Hb A2 β thalassemia (type I).
δβ thalasemia and High Persistent Fetal Hemoglobin (HPFH): Homozygous (G) γ, δβ thalasemia. Heterozygous β (o) or β+ thalasemia/δβ thalasemia. Heterozygous Greek HPFH/β thalassemia.
β or δβ thalasemia with structural hemoglobin variants: Hemoglobin S,C,E in association with β (o), β+ or δβ thalasemia. Hb E thalassemia. Hb E/β thalassemia
Severe heterozygous β thalasemia
Interactions of α and β thalasemia: α- and β-thalassemia
Interactions of α thalasemia and Hemoglobin S
(b) Sickle cell anemia
(c) Hematologic categories: Myelodysplastic syndrome, aplastic anemia, Fanconi anemia, Blackfan-Diamond anemia, pyruvate kinase deficiency, hematopoietic stem cell transplantation, hereditary sideroblastic anemia.
(d) Oncologic and other categories: Cancer, renal dialysis
B. Idiopathic hemochromatosis and other diseases related to increased iron absorption
Iron overload in liver disease. Iron poisoning. Dietary or iatrogenic iron intake.
C. Hemolytic and other anemias: Hemolytic diseases. Pernicious anemia. Prophyria cutanea tarda. Hereditary spherocytosis. Congenital dyserythropoietic anemia. Congenital atransferrinemia. Hereditary hypochromic anemia.
D. Anemia of chronic disease: Inflammatory diseases, chronic kidney disease, cancer etc.
E. Neurodegenerative diseases: Friedreich’s ataxia, Alzheimer’s disease, Parkinson’s disease, etc.
F. Drug and natural product iron toxicity: Drug toxicity, e.g., doxorubicin. Natural product toxicity, e.g., ascorbic acid
G. Dietary toxins: Processed meat carcinogens. Red meat carcinogens. Nitroso heme complexes. Other heme and iron complexes.
H. Environmental toxins: Toxic iron particles from gas fumes. Asbestos.
I. Free radical toxicity: Diseases of free radical pathology. Ischemia reperfusion injury. Storage of organs for transplantation.
K. Ferroptotic cell death related diseases: Almost all common and rare diseases.
L. Iron toxicity associated with abnormalities related to regulatory proteins of iron metabolism
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kontoghiorghes, G.J. Iron Load Toxicity in Medicine: From Molecular and Cellular Aspects to Clinical Implications. Int. J. Mol. Sci. 2023, 24, 12928. https://doi.org/10.3390/ijms241612928

AMA Style

Kontoghiorghes GJ. Iron Load Toxicity in Medicine: From Molecular and Cellular Aspects to Clinical Implications. International Journal of Molecular Sciences. 2023; 24(16):12928. https://doi.org/10.3390/ijms241612928

Chicago/Turabian Style

Kontoghiorghes, George J. 2023. "Iron Load Toxicity in Medicine: From Molecular and Cellular Aspects to Clinical Implications" International Journal of Molecular Sciences 24, no. 16: 12928. https://doi.org/10.3390/ijms241612928

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop