Abstract
Iron is biologically essential, but also potentially toxic; as such it is tightly controlled at cell and systemic levels to prevent both deficiency and overload. Iron regulatory proteins post-transcriptionally control genes encoding proteins that modulate iron uptake, recycling and storage and are themselves regulated by iron. The master regulator of systemic iron homeostasis is the liver peptide hepcidin, which controls serum iron through degradation of ferroportin in iron-absorptive enterocytes and iron-recycling macrophages. This review emphasizes the most recent findings in iron biology, deregulation of the hepcidin-ferroportin axis in iron disorders and how research results have an impact on clinical disorders. Insufficient hepcidin production is central to iron overload while hepcidin excess leads to iron restriction. Mutations of hemochro-matosis genes result in iron excess by downregulating the liver BMP-SMAD signaling pathway or by causing hepcidin-resistance. In iron-loading anemias, such as β-thalassemia, enhanced albeit ineffective ery-thropoiesis releases erythroferrone, which sequesters BMP receptor ligands, thereby inhibiting hepcidin. In iron-refractory, iron-deficiency ane-mia mutations of the hepcidin inhibitor TMPRSS6 upregulate the BMP-SMAD pathway. Interleukin-6 in acute and chronic inflammation increases hepcidin levels, causing iron-restricted erythropoiesis and ane-mia of inflammation in the presence of iron-replete macrophages. Our improved understanding of iron homeostasis and its regulation is having an impact on the established schedules of oral iron treatment and the choice of oral versus intravenous iron in the management of iron deficiency. Moreover it is leading to the development of targeted therapies for iron overload and inflammation, mainly centered on the manipulation of the hepcidin-ferroportin axis.Introduction
Research advances in understanding the biological functions and homeostasis of iron have clarified its role in physiology and disease. Iron, essential for hemoglobin synthesis, is indispensable to all cells for the production of heme and iron-sulfur (Fe/S) clusters, which are components of proteins/enzymes involved in vital biological processes such as respiration, nucleic acid replication and repair, metabolic reactions and host defense. While essential for life, excess iron is toxic. The ability to accept/release electrons explains the propensity of iron to damage cell components and is the reason why body iron must be tightly regulated. The two-faced nature of iron is also evident in its disorders, which span from iron excess to iron deficiency and maldistribution, when some tissues are iron-loaded and others are iron-deficient.
In the new millennium studies of genetic and acquired iron disorders and the development of their corresponding murine models have identified novel iron genes, proteins and pathways and unveiled the central role of the hepcidinferro-portin axis in systemic iron homeostasis. This review summarizes recent advances in the understanding of iron trafficking, utilization and regulation, emphasizing the implications for iron disorders of hematologic interest; for further insights readers are directed to specific reviews.31
Iron trafficking
Iron trafficking is an example of circular economy. Only 1-2 mg iron are absorbed daily in the gut, compensating for an equal loss; most iron (20-25 mg/daily) is recycled by macrophages upon phagocytosis of erythrocytes. The site of regulated non-heme iron uptake is the duodenum: non-heme iron is imported from the lumen by the apical divalent metal transporter 1 (DMT1) after reduction from ferric to ferrous iron by duodenal cytochrome B reductase (DCYTB). Absorption of heme exceeds that of non-heme iron, though the mechanisms remain obscure. In enterocytes non-utilized iron is stored in ferritin - and lost with mucosal shedding - or exported to plasma by basolateral membrane ferroportin according to the body’s needs (Figure 1).
The role of transferrin and its receptors
The plasma iron pool is only 3-4 mg and must turn over several times daily to meet the high (20-25 mg) demand of erythropoiesis and other tissues. The iron carrier transferrin is central to iron trafficking. Binding to its ubiquitous receptor TFR1, transferrin delivers iron to cells through the well-known endosomal cycle.1 This function is crucial not only for erythropoiesis, but also for muscle4 and for B- and T-lymphocytes, as highlighted by a TFR1 homozygous mutation that causes combined immunodeficiency with only mild anemia.5 TFR1 is also essential in the gut to maintain epithelial homeostasis independently of its function of an iron importer;6 in hepatocytes TFR1 is dispensable for basal iron uptake, but essential in iron loading to finely tune the hepcidin increase.7
Transferrin is emerging as a key regulator of iron homeostasis through binding to its second receptor TFR2, which has a lower binding affinity than TFR18 and whose expression is restricted to hepatocytes and erythroblasts. When plasma iron concentration is high, diferric transferrin binds TFR2 inducing upregulation of hepcidin in hepatocytes and a reduction of erythropoietin responsiveness in erythroid cells9 where TFR2 binds erythropoietin receptors.10 The reverse occurs in iron deficiency. The dual function of transferrin as an iron cargo and regulator seems to be dependent on the unequal ability of iron binding of the N and C terminal lobes and operates through the differential interaction of monoferric transferrin with the two receptors.11
Iron recycling
Macrophages phagocytize senescent and damaged erythrocytes, recover iron from heme through heme oxygenase (HMOX) 1 and may utilize, conserve or recycle the metal. The relevance of their role is strengthened by the severity of conditions in which recycling is altered. HMOX1 mutations in children cause a rare, severe disorder12 and reduced recycling in inflammation causes anemia. Macrophage ferroportin is crucial for iron balance. Its expression is upregulated by heme and downregulated by inflammatory cytokines contributing to iron sequestration and its translation is repressed by iron. The protein is ultimately controlled at the post-translational level by hep-cidin.13
Cell iron import
Intracellular iron is used for multiple functions; if not utilized it is stored in ferritin, or exported by ferroportin, in order to maintain the labile iron pool within narrow limits to avoid toxicity. Although all cells may import, export or store iron, some have specific functions:1 e.g., erythroblasts are specialized in iron uptake, macrophages and enterocytes in iron export, and hepatocytes in iron storage. Within cells most iron is transferred to mitochondria for heme and Fe/S cluster production. Heme is indispensable for hemoglobin, cytochromes and enzyme activity. Biogenesis of Fe/S clusters is a process conserved from yeast to humans: this prosthetic group is essential to proteins involved in genome maintenance, energy conversion, iron regulation and protein translation.1514 In erythroblasts >80% iron is directed to mitochondria through a “kiss and run” mechanism between endosomes and mitochondria.16 Mitoferrin 1 and 2 are iron transporters of the inner mitochondrial membrane, the former being essential for zebrafish and murine erythropoiesis.17
Ferritin may store up to 4,500 iron atoms in a shell-like structure formed by 24 chains, comprising both heavy (H) chains, with ferroxidase activity, and light (L) chains.18 Ferritin storage of iron provides protection from oxidative damage, and also saves an essential element for future needs. H-ferritin deletion is incompatible with life and its conditional deletion in the gut deregulates the fine mechanism of iron absorption causing iron overload.19 L-ferritin heterozygous mutations are rare and limited to the 5’ iron regulatory element (IRE) - leading to escape from iron regulatory protein (IRP) control and constitutive high ferritin synthesis in hyperferritinemia-cataract syndrome.20 Rare dominant mutations lead to elongated proteins and neuro-ferritinopathies, a type of neurodegeneration caused by abnormal ferritin aggregates in the basal ganglia and other areas of the brain21 (Table 1).
In the clinical setting serum ferritin is a marker of iron deficiency when its level is low, and of iron overload/inflammation when its level is increased, reflecting macrophage ferritin content. However, both the origin and the function of serum ferritin remain largely unexplored. One hypothesis is that the secreted ferritin22 may be re-uptaken by cells as an alternative mechanism of iron recycling, e.g., when iron release from macrophages is limited in inflammation.
The cytosolic chaperon Poly (rC) binding protein 1 (PCBP1) delivers iron to ferritin,23 and Pcbp1 null mice have microcytic anemia.24 Ferritin turnover occurs through “ferritinophagy”, an autophagic process orchestrated by nuclear receptor co-activator (NCOA)4, a cargo molecule that directs ferritin to lysosomal degradation, to recover iron when needed.2625 PCBP1 also delivers iron to prolylhydroxylase (PHD2) the enzyme that induces degradation of hypoxia inducible factors (HIF), one of the several links between iron and the hypoxia pathways.27
Iron export
The ubiquitous iron exporter ferroportin cooperates with the oxidases ceruloplasmin or hephaestin, to release ferric iron to transferrin. Enterocytes, macrophages, hepatocytes and trophoblasts express high ferroportin levels for their specific functions in iron homeostasis. Blocking iron export may be dangerous in some cells. For example, conditional ferroportin deletion in murine cardiomyocytes leads to local iron overload and cardiac failure;28 furthermore, specific deletion of ferroportin in erythroblasts and erythrocytes leads to hemolytic anemia, due to the toxicity of iron derived from hemoglobin oxidation in an environment (red blood cells) with limited antioxidant capacity.3029
Erythroblasts may also export heme through feline leukemia virus C receptor (FLVCR).3231 The latter two export mechanisms seem counterintuitive in cells that need iron/heme for the production of hemoglobin, but are likely biological safeguard mechanisms that protect erythroid cells from iron/heme excess.
Iron homeostasis
Maintaining iron balance requires tight regulation at cellular, systemic and tissue levels.
Cell iron homeostasis
This system is based on the post-transcriptional control of iron genes mediated by the interaction of IRP with IRE of their mRNA untranslated regions. In iron-deficiency states, IRP1 and 2 increase iron uptake by stabilizing TFR1 mRNA and blocking iron storage and export by suppressing ferritin and ferroportin translation. In iron-replete cells, Fe/S clusters convert IRP1 into cytosolic aconitase, while IRP2 undergoes iron-dependent proteasomal degradation. The IRP1/aconitase interconversion on the one hand links iron to tricarboxylic acid and cell metabolism, and on the other hand highlights that, through Fe/S clusters, iron controls its own availability. IRP2 binds IRE at normal tissue oxygen, IRP1 acts in hypoxic tissues, such as the duodenum and kidneys.
Murine models of total and tissue-specific IRP inactiva tion are providing further insights into the local function of these proteins. Deletion of both Irps in mice is incompatible with life; loss of Irp2 results in mild anemia, erythropoietic protoporphyria and adult-onset neurodegeneration in mice33 and in patients,34 likely because of functional iron deficiency. Adult Irp1-knockout mice have a normal phenotype in basal conditions. Intestinal epithelium Irp1 and 2 deletion in adult mice leads to impaired iron absorption and local iron retention because of ferritin-mediated mucosal block.35
To escape IRP control, both enterocytes and erythroid cells also have a ferroportin isoform that, lacking the 5’ IRE, ensures iron export in iron deficiency while remaining sensitive to degradation by hepcidin.36
In iron deficiency, cells may recover iron through ferritinophagy, a process mediated by the multifunctional protein NCOA4.2625 First described as a transcriptional co-activator of androgen nuclear receptor, this protein is iron-regulated at the post-translational level. In iron-replete cells NCOA4 is bound by the E3 ubiquitin ligase HERC2 and degraded by the proteasome.37 In iron-deficient cells NCOA4 binds ferritin inducing its degradation. NCOA4 also controls DNA duplication origins and its loss in vitro predisposes cells to replication stress and senescence,38 coupling cell duplication and iron availability. Ncoa4-knockout mice accumulate ferritin in the liver and spleen, have reduced iron recycling and demonstrate increased susceptibility to iron-deficiency anemia.39 The high NCOA4 expression in erythroblasts24 suggested a role for ferritinophagy in hemoglobinization in vitro,24 in zebrafish embryos37 and in mice.40 However, the major relevance of the process is in iron-storing macrophages (Nai A. et al., unpublished data) contributing to systemic homeostasis.
Systemic iron homeostasis: the hepcidin-ferroportin axis
The identification of hepcidin was a breakthrough in understanding how the liver is the central regulator of iron homeostasis and how its deregulation leads to iron disorders. The 25 amino acid mature hepcidin peptide controls iron export to the plasma by inducing lysosomal degradation of the iron exporter ferroportin in enterocytes, macrophages and hepatocytes;13 moreover, hepcidin also occludes the central cavity that exports iron in ferroportin.41
Hepcidin transcription is upregulated in hepatocytes by circulating and tissue iron, through a crosstalk with liver sinusoidal endothelial cells, which produce the ligands (BMP6 and 2) that activate the hepatocyte BMP-SMAD pathway. BMP6 expression is regulated by iron,42 possibly in the context of an antioxidant response, controlled by NRF2.43 BMP2 is less iron sensitive and is highly expressed in basal conditions.4544
Inflammatory cytokines such as interleukin (IL)-6 upregulate hepcidin expression by activating the IL-6R-JAK2-STAT3 pathway. High hepcidin levels induce iron retention in macrophages, high serum ferritin levels and iron-restricted erythropoiesis, all features of anemia of inflammation. For full hepcidin activation the IL-6 pathway requires functional BMP-SMAD signaling.46
Hepcidin expression is inhibited by iron deficiency, expansion of erythropoiesis, anemia/hypoxia, testosterone and possibly other factors.471 The most powerful inhibitor is the liver transmembrane serine protease matriptase 2, encoded by TMPRSS6,48 which cleaves the BMP co-receptor hemojuvelin,49 thereby attenuating BMP-SMAD signaling and hepcidin transcription. The relevance of TMPRSS6 is highlighted by iron-refractory, iron-deficiency anemia (IRIDA), which results from TMPRSS6 mutations in patients50 and inactivation in mice.48 Deregulated, persistently high hepcidin blocks iron entry into the plasma and leads to iron deficiency. Another local inhibitor, the immunophillin FKBP12, binds the BMP receptor ALK2, suppressing the pathway activation.51
Erythroferrone (ERFE) is released by erythroid precursors stimulated by erythropoietin to suppress hepcidin expression and favor iron acquisition for hemoglobin synthesis.52 In hypoxia hepcidin is also suppressed in vitro by soluble hemojuvelin, released by furin,53 an effect unclear in vivo, and by platelet-derived growth factor-BB in volunteers exposed to hypoxia.54 Proposed models of hepcidin regulation in different conditions are depicted in Figure 2A-D.
Macrophages produce hepcidin in inflammation, potentiating the systemic effect on iron sequestration.55
Local effects of hepcidin
As an antimicrobial peptide hepcidin is induced in the skin of patients with necrotizing fasciitis caused by group A streptococcal infections. Mice without hepcidin in keratinocytes fail to block the spread of infection because of a reduction of the neutrophils recruiting chemokine CXCL1.56
Cardiomyocytes produce hepcidin with local effect on ferroportin. Conditional cardiomyocyte hepcidin deletion in mice does not affect systemic iron homeostasis but leads to excess iron export, severe contractile dysfunction and heart failure.57
Emerging evidence links iron with lipid and glucose metabolism. Genome-wide association studies found overlapping associations for iron and lipid traits.58 Adipocytes produce hepcidin in severe obesity59 and hepcidin and gluconeogenesis are concomitantly upregulated in conditions of insulin-resistance.60 Finally Tmprss6-knockout mice with high hepcidin levels are protected from obesity induced by a high-fat diet.61
Crosstalk between iron, oxygen and erythropoiesis
The hepcidin-ferroportin axis intersects other biological systems, such as IRP, hypoxia responsive pathways and erythropoietin signaling.
Iron absorption is a physiological example of crosstalk between IRP-hypoxia and the hepcidin-ferroportin axis. In the hypoxic duodenal environment, IRP1 controls translation of hypoxia-inducible factor 2α (HIF-2α) which, stabilized by prolyl hydroxylase, upregulates the expression of apical (DMT1) and basolateral (ferroportin) enterocyte iron transporters.62 In iron deficiency, absorption is enhanced by hepcidin downregulation, which, favoring export, depletes enterocytes of iron, further stabilizing HIF-2α63 (Figure 1). In iron overload, high hepcidin increases enterocyte iron and impairs luminal uptake. In addition, the rapid cell turnover with shedding of ferritin-loaded enterocytes further limits iron absorption. In this way the interaction between local (hypoxia, IRP) and systemic (hepcidin) mechanisms optimizes iron balance.
Iron and erythropoiesis are interconnected at multiple levels and are reciprocally regulated. First, iron tunes renal production of erythropoietin, the growth factor essential for proliferation and differentiation of erythroid cells, The synthesis of erythropoietin is mediated by HIF-2α. In iron deficiency, IRP1 binding to HIF-2α 5’IRE represses the latter’s translation and decreases erythropoietin production, to limit erythropoiesis and iron consumption. When the mechanism fails, as in Irp1-knockout mice, a transient polycythemia occurs in the relatively iron-deficient young animals, which reverts in adult mice with iron sufficiency.6564 In this context prolyl hydroxylase, the enzyme that induces degradation of HIF-2α, is iron-dependent and is thus inactive in iron deficiency.
Second, in in vitro studies, iron deprivation induced a block of early erythroid progenitors, by inactivating mitochondrial aconitase;66 this block could be overcome by isocitrate supplementation.67 Finally the erythroid response to iron restriction is optimized by the iron sensor TFR2, a partner of erythropoietin receptor.10 Loss of bone marrow TFR2 in mice increases the sensitivity of erythroblasts to erythropoietin, which causes erythrocytosis, especially in iron deficiency.68 Liver TFR2 upregulates hepcidin and TFR2 mutations cause hemochromato-sis.69 As a sensor of iron-bound transferrin, erythroid TFR2 regulates erythropoiesis, while liver TFR2, controlling hepcidin, modulates iron acquisition according to erythropoietic needs. The recently demonstrated TFR2 expression in osteoclasts and osteoblasts70 places this iron sensor at the crossroads of red cell production, iron homeostasis and bone turnover.
While iron regulates erythropoiesis, the reverse is also true. The old hypothesis that the erythropoietic drive controls iron absorption through an erythroid regulator71 was confirmed by the discovery of ERFE, the best example of tissue-mediated regulation of hepcidin. ERFE is a member of the tumor-necrosis factor (TNF)-α family, produced by several tissues, but increased in response to erythropoietin only in erythroid precursors. ERFE sequesters BMP receptor ligands, especially BMP6,72 inhibiting BMP-SMAD signaling and hepcidin. However, ERFE fails to suppress hepcidin when the BMP pathway is overactive.73 Erfe-knockout mice are not anemic, indicating that ERFE has a modest effect on hepcidin repression in steady state. However, ERFE contributes to iron loading in mice with β-thalassemia.74
Iron disorders
The improved understanding of iron physiology has profoundly changed the modern approach to iron disorders, known historically for centuries as iron deficiency (chlorosis) in young females and hemochromatosis (bronze diabetes) in middle-age males. We now suspect hemochromatosis based on iron parameters and confirm the diagnosis by genetic testing well before the development of iron overload and organ damage. We are aware that anemia is a complication of iron deficiency, though not the only one, since other tissues/organs may be iron-depleted before anemia develops, as occurs in chronic heart failure.75
Hepcidin is tightly controlled to maintain body iron balance. Loss of this control leads to opposite genetic or acquired disorders (Table 1).
Genetic disorders
The pathophysiology and diagnosis of hemochromatosis were profoundly influenced by the cloning of the HFE gene76 and the definition of the genetic heterogeneity of the disease (Table 1). Overall hemochromatosis is due to “insufficient hepcidin production” or exceptionally to “hepcidin resistance”.77 Mutations in genes of the hepcidin-ferroportin axis disrupt iron homeostasis, leading to increased iron absorption, high transferrin saturation and increased toxicity from non-transferrin bound iron (NTBI) species.78 The commonest form of hemochromatosis in Caucasians is due to homozygous HFE(C282Y) mutations. Genetic tests allow early diagnosis so that individuals with the affected genotypes show high biochemical penetrance (increased transferrin saturation ± increased serum ferritin) but low clinical expression. Loss-of-function mutations of HJV, TFR2 and HAMP (encoding hepcidin) lead to more severe diseases, collectively called “non-HFE hemochromatosis”. In all the recessive forms hepcidin is inappropriately low in comparison with iron excess and the onset and severity of iron overload correlate with the hormone deficiency.79 The implementation of next-generation sequencing in familial and isolated cases of iron overload80 has enabled the identification of mutations in more than one gene and provided examples of digenic inheritance. Both genetic and acquired modifiers contribute to the penetrance of HFE-hemochromatosis, interfering with hepcidin expression: e.g., alcohol aggravates the iron burden, whereas blood donations attenuates it.77
Ferroportin mutations are inherited in a dominant manner (Table 1). The different effects of these mutations account for the controversy in disease nomenclature. Loss-of-function mutations impair iron export, are associated with iron accumulation in Küpffer cells and require no or reduced phlebotomy therapy, representing the true “ferroportin disease”.81 Gain-of-function mutations lead to hepcidin resistance and the release of too much iron, as occurs in hemochromatosis.
The clinical severity of hemochromatosis is related to NTBI, a toxic iron species bound to low molecular weight molecules, easily taken up by hepatocytes and pancreatic cells via ZIP14 transporter82 and by cardiomyocytes through other transporters.83 While iron uptake by transferrin receptor is tightly regulated, the uptake of NTBI is not and persists in iron overload. NTBI leads to the generation of reactive oxygen species and cell damage, causing liver fibrosis (which may progress to cirrhosis and hepatocellular carcinoma), chronic heart failure, diabetes, hypopituitarism and other complications of iron loading.7977
This form of iron deficiency anemia (IRIDA) was recognized after the discovery of hepcidin as being due to mutations of its inhibitor TMPRSS6.50 High hepcidin levels lead to a phenotype opposite to that of hemochromatosis, reminiscent of anemia of inflammation.84 The anemia is refractory to oral iron and may require intravenous therapy, especially when iron demand is high.85 TMPRSS6 genetic variants modulate iron and hematologic traits in several genome-wide association studies,58 alter hepcidin levels in normal subjects86 and might confer susceptibility/resistance to iron deficiency, as observed in blood donors.87
Other rare recessive disorders of the transferrin receptor pathway – such as hypotransferrinemia and DMT1 mutations – lead to “atypical microcytic anemia” with increased transferrin saturation and iron stores, because of decreased iron utilization by blunted erythropoiesis.85
Ringed sideroblasts are erythroblasts with iron-loaded mitochondria that, clustering around the nucleus, confer the appearance of a ring at Perls iron staining. Hereditary sideroblastic anemias are usually due to heme deficiency: X-linked sideroblastic anemia is caused by mutations in ALAS2, the first, rate-limiting enzyme of heme biosynthesis, while recessive forms are associated with mutations of mitochondria glycine importer solute carrier family 25 member 38.88 Rare severe cases result from mutations of Fe/S cluster proteins, such as GLRX589 or HSPA9,90 which decrease Fe/S groups and the activity of ferrochelatase, the last enzyme of the heme pathway. Another cause of low heme in GLRX5 deficiency is the overactive IRP1 that, not being converted to aconitase because of the Fe/S cluster deficit, blocks ALAS2 translation, thereby preventing heme formation. These disorders reveal the tight connection of heme-Fe/S metabolism. Among syndromic forms, the X-linked ABCB7 deficiency reduces export of Fe/S clusters to the cytosol,91 while others are associated with immunodeficiency92 strengthening the need of Fe/S clusters in other cell types (Table 1). Those due to mitochondrial protein mutations91 are not discussed here.
Acquired iron disorders
Iron deficiency, both isolated and associated with anemia, represents one of the five major causes of disability burden worldwide, especially in women.93 For discussions of the etiology, clinical presentation and treatment of iron deficiency with or without anemia readers are directed to specific reviews.9694 In absolute iron deficiency low total body and serum iron fully suppress hepcidin, a mechanism of adaptation to increase iron absorption. In functional iron deficiency (e.g., in inflammation) total body iron is not decreased, but iron is sequestered in stores by the high hepcidin levels.9784 This distinction strongly influences the route of iron administration required to treat iron deficiency, as discussed below.
Proinflammatory cytokines such as IL-6 and IL-1β, produced in chronic infections, autoimmunity, cancer, renal failure and other chronic disorders activate hepcidin expression leading to iron-restricted erythropoiesis and anemia of inflammation, once named anemia of chronic diseases.989784 By withholding iron in macrophages, extracellular Gram-negative microorganisms are deprived of this essential nutrient.10099 This is an innate defense mechanism known as ‘nutritional immunity’.101 A recent interpretation is that hypoferremia prevents the generation of NTBI that potentiates the pathogenicity of Gram-negative bacteria.102 Anemia, usually moderate and normocytic, is multifactorial, because of concomitant insufficient erythropoietin production and impaired early erythroid commitment.98 Microcytosis occurs in longstanding severe inflammation such as in Castleman disease, a lymphoproliferative disorder in which high IL-6 production strongly enhances hepcidin synthesis103 or in patients with ectopic hepcidin expression by liver adenomas.104 Anemia reverts after anti-IL6 receptor treatment in Castleman disease or after surgical removal of the tumor in the case of adenoma.
Anemia of inflammation regresses with control of the disease. In selected cases intravenous iron or erythropoiesis-stimulating agents are used. Since treatment is often unsatisfactory, manipulation of the hepcidin pathway (blocking either its production or function) is proposed as a novel therapeutic opportunity.97
Low hepcidin levels explain the iron overload that develops in the absence of blood transfusions in “iron-loading anemias”, i.e., anemias with ineffective erythropoiesis (Table 1). ERFE, released by erythropoietin-stimulated erythroblasts, inhibits hepcidin, despite iron overload. In non-transfusion-dependent β-thalassemia patients, serum ERFE levels are high,105 to ensure iron acquisition for the expanded erythropoiesis.106 However, since the erythropoiesis is inefficient, excess iron interferes with erythroblast maturation aggravating anemia in a vicious cycle.107 In patients with transfusion-dependent thalassemia, hepcidin increases following transfusions which partially suppress erythropoiesis.
ERFE contributes to the iron loading of some clonal myelodysplastic syndromes. Patients with the ringed sideroblasts subtype of myelodysplastic syndrome (once called refractory anemia with ringed sideroblasts) carry a somatic mutation in the spliceosome gene SF3B1.108 Among other abnormally spliced products, an elongated variant of ERFE is more efficient than the wildtype hormone in hepcidin repression.109
Diagnostic implications
Notwithstanding spectacular advances in our understanding of iron metabolism and homeostasis our diagnostic approach to iron disorders still relies mainly on three historical tests: serum iron, transferrin (or total iron binding capacity) and ferritin. Transferrin saturation (Tsat), i.e. the ratio of serum iron/total iron binding capacity and serum ferritin coupled with genetic testing and non-invasive magnetic resonance imaging measurements of liver iron content, define the nature and severity of iron loading in both hemochromatosis77 and thalassemia.110 Other useful markers are the level of serum soluble transferrin receptor (sTFRC), related to the expansion of erythropoiesis or iron deficiency, the sTFRC/log ferritin ratio for the diagnosis of iron deficiency in inflammation98 and the Tsat/log hepcidin ratio to suspect IRIDA.111
Enzyme-linked immunosorbent assay kits can measure serum hepcidin levels. However, this does not provide any information additional to serum ferritin, since the two variables are tightly related.113112 Some researchers propose determining hepcidin levels in order to choose the better therapeutic route of administration of iron supplementation (oral vs. intravenous),114 as well as its correct timing115 and schedule.116 However, besides being subject to circadian oscillations, hepcidin levels change rapidly in response to activating and inhibitory signals, making their measurement useful in only a limited number of conditions.47 A kit to measure human serum ERFE concentration is available for research purposes. Whether the elongated ERFE identified in individuals with SF3B1 mutations will become a biomarker of ringed sideroblast myelodysplastic syndrome109 remains to be tested.
Therapeutic implications
Hepcidin levels favor response (when low) or resistance (when high) to oral iron administration, explaining part of iron refractoriness.117 The dynamics of the increase in hepcidin levels after oral iron therapy have suggested that alternate-day administration of iron salts is an alternative to daily refracted doses, with the former being a protocol that increases both efficacy and tolerability, at least in women with iron deficiency without or with mild anemia.119118116 The availability of more tolerated, iron-stable and efficacious preparations has increased the use of intravenous iron, especially of the high-dose single-injection compounds.120 However, when used to correct iron deficiency in inflammation, intravenous iron may lead to macrophage iron accumulation whose long-term effects are unknown.
Manipulation of the hepcidin-ferroportin axis is the most logical experimental approach to iron disorders. The rationale is to use hepcidin agonists for iron overload disorders caused by inappropriate/low hepcidin and hepcidin antagonists to release sequestered iron in IRIDA and in anemia of inflammation (Table 2).
Increasing hepcidin levels/decreasing ferroportin activity
In preclinical studies, increasing hepcidin levels prevented iron overload or redistributed iron to sites of safe stor age. Potentially useful in hemochromatosis, whose treatment is still based on phlebotomy,77 hepcidin agonists are of interest in disorders with ineffective erythropoiesis, such as β-thalassemia.107 Agonists include hepcidin analogues, minihepcidins, inhibitors of hepcidin repressors such as anti-TMPRSS6 molecules or compounds that block ferroportin activity. By inducing iron restriction hepcidin agonists ameliorated anemia and iron overload in preclinical studies of thalassemia models;121106 a few hepcidin agonists are currently being investigated in phase I-II clinical trials (Table 2). Hepcidin mimics could also be useful to induce iron restriction in polycythemia.122 According to recent findings hepcidin might have a role as an antimicrobial peptide in the treatment of Gram-negative sepsis102 and streptococcal necrotizing fasciitis.56
Other approaches
In non-transfusion-dependent β-thalassemia (Hbbth1/th1 and Hbbth3/+) mice transferrin infusions improve the phenotype, increasing hepcidin and hemoglobin levels, improving erythrocyte survival and limiting splenomegaly,124123 effects similar to those observed when Tfr1 expression is decreased.125 Selective inactivation of bone marrow Tfr2 improves anemia in a non-transfusion-dependent Hbbth3/+ model, enhancing the sensitivity of erythroid cells to ery-thropoietin.126
Short interfering RNA against DMT1, administered in nanoparticles to target intestinal absorption,127 established a proof of principle of reducing dietary iron uptake. Another approach might be to block intestinal HIF-2α by specific antagonists.
Clinical trials are showing that correcting ineffective erythropoiesis by activin ligand traps128 not only improves anemia but, in the long-term, also iron loading in both non-transfusion-dependent and transfusion-dependent thalassemia129 and ringed sideroblast myelodysplastic syndrome.130 Some thiazolidinones have been shown to stimulate hepcidin activity in preclinical studies.131 The use of proton pump inhibitors reduced the need for phlebotomy in patients with hemochromatosis.132
Decreasing hepcidin levels/increasing ferroportin function
In preclinical models of anemia of inflammation, hepcidin antagonists decreased hepcidin expression, an effect verified in clinical trials for some compounds.133 Another option is to interfere with the hepcidin-ferroportin interaction (Table 2). However, targeting the hepcidin-ferroportin axis may not fully correct this multifactorial anemia characterized by low erythropoietin and a blunted erythropoietic response.9897 Another approach is based on manipulation of the hypoxia-responsive pathway.63 Prolyl hydroxylase inhibitors or HIF stabilizers, now tested in chronic kidney disease, by increasing HIF-2α might target two abnormal processes enhancing both erythropoietin synthesis and iron absorption.134
Unresolved issues
Notwithstanding significant advances many questions about iron metabolism and homeostasis remain unanswered. The mechanisms of intestinal heme absorption are mysterious, as are the roles of secreted ferritin and soluble transferrin receptor. We have just started exploring the autonomous regulation of iron in the heart and vascular wall; the role of iron (deficiency or excess) as a cofactor of metabolic disorders, chronic liver disease, heart failure, pulmonary hypertension and neurodegeneration still requires elucidation. We need to be able to diagnose isolated tissue iron deficiency better and to increase the limited number of iron status markers.
In hematology we need to clarify the relationship between iron and platelet production considering that iron deficiency directs the common erythroid-megakaryocyte precursor towards the platelet lineage.135 More information is required on the role of iron in B-lymphocyte development and function, in B-cell malignancies, such as multiple myeloma,136 and in response to infectious diseases. We have to explore better how iron/TFR2 intersects the erythropoietin signaling pathway and bone metabolism.
We need novel protocols of iron supplementation and clear indications regarding high-dose intravenous iron to optimize iron therapy. Targeted approaches, now in clinical trials, have the potential to change traditional treatment – such as the time-honored phlebotomy-based regimen – for disorders such as hemochromatosis. Repurposing commercially available compounds, developed for other conditions, to iron/erythroid disorders is another option. All these approaches will, it is hoped, enable a more personalized treatment of iron disorders in the near future.
Acknowledgments
This work was supported in part by an ASH Global Research Award 2017 to AN.
Footnotes
- Check the online version for the most updated information on this article, online supplements, and information on authorship & disclosures: www.haematologica.org/content/105/2/260
- Received November 4, 2019.
- Accepted December 18, 2019.
References
- Hentze MW, Muckenthaler MU, Galy B, Camaschella C. Two to tango: regulation of Mammalian iron metabolism. Cell. 2010; 142(1):24-38. PubMedhttps://doi.org/10.1016/j.cell.2010.06.028Google Scholar
- Muckenthaler MU, Rivella S, Hentze MW, Galy B. A red carpet for iron metabolism. Cell. 2017; 168(3):344-361. PubMedhttps://doi.org/10.1016/j.cell.2016.12.034Google Scholar
- Wang CY, Babitt JL. Liver iron sensing and body iron homeostasis. Blood. 2019; 133(1):18-29. PubMedhttps://doi.org/10.1182/blood-2018-06-815894Google Scholar
- Barrientos T, Laothamatas I, Koves TR. Metabolic catastrophe in mice lacking trans-ferrin receptor in muscle. EBioMedicine. 2015; 2(11):1705-1717. Google Scholar
- Jabara HH, Boyden SE, Chou J. A missense mutation in TFRC, encoding transfer-rin receptor 1, causes combined immunodeficiency. Nat Genet. 2016; 48(1):74-78. Google Scholar
- Chen AC, Donovan A, Ned-Sykes R, Andrews NC. Noncanonical role of transfer-rin receptor 1 is essential for intestinal home-ostasis. Proc Natl Acad Sci U S A. 2015; 112(37):11714-11719. PubMedhttps://doi.org/10.1073/pnas.1511701112Google Scholar
- Fillebeen C, Charlebois E, Wagner J. Transferrin receptor 1 controls systemic iron homeostasis by fine-tuning hepcidin expression to hepatocellular iron load. Blood. 2019; 133(4):344-355. PubMedhttps://doi.org/10.1182/blood-2018-05-850404Google Scholar
- Kawabata H, Yang R, Hirama T. Molecular cloning of transferrin receptor 2. A new member of the transferrin receptor-like family. J Biol Chem. 1999; 274(30):20826-20832. PubMedhttps://doi.org/10.1074/jbc.274.30.20826Google Scholar
- Camaschella C, Pagani A, Nai A, Silvestri L. The mutual control of iron and erythro-poiesis. Int J Lab Hematol. 2016; 38(Suppl 1):20-26. https://doi.org/10.1111/ijlh.12505Google Scholar
- Forejtnikova H, Vieillevoye M, Zermati Y. Transferrin receptor 2 is a component of the erythropoietin receptor complex and is required for efficient erythropoiesis. Blood. 2010; 116(24):5357-5367. PubMedhttps://doi.org/10.1182/blood-2010-04-281360Google Scholar
- Parrow NL, Li Y, Feola M. Lobe specificity of iron-binding to transferrin modulates murine erythropoiesis and iron homeostasis. Blood. 2019; 134(17):1373-1384. Google Scholar
- Yachie A, Niida Y, Wada T. Oxidative stress causes enhanced endothelial cell injury in human heme oxygenase-1 deficiency. J Clin Invest. 1999; 103(1):129-135. PubMedhttps://doi.org/10.1172/JCI4165Google Scholar
- Nemeth E, Tuttle MS, Powelson J. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its inter-nalization. Science. 2004; 306(5704):2090-2093. PubMedhttps://doi.org/10.1126/science.1104742Google Scholar
- Lill R, Broderick JB, Dean DR. Special issue on iron-sulfur proteins: Structure, function, biogenesis and diseases. Biochim Biophys Acta. 2015; 1853(6):1251-1252. Google Scholar
- Braymer JJ, Lill R. Iron-sulfur cluster biogenesis and trafficking in mitochondria. J Biol Chem. 2017; 292(31):12754-12763. PubMedhttps://doi.org/10.1074/jbc.R117.787101Google Scholar
- Hamdi A, Roshan TM, Kahawita TM, Mason AB, Sheftel AD, Ponka P. Erythroid cell mitochondria receive endosomal iron by a "kiss-and-run" mechanism. Biochim Biophys Acta. 2016; 1863(12):2859-2867. https://doi.org/10.1016/j.bbamcr.2016.09.008Google Scholar
- Shaw GC, Cope JJ, Li L. Mitoferrin is essential for erythroid iron assimilation. Nature. 2006; 440(7080):96-100. PubMedhttps://doi.org/10.1038/nature04512Google Scholar
- Arosio P, Carmona F, Gozzelino R, Maccarinelli F, Poli M. The importance of eukaryotic ferritins in iron handling and cytoprotection. Biochem J. 2015; 472(1):1-15. PubMedhttps://doi.org/10.1042/BJ20150787Google Scholar
- Vanoaica L, Darshan D, Richman L, Schumann K, Kuhn LC. Intestinal ferritin H is required for an accurate control of iron absorption. Cell Metab. 2010; 12(3):273-282. PubMedhttps://doi.org/10.1016/j.cmet.2010.08.003Google Scholar
- Castiglioni E, Soriani N, Girelli D. High resolution melting for the identification of mutations in the iron responsive element of the ferritin light chain gene. Clin Chem Lab Med. 2010; 48(10):1415-1418. PubMedGoogle Scholar
- Luscieti S, Santambrogio P, Langlois d’Estaintot B. Mutant ferritin L-chains that cause neurodegeneration act in a dominant-negative manner to reduce ferritin iron incorporation. J Biol Chem. 2010; 285(16):11948-11957. PubMedhttps://doi.org/10.1074/jbc.M109.096404Google Scholar
- Truman-Rosentsvit M, Berenbaum D, Spektor L. Ferritin is secreted via 2 distinct nonclassical vesicular pathways. Blood. 2018; 131(3):342-352. PubMedhttps://doi.org/10.1182/blood-2017-02-768580Google Scholar
- Leidgens S, Bullough KZ, Shi H. Each member of the poly-r(C)-binding protein 1 (PCBP) family exhibits iron chaperone activity toward ferritin. J Biol Chem. 2013; 288(24):17791-17802. PubMedhttps://doi.org/10.1074/jbc.M113.460253Google Scholar
- Ryu MS, Zhang D, Protchenko O, Shakoury-Elizeh M, Philpott CC. PCBP1 and NCOA4 regulate erythroid iron storage and heme biosynthesis. J Clin Invest. 2017; 127(5):1786-1797. Google Scholar
- Mancias JD, Wang X, Gygi SP, Harper JW, Kimmelman AC. Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature. 2014; 509(7498):105-109. PubMedhttps://doi.org/10.1038/nature13148Google Scholar
- Dowdle WE, Nyfeler B, Nagel J. Selective VPS34 inhibitor blocks autophagy and uncovers a role for NCOA4 in ferritin degradation and iron homeostasis in vivo. Nat Cell Biol. 2014; 16(11):1069-1079. PubMedhttps://doi.org/10.1038/ncb3053Google Scholar
- Nandal A, Ruiz JC, Subramanian P. Activation of the HIF prolyl hydroxylase by the iron chaperones PCBP1 and PCBP2. Cell Metab. 2011; 14(5):647-657. PubMedhttps://doi.org/10.1016/j.cmet.2011.08.015Google Scholar
- Lakhal-Littleton S, Wolna M, Carr CA. Cardiac ferroportin regulates cellular iron homeostasis and is important for cardiac function. Proc Natl Acad Sci U S A. 2015; 112(10):3164-3169. PubMedhttps://doi.org/10.1073/pnas.1422373112Google Scholar
- Zhang DL, Ghosh MC, Ollivierre H, Li Y, Rouault TA. Ferroportin deficiency in erythroid cells causes serum iron deficiency and promotes hemolysis due to oxidative stress. Blood. 2018; 132(19):2078-2087. https://doi.org/10.1182/blood-2018-99-116276Google Scholar
- Zhang DL, Wu J, Shah BN. Erythrocytic ferroportin reduces intracellular iron accumulation, hemolysis, and malaria risk. Science. 2018; 359(6383):1520-1523. PubMedhttps://doi.org/10.1126/science.aal2022Google Scholar
- Keel SB, Doty RT, Yang Z. A heme export protein is required for red blood cell differentiation and iron homeostasis. Science. 2008; 319(5864):825-828. PubMedhttps://doi.org/10.1126/science.1151133Google Scholar
- Chiabrando D, Marro S, Mercurio S. The mitochondrial heme exporter FLVCR1b mediates erythroid differentiation. J Clin Invest. 2012; 122(12):4569-4579. PubMedhttps://doi.org/10.1172/JCI62422Google Scholar
- Zhang DL, Ghosh MC, Rouault TA. The physiological functions of iron regulatory proteins in iron homeostasis - an update. Front Pharmacol. 2014; 5:124. PubMedhttps://doi.org/10.3389/fphar.2014.00124Google Scholar
- Costain G, Ghosh MC, Maio N. Absence of iron-responsive element-binding protein 2 causes a novel neurodegenerative syndrome. Brain. 2019; 142(5):1195-1202. Google Scholar
- Galy B, Ferring-Appel D, Becker C. Iron regulatory proteins control a mucosal block to intestinal iron absorption. Cell Rep. 2013; 3(3):844-857. PubMedhttps://doi.org/10.1016/j.celrep.2013.02.026Google Scholar
- Zhang DL, Hughes RM, Ollivierre-Wilson H, Ghosh MC, Rouault TA. A ferroportin transcript that lacks an iron-responsive element enables duodenal and erythroid precursor cells to evade translational repression. Cell Metab. 2009; 9(5):461-473. PubMedhttps://doi.org/10.1016/j.cmet.2009.03.006Google Scholar
- Mancias JD, Pontano Vaites L, Nissim S. Ferritinophagy via NCOA4 is required for erythropoiesis and is regulated by iron dependent HERC2-mediated proteolysis. Elife. 2015; 4Google Scholar
- Bellelli R, Castellone MD, Guida T. NCOA4 transcriptional coactivator inhibits activation of DNA replication origins. Mol Cell. 2014; 55(1):123-137. PubMedhttps://doi.org/10.1016/j.molcel.2014.04.031Google Scholar
- Bellelli R, Federico G, Matte A. NCOA4 deficiency impairs systemic iron homeostasis. Cell Rep. 2016; 14(3):411-421. https://doi.org/10.1016/j.celrep.2015.12.065Google Scholar
- Gao X, Lee HY, Li W. Thyroid hormone receptor beta and NCOA4 regulate terminal erythrocyte differentiation. Proc Natl Acad Sci U S A. 2017; 114(38):10107-10112. PubMedhttps://doi.org/10.1073/pnas.1711058114Google Scholar
- Aschemeyer S, Qiao B, Stefanova D. Structure-function analysis of ferroportin defines the binding site and an alternative mechanism of action of hepcidin. Blood. 2018; 131(8):899-910. PubMedhttps://doi.org/10.1182/blood-2017-05-786590Google Scholar
- Kautz L, Meynard D, Monnier A. Iron regulates phosphorylation of Smad1/5/8 and gene expression of Bmp6, Smad7, Id1, and Atoh8 in the mouse liver. Blood. 2008; 112(4):1503-1509. PubMedhttps://doi.org/10.1182/blood-2008-03-143354Google Scholar
- Lim PJ, Duarte TL, Arezes J. Nrf2 controls iron homeostasis in haemochromatosis and thalassaemia via Bmp6 and hepcidin. Nat Metab. 2019; 1(5):519-531. Google Scholar
- Koch PS, Olsavszky V, Ulbrich F. Angiocrine Bmp2 signaling in murine liver controls normal iron homeostasis. Blood. 2017; 129(4):415-419. PubMedhttps://doi.org/10.1182/blood-2016-07-729822Google Scholar
- Canali S, Wang CY, Zumbrennen-Bullough KB, Bayer A, Babitt JL. Bone morphogenetic protein 2 controls iron homeostasis in mice independent of Bmp6. Am J Hematol. 2017; 92(11):1204-1213. PubMedhttps://doi.org/10.1002/ajh.24888Google Scholar
- Theurl I, Schroll A, Sonnweber T. Pharmacologic inhibition of hepcidin expression reverses anemia of chronic inflammation in rats. Blood. 2011; 118(18):4977-4984. PubMedhttps://doi.org/10.1182/blood-2011-03-345066Google Scholar
- Girelli D, Nemeth E, Swinkels DW. Hepcidin in the diagnosis of iron disorders. Blood. 2016; 127(23):2809-2813. PubMedhttps://doi.org/10.1182/blood-2015-12-639112Google Scholar
- Du X, She E, Gelbart T. The serine protease TMPRSS6 is required to sense iron deficiency. Science. 2008; 320(5879):1088-1092. PubMedhttps://doi.org/10.1126/science.1157121Google Scholar
- Silvestri L, Pagani A, Nai A, De Domenico I, Kaplan J, Camaschella C. The serine protease matriptase-2 (TMPRSS6) inhibits hep-cidin activation by cleaving membrane hemojuvelin. Cell Metab. 2008; 8(6):502-511. PubMedhttps://doi.org/10.1016/j.cmet.2008.09.012Google Scholar
- Finberg KE, Heeney MM, Campagna DR. Mutations in TMPRSS6 cause iron-refractory iron deficiency anemia (IRIDA). Nat Genet. 2008; 40(5):569-571. PubMedhttps://doi.org/10.1038/ng.130Google Scholar
- Colucci S, Pagani A, Pettinato M. The immunophilin FKBP12 inhibits hepcidin expression by binding the BMP type I receptor ALK2 in hepatocytes. Blood. 2017; 130(19):2111-2120. PubMedhttps://doi.org/10.1182/blood-2017-04-780692Google Scholar
- Kautz L, Jung G, Valore EV, Rivella S, Nemeth E, Ganz T. Identification of erythro-ferrone as an erythroid regulator of iron metabolism. Nat Genet. 2014; 46(7):678-684. PubMedhttps://doi.org/10.1038/ng.2996Google Scholar
- Silvestri L, Pagani A, Camaschella C. Furin-mediated release of soluble hemojuvelin: a new link between hypoxia and iron homeostasis. Blood. 2008; 111(2):924-931. PubMedhttps://doi.org/10.1182/blood-2007-07-100677Google Scholar
- Sonnweber T, Nachbaur D, Schroll A. Hypoxia induced downregulation of hepcidin is mediated by platelet derived growth factor BB. Gut. 2014; 63(12):1951-1959. PubMedhttps://doi.org/10.1136/gutjnl-2013-305317Google Scholar
- Theurl I, Theurl M, Seifert M. Autocrine formation of hepcidin induces iron retention in human monocytes. Blood. 2008; 111(4):2392-2399. PubMedhttps://doi.org/10.1182/blood-2007-05-090019Google Scholar
- Malerba M, Louis S, Cuvellier S. Epidermal hepcidin is required for neutrophil response to bacterial infection. J Clin Invest. 2019. Google Scholar
- Lakhal-Littleton S, Wolna M, Chung YJ. An essential cell-autonomous role for hepcidin in cardiac iron homeostasis. Elife. 2016; 5Google Scholar
- Benyamin B, Esko T, Ried JS. Novel loci affecting iron homeostasis and their effects in individuals at risk for hemochromatosis. Nat Commun. 2014; 5:4926. PubMedhttps://doi.org/10.1038/ncomms5926Google Scholar
- Bekri S, Gual P, Anty R. Increased adipose tissue expression of hepcidin in severe obesity is independent from diabetes and NASH. Gastroenterology. 2006; 131(3):788-796. PubMedhttps://doi.org/10.1053/j.gastro.2006.07.007Google Scholar
- Vecchi C, Montosi G, Garuti C. Gluconeogenic signals regulate iron homeostasis via hepcidin in mice. Gastroenterology. 2014; 146(4):1060-1069. PubMedhttps://doi.org/10.1053/j.gastro.2013.12.016Google Scholar
- Folgueras AR, Freitas-Rodriguez S, Ramsay AJ. Matriptase-2 deficiency protects from obesity by modulating iron homeostasis. Nat Commun. 2018; 9(1):1350. Google Scholar
- Mastrogiannaki M, Matak P, Peyssonnaux C. The gut in iron homeostasis: role of HIF-2 under normal and pathological conditions. Blood. 2013; 122(6):885-892. PubMedhttps://doi.org/10.1182/blood-2012-11-427765Google Scholar
- Schwartz AJ, Das NK, Ramakrishnan SK. Hepatic hepcidin/intestinal HIF-2alpha axis maintains iron absorption during iron deficiency and overload. J Clin Invest. 2019; 129(1):336-348. PubMedGoogle Scholar
- Ghosh MC, Zhang DL, Jeong SY. Deletion of iron regulatory protein 1 causes polycythemia and pulmonary hypertension in mice through translational derepression of HIF-2alpha. Cell Metab. 2013; 17(2):271-281. PubMedhttps://doi.org/10.1016/j.cmet.2012.12.016Google Scholar
- Anderson SA, Nizzi CP, Chang YI. The IRP1-HIF-2alpha axis coordinates iron and oxygen sensing with erythropoiesis and iron absorption. Cell Metab. 2013; 17(2):282-290. PubMedhttps://doi.org/10.1016/j.cmet.2013.01.007Google Scholar
- Bullock GC, Delehanty LL, Talbot AL. Iron control of erythroid development by a novel aconitase-associated regulatory pathway. Blood. 2010; 116(1):97-108. PubMedhttps://doi.org/10.1182/blood-2009-10-251496Google Scholar
- Richardson CL, Delehanty LL, Bullock GC. Isocitrate ameliorates anemia by suppressing the erythroid iron restriction response. J Clin Invest. 2013; 123(8):3614-3623. PubMedhttps://doi.org/10.1172/JCI68487Google Scholar
- Nai A, Lidonnici MR, Rausa M. The second transferrin receptor regulates red blood cell production in mice. Blood. 2015; 125(7):1170-1179. PubMedhttps://doi.org/10.1182/blood-2014-08-596254Google Scholar
- Camaschella C, Roetto A, Cali A. The gene TFR2 is mutated in a new type of haemochromatosis mapping to 7q22. Nat Genet. 2000; 25(1):14-15. PubMedhttps://doi.org/10.1038/75534Google Scholar
- Rauner M, Baschant U, Roetto A. Transferrin receptor 2 controls bone mass and pathological bone formation via BMP and Wnt signaling. Nat Metab. 2019; 1(1):111-124. Google Scholar
- Finch CA. Erythropoiesis, erythropoietin, and iron. Blood. 1982; 60(6):1241-1246. PubMedGoogle Scholar
- Arezes J, Foy N, McHugh K. Erythroferrone inhibits the induction of hepcidin by BMP6. Blood. 2018; 132(14):1473-1477. PubMedhttps://doi.org/10.1182/blood-2018-06-857995Google Scholar
- Nai A, Rubio A, Campanella A. Limiting hepatic Bmp-Smad signaling by matriptase-2 is required for erythropoietin-mediated hepcidin suppression in mice. Blood. 2016; 127(19):2327-2336. PubMedhttps://doi.org/10.1182/blood-2015-11-681494Google Scholar
- Kautz L, Jung G, Du X. Erythroferrone contributes to hepcidin suppression and iron overload in a mouse model of beta-tha-lassemia. Blood. 2015; 126(17):2031-2037. PubMedhttps://doi.org/10.1182/blood-2015-07-658419Google Scholar
- Anker SD, Comin Colet J, Filippatos G. Investigators F-HT. Ferric carboxymaltose in patients with heart failure and iron deficiency. N Engl J Med. 2009; 361(25):2436-2448. PubMedhttps://doi.org/10.1056/NEJMoa0908355Google Scholar
- Feder JN, Gnirke A, Thomas W. A novel MHC class I-like gene is mutated in patients with hereditary haemochromatosis. Nat Genet. 1996; 13(4):399-408. PubMedhttps://doi.org/10.1038/ng0896-399Google Scholar
- Brissot P, Pietrangelo A, Adams PC, de Graaff B, McLaren CE, Loreal O. Haemochromatosis. Nat Rev Dis Primers. 2018; 4:18016. Google Scholar
- Le Lan C, Loreal O, Cohen T. Redox active plasma iron in C282Y/C282Y hemochromatosis. Blood. 2005; 105(11):4527-4531. PubMedhttps://doi.org/10.1182/blood-2004-09-3468Google Scholar
- Sandhu K, Flintoff K, Chatfield MD. Phenotypic analysis of hemochromatosis subtypes reveals variations in severity of iron overload and clinical disease. Blood. 2018; 132(1):101-110. PubMedhttps://doi.org/10.1182/blood-2018-02-830562Google Scholar
- McDonald J, Wooderchak-Donahue W, VanSant Webb C, Whitehead K, Stevenson DA, Bayrak-Toydemir P. Hereditary hemorrhagic telangiectasia: genetics and molecular diagnostics in a new era. Front Genet. 2015; 6:1. PubMedhttps://doi.org/10.3389/fgene.2015.00001Google Scholar
- Pietrangelo A. Ferroportin disease: pathogenesis, diagnosis and treatment. Haematologica. 2017; 102(12):1972-1984. PubMedhttps://doi.org/10.3324/haematol.2017.170720Google Scholar
- Jenkitkasemwong S, Wang CY, Coffey R. SLC39A14 is required for the development of hepatocellular iron overload in murine models of hereditary hemochromatosis. Cell Metab. 2015; 22(1):138-150. PubMedhttps://doi.org/10.1016/j.cmet.2015.05.002Google Scholar
- Oudit GY, Sun H, Trivieri MG. L-type Ca2+ channels provide a major pathway for iron entry into cardiomyocytes in iron-overload cardiomyopathy. Nat Med. 2003; 9(9):1187-1194. PubMedhttps://doi.org/10.1038/nm920Google Scholar
- Weiss G, Ganz T, Goodnough LT. Anemia of inflammation. Blood. 2019; 133(1):40-50. PubMedhttps://doi.org/10.1182/blood-2018-06-856500Google Scholar
- Camaschella C. How I manage patients with atypical microcytic anaemia. Br J Haematol. 2013; 160(1):12-24. PubMedhttps://doi.org/10.1111/bjh.12081Google Scholar
- Nai A, Pagani A, Silvestri L. TMPRSS6 rs855791 modulates hepcidin transcription in vitro and serum hepcidin levels in normal individuals. Blood. 2011; 118(16):4459-4462. PubMedhttps://doi.org/10.1182/blood-2011-06-364034Google Scholar
- Kiss JE, Vassallo RR. How do we manage iron deficiency after blood donation?. Br J Haematol. 2018; 181(5):590-603. Google Scholar
- Guernsey DL, Jiang H, Campagna DR. Mutations in mitochondrial carrier family gene SLC25A38 cause nonsyndromic auto-somal recessive congenital sideroblastic ane-mia. Nat Genet. 2009; 41(6):651-653. PubMedhttps://doi.org/10.1038/ng.359Google Scholar
- Camaschella C, Campanella A, De Falco L. The human counterpart of zebrafish shiraz shows sideroblastic-like microcytic anemia and iron overload. Blood. 2007; 110(4):1353-1358. PubMedhttps://doi.org/10.1182/blood-2007-02-072520Google Scholar
- Schmitz-Abe K, Ciesielski SJ, Schmidt PJ. Congenital sideroblastic anemia due to mutations in the mitochondrial HSP70 homologue HSPA9. Blood. 2015; 126(25):2734-2738. PubMedhttps://doi.org/10.1182/blood-2015-09-659854Google Scholar
- Ducamp S, Fleming MD. The molecular genetics of sideroblastic anemia. Blood. 2019; 133(1):59-69. PubMedhttps://doi.org/10.1182/blood-2018-08-815951Google Scholar
- Chakraborty PK, Schmitz-Abe K, Kennedy EK. Mutations in TRNT1 cause congenital sideroblastic anemia with immunodeficiency, fevers, and developmental delay (SIFD). Blood. 2014; 124(18):2867-2871. PubMedhttps://doi.org/10.1182/blood-2014-08-591370Google Scholar
- Disease GBD, Injury I, Prevalence C. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet. 2017; 390(10100):1211-1259. PubMedhttps://doi.org/doi:10.1016/S0140-6736(17)32154-2Google Scholar
- Camaschella C. Iron-deficiency anemia. N Engl J Med. 2015; 372(19):1832-1843. PubMedhttps://doi.org/10.1056/NEJMra1401038Google Scholar
- Lopez A, Cacoub P, Macdougall IC, Peyrin-Biroulet L. Iron deficiency anaemia. Lancet. 2016; 387(10021):907-916. PubMedhttps://doi.org/10.1016/S0140-6736(15)60865-0Google Scholar
- Camaschella C. Iron deficiency. Blood. 2019; 133(1):30-39. PubMedhttps://doi.org/10.1182/blood-2018-05-815944Google Scholar
- Ganz T. Anemia of inflammation. N Engl J Med. 2019; 381(12):1148-1157. Google Scholar
- Weiss G, Goodnough LT. Anemia of chronic disease. N Engl J Med. 2005; 352(10):1011-1023. PubMedhttps://doi.org/10.1056/NEJMra041809Google Scholar
- Drakesmith H, Prentice AM. Hepcidin and the iron-infection axis. Science. 2012; 338(6108):768-772. PubMedhttps://doi.org/10.1126/science.1224577Google Scholar
- Nairz M, Dichtl S, Schroll A. Iron and innate antimicrobial immunity-Depriving the pathogen, defending the host. J Trace Elem Med Biol. 2018; 48:118-133. https://doi.org/10.1016/j.jtemb.2018.03.007Google Scholar
- Hood MI, Skaar EP. Nutritional immunity: transition metals at the pathogen-host interface. Nat Rev Microbiol. 2012; 10(8):525-537. PubMedhttps://doi.org/10.1038/nrmicro2836Google Scholar
- Stefanova D, Raychev A, Arezes J. Endogenous hepcidin and its agonist mediate resistance to selected infections by clearing non-transferrin-bound iron. Blood. 2017; 130(3):245-257. PubMedhttps://doi.org/10.1182/blood-2017-03-772715Google Scholar
- Song SN, Tomosugi N, Kawabata H, Ishikawa T, Nishikawa T, Yoshizaki K. Down-regulation of hepcidin resulting from long-term treatment with an anti-IL-6 receptor antibody (tocilizumab) improves anemia of inflammation in multicentric Castleman disease. Blood. 2010; 116(18):3627-3634. PubMedhttps://doi.org/10.1182/blood-2010-03-271791Google Scholar
- Weinstein DA, Roy CN, Fleming MD, Loda MF, Wolfsdorf JI, Andrews NC. Inappropriate expression of hepcidin is associated with iron refractory anemia: implications for the anemia of chronic disease. Blood. 2002; 100(10):3776-3781. PubMedhttps://doi.org/10.1182/blood-2002-04-1260Google Scholar
- Ganz T, Jung G, Naeim A. Immunoassay for human serum erythroferrone. Blood. 2017; 130(10):1243-1246. PubMedhttps://doi.org/10.1182/blood-2017-04-777987Google Scholar
- Rivella S. Iron metabolism under conditions of ineffective erythropoiesis in beta-tha-lassemia. Blood. 2019; 133(1):51-58. PubMedhttps://doi.org/10.1182/blood-2018-07-815928Google Scholar
- Camaschella C, Nai A. Ineffective erythropoiesis and regulation of iron status in iron loading anaemias. Br J Haematol. 2016; 172(4):512-523. Google Scholar
- Papaemmanuil E, Cazzola M, Boultwood J. Chronic Myeloid Disorders Working Group of the International Cancer Genome C. Somatic SF3B1 mutation in myelodysplasia with ring sideroblasts. N Engl J Med. 2011; 365(15):1384-1395. PubMedhttps://doi.org/10.1056/NEJMoa1103283Google Scholar
- Bondu S, Alary AS, Lefevre C. A variant erythroferrone disrupts iron homeostasis in SF3B1-mutated myelodysplastic syndrome. Sci Transl Med. 2019; 11(500)Google Scholar
- Taher AT, Weatherall DJ, Cappellini MD. Thalassaemia. Lancet. 2018; 391(10116):155-167. Google Scholar
- Heeney MM, Guo D, De Falco L. Normalizing hepcidin predicts TMPRSS6 mutation status in patients with chronic iron deficiency. Blood. 2018; 132(4):448-452. PubMedhttps://doi.org/10.1182/blood-2017-03-773028Google Scholar
- Traglia M, Girelli D, Biino G. Association of HFE and TMPRSS6 genetic variants with iron and erythrocyte parameters is only in part dependent on serum hepcidin concentrations. J Med Genet. 2011; 48(9):629-634. PubMedhttps://doi.org/10.1136/jmedgenet-2011-100061Google Scholar
- Galesloot TE, Vermeulen SH, Geurts-Moespot AJ. Serum hepcidin: reference ranges and biochemical correlates in the general population. Blood. 2011; 117(25):e218-225. PubMedhttps://doi.org/10.1182/blood-2011-02-337907Google Scholar
- Bregman DB, Morris D, Koch TA, He A, Goodnough LT. Hepcidin levels predict non-responsiveness to oral iron therapy in patients with iron deficiency anemia. Am J Hematol. 2013; 88(2):97-101. PubMedhttps://doi.org/10.1002/ajh.23354Google Scholar
- Prentice AM, Doherty CP, Abrams SA. Hepcidin is the major predictor of erythrocyte iron incorporation in anemic African children. Blood. 2012; 119(8):1922-1928. PubMedhttps://doi.org/10.1182/blood-2011-11-391219Google Scholar
- Stoffel NU, Zeder C, Brittenham GM, Moretti D, Zimmermann MB. Iron absorption from supplements is greater with alternate day than with consecutive day dosing in iron-deficient anemic women. Haematologica. 2019. Google Scholar
- Hershko C, Camaschella C. How I treat unexplained refractory iron deficiency ane-mia. Blood. 2014; 123(3):326-333. PubMedhttps://doi.org/10.1182/blood-2013-10-512624Google Scholar
- Moretti D, Goede JS, Zeder C. Oral iron supplements increase hepcidin and decrease iron absorption from daily or twice-daily doses in iron-depleted young women. Blood. 2015; 126(17):1981-1929. PubMedhttps://doi.org/10.1182/blood-2015-05-642223Google Scholar
- Stoffel NU, Cercamondi CI, Brittenham G. Iron absorption from oral iron supplements given on consecutive versus alternate days and as single morning doses versus twice-daily split dosing in iron-depleted women: two open-label, randomised controlled trials. Lancet Haematol. 2017; 4(11):e524-e533. Google Scholar
- Auerbach M, Deloughery T. Single-dose intravenous iron for iron deficiency: a new paradigm. Hematology Am Soc Hematol Educ Program. 2016; 2016(1):57-66. PubMedhttps://doi.org/10.1182/asheducation-2016.1.57Google Scholar
- Casu C, Chessa R, Liu A. Minihepcidins improve ineffective erythropoiesis and splenomegaly in a new mouse model of adult beta-thalassemia major. Haematologica. 2019. Google Scholar
- Casu C, Nemeth E, Rivella S. Hepcidin agonists as therapeutic tools. Blood. 2018; 131(16):1790-1794. PubMedhttps://doi.org/10.1182/blood-2017-11-737411Google Scholar
- Li H, Rybicki AC, Suzuka SM. Transferrin therapy ameliorates disease in beta-thalassemic mice. Nat Med. 2010; 16(2):177-182. PubMedhttps://doi.org/10.1038/nm.2073Google Scholar
- Gelderman MP, Baek JH, Yalamanoglu AP. Reversal of hemochromatosis by apo-transferrin in non-transfused and transfused Hbbth3/+ (heterozygous B1/B2 globin gene deletion) mice. Haematologica. 2015; 100(5):611-622. PubMedhttps://doi.org/10.3324/haematol.2014.117325Google Scholar
- Li H, Choesang T, Bao W. Decreasing TfR1 expression reverses anemia and hep-cidin suppression in β-thalassemic mice. Blood. 2017; 129(11):1514-1526. PubMedhttps://doi.org/10.1182/blood-2016-09-742387Google Scholar
- Artuso I, Lidonnici MR, Altamura S. Transferrin receptor 2 is a potential novel therapeutic target for β-thalassemia: evidence from a murine model. Blood. 2018; 132(21):2286-2297. PubMedhttps://doi.org/10.1182/blood-2018-05-852277Google Scholar
- Wang X, Zhang M, Flores SRL. Oral gavage of ginger nanoparticle-derived lipid vectors carrying Dmt1 siRNA blunts iron loading in murine hereditary hemochromatosis. Mol Ther. 2019; 27(3):493-506. Google Scholar
- Dussiot M, Maciel TT, Fricot A. An activin receptor IIA ligand trap corrects ineffective erythropoiesis in β-thalassemia. Nat Med. 2014; 20(4):398-407. PubMedhttps://doi.org/10.1038/nm.3468Google Scholar
- Piga A, Perrotta S, Gamberini MR. Luspatercept improves hemoglobin levels and blood transfusion requirements in a study of patients with β-thalassemia. Blood. 2019; 133(12):1279-1289. PubMedhttps://doi.org/10.1182/blood-2018-10-879247Google Scholar
- Platzbecker U, Germing U, Gotze KS. Luspatercept for the treatment of anaemia in patients with lower-risk myelodysplastic syndromes (PACE-MDS): a multicentre, open-label phase 2 dose-finding study with long-term extension study. Lancet Oncol. 2017; 18(10):1338-1347. https://doi.org/10.1016/S1470-2045(17)30615-0Google Scholar
- Liu J, Liu W, Liu Y. New thiazolidi-nones reduce iron overload in mouse models of hereditary hemochromatosis and β-thalassemia. Haematologica. 2019; 104(9):1768-1781. PubMedhttps://doi.org/10.3324/haematol.2018.209874Google Scholar
- Vanclooster A, van Deursen C, Jaspers R, Cassiman D, Koek G. Proton pump inhibitors decrease phlebotomy need in HFE hemochromatosis: double-blind randomized placebo-controlled trial. Gastroenterology. 2017; 153(3):678-680.e2. Google Scholar
- Crielaard BJ, Lammers T, Rivella S. Targeting iron metabolism in drug discovery and delivery. Nat Rev Drug Discov. 2017; 16(6):400-423. PubMedhttps://doi.org/10.1038/nrd.2016.248Google Scholar
- Chen N, Hao C, Peng X. Roxadustat for anemia in patients with kidney disease not receiving dialysis. N Engl J Med. 2019; 381(11):1001-1010. Google Scholar
- Xavier-Ferrucio J, Scanlon V, Li X. Low iron promotes megakaryocytic commitment of megakaryocytic-erythroid progenitors in humans and mice. Blood. 2019; 134(18):1547-1557. Google Scholar
- Bordini J, Galvan S, Ponzoni M. Induction of iron excess restricts malignant plasma cells expansion and potentiates bortezomib effect in models of multiple myeloma. Leukemia. 2017; 31(4):967-970. Google Scholar
- Guo S, Casu C, Gardenghi S. Reducing TMPRSS6 ameliorates hemochromatosis and β-thalassemia in mice. J Clin Invest. 2013; 123(4):1531-1541. PubMedhttps://doi.org/10.1172/JCI66969Google Scholar
- Garcia-Santos D, Hamdi A, Saxova Z. Inhibition of heme oxygenase ameliorates anemia and reduces iron overload in a β-tha-lassemia mouse model. Blood. 2018; 131(2):236-246. PubMedhttps://doi.org/10.1182/blood-2017-07-798728Google Scholar
- Sheetz M, Barrington P, Callies S. Targeting the hepcidin-ferroportin pathway in anaemia of chronic kidney disease. Br J Clin Pharmacol. 2019; 85(5):935-948. Google Scholar
- Asshoff M, Petzer V, Warr MR. Momelotinib inhibits ACVR1/ALK2, decreases hepcidin production, and ameliorates anemia of chronic disease in rodents. Blood. 2017; 129(13):1823-1830. PubMedhttps://doi.org/10.1182/blood-2016-09-740092Google Scholar
- Kovac S, Boser P, Cui Y. Anti-hemoju-velin antibody corrects anemia caused by inappropriately high hepcidin levels. Haematologica. 2016; 101(5):e173-176. PubMedhttps://doi.org/10.3324/haematol.2015.140772Google Scholar
- Poli M, Asperti M, Naggi A. Glycol-split nonanticoagulant heparins are inhibitors of hepcidin expression in vitro and in vivo. Blood. 2014; 123(10):1564-1573. PubMedhttps://doi.org/10.1182/blood-2013-07-515221Google Scholar
- Vadhan-Raj S, Abonour R, Goldman JW. A first-in-human phase 1 study of a hep-cidin monoclonal antibody, LY2787106, in cancer-associated anemia. J Hematol Oncol. 2017; 10(1):73. Google Scholar
- Boyce M, Warrington S, Cortezi B. Safety, pharmacokinetics and pharmacodynamics of the anti-hepcidin Spiegelmer lexaptepid pegol in healthy subjects. Br J Pharmacol. 2016; 173(10):1580-1588. Google Scholar
- Hohlbaum AM, Gille H, Trentmann S. Sustained plasma hepcidin suppression and iron elevation by anticalin-derived hepcidin antagonist in cynomolgus monkey. Br J Pharmacol. 2018; 175(7):1054-1065. Google Scholar
- Angmo S, Tripathi N, Abbat S. Identification of guanosine 5’-diphosphate as potential iron mobilizer: preventing the hepcidin-ferroportin interaction and modulating the interleukin-6/Stat-3 pathway. Sci Rep. 2017; 7:40097. Google Scholar
- Silvestri L, Nai A, Dulja A, Pagani A. Hepcidin and the BMP-SMAD pathway: an unexpected liaison. Vitam Horm. 2019; 110:71-99. Google Scholar
- Latour C, Besson-Fournier C, Meynard D. Differing impact of the deletion of hemochromatosis-associated molecules HFE and transferrin receptor-2 on the iron phenotype of mice lacking bone morpho- genetic protein 6 or hemojuvelin. Hepatology. 2016; 63(1):126-137. PubMedhttps://doi.org/10.1002/hep.28254Google Scholar
- Zhao N, Nizzi CP, Andrerson SA. Low intracellular iron increases the stability of matriptase-2. J Biol Chem. 2015; 290(7):4432-4446. PubMedhttps://doi.org/10.1074/jbc.M114.611913Google Scholar
- Schmidt PJ, Toran PT, Giannetti AM, Bjorkman PJ, Andrews NC. The transferrin receptor modulates Hfe-dependent regulation of hepcidin expression. Cell Metab. 2008; 7(3):205-214. PubMedhttps://doi.org/10.1016/j.cmet.2007.11.016Google Scholar
- Pasricha SR, Lim PJ, Duarte TL. Hepcidin is regulated by promoter-associated histone acetylation and HDAC3. Nat Commun. 2017; 8(1):403. Google Scholar
- Artuso I, Pettinato M, Nai A. Transient decrease of serum iron after acute erythro-poietin treatment contributes to hepcidin inhibition by ERFE in mice. Haematologica. 2019; 104(3):e87-e90. PubMedhttps://doi.org/10.3324/haematol.2018.199810Google Scholar
- Xiao X, Dev S, Canali S. Endothelial Bmp2 knockout exacerbates hemochro-matosis in Hfe knockout mice but not Bmp6 knockout mice. Hepatology. 2019. Google Scholar
- Wang CY, Xu Y, Traeger L. Erythroferrone lowers hepcidin by sequestering BMP2/6 heterodimer from binding to the BMP type I receptor ALK3. Blood. 2019. Google Scholar
- Manolova V, Nyffenegger N, Flace A. Oral ferroportin inhibitor ameliorates ineffective erythropoiesis in a model of β-tha-lassemia. J Clin Invest. 2019; 130(1):491-506. Google Scholar