Iron is an ubiquitous metal of vital importance to the normal physiologic processes of many organisms. Over the last 2 decades, the discovery of mutations in genes leading to hereditary disorders of iron overload, iron deficiency, and iron maldistribution have accelerated our understanding of human iron homeostasis. This chapter provides an updated overview of the human iron cycle, regulation of iron homeostasis, and how perturbations in these homeostatic mechanisms lead to iron overload disease and provides strategies for the diagnosis of hereditary iron overload.

Learning Objectives
  • To appreciate the clinical and molecular modifiers of iron homeostasis

  • To develop an approach to the diagnosis of hereditary hemochromatosis

Iron is an essential metal for many biological processes due to its ability to transfer electrons in reduction/oxidation reactions. This reactivity also provides the potential for great damage to biological systems if iron is not chaperoned through a tightly regulated network of iron-binding proteins and transporters. If the capacity of these iron-binding proteins is exceeded, “free” iron is capable of forming reactive oxygen species that may damage macromolecular cellular components such as nucleic acids, proteins, and lipids and lead to tissue damage. Iron homeostasis in humans is maintained almost exclusively at the level of intestinal absorption because evolution has not provided a physiologically regulated mechanism for iron excretion.

Iron recycling and storage

The primary role of iron in mammals is to provide a binding site for oxygen in the heme moiety of hemoglobin. The average adult male has a total body iron content of ∼4 g, ∼2/3 of which is contained in the erythroid compartment. Under normal physiologic conditions, only 1-2 mg of the daily 20-25 mg of iron required to maintain erythropoiesis enters the body through carefully regulated intestinal absorption.1  Therefore, most iron required for the developing erythroid precursors in the BM is derived from the recycling of heme iron through the reticuloendothelial macrophage phagocytosis of senescent RBCs.

Most nonerythroid iron, 0.5–1 g in adult males, is stored in hepatocytes and macrophages and is easily mobilized for erythropoiesis. When iron stores are depleted through decreased dietary intake or intestinal absorption, increased requirements (eg, pregnancy) or loss (eg, intestinal blood loss), erythropoiesis becomes iron restricted and eventually results in the characteristic microcytic, hypochromic anemia of iron deficiency. In contrast, erythropoiesis in most iron overload states is relatively unperturbed; however, HFE hemochromatosis is associated with increases in hemoglobin, mean corpuscular volume, mean corpuscular hemoglobin, and mean corpuscular hemoglobin concentration that may be related to increased iron uptake and hemoglobin production in erythroid precursors.2  The excess iron absorbed in hereditary hemochromatosis is deposited in hepatocytes, cardiac myocytes, and multiple endocrine organs. This excess cellular iron deposition ultimately exceeds the capacity of iron-binding proteins, thereby leading to cellular damage, organ dysfunction, and eventually to clinical symptoms.

Ferritin is the primary intracellular iron storage protein, which forms multimeric complexes that facilitate iron sequestration and mobilization depending on cellular need. In contrast, hemosiderin is an amorphous and poorly bioavailable iron-containing conglomerate the presence of which is generally a pathologic sign of cellular iron excess. A soluble form of ferritin is also found in the plasma and is expelled primarily from reticuloendothelial macrophages and the liver; however, its biological role remains unclear. Serum ferritin correlates with iron stores in many conditions; however, caution must be taken before interpreting in the setting of inflammation and cellular injury.3 

The large size and electrochemical properties of iron require that it be transported across the mammalian cell membranes by specific transmembrane proteins that mediate cellular iron uptake (import) and iron release (export). Although hepatocytes, intestinal mucosal cells, and macrophages possess specific carriers for iron import and export, erythroid cells only import iron and do not “release” iron until they are lysed or phagocytosed within the reticuloendothelial system.

Cellular iron import

Dietary nonheme iron, primarily in the ferric (Fe3+) state, is reduced to the ferrous (Fe2+) state in the acidic environment of the proximal duodenum by a brush border ferrireductase named Dcytb.4  Ferrous (Fe2+) iron is then cotransported with protons through the apical membrane of the duodenal enterocyte by divalent metal transporter 1 (DMT1).5  Some imported iron remains stored within the enterocyte cytoplasm as ferritin and the remainder is exported through the enterocyte basolateral membrane into the plasma via the transmembrane iron exporter ferroportin (FPN; described more fully in the Cellular iron export section).

Nonheme iron is imported into nonintestinal cell types from the plasma by the transferrin (TF) cycle (for review, see Chen and Paw6 ). Almost all plasma iron exists bound to the abundant glycoprotein TF. Each TF molecule binds 2 Fe3+ iron atoms with high affinity, thereby maintaining its solubility and nonreactivity and allowing it to circulate in a safe form. The TF cycle begins with iron-loaded plasma TF binding with high affinity to TF receptor 1 (TFR1) on the cell surface and being endocytosed. The endosome is then acidified, prompting the release of iron from TF and the empty TF and TFR1 return to the cell surface and are available to repeat the cycle again. The unbound Fe3+ iron in the acidified endosome is reduced and transported out of the endosome into the cytoplasm by DMT1.

Despite significant advances in our understanding of absorption and regulatory mechanisms of nonheme iron, the mediators of heme iron import, particularly during intestinal absorption, remains enigmatic (for review, see Yuan et al7  and Hamza and Korolnek8 ). It is unclear whether intestinal heme absorption uses membrane-bound transporters or endocytic uptake.8  The only membrane-bound heme importer characterized is the heme-responsive gene-1, (HRG-1), initially identified by homology to heme transporters found in Caenorhabditis elegans.9,10  HRG-1 localizes to the phagolysosomes of macrophages,9,11  suggesting a role in the import of heme during the recycling of senescent erythrocytes. Most intracellular heme will have its iron liberated by hemoxygenase, allowing storage in ferritin or export via FPN. However, cellular heme export has been postulated as a method of cellular detoxification and transfer of heme between cells.12  Feline Leukemia Virus sub-group C Receptor 1a (FLVCR1a), ATP-binding cassette transporter sub-family G member 2 (ABCG2),12  and, most recently, the multidrug resistance protein (MRP-5)13  have been implicated as heme exporters.

Cellular iron export

Some imported iron remains stored intracellularly as ferritin and the remainder is exported. The only known mammalian iron exporter is FPN,14,15  which is expressed at all sites involved in cellular Fe2+ iron export to the plasma, including the basolateral membranes of duodenal enterocyte, macrophages, hepatocytes, and in the placenta.16 

Cellular iron export is dependent on the copper-containing ferroxidases ceruloplasmin and hephaestin.17  These enzymes oxidize exported Fe2+ iron, returning it to its Fe3+ state and thus promoting its binding to plasma TF. Deficiencies of these ferroxidases appear to result in impaired iron export from the cells/tissue where they are expressed and appear to result in cellular iron accumulation.16 

Regulation of iron homeostasis

Iron homeostasis requires carefully coordinated regulation of intestinal iron absorption, cellular iron import/export, and iron storage. Because humans have no physiological mechanism for iron excretion, the control of intestinal iron absorption is the primary mechanism for determining overall iron balance. A small fraction of dietary iron is imported into the enterocyte and only a fraction is exported from the enterocyte into the plasma. Iron remaining within enterocytes is lost from the body through their physiologic sloughing into the gut lumen.

Intestinal iron absorption is regulated primarily through the expression of hepcidin, a circulating 25-aa hormone synthesized by the liver and filtered into the urine.18  Hepcidin acts as a negative regulator of cellular iron export from cells, thus limiting iron availability to the plasma. As a negative regulator, decreased hepcidin leads to enhanced intestinal absorption and macrophage iron release, elevated plasma iron levels, and iron loading.

Hepcidin exerts its central role in iron homeostasis through its effect on FPN, the only known receptor for hepcidin and cellular iron exporter. Hepcidin binds to FPN at the cell surface, resulting in endocytosis and lysosomal degradation,19  blocking of iron efflux into the plasma, hypoferremia, and iron-restricted erythropoiesis.

Clinically, plasma hepcidin concentration is modulated by several “regulators.”20  Hepcidin is increased by increased iron stores or inflammation and is decreased by hypoxia, increased erythropoietic activity, or testosterone.21  In the setting of conflicting influences on hepcidin expression, the negative regulation by erythropoietic activity is dominant (eg, non-transfusion-dependent thalassemia). The physiologic regulation of hepcidin expression is controlled at the level of transcription by a complex network resulting from the interaction of multiple proteins and cascades (Figure 1). Although a great deal remains to be elucidated regarding the molecular control of hepcidin expression, much has been learned through studies of human diseases or mouse models in which hepcidin is dysregulated.22 

Figure 1.

Regulators of hepcidin expression. Hepcidin is a common effector of 4 known regulators of iron homeostasis. Iron stores, erythropoietic demand, hypoxia, and inflammation all act by modulating hepatocyte production of hepcidin. Increased iron stores and inflammation both appear to increase hepcidin expression, primarily through signal transduction via the SMAD and JAK/STAT pathways, respectively. Increased/ineffective erythropoietic drive and hypoxia appear to decrease hepcidin expression, although the mechanisms of their control of hepcidin expression remain to be fully elucidated.

Figure 1.

Regulators of hepcidin expression. Hepcidin is a common effector of 4 known regulators of iron homeostasis. Iron stores, erythropoietic demand, hypoxia, and inflammation all act by modulating hepatocyte production of hepcidin. Increased iron stores and inflammation both appear to increase hepcidin expression, primarily through signal transduction via the SMAD and JAK/STAT pathways, respectively. Increased/ineffective erythropoietic drive and hypoxia appear to decrease hepcidin expression, although the mechanisms of their control of hepcidin expression remain to be fully elucidated.

Close modal

Hepcidin regulation by plasma iron and tissue iron stores is mediated through the bone morphogenic protein (BMP) in the sons of mothers against decapentaplegic (SMAD) pathway. In the iron-replete state, BMP6 binds to its receptor (BMPR) and their coreceptor hemojuvelin (HJV), which triggers a SMAD-dependent signaling cascade to increase hepcidin transcription in hepatocytes. The sensor for iron repletion is not nearly as clear, but appears to involve interactions among the BMPR/HJV complex, HFE, and TF receptor 2 (TFR2).16  Positive flux through this BMP/SMAD pathway can be dampened by the cleavage of HJV by the membrane-associated serine protease matriptase-2 encoded by TMPRSS6.22 

Hepcidin up-regulation by inflammation appears independent of BMP6 and HJV and mediated by inflammatory cytokines, especially IL-6, which binds to its receptor, activating JAK2 signaling and STAT3 phosphorylation.23  The inflammatory pathway requires the integrity of the BMP-SMAD pathway (particularly SMAD4) to fully activate hepcidin.24  The inflammatory cytokine Activin B also induces SMAD signaling and acts synergistically with IL-6 JAK2/STAT-3 signaling to up-regulate hepcidin expression.25 

Hepcidin is clearly down-regulated in the setting of hypoxia, anemia, and increased/ineffective erythropoietic activity; however, the identification of the mediator(s) and molecular mechanism remains unknown. The most commonly held hypothesis is that the BM produces a hepcidin-suppressing mediator in response to physiologic erythropoietin or pathophysiologic ineffective erythropoiesis. Growth differentiation factor 15 (GDF15) and twisted-gastrulation 1 (TWSG1) are BMP family members postulated to be suppressors of hepcidin in thalassemia26,27 ; however, their contribution remains uncertain.28  Erythroferrone (ERFE) has recently been described as a protein that mediates hepcidin suppression during stress erythropoiesis and ineffective erythropoiesis.29 

Dysregulation of hepcidin expression results in several hereditary and acquired disease entities. Inappropriately elevated levels of hepcidin can result in iron-restricted anemia that is acquired (eg, anemia of chronic inflammation) or more rarely hereditary (eg, iron-refractory iron deficiency anemia). Conversely, inappropriately low levels of hepcidin result in iron overload that can be hereditary (eg, hereditary hemochromatosis) or secondary (eg, anemias with ineffective erythropoiesis). The remainder of this review will concentrate on hereditary disorders of iron overload.

Although the earth is an iron-rich environment, acquired iron deficiency remains a global public health concern, with an estimated 1 billion persons affected. In contrast, most hereditary defects of iron metabolism result in iron overload rather than iron deficiency. The known genetic defects in many hereditary disorders of iron overload result in the aberrant regulation of iron absorption and storage. Clinically significant hereditary iron overload is almost exclusively an adult phenomenon, but there are rare pediatric forms. A summary of these disorders and their genetics can be found in Table 1.

HFE hemochromatosis

Originally recognized in the mid-19th century, this form of hereditary hemochromatosis was not linked to mutations in the HFE gene until 1996.30 HFE encodes atypical HLA class I-like membrane associated protein with a similar structure to other class I HLA membrane-associated proteins, but has no known function in antigen presentation. HFE is widely expressed and is known to bind with TFR131  to decrease its affinity for iron-loaded TF.32  Although the role of HFE in the regulation of hepcidin transcription remains unclear, patients with HFE hemochromatosis have inappropriately low hepcidin expression,33  which explains their increased intestinal iron absorption and increased reticuloendothelial iron release into the plasma.

Although HFE hemochromatosis has a worldwide distribution, it is the most common autosomal recessive disorder of northern European heritage, apparently having descended from a common Celtic ancestor.34  Most HFE hemochromatosis patients are homozygous for a p.Cys282Tyr (C282Y) amino acid substitution. The estimated prevalence of the C282Y allele in Caucasian Americans is 10%–15%, consistent with a homozygote prevalence estimated at 1 in every 200-250 persons.35-37 

Some clinical laboratories commonly report other HFE mutations. The p.His63Asp (H63D) amino acid substitution is occurs in ∼20% of the global population.36,38  The p.Ser65Cys (S65C) amino acid substitution is also reported by some, but the clinical relevance is not clear. Other very rare (“private”) HFE mutations can be found in trans to the C282Y and can explain some cases of rapid iron overload out of keeping with the C282Y carrier status.39 HFE sequencing (beyond genotyping for C282Y and H63D) may be difficult to obtain clinically and reimbursement by insurance payer may also be challenging.

Despite the relatively high prevalence of the HFE mutations in some populations, the penetrance of HFE hemochromatosis is low. Although C282Y homozygotes may develop biochemical evidence of significant iron loading in adolescence (an elevation in TF saturation usually precedes elevation of ferritin), usually only a small proportion ever develop sufficient iron overload to cause symptomatic clinical manifestations related to tissue or organ damage.40  Depending on the diagnostic criteria used, clinically significant iron overload is observed in only 1%–75% of C282Y homozygotes,37,41  with a recent European meta-analysis showing an overall penetrance of 13.5%.42 

The pathophysiologic consequences of the H63D mutation, and particularly the implications for C282Y/H63D compound heterozygotes, remain somewhat less certain. Only a small fraction of C282Y/H63D heterozygotes appear to develop biochemical evidence of iron loading and generally do not develop overt iron overload43  and clinicians should consider the contribution of other factors leading to iron overload in patients of this genotype.

HFE hemochromatosis was classically described as the triad of cirrhosis, diabetes mellitus, and skin hyperpigmentation developing in the in the 4th-7th decade of life and thus earning the description “bronze diabetes.” However, with the availability of genetic diagnosis since the late 1990s, the clinical presentation of HFE hemochromatosis has changed as a result of earlier diagnosis due to routine chemistry screening and screening relatives of affected individuals. Evaluations of large populations of in C282Y homozygotes or C282Y/H63D compound heterozygotes has revealed that the frequency of classically related nonspecific symptoms (eg, fatigue, arthralgias, affective disorders) and symptoms of end organ damage (eg, cardiomyopathy, arrhythmias, diabetes mellitus, sexual dysfunction) are uncommon,41  and in one study they did not differ significantly from wild-type controls.37  Men are significantly more likely to have symptomatic iron overload than women41  and, although this has been attributed to physiologic blood loss of menstruation and pregnancy or sex-related disease modifier genes,44  it may be more related to the testosterone-mediated suppression of hepcidin in males.21 

Juvenile hemochromatosis (HJV hemochromatosis and HAMP hemochromatosis)

Juvenile hemochromatosis is a rare, autosomal-recessive disorder with a phenotype of accelerated iron loading that usually presents before or during the 2nd decade of life. Genetic linkage analysis of multiple affected families revealed 2 distinct genetic entities with a common phenotype. Homozygous or compound heterozygous mutations in HJV or hepcidin (HAMP)45  result in the juvenile hemochromatosis phenotype. Both genotypes appear to have high penetrance, but HJV mutations are responsible for a majority of reported juvenile hemochromatosis cases.46 

As discussed previously, hemojuvelin is a critical BMPR coreceptor critical for SMAD-dependent signaling and hepcidin transcription. Mutations in HJV or hepcidin itself results in severely decreased hepcidin transcription and plasma levels with increased intestinal iron absorption and reticuloendothelial iron release, leading to rapid iron accumulation in target organs.

In contrast to HFE hemochromatosis, patients with juvenile hemochromatosis are more likely to present with endocrine failure (hypogonadism) or cardiomyopathy and hepatic disease is less prominent.47  Affected patients usually die from cardiac failure by age 30 years if untreated.48  The most effective current treatment is aggressive serial phlebotomy.

TFR2 hemochromatosis

TFR2 shares significant sequence homology with TFR1 and, although it can bind TF, it does so with lower affinity.49  The normal physiologic role of TFR2 in iron metabolism is not yet well understood, but appears to be involved as the “sensor” of iron status that regulates hepcidin expression.16,22  The prevalence of TFR2 mutations is low relative to HFE mutations, but they appear to have a more global ethnic distribution.46  One might expect that the loss of TFR2 function would result in decreased cellular iron uptake, but mutations in TFR2 are associated with an autosomal-recessive, adult-onset iron loading phenotype very similar to HFE hemochromatosis, whichi s associated with a predilection for hepatocellular iron deposition.50  The most effective current treatment is also serial phlebotomy.

FPN1 hemochromatosis

FPN1 hemochromatosis appears to be the most common form of non-HFE hemochromatosis.51  It is distinguished from other iron overload disorders by its autosomal-dominant inheritance and 2 phenotypic subtypes depending on the functional consequences of the pathological mutations in FPN1, the only cellular iron exporter.

Loss-of-function FPN1 hemochromatosis (“classical”) is associated with impaired iron release from macrophages, with reticuloendothelial accumulation of iron and iron-restricted erythropoiesis resulting in a mild, hypochromic anemia. Serum ferritin levels rise early and out of proportion to TF saturation, a pattern in contrast to the early elevation in TF saturation seen in HFE hemochromatosis. Although FPN1 hemochromatosis is generally milder than HFE hemochromatosis, the complete spectrum of iron overload morbidity has been demonstrated. Phlebotomy is a generally effective therapy; however, given the frequent presence of mild anemia and sequestration of iron within macrophages, patients usually require a gentler iron depletion program.52 

Rarer cases of gain-of-function FPN1 hemochromatosis (“nonclassical”) are related to mutations in FPN1 that lead to impaired hepcidin binding (“resistance”) and constitutive iron export from enterocytes and macrophages.51  Gain-of-function FPN1 hemochromatosis mutations lead to elevated TF saturation and hepatocellular/parenchymal iron overload that is phenotypically very similar to HFE hemochromatosis.

Aceruloplasminemia

Ceruloplasmin is a plasma copper-containing protein synthesized primarily in the liver. It is also expressed in a glycosylphosphatidylinositol-linked isoform on glial cells of the CNS.53  Ceruloplasmin is a ferrioxidase that has a vital function to iron mobilization from the tissues by converting Fe2+ to Fe3+.

Aceruloplasminemia is an extremely rare, autosomal-recessive disorder caused by missense/nonsense mutations in the ceruloplasmin gene (CP) found on chromosome 3q21.54  The absence of ferrioxidase ceruloplasmin does not affect copper homeostasis, but impairs mobilization of recycled iron stores from the reticuloendothelial system, resulting in low TF saturation, iron-restricted anemia, and visceral iron retention similar to gain-of-function FPN1 hemochromatosis. However, aceruloplasminemia can be clinically distinguished from FPN disease and the more common HFE hemochromatosis by the unique triad of early retinal degeneration, diabetes mellitus, and CNS iron deposition with neurological dysfunction. Onset of retinal degeneration and diabetes mellitus may occur as early as the 3rd decade, followed by neurological symptoms as much as a decade later. Neurological symptoms, including ataxia, involuntary movements, and cognitive dysfunction, are likely related to direct neurotoxicity of iron deposition in the CNS.55  Treatment with iron chelators, specifically improving the hepatic iron overload and diabetes, has met with some success, but is ineffective for mobilization of brain iron and neurological symptoms. In addition, chelation may be limited by accentuation of the associated iron-restricted anemia.54 

Atransferrinemia

Congenital deficiency of TF, the primary plasma iron-binding protein, is another extremely rare autosomal-recessive disorder due to mutations of the TF gene.56  Without plasma TF to deliver iron to erythroid precursors, affected individuals develop a severe, microcytic, hypochromic, iron-deficiency anemia phenotype requiring erythrocyte transfusions from birth.57  However, this iron-restricted state leads to enhanced intestinal absorption of iron that is ineffectively transported in the plasma and imported into erythroid precursors. The resulting non-TF-bound iron is avidly imported by parenchymal cells by an unclear mechanism and leads to a paradoxical iron overload of nonerythropoietic tissues. Tissues affected are similar to those seen in hemochromatosis, including liver, pancreas, heart, thyroid, and kidneys. Reticuloendothelial macrophages may be spared until the development of transfusion-related hemosiderosis. Serum ferritin is elevated and serum iron and total iron-binding capacity are very low. Supportive care with erythrocyte transfusions may relieve the anemia. Recurrent infusions of fresh-frozen plasma containing TF combined with chelation therapy or phlebotomy may be effective.56 

The diagnosis of hereditary iron overload relies on the careful evaluation of clinical phenotype, family history, and laboratory assessment. The rational use of a genetic testing algorithm will prevent the excessive use of resources (Figure 2).43,58 

Figure 2.

Guide to the evaluation of hereditary iron overload. After ruling out acquired causes of hyperferritinemia, TF saturation and clinical phenotype directs further rational genetic evaluation of hereditary iron overload. See text for more details. MAS indicates macrophage activation syndrome; HLH, hemophagocytic lymphohistiocytosis; MDS, myelodysplastic syndrome; LIC, liver iron concentration; and HH, hereditary hemochromatosis.

Figure 2.

Guide to the evaluation of hereditary iron overload. After ruling out acquired causes of hyperferritinemia, TF saturation and clinical phenotype directs further rational genetic evaluation of hereditary iron overload. See text for more details. MAS indicates macrophage activation syndrome; HLH, hemophagocytic lymphohistiocytosis; MDS, myelodysplastic syndrome; LIC, liver iron concentration; and HH, hereditary hemochromatosis.

Close modal

Large screening studies in HFE hemochromatosis have revealed that only ∼70% of adult C282Y homozygotes have elevated ferritin, and only a very small fraction of these have a symptomatic iron overload phenotype.36,37,41  Although uncommon, the clinician should still be able to recognize the symptomatic iron overload phenotype (hepatic or cardiac dysfunction, early onset atypical arthropathy, diabetes, sexual dysfunction, affective disorders, or hyperpigmentation).40  The clinician must also rule out secondary causes of iron overload (eg, chronic transfusion therapy, ineffective erythropoiesis, chronic supplemental iron) and alternative causes for elevated ferritin and modification of iron burden (eg, metabolic syndrome, inflammatory states, cell damage).

Family history suggestive of an inherited disease is also very important. Specific questioning regarding iron or liver abnormalities, race/ethnicity, and possible consanguinity are required. If a family history is positive, the clinician should attempt to determine the mode of inheritance (eg, recessive/dominant).

Laboratory documentation of increased iron stores is often the first step in screening/diagnosing hereditary hemochromatosis and include TF saturation (TFSat), unsaturated iron-binding capacity, and serum ferritin. TFSat is the ratio of serum iron to total iron-binding capacity expressed as a percentage. Previous recommendations to obtain fasting samples for TfSat have been shown to be unnecessary because fasting does not affect the sensitivity or specificity of detecting HFE hemochromatosis.59  Unsaturated iron-binding capacity has been shown to be a modestly better predictor of HFE hemochromatosis than TFSat, with the added advantage of lower cost and being a single direct measurement rather than a ratio60 ; however, the most recent American Association for the Study of Liver Diseases (AASLD) practice guideline uses TFSat in its diagnostic algorithm. Persistently elevated early morning fasting TFSat >45% is generally considered diagnostic of iron overload.40 

Serum ferritin provides a useful assessment body iron stores and confirmatory evidence of iron overload in the setting of an elevated TFSat. However, serum ferritin can be falsely elevated in some clinical settings such as inflammation, hepatic injury, and rapid cell turnover. Therefore, interpretation of an elevated ferritin must made in the broader clinical context. In Caucasians, an elevated TFSat should prompt assessment of the common HFE genotypes. Discovery of C282Y homozygosity confirms HFE hemochromatosis. Compound heterozygotes for HFE C282Y/H63D generally do not develop overt iron overload and clinicians should consider the contribution of other factors leading to iron overload. If the HFE genotyping is negative, one must consider rare private HFE mutations with complete HFE sequencing and non-HFE form of hereditary hemochromatosis. Non-HFE hemochromatosis etiologies can generally be divided into those with rapid loading and “juvenile” onset (<30 years of age) and others mimicking the natural history of HFE hemochromatosis. Younger patients should have sequencing of HJV and HAMP to assess for these forms of juvenile hemochromatosis, respectively. Older patients should have sequencing of TFR2 to assess for TFR2 hemochromatosis and FPN1 sequencing for mutations leading to FPN “resistance” and loss-of-function FPN1 hemochromatosis type 4B.

Much less commonly, a patient without acquired causes will present with isolated hyperferritinemia and a normal TF saturation. Confirmation of increased liver iron content by MRI or liver biopsy should prompt further careful evaluation of the hematologic and neurologic phenotype. Predominant macrophage iron loading on liver biopsy should prompt FPN1 sequencing for loss-of-function FPN1 hemochromatosis. The presence of microcytic, hypochromic anemia should prompt measurement of serum TF for consideration of hereditary hypo/atransferrinemia. If there are neurological symptoms in addition to anemia, ceruloplasmin should be measured in consideration of hereditary aceruloplasminemia. If isolated hyperferritinemia is discovered without an elevated liver iron concentration but with the presence of cataracts or brain iron loading, consideration should be given to sequencing ferritin light chain (FTL) for hyperferritinemia-cataract syndrome. Hyperferritinemia-cataract syndrome is more common than hereditary hypo/atransferrinemia and, although it is autosomal recessive, it can mimic the autosomal-dominant FPN1 loss-of-function hemochromatosis with elevated ferritin and normal TFSat.

Occasionally, a patient presents with what appears to be a hereditary hemochromatosis that does not fall neatly into the algorithm presented in Figure 2. Some of these individuals may harbor mutations in an unknown gene related to iron metabolism or hepcidin expression and others may have complex genotypes with single mutated/polymorphic alleles of 2 or more of the known iron homeostasis genes presented.

Conflict-of-interest disclosure: The author declares no competing financial interests. Off-label drug use: None disclosed.

Matthew M. Heeney, MD, Boston Children's Hospital, 300 Longwood Ave., Fegan 702, Boston, MA 02115; Phone: (617)919-3242; Fax: (617)730-0641; e-mail: matthew.heeney@childrens.harvard.edu.

1
Cook
 
JD
Barry
 
WE
Hershko
 
C
Fillet
 
G
Finch
 
CA
Iron kinetics with emphasis on iron overload
Am J Pathol
1973
, vol. 
72
 
2
(pg. 
337
-
344
)
2
Barton
 
JC
Bertoli
 
LF
Rothenberg
 
BE
Peripheral blood erythrocyte parameters in hemochromatosis: evidence for increased erythrocyte hemoglobin content
J Lab Clin Med
2000
, vol. 
135
 
1
(pg. 
96
-
104
)
3
Wood
 
JC
Use of magnetic resonance imaging to monitor iron overload
Hematol Oncol Clin North Am
2014
, vol. 
28
 
4
(pg. 
747
-
764
)pg. 
vii
 
4
Latunde-Dada
 
GO
Van der Westhuizen
 
J
Vulpe
 
CD
Anderson
 
GJ
Simpson
 
RJ
McKie
 
AT
Molecular and functional roles of duodenal cytochrome B (Dcytb) in iron metabolism
Blood Cells Mol Dis
2002
, vol. 
29
 
3
(pg. 
356
-
360
)
5
Fleming
 
MD
Trenor
 
CC
Su
 
MA
, et al. 
Microcytic anemia mice have a mutation in Nramp2, a candidate iron transporter gene
Nat Genet
1997
, vol. 
16
 (pg. 
383
-
386
)
6
Chen
 
C
Paw
 
BH
Cellular and mitochondrial iron homeostasis in vertebrates
Biochim Biophys Acta
2012
, vol. 
1823
 
9
(pg. 
1459
-
1467
)
7
Yuan
 
X
Fleming
 
MD
Hamza
 
I
Heme transport and erythropoiesis
Curr Opin Chem Biol
2013
, vol. 
17
 
2
(pg. 
204
-
211
)
8
Hamza
 
I
Korolnek
 
T
Like iron in the blood of the people: the requirement for heme trafficking in iron metabolism
Front Pharmacol
2014
, vol. 
5
 pg. 
126
 
9
Rajagopal
 
A
Rao
 
AU
Amigo
 
J
, et al. 
Haem homeostasis is regulated by the conserved and concerted functions of HRG-1 proteins
Nature
2008
, vol. 
453
 
7198
(pg. 
1127
-
1131
)
10
Severance
 
S
Hamza
 
I
Trafficking of heme and porphyrins in metazoa
Chem Rev
2009
, vol. 
109
 
10
(pg. 
4596
-
4616
)
11
Delaby
 
C
Rondeau
 
C
Pouzet
 
C
, et al. 
Subcellular localization of iron and heme metabolism related proteins at early stages of erythrophagocytosis
PLoS One
2012
, vol. 
7
 
7
pg. 
e42199
 
12
Chiabrando
 
D
Vinchi
 
F
Fiorito
 
V
Mercurio
 
S
Tolosano
 
E
Heme in pathophysiology: a matter of scavenging, metabolism and trafficking across cell membranes
Front Pharmacol
2014
, vol. 
5
 pg. 
61
 
13
Korolnek
 
T
Zhang
 
J
Beardsley
 
S
Scheffer
 
GL
Hamza
 
I
Control of metazoan heme homeostasis by a conserved multidrug resistance protein
Cell Metab
2014
, vol. 
19
 
6
(pg. 
1008
-
1019
)
14
Abboud
 
S
Haile
 
DJ
A novel mammalian iron-regulated protein involved in intracellular iron metabolism
J Biol Chem
2000
, vol. 
275
 
26
(pg. 
19906
-
19912
)
15
McKie
 
AT
Marciani
 
P
Rolfs
 
A
, et al. 
A novel duodenal iron-regulated transporter, IREG1, implicated in the basolateral transfer of iron to the circulation
Mol Cell
2000
, vol. 
5
 
2
(pg. 
299
-
309
)
16
Ganz
 
T
Systemic iron homeostasis
Physiol Rev
2013
, vol. 
93
 
4
(pg. 
1721
-
1741
)
17
Kosman
 
DJ
Redox cycling in iron uptake, efflux, and trafficking
J Biol Chem
2010
, vol. 
285
 
35
(pg. 
26729
-
26735
)
18
Ganz
 
T
Hepcidin and iron regulation, 10 years later
Blood
2011
, vol. 
117
 
17
(pg. 
4425
-
4433
)
19
Nemeth
 
E
Tuttle
 
MS
Powelson
 
J
, et al. 
Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization
Science
2004
, vol. 
306
 
5704
(pg. 
2090
-
2093
)
20
Heeney
 
MM
Andrews
 
NC
Iron homeostasis and inherited iron overload disorders: an overview
Hematol Oncol Clin North Am
2004
, vol. 
18
 
6
(pg. 
1379
-
1403
)pg. 
ix
 
21
Bachman
 
E
Feng
 
R
Travison
 
T
, et al. 
Testosterone suppresses hepcidin in men: a potential mechanism for testosterone-induced erythrocytosis
J Clin Endocrinol Metab
2010
, vol. 
95
 
10
(pg. 
4743
-
4747
)
22
Camaschella
 
C
Silvestri
 
L
Molecular mechanisms regulating hepcidin revealed by hepcidin disorders
ScientificWorldJournal
2011
, vol. 
11
 (pg. 
1357
-
1366
)
23
Verga Falzacappa
 
MV
Vujic Spasic
 
M
Kessler
 
R
Stolte
 
J
Hentze
 
MW
Muckenthaler
 
MU
STAT3 mediates hepatic hepcidin expression and its inflammatory stimulation
Blood
2007
, vol. 
109
 
1
(pg. 
353
-
358
)
24
Wang
 
RH
Li
 
C
Xu
 
X
, et al. 
A role of SMAD4 in iron metabolism through the positive regulation of hepcidin expression
Cell Metab
2005
, vol. 
2
 
6
(pg. 
399
-
409
)
25
Besson-Fournier
 
C
Latour
 
C
Kautz
 
L
, et al. 
Induction of activin B by inflammatory stimuli up-regulates expression of the iron-regulatory peptide hepcidin through Smad1/5/8 signaling
Blood
2012
, vol. 
120
 
2
(pg. 
431
-
439
)
26
Tanno
 
T
Bhanu
 
NV
Oneal
 
PA
, et al. 
High levels of GDF15 in thalassemia suppress expression of the iron regulatory protein hepcidin
Nat Med
2007
, vol. 
13
 
9
(pg. 
1096
-
1101
)
27
Tanno
 
T
Porayette
 
P
Sripichai
 
O
, et al. 
Identification of TWSG1 as a second novel erythroid regulator of hepcidin expression in murine and human cells
Blood
2009
, vol. 
114
 
1
(pg. 
181
-
186
)
28
Casanovas
 
G
Vujic Spasic
 
M
Casu
 
C
, et al. 
The murine growth differentiation factor 15 is not essential for systemic iron homeostasis in phlebotomized mice
Haematologica
2013
, vol. 
98
 
3
(pg. 
444
-
447
)
29
Kautz
 
L
Jung
 
G
Valore
 
EV
Rivella
 
S
Nemeth
 
E
Ganz
 
T
Identification of erythroferrone as an erythroid regulator of iron metabolism
Nat Genet
2014
, vol. 
46
 
7
(pg. 
678
-
684
)
30
Feder
 
JN
Gnirke
 
A
Thomas
 
W
, et al. 
A novel MHC class I-like gene is mutated in patients with hereditary haemochromatosis
Nat Genet
1996
, vol. 
13
 
4
(pg. 
399
-
408
)
31
Lebron
 
JA
West
 
AP
Bjorkman
 
PJ
The hemochromatosis protein HFE competes with transferrin for binding to the transferrin receptor
J Mol Biol
1999
, vol. 
294
 
1
(pg. 
239
-
245
)
32
Lebron
 
JA
Bennett
 
MJ
Vaughn
 
DE
, et al. 
Crystal structure of the hemochromatosis protein HFE and characterization of its interaction with transferrin receptor
Cell
1998
, vol. 
93
 (pg. 
111
-
123
)
33
Bridle
 
KR
Frazer
 
DM
Wilkins
 
SJ
, et al. 
Disrupted hepcidin regulation in HFE-associated haemochromatosis and the liver as a regulator of body iron homoeostasis
Lancet
2003
, vol. 
361
 
9358
(pg. 
669
-
673
)
34
Ajioka
 
RS
Jorde
 
LB
Gruen
 
JR
, et al. 
Haplotype analysis of hemochromatosis: evaluation of different linkage-disequilibrium approaches and evolution of disease chromosomes
Am J Hum Genet
1997
, vol. 
60
 (pg. 
1439
-
1447
)
35
Olynyk
 
JK
Cullen
 
DJ
Aquilia
 
S
Rossi
 
E
Summerville
 
L
Powell
 
LW
A population-based study of the clinical expression of the hemochromatosis gene
N Engl J Med
1999
, vol. 
341
 
10
(pg. 
718
-
724
)
36
Adams
 
PC
Reboussin
 
DM
Barton
 
JC
, et al. 
Hemochromatosis and iron-overload screening in a racially diverse population
N Engl J Med
2005
, vol. 
352
 
17
(pg. 
1769
-
1778
)
37
Beutler
 
E
Felitti
 
VJ
Koziol
 
JA
Ho
 
NJ
Gelbart
 
T
Penetrance of 845G–> A (C282Y) HFE hereditary haemochromatosis mutation in the USA
Lancet
2002
, vol. 
359
 
9302
(pg. 
211
-
218
)
38
Merryweather-Clarke
 
AT
Pointon
 
JJ
Shearman
 
JD
Robson
 
KJ
Global prevalence of putative haemochromatosis mutations
J Med Genet
1997
, vol. 
34
 
4
(pg. 
275
-
278
)
39
Aguilar-Martinez
 
P
Grandchamp
 
B
Cunat
 
S
, et al. 
Iron overload in HFE C282Y heterozygotes at first genetic testing: a strategy for identifying rare HFE variants
Haematologica
2011
, vol. 
96
 
4
(pg. 
507
-
514
)
40
Bacon
 
BR
Adams
 
PC
Kowdley
 
KV
Powell
 
LW
Tavill
 
AS
American Association for the Study of Liver Diseases
Diagnosis and management of hemochromatosis: 2011 practice guideline by the American Association for the Study of Liver Diseases
Hepatology
2011
, vol. 
54
 
1
(pg. 
328
-
343
)
41
Allen
 
KJ
Gurrin
 
LC
Constantine
 
CC
, et al. 
Iron-overload-related disease in HFE hereditary hemochromatosis
N Engl J Med
2008
, vol. 
358
 
3
(pg. 
221
-
230
)
42
EASL
EASL clinical practice guidelines for HFE hemochromatosis
J Hepatol
2010
, vol. 
53
 
1
(pg. 
3
-
22
)
43
Bardou-Jacquet
 
E
Brissot
 
P
Diagnostic evaluation of hereditary hemochromatosis (HFE and non-HFE)
Hematol Oncol Clin North Am
2014
, vol. 
28
 
4
(pg. 
625
-
635
)
44
Barton
 
JC
Wiener
 
HW
Acton
 
RT
Go
 
RC
HLA haplotype A*03-B*07 in hemochromatosis probands with HFE C282Y homozygosity: frequency disparity in men and women and lack of association with severity of iron overload
Blood Cells Mol Dis
2005
, vol. 
34
 
1
(pg. 
38
-
47
)
45
Roetto
 
A
Papanikolaou
 
G
Politou
 
M
, et al. 
Mutant antimicrobial peptide hepcidin is associated with severe juvenile hemochromatosis
Nat Genet
2003
, vol. 
33
 
1
(pg. 
21
-
22
)
46
Pietrangelo
 
A
Caleffi
 
A
Corradini
 
E
Non-HFE hepatic iron overload
Semin Liver Dis
2011
, vol. 
31
 
3
(pg. 
302
-
318
)
47
De Gobbi
 
M
Roetto
 
A
Piperno
 
A
, et al. 
Natural history of juvenile haemochromatosis
Br J Haematol
2002
, vol. 
117
 
4
(pg. 
973
-
979
)
48
Pietrangelo
 
A
Non-HFE hemochromatosis
Hepatology
2004
, vol. 
39
 
1
(pg. 
21
-
29
)
49
West
 
AP
Bennett
 
MJ
Sellers
 
VM
Andrews
 
NC
Enns
 
CA
Bjorkman
 
PJ
Comparison of the interactions of transferrin receptor and transferrin receptor 2 with transferrin and the hereditary hemochromatosis protein HFE
J Biol Chem
2000
, vol. 
275
 
49
(pg. 
38135
-
38138
)
50
Camaschella
 
C
Roetto
 
A
Cali
 
A
, et al. 
The gene TFR2 is mutated in a new type of haemochromatosis mapping to 7q22
Nat Genet
2000
, vol. 
25
 (pg. 
14
-
15
)
51
Mayr
 
R
Janecke
 
AR
Schranz
 
M
, et al. 
Ferroportin disease: a systematic meta-analysis of clinical and molecular findings
J Hepatol
2010
, vol. 
53
 
5
(pg. 
941
-
949
)
52
Pietrangelo
 
A
The ferroportin disease
Blood Cells Mol Dis
2004
, vol. 
32
 
1
(pg. 
131
-
138
)
53
Klomp
 
LW
Gitlin
 
JD
Expression of the ceruloplasmin gene in the human retina and brain: implications for a pathogenic model in aceruloplasminemia
Hum Mol Genet
1996
, vol. 
5
 
12
(pg. 
1989
-
1996
)
54
Kono
 
S
Aceruloplasminemia: an update
Int Rev Neurobiol
2013
, vol. 
110
 (pg. 
125
-
151
)
55
Miyajima
 
H
Takahashi
 
Y
Kono
 
S
Aceruloplasminemia, an inherited disorder of iron metabolism
Biometals
2003
, vol. 
16
 
1
(pg. 
205
-
213
)
56
Beutler
 
E
Gelbart
 
T
Lee
 
P
Trevino
 
R
Fernandez
 
MA
Fairbanks
 
VF
Molecular characterization of a case of atransferrinemia
Blood
2000
, vol. 
96
 
13
(pg. 
4071
-
4074
)
57
Hamill
 
RL
Woods
 
JC
Cook
 
BA
Congenital atransferrinemia: a case report and review of the literature
Am J Clin Pathol
1991
, vol. 
96
 (pg. 
215
-
218
)
58
European Association For The Study Of The L
EASL clinical practice guidelines for HFE hemochromatosis
J Hepatol
2010
, vol. 
53
 
1
(pg. 
3
-
22
)
59
Adams
 
PC
Reboussin
 
DM
Press
 
RD
, et al. 
Biological variability of transferrin saturation and unsaturated iron-binding capacity
Am J Med
2007
, vol. 
120
 
11
(pg. 
999.e1
-
e7
)
60
Adams
 
PC
Reboussin
 
DM
Leiendecker-Foster
 
C
, et al. 
Comparison of the unsaturated iron-binding capacity with transferrin saturation as a screening test to detect C282Y homozygotes for hemochromatosis in 101,168 participants in the hemochromatosis and iron overload screening (HEIRS) study
Clin Chem
2005
, vol. 
51
 
6
(pg. 
1048
-
1052
)
61
Adams
 
PC
Barton
 
JC
How I treat hemochromatosis
Blood
2010
, vol. 
116
 
3
(pg. 
317
-
325
)