You are on page 1of 21

Blood Iron Homeostasis: Newly Discovered Proteins and

Iron Imbalance
Mark R. Bleackley1, Ann Y.K. Wong1, David M. Hudson, Christopher H-Y. Wu, and Ross T.A. MacGillivray

In biological systems, iron exerts 2 contrasting effects. evolved. As shown in the thalassemias, iron imbalance
The chemical reactivity of iron is essential for the can have devastating effects on human health. Recently,
biological activities of proteins such as hemoglobin, several new proteins have been described that play
ribonucleotide reductase, the cytochromes, and aconi- critical roles in iron regulation including the master
tases. However, free iron in a cell has the propensity to regulator of iron metabolism (hepcidin). In this review,
generate free radicals which can damage cellular we discuss the new knowledge that has arisen from
components containing proteins, lipids, and nucleic studies in yeast and in humans, and we show how these
acids. To maintain the balance between iron as an studies are shedding new light on some well-known
essential nutrient and iron as a potential cytotoxin, a human disorders.
number of biological protective mechanisms have © 2009 Elsevier Inc. All rights reserved.

ORE THAN HALF of the 3 to 4 g of iron in transporter 1, duodenal cytochrome b, hemojuvelin,


M a well-nourished human is bound to hemo-
globin in red blood cells (RBCs) and their
and hepcidin. We then discuss the proposed
functions of these proteins in iron homeostasis
precursors. Each day, 20 to 25 mg of iron is while identifying gaps in our current knowledge.
required to support the synthesis of hemoglobin in
reticulocytes. The chemical properties of iron that
make it such a beneficial element also lead to a
Abbreviations: Aα, alpha-amyloid peptide; APP, amyloid β
propensity of free iron and the subsequent forma-
precursor protein; β2M, beta 2 microglobulin; BMP, bone
tion of damaging free radicals via the Fenton series morphogenetic protein; Cp, ceruloplasmin; cytb561, cytochrome
of reactions. As a result, organisms have evolved b561; DCT1, divalent cation transporter; Dcytb, duodenal
complex biochemical mechanisms for maintaining cytochrome b; DMT1, divalent metal transporter 1; FPN1,
a balance between iron as an essential element and ferroportin-1; FRDA, Friedreich's ataxia; FXN, frataxin; GPI,
glycosylphosphatidylinositol; HCP1, heme carrier protein 1; HFE,
iron as a cytotoxic agent. With the exception of a
hemochromatosis protein; HH, hereditary hemochromatosis;
few members of the genus Lactobacillus and some HIF1α, hypoxia inducible factor-1 α; HJV, hemojuvelin; Hp,
strains of Bacillus, iron is essential for the viability hephaestin; IMP, integrin-mobilferrin pathway; IRE, iron response
of all organisms.1 Two major redox states exist for element; IRP, iron response protein; Lf, lactoferrin; LIP, labile iron
iron (ferrous or Fe(II) and ferric or Fe(III)), but both pool; mHJV, glycosylphosphatidylinositol-anchored hemojuvelin;
MTf, melanotransferrin; PCFT, proton-coupled folate transporter;
higher and lower valences have been observed in
RBCs, red blood cells; ROS, reactive oxygen species; sHJV, soluble
the laboratory. Under physiologic conditions, iron hemojuvelin; SLA, sex-linked anemia; sMTf, soluble
exists in a number of biochemically available redox melanotransferrin; Tf, transferrin; TfR, transferrin receptor; TM,
states that allow it to participate in single electron transmembrane; UTR, untranslated region.
transfer reactions. Binding of different ligands to From the Centre for Blood Research and Department of
Biochemistry and Molecular Biology, University of British
iron can vary the potential of the Fe(II)/Fe(III)
Columbia, Vancouver, BC, Canada.
redox couple allowing iron to meet the require- This study was supported in part by a grant from the
ments of a wide range of cellular reactions. Iron in Canadian Blood Services–Canadian Institutes of Health
proteins is often found complexed in heme (such as Research (CIHR) Program in Blood Utilization and
hemoglobin) or in iron-sulfur (Fe-S) clusters (such Conservation awarded to RTAM. MRB was supported by a
graduate studentship from the Strategic Training Program in
as aconitases). Other proteins (nonheme iron
Transfusion Science (funded by the CIHR and the Heart and
proteins including ribonucleotide reductase and Stroke Foundation of Canada). AYKW was supported by a
transferrin [Tf]) bind iron in a variety of different graduate studentship from CBS.
1
ways. These authors contributed equally to this work.
During the past few years, several molecular Address reprint requests to Ross T.A. MacGillivray, Centre for
Blood Research, University of British Columbia, Vancouver, BC,
genetic approaches have revealed the functions of
Canada V6T 1Z3. E-mail: macg@interchange.ubc.ca
many new iron-related proteins. In this review, we 0887-7963/08/$ - see front matter
discuss the strategies used to identify proteins such © 2009 Elsevier Inc. All rights reserved.
as hephaestin (Hp), ferroportin, divalent metal doi:10.1016/j.tmrv.2008.12.001

Transfusion Medicine Reviews, Vol 23, No 2 (April), 2009: pp 103-123 103


104 BLEACKLEY ET AL

Finally, we discuss the disorders that result from a phenomena explain why there is so much iron on
deficiency of these new proteins including effects and within the earth.2
within iron regulation as well as some newly When life first evolved on earth, the atmosphere
recognized neurodegenerative diseases. is believed to have been anoxic. Under these
conditions, Fe(II) was stable (as it did not auto-
CHEMISTRY AND BIOCHEMISTRY OF IRON oxidize) and the concentration of dissolved ferrous
iron in the oceans was around 50 μmol/L. The
Located in the middle of the periodic table, iron
abundance of iron in the oceans, combined with
is a transition metal that exists in oxidation states
chemical properties of iron, made it well suited for
ranging from −II, as in the Fe(CO)42− anion, to +VI,
biological systems. In the absence of oxygen,
as in the ferrate ion FeO42−.2 Biochemically, Fe(II)
copper (which is also capable of single electron
and Fe(III) are the most relevant oxidation states.
transfer reactions) existed predominantly in the
Transfer of a single electron between these 2 states
insoluble and thus nonbioavailable Cu(I) state.
is readily accomplished with ascorbate or molecular
Early life exploited Fe(II) chemistry and avail-
oxygen. Fe(IV) and Fe(V) are encountered in
ability to carry out chemical reactions necessary for
biology but only as reactive intermediates in the
survival. Evolution of photosynthetic bacteria 109
catalytic mechanisms of some iron-containing
years ago began the shift to the aerobic atmosphere
enzymatic processes.3 The potential of the Fe(III)/
found today. However, it is estimated that it took
Fe(II) redox couple (0.77 V)2 can vary by up to 1 V
200 to 300 million years to oxidize all of the iron in
in response to different environments,4,5 particu-
the oceans with the resulting precipitation and
larly when influenced by protein-bound coordina-
sedimentation of the Fe(III). Today, evidence of the
tion ligands. This variability in redox potential
shift to an aerobic atmosphere can be seen as red
contributes significantly to the role of iron as an
bands in sedimentary rock. In contrast, the oxida-
essential biological metal. Within the range of
tion of copper to Cu(II) increased solubility and
biological oxidants and reductants (+0.82V to
thus bioavailability allowing for the evolution of
−0.32V, molecular oxygen and pyridine nucleo-
copper-catalyzed biological reactions. Bioavailable
tides, respectively), electron transfers facilitated by
Fe(II) was no longer prevalent, leading to a mass
iron are reversible.6
extinction of many life forms. Surviving species
had evolved mechanisms for scavenging and
PREVALENCE OF IRON IN BIOLOGICAL solubilizing Fe(III) and reducing it to Fe(II).
SYSTEMS However, Fe(II) can react with environmental
On examination of the history of the earth and oxygen, causing production of hydroxyl radicals
evolution of life, it is clear that abundance, through the Haber-Weiss-Fenton reaction sequence.
availability, and suitability of iron have been critical Extracellular matrices of unicellular organisms
to life. In the earth's crust, iron is the fourth most were subject to destruction by these damaging
abundant element and second most abundant metal. radicals. In turn, organisms evolved matrices of
Stars, such as our sun, produce energy through the crosslinked connective tissue using free radical
formation of helium via nuclear fusion of hydro- polymerization. This is thought to correspond with
gen.7 Helium builds up in the core of the star the beginning of multicellular life.1
leading to the formation of a red giant. As the
temperature increases to 108 K, helium fuses to IRON METABOLISM IN YEAST
form large quantities of carbon, and oxygen Baker's yeast (Saccharomyces cerevisiae) has
becomes the major energy source.8 In larger stars, proved to be a valuable model organism in the
this process occurs more rapidly (10 million years). study of iron metabolism. A high degree of
The additional heat from the fusion reactions (109 conservation exists between the cellular components
K) provides the energy7 to fuse nuclei up to the of iron metabolism in humans and S cerevisiae (see
mass of iron but is insufficient for fusions forming Table 1). As shown in Figure 1, there are 3 iron
heavier elements. As a result, large amounts of iron uptake pathways in S cerevisiae: 2 low affinity iron
accumulate in the core. Our solar system was pathways (Fet4 and Smf1) and one high affinity
formed via aggregation of material from an earlier pathway (Fet3/Ftr1). There is also a siderophore
star that exploded. Thus, these cosmochemical scavenging pathway that is not shown in Figure 1.
IRON IMBALANCE AND DISEASE 105

Table 1. Proteins of Human Iron Metabolism and Homologs in S cerevisiae


Human Gene/
Protein Function in Humans Yeast Homolog Function in Yeast

Dcytb Membrane ferric reductase None but yeast contains Not applicable
nonhomologous ferric
reductases such as Fre1/2
DMT1 Dietary iron absorption at the SMF1/2 Broad specificity divalent and
apical membrane of duodenal trivalent metal transporter
enterocytes, release of iron
from transferrin cycle
endosomes
Ferritin Multimeric iron storage protein None Not applicable
Ferroportin-1 Exports iron out of duodenal None Not applicable
enterocytes, macrophages,
and a variety of neural cells
Frataxin Regulates mitochondrial iron Yfh1 Regulates mitochondrial iron
accumulation, thought to accumulation, thought to
promote Fe(II) availability, promote Fe(II) availability,
involved in maturation of heme involved in maturation of heme
and Fe-S cluster proteins and Fe-S cluster proteins
Human ATX1 homologue Cytoplasmic Cu Atx1 Cytoplasmic Cu
metallochaperone metallochaperone that
transports copper to Ccc2
Hephaestin Multicopper ferroxidase Fet3 Multicopper ferroxidase
involved in iron export from required for high affinity
duodenal enterocytes iron uptake
Heme carrier protein 1 Apical membrane heme None Not applicable
transporter
Mitoferrin Mitochondrial membrane iron Mrs3/4 Mitochondrial membrane iron
transporter transporter
Transferrin Binding of Fe3+ in blood None Not applicable
Transferrin receptor 1 Receptor for Tf-Fe, essential for None Not applicable
iron uptake from blood
Wilson/Menkes protein Golgi Cu transporters Ccc2 Cu transporting P-type ATPase,
responsible for Cu uptake into
the late Golgi for incorporation
into Fet3

Once inside the cell, iron is trafficked to mitochondria not have homologs in yeast. As shown in Figure 2,
where it is transported across the membrane by however, some aspects of iron uptake and utilization
proteins such as Mrs3/4. Inside the mitochondrion, are well conserved between yeast and human cells.
Yfh1 binds iron and takes it to sites of heme and Fe-S
cluster synthesis. Excess iron is trafficked to the DIETARY IRON UPTAKE IN HUMANS
vacuole where it is transported across the vacuolar
membrane by Ccc1 for storage. Release of iron from Bioavailability of Iron
the vacuole is accomplished by Fet5/Fth1 which are In food, iron is found in 2 basic forms: inorganic
homologous to Fet3/Ftr1 (Fig 1).9 Studies of yeast iron (both Fe(II) and Fe(III)) and heme where iron
proteins such as Fet3, Yfh1, Ccc2, Atx1, and other is complexed to protoporphyrin IX. In an average
proteins have brought considerable insight to the diet, inorganic iron accounts for approximately
functions of their human homologs. There are 90% of total dietary iron content, whereas heme
additional levels of complexity in human iron makes up the remaining 10%.10,11 Iron absorption
metabolism that are caused by the additional varies significantly with diet composition, iron
transport, absorption, and regulation steps that are status of the individual, and, most importantly,
required in a multicellular organism. As a result, a bioavailability of the different iron forms. It has
regulatory hormone such as hepcidin, a storage been estimated that in the developed world, where
protein like ferritin, and a transport protein like Tf do meat consumption is relatively high, more than half
106 BLEACKLEY ET AL

Fig 1. Iron metabolism in the yeast S cerevisiae. Starting at the bottom and going clockwise, the organelles within the yeast cell are the
nucleus, the Golgi apparatus, the vacuole, and the mitochondrion. Yeast proteins are indicated by beads containing the abbreviated protein
name. Involvement of a putative intracellular chaperone is indicated with the 2 question marks. Specific transporter proteins are shown as
beads containing a channel. The oxidation state of iron and copper is indicated as suffixes. Three common uptake pathways are shown
involving Fet3/Ftr1, Smf1, and Fet4. Further details of the yeast proteins can be found in the text and at http://www.yeastgenome.org.

of all absorbed iron comes from the heme in vailability of inorganic iron is dependent on other
hemoglobin or myoglobin in dietary meats, dietary components. Enhancers, such as ascorbic
whereas the rest of the world obtains iron mostly acid, increase inorganic iron bioavailability by
as Fe(III) from plant sources.10-14 An alternative promoting the reduction of Fe(III) to soluble Fe
iron source in some diets comes from pharmaceu- (II). Inhibitors such as phytates in cereal and
tical iron supplements such as Fe(II).12,13 Among polyphenols in plants form insoluble complexes
all natural iron sources, heme is the most bioavail- with inorganic iron and thereby reduce its absorp-
able and its level of absorption remains unaffected tion.11,15 In addition, inorganic iron absorption is
regardless of diet composition. In contrast, bioa- influenced by the level of gastric acid secretion in
IRON IMBALANCE AND DISEASE 107

Fig 2. Iron absorption in a human duodenal enterocyte. A stylized enterocyte cell is shown with the villi-containing apical membrane at
the top and the basolateral membrane and the blood vessels at the bottom. Within the cell, the organelles surrounding the nucleus include 2
mitochondria, a recycling endosome and the Golgi apparatus. Proteins are indicated by beads containing the abbreviated protein name (see
Table 1). Transporter proteins are shown as beads containing a channel. The involvement of a putative intracellular chaperone is indicated
by the question mark. Uptake of Fe(III) involves DCytB and the transporter DMT1 together with a proton gradient (Δ [H+]). Uptake of heme
iron involves HCP1. Within the cell, iron stores are shown as ferritin (Fr). The putative functions of FPN1 and Hp are indicated by their
supplying Fe(III) to apotransferrin (Tf). See text for details.

the stomach with acidity increasing the solubility of Dietary iron absorption occurs in the proximal
inorganic iron.16 An overview of dietary iron small intestine by specialized epithelial cells called
uptake in the human gut is shown in Figure 2. duodenal enterocytes. Mechanisms for inorganic
108 BLEACKLEY ET AL

iron uptake allow both Fe(II) and Fe(III) to be form I is proposed to be the primary functional
imported into the enterocyte. To date, 3 pathways isoform 11 and is found mainly in the apical
for iron uptake have been identified, 2 of which are membrane of epithelial cells.31 The isoform I
shown in Figure 2. The third pathway involves mRNA is stabilized by the binding of iron response
uptake of Fe(III) through the β3 integrin mobilferrin protein (IRP) to the IRE resulting in increased
pathway13,17,18; this pathway is not well character- expression.29,32 Motifs in the carboxy termini of
ized and thus has been omitted from Figure 2 but a isoforms I and II have recently been found to be
brief overview is presented later in this review. responsible for protein targeting and trafficking.
Isoform I is more stable in the plasma membrane
Divalent Metal Transporter 1 compared to isoform II. Isoform II contains a
Inorganic iron is transported across the apical sequence that signals recycling of the protein back
membrane of enterocytes as Fe(II) by a nonspecific to the plasma membrane after internalization,
divalent metal transporter 1 (DMT1; Fig 2). whereas isoform I contains a signal that targets it
Divalent metal transporter 1 is also known as to lysosomes.33
SLC11A2, natural resistance–associated macro-
phage protein 2,19 and divalent cation transporter Duodenal Cytochrome b
1,20 and is necessary for dietary iron absorption.21 Duodenal cytochrome b (Dcytb) is a highly
Significant reduction in DMT1 results in severe regulated ferrireductase in the apical membrane of
anemia that is untreatable by oral or intravenous duodenal enterocytes that reduces dietary Fe(III) to
iron supplement demonstrating the importance of Fe(II).34 The mRNA and protein expression levels
DMT1.19,22 Protons are required by DMT1 to drive of Dcytb both increase in response to iron
the transport of divalent metal ions including Fe(II), deficiency and hypoxia. 34,35 Unlike the iron-
Mn(II), and Co(II).20 The stoichiometry between regulated transporter DMT1, Dcytb transcripts do
protons and divalent metal ion transported by not contain IREs, and so the mechanism whereby
DMT1 is dependent on extracellular pH.23 Divalent iron regulates Dcytb expression remains unknown.
metal transporter 1 is predicted to have 12 It has been proposed that a transcription factor may
transmembrane (TM) domains.20 Mutation studies be involved, similar to Aft1p regulation of
were performed to identify regions important for ferrireductases FRE1p and FRE2p expression in
DMT1 function. Amino acids near the end of TM1 yeast.36 Over a 218–amino acid region, Dcytb
and in the extracellular loop between TM1 and shows 40% to 50% sequence identity to sheep
TM2 were shown to interact with both divalent cytochrome b561 (cytb561), whereas the amino-
cations and protons.24 A spontaneous mutation in terminal region is homologous to hemoprotein p30.
the DMT1 gene (G185R) is located on the polar However, no homology is found between Dcytb
side of the amphipathic TM4 and is likely to have and ferrireductases isolated from plants and
an effect on contacts between TM domains.25 In yeast.34,36 Dcytb is predicted to contain 6 trans-
addition, TM4 26 and His272 in TM6 27 are membrane domains and the sequence comparison
important for coupling divalent cation and proton with cytb561 shows 4 conserved histidine residues
transport. The proton-induced inward current which are thought to be involved in heme
obtained from Fe(II) transport studies by a H272A binding.34,37 Partial conservation of the cytb561
DMT1 mutant suggests that proton coupling ascorbic acid and dehydroascorbic acid binding
increases DMT1 affinity for Fe(II) in addition to sites in Dcytb suggests that Dcytb may react with
providing thermodynamic force for transport.27 one or both of these compounds. 34 Cytb561
Multiple isoforms of DMT1 result from alter- reduces dehydroascorbate to ascorbate by trans-
native splicing in exon 16 and the use of different porting an electron donated by cytoplasmic ascor-
transcription start sites (1A or 1B) in exon 1.28,29 In bate across the membrane; thus, Dcytb may be able
rodents, differential expression and subcellular to reduce Fe(III) to Fe(II) using ascorbate as an
localization of the different isoforms are regulated electron donor.38 The requirement of Dcytb for
both transcriptionally and posttranscriptionally.30 dietary iron absorption is questionable; Dcytb null
There are 2 common isoforms: isoform I, which has mice do not present any iron-related disorders when
an additional iron response element (IRE) in the 3′ fed a normal diet21 and no mutations in human
untranslated region (UTR), and isoform II.28 Iso- Dcytb have been reported to date.39
IRON IMBALANCE AND DISEASE 109

Heme Absorption degradation by heme oxygenase 1.48 Heme carrier


protein 1 was recently shown to play a major role in
Until recently, knowledge about heme absorption folate transport in the presence of a proton gradient
by enterocytes was limited to the idea that heme and was given the additional name proton-coupled
binds to an apical membrane protein and is absorbed folate transporter (PCFT)/HCP1.49
by enterocytes as an intact molecule.40-42 A specific
pathway for heme absorption is evident when Alternative Iron Pathways
studying the import of zinc mesoporphyrin, a The third pathway for the transport of iron into
fluorescent heme analog, by human intestinal enterocytes is the integrin-mobilferrin pathway
Caco2 cells.43 Our understanding of heme absorp- (IMP).50 Mobilferrin is a homologue of calreticulin,
tion has increased considerably with the discovery of a common chaperone protein within the endoplas-
heme carrier protein 1 (HCP1) by cDNA suppressive mic reticulum, whereas β3 integrin is a known
subtraction using hypotransferrinemic mice.44 The adhesion protein.13 Despite having been studied for
human HCP1 gene (2097 base pair) on chromosome more than a decade, the IMP pathway remains
17q11.1 encodes a 459–amino acid polypeptide that poorly understood. It is now known that the IMP
is predicted to have 9 transmembrane domains and pathway only transports iron in an inorganic Fe(III)
belongs to the major facilitator superfamily of state.17 It is believed that during iron deficiency,
transporter proteins (reviewed in reference 45). mobilferrin is secreted into the lumen of the small
Within this superfamily, HCP1 most resembles the intestine along with the mucosal surface protein
bacterial metal-tetracycline transporter.44 Heme mucin and the 2 proteins chelate Fe(III).13 The
carrier protein 1 contains a highly conserved motif soluble Fe(III)-mucin-mobilferrin complex is trans-
(GXXSDRXGRR) between TM2 and TM3, which ported to the plasma membrane and Fe(III) is taken
is also found in the metal-tetracycline transporter and up into the enterocyte by β3 integrin.51 Inside the
the heme exporter protein, feline leukemia virus cytosol, the assembly of a complex of β3 integrin,
receptor C. However, no known heme binding mobilferrin, flavin mono-oxygenase, and DMT1
motifs, such as CXXCH in cytochrome c, are has been proposed.13 This complex, known as
found in HCP1 (see references 46 and 47). paraferritin, was shown to have ferrireductase
Expression of HCP1 in Xenopus oocytes and activity capable of reducing the newly transported
HeLa cells results in a 2- to 3-fold increase in heme Fe(III).52 Soluble Fe(II) is then free to join the pool
uptake, which is abolished when the gene is silenced of Fe(II) transported across the plasma membrane
by HCP1-specific siRNA or in the presence of via the DMT1 pathway.
HCP1 antisera. The temperature-dependent and
saturable activity of HCP1 suggests an energy Ferroportin
requirement for heme transport.44 However, this Duodenal basolateral iron export into blood is
energy requirement is presently undefined but may mediated by the transmembrane protein ferroportin
involve a proton or anion gradient similar to the 1 (FPN1) that is also known as iron regulatory
mechanism used by the major facilitator super- transporter 1 and metal transporter protein 1.
family transporters (see reference 45). Heme carrier Ferroportin was simultaneously discovered by
protein 1 is regulated both transcriptionally and 3 groups: one group mapped the gene through
posttranslationally. Expression is induced by positional cloning in zebrafish exhibiting severe
hypoxia, whereas subcellular localization of HCP1 microcytic anemia,53 another performed mRNA
appears to correlate to the iron status in the body. hybridization using hypotransferrinemic mice,54
Under normal conditions, HCP1 is found in both the and the third group enriched a library with IRP-1
apical membrane and cytosol. In response to iron binding.55 Ferroportin 1 is a 571–amino acid protein
deficiency, HCP1 migrates to the apical membrane, with a 90% amino acid sequence identity between
whereas cytosolic distribution peaks with the mice, rats, and humans.54 Encoded by the SLC40 A1
hepatic inorganic iron store.44 Induction of heme gene,56 FPN1 is also found on the basal membrane
oxygenase 1 expression by CdCl2 also causes an of the placental syncytiotrophoblast as well as in
increase in HCP1 expression and heme uptake. macrophages including the Kupffer cells in the liver,
Thus, it is proposed that HCP1 expression and spleen, and hepatocytes.53 Ferroportin 1 is the sole
function are proportional to the rate of heme iron exporter in tissues and cells where major iron
110 BLEACKLEY ET AL

flow is regulated57 including dietary iron transport linked anemia, it was found that sex-linked anemia
into the blood, transport of iron from mother to fetus, mice had a 582–base pair deletion in the heph gene
and iron export after the degradation of hemoglobin resulting in a 194–amino acid truncation of the Hp
from recycled RBCs.58 Experiments in Xenopus protein.64,65 Although apical uptake of iron was
oocytes demonstrated the iron export ability of normal, the deletion resulted in impaired basolateral
FPN154; however, there is still some debate whether intestinal iron transport, causing moderate to severe
an associated ferroxidase is necessary for transport. microcytic hypochromic anemia.64,66 Hephaestin
Studies indicating a colocalization of FPN1 with a was proposed as the limiting factor in intestinal iron
glycosylphosphatidylinositol (GPI)-linked form of export. Significant sequence homology with Cp and
the ferroxidase ceruloplasmin (Cp) facilitates iron the yeast homologue Fet3 (see Fig 1) led to
efflux from brain astrocytes59 together with the predictions of multicopper oxidase activity.60,67
colocalization of the ferroxidase Hp with FPN1 on Multicopper oxidase activity was confirmed through
the basolateral membrane of duodenal enterocytes60 complementation of the low-iron growth defect
suggest that a protein complex is formed in vivo. Δfet3 yeast strains. Hephaestin is primarily expressed
However, FPN1 retains iron export ability even in in the small intestine in contrast to expression of Cp
the absence of a ferroxidase.53 Upregulation of in the liver. Similar to Fet3, Hp also possesses a C-
FPN1 expression may be mediated by the presence terminal TM domain which anchors the protein to the
of an IRE in the 5′UTR of the FPN1 mRNA.61 The basolateral membrane.68 It is known that high affinity
prepropeptide hepcidin controls blood iron levels by iron uptake in yeast is mediated by a 2-component
binding FPN1 and inducing endocytosis and interaction between ferroxidase Fet3p and the iron
degradation as a homeostatic mechanism.62 Cou- permease Ftr1p, providing a paradigm for mamma-
pling of Tf receptor (TfR)–mediated endocytosis of lian iron transport.69
plasma iron through the intestinal basolateral
membrane with hepcidin-mediated FPN1 endocy- Ferroportin-Hephaestin Interactions
tosis and degradation under iron replete or overload A study on glioma cells and astrocytes has shown
conditions leads to the iron loading, and subsequent that an additional function of multicopper oxidases
shedding of intestinal epithelial cells into the is to provide stability for FPN1 at the cell surface.70
digestive tract at the end of their 1- to 2-day lifespan Loss of GPI-linked Cp resulted in loss of FPN1-
has been hypothesized.63 Thus, iron excretion in the mediated iron transport as well as increased FPN1
stool also plays a role in iron homeostasis. Studies in internalization and ubiquitination leading to degra-
zebrafish have shown that loss of FPN1 expression dation. In addition, lowering the extracellular
is embryonic lethal, with rescue only by supple- ferrous iron levels by ferroxidases played a large
mentary iron injections. However, rescued zebrafish role in the stability of FPN1 at the cell surface.70
develop severe iron deficiency anemia as a result of This suggests that the steep Fe(II) concentration
the inability to absorb dietary iron.53 Mice with total gradient formed by Hp may not only promote
postnatal ablation of FPN1 and intestine-specific efficient iron transport, but may also stabilize the
ablation of FPN1 both present with severe systemic FPN1 transport complex at the basolateral surface.
iron deficiency and iron-loaded enterocytes, provid- Hephaestin colocalizes with FPN1 on the basolat-
ing insight into the importance of FPN1 in intestinal eral surface of intestinal absorptive cells, suggest-
iron absorption.61 The role of FPN1 in dietary iron ing that Hp and FPN1 may interact in a similar
metabolism has shed light on previously unex- manner.60 It is still not known whether FPN1 is iron
plained cases of hemochromatosis. Ferroportin-1 specific or whether Hp association provides
mutation causing iron overload disease is now called specificity.71 The requirement for copper in Cp
type IV hemochromatosis or ferroportin disease.58 and Hp function emphasizes the importance of
copper in dietary iron uptake and iron mobilization
Hephaestin from tissues.72 Currently, the role of Hp in dietary
Hephaestin is a membrane bound multicopper iron uptake is not clear.73 Functional studies on the
oxidase that is highly expressed in the basolateral role of Cp on iron mobilization from iron storage
membrane of the duodenal enterocytes and, to a cells (eg, reticuloendothelial system) showed that
lesser extent, in the colon, brain, spleen, lung, and ferroxidase activity generates a steep concentration
placenta.64 Originally implicated in murine sex- gradient of ferrous iron. This in turn allows for
IRON IMBALANCE AND DISEASE 111

highly efficient iron efflux, providing some insight Fenton reaction, the sequestering of iron is funda-
to the possible role of Hp in intestinal iron transport. mental to the prevention of cell toxicity.6 Once
Hephaestin ferroxidase activity may also be bound to Tf, Fe(III) is also unavailable for the
required to efficiently load exported iron onto Tf growth of most pathogens.78 Human serum Tf
which carries only Fe(III). circulates through blood at very high concentrations
(35-50 μmol/L). However, about 70% of circulating
IRON IN BLOOD Tf is in an iron-deplete state and available to act as a
Red Blood Cells buffer against excess free iron.79 Transferin delivers
iron to cells via an interaction with the TfR. The
Red blood cells are the predominant form of importance of these proteins in iron homeostasis
blood cells in humans. Mature mammalian RBCs was demonstrated by using knockout mice80,81,
are fully differentiated anucleate cells that do not with gene deletions in Tf and TfR causing death
possess the subcellular organelles required for shortly after birth and in utero, respectively.
protein or lipid biosynthesis. 30 Before RBCs
reach maturity, they must gather enough iron for Ceruloplasmin
hemoglobin production. Hemoglobin, the major Ceruloplasmin belongs to the multicopper oxi-
iron carrying metalloprotein in humans, makes up dases family and plays a vital role in mammalian
more than 95% of the dry weight of RBCs.74 More iron homeostasis as the major multicopper ferrox-
than two thirds of total iron in a normal human idase in blood.82 Ceruloplasmin requires 6 copper
being is bound to hemoglobin in resting condi- atoms for functionality, accounting for more than
tions.46 The major physiologic role of hemoglobin 95% of the copper in human plasma. Ceruloplasmin
is oxygen transport as hemoglobin represents the is postulated to have several physiologic functions
only method of oxygen transport and delivery from besides ferroxidase activity, including activity as a
lungs to peripheral tissues and cells. In addition to glutathione peroxidase, an ascorbate oxidase, and
the 4 globin polypeptides, each hemoglobin an antioxidant, as well as roles as a copper
molecule contains 4 iron-containing heme prosthe- transporter and regulator of cellular iron concentra-
tic groups, each of which reversibly binds an tions.83 Ceruloplasmin functions in iron home-
oxygen molecule. More than 80% of the iron ostasis by controlling the rate of cellular iron
transported daily by Tf is used in heme synthesis. efflux.84 In the absence of Cp, cellular iron egress is
The majority of this iron comes from senescent inhibited with subsequent sequestration of iron
RBCs that have been erythrophagocytosed by within cells.84 This regulatory function of Cp is
macrophages. 74 Iron deficiency compromises connected with its role in loading apo-Tf with Fe
RBC production and can result in a microcytic (III), a process that requires the catalytic oxidation
hypochromic anemia. The principal regulator in the of Fe(II) released from cells.77 However, it is
rate of eythropoiesis is the glycoprotein hormone perhaps its biological function within the central
erythropoietin, the broad scope of which is beyond nervous system that Cp is most noted. Deficiencies
this review (reviewed in references 46, 75, and 76). in Cp result in iron accumulation within the basal
ganglia that can lead to progressive neurodegen-
Transferrin erative disease.85 Aceruloplasminemia is an auto-
Although most of the human body's iron is found somal recessive disorder caused by a loss-of-
within circulating RBCs, Tf is the primary iron function mutation in the gene for Cp.86 Affected
transport protein in blood. Transferrin is composed individuals have a complete absence of functional
of a single polypeptide composed of 2 homologous serum Cp and elevated levels of serum ferritin and
globular lobes (the N-lobe and C-lobes) that are can also present with anemia, diabetes, retinal
each divided into 2 subdomains. Each lobe contains degeneration, and neuronal defects.87 The role of
a metal-binding cleft that is capable of binding one Cp in brain iron metabolism and neuronal survival
iron atom. Transferrin binds Fe(III) very tightly has been reviewed recently.82
with a Kd of ∼10−24 mol/L at physiologic pH.77
Owing to the insoluble nature of free Fe(III) under Iron Recycling by Macrophages
physiologic conditions and the potential for free Macrophages are phagocytic white blood cells
iron to catalyze superoxide radical formation via the with roles in innate and cell-specific immunity.
112 BLEACKLEY ET AL

Macrophages also play an important role in iron receptor binding to endosomal release occurs in less
recycling. Plasma iron levels are largely regulated than 3 minutes.93
by the rate of iron release from macrophages.63
Macrophages extract iron from hemoglobin that has Lactoferrin and Melanotransferrin
been acquired from phagocytosed senescent RBCs. The role of Tf homologs in iron delivery has
Iron is transported into macrophages through always been poorly understood. Lactoferrin (Lf)
phagocytic vacuoles that contain a divalent metal and melanotransferrin (MTf) share 37% to 39%
transporter, Nramp1, a homolog of the duodenal sequence similarity with Tf. Unlike soluble Tf, the
transporter DMT1.88 Nramp1 transports iron to the majority of MTf is tethered to the cell membrane by
cytoplasm of the macrophage where it is seques- a GPI anchor.100 However, a small amount of
tered until needed. Ferroportin-1, the duodenal iron soluble MTf (sMTf) exists in serum, saliva,
exporter, also functions to mobilize iron from cerebrospinal fluid, and urine.101 Another differ-
macrophages. 63 Macrophage iron recycling is ence between Tf and MTf occurs with iron binding;
regulated by hepcidin levels. Hepcidin interacts MTf contains only one Fe-binding site.102 The
with FPN1 causing its internalization and degrada- tissue expression patterns of MTf and sMTf are
tion.89,90 The degradation of FPN1 results in iron quite different from Tf and TfR.101 Melanotrans-
sequestering in macrophages.91 The iron-binding ferrin was originally identified at high levels in
properties of macrophages also play an important melanoma cells and fetal tissue.101,103,104 More
role during infection, as iron sequestration by host recently, MTf has been found highly expressed in
macrophages can suppress pathogen proliferation.92 an array of tissues including brain, salivary gland
epithelium, pancreas, testis, kidney, and sweat
IRON UPTAKE BY OTHER CELLS gland ducts.105 The most significant difference
between these homologs and Tf is that neither
Transferrin Receptor 1 membrane-bound MTf101 nor sMTf106 has been
Cellular iron uptake is a well-characterized, pH- shown to bind to TfR1 and deliver iron to cells.
dependent process of receptor-mediated endocyto- Knockout mice, with a gene deletion for MTf,
sis.93 The TfR is a ubiquitous protein, anchored to showed no morphological, histologic, behavioral,
cell membranes by a short cytoplasmic and TM N- hematological, or Fe status variations compared to
terminal region. The TfR forms a homodimer, wild-type mice.107 Although several hypotheses
joined by 2 intermolecular disulfide linkages in this have been proposed for potential MTf functions,107
anchored region. The soluble extracellular domain it is probable that MTf does not play a significant
of the receptor extends into the plasma and is role in Fe transport and delivery.108
available for Tf binding. Fe(III)-bound Tf enters Lactoferrin, a soluble homologue of Tf, pos-
cells by preferentially binding to the TfR with high sesses both Fe-binding sites and is able to bind Fe
affinity (KD, ∼0.1-10 nmol/L).94-96 The Tf-TfR (III) with a higher affinity than Tf.109 Human Lf is
complex is internalized by endocytosis into a expressed in glandular epithelial cells and found in
clathrin-coated pit. The endosome is acidified to breast milk and other mucosal secretions.110 There
pH ∼5.6 through the action of an ATPase pump.94 exists some debate regarding the role of the Lf in
At this lower pH, Fe(III) is released from Tf, iron metabolism. Like the MTf knockout, deletion
whereas apo-Tf remains bound to the receptor. The of the Lf gene results in no change in phenotype.111
removal of Fe(III) from Tf is a complex and Although Lf has been postulated to have a function
incompletely understood process involving salt in neonate iron absorption, it is generally believed
concentration, pH, and a potential unknown to have a larger role in immunity acting as an
chelator inside the endosome.97 Upon release, it is antimicrobial agent.110
thought that Fe(III) is reduced to Fe(II) by the
ferrireductase Steap3.98 The Fe(II) can then be INTRACELLULAR IRON METABOLISM
transported across the endosomal membrane by Once taken into a cell, iron has 2 possible fates:
DMT1 and released into the intracellular environ- incorporation into iron proteins usually as heme or
ment.99 The Tf-TfR complex is recycled to the Fe-S clusters, or storage in ferritin for later use
outside of the cell and apo-Tf is released. The entire during iron deficiency. Intracellular iron proteins
process of cellular iron uptake from extracellular have a wide range of functions. Cytochromes
IRON IMBALANCE AND DISEASE 113

including those in the electron transport chain and L types in varying ratios depending on the type of
cytochrome P450s in the oxidative metabolism of cell and physiologic conditions. Iron is stored in the
drugs and other compounds112 are heme proteins. lumen of the multisubunit sphere. Up to 4500 iron
Aconitase, the enzyme responsible for isomeriza- atoms can be stored in each ferritin multimer. As
tion of citrate to aconitate, is an Fe-S protein, iron is transported into the ferritin multimer core, it
whereas other Fe-S proteins including hydroge- is oxidized from Fe(II) to Fe(III) by the H subunit
nases act as electron carriers. Ribonucleotide allowing formation of nonreactive ferrihydrate
reductases, the enzymes that control the pool of which assembles as a solid in the core in a process
deoxyribonucelotides for DNA synthesis, are involving the L-subunit.134-136 Although ferritin is
examples of nonheme iron proteins.113 Nonheme a predominantly cytoplasmic protein, recent evi-
iron proteins contain iron but not complexed in dence has shown that it can also be found in the
heme or Fe-S clusters. nucleus137 and mitochondria.138 A small quantity
of glycosylated ferritin is secreted into the
Mitochondrial Iron Metabolism serum.139 Cytoplasmic ferritin has been shown to
Both heme and Fe-S clusters are synthesized in associate with microtubules.140 Studies into the
mitochondria making this organelle a major target for distribution of microtubule-associated ferritin com-
intracellular iron metabolism. Mitoferrin, encoded bined with evidence that association with micro-
by the mfrn gene, is a mitochondrial membrane tubules increases ferritin iron binding in vitro
protein responsible for iron uptake into the mito- support a role for microtubules in the distribution
chondria for heme biosynthesis during eythropoi- of intracellular ferritin and thus iron metabolism as
esis.114 A second gene, mfrn2, encodes a similar a whole.141
protein that is sufficient to provide basal level of iron
required by nonerythroid cells.115 Characterization Regulation of Iron Metabolism
of the orthologous pair MRS3/4 in S cerevisiae The cell must regulate all stages of iron
provided much of the basis for the identification and metabolism including uptake, storage, and utiliza-
characterization of mfrn/mfrn2.116-119 Inside the tion. In vertebrates, expression of proteins such as
mitochondria, iron is bound by the mitochondrial TfR and ferritin is regulated by cellular iron levels at
iron chaperone frataxin (FXN),120 a protein that the posttranscriptional level.142 Briefly, mRNAs of
forms multimers with iron stored in the core.121 some proteins of iron metabolism contain IREs in
Frataxin is involved in mitochondrial iron storage the UTRs. Iron response elements are 30-nucleotide
while preventing the formation of damaging free hairpin structures that are conserved in vertebrates
radicals,122-124 maintaining a pool of Fe(II) in the and some insects and bacteria. Under iron-starved
mitochondria, and delivering iron to sites of heme conditions 2 IRP1 and IRP2 bind to the IREs. In
and Fe-S cluster assembly.125-127 Monomeric FXN mRNAs for proteins involved in iron uptake such as
has been shown to bind iron and interact with TfR, the IREs are found in the 3′UTR; IRP binding
proteins involved in heme and Fe-S cluster synthesis to IREs in the 3′UTR stabilizes the mRNA
providing an alternate model to the multimeric model increasing translation of the proteins. Proteins
previously described.128 Mutations in the FXN gene such as ferritin that are involved in sequestering
cause the neurodegenerative disorder Friedreich's iron have IREs in the 5′UTR; IRP binding to 5′IREs
ataxia (FRDA). 129 Studies of the yeast FXN results in translational inhibition and decreased
homolog Yfh1 have contributed significantly to the synthesis of the proteins. RNA binding activity of
understanding of the role of FXN in mitochondrial IRP1 is regulated by the formation of an Fe-S cluster
iron metabolism. Heme and Fe-S cluster synthesis which inhibits IRE binding in iron replete condi-
are both complicated procedures, and have been tions.143 The IRE/IRP regulation of iron metabolism
reviewed recently.130-133 has been reviewed recently.144,145

Ferritin Intracellular Iron Transport


Ferritin is the major intracellular iron storage Intracellular iron transport between sites of
protein in animals and prokaryotes. Vertebrate uptake, storage, and utilization is not well under-
ferritin, which is expressed ubiquitously, is a stood. Iron that is not complexed into proteins, not
hollow sphere composed of 24 subunits of H and stored in ferritin, nor localized to the mitochondria
114 BLEACKLEY ET AL

has been termed the labile iron pool (LIP). The intracellular iron level lowers the expression of
oxidation states of iron in the LIP remain unclear. DMT1, resulting in a decrease in iron uptake.35,158
Evidence supports the hypothesis that the LIP is Hepcidin expression is modulated at the tran-
bound to low-molecular-weight proteins as scription level by multiple factors including inflam-
opposed to small molecules such as citrate.146,147 mation, Tf saturation, iron deficiency, and hypoxia.
Intracellular copper transport occurs via a number In response to inflammation, the cytokine inter-
of copper metallo-chaperone proteins.148-150 From leukin-6 interacts with its receptor on hepatocyte
these 2 lines of evidence, some have hypothesized membranes and activates Stat3, which upregulates
that intracellular iron is transported via protein hepcidin expression by binding to a regulatory
chaperones in a manner similar to copper, and it is element on the hepcidin gene.159-161 A better
these small iron chaperone proteins that are being characterized pathway for regulation of hepcidin
detected in the analysis of the LIP. In erythroid expression involves the interaction of hemojuvelin
cells, where 85% of heme synthesis occurs, (HJV) with the bone morphogenetic protein (BMP)/
inhibition of endosome mobility decreases the Smad signaling pathway. 162 Hemojuvelin is
incorporation of 59Fe into heme, supporting the expressed by liver, heart, and skeletal muscles,163
hypothesis that there is a direct interaction between and is found in 2 forms: GPI-anchored HJV (mHJV)
iron-loaded vesicle and the mitochondria in and soluble HJV (sHJV).164 The 2 forms recipro-
RBCs.130,151 However, vesicular trafficking cannot cally regulate hepcidin expression in response to the
be the only form of intracellular iron transport. In a iron requirement of the body. As a BMP coreceptor,
S cerevisiae strain with a deletion of end4 which is mHJV interacts with BMP2/BMP4, enhancing
defective in endocytosis, iron is transported from BMP binding to type I and type II receptor serine/
the cytosol to the vacuole for storage (S cerevisiae threonine kinases. Type II receptor kinase activates
lack an expressed ferritin gene and store iron in the type I receptor kinase, which in turn phosphorylates
vacuole).152 Thus, another intracellular iron trans- receptor-regulated Smad. Phosphorylated receptor-
port mechanism must exist, and this remains the regulated Smad complexes with Smad4 and the
subject of intense investigation. complex translocates into the nucleus promoting
hepcidin transcription.165,166 Soluble HJV com-
REGULATION OF IRON ABSORPTION petes for binding to BMP2/BMP4 against mHJV to
inhibit BMP/Smad signaling and, hence, blocks
Hepcidin and Hemojuvelin hepcidin production.159,166
Hepcidin is synthesized by the liver as a Hepcidin expression is determined by the relative
prepropeptide consisting of 84 amino acid residues. levels of mHJV and sHJV. Expression of both forms
Removal of a 24–amino acid signal peptide and of HJV depends on the level of Tf-bound iron but not
cleavage at Arg-59 by furin result in the release of other chelated iron.166 The release of mHJV from
the active peptide hormone that is 25 amino acids in membrane upon interaction with its receptor,
length.153 Hepcidin is rich in cysteine residues and neogenin, is suggested to downregulate hepcidin
forms a hairpin with β-sheet structure stabilized by 4 expression and is inhibited by holo-Tf.167 Contrarily,
disulfide bonds.154 Hepcidin was identified as a iron deficiency and hypoxia stimulate sHJV produc-
negative regulator of iron absorption based on iron tion by stabilizing hypoxia inducible factor-1 α.
level and distribution in hepcidin-deficient or Hypoxia inducible factor-1 α induces the expression
-overexpressing mouse models.155,156 Hepcidin of furin, which cleaves HJV at Arg-335 and releases
downregulates iron absorption by binding FPN1 the mature sHJV into cell culture media.168
inducing its internalization and degradation.62 After
hepcidin incubation, FPN1 is phosphorylated at 2 Hemochromatosis Protein
adjacent tyrosine residues (T302-T303) in an The precise role of hemochromatosis protein
extracellular loop between TM6 and TM7.157 The (HFE) in the regulation of iron absorption remains
trafficking of internalized FPN1 through multi- to be elucidated. Hemochromatosis protein is
vesicular body to lysosomes for degradation expressed ubiquitously at low levels in tissues169
involves the protein complex known as the Endo- and is highly expressed in intestinal crypt cells and
some Sorting Complex Required for Transport. As a liver.170 The level of HFE expression inversely
secondary effect of FPN1 downregulation, increased correlates to the level of iron uptake.171 The
IRON IMBALANCE AND DISEASE 115

absence of HFE is associated with iron overloading, regulate duodenal iron absorption directly.171 Ecto-
whereas overexpression of HFE reduces iron pic expression of HFE increases HFE-β2M apical
absorption (reviewed in reference 172). The HFE distribution, which in turn reduces the rate of apical
polypeptide is composed of 3 extracellular loops iron absorption.183
(α1, α2, α3), a TM domain, and a short cytosolic
tail. Its 3-dimensional structure is stabilized by 2 DISEASES OF IRON IMBALANCE
disulfide bonds, one of which gives rise to the α3 Divalent Metal Transporter 1 Deficiency
loop for noncovalent interaction with β2 micro-
globulin (β2M).173,174 The HFE-β2M interaction is Mutations of transporter proteins involved in
essential for HFE translocation across the plasma dietary iron uptake result in anemia. DMT1
membrane for cell surface expression of the mutations are autosomal recessive and cause
complex. 174 The HFE-β2M complex interacts hypochromic microcytic anemia and hepatic iron
with the helical domain of TfR1.95,175,176 Coloca- accumulation. Defective DMT1 loses its function
lization95 and interaction177 with TfR2, which is owing point mutations, in-frame deletion of a single
selectively expressed in crypt cells, have also been amino acid, or the complete deletion of an exon due
reported recently. to alternative splicing. Mutations that give rise to the
Hemochromatosis protein is believed to act as a exon skipping DMT1 variants do not completely
sensor of the iron status in the body. Various models remove DMT1 function.184-186 DMT1 exon 12
regarding HFE-mediated regulation of iron home- skipping normally occurs at low levels (∼10%
ostasis have been postulated. Studies have indicated mRNA); however, the DMT1 G1285C mutation
that HFE sequesters TfR1 when Tf saturation falls increases the tendency of exon skipping to ∼90% of
within the normal range39 but competes with holo-Tf DMT1 mRNA, giving a nonfunctional splice variant
for binding to TfR1 with increased Tf saturation.178 that lacks TM8. The remaining full-length mRNA
Thus, HFE is proposed to sense the body's iron encodes a fully functional mutant DMT1, where a
requirement based on the level of Tf saturation with glutamate is replaced by aspartate in an intracellular
iron. The original model assumed that HFE forms a loop. The small amount of functional DMT1 is
heterotrimer with β2M and TfR1 in the basolateral thought to be responsible for hepatic iron overload
membrane of enterocytes and signals expression of observed in the patients.187 As discussed previously,
iron transport proteins through the action of IRP,179 PCFT/HCP1 is shown to be involved primarily in
or in the plasma membrane of hepatocytes to folate absorption rather than heme absorption. No
modulate iron absorption by inducing hepcidin mutation in PCFT/HCP1 that affects heme uptake
production.180 A recent study demonstrated the has been identified to date. However, mutation of
interaction between HFE and TfR2.177 Because PCFT/HCP1 has been shown to lower the stability
TfR2 is distributed in lipid rafts, it is believed to play or affect membrane trafficking of the protein,
a role in signal transduction upon holo-Tf binding. In impairing folate dietary absorption and transport to
stimulated erthroleukemic cells, TfR2 activates central nervous system. Folate deficiency leads to
extracellular signal-regulated kinases 1 and 2 and erythroblast apoptosis, resulting in megaloblastic
p38 mitogen-activated protein kinases.181 The latest anemia from compromised erythropoiesis.74,188
model for the mechanism of action of HFE, again,
presumes HFE-β2M to be involved in sensing Tf Hemochromatosis
saturation and is complexed to TfR1 under normal or Hereditary hemochromatosis is a primary iron
low serum iron condition. As Tf saturation increases, overload disease with consequent tissue damage
competitive binding to TfR1 occurs between HFE from iron-induced oxidative stress. Hereditary
and holo-Tf. The resulting free HFE-β2M then binds hemochromatosis is caused by one or more
TfR2 to signal hepcidin production via the extra- mutations in genes encoding proteins that are
cellular signal-regulated kinase/mitogen-activated involved in maintaining iron homeostasis. Hemo-
protein kinase pathway.181,182 However, the require- chromatosis type I is caused by mutations in HFE
ment for HFE in TfR2-mediated signal transduction with most patients being homozygous for the
is unknown.177 Colocalization of HFE-β2M with C282Y mutation. C282Y disrupts a disulfide
DMT1 in apical membrane of enterocytes during bond required for the formation of the α3 domain,
iron deficiency implies that HFE may function to disrupting HFE-β2M interaction and thereby
116 BLEACKLEY ET AL

inhibiting surface expression of the protein.169 Not with FPN1 on astrocyte surface suggests a role in
all individuals who are homozygous for HFE iron transport in the brain.59 Ceruloplasmin is
C282Y manifest iron overloading, suggesting essential for brain iron metabolism and neuronal
incomplete penetrance of this mutant. 172 Iron survival and functions as an antioxidant by
overloading due to mutations in HJV or hepcidin preventing Fe(II)-induced lipid peroxidation and
is known as hemochromatosis type II or juvenile reactive oxygen species (ROS) generation. The
hemochromatosis. Juvenile hemochromatosis is a level of iron accumulation parallels the extent of
severe form of hereditary hemochromatosis and is astrocyte deformity and formation of globular
characterized by an early onset, usually before structures within these cells. Fe(II)-stimulated free
30 years of age. The majority of type II cases are radical production and lipid peroxidation, as
caused by mutations in HJV. A rare form of indicated by a significant increase in the levels of
hemochromatosis type III is associated with muta- 4-hydroxynonenal and malondialdehyde (markers
tions in TfR2 which is implicated in regulation of of lipid peroxidation) in astrocytes, cause glial cell
iron transport by interacting with HFE. Patients death. The loss of glia function lowers the survival
with one of the 3 types of hemochromatosis of neurons and accentuates oxidative neurodegen-
discussed above show autosomal recessive inheri- eration with Fe(II)-induced oxidative stress.192
tance and are found to have abnormally low levels
of hepcidin, suggesting that these mutations act NEURODEGENERATIVE DISEASES RESULTING
through a common mechanism.189 Unlike hemo- FROM THE LOSS OF REGULATION OF
chromatosis types I, II, and III, hemochromatosis INTRACELLULAR IRON
type IV results from mutations in FPN1 and is also
known as ferroportin disease.71,190 Ferroportin-1 FriedreichTs Ataxia
mutations display autosomal dominant inheritance Friedreich's ataxia is an autosomal recessive
and are heterozygous in clinical presentation. All neurodegenerative disease characterized by degen-
FPN1 mutations reported to date involve either a eration of Purkinje neurons of the cerebel-
single base change or a deletion. Recently, FPN1 lum.194,195 It is the most common hereditary
has been characterized as a homodimer which ataxia among whites with a prevalence of 1 to 2
probably explains in part the autosomal dominant in every 50 000 people.196 The majority (∼97%) of
nature of ferroportin disease as the mutant subunit patients with FRDA are homozygous for an
may affect the assembly of the dimer and possibly abnormal expansion of GAA triplet repeats in
the subsequent membrane localization or response intron 1 of the FXN gene, whereas the remainder
to hepcidin regulation.191 are compound heterozygotes for a point mutation or
microdeletion in one allele and a GAA repeat
Aceruloplasminemia expansion in the other.197 The extra repeats in the
Aceruloplasminemia is an iron metabolic dis- FXN gene are shown to interfere with transcription
order caused by recessive mutations in the Cp gene by forming a triplex DNA structure called “sticky
that result in a complete absence of Cp ferroxidase DNA,” and the size of the repeats parallels the time
activity. Some mutations in Cp cause premature of onset and the rate of FRDA progression.198
termination of Cp translation. The resulting trun- Frataxin deficiency causes iron accumulation in
cated product is not or partially copper loaded.192 the mitochondria, reduction in iron-sulfur cluster–
Patients with aceruloplasminemia are anemic dependent enzymes, and altered antioxidant
owing to the low serum iron concentration and defense. 195 The excess iron in mitochondria
high serum ferritin level. Despite the low serum displaces manganese in superoxide dismutase and
iron level, iron accumulates in the brain and visceral renders the enzyme inactive.199 Superoxide dis-
organs such as the liver and pancreas. Excess iron mutase inactivity causes oxidative stress and lipid
deposition in the brain leads to loss of neurons in peroxidation in patients with FRDA, as indicated
basal ganglia and Purkinje cells in cerebellar by increased levels of urinary 8-hydroxy-2′-deox-
cortex.193 As discussed in detail earlier, Cp is a yguanosine, a marker for oxidative DNA damage,
multicopper ferroxidase that is synthesized by and plasma malondialdehyde, respectively.198 The
astrocytes in the mammalian central nervous effect of decreased frataxin is not limited to the
system as a GPI-anchored protein. Colocalization mitochondria as FXN deletion in mouse tissues is
IRON IMBALANCE AND DISEASE 117

recently shown to impair extramitochondrial iron- precursor protein (APP) whose translation may be
sulfur proteins.200 regulated by intracellular iron levels via a putative
IRE in the 5′UTR of APP.208 At low concentra-
ParkinsonTs Disease tions, Aα exhibits antioxidant activity; however,
Parkinson's disease results from the death of Aα is toxic to the cells at high concentrations.
dopamine-producing neurons in substantia nigra. Accumulation of Aα in patients with Alzheimer's
Parkinson's disease is characterized by the pre- disease is thought to be caused by increased APP
sence of insoluble fibrillar structures, called Lewy production or inefficient clearance of Aα. Fe(III)
bodies, in the presynaptic nerve terminals.201 A binds to Aα at high affinity and induces its
major component of Lewy bodies is α-synuclein, a aggregation, whereas heme binds and prevents Aα
small acidic protein of 140 amino acid residues.202 aggregation. Fe(III)-bound Aα plaques possess
Native α-synuclein displays an unfolded random high reduction potential and reduce Fe(III) to Fe
coil structure. Interaction with factors such as (II) at high efficiency. The reduced iron reacts with
proteins, lipids, oxidants, or metal ions promotes trapped oxygen in the senile plaques to ROS via
α-synuclein folding and aggregation, leading to Fenton reactions. Similarly, the heme-Aα complex
Lewy body formation.203 A potential IRE in the 5′ functions as a peroxidase to produce ROS.209,210
UTR of α-synuclein has been described, indicating Met35 of Aα is believed to serve as an electron
that iron may also function to regulate α-synuclein donor in the generation of hydrogen peroxide from
production at the translational level.204 Iron oxygen as oxidized-Met35-Aα is isolated from
accumulates at the site of substantia nigra degen- amyloid plaques. The removal of hydrogen per-
eration in patients with Parkinson's disease.205 The oxide inhibits Aα neurotoxicity, suggesting that Aα
precise mechanism through which iron promotes plaques cause neurodegeneration through ROS
neurodegeneration is not known. However, it has production. As a result of free radical reactions,
been postulated that iron interacts with α-synuclein irreversible dityrosine linkages are formed between
to induce its oligomerization through the genera- Aα. Thus, it has been hypothesized that toxicity
tion of free radicals, resulting in Lewy body may require oxidative modification of Aα.211
formation.206 A recent study has also shown that
Fe(III) promotes the production of short and thick Neuroferritinopathy
fibrils when it interacts with both wild-type and Neuroferritinopathy is a cognitive and motor
mutant α-synuclein.203 Although harmful, iron disorder caused by neuronal degeneration and free
cannot be eliminated entirely from these dopami- radical toxicity from iron and ferritin deposition in
nergic cells because iron is required for tyrosine the brain, particularly the basal ganglia. Neurofer-
hydroxylase activity during the synthesis of ritinopathy is a dominant disorder in which patients
dopamine; iron is also involved in the conversion have a single nucleotide insertion in the carboxyl
of excess dopamine to neuromelanin. However, terminal of the ferritin light chain. Insertion of an
when bound to excess iron, neuromelanin acquires adenine at position 460 to 461 or thymine/cytosine
ferrireductase activity and converts Fe(III) to Fe(II) at position 498 to 499 in exon 4 of the ferritin light
which is released owing to its poor affinity for chain results in a frame shift causing formation of a
neuromelanin. Iron-induced oxidation of dopamine new stop codon that is downstream of the normal
releases hydrogen peroxide, which reacts with Fe position. Consequently, this replaces the last 22 or 9
(II) via the Fenton reactions to generate harmful amino acids with a different 26 or 25 amino acids,
radicals which can cause cell death.205 respectively.212,213 Based on protein modeling data,
the C-terminal of ferritin light chain is predicted to
Alzheimer's Disease be involved in pore formation in the ferritin
The main features of Alzheimer's disease are the dodecahedron complex. The mutant C-terminal
formation of senile neuritic plaques and neurofi- sequence likely affects ferritin subunit interaction
brillary triangles. Senile neuritic plaques are and stability, and thereby increases iron availability
composed of insoluble α-amyloid peptide (Aα) and the cell's sensitivity to oxidative stress.214 The
which aggregates spontaneously in the presence of exact mechanism through which mutations in
divalent metals, such as Zn(II), Cu(II), and Fe ferritin light chain induce neurodegeneration is
(II).207 Aα is a cleavage product of amyloid β undefined. However, it has been postulated that the
118 BLEACKLEY ET AL

incorporation of mutant light chains partially However, the chemical reactivity that enables iron
compromises the structure and function of ferritin. to be used in metalloproteins also requires careful
Iron and ferritin aggregation suggests that the regulation to avoid cellular toxicity. Over the past
mutant subunit may promote the release of iron decade, many advances have been made in
by ferritin, leading to oxidative stress and cell identifying new proteins that are involved in iron
death.205 It has been shown that ferritin light chain homeostasis allowing some of the intricacies of the
level is not critical in the pathogenesis of complex regulation of iron to be elucidated. There
neuroferritinopathy as neurodegeneration is unde- are still many facets of iron metabolism that have
tectable in an individual with decreased ferritin yet to be fully understood. However, the application
light chain expression due to a heterozygous start of genetic screens in yeast and the use of knockout
codon mutation.215 mice have become valuable tools for the discovery
of iron-related pathways in yeast that are conserved
CONCLUDING REMARKS in humans. With more research, the discoveries
Because of its roles in oxygen transport, the outlined in this review will lead to new approaches
respiratory chain, and deoxyribonucleotide bio- for the identification, diagnosis, and treatment of
synthesis, iron is an essential element in blood cells. diseases related to imbalances in iron metabolism.
REFERENCES
1. Crichton RR, Pierre JL: Old iron, young copper: From 16. Skikne BS, Lynch SR, Cook JD: Role of gastric acid in
Mars to Venus. Biometals 14:99-1128 2001 food iron absorption. Gastroenterology 81:1068-1071, 1981
2. Harris WR: In: Templeton D, editor. Molecular and 17. Conrad ME, Umbreit JN: Iron absorption and transport
cellular iron transport. New York, Marcel Dekker Inc., 2002, —An update. Am J Hematol 64:287-298, 2000
pp 1-40 18. Umbreit JN, Conrad ME, Hainsworth LN, et al: The
3. Pierre JL, Fontecave M: Iron and activated oxygen ferrireductase paraferritin contains divalent metal transporter as
species in biology: The basic chemistry. Biometals 12:195-199, well as mobilferrin. Am J Physiol Gastrointest Liver Physiol
1999 282:G534-G539, 2002
4. Lippard SJ, Beng JM: Principles of Bioinorganic 19. Fleming MD, Trenor III CC, Su MA, et al: Microcytic
Chemistry. Mill Valley (Calif), University Science Books; 1994 anaemia mice have a mutation in Nramp2, a candidate iron
5. Radisky D, Kaplan J: Regulation of transition metal transporter gene. Nat Genet 16:383-386, 1997
transport across the yeast plasma membrane. J Biol Chem 274: 20. Gunshin H, Mackenzie B, Berger UV, et al: Cloning and
4481-4484, 1999 characterization of a mammalian proton-coupled metal-ion
6. Aisen P, Enns C, Wessling-Resnick M: Chemistry and transporter. Nature 388:482-488, 1997
biology of eukaryotic iron metabolism. Int J Biochem Cell Biol 21. Gunshin H, Fujiwara Y, Custodio AO, et al: Slc11a2 is
33:940-959, 2001 required for intestinal iron absorption and erythropoiesis but
7. Mason SF: Origin of the Elements, Molecules and dispensable in placenta and liver. J Clin Invest 115:1258-1266,
Living Systems. Oxford, UK, Clarendon Press; 1991 2005
8. Burbridge E, Burbridge G, Fowler W, et al: Synthesis of 22. Lam-Yuk-Tseung S, Govoni G, Forbes J, et al: Iron
the elements in stars. Rev Mod Phys 29:547-650, 1957 transport by Nramp2/DMT1: pH regulation of transport by 2
9. Askwith C, Kaplan J: Iron and copper transport in yeast histidines in transmembrane domain 6. Blood 101:3699-3707,
and its relevance to human disease. Trends Biochem Sci 23: 2003
135-138, 1998 23. Courville P, Chaloupka R, Cellier MF: Recent progress
10. Anderson GJ, Frazer DM, McKie AT, et al: Mechanisms in structure-function analyses of Nramp proton-dependent metal-
of haem and non-haem iron absorption: Lessons from inherited ion transporters. Biochem Cell Biol 84:960-978, 2006
disorders of iron metabolism. Biometals 18:339-348, 2005 24. Cohen A, Nevo Y, Nelson N: The first external loop of
11. Sharp P, Srai SK: Molecular mechanisms involved in the metal ion transporter DCT1 is involved in metal ion binding
intestinal iron absorption. World J Gastroenterol 13:4716-4724, and specificity. Proc Natl Acad Sci U S A 100:10694-10699,
2007 2003
12. Umbreit J: Iron deficiency: A concise review. Am J 25. Li H, Li F, Kwan M, et al: NMR Structures and
Hematol 78:225-231, 2005 orientation of the fourth transmembrane domain of the rat
13. Conrad ME, Umbreit JN: Pathways of iron absorption. divalent metal transporter (DMT1) with G185D mutation in SDS
Blood Cells Mol Dis 29:336-355, 2002 micelles. Biopolymers 77:173-183, 2005
14. Fleming RE: Advances in understanding the molecular 26. Nevo Y, Nelson N: The mutation F227I increases the
basis for the regulation of dietary iron absorption. Curr Opin coupling of metal ion transport in DCT1. J Biol Chem 279:
Gastroenterol 21:201-206, 2005 53056-53061, 2004
15. Bothwell TH, Baynes RD, MacFarlane BJ, et al: 27. Mackenzie B, Ujwal ML, Chang MH, et al: Divalent
Nutritional iron requirements and food iron absorption. J Intern metal-ion transporter DMT1 mediates both H+-coupled Fe2+
Med 226:357-365, 1989 transport and uncoupled fluxes. Pflugers Arch 451:544-558, 2006
IRON IMBALANCE AND DISEASE 119

28. Lee PL, Gelbart T, West C, et al: The human Nramp2 48. Latunde-Dada GO, Takeuchi K, Simpson RJ, et al:
gene: characterization of the gene structure, alternative splicing, Haem carrier protein 1 (HCP1): Expression and functional
promoter region and polymorphisms. Blood Cells Mol Dis 24: studies in cultured cells. FEBS Lett 580:6865-6870, 2006
199-215, 1998 49. Qiu A, Jansen M, Sakaris A, et al: Identification of an
29. Hubert N, Hentze MW: Previously uncharacterized intestinal folate transporter and the molecular basis for hereditary
isoforms of divalent metal transporter (DMT)-1: Implications folate malabsorption. Cell 127:917-928, 2006
for regulation and cellular function. Proc Natl Acad Sci U S A 50. Conrad ME, Umbreit JN, Moore EG, et al: Alternate
99:12345-12350, 2002 iron transport pathway. Mobilferrin and integrin in K562 cells.
30. Hentze MW, Muckenthaler MU, Andrews NC: Balan- J Biol Chem 269:7169-7173, 1994
cing acts: Molecular control of mammalian iron metabolism. 51. Simovich M, Hainsworth LN, Fields PA, et al: Localiza-
Cell 117:285-297, 2004 tion of the iron transport proteins mobilferrin and DMT-1 in the
31. Tandy S, Williams M, Leggett A, et al: Nramp2 duodenum: The surprising role of mucin. Am J Hematol 74:
expression is associated with pH-dependent iron uptake across 32-45, 2003
the apical membrane of human intestinal Caco-2 cells. J Biol 52. Umbreit JN, Conrad ME, Moore EG, et al: Paraferritin:
Chem 275:1023-1029, 2000 A protein complex with ferrireductase activity is associated with
32. Gunshin H, Allerson CR, Polycarpou-Schwarz M, et al: iron absorption in rats. Biochemistry 35:6460-6469, 1996
Iron-dependent regulation of the divalent metal ion transporter. 53. Donovan A, Brownlie A, Zhou Y, et al: Positional
FEBS Lett 509:309-316, 2001 cloning of zebrafish ferroportin1 identifies a conserved verte-
33. Lam-Yuk-Tseung S, Gros P: Distinct targeting and brate iron exporter. Nature 403:776-781, 2000
recycling properties of two isoforms of the iron transporter 54. McKie AT, Marciani P, Rolfs A, et al: A novel duodenal
DMT1 (NRAMP2, Slc11A2). Biochemistry 45:2294-2301, 2006 iron-regulated transporter, IREG1, implicated in the basolateral
34. McKie AT, Barrow D, Latunde-Dada GO, et al: An iron- transfer of iron to the circulation. Mol Cell 5:299-309, 2000
regulated ferric reductase associated with the absorption of 55. Abboud S, Haile DJ: A novel mammalian iron-regulated
dietary iron. Science 291:1755-1759, 2001 protein involved in intracellular iron metabolism. J Biol Chem
35. Frazer DM, Wilkins SJ, Becker EM, et al: A rapid 275:19906-19912, 2000
decrease in the expression of DMT1 and Dcytb but not Ireg1 or 56. Pietrangelo A: The ferroportin disease. Blood Cells Mol
hephaestin explains the mucosal block phenomenon of iron Dis 32:131-138, 2004
absorption. Gut 52:340-346, 2003 57. McKie AT, Barlow DJ: The SLC40 basolateral iron
36. Latunde-Dada GO, Van der Westhuizen J, Vulpe CD, transporter family (IREG1/ferroportin/MTP1). Pflugers Arch
et al: Molecular and functional roles of duodenal cytochrome b 447:801-806, 2004
(Dcytb) in iron metabolism. Blood Cells Mol Dis 29:356-360, 58. Schimanski LM, Drakesmith H, Merryweather-Clarke
2002 AT, et al: In vitro functional analysis of human ferroportin (FPN)
37. Gunshin H, Starr CN, Direnzo C, et al: Cybrd1 and hemochromatosis-associated FPN mutations. Blood 105:
(duodenal cytochrome b) is not necessary for dietary iron 4096-4102, 2005
absorption in mice. Blood 106:2879-2883, 2005 59. Jeong SY, David S: Glycosylphosphatidylinositol-
38. McKie AT, Latunde-Dada GO, Miret S, et al: Molecular anchored ceruloplasmin is required for iron efflux from cells
evidence for the role of a ferric reductase in iron transport. in the central nervous system. J Biol Chem 278:27144-27148,
Biochem Soc Trans 30:722-724, 2002 2003
39. Andrews NC, Schmidt PJ: Iron homeostasis. Annu Rev 60. Han O, Kim EY: Colocalization of ferroportin-1 with
Physiol 69:69-85, 2007 hephaestin on the basolateral membrane of human intestinal
40. Hallberg L: Bioavailability of dietary iron in man. Annu absorptive cells. J Cell Biochem 101:1000-1010, 2007
Rev Nutr 1:123-147, 1981 61. Mok H, Jelinek J, Pai S, et al: Disruption of ferroportin 1
41. Wheby MS, Suttle GE, Ford III KT: Intestinal absorption regulation causes dynamic alterations in iron homeostasis and
of hemoglobin iron. Gastroenterology 58:647-654, 1970 erythropoiesis in polycythaemia mice. Development 131:
42. Grasbeck R, Kouvonen I, Lundberg M, et al: An 1859-1868, 2004
intestinal receptor for heme. Scand J Haematol 23:5-9, 1979 62. Nemeth E, Tuttle MS, Powelson J, et al: Hepcidin
43. Worthington MT, Cohn SM, Miller SK, et al: Char- regulates cellular iron efflux by binding to ferroportin and
acterization of a human plasma membrane heme transporter in inducing its internalization. Science 306:2090-2093, 2004
intestinal and hepatocyte cell lines. Am J Physiol Gastrointest 63. Donovan A, Lima CA, Pinkus JL, et al: The iron
Liver Physiol 280:G1172-G1177, 2001 exporter ferroportin/Slc40a1 is essential for iron homeostasis.
44. Shayeghi M, Latunde-Dada GO, Oakhill JS, et al: Cell Metab 1:191-200, 2005
Identification of an intestinal heme transporter. Cell 122: 64. Vulpe CD, Kuo YM, Murphy TL, et al: Hephaestin, a
789-801, 2005 ceruloplasmin homologue implicated in intestinal iron transport,
45. Rouault TA: The intestinal heme transporter revealed. is defective in the sla mouse. Nat Genet 21:195-199, 1999
Cell 122:649-651, 2005 65. Chen H, Su T, Attieh ZK, et al: Systemic regulation of
46. Latunde-Dada GO, Simpson RJ, McKie AT: Recent hephaestin and Ireg1 revealed in studies of genetic and
advances in mammalian haem transport. Trends Biochem Sci 31: nutritional iron deficiency. Blood 102:1893-1899, 2003
182-188, 2006 66. Bannerman RM, Cooper RG: Sex-linked anemia: A
47. Krishnamurthy P, Xie T, Schuetz JD: The role of hypochromic anemia of mice. Science 151:581-582, 1966
transporters in cellular heme and porphyrin homeostasis. 67. Kuo YM, Su T, Chen H, et al: Mislocalisation of
Pharmacol Ther 114:345-358, 2007 hephaestin, a multicopper ferroxidase involved in basolateral
120 BLEACKLEY ET AL

intestinal iron transport, in the sex linked anaemia mouse. Gut 89. Nemeth E, Rivera S, Gabayan V, et al: IL-6 mediates
53:201-206, 2004 hypoferremia of inflammation by inducing the synthesis of the
68. Li L, Vulpe CD, Kaplan J: Functional studies of iron regulatory hormone hepcidin. J Clin Invest 113:1271-1276,
hephaestin in yeast: evidence for multicopper oxidase activity 2004
in the endocytic pathway. Biochem J 375:793-798, 2003 90. Nemeth E, Ganz T: Regulation of iron metabolism by
69. Bonaccorsi di Patti MC, Miele R, Eugenia Schinina M, hepcidin. Annu Rev Nutr 26:323-342, 2006
et al: The yeast multicopper oxidase Fet3p and the iron permease 91. Knutson MD, Oukka M, Koss LM, et al: Iron release
Ftr1p physically interact. Biochem Biophys Res Commun 333: from macrophages after erythrophagocytosis is up-regulated by
432-437, 2005 ferroportin 1 overexpression and down-regulated by hepcidin.
70. De Domenico I, Ward DM, di Patti MC, et al: Proc Natl Acad Sci U S A 102:1324-1328, 2005
Ferroxidase activity is required for the stability of cell surface 92. Nairz M, Theurl I, Ludwiczek S, et al: The co-ordinated
ferroportin in cells expressing GPI-ceruloplasmin. Embo J 26: regulation of iron homeostasis in murine macrophages limits the
2823-2831, 2007 availability of iron for intracellular Salmonella typhimurium.
71. McGregor JA, Shayeghi M, Vulpe CD, et al: Impaired Cell Microbiol 9:2126-2140, 2007
iron transport activity of ferroportin 1 in hereditary iron overload. 93. Enns C: In: Templeton D, editor. Molecular and Cellular
J Membr Biol 206:3-7, 2005 Iron Transport. New York, Marcel Decker Inc., 2002, pp 71-94
72. Hart EB, Steenbock H, Waddell J, et al: Iron in nutrition: 94. Dautry-Varsat A, Ciechanover A, Lodish HF: pH and the
VII. Copper as a supplement to iron for hemoglobin building in recycling of transferrin during receptor-mediated endocytosis.
the rat. 1928. J Biol Chem 277:e228 2002 Proc Natl Acad Sci U S A 80:2258-2262, 1983
73. Roy CN, Enns CA: Iron homeostasis: New tales from 95. Bennett MJ, Lebron JA, Bjorkman PJ: Crystal structure
the crypt. Blood 96:4020-4027, 2000 of the hereditary haemochromatosis protein HFE complexed
74. Koury MJ, Ponka P: New insights into erythropoiesis: with transferrin receptor. Nature 403:46-53, 2000
The roles of folate, vitamin B12, and iron. Annu Rev Nutr 24: 96. Giannetti AM, Halbrooks PJ, Mason AB, et al: The
105-131, 2004 molecular mechanism for receptor-stimulated iron release from
75. Jelkmann W: Erythropoietin after a century of research: the plasma iron transport protein transferrin. Structure 13:
Younger than ever. Eur J Haematol 78:183-205, 2007 1613-1623, 2005
76. Goodnough LT: Erythropoietin and iron-restricted 97. McKie AT: A ferrireductase fills the gap in the
erythropoiesis. Exp Hematol 35:167-172, 2007 transferrin cycle. Nat Genet 37:1159-1160, 2005
77. Richardson DR, Ponka P: The molecular mechanisms of 98. Ohgami RS, Campagna DR, Greer EL, et al: Identifica-
the metabolism and transport of iron in normal and neoplastic tion of a ferrireductase required for efficient transferrin-
cells. Biochim Biophys Acta 1331:1-40 dependent iron uptake in erythroid cells. Nat Genet 37:
78. Bullen J, Griffiths E, Rogers H, et al: Sepsis: The critical 1264-1269, 2005
role of iron. Microbes Infect 2:409-415, 2000 99. Fleming RE, Migas MC, Zhou X, et al: Mechanism of
79. Johnson MB, Enns CA: Diferric transferrin regulates increased iron absorption in murine model of hereditary
transferrin receptor 2 protein stability. Blood 104:4287-4293, hemochromatosis: Increased duodenal expression of the iron
2004 transporter DMT1. Proc Natl Acad Sci U S A 96:3143-3148,
80. Rothenberg BE, Voland JR: Beta2 knockout mice 1999
develop parenchymal iron overload: A putative role for class I 100. Food MR, Rothenberger S, Gabathuler R, et al: Trans-
genes of the major histocompatibility complex in iron metabo- port and expression in human melanomas of a transferrin-like
lism. Proc Natl Acad Sci U S A 93:1529-1534, 1996 glycosylphosphatidylinositol-anchored protein. J Biol Chem
81. Levy JE, Jin O, Fujiwara Y, et al: Transferrin receptor is 269:3034-3040, 1994
necessary for development of RBCs and the nervous system. Nat 101. Richardson DR: The role of the membrane-bound
Genet 21:396-399, 1999 tumour antigen, melanotransferrin (p97), in iron uptake by the
82. Hellman NE, Gitlin JD: Ceruloplasmin metabolism and human malignant melanoma cell. Eur J Biochem 267:1290-1298,
function. Annu Rev Nutr 22:439-458, 2002 2000
83. Healy J, Tipton K: Ceruloplasmin and what it might do. 102. Dunn LL, Rahmanto YS, Richardson DR: Iron uptake
J Neural Transm 114:777-781, 2007 and metabolism in the new millennium. Trends Cell Biol 17:
84. Harris ZL, Klomp LW, Gitlin JD: Aceruloplasminemia: 93-100
An inherited neurodegenerative disease with impairment of iron 103. Woodbury RG, Brown JP, Yeh MY, et al: Identification
homeostasis. Am J Clin Nutr 67:972S-977S, 1998 of a cell surface protein, p97, in human melanomas and certain
85. Harris ZL, Takahashi Y, Miyajima H, et al: Acerulo- other neoplasms. Proc Natl Acad Sci U S A 77:2183-2187, 1980
plasminemia: Molecular characterization of this disorder of iron 104. Brown JP, Woodbury RG, Hart CE, et al: Quantitative
metabolism. Proc Natl Acad Sci U S A 92:2539-2543, 1995 analysis of melanoma-associated antigen p97 in normal and
86. Gitlin JD: Aceruloplasminemia. Pediatr Res 44:271-276, neoplastic tissues. Proc Natl Acad Sci U S A 78:539-543, 1981
1998 105. Sekyere EO, Dunn LL, Richardson DR: Examination of
87. Madsen E, Gitlin JD: Copper and iron disorders of the the distribution of the transferrin homologue, melanotransferrin
brain. Annu Rev Neurosci 30:317-337, 2007 (tumour antigen p97), in mouse and human. Biochim Biophys
88. Jabado N, Canonne-Hergaux F, Gruenheid S, et al: Iron Acta 1722:131-142, 2005
transporter Nramp2/DMT-1 is associated with the membrane of 106. Food MR, Sekyere EO, Richardson DR: The soluble
phagosomes in macrophages and Sertoli cells. Blood 100: form of the membrane-bound transferrin homologue, melano-
2617-2622, 2002 transferrin, inefficiently donates iron to cells via nonspecific
IRON IMBALANCE AND DISEASE 121

internalization and degradation of the protein. Eur J Biochem 126. Yoon T, Cowan JA: Iron-sulfur cluster biosynthesis.
269:4435-4445, 2002 Characterization of frataxin as an iron donor for assembly of
107. Dunn LL, Sekyere EO, Rahmanto YS, et al: The [2Fe-2S] clusters in ISU-type proteins. J Am Chem Soc 125:
function of melanotransferrin: a role in melanoma cell prolifera- 6078-6084, 2003
tion and tumorigenesis. Carcinogenesis 27:2157-2169, 2006 127. Lesuisse E, Santos R, Matzanke BF, et al: Iron use for
108. Sekyere E, Richardson DR: The membrane-bound haeme synthesis is under control of the yeast frataxin homologue
transferrin homologue melanotransferrin: Roles other than iron (Yfh1). Hum Mol Genet 12:879-889, 2003
transport? FEBS Lett 483:11-16, 2000 128. Cook JD, Bencze KZ, Jankovic AD, et al: Monomeric
109. MacGillivray RT, Mason AB: In: Templeton D, editor. yeast frataxin is an iron-binding protein. Biochemistry 45:
Molecular and Cellular Iron Transport. New York, Marcel 7767-7777, 2006
Dekker Inc., 2002, pp 95-123 129. Gibson TJ, Koonin EV, Musco G, et al: Friedreich's
110. Ward PP, Paz E, Conneely OM: Multifunctional roles of ataxia protein: Phylogenetic evidence for mitochondrial dysfunc-
lactoferrin: A critical overview. Cell Mol Life Sci 62:2540-2548, tion. Trends Neurosci 19:465-468, 1996
2005 130. Ponka P: Tissue-specific regulation of iron metabolism
111. Ward PP, Mendoza-Meneses M, Cunningham GA, et al: and heme synthesis: Distinct control mechanisms in erythroid
Iron status in mice carrying a targeted disruption of lactoferrin. cells. Blood 89:1-258 1997
Mol Cell Biol 23:178-185, 2003 131. Ponka P: Cell biology of heme. Am J Med Sci 318:
112. Coon MJ, Ding XX, Pernecky SJ, et al: Cytochrome 241-256, 1999
P450: Progress and predictions. Faseb J 6:669-673, 1992 132. Lill R, Dutkiewicz R, Elsasser HP, et al: Mechanisms of
113. Eklund H, Uhlin U, Farnegardh M, et al: Structure and iron-sulfur protein maturation in mitochondria, cytosol and
function of the radical enzyme ribonucleotide reductase. Prog nucleus of eukaryotes. Biochim Biophys Acta 1763:652-667,
Biophys Mol Biol 77:177-268, 2001 2006
114. Shaw GC, Cope JJ, Li L, et al: Mitoferrin is essential for 133. Lill R, Muhlenhoff U: Iron-sulfur protein biogenesis in
erythroid iron assimilation. Nature 440:96-100 eukaryotes: components and mechanisms. Annu Rev Cell Dev
115. Li FY, Nikali K, Gregan J, et al: Characterization of a Biol 22:457-486, 2006
novel human putative mitochondrial transporter homologous to 134. Torti FM, Torti SV: Regulation of ferritin genes and
the yeast mitochondrial RNA splicing proteins 3 and 4. FEBS protein. Blood 99:3505-3516, 2002
Lett 494:79-84, 2001 135. Carrondo MA: Ferritins, iron uptake and storage from
116. Muhlenhoff U, Stadler JA, Richhardt N, et al: A specific the bacterioferritin viewpoint. EMBO J 22:1959-1968, 2003
role of the yeast mitochondrial carriers MRS3/4p in mitochon- 136. Harrison PM: Ferritin: An iron-storage molecule. Semin
drial iron acquisition under iron-limiting conditions. J Biol Chem Hematol 14:55-70, 1977
278:40612-40620, 2003 137. Surguladze N, Patton S, Cozzi A, et al: Characterization
117. Zhang Y, Lyver ER, Knight SA, et al: Mrs3p, Mrs4p, of nuclear ferritin and mechanism of translocation. Biochem J
and frataxin provide iron for Fe-S cluster synthesis in 388:731-740, 2005
mitochondria. J Biol Chem 281:22493-22502, 2006 138. Levi S, Arosio P: Mitochondrial ferritin. Int J Biochem
118. Zhang Y, Lyver ER, Knight SA, et al: Frataxin and Cell Biol 36:1887-1889, 2004
mitochondrial carrier proteins, Mrs3p and Mrs4p, cooperate in 139. Ghosh S, Hevi S, Chuck SL: Regulated secretion of
providing iron for heme synthesis. J Biol Chem 280: glycosylated human ferritin from hepatocytes. Blood 103:
19794-19807, 2005 2369-2376, 2004
119. Li L, Kaplan J: A mitochondrial-vacuolar signaling 140. Hasan MR, Morishima D, Tomita K, et al: Identification
pathway in yeast that affects iron and copper metabolism. J Biol of a 250 kDa putative microtubule-associated protein as bovine
Chem 279:33653-33661, 2004 ferritin. Evidence for a ferritin-microtubule interaction. FEBS J
120. Bulteau AL, O'Neill HA, Kennedy MC, et al: Frataxin 272:822-831, 2005
acts as an iron chaperone protein to modulate mitochondrial 141. Hasan MR, Koikawa S, Kotani S, et al: Ferritin forms
aconitase activity. Science 305:242-245, 2004 dynamic oligomers to associate with microtubules in vivo:
121. Nichol H, Gakh O, O'Neill HA, et al: Structure of Implication for the role of microtubules in iron metabolism. Exp
frataxin iron cores: An X-ray absorption spectroscopic study. Cell Res 312:1950-1960, 2006
Biochemistry 42:5971-5976, 2003 142. Hentze MW, Kuhn LC: Molecular control of vertebrate
122. Adamec J, Rusnak F, Owen WG, et al: Iron-dependent iron metabolism: mRNA-based regulatory circuits operated by
self-assembly of recombinant yeast frataxin: Implications for iron, nitric oxide, and oxidative stress. Proc Natl Acad Sci U S A
Friedreich ataxia. Am J Hum Genet 67:549-562, 2000 93:8175-8182, 1996
123. Cavadini P, O'Neill HA, Benada O, et al: Assembly and 143. Haile DJ, Rouault TA, Harford JB, et al: Cellular
iron-binding properties of human frataxin, the protein deficient regulation of the iron-responsive element binding protein:
in Friedreich ataxia. Hum Mol Genet 11:217-227, 2002 Disassembly of the cubane iron-sulfur cluster results in high-
124. Gakh O, Adamec J, Gacy AM, et al: Physical evidence affinity RNA binding. Proc Natl Acad Sci U S A 89:
that yeast frataxin is an iron storage protein. Biochemistry 41: 11735-11739, 1992
6798-6804, 2002 144. Pantopoulos K: Iron metabolism and the IRE/IRP
125. Park S, Gakh O, O'Neill HA, et al: Yeast frataxin regulatory system: An update. Ann N Y Acad Sci 1012:1-138 2004
sequentially chaperones and stores iron by coupling protein 145. Rouault TA: The role of iron regulatory proteins in
assembly with iron oxidation. J Biol Chem 278:31340-31351, mammalian iron homeostasis and disease. Nat Chem Biol 2:
2003 406-414, 2006
122 BLEACKLEY ET AL

146. Vyoral D, Petrak J: Iron transport in K562 cells: A 165. Wang RH, Li C, Xu X, et al: A role of SMAD4 in iron
kinetic study using native gel electrophoresis and 59Fe metabolism through the positive regulation of hepcidin expres-
autoradiography. Biochim Biophys Acta 1403:179-188, 1998 sion. Cell Metab 2:399-409, 2005
147. Petrak JV, Vyoral D: Detection of iron-containing 166. Lin L, Valore EV, Nemeth E, et al: Iron transferrin
proteins contributing to the cellular labile iron pool by a native regulates hepcidin synthesis in primary hepatocyte culture
electrophoresis metal blotting technique. J Inorg Biochem 86: through hemojuvelin and BMP2/4. Blood 110:2182-2189, 2007
669-675, 2001 167. Zhang AS, Anderson SA, Meyers KR, et al: Evidence
148. Pufahl RA, Singer CP, Peariso KL, et al: Metal ion that inhibition of hemojuvelin shedding in response to iron is
chaperone function of the soluble Cu(I) receptor Atx1. Science mediated through neogenin. J Biol Chem 282:12547-12556,
278:853-856, 1997 2007
149. Culotta VC, Klomp LW, Strain J, et al: The copper 168. Silvestri L, Pagani A, Camaschella C: Furin mediated
chaperone for superoxide dismutase. J Biol Chem 272: release of soluble hemojuvelin: A new link between hypoxia and
23469-23472, 1997 iron homeostasis. Blood8 2007
150. Beers J, Glerum DM, Tzagoloff A: Purification, 169. Feder JN, Gnirke A, Thomas W, et al: A novel MHC
characterization, and localization of yeast Cox17p, a mitochon- class I-like gene is mutated in patients with hereditary
drial copper shuttle. J Biol Chem 272:33191-33196, 1997 haemochromatosis. Nat Genet 13:399-408, 1996
151. Ponka P, Sheftel AD, Zhang AS: Iron targeting to 170. Waheed A, Parkkila S, Saarnio J, et al: Association of
mitochondria in erythroid cells. Biochem Soc Trans 30:735-738, HFE protein with transferrin receptor in crypt enterocytes of
2002 human duodenum. Proc Natl Acad Sci U S A 96:1579-1584,
152. Li L, Chen OS, McVey Ward D, et al: CCC1 is a 1999
transporter that mediates vacuolar iron storage in yeast. J Biol 171. West AR, Thomas C, Sadlier J, et al: Haemochromatosis
Chem 276:29515-29519, 2001 protein is expressed on the terminal web of enterocytes in
153. Valore EV, Ganz T: Posttranslational processing of proximal small intestine of the rat. Histochem Cell Biol 125:
hepcidin in human hepatocytes is mediated by the prohormone 283-292, 2006
convertase furin. Blood Cells Mol Dis8 2007 172. Sebastiani G, Walker AP: HFE Gene in primary and
154. Hunter HN, Fulton DB, Ganz T, et al: The solution secondary hepatic iron overload. World J Gastroenterol 13:
structure of human hepcidin, a peptide hormone with anti- 4673-4689, 2007
microbial activity that is involved in iron uptake and hereditary 173. Lebron JA, Bennett MJ, Vaughn DE, et al: Crystal
hemochromatosis. J Biol Chem 277:37597-37603, 2002 structure of the hemochromatosis protein HFE and characteriza-
155. Nicolas G, Bennoun M, Devaux I, et al: Lack of tion of its interaction with transferrin receptor. Cell 93:111-123,
hepcidin gene expression and severe tissue iron overload in 1998
upstream stimulatory factor 2 (USF2) knockout mice. Proc Natl 174. Feder JN, Tsuchihashi Z, Irrinki A, et al: The
Acad Sci U S A 98:8780-8785, 2001 hemochromatosis founder mutation in HLA-H disrupts beta2-
156. Nicolas G, Bennoun M, Porteu A, et al: Severe iron microglobulin interaction and cell surface expression. J Biol
deficiency anemia in transgenic mice expressing liver hepcidin. Chem 272:14025-14028, 1997
Proc Natl Acad Sci U S A 99:4596-4601, 2002 175. Parkkila S, Waheed A, Britton RS, et al: Association of
157. De Domenico I, Ward DM, Langelier C, et al: The the transferrin receptor in human placenta with HFE, the protein
molecular mechanism of hepcidin-mediated ferroportin down- defective in hereditary hemochromatosis. Proc Natl Acad Sci U S
regulation. Mol Biol Cell 18:2569-2578, 2007 A 94:13198-13202, 1997
158. Frazer DM, Wilkins SJ, Becker EM, et al: Hepcidin 176. Feder JN, Penny DM, Irrinki A, et al: The hemochro-
expression inversely correlates with the expression of duodenal matosis gene product complexes with the transferrin receptor and
iron transporters and iron absorption in rats. Gastroenterology lowers its affinity for ligand binding. Proc Natl Acad Sci U S A
123:835-844, 2002 95:1472-1477, 1998
159. Babitt JL, Huang FW, Xia Y, et al: Modulation of bone 177. Goswami T, Andrews NC: Hereditary hemochromatosis
morphogenetic protein signaling in vivo regulates systemic iron protein, HFE, interaction with transferrin receptor 2 suggests a
balance. J Clin Invest 117:1933-1939, 2007 molecular mechanism for mammalian iron sensing. J Biol Chem
160. Wrighting DM, Andrews NC: Interleukin-6 induces 281:28494-28498, 2006
hepcidin expression through STAT3. Blood 108:3204-3209, 178. West Jr AP, Giannetti AM, Herr AB, et al: Mutational
2006 analysis of the transferrin receptor reveals overlapping HFE and
161. Verga Falzacappa MV, Vujic Spasic M, Kessler R, et al: transferrin binding sites. J Mol Biol 313:385-397, 2001
STAT3 mediates hepatic hepcidin expression and its inflamma- 179. Waheed A, Grubb JH, Zhou XY, et al: Regulation of
tory stimulation. Blood 109:353-358, 2007 transferrin-mediated iron uptake by HFE, the protein defective in
162. Babitt JL, Huang FW, Wrighting DM, et al: Bone hereditary hemochromatosis. Proc Natl Acad Sci U S A 99:
morphogenetic protein signaling by hemojuvelin regulates 3117-3122, 2002
hepcidin expression. Nat Genet 38:531-539, 2006 180. Fleming RE, Sly WS: Mechanisms of iron accumulation
163. Papanikolaou G, Samuels ME, Ludwig EH, et al: in hereditary hemochromatosis. Annu Rev Physiol 64:663-680,
Mutations in HFE2 cause iron overload in chromosome 1q- 2002
linked juvenile hemochromatosis. Nat Genet 36:77-82, 2004 181. Calzolari A, Raggi C, Deaglio S, et al: TfR2 localizes in
164. Lin L, Goldberg YP, Ganz T: Competitive regulation of lipid raft domains and is released in exosomes to activate signal
hepcidin mRNA by soluble and cell-associated hemojuvelin. transduction along the MAPK pathway. J Cell Sci 119:
Blood 106:2884-2889, 2005 4486-4498, 2006
IRON IMBALANCE AND DISEASE 123

182. Chua AC, Graham RM, Trinder D, et al: The regulation 199. Yang M, Cobine PA, Molik S, et al: The effects of
of cellular iron metabolism. Crit Rev Clin Lab Sci 44:413-459, mitochondrial iron homeostasis on cofactor specificity of
2007 superoxide dismutase 2. EMBO J 25:1775-1783, 2006
183. Arredondo M, Tapia V, Rojas A, et al: Apical 200. Martelli A, Wattenhofer-Donze M, Schmucker S, et al:
distribution of HFE-beta2-microglobulin is associated with Frataxin is essential for extramitochondrial Fe S cluster proteins
inhibition of apical iron uptake in intestinal epithelia cells. in mammalian tissues. Hum Mol Genet 16:2651-2658, 2007
Biometals 19:379-388, 2006 201. Goedert M: Alpha-synuclein and neurodegenerative
184. Lam-Yuk-Tseung S, Camaschella C, Iolascon A, et al: A diseases. Nat Rev Neurosci 2:492-501, 2001
novel R416C mutation in human DMT1 (SLC11A2) displays 202. Spillantini MG, Crowther RA, Jakes R, et al: Alpha-
pleiotropic effects on function and causes microcytic anemia and synuclein in filamentous inclusions of Lewy bodies from
hepatic iron overload. Blood Cells Mol Dis 36:347-354, 2006 Parkinson's disease and dementia with Lewy bodies. Proc Natl
185. Iolascon A, d'Apolito M, Servedio V, et al: Microcytic Acad Sci U S A 95:6469-6473, 1998
anemia and hepatic iron overload in a child with compound 203. Bharathi, Indi SS, Rao KS: Copper- and iron-induced
heterozygous mutations in DMT1 (SCL11A2). Blood 107: differential fibril formation in alpha-synuclein: TEM study.
349-354, 2006 Neurosci Lett 424:78-82, 2007
186. Beaumont C, Delaunay J, Hetet G, et al: Two new 204. Friedlich AL, Tanzi RE, Rogers JT: The 5′-untranslated
human DMT1 gene mutations in a patient with microcytic region of Parkinson's disease alpha-synuclein messenger RNA
anemia, low ferritinemia, and liver iron overload. Blood 107: contains a predicted iron responsive element. Mol Psychiatry 12:
4168-4170, 2006 222-223, 2007
187. Lam-Yuk-Tseung S, Mathieu M, Gros P: Functional 205. Kaur D, Andersen JK: Ironing out Parkinson's disease:
characterization of the E399D DMT1/NRAMP2/SLC11A2 Is therapeutic treatment with iron chelators a real possibility?
protein produced by an exon 12 mutation in a patient with Aging Cell 1:17-21, 2002
microcytic anemia and iron overload. Blood Cells Mol Dis 35: 206. Ostrerova-Golts N, Petrucelli L, Hardy J, et al: The
212-216, 2005 A53T alpha-synuclein mutation increases iron-dependent aggre-
188. Zhao R, Min SH, Qiu A, et al: The spectrum of gation and toxicity. J Neurosci 20:6048-6054, 2000
mutations in the PCFT gene, coding for an intestinal folate 207. Atwood CS, Obrenovich ME, Liu T, et al: Amyloid-
transporter, that are the basis for hereditary folate malabsorption. beta: A chameleon walking in two worlds: a review of the trophic
Blood 110:1147-1152, 2007 and toxic properties of amyloid-beta. Brain Res Brain Res Rev
189. Robson KJ, Merryweather-Clarke AT, Cadet E, et al: 43:1-168 2003
Recent advances in understanding haemochromatosis: A transi- 208. Rogers JT, Randall JD, Cahill CM, et al: An iron-
tion state. J Med Genet 41:721-730, 2004 responsive element type II in the 5′-untranslated region of the
190. De Domenico I, McVey Ward D, Nemeth E, et al: Alzheimer's amyloid precursor protein transcript. J Biol Chem
Molecular and clinical correlates in iron overload associated with 277:45518-45528, 2002
mutations in ferroportin. Haematologica 91:1092-1095, 2006 209. Bush AI: The metallobiology of Alzheimer's disease.
191. De Domenico I, Ward DM, Musci G, et al: Evidence for Trends Neurosci 26:207-214, 2003
the multimeric structure of ferroportin. Blood 109:2205-2209, 2007 210. Atamna H: Heme binding to amyloid-beta peptide:
192. Vassiliev V, Harris ZL, Zatta P: Ceruloplasmin in Mechanistic role in Alzheimer's disease. J Alzheimers Dis 10:
neurodegenerative diseases. Brain Res Brain Res Rev 49: 255-266, 2006
633-640, 2005 211. Smith DG, Cappai R, Barnham KJ: The redox chemistry
193. Miyajima H: Aceruloplasminemia, an iron metabolic of the Alzheimer's disease amyloid beta peptide. Biochim
disorder. Neuropathology 23:345-350, 2003 Biophys Acta 1768:1976-1990, 2007
194. Durr A, Cossee M, Agid Y, et al: Clinical and genetic 212. Curtis AR, Fey C, Morris CM, et al: Mutation in the
abnormalities in patients with Friedreich's ataxia. N Engl J Med gene encoding ferritin light polypeptide causes dominant adult-
335:1169-1175, 1996 onset basal ganglia disease. Nat Genet 28:350-354, 2001
195. Baron M, Kudin AP, Kunz WS: Mitochondrial dysfunc- 213. Vidal R, Ghetti B, Takao M, et al: Intracellular ferritin
tion in neurodegenerative disorders. Biochem Soc Trans 35: accumulation in neural and extraneural tissue characterizes a
1228-1231, 2007 neurodegenerative disease associated with a mutation in the
196. Krasilnikova MM, Kireeva ML, Petrovic V, et al: Effects ferritin light polypeptide gene. J Neuropathol Exp Neurol 63:
of Friedreich's ataxia (GAA)n⁎(TTC)n repeats on RNA synth- 363-380, 2004
esis and stability. Nucleic Acids Res 35:1075-1084, 2007 214. Burn J, Chinnery PF: Neuroferritinopathy. Semin
197. Campuzano V, Montermini L, Molto MD, et al: Friedreich's Pediatr Neurol 13:176-181, 2006
ataxia: Autosomal recessive disease caused by an intronic GAA 215. Cremonesi L, Foglieni B, Fermo I, et al: Identification of
triplet repeat expansion. Science 271:1423-1427, 1996 two novel mutations in the 5′-untranslated region of H-ferritin
198. Wilson RB: Iron dysregulation in Friedreich ataxia. using denaturing high performance liquid chromatography
Semin Pediatr Neurol 13:166-175, 2006 scanning. Haematologica 88:1110-1116, 2003

You might also like