Sei sulla pagina 1di 9

PHYSIOLOGY 21: 115–123, 2006; 10.1152/physiol.00052.

2005
REVIEWS
The Ins and Outs of Iron Homeostasis Adriana Donovan,1* Cindy N.
Roy,1* and Nancy C. Andrews1,2
1Children’sHospital Boston, Dana-Farber Cancer Institute,
Iron is an essential element that is toxic when it accumulates in excess. Intricate reg- Harvard Medical School, and 2Howard Hughes Medical
Institute, Boston, Massachusetts
nancy_andrews@hms.harvard.edu
ulatory mechanisms have evolved to maintain iron homeostasis within cells and *A. Donovan and C. N. Roy contributed equally to this
work.
between different tissues of complex organisms. This review discusses the proteins
involved in iron transport and storage and their regulation in health and disease.

Iron is an essential metal, required as a co-factor in Proteins involved in the uptake of heme and inor-
proteins that transfer electrons and manage oxygen. ganic iron reside on the brush-border membrane
Until recently, there was little molecular understand- (FIGURE 1). Most non-heme iron in the diet is present
ing of how mammalian tissues meet the challenge of as the Fe3+ form. The major transporter involved in
acquiring adequate amounts of iron without risking cellular non-heme iron uptake (import) is divalent
the toxic effects of iron excess. Our current knowledge metal transporter 1 (DMT1; also known as Nramp2,
of mammalian iron physiology and homeostasis is DCT1, and SLC11A2), which has 12 predicted trans-
painted in broad brushstrokes, with many details membrane segments (26, 33, 35). As its name implies,
remaining to be discovered. This review summarizes DMT1 also transports other divalent metals including
recent progress in identifying key protein transporters Mn2+, Co2+, Cu2+, and Zn2+ (35). DMT1 is active in a
and the molecules that regulate their activities. low-pH environment, as found in the duodenum,
Although all cells need a small amount of iron, ery- because it requires proton cotransport (35). A require-
throid precursors require substantial amounts to pro- ment for DMT1 in intestinal iron absorption is clearly
duce hemoglobin. Accordingly, anemia is a prominent supported by animal models with spontaneous and
manifestation of iron deficiency. Other cell types also induced mutations (20, 25, 26, 34). DMT1 exclusively
have specialized roles that are important to consider transports divalent metals, necessitating luminal con-
in assembling a systemic model of iron homeostasis. version of Fe3+ to Fe2+. McKie et al. (60) identified a
Intestinal epithelial cells (enterocytes), extraembryon- candidate intestinal iron reductase, duodenal
ic visceral endoderm cells, and placental syncytiotro- cytochrome b (Dcytb; also known as Cybrd1).
phoblasts serve in the acquisition of iron from the Expression of this putative transmembrane, di-heme
external environment. Of these, iron handling by ente- protein is induced in the intestinal mucosa of mice
rocytes is best understood. Liver hepatocytes serve a with increased intestinal iron absorption due to ane-
depot function, removing excess iron from circulating mia, iron deficiency, or hypoxia (60). However, it was
plasma and safely storing it until it is needed. Tissue recently reported that disruption of the murine Dcytb
macrophages recognize and phagocytose old and gene did not significantly impair intestinal iron
damaged erythrocytes, recovering their iron for reuse absorption under normal conditions (36), suggesting
and storage. Molecular signals must coordinate the that other intestinal reductases may substitute or that
operations of each of these cell types. To date, no effi- mice have an efficient mechanism for non-enzymatic
cient, regulated excretion mechanism for iron has iron reduction.
been described, underscoring the importance of The intestine also absorbs heme iron from the diet.
meticulous regulation of iron acquisition and distribu- Cell-culture studies of the intestinal cell line Caco-2
tion. suggested that heme absorption is a saturable, carrier-
mediated process (103). Other studies have described a
Intestinal Iron Absorption heme receptor on duodenal brush-border membranes
and erythroleukemia cells (28, 32). Recently, an entero-
Mammals absorb dietary iron through the duodenal cyte heme importer was described that likely mediates
epithelium of the small intestine (29, 66), which is dietary heme uptake (92). This molecule, termed heme
organized in villous structures to maximize its absorp- carrier protein 1 (HCP1), resembles bacterial proteins
tive surface area. Enterocyte precursors are present in that transport metal-tetracycline complexes. It has no
crypts at the bases of villi, migrating up the villous axis close mammalian homologs. Once dietary heme has
as they differentiate. Membrane extensions at the api- entered the intestinal epithelial cell, it is likely cleaved
cal surface of enterocytes form a brush border that fur- by intracellular heme oxygenase 1 to release iron (85).
ther increases the surface area available for absorp- Subsequently, it probably joins the same intracellular
tion. Mature enterocytes live for only 1–2 days. Iron pool as non-heme iron. Two other proteins, FLVCR and
that accumulates within them is lost from the body Bcrp, both function as cellular heme exporters (55, 84),
when senescent enterocytes are shed into the gut but neither has been shown to be involved in intestinal
lumen. heme transport.

1548-9213/06 8.00 ©2006 Int. Union Physiol. Sci./Am. Physiol. Soc. 115
REVIEWS
1 Dietary non-heme iron 2 DMT1 mediates proton- across the basolateral
(Fe3+) must be reduced for dependent Fe2+ import. membrane. A sponta-
transport across the apical
brush border. neous mouse mutant,
sex-linked anemia (sla),
Fe2+ Heme
has impaired iron
Fe3+
H+ DMT1 3 HCP1 transports absorption and accumu-
dietary heme iron. lation of iron in duodenal
Apical HCP1
enterocytes attributable
to a mutation in the gene
Ferri- encoding hephaestin (5,
reductase 82, 98). This phenotype is
(?Dcytb) similar to, but less severe
Heme
oxygenase than, the phenotype seen
in mice with intestine-
specific inactivation of
ferroportin (22).
4 Heme oxygenase releases Hephaestin is a mem-
Ferritin iron from protoporphyrin, brane-bound homolog of
allowing it to enter the same
pool as non-heme iron. the serum multi-copper
5 Some iron is used or stored oxidase ceruloplasmin
within the enterocyte in (40). The current model
ferritin, and is later lost suggests that hephaestin
when the intestinal
oxidizes Fe2+ released by
mucosa is sloughed.
ferroportin, facilitating
its incorporation into
Ferroxidase transferrin, the major
(hephaestin) serum iron carrier pro-
tein. The anemia of sla
mice resolves after the
Ferroportin Fe3+ neonatal period, sug-
Basolateral Fe2+
gesting that hephaestin is
primarily needed for iron
transfer during the accu-
6 Ferroportin exports some
iron, where it is oxidized to mulation of initial stores
Fe3+ for incorporation into and that serum cerulo-
serum transferrin. plasmin may substitute
later.
FIGURE 1. Intestinal iron absorption
An individual enterocyte is depicted. Dietary non-heme iron (Fe3+) must be reduced for
transport across the apical brush border. DMT1 mediates proton-dependent Fe2+ import. Intercellular and
Dietary heme iron is transported by HCP1. Once inside the cell, heme oxygenase releases Intracellular Iron
iron from protoporphyrin, presumably allowing it to enter the same pool as non-heme iron.
Some iron is used or stored within the enterocyte in ferritin. This iron is later lost from the
Handling
body when the intestinal mucosa is sloughed. Some iron is exported across the basolateral
membrane by ferroportin and oxidized to Fe3+ for incorporation into serum transferrin. Iron circulates bound to
Intracellular enterocyte iron can either be stored in the 80-kDa serum glycoprotein transferrin (TF), which
the multimeric protein ferritin (95) or transported has two high-affinity iron binding sites. Diferric TF
across the basolateral membrane of the enterocyte binds to a highly specific TF receptor (TFR1), allowing
into the circulation. Ferroportin (also known as cellular uptake by receptor-mediated endocytosis.
IREG1, MTP1, and SLC40A1), a distinct transporter The receptors collect in clathrin-coated pits and facil-
with 10–12 transmembrane segments, was identified itate transferrin internalization into endocytic vesi-
as a compelling candidate for the basolateral iron cles. The endosomes become acidified, likely through
exporter (1, 21, 61). Intestinal expression of ferroportin the pump action of an Na+-H+-ATPase (73). As the
mRNA and protein increases in response to iron defi- endosomes reach pH 5.5, acidification and protein
ciency and hypoxia (61). Recently, selective inactiva- conformation changes cause iron to dissociate from
tion of the murine ferroportin gene in intestinal cells TF (18). Fe3+ is reduced to Fe2+, likely by the endosomal
established that ferroportin is the major, if not only, reductase Steap3 (75), for transport from the endo-
intestinal iron exporter (22). Ferroportin is probably some to the cytoplasm by the transporter DMT1 (25).
also selective for Fe2+. Multicopper oxidases that oxi- DMT1 localizes to recycling endosomes through a sig-
dize Fe2+ to Fe3+ also play a role in the transport of iron nal found in one of two carboxy-termini generated by

116 PHYSIOLOGY • Volume 21 • April 2006 • www.physiologyonline.org


REVIEWS
alternative splicing (56). The TF cycle is completed tions associated with anemia have been identified in
when the endosome returns to and fuses with the plas- both the TF and DMT1 genes (7, 46, 62). However,
ma membrane, returning apoTF to the circulation and studies of non-erythroid cells indicate that there must
TFR1 to the plasma membrane and allowing both be distinct mechanisms for non-TF-bound iron
molecules to start the cycle again. (NTBI) uptake. Although the proteins normally
Cellular iron in excess of immediate needs is stored involved in this process have not been identified (9),
as an iron oxide solid within the central cavity of fer- two possible mechanisms for mammalian NTBI
ritin, a polymeric protein composed of varying ratios uptake have been described.
of heavy (H) and light (L) ferritin polypeptides (94). To Purified lipocalin 2 (also known as Ngal and 24p3)
ensure that very little iron is free within cells, iron reg- (30, 106) contains iron complexed in bacterial
ulatory proteins (IRP1 and IRP2) control the posttran- siderophores. It is not yet clear whether this form has a
scriptional expression of genes modulating cellular physiological role in iron transport. Nonetheless, the
iron uptake and storage. Under low iron conditions, complex is taken up by cells through an as yet uniden-
both proteins bind to conserved, hairpin-like iron reg- tified receptor-mediated process. To date, studies
ulatory elements (IREs) present in the untranslated implicate the iron-binding activity of lipocalin 2 in
regions (UTRs) of target mRNAs. Classical studies early renal development and in innate immunity, but
defined the roles of IRPs and IREs in regulating further study will be necessary to determine whether it
mRNAs encoding TFR1 and ferritin subunits. The plays a general role in NTBI uptake (27, 106).
TFR1 mRNA contains five IRE elements in its 3’-UTR. L-type calcium channels have also been implicated
IRP binding protects the mRNA from endonucleolytic in NTBI uptake. Mammals express four highly homol-
degradation (42). Thus, when the cell is iron depleted, ogous L-type calcium channels (Cav1.1-1.4). L-type cal-
more TFR1 protein is made, resulting in increased iron cium channels have been shown to mediate cellular
acquisition. In contrast, binding of IRPs to the single iron uptake in vitro (96) and to be involved in iron dep-
IREs in the 5’-UTRs of ferritin mRNAs does not affect osition in the murine heart in the setting of iron over-
mRNA stability but rather inhibits translation of the load in vivo (76). Pharmacological calcium channel
mRNA into protein (42). Accordingly, less ferritin pro- blockers interfere with this activity. Interestingly, the
tein is made when it is not needed for iron storage. spontaneous mk mutation in murine DMT1 effective-
IRP1 and IRP2 activities are regulated by iron, but ly converts the iron transporter to an efficient calcium
through distinct mechanisms. IRP1 is inactivated by channel, consistent with the idea that similar trans-
the incorporation of a 4Fe•4S cluster, which converts membrane pores conduct Fe2+ and Ca2+ (104). L-type
the protein into a cytoplasmic aconitase enzyme (38). calcium channels are widely expressed and thus could
IRP2 is inactivated by iron-dependent degradation be involved in NTBI uptake in other tissues.
(37, 47). In addition to interpreting cellular iron status,
IRP1 and IRP2 both respond to nitric oxide levels, IRP1 Iron Utilization and Recovery
responds to H2O2, and IRP2 is regulated by hypoxia
(39, 42, 77). Hemoglobin production in erythroid precursors is a
Several other iron-related mRNAs also contain IRE complex process requiring meticulous coordination of
elements. A single IRE is present in the 3’-UTR of one iron acquisition, protoporphyrin biosynthesis, and
splice isoform of DMT1 mRNA (45). 5’-IREs are pres- globin protein production. Perturbation of this bal-
ent in mRNAs encoding the erythroid form of the anced process invariably results in disease. Disorders
heme biosynthetic aminolevulinic acid synthase of hemoglobin synthesis give insight into iron home-
(eALAS; Refs. 14, 17) and the iron transporter ferro- ostasis.
portin (1, 21, 61). The 5’-IRE in the eALAS mRNA like- The protoporphyrin precursor of heme is construct-
ly serves to coordinate initiation of heme biosynthesis ed through a series of enzymatic reactions that take
with iron availability. The presence of IREs in DMT1 place alternately in the mitochondrion and the cyto-
and ferroportin mRNAs suggests that their expression plasm. Mutations in the enzymes involved in heme
may be controlled, at least in part, by cellular iron con- biosynthesis lead to a spectrum of diseases, several of
tent. Although this is not yet understood in detail, evi- which have iron phenotypes. Mutations in the first
dence for in vivo activity of the IRE in the ferroportin enzyme, ALAS2, result in sideroblastic anemia (15),
mRNA comes from the spontaneous mouse mutant characterized by accumulation of unused iron in the
polycythemia (pcm), which has a genomic deletion mitochondria of erythroid precursors, likely in the
that inactivates the ferroportin IRE, resulting in a com- form of mitochondrial ferritin (11). The final enzyme
plex phenotype with transient polycythemia in het- of heme biosynthesis, ferrochelatase, introduces iron
erozygotes (pcm/+) and iron deficiency anemia in into protoporphyrin IX to produce heme (16).
homozygotes (pcm/pcm) (63). Mutations in this protein result in erythropoietic pro-
Data from multiple animal models establish that the toporphyria, a rare disease resulting from accumula-
TF cycle is critical for iron uptake into erythroid cells tion of protoporphyrin lacking iron (12, 89, 91, 97).
(6, 20, 25, 26, 34, 58, 102). In addition, human muta- These patients can also have sideroblasts, similar to

PHYSIOLOGY • Volume 21 • April 2006 • www.physiologyonline.org 117


REVIEWS
patients with ALAS2 mutations. Sideroblastic anemia per oxidase, facilitates movement of iron out of tissue
can also result from mutations in proteins with less stores and into TF (40, 41).
direct roles in heme biosynthesis (24). For example, X-
linked sideroblastic anemia with spinocerebellar atax- Systemic Regulation and Regulatory
ia results from mutations in the mitochondrial trans- Factors
porter ABC7 (4), which is required for the maturation
of cytosolic iron sulfur (Fe•S) clusters. Control of iron balance in the whole organism requires
Recycling of iron from senescent erythrocytes pro- communication between sites of uptake, utilization,
vides most of the iron utilized by developing red blood and storage. In recent years, hepcidin (encoded by the
cells. Specialized macrophages in the spleen, bone HAMP gene) has emerged as a primary regulator of
marrow, and liver remove effete red cells from circula- iron homeostasis. Hepcidin is a circulating peptide
tion for recovery of their iron. Neither the signal on the secreted by hepatocytes (54, 79, 81) that binds to fer-
effete erythrocyte nor the macrophage receptor has roportin at the cell surface to initiate ferroportin inter-
been clearly identified to date, but several promising nalization and degradation (69). In the duodenal ente-
candidates have been described. Accumulation of rocyte, hepcidin-dependent regulation of ferroportin
phosphatidylserine in the outer leaflet of the erythro- reduces dietary iron absorption. In the macrophage
cyte membrane seems to be a primary event (13), but (and possibly the hepatocyte), hepcidin activity atten-
Ca2+ flux (57), removal of sialic acids on the cell sur- uates cellular iron release. Loss of hepcidin protein
face, and opsonization of red blood cells by autoanti- results in severe iron overload in mice (70) and human
bodies are all possible signals for turnover (8). The patients (78, 88), underscoring the central role of hep-
scavenger receptor CD36 (53, 93) is a plausible candi- cidin in regulating iron balance.
date for the macrophage-specific receptor. Consistent with its role as a negative regulator of
After the red cell has been internalized into an iron absorption, hepcidin expression is decreased in
acidic phagosome, heme oxygenase liberates iron response to anemia and hypoxia (71) (FIGURE 2).
from heme (83). The iron is likely transferred out of the This is advantageous, because it makes more iron
phagosome by DMT1 (48) for cellular storage or return available for erythropoiesis. In contrast, hepcidin
to the serum. The factors that influence whether the expression is increased in response to inflammation
macrophage retains or releases iron are not known, (67) and nongenetic iron overload (81). This response
but intracellular iron content (99), cytokine activity minimizes the availability of iron, inhibiting the
(86, 101), and reactive oxygen species such as nitric growth of pathogens and limiting the iron burden.
oxide (31) may be involved. Iron egress from Neither the factor that induces hepcidin in response to
macrophages is partly, if not entirely, mediated by fer- iron loading nor the factor that inhibits hepcidin in
roportin (22, 105). Ceruloplasmin, a serum multicop- response to anemia or hypoxia has been identified.
The unidentified ery-
NEGATIVE REGULATORS POSITIVE REGULATORS throid suppressor of hep-
cidin expression appears
Hypoxia Iron
overload to overcome the putative
sensor for iron loading,
Hepatocyte ?Fe2-TF
however, because hep-
cidin expression is sup-
Nucleus Mutations in HFE,
Soluble HJV TFR2, HJV, and HAMP pressed in anemias com-
result in decreased plicated by iron overload
hepcidin expression. such as hypotransfer-
rinemia (100) and tha-
HAMP lassemia (2). Although
IL-6
we have a general knowl-
Increased Inflammation edge of the conditions
erythroid drive that regulate hepcidin
HAMP = hepcidin gene expression, little is
known about the molec-
FIGURE 2. Regulation of hepcidin expression ular mechanisms of hep-
Hepatic production of the peptide hormone hepcidin is influenced by iron needs and cidin regulation. The dis-
stores. Anemia and hypoxia result in decreased hepcidin synthesis; inflammation, the covery of such factors
inflammatory cytokine interleukin-6 (IL-6), and increased iron stores (possibly through difer-
ric-TF) result in increased hepcidin synthesis. The molecular pathways involved in signal
will probably result from
transduction in response to these conditions have not yet been elucidated. The protein studies of hepatocytes,
products of hemochromatosis disease genes (HFE, TFR2, and HJV) are presumed to act as where several membrane
regulators of hepcidin expression because mutations in each result in inadequate hepcidin
production. A soluble cleavage product of HJV (sHJV) also has been shown to inhibit hep-
proteins (HFE, TFR2, and
cidin production. HJV) have already been

118 PHYSIOLOGY • Volume 21 • April 2006 • www.physiologyonline.org


REVIEWS
NORMAL REGULATION HEMOCHROMATOSIS

Fe Fe

DMT1 DMT1

Ferritin
1 Under normal conditions,
circulating hepcidin regulates
cellular iron export by binding
to ferroportin and inducing
its degradation in lysosomes. Lysosome

Lysosomal
degradation
of ferroportin Enterocyte

Hepcidin Ferroportin
Fe
2 In balance, some iron is 3 In hemochromatosis, hepcidin is
retained within enterocytes deficient or absent, resulting in
and macrophages and some increased ferroportin on the cell
iron is exported into the serum. surface and accelerated iron release.

Macrophage

Fe Ferroportin Lysosomal
4 Consequently, intestinal
degradation
iron absorption is
of ferroportin
increased and serum
iron levels rise.

FIGURE 3. The hepcidin-ferroportin axis


In hemochromatosis, hepcidin is deficient or absent, resulting in increased ferroportin on the cell surface and acceler-
ated iron release. Consequently, intestinal iron absorption is increased and serum iron levels rise.

implicated in the modulation of hepcidin expression of HFE expression remains controversial. The fact that
in vivo. Homozygous or compound heterozygous HFE forms a protein complex with TFR has led to the
mutations in any of these three proteins result in attractive hypothesis that a soluble factor such as
genetic hemochromatosis, a common iron-overload diferric TF (which competes with HFE for TFR bind-
disorder in human populations. ing) might modulate HFE activity and regulate a
HFE is an atypical major histocompatibility class I potential pathway signaling to the hepcidin (HAMP)
protein that is mutated in the majority of patients with promoter. To date, direct evidence supporting this
hemochromatosis (23). Mice (3, 65) and humans (10) hypothesis is lacking.
deficient in HFE have reduced hepatic expression of Transferrin receptor 2 (TFR2; a homolog of TFR) is
hepcidin despite iron overload. HFE is highly mutated in a small subset of patients with genetic
expressed in hepatocytes and is also expressed in hemochromatosis. Its normal function is unclear.
Kupffer cells (107). The physiologically significant site However, like HFE, deficiency of TFR2 attenuates hep-

PHYSIOLOGY • Volume 21 • April 2006 • www.physiologyonline.org 119


REVIEWS
cidin expression in mice (50) and humans (68) and iron in tissue macrophages (80). Each of these features
results in moderate iron overload. TFR2 is highly can be explained by modest hepcidin deficiency in
expressed in hepatocytes (52) but is also expressed in these individuals. Without hepcidin, there is inade-
erythroblasts (51). No erythropoietic abnormalities quate regulation of ferroportin at the basolateral sur-
have been observed in mice or humans with TFR2 face of enterocytes, contributing to increased dietary
deficiency, suggesting that the TFR2 requirement for iron uptake. Likewise, ferroportin activity remains ele-
hepcidin regulation is intrinsic to the hepatocyte and vated in macrophages, explaining their relative iron
not the result of feedback through the anemia/hypox- deficiency (FIGURE 3).
ia regulatory pathway. Diferric TF is also an attractive Patients with mutations in hepcidin and HJV pres-
candidate for communicating iron status to regulate ent in the second or third decade of life with elevated
hepcidin expression via TFR2 as TF induces TFR2 transferrin saturation and severe iron loading. The
expression in hepatocytes (49, 87). clinical picture is dominated by cardiomyopathy and
Hemojuvelin (HJV) is a homolog of proteins endocrinopathies. If untreated, iron-related organ
involved in neural patterning. It was recently damage is lethal by the fourth decade of life. Their
described as a hemochromatosis disease gene (78). hepcidin levels are lower than those in patients with
HJV deficiency in mice (43, 72) and humans (44, 78) HFE or TFR2-related hemochromatosis, consistent
reduces hepcidin levels further than either HFE or with the accelerated iron loading in these individuals.
TFR2 deficiency and, accordingly, results in more Mutations in ferroportin result in either of two dis-
severe iron loading. HJV is expressed in a number of tinct iron overload diseases, each inherited in an auto-
tissues including liver, heart, and skeletal muscle, and somal-dominant pattern (19, 64, 74, 90). Similar to
found in plasma in a soluble form. Recent data suggest other forms of hemochromatosis, some patients pres-
that cell-associated HJV is required for normal expres- ent with parenchymal iron overload and elevated
sion of hepcidin in hepatocytes and that soluble HJV transferrin saturation. These patients have mutant fer-
negatively regulates hepcidin expression in hepato- roportin that is expressed at the cell surface and is
cytes (59). The cellular source of soluble HJV and the capable of iron egress but resistant to regulation by
conditions that regulate its production are currently hepcidin. In contrast, other ferroportin mutations
unknown, but high expression of HJV in skeletal and cause mislocalization and/or loss of transporter func-
cardiac muscle suggests that these iron-rich tissues tion. These patients have little or no parenchymal iron
may communicate their iron needs through soluble loading and low to normal serum iron levels, but tissue
HJV. macrophages are iron-laden.
Induction of hepcidin in response to inflammation
is mediated, at least in part, by interleukin-6 (IL-6) (67) Future Directions
(FIGURE 2). The relationship between IL-6 induction
of hepcidin expression and regulation by HFE, TFR2, Now that most of the key proteins involved in iron
and HJV remains uncertain. Hepcidin induction caus- homeostasis have been identified, future investiga-
es hypoferremia through cellular iron-withholding, a tions must focus on understanding their molecular
response that contributes to innate immunity (67). functions. To date, there have been few structural
However, there is a co-existing deleterious effect. studies of DMT1, ferroportin, or HCP1, and it is not yet
Increased hepcidin levels also restrict the availability known how they carry out transmembrane iron trans-
of iron for erythropoiesis, resulting in the anemia of port. Although the stimuli that modulate hepcidin
inflammation, a common disorder observed in expression are well described as phenomena, we do
patients with inflammation, infection, organ failure, or not yet understand how physiological signals are
recent trauma. transduced to regulate hepcidin production. The
potent erythroid suppressor of hepcidin synthesis has
Pathogenesis of Iron-Overload not yet been identified. The detailed molecular activi-
Disorders ties of hemochromatosis disease proteins HFE, TFR2,
and HJV have not yet been reported. Undoubtedly, the
Studies focused on the regulation of iron homeostasis next decade should yield a more comprehensive
have converged on hepcidin as a common effector understanding of iron metabolism and likely inform
molecule, acting through its modulation of ferroportin other studies of the whole organism. 
activity (FIGURE 2). All known heritable disorders of References
iron overload involve mutations affecting hepcidin 1. Abboud S and Haile DJ. A novel mammalian iron-regulated
itself, regulators of hepcidin (HFE, TFR2, HJV), or the protein involved in intracellular iron metabolism. J Biol Chem
275: 19906-19912, 2000.
hepcidin target ferroportin.
2. Adamsky K, Weizer O, Amariglio N, Breda L, Harmelin A,
Patients with mutations in HFE and TFR2, pre- Rivella S, Rachmilewitz E, and Rechavi G. Decreased hepcidin
sumed regulators of hepcidin expression, generally mRNA expression in thalassemic mice. Br J Haematol 124:
123-124, 2004.
present in midlife with elevated transferrin saturation,
parenchymal iron loading, and proportionally less

120 PHYSIOLOGY • Volume 21 • April 2006 • www.physiologyonline.org


REVIEWS
3. Ahmad KA, Ahmann JR, Migas MC, Waheed A, 19. De Domenico I, Ward DM, Nemeth E, Vaughn 35. Gunshin H, Mackenzie B, Berger UV, Gunshin Y,
Britton RS, Bacon BR, Sly WS, and Fleming RE. MB, Musci G, Ganz T, and Kaplan J. The molecu- Romero MF, Boron WF, Nussberger S, Gollan JL,
Decreased liver hepcidin expression in the Hfe lar basis of ferroportin-linked hemochromatosis. and Hediger MA. Cloning and characterization of
knockout mouse. Blood Cells Mol Dis 29: 361- Proc Natl Acad Sci USA 102: 8955-8960, 2005. a mammalian proton-coupled metal-ion trans-
366, 2002. porter. Nature 388: 482-488, 1997.
20. Donovan A, Brownlie A, Dorschner MO, Zhou Y,
4. Allikmets R, Raskind WH, Hutchinson A, Schueck Pratt SJ, Paw BH, Phillips RB, Thisse C, Thisse B, 36. Gunshin H, Starr CN, Direnzo C, Fleming MD, Jin
ND, Dean M, and Koeller DM. Mutation of a puta- and Zon LI. The zebrafish mutant gene chardon- J, Greer EL, Sellers VM, Galica SM, and Andrews
tive mitochondrial iron transporter gene (ABC7) in nay (cdy) encodes divalent metal transporter 1 NC. Cybrd1 (duodenal cytochrome b) is not nec-
X-linked sideroblastic anemia and ataxia (DMT1). Blood 100: 4655-4659, 2002. essary for dietary iron absorption in mice. Blood
(XLSA/A). Hum Mol Genet 8: 743-749, 1999. 106: 2879-2883, 2005.
21. Donovan A, Brownlie A, Zhou Y, Shepard J, Pratt
5. Bannerman RM and Cooper RG. Sex-linked ane- SJ, Moynihan J, Paw BH, Drejer A, Barut B, Zapata 37. Guo B, Phillips JD, Yu Y, and Leibold EA. Iron reg-
mia: a hypochromic anemia of mice. Science 151: A, Law TC, Brugnara C, Lux SE, Pinkus GS, Pinkus ulates the intracellular degradation of iron regula-
581-582, 1966. JL, Kingsley PD, Palis J, Fleming MD, Andrews tory protein 2 by the proteasome. J Biol Chem
NC, and Zon LI. Positional cloning of zebrafish fer- 270: 21645-21651, 1995.
6. Bernstein SE. Hereditary hypotransferrinemia with roportin1 identifies a conserved vertebrate iron
hemosiderosis, a murine disorder resembling exporter. Nature 403: 776-781, 2000. 38. Haile DJ, Rouault TA, Tang CK, Chin J, Harford JB,
human atransferrinemia. J Lab Clin Med 110: 690- and Klausner RD. Reciprocal control of RNA-bind-
705, 1987. 22. Donovan A, Lima CA, Pinkus JL, Pinkus GS, Zon LI, ing and aconitase activity in the regulation of the
Robine S, and Andrews NC. The iron exporter fer- iron-responsive element binding protein: role of
7. Beutler E, Gelbart T, Lee P, Trevino R, Fernandez roportin (Slc40a1) is essential for iron homeosta- the iron-sulfur cluster. Proc Natl Acad Sci USA 89:
MA, and Fairbanks VF. Molecular characterization sis. Cell Metabolism 1: 191-200, 2005. 7536-7540, 1992.
of a case of atransferrinemia. Blood 96: 4071-
4074, 2000. 23. Feder JN, Gnirke A, Thomas W, Tsuchihashi Z, 39. Hanson ES, Foot LM, and Leibold EA. Hypoxia
Ruddy DA, Basava A, Dormishian F, Domingo R Jr, post-translationally activates iron-regulatory pro-
8. Bratosin D, Mazurier J, Tissier JP, Estaquier J, Ellis MC, Fullan A, Hinton LM, Jones NL, Kimmel tein 2. J Biol Chem 274: 5047-5052, 1999.
Huart JJ, Ameisen JC, Aminoff D, and Montreuil BE, Kronmal GS, Lauer P, Lee VK, Loeb DB, Mapa
J. Cellular and molecular mechanisms of senes- FA, McClelland E, Meyer NC, Mintier GA, Moeller 40. Harris ZL, Durley AP, Man TK, and Gitlin JD.
cent erythrocyte phagocytosis by macrophages. A N, Moore T, Morikang E, Wolff RK, et al. A novel Targeted gene disruption reveals an essential role
review. Biochimie 80: 173-195, 1998. MHC class I-like gene is mutated in patients with for ceruloplasmin in cellular iron efflux. Proc Natl
hereditary haemochromatosis. Nat Genet 13: 399- Acad Sci USA 96: 10812-10817, 1999.
9. Breuer W, Hershko C, and Cabantchik ZI. The 408, 1996.
importance of non-transferrin bound iron in disor- 41. Harris ZL, Takahashi Y, Miyajima H, Serizawa M,
ders of iron metabolism. Transfus Sci 23: 185-192, 24. Fleming MD. The genetics of inherited sideroblas- MacGillivray RT, and Gitlin JD.
2000. tic anemias. Semin Hematol 39: 270-281, 2002. Aceruloplasminemia: molecular characterization
of this disorder of iron metabolism. Proc Natl
10. Bridle KR, Frazer DM, Wilkins SJ, Dixon JL, Purdie 25. Fleming MD, Romano MA, Su MA, Garrick LM, Acad Sci USA 92: 2539-2543, 1995.
DM, Crawford DH, Subramaniam VN, Powell LW, Garrick MD, and Andrews NC. Nramp2 is mutat-
Anderson GJ, and Ram GA. Disrupted hepcidin ed in the anemic Belgrade (b) rat: evidence of a 42. Hentze MW and Kuhn LC. Molecular control of
regulation in HFE-associated haemochromatosis role for Nramp2 in endosomal iron transport. Proc vertebrate iron metabolism: mRNA-based regula-
and the liver as a regulator of body iron Natl Acad Sci USA 95: 1148-1153, 1998. tory circuits operated by iron, nitric oxide, and
homoeostasis. Lancet 361: 669-673, 2003. oxidative stress. Proc Natl Acad Sci USA 93: 8175-
26. Fleming MD, Trenor CC, 3rd Su MA, Foernzler D, 8182, 1996.
11. Cazzola M, Invernizzi R, Bergamaschi G, Levi S, Beier DR, Dietrich WF, and Andrews NC.
Corsi B, Travaglino E, Rolandi V, Biasiotto G, Microcytic anaemia mice have a mutation in 43. Huang FW, Pinkus JL, Pinkus GS, Fleming MD,
Drysdale J, and Arosio P. Mitochondrial ferritin Nramp2, a candidate iron transporter gene. Nat and Andrews NC. A mouse model of juvenile
expression in erythroid cells from patients with Genet 16: 383-386, 1997. hemochromatosis. J Clin Invest 115: 2187-2191,
sideroblastic anemia. Blood 101: 1996-2000, 2005.
2003. 27. Flo TH, Smith KD, Sato S, Rodriguez DJ, Holmes
MA, Strong RK, Akira S, and Aderem A. Lipocalin 44. Huang FW, Rubio-Aliaga I, Kushner JP, Andrews
12. Childs S, Weinstein BM, Mohideen MA, Donohue 2 mediates an innate immune response to bacter- NC, and Fleming MD. Identification of a novel
S, Bonkovsky H, and Fishman MC. Zebrafish drac- ial infection by sequestrating iron. Nature 432: mutation (C321X) in HJV. Blood 104: 2176-2177,
ula encodes ferrochelatase and its mutation pro- 917-921. 2004.
vides a model for erythropoietic protoporphyria.
Curr Biol 10: 1001-1004, 2000. 28. Galbraith RA, Sassa S, and Kappas A. Heme bind- 45. Hubert N and Hentze MW. Previously uncharac-
ing to murine erythroleukemia cells. Evidence for terized isoforms of divalent metal transporter
13. Connor J, Pak CC, and Schroit AJ. Exposure of a heme receptor. J Biol Chem 260: 12198-12202, (DMT)-1: implications for regulation and cellular
phosphatidylserine in the outer leaflet of human 1985. function. Proc Natl Acad Sci USA 99: 12345-
red blood cells. Relationship to cell density, cell 12350, 2002.
age, and clearance by mononuclear cells. J Biol 29. Gitlin D and Cruchaud A. On the kinetics of iron
Chem 269: 2399-2404, 1994. absorption in mice. J Clin Invest 41: 344-350, 46. Iolascon A, d’Apolito M, Servedio V, Cimmino F,
1962. Piga A, and Camaschella C. Microcytic anemia
14. Cox TC, Bawden MJ, Martin A, and May BK. and hepatic iron overload in a child with com-
Human erythroid 5’-aminolevulinate synthase: 30. Goetz DH, Holmes MA, Borregaard N, Bluhm ME, pound heterozygous mutations in DMT1. Blood
promoter analysis and identification of an iron- Raymond KN, and Strong RK. The neutrophil 107: 349-354, 2006.
responsive element in the mRNA. EMBO J 10: lipocalin NGAL is a bacteriostatic agent that inter-
1891, 1991. feres with siderophore-mediated iron acquisition. 47. Iwai K, Klausner RD, and Rouault TA.
Mol Cell 10: 1033-1043, 2002. Requirements for iron-regulated degradation of
15. Cox TC, Bottomley SS, Wiley JS, Bawden MJ, the RNA binding protein, iron regulatory protein
Matthews CS, and May BK. X-linked pyridoxine- 31. Gonzalez D, Drapier JC, and Bouton C. 2. EMBO J 14: 5350-5357, 1995.
responsive sideroblastic anemia due to a Thr388- Endogenous nitration of iron regulatory protein-1
to-Ser substitution in erythroid 5-aminolevulinate (IRP-1) in nitric oxide-producing murine 48. Jabado N, Canonne-Hergaux F, Gruenheid S,
synthase [see comments]. N Engl J Med 330: 675- macrophages: further insight into the mechanism Picard V, and Gros P. Iron transporter
679, 1994. of nitration in vivo and its impact on IRP-1 func- Nramp2/DMT-1 is associated with the membrane
tions. J Biol Chem 279: 43345-43351, 2004. of phagosomes in macrophages and Sertoli cells.
16. Dailey HA. Terminal steps of haem biosynthesis. Blood 100: 2617-2622, 2002.
Biochem Soc Trans 30: 590-595, 2002. 32. Grasbeck R, Majuri R, Kouvonen I, and Tenhunen
R. Spectral and other studies on the intestinal 49. Johnson MB and Enns CA. Regulation of transfer-
17. Dandekar T, Stripecke R, Gray NK, Goossen B, haem receptor of the pig. Biochim Biophys Acta rin receptor 2 by transferrin: diferric transferrin
Constable A, Johansson HE, and Hentze MW. 700: 137-142, 1982. regulates transferrin receptor 2 protein stability.
Identification of a novel iron-responsive element Blood 104: 4287-4293, 2004.
in murine and human erythroid delta-aminole- 33. Gruenheid S, Cellier M, Vidal S, and Gros P.
vulinic acid synthase mRNA. EMBO J 10: 1903- Identification and characterization of a second 50. Kawabata H, Fleming RE, Gui D, Moon SY, Saitoh
1909, 1991. mouse Nramp gene. Genomics 25: 514-525, T, O’Kelly J, Umehara Y, Wano Y, Said JW, and
1995. Koeffler HP. Expression of hepcidin is down-regu-
18. Dautry-Varsat A, Ciechanover A, and Lodish HF. lated in TfR2 mutant mice manifesting a pheno-
pH and the recycling of transferrin during recep- 34. Gunshin H, Fujiwara Y, Custodio AO, Direnzo C, type of hereditary hemochromatosis. Blood 105:
tor-mediated endocytosis. Proc Natl Acad Sci Robine S, and Andrews NC. Slc11a2 is required 376-381, 2005.
USA 80: 2258-2262, 1983. for intestinal iron absorption and erythropoiesis
but dispensable in placenta and liver. J Clin Invest
115: 1258-1266, 2005.

PHYSIOLOGY • Volume 21 • April 2006 • www.physiologyonline.org 121


REVIEWS
51. Kawabata H, Nakamaki T, Ikonomi P, Smith RD, 66. Muir A and Hopfer U. Regional specificity of iron 82. Pinkerton PH. Histological evidence of disordered
Germain RS, and Koeffler HP. Expression of trans- uptake by small intestinal brush-border mem- iron transport in the x-linked hypochromic
ferrin receptor 2 in normal and neoplastic branes from normal and iron-deficient mice. Am J anaemia of mice. J Pathol Bacteriol 95: 155-165,
hematopoietic cells. Blood 98: 2714-2719, 2001. Physiol Gastrointest Liver Pathol 248: G376-G379, 1968.
1985.
52. Kawabata H, Yang R, Hirama T, Vuong PT, Kawano 83. Poss KD and Tonegawa S. Heme oxygenase 1 is
S, Gombart AF, and Koeffler HP. Molecular cloning 67. Nemeth E, Rivera S, Gabayan V, Keller C, Taudorf required for mammalian iron reutilization. Proc
of transferrin receptor 2. A new member of the S, Pedersen BK, and Ganz T. IL-6 mediates hypo- Natl Acad Sci USA 94: 10919-10924, 1997.
transferrin receptor-like family. J Biol Chem 274: ferremia of inflammation by inducing the synthe-
20826-20832, 1999. sis of the iron regulatory hormone hepcidin. J Clin 84. Quigley JG, Yang Z, Worthington MT, Phillips JD,
Invest 113: 1271-1276, 2004. Sabo KM, Sabath DE, Berg CL, Sassa S, Wood BL,
53. Kiefer CR and Snyder LM. Oxidation and erythro- and Abkowitz JL. Identification of a human heme
cyte senescence. Curr Opin Hematol 7: 113-116, 68. Nemeth E, Roetto A, Garozzo G, Ganz T, and exporter that is essential for erythropoiesis. Cell
2000. Camaschella C. Hepcidin is decreased in TFR2 118: 757-766, 2004.
hemochromatosis. Blood 105: 1803-1806, 2005.
54. Krause A, Neitz S, Magert HJ, Schulz A, 85. Raffin SB, Woo CH, Roost KT, Price DC, and
Forssmann WG, Schulz-Knappe P, and Adermann 69. Nemeth E, Tuttle MS, Powelson J, Vaughn MB, Schmid R. Intestinal absorption of hemoglobin
K. LEAP-1, a novel highly disulfide-bonded human Donovan A, Ward DM, Ganz T, and Kaplan J. iron-heme cleavage by mucosal heme oxygenase.
peptide, exhibits antimicrobial activity. FEBS Lett Hepcidin regulates cellular iron efflux by binding J Clin Invest 54: 1344-1352, 1974.
480: 147-150, 2000. to ferroportin and inducing its internalization.
Science 306: 2090-2093, 2004. 86. Recalcati S, Taramelli D, Conte D, and Cairo G.
55. Krishnamurthy P, Ross DD, Nakanishi T, Bailey-Dell Nitric oxide-mediated induction of ferritin synthe-
K, Zhou S, Mercer KE, Sarkadi B, Sorrentino BP, 70. Nicolas G, Bennoun M, Devaux I, Beaumont C, sis in J774 macrophages by inflammatory
and Schuetz JD. The stem cell marker Grandchamp B, Kahn A, and Vaulont S. Lack of cytokines: role of selective iron regulatory protein-
Bcrp/ABCG2 enhances hypoxic cell survival hepcidin gene expression and severe tissue iron 2 downregulation. Blood 91: 1059-1066, 1998.
through interactions with heme. J Biol Chem 279: overload in upstream stimulatory factor 2 (USF2)
24218-24225, 2004. knockout mice. Proc Natl Acad Sci USA 98: 8780- 87. Robb A and Wessling-Resnick M. Regulation of
8785, 2001. transferrin receptor 2 protein levels by transferrin.
56. Lam-Yuk-Tseung S, Touret N, Grinstein S, and Blood 104: 4294-4299, 2004.
Gros P. Carboxyl-terminus determinants of the 71. Nicolas G, Chauvet C, Viatte L, Danan JL, Bigard
iron transporter DMT1/SLC11A2 isoform II X, Devaux I, Beaumont C, Kahn A, and Vaulont S. 88. Roetto A, Papanikolaou G, Politou M, Alberti F,
(–IRE/1B) mediate internalization from the plasma The gene encoding the iron regulatory peptide Girelli D, Christakis J, Loukopoulos D, and
membrane into recycling endosomes. hepcidin is regulated by anemia, hypoxia, and Camaschella C. Mutant antimicrobial peptide
Biochemistry 44: 12149-12159, 2005. inflammation. J Clin Invest 110: 1037-1044, 2002. hepcidin is associated with severe juvenile
hemochromatosis. Nat Genet 33: 21-22, 2003.
57. Lang F, Lang KS, Wieder T, Myssina S, Birka C, 72. Niederkofler V, Salie R, and Arber S. Hemojuvelin
Lang PA, Kaiser S, Kempe D, Duranton C, and is essential for dietary iron sensing, and its muta- 89. Rufenacht UB, Gouya L, Schneider-Yin X, Puy H,
Huber SM. Cation channels, cell volume and the tion leads to severe iron overload. J Clin Invest Schafer BW, Aquaron R, Nordmann Y, Minder EI,
death of an erythrocyte. Pflügers Arch 447: 121- 115: 2180-2186, 2005. and Deybach JC. Systematic analysis of molecular
125, 2003. defects in the ferrochelatase gene from patients
73. Nishi T and Forgac M. The vacuolar (H+)-ATPases: with erythropoietic protoporphyria. Am J Hum
58. Levy JE, Jin O, Fujiwara Y, Kuo F, and Andrews nature’s most versatile proton pumps. Nat Rev Genet 62: 1341-1352, 1998.
NC. Transferrin receptor is necessary for develop- Mol Cell Biol 3: 94-103, 2002.
ment of erythrocytes and the nervous system. Nat 90. Schimanski LM, Drakesmith H, Merryweather-
Genet 21: 396-399, 1999. 74. Njajou OT, Vaessen N, Joosse M, Berghuis B, van Clarke AT, Viprakasit V, Edwards JP, Sweetland E,
Dongen JW, Breuning MH, Snijders PJ, Rutten Bastin JM, Cowley D, Chinthammitr Y, Robson KJ,
59. Lin L, Goldberg YP, and Ganz T. Competitive reg- WP, Sandkuijl LA, Oostra BA, van Duijn CM, and and Townsend AR. In vitro functional analysis of
ulation of hepcidin mRNA by soluble and cell- Heutink P. A mutation in SLC11A3 is associated human ferroportin (FPN) and hemochromatosis-
associated hemojuvelin. Blood 106: 2884-2889, with autosomal dominant hemochromatosis. Nat associated FPN mutations. Blood 105: 4096-4102,
2005. Genet 28: 213-214, 2001. 2005.
60. McKie AT, Barrow D, Latunde-Dada GO, Rolfs A, 75. Ohgami RS, Campagna DR, Greer EL, Antiochos 91. Sellers VM, Dailey TA, and Dailey HA. Examination
Sager G, Mudaly E, Mudaly M, Richardson C, B, McDonald A, Chen J, Sharp JJ, Fujiwara Y, of ferrochelatase mutations that cause erythropoi-
Barlow D, Bomford A, Peters TJ, Raja KB, Shirali S, Barker JE, and Fleming MD. Identification of a fer- etic protoporphyria. Blood 91: 3980-3985, 1998.
Hediger MA, Farzaneh F, and Simpson RJ. An rireductase required for efficient transferrin-
iron-regulated ferric reductase associated with the dependent iron uptake in erythroid cells. Nat 92. Shayeghi M, Latunde-Dada GO, Oakhill JS, Laftah
absorption of dietary iron. Science 291: 1755- Genet 37: 1264-1269, 2005. AH, Takeuchi K, Halliday N, Khan Y, Warley A,
1759, 2001. McCann FE, Hider RC, Frazer DM, Anderson GJ,
76. Oudit GY, Sun H, Trivieri MG, Koch SE, Dawood F, Vulpe CD, Simpson RJ, and McKie AT.
61. McKie AT, Marciani P, Rolfs A, Brennan K, Wehr K, Ackerley C, Yazdanpanah M, Wilson GJ, Schwartz Identification of an intestinal heme transporter.
Barrow D, Miret S, Bomford A, Peters TJ, A, Liu PP, and Backx PH. L-type Ca2+ channels pro- Cell 122: 789-801, 2005.
Farzaneh F, Hediger MA, Hentze MW, and vide a major pathway for iron entry into cardiomy-
Simpson RJ. A novel duodenal iron-regulated ocytes in iron-overload cardiomyopathy. Nat Med 93. Smith TG, Serghides L, Patel SN, Febbraio M,
transporter, IREG1, implicated in the basolateral 9: 1187-1194, 2003. Silverstein RL, and Kain KC. CD36-mediated
transfer of iron to the circulation. Mol Cell 5: 299- nonopsonic phagocytosis of erythrocytes infected
309, 2000. 77. Pantopoulos K and Hentze MW. Activation of iron with stage I and IIA gametocytes of Plasmodium
regulatory protein-1 by oxidative stress in vitro. falciparum. Infect Immun 71: 393-400, 2003.
62. Mims MP, Guan Y, Pospisilova D, Priwitzerova M, Proc Natl Acad Sci USA 95: 10559-10563, 1998.
Indrak K, Ponka P, Divoky V, and Prchal JT. 94. Theil EC. Ferritin: at the crossroads of iron and
Identification of a human mutation of DMT1 in a 78. Papanikolaou G, Samuels ME, Ludwig EH, oxygen metabolism. J Nutr 133: 1549-1553, 2003.
patient with microcytic anemia and iron overload. MacDonald ML, Franchini PL, Dube MP, Andres L,
Blood 105: 1337-1342, 2005. MacFarlane J, Sakellaropoulos N, Politou M, 95. Torti FM and Torti SV. Regulation of ferritin genes
Nemeth E, Thompson J, Risler JK, Zaborowska C, and protein. Blood 99: 3505-3516, 2002.
63. Mok H, Jelinek J, Pai S, Cattanach BM, Prchal JT, Babakaiff R, Radomski CC, Pape TD, Davidas O,
Youssoufian H, and Schumacher A. Disruption of Christakis J, Brissot P, Lockitch G, Ganz T, Hayden 96. Tsushima RG, Wickenden AD, Bouchard RA, Oudit
ferroportin 1 regulation causes dynamic alter- MR, and Goldberg YP. Mutations in HFE2 cause GY, Liu PP, and Backx PH. Modulation of iron
ations in iron homeostasis and erythropoiesis in iron overload in chromosome 1q-linked juvenile uptake in heart by L-type Ca2+ channel modifiers:
polycythaemia mice. Development 131: 1859- hemochromatosis. Nat Genet 36: 77-82, 2004. possible implications in iron overload. Circ Res 84:
1868, 2004. 1302-1309, 1999.
79. Park CH, Valore EV, Waring AJ, and Ganz T.
64. Montosi G, Donovan A, Totaro A, Garuti C, Hepcidin, a urinary antimicrobial peptide synthe- 97. Tutois S, Montagutelli X, Da Silva V, Jouault H,
Pignatti E, Cassanelli S, Trenor CC, Gasparini P, sized in the liver. J Biol Chem 276: 7806-7810, Rouyer-Fessard P, Leroy-Viard K, Guenet JL,
Andrews NC, and Pietrangelo A. Autosomal-dom- 2001. Nordmann Y, Beuzard Y, and Deybach JC.
inant hemochromatosis is associated with a muta- Erythropoietic protoporphyria in the house
tion in the ferroportin (SLC11A3) gene. J Clin 80. Pietrangelo A. Hereditary hemochromatosis: a mouse. A recessive inherited ferrochelatase defi-
Invest 108: 619-623, 2001. new look at an old disease. N Engl J Med 350: ciency with anemia, photosensitivity, and liver dis-
2383-2397, 2004. ease. J Clin Invest 88: 1730-1736, 1991.
65. Muckenthaler M, Roy CN, Custodio AO, Minana
B, deGraaf J, Montross LK, Andrews NC, and 81. Pigeon C, Ilyin G, Courselaud B, Leroyer P, Turlin 98. Vulpe CD, Kuo YM, Murphy TL, Cowley L, Askwith
Hentze MW. Regulatory defects in liver and intes- B, Brissot P, and Loreal O. A new mouse liver-spe- C, Libina N, Gitschier J, and Anderson GJ.
tine implicate abnormal hepcidin and Cybrd1 cific gene, encoding a protein homologous to Hephaestin, a ceruloplasmin homologue implicat-
expression in mouse hemochromatosis. Nat human antimicrobial peptide hepcidin, is overex- ed in intestinal iron transport, is defective in the
Genet 34: 102-107, 2003. pressed during iron overload. J Biol Chem 276: sla mouse. Nat Genet 21: 195-199, 1999.
7811-7819, 2001.

122 PHYSIOLOGY • Volume 21 • April 2006 • www.physiologyonline.org


REVIEWS
99. Wardrop SL and Richardson DR. The effect of 102. Wingert RA, Brownlie A, Galloway JL, Dooley K, 105. Yang F, Liu XB, Quinones M, Melby PC, Ghio A,
intracellular iron concentration and nitrogen Fraenkel P, Axe JL, Davidson AJ, Barut B, Noriega and Haile DJ. Regulation of reticuloendothelial
monoxide on Nramp2 expression and non-trans- L, Sheng X, Zhou Y, and Zon LI. The chianti iron transporter MTP1 (Slc11a3) by inflammation.
ferrin-bound iron uptake. Eur J Biochem 263: 41- zebrafish mutant provides a model for erythroid- J Biol Chem 277: 39786-39791, 2002.
49, 1999. specific disruption of transferrin receptor 1.
Development 131: 6225-6235, 2004. 106. Yang J, Goetz D, Li JY, Wang W, Mori K, Setlik D,
100. Weinstein DA, Roy CN, Fleming MD, Loda MF, Du T, Erdjument-Bromage H, Tempst P, Strong R,
Wolfsdorf JI, and Andrews NC. Inappropriate 103. Worthington MT, Cohn SM, Miller SK, Luo RQ, and Barasch J. An iron delivery pathway mediated
expression of hepcidin is associated with iron and Berg CL. Characterization of a human plasma by a lipocalin. Mol Cell 10: 1045-1056, 2002.
refractory anemia: implications for the anemia of membrane heme transporter in intestinal and
chronic disease. Blood 100: 3776-3781, 2002. hepatocyte cell lines. Am J Physiol Gastrointest 107. Zhang AS, Xiong S, Tsukamoto H, and Enns CA.
Liver Physiol 280: G1172-G1177, 2001. Localization of iron metabolism-related mRNAs in
101. Weiss G, Bogdan C, and Hentze MW. Pathways rat liver indicate that HFE is expressed predomi-
for the regulation of macrophage iron metabolism 104. Xu H, Jin J, DeFelice LJ, Andrews NC, and nantly in hepatocytes. Blood 103: 1509-1514,
by the anti-inflammatory cytokines IL-4 and IL-13. Clapham DE. A spontaneous, recurrent mutation 2004.
J Immunol 158: 420-425, 1997. in divalent metal transporter-1 exposes a calcium
entry pathway. PLoS Biol 2: E50, 2004.

PHYSIOLOGY • Volume 21 • April 2006 • www.physiologyonline.org 123

Potrebbero piacerti anche