You are on page 1of 6

Biochimie 91 (2009) 1223–1228

Contents lists available at ScienceDirect

Biochimie
journal homepage: www.elsevier.com/locate/biochi

Review

Hepcidin, the iron watcher


Lydie Viatte, Sophie Vaulont*
Institut Cochin, Département Endocrinologie, Métabolisme et Cancer, Paris F-75014 France; Inserm, U567; Université Paris Descartes, Faculté de Médecine René Descartes;
CNRS, UMR-S 8104

a r t i c l e i n f o a b s t r a c t

Article history: Hepcidin, a peptide hormone produced by the liver, constitutes the master regulator of iron homeostasis
Received 10 April 2009 in mammals allowing iron adaptation according to the body iron needs. In recent years there has been
Accepted 15 June 2009 important breakthrough in our knowledge of hepcidin regulation that has also implications for under-
Available online 23 June 2009
standing the physiopathology of human iron disorders. Different aspects of hepcidin regulation will be
considered in this review, including regulation by the iron status and the BMP6/HJV/SMAD pathway.
Keywords:
Hepcidin dysregulation in iron disorders will be also discussed. Although much can already be accom-
Metal
plished for treating iron disorders using the knowledge that has currently been developed, additional
Hepcidin
Iron disorders issues will be challenging for the coming years.
BMP/hemojuvelin Ó 2009 Published by Elsevier Masson SAS.
HFE/TfR

1. Introduction 2. Discovery of hepcidin

Iron is a vital biocatalyst that coexists in an oxidised insoluble Although dysregulations of iron metabolism were first
(Fe3þ) or a reduced soluble form (Fe2þ). The chemical properties of described in the XIXth century, little was known until recently
iron makes it an essential element for many functions in particular about the molecules involved in the regulation of iron homeostasis.
transport of oxygen. In mammals, 1–2 mg of iron are absorbed daily In 2001, two independent laboratories purified a new antimicrobial
by the duodenum, this represents the amount lost daily through peptide from blood [2] and urine [3]. This small 25 amino acid
normal blood loss and desquamation. The main consumer of iron in peptide produced by the liver proved to be the iron hormone
the body is the bone marrow for the production of erythrocytes sought for many years. It was baptised hepcidin, ‘‘hep’’ for hepatic
(approximately 20 mg iron/day). This iron is recycled by the and ‘‘cidin’’ for its antimicrobial activity. As expected for an iron-
macrophages of the reticulo-endothelial system that phagocytose regulatory hormone, hepcidin was found to be up-regulated by iron
senescent red blood cells, degrade hemoglobin containing iron and overload [4] and downregulated by iron deficiency [5]. Studies in
release iron in the bloodstream [1]. transgenic mice showed that hepcidin deficiency leads to iron
The dark side of the metal is that, in excess, it can be toxic and overload [6] and that overexpression of hepcidin leads to severe
produce free radicals through the Fenton reaction. Human iron iron deficiency and anemia [7]. Thus, hepcidin maintains iron
disorders unfortunately illustrate these two aspects of iron: homeostasis by inhibiting the intestinal absorption and the release
patients with iron deficiency due to poor iron content in the diet, of iron by macrophages. This hyposideremic activity of hepcidin is
abnormal blood loss, or chronic disease, but also, in rare cases, with mediated through the control of the surface expression of the iron
inherited diseases, present with anemia. On the other side of the exporter ferroportin on duodenal enterocytes and iron recycling
spectrum, patients suffering from iron overload, mainly hereditary macrophages. Hepcidin was shown to be able to bind to ferroportin,
hemochromatosis (HH), have multiple symptoms due to organs leading to the internalization and degradion of the iron exporter
dysfunction such as diabetes, arthritis, liver fibrosis, cirrhosis, [8], thereby decreasing iron availability in the circulation.
cardiomyopathy and hypogonadism. Iron homeostasis must
therefore be finely tuned. First discovered as an antimicrobial 3. Regulation of hepcidin
peptide, hepcidin proved to be the long-sought iron hormone.
Hepcidin is mainly produced by the liver but recent studies
describe production of hepcidin in other tissues: macrophages
* Corresponding author. Institut Cochin, Faculté de Médecine Cochin Port Royal,
24, rue du Faubourg Saint Jacques 75014 Paris, France. Tel.: þ33 1 44412408; fax:
[9–11], pancreatic beta cells [12], kidney [13], and adipocytes [14].
þ33 1 44412421. However, the contribution of extrahepatic hepcidin to the circulating
E-mail address: sophie.vaulont@inserm.fr (S. Vaulont). pool is unknown and, in contrast to liver hepcidin, hepcidin gene

0300-9084/$ – see front matter Ó 2009 Published by Elsevier Masson SAS.


doi:10.1016/j.biochi.2009.06.012
1224 L. Viatte, S. Vaulont / Biochimie 91 (2009) 1223–1228

expression in these cell types seems to be more sensitive to inflam- 3.1.2. HJV, BMP and TMPRSS6
mation than to iron [10,11,14]. A possible role of extrahepatic hepci- Hemojuvelin (HJV or RGMc) was identified in patients pre-
din could be the control of local iron fluxes under particular stimuli. senting a severe form of HH: hemochromatosis type 2 or juvenile
Hepcidin is produced as a pre-pro-protein of 84 amino-acids hemochromatosis [39]. HJV is a Bone Morphogenetic Protein (BMP)
composed of a signal peptide, a pro-region and the mature 25 co-receptor [40]. BMPs are members of the TGFb family; they
amino acid peptide. Pro-hepcidin was shown to be biologically induce hepcidin gene transcription through the SMAD1,5,8/SMAD4
inactive [15]. In the hepatocyte, hepcidin maturation occurs via pathway [41](see Fig. 1). Ablation of SMAD4 specifically in the liver,
a furin cleavage site recognized mainly by the prohormone con- triggers an iron overload in multiple organs due to decreased levels
vertase furin [16–18]. The exact conditions and mechanisms of of liver hepcidin [42]. Several different BMPs were able in vitro
regulation of hepcidin maturation and secretion are largely [40,42,43] and in vivo [41] to induce hepcidin but recent data
unknown, and the role for the presence of pro-hepcidin in the suggest that BMP6 is essential for hepcidin up-regulation by iron.
serum remains elusive. Only one commercially available ELISA is BMP6 gene expression is directly regulated by the amount of iron
used to detect serum pro-hepcidin. However, the antibody is and increasing amount of phosphorylation of Smad1,5,8 could be
detecting a non-matured form of 10 kDa [19] which would corre- directly related to iron-dependent expression of hepcidin in vivo
spond to the pre-pro-hepcidin containing the peptide signal rather [44,45]. Furthermore, recent reports demonstrate that BMP6
than pro-hepcidin estimated at 7.8 kDa. Furthermore, to date, the knockout mice present the same iron overload phenotype as
physiological significance of this assay, and thus its interest in SMAD4 and HJV KO mice due to hepcidin deficiency [46,47]
clinical studies, is not validated and no relatioship between pro- demonstrating that other BMPs cannot compensate for the loss of
hepcidin and iron absorption was demonstrated (for review [20]). BMP6. Two functional BMP response elements (RE) have been
described in hepcidin promoter: one located at position 84
3.1. Regulation by iron status nucleotides from transcription start, and one distal element located
at position 2301 [48,49]. Very recently a patient with a mutation
To limit iron toxicity, hepcidin is physiologically induced in in the proximal BMP-RE was described [50]. This patient has low
situation of iron overload to decrease circulating iron levels [4,21]. serum hepcidin level and massive iron overload associated with
Conversely, during iron deficiency, hypoxia or erythroid expansion hypogonadism, melanodermia and osteoporosis, highlighting the
[5,22–24], hepcidin expression is reduced to allow efficient iron critical role of BMP/HJV pathway to regulate hepcidin expression.
mobilization. HJV is a GPI-anchored protein that can be cleaved and released
Identification of the proteins involved in HH, namely HFE, TfR2 in a soluble form, sHJV[51]. Interestingly, as for hepcidin matura-
and HJV, shed a new light in understanding, at the molecular level, tion, sHJV is produced after cleavage by the prohormone convertase
the iron regulation of hepcidin. Mutations in these proteins lead to furin [52]. Release of sHJV is regulated by iron [51,53] and hypoxia
increased iron absorption and recycling due to inappropriate levels [54]. In contrast to membrane-bound HJV, sHJV inhibits iron-
of hepcidin [25]. induced hepcidin production and when injected into mice,
increases serum iron through reduction of phosphorylated
3.1.1. HFE and transferrin receptors (TfR1 & TfR2) SMAD1,5,8 levels [41]. A possible mechanism for sHJV inhibition of
HFE is the main gene responsible for HH. It is a member of HLA hepcidin is that it competes with HJV for binding with BMPs
class I family. HFE is expressed in many different tissues, including (see Fig. 1).
enterocytes and hepatocytes. With the increased iron absorption in A new partner of the HJV/BMP pathway was recently involved in
hemochromatotic patients and evidence that HFE could interact hepcidin regulation: the serine protease TMPRSS6 (also known as
with transferrin receptor 1 (TfR1) [26], HFE was first thought to act matriptase-2, for review, see [55]). This transmembrane serine
as a regulator of iron homeostasis by sensing iron at the baso-lateral protease is mainly expressed in the liver. Mice with ‘‘mask’’ muta-
surface of immature enterocytes, also referred to as the ‘‘crypt tion of Tmprss6 are characterized by abnormal hair distribution and
model’’ [27]. Recently, with the discovery of hepcidin, attention was iron-deficiency anemia associated with inappropriately high levels
brought to the liver as the center for iron regulation. Tissue-specific of hepcidin [56]. Tmprss6 KO mice present a similar phenotype [57].
HFE knockout mouse models demonstrated that, while ablation of In humans, TMPRSS6 deficiency is responsible for iron-refractory
HFE in enterocytes had no effect on iron metabolism [28], liver- iron-deficient anemia (IRIDA) [58–60]. Anemia of these patients is
specific knockout of HFE recapitulated the phenotype of total HFE poorly improved by oral iron therapy due to inhibited iron
KO [29], i.e. multivisceral iron overload. absorption. The high levels of hepcidin in spite of iron-deficiency
While the exact mechanisms by which HFE regulates hepcidin anemia suggest that TMPRSS6 is an inhibitor of hepcidin synthesis
expression in the liver are not completely known, recent data help [56]. In vitro data suggest that TMPRSS6 has proteolytic activity
to uncover possible pathways. First, it was shown that HFE can also against membrane-bound HJV [61] (see Fig. 1). How TMPRSS6 is
interact with TfR2 [30] in addition to TfR1. While TfR1 is ubiquitous regulated by iron remains to be determined.
and used by most of the cells for uptake of iron bound to transferrin
(Tf-Fe2), TfR2 expression is more restricted, with high levels found 3.2. Regulation by inflammation
in the liver [31], its affinity toward Tf-Fe2 is much weaker and, in
contrast to TfR1, TfR2 levels respond to Tf-Fe2 independent of Linked to its function as an antimicrobial peptide, hepcidin is
changes in cellular iron status [32,33]. TfR2 was found to be also induced by inflammation. The induction of hepcidin in
mutated in patients suffering from HH [34] demonstrating that it is inflammatory context is thought to play a major role in the setting
not essential for iron uptake by the hepatocytes. TfR2 is thought to of anemia of inflammation (also called anemia of chronic diseases).
act with HFE to regulate hepcidin expression. A recent model is When mice are treated with turpentine oil or LPS, liver hepcidin
proposing that when Tf-Fe2 is present in high amounts, it binds to [4,22] but also macrophage hepcidin [62] is induced. In humans,
TfR1, releasing HFE that can then bind to TfR2 [35,36]. Conse- LPS was also shown to increase urinary hepcidin [63]. While IL6
quently, the HFE-TfR2 complex would signal, possibly through [21,64,65] and IL1 b [65,66] induce hepcidin transcription, TNFa
the MAPK/ERK pathway [37], to induce hepcidin expression seems to have no effect [9,64,67] or even repress hepcidin [21,66].
(see Figure). It remains unclear whether HFE is necessary for IL6-induced expression of hepcidin is dependent on the JAK/STAT3
iron-sensing or just for basal expression of hepcidin [38]. pathway, and the STAT3 binding site has been identified in hepcidin
L. Viatte, S. Vaulont / Biochimie 91 (2009) 1223–1228 1225

Fig. 1. The expression of hepcidin is dependent on opposing signalling pathways: the combined effects of the various pathways will determine hepcidin levels. In conditions of iron
deficiency, sHJV inhibitor is high, BMP6 activator is low and mHJV is degraded by the activated serine protease TMPRSS6, contributing to decreased hepcidin gene expression.
The activity of an unknown factor mediated by increased erythropoiesis has also been postulated to contribute to hepcidin decrease to allow iron mobilization. Similarly, by
a yet unknown mechanism, has GDF15 been proposed as a direct repressor of hepcidin in cultured cells. EPO was proposed to act either directly (through EPOR-mediating decreased
C/EBPa activity) or indirectly, by inhibiting both inflammatory and iron-sensing pathway by decreasing STAT3 and SMAD signaling. Finally, stabilization of HIFa by iron deficiency or
hypoxia was shown to downregulate hepcidin expression. As a consequence of iron deficiency, TfR1 is bound to HFE. In conditions of increased holotransferrin, BMP6 is increased,
sHJV is decreased and the HJV/SMAD pathway is fully activated leading to enhanced hepcidin gene expression. As serum iron saturation increases, HFE is dislodged from its
overlapping binding site on TfR1 by holotransferrin. HFE is then freed to interact with TfR2 and to signal, possibly through the activation of ERK1/2, for the up-regulation of hepcidin.
In inflammatory conditions, the classical IL6/JAK/STAT3 pathway has been characterized to be responsible for hepcidin gene activation (the crosstalk between the inflammatory
signals and the iron-sensing pathway is not represented).

promoter next to the proximal BMP-RE [68–70] (see Fig. 1). It seems recently proposed that EPO could act directly to repress hepcidin in
that there is some crosstalk between the BMP/SMAD and JAK/STAT hepatocytes through EPOR-mediated regulation of the transcrip-
pathways to control hepcidin levels: mutation of the proximal tion factor C/EBPa [74]. Alternatively, Huang et al. recently
BMP-RE decreases IL6 response [70] as well as sHJV treatment [41]. proposed that EPO can inhibit hepcidin expression indirectly via
In contrast, BMP2 response is increased when STAT3 binding site is the suppression of STAT3 and SMAD4 signaling in vivo [75].
deleted [70] and conversely hepcidin response to LPS is increased in The major hypoxia-mediated gene regulation system, involving
BMP6 KO mice [46]. Hypoxia Inducible Factors, HIFa was recently reported to control
hepcidin synthesis [76]. In normoxia, the regulatory subunit a is
hydroxylated by the oxygen- and iron-dependent prolyl-hydroxy-
3.3. Regulation by anemia, hypoxia and increased erythropoiesis
lases (PHD), and degraded through the ubiquitin-proteasome
pathway via its interaction with the von Hippel-Lindau (VHL)
Iron-deficient anemia and hypoxia inhibit the synthesis of
tumor suppressor protein. Conversely, under hypoxia or iron
hepcidin allowing iron supply to match erythropoietic demand
depletion, hydroxylation is inhibited resulting in stabilization of the
[22]. This response is most likely multifactorial and some of the
a-subunit, heterodimerization with ARNT, nuclear translocation
signals require active erythropoiesis [71] (see Fig. 1).
and trans-activation of HIF-target genes such as EPO [77]. Hepcidin
Recently, Tanno et al.[72] reported that GDF15, a member of the
was recently shown to be a direct negative target of HIF-1: HIF-1
TGF-b family, mediates hepcidin repression in thalassemia. Thal-
binds to the hepcidin promoter, decreasing hepcidin levels.
assemia patients present anemia due to hemolysis of abnormal red
Consequently, in mice harboring hepatic deletion of vHL, HIF 1 is
blood cells and subsequent expansion of the erythroid compart-
stabilized and mice presented with dramatic decreased hepcidin
ment. These patients have high concentrations of GDF15 in their
levels [76] (see Fig. 1).
serum and Tanno et al. further showed that GDF15 could repress
Finally, an increase in reactive oxygen species (ROS) levels in
hepcidin expression directly on hepatoma cells. Interestingly,
hypoxic conditions has also been proposed as a possible mecha-
GDF15 was recently shown to be up-regulated by stimuli that
nism responsible for hepcidin repression via alteration of C/EBPa
deplete cells of iron [73]. In agreement, induction of serum GDF15
and STAT-3 activities [78].
was observed in iron-deficient subjects as well as following iron
chelator administration in normal subjects [73].
Erythropoietin (EPO) is synthesized by the kidney in hypoxic 4. Mode of action of hepcidin
conditions and acts as an anti-apoptotic agent on erythroblasts
through its receptor EPOR. As observed in mice under hypoxia [22], Hepcidin was identified for its antimicrobial properties and
injection of EPO inhibits hepcidin expression [23]. Pinto el al. classified as member of the defensin family [2,3]. The role of
1226 L. Viatte, S. Vaulont / Biochimie 91 (2009) 1223–1228

hepcidin as an antimicrobial peptide in mammals has been regar- Furthermore, studies in mice have shown that overexpression of
ded as unlikely since this antimicrobial activity was efficient at hepcidin can prevent [99] or cure [100] iron overload of Hfe KO
concentrations much higher than those found in the blood [2,3] and mice.
by the fact that physiological blood concentrations of NaCl was The iron exporter ferroportin itself was found to be mutated in
shown to inhibit this activity [3]. Furthermore, exogenous hepcidin the iron overload disorder called the ferroportin disease (for
added to infected macrophages enhanced bacterial growth in the review, [101]). Ferroportin disease has an autosomal dominant
macrophages [79]. This effect is thought to be mediated by iron mode of inheritance and phenotypic characteristics that differen-
retention caused by decreased ferroportin levels of the macro- tiate it from the other forms of HH.
phages, the intracellular pathogens using this iron for their Interestingly, it was shown that specific mutations are respon-
proliferation. sible for the two main phenotypes of the ferroportin disease.
While circulating hepcidin seems to be inefficient as an anti- Mutations that result in loss or reduction in iron transport activity
microbial peptide, it was recently proposed that hepcidin produced lead to the typical macrophage iron loading with high serum
endogenously by the macrophages and localized in the phagosome ferritin and normal transferrin saturation. Other mutations retain
could play a role in host defense against tuberculosis by causing normal iron transport activity but are insensitive to hepcidin-
structural damage to the mycobacteria [11]. Of note, while hepcidin induced internalization leading thus to an elevated transferrin
peptide of 20 amino-acids purified from the urine have similar saturation, moderately increased serum ferritin and hepatocyte
antimicrobial activity as the 25 amino acid peptide [3], it it is unable iron accumulation as seen in classical HH.
to degrade ferroportin in vitro and in vivo [80], showing that these
two functions of hepcidin are not overlapping. 5.2. Anemia of inflammation (or anemia of chronic diseases)
The role of hepcidin as a regulator of homeostasis has been
extensively studied and a mechanism for ferroportin degradation Low-grade inflammation is usually associated with anemia.
has been proposed. It was first shown that overexpressed tagged- While proinflammatory cytokines can directly inhibit erythropoi-
ferroportin could be internalized and degraded in vitro by hepcidin esis, EPO synthesis or downregulate ferroportin, interests slowly
treatment [8]. This observation could be reproduced by other grew on the contribution of hepcidin in the setting of anemia of
groups in vitro [81,82] and in the more physiological conditions of inflammation (for review, see [102]). In mice, decreased serum iron
bone-marrow derived macrophages [83]. In vivo, hepcidin defi- consequently to an acute inflammation caused by turpentine
ciency was shown to be associated with increased ferroportin injection was prevented in hepcidin-deficient mice [22]. In
protein levels [84]. The mechanism by which hepcidin binding to humans, although up-regulation of hepcidin was found in patients
ferroportin leads to degradation of the iron exporter was suggested with anemia of chronic diseases [24,64,103], further investigations
by the work of De Domenico et al. They showed that hepcidin are needed to determine the precise role of increased hepcidin
binding to its binding site (HBD for Hepcidin Binding Site) on the levels in the physiopathogeny of anemia of inflammation.
exporter [85] leads to JAK2-dependent tyrosines phosphorylation
[86,87]. Once internalized, ferroportin is dephosphorylated and 5.3. Detection of hepcidin
degraded after ubiquitination [86]. Whether ferroportin is
a monomer or a multimer is still a point of controversy [88]. The discovery of the hormonal iron-regulating role of hepcidin,
Recently ferroportin was proposed to be a dimer and it was shown followed by the elucidation of its mechanism of action -to regulate
that both monomers have to bind hepcidin for JAK2 to bind to and ferroportin levels- herald exciting times in the field of iron
to phosphorylate ferroportin [87]. These results, showing that metabolism and related disorders and offer new clinical potential in
cooperation between the ferroportin monomers is required for terms of diagnosis and therapeutics. It became rapidly obvious that
hepcidin-mediated ferroportin down-regulation, provide a molec- a reliable method to detect mature hepcidin in human samples was
ular explanation for the dominant inheritance of hepcidin-resistant highly desirable for the diagnosis and clinical management of iron
iron overload disease (see below). disorders. However, the generation of hepcidin antibodies has been
hampered due to the very low immunogenicity of the peptide. To
5. Hepcidin at the center of iron disorders date, only one team has produced antibodies against the biologi-
cally active peptide. They first developed a dot blot system for
5.1. Hereditary hemochromatosis (HH) urinary hepcidin measurement [21], and recently succeeded in
elaborating an ELISA detecting serum hepcidin [103]. To bypass the
Hemochromatosis was first described and named in the XIXth difficulty of measuring serum hepcidin levels, some laboratories
century. But it’s only in 1996 that mutations in HFE were identified have developed semi-quantitative methods using mass spectrom-
as responsible for genetic hemochromatosis [89]. C282Y mutation etry [104–107] (for review, see [108]). However, these methods
of HFE is very frequent and when at homozygous state, is respon- have reported limits of detection and reproducibility. Interestingly,
sible for the most common form of hemochromatosis. Later, iden- De Domenico et al. used an innovating assay to measure hepcidin in
tification of mutations in TFR2, HJV, hepcidin and ferroportin genes the serum. Using the synthetic peptide corresponding to the HBD of
followed(for review, see [90]). Hemochromatosis is characterized ferroportin, they developed a competitive assay with radiolabeled
by iron overload of multiple organs causing cirrhosis, cardiomy- hepcidin to measure hepcidin levels in biological fluids [85].
opathy, arthritis, diabetes when not treated. In the most severe
forms, due to mutations in HJV or hepcidin genes, patients present 6. Conclusion
severe cardiac disease and hypogonadism.
All forms of HH are supposed to result from either inappropriate The discovery of hepcidin has revolutionized the field of iron
levels of hepcidin [39,91–95] or, for mutations in ferroportin gene, homeostasis. This small antimicrobial peptide proved to be the
from resistance to hepcidin. It is now assumed that the severity of long-sought iron hormone and the liver became the center of
symptoms is associated to the remaining hepcidin levels found in interest, especially to understand the many complexes pathways
the patients. This hypothesis is reinforced by the fact that muta- regulating hepcidin expression (see Fig. 1). Its role in hemochro-
tions in hepcidin genes in the context of patients homozygotes matosis helped to sort out a heterogenous family of diseases into
for C282Y aggravates the phenotype of the patients [96–98]. one common etiology: dysregulation of hepcidin.
L. Viatte, S. Vaulont / Biochimie 91 (2009) 1223–1228 1227

Signalling from tissue iron stores or erythropoietic activity of the [34] C. Camaschella, A. Roetto, A. Cali, M. De Gobbi, G. Garozzo, M. Carella,
N. Majorano, A. Totaro, P. Gasparini, Nat. Genet. 25 (2000) 14–15.
bone marrow ends up by regulating hepcidin synthesis, revealing
[35] J. Gao, J. Chen, M. Kramer, H. Tsukamoto, A.S. Zhang, C.A. Enns, Cell Metab. 9
unexpected pathways and much complexity. However, a significant (2009) 217–227.
amount of work remains to be carried out to precise the molecular [36] P.J. Schmidt, P.T. Toran, A.M. Giannetti, P.J. Bjorkman, N.C. Andrews, Cell
mechanisms of hepcidin action, its synthesis, maturation, secretion Metab. 7 (2008) 205–214.
[37] G. Ramey, J.C. Deschemin, S. Vaulont, Haematologica, in press.
and transport and to understand the crosstalk between all the [38] R.E. Fleming, Cell Metab. 9 (2009) 211–212.
regulators identified by genetics. There is no doubt that hepcidin [39] G. Papanikolaou, M.E. Samuels, E.H. Ludwig, M.L. MacDonald, P.L. Franchini,
measurements will be used for the diagnosis, classification and M.P. Dube, L. Andres, J. MacFarlane, N. Sakellaropoulos, M. Politou,
E. Nemeth, J. Thompson, J.K. Risler, C. Zaborowska, R. Babakaiff,
follow-up of disorders of iron metabolism. Lastly, hepcidin and its C.C. Radomski, T.D. Pape, O. Davidas, J. Christakis, P. Brissot, G. Lockitch,
regulators constitute considerable potential therapeutic targets for T. Ganz, M.R. Hayden, Y.P. Goldberg, Nat. Genet. 36 (2004) 77–82.
treatment of iron disorders with, at the two extremes, iron overload [40] J.L. Babitt, F.W. Huang, D.M. Wrighting, Y. Xia, Y. Sidis, T.A. Samad,
J.A. Campagna, R.T. Chung, A.L. Schneyer, C.J. Woolf, N.C. Andrews, H.Y. Lin,
and anaemia of inflammation. Nat. Genet. 38 (2006) 531–539.
[41] J.L. Babitt, F.W. Huang, Y. Xia, Y. Sidis, N.C. Andrews, H.Y. Lin, J. Clin. Invest.
117 (2007) 1933–1939.
References [42] R.H. Wang, C. Li, X. Xu, Y. Zheng, C. Xiao, P. Zerfas, S. Cooperman,
M. Eckhaus, T. Rouault, L. Mishra, C.X. Deng, Cell Metab. 2 (2005) 399–409.
[1] M. Knutson, M. Wessling-Resnick, Crit. Rev. Biochem. Mol. Biol. 38 (2003) [43] J. Truksa, H. Peng, P. Lee, E. Beutler, Proc. Natl. Acad. Sci. U S A 103 (2006)
61–88. 10289–10293.
[2] A. Krause, S. Neitz, H.J. Magert, A. Schulz, W.G. Forssmann, P. Schulz- [44] L. Kautz, D. Meynard, A. Monnier, V. Darnaud, R. Bouvet, R.H. Wang, C. Deng,
Knappe, K. Adermann, FEBS Lett. 480 (2000) 147–150. S. Vaulont, J. Mosser, H. Coppin, M.P. Roth, Blood 112 (2008) 1503–1509.
[3] C.H. Park, E.V. Valore, A.J. Waring, T. Ganz, J. Biol. Chem. 276 (2001) [45] P.B. Yu, C.C. Hong, C. Sachidanandan, J.L. Babitt, D.Y. Deng, S.A. Hoyng,
7806–7810. H.Y. Lin, K.D. Bloch, R.T. Peterson, Nature Chemical Biology 4 (2008) 33–41.
[4] C. Pigeon, G. Ilyin, B. Courselaud, P. Leroyer, B. Turlin, P. Brissot, O. Loreal, [46] D. Meynard, L. Kautz, V. Darnaud, F. Canonne-Hergaux, H. Coppin, M.P. Roth,
J. Biol. Chem. 276 (2001) 7811–7819. Nat. Genet. (2009).
[5] D.M. Frazer, S.J. Wilkins, E.M. Becker, C.D. Vulpe, A.T. McKie, D. Trinder, [47] B. Andriopoulos Jr., E. Corradini, Y. Xia, S.A. Faasse, S. Chen, L. Grgurevic,
G.J. Anderson, Gastroenterology 123 (2002) 835–844. M.D. Knutson, A. Pietrangelo, S. Vukicevic, H.Y. Lin, J.L. Babitt, Nat. Genet.
[6] G. Nicolas, M. Bennoun, I. Devaux, C. Beaumont, B. Grandchamp, A. Kahn, (2009).
S. Vaulont, Proc. Natl. Acad. Sci. U.S.A. 98 (2001) 8780–8785. [48] J. Truksa, P. Lee, H. Peng, J. Flanagan, E. Beutler, Blood 110 (2007)
[7] G. Nicolas, M. Bennoun, A. Porteu, S. Mativet, C. Beaumont, B. Grandchamp, 3436–3437.
M. Sirito, M. Sawadogo, A. Kahn, S. Vaulont, Proc. Natl. Acad. Sci. U.S.A. 99 [49] G. Casanovas, K. Mleczko-Sanecka, S. Altamura, M.W. Hentze,
(2002) 4596–4601. M.U. Muckenthaler, J. Mol. Med. (2009).
[8] E. Nemeth, M.S. Tuttle, J. Powelson, M.B. Vaughn, A. Donovan, D.M. Ward, [50] M.L. Island, A.M. Jouanolle, A. Mosser, Y. Deugnier, V. David, P. Brissot,
T. Ganz, J. Kaplan, Science 306 (2004) 2090–2093. O. Loreal, Haematologica (2009).
[9] X.B. Liu, N.B. Nguyen, K.D. Marquess, F. Yang, D.J. Haile, Blood Cells Mol. Dis. [51] L. Lin, Y.P. Goldberg, T. Ganz, Blood 106 (2005) 2884–2889.
35 (2005) 47–56. [52] L. Lin, E. Nemeth, J.B. Goodnough, D.R. Thapa, V. Gabayan, T. Ganz, Blood
[10] C. Peyssonnaux, A.S. Zinkernagel, V. Datta, X. Lauth, R.S. Johnson, V. Nizet, Cells Mol. Dis. 40 (2008) 122–131.
Blood 107 (2006) 3727–3732. [53] L. Silvestri, A. Pagani, C. Fazi, G. Gerardi, S. Levi, P. Arosio, C. Camaschella,
[11] F.B. Sow, W.C. Florence, A.R. Satoskar, L.S. Schlesinger, B.S. Zwilling, Blood 109 (2007) 4503–4510.
W.P. Lafuse, J. Leukoc. Biol. 82 (2007) 934–945. [54] L. Silvestri, A. Pagani, C. Camaschella, Blood 111 (2008) 924–931.
[12] H. Kulaksiz, E. Fein, P. Redecker, W. Stremmel, G. Adler, Y. Cetin, J. Endo- [55] A.J. Ramsay, J.C. Reid, G. Velasco, J.P. Quigley, J.D. Hooper, Front. Biosci. 13
crinol. 197 (2008) 241–249. (2008) 569–579.
[13] H. Kulaksiz, F. Theilig, S. Bachmann, S.G. Gehrke, D. Rost, A. Janetzko, [56] X. Du, E. She, T. Gelbart, J. Truksa, P. Lee, Y. Xia, K. Khovananth, S. Mudd,
Y. Cetin, W. Stremmel, J. Endocrinol. 184 (2005) 361–370. N. Mann, E.M. Moresco, E. Beutler, B. Beutler, Science 320 (2008)
[14] S. Bekri, P. Gual, R. Anty, N. Luciani, M. Dahman, B. Ramesh, A. Iannelli, 1088–1092.
A. Staccini-Myx, D. Casanova, I. Ben Amor, M.C. Saint-Paul, P.M. Huet, [57] A.R. Folgueras, F.M. de Lara, A.M. Pendas, C. Garabaya, F. Rodriguez,
J.L. Sadoul, J. Gugenheim, S.K. Srai, A. Tran, Y. Le Marchand-Brustel, A. Astudillo, T. Bernal, R. Cabanillas, C. Lopez-Otin, G. Velasco, Blood 112
Gastroenterology 131 (2006) 788–796. (2008) 2539–2545.
[15] B. Gagliardo, A. Faye, M. Jaouen, J.C. Deschemin, F. Canonne-Hergaux, [58] K.E. Finberg, M.M. Heeney, D.R. Campagna, Y. Aydinok, H.A. Pearson,
S. Vaulont, M.A. Sari, FEBS J. 275 (2008) 3793–3803. K.R. Hartman, M.M. Mayo, S.M. Samuel, J.J. Strouse, K. Markianos,
[16] E.V. Valore, T. Ganz, Blood Cells Mol. Dis. 40 (2008) 132–138. N.C. Andrews, M.D. Fleming, Nat. Genet. 40 (2008) 569–571.
[17] N. Scamuffa, A. Basak, C. Lalou, A. Wargnier, J. Marcinkiewicz, G. Siegfried, [59] M.A. Melis, M. Cau, R. Congiu, G. Sole, S. Barella, A. Cao, M. Westerman,
M. Chretien, F. Calvo, N.G. Seidah, A.M. Khatib, Gut 57 (2008) 1573–1582. M. Cazzola, R. Galanello, Haematologica 93 (2008) 1473–1479.
[18] A.E. Kartikasari, R. Roelofs, R.M. Schaeps, E.H. Kemna, W.H. Peters, [60] F. Guillem, S. Lawson, C. Kannengiesser, M. Westerman, C. Beaumont,
D.W. Swinkels, H. Tjalsma, Biochim. Biophys. Acta 1784 (2008) 2029–2037. B. Grandchamp, Blood 112 (2008) 2089–2091.
[19] H. Kulaksiz, S.G. Gehrke, A. Janetzko, D. Rost, T. Bruckner, B. Kallinowski, [61] L. Silvestri, A. Pagani, A. Nai, I. De Domenico, J. Kaplan, C. Camaschella, Cell
W. Stremmel, Gut 53 (2004) 735–743. Metab. 8 (2008) 502–511.
[20] E.H. Kemna, A.E. Kartikasari, L.J. van Tits, P. Pickkers, H. Tjalsma, [62] L. Viatte, H.-J. Gröne, M.W. Hentze, B. Galy, J Mol Med ,in press.
D.W. Swinkels, Blood Cells Mol. Dis. 40 (2008) 339–346. [63] E. Kemna, P. Pickkers, E. Nemeth, H. van der Hoeven, D. Swinkels, Blood 106
[21] E. Nemeth, S. Rivera, V. Gabayan, C. Keller, S. Taudorf, B.K. Pedersen, T. Ganz, (2005) 1864–1866.
J. Clin. Invest. 113 (2004) 1271–1276. [64] E. Nemeth, E.V. Valore, M. Territo, G. Schiller, A. Lichtenstein, T. Ganz, Blood
[22] G. Nicolas, C. Chauvet, L. Viatte, J.L. Danan, X. Bigard, I. Devaux, C. Beaumont, 101 (2003) 2461–2463.
A. Kahn, S. Vaulont, J. Clin. Invest. 110 (2002) 1037–1044. [65] P. Lee, H. Peng, T. Gelbart, L. Wang, E. Beutler, Proc. Natl. Acad. Sci. U S A 102
[23] G. Nicolas, L. Viatte, M. Bennoun, C. Beaumont, A. Kahn, S. Vaulont, Blood (2005) 1906–1910.
Cells Mol. Dis. 29 (2002) 327–335. [66] J. Inamura, K. Ikuta, J. Jimbo, M. Shindo, K. Sato, Y. Torimoto, Y. Kohgo,
[24] E. Kemna, H. Tjalsma, C. Laarakkers, E. Nemeth, H. Willems, D. Swinkels, Hepatol. Res. (2005).
Blood 106 (2005) 3268–3270. [67] D.D. Harrison-Findik, E. Klein, J. Evans, J. Gollan, Am. J. Physiol. Gastrointest.
[25] S. Vaulont, D.Q. Lou, L. Viatte, A. Kahn, J. Clin. Invest. 115 (2005) 2079–2082. Liver Physiol. 296 (2009) G112–G118.
[26] J.A. Lebron, M.J. Bennett, D.E. Vaughn, A.J. Chirino, P.M. Snow, G.A. Mintier, [68] D.M. Wrighting, N.C. Andrews, Blood 108 (2006) 3204–3209.
J.N. Feder, P.J. Bjorkman, Cell 93 (1998) 111–123. [69] M.V. Verga Falzacappa, M. Vujic Spasic, R. Kessler, J. Stolte, M.W. Hentze,
[27] C.N. Roy, C.A. Enns, Blood 96 (2000) 4020–4027. M.U. Muckenthaler, Blood 109 (2007) 353–358.
[28] M. Vujic Spasic, J. Kiss, T. Herrmann, R. Kessler, J. Stolte, B. Galy, B. Rathkolb, [70] M.V. Verga Falzacappa, G. Casanovas, M.W. Hentze, M.U. Muckenthaler,
E. Wolf, W. Stremmel, M.W. Hentze, M.U. Muckenthaler, Blood 109 (2007) J. Mol. Med. 86 (2008) 531–540.
4511–4517. [71] M. Pak, M.A. Lopez, V. Gabayan, T. Ganz, S. Rivera, Blood 108 (2006)
[29] M. Vujic Spasic, J. Kiss, T. Herrmann, B. Galy, S. Martinache, J. Stolte, 3730–3735.
H.J. Grone, W. Stremmel, M.W. Hentze, M.U. Muckenthaler, Cell Metab. 7 [72] T. Tanno, N.V. Bhanu, P.A. Oneal, S.H. Goh, P. Staker, Y.T. Lee, J.W. Moroney,
(2008) 173–178. C.H. Reed, N.L. Luban, R.H. Wang, T.E. Eling, R. Childs, T. Ganz, S.F. Leitman,
[30] T. Goswami, N.C. Andrews, J. Biol. Chem. 281 (2006) 28494–28498. S. Fucharoen, J.L. Miller, Nature Medicine 13 (2007) 1096–1101.
[31] H. Kawabata, R. Yang, T. Hirama, P.T. Vuong, S. Kawano, A.F. Gombart, [73] S. Lakhal, N.P. Talbot, A. Crosby, C. Stoepker, A.R. Townsend, P.A. Robbins,
H.P. Koeffler, J. Biol. Chem. 274 (1999) 20826–20832. C.W. Pugh, P.J. Ratcliffe, D.R. Mole, Blood 113 (2009) 1555–1563.
[32] A. Robb, M. Wessling-Resnick, Blood 104 (2004) 4294–4299. [74] J.P. Pinto, S. Ribeiro, H. Pontes, S. Thowfeequ, D. Tosh, F. Carvalho, G. Porto,
[33] M.B. Johnson, C.A. Enns, Blood 104 (2004) 4287–4293. Blood 111 (2008) 5727–5733.
1228 L. Viatte, S. Vaulont / Biochimie 91 (2009) 1223–1228

[75] H. Huang, M. Constante, A. Layoun, M.M. Santos, Blood (2009). [92] K.R. Bridle, D.M. Frazer, S.J. Wilkins, J.L. Dixon, D.M. Purdie, D.H. Crawford,
[76] C. Peyssonnaux, A.S. Zinkernagel, R.A. Schuepbach, E. Rankin, S. Vaulont, V.N. Subramaniam, L.W. Powell, G.J. Anderson, G.A. Ramm, Lancet 361
V.H. Haase, V. Nizet, R.S. Johnson, J. Clin. Invest. 117 (2007) 1926–1932. (2003) 669–673.
[77] G.L. Semenza, Curr. Opin. Cell Biol. 13 (2001) 167–171. [93] H. Kawabata, R.E. Fleming, D. Gui, S.Y. Moon, T. Saitoh, J. O’Kelly, Y. Umehara,
[78] S.O. Choi, Y.S. Cho, H.L. Kim, J.W. Park, Biochem. Biophys. Res. Commun. 356 Y. Wano, J.W. Said, H.P. Koeffler, Blood 105 (2005) 376–381.
(2007) 312–317. [94] G. Porto, A. Roetto, F. Daraio, J.P. Pinto, S. Almeida, C. Bacelar, E. Nemeth,
[79] P.N. Paradkar, I. De Domenico, N. Durchfort, I. Zohn, J. Kaplan, D.M. Ward, T. Ganz, C. Camaschella, Blood 106 (2005) 2922–2923.
Blood 112 (2008) 866–874. [95] G. Papanikolaou, M. Tzilianos, J.I. Christakis, D. Bogdanos, K. Tsimirika,
[80] E. Nemeth, G.C. Preza, C.L. Jung, J. Kaplan, A.J. Waring, T. Ganz, Blood 107 J. MacFarlane, Y.P. Goldberg, N. Sakellaropoulos, T. Ganz, E. Nemeth, Blood
(2006) 328–333. 105 (2005) 4103–4105.
[81] H. Drakesmith, L.M. Schimanski, E. Ormerod, A.T. Merryweather-Clarke, [96] A.T. Merryweather-Clarke, E. Cadet, A. Bomford, D. Capron, V. Viprakasit,
V. Viprakasit, J.P. Edwards, E. Sweetland, J.M. Bastin, D. Cowley, A. Miller, P.J. McHugh, R.W. Chapman, J.J. Pointon, V.L. Wimhurst,
Y. Chinthammitr, K.J. Robson, A.R. Townsend, Blood 106 (2005) 1092–1097. K.J. Livesey, V. Tanphaichitr, J. Rochette, K.J. Robson, Hum. Mol. Genet. 12
[82] M.D. Knutson, M. Oukka, L.M. Koss, F. Aydemir, M. Wessling-Resnick, Proc. (2003) 2241–2247.
Natl. Acad. Sci. U S A 102 (2005) 1324–1328. [97] S. Jacolot, G. Le Gac, V. Scotet, I. Quere, C. Mura, C. Ferec, Blood 103 (2004)
[83] C. Delaby, N. Pilard, A.S. Goncalves, C. Beaumont, F. Canonne-Hergaux, 2835–2840.
Blood 106 (2005) 3979–3984. [98] V. Bach, A. Altés, M.J. Barcelo, A. Cortés, P. Sarda, C. Canals, J. Felez, M. Baiget,
[84] L. Viatte, J.C. Lesbordes-Brion, D.Q. Lou, M. Bennoun, G. Nicolas, A. Kahn, A. Remacha, European Iron Club, Rennes, France, 2004.
F. Canonne-Hergaux, S. Vaulont, Blood 105 (2005) 4861–4864. [99] G. Nicolas, L. Viatte, D.Q. Lou, M. Bennoun, C. Beaumont, A. Kahn,
[85] I. De Domenico, E. Nemeth, J.M. Nelson, J.D. Phillips, R.S. Ajioka, N.C. Andrews, S. Vaulont, Nat. Genet. 34 (2003) 97–101.
M.S. Kay, J.P. Kushner, T. Ganz, D.M. Ward, J. Kaplan, Cell Metab. 8 [100] L. Viatte, G. Nicolas, D.Q. Lou, M. Bennoun, J.C. Lesbordes-Brion, F. Canonne-
(2008) 146–156. Hergaux, K. Schonig, H. Bujard, A. Kahn, N.C. Andrews, S. Vaulont, Blood 107
[86] I. De Domenico, D.M. Ward, C. Langelier, M.B. Vaughn, E. Nemeth, (2006) 2952–2958.
W.I. Sundquist, T. Ganz, G. Musci, J. Kaplan, Mol. Biol. Cell 18 (2007) [101] D.F. Wallace, V.N. Subramaniam, World J. Gastroenterol. 13 (2007)
2569–2578. 4690–4698.
[87] I. De Domenico, E. Lo, D.M. Ward, J. Kaplan, Proc. Natl. Acad. Sci. U S A 106 [102] G. Weiss, L.T. Goodnough, N. Engl. J. Med. 352 (2005) 1011–1023.
(2009) 3800–3805. [103] T. Ganz, G. Olbina, D. Girelli, E. Nemeth, M. Westerman, Blood 112 (2008)
[88] A.E. Rice, M.J. Mendez, C.A. Hokanson, D.C. Rees, P.J. Bjorkman, J. Mol. Biol. 4292–4297.
386 (2009) 717–732. [104] D.W. Swinkels, D. Girelli, C. Laarakkers, J. Kroot, N. Campostrini, E.H. Kemna,
[89] J.N. Feder, A. Gnirke, W. Thomas, Z. Tsuchihashi, D.A. Ruddy, A. Basava, H. Tjalsma, PLoS ONE 3 (2008) e2706.
F. Dormishian, R. Domingo Jr., M.C. Ellis, A. Fullan, L.M. Hinton, N.L. Jones, [105] D.G. Ward, K. Roberts, P. Stonelake, P. Goon, C.G. Zampronio, A. Martin,
B.E. Kimmel, G.S. Kronmal, P. Lauer, V.K. Lee, D.B. Loeb, F.A. Mapa, P.J. Johnson, T. Iqbal, C. Tselepis, Proteome Sci. 6 (2008) 28.
E. McClelland, N.C. Meyer, G.A. Mintier, N. Moeller, T. Moore, E. Morikang, [106] U. Kobold, T. Dulffer, M. Dangl, A. Escherich, M. Kubbies, R. Roddiger,
R.K. Wolff, et al., Nat. Genet. 13 (1996) 399–408. J.A. Wright, Clin. Chem. 54 (2008) 1584–1586.
[90] A. Pietrangelo, N. Engl. J. Med. 350 (2004) 2383–2397. [107] S.S. Bansal, J.M. Halket, A. Bomford, R.J. Simpson, N. Vasavda, S.L. Thein,
[91] M. Muckenthaler, C.N. Roy, A.O. Custodio, B. Minana, J. deGraaf, R.C. Hider, Anal. Biochem. 384 (2009) 245–253.
L.K. Montross, N.C. Andrews, M.W. Hentze, Nat. Genet. 34 (2003) [108] A. Piperno, R. Mariani, P. Trombini, D. Girelli, World J. Gastroenterol. 15
102–107. (2009) 538–551.

You might also like