Está en la página 1de 7

Med Clin (Barc).

2017;148(5):218–224

www.elsevier.es/medicinaclinica

Review

Current status of iron metabolism: Clinical and therapeutic


implications夽
Susana Conde Diez a , Ricardo de las Cuevas Allende b , Eulogio Conde García c,∗
a
Medicina de Familia, Servicio Cántabro de Salud, Santander (Cantabria), Spain
b
Medicina de Familia, Servicio Cántabro de Salud, Centro de Salud Bajo Asón, Ampuero (Cantabria), Spain
c
Servicio de Hematología, Hospital Universitario Marqués de Valdecilla, Universidad de Cantabria, Santander (Cantabria), Spain

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

Article history: Hepcidin is the main regulator of iron metabolism and a pathogenic factor in iron disorders. Hepcidin
Received 22 September 2016 deficiency causes iron overload, whereas hepcidin excess causes or contributes to the development of
Accepted 26 October 2016 iron-restricted anaemia in chronic inflammatory diseases. We know the mechanisms involved in the
Available online 23 March 2017
synthesis of hepcidin and, under physiological conditions, there is a balance between activating signals
and inhibitory signals that regulate its synthesis. The former include those related to plasmatic iron level
Keywords: and also those related to chronic inflammatory diseases. The most important inhibitory signals are related
Iron metabolism
to active erythropoiesis and to matriptase-2. Knowing how hepcidin is synthesized has helped design
Hepcidin
Ferroportin
new pharmacological treatments whose main target is the hepcidin. In the near future, there will be
Hepcidin agonists effective treatments aimed at correcting the defect of many of these iron metabolism disorders.
Hepcidin antagonists © 2016 Elsevier España, S.L.U. All rights reserved.

Estado actual del metabolismo del hierro: implicaciones clínicas y terapéuticas

r e s u m e n

Palabras clave: La hepcidina es el principal regulador del metabolismo del hierro y el factor patogénico más importante en
Metabolismo del hierro sus trastornos. La deficiencia de hepcidina provoca sobrecarga de hierro, mientras que su exceso da lugar
Hepcidina o contribuye al desarrollo de anemias por déficit o restricción de hierro en las enfermedades crónicas.
Ferroportina
Conocemos los mecanismos implicados en la síntesis de hepcidina y, en condiciones fisiológicas, hay un
Agonistas de la hepcidina
equilibrio entre las señales activadoras e inhibidoras que regulan su síntesis. Las primeras incluyen las
Antagonistas de la hepcidina
relacionadas con la concentración plasmática de hierro y con las enfermedades inflamatorias. Las señales
inhibidoras más importantes están relacionadas con la eritropoyesis activa y con la matriptasa-2. Conocer
cómo se sintetiza la hepcidina ha servido para diseñar nuevos tratamientos farmacológicos cuya diana
principal es la hepcidina. En un futuro próximo, se dispondrá de tratamientos eficaces dirigidos a corregir
el defecto de muchos de los trastornos del metabolismo del hierro.
© 2016 Elsevier España, S.L.U. Todos los derechos reservados.

Introduction cellular immune response.1 In the adult, the total amount of iron in
the body is 3–4 g, of which 65% is in haemoglobin, 25% in deposit
Iron is an essential nutrient in the body which plays a central organs (liver, reticuloendothelial system macrophages and bone
role in cellular energy metabolism, anaerobic respiration, synthe- marrow) and the remaining 10% in myoglobin, cytochromes, per-
sis of haemoglobin and nucleotide synthesis, additionally, it is also oxidase and catalases.2 Every day, 1–2 mg/d of iron is absorbed
involved in many other processes of exudative metabolism and from the diet, which is the same amount lost daily, but it should
be noted that the organism does not have an active iron excretion
mechanism, so that control of the duodenal absorption plays a vital
role in iron homeostasis.3 Plasma iron circulates bound to trans-
夽 Please cite this article as: Conde Diez S, de las Cuevas Allende R, Conde García
ferrin and it comes from the iron which is absorbed and the iron
E. Estado actual del metabolismo del hierro: implicaciones clínicas y terapéuticas.
that comes from deposit organs, which release it into the plasma
Med Clin (Barc). 2017;148:218–224.
∗ Corresponding author. through ferroportin.1–3 Their accumulation leads to iron overload
E-mail addresses: euconde@humv.es, condee@unican.es (E. Conde García). that is toxic and can damage tissues and cause cell death by free

2387-0206/© 2016 Elsevier España, S.L.U. All rights reserved.


S. Conde Diez et al. / Med Clin (Barc). 2017;148(5):218–224 219

Hepcidin macrophages, which contain the iron coming from recycled senes-
cent red blood cells, and that released from the deposits (Fig. 2).
1-2 mg/d Ferroportin is responsible for the efflux of iron from enterocytes,
FPN Macrophage
Enterocyte FPN macrophages and hepatocytes into the plasma. Hepcidin binds
to ferroportin for its destruction by endocytosis into the cell’s
lysosomes resulting, on the one hand, in a hyposideremia by low-
ering the iron transferred to the plasma and, on the other hand,
to the accumulation of iron as ferritin enterocytes, macrophages
Liver FPN Plasma 20-30 RBCs
1000mg 3-4mg 2500 mg and hepatocytes.1–3 The control of iron homeostasis by hepcidin
mg/d
is a classic endocrine regulation system; in the words of Ganz,
the relationship of hepcidin with iron is similar to that of insulin
with glucose.4 Hepcidin is therefore the principal regulator of iron
and plays a key role in all its abnormalities, whether having to do
Fe losses
BM with deficiency or excess. Hepcidin deficiency causes iron over-
1-2 mg/d
load, while its excess favours iron sequestration in the liver and
Fig. 1. Body iron homeostasis. The interaction of hepcidin with ferroportin (FPN) macrophages and contributes to the development of iron deficiency
controls the main efflux of iron into plasma. anaemias or because of its misuse in anaemia of chronic disease.5
In these cases, a functional iron deficiency anaemia occurs because
iron reserves are not available for erythropoiesis.5,6 Increased
radical formation and lipid peroxidation. For this reason, the circu-
hepcidin also occurs in chronic inflammatory processes by an
lating iron is never found in free form, it is always attached to other
increase in IL6, which also represents a host defence mechanism
molecules, mainly transferrin, but when concentrations of plasma
against infection by limiting the availability of extracellular iron
iron are high and transferrin is saturated, the excess iron is bound
to microorganisms.5 Hepcidin production is negatively regulated
to other plasma molecules of low molecular weight such as citrate,
by erythropoiesis through mediators that prevent its production
acetate and albumin.2
when iron is required for haemoglobin synthesis.5 Hepcidin is an
The iron in the body follows a cycle consisting of duodenal
acute phase reactant that responds to a variety of inflammatory
absorption, distribution through the plasma bound to transferrin
mediators and signals that activate transcription through different
and transfer to cells via the transferrin receptor, located in the cyto-
signalling pathways.5
plasmic membrane, for use in different metabolic processes or for
storing it in deposit organs.1–3 When red blood cells age, they are
destroyed in macrophages, mainly in the spleen, and iron is reused
Regulation of hepcidin expression
after passing through the plasma (Fig. 1). Foods containing ferric
iron which, after being reduced to the ferrous form, it is absorbed
Hepcidin, encoded by the gene HAMP, is the hormone that reg-
by duodenal enterocytes and subsequently released into plasma
ulates iron metabolism. It is a 25-amino acid peptide produced by
through ferroportin (Fig. 2).
hepatocytes interacting with the ferroportin found in the cell mem-
brane of enterocytes, macrophages and hepatocytes.7 Regulation
Regulation of iron homeostasis. Hepcidin-ferroportin axis of hepcidin is a multifactorial process involving different stimu-
latory and inhibitory signals which, in various ways, control its
Hepcidin is the main hormone that regulates iron metabolism. final transcript.7,8 Hepcidin is regulated by plasma iron through
It is synthesized in the liver and its main mission is to con- a feedback mechanism which involves intra and extracellular iron
trol the arrival of iron in plasma from food through enterocytes, sensors coupled to one or more signal transduction pathways.

Enterocyte BM: Erythropoiesis


Ferritin
Fe3+
DCYTB Hephaestin
Fe2+
FPN
DMT1

Fer
ritin

Hepcidin
FPN

FPN

Liver Vein Macrophage


DCYTB: ferrireductase; DMT1: Divalent Iron Metal Transporter 1; ferroxidase: hephaestin
Free transferrin and iron bound ; Ferritin ; Hepcidin ; Ferroportin FPN

Fig. 2. Dietary iron is absorbed by duodenal enterocytes. Nonheme iron occurs primarily in the ferric state (Fe 3+) in the intestine and is reduced to ferrous iron (Fe 2+) by the
action of ferrireductases, especially duodenal cytochrome b (Dcytb). Ferrous iron passes through the duodenal enterocytes of the divalent metal transporter-1 (DMT1). Once
in the enterocyte, the ferrous iron can be stored in it as ferritin or can be released into the bloodstream through ferroportin (FPN). Ferrous iron is oxidized by a ferroxidase
identified as hephaestin, which after converting into ferric iron, it binds to transferrin and thus circulates in plasma. Furthermore, senescent erythrocytes are phagocytosed
by macrophages, primarily in the spleen, but also in the liver and the bone marrow (BM). During erythropoiesis, erythroblasts acquire iron for haemoglobin synthesis from
transferrin via transferrin receptors. Excess iron is stored in the liver and in macrophages as ferritin, which is oxidized to hemosiderin. Hepcidin plays a key role in the release
of iron from the deposits depending on the requirements (e.g.: increased erythropoiesis, etc.).
220 S. Conde Diez et al. / Med Clin (Barc). 2017;148(5):218–224

tion Iron activation Inflammation in the presence of neoginin, promoting SMAD1/5/8 phosphoryla-
nhibi
sis i act tion, HAMP transcription and finally hepcidin synthesis12,13 (Fig. 3).
poie Tf - Fe2 iva
ro ti Another important mediator in hepcidin synthesis is the inflamma-
Eryth

on
Tf - Fe2
Tf - Fe2 BMP6 tion through IL6 and other cytokines (IL1, IL22) and activin B (Act-B)
which activate the JAK-STAT3 and BMP signalling pathway7,9

B
MT - 2

Act -
TfR2 B
e TfR1 B IL6

HFE
ErF M HJV
M
P
R
P (Fig. 3).
NEO R
1
2

Inhibitory signals

SMAD
1/5/8 The main hepcidin inhibitory signal comes from erythropoiesis
and is related to different proteins such as erythroferrone, growth
differentiation factor 15 (GDF15) and twisted gastrulation BMP
EPO P SMAD
JAK - STAT3 signalling modulator (TWSG1) that inhibits the SMAD pathway7
Hypoxia 1/5/8
(Fig. 3). Matriptase-2 also has an inhibiting effect on hepcidin,
encoded by the TMPRSS6 gene, as it blocks HJV and prevents BMP
complex activation.10 Other inhibitory signals of hepcidin synthe-
sis are tissue hypoxia and erythropoiesis stimulating factors such
as erythropoietin (EPO).1,2
SMAD4
STAT3 Hepcidin Disorders associated with hepcidin abnormalities
HAMP

Ferroportin and hepcidin abnormalities, whether by interaction


Fig. 3. Hepcidin synthesis. The main axis that controls the synthesis of hepcidin or aberrant expression, cause or contribute to triggering a large
is bone morphogenetic protein-hemojuvelin-sons of mothers against decapentaplegic number of iron related disorders ranging from anaemia due to iron
(BMP-HJV-SMAD). Transferrin bound to iron (Fe2-Tf) is a hepatocyte sensor for deficiency to iron overload diseases.14,15
hepcidin transcription control. The binding of Fe2-Tf with transferrin receptors
(TfR1 and TfR2) and human hemochromatosis protein (HFE) is inversely corre-
lated with levels of transferrin saturation by iron, so that, if levels increase, HFE Diseases associated with an excess of hepcidin
is displaced from TfR1 to TfR2 to activate BMP. This activation facilitates the bind-
ing of BMP6 with its receptors 1 and 2 (BMPR1 and BMPR2) in the presence of
A high level of hepcidin generates hyposideremia with the
hemojuvelin (HJV) and neoginin (NEO), and launches SMAD 1/5/8 phosphorylation,
hepcidin antimicrobial peptide (HAMP) gene transcription and the synthesis of hep-
consequent reduction in erythropoiesis due to insufficient sup-
cidin. Inflammation is another important mediator in hepcidin synthesis through IL6 ply of iron, which leads to the development of anaemia in chronic
and activin B (Act-B) via janus kinase-signal transducer and activator of transcription diseases.15,16
(JAK-STAT3) and BMP. The main hepcidin inhibitory signal comes from the bone Chronic diseases present with moderate normocytic and nor-
marrow during active erythropoiesis and takes place through different proteins,
mochromic anaemia but sometimes it may be microcytic and
highlighting erythroferrone (ErFe), growth differentiation factor (GDF15) and twisted
gastrulation BMP signalling modulator (TWSG1), which inhibit the SMAD pathway. hypochromic. Anaemia is not only the result of elevated hepcidin,
Matriptase-2 (MT-2), encoded by the gene TMPRSS6, also has an inhibitory effect on there is also a direct effect of cytokines on the production and
hepcidin transcription through HJV cleavage, preventing BMP complex activation. half-life of red blood cells.5 In chronic kidney disease (CKD), an
Other inhibitory signals of hepcidin synthesis come from situations that generate elevated hepcidin is the result of a combination of inflammation
tissue hypoxia and from the administration of erythropoiesis stimulating factors
such as erythropoietin (EPO).
and inadequate kidney clearance.10,17 In neoplasms, anaemia is
associated with disease and treatments, but high levels of hep-
cidin and cytokines also play an important role.2 In addition, 10%
Iron-bound transferrin (Fe2-Tf) (holotransferrin) is a hepatocyte of the elderly develop anaemia due to nutritional defects, bleeding,
sensor converging in a complex heterotetrameric signalling asso- chronic inflammation, renal failure, MDS and increased hepcidin.1,2
ciated with the hepatocyte membrane made up of transferrin Finally, iron-refractory iron deficiency anaemia must be included in
receptors (TfR1 and TfR2), human hemochromatosis protein (HFE), this section, a genetic anaemia characterized by a mutation in the
BMP ligands (bone morphogenetic protein), 2 kinase receptors of TMPRR6 gene encoding matriptase-2.10 The genetic defect is auto-
BMP (BMPR1 and BMPR2), a BMP coreceptor (hemojuvelin [HJV]) somal recessive and occurs with elevated hepcidin levels due to the
and a facilitator (neoginin).7–10 There is also an increase in hepcidin absence of matriptase-2, resulting in an accumulation of HJV and
in IL-6-mediated inflammatory processes. Conversely, an effective activation of BMP pathway.10
erythropoiesis in bone marrow decreases hepcidin levels, ensuring
the supply of iron for erythrocyte production. Therefore, hepcidin Diseases associated with hepcidin decrease or resistance
levels reflect the integration of multiple activating and inhibitory
signals involved in iron regulation (Fig. 3). A hepcidin production deficiency results in iron overload which
is usually genetic and is due to mutations in the HFE or TfR2
Activating signals gene (adult hemochromatosis) or the HJV or HAMP genes (juve-
nile hemochromatosis).18,19 This decrease of hepcidin contributes
BMP-HJV-SMAD is the main axis controlling hepcidin synthe- to the normal operation of ferroportin, which results in enterocyte
sis. In situations of iron deficiency, the Fe2-Tf complex binds iron absorption and increased release of iron from macrophages,
to the TfR1 receptor, which is not capable of activating the which causes hypersideremia with subsequent increase in trans-
BMP-SMAD pathway and therefore hepcidin is not synthesized, ferrin saturation, and the appearance of iron bound to proteins
something which facilitates the arrival of iron into plasma. When other than transferrin, called non-transferrin-bound iron (NTBI).7,15
iron deficiency is corrected, holotransferrin joins the TfR2 recep- As there is no regulatory mechanism for excreting iron in humans,
tor, forming a complex with HFE1,7–11 (Fig. 3). The role of HFE excess iron is deposited in tissues that express carriers for NTBI,
is to promote the association and stabilization with TfR2, acti- mainly the liver, heart, pancreas and other endocrine glands, caus-
vate HJV and BMP to bind to its receptors (BMPR1 and BMPR2), ing function failure in the affected organ.
S. Conde Diez et al. / Med Clin (Barc). 2017;148(5):218–224 221

Hemochromatosis is the most common cause of genetic iron hepcidin, without distinguishing between the complete hepcidin
overload. There are many mutations that lead to the clinical syn- (hepcidin-25) and the smaller isoforms (hepcidin-20, -22, -23 and
drome previously mentioned, but the most common is inherited as -24), besides, its concentrations are usually higher in CKD due to
an autosomal recessive trait, affecting the HFE gene, which encodes a dysfunction in hepcidin clearance.24 Urinary hepcidin measure-
the HFE protein (Type 1 Hemochromatosis). The most common sub- ment correlates with plasma hepcidin, but urinary detection may
type affects homozygous patients with Cys282Tyr mutation (type be distorted by the high concentration of small isoforms and in
1A). Heterozygous patients may have mixed Cys282Tyr/His63Asp CKD.24
mutations (type 1B). There are also cases that have mutations in
genotypes other than HFE, for example, Ser65Cys (Type 1C).13,18,19
Hepcidin as a therapeutic target
More penetrating and severe forms of hereditary hemochromato-
sis are rare and are caused by mutations in genes HJV (type 2A),
Hepcidin is the main target of diseases related to iron
hepcidin (type 2B), or TfR2 (type 3).9,18 Mutations in the gene that
metabolism disorders and, therefore, new hepcidin agonists or
controls TfR2 prevent this from binding to HFE and consequently,
antagonist drug therapies are being researched.10,11 The interest of
the BMP-HJV-SMAD complex is not activated and hepcidin is not
these studies is huge, as evidenced by the fact that in July 2016 there
synthesized.18 Another rare form of hemochromatosis is due to
were 120 registered clinical trials of hepcidin in ClinicalTrials.gov.
mutations in the ferroportin gene (type 4). These mutations, C326S
and SLC11A3, lead to a hepcidin-resistant ferroportin, so that the
iron is exported to the circulation, even in the presence of ele- Hepcidin agonists
vated levels of hepcidin.18,20 Phlebotomy is the treatment of choice
in iron overload and it is estimated that 1 mg of iron is lost with Increase levels of circulating hepcidin may be beneficial in
each millilitre of red cells removed and, therefore, the mobilization patients with processes that occur with iron overload, such as
of excess iron accumulated in the tissues is promoted to restore hemochromatosis and thalassemia. This could be achieved with
erythropoiesis.2 drugs that have a hepcidin-like activity or stimulating its endoge-
ˇ-Thalassemia is another disease that occurs with anaemia, nous production, but the design of a hepcidin similar to that of
iron overload and low levels of hepcidin. Iron overload is the oneself does not seem to be the solution, because its half-life is
leading cause of morbidity and mortality in these patients, very short and because of the costs involved in its production.10
both in non-transfusion-dependent thalassemia as well as in
the dependent one. Defective ␤-globin chain production during Minihepcidins
erythropoiesis results in the precipitation of ␣-chains produced They have a hepcidin-like action with a better bioavailability
in excess and many erythroid precursors may die early, which and a longer circulating half-life. Their action is exerted by bind-
results in an ineffective erythropoiesis.21,22 To minimize anaemia, ing to ferroportin and blocking the efflux of iron from enterocytes,
a series of compensatory mechanisms are activated, such as an macrophages and hepatocytes, leading to decreased plasma iron
increase in erythropoietin which causes a erythroblastic hyperpla- levels.11,25–27 Its efficacy has been demonstrated in knockout HAMP
sia, extramedullary haematopoiesis, splenomegaly and increased −/− mice, a model of severe hemochromatosis with lack of hep-
intestinal absorption of iron which, in the absence of transfusion, cidin, in which a plasma iron reduction and tissue iron overload
is the cause of iron overload.23 The decrease in hepcidin levels normalization is achieved.27 In thalassaemic mice, treatment with
is the result of increased suppressor factors, erythroferrone and minihepcidins decreases iron overload and improves all haemato-
GDF15 during the development of erythroblasts.8 Transfusions par- logical parameters.25–27 This is the result of iron restriction, leading
tially correct anaemia and hepcidin suppression, but provide very to a decrease in aggregate formation and intracellular inclusion
high amounts of iron with the RBCs transfused. Iron overload in bodies and prevents oxidative stress in erythrocytes, damage in
␤-thalassemia is treated with iron chelators. DNA, organelle and cell membrane, with improved erythropoiesis
and anaemia correction.2,20

Hepcidin in the diagnosis of iron abnormalities


Hepcidin endogenous production stimulators
Another way to increase hepcidin production is based on the
If hepcidin plays a central role in the pathogenesis of many iron
design of TMPRSS6 antagonists. RNA technologies are used, mainly
disorders, it seems logical that its quantification should become a
antisense oligonucleotides that could silence the mRNA of TMPRSS6
useful tool for the diagnosis and clinical management of associated
thereby stabilizing HJV, stimulating BMP-HJV-SMAD pathway and
diseases. The size of hepcidin in its circulating bioactive form is 25
increasing the synthesis of endogenous hepcidin.11,28 These drugs
amino acids and the N-terminal degradation or storage of samples
have also been effective in treating iron overload in knockout mice
at room temperature results in smaller isoforms (hepcidin-24, -23,
and in the treatment of anaemia and iron overload in thalassemia
-22 and -20 amino acids) whose meaning is not known.1 Circulating
mouse model (th3/+).29 In addition to increasing the levels of
hepcidin binds to one alpha-2-macroglobulin and to albumin, it is
hepcidin, they improve iron overload, reduce splenomegaly and,
excreted by the kidney and reabsorbed in the proximal tubules.24
surprisingly, ineffective erythropoiesis is corrected in thalassaemic
The development of tests to quantify hepcidin levels in biologi-
mice with homozygous inactivation of TMPRSS6.29,30
cal samples is a not yet resolved challenge. Absolute hepcidin levels
differ widely, up to 10 times in the general population and between
the different clinical trials, so each laboratory should establish its Hepcidin antagonists
own reference values. These differences may be due to the fact
that hepcidin is influenced by many physiological and patholog- The high concentration of hepcidin causes iron entrapment
ical stimuli that promote or inhibit its synthesis and because it in the cells of the reticuloendothelial system and is the leading
adheres to plastics.24 Hepcidin determination can be performed cause of effective iron deficiency in anaemia of chronic disease,
on serum, plasma or urine by immunoassay or by mass spectrom- therefore, pharmacological suppression of hepcidin would facil-
etry. The quantification of hepcidin by immunoassay may be more itate iron mobilization, promoting erythropoiesis and anaemia
appropriate for large-scale studies due to its high performance correction. The strategies that are being considered for this pur-
and relatively low cost. However, immunoassays quantify the total pose are directed to change the BMP-SMAD or IL6/STAT3 axes,
222 S. Conde Diez et al. / Med Clin (Barc). 2017;148(5):218–224

Table 1
Hepcidin inhibitors and their corresponding targets.

Inhibitor Action and target References

BMPs/BMPR/HJV complex
• Anti-HJV MAb (ABT-207, h5F9-AM8) Inhibitor of BMPs/SMAD pathway 33,34

• sHJV-Fc Inhibitor of BMPs/SMAD pathway 11,35

• LDN-193189 BMPR1 phosphorylation inhibitor 36

• Anti-BPM6 MAb BMP6 sequestration 37

• siHep, siHJV, siTfR2 Hepcidin mRNA degradation, HJV or TfR2 36,38

• Modified heparin Inhibitors of BMPs/SMAD pathway 40,41

IL6/SATAT3 pathway
• Anti-IL6 (xiltuximab) IL6 sequestration 42

• Anti-IL6R (tocilizumab) IL6 receptor sequestration 43

• AG490 STAT3 phosphorylation inhibitor 44

Antihepcidin agents
• Antihepcidin MAb (12B9m) Hepcidin protein sequestration 48

• Anticalines (PRS-080) Hepcidin protein sequestration 49

• Aptamers (Spiegelmers: NOX-H94) Hepcidin protein sequestration 50

Hepcidin-ferroportin interaction
• Antiferroportin MAb (LY2928057) Hepcidin-ferroportin binding interference 51

• Fursultiamine Cys326-HS sequestration (FPN-hepcidin binding) 52

Other inhibitors
• TNF-␣ MAb (infliximab, golimumab) Indirect effect of IL6 suppression 53,54

• Vitamin D SMAD 1/5/8 phosphorylation suppressed. Vitamin D recep 55

• 17-Estradiol SMAD 1/5/8 phosphorylation suppressed 56

• Testosterone SMAD 1/5/8 phosphorylation suppressed 57

• Chinese medicinal plant extract: Caulis spatholobi (Jixueteng) SMAD 1/5/8 phosphorylation suppressed 58

MAb: monoclonal antibody; MRNA: messenger ribonucleic acid; BMP: bone morphogenetic protein; BMPR: bone morphogenetic protein receptor; FPN: ferroportin; HJV:
hemojuvelin; sHJV-Fc: fusion protein between the soluble HJV and the Fc fragment of immunoglobulins; IL: interleukin; SMAD: sons of mothers against decapentaplegic;
STAT3: signal transducer and activator of transcription 3; TfR2: transferrin receptor 2; TNF: tumour necrosis factor.
siHep, siHJV, siTfR2: oligonucleotides silencing mRNA expression in genes encoding hepcidin, HJV or TfR2.

interfere in hepcidin-ferroportin binding and enhance their sup- modified heparin analogues have been designed to minimize its
pressive mechanisms10,11,16,31,32 (Table 1). anticoagulant effect, maintaining their inhibitory effect on hepcidin
expression to treat anaemia of inflammatory processes.40,41

Inhibition of bone morphogenetic protein pathway Suppression of the inflammatory pathway


The BMP-HJV-SMAD pathway is the main axis which con- Blocking IL6 with xiltuximab42 and IL6 receptor with
trols the synthesis of hepcidin and, therefore, inhibiting the BMP tocilizumab43 prevent STAT3 phosphorylation and hepcidin con-
pathway would decrease HAMP expression and blood concentra- centration decrease and blood iron (level) normalization in
tions of hepcidin. Various molecules have been directed against monkeys with arthritis and in patients with Castleman’s disease.
HJV as a target. On the one hand, several monoclonal antibodies Similar effects were achieved with a chemical inhibitor of JAK2,
(mAbs) targeting the HJV (ABT-207, h5F9-AM8 and h5F9-23) pro- the AG490, blocking the IL6-STAT3 pathway.44 The main drawback
tein that prevent the binding of BMP with BMPR, block the SMAD of anti-cytokine treatments is that they induce immunosuppres-
pathway and correct anaemia in rats with chronic inflammatory sion, with impairment of the host defences and an increased risk of
processes33,34 have been investigated. A similar effect is achieved infections.11
with a fusion protein between the soluble HJV (sHJV) and the Fc
fragment of immunoglobulins (sHJV-Fc) that binds to BMP and pre- Potentiation of erythropoiesis
vents BMPR activation.11,35 The same applies to a BMP receptor 1 Increased erythropoiesis is accompanied by a suppression of
(BMPR1) inhibitor, the LDN-193189, in rats and human hepatoma hepcidin, which could represent a new strategy for the treatment of
cells36 and with an anti-BPM6 MAb.37 Another way of inhibiting anaemia of chronic disease. High doses of EPO may be able to over-
the BMP pathway is blocking or silencing the genes regulating hep- come the observed resistance to EPO in these diseases, probably
cidin synthesis with antisense oligonucleotides that interfere with by partial suppression of hepcidin.45 Similarly, prolylhydroxylase
RNA and silence the mRNA (siRNA) of hepcidin-encoding genes, inhibitors, which stabilize the factors, induce hypoxia and increase
TfR2 and HJV.36,38 The administration of these siRNAs decreases EPO synthesis, decrease hepcidin levels and increase haemoglobin
hepcidin concentrations and corrects anaemia in a chronic inflam- in patients with CKD.46
matory anaemia mouse model.16,39 Furthermore, it is known that
human mutations in the genes encoding hepcidin, the HJV and TfR2, Neutralization of circulating hepcidin
are associated with iron overload diseases, so in these cases, the The bioactive neutralization of hepcidin may be achieved by
effects on the corresponding targets of these oligonucleotides and direct binding or sequestration of the circulating hormone with
siRNAs may not be effective.10 MAb, anticalines or L-RNA aptamers (called Spiegelmers, in Ger-
Heparin is another known hepcidin inhibitor through BMP6 man, ‘mirror’). The main advantage of these hepcidin neutralizers
sequestration and the subsequent blocking of the SMAD pathway, is their high specificity and picomolar affinity to bind to their tar-
but its anticoagulant activity limits the therapeutic use as an hep- gets, but the main challenge they must overcome to demonstrate
cidin inhibitor. This action of heparin has been demonstrated in their efficacy is the high rate of hepcidin production present in
mice and in patients receiving heparin to prevent deep venous humans, so that would require massive doses of therapeutic agents
thrombosis which has achieved an increase in serum iron and trans- in order to achieve their goal.10 However, it has been shown that the
ferrin saturation and a reduction in C-reactive protein.40 Several intermittent or partial neutralization of hepcidin activity may be
S. Conde Diez et al. / Med Clin (Barc). 2017;148(5):218–224 223

sufficient to achieve the desired therapeutic effects.10,47 A human References


anti-hepcidin antibody, 12B9m, has been evaluated in transgenic
1. Ganz T. Systemic iron homeostasis. Physiol Rev. 2013;93:1721–41.
mice with inflammatory anaemia caused by Brucella abortus and 2. Arezes J, Nemeth E. Hepcidin and iron disorders: new biology and clinical
in monkeys. Neutralization of hepcidin by MAb increased ery- approches. Int J Lab Hem. 2015;37:92–8.
thropoiesis and haemoglobin levels.48 Currently, clinical trials are 3. Muñoz M, García-Erce JA, Remacha AF. Disorders of iron metabolism. Part I:
molecular basis of iron homeostasis. J Clin Pathol. 2011;64:281–6.
being conducted with another anti-hepcidin antibody, LY2787106, 4. Ganz T. Iron, erythropoiesis, and host defense: A ménage à trois. Am Soc Hematol
in patients with anaemia associated with cancer.10 Anticalines are E Donnall Thomas Lecture. 2014.
small proteins designed by genetic engineering targeting various 5. Nemeth E, Ganz T. Anemia of inflammation. Hematol Oncol Clin North Am.
2014;28:671–81.
biological ligands.47 One of these compounds is the PRS-080, which 6. Waldvogel-Abramowski S, Waeber G, Gassner C, Buser A, Frey BM, Favrat B, et al.
binds to human hepcidin with subnanomolar affinity, achieving an Physiology of iron metabolism. Transfus Med Hemother. 2014;41:213–21.
effective mobilization of iron and hypersideremia in primates.49 7. Ganz T, Nemeth E. Hepcidina and iron homeostasis. Biochim Biophys Acta.
2012;1823:1434–43.
Aptamers (Spiegelmers) are l-oligonucleotides designed against the
8. Kautz L, Jung G, Valore EV, Rivella S, Nemeth E, Ganz T. Identification of
mirror image of the target, to which they bind with high affin- erythroferrone as an erythroid regulator of iron metabolism. Nat Genet.
ity. They are stable in the circulation and immunologically passive 2014;46:678–84.
9. Pietrangelo A. Genetics, genetic testing, and management of hemochromatosis:
because their structure contains l-ribose, instead of d-ribose, which
15 years since hepcidin. Gastroenterology. 2015;149:1240–51.
gives them a high degradation resistance due to nucleases.50 NOX- 10. Ruchala P, Nemeth E. The pathophisiology and pharmacology of hepcidin. Trends
H94 neutralizes human hepcidin and increases plasma iron and Pharmacol Sci. 2014;35:155–61.
transferrin saturation.50 11. Liu J, Sun B, Yin H, Liu S. Hepcidin A promising therapeutic tar-
get for iron disorders. A systematic review. Medicine (Baltimore).
2016;95:e3150, http://dx.doi.org/10.1097/MD.0000000000003150. Available
in: www.md-journal.com
Ferroportin as a target. Blocking hepcidin-ferroportin interaction
12. Wu XG, Wang Y, Wu Q, Cheng WH, Liu W, Zhao Y, et al. HFE interacts
Hepcidin binding to ferroportin takes place in an extracellu- with the BMP type I receptor ALK3 to regulate hepcidin expression. Blood.
lar region of ferroportin containing the sulfhydryl residue Cys326 2014;124:1335–43.
13. Muckenthaler MU. How mutant HFE causes hereditary hemochromatosis. Blood.
surrounded by hydrophobic residues.10 Blocking this interaction
2014;124:1212–3.
prevents endocytosis and ferroportin destruction, which favours 14. Camaschella C. Iron-deficiency anemia. N Engl J Med. 2015;372:1832–43.
the efflux of iron from cells into plasma. A humanized antifer- 15. Muñoz M, García-Erce JA, Remacha AF. Disorders of iron metabolism. Part II:
roportin MAb, the LY2928057, is being evaluated. It targets this iron deficiency and iron overlad. J Clin Pathol. 2011;64:287–96.
16. Wang CY, Babitt JL. Hepcidin regulation in the anemia of inflammation. Curr
region and prevents hepcidin binding the carrier without interfer- Opin Hematol. 2016;23:189–97.
ing with the efflux of iron through ferroportin,51 increasing plasma 17. López-Gómez JM, Abad S, Vega A. Nuevas expectativas en el tratamiento de la
iron levels in cynomolgus monkeys. Fursultiamine is a thiol com- anemia en la enfermedad renal crónica. Nefrologia. 2016;36:232–6.
18. Powell LW, Seckington RC, Deugnier Y. Haemochromatosis. Lancet.
pound that prevents hepcidin binding to ferroportin in vitro and 2016;388:706–16.
in vivo in mice.52 19. Altés A, Sanz C, Bruguera M. Hemocromatosis hereditaria. Problemas en el diag-
nóstico y tratamiento. Med Clin (Barc). 2015;144:424–8.
20. Fernandes A, Preza GC, Phung Y, Kaplan J, Ganz T, Nemeth E. The molecular basis
Other inhibitors of hepcidin of hepcidin-resistant hereditary hemochromatosis. Blood. 2009;114:437–43.
TNF-␣ inhibitors (golimumab or infliximab) also decrease the 21. Rivella S. The role of ineffective erythropoiesis in non-transfusion-dependent
thalassemia. Blood Rev. 2012;26:S12–5.
synthesis of hepcidin and improve the anaemia of chronic disease 22. Camaschella C, Nai A. Ineffective erythropoiesis and regulation of iron status in
through an indirect effect on IL653,54 and vitamin D, testosterone iron loading anaemias. Br J Haematol. 2016;172:512–23.
and 17-estradiol, which prevents SMAD55-57 phosphorylation. 23. Saliba AN, Taher AT. Morbidities in non-transfusion-dependent thalassemia.
Ann N Y Acad Sci. 2016;1368:82–94.
Finally, it should be noted that some extracts of Chinese medici- 24. Girelli D, Nemeth E. Swinkels hepcidin in the diagnosis of iron disorders. Blood.
nal plants, such as Caulis spatholobi, called jixueteng, have a potent 2016;127:2809–13.
inhibitor effect on HAMP expression through SMAD 1/5/858 phos- 25. Fung E, Nemeth E. Manipulation of the hepcidin pathway for therapeutics pur-
pose. Haematologica. 2013;98:1667–76.
phorylation suppression. 26. Sabastiani G, Wilkinson N, Pantopoulos K. Pharmacological targeting of the hep-
cidin/ferroportin axis. Front Pharmacol. 2016;7:160.
27. Palaneeswari MS, Ganesh M, Karthikeyan T, Devi AJ, Mythili SV. Hepcidin.
Conclusions Minireview. J Clin Diagn Res. 2013;7:1767–71.
28. Nai A, Pagani A, Mandelli G, Lidonnici MR, Silvestri L, Ferrari G, Camaschell
We have reviewed the mechanisms that regulate iron homeo- C. Deletion of TMPRSS6 attenuates the phenotype in a mouse model of beta-
thalassemia. Blood. 2012;119:5021–9.
stasis and hepcidin expression, as well as diseases related to its 29. Schmidt PJ, Toudjarska I, Sendamarai AK, Racie T, Milstein S, Bettencourt BR,
excess or decrease and the advantages of its quantification in the et al. An RNAi therapeutic targeting Tmprss6 decreases iron overload in Hfe(−/−)
diagnosis of these diseases. The importance of hepcidin as a thera- mice and ameliorates anemia and iron overload in murine ␤-thalassemia inter-
media. Blood. 2013;121:1200–8.
peutic target focusing on different compounds aimed at correcting 30. Silvestri L. Inhibiting the hepcidin inhibitor for treatment of iron overload. Blood.
iron overload or block/inhibit hepcidin in order to cure the anaemia 2013;121:1068–9.
of chronic disease has been analyzed. Numerous clinical trials are 31. Ganz T, Nemeth E. The hepcidin-ferroportin system as a therapeutic target in
anemias and iron overload disorders. Hematology Am Soc Hematol Educ Pro-
being conducted and it is expected that they will be soon applied to
gram. 2011:538–42.
clinical practice, but which of the different therapeutic approaches 32. Blanchette NL, Manz DH, Torti FM, Torti SV. Modulation of hepcidin to
will have more chances of success is not yet known. treat iron deregulation: potential clinical applications. Expert Rev Hematol.
2016;9:169–86.
33. Böser P, Seemann D, Liguori MJ, Fan L, Huang L, Hafner M, et al. Anti-
Authorship/collaboration repulsive guidance molecule C (RGMc) antibodies increases serum iron in
rats and cynomolgus monkeys by hepcidin downregulation. AAPS J. 2015;17:
930–8.
SCD, RCA and ECG have contributed to the literature review, 34. Kovac S, Böser P, Cui Y, Ferring-Appel D, Casarrubea D, Huang L, et al. Anti-
writing and discussion of the content of the work. ECG has made hemojuvelin antibody corrects anemia caused by inappropriately high hepcidin
levels. Haematologica. 2016;101:e173–6.
the final text review. 35. Theurl I, Schroll A, Sonnweber T, Nairz M, Theurl M, Willenbacher W, et al.
Pharmacologic inhibition of hepcidin expression reverses anemia of chronic
inflammation in rats. Blood. 2011;118:4977–84.
Conflicts of interest 36. Steinbicker AU, Sachidanandan C, Vonner AJ, Yusuf RZ, Deng DY, Lai CS, et al. Inhi-
bition of bone morphogenetic protein signaling attenuates anemia associated
There are no conflicts of interest. with inflammation. Blood. 2011;117:4915–23.
224 S. Conde Diez et al. / Med Clin (Barc). 2017;148(5):218–224

37. Corradini E, Schmidt PJ, Meynard D, Garuti C, Montosi G, Chen S, et al. BMP6 treat- 48. Cooke KS, Hinkle B, Salimi-Moosavi H, Foltz I, King C, Rathanaswami P, et al. A
ment compensates for the molecular defect and ameliorates hemochromatosis fully human anti-hepcidin antibody modulates iron metabolism in both mice
in Hfe knockout mice. Gastroenterology. 2010;139:1721–9. and nonhuman primates. Blood. 2013;122:3054–61.
38. Poli M, Asperti M, Ruzzenenti P, Regoni M, Arosio P. Hepcidin antagonists 49. Hohlbaum A, Gille H, Christian J, Allersdorfer A, Jaworski J, Burrows J, et al.
for potential treatments of disorders with hepcidin excess. Front Pharmacol. Iron mobilization and pharmacodynamic marker measurements in non-human
2014;5:86, http://dx.doi.org/10.3389/fphar.2014.00086. www.frontiersin.org primates following administration of PRS-080, a novel and highly specific anti-
39. Gilleron J, Paramasivam P, Zeigerer A, Querbes W, Marsico G, Andree C, et al. hepcidin therapeutic [abstract]. Am J Hematol. 2013;5:E41.
Identification of siRNA delivery enhancers by a chemical library screen. Nucleic 50. Schwoebel F, van Eijk LT, Zboralski D, Sell S, Buchner K, Maasch C, et al. The effects
Acids Res. 2015;43:7984–8001. of the anti-hepcidin Spiegelmer NOX-H94 on inflammation-induced anemia in
40. Poli M, Asperti M, Naggi A, Campostrini N, Girelli D, Corbella M, et al. Glycol-split cynomolgus monkeys. Blood. 2013;121:2311–5.
nonanticoagulant heparins are inhibitors of hepcidin expression in vitro and in 51. Witcher DR, Leung D, Hill KA, de Rosa DC, Xu J, Manetta J, et al. LY2928057,
vivo. Blood. 2014;123:1564–73. an antibody targeting ferroportin, is a potent inhibitor of hepcidin activ-
41. Poli M, Asperti M, Ruzzenenti P, Mandelli L, Campostrini N, Martini G, et al. Over- ity and increases iron mobilization in normal cynomolgus monkeys. Blood.
sulfated heparins with low anticoagulant activity are strong and fast inhibitors 2013;122:3433.
of hepcidin expression in vitro and in vivo. Biochem Pharmacol. 2014;92:467–75. 52. Fung E, Sugianto P, Hsu J, Damoiseaux R, Ganz T, Nemeth E. High-throughput
42. Casper C, Chaturvedi S, Munshi N, Wong R, Qi M, Schaffer M, et al. screening of small molecules identifies hepcidin antagonists. Mol Pharmacol.
Analysis of inflammatory and anemia-related biomarkers in a randomized, 2013;83:681–90.
double-blind, placebo-controlled study of siltuximab (anti-IL6 monoclonal 53. Doyle MK, Rahman MU, Frederick B, Birbara CA, de Vries D, Teodter G, et al.
antibody) in patients with multicentric Castleman disease. Clin Cancer Res. Effects of subcutaneous and intravenous golimumab on inflammatory biomark-
2015;21:4294–304. ers in patients with rheumatoid arthritis: results of a phase 1, randomized,
43. Song SN, Tomosugi N, Kawabata H, Ishikawa T, Nishikawa T, Yoshizaki K. Down- open-label trial. Rheumatology. 2013;52:1214–9.
regulation of hepcidin resulting from long-term treatment with an anti-IL-6 54. Salama MF, Bayele HK, Srai SS. Tumour necrosis factor alpha downregulates
receptor antibody (tocilizumab) improves anemia of inflammation in multicen- human hemojuvelin expression via a novel response element within its pro-
tric Castleman disease. Blood. 2010;116:3627–34. moter. J Biomed Sci. 2012;19:1–12.
44. Zhang SP, Wang Z, Wang LX, Liu SJ. AG490: an inhibitor of hepcidin expression 55. Bacchetta J, Zaritsky JJ, Sea JL, Chun RF, Lisse TS, Zavala K, et al. Suppression of
in vivo. World J Gastroenterol. 2011;17:5032–4. iron-regulatory hepcidin by vitamin D. J Am Soc Nephrol. 2014;25:564–72.
45. Elliott J, Mishler D, Agarwal R. Hyporesponsiveness to erythropoietin: causes 56. Yang Q, Jian J, Katz S, Abramson SB, Huang X. 17␤-Estradiol inhibits
and management. Adv Chronic Kidney Dis. 2009;16:94–100. iron hormone hepcidin through an estrogen responsive element half-site.
46. Provenzano R, Besarab A, Sun CH, Diamond SA, Durham JH, Cangiano JL, et al. Endocrinology. 2012;153:3170–8.
Oral hypoxia-inducible factor prolyl hydroxylase inhibitor roxadustat (FG-4592) 57. Guo W, Bachman E, Li M, Roy CN, Blusztajn J, Wong S, et al. Testosterone admin-
for the treatment of anemia in patients with CKD. Clin J Am Soc Nephrol. istration inhibits hepcidin transcription and is associated with increased iron
2016;11:982–91. incorporation into red blood cells. Aging Cell. 2013;12:280–91.
47. Laarakkers CM, Wiegerinck ET, Klaver S, Kolodziejczyk M, Gille H, Hohlbaum 58. Guan Y, An P, Zhang Z, Zhang F, Tu Y, Wu Q, et al. Screening identifies the Chinese
AM, et al. Improved mass spectrometry assay for plasma hepcidin: detection medicinal plant caulis spatholobi as an effective HAMP expression inhibitor. J
and characterization of a novel hepcidin isoform. PLoS One. 2013;8:e75518. Nutr. 2013;143:1061–6.

También podría gustarte