You are on page 1of 7

Anemia of Chronic Disease

Naseema Gangat and Alexandra P. Wolanskyj

Anemia of chronic disease (ACD) or inammation may be secondary to infections, autoimmune


disorders, chronic renal failure, or malignancies. It is characterized by an immune activation with an
increase in inammatory cytokines and resultant increase in hepcidin levels. In addition, inappropriate
erythropoietin levels or hyporesponsiveness to erythropoietin and reduced red blood cell survival
contribute to the anemia. Hepcidin being the central regulator of iron metabolism plays a key role in
the pathophysiology of ACD. Hepcidin binds to the iron export protein, ferroportin, present on
macrophages, hepatocytes, and enterocytes, causing degradation of the latter. This leads to iron trapping
within the macrophages and hepatocytes, resulting in functional iron deciency. Production of hepcidin
is in turn regulated by iron stores, inammation, and erythropoiesis via the BMP-SMAD and JAK-STAT
signaling pathways. Treatment of anemia should primarily be directed at the underlying disease, and
conventional therapy such as red blood cell transfusions, iron, erythropoietin, and novel agents targeting
the hepcidin-ferroportin axis and signaling pathways (BMP-SMAD, JAK-STAT) involved in hepcidin
production also may be considered.
Semin Hematol 50:232238. C 2013 Elsevier Inc. All rights reserved.

A nemia of chronic disease (ACD) or anemia of


chronic inammation is the leading cause of
anemia in hospitalized patients and is the second
most common cause of anemia after iron deciency
anemia.1 The major etiologies are acute and chronic
hepcidin generation; and standard and new therapeutic
agents under development for ACD.

IRON HOMEOSTASIS
infections, autoimmune disorders, chronic renal insuf- Iron Metabolism
ciency, and malignancies (both hematologic and solid
tumors). In most instances, the anemia is mild normo- Iron is an essential micronutrient required for heme
cytic/normochromic, but it may be microcytic/hypochro- biosynthesis, which is subsequently incorporated into
mic in a few cases, making it indistinguishable from iron hemoglobin in red blood cells. In addition, iron is present
deciency anemia. Pathophysiology is multifactorial and in proteins such as cytochromes involved in respiration,
complex, involving disordered iron metabolism and dis- myoglobin, and other iron-containing proteins. The total
tribution secondary to increased hepcidin, a peptide iron content in the body is tightly regulated since excess
hormone that is the central regulator of iron metabolism.2 iron is toxic via generation of free radicals and its
Given the key role hepcidin plays in iron homeostasis, propensity to deposit in various organs such as liver,
several novel agents targeting the hepcidinferroportin axis cardiac myocytes, and endocrine organs.4 The body
as well as signaling pathways (BMP6-HJV-SMAD and IL- requires more than 20 mg of iron daily, of which only
6-JAK-STAT) involved in hepcidin production are under 1 to 2 mg is derived from intestinal absorption. The
development for treating this disorder.3 In this review, we majority of iron is provided through recycling by the
will provide a comprehensive update on the causes and degradation of senescent red blood cells within macro-
pathophysiology of ACD; its laboratory characteristics; phages in the liver, spleen, and bone marrow (reticulo-
iron metabolism and its regulation by hepcidin, regulation endothelial system).5 The major iron storage sites are the
of hepcidin, and key signaling pathways involved in hepatocytes and macrophages, and along with duodenal
enterocytes involved in iron absorption, they release iron
into the extracellular uid or plasma. Since iron is not
excreted from the body, the entire process of intestinal
Division of Hematology, Mayo Clinic, Rochester, MN.
Conicts of interest: iron absorption and release of iron from macrophages and
Address correspondence to Alexandra P. Wolanskyj, MD, Division of hepatocytes is stringently regulated by the peptide hor-
Hematology, Mayo Clinic, 200 First St SW, Rochester, MN 55905. mone hepcidin (Figure 1).6 Disordered iron metabolism is
E-mail: wolanskyj.alexandra@mayo.edu
0037-1963/$ - see front matter
the hallmark of ACD as a consequence of increased
& 2013 Elsevier Inc. All rights reserved. hepcidin levels. The increase in hepcidin levels by either
http://dx.doi.org/10.1053/j.seminhematol.2013.06.006 increased production or decreased clearance leads to a

232 Seminars in Hematology, Vol 50, No 3, July 2013, pp 232238


Anemia of chronic disease 233

Figure 1. Regulation of iron metabolism by hepcidin. Hepcidin production by the liver is regulated by inammation via
cytokines, iron levels/stores, and erythropoiesis. Hepcidin binds to its receptor ferroportin (iron export protein) present on
hepatocytes, macrophages and duodenal enterocytes, resulting in internalization and degradation of ferroportin. In
addition, hepcidin inhibits intestinal absorption of iron by duodenal enterocytes. The end result is that iron is trapped
within the hepatocytes and macrophages resulting in hypoferremia, functional iron deciency and iron-restricted
erythropoiesis. Abbreviations: Fpn, ferroportin; Tf, transferring; TfR, transferrin receptor. Modied with permission from
Ganz et al. Cold Spring Harb Perspect Med 2012;2:a011668.

block in release of iron from iron storage cells (ie, and placental syncytiotrophoblasts.18,19 Hepcidin binding
enterocytes, macrophages, hepatocytes) to the plasma to ferroportin leads to ubiquitinization, followed by
and hence hypoferremia, resulting in functional iron internalization and degradation of ferroportin.20,21 The
deciency and iron-restricted erythropoiesis.7,8 end result is that iron cannot be released into the plasma
and remains trapped inside the macrophages and hepato-
cytes, resulting in an increase in iron stores reected in
Hepcidin: Regulator of Iron Metabolism high levels of serum ferritin.22,23 In addition, hepcidin
Hepcidin is a 25amino acid peptide with anti- inhibits intestinal absorption of iron.24 Therefore, ACD is
microbial properties that is produced by the liver in characterized by low serum iron, transferrin, and total iron
response to increases in total body iron and inamma- binding capacity, by normal transferrin saturation, and by
tion.2,9,10 The production of hepcidin is regulated by increased ferritin, the latter in contrast to iron deciency
iron stores, pro-inammatory cytokines, and the anemia/ anemia. The low transferrin levels are due to down-
erythroid response axis.11,12 regulation of transferrin synthesis as a result of an increase
The central role of hepcidin in iron metabolism in ferritin.
(reviewed in Figure 1) was elucidated from studies
performed in iron overload disorders such as hereditary
hemochromatosis, characterized by low hepcidin levels.
Regulation of Hepcidin by Iron
Hemochromatosis (HFE), hemojuvelin (HJV), transferrin Production of hepcidin is transcriptionally regulated by
receptor2 (TfR2), and hepcidin (HAMP) are the major total body iron, which includes serum iron and iron stores.
genes involved.1316 In juvenile hemochromatosis, muta- In iron overload conditions, such as hereditary hemochro-
tions in either HJV or HAMP occur, which results in low matosis, the increased total body iron, because of the
hepcidin levels.16,17 existence of a mutated gene involved in hepcidin gene
The increase in hepcidin production in response to regulation (HFE, HJV, TfR2) inhibits hepcidin produc-
inammation is a protective mechanism in the case of tion. However, in ACD, high levels of interleukin-6 (IL-6)
infections in which iron restriction would limit bacterial lead to an increase in hepcidin production, with low serum
growth. However, in ACD or inammation, it is a iron levels as a consequence. The major signaling pathway
maladaptive mechanism whereby iron remains sequestered involved in hepcidin generation is the bone morphoge-
in the macrophages and hepatocytes and is not available netic protein/sons of mothers against decapentaplegic
for erythropoiesis, resulting in anemia. Hepcidin produced (BMP)/SMAD pathway, which is also key in develop-
by the hepatocytes binds to its receptor ferroportin, which ment, morphogenesis, and osteogenesis (Figure 2).2527
is an iron export protein that is present on limited cells Low intracellular iron levels within the hepatocytes leads
such as macrophages, hepatocytes, duodenal enterocytes, to an increase in BMP-6 production. BMP-6 belongs to
234 N. Gangat and A.P. Wolanskyj

Figure 2. Signaling pathways regulating hepcidin production. BMP-SMAD and IL-6-JAK-STAT signaling pathways are the
two major pathways involved in hepcidin production. Low intracellular iron results in increased BMP-6 production. BMP-6
binds to the BMP-R, activating a series of phosphorylation events that result in generation of the SMAD 1/5/8-SMAD4
complex that translocates to the nucleus, binds to the hepcidin gene promoter, and turns on transcription. Extracellular
iron is bound to transferrin and when iron binds to transferrin receptor 1(TfR1), HFE-TfR2 sends a signal to the BMP
receptor complex. In addition, hemojuvelin (HJV) potentiates BMP-SMAD signaling. Production of inammatory cytokines
(IL-6) activates the JAK-STAT3 signaling pathway. IL-6 binds to the IL-6 receptor. JAK1/JAK2 are tyrosine kinases associated
with the IL-6-R. A series of phosphorylation events leads to phosphorylation of STAT3 which forms homodimers and
translocates to the nucleus, binding to the hepcidin gene promoter, and turns on hepcidin production. Modied with
permission from Ganz et al, Cold Spring Harb Perspect Med 2012; 2:a011668.

the transforming growth factor-beta family of ligands and malignancy, serves as a potent stimulus for hepcidin
binds to its receptor BMP (BMP-R) located on the surface production.30 Herein, the JAK-STAT signaling pathway
of hepatocytes. Hemojuvelin (HJV) serves as a co-receptor plays a key role (Figure 2).31,32 Pro-inammatory cyto-
for BMP and potentiates BMP/SMAD signaling.28 Ligand kines such as IL-6, IL-1, and tumor necrosis factor-alpha
binding to its receptor (BMP-R), which is a serine- are released and serve as ligands for this signaling pathway.
threonine kinase, triggers a series of phosphorylation JAK1/2 are non-receptor tyrosine kinases that are associ-
events with resultant phosphorylation of SMAD 1/5/8, ated with the IL-6 receptor (IL-6-R). IL-6 is the major
which in turn leads to phosphorylation of SMAD4. ligand involved.12 When IL-6 binds to its receptor, there is
SMAD 1/5/8/SMAD4 complex translocates to the nucleus activation of the JAK-STAT signaling pathway by a series
and binds to the promoter of the hepcidin gene and turns of phosphorylation events. JAK leads to phosphorylation
on hepcidin gene expression. Extracellular iron, which is of STAT3. Phosphorylated STAT3 homodimerizes and
bound to transferrin, the iron carrier protein, also serves to translocates to the nucleus, binding to the promoter of the
regulate hepcidin gene expression. Transferrin receptors hepcidin gene and turning on hepcidin gene expression. In
TfR1 and 2 are located on the hepatocyte surface in mice studies, injection with lipopolysaccharides (LPS)
association with HFE. When transferrin-iron binds to mimics a similar series of events.33 It is to be noted that
TfR1 and is internalized into the hepatocyte, TfR2 intact BMP-SMAD signaling is required for IL-6/JAK/
associates with HFE and sends a signal to potentiate STAT signaling to act on hepcidin gene expression, since
BMP-SMAD signaling and hence increases hepcidin gene the ability of IL-6 to increase hepcidin gene expression is
expression. The critical role of the BMP-SMAD signaling considerably diminished in SMAD4 knockout mice.34
pathway in regulating hepcidin expression was determined
by studies performed in SMAD knockout mice that failed
Regulation of Hepcidin by Erythropoiesis and
to produce hepcidin (Figure 2).29
Hypoxia
With certain types of anemia such as thalassemia and
Regulation of Hepcidin by Inammation the sideroblastic anemias, the anemia suppresses hepcidin
Inammation, both acute and chronic such as seen in production indirectly by an increase in erythropoietin and
the case of infections, autoimmune conditions, and subsequent expansion of erythroid progenitors in the bone
Anemia of chronic disease 235

marrow and by another mechanism(s) still not under- deciency anemia, which is microcytic, hypochromic with
stood. It is possible that in thalassemia overexpression of anisocytosis and poikilocytosis noted on peripheral blood
growth differentiation factor 15 (GDF15), a member of smear. Serum iron, total iron binding capacity, and
the transforming growth factor-beta superfamily occurring transferrin saturation are all low in ACD, and are
as a consequence of erythroid expansion, may inhibit accompanied by an increase in serum ferritin and bone
hepcidin expression.35 The regulation of hepcidin by marrow iron stores. On the other hand, in iron deciency
hypoxia is modulated via the hypoxia inducible factor anemia, serum iron, transferrin saturation, and ferritin are
von Hippel Lindau (HIF-1VHL) pathway. Under low, with an increase in total iron binding capacity. It is
hypoxic conditions HIF-1 is unable to undergo hydrox- often challenging to distinguish between ACD and iron
ylation by proline/lysine hydroxylase, escaping ubiquitina- deciency anemia based on available laboratory tests, and
tion and degradation. Under these conditions. HIF-1 even more complex are situations in which the two
translocates to the nucleus where it increases production of conditions coexist. Measurement of soluble transferrin
erythropoietin and red blood cell expansion; the latter in receptor, a truncated form of the membrane receptor,
turn suppresses hepcidin production.36,37 may be used to distinguish between the two entities. In
iron deciency, soluble transferrin receptor levels are high
since the availability of iron is low, but in ACD soluble
PATHOPHYSIOLOGY OF ACD
transferrin receptor levels are normal.38,39 Given the
Pathophysiology of ACD is complex but can be central role of hepcidin in regulating iron metabolism,
summarized as three main causations, rooted in the measuring serum hepcidin levels would serve to distin-
increase in pro-inammatory cytokines. Increase in hepci- guish between the two entities, but the issue remains the
din plays a key role.1 In addition, inappropriate eryth- lack of availability of a standard hepcidin assay. Hepcidin
ropoietin levels or hyporesponsiveness to erythropoietin, levels are reduced in iron deciency, and measurement of
and suppression of erythropoiesis in the bone marrow blood or urine hepcidin levels may be indicative of true
coupled with reduced red blood cell survival, contribute to iron deciency.39 Mass spectrometry and, more recently,
the anemia seen in chronic disease, as illustrated in enzyme-linked immunoassays (ELISA) for quantitation of
Figure 3. hepcidin in serum, plasma, and urine have been devel-
oped.40,41 A comparative study of different assays for
hepcidin levels was performed, which found that despite
LABORATORY CHARACTERISTICS OF ACD
signicant differences in the absolute value at each
ACD is a mild to moderate normocytic normochromic laboratory, results for samples correlated well and analyt-
anemia, with less than 25% of cases depicting a microcytic ical variance was low.40 Furthermore, issues in interpreting
hypochromic anemia, in which case the mean corpuscular hepcidin levels include diurnal uctuations of hepcidin
volume is rarely less than 70.1 This is in contrast to iron (lower in the morning, higher in the afternoon), and
relative sensitivity to the iron content of the diet.41,42

TREATMENT OF ACD
Treatment is primarily directed at the underlying
disease in the case of infections, autoimmune disorders,
and malignancy, but most of these conditions are chronic
and eradication of the underlying disease is difcult.
Improvement of the anemia contributes to improvement
in the quality of life of these patients.43,44 Currently
available treatments may be categorized either as conven-
tional therapy or novel agents as summarized in Table 1.

Conventional Therapy
Red blood cell transfusions should be provided for
hemoglobin o8 g/dL, particularly for patients with
known coronary artery disease. It is to be kept in mind
Figure 3. Pathophysiology of anemia of chronic disease. that repeated use of blood transfusions leads to iron
Summarized as three main causations rooted in increase overload, risk of transmission of infections, and allo-
in pro-inammatory cytokines: (i) increase in hepcidin
levels, (ii) inappropriate erythropoietin levels or hypor-
immunization.1
esponsive to erythropoietin, (and iii) suppression of Since ACD is accompanied by functional or in some
erythropoiesis in the bone marrow coupled with reduced cases absolute iron deciency, iron supplementation might
red blood cell survival. Abbreviation: EPO; erythropoietin. be of benet particularly for patients with low ferritin and
236 N. Gangat and A.P. Wolanskyj

Table 1. Treatment Options for Anemia of Chronic Disease*


Therapy Types Description Dosing
Conventional Red blood cell transfusions 12 U, at the physicians discretion
Intravenous iron with Iron sucrose 100 mg/dose
erythropoiesis-stimulating agents Iron dextran 25 mg test dose followed by
500 mg to 2,000 mg
Erythropoiesis-stimulating Epoetin-alpha, -beta 20,000 to 60,000 U
agents weekly
Darbepoetin 300 g every 3 weeks
if hemoglobin o11 g/dL. Hold dose if
hemoglobin 412 g/dL.
Novel Direct hepcidin antagonists
Monoclonal antibodies mAb2.7, Ab12B9m
siRNA ALN-HPN
Anticalins PRS-080
Spiegelmers NOX-H94
BMP-HJV-SMAD inhibitors LDN-193189
IL-6 antagonists Tocilizumab
Siltuximab
JAK-STAT inhibitors AG490
PpYLKTK
Ferroportin agonists/stabilizers Anti-ferroportin monoclonal antibody
*Therapy should primarily be directed towards the underlying disease.

hyporesponsiveness to erythropoeisis-stimulating agents HJV and could potentially be effective therapeutic agents
(ESAs).45 Intravenous iron is used instead of oral iron except for technical challenges encountered in the delivery
because of the increase in hepcidin levels and inhibition of of RNAi.51,52 In addition, hepcidin binding proteins,
intestinal absorption of iron. anticalins, and spiegelmers are also being developed.53,54
ESAs such as epoetin-alpha, epoetin-beta, and Spiegelmers are mirror image aptamers that are single-
darbopoeitin-alpha are widely used in patients with stranded synthetic oligonucleotides that bind with high
anemia secondary to chronic renal failure.46 However, afnity and specicity to a wide range of targets such as
studies have shown an increased risk of cardiovascular peptides, proteins, etc, being produced by NOXXON
events with liberal use of ESAs.47,48 Therefore, it is critical Pharma per their website. The anti-hepcidin Spiegelmer
to adjust the ESA dose to keep hemoglobin o13 g/dL.49 NOX-H94 is in a phase IIa clinical trial to treat anemia
In addition, ESA use has been associated with disease associated with chronic disease. This phase IIa study was
progression in a variety of tumors.50 initiated following the successful completion of the clinical
Since blood transfusions, iron therapy, and ESAs are phase I program, which was presented at the American
not without detrimental effects, the search continues for Society of Hematology (ASH) meeting in Atlanta, GA in
better therapies targeting the underlying mechanism that December 2012.55,56 The phase I study consisted of a
contributes to ACD. comprehensive single and multiple ascending dose study
in healthy volunteers and a subsequent human pharma-
codynamic study to assess the ability of NOX-H94 to
Novel Agents prevent endotoxin-induced hypoferremia in healthy sub-
Hepcidin antagonists (monoclonal antibodies, small jects. This endotoxemia study delivered the rst clinical
interfering RNA [siRNA], antisense oligonucleotides, evidence that NOX-H94 is capable of neutralizing high
hepcidin binding proteins, and aptamers) are being levels of hepcidin in humans and maintaining higher
developed. Anti-hepcidin monoclonal antibodies are under serum iron concentrations compared to subjects receiving
development by Amgen and Eli Lilly according to their placebo.
website. These antibodies bind to hepcidin and prevent it The increased production of hepcidin in ACD serves as
from binding to ferroportin.51 RNA interference (RNAi) the rationale for development of inhibitors targeting the
and antisense oligonucleotides interfere with either tran- BMP-HJV-SMAD and IL-6-JAK-STAT pathways that are
scription or translation of hepcidin or its regulators such as involved in hepcidin synthesis. Dorsomorphin is a small
Anemia of chronic disease 237

molecular inhibitor of BMP receptor, and a derivative of 10. Krause A, Neitz S, Magert HJ, et al. LEAP-1, a novel highly
dorsomorphin, LDN-193189, is a more selective BMP disulde-bonded human peptide, exhibits antimicrobial
inhibitor. The latter has been shown to reverse anemia activity. FEBS Lett. 2000;480:147-50.
11. Finch C. Regulators of iron balance in humans. Blood.
associated with chronic arthritis in rats.57 Other agents
1994;84:1697-702.
include anti-BMP6 monoclonal antibody and soluble
12. Nemeth E, Rivera S, Gabayan V, et al. IL-6 mediates
HJV.25,27 In addition, heparin also has been shown to hypoferremia of inammation by inducing the synthesis of
decrease BMP-SMAD signaling and inhibit hepcidin the iron regulatory hormone hepcidin. J Clin Invest.
transcription.58 2004;113:1271-6.
In regards to the IL-6-JAK-STAT pathway, monoclo- 13. Feder JN. The hereditary hemochromatosis gene (HFE): a
nal IL-6 and IL-6R antibodies and the JAK2 and STAT3 MHC class I-like gene that functions in the regulation of
inhibitors all have been shown to downregulate hepcidin iron homeostasis. Immunol Res. 1999;20:175-85.
expression.5961 14. Papanikolaou G, Samuels ME, Ludwig EH, et al. Mutations
Vitamin D deciency is associated with an increased in HFE2 cause iron overload in chromosome 1q-linked
prevalence of ACD, and vitamin D replacement lowers juvenile hemochromatosis. Nat Genet. 2004;36:77-82.
hepcidin levels.62 15. Camaschella C, Roetto A, Cali A, et al. The gene TFR2 is
mutated in a new type of haemochromatosis mapping to
Since hepcidin excess blocks ferroportin by causing
7q22. Nat Genet. 2000;25:14-5.
degradation of the latter, agents that stabilize ferroportin 16. Roetto A, Papanikolaou G, Politou M, et al. Mutant
or inhibit the interaction with hepcidin are an attractive antimicrobial peptide hepcidin is associated with severe
target.63 Eli Lily has developed an antiferroportin mono- juvenile hemochromatosis. Nat Genet. 2003;33:21-2.
clonal antibody that blocks hepcidinferroportin 17. Lanzara C, Roetto A, Daraio F, et al. Spectrum of
interaction. hemojuvelin gene mutations in 1q-linked juvenile hemo-
chromatosis. Blood. 2004;103:4317-21.
18. Donovan A, Brownlie A, Zhou Y, et al. Positional cloning of
CONCLUSION zebrash ferroportin1 identies a conserved vertebrate iron
exporter. Nature. 2000;403:776-81.
In the last decade, we have gained immense insight 19. Donovan A, Lima CA, Pinkus JL, et al. The iron exporter
regarding the complex pathophysiology of ACD with ferroportin/Slc40a1 is essential for iron homeostasis. Cell
increase in hepcidin levels and resultant disordered iron Metab. 2005;1:191-200.
metabolism playing a key role. Based on the central role of 20. Nemeth E, Tuttle MS, Powelson J, et al. Hepcidin regulates
hepcidin and its regulators in ACD, several novel targeted cellular iron efux by binding to ferroportin and inducing its
agents are currently under development; however, therapy internalization. Science. 2004;306:2090-3.
remains primarily directed at the underlying cause. 21. De Domenico I, Ward DM, Langelier C, et al. The
molecular mechanism of hepcidin-mediated ferroportin
down-regulation. Mol Biol Cell. 2007;18:2569-78.
REFERENCES 22. Rivera S, Nemeth E, Gabayan V, et al. Synthetic hepcidin
1. Weiss G, Goodnough LT. Anemia of chronic disease. causes rapid dose-dependent hypoferremia and is concen-
N Engl J Med. 2005;352:1011-23. trated in ferroportin-containing organs. Blood. 2005;
2. Pigeon C, Ilyin G, Courselaud B, et al. A new mouse liver- 106:2196-9.
specic gene, encoding a protein homologous to human 23. Rivera S, Liu L, Nemeth E, et al. Hepcidin excess induces
antimicrobial peptide hepcidin, is overexpressed during iron the sequestration of iron and exacerbates tumor-associated
overload. J Biol Chem. 2001;276:7811-9. anemia. Blood. 2005;105:1797-802.
3. Sun CC, Vaja V, Babitt JL, et al. Targeting the hepcidin- 24. Laftah AH, Ramesh B, Simpson RJ, et al. Effect of hepcidin
ferroportin axis to develop new treatment strategies for on intestinal iron absorption in mice. Blood. 2004;103:
anemia of chronic disease and anemia of inammation. 3940-4.
Am J Hematol. 2012;87:392-400. 25. Andriopoulos B Jr., Corradini E, Xia Y, et al. BMP6 is a key
4. McCord JM. Iron, free radicals, and oxidative injury. J Nutr. endogenous regulator of hepcidin expression and iron
2004;134:3171S-2S. metabolism. Nat Genet. 2009;41:482-7.
5. Andrews NC. Disorders of iron metabolism. N Engl J Med. 26. Corradini E, Meynard D, Wu Q, et al. Serum and liver iron
1999;341:1986-95. differently regulate the bone morphogenetic protein 6
6. Ganz T. Hepcidin and iron regulation, 10 years later. Blood. (BMP6)-SMAD signaling pathway in mice. Hepatology.
2011;117:4425-33. 2011;54:273-84.
7. Nicolas G, Bennoun M, Porteu A, et al. Severe iron deciency 27. Babitt JL, Huang FW, Xia Y, et al. Modulation of bone
anemia in transgenic mice expressing liver hepcidin. Proc Natl morphogenetic protein signaling in vivo regulates systemic
Acad Sci U S A. 2002;99:4596-601. iron balance. J Clin Invest. 2007;117:1933-9.
8. Yamaji S, Sharp P, Ramesh B, et al. Inhibition of iron 28. Babitt JL, Huang FW, Wrighting DM, et al. Bone
transport across human intestinal epithelial cells by hepcidin. morphogenetic protein signaling by hemojuvelin regulates
Blood. 2004;104:2178-80. hepcidin expression. Nat Genet. 2006;38:531-9.
9. Park CH, Valore EV, Waring AJ, et al. Hepcidin, a urinary 29. Wang RH, Li C, Xu X, et al. A role of SMAD4 in iron
antimicrobial peptide synthesized in the liver. J Biol Chem. metabolism through the positive regulation of hepcidin
2001;276:7806-10. expression. Cell Metab. 2005;2:399-409.
238 N. Gangat and A.P. Wolanskyj

30. Nemeth E, Valore EV, Territo M, et al. Hepcidin, a putative 47. Pfeffer MA, Burdmann EA, Chen CY, et al. A trial of
mediator of anemia of inammation, is a type II acute-phase darbepoetin alfa in type 2 diabetes and chronic kidney
protein. Blood. 2003;101:2461-3. disease. N Engl J Med. 2009;361:2019-32.
31. Verga Falzacappa MV, Vujic Spasic M, Kessler R, et al. 48. Singh AK, Szczech L, Tang KL, et al. Correction of anemia
STAT3 mediates hepatic hepcidin expression and its inam- with epoetin alfa in chronic kidney disease. N Engl J Med.
matory stimulation. Blood. 2007;109:353-8. 2006;355:2085-98.
32. Wrighting DM, Andrews NC. Interleukin-6 induces hepci- 49. Palmer SC, Navaneethan SD, Craig JC, et al. Meta-analysis:
din expression through STAT3. Blood. 2006;108:3204-9. erythropoiesis-stimulating agents in patients with chronic
33. Roy CN, Custodio AO, de Graaf J, et al. An Hfe-dependent kidney disease. Ann Intern Med. 2010;153:23-33.
pathway mediates hyposideremia in response to lipopoly- 50. Bohlius J, Schmidlin K, Brillant C, et al. Recombinant
saccharide-induced inammation in mice. Nat Genet. human erythropoiesis-stimulating agents and mortality in
2004;36:481-5. patients with cancer: a meta-analysis of randomised trials.
34. Verga Falzacappa MV, Casanovas G, Hentze MW, et al. A
Lancet. 2009;373:1532-42.
bone morphogenetic protein (BMP)-responsive element in
51. Sasu BJ, Cooke KS, Arvedson TL, et al. Antihepcidin
the hepcidin promoter controls HFE2-mediated hepatic
antibody treatment modulates iron metabolism and is
hepcidin expression and its response to IL-6 in cultured
effective in a mouse model of inammation-induced anemia.
cells. J Mol Med (Berl). 2008;86:531-40.
Blood. 2010;115:3616-24.
35. Tanno T, Bhanu NV, Oneal PA, et al. High levels of
52. Wang J, Lu Z, Wientjes MG, et al. Delivery of siRNA
GDF15 in thalassemia suppress expression of the iron
regulatory protein hepcidin. Nat Med. 2007;13:1096-101. therapeutics: barriers and carriers. AAPS J. 2010;12:
36. Nicolas G, Chauvet C, Viatte L, et al. The gene encoding 492-503.
the iron regulatory peptide hepcidin is regulated by 53. Schlehuber S, Skerra A. Lipocalins in drug discovery: from
anemia, hypoxia, and inammation. J Clin Invest. 2002; natural ligand-binding proteins to "anticalins". Drug Discov
110:1037-44. Today. 2005;10:23-33.
37. Vokurka M, Krijt J, Sulc K, et al. Hepcidin mRNA levels in 54. Vater A, Klussmann S. Toward third-generation aptamers:
mouse liver respond to inhibition of erythropoiesis. Physiol Spiegelmers and their therapeutic prospects. Curr Opin
Res. 2006;55:667-74. Drug Discov Dev. 2003;6:253-61.
38. Brugnara C. Iron deciency and erythropoiesis: new diag- 55. Riecke K, ZS, Boyce M, Zollner S, et al. Single and repeated
nostic approaches. Clin Chem. 2003;49:1573-8. dose rst-in-human study with the anti-hepcidin spiegelmer
39. Kemna EH, Tjalsma H, Willems HL, et al. Hepcidin: from Nox-H94. Blood. 2012;120:2342.
discovery to differential diagnosis. Haematologica. 2008; 56. van Eijk L, SD, Aaron J, Swinkels DW, et al. Randomized
93:90-7. double blind placebo controlled PK/PD study on the effects
40. Kroot JJ, Kemna EH, Bansal SS, et al. Results of the rst of a single intravenous dose of the anti-hepcidin spiegelmer
international round robin for the quantication of urinary Nox-H94 on serum iron during experimental human
and plasma hepcidin assays: need for standardization. endotoxemia. Blood. 2012;120:3452.
Haematologica. 2009;94:1748-52. 57. Theurl I, Schroll A, Sonnweber T, et al. Pharmacologic
41. Ganz T, Olbina G, Girelli D, et al. Immunoassay for human inhibition of hepcidin expression reverses anemia of chronic
serum hepcidin. Blood. 2008;112:4292-7. inammation in rats. Blood. 2011;118:4977-84.
42. Kroot JJ, Hendriks JC, Laarakkers CM, et al. (Pre)analytical 58. Poli M, Girelli D, Campostrini N, et al. Heparin: a potent
imprecision, between-subject variability, and daily variations inhibitor of hepcidin expression in vitro and in vivo. Blood.
in serum and urine hepcidin: implications for clinical 2011;117:997-1004.
studies. Anal Biochem. 2009;389:124-9. 59. Hashizume M, Uchiyama Y, Horai N, et al. Tocilizumab, a
43. Moreno F, Sanz-Guajardo D, Lopez-Gomez JM, et al. humanized anti-interleukin-6 receptor antibody, improved
Increasing the hematocrit has a benecial effect on quality anemia in monkey arthritis by suppressing IL-6-induced
of life and is safe in selected hemodialysis patients. Spanish
hepcidin production. Rheumatol Int. 2010;30:917-23.
Cooperative Renal Patients Quality of Life Study Group of
60. van Rhee F, Fayad L, Voorhees P, et al. Siltuximab, a novel
the Spanish Society of Nephrology. J Am Soc Nephrol.
anti-interleukin-6 monoclonal antibody, for Castleman's
2000;11:335-42.
disease. J Clin Oncol. 2010;28:3701-8.
44. Littlewood TJ, Bajetta E, Nortier JW, et al. Effects of
61. Fatih N, Camberlein E, Island ML, et al. Natural and
epoetin alfa on hematologic parameters and quality of life in
synthetic STAT3 inhibitors reduce hepcidin expression in
cancer patients receiving nonplatinum chemotherapy: results
of a randomized, double-blind, placebo-controlled trial. differentiated mouse hepatocytes expressing the active phos-
J Clin Oncol. 2001;19:2865-74. phorylated STAT3 form. J Mol Med (Berl). 2010;88:
45. Goodnough LT, Skikne B, Brugnara C. Erythropoietin, 477-86.
iron, and erythropoiesis. Blood. 2000;96:823-33. 62. Perlstein TS, Pande R, Berliner N, et al. Prevalence of
46. Hertel J, Locay H, Scarlata D, et al. Darbepoetin alfa 25-hydroxyvitamin D deciency in subgroups of elderly
administered every other week maintains hemoglobin levels persons with anemia: association with anemia of inamma-
over 52 weeks in patients with chronic kidney disease tion. Blood. 2011;117:2800-6.
converting from once-weekly recombinant human erythro- 63. Fernandes A, Preza GC, Phung Y, et al. The molecular basis
poietin: results from simplify the treatment of anemia with of hepcidin-resistant hereditary hemochromatosis. Blood.
Aranesp (STAAR). Am J Nephrol. 2006;26:149-56. 2009;114:437-43.

You might also like