The discovery of the iron-regulatory hormone hepcidin in 2001 has revolutionized our understanding of iron disorders, and its measurement should advance diagnosis/treatment of these conditions. Although several assays have been developed, a gold standard is still lacking, and efforts toward harmonization are ongoing. Nevertheless, promising applications can already be glimpsed, ranging from the use of hepcidin levels for diagnosing iron-refractory iron deficiency anemia to global health applications such as guiding safe iron supplementation in developing countries with high infection burden.

Hepcidin, a liver-derived peptide hormone, is a key regulator of systemic iron homeostasis, and its unbalanced production contributes to the pathogenesis of a spectrum of iron disorders. Hepcidin functions by blocking the following iron flows into plasma: duodenal absorption, release from macrophages recycling old red blood cells, and mobilization of stored iron from hepatocytes (for extensive reviews, see Ganz1  and Hentze et al2 ). This blocking of iron flows is achieved by hepcidin causing degradation of its receptor, the iron transporter ferroportin. Hepcidin production is tightly regulated: it is (1) increased by plasma and liver iron as a feedback mechanism to maintain stable body iron levels, (2) decreased by erythroid activity to ensure iron supply for erythropoiesis, and (3) increased by inflammation as a host defense mechanism to limit extracellular iron availability to microbes.1-3  Because hepcidin levels reflect the integration of multiple key signals involved in iron regulation, and hepcidin directly controls iron absorption and bioavailability in circulation,4-6  its measurement should be a useful clinical tool for the management of iron disorders. Although a thorough understanding of its unique advantages over traditional biomarkers of iron status in different conditions will require larger studies with head-to-head comparisons, we discuss here some studies already supporting the distinct utility of hepcidin measurements.

The bioactive circulating form of hepcidin is 25 amino acids in size. N-terminal degradation leads to smaller isoforms (hepcidin-24, -23, -22, and -20) of unknown significance.7-9  These isoforms are generally present in diseases with elevated hepcidin-25 levels, including chronic kidney disease and sepsis.10,11  Circulating hepcidin is bound to α-2-macroglobulin and albumin, but estimates of this binding vary from <3% to 89% of total.12,13  The impact of binding on hepcidin activity or assay performance is unclear. Hepcidin is rapidly excreted by the kidney and reabsorbed in the proximal tubules by megalin-dependent endocytosis, but urinary hepcidin levels generally correlate with serum levels,14,15  with the exception of renal diseases.16  Nevertheless, urinary hepcidin measurements may have utility in specific conditions, such as noninvasive measurement in children, screening in low-resource settings, or prediction of acute kidney injury.17 

The development of assays to quantify hepcidin in biological samples has proved challenging. Because hepcidin is a small, evolutionarily conserved peptide, it is difficult to generate antibodies for laboratory assays. Hepcidin quantification is further complicated by its tendency to aggregate and stick to laboratory plastics.18  Nonetheless, a number of well-performing assays have been established and are listed in supplemental Table 1, available on the Blood Web site. They are divided into two major groups: mass spectrometry–based and classical immunoassays.19  Mass spectrometry–based assays require relatively expensive equipment but are able to distinguish the hepcidin isoforms. Immunoassays generally lack specificity for hepcidin-25 and measure total hepcidin levels. The relevance of specifically measuring hepcidin-25 instead of total hepcidin for clinical decision making, however, is unclear.

In the absence of a primary reference material, a reference method, and a commutable calibrator, absolute hepcidin levels differ widely between assays (up to 10-fold).20  Studies aiming at harmonization are ongoing,21  but for now, these differences preclude data comparability and the establishment of a universal reference range. Rather, each method/laboratory needs to establish rigorous age- and sex-specific reference ranges, preferably not only for hepcidin but also for hepcidin-to-transferrin saturation and hepcidin-to-serum ferritin ratios. To date, only 2 large studies in Holland (n = 2998)22  and Italy (n = 1577)23  have evaluated serum hepcidin variations at the population level, clearly showing that hepcidin levels are lower in premenopausal vs postmenopausal women and are highly correlated with serum ferritin levels. In smaller studies, the within-subject variation of serum hepcidin was relatively high; hepcidin increased with prolonged fasting24  and showed both circadian rhythm and considerable (27%-50%) day-to-day variation.25  Hepcidin-25 values decrease within 1 to 2 days with storage at room temperature but are stable at 4°C, −20°C, and −80°C for at least 1 week, 4 weeks, and ∼2 years, respectively.8,26 

Like every hormone, hepcidin is under the influence of many different stimuli. Figure 1 summarizes the opposing effects exerted by a number of physiological and pathological conditions. Of note, the response is often quite rapid, making circulating hepcidin a very dynamic compartment. For example, hepcidin production increases substantially within a few hours after iron administration or inflammatory stimulation.15,28,29,33,34  Because many of the conditions listed in Figure 1 are common, several stimuli may be present simultaneously, with hepcidin output depending on the relative strength of each. For example, in severe ID, hepcidin production tends to be low, even in the presence of inflammation.46,47  Similarly, in conditions of ineffective/expanded erythropoiesis, like in non-transfusion-dependent thalassemias, signals released by bone marrow erythroid precursors tend to override those from replete iron stores.48  This results in relative hepcidin suppression in non-transfusion-dependent thalassemias,27,49,50  other iron-loading anemias,3,51  and even in β-thalassemia trait.50  One such erythroid signal, erythroferrone, has been recently identified in a mouse model.52  In line with this pathophysiological model, serum hepcidin fluctuates in transfusion-dependent β-thalassemia, with levels increasing soon after red blood cell transfusions (which suppress ineffective erythropoiesis) and declining during intertransfusional periods.53  Because of the highly dynamic and multifactorial regulation, a key practical message is that, in a given individual, the correct interpretation of hepcidin levels requires accurate knowledge of the overall clinical context (Table 18,19,22,23,25,26 )

Figure 1

Clinical conditions known to influence circulating hepcidin levels. Clinically relevant conditions include CKD,11,16  RBC transfusions,27  iron administration,28.29  replete iron stores,1 TMPRSS6 variants,30,31  infections/inflammatory disorders,32-34  ineffective erythropoiesis,3,49  hypoxia,35,36  administration of erythropoietic stimulating agents,37  chronic liver diseases,38  alcohol abuse,39  HCV,40  hemochromatosis-related mutations,1,28,41,42  and administration of the sex hormones testosterone43  and estrogens.44,45  CKD, chronic kidney disease; GFR, glomerular filtration rate; HCV, hepatitis C virus; HH, hereditary hemochromatosis; IDA, iron deficiency anemia; RBC, red blood cell; TMPRSS6 (transmembrane protease serine 6), the gene encoding for matriptase-2.

Figure 1

Clinical conditions known to influence circulating hepcidin levels. Clinically relevant conditions include CKD,11,16  RBC transfusions,27  iron administration,28.29  replete iron stores,1 TMPRSS6 variants,30,31  infections/inflammatory disorders,32-34  ineffective erythropoiesis,3,49  hypoxia,35,36  administration of erythropoietic stimulating agents,37  chronic liver diseases,38  alcohol abuse,39  HCV,40  hemochromatosis-related mutations,1,28,41,42  and administration of the sex hormones testosterone43  and estrogens.44,45  CKD, chronic kidney disease; GFR, glomerular filtration rate; HCV, hepatitis C virus; HH, hereditary hemochromatosis; IDA, iron deficiency anemia; RBC, red blood cell; TMPRSS6 (transmembrane protease serine 6), the gene encoding for matriptase-2.

Close modal
Table 1

Hepcidin measurement in clinical practice: a decalogue for the hematologist

CommentsReference
Checklist before ordering the assay   
 1. Ensure local availability of a validated assay See text and supplemental Table 1 19 
 2. Ensure control of preanalytical conditions (including diurnal rhythm) See text 8, 25, 26 
 3. Refer to age- and sex-specific ranges Significant differences between males and females, particularly during fertile period 22, 23 
 4. Interpret hepcidin value into a minimum laboratory context (CBC, ferritin, transferrin saturation, CRP, serum creatinine, and liver function tests) See Figure 1 — 
 5. Be aware of any potential confounders/comorbidities in the individual patient See Figure 1 — 
Most promising applications   
 6. Evaluation of suspected IRIDA Virtually diagnostic in an appropriate clinical context 54, 55 
 7. Evaluation of IO disorders For example, ferroportin disease due to hepcidin resistant mutations (see text) 41, 42, 49, 51, 56, 57 
 8. Diagnosis of concomitant ID in patients with ACD Promising reports in rheumatoid arthritis and inflammatory bowel disease patients, and in African children 32, 58-60 
 9. Guide for iron therapy For example, selection of patients for direct IV supplementation; oral administration in children from developing countries with high prevalence of infectious diseases (see text) 6, 32, 58, 61-63 
 10. Monitoring of treatments targeting the hepcidin/ferroportin axis To be confirmed by further studies 64 
CommentsReference
Checklist before ordering the assay   
 1. Ensure local availability of a validated assay See text and supplemental Table 1 19 
 2. Ensure control of preanalytical conditions (including diurnal rhythm) See text 8, 25, 26 
 3. Refer to age- and sex-specific ranges Significant differences between males and females, particularly during fertile period 22, 23 
 4. Interpret hepcidin value into a minimum laboratory context (CBC, ferritin, transferrin saturation, CRP, serum creatinine, and liver function tests) See Figure 1 — 
 5. Be aware of any potential confounders/comorbidities in the individual patient See Figure 1 — 
Most promising applications   
 6. Evaluation of suspected IRIDA Virtually diagnostic in an appropriate clinical context 54, 55 
 7. Evaluation of IO disorders For example, ferroportin disease due to hepcidin resistant mutations (see text) 41, 42, 49, 51, 56, 57 
 8. Diagnosis of concomitant ID in patients with ACD Promising reports in rheumatoid arthritis and inflammatory bowel disease patients, and in African children 32, 58-60 
 9. Guide for iron therapy For example, selection of patients for direct IV supplementation; oral administration in children from developing countries with high prevalence of infectious diseases (see text) 6, 32, 58, 61-63 
 10. Monitoring of treatments targeting the hepcidin/ferroportin axis To be confirmed by further studies 64 

ACD, anemia of chronic disease; CBC, complete blood count; CRP, C-reactive protein; ID, iron deficiency; IO, iron overload; IRIDA, iron-refractory iron deficiency anemia.

Diagnosis of IRIDA

This genetic disease, due to mutations in the hepcidin inhibitor TMPRSS6 (encoding matriptase-2),30  is characterized by IDA with inappropriately normal or high hepcidin levels.54,55  Thus, oral iron is ineffective and parenteral administration is needed to achieve at least a partial response. In classical IDA, hepcidin is suppressed below the limit of detection in biological fluids.15,19  By contrast, detection of pseudo-normal/elevated serum hepcidin in an appropriate clinical context can be considered virtually diagnostic of IRIDA, even without confirmation by TMPRSS6 sequencing (eg, in a young patient with unexplained IDA not responding to oral iron and with positive family history). Measurement of hepcidin may also help in the diagnosis of other atypical microcytic anemias due to rare genetic disorders of iron metabolism or heme synthesis (reviewed in Donker et al55 ), although data are presently insufficient (Table 1).

Diagnosis of IO disorders

Classical type 1 (HFE-related) HH is by far the commonest genetic disorder leading to IO in populations of northern European descent,65  but in other ethnicities, “atypical” IO accounts for up to 35% to 40% of cases.66  In type 1 HH at diagnosis, serum hepcidin levels are typically in the low to normal range and inappropriate for the degree of IO (manifesting as a low hepcidin-to-ferritin ratio).41  Hepcidin is further suppressed after normalization of iron stores by phlebotomies15,41  and shows a blunted response to oral iron.28  This blunted response is also seen in type 3 (TFR2-related) HH,28  whereas in type 2A/B or “juvenile” hemochromatosis (HJV or HAMP related, respectively), hepcidin levels are markedly suppressed/undetectable.19,42  By contrast, in type 4B HH or atypical ferroportin disease due to mutations in SLC40A1 conferring partial or complete resistance to hepcidin,67  serum hepcidin levels are substantially increased.56  Some data support using hepcidin measurement as a guide for the follow-up genetic testing of IO patients, particularly in populations in whom classical type 1 HFE-related HH is rare.57  Furthermore, during treatment of type 1 HH, monitoring hepcidin to prevent its complete suppression may curb iron hyperabsorption in the maintenance phase, possibly decreasing the need for phlebotomies. In iron-loading anemias, hepcidin measurement may also be valuable for identifying the most severely affected patients, helping to predict the development of IO and guiding the therapy.

Diagnosis and management of IDA

In IDA, hepcidin levels are generally suppressed to allow maximal iron absorption,15,19  but some patients, particularly the elderly,61  may have detectable hepcidin levels because of comorbidities like renal, inflammatory, or neoplastic diseases. Because hepcidin directly controls iron absorption,4,5  serum hormone levels have the potential to predict poor responsiveness to oral iron, preventing possible detrimental effects of oral iron on the gut microbiome and metabolome68  and eliminating delays before switching to IV iron. The usefulness of measuring basal hepcidin to personalize the optimal route of iron administration has been recently showed in patients with IDA,62  chronic rheumatic anemia,32  and chemotherapy-associated anemia.63  However, large prospective trials are needed to confirm this attractive hypothesis. Hepcidin has also proved effective for detecting ID in blood donors,69  the prevention of which is a major concern for blood services.

Notably, determination of serum hepcidin may help to solve a major global health problem; that is, the appropriateness of oral iron supplementation in children from regions with high infection burden. ID is highly prevalent in these regions and affects children’s physical growth and cognitive performance.70  Nonetheless, systematic iron supplementation has been associated with serious adverse outcomes, including increased mortality.71-73  Such detrimental effects have been mainly attributed to increased vulnerability to malaria and microbial agents, which are engaged in the host–pathogen battle for essential iron.68,74,75  Hepcidin capability to integrate competing signals (anemia, ID, and infection) makes its measurement promising in this setting. Indeed, 2 recent elegant studies using iron isotopes showed that low serum hepcidin was a good predictor of erythrocyte iron incorporation in African children, and suggest hepcidin as a guide for distinguishing individuals “ready to receive iron” from those in whom it should be avoided.6,58  This fascinating hypothesis, requiring the development of a validated point-of-care hepcidin assay, is actively under evaluation. However, because iron-induced reticulocytosis could increase susceptibility to malaria,76  and low hepcidin values do not necessarily exclude a concurrent infection,46  safe iron supplementation in malaria-endemic areas should entail not only the ability to absorb and incorporate iron but also effective measures to prevent exacerbations of infections.

Distinction between ID and ACD

ACD is a common condition with complex pathogenesis.77  Hepcidin-driven iron maldistribution plays a substantial role because iron is trapped in macrophages and less available for erythropoiesis. Patients with chronic inflammatory disorders are also at risk of developing ID. For example, occult/overt blood loss frequently occurs in conditions like inflammatory bowel diseases, chronic hemodialysis, and concomitant use of nonsteroidal anti-inflammatory drugs or antithrombotic drugs. In such settings, ID is difficult to detect using traditional iron biomarkers,78  but hepcidin may be helpful. Patients with inflammatory disorders and concomitant ID typically have lower hepcidin levels as compared with those with “pure” ACD.59  Indeed, hepcidin has proved effective in distinguishing IDA from ACD in patients with rheumatoid arthritis,32  inflammatory bowel diseases,60  cancer-related anemia,79  and critical illnesses,47  as well as in African children.58  This, again, can help in personalizing iron therapy to avoid delays or unnecessary/harmful treatment. However, because of the highly variable cutoff reported with different assays, harmonization of hepcidin assays is necessary before giving practical recommendations. Supplemental Table 2 shows examples of the diagnostic performance of hepcidin (eg, area under the curve of receiver operating characteristic, sensitivity, and specificity) in different clinical settings.

Companion diagnostic for novel therapies

Hepcidin discovery has opened unprecedented therapeutic opportunities for iron disorders. Hepcidin pharmacology is an active field with a number of drugs in the pipeline, including hepcidin agonists for the treatment of IO, and hepcidin antagonists for the treatment of iron restriction in ACD (reviewed in Ruchala and Nemeth64  and Camaschella80 ). Measuring hepcidin levels is anticipated to be helpful both in selecting patients and in monitoring the effects of novel targeted therapies.

Hepcidin is a promising tool to be added to the present battery of diagnostic tests for iron status, especially in ID, because it has the potential to differentiate from ACD and to inform about the ability to respond to oral iron. However, full inclusion in clinical practice and public health requires further efforts to harmonize the assays, assess the relevance of measuring specific hepcidin isoforms, define clinical decision limits, and make validated assays universally available.

The online version of this article contains a data supplement.

Contribution: D.G., E.N., and D.W.S. conceived of and cowrote the manuscript.

Conflict-of-interest disclosure: D.W.S. is an employee of Radboud University Medical Center, which offers high-quality hepcidin measurements to the medical, scientific, and pharmaceutical communities via the www.hepcidinanalysis.com initiative on a fee-for-service basis. E.N. is a consultant and stock holder for Intrinsic LifeSciences, Merganser Biotech, and Silarus Therapeutics. D.G. declares no competing financial interests.

Correspondence: Domenico Girelli, Department of Medicine, University of Verona, Veneto Region Referral Center for Iron Metabolism Disorders, Azienda Ospedaliera Universitaria Integrata Verona, Policlinico G.B. Rossi, 37134 Verona, Italy; e-mail: domenico.girelli@univr.it; and Dorine W. Swinkels, Department of Laboratory Medicine, Translational Metabolic Laboratory 830, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands; e-mail: dorine.swinkels@radboudumc.nl.

1
Ganz
 
T
Hepcidin and iron regulation, 10 years later.
Blood
2011
, vol. 
117
 
17
(pg. 
4425
-
4433
)
2
Hentze
 
MW
Muckenthaler
 
MU
Galy
 
B
Camaschella
 
C
Two to tango: regulation of mammalian iron metabolism.
Cell
2010
, vol. 
142
 
1
(pg. 
24
-
38
)
3
Kautz
 
L
Nemeth
 
E
Molecular liaisons between erythropoiesis and iron metabolism.
Blood
2014
, vol. 
124
 
4
(pg. 
479
-
482
)
4
Young
 
MF
Glahn
 
RP
Ariza-Nieto
 
M
, et al. 
Serum hepcidin is significantly associated with iron absorption from food and supplemental sources in healthy young women.
Am J Clin Nutr
2009
, vol. 
89
 
2
(pg. 
533
-
538
)
5
Roe
 
MA
Collings
 
R
Dainty
 
JR
Swinkels
 
DW
Fairweather-Tait
 
SJ
Plasma hepcidin concentrations significantly predict interindividual variation in iron absorption in healthy men.
Am J Clin Nutr
2009
, vol. 
89
 
4
(pg. 
1088
-
1091
)
6
Prentice
 
AM
Doherty
 
CP
Abrams
 
SA
, et al. 
Hepcidin is the major predictor of erythrocyte iron incorporation in anemic African children.
Blood
2012
, vol. 
119
 
8
(pg. 
1922
-
1928
)
7
Campostrini
 
N
Traglia
 
M
Martinelli
 
N
, et al. 
Serum levels of the hepcidin-20 isoform in a large general population: the Val Borbera study.
J Proteomics
2012
, vol. 
76
 
Spec No
(pg. 
28
-
35
)
8
Laarakkers
 
CM
Wiegerinck
 
ET
Klaver
 
S
, et al. 
Improved mass spectrometry assay for plasma hepcidin: detection and characterization of a novel hepcidin isoform.
PLoS One
2013
, vol. 
8
 
10
pg. 
e75518
 
9
Moe
 
MK
Hardang
 
IM
Hagve
 
TA
Novel circulating isoforms of hepcidin.
Clin Chem
2013
, vol. 
59
 
9
(pg. 
1412
-
1414
)
10
Tomosugi
 
N
Kawabata
 
H
Wakatabe
 
R
, et al. 
Detection of serum hepcidin in renal failure and inflammation by using ProteinChip System.
Blood
2006
, vol. 
108
 
4
(pg. 
1381
-
1387
)
11
Peters
 
HP
Laarakkers
 
CM
Swinkels
 
DW
Wetzels
 
JF
Serum hepcidin-25 levels in patients with chronic kidney disease are independent of glomerular filtration rate.
Nephrol Dial Transplant
2010
, vol. 
25
 
3
(pg. 
848
-
853
)
12
Peslova
 
G
Petrak
 
J
Kuzelova
 
K
, et al. 
Hepcidin, the hormone of iron metabolism, is bound specifically to alpha-2-macroglobulin in blood.
Blood
2009
, vol. 
113
 
24
(pg. 
6225
-
6236
)
13
Itkonen
 
O
Stenman
 
UH
Parkkinen
 
J
Soliymani
 
R
Baumann
 
M
Hämäläinen
 
E
Binding of hepcidin to plasma proteins.
Clin Chem
2012
, vol. 
58
 
7
(pg. 
1158
-
1160
)
14
Kemna
 
EH
Tjalsma
 
H
Podust
 
VN
Swinkels
 
DW
Mass spectrometry-based hepcidin measurements in serum and urine: analytical aspects and clinical implications.
Clin Chem
2007
, vol. 
53
 
4
(pg. 
620
-
628
)
15
Ganz
 
T
Olbina
 
G
Girelli
 
D
Nemeth
 
E
Westerman
 
M
Immunoassay for human serum hepcidin.
Blood
2008
, vol. 
112
 
10
(pg. 
4292
-
4297
)
16
Peters
 
HP
Laarakkers
 
CM
Pickkers
 
P
, et al. 
Tubular reabsorption and local production of urine hepcidin-25.
BMC Nephrol
2013
, vol. 
14
 pg. 
70
 
17
Ho
 
J
Reslerova
 
M
Gali
 
B
, et al. 
Urinary hepcidin-25 and risk of acute kidney injury following cardiopulmonary bypass.
Clin J Am Soc Nephrol
2011
, vol. 
6
 
10
(pg. 
2340
-
2346
)
18
Hunter
 
HN
Fulton
 
DB
Ganz
 
T
Vogel
 
HJ
The solution structure of human hepcidin, a peptide hormone with antimicrobial activity that is involved in iron uptake and hereditary hemochromatosis.
J Biol Chem
2002
, vol. 
277
 
40
(pg. 
37597
-
37603
)
19
Kroot
 
JJ
Laarakkers
 
CM
Geurts-Moespot
 
AJ
, et al. 
Immunochemical and mass-spectrometry-based serum hepcidin assays for iron metabolism disorders.
Clin Chem
2010
, vol. 
56
 
10
(pg. 
1570
-
1579
)
20
Kroot
 
JJ
van Herwaarden
 
AE
Tjalsma
 
H
Jansen
 
RT
Hendriks
 
JC
Swinkels
 
DW
Second round robin for plasma hepcidin methods: first steps toward harmonization.
Am J Hematol
2012
, vol. 
87
 
10
(pg. 
977
-
983
)
21
van der Vorm
 
LN
Hendriks
 
JCM
Laarakkers
 
CM
, et al. 
Towards worldwide hepcidin assay harmonization: identification of a commutable secondary reference material.
Clin Chem
 
In press
22
Galesloot
 
TE
Vermeulen
 
SH
Geurts-Moespot
 
AJ
, et al. 
Serum hepcidin: reference ranges and biochemical correlates in the general population.
Blood
2011
, vol. 
117
 
25
(pg. 
e218
-
e225
)
23
Traglia
 
M
Girelli
 
D
Biino
 
G
, et al. 
Association of HFE and TMPRSS6 genetic variants with iron and erythrocyte parameters is only in part dependent on serum hepcidin concentrations.
J Med Genet
2011
, vol. 
48
 
9
(pg. 
629
-
634
)
24
Troutt
 
JS
Rudling
 
M
Persson
 
L
, et al. 
Circulating human hepcidin-25 concentrations display a diurnal rhythm, increase with prolonged fasting, and are reduced by growth hormone administration.
Clin Chem
2012
, vol. 
58
 
8
(pg. 
1225
-
1232
)
25
Kroot
 
JJ
Hendriks
 
JC
Laarakkers
 
CM
, et al. 
(Pre)analytical imprecision, between-subject variability, and daily variations in serum and urine hepcidin: implications for clinical studies.
Anal Biochem
2009
, vol. 
389
 
2
(pg. 
124
-
129
)
26
Itkonen
 
O
Parkkinen
 
J
Stenman
 
UH
Hamalainen
 
E
 
Preanalytical factors and reference intervals for serum hepcidin LC-MS/MS method. Clin Chim Acta. 2012;413(7-8):696-701
27
Origa
 
R
Galanello
 
R
Ganz
 
T
, et al. 
Liver iron concentrations and urinary hepcidin in beta-thalassemia.
Haematologica
2007
, vol. 
92
 
5
(pg. 
583
-
588
)
28
Girelli
 
D
Trombini
 
P
Busti
 
F
, et al. 
A time course of hepcidin response to iron challenge in patients with HFE and TFR2 hemochromatosis.
Haematologica
2011
, vol. 
96
 
4
(pg. 
500
-
506
)
29
Moretti
 
D
Goede
 
JS
Zeder
 
C
, et al. 
Oral iron supplements increase hepcidin and decrease iron absorption from daily or twice-daily doses in iron-depleted young women.
Blood
2015
, vol. 
126
 
17
(pg. 
1981
-
1989
)
30
Finberg
 
KE
Heeney
 
MM
Campagna
 
DR
, et al. 
Mutations in TMPRSS6 cause iron-refractory iron deficiency anemia (IRIDA).
Nat Genet
2008
, vol. 
40
 
5
(pg. 
569
-
571
)
31
Nai
 
A
Pagani
 
A
Silvestri
 
L
, et al. 
TMPRSS6 rs855791 modulates hepcidin transcription in vitro and serum hepcidin levels in normal individuals.
Blood
2011
, vol. 
118
 
16
(pg. 
4459
-
4462
)
32
van Santen
 
S
van Dongen-Lases
 
EC
de Vegt
 
F
, et al. 
Hepcidin and hemoglobin content parameters in the diagnosis of iron deficiency in rheumatoid arthritis patients with anemia.
Arthritis Rheum
2011
, vol. 
63
 
12
(pg. 
3672
-
3680
)
33
Nemeth
 
E
Rivera
 
S
Gabayan
 
V
, et al. 
IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin.
J Clin Invest
2004
, vol. 
113
 
9
(pg. 
1271
-
1276
)
34
Kemna
 
E
Pickkers
 
P
Nemeth
 
E
van der Hoeven
 
H
Swinkels
 
D
Time-course analysis of hepcidin, serum iron, and plasma cytokine levels in humans injected with LPS.
Blood
2005
, vol. 
106
 
5
(pg. 
1864
-
1866
)
35
Piperno
 
A
Galimberti
 
S
Mariani
 
R
, et al. 
HIGHCARE investigators
Modulation of hepcidin production during hypoxia-induced erythropoiesis in humans in vivo: data from the HIGHCARE project.
Blood
2011
, vol. 
117
 
10
(pg. 
2953
-
2959
)
36
Goetze
 
O
Schmitt
 
J
Spliethoff
 
K
, et al. 
Adaptation of iron transport and metabolism to acute high-altitude hypoxia in mountaineers.
Hepatology
2013
, vol. 
58
 
6
(pg. 
2153
-
2162
)
37
Robach
 
P
Recalcati
 
S
Girelli
 
D
, et al. 
Serum hepcidin levels and muscle iron proteins in humans injected with low- or high-dose erythropoietin.
Eur J Haematol
2013
, vol. 
91
 
1
(pg. 
74
-
84
)
38
Tan
 
TC
Crawford
 
DH
Franklin
 
ME
, et al. 
 
The serum hepcidin:ferritin ratio is a potential biomarker for cirrhosis. Liver Int. 2012;32(9):1391-1399
39
Dostalikova-Cimburova
 
M
Balusikova
 
K
Kratka
 
K
, et al. 
Role of duodenal iron transporters and hepcidin in patients with alcoholic liver disease.
J Cell Mol Med
2014
, vol. 
18
 
9
(pg. 
1840
-
1850
)
40
Girelli
 
D
Pasino
 
M
Goodnough
 
JB
, et al. 
Reduced serum hepcidin levels in patients with chronic hepatitis C.
J Hepatol
2009
, vol. 
51
 
5
(pg. 
845
-
852
)
41
van Dijk
 
BA
Laarakkers
 
CM
Klaver
 
SM
, et al. 
Serum hepcidin levels are innately low in HFE-related haemochromatosis but differ between C282Y-homozygotes with elevated and normal ferritin levels.
Br J Haematol
2008
, vol. 
142
 
6
(pg. 
979
-
985
)
42
Papanikolaou
 
G
Tzilianos
 
M
Christakis
 
JI
, et al. 
Hepcidin in iron overload disorders.
Blood
2005
, vol. 
105
 
10
(pg. 
4103
-
4105
)
43
Bachman
 
E
Feng
 
R
Travison
 
T
, et al. 
Testosterone suppresses hepcidin in men: a potential mechanism for testosterone-induced erythrocytosis.
J Clin Endocrinol Metab
2010
, vol. 
95
 
10
(pg. 
4743
-
4747
)
44
Yang
 
Q
Jian
 
J
Katz
 
S
Abramson
 
SB
Huang
 
X
17β-Estradiol inhibits iron hormone hepcidin through an estrogen responsive element half-site.
Endocrinology
2012
, vol. 
153
 
7
(pg. 
3170
-
3178
)
45
Lehtihet
 
M
Bonde
 
Y
Beckman
 
L
, et al. 
Circulating hepcidin-25 is reduced by endogenous estrogen in humans.
PLoS One
2016
, vol. 
11
 
2
pg. 
e0148802
 
46
Jonker
 
FA
Calis
 
JC
Phiri
 
K
, et al. 
Low hepcidin levels in severely anemic malawian children with high incidence of infectious diseases and bone marrow iron deficiency.
PLoS One
2013
, vol. 
8
 
12
pg. 
e78964
 
47
Lasocki
 
S
Baron
 
G
Driss
 
F
, et al. 
Diagnostic accuracy of serum hepcidin for iron deficiency in critically ill patients with anemia.
Intensive Care Med
2010
, vol. 
36
 
6
(pg. 
1044
-
1048
)
48
Kemna
 
EH
Kartikasari
 
AE
van Tits
 
LJ
Pickkers
 
P
Tjalsma
 
H
Swinkels
 
DW
Regulation of hepcidin: insights from biochemical analyses on human serum samples.
Blood Cells Mol Dis
2008
, vol. 
40
 
3
(pg. 
339
-
346
)
49
Origa
 
R
Cazzola
 
M
Mereu
 
E
, et al. 
Differences in the erythropoiesis-hepcidin-iron store axis between hemoglobin H disease and β-thalassemia intermedia.
Haematologica
2015
, vol. 
100
 
5
(pg. 
e169
-
e171
)
50
Jones
 
E
Pasricha
 
SR
Allen
 
A
, et al. 
Hepcidin is suppressed by erythropoiesis in hemoglobin E β-thalassemia and β-thalassemia trait.
Blood
2015
, vol. 
125
 
5
(pg. 
873
-
880
)
51
Nemeth
 
E
Ganz
 
T
Hepcidin and iron-loading anemias.
Haematologica
2006
, vol. 
91
 
6
(pg. 
727
-
732
)
52
Kautz
 
L
Jung
 
G
Valore
 
EV
Rivella
 
S
Nemeth
 
E
Ganz
 
T
Identification of erythroferrone as an erythroid regulator of iron metabolism.
Nat Genet
2014
, vol. 
46
 
7
(pg. 
678
-
684
)
53
Pasricha
 
SR
Frazer
 
DM
Bowden
 
DK
Anderson
 
GJ
Transfusion suppresses erythropoiesis and increases hepcidin in adult patients with β-thalassemia major: a longitudinal study.
Blood
2013
, vol. 
122
 
1
(pg. 
124
-
133
)
54
De Falco
 
L
Silvestri
 
L
Kannengiesser
 
C
, et al. 
Functional and clinical impact of novel TMPRSS6 variants in iron-refractory iron-deficiency anemia patients and genotype-phenotype studies.
Hum Mutat
2014
, vol. 
35
 
11
(pg. 
1321
-
1329
)
55
Donker
 
AE
Raymakers
 
RA
Vlasveld
 
LT
, et al. 
Practice guidelines for the diagnosis and management of microcytic anemias due to genetic disorders of iron metabolism or heme synthesis.
Blood
2014
, vol. 
123
 
25
(pg. 
3873
-
3886, quiz 4005
)
56
Sham
 
RL
Phatak
 
PD
Nemeth
 
E
Ganz
 
T
Hereditary hemochromatosis due to resistance to hepcidin: high hepcidin concentrations in a family with C326S ferroportin mutation.
Blood
2009
, vol. 
114
 
2
(pg. 
493
-
494
)
57
Kaneko
 
Y
Miyajima
 
H
Piperno
 
A
, et al. 
Measurement of serum hepcidin-25 levels as a potential test for diagnosing hemochromatosis and related disorders.
J Gastroenterol
2010
, vol. 
45
 
11
(pg. 
1163
-
1171
)
58
Pasricha
 
SR
Atkinson
 
SH
Armitage
 
AE
, et al. 
Expression of the iron hormone hepcidin distinguishes different types of anemia in African children.
Sci Transl Med
2014
, vol. 
6
 
235
pg. 
235re3
 
59
Theurl
 
I
Aigner
 
E
Theurl
 
M
, et al. 
Regulation of iron homeostasis in anemia of chronic disease and iron deficiency anemia: diagnostic and therapeutic implications.
Blood
2009
, vol. 
113
 
21
(pg. 
5277
-
5286
)
60
Bergamaschi
 
G
Di Sabatino
 
A
Albertini
 
R
, et al. 
Serum hepcidin in inflammatory bowel diseases: biological and clinical significance.
Inflamm Bowel Dis
2013
, vol. 
19
 
10
(pg. 
2166
-
2172
)
61
Busti
 
F
Campostrini
 
N
Martinelli
 
N
Girelli
 
D
Iron deficiency in the elderly population, revisited in the hepcidin era.
Front Pharmacol
2014
, vol. 
5
 pg. 
83
 
62
Bregman
 
DB
Morris
 
D
Koch
 
TA
He
 
A
Goodnough
 
LT
Hepcidin levels predict nonresponsiveness to oral iron therapy in patients with iron deficiency anemia.
Am J Hematol
2013
, vol. 
88
 
2
(pg. 
97
-
101
)
63
Steensma
 
DP
Sasu
 
BJ
Sloan
 
JA
Tomita
 
DK
Loprinzi
 
CL
Serum hepcidin levels predict response to intravenous iron and darbepoetin in chemotherapy-associated anemia.
Blood
2015
, vol. 
125
 
23
(pg. 
3669
-
3671
)
64
Ruchala
 
P
Nemeth
 
E
The pathophysiology and pharmacology of hepcidin.
Trends Pharmacol Sci
2014
, vol. 
35
 
3
(pg. 
155
-
161
)
65
Fleming
 
RE
Ponka
 
P
Iron overload in human disease.
N Engl J Med
2012
, vol. 
366
 
4
(pg. 
348
-
359
)
66
McDonald
 
CJ
Wallace
 
DF
Crawford
 
DH
Subramaniam
 
VN
Iron storage disease in Asia-Pacific populations: the importance of non-HFE mutations.
J Gastroenterol Hepatol
2013
, vol. 
28
 
7
(pg. 
1087
-
1094
)
67
Drakesmith
 
H
Nemeth
 
E
Ganz
 
T
Ironing out Ferroportin.
Cell Metab
2015
, vol. 
22
 
5
(pg. 
777
-
787
)
68
Jaeggi
 
T
Kortman
 
GA
Moretti
 
D
, et al. 
Iron fortification adversely affects the gut microbiome, increases pathogen abundance and induces intestinal inflammation in Kenyan infants.
Gut
2015
, vol. 
64
 
5
(pg. 
731
-
742
)
69
Pasricha
 
SR
McQuilten
 
Z
Westerman
 
M
, et al. 
Serum hepcidin as a diagnostic test of iron deficiency in premenopausal female blood donors.
Haematologica
2011
, vol. 
96
 
8
(pg. 
1099
-
1105
)
70
Zimmermann
 
MB
Hurrell
 
RF
Nutritional iron deficiency.
Lancet
2007
, vol. 
370
 
9586
(pg. 
511
-
520
)
71
Sazawal
 
S
Black
 
RE
Ramsan
 
M
, et al. 
Effects of routine prophylactic supplementation with iron and folic acid on admission to hospital and mortality in preschool children in a high malaria transmission setting: community-based, randomised, placebo-controlled trial.
Lancet
2006
, vol. 
367
 
9505
(pg. 
133
-
143
)
72
Soofi
 
S
Cousens
 
S
Iqbal
 
SP
, et al. 
Effect of provision of daily zinc and iron with several micronutrients on growth and morbidity among young children in Pakistan: a cluster-randomised trial.
Lancet
2013
, vol. 
382
 
9886
(pg. 
29
-
40
)
73
Zlotkin
 
S
Newton
 
S
Aimone
 
AM
, et al. 
Effect of iron fortification on malaria incidence in infants and young children in Ghana: a randomized trial.
JAMA
2013
, vol. 
310
 
9
(pg. 
938
-
947
)
74
Drakesmith
 
H
Prentice
 
AM
Hepcidin and the iron-infection axis.
Science
2012
, vol. 
338
 
6108
(pg. 
768
-
772
)
75
Ganz
 
T
Nemeth
 
E
Iron homeostasis in host defence and inflammation.
Nat Rev Immunol
2015
, vol. 
15
 
8
(pg. 
500
-
510
)
76
Clark
 
MA
Goheen
 
MM
Fulford
 
A
, et al. 
Host iron status and iron supplementation mediate susceptibility to erythrocytic stage Plasmodium falciparum.
Nat Commun
2014
, vol. 
5
 pg. 
4446
 
77
Weiss
 
G
Anemia of chronic disorders: new diagnostic tools and new treatment strategies.
Semin Hematol
2015
, vol. 
52
 
4
(pg. 
313
-
320
)
78
Brugnara
 
C
Adamson
 
J
Auerbach
 
M
Kane
 
R
Macdougall
 
I
Mast
 
A
Iron deficiency: what are the future trends in diagnostics and therapeutics?
Clin Chem
2013
, vol. 
59
 
5
(pg. 
740
-
745
)
79
Shu
 
T
Jing
 
C
Lv
 
Z
Xie
 
Y
Xu
 
J
Wu
 
J
Hepcidin in tumor-related iron deficiency anemia and tumor-related anemia of chronic disease: pathogenic mechanisms and diagnosis.
Eur J Haematol
2015
, vol. 
94
 
1
(pg. 
67
-
73
)
80
Camaschella
 
C
Treating iron overload.
N Engl J Med
2013
, vol. 
368
 
24
(pg. 
2325
-
2327
)

Supplemental data

Sign in via your Institution