Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Mar 1.
Published in final edited form as: Med Clin North Am. 2016 Dec 24;101(2):285–296. doi: 10.1016/j.mcna.2016.09.005

Anemia of Inflammation: A Review

Paula G Fraenkel 1,2,*
PMCID: PMC5308549  NIHMSID: NIHMS819021  PMID: 28189171

Abstract

Impaired iron homeostasis and the suppressive effects of proinflammatory cytokines on erythropoiesis, together with alterations of the erythrocyte membrane that impair its survival, cause the anemia of inflammation. Recent epidemiologic studies have connected inflammatory anemia with critical illness, obesity, aging, and kidney failure, as well as with cancer, chronic infection, and autoimmune disease. The proinflammatory cytokine, interleukin-6, the iron regulatory hormone, hepcidin, and the iron exporter, ferroportin, interact to cause iron sequestration in the setting of inflammation. While severe anemia is associated with adverse outcomes in critical illness, experimental models suggest that iron sequestration is part of a natural defense against pathogens. In animal models and human patients, experimental therapeutic approaches targeting interleukin-6 or the ferroportin-hepcidin axis have shown efficacy in reversing anemia, although these agents have not yet been approved for the treatment of the anemia of inflammation.

Keywords: anemia, inflammation, hepcidin, ferroportin, interleukin-6, erythroferrone, iron, cancer

EPIDEMIOLOGY

Inflammation is one of the most common causes of anemia in the elderly and chronically ill. In NHANES III, anemia of inflammation was defined as a low serum iron level (<10.74 µM or <60 µg/dL) without evidence of low iron stores, i.e. transferrin saturation >15%, serum ferritin >12 ng/mL, or erythrocyte protoporphyrin concentration >1.24 µM [1]. Other features of anemia of inflammation include inappropriately low levels of erythropoietin, and elevated measures of inflammatory markers, such as C-reactive protein [2]. In the National Health and Nutrition Examination Study (NHANES III), investigators discovered that about 1 million Americans over age 65 exhibit anemia related to inflammation.

Anemia in Critical Illness

The anemia of inflammation can occur in the setting of acute or chronic inflammation. The CRIT study of anemia and blood transfusion in the critically ill was an observational cohort analysis of 4892 patients in intensive care units across the United States [3]. The CRIT study revealed that anemia of inflammation develops in critically ill patients even within 30 days. The mean hemoglobin levels in critically ill individuals decreased over a 30-day period despite the administration of blood transfusions. The CRIT study also demonstrated that severe anemia, hemoglobin <9 g/dL, is an independent predictor of increased mortality and length of stay in critically ill patients.

Anemia in the elderly is frequently linked to inflammatory conditions. In a recent study of 191 consecutive hospitalized elderly patients with anemia, 70% of the patients were found to have anemia related to inflammation. Sixteen percent of the patients with anemia of inflammation had concomitant chronic renal failure [4]. Of the patients with inflammatory anemia, 71% were suffering from an acute infection, 12% had cancer, and 16% had a chronic infection, such as a pressure ulcer, or a chronic autoimmune inflammatory disease. [4]

Anemia in Obesity

As the prevalence of obesity is rising in the United States, increasing attention is being paid to the potential effect of obesity on erythropoiesis. Obese patients exhibit higher plasma levels of pro-inflammatory cytokines and acute phase reactants, as well as higher rates of iron-restricted erythropoiesis that can result in anemia [5]. Functional iron deficiency is defined as inappropriately low iron stores, despite the presence of inflammation, i.e. a normal serum ferritin (12–100 ng/mL for females or 15–100 ng/mL for males) and a serum C-reactive protein >3 mg/L [6][7] A recent cross-sectional study of 947 obese patients under evaluation for bariatric surgery revealed that 52.5% exhibited functional iron deficiency. The majority of obese patients with functional iron deficiency appear to have sequestration of iron as manifest by a serum transferrin saturation <20% [6]. Weight loss has been associated with an increase in transferrin saturation in overweight individuals [8], which supports the hypothesis that obesity causes iron sequestration.

Anemia in Cancer

Anemia of inflammation is a common manifestation of advanced cancer. A recent prospective, observational study of 888 patients with a variety of carcinomas revealed that 63.4% of the patients were anemic [9]. The prevalence and severity of anemia correlated with the stage of cancer [9]. The prevalence of increased mean plasma levels of markers of inflammation, including C-reactive protein, fibrinogen, IL-6, TNFα, IL-1β, ferritin, hepcidin, erythropoietin and reactive oxygen species, was significantly increased in advanced stage compared to early stage cancer patients. Supporting a role for inflammation in iron sequestration, serum iron levels were significantly reduced in advanced stage patients [9].

PATHOPHYSIOLOGY

Recent discoveries indicate that both iron sequestration and impaired erythropoiesis cause the anemia of inflammation. Erythroid progenitors mature to erythrocytes through a series of stages that require co-ordination of iron acquisition and cell proliferation. As erythroid progenitors mature to the polychromatophilic stage, transferrin receptor 1 expression on the surface of the red cell membrane increases [10]. Macrophages export iron via ferroportin (fpn). The carrier protein, transferrin, binds free iron with high avidity. Transferrin-bound iron attaches to transferrin receptor 1. In an acidified vacuole, the iron is released from transferrin and exported to the cytoplasm by divalent metal transporter 1 (DMT1), while transferrin receptor is recycled to the cell surface (reviewed in [11]). The iron then enters the mitochondria where it is attached to protoporphyrin IX in the final step of heme synthesis.

Iron is effectively salvaged in the human body. Macrophages consume senescent erythrocytes, liberate iron from hemoglobin, and store the iron in ferritin for subsequent release of iron to developing erythrocytes via fpn.[10] Studies of the fpn-deficient zebrafish, weissherbst [12, 13], and the fpn-knockout mouse [14] revealed that fpn is required to export iron from macrophages to developing erythrocytes. In fpn-deficiency, iron remains sequestered in macrophages. This results in impaired iron delivery to developing erythrocytes and erythroid maturation arrest [12, 13].

Hepcidin is a short, cysteine-rich peptide hormone that regulates the intestinal absorption of iron and its release from macrophage iron stores. Human hepatocytes secrete the iron-regulatory peptide hormone, hepcidin [1517] in response to iron overload or inflammation. Hepcidin binds fpn on the surface of macrophages resulting in the internalization and degradation of both proteins [18] and sequestration of iron in macrophages, away from the developing erythrocytes (Figure 1). Hepcidin appears to be the key regulator of iron homeostasis, because loss of function mutations in genes that regulate Hepcidin expression, e.g. Transferrin receptor 2, HFE, Hemojuvelin, or in Hepcidin itself have each been associated with hereditary iron overload syndromes [19].

Figure 1.

Figure 1

Inflammation stimulates increased production of the iron-regulatory peptide, hepcidin, by hepatocytes and the pro-inflammatory cytokine, IL-6, which suppresses erythropoiesis. Hepcidin binds the iron exporter, ferroportin (fpn), causing internalization and degradation of both proteins and decreasing delivery of iron from macrophages to developing erythrocytes. This impairs erythroid development and leads to anemia. Increased erythropoietic drive stimulates erythroid progenitors to release erythroferrone, a hormone that suppresses hepcidin expression. When inflammation resolves, hepcidin and IL-6 levels decline, allowing iron to be exported to from macrophages to erythrocytes and promoting erythropoiesis.

Regulation of hepcidin

The BMP co-receptor, hemojuvelin, and TMPRSS6, [2026] participate in regulating hepcidin transcription in the setting of iron overload. Iron overload enhances bone morphogenic protein (BMP) signaling and Smad protein phosphorylation that enhances hepcidin transcription. On the other hand, iron deficiency or the acute onset of anemia stimulates release of the hormone, erythroferrone, by erythroid progenitors, and erythropoietin by the kidney [11, 27] Erythroferrone suppresses hepcidin expression to promote intestinal iron absorption and macrophage iron release, while erythropoietin stimulates proliferation of committed erythroid progenitors [11, 27].

Interleukin-6 (IL-6) and interleukin-1β (IL-1β) [28] are key cytokines that mediate the effects of inflammation on the developing erythrocyte. The transcription factors that mediate the effects of inflammation include Stat3, C/EBPα, and p53 [29, 30] (Figure 2). IL-6 increases JAK/Stat signaling [31, 32], which promotes phosphorylation of Stat3 and Stat3 binding to the Hepcidin promoter [30, 33]. IL-1β induces Hepcidin expression via the C/EBPα and BMP/SMAD signaling pathways. Hepatocyte damage, via endoplasmic reticulum stress or oxidation, enhances C/EBPα [34, 35] or Stat3 activity [36], respectively, leading to increased Hepcidin expression. Lipopolysaccharide (LPS), released by severe bacterial infection, activates toll-like receptor 4 (TLR4) signaling, which, in turn, enhances production of IL-6 [37] by macrophages that stimulates hepcidin expression. HMGB1, a proinflammatory cytokine that is produced in patients with critical illness, is associated with increased in-hospital mortality. HMGB1 has been shown to bind the MD2-TLR4 complex [38, 39] and to increase expression of TNF and IL-6, cytokines that impair erythropoiesis [40, 41]. Inhibition of HMGB1 activity ameliorates anemia of inflammation in mouse models [38]. Recent studies [42] indicate that inflammation in mouse models increases transcript levels of Activin B, a member of the TGF-β superfamily of signaling molecules. Activin B has been shown to increase hepcidin transcription in cultured hepatocytes in vitro via Smad signaling [42].

Figure 2.

Figure 2

Inflammation and hepatocyte damage augment Hepcidin transcription and iron sequestration via several pathways. Lipopolysaccharide (LPS), released by bacterial infection, and the proinflammatory cytokine, HMGB1, activate toll-like receptor 4 (TLR-4) signaling, which increases IL-6 release by macrophages, while leptin and obesity also promote IL-6 release. IL-6 signaling leads to phosphorylation of Stat3 and increased Stat3 binding to the Hepcidin promoter, while endoplasmic reticulum (ER) stress in hepatocytes promotes CEBP-α binding to the Hepcidin promoter. Bone morphogenic protein (BMP) or activin signaling via ligands, including BMP2,4,6, and 9 and Activin B, activate receptors, such as BMP receptor-I, causing Smad phosphorylation and Smad binding to the Hepcidin promoter, which is required for Hepcidin transcription. The BMP co-receptor, hemojuvelin (HJV) interacts with the BMP receptor to enhance BMP signaling. Inflammation or obesity also promotes macrophage release of lipocalin, which can interact with bacterial siderophores to sequester iron.

Iron sequestration

Hepcidin is not the only protein causing iron sequestration during bacterial infection. Recent studies indicate that stimulation of toll-like receptors 2 and 6 (TLR2 and TLR6) in mouse models decreases expression of fpn in macrophages and causes iron sequestration without increasing macrophage hepcidin transcript levels [43]. LPS stimulates macrophages to produce lipocalin 2, which sequesters iron by binding bacterially produced siderophores [44]. Furthermore, infection or inflammation stimulates neutrophil release of the iron binding protein, Lactoferrin. Lactoferrin is internalized by bacteria, sequesters iron, and arrests microbial growth [45].

Obese individuals exhibit increased plasma levels of pro-inflammatory cytokines, including leptin, hepcidin, and the iron sequestering protein, lipocalin-2. There are two proposed mechanisms by which obesity may contribute to functional iron deficiency and anemia, based on experimental models: (1) Leptin and pro-inflammatory cytokines stimulate hepcidin production in adipocytes and hepatocytes [46]; (2) Adipocytes and peripheral blood mononuclear cells in obese patients produce lipocalin 2, which restricts iron availability to developing erythroid cells [47].

Changes in erythrocyte membrane

In addition to effects on iron metabolism, inflammatory cytokines diminish erythropoietin synthesis, impair the differentiation of erythroid progenitors, and shorten the lifespan of mature red blood cells [48]. Incubation of erythrocytes from healthy donors with plasma from patients with septic shock induced hydrolysis of red blood cell membrane phosphatidyl choline to lysophosphatidyl choline and increased erythrocyte phosphatidyl serine exposure [49], changes that may shorten erythrocyte survival. The presence of renal failure can exacerbate the effects of inflammation. Uremia is associated with reduced levels of albumin, increased levels of nitric oxide, and changes in the levels of erythrocyte membrane proteins that promote the accumulation of reactive oxygen species and oxidation of erythrocyte membrane proteins (reviewed in [50]). These alterations are thought to accelerate erythrocyte destruction and tissue injury.

NEW EXPERIMENTAL MODELS

In order to develop new therapies for the anemia of inflammation, animal models are needed that resemble the findings in human patients. In one recent model, killed Brucella abortus (KBA) bacteria are injected once by intraperitoneal injection. [40, 51] Fourteen days later, mean murine hemoglobin levels decline by 50%.. Erythropoiesis gradually recovers following the injection, just as human patients may recover from the insult of a critical illness, such as sepsis. Hepcidin deficiency blunted the severity of anemia following injection of KBA, while IL-6 deficiency protected against hypoferremia and anemia in murine models. Hepcidin-knockout mice exhibited significantly impaired survival compared to wild type controls following injection of KBA [51], which supports the long-held theory that anemia of inflammation is protective in critical illness.

EXPERIMENTAL TREATMENTS

The best treatment for the anemia of inflammation is to eradicate the underlying disease. When that is not possible, transfusions, intravenous iron supplementation, and erythropoiesis stimulating agents [52] may ameliorate the condition. Newer, experimental approaches target IL-6 activity and the hepcidin-ferroportin axis. In 1993, researchers observed that treating patients with metastatic renal cell carcinoma with a murine anti-IL-6 antibody improved paraneoplastic thrombocytosis and anemia [53, 54]. These observations led to the evaluation of antibodies against IL-6 in human patients with inflammatory anemia secondary to Multicentric Castleman Disease (MCD).

Patients with MCD exhibit generalized lymphadenopathy, systemic chronic inflammation, increased IL-6 activity and anemia of inflammation [55]. Siltuximab, a human-mouse chimeric anti-IL-6 antibody has been shown to improve anemia in patients with this condition [56]. Siltuximab was subsequently evaluated in a Phase II trial in patients with transfusion-dependent, low or intermediate risk myelodysplastic syndrome, whose levels of pro-inflammatory cytokines are often elevated, however siltuximab failed to reduce transfusion dependence in these patients [57]. Tocilizumab, a humanized monoclonal antibody against the IL-6 receptor that has been approved to treat rheumatoid arthritis, has been found to reduce serum hepcidin levels and improve anemia in patients with Castleman disease and rheumatoid arthritis [52, 55].

Inhibitors of hepcidin production, blockers of hepcidin peptide activity, and antibodies to fpn have also been used to modulate the anemia of inflammation. A potent, orally available BMP receptor antagonist, LDN-193189, has been shown to decrease BMP signaling and reduce hepcidin mRNA levels in mice in a dose-dependent manner. Furthermore, LDN-193189 ameliorated the anemia of inflammation in mice that had been injected with turpentine to generate sterile abscesses [58]. Other efforts have focused on agents that are administered parenterally. NOX-H94, a structured L-oligoribonucleotide that binds human hepcidin with a high affinity, blocked IL-6 induced hypoferremia and moderated the development of anemia in cynomolgus monkey models [59]. In human trials, NOX-H94 prevented LPS-induced hypoferremia [60]. The human anti-hepcidin antibody, 12B9M, increased serum iron levels in cynomolgus monkeys, but has not been evaluated in a primate model of anemia of inflammation. [61] A Phase I trial to evaluate the effect of an antibody against ferroportin, LY2928057, has been completed, but the results are not yet available (clinicaltrials.gov). Table 1 summarizes other experimental therapies in development. Conversely, small molecules that increase hepcidin expression may be developed into treatments for iron overload [6264].

Table 1.

Agents with potential activity against the anemia of inflammation

Class of Agent Name Stage of
development
Route of
administration
Citation
Anti-IL-6 Elsilimomab
or BE-8
Decreased
thrombocytosis and
anemia in patients
with metastatic
renal cell
carcinoma.
Intravenous [53, 54]
Anti-IL-6 Siltuximab Approved to treat
Castleman disease;
Decreased anemia
in patients with
Castleman disease;
Not effective in
patients with early
stage
myelodysplastic
syndrome.
Intravenous [56, 57]
Anti-IL-6 Receptor Tocilizumab Approved to treat
rheumatoid arthritis;
Decreased anemia
in patients with
Castleman disease
Intravenous [55]
BMP receptor
antagonist
LDN-193189 Evaluated in mouse
models.
Oral [58]
Anti-hepcidin
oligoribonucleotide
Spiegelmer
NOX-H94
Evaluated in
cynomolgus
monkeys and Phase
I trial in humans
published.
Intravenous [59, 60]
Anti-hepcidin
antibody
12B9M Evaluated in
cynomolgus
monkeys.
Intravenous [61]
Anti-ferroportin
antibody
LY2928057 Phase I trial in
humans completed.
Results
unpublished.
Intravenous Clinicaltrials.gov
Anti-hemojuvelin
antibody
ABT-207 and
h5F9-AM8
Preclinical study in
rats.
Intravenous [66]
Soluble hemojuvelin
extracellular domain
fused with
immunoglobulin Fc
HJV.fc Preclinical study in
rats.
Intravenous [67]
siRNA against
hepatic EglN prolyl
hydroxylases
EglN1+2+3
siRNA lipid
nanoparticles
Preclinical study in
mice.
Intravenous [68]
Suppressor of
erythroid iron
restriction response,
bypasses effect of
aconitase
inactivation
Isocitrate Preclinical study in
rats.
Intravenous [69]
Inhibitor of activin
signaling, decreases
LPS or KBA-induced
hepcidin expression
in mice
Follistatin-
315
Preclinical study in
mice
Intraperitoneal [42]

Adapted from Fraenkel PG. Understanding anemia of chronic disease. Hematology Am Soc Hematol Educ Program 2015;14-8; with permission.

Our knowledge of the interaction between inflammation, iron metabolism, and erythropoiesis has improved our ability to understand the anemia of inflammation. While drugs have not yet been approved to treat this condition, several agents are under investigation, and some agents improve anemia of inflammation in patients with Castleman disease or rheumatoid arthritis. In some cases, the anemia of inflammation may be protective, for instance in animal models of the anemia of critical illness, hepcidin-deficient mice exhibited significantly lower rates of survival than wild type animals [51]. Thus the best course of action continues to be to identify and treat the underlying causes of the anemia of chronic disease.

KEY POINTS.

  • In the anemia of inflammation, proinflammatory cytokines suppress erythropoiesis, cause iron sequestration via effects on the iron regulatory hormone, hepcidin, and promote alteration in the erythrocyte cell membrane leading to shortened red blood cell survival.

  • In the setting of critical illness, severe anemia is associated with increased mortality.

  • Advanced age, cancer, rheumatologic diseases, chronic infection, and kidney failure are also associated with the anemia of inflammation.

  • Experimental therapies reverse the effects of the anemia of inflammation in animal models, but have not been approved for human use.

Acknowledgments

Dr. Fraenkel’s research is supported by the National Institutes of Health (R01 DK085250 and R01 GM083198). The funding agencies did not influence the design or interpretation of the studies.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Dr. Fraenkel has no relevant financial conflicts of interest.

REFERENCES

  • 1.Guralnik JM, et al. Prevalence of anemia in persons 65 years and older in the United States: evidence for a high rate of unexplained anemia. Blood. 2004;104(8):2263–2268. doi: 10.1182/blood-2004-05-1812. [DOI] [PubMed] [Google Scholar]
  • 2.van Iperen CE, et al. Response of erythropoiesis and iron metabolism to recombinant human erythropoietin in intensive care unit patients. Crit Care Med. 2000;28(8):2773–2778. doi: 10.1097/00003246-200008000-00015. [DOI] [PubMed] [Google Scholar]
  • 3.Corwin HL, et al. The CRIT Study: Anemia and blood transfusion in the critically ill--current clinical practice in the United States. Crit Care Med. 2004;32(1):39–52. doi: 10.1097/01.CCM.0000104112.34142.79. [DOI] [PubMed] [Google Scholar]
  • 4.Joosten E, Lioen P. Iron deficiency anemia and anemia of chronic disease in geriatric hospitalized patients: How frequent are comorbidities as an additional explanation for the anemia? Geriatr Gerontol Int. 2014 doi: 10.1111/ggi.12371. 10.1111/ggi.12371. [DOI] [PubMed] [Google Scholar]
  • 5.Coimbra S, Catarino C, Santos-Silva A. The role of adipocytes in the modulation of iron metabolism in obesity. Obes Rev. 2013;14(10):771–779. doi: 10.1111/obr.12057. [DOI] [PubMed] [Google Scholar]
  • 6.Careaga M, et al. Inflammation and iron status in bariatric surgery candidates. Surg Obes Relat Dis. 2015 doi: 10.1016/j.soard.2014.09.028. 10.1016/j.soard.2014.09.028. [DOI] [PubMed] [Google Scholar]
  • 7.Thomas DW, et al. Guideline for the laboratory diagnosis of functional iron deficiency. Br J Haematol. 2013;161(5):639–648. doi: 10.1111/bjh.12311. [DOI] [PubMed] [Google Scholar]
  • 8.Anty R, et al. Bariatric surgery can correct iron depletion in morbidly obese women: a link with chronic inflammation. Obes Surg. 2008;18(6):709–714. doi: 10.1007/s11695-007-9276-y. [DOI] [PubMed] [Google Scholar]
  • 9.Maccio A, et al. The role of inflammation, iron, and nutritional status in cancer-related anemia: results of a large, prospective, observational study. Haematologica. 2015;100(1):124–132. doi: 10.3324/haematol.2014.112813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Andrews NC. Forging a field: the golden age of iron biology. Blood. 2008;112(2):219–230. doi: 10.1182/blood-2007-12-077388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ganz T, Nemeth E. Iron Balance and the Role of Hepcidin in Chronic Kidney Disease. Semin Nephrol. 2016;36(2):87–93. doi: 10.1016/j.semnephrol.2016.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Donovan A, et al. Positional cloning of zebrafish ferroportin1 identifies a conserved vertebrate iron exporter. Nature. 2000;403(6771):776–781. doi: 10.1038/35001596. [DOI] [PubMed] [Google Scholar]
  • 13.Fraenkel PG, et al. Ferroportin1 is required for normal iron cycling in zebrafish. J Clin Invest. 2005;115(6):1532–1541. doi: 10.1172/JCI23780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Donovan A, et al. The iron exporter ferroportin/Slc40a1 is essential for iron homeostasis. Cell Metab. 2005;1(3):191–200. doi: 10.1016/j.cmet.2005.01.003. [DOI] [PubMed] [Google Scholar]
  • 15.Hentze MW, Muckenthaler MU, Andrews NC. Balancing acts: molecular control of mammalian iron metabolism. Cell. 2004;117(3):285–297. doi: 10.1016/s0092-8674(04)00343-5. [DOI] [PubMed] [Google Scholar]
  • 16.Pigeon C, et al. A new mouse liver-specific gene, encoding a protein homologous to human antimicrobial peptide hepcidin, is overexpressed during iron overload. J Biol Chem. 2001;276(11):7811–7819. doi: 10.1074/jbc.M008923200. [DOI] [PubMed] [Google Scholar]
  • 17.Bolondi G, et al. Altered hepatic BMP signaling pathway in human HFE hemochromatosis. Blood Cells Mol Dis. 2010;45(4):308–312. doi: 10.1016/j.bcmd.2010.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Nemeth E, et al. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science. 2004;306(5704):2090–2093. doi: 10.1126/science.1104742. [DOI] [PubMed] [Google Scholar]
  • 19.Ganz T, Nemeth E. Iron metabolism: interactions with normal and disordered erythropoiesis. Cold Spring Harb Perspect Med. 2012;2(5):a011668. doi: 10.1101/cshperspect.a011668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Arndt S, et al. Iron-induced expression of bone morphogenic protein 6 in intestinal cells is the main regulator of hepatic hepcidin expression in vivo. Gastroenterology. 2010;138(1):372–382. doi: 10.1053/j.gastro.2009.09.048. [DOI] [PubMed] [Google Scholar]
  • 21.Babitt JL, et al. Bone morphogenetic protein signaling by hemojuvelin regulates hepcidin expression. Nat Genet. 2006;38(5):531–539. doi: 10.1038/ng1777. [DOI] [PubMed] [Google Scholar]
  • 22.Gibert Y, et al. BMP signaling modulates hepcidin expression in zebrafish embryos independent of hemojuvelin. PLoS One. 2011;6(1):e14553. doi: 10.1371/journal.pone.0014553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Niederkofler V, Salie R, Arber S. Hemojuvelin is essential for dietary iron sensing, and its mutation leads to severe iron overload. J Clin Invest. 2005;115(8):2180–2186. doi: 10.1172/JCI25683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Zhang AS, et al. Evidence that inhibition of hemojuvelin shedding in response to iron is mediated through neogenin. J Biol Chem. 2007;282(17):12547–12556. doi: 10.1074/jbc.M608788200. [DOI] [PubMed] [Google Scholar]
  • 25.Finberg KE, et al. Mutations in TMPRSS6 cause iron-refractory iron deficiency anemia (IRIDA) Nat Genet. 2008;40(5):569–571. doi: 10.1038/ng.130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Melis MA, et al. A mutation in the TMPRSS6 gene, encoding a transmembrane serine protease that suppresses hepcidin production, in familial iron deficiency anemia refractory to oral iron. Haematologica. 2008;93(10):1473–1479. doi: 10.3324/haematol.13342. [DOI] [PubMed] [Google Scholar]
  • 27.Kautz L, et al. Identification of erythroferrone as an erythroid regulator of iron metabolism. Nat Genet. 2014;46(7):678–684. doi: 10.1038/ng.2996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lee P, et al. Regulation of hepcidin transcription by interleukin-1 and interleukin-6. Proc Natl Acad Sci U S A. 2005;102(6):1906–1910. doi: 10.1073/pnas.0409808102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Weizer-Stern O, et al. Hepcidin, a key regulator of iron metabolism, is transcriptionally activated by p53. Br J Haematol. 2007;138(2):253–262. doi: 10.1111/j.1365-2141.2007.06638.x. [DOI] [PubMed] [Google Scholar]
  • 30.Gkouvatsos K, Papanikolaou G, Pantopoulos K. Regulation of iron transport and the role of transferrin. Biochim Biophys Acta. 2012;1820(3):188–202. doi: 10.1016/j.bbagen.2011.10.013. [DOI] [PubMed] [Google Scholar]
  • 31.Kemna E, et al. Time-course analysis of hepcidin, serum iron, and plasma cytokine levels in humans injected with LPS. Blood. 2005;106(5):1864–1866. doi: 10.1182/blood-2005-03-1159. [DOI] [PubMed] [Google Scholar]
  • 32.Nemeth E, et al. IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J Clin Invest. 2004;113(9):1271–1276. doi: 10.1172/JCI20945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ganz T, Nemeth E. Hepcidin and iron homeostasis. Biochim Biophys Acta. 2012;1823(9):1434–1443. doi: 10.1016/j.bbamcr.2012.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Oliveira SJ, et al. ER stress-inducible factor CHOP affects the expression of hepcidin by modulating C/EBPalpha activity. PLoS One. 2009;4(8):e6618. doi: 10.1371/journal.pone.0006618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Vecchi C, et al. ER stress controls iron metabolism through induction of hepcidin. Science. 2009;325(5942):877–880. doi: 10.1126/science.1176639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Millonig G, et al. Sustained submicromolar H2O2 levels induce hepcidin via signal transducer and activator of transcription 3 (STAT3) J Biol Chem. 2012;287(44):37472–37482. doi: 10.1074/jbc.M112.358911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Constante M, et al. Distinct requirements for Hfe in basal and induced hepcidin levels in iron overload and inflammation. Am J Physiol Gastrointest Liver Physiol. 2006;291(2):G229–G237. doi: 10.1152/ajpgi.00092.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Valdes-Ferrer SI, et al. HMGB1 mediates anemia of inflammation in murine sepsis survivors. Mol Med. 2015 doi: 10.2119/molmed.2015.00243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Yang H, et al. MD-2 is required for disulfide HMGB1-dependent TLR4 signaling. J Exp Med. 2015;212(1):5–14. doi: 10.1084/jem.20141318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Gardenghi S, et al. Distinct roles for hepcidin and interleukin-6 in the recovery from anemia in mice injected with Heat-Killed Brucella abortus. Blood. 2014;123(8):1137–1145. doi: 10.1182/blood-2013-08-521625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Tracey KJ, et al. Cachectin/tumor necrosis factor induces cachexia, anemia, and inflammation. J Exp Med. 1988;167(3):1211–1227. doi: 10.1084/jem.167.3.1211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Canali S, et al. Activin B Induces Noncanonical SMAD1/5/8 Signaling via BMP Type I Receptors in Hepatocytes: Evidence for a Role in Hepcidin Induction by Inflammation in Male Mice. Endocrinology. 2016;157(3):1146–1162. doi: 10.1210/en.2015-1747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Guida C, et al. A novel inflammatory pathway mediating rapid hepcidin-independent hypoferremia. Blood. 2015;125(14):2265–2275. doi: 10.1182/blood-2014-08-595256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Flo TH, et al. Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron. Nature. 2004;432(7019):917–921. doi: 10.1038/nature03104. [DOI] [PubMed] [Google Scholar]
  • 45.Kanwar JR, et al. Multifunctional iron bound lactoferrin and nanomedicinal approaches to enhance its bioactive functions. Molecules. 2015;20(6):9703–9731. doi: 10.3390/molecules20069703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Chung B, et al. Leptin increases the expression of the iron regulatory hormone hepcidin in HuH7 human hepatoma cells. J Nutr. 2007;137(11):2366–2370. doi: 10.1093/jn/137.11.2366. [DOI] [PubMed] [Google Scholar]
  • 47.Catalan V, et al. Peripheral mononuclear blood cells contribute to the obesity-associated inflammatory state independently of glycemic status: involvement of the novel proinflammatory adipokines chemerin, chitinase-3-like protein 1, lipocalin-2 and osteopontin. Genes Nutr. 2015;10(3):460. doi: 10.1007/s12263-015-0460-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Weiss G, Goodnough LT. Anemia of chronic disease. N Engl J Med. 2005;352(10):1011–1023. doi: 10.1056/NEJMra041809. [DOI] [PubMed] [Google Scholar]
  • 49.Dinkla S, et al. Inflammation-associated changes in lipid composition and the organization of the erythrocyte membrane. BBA Clin. 2016;5:186–192. doi: 10.1016/j.bbacli.2016.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Georgatzakou HT, et al. Red blood cell abnormalities and the pathogenesis of anemia in end stage renal disease. Proteomics Clin Appl. 2016 doi: 10.1002/prca.201500127. [DOI] [PubMed] [Google Scholar]
  • 51.Kim A, et al. A mouse model of anemia of inflammation: complex pathogenesis with partial dependence on hepcidin. Blood. 2014;123(8):1129–1136. doi: 10.1182/blood-2013-08-521419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Nemeth E, Ganz T. Anemia of inflammation. Hematol Oncol Clin North Am. 2014;28(4):671–681. doi: 10.1016/j.hoc.2014.04.005. vi. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Blay JY, et al. Role of interleukin-6 in paraneoplastic thrombocytosis. Blood. 1993;82(7):2261–2262. [PubMed] [Google Scholar]
  • 54.Rossi JF, et al. Interleukin-6 as a therapeutic target. Clin Cancer Res. 2015;21(6):1248–1257. doi: 10.1158/1078-0432.CCR-14-2291. [DOI] [PubMed] [Google Scholar]
  • 55.Song SN, et al. Down-regulation of hepcidin resulting from long-term treatment with an anti-IL-6 receptor antibody (tocilizumab) improves anemia of inflammation in multicentric Castleman disease. Blood. 2010;116(18):3627–3634. doi: 10.1182/blood-2010-03-271791. [DOI] [PubMed] [Google Scholar]
  • 56.van Rhee F, et al. Siltuximab for multicentric Castleman's disease: a randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2014;15(9):966–974. doi: 10.1016/S1470-2045(14)70319-5. [DOI] [PubMed] [Google Scholar]
  • 57.Garcia-Manero G, et al. A phase 2, randomized, double-blind, multicenter study comparing siltuximab plus best supportive care (BSC) with placebo plus BSC in anemic patients with International Prognostic Scoring System low- or intermediate-1-risk myelodysplastic syndrome. Am J Hematol. 2014;89(9):E156–E162. doi: 10.1002/ajh.23780. [DOI] [PubMed] [Google Scholar]
  • 58.Mayeur C, et al. Oral administration of a bone morphogenetic protein type I receptor inhibitor prevents the development of anemia of inflammation. Haematologica. 2015;100(2):e68–e71. doi: 10.3324/haematol.2014.111484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Schwoebel F, et al. The effects of the anti-hepcidin Spiegelmer NOX-H94 on inflammation-induced anemia in cynomolgus monkeys. Blood. 2013;121(12):2311–2315. doi: 10.1182/blood-2012-09-456756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.van Eijk LT, et al. Effect of the antihepcidin Spiegelmer lexaptepid on inflammation-induced decrease in serum iron in humans. Blood. 2014;124(17):2643–2646. doi: 10.1182/blood-2014-03-559484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Cooke KS, et al. A fully human anti-hepcidin antibody modulates iron metabolism in both mice and nonhuman primates. Blood. 2013;122(17):3054–3061. doi: 10.1182/blood-2013-06-505792. [DOI] [PubMed] [Google Scholar]
  • 62.Gaun V, et al. A chemical screen identifies small molecules that regulate hepcidin expression. Blood Cells Mol Dis. 2014;53(4):231–240. doi: 10.1016/j.bcmd.2014.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Zhen AW, et al. The small molecule, genistein, increases hepcidin expression in human hepatocytes. Hepatology. 2013;58(4):1315–1325. doi: 10.1002/hep.26490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Alkhateeb AA, et al. The small molecule ferristatin II induces hepatic hepcidin expression in vivo and in vitro. Am J Physiol Gastrointest Liver Physiol. 2015;308(12):G1019–G1026. doi: 10.1152/ajpgi.00324.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Fraenkel PG. Understanding anemia of chronic disease. Hematology Am Soc Hematol Educ Program. 2015;2015:14–18. doi: 10.1182/asheducation-2015.1.14. [DOI] [PubMed] [Google Scholar]
  • 66.Boser P, et al. Anti-repulsive Guidance Molecule C (RGMc) Antibodies Increases Serum Iron in Rats and Cynomolgus Monkeys by Hepcidin Downregulation. AAPS J. 2015;17(4):930–938. doi: 10.1208/s12248-015-9770-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Theurl I, et al. Pharmacologic inhibition of hepcidin expression reverses anemia of chronic inflammation in rats. Blood. 2011;118(18):4977–4984. doi: 10.1182/blood-2011-03-345066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Querbes W, et al. Treatment of erythropoietin deficiency in mice with systemically administered siRNA. Blood. 2012;120(9):1916–1922. doi: 10.1182/blood-2012-04-423715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Richardson CL, et al. Isocitrate ameliorates anemia by suppressing the erythroid iron restriction response. J Clin Invest. 2013;123(8):3614–3623. doi: 10.1172/JCI68487. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES