Abstract

Atopic individuals tend to develop a Th2 dominant immune response, resulting in hyperresponsiveness to harmless antigens, termed allergens. In the last decade, epidemiological studies have emerged that connected allergy with a deficient iron-status. Immune activation under iron-deficient conditions results in the expansion of Th2-, but not Th1 cells, can induce class-switching in B-cells and hampers the proper activation of M2, but not M1 macrophages. Moreover, many allergens, in particular with the lipocalin and lipocalin-like folds, seem to be capable of binding iron indirectly via siderophores harboring catechol moieties. The resulting locally restricted iron-deficiency may then lead during immune activation to the generation of Th2-cells and thus prepare for allergic sensitization. Moreover, iron-chelators seem to also influence clinical reactivity: mast cells accumulate iron before degranulation and seem to respond differently depending on the type of the encountered siderophore. Whereas deferoxamine triggers degranulation of connective tissue-type mast cells, catechol-based siderophores reduce activation and degranulation and improve clinical symptoms. Considering the complex interplay of iron, siderophores and immune molecules, it remains to be determined whether iron-deficiencies are the cause or the result of allergy.

Graphical Abstract

Atopic individuals are often iron-deficient and tend to develop a Th2 dominant immune response, resulting in hyperresponsiveness to harmless antigens, termed allergens.

Introduction

Iron is an essential nutrient utilized in almost every aspect of normal cell function. All cells require iron to proliferate, iron being essential for DNA biosynthesis, protein function and cell cycle progression. In humans, iron is critical for a wide variety of biological processes as it allows transportation of oxygen, aids in the energy household and is essential for a healthy immune system.

Iron deficiency is probably the most common cause for anaemia, which by clinical definition is an insufficient mass of circulating red blood cells, and in public health terms is defined as a haemoglobin concentration below the thresholds given by the WHO, UNICEF and UNU.1,2 As such, iron deficiency can exist in the absence of anaemia, if it is mild or if the deficiency lasts not long enough.2

Iron deficiency can be absolute or functional, though combinations also exist. In absolute iron deficiency, the body has to cope with increased iron demand (e.g. during growth, blood donations, bleedings, and infections) that cannot sufficiently be compensated by dietary iron absorption and the release of recycled iron from senescent erythrocytes by macrophages.3 In contrast, during functional iron deficiency, the body has enough iron stores in the form of iron-laden ferritin in the liver, spleen and bone marrow, but iron-trafficking is on hold and/or reversed.

The different settings of iron metabolism are illustrated in Fig. 1. Under normal steady-state conditions, recycled iron is continuously released into the circulation by macrophages and dietary iron by enterocytes via the iron-exporter ferroportin in the form of Fe(ii) (Fig. 1A). Subsequently, ceruloplasmin or membrane-bound hephaestin oxidizes iron to Fe(iii) for transport via transferrin.4 Functional iron deficiency (Fig. 1B) is the consequence of an activated immune system that is triggered by danger signals derived from pathogens or damaged tissue and which results in a stop of the continuous iron release from enterocytes and macrophages and the accumulation of circulating iron into macrophages.

Fig. 1

Iron homeostasis under steady-state and inflammation. (A) Under steady-state conditions, dietary iron absorption occurs via (1) the divalent metal-ion transporter 1 (Dmt1) upon reduction of ferric iron in its ferrous form by ascorbic acid or duodenal cytochrome b (Dcytb) and (2) the heme transporter (Hcp1) in the form of heme. Iron is released into the circulation by (3) macrophages recycling senescent erythrocytes with the help of heme oxygenase 1 (HO1) and dietary iron by enterocytes via the iron-exporter ferroportin (Fpn1) in the form of Fe(ii). Subsequently, ceruloplasmin (Cp) or membrane-bound hephaestin (Heph) oxidizes iron to Fe(iii) for transport via transferrin (Tf). Intracellularly iron concentrations are regulated by the iron regulatory proteins (Irp)1/2. Binding of these proteins to iron results in the transcription of transferrin and ferritin (Ft). Also, a decrease of oxygen can activate hypoxia-inducible factor (Hif), leading to transcription of Tf and the transferrin receptor (TfR). (B) Under inflammation, not only hepcidin (Hepc), but also innate proteins like lipocalin 2 (Lcn2) and ferritin are released into the circulation, Hepc binding to ferroportin (FpN1), thereby initiating its degradation and leading to cytoplasmic accumulation of iron. The immune response differs depending on the human's iron status. (C) Under limited iron supply Th2-, but not Th1-cells, will expand due to their larger intracellular iron pool and secrete IL-4 and IL-13. M2 macrophages will differentiate into an allergic subtype, further providing a Th2 environment. B cells expand via transferrin receptor-independent mechanisms and induce class-switching in the presence of IL-4 and IL-13 towards IgE. Apo-allergens may cause local iron-deprivation or interfere with the regulatory functions of Lcn2 and further modulate the immune-activation. (D) In contrast, under sufficient iron-supply, macrophages may release Lcn2, Th1 cells will expand, secreting IFN-γ and TNF-α, and B cells will generate IgG antibodies.

Discrimination between the two forms is complicated by the existence of various degrees and levels of absolute and functional iron deficiency and their smooth transition into anemia.3 Moreover, many proteins contributing to iron homeostasis are also innate proteins, e.g. ferritin, lactoferrin, and lipocalin 2, which are released into the circulation upon immune activation to impede iron-sequestration by pathogens. This all complicates the assessment of the true iron status.

An interesting aspect of iron deficiency it that a decrease in red blood cells is often accompanied by a relative increase of the white blood cell population representing the immune cells per se.

With respect to allergy, several epidemiological studies have correlated a greater degree of iron deficiency in allergic subjects than in non-allergic individuals.5,6 Whether this is the result of an overboard in the immune response, in which a functional iron-deficiency becomes absolute, or whether an absolute iron-deficiency lays the ground for the generation of allergy remains to be determined.

This review aims to address several important environmental, immunological and physiological aspects that may influence an individual's iron homeostatic status and may contribute to the establishment of allergy.

Humans and iron

Redox chemistry of iron

Under physiological conditions, iron is largely found in the ferrous Fe(ii) or ferric Fe(iii) form and the metal's redox cycling properties make it highly suited to act as a biocatalyst in proteins or as an electron carrier. In general, Fe(iii) prefers oxygen ligands, while Fe(ii) favors nitrogen and sulfur ligands.7 In the human body typical examples of Fe(iii) binding proteins are lactotransferrin, transferrin and ferritin oxidizing Fe(ii) to Fe(iii) upon binding, and low molecular weight compounds in the blood also bind Fe(iii), with citric acid being the major representative.8 Also, amino acids, ATP/AMP, inositol phosphates and 2,5-dihydroxybenzoic acid have been described to chelate Fe(iii) but not Fe(ii).9 A different picture emerges in the cytosolic compartment of cells, in which about 1 μM of Fe(ii) predominates the labile iron pool, with glutathione in cellular concentrations ranging from 0.5 to 10 mM10,11 acting as a buffer9 and thus serving as a means for the subsequent incorporation of Fe(ii) into a wide range of iron-dependent enzymes and electron transfer proteins.9

Importantly, free redox-active iron can be very toxic under aerobic conditions due to the Haber–Weiss cycle.12 In this cycle Fe(iii) is reduced to Fe(ii) by superoxide or other reducing agents and the oxidation of Fe(ii) produces Fe(iii) and hydroxyl radicals. As such, iron serves as a catalyst and minute amounts of free iron are sufficient to produce significant levels of reactive oxygen species (ROS).

Human iron homeostasis

An 80 kg human body contains approximately 4 g of iron. Hemoglobin iron accounts for approximately 60% of total iron, the vast majority of which is found within circulating erythrocytes.13 Most of the remaining iron is stored in the liver within ferritin (≈1 g). From an immunological point of view, it is interesting to note that the next largest iron-stores are the macrophages (≈0.6 g) in the spleen, liver and bone marrow.14 Around 0.3 g of iron is stored as myoglobin of muscles. All other cellular iron-containing proteins and enzymes are estimated to bind a total of ≈8 mg of iron. Iron is delivered to most tissues via circulating transferrin, which carries ≈2 mg of this metal in the steady state. Iron mass in the total extracellular fluid volume is about 10 mg, implicating that the transferrin iron pool turns over several times a day.15 In healthy men plasma iron turnover ranges from 25 to 35 mg16 per day, of which only 5 to 10% is provided by absorption of dietary iron in the gut, and the rest is predominantly iron recycled from monocytes and macrophages of the liver, adipose tissue, bone marrow, spleen and lymph nodes.14

Nutritional iron uptake

Dietary iron requirements, as well as bioavailability, are mainly determined by an individual's iron homeostatic status, affected by physiological conditions and reflected to a large extent in serum hepcidin levels.17 The chief area of iron absorption is the duodenum and the proximal jejunum.18 The duodenum has some unique characteristics as its pH is more acidic, with a pH ranging from 4 to 5 compared to the rest of the small intestine that has a pH-range between 7 and 9, and is the site where pancreatic juices and bile enter the small intestine. Depending on its form, iron will also be transported,

(a) as heme (from meat) into the enterocytes via the high-affinity folate transporter, which is also the intestinal heme transporter PCP/HCP1 (SLC46A1).19–21 Interestingly, the duodenal cytochrome b, Dcytb, is also able to bind on the lumen and on the cytoplasmic side to heme molecules,22–25 though the implications of these binding sites have so far been investigated only for the cytoplasmic heme binding site.26

(b) as non-heme iron, typically through low molecular weight chelates of ferric iron, which can derive from meat or plants. After reduction by ascorbic acid and/or duodenal cytochrome b, Dcytb,24,25 iron enters the cells (enterocytes, macrophages, T cells) via the divalent metal-ion transporter 1, DMT1, pathway.26

(c) via other uptake-pathways that seem to exist – e.g. bile itself in “premicellar” concentrations has been shown to interact with iron(ii)27 and contribute to iron absorption.28,29 Iron-carrying proteins like ferritin from food are efficiently absorbed without depending on reduction or the heme transporter via receptor-mediated, clathrin-dependent endocytosis.30 Absorption and increased iron accumulation were also found in the liver, when Fe(ii) was ingested with glycine and asparagine, but not with other amino acids.31

Once in the cell, iron is exported by ferroportin 1, also known as IREG1, MTP1, SLC40A1, FPN1, and HFE4, into the circulation.32 In general, iron excretion is suppressed by iron deficiency and anaemia and enhanced during erythropoiesis and hypoxia.26

Known inhibitors of bioavailability for non-heme iron are phytates, which are inositol polyphosphates found predominantly in nuts, seeds and grains that form insoluble precipitates with iron,33 and polyphenolic compounds. Many of these fruit- and plant-derived polyphenols bind with high affinity to iron34 and can greatly affect iron homeostasis,35,36 as upon consumption, the plasma concentration of polyphenols can commonly reach values about 1–10 μM.37 In most studies, the consumption of large quantities of purified polyphenols has been reported to decrease the volunteers’ iron status. However, under more natural circumstances, it is reasonable to assume that polyphenols will be present in the food matrix already complexed with iron. As such, one study demonstrated that, while purified polyphenols decreased the iron parameters in the subjects, ingestion of polyphenols in context with iron significantly improved the iron and redox status in vivo.38

Regulation of iron homeostasis

Hepcidin, a 25 amino acid-long peptide, is the major regulator of iron homeostasis. It is mainly expressed in the liver, but can also be produced by parietal cells of the stomach39 and by macrophages.

As schematically presented in Fig. 1A, under steady-state conditions only a low concentration of hepcidin is present in circulation, with a median concentration of 7.8 nM found in men and 4.1 nM vs. 8.5 nM, in pre- and post-menopausal women, respectively.40 Dietary and recycled iron is continuously released into the circulation to meet the daily iron requirements of the human body. Excretion of ferrous iron into the circulation is mediated by ferroportin, and subsequently, ceruloplasmin or membrane-bound hephaestin oxidizes Fe(ii) to Fe(iii) for further transport via transferrin.

During inflammation (Fig. 1B), iron is removed from the circulation, mainly by upregulation of hepcidin, and many innate proteins like Lcn2 and ferritin are also secreted into the circulation to sequester iron. Importantly, also hepcidin-independent processes have been described in humans with iron-deficiency.40 Hepcidin binds to the iron-exporter ferroportin, leading to its degradation, while iron is retained within the cells.40,41 Thus, increasing body iron levels cause increased hepcidin expression, resulting in increased macrophage and liver cell iron sequestration, and decreased dietary iron absorption; the result is a reduction in serum iron.42 In contrast, decreasing body iron levels cause decreased hepcidin expression, resulting in an increased release of macrophage iron and accelerated dietary iron absorption.

Under absolute iron deficiency, a decreased hepcidin concentration will result in the release of the remaining iron stores and in an increase in the dietary iron absorption. Intracellular iron is also regulated by iron regulatory proteins (IRPs) 1 and 2 with iron-responsive elements, in which upregulation of these proteins reflects low body iron stores and an increase of dietary iron absorption.43 Moreover, oxygen can regulate iron homeostasis. During hypoxia, the hypoxia-inducible factors (HIF) can target genes encoding transferrin and the transferrin receptor, leading to increased expression of transferrin and thus increased transport of ferric iron into cells.44

Non-transferrin bound iron and the labile iron pool

Excreted iron is usually bound to transferrin, ferritin or heme. In the circulation, also non-transferrin-bound iron, NTBI, is present, which represents a heterogeneous population that comprises organic anions with low affinity to iron (e.g. citrate, phosphates and carboxylates), polyfunctional ligands (chelates, siderophores and polypeptides), albumin45 and surface components of membranes (e.g. glycans and sulfonates) able to bind Fe(ii) and Fe(iii). The NTBI is typically present in concentrations up to 10 μM and its existence has been correlated with high levels of transferrin saturation. Similarly, intracellularly, this heterogeneous population of redox-active Fe complexes is called the labile iron pool, LIP. In cells, the iron concentration usually ranges from 20 to 100 μM and is largely associated with proteins, whereas only a minor fraction of the cellular iron is present as the LIP (>1%, up to 5 μM46). In the cytosol, Fe(ii) is prevalent, with glutathione probably acting as a major buffer. The LIP is primarily found in erythroid and myeloid cells, as well as in neuronal cells.

Importantly, though the NTBI or LIP represents only a fraction of the total intra- and extracellular iron, fine-tuning the LIP-levels has physiologically and immunologically wide-reaching consequences. Both iron forms are the immediate therapeutic targets of diseases associated with a misbalance in the iron homeostasis, e.g. hereditary hemochromatosis, myelodysplastic syndromes and sickle-cell disease.47

The immune system

The immune system and iron

Iron is essential for many peroxide- and nitrous oxide-generating enzymes,48 and regulates cell growth, cell differentiation, and cytokine production. Iron can activate protein kinase C, which leads to phosphorylation of compounds regulating cell proliferation. In addition, iron is necessary for myeloperoxidase activity to form hydroxyl radicals, enabling neutrophils to efficiently eliminate bacteria.49 Thus, any misbalance in the iron homeostasis towards either deficiency or overload has wide-reaching immunological consequences.

Macrophages are immune cells known to store iron. The reason for the high presence of iron in macrophages is their pivotal role in systemic iron recycling, where senescent erythrocytes are cleared by predominantly splenic macrophages.50 In addition to their important role in erythrocyte clearance and maintenance, their anti-inflammatory or inflammatory state seems to be dependent on the iron content. Anti-inflammatory macrophages have a lower iron-content and an increased expression of proteins associated with iron efflux and can be discriminated from inflammatory macrophages, which harbour high iron-levels.51 Macrophage phagocytosis is generally unaffected by iron deficiency, even though macrophage bactericidal activity is affected.48 Neutrophils under iron-limited conditions show impaired or lower killing activity due to the reduced activity of myeloperoxidase and reduced mobility to inflamed sites. Likewise, NK-cells exhibit lower activity due to reduced differentiation and proliferation under iron-restricted conditions. Importantly, T lymphocytes can also actively modulate the NTBI pool by uptake and export, with T cell deficiency associated with an accumulation of iron in the liver and pancreas.52 Impaired iron-uptake via transferrin receptor 1 (TfR1) caused by mutations can result in severe B- and T-cell deficiencies due to the lack of activation, which can partly be compensated through the internalization of iron via NTBI-pathways.53

Allergy and iron

Allergy is an immune-mediated disease, caused by an aberrant immune response towards exogenous, normally harmless antigens derived from pollen, house dust mites, animal dander, insect venom and food components. In particular, in the westernized world the prevalence of allergy seems to increase, with almost 20% of the adults in Germany having at least one self-reported doctor diagnosed allergy.54 Also in the United States, the prevalence of respiratory allergies is approximately 20%, whereas the prevalence of food allergies has increased from 3 to 5% and the prevalence of skin allergies from 7 to 12%.55 Allergy defined as type I hypersensitivity causes an immediate reaction usually within minutes upon re-exposure to the antigen in an individual with preformed antigen-specific IgE antibodies. Allergic symptoms may affect the eyes, nose, skin, the lungs or the gastrointestinal tract, leading to red eyes, an itchy rash, runny nose, shortness of breath, swelling or diarrhea. In severe cases, a systemic reaction can occur, resulting in an anaphylactic shock.

Mechanistically, it is important to note that the presence of specific IgE antibodies, referred to as allergic sensitization, is a precondition of allergic symptoms. They are a product from plasma cells when stimulated by T-helper 2 cell (Th2-) cytokines IL-4 and IL-13. A Th2 bias in the immune response is typically associated with allergies. Re-exposure to the allergen results in binding and cross-linking of IgE antibodies bound via high-affinity receptors on effector cells. Mast cells then discharge their granules containing histamine, leukotrienes and other active agents by exocytosis, causing allergic reactions.56 However, many people may just be sensitized, thus already having IgE-antibodies, but not yet an allergic reaction upon exposure to the respective allergen.54

As such, the events leading to the initiation of IgE formation are poorly understood, though it is generally accepted that the shift in the immune balance towards Th2 directly correlates with the overproduction of allergen-specific IgE.

Epidemiological and experimental evidence

Iron and allergy

The poor iron status at birth was associated with an increased risk of developing allergic diseases.57 It has been argued that a massive perinatal lymphocyte expansion may have led to a prioritization of erythrocytes at the expense of other vital developing tissues58 due to the restricted iron supply.57 This may then have compromised vulnerable Th1 lymphocytes having lower intracellular iron stores and may have promoted eosinophilia. Accordingly, when the maternal iron status during pregnancy was reduced, this was adversely associated with childhood wheezing, altered lung function and atopic sensitisation in the first 10 years of life.59

Along with a lower iron status increasing the risk of atopy, high iron concentration in umbilical cord samples was associated with a decreased risk of wheezing and eczema in the population-based Avon Longitudinal Study of Parents and Children.60 Similarly, in a British study decreased serum ferritin levels were found in children with atopic eczema.61 The lower iron-status is a consistent and a reproducible finding in multiple US cohorts, which clearly associates atopy with anemia.5 Also in a case-controlled, population-based Korean study, low beta-carotene, iron, folic acid, and dietary vitamin E were associated with atopic dermatitis in young children.62 Another study showed a greatly reduced incidence of wheezing and asthma in infants of mothers who were supplemented during pregnancy with vitamin C, a known contributor to increasing iron bioavailability.63 The change in the iron status impacted allergy also in vivo in a murine model of allergic asthma, in which oral iron supplementation, as well as systemic iron administration, suppressed airway manifestations.64

Misregulated iron-metabolism can affect atopy or the generation of allergies. Patients with a much too high iron load due to frequent blood transfusions have a decreased CD4/CD8 ratio,65,66 and their increased serum ferritin levels significantly correlate with the number of transfusions.

More female allergy sufferers: association with iron?

Finally, as iron homeostasis differs between the genders67 and before and after adolescence, it may contribute to the differences seen in the prevalence of allergy in the various groups. During childhood, boys are more affected by allergy than girls, but this changes in adulthood and women are more likely to be affected than men. In a German evaluation, 24% of men, but 35% of women suffer from at least one allergy and 2.9% of men, but 6.4% of women suffer from food allergies.68 Over 20% of Portuguese women were found iron-deficient and other studies have also confirmed that females present more often iron deficiency.69,70

All in all, there is consistent evidence that the iron-status of allergic subjects is reduced and may be linked with the disease.

Iron and cancer

It should be considered that cancer is in general associated with a highly suppressed immune defense (tumor tolerance phenomenon) and thus cancer and allergy have opposite immunological features.71 In fact, epidemiological studies have suggested that an inverse association between allergic diseases and cancer exists, indicating that allergic and/or atopic subjects with elevated IgE levels have a higher risk for allergies, but a lower risk of developing certain cancer types.72 As far as the iron content is concerned, while decreased iron levels predispose to allergies, increased serum iron correlates with an increased cancer risk.73–78

Therefore, iron levels affect in opposite ways the immune regulation in allergy and cancer.

Immune effects of iron

Iron deficiency affects more Th1 than Th2 immune cells

The proliferative phase of T-cells is dependent on iron supply. Activation of T cells leads to the expression of TfR via an IL2-dependent pathway, which facilitates iron uptake. The iron availability is known to differentially modulate the proliferation of different Th cell subsets. Th1 cells usually associated with inflammation have a lower labile iron pool and activation of Th1 cells can be readily blocked79 by inhibiting iron-uptake via TfR and/or deferoxamine, whereas Th2 cells seem to maintain a larger amount of iron in an assumable less labile and less readily chelatable pool that is only partly affected by blocking TfR and/or deferoxamine.80 As such, under iron-restricted conditions, DNA synthesis in Th1, but not Th2 cells, was inhibited. In line with these experiments, the Th1-associated cytokines IFN-γ and the IL-12/IL-18 mediated proliferation was found to be severely affected by iron chelators, whereas Th2-associated cytokines IL-13, which is IL-4 mediated, were resistant to potent iron chelators.81

Thus, it is very likely that under conditions of lymphocyte expansion the limited iron supply in allergic subjects will favour the development of a Th2-environment, thereby preparing for later allergic sensitisation.

M2 macrophages are more susceptible to iron-deprivation

Classical activated M1 macrophages are characterized by secretion of inflammatory cytokines, harbour high iron-levels51 and are important contributors to the classical Th1 response. In contrast, alternative activated M2 macrophages usually residing in the tissues have immunoregulatory functions, have a lower iron-content and participate in Th2 reactions and in wound healing processes.

Corna et al. demonstrated that under iron deprivation by deferoxamine both M1 and M2 macrophages enhanced IRP1 activity, whereas IRP2 was more strongly enhanced in M2 macrophages.82 Deferoxamine treatment also does not lead to the suppression of ferritin heavy chain expression in M1 macrophages, indicating high enough iron stores, whereas in M2 macrophages TfR1 was upregulated for continuous iron supply.51 As such, M1 macrophages are not as affected by iron-deficient conditions as M2 macrophages. Importantly, M2 cells are not as efficient in expressing molecules involved in antigen presentation, such as MHC class II (I-Ab), or the costimulatory molecule CD86 after T-cell stimulation under iron-deficient conditions, whereas M1 macrophages seem unaffected by iron chelators.

Iron suppresses class-switching in B-cells

Resting B-cells express much lower levels of TfR on the plasma membrane than on T-cells83 and seem to have evolved also transferrin-independent iron uptake mechanisms.84 B-cell proliferation and plasmacytic differentiation are not affected by Fe(ii) although ferrous iron seems to inhibit activation-induced cytidine deaminase (AID), an important enzyme to initiate class-switching, in a dose-dependent and specific manner, while other bivalent ions such as Zn(ii), Mn(ii), Mg(ii), and Ni(ii) did not inhibit class-switching.85 As such, iron-deficiency may facilitate IgE class switching in the presence of Th2-associated cytokines like IL-4 and IL-13. Alternatively, B-cells can be stimulated by TGF-β to secrete IgA, and the iron-carrying protein lactoferrin is also able to directly interact with betaglycan (TGF-β receptor III, TβRIII) and initiate class-switching to IgA and IgG2b.86 Thus, though proliferation and plasmacytic differentiation are not affected,49 the AID enzyme responsible for class switching can be active under iron-deficient conditions.85

Flavonoids impair mast cell function

Mast cells are important effector cells in triggering IgE-mediated allergic reactions and impeding mast cell release is a strategy of allergy treatment. It is known that, after priming with IgE and antigens, mast cells accumulate Fe(iii)87 before degranulation. On the other hand, the iron chelator desferrioxamine can similarly activate connective tissue-type mast cells via non-IgE mediated mechanisms as compound 80/40, but fails to activate basophils and mucosal-type mast cells and does not elicit late phase reaction similar to compound 80/40. As such, desferrioxamine has been suggested as a positive control in intradermal skin tests.88 Still, many flavonoids such as apigenin, quercetin, luteolin, fisetin89 and epicatechin that are strong iron chelators (Fig. 2) have been shown to exhibit anti-allergic activity in vitro and in in vivo models.90,91 Luteolin inhibited mast cell activation,92 naringenin inhibited allergen-induced airway inflammation,93 baicalin suppressed food allergy by the generation of regulatory T-cells,94 cacao-bean extract containing catechins ameliorated house dust mite induced atopic dermatitis,95 rutin suppressed atopic dermatitis,96 afzelin ameliorated asthma,97 and proanthocyanidins, oligomers consisting of catechin, epicatechin and gallic esters,98 and apigenin99 inhibited airway inflammation. In double-blind placebo controlled clinical trials, O-methylated catechins reduced symptoms of Japanese cedar pollinosis100 and catechins also reduced symptoms in mild and moderate atopic dermatitis.101 From this, it becomes apparent that iron levels and chelators are able to regulate mast cells and thus have an impact on the severeness of allergic symptoms.

Fig. 2

Structures with affinity to iron. (A) Siderophores can be classified by their ligands interacting with iron as catecholates, hydroxamates, and hydroxyl-carboxylates. (B) The four different subclasses of flavonoids namely flavonols, flavan-3-ols, flavones and flavanones, with described ligand-binding activity to allergens or described as having anti-allergic properties.

Allergens

Structure–function relationships: role for siderophores and iron

One of the fundamental riddles in allergy is why certain proteins emerge as allergens. It is assumed that they are directly related to the critical events triggering the Th2 bias. Despite the existence of thousands of protein families, the structures of major allergens can be restricted to few protein families. Although many allergens have been characterized in terms of secondary and tertiary structures, it is still uncertain whether common structural, functional or biochemical features underlie their ability to generate an allergic response.

Nearly all major allergens from mammals belong to the lipocalin family,102 while plant allergens usually originate from the prolamin (2S albumin, lipid binding proteins (LTPs)) and cupin (7S, 11S) superfamilies or from the pathogenesis-related (PR)-10 family.103 All members of these families share certain characteristics like their great structural stability and their ability to serve as carriers for a variety of compounds with lipidic segments.104,105

Allergens deriving from mammalian sources usually belong to the lipocalin family. Lipocalins show unusually low levels of overall sequence conservation with pairwise sequence identities often below 20%. Nevertheless, as illustrated in Fig. 3 the lipocalin fold is highly conserved.106 This β-barrel structure shapes a calyx-like site which gives the name to the protein family and is the main feature regarding the binding abilities of the lipocalin fold.107 While the wider end of the barrel is open to the solvent and rich in polar and charged amino acids, the narrower end is an inner, buried region rich in hydrophobic amino acids. Loops flanking the calyx display a great sequence variability that endows lipocalins with the ability to bind a large variety of ligands having polar and non-polar moieties. This property has been exploited in protein design that uses lipocalins as scaffolds to engineer novel binding proteins (“anticalins”).108

Fig. 3

Sequence and structural alignment of lipocalins. (A) Clustal multiple sequence alignment of the following lipocalins. hLcn2: human Lcn2, Bos d 5: bovine β-lactoglobulin, Can f 1: dog allergen, Fel d 4: cat allergen, and Mus m 1: pheromone binding rodent major urinary protein from a mouse. The seven residues strictly conserved are boxed. (B) Percent identity matrix for the multiple sequence alignment in A. (C) Three views of the structural superposition of the hLcn2 (orange) crystal structure PDB id 1l6 m, Bos d 5 (blue) crystal structure PDB id 3NPO, Can f 1 (red) and Fel d 4 (green) homology model structures (ref. 100), and Mus m 1 (gray) crystal structure PDB id 1MUP. The percentages of allergen residues and RMSD values for backbone atoms in the structural superposition with hLcn2 are the following – Bos d 5: 67%, 1.362 Å; Can f 1: 66%, 1.574 Å; Fel d 4: 68%, 1.235 Å; Mus m 1: 73%, 1.158 Å.

Lipocalins are usually secreted and can be found in the dander, urine, fur, and saliva of animals.109 They have been described as powerful bacteriostatic agents against various microorganisms, by impeding their iron sequestration and binding to siderophore–iron complexes,110,111 but in accordance with their ligand binding plasticity, lipocalins also seem to act as carriers for lipids and hormones.

The human homologue lipocalin 2 (Lcn2, NGAL) has immune-regulatory function, but is also a growth factor112 and a stress protein that is released under various inflammatory conditions and in cancer.113 The binding of Lcn2 to bacterial siderophores, which are low molecular weight compounds that are amongst the strongest soluble Fe(iii) binding agents known, is considered a key feature against bacterial infections. Secreted Lcn2 seems to have specialized towards siderophores with catechol-moieties that facilitate their binding in the calyx site (see Fig. 4).114 Moreover, Lcn2 has been described to bind to endogenous siderophores like 2,5-dihydroxybenzoic acid (2,5-DHBA),115 thereby probably ensuring that excess free iron does not accumulate in the cytoplasm. Mammalian cells lacking this endogenous siderophore accumulate abnormally high levels of intracellular iron, leading to high levels of reactive oxygen species.115

Fig. 4

Structure of the Bos d 5–Fe(catechin)3 complex. (A) The structural formula of catechin. Catechol hydroxyls participating in the chelation of iron are marked with arrows. (B) 3D structure of the Fe(catechin)3 ligand showing the spatial arrangement of catechols to set the hexadentate octahedral coordination of iron (carbons in yellow, oxygen in red, and iron in dark gray). (C) 3D structure of the Bos d 5–Fe(catechin)3 complex showing the location of the ligand at the calyx of the lipocalin. Residues within a 3.5 Å distance from the ligand are displayed as sticks with the following color code: nine non-polar in pale cyan, two polar in cyan and one acidic in red.

Siderophores and iron modulate the Th2-potency of allergens

As iron is essential for almost all life, microbes and plants have evolved efficient iron sequestration strategies by producing siderophores that usually form a stable, hexadentate, octahedral complex predominantly with Fe(iii).116 Siderophores are usually classified by the ligands used to chelate the ferric iron with the major types of siderophores having catechol, hydroxamate or α-hydroxycarboxylate moieties, as depicted in Fig. 2. Also, combinations of the chemical groups are possible with microbes often employing non-ribosomal peptides to act as siderophores.117

Importantly, also many fruits and plants feature the presence of polyphenols/flavonoids known to behave as high-affinity iron chelators, a fact that is commonly overlooked,34 but that may contribute to their mast cell stabilizing ability. In addition, the ability of those compounds to chelate iron has been proposed to be a key element in their anti-oxidant properties.118

With regard to allergy, there seems to exist a particular role for catechol-type siderophores, which may also be related to the fact that catechol groups chelate iron119,120 at physiologically relevant pHs.

In this respect, it is relevant that lipocalin 2 can bind to only catechol-containing siderophores and not to others. This is an important characteristic that also seems to be extendable to many major allergens. Many lipocalin allergens such as the major milk allergen Bos d 5, or the major birch allergen Bet v 1, a prototype for the PR-10 protein family with a lipocalin-like architecture, are capable of transporting iron via catechol-type siderophores.121,122 Importantly, their loading state, apo- (empty) or holo- (filled), seemed to be decisive for the subsequent immune response. Apo-allergens can mount a Th2-response in vitro, whereas the holo forms are rather immune-suppressive, indicating that the iron-carrying form impedes allergic sensitization.121,122

As such, the natural ligand of the major birch pollen allergen Bet v 1 has been identified to be the flavonoid quercetin-3-O-sophoroside;123 the major peanut allergens Ara h 2 and Ara h 6, belonging to the 2S family, bind the flavonoid epigallocatechin-3-gallate;124 the major peanut allergen Ara h 1 from the 7S family forms large complexes by binding proanthocyanidins, which are oligomers consisting of catechin and epicatechin and their gallic acid esters.125 Accordingly, the pathogenesis-related PR-10 proteins and major allergens in strawberries, Fra a 1 and Fra a 3, have been crystallized with catechin ligands.126

In summary, there is solid evidence that allergens are capable of binding in particular catechol-type structures like quercetin and rutin with high iron affinities that surpass iron-affinity constants of deferoxamine.127,128

Importantly, a great number of these polyphenols have been described in the literature to exhibit anti-allergic properties. Taking lipocalins as an example, allergens are not only structurally at the border between self and non-self, but may also functionally interfere with the human immune-regulatory processes.

How normally harmless antigens become allergens with their characteristic Th2 skewing capacity is not known. There are several possible scenarios: (i) iron deficiency turns holo-lipocalins into apo-allergens, or (ii) exogenous lipocalins enter the body already devoid of a ligand in their calyx. In this case, they – due to their high affinity – will immediately sequester endogenous siderophores or comparable ligands directly at the mucosal sites, thereby contributing locally to an iron-deficient state. Additional triggers then may activate lymphocytes to become Th2 rather than Th1 cells.121,122,128 (iii) Allergenic lipocalins may in an ongoing immune response interfere with the function of Lcn2, e.g. during infections either by providing or by sequestering ligands for Lcn2. In any of these cases, the immune function of Lcn2 may be skewed by allergens towards Th2.

Can microbiota interfere with iron homeostasis?

From hygiene hypothesis to colonizing microbiota

The so-called hygiene hypothesis originated from observations that hay fever is more common in small families having few members than in large families and is associated with a less “dirty” environment, hence less microbial exposure.129 Moreover, living on a farm protects against atopy, hay fever, and asthma, especially in children.130 As such, cumulative evidence suggests that decreased exposure to microorganisms promotes allergy. The microbiota that colonizes humans has not merely a commensal, but rather a mutualistic relationship with their human host. It is accepted today that as such the bacterial composition has a profound impact on the immune system and vice versa.

Facts on microbiota in healthy versus allergic subjects

In the human healthy gut over 1000 bacterial species have been described,131 with an individual having at least 160 species, of which 52 species account for over 90%.132 Aerobic organisms including Streptococci and Lactobacilli, occasionally with Candida spp., predominantly colonize the human stomach, duodenum, and proximal jejunum,133 whereas an anaerobic predominance prevails in the distal ileum and colon. Dominant colonic organisms in humans include Clostridium spp., Bacteroides and Bifidobacterium.133 Thus, the Firmicutes and Proteobacteria phyla predominate in the duodenum, whereas Firmicutes and Bacteroidetes are the predominant phyla in the distal colon.

Microbiome depletion as a result of broad-spectrum antibiotic treatment led not only to anaemia and thrombocytosis but also to an altered immune homeostasis, resulting in diminished granulocytes and B cells116 in the bone marrow and increased CD8+ T-cells in mice. Importantly, faecal microbiota transfer partly rescued these hematopoietic changes.134 In rats inoculated with human faecal microbiota, provision of an iron-fortified diet after feeding an iron-deficient diet significantly increased the abundance of dominant bacterial groups such as Bacteroides spp. and Clostridium cluster IV members compared with rats on an iron-deficient diet only. Moreover, iron supplementation increased the gut microbial butyrate concentration 6-fold compared with iron depletion and did not affect histological colitis scores, suggesting that iron supplementation enhanced the concentration of beneficial gut microbiota metabolites and may contribute to gut health.135

Importantly, microbes able to inhabit the upper G/I tract seemed to be reduced in allergic subjects compared to non-allergic subjects.

There are only a few studies on the microbiota conducted in humans. As such, food allergic patients seem to have an increased abundance of bacteria of the order Clostridiales (Lachnospiraceae, Ruminococcaceae)136–139 and a decreased abundance of the order Bacteroidales.136,140,141

Microbiota interfere with iron levels via siderophores

An interesting aspect here is that Proteobacteria, Bacteroidetes and some family members of Firmicutes (see Table 1) are more likely to influence dietary iron uptake in the host due to the fact that their site of residency coincides with the site of iron-uptake. When screening these bacteria for their ability to secrete or utilize siderophores, it is apparent that indeed most of these organisms can acquire iron by siderophore-mediated mechanisms. As no data indicated the increased or decreased abundance of certain bacterial order, one can only speculate on their impact on iron homeostasis and on the immune cells.

Table 1

Organisms in the human microbiota associated with allergy

PhylumFamilyGenusSiderophore useReported abundancy in allergy
Actinobacteria137Bifidobacteriaceae136,140,141Bifidobacterium140+/−, Fe(ii)Increased140/decreased136–138,141
Coriobacteriaceae140Collinsella140+Increased140/decreased137,141
Bacteriodetes137,140,142BacteroidaceaeBacteroides+/hrDecreased137,140,141
RikenellaceaeRickenella+/hemeDecreased141
PorphyromonadaceaePorphyromonasFe(ii)/heme
Parabacteroides140?Decreased140, 141
PrevotellaceaePrevotella+, heme, tr −Decreased140, 141, 143
FirmicutesLactobacillaceaeLactobacillus140,141−/MnIncreased140
StreptococcaceaeStreptococcus140+, fr, lr, heme, hrDecreased138,140,141
EnterococcaceaeEnterococcus+, hemeDecreased138
StaphylococcaceaeStaphylocoocus+Decreased138,145
Clostridiaceae/cluster I140,141Clostridium+, hemeIncreased136,137,141
Clostridiaceae/cluster IV140,141Butyricicoccus?n/a
Anaerobacter140?Increased137,140/decreased139
Lachnospiraceae138,140,143,146,147Lactonifactor?n/a
Blautia+Decreased143/increased138
Roseburia+Increased138,140,141
Ruminococcaceae/cluster IV140,143Ruminococcus+Increased138,140,141
Faecalibacterium+Increased138,140/decreased139,141,143
SubdoligranulumFe(ii)Increased138,140/decreased141,143
VeillonellaceaeDialister141+Decreased141
Megamonas+n/a
Megasphaera141+Decreased141
Veillonella140+Decreased140,141
Proteobacteria137Enterobacteriaceae139Escherichia+Decreased138
Citrobacter+Increased139
PseudomonadaceaePseudomonas+
MoraxellaceaeAcinetobacter144+148Decreased144
CampylobacteraceaeCampylobacter149+Decreased150
Arcobacter+
VerrucomicrobiaAkkermansiaceae140Akkermansia+Increased140/decreased139,141
Kingdom Fungi: AscomycotaSaccharomycetaceaeCandida+Increased141
PhylumFamilyGenusSiderophore useReported abundancy in allergy
Actinobacteria137Bifidobacteriaceae136,140,141Bifidobacterium140+/−, Fe(ii)Increased140/decreased136–138,141
Coriobacteriaceae140Collinsella140+Increased140/decreased137,141
Bacteriodetes137,140,142BacteroidaceaeBacteroides+/hrDecreased137,140,141
RikenellaceaeRickenella+/hemeDecreased141
PorphyromonadaceaePorphyromonasFe(ii)/heme
Parabacteroides140?Decreased140, 141
PrevotellaceaePrevotella+, heme, tr −Decreased140, 141, 143
FirmicutesLactobacillaceaeLactobacillus140,141−/MnIncreased140
StreptococcaceaeStreptococcus140+, fr, lr, heme, hrDecreased138,140,141
EnterococcaceaeEnterococcus+, hemeDecreased138
StaphylococcaceaeStaphylocoocus+Decreased138,145
Clostridiaceae/cluster I140,141Clostridium+, hemeIncreased136,137,141
Clostridiaceae/cluster IV140,141Butyricicoccus?n/a
Anaerobacter140?Increased137,140/decreased139
Lachnospiraceae138,140,143,146,147Lactonifactor?n/a
Blautia+Decreased143/increased138
Roseburia+Increased138,140,141
Ruminococcaceae/cluster IV140,143Ruminococcus+Increased138,140,141
Faecalibacterium+Increased138,140/decreased139,141,143
SubdoligranulumFe(ii)Increased138,140/decreased141,143
VeillonellaceaeDialister141+Decreased141
Megamonas+n/a
Megasphaera141+Decreased141
Veillonella140+Decreased140,141
Proteobacteria137Enterobacteriaceae139Escherichia+Decreased138
Citrobacter+Increased139
PseudomonadaceaePseudomonas+
MoraxellaceaeAcinetobacter144+148Decreased144
CampylobacteraceaeCampylobacter149+Decreased150
Arcobacter+
VerrucomicrobiaAkkermansiaceae140Akkermansia+Increased140/decreased139,141
Kingdom Fungi: AscomycotaSaccharomycetaceaeCandida+Increased141

Code: predominantly found in colon (bold face), upper G/I (regular font). Ferritin – fr, transferrin – tr, lactoferrin – lr, hemophores – hr.

Table 1

Organisms in the human microbiota associated with allergy

PhylumFamilyGenusSiderophore useReported abundancy in allergy
Actinobacteria137Bifidobacteriaceae136,140,141Bifidobacterium140+/−, Fe(ii)Increased140/decreased136–138,141
Coriobacteriaceae140Collinsella140+Increased140/decreased137,141
Bacteriodetes137,140,142BacteroidaceaeBacteroides+/hrDecreased137,140,141
RikenellaceaeRickenella+/hemeDecreased141
PorphyromonadaceaePorphyromonasFe(ii)/heme
Parabacteroides140?Decreased140, 141
PrevotellaceaePrevotella+, heme, tr −Decreased140, 141, 143
FirmicutesLactobacillaceaeLactobacillus140,141−/MnIncreased140
StreptococcaceaeStreptococcus140+, fr, lr, heme, hrDecreased138,140,141
EnterococcaceaeEnterococcus+, hemeDecreased138
StaphylococcaceaeStaphylocoocus+Decreased138,145
Clostridiaceae/cluster I140,141Clostridium+, hemeIncreased136,137,141
Clostridiaceae/cluster IV140,141Butyricicoccus?n/a
Anaerobacter140?Increased137,140/decreased139
Lachnospiraceae138,140,143,146,147Lactonifactor?n/a
Blautia+Decreased143/increased138
Roseburia+Increased138,140,141
Ruminococcaceae/cluster IV140,143Ruminococcus+Increased138,140,141
Faecalibacterium+Increased138,140/decreased139,141,143
SubdoligranulumFe(ii)Increased138,140/decreased141,143
VeillonellaceaeDialister141+Decreased141
Megamonas+n/a
Megasphaera141+Decreased141
Veillonella140+Decreased140,141
Proteobacteria137Enterobacteriaceae139Escherichia+Decreased138
Citrobacter+Increased139
PseudomonadaceaePseudomonas+
MoraxellaceaeAcinetobacter144+148Decreased144
CampylobacteraceaeCampylobacter149+Decreased150
Arcobacter+
VerrucomicrobiaAkkermansiaceae140Akkermansia+Increased140/decreased139,141
Kingdom Fungi: AscomycotaSaccharomycetaceaeCandida+Increased141
PhylumFamilyGenusSiderophore useReported abundancy in allergy
Actinobacteria137Bifidobacteriaceae136,140,141Bifidobacterium140+/−, Fe(ii)Increased140/decreased136–138,141
Coriobacteriaceae140Collinsella140+Increased140/decreased137,141
Bacteriodetes137,140,142BacteroidaceaeBacteroides+/hrDecreased137,140,141
RikenellaceaeRickenella+/hemeDecreased141
PorphyromonadaceaePorphyromonasFe(ii)/heme
Parabacteroides140?Decreased140, 141
PrevotellaceaePrevotella+, heme, tr −Decreased140, 141, 143
FirmicutesLactobacillaceaeLactobacillus140,141−/MnIncreased140
StreptococcaceaeStreptococcus140+, fr, lr, heme, hrDecreased138,140,141
EnterococcaceaeEnterococcus+, hemeDecreased138
StaphylococcaceaeStaphylocoocus+Decreased138,145
Clostridiaceae/cluster I140,141Clostridium+, hemeIncreased136,137,141
Clostridiaceae/cluster IV140,141Butyricicoccus?n/a
Anaerobacter140?Increased137,140/decreased139
Lachnospiraceae138,140,143,146,147Lactonifactor?n/a
Blautia+Decreased143/increased138
Roseburia+Increased138,140,141
Ruminococcaceae/cluster IV140,143Ruminococcus+Increased138,140,141
Faecalibacterium+Increased138,140/decreased139,141,143
SubdoligranulumFe(ii)Increased138,140/decreased141,143
VeillonellaceaeDialister141+Decreased141
Megamonas+n/a
Megasphaera141+Decreased141
Veillonella140+Decreased140,141
Proteobacteria137Enterobacteriaceae139Escherichia+Decreased138
Citrobacter+Increased139
PseudomonadaceaePseudomonas+
MoraxellaceaeAcinetobacter144+148Decreased144
CampylobacteraceaeCampylobacter149+Decreased150
Arcobacter+
VerrucomicrobiaAkkermansiaceae140Akkermansia+Increased140/decreased139,141
Kingdom Fungi: AscomycotaSaccharomycetaceaeCandida+Increased141

Code: predominantly found in colon (bold face), upper G/I (regular font). Ferritin – fr, transferrin – tr, lactoferrin – lr, hemophores – hr.

The microbiota strongly manipulates the immune system. It is tempting to speculate that the composition and localization of the commensal microbiota in allergic subjects may directly impact the homeostatic iron status of the host, but more studies need to be done.

Conclusions

There is a clear epidemiological connection between a poor iron status and allergy risk, especially in females. Of note, iron-deficient conditions seem to promote a Th2-environment, which is a prerequisite for allergy. Potential contributing factors are endogenous iron levels, allergens capable of binding to iron chelators, and likely a skewed microbiota in allergic subjects.

Conflicts of interest

The authors declare no conflicts of interest.

Acknowledgements

This work was supported by the Austrian Science Fund FWF grant SFB F4606-B28 to EJJ.

References

United Nations Children's Fund, United Nations University, World Health Organization Iron Deficiency Anaemia Assessment, Prevention, and Control: A guide for programme managers, WHO/NHD/01.3
,
2001
, p.
114
.

World Health Organization, Centers for Disease Control and Prevention Technical Consultation. Assessing the iron status of populations, WD 105
,
2007
, p.
108
.

M.
 
Nairz
,
I.
 
Theurl
,
D.
 
Wolf
and
G.
 
Weiss
,
Iron deficiency or anemia of inflammation?: Differential diagnosis and mechanisms of anemia of inflammation
,
Wien. Med. Wochenschr.
,
2016
,
166
,
411
423
.

G.
 
Papanikolaou
and
K.
 
Pantopoulos
,
Systemic iron homeostasis and erythropoiesis
,
IUBMB Life
,
2017
,
69
,
399
413
.

K. E.
 
Drury
,
M.
 
Schaeffer
and
J. I.
 
Silverberg
,
Association Between Atopic Disease and Anemia in US Children
,
JAMA Pediatr.
,
2016
,
170
,
29
34
.

T.
 
Haahtela
,
G. J.
 
Burbach
,
C.
 
Bachert
,
C.
 
Bindslev-Jensen
,
S.
 
Bonini
,
J.
 
Bousquet
,
L.
 
Bousquet-Rouanet
,
P. J.
 
Bousquet
,
M.
 
Bresciani
,
A.
 
Bruno
,
G. W.
 
Canonica
,
U.
 
Darsow
,
P.
 
Demoly
,
S. R.
 
Durham
,
W. J.
 
Fokkens
,
S.
 
Giavi
,
M.
 
Gjomarkaj
,
C.
 
Gramiccioni
,
M. L.
 
Kowalski
,
G.
 
Losonczy
,
M.
 
Orosz
,
N. G.
 
Papadopoulos
,
G.
 
Stingl
,
A.
 
Todo-Bom
,
E.
 
von Mutius
,
A.
 
Kohli
,
S.
 
Wohrl
,
S.
 
Jarvenpaa
,
H.
 
Kautiainen
,
L.
 
Petman
,
O.
 
Selroos
,
T.
 
Zuberbier
and
L. M.
 
Heinzerling
,
Clinical relevance is associated with allergen-specific wheal size in skin prick testing
,
Clin. Exp. Allergy
,
2014
,
44
,
407
416
.

J. R.
 
Chipperfield
and
C.
 
Ratledge
,
Salicylic acid is not a bacterial siderophore: a theoretical study
,
Biometals
,
2000
,
13
,
165
168
.

F. I.
 
Adam
,
P. L.
 
Bounds
,
R.
 
Kissner
and
W. H.
 
Koppenol
,
Redox properties and activity of iron–citrate complexes: evidence for redox cycling
,
Chem. Res. Toxicol.
,
2015
,
28
,
604
614
.

R. C.
 
Hider
and
X. L.
 
Kong
,
Glutathione: a key component of the cytoplasmic labile iron pool
,
Biometals
,
2011
,
24
,
1179
1187
.

F.
 
Roth-Walter
,
P.
 
Starkl
,
T.
 
Zuberbier
,
K.
 
Hummel
,
K.
 
Nobauer
,
E.
 
Razzazi-Fazeli
,
R.
 
Brunner
,
I.
 
Pali-Scholl
,
J.
 
Kinkel
,
F.
 
Felix
,
E.
 
Jensen-Jarolim
and
T.
 
Kinaciyan
,
Glutathione exposes sequential IgE-epitopes in ovomucoid relevant in persistent egg allergy
,
Mol. Nutr. Food Res.
,
2013
,
57
,
536
544
.

A.
 
Meister
,
Glutathione metabolism and its selective modification
,
J. Biol. Chem.
,
1988
,
263
,
17205
17208
.

G. A.
 
Kortman
,
M.
 
Raffatellu
,
D. W.
 
Swinkels
and
H.
 
Tjalsma
,
Nutritional iron turned inside out: intestinal stress from a gut microbial perspective
,
FEMS Microbiol. Rev.
,
2014
,
38
,
1202
1234
.

G.
 
Pishchany
and
E. P.
 
Skaar
,
Taste for blood: hemoglobin as a nutrient source for pathogens
,
PLoS Pathog.
,
2012
,
8
,
e1002535
.

K.
 
Pantopoulos
,
S. K.
 
Porwal
,
A.
 
Tartakoff
and
L.
 
Devireddy
,
Mechanisms of mammalian iron homeostasis
,
Biochemistry
,
2012
,
51
,
5705
5724
.

T.
 
Ganz
,
Macrophages and systemic iron homeostasis
,
J. Innate Immun.
,
2012
,
4
,
446
453
.

D. D.
 
Funk
,
Plasma iron turnover in normal subjects
,
J. Nucl. Med.
,
1970
,
11
,
107
111
.

D.
 
Demeyer
,
S.
 
De Smet
and
M.
 
Ulens
,
The near equivalence of haem and non-haem iron bioavailability and the need for reconsidering dietary iron recommendations
,
Eur. J. Clin. Nutr.
,
2014
,
68
,
750
751
.

H.
 
Huebers
,
E.
 
Huebers
,
W.
 
Forth
and
W.
 
Rummel
,
and iron-deficient rats during absorption
,
Life Sci.
,
1971
,
10
,
1141
1148
.

Y.
 
Nakai
,
K.
 
Inoue
,
N.
 
Abe
,
M.
 
Hatakeyama
,
K. Y.
 
Ohta
,
M.
 
Otagiri
,
Y.
 
Hayashi
and
H.
 
Yuasa
,
Functional characterization of human proton-coupled folate transporter/heme carrier protein 1 heterologously expressed in mammalian cells as a folate transporter
,
J. Pharmacol. Exp. Ther.
,
2007
,
322
,
469
476
.

G. O.
 
Latunde-Dada
,
K.
 
Takeuchi
,
R. J.
 
Simpson
and
A. T.
 
McKie
,
Haem carrier protein 1 (HCP1): expression and functional studies in cultured cells
,
FEBS Lett.
,
2006
,
580
,
6865
6870
.

S.
 
Le Blanc
,
M. D.
 
Garrick
and
M.
 
Arredondo
,
Heme carrier protein 1 transports heme and is involved in heme-Fe metabolism
,
Am. J. Physiol.: Cell Physiol.
,
2012
,
302
,
C1780
C1785
.

G. F.
 
da Silva
,
V. P.
 
Shinkarev
,
Y. A.
 
Kamensky
and
G.
 
Palmer
,
Spectroscopic evidence of the role of an axial ligand histidinate in the mechanism of adrenal cytochrome b561
,
Biochemistry
,
2012
,
51
,
8730
8742
.

A. T.
 
McKie
,
D.
 
Barrow
,
G. O.
 
Latunde-Dada
,
A.
 
Rolfs
,
G.
 
Sager
,
E.
 
Mudaly
,
M.
 
Mudaly
,
C.
 
Richardson
,
D.
 
Barlow
,
A.
 
Bomford
,
T. J.
 
Peters
,
K. B.
 
Raja
,
S.
 
Shirali
,
M. A.
 
Hediger
,
F.
 
Farzaneh
and
R. J.
 
Simpson
,
An iron-regulated ferric reductase associated with the absorption of dietary iron
,
Science
,
2001
,
291
,
1755
1759
.

J. S.
 
Oakhill
,
S. J.
 
Marritt
,
E. G.
 
Gareta
,
R.
 
Cammack
and
A. T.
 
McKie
,
Functional characterization of human duodenal cytochrome b (Cybrd1): redox properties in relation to iron and ascorbate metabolism
,
Biochim. Biophys. Acta
,
2008
,
1777
,
260
268
.

S.
 
Ludwiczek
,
F. I.
 
Rosell
,
M. L.
 
Ludwiczek
and
A. G.
 
Mauk
,
Recombinant expression and initial characterization of the putative human enteric ferric reductase Dcytb
,
Biochemistry
,
2008
,
47
,
753
761
.

D. J.
 
Lane
,
D. H.
 
Bae
,
A. M.
 
Merlot
,
S.
 
Sahni
and
D. R.
 
Richardson
,
Duodenal cytochrome b (DCYTB) in iron metabolism: an update on function and regulation
,
Nutrients
,
2015
,
7
,
2274
2296
.

A.
 
Fini
,
G.
 
Feroci
,
G.
 
Fazio
and
P.
 
Zuman
,
Interaction of iron(ii) with bile salts
,
J. Inorg. Biochem.
,
1997
,
68
,
251
256
.

A. J.
 
Sanyal
,
M. L.
 
Shiffmann
,
J. I.
 
Hirsch
and
E. W.
 
Moore
,
Premicellar taurocholate enhances ferrous iron uptake from all regions of rat small intestine
,
Gastroenterology
,
1991
,
101
,
382
389
.

A. J.
 
Sanyal
,
J. I.
 
Hirsch
and
E. W.
 
Moore
,
Evidence that bile salts are important for iron absorption
,
Am. J. Physiol.
,
1994
,
266
,
G318
G323
.

E. C.
 
Theil
,
H.
 
Chen
,
C.
 
Miranda
,
H.
 
Janser
,
B.
 
Elsenhans
,
M. T.
 
Nunez
,
F.
 
Pizarro
and
K.
 
Schumann
,
Absorption of iron from ferritin is independent of heme iron and ferrous salts in women and rat intestinal segments
,
J. Nutr.
,
2012
,
142
,
478
483
.

J. M.
 
Christensen
,
M.
 
Ghannam
and
J. W.
 
Ayres
,
Effects of divalent amino acids on iron absorption
,
J. Pharm. Sci.
,
1984
,
73
,
1245
1248
.

A.
 
Donovan
,
A.
 
Brownlie
,
Y.
 
Zhou
,
J.
 
Shepard
,
S. J.
 
Pratt
,
J.
 
Moynihan
,
B. H.
 
Paw
,
A.
 
Drejer
,
B.
 
Barut
,
A.
 
Zapata
,
T. C.
 
Law
,
C.
 
Brugnara
,
S. E.
 
Lux
,
G. S.
 
Pinkus
,
J. L.
 
Pinkus
,
P. D.
 
Kingsley
,
J.
 
Palis
,
M. D.
 
Fleming
,
N. C.
 
Andrews
and
L. I.
 
Zon
,
Positional cloning of zebrafish ferroportin1 identifies a conserved vertebrate iron exporter
,
Nature
,
2000
,
403
,
776
781
.

S.
 
Clemens
,
Zn and Fe biofortification: the right chemical environment for human bioavailability
,
Plant Sci.
,
2014
,
225
,
52
57
.

M.
 
Dell'mour
,
W.
 
Schenkeveld
,
E.
 
Oburger
,
L.
 
Fischer
,
S.
 
Kraemer
,
M.
 
Puschenreiter
,
M.
 
Lammerhofer
,
G.
 
Koellensperger
and
S.
 
Hann
,
Analysis of iron-phytosiderophore complexes in soil related samples: LC-ESI-MS/MS versus CE-MS
,
Electrophoresis
,
2012
,
33
,
726
733
.

J. R.
 
Hunt
and
Z. K.
 
Roughead
,
Nonheme-iron absorption, fecal ferritin excretion, and blood indexes of iron status in women consuming controlled lactoovovegetarian diets for 8 wk
,
Am. J. Clin. Nutr.
,
1999
,
69
,
944
952
.

J.
 
Suliburska
,
P.
 
Bogdanski
,
M.
 
Szulinska
,
M.
 
Stepien
,
D.
 
Pupek-Musialik
and
A.
 
Jablecka
,
Effects of green tea supplementation on elements, total antioxidants, lipids, and glucose values in the serum of obese patients
,
Biol. Trace Elem. Res.
,
2012
,
149
,
315
322
.

N. R.
 
Perron
and
J. L.
 
Brumaghim
,
A review of the antioxidant mechanisms of polyphenol compounds related to iron binding
,
Cell Biochem. Biophys.
,
2009
,
53
,
75
100
.

A.
 
Imessaoudene
,
H.
 
Merzouk
,
F.
 
Berroukeche
,
N.
 
Mokhtari
,
B.
 
Bensenane
,
S.
 
Cherrak
,
S. A.
 
Merzouk
and
M.
 
Elhabiri
,
Beneficial effects of quercetin-iron complexes on serum and tissue lipids and redox status in obese rats
,
J. Nutr. Biochem.
,
2016
,
29
,
107
115
.

P.
 
Schwarz
,
J. A.
 
Kubler
,
P.
 
Strnad
,
K.
 
Muller
,
T. F.
 
Barth
,
A.
 
Gerloff
,
P.
 
Feick
,
C.
 
Peyssonnaux
,
S.
 
Vaulont
,
G.
 
Adler
and
H.
 
Kulaksiz
,
Hepcidin is localised in gastric parietal cells, regulates acid secretion and is induced by Helicobacter pylori infection
,
Gut
,
2012
,
61
,
193
201
.

T. E.
 
Galesloot
,
S. H.
 
Vermeulen
,
A. J.
 
Geurts-Moespot
,
S. M.
 
Klaver
,
J. J.
 
Kroot
,
D.
 
van Tienoven
,
J. F.
 
Wetzels
,
L. A.
 
Kiemeney
,
F. C.
 
Sweep
,
M.
 
den Heijer
and
D. W.
 
Swinkels
,
Serum hepcidin: reference ranges and biochemical correlates in the general population
,
Blood
,
2011
,
117
,
e218
e225
.

G.
 
Ramey
,
J. C.
 
Deschemin
,
B.
 
Durel
,
F.
 
Canonne-Hergaux
,
G.
 
Nicolas
and
S.
 
Vaulont
,
Hepcidin targets ferroportin for degradation in hepatocytes
,
Haematologica
,
2010
,
95
,
501
504
.

T.
 
Nakanishi
,
Y.
 
Hasuike
,
M.
 
Nanami
,
M.
 
Yahiro
and
T.
 
Kuragano
,
Novel iron-containing phosphate binders and anemia treatment in CKD: oral iron intake revisited
,
Nephrol., Dial., Transplant.
,
2016
,
31
,
1588
1594
.

M.
 
Munoz
,
J. A.
 
Garcia-Erce
and
A. F.
 
Remacha
,
Disorders of iron metabolism. Part 1: molecular basis of iron homoeostasis
,
J. Clin. Pathol.
,
2011
,
64
,
281
286
.

A.
 
Rolfs
,
I.
 
Kvietikova
,
M.
 
Gassmann
and
R. H.
 
Wenger
,
Oxygen-regulated transferrin expression is mediated by hypoxia-inducible factor-1
,
J. Biol. Chem.
,
1997
,
272
,
20055
20062
.

A. M.
 
Silva
and
R. C.
 
Hider
,
Influence of non-enzymatic post-translation modifications on the ability of human serum albumin to bind iron. Implications for non-transferrin-bound iron speciation
,
Biochim. Biophys. Acta
,
2009
,
1794
,
1449
1458
.

Y.
 
Ma
,
H.
 
de Groot
,
Z.
 
Liu
,
R. C.
 
Hider
and
F.
 
Petrat
,
Chelation and determination of labile iron in primary hepatocytes by pyridinone fluorescent probes
,
Biochem. J.
,
2006
,
395
,
49
55
.

L.
 
de Swart
,
J. C.
 
Hendriks
,
L. N.
 
van der Vorm
,
Z. I.
 
Cabantchik
,
P. J.
 
Evans
,
E. A.
 
Hod
,
G. M.
 
Brittenham
,
Y.
 
Furman
,
B.
 
Wojczyk
,
M. C.
 
Janssen
,
J. B.
 
Porter
,
V. E.
 
Mattijssen
,
B. J.
 
Biemond
,
M. A.
 
MacKenzie
,
R.
 
Origa
,
R.
 
Galanello
,
R. C.
 
Hider
and
D. W.
 
Swinkels
,
Second international round robin for the quantification of serum non-transferrin-bound iron and labile plasma iron in patients with iron-overload disorders
,
Haematologica
,
2016
,
101
,
38
45
.

J. L.
 
Beard
,
Iron biology in immune function, muscle metabolism and neuronal functioning
,
J. Nutr.
,
2001
,
131
,
568S
579S
, discussion 580S.

E. S.
 
Wintergerst
,
S.
 
Maggini
and
D. H.
 
Hornig
,
Contribution of selected vitamins and trace elements to immune function
,
Ann. Nutr. Metab.
,
2007
,
51
,
301
323
.

D. Z.
 
de Back
,
E. B.
 
Kostova
,
M.
 
van Kraaij
,
T. K.
 
van den Berg
and
R.
 
van Bruggen
,
Of macrophages and red blood cells; a complex love story
,
Front. Physiol.
,
2014
,
5
,
9
.

S.
 
Recalcati
,
M.
 
Locati
,
A.
 
Marini
,
P.
 
Santambrogio
,
F.
 
Zaninotto
,
M.
 
De Pizzol
,
L.
 
Zammataro
,
D.
 
Girelli
and
G.
 
Cairo
,
Differential regulation of iron homeostasis during human macrophage polarized activation
,
Eur. J. Immunol.
,
2010
,
40
,
824
835
.

J. P.
 
Pinto
,
J.
 
Arezes
,
V.
 
Dias
,
S.
 
Oliveira
,
I.
 
Vieira
,
M.
 
Costa
,
M.
 
Vos
,
A.
 
Carlsson
,
Y.
 
Rikers
,
M.
 
Rangel
and
G.
 
Porto
,
Physiological implications of NTBI uptake by T lymphocytes
,
Front. Pharmacol.
,
2014
,
5
,
24
.

H. H.
 
Jabara
,
S. E.
 
Boyden
,
J.
 
Chou
,
N.
 
Ramesh
,
M. J.
 
Massaad
,
H.
 
Benson
,
W.
 
Bainter
,
D.
 
Fraulino
,
F.
 
Rahimov
,
C.
 
Sieff
,
Z. J.
 
Liu
,
S. H.
 
Alshemmari
,
B. K.
 
Al-Ramadi
,
H.
 
Al-Dhekri
,
R.
 
Arnaout
,
M.
 
Abu-Shukair
,
A.
 
Vatsayan
,
E.
 
Silver
,
S.
 
Ahuja
,
E. G.
 
Davies
,
M.
 
Sola-Visner
,
T. K.
 
Ohsumi
,
N. C.
 
Andrews
,
L. D.
 
Notarangelo
,
M. D.
 
Fleming
,
W.
 
Al-Herz
,
L. M.
 
Kunkel
and
R. S.
 
Geha
,
A missense mutation in TFRC, encoding transferrin receptor 1, causes combined immunodeficiency
,
Nat. Genet.
,
2016
,
48
,
74
78
.

K. C.
 
Bergmann
,
J.
 
Heinrich
and
H.
 
Niemann
,
Current status of allergy prevalence in Germany: position paper of the Environmental Medicine Commission of the Robert Koch Institute
,
Allergol. J. Int.
,
2016
,
25
,
6
10
.

K. D.
 
Jackson
,
L. D.
 
Howie
and
L. J.
 
Akinbami
,
Trends in allergic conditions among children: United States, 1997–2011
,
NCHS Data Brief
,
2013
,
1
8
.

I.
 
Pali-Scholl
,
J.
 
Namazy
and
E.
 
Jensen-Jarolim
,
Allergic diseases and asthma in pregnancy, a secondary publication
,
World Allergy Organ. J.
,
2017
,
10
,
10
.

R.
 
Weigert
,
N. C.
 
Dosch
,
M. E.
 
Bacsik-Campbell
,
T. W.
 
Guilbert
,
C. L.
 
Coe
and
P. J.
 
Kling
,
Maternal pregnancy weight gain and cord blood iron status are associated with eosinophilia in infancy
,
J. Perinatol.
,
2015
,
35
,
621
626
.

M. K.
 
Georgieff
,
R. L.
 
Schmidt
,
M. M.
 
Mills
,
W. J.
 
Radmer
and
J. A.
 
Widness
,
Fetal iron and cytochrome c status after intrauterine hypoxemia and erythropoietin administration
,
Am. J. Physiol.
,
1992
,
262
,
R485
R491
.

B. I.
 
Nwaru
,
H.
 
Hayes
,
L.
 
Gambling
,
L. C.
 
Craig
,
K.
 
Allan
,
N.
 
Prabhu
,
S. W.
 
Turner
,
G.
 
McNeill
,
M.
 
Erkkola
,
A.
 
Seaton
,
H. J.
 
McArdle
and
G.
 
Devereux
,
An exploratory study of the associations between maternal iron status in pregnancy and childhood wheeze and atopy
,
Br. J. Nutr.
,
2014
,
112
,
2018
2027
.

S. O.
 
Shaheen
,
R. B.
 
Newson
,
A. J.
 
Henderson
,
P. M.
 
Emmett
,
A.
 
Sherriff
,
M.
 
Cooke
and
A. S.
 
Team
,
Umbilical cord trace elements and minerals and risk of early childhood wheezing and eczema
,
Eur. Respir. J.
,
2004
,
24
,
292
297
.

T. J.
 
David
,
F. E.
 
Wells
,
T. C.
 
Sharpe
,
A. C.
 
Gibbs
and
J.
 
Devlin
,
Serum levels of trace metals in children with atopic eczema
,
Br. J. Dermatol.
,
1990
,
122
,
485
489
.

S. Y.
 
Oh
,
J.
 
Chung
,
M. K.
 
Kim
,
S. O.
 
Kwon
and
B. H.
 
Cho
,
Antioxidant nutrient intakes and corresponding biomarkers associated with the risk of atopic dermatitis in young children
,
Eur. J. Clin. Nutr.
,
2010
,
64
,
245
252
.

C. T.
 
McEvoy
,
D.
 
Schilling
,
N.
 
Clay
,
K.
 
Jackson
,
M. D.
 
Go
,
P.
 
Spitale
,
C.
 
Bunten
,
M.
 
Leiva
,
D.
 
Gonzales
,
J.
 
Hollister-Smith
,
M.
 
Durand
,
B.
 
Frei
,
A. S.
 
Buist
,
D.
 
Peters
,
C. D.
 
Morris
and
E. R.
 
Spindel
,
Vitamin C supplementation for pregnant smoking women and pulmonary function in their newborn infants: a randomized clinical trial
,
JAMA
,
2014
,
311
,
2074
2082
.

H.
 
Maazi
,
S.
 
Shirinbak
,
N.
 
Bloksma
,
M. C.
 
Nawijn
and
A. J.
 
van Oosterhout
,
Iron administration reduces airway hyperreactivity and eosinophilia in a mouse model of allergic asthma
,
Clin. Exp. Immunol.
,
2011
,
166
,
80
86
.

M.
 
Gharagozloo
,
M.
 
Karimi
and
Z.
 
Amirghofran
,
Double-faced cell-mediated immunity in beta-thalassemia major: stimulated phenotype versus suppressed activity
,
Ann. Hematol.
,
2009
,
88
,
21
27
.

G.
 
Pardalos
,
F.
 
Kanakoudi-Tsakalidis
,
M.
 
Malaka-Zafiriu
,
H.
 
Tsantali
,
M.
 
Athanasiou-Metaxa
,
G.
 
Kallinikos
and
G.
 
Papaevangelou
,
Iron-related disturbances of cell-mediated immunity in multitransfused children with thalassemia major
,
Clin. Exp. Immunol.
,
1987
,
68
,
138
145
.

E.
 
Jensen-Jarolim
and
E.
 
Untersmayr
,
Gender-medicine aspects in allergology
,
Allergy
,
2008
,
63
,
610
615
.

U.
 
Langen
,
R.
 
Schmitz
and
H.
 
Steppuhn
,
Prevalence of allergic diseases in Germany: results of the German Health Interview and Examination Survey for Adults (DEGS1)
,
Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz
,
2013
,
56
,
698
706
.

C.
 
Fonseca
,
F.
 
Marques
,
A.
 
Robalo Nunes
,
A.
 
Belo
,
D.
 
Brilhante
and
J.
 
Cortez
,
Prevalence of anaemia and iron deficiency in Portugal: the EMPIRE study
,
Intern. Med. J.
,
2016
,
46
,
470
478
.

G.
 
Alvarez-Uria
,
P. K.
 
Naik
,
M.
 
Midde
,
P. S.
 
Yalla
and
R.
 
Pakam
,
Prevalence and severity of anaemia stratified by age and gender in rural India
,
Anemia
,
2014
,
2014
,
176182
.

E.
 
Jensen-Jarolim
,
M. C.
 
Turner
and
S. N.
 
Karagiannis
,
AllergoOncology: IgE- and IgG4-mediated immune mechanisms linking allergy with cancer and their translational implications
,
J. Allergy Clin. Immunol.
,
2017
,
140
,
982
984
.

E.
 
Jensen-Jarolim
,
G.
 
Achatz
,
M. C.
 
Turner
,
S.
 
Karagiannis
,
F.
 
Legrand
,
M.
 
Capron
,
M. L.
 
Penichet
,
J. A.
 
Rodriguez
,
A. G.
 
Siccardi
,
L.
 
Vangelista
,
A. B.
 
Riemer
and
H.
 
Gould
,
AllergoOncology: the role of IgE-mediated allergy in cancer
,
Allergy
,
2008
,
63
,
1255
1266
.

A. C.
 
Chua
,
M. W.
 
Knuiman
,
D.
 
Trinder
,
M. L.
 
Divitini
and
J. K.
 
Olynyk
,
Higher concentrations of serum iron and transferrin saturation but not serum ferritin are associated with cancer outcomes
,
Am. J. Clin. Nutr.
,
2016
,
104
,
736
742
.

H.
 
Cao
,
C.
 
Wang
,
R.
 
Chai
,
Q.
 
Dong
and
S.
 
Tu
,
Iron intake, serum iron indices and risk of colorectal adenomas: a meta-analysis of observational studies
,
Eur. J. Cancer Care
,
2017
,
26
,
e12486
.

C. P.
 
Wen
,
J. H.
 
Lee
,
Y. P.
 
Tai
,
C.
 
Wen
,
S. B.
 
Wu
,
M. K.
 
Tsai
,
D. P.
 
Hsieh
,
H. C.
 
Chiang
,
C. A.
 
Hsiung
,
C. Y.
 
Hsu
and
X.
 
Wu
,
High serum iron is associated with increased cancer risk
,
Cancer Res.
,
2014
,
74
,
6589
6597
.

A. G.
 
Mainous, 3rd
,
J. M.
 
Gill
and
C. J.
 
Everett
,
Transferrin saturation, dietary iron intake, and risk of cancer
,
Ann. Fam. Med.
,
2005
,
3
,
131
137
.

A. G.
 
Mainous, 3rd
,
B. J.
 
Wells
,
R. J.
 
Koopman
,
C. J.
 
Everett
and
J. M.
 
Gill
,
Iron, lipids, and risk of cancer in the Framingham Offspring cohort
,
Am. J. Epidemiol.
,
2005
,
161
,
1115
1122
.

B. J.
 
Wells
,
A. G.
 
Mainous, 3rd
,
C. J.
 
Everett
and
J. M.
 
Gill
,
Iron, cholesterol, and the risk of cancer in an 18-year cohort
,
Asian Pac. J. Cancer Prev.
,
2005
,
6
,
505
509
.

A.
 
Sica
and
A.
 
Mantovani
,
Macrophage plasticity and polarization: in vivo veritas
,
J. Clin. Invest.
,
2012
,
122
,
787
795
.

J. A.
 
Thorson
,
K. M.
 
Smith
,
F.
 
Gomez
,
P. W.
 
Naumann
and
J. D.
 
Kemp
,
Role of iron in T cell activation: TH1 clones differ from TH2 clones in their sensitivity to inhibition of DNA synthesis caused by IgG Mabs against the transferrin receptor and the iron chelator deferoxamine
,
Cell. Immunol.
,
1991
,
134
,
126
137
.

S.
 
Leung
,
A.
 
Holbrook
,
B.
 
King
,
H. T.
 
Lu
,
V.
 
Evans
,
N.
 
Miyamoto
,
C.
 
Mallari
,
S.
 
Harvey
,
D.
 
Davey
,
E.
 
Ho
,
W. W.
 
Li
,
J.
 
Parkinson
,
R.
 
Horuk
,
S.
 
Jaroch
,
M.
 
Berger
,
W.
 
Skuballa
,
C.
 
West
,
R.
 
Pulk
,
G.
 
Phillips
,
J.
 
Bryant
,
B.
 
Subramanyam
,
C.
 
Schaefer
,
H.
 
Salamon
,
E.
 
Lyons
,
D.
 
Schilling
,
H.
 
Seidel
,
J.
 
Kraetzschmar
,
M.
 
Snider
and
D.
 
Perez
,
Differential inhibition of inducible T cell cytokine secretion by potent iron chelators
,
J. Biomol. Screening
,
2005
,
10
,
157
167
.

G.
 
Corna
,
L.
 
Campana
,
E.
 
Pignatti
,
A.
 
Castiglioni
,
E.
 
Tagliafico
,
L.
 
Bosurgi
,
A.
 
Campanella
,
S.
 
Brunelli
,
A. A.
 
Manfredi
,
P.
 
Apostoli
,
L.
 
Silvestri
,
C.
 
Camaschella
and
P.
 
Rovere-Querini
,
Polarization dictates iron handling by inflammatory and alternatively activated macrophages
,
Haematologica
,
2010
,
95
,
1814
1822
.

J. W.
 
Larrick
and
P.
 
Cresswell
,
Transferrin receptors on human B and T lymphoblastoid cell lines
,
Biochim. Biophys. Acta
,
1979
,
583
,
483
490
.

P. A.
 
Seligman
,
J.
 
Kovar
,
R. B.
 
Schleicher
and
E. W.
 
Gelfand
,
Transferrin-independent iron uptake supports B lymphocyte growth
,
Blood
,
1991
,
78
,
1526
1531
.

G.
 
Li
,
E. J.
 
Pone
,
D. C.
 
Tran
,
P. J.
 
Patel
,
L.
 
Dao
,
Z.
 
Xu
and
P.
 
Casali
,
Iron inhibits activation-induced cytidine deaminase enzymatic activity and modulates immunoglobulin class switch DNA recombination
,
J. Biol. Chem.
,
2012
,
287
,
21520
21529
.

Y. S.
 
Jang
,
G. Y.
 
Seo
,
J. M.
 
Lee
,
H. Y.
 
Seo
,
H. J.
 
Han
,
S. J.
 
Kim
,
B. R.
 
Jin
,
H. J.
 
Kim
,
S. R.
 
Park
,
K. J.
 
Rhee
,
W. S.
 
Kim
and
P. H.
 
Kim
,
Lactoferrin causes IgA and IgG2b isotype switching through betaglycan binding and activation of canonical TGF-beta signaling
,
Mucosal Immunol.
,
2015
,
8
,
906
917
.

K.
 
Nakashima
,
T.
 
Takeuchi
and
T.
 
Shirakawa
,
Differentiation, distribution, and chemical state of intracellular trace elements in LAD2 mast cell line
,
Biol. Trace Elem. Res.
,
2005
,
108
,
105
114
.

M.
 
Shalit
,
A.
 
Tedeschi
,
A.
 
Miadonna
and
F.
 
Levi-Schaffer
,
Desferal (desferrioxamine)–a novel activator of connective tissue-type mast cells
,
J. Allergy Clin. Immunol.
,
1991
,
88
,
854
860
.

S.
 
Higa
,
T.
 
Hirano
,
M.
 
Kotani
,
M.
 
Matsumoto
,
A.
 
Fujita
,
M.
 
Suemura
,
I.
 
Kawase
and
T.
 
Tanaka
,
Fisetin, a flavonol, inhibits TH2-type cytokine production by activated human basophils
,
J. Allergy Clin. Immunol.
,
2003
,
111
,
1299
1306
.

A.
 
Singh
,
A.
 
Demont
,
L.
 
Actis-Goretta
,
S.
 
Holvoet
,
A.
 
Leveques
,
M.
 
Lepage
,
S.
 
Nutten
and
A.
 
Mercenier
,
Identification of epicatechin as one of the key bioactive constituents of polyphenol-enriched extracts that demonstrate an anti-allergic effect in a murine model of food allergy
,
Br. J. Nutr.
,
2014
,
112
,
358
368
.

D. F.
 
Finn
and
J. J.
 
Walsh
,
Twenty-first century mast cell stabilizers
,
Br. J. Pharmacol.
,
2013
,
170
,
23
37
.

S. K.
 
Kritas
,
A.
 
Saggini
,
G.
 
Varvara
,
G.
 
Murmura
,
A.
 
Caraffa
,
P.
 
Antinolfi
,
E.
 
Toniato
,
A.
 
Pantalone
,
G.
 
Neri
,
S.
 
Frydas
,
M.
 
Rosati
,
M.
 
Tei
,
A.
 
Speziali
,
R.
 
Saggini
,
F.
 
Pandolfi
,
G.
 
Cerulli
,
T. C.
 
Theoharides
and
P.
 
Conti
,
Luteolin inhibits mast cell-mediated allergic inflammation
,
J. Biol. Regul. Homeostatic Agents
,
2013
,
27
,
955
959
.

Y.
 
Shi
,
J.
 
Dai
,
H.
 
Liu
,
R. R.
 
Li
,
P. L.
 
Sun
,
Q.
 
Du
,
L. L.
 
Pang
,
Z.
 
Chen
and
K. S.
 
Yin
,
Naringenin inhibits allergen-induced airway inflammation and airway responsiveness and inhibits NF-kappaB activity in a murine model of asthma
,
Can. J. Physiol. Pharmacol.
,
2009
,
87
,
729
735
.

M. J.
 
Bae
,
H. S.
 
Shin
,
H. J.
 
See
,
S. Y.
 
Jung
,
D. A.
 
Kwon
and
D. H.
 
Shon
,
Baicalein induces CD4(+)Foxp3(+) T cells and enhances intestinal barrier function in a mouse model of food allergy
,
Sci. Rep.
,
2016
,
6
,
32225
.

H.
 
Kang
,
C. H.
 
Lee
,
J. R.
 
Kim
,
J. Y.
 
Kwon
,
M. J.
 
Son
,
J. E.
 
Kim
and
K. W.
 
Lee
,
Theobroma cacao extract attenuates the development of Dermatophagoides farinae-induced atopic dermatitis-like symptoms in NC/Nga mice
,
Food Chem.
,
2017
,
216
,
19
26
.

J. K.
 
Choi
and
S. H.
 
Kim
,
Rutin suppresses atopic dermatitis and allergic contact dermatitis
,
Exp. Biol. Med.
,
2013
,
238
,
410
417
.

W.
 
Zhou
and
X.
 
Nie
,
Afzelin attenuates asthma phenotypes by downregulation of GATA3 in a murine model of asthma
,
Mol. Med. Rep.
,
2015
,
12
,
71
76
.

D. Y.
 
Zhou
,
S. R.
 
Fang
,
C. F.
 
Zou
,
Q.
 
Zhang
and
W.
 
Gu
,
Proanthocyanidin from grape seed extract inhibits airway inflammation and remodeling in a murine model of chronic asthma
,
Nat. Prod. Commun.
,
2015
,
10
,
257
262
.

R. R.
 
Li
,
L. L.
 
Pang
,
Q.
 
Du
,
Y.
 
Shi
,
W. J.
 
Dai
and
K. S.
 
Yin
,
Apigenin inhibits allergen-induced airway inflammation and switches immune response in a murine model of asthma
,
Immunopharmacol. Immunotoxicol.
,
2010
,
32
,
364
370
.

S.
 
Masuda
,
M.
 
Maeda-Yamamoto
,
S.
 
Usui
and
T.
 
Fujisawa
,
‘Benifuuki’ green tea containing O-methylated catechin reduces symptoms of Japanese cedar pollinosis: a randomized, double-blind, placebo-controlled trial
,
Allergol. Int.
,
2014
,
63
,
211
217
.

A.
 
Patrizi
,
B.
 
Raone
,
I.
 
Neri
,
C.
 
Gurioli
,
M.
 
Carbonara
,
N.
 
Cassano
and
G. A.
 
Vena
,
Randomized, controlled, double-blind clinical study evaluating the safety and efficacy of MD2011001 cream in mild-to-moderate atopic dermatitis of the face and neck in children, adolescents and adults
,
J. Dermatol. Treat.
,
2016
,
27
,
346
350
.

T.
 
Virtanen
,
Lipocalin allergens
,
Allergy
,
2001
,
56
(
suppl 67
)
48
51
.

H.
 
Breiteneder
and
E. N.
 
Mills
,
Molecular properties of food allergens
,
J. Allergy Clin. Immunol.
,
2005
,
115
,
14
23
, quiz 24.

G. A.
 
Stewart
and
P. J.
 
Thompson
,
The biochemistry of common aeroallergens
,
Clin. Exp. Allergy
,
1996
,
26
,
1020
1044
.

F.
 
Roth-Walter
,
M. C.
 
Berin
,
P.
 
Arnaboldi
,
C. R.
 
Escalante
,
S.
 
Dahan
,
J.
 
Rauch
,
E.
 
Jensen-Jarolim
and
L.
 
Mayer
,
Pasteurization of milk proteins promotes allergic sensitization by enhancing uptake through Peyer's patches
,
Allergy
,
2008
,
63
,
882
890
.

D. R.
 
Flower
,
A. C.
 
North
and
T. K.
 
Attwood
,
Structure and sequence relationships in the lipocalins and related proteins
,
Protein Sci.
,
1993
,
2
,
753
761
.

D. R.
 
Flower
,
A. C.
 
North
and
C. E.
 
Sansom
,
The lipocalin protein family: structural and sequence overview
,
Biochim. Biophys. Acta
,
2000
,
1482
,
9
24
.

C.
 
Jost
and
A.
 
Pluckthun
,
Engineered proteins with desired specificity: DARPins, other alternative scaffolds and bispecific IgGs
,
Curr. Opin. Struct. Biol.
,
2014
,
27
,
102
112
.

T.
 
Schafer
,
J.
 
Merkl
,
E.
 
Klemm
,
H. E.
 
Wichmann
and
J.
 
Ring
,
We and our pets: allergic together?
,
Acta Vet. Hung.
,
2008
,
56
,
153
161
.

M. A.
 
Holmes
,
W.
 
Paulsene
,
X.
 
Jide
,
C.
 
Ratledge
and
R. K.
 
Strong
,
Siderocalin (Lcn 2) also binds carboxymycobactins, potentially defending against mycobacterial infections through iron sequestration
,
Structure
,
2005
,
13
,
29
41
.

J. J.
 
Rodvold
,
N. R.
 
Mahadevan
and
M.
 
Zanetti
,
Lipocalin 2 in cancer: when good immunity goes bad
,
Cancer Lett.
,
2012
,
316
,
132
138
.

K. M.
 
Schmidt-Ott
,
K.
 
Mori
,
J. Y.
 
Li
,
A.
 
Kalandadze
,
D. J.
 
Cohen
,
P.
 
Devarajan
and
J.
 
Barasch
,
Dual action of neutrophil gelatinase-associated lipocalin
,
J. Am. Soc. Nephrol.
,
2007
,
18
,
407
413
.

V.
 
Catalan
,
J.
 
Gomez-Ambrosi
,
A.
 
Rodriguez
,
B.
 
Ramirez
,
C.
 
Silva
,
F.
 
Rotellar
,
M. J.
 
Gil
,
J. A.
 
Cienfuegos
,
J.
 
Salvador
and
G.
 
Fruhbeck
,
Increased adipose tissue expression of lipocalin-2 in obesity is related to inflammation and matrix metalloproteinase-2 and metalloproteinase-9 activities in humans
,
J. Mol. Med.
,
2009
,
87
,
803
813
.

T. H.
 
Flo
,
K. D.
 
Smith
,
S.
 
Sato
,
D. J.
 
Rodriguez
,
M. A.
 
Holmes
,
R. K.
 
Strong
,
S.
 
Akira
and
A.
 
Aderem
,
Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron
,
Nature
,
2004
,
432
,
917
921
.

L. R.
 
Devireddy
,
D. O.
 
Hart
,
D. H.
 
Goetz
and
M. R.
 
Green
,
A mammalian siderophore synthesized by an enzyme with a bacterial homolog involved in enterobactin production
,
Cell
,
2010
,
141
,
1006
1017
.

R. C.
 
Hider
and
X.
 
Kong
,
Chemistry and biology of siderophores
,
Nat. Prod. Rep.
,
2010
,
27
,
637
657
.

S. M.
 
Barry
and
G. L.
 
Challis
,
Recent advances in siderophore biosynthesis
,
Curr. Opin. Chem. Biol.
,
2009
,
13
,
205
215
.

M.
 
Guo
,
C.
 
Perez
,
Y.
 
Wei
,
E.
 
Rapoza
,
G.
 
Su
,
F.
 
Bou-Abdallah
and
N. D.
 
Chasteen
,
Iron-binding properties of plant phenolics and cranberry's bio-effects
,
Dalton Trans.
,
2007
,
4951
4961
, DOI: .

P.
 
Mladenka
,
K.
 
Macakova
,
T.
 
Filipsky
,
L.
 
Zatloukalova
,
L.
 
Jahodar
,
P.
 
Bovicelli
,
I. P.
 
Silvestri
,
R.
 
Hrdina
and
L.
 
Saso
,
In vitro analysis of iron chelating activity of flavonoids
,
J. Inorg. Biochem.
,
2011
,
105
,
693
701
.

S. G.
 
Parkar
,
D. E.
 
Stevenson
and
M. A.
 
Skinner
,
influence of fruit polyphenols on colonic microflora and human gut health
,
Int. J. Food Microbiol.
,
2008
,
124
,
295
298
.

F.
 
Roth-Walter
,
C.
 
Gomez-Casado
,
L. F.
 
Pacios
,
N.
 
Mothes-Luksch
,
G. A.
 
Roth
,
J.
 
Singer
,
A.
 
Diaz-Perales
and
E.
 
Jensen-Jarolim
,
Bet v 1 from Birch Pollen is a Lipocalin-like Protein acting as Allergen only when devoid of Iron by promoting Th2 lymphocytes
,
J. Biol. Chem.
,
2014
,
289
,
17416
17421
.

F.
 
Roth-Walter
,
L. F.
 
Pacios
,
C.
 
Gomez-Casado
,
G.
 
Hofstetter
,
G. A.
 
Roth
,
J.
 
Singer
,
A.
 
Diaz-Perales
and
E.
 
Jensen-Jarolim
,
The major cow milk allergen Bos d 5 manipulates T-helper cells depending on its load with siderophore-bound iron
,
PLoS One
,
2014
,
9
,
e104803
.

C.
 
Seutter von Loetzen
,
T.
 
Hoffmann
,
M. J.
 
Hartl
,
K.
 
Schweimer
,
W.
 
Schwab
,
P.
 
Rosch
and
O.
 
Hartl-Spiegelhauer
,
Secret of the major birch pollen allergen Bet v 1: identification of the physiological ligand
,
Biochem. J.
,
2014
,
457
,
379
390
.

J.
 
Vesic
,
I.
 
Stambolic
,
D.
 
Apostolovic
,
M.
 
Milcic
and
D.
 
Stanic-Vucinic
,
and T. Cirkovic Velickovic, Complexes of green tea polyphenol, epigalocatechin-3-gallate, and 2S albumins of peanut
,
Food Chem.
,
2015
,
185
,
309
317
.

E. L.
 
van Boxtel
,
L. A.
 
van den Broek
,
S. J.
 
Koppelman
,
J. P.
 
Vincken
and
H.
 
Gruppen
,
Peanut allergen Ara h 1 interacts with proanthocyanidins into higher molecular weight complexes
,
J. Agric. Food Chem.
,
2007
,
55
,
8772
8778
.

A.
 
Casanal
,
U.
 
Zander
,
F.
 
Dupeux
,
V.
 
Valpuesta
and
J. A.
 
Marquez
,
Purification, crystallization and preliminary X-ray analysis of the strawberry allergens Fra a 1E and Fra a 3 in the presence of catechin
,
Acta Crystallogr., Sect. F: Struct. Biol. Cryst. Commun.
,
2013
,
69
,
510
514
.

G. M.
 
Escandar
and
L. F.
 
Sala
,
Complexing behavior of rutin and quercetin
,
Can. J. Chem.
,
1991
,
69
,
1994
2001
.

E.
 
Jensen-Jarolim
,
L. F.
 
Pacios
,
R.
 
Bianchini
,
G.
 
Hofstetter
and
F.
 
Roth-Walter
,
Structural similarities of human and mammalian lipocalins, and their function in innate immunity and allergy
,
Allergy
,
2016
,
71
,
286
294
.

D. P.
 
Strachan
,
Hay fever, hygiene, and household size
,
BMJ
,
1989
,
299
,
1259
1260
.

E.
 
von Mutius
and
D.
 
Vercelli
,
Farm living: effects on childhood asthma and allergy
,
Nat. Rev. Immunol.
,
2010
,
10
,
861
868
.

M.
 
Rajilic-Stojanovic
and
W. M.
 
de Vos
,
The first 1000 cultured species of the human gastrointestinal microbiota
,
FEMS Microbiol. Rev.
,
2014
,
38
,
996
1047
.

J.
 
Qin
,
R.
 
Li
,
J.
 
Raes
,
M.
 
Arumugam
,
K. S.
 
Burgdorf
,
C.
 
Manichanh
,
T.
 
Nielsen
,
N.
 
Pons
,
F.
 
Levenez
,
T.
 
Yamada
,
D. R.
 
Mende
,
J.
 
Li
,
J.
 
Xu
,
S.
 
Li
,
D.
 
Li
,
J.
 
Cao
,
B.
 
Wang
,
H.
 
Liang
,
H.
 
Zheng
,
Y.
 
Xie
,
J.
 
Tap
,
P.
 
Lepage
,
M.
 
Bertalan
,
J. M.
 
Batto
,
T.
 
Hansen
,
D.
 
Le Paslier
,
A.
 
Linneberg
,
H. B.
 
Nielsen
,
E.
 
Pelletier
,
P.
 
Renault
,
T.
 
Sicheritz-Ponten
,
K.
 
Turner
,
H.
 
Zhu
,
C.
 
Yu
,
S.
 
Li
,
M.
 
Jian
,
Y.
 
Zhou
,
Y.
 
Li
,
X.
 
Zhang
,
S.
 
Li
,
N.
 
Qin
,
H.
 
Yang
,
J.
 
Wang
,
S.
 
Brunak
,
J.
 
Dore
,
F.
 
Guarner
,
K.
 
Kristiansen
,
O.
 
Pedersen
,
J.
 
Parkhill
,
J.
 
Weissenbach
,
H. I. T. C.
 
Meta
,
P.
 
Bork
,
S. D.
 
Ehrlich
and
J.
 
Wang
,
A human gut microbial gene catalogue established by metagenomic sequencing
,
Nature
,
2010
,
464
,
59
65
.

R.
 
Balfour Sartor
,
Bacteria in Crohn's disease: mechanisms of inflammation and therapeutic implications
,
J. Clin. Gastroenterol.
,
2007
,
41
(
suppl 1
)
S37
S43
.

K. S.
 
Josefsdottir
,
M. T.
 
Baldridge
,
C. S.
 
Kadmon
and
K. Y.
 
King
,
Antibiotics impair murine hematopoiesis by depleting the intestinal microbiota
,
Blood
,
2017
,
129
,
729
739
.

A.
 
Dostal
,
C.
 
Lacroix
,
V. T.
 
Pham
,
M. B.
 
Zimmermann
,
C.
 
Del'homme
,
A.
 
Bernalier-Donadille
and
C.
 
Chassard
,
Iron supplementation promotes gut microbiota metabolic activity but not colitis markers in human gut microbiota-associated rats
,
Br. J. Nutr.
,
2014
,
111
,
2135
2145
.

M.
 
Kalliomaki
,
P.
 
Kirjavainen
,
E.
 
Eerola
,
P.
 
Kero
,
S.
 
Salminen
and
E.
 
Isolauri
,
Distinct patterns of neonatal gut microflora in infants in whom atopy was and was not developing
,
J. Allergy Clin. Immunol.
,
2001
,
107
,
129
134
.

Z.
 
Ling
,
Z.
 
Li
,
X.
 
Liu
,
Y.
 
Cheng
,
Y.
 
Luo
,
X.
 
Tong
,
L.
 
Yuan
,
Y.
 
Wang
,
J.
 
Sun
,
L.
 
Li
and
C.
 
Xiang
,
Altered fecal microbiota composition associated with food allergy in infants
,
Appl. Environ. Microbiol.
,
2014
,
80
,
2546
2554
.

R.
 
Berni Canani
,
N.
 
Sangwan
,
A. T.
 
Stefka
,
R.
 
Nocerino
,
L.
 
Paparo
,
R.
 
Aitoro
,
A.
 
Calignano
,
A. A.
 
Khan
,
J. A.
 
Gilbert
and
C. R.
 
Nagler
,
Lactobacillus rhamnosus GG-supplemented formula expands butyrate-producing bacterial strains in food allergic infants
,
ISME J.
,
2016
,
10
,
742
750
.

M.
 
Candela
,
S.
 
Rampelli
,
S.
 
Turroni
,
M.
 
Severgnini
,
C.
 
Consolandi
,
G.
 
De Bellis
,
R.
 
Masetti
,
G.
 
Ricci
,
A.
 
Pession
and
P.
 
Brigidi
,
Unbalance of intestinal microbiota in atopic children
,
BMC Microbiol.
,
2012
,
12
,
95
.

C. C.
 
Chen
,
K. J.
 
Chen
,
M. S.
 
Kong
,
H. J.
 
Chang
and
J. L.
 
Huang
,
Alterations in the gut microbiotas of children with food sensitization in early life
,
Pediatr. Allergy Immunol.
,
2016
,
27
,
254
262
.

K. E.
 
Fujimura
,
A. R.
 
Sitarik
,
S.
 
Havstad
,
D. L.
 
Lin
,
S.
 
Levan
,
D.
 
Fadrosh
,
A. R.
 
Panzer
,
B.
 
LaMere
,
E.
 
Rackaityte
,
N. W.
 
Lukacs
,
G.
 
Wegienka
,
H. A.
 
Boushey
,
D. R.
 
Ownby
,
E. M.
 
Zoratti
,
A. M.
 
Levin
,
C. C.
 
Johnson
and
S. V.
 
Lynch
,
Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation
,
Nat. Med.
,
2016
,
22
,
1187
1191
.

M.
 
Rajilic-Stojanovic
,
H. G.
 
Heilig
,
S.
 
Tims
,
E. G.
 
Zoetendal
and
W. M.
 
de Vos
,
Long-term monitoring of the human intestinal microbiota composition
,
Environ. Microbiol.
,
2013
,
15
,
1146
1159
.

X.
 
Hua
,
J. J.
 
Goedert
,
A.
 
Pu
,
G.
 
Yu
and
J.
 
Shi
,
Allergy associations with the adult fecal microbiota: analysis of the American Gut Project
,
EBioMedicine
,
2016
,
3
,
172
179
.

N.
 
Fyhrquist
,
L.
 
Ruokolainen
,
A.
 
Suomalainen
,
S.
 
Lehtimaki
,
V.
 
Veckman
,
J.
 
Vendelin
,
P.
 
Karisola
,
M.
 
Lehto
,
T.
 
Savinko
,
H.
 
Jarva
,
T. U.
 
Kosunen
,
J.
 
Corander
,
P.
 
Auvinen
,
L.
 
Paulin
,
L.
 
von Hertzen
,
T.
 
Laatikainen
,
M.
 
Makela
,
T.
 
Haahtela
,
D.
 
Greco
,
I.
 
Hanski
and
H.
 
Alenius
,
Acinetobacter species in the skin microbiota protect against allergic sensitization and inflammation
,
J. Allergy Clin. Immunol.
,
2014
,
134
,
1301
1309
, e1311.

E. A.
 
Kennedy
,
J.
 
Connolly
,
J. O.
 
Hourihane
,
P. G.
 
Fallon
,
W. H.
 
McLean
,
D.
 
Murray
,
J. H.
 
Jo
,
J. A.
 
Segre
,
H. H.
 
Kong
and
A. D.
 
Irvine
,
Skin microbiome before development of atopic dermatitis: Early colonization with commensal staphylococci at 2 months is associated with a lower risk of atopic dermatitis at 1 year
,
J. Allergy Clin. Immunol.
,
2017
,
139
,
166
172
.

J.
 
Jalanka-Tuovinen
,
A.
 
Salonen
,
J.
 
Nikkila
,
O.
 
Immonen
,
R.
 
Kekkonen
,
L.
 
Lahti
,
A.
 
Palva
and
W. M.
 
de Vos
,
Intestinal microbiota in healthy adults: temporal analysis reveals individual and common core and relation to intestinal symptoms
,
PLoS One
,
2011
,
6
,
e23035
.

J.
 
Jalanka-Tuovinen
,
J.
 
Salojarvi
,
A.
 
Salonen
,
O.
 
Immonen
,
K.
 
Garsed
,
F. M.
 
Kelly
,
A.
 
Zaitoun
,
A.
 
Palva
,
R. C.
 
Spiller
and
W. M.
 
de Vos
,
Faecal microbiota composition and host-microbe cross-talk following gastroenteritis and in postinfectious irritable bowel syndrome
,
Gut
,
2014
,
63
,
1737
1745
.

J. A.
 
Shapiro
and
T. A.
 
Wencewicz
,
Acinetobactin Isomerization Enables Adaptive Iron Acquisition in Acinetobacter baumannii through pH-Triggered Siderophore Swapping
,
ACS Infect. Dis.
,
2016
,
2
,
157
168
.

B. H.
 
Baig
,
I. K.
 
Wachsmuth
and
G. K.
 
Morris
,
Utilization of exogenous siderophores by Campylobacter species
,
J. Clin. Microbiol.
,
1986
,
23
,
431
433
.

D.
 
Jovanovic
,
N.
 
Ilic
,
B.
 
Miljkovic-Selimovic
,
D.
 
Djokic
,
T.
 
Relic
,
Z.
 
Tambur
,
R.
 
Doder
and
G.
 
Kostic
,
Campylobacter jejuni infection and IgE sensitization in up to 2-year-old infants
,
Vojnosanit. Pregl.
,
2015
,
72
,
140
147
.

Franziska Roth-Walter studied Chemistry and completed her PhD at the University of Vienna. She did her post-doctoral training in Immunology at the Medical University of Vienna, Austria, and the Mount Sinai School of Medicine, New York, USA. Since 2011 she has been working in the Interuniversity Messerli Research Institute in Vienna. She habilitated for Immunology in 2014 at the Medical University of Vienna. Her research is focused on understanding the mechanisms in mucosal immunity and allergy.

Luis F. Pacios is a physical chemist, obtained his PhD in Quantum Chemistry at the University Complutense of Madrid and completed his training in Clarkson University, New York, USA. Since 1987 he has been Associate Professor in the Technic University of Madrid where he currently teaches Protein Structure and Computational Structural Biology. His research focuses on the computational modeling of molecular structures and interactions in protein–ligand systems.

Rodolfo Bianchini is a senior scientist at the Interuniversity Messerli Research Institute in the Department of Comparative Medicine, Vienna, Austria. He obtained his PhD in Clinical and Experimental Pharmacology at the University of Perugia, Italy, and completed his post-doctoral training at the University of Perugia and the University of Salzburg in Austria. His research is primarily focused on modulating macrophage responses in allergy and cancer.

Erika Jensen-Jarolim, MD, immunologist, holds a professorship at the Medical University of Vienna, affiliated to the Interuniversity Messerli Research Institute where she directs the Department for Comparative Medicine. She is active on multiple boards and councils. She sees allergic patients and applies molecular allergy diagnosis. Her research focuses on immune mechanisms of molecular allergens and the immune regulation in allergen immunotherapy and cancer, in line with the concept of AllergoOncology.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by/3.0/), which permits non-commercial reuse, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com