Skip to main content

Biomarkers of the involvement of mast cells, basophils and eosinophils in asthma and allergic diseases

Abstract

Biomarkers of disease activity have come into wide use in the study of mechanisms of human disease and in clinical medicine to both diagnose and predict disease course; as well as to monitor response to therapeutic intervention. Here we review biomarkers of the involvement of mast cells, basophils, and eosinophils in human allergic inflammation. Included are surface markers of cell activation as well as specific products of these inflammatory cells that implicate specific cell types in the inflammatory process and are of possible value in clinical research as well as within decisions made in the practice of allergy-immunology.

Introduction

Eosinophils, mast cells and basophils are major effector cells of innate immunity and play a fundamental role in the mechanisms of defense against bacterial, viral and parasitic infections. These cells are distinct in terms of development, maturation and location within tissues. Eosinophils and basophils fully mature in the bone marrow and under physiological circumstances circulate as blood cells. Mast cells leave the bone marrow as progenitors and reach their final maturation within peripheral tissues. However, these three types of cells are often involved together in a variety of pathological conditions when they are activated and accumulate in inflamed tissues where they release a wide spectrum of chemical mediators of inflammation.

Given their strategic location at the major body surfaces including the skin and the lining of the lung and gastrointestinal tract, mast cells are among the first cells to recognize danger signals coming from the external environment. Once activated, mast cells help orchestrate the early phases of the innate immune response, promote the recruitment of other inflammatory cells and participate in the initiation and regulation of adaptive immunity. Eosinophils and basophils are recruited within organs and tissues following inflammatory and chemotactic stimuli that are produced by inflammation. Mast cells, eosinophils and basophils are therefore activated simultaneously in many diseases including infections, allergic and autoimmune disorders and cancer. Investigation of the role and function of mast cells, eosinophils and basophils in human disease has been hampered by difficulties in obtaining cells from human sources in sufficient number to perform suitable in vitro studies. Mature human mast cells can be retrieved only from tissues whereas eosinophils and basophils circulate in the blood in low numbers. Fortunately, new and efficient methods to purify these cells from human blood and tissues have been developed and now allow reproducible studies to explore their role in various pathophysiological conditions. At the same time, highly sensitive techniques are now available to measure mediators and to detect cell surface molecules, even from limited number of cells. These major improvements in cell purification, flow cytometry and mediator measurement have contributed to the identification of molecules that are either released from or expressed on the surface of activated mast cells, eosinophils and basophils and have been associated with involvement of these cells in human disease. In some cases, these molecules have provided important clues into the mechanisms by which mast cells, eosinophils and basophils participate in the development of human disease.

Eosinophils

Surface markers of activation

Recruitment of eosinophils from the circulation to tissues in disease requires that eosinophils become activated [13]. Up-regulation, and in some cases down-regulation, of surface proteins on blood eosinophils and activated conformations of integrins or Fc receptors have been associated with aspects of allergic disease (Table 1) [14]. Up-regulation or down-regulation in the table refers to altered cell-surface protein expression regardless of the mechanism, which may be mobilization from intracellular stores or the result of altered transcription or translation and may mean a change in average level on all cells or a change in the proportion of positive cells [1]. Some studies are on purified eosinophils, whereas some are on unfractionated blood cells. The latter has advantages, including the requirement for only a small amount of blood.

Table 1 Eosinophil surface proteins associated with allergic disease or aspects of allergic disease

Several proteins, including CD48, CD137, CRTH2, ICAM-1, IL-25R, integrins, and Fc receptors have been reported to be up- or down-regulated in one or more diseases when compared to normal, non-allergic healthy individuals (Table 1). At least some changes appear disease-specific. αX integrin, CRTH2, FcεRII (CD23), and ICAM-1 are up-regulated on eosinophils in eosinophilic eosophagitis (EoE) but not in airway allergies like atopic asthma or allergic rhinitis [5]; IL-25R is up-regulated in mild allergic asthma but not in atopic non-asthmatic subjects [1]; CD44 and CCR3 are down-regulated in EoE but not in airway allergies [5]. There are conflicting reports in the case of some proteins. Some authors found FcγRIII (CD16) to be up-regulated in airway allergies [1], whereas others [5] found no change compared to normal healthy individuals.

Expression or activation of some proteins has been found to be associated with clinical findings including extent of disease (Table 1). β2 integrin positively correlates while CD40 and CCR3 inversely correlate with tissue eosinophilia in EoE [6]. Activated β1 integrin, specifically the number of β1 integrins in the intermediate-activity conformation which is recognized by mAb N29, is significantly increased in mild to moderate (less severe) asthma compared to healthy donors, but is not significantly increased in severe asthma [1, 2]. Expression of the N29 epitope correlates inversely with lung function in non-severe asthma, i.e., higher N29 reactivity is associated with lower pulmonary function [1, 2]. In addition, by ROC curve analysis, N29 reactivity predicts decreased lung function in non-severe asthma [1, 2]. Partially activated β2 integrin correlates with broncheoalveolar lavage (BAL) eosinophilia in patients with mild allergic asthma [1, 2]. CD25 can be upregulated by IL-5 and GM-CSF in vitro and its expression is increased on BAL eosinophils in eosinophilic asthma and tissue eosinophils in atopic dermatitis and EoE [7, 8]. Moreover, HLA-DR expression was observed on sputum eosinophils of patients with asthma [9]. CD69 and neuropeptide S receptor are up-regulated on blood eosinophils in severe disease compared to non-severe disease (Table 1) [1, 10]. In contrast, CD44 and activated β1 integrin are increased in well-controlled/non-severe, but not in poorly controlled/severe, asthma. This is suggested to be due to continuous extravasation of the high-expressing eosinophils in severe disease [1].

The expression of some proteins has been found to change in response to intervention, such as administration of corticosteroids or an anti-IL-5 antibody (Table 2) [1, 6, 7, 11]. Specifically, the expression of β2 integrin is decreased by corticosteroids and CCR3 is decreased by anti-IL-5 (in EoE) [6, 11]. Anti-IL-5 causes decreased β2 activation and, after segmental lung antigen challenge, decreased levels of αL, αM, and β2 integrins as well as PSGL-1 (in mild allergic asthma) [1]. The utility of these potential markers, e.g., as predictors of aspects of allergic disease or of response to intervention, and differences and similarities in the behavior of markers among different allergic diseases, need to be explored further in clinical studies.

Table 2 Eosinophil surface proteins reported to respond to intervention

Major basic protein

Major basic protein (MBP)-1 and MBP-2 are two of the five basic proteins found within secondary eosinophil granules, the others being eosinophil-derived neurotoxin (EDN) also known as ribonuclease (RNase) 2, eosinophil cationic protein (ECP) also known as RNase3, and eosinophil peroxidase (EPX) (Table 3) [12]. MBP comprises the crystalloid core of the specific eosinophil granule, accounting for a major portion of the eosinophil granule protein, and has a high isoelectric point, calculated at greater than pH 11, so strongly basic that it cannot be measured accurately [13]. It is now known that MBP is expressed as two homologs, MBP-1 and MBP-2, coded by different genes on chromosome 11. MBP-1 and presumably MBP-2, is translated as a proprotein with a strongly acidic pro-portion [14]. The combination of the pro-section with MBP-1 or MBP-2 yields a molecule with a relatively neutral pI, 6.2. ProMBP-1 lacks the cytotoxic properties of MBP-1 and MBP2. Synthesis as a neutral pro-protein may function to protect the cell during transport from the Golgi apparatus to the granule. ProMBP-1 is the predominant form of MBP-1 in the blood of pregnant women and circulates as a complex with pregnancy-associated plasma protein-A (PAPP-A), with angiotensin, and with complement component, C3dg. It localizes to placental X cells and functions as a novel enzyme inhibitor of PAPP-A to modify its ability to release active insulin-like growth factor during primate pregnancy.

Table 3 Biologic activities of human eosinophil granule proteins

Both MBP-1 and MBP-2 are present in eosinophil granules [15]; MBP-1 is also present in basophils in a much lesser concentration than in eosinophils, but MBP-2 is not present in basophils. Mature eosinophils lose their ability to transcribe mRNA encoding MBP-1, indicating that all MBP-1 stored in the crystalloid granule cores is synthesized during early eosinophil development. MBP-2 is approximately 100 times less basic than MBP-1, with a calculated pI of 8.7. MBP-1 and MBP-2 have 42 identical amino acids of the approximately 117 in each of these proteins. The crystal structure of MBP-1 indicates that it is a member of the C-type lectin family. Comparative analyses of the biological effects of MBP-1 and MBP-2 demonstrate that they are similar in cytotoxic and cytostimulatory effects, but with reduced potency of MBP-2. Most of the eosinophil granule protein activities have been characterized for MBP-1. MBP-1 directly damages helminths and bacteria, and lethally damages mammalian cells and tissues. MBP-1 and MBP-2 stimulate histamine and LTC 4 release from human basophils and MBP-1 activates primed mast cells. Both MBP-1 and MBP-2 stimulate neutrophils. MBP-1 is a potent platelet agonist, causing release of 5-hydroxytryptamine and promoting clotting. MBP-1 is a potent vasodilator and increases vascular permeability. MBP-1 is elevated in the sputum of patients with asthma and is deposited in cardiac and skin lesions of patients with hypereosinophilic syndromes. Recently it has been shown that the toxicity of MBP-1 is regulated by aggregation [16].

Eosinophil Cationic Protein (ECP)

ECP occurs as a single chain protein with apparent molecular masses ranging from 15.7 to 22 kDa largely due to glycosylation and is stored in the granules as a highly glycosylated, non-toxic protein. ECP is expressed by eosinophils and is present only in humans and primates. Upon eosinophil degranulation, ECP is deglycosylated and acquires cytotoxic capabilities [17].

Among the four eosinophil basic proteins, ECP is the most widely used marker, to monitor diseases involving eosinophils and can be measured in serum/plasma and BAL. The majority of reports indicate that ECP levels provide useful information allowing not only monitoring of the disease course, but also treatment stratification [18, 19]. Levels of ECP in serum are consistently higher than in plasma, since eosinophils release ECP during the coagulation process, which is both time- and temperature- dependent [20]. Compared to eosinophil numbers, serum ECP levels decline with a delay following anti-IL-5 antibody therapy in patients with eosinophilia [21], pointing to the possibility that released ECP may remain in the circulation longer than the cellular source.

The levels of ECP in plasma likely reflect the ECP concentrations in blood at the time of sampling. Therefore, plasma ECP levels may better reflect the intensity of eosinophil activation than serum ECP levels, but no data are available that would support this assumption. ECP concentrations in other body fluids also likely reflect the activation stage of eosinophils. Taken together, the levels in serum/plasma and/or other body fluids largely correlate with the intensity of eosinophilic inflammation that is characterized by both numbers and activation of eosinophils. It should be noted, however, that ECP has been reported to be present in neutrophils which should be taken into consideration [22].

Eosinophil Peroxidase (EPX)

EPX is an eosinophil-specific cationic protein that is localized in the matrix of the secondary granules. Synthesized as a single chain precursor, EPX is proteolytically processed into a heavy chain (50–57 kDa) and a light chain (11–15 kDa). Although EPX has 68 % sequence identity to the neutrophil myeloperoxidase (MPO), patients with MPO deficiency have normal levels of EPX, consistent with differing mechanisms of expression of the two proteins.

In the presence of hydrogen peroxide, EPX catalyzes the oxidation of halides, pseudohalides and nitric oxide to form hypohalous acids, nitric dioxide and other reactive species. These products have been implicated in both inflammatory and anti-inflammatory processes including killing of pathogens [23], regulation of cholinergic nerve plasticity and inactivation of leukotrienes [24]. EPX has also been shown to be a useful biomarker of eosinophilic inflammation in a wide variety of clinical samples. Intracellularly, EPX mediates post-translational tyrosine nitration of all of the eosinophil granule proteins, including itself, during eosinophil maturation [25]. Despite these seemingly important functions, genetic deficiency in EPX has been described in humans and does not appear to be associated with a clinical phenotype.

Charcot-Leyden crystal protein

Charcot-Leyden Crystal Protein (CLC/Galectin-10) forms hexagonal bipyramidal crystals, hallmarks of eosinophil participation in allergic inflammatory reactions, found in tissues, body fluids and secretions. CLC, a 142 amino acid protein of 16.5 kDa [26], was initially identified as eosinophil lysophospholipase [27], but is now assigned to the galectin superfamily (Galectin-10) [28, 29]. Importantly, CLC/Gal-10 lacks lysophospholipase activity, and the weak enzymatic activity initially attributed to CLC was due to contamination by other lysophospholipases expressed by eosinophils [30]. In addition to eosinophils and basophils [29], CLC/Gal-10 is expressed by human regulatory T cells (Tregs) [31]. Proteomics of CD4 + CD25+ Tregs identified CLC/Gal-10 as a novel biomarker and essential for Treg anergy and suppressive functions on T cell activation [31]. CLC/Gal-10 is a potentially useful biomarker of eosinophil/basophil involvement in asthma, allergic rhinitis and other eosinophil-associated inflammatory diseases [32]. CLC/Gal-10 levels measured by ELISA are a sensitive and specific biomarker of active eosinophilic inflammation in EoE [33], differentiating patients with active EoE from treated patients in remission and those with gastroesophageal reflux disease, and is highly correlated with the numbers of tissue eosinophils. CLC/Gal-10 is also a potential biomarker for eosinophilic airway inflammation in induced sputum [34]. In celiac disease, CLC/Gal-10 expression is related to disease activity and tissue eosinophils in intestinal lesions [35]. Genetic variations (single nucleotide polymorphisms [SNPs]) in the promoter region of the CLC gene identify potential susceptibility biomarkers for allergic rhinitis [36], with variation compatible with recessive inheritance and increased levels of CLC/Gal-10 in nasal fluid of patients with allergic rhinitis during allergy season.

Eosinophil-derived neurotoxin (EDN, RNase2)

EDN, a small cationic protein belonging to the ribonuclease A (RNase A) superfamily, is localized to the matrix of the eosinophil’s large secondary granule, and is also known as RNase-2, non-secretory RNase, and RNase-Us, the latter based on its specificity toward uridine-containing nucleotides. EDN has been isolated from a wide variety of sources including eosinophils, placenta, liver, and urine. There is ~3.3 μg of EDN/106 eosinophils, but it is not specific to eosinophils, being expressed in smaller amounts by both basophils and neutrophils [37]. EDN was first identified based on its induction of a paralytic syndrome of ataxia, incoordination, and spasmodic paralysis when injected intrathecally into rabbits [38]. The RNase activity of EDN is a prerequisite for its cytotoxic, neurotoxic and antiviral activities [39, 40]. Unlike other eosinophil granule proteins, EDN is a poor cationic toxin with limited toxicity for helminth parasites and mammalian cells, but as a ribonuclease, it is considerably more effective against single-stranded RNA viruses [39]. EDN activates human dendritic cells (DCs) [41], induces phenotypic and functional maturation of DCs, and acts as an alarmin that activates the TLR2-MyD88 signaling pathway [42]. As a measure of eosinophil activation in vitro, EDN has been a biomarker of choice, since eosinophils have been shown to secrete >80 % of available EDN compared to the other granule cationic proteins. As a biomarker of eosinophil-associated diseases, EDN levels are significantly increased in blood (serum/plasma), urine and other body fluids, and tissue biopsies and esophageal luminal secretions [33]. EDN levels are significantly increased in the blood of patients with Onchocerciasis during an eosinopenia induced by treatment with diethylcarbamazine [43]. In EoE, plasma EDN levels are significantly elevated in active EoE, with moderately good sensitivity and specificity for differentiating EoE from patients with GERD and normal subjects, but only when combined with absolute blood eosinophil counts [44, 45]. Studies comparing EDN levels in esophageal biopsy extracts with the minimally invasive Esophageal String Test (EST), show elevated levels of EDN that distinguish patients with active EoE from those in treatment-induced remission, GERD and normal esophagus [33]. Serum EDN levels measured 3 months post-respiratory syncytial virus (RSV)-induced bronchiolitis may be a predictive biomarker of recurrent wheezing in infants [46]. In summary, for already diagnosed eosinophil-associated inflammatory diseases, e.g., EoE, EDN may have utility as a blood and/or tissue biomarker for following patient responses to treatment; however, EDN’s value may be limited by its expression in neutrophils, e.g., for distinguishing eosinophilic vs. neutrophilic phenotypes of asthma using induced sputum.

Mast cells

Histamine

Histamine is the main biogenic amine released upon IgE-receptor activation by human mast cells (3–8 pg/cell) and basophils [47]. Histamine measured in blood in association with an allergic reaction cannot thus be considered a specific marker of mast cell activation. Histamine is present in substantial amounts in many fermented foods and beverages, and this must be taken into consideration when measuring histamine levels. Histamine can be measured in several body fluids, and been shown to be increased in bronchoalveolar lavage fluid from patients with allergic asthma and plasma from patients with atopic dermatitis or chronic urticaria [48, 49]. Measurement of plasma histamine is problematic due to the short half-life of histamine (10–30 min) [50]. Histamine is rapidly metabolized either by methylation into methyl histamine catalyzed by histamine N-methyltransferase, or oxidative deamination into imidazole acetaldehyde catalyzed by diamine oxidase. The metabolite, 1-methyl-4-imidazole acetic acid (tele-MIAA), represents 70–80 % of metabolized histamine [51] and is excreted in urine. Determination of levels of histamine metabolites in urine provides a non-invasive means by which to monitor global histamine turnover in humans. Such an analysis is usually performed on urine collected over 24 h, but shorter collection times and even spot samples may be used. Increased levels of histamine metabolites in the urine can be sign of systemic mastocytosis and/or mast cell and basophil activation [52]. There are several methods available to measure histamine, including ELISA and solid phases, e.g., glass fibers. These are primarily useful for in vitro measurement of histamine released from mast cells and basophils. Histamine metabolites in the urine may also be measured by high-performance liquid chromatography (HPLC) and HPLC coupled to mass spectrometry (LC – MS) [51, 53].

Heparin

Heparin is produced by mast cells and human lung mast cells contain approximately 2.4 to 7.8 micrograms of heparin per 106 cells [54]. Human heparin is associated with the collections of mast cells associated with urticaria pigmentosa (maculopapular lesions) [55]. In rare cases of advanced systemic mastocytosis, a heparin-like anticoagulant may be released which leads to hemorrhagic complications [56]. However, in most cases, the thrombin time and partial thromboplastin time remains normal in those with mast cell proliferative disorders. Basal endogenous heparin levels in blood have been reported to be above the laboratory-specific range in approximately 40 % of patients classified as having mast cell activation disease [57]. It has also been suggested that heparin-initiated bradykinin formation plays a role in mast cell-mediated diseases [58]. Thus measurement of mast cell-derived heparin should be considered in mast cell mediated diseases where there is clinical evidence of hemorrhagic complications. However, in the vast majority of mast cell mediated inflammatory reactions, blood levels of heparin will remain within the normal range.

Tryptase

The human tryptase locus on chromosome 16 includes genes that encode α- and β- tryptases [59], which are major protein products of human mast cells, much lesser amounts being expressed by basophils [60]. α/β-Protryptases are processed to maturity by cathepsins B and L, while β-protryptase also can be sequentially processed by autocatalysis and cathepsin C [61, 62]. Mature β-tryptase is proteolytically active as a homotetramer, whereas mature α-tryptase is not, despite being 93 % identical in amino acid sequence. Protryptases are constitutively secreted by resting mast cells, whereas mature tryptases, which are stored in secretory granules, are only secreted by activated mast cells. Baseline serum levels of α/β-tryptases (pro + mature), ranging from 1–11 ng/ml in healthy subjects, reflecting primarily their genetic background [63], serve as a minor diagnostic criterion for systemic mastocytosis when >20 ng/ml (Table 4). However, in subjects with systemic anaphylaxis to insect stings, serum basal tryptase levels in the upper normal range [64] or when between 11 and 20 [65], also raise suspicion for an underlying clonal mast cell disorder. Acute serum tryptase levels, collected within 4 h of a putative anaphylactic event, if increased > (2 + 1.2*baseline level), provide laboratory evidence for mast cell activation [66].

Table 4 Tryptase levels in relation to mastocytosis amd anaphylaxis

Chymase

Human chymase is a chymotrypsin-like serine protease specific for mast cells. It is found in a subset of human mast cells, usually in conjunction with human mast cell carboxypeptidase A3. It is released from mast cells in large complexes containing heparin proteoglycan and carboxypeptidase and distinct from complexes containing tryptase [67]. Human chymase is the major non-angiotensin-converting enzyme (ACE) that generates angiotensin II as well as a non-endothelin converting-enzyme (ECE) that generates endothelin-1. As such, chymase is thought to participate in multiple inflammatory responses in the vasculature, including blood pressure regulation and plaque instability [68]. Chymase degrades lipoproteins which promotes macrophage foam cell formation. Chymase can also degrade extracellular matrix, generate fibronectin and transforming growth factor-β and activate IL-1β, and has been implicated in the pathogenesis of tissue fibrosis and wound healing [69, 70]. In human serum, chymase is subject to inhibition by endogenous circulating inhibitors, including α-1 antitrypsin, α-1 antichymotrypsin, α-2 macroglobulin and locally secreted inhibitors including secretory leukocyte protease inhibitor (SLPI) [71]. An immunoassay for the detection of released chymase in biologic fluids is not currently available. Raymond et al. developed an α-2 macroglobulin capture assay using a synthetic substrate which detects enzymatic activity in chymase-spiked serum with a threshold of approximately 30 pg/ml, and reveals detectable chymase activity in serum of most subjects with systemic mastocytosis [72].

Carboxypeptidase A3

Carboxypeptidase was first identified in human mast cells in 1989, with enzyme activity varying from 0.5 to 16 ug per 106 mast cells depending on the mast cell population [73]. Mast cells containing Carboxypeptidase A3 (CA3) have been reported in association with allergic disease of both the lower and upper airways [74, 75]. CPA3 is also one of 10 genes overexpressed in the bone marrow mononuclear cells of adult patients with systemic mastocytosis [76].

Plasma levels of mast cell carboxypeptidase have been reported to be elevated in children with allergic diseases compared to normal children (1.2+/− 0.8 vs 0.06 +/− 0.5 ng/ml) [77]. Serum CPA3 levels have also been reported to be elevated in those with a clinical diagnosis of anaphylaxis but not in the serum of adult healthy blood donors or individuals with a diagnosis of asthma of another IgE-mediated allergic disease. The serum levels of tryptase and of CPA3 after anaphylaxis do not necessarily correlate [78]. CPA3 levels appear to remain elevated longer than the tryptase levels. Further, CPA3 serum levels have also been reported to be detected in individuals with anaphylaxis where elevations in total serum tryptase levels were not observed.

Prostaglandin D2 and cysteinyl leukotrienes

Prostaglandin D2 (PGD2) and cysteinyl leukotrienes (Cyst LTs) are the major lipid mediators synthesized after mast cell activation [79]. They are released shortly after the granule contents as part of the immediate mast cell response. Prostaglandins and leukotrienes are synthesized from arachidonic acid (AA) which is released by the action of cytosolic phospholipase A2 on membrane phospholipids [80].

In the PG pathway, AA is first converted to PGG2 by cycloxygenase (COX)-1 and COX-2, and then reduced to PGH2. The latter serves as the precursor for PGD2 as well as other prostanoids PGE2, PGF2a, PGI2, and thromboxane A2 through terminal PG synthases. PGD2 is also produced by eosinophils and in lesser quantities by other immune cells such as Th2 cells and dendritic cells. Non-hematopoietic tissues such as brain, heart, lungs and kidneys can also produce PGD2 via lipocalin type PGD2 synthase. PGD2 exerts its biologic actions by binding to two receptors named DP1 and DP2. End organ functions of PGD2 in humans include vasodilation and bronchoconstriction. Pulmonary, nasal and ocular allergen challenges result in increased levels of PGD2 in relevant biologic fluids. PGD2 and its metabolite 11b-PGF2a are found increased in the urine of patients with mastocytosis and mast cell activation syndrome [81, 82]. The diagnostic and therapeutic clinical utility of PGD2 as a marker of mast cell activation has been limited by its production by cells other than mast cells.

Historically known as the slow reacting substance of anaphylaxis [83], LTC4, LTD4 and LTE4 are collectively termed as CysLTs. 5 lipoxygenase (5-LO) along with the perinuclear membrane protein called 5-LO activating protein convert AA to 5-hydroxyperoxyeicosotetraenoic acid which then gets dehydrated to the unstable leukotriene precursor LTA4. LTA4 is then conjugated to reduced gluthatione by LTC4 synthase, which is secreted out of the cells. LTC4 is converted first to LTD4, then to the most stable form LTE4 extracellularly. CysLTs are generated by mast cells, basophils, eosinophils, macrophages and myeloid dendritic cells. There are at least 3 CysLT receptors: CysLT1R, CysLT2R and CysLT3R (GPR99) [84]. As a mediator associated with allergic inflammation, LTC4’s biological importance resides in its capacity to induce smooth muscle contraction at a concentration that is on the order of 100–6000 times lower than for histamine, and this contraction also lasts substantially longer when initiated with LTC4 than with histamine. CysLTs also induce wheal and flare reactions (increased vascular permeability) in humans. Urinary LTE4 was found to be elevated in systemic mastocytosis and correlated with 24 h urine N-methylhistamine and serum tryptase levels [85].

CD25, CD63, CD203c, FcεR1

The stem cell factor receptor KIT (CD117) is expressed on on mast cells independent of tissue site, maturation or activation status. In systemic mastocytosis, KIT is often expressed in mast cells in a mutated and constitutively activated form. CD63 and CD203c expression are upregulated following aggregation of FcεRI, and they are overexpressed on neoplastic mast cells in systemic mastocytosis [86]. Mast cells in systemic mastocytosis express CD30 and CD25.

CD63 belongs to a family of tetraspanins, which comprise a superfamily of cell surface-associated membrane proteins characterized by four transmembrane domains [87]. At the cell surface, tetraspanins form networks with a diversity of proteins, including cell surface receptors (MHCII, CD3, FcεRI, CXCR4), kinases (phosphatidylinositol 4-kinase and the Src family tyrosine kinases Lyn and Hck), integrins (α4β1, α3β1, α6β1, LFA-1 and β2), and other tetraspanins (CD81, CD82, CD9, CD151) [87]. The networks of tetraspanins with proteins are called tetraspanin-enriched microdomains. CD63 is also abundantly present in late endosomes and lysosomes [87]. CD63 at the cell surface is endocytosed via a clathrin-dependent pathway. In late endosomes, CD63 is enriched on the intraluminal vesicles, which are secreted by specialized cells as exosomes through fusion of endosomes with the plasma membrane [87]. CD63 has been identified as an activation marker for basophils and mast cells [88]. CD63 expression is rapidly upregulated following allergen challenge and reaches a maximum after 20–30 min [89]. CD63 is also expressed on neutrophils, dendritic cells, T cells and eosinophils [88]. Anti-CD63 antibodies inhibit FcεRI-induced mast cell degranulation in vitro and FcεRI-induced allergic reaction in vivo [90, 91].

CD203c (E-NPP3) belongs to a family of ecto-nucleotide pyrophosphates/phosphodiesterases (E-NPPs). E-NPPs catalyze the cleavage of phosphodiester and phosphosulfate bonds of molecules, including deoxynucleotides, NAD and nucleotide sugars [92]. E-NPP3 is a member of the family of type II transmembrane proteins, which are composed of a short N-terminal cytoplasmic domain, followed by the transmembrane region, two somatomedin-like domains, the catalytic domain and a C-terminal endonuclease-like domain. CD203c is associated with malignant subversion and invasive properties [93]. CD203c has been defined as an activation-linked surface antigen on basophils and mast cells that is upregulated in response to IgE receptor cross-linking and is overexpressed on neoplastic mast cells in patients with systemic mastocytosis [94]. CD203c expression on basophils may implicate these cells in some allergic diseases [95].

CD30 is a member of the tumor necrosis factor receptor/nerve growth factor (TNFR/NGFR) superfamily [96]. Ligation of CD30 ligand (CD30L or CD153) to CD30 elicits multidirectional signals leading to either cell activation or apoptosis [96]. Under physiological conditions, expression of CD30 is restricted to T and B cells, mainly to activated Th2 cells. CD30 is expressed typically on the surface of Hodgkin’s Reed-Sternberg cells and anaplastic large cell lynphomas [96]. Human mast cells from normal donors do not express CD30. CD30 expression is upregulated aberrantly in most indolent and aggressive forms of systemic mastocytosis [97].

CD25 is α chain of IL-2 receptor. The high affinity IL-2 receptor (IL-2R) is a heterotrimer consisting of the IL-2R α chain (IL-2Rα, CD25) and the IL-2R β- and γ chains (IL-2Rβ and IL-2Rγ) [98]. CD25 serves as a major growth factor receptor by binding IL-2. The IL-2Rα does not contain an intracellular signaling domain, therefore binding to IL-2Rα alone does not result in T cell activation [98]. The high affinity IL-2R heterotrimer is expressed on activated T cells and regulatory T cells [98]. Mast cells from normal donors do not express CD25 during maturation and activation [99]. However, mast cells in systemic mastocytosis aberrantly display CD25, which is a diagnostic marker of neoplastic mast cells in systemic mastocytosis variants and in platelet-derived growth factor receptor alpha (PDGFRA)-associated myeloproliferative disorders [86].

Under allergic inflammatory conditions, “primed” mast cells produce IL-4 and IL-13 and express high levels of the FcεRI and the ligand for the surface antigen CD40, involved in T/B cell interactions leading to IgE production [100, 101]. IL-4 from mast cells can direct uncommitted helper T lymphocytes toward Th2 and also upregulate the FcεRI expression in mast cells [100]. Furthermore, locally synthesized IgE can upregulate the FcεRI expression on mast cells [102]. The augmented FcεRI on mast cells can bind an increased number of IgE-Ag complexes which in turn can enhance the sensitivity of mast cells to allergen resulting in the enhancement of the production of immunomodulatory cytokines /chemical mediators [103]. The upregulation of FcεRI is observed in mast cells from atopic asthma [104, 105]. By contrast, alveolar mast cells may lack expression of FcεRI in healthy subjects [106]. Supporting the emerging concept of alveolar inflammation in asthma, alveolar mast cells shift to a FcεRI-expressing phenotype in atopic and uncontrolled asthma [107]. Site-specific and disease-associated mast cell changes have also recently been described in most other inflammatory conditions of the lung.

Thus a robust upregulation of FcεRI-expression (and surface-bound IgE) on alveolar mast cells and nasal mast cells is a novel disease-specific feature of atopic asthma and AR. Changes in the alveolar mast cells in allergic asthma are associated with a Th2-skewed alveolar cell profile, which correlates significantly with the degree of clinical control [108, 109].

Basophils

Histamine and its metabolites as biomarkers of basophils

Histamine [2-(4-Imidazolyl)-ethylamine] is stored in the granules of basophils, as well as in mast cells, neurons and enterochromaffin-like (ECL) cells in the stomach. Platelets also contain histamine, and it can be produced also by certain bacteria, including those commensal in the upper respiratory tract. Oxidative deamination of histamine by diamine oxidase, yields ammonia, hydrogen peroxide, and imidazole acetaldehyde. Neither degradation product binds to one of the four G-protein coupled histamine receptors (HR1-4), and thus these products have no known pharmacological activities.

Due to its storage in secretory basophil granules and release during immunological activation, histamine can be considered as a biomarker of basophil activation in vitro. However, its practical usefulness as a basophil activation marker is limited by several issues. Technically, histamine assays are relatively laborious and expensive, as in the histamine ELISA detection methods, or require specialised equipment such as histamine autoanalyzers. Moreover, collecting plasma at an optimal time is challenging, because histamine undergoes first pass metabolism in the circulation, peaking about five minutes after onset of insect sting-initiated anaphylaxis and returning to baseline within 15 to 30 min. Also, plasma rather than serum is preferred for histamine measurements, because of concerns over basophil activation during blood clotting, and avoidance of small bore venipuncture needles and vacuum tubes is also recommended to decrease the likelihood of basophil shearing during blood collection. Further, traditional histamine measurements are bulk-type measurements which average the content or release of histamine across the studied cell population. Finally, while histamine is stored in large amounts in the granules of mature basophils (and mast cells), it is also secreted by haematopoietic progenitors [110] before being stored in the granules. Accordingly, it is also produced by other non-mast cell/non-basophil cells such as eosinophils, platelets, neutrophils, in addition to the aforementioned neurons, microglia and enterochromaffin-like cells. Thus, histamine elevations per se do not unequivocally identify the basophil as the source of the histamine. Increased histamine concentrations in the absence of concomitant rises in levels of other mast cell mediators, e.g., PGD2, can implicate infiltrating basophils as the primary source in allergen-induced late phase reactions [111]. Histamine released from blood cells will in most cases be of basophil origin, though assessment of basophil activation to allergens in vitro is more commonly carried out at present by measuring membrane expression of CD63 and/or CD203c by flow cytometry [112].

As an alternative to measurements of extracellular histamine or its metabolites, a HistaFlow technique using an enzyme-affinity-fluorochrome method using fluorescently labelled DAO to measure intracellular histamine [113], is able to capture data at the single cell level, has been used to monitor basophil responsiveness during immunotherapy [114], potentially serving as a surrogate biomarker for monitoring immunotherapy efficacy.

LTC4 and its metabolites

Basophils rapidly synthesize LTC4 within minutes upon allergen binding to specific IgE, occupying high affinity receptors (FcεRI), as well as to other diverse stimuli [115, 116]. Phospholipids in basophils, including phosphatidylcholine and phosphatidylinositol, likely contribute to the arachidonic acid source from which LTC4 is then derived via the actions of calcium-sensitive 5-lipoxigenase. However, there are a variety of other cell types potentially contributing to the production of LTC4 in vivo, including mast cells, eosinophils and macrophages [117]. Nonetheless, its synthesis and secretion from washed leukocytes has long functioned as an in vitro/ex vivo assay to evaluate the allergic status, since basophils within these suspensions are the sole source of LTC4 following IgE/FcεRI-dependent activation [118]. The relative ease of measuring LTC4 in culture supernatants using commercially available enzymatic immunoassays (EIA) further enhances the utility of this mediator as a marker of basophil activation in vitro.

Monoclonal antibody, J175-7D4, to pro-major basic protein 1

Studies of basophils [119] have been hampered both by the paucity of this cell type and by the difficulty of finding it in tissue. The monoclonal J175-7D4 antibody binds the propeptide region of bone marrow proteoglycan (gene name PRG2), but not the mature eosinophil granule major basic protein after removal of the propeptide [120]. Immunostaining with J175-7D4 is basophil-specific in human skin, nasal polyps, and esophagus. J175-7D4 does not stain mature mast cells or eosinophils; however, it does stain eosinophil precursors. Clone 2D7 antibody, commercially available, also recognizes a precursor form of eosinophil granule major basic protein, labelling only basophils, and has been used to identify basophils in tissues, including formalin-fixed, paraffin-embedded specimens [121], and in suspension by flow cytometry [122]. However, in our experience, 2D7 staining is inconsistent and can be weak in formalin-fixed paraffin-embedded tissue, and basophil staining is consistently brighter with J175-7D4 than with 2D7. In formalin-fixed, paraffin-embedded tissue, 7D4 antibody works well after basic EDTA antigen retrieval.

Surface activation markers

Basophils are the least common of the classically described peripheral blood leukocyte populations. Because of their expression of FcεRI and easy accessibility in peripheral blood, basophils are often used as an experimental system to examine IgE mediated cell activation. Additionally, basophils are clearly an important allergic effector cell population in their own right [123].

Basophils can be physically separated from peripheral blood mononuclear cells and their production of soluble inflammatory mediators studied in bulk culture, in a manner similar to that noted above for mast cells. The ability to identify basophils using multicolor flow cytometry has facilitated their study in whole blood without such labor-intensive separation. In such work, basophils are typically identified by their expression of cell surface markers such as CD123, CCR3, or CRTH2 [124, 125], usually in combination with their low side-scatter properties [126].

The two best-described markers of human basophil activation are CD63 and CD203c [112, 124, 125]. In resting basophils, CD63 is located primarily in secretory granule membranes, and CD203c, reportedly in membranes of vesicles that are rapidly expressed after activation, are both translocated to the cell surface when basophils are activated to degranulate, e.g., after aggregation of FcεRI, where they can be detected by flow cytometry [89]. Bimodal surface expression of CD63 is typically detected on allergen-activated basophils in whole blood, whereas CD203c expression is seen on nearly all basophils regardless of their purity. Surface CD203c but not CD63 may be detected after activation that does not cause degranulation, e.g., after stimulation with IL-3 alone [127]. Thus, CD63 and CD203c are complementary for assessing basophil activation. In addition to CD63 and CD203c, multiple other granule membrane associated proteins, including CD13, CD107a, and CD164, exhibit similar upregulation [128]. These markers fall into two families that respectively represent anaphylactic and piecemeal degranulation processes [129].

Basogranulin

Basogranulin was initially described as a high molecular weight granule protein that was a specific marker of basophils [130]. Basogranulin is secreted from granules after both IgE- and non-IgE mediated stimuli [131]. Basogranulin can be used as a specific immunohistochemical marker to identify basophils in tissue [132].

Conclusions

Mast cells, eosinophils and basophils play a fundamental role in several inflammatory, allergic and autoimmune diseases and they contribute to defense against infectious organisms and to development of many types of cancer. The role of these cells in human diseases may be beneficial, for example in infectious diseases, or detrimental, as in allergic disorders. Thus, proper assessment of the degree of activation of these cells in vivo is desirable to monitor the course of disease and to assess the disease response to therapy. In this review, a number of molecules have been discussed that are biomarkers of the activation of mast cells, eosinophils and basophils. There is good evidence that these surrogate markers are useful in assessing the state of activation in vivo of mast cells, eosinophils and basophils in a variety of human diseases. In most cases these markers reflect the involvement of a given cell in a particular disease and their levels in blood or body fluids often correlate to the severity of the disease. Some of the markers are relatively cell-specific, e.g., cationic proteins for eosinophils and tryptase for mast cells. Others, such as histamine and cysteinyl leukotrienes, are not cell-specific.

In some cases the biomarkers are clearly reflective of their involvement in the pathogenesis of a disease. For example, histamine in allergic rhinitis and cysteinyl leukotrienes in asthma are both associated with pathology and respond to pharmacologic intervention. In the case of other markers, their role, if any, in the disease pathogenesis is still unclear. Surface markers and activation molecules expressed on the membrane of mast cells, eosinophils and basophils can, in some cases, be used for in vitro diagnosis of allergic diseases. Tryptase and ECP are considered useful diagnostic tools to measure mast cell and eosinophil involvement in mastocytosis and in hypereosinophilic syndromes. Many biomarkers of mast cell, eosinophil and basophil activation are modulated by pharmacologic treatments and, therefore, in some instances are used to monitor the response of disease to treatment.

While there are currently many good candidates as surrogate markers of mast cell, eosinophil and basophil activation in vivo, there is still work that must be done to fully establish the usefulness of these markers in clinical practice. First, the best techniques to measure these molecules should be employed and made more widely available for use in clinical studies and in the management of clinical disease. Second, we should better understand the significance of biomarkers of human inflammation and we should define how they correlate with specific clinical phenotypes. Finally, a better understanding of the mechanisms regulating the secretion and expression of biomarkers and their pharmacological modulation will have practical application in the management of diseases associated with mast cell, eosinophil and basophil activation.

Abbreviations

ACE:

angiotensin-converting enzyme

BAL:

bronchoalveolar lavage

CA3:

carboxypeptidase A3

CCR:

c-c motif receptor

CD:

cluster of differentiation

CEACAM:

carcinoembryonic antigen-related cell adhesion molecule

CLC:

Charcot-Leyden crystal protein

COX-1:

cycloxygenase-1

CRTH2:

chemoattractant receptor-homologous molecule expressed on th2 cells

Cyst LTs:

cysteinyl leukotrienes

DCs:

human dendritic cells

ECE:

endothelin converting-enzyme

ECL:

enterochromaffin-like cells

ECP:

eosinophil cationic protein

EDN:

eosinophil-derived neurotoxin

EIA:

enzymatic immunoassays

EoE:

eosinophilic eosophagitis

EPX:

eosinophil peroxidase

Fc:

fragment crystallizable (of immunoglobulin)

FENO:

fraction of exhaled nitric oxide

FEV1:

forced expiratory volume in 1 s

FVC:

forced vital capacity

HPLC:

high-performance liquid chromatography

ICAM:

intercellular adhesion molecule

ICS:

inhaled corticosteroid

IL:

interleukin

ILA:

induced by lymphocyte activation

KIT:

stem cell factor receptor

LO:

lipoxygenase

LT:

leukotriene

mAb:

monoclonal antibody

MBP:

major basic protein

PAPP-A:

pregnancy-associated plasma protein-A

PC20:

provocative concentration of methacholine or histamine producing a 20 % fall in FEV1

PDGFRA:

platelet-derived growth factor receptor alpha

PGD2:

prostaglandin D2

PSGL:

P-selectin glycoprotein ligand

R:

receptor

RNase:

ribonuclease

ROC:

receiver-operating characteristic

SLPI:

secretory leukocyte protease inhibitor

SNPs:

single nucleotide polymorphisms

Tele-MIAA:

1-methyl-4-imidazole acetic acid

TNFR:

tumor necrosis factor receptor

TNFRSF:

tumor necrosis factor receptor superfamily member.

Tregs:

human regulatory T cells

References

  1. Johansson MW. Activation states of blood eosinophils in asthma. Clin Exp Allergy. 2014;44(4):482–98. doi:10.1111/cea.12292.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  2. Johansson MW, Mosher DF. Integrin activation states and eosinophil recruitment in asthma. Front Pharmacol. 2013;4:33. doi:10.3389/fphar.2013.00033.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  3. Rosenberg HF, Dyer KD, Foster PS. Eosinophils: changing perspectives in health and disease. Nat Rev Immunol. 2013;13(1):9–22. doi:10.1038/nri3341.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  4. Driss V, Legrand F, Capron M. Eosinophils receptor profile. In: Lee J, Rosenberg HF, editors. Eosinophils in health and disease. 1st ed. Waltham: Elsevier/Academic Press; 2013. p. 30–8.

    Google Scholar 

  5. Johnsson M, Bove M, Bergquist H, Olsson M, Fornwall S, Hassel K, et al. Distinctive blood eosinophilic phenotypes and cytokine patterns in eosinophilic esophagitis, inflammatory bowel disease and airway allergy. J Innate Immun. 2011;3(6):594–604. doi:10.1159/000331326.

    Article  CAS  PubMed  Google Scholar 

  6. Lingblom C, Bergquist H, Johnsson M, Sundstrom P, Quiding-Jarbrink M, Bove M, et al. Topical corticosteroids do not revert the activated phenotype of eosinophils in eosinophilic esophagitis but decrease surface levels of CD18 resulting in diminished adherence to ICAM-1, ICAM-2, and endothelial cells. Inflammation. 2014;37(6):1932–44. doi:10.1007/s10753-014-9926-x.

    Article  CAS  PubMed  Google Scholar 

  7. Simon HU, Plotz S, Simon D, Seitzer U, Braathen LR, Menz G, et al. Interleukin-2 primes eosinophil degranulation in hypereosinophilia and Wells’ syndrome. Eur J Immunol. 2003;33(4):834–9. doi:10.1002/eji.200323727.

    Article  CAS  PubMed  Google Scholar 

  8. Straumann A, Kristl J, Conus S, Vassina E, Spichtin HP, Beglinger C, et al. Cytokine expression in healthy and inflamed mucosa: probing the role of eosinophils in the digestive tract. Inflamm Bowel Dis. 2005;11(8):720–6.

    Article  PubMed  Google Scholar 

  9. Hansel TT, Braunstein JB, Walker C, Blaser K, Bruijnzeel PL, Virchow Jr JC, et al. Sputum eosinophils from asthmatics express ICAM-1 and HLA-DR. Clin Exp Immunol. 1991;86(2):271–7.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  10. Toma T, Mizuno K, Okamoto H, Kanegane C, Ohta K, Ikawa Y, et al. Expansion of activated eosinophils in infants with severe atopic dermatitis. Pediatr Int. 2005;47(1):32–8. doi:10.1111/j.1442-200x.2004.02004.x.

    Article  PubMed  Google Scholar 

  11. Stein ML, Collins MH, Villanueva JM, Kushner JP, Putnam PE, Buckmeier BK, et al. Anti-IL-5 (mepolizumab) therapy for eosinophilic esophagitis. J Allergy Clin Immunol. 2006;118(6):1312–9. doi:10.1016/j.jaci.2006.09.007.

    Article  CAS  PubMed  Google Scholar 

  12. Kita H, Adolphoson CR, Gleich GJ. Biology of eosinophils. In: Adkinson NF, Yunginger JW, Busse WW, Bochner BS, Holgate ST, Simons FER, editors. Middleton’s allergy: principles & practice. 6th ed. St. Louis: Mosby; 2003. p. 305–32.

    Google Scholar 

  13. Gleich GJ, Loegering DA, Mann KG, Maldonado JE. Comparative properties of the Charcot-Leyden crystal protein and the major basic protein from human eosinophils. J Clin Invest. 1976;57(3):633–40. doi:10.1172/JCI108319.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  14. Barker RL, Gleich GJ, Pease LR. Acidic precursor revealed in human eosinophil granule major basic protein cDNA. J Exp Med. 1988;168(4):1493–8.

    Article  CAS  PubMed  Google Scholar 

  15. Plager DA, Loegering DA, Weiler DA, Checkel JL, Wagner JM, Clarke NJ, et al. A novel and highly divergent homolog of human eosinophil granule major basic protein. J Biol Chem. 1999;274(20):14464–73.

    Article  CAS  PubMed  Google Scholar 

  16. Soragni A, Yousefi S, Stoeckle C, Soriaga AB, Sawaya MR, Kozlowski E, et al. Toxicity of eosinophil MBP is repressed by intracellular crystallization and promoted by extracellular aggregation. Mol Cell. 2015;57(6):1011–21. doi:10.1016/j.molcel.2015.01.026.

    Article  CAS  PubMed  Google Scholar 

  17. Woschnagg C, Rubin J, Venge P. Eosinophil cationic protein (ECP) is processed during secretion. J Immunol. 2009;183(6):3949–54. doi:10.4049/jimmunol.0900509.

    Article  CAS  PubMed  Google Scholar 

  18. Koh GC, Shek LP, Goh DY, Van Bever H, Koh DS. Eosinophil cationic protein: is it useful in asthma? A systematic review. Respir Med. 2007;101(4):696–705. doi:10.1016/j.rmed.2006.08.012.

    Article  PubMed  Google Scholar 

  19. Lowhagen O, Wever AM, Lusuardi M, Moscato G, De Backer WA, Gandola L, et al. The inflammatory marker serum eosinophil cationic protein (ECP) compared with PEF as a tool to decide inhaled corticosteroid dose in asthmatic patients. Respir Med. 2002;96(2):95–101.

    Article  CAS  PubMed  Google Scholar 

  20. Bjork A, Venge P, Peterson CG. Measurements of ECP in serum and the impact of plasma coagulation. Allergy. 2000;55(5):442–8.

    Article  CAS  PubMed  Google Scholar 

  21. Plotz SG, Simon HU, Darsow U, Simon D, Vassina E, Yousefi S, et al. Use of an anti-interleukin-5 antibody in the hypereosinophilic syndrome with eosinophilic dermatitis. N Engl J Med. 2003;349(24):2334–9. doi:10.1056/NEJMoa031261.

    Article  PubMed  Google Scholar 

  22. Sur S, Glitz DG, Kita H, Kujawa SM, Peterson EA, Weiler DA, et al. Localization of eosinophil-derived neurotoxin and eosinophil cationic protein in neutrophilic leukocytes. J Leukoc Biol. 1998;63(6):715–22.

    CAS  PubMed  Google Scholar 

  23. Wang JG, Mahmud SA, Thompson JA, Geng JG, Key NS, Slungaard A. The principal eosinophil peroxidase product, HOSCN, is a uniquely potent phagocyte oxidant inducer of endothelial cell tissue factor activity: a potential mechanism for thrombosis in eosinophilic inflammatory states. Blood. 2006;107(2):558–65. doi:10.1182/blood-2005-05-2152.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  24. Henderson WR, Jorg A, Klebanoff SJ. Eosinophil peroxidase-mediated inactivation of leukotrienes B4, C4, and D4. J Immunol. 1982;128(6):2609–13.

    CAS  PubMed  Google Scholar 

  25. Ulrich M, Petre A, Youhnovski N, Promm F, Schirle M, Schumm M, et al. Post-translational tyrosine nitration of eosinophil granule toxins mediated by eosinophil peroxidase. J Biol Chem. 2008;283(42):28629–40. doi:10.1074/jbc.M801196200.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  26. Ackerman SJ, Corrette SE, Rosenberg HF, Bennett JC, Mastrianni DM, Nicholson-Weller A, et al. Molecular cloning and characterization of human eosinophil Charcot-Leyden crystal protein (lysophospholipase). Similarities to IgE binding proteins and the S-type animal lectin superfamily. J Immunol. 1993;150(2):456–68.

    CAS  PubMed  Google Scholar 

  27. Weller PF, Bach DS, Austen KF. Biochemical characterization of human eosinophil Charcot-Leyden crystal protein (lysophospholipase). J Biol Chem. 1984;259(24):15100–5.

    CAS  PubMed  Google Scholar 

  28. Leonidas DD, Elbert BL, Zhou Z, Leffler H, Ackerman SJ, Acharya KR. Crystal structure of human Charcot-Leyden crystal protein, an eosinophil lysophospholipase, identifies it as a new member of the carbohydrate-binding family of galectins. Structure. 1995;3(12):1379–93.

    Article  CAS  PubMed  Google Scholar 

  29. Ackerman SJ, Weil GJ, Gleich GJ. Formation of Charcot-Leyden crystals by human basophils. J Exp Med. 1982;155(6):1597–609.

    Article  CAS  PubMed  Google Scholar 

  30. Ackerman SJ, Liu L, Kwatia MA, Savage MP, Leonidas DD, Swaminathan GJ, et al. Charcot-Leyden crystal protein (galectin-10) is not a dual function galectin with lysophospholipase activity but binds a lysophospholipase inhibitor in a novel structural fashion. J Biol Chem. 2002;277(17):14859–68. doi:10.1074/jbc.M200221200.

    Article  CAS  PubMed  Google Scholar 

  31. Kubach J, Lutter P, Bopp T, Stoll S, Becker C, Huter E, et al. Human CD4 + CD25+ regulatory T cells: proteome analysis identifies galectin-10 as a novel marker essential for their anergy and suppressive function. Blood. 2007;110(5):1550–8. doi:10.1182/blood-2007-01-069229.

    Article  CAS  PubMed  Google Scholar 

  32. Acharya KR, Ackerman SJ. Eosinophil granule proteins: form and function. J Biol Chem. 2014;289(25):17406–15. doi:10.1074/jbc.R113.546218.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  33. Furuta GT, Kagalwalla AF, Lee JJ, Alumkal P, Maybruck BT, Fillon S, et al. The oesophageal string test: a novel, minimally invasive method measures mucosal inflammation in eosinophilic oesophagitis. Gut. 2013;62(10):1395–405. doi:10.1136/gutjnl-2012-303171.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  34. Chua JC, Douglass JA, Gillman A, O’Hehir RE, Meeusen EN. Galectin-10, a potential biomarker of eosinophilic airway inflammation. PLoS One. 2012;7(8):e42549. doi:10.1371/journal.pone.0042549.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  35. De Re V, Simula MP, Cannizzaro R, Pavan A, De Zorzi MA, Toffoli G, et al. Galectin-10, eosinophils, and celiac disease. Ann N Y Acad Sci. 2009;1173:357–64. doi:10.1111/j.1749-6632.2009.04627.x.

    Article  PubMed  CAS  Google Scholar 

  36. Bryborn M, Hallden C, Sall T, Cardell LO. CLC- a novel susceptibility gene for allergic rhinitis? Allergy. 2010;65(2):220–8. doi:10.1111/j.1398-9995.2009.02141.x.

    Article  CAS  PubMed  Google Scholar 

  37. Abu-Ghazaleh RI, Dunnette SL, Loegering DA, Checkel JL, Kita H, Thomas LL, et al. Eosinophil granule proteins in peripheral blood granulocytes. J Leukoc Biol. 1992;52(6):611–8.

    CAS  PubMed  Google Scholar 

  38. Durack DT, Ackerman SJ, Loegering DA, Gleich GJ. Purification of human eosinophil-derived neurotoxin. Proc Natl Acad Sci U S A. 1981;78(8):5165–9.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  39. Domachowske JB, Dyer KD, Bonville CA, Rosenberg HF. Recombinant human eosinophil-derived neurotoxin/RNase 2 functions as an effective antiviral agent against respiratory syncytial virus. J Infect Dis. 1998;177(6):1458–64.

    Article  CAS  PubMed  Google Scholar 

  40. Sorrentino S, Glitz DG, Hamann KJ, Loegering DA, Checkel JL, Gleich GJ. Eosinophil-derived neurotoxin and human liver ribonuclease. Identity of structure and linkage of neurotoxicity to nuclease activity. J Biol Chem. 1992;267(21):14859–65.

    CAS  PubMed  Google Scholar 

  41. Yang D, Chen Q, Rosenberg HF, Rybak SM, Newton DL, Wang ZY, et al. Human ribonuclease A superfamily members, eosinophil-derived neurotoxin and pancreatic ribonuclease, induce dendritic cell maturation and activation. J Immunol. 2004;173(10):6134–42.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  42. Yang D, Chen Q, Su SB, Zhang P, Kurosaka K, Caspi RR, et al. Eosinophil-derived neurotoxin acts as an alarmin to activate the TLR2-MyD88 signal pathway in dendritic cells and enhances Th2 immune responses. J Exp Med. 2008;205(1):79–90. doi:10.1084/jem.20062027.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  43. Ackerman SJ, Kephart GM, Francis H, Awadzi K, Gleich GJ, Ottesen EA. Eosinophil degranulation. An immunologic determinant in the pathogenesis of the Mazzotti reaction in human onchocerciasis. J Immunol. 1990;144(10):3961–9.

    CAS  PubMed  Google Scholar 

  44. Konikoff MR, Blanchard C, Kirby C, Buckmeier BK, Cohen MB, Heubi JE, et al. Potential of blood eosinophils, eosinophil-derived neurotoxin, and eotaxin-3 as biomarkers of eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2006;4(11):1328–36. doi:10.1016/j.cgh.2006.08.013.

    Article  CAS  PubMed  Google Scholar 

  45. Subbarao G, Rosenman MB, Ohnuki L, Georgelas A, Davis M, Fitzgerald JF, et al. Exploring potential noninvasive biomarkers in eosinophilic esophagitis in children. J Pediatr Gastroenterol Nutr. 2011;53(6):651–8. doi:10.1097/MPG.0b013e318228cee6.

    CAS  PubMed  Google Scholar 

  46. Kim CK, Seo JK, Ban SH, Fujisawa T, Kim DW, Callaway Z. Eosinophil-derived neurotoxin levels at 3 months post-respiratory syncytial virus bronchiolitis are a predictive biomarker of recurrent wheezing. Biomarkers. 2013;18(3):230–5. doi:10.3109/1354750X.2013.773078.

    Article  CAS  PubMed  Google Scholar 

  47. Schulman ES, Kagey-Sobotka A, MacGlashan Jr DW, Adkinson Jr NF, Peters SP, Schleimer RP, et al. Heterogeneity of human mast cells. J Immunol. 1983;131(4):1936–41.

    CAS  PubMed  Google Scholar 

  48. Casale TB, Wood D, Richerson HB, Trapp S, Metzger WJ, Zavala D, et al. Elevated bronchoalveolar lavage fluid histamine levels in allergic asthmatics are associated with methacholine bronchial hyperresponsiveness. J Clin Invest. 1987;79(4):1197–203. doi:10.1172/JCI112937.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  49. Kaplan AP, Horakova Z, Katz SI. Assessment of tissue fluid histamine levels in patients with urticaria. J Allergy Clin Immunol. 1978;61(6):350–4.

    Article  CAS  PubMed  Google Scholar 

  50. Pollock I, Murdoch RD, Lessof MH. Plasma histamine and clinical tolerance to infused histamine in normal, atopic and urticarial subjects. Agents Actions. 1991;32(3–4):359–65.

    Article  CAS  PubMed  Google Scholar 

  51. Granerus G, Lonnqvist B, Wass U. Determination of the histamine metabolite tele-methylimidazoleacetic acid and of creatinine in urine by the same HPLC system. Inflamm Res. 1999;48(2):75–80.

    Article  CAS  PubMed  Google Scholar 

  52. Keyzer JJ, de Monchy JG, van Doormaal JJ, van Voorst Vader PC. Improved diagnosis of mastocytosis by measurement of urinary histamine metabolites. N Engl J Med. 1983;309(26):1603–5. doi:10.1056/NEJM198312293092603.

    Article  CAS  PubMed  Google Scholar 

  53. Kolmert J, Forngren B, Lindberg J, Ohd J, Aberg KM, Nilsson G, et al. A quantitative LC/MS method targeting urinary 1-methyl-4-imidazoleacetic acid for safety monitoring of the global histamine turnover in clinical studies. Anal Bioanal Chem. 2014;406(6):1751–62. doi:10.1007/s00216-013-7594-6.

    Article  CAS  PubMed  Google Scholar 

  54. Metcalfe DD, Lewis RA, Silbert JE, Rosenberg RD, Wasserman SI, Austen KF. Isolation and characterization of heparin from human lung. J Clin Invest. 1979;64(6):1537–43. doi:10.1172/JCI109613.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  55. Metcalfe DD, Soter NA, Wasserman SI, Austen KF. Identification of sulfated mucopolysaccharides including heparin in the lesional skin of a patient with mastocytosis. J Invest Dermatol. 1980;74(4):210–5.

    Article  CAS  PubMed  Google Scholar 

  56. Sucker C, Mansmann G, Steiner S, Gattermann N, Schmitt-Graeff A, Loncar R, et al. Fatal bleeding due to a heparin-like anticoagulant in a 37-year-old woman suffering from systemic mastocytosis. Clin Appl Thromb Hemost. 2008;14(3):360–4. doi:10.1177/1076029607309173.

    Article  CAS  PubMed  Google Scholar 

  57. Seidel H, Molderings GJ, Oldenburg J, Meis K, Kolck UW, Homann J, et al. Bleeding diathesis in patients with mast cell activation disease. Thromb Haemost. 2011;106(5):987–9. doi:10.1160/TH11-05-0351.

    Article  CAS  PubMed  Google Scholar 

  58. Oschatz C, Maas C, Lecher B, Jansen T, Bjorkqvist J, Tradler T, et al. Mast cells increase vascular permeability by heparin-initiated bradykinin formation in vivo. Immunity. 2011;34(2):258–68. doi:10.1016/j.immuni.2011.02.008.

    Article  CAS  PubMed  Google Scholar 

  59. Pallaoro M, Fejzo MS, Shayesteh L, Blount JL, Caughey GH. Characterization of genes encoding known and novel human mast cell tryptases on chromosome 16p13.3. J Biol Chem. 1999;274(6):3355–62.

    Article  CAS  PubMed  Google Scholar 

  60. Schwartz LB. Diagnostic value of tryptase in anaphylaxis and mastocytosis. Immunol Allergy Clin North Am. 2006;26(3):451–63. doi:10.1016/j.iac.2006.05.010.

    Article  PubMed  Google Scholar 

  61. Le QT, Gomez G, Zhao W, Hu J, Xia HZ, Fukuoka Y, et al. Processing of human protryptase in mast cells involves cathepsins L, B, and C. J Immunol. 2011;187(4):1912–8. doi:10.4049/jimmunol.1001806.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  62. Le QT, Min HK, Xia HZ, Fukuoka Y, Katunuma N, Schwartz LB. Promiscuous processing of human alphabeta-protryptases by cathepsins L, B, and C. J Immunol. 2011;186(12):7136–43. doi:10.4049/jimmunol.1001804.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  63. Sverrild A, van der Sluis S, Kyvik KO, Garvey LH, Porsbjerg C, Backer V, et al. Genetic factors account for most of the variation in serum tryptase--a twin study. Ann Allergy Asthma Immunol. 2013;111(4):286–9. doi:10.1016/j.anai.2013.07.011.

    Article  CAS  PubMed  Google Scholar 

  64. Alvarez-Twose I, Zanotti R, Gonzalez-de-Olano D, Bonadonna P, Vega A, Matito A, et al. Nonaggressive systemic mastocytosis (SM) without skin lesions associated with insect-induced anaphylaxis shows unique features versus other indolent SM. J Allergy Clin Immunol. 2014;133(2):520–8. doi:10.1016/j.jaci.2013.06.020.

    Article  PubMed  Google Scholar 

  65. Bonadonna P, Perbellini O, Passalacqua G, Caruso B, Colarossi S, Dal Fior D, et al. Clonal mast cell disorders in patients with systemic reactions to Hymenoptera stings and increased serum tryptase levels. J Allergy Clin Immunol. 2009;123(3):680–6. doi:10.1016/j.jaci.2008.11.018.

    Article  CAS  PubMed  Google Scholar 

  66. Valent P, Akin C, Arock M, Brockow K, Butterfield JH, Carter MC, et al. Definitions, criteria and global classification of mast cell disorders with special reference to mast cell activation syndromes: a consensus proposal. Int Arch Allergy Immunol. 2012;157(3):215–25. doi:10.1159/000328760.

    Article  PubMed Central  PubMed  Google Scholar 

  67. Hsu FI, Boyce JA. Biology of mast cells and their mediators. In: Adkinson Jr NF, Busse WW, Bochner BS, Holgate ST, Simons FER, Lemanske Jr RF, editors. Middleton’s allergy: principles & practice. 7th ed. Philadelphia: Mosby/Elsevier; 2009. p. 311–28.

    Chapter  Google Scholar 

  68. He A, Shi GP. Mast cell chymase and tryptase as targets for cardiovascular and metabolic diseases. Curr Pharm Des. 2013;19(6):1114–25.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  69. Oskeritzian CA. Mast cells and wound healing. Adv Wound Care (New Rochelle). 2012;1(1):23–8. doi:10.1089/wound.2011.0357.

    Article  Google Scholar 

  70. Takato H, Yasui M, Ichikawa Y, Waseda Y, Inuzuka K, Nishizawa Y, et al. The specific chymase inhibitor TY-51469 suppresses the accumulation of neutrophils in the lung and reduces silica-induced pulmonary fibrosis in mice. Exp Lung Res. 2011;37(2):101–8. doi:10.3109/01902148.2010.520815.

    Article  CAS  PubMed  Google Scholar 

  71. Walter M, Sutton RM, Schechter NM. Highly efficient inhibition of human chymase by alpha(2)-macroglobulin. Arch Biochem Biophys. 1999;368(2):276–84. doi:10.1006/abbi.1999.1309.

    Article  CAS  PubMed  Google Scholar 

  72. Raymond WW, Su S, Makarova A, Wilson TM, Carter MC, Metcalfe DD, et al. Alpha 2-macroglobulin capture allows detection of mast cell chymase in serum and creates a reservoir of angiotensin II-generating activity. J Immunol. 2009;182(9):5770–7. doi:10.4049/jimmunol.0900127.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  73. Goldstein SM, Kaempfer CE, Kealey JT, Wintroub BU. Human mast cell carboxypeptidase. Purification and characterization. J Clin Invest. 1989;83(5):1630–6. doi:10.1172/JCI114061.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  74. Fajt ML, Wenzel SE. Mast cells, their subtypes, and relation to asthma phenotypes. Ann Am Thorac Soc. 2013;10(Suppl):S158–64. doi:10.1513/AnnalsATS.201303-064AW.

    Article  CAS  PubMed  Google Scholar 

  75. Takabayashi T, Kato A, Peters AT, Suh LA, Carter R, Norton J, et al. Glandular mast cells with distinct phenotype are highly elevated in chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol. 2012;130(2):410–20. doi:10.1016/j.jaci.2012.02.046. e5.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  76. D’Ambrosio C, Akin C, Wu Y, Magnusson MK, Metcalfe DD. Gene expression analysis in mastocytosis reveals a highly consistent profile with candidate molecular markers. J Allergy Clin Immunol. 2003;112(6):1162–70. doi:10.1016/j.jaci.2003.07.008.

    Article  PubMed  CAS  Google Scholar 

  77. Pan Q, Ding MF, Zhang S, Ning Y, Liu HW, Wei H, et al. Measurement of plasma mast cell carboxypeptidase and chymase levels in children with allergic diseases. Zhongguo Dang Dai Er Ke Za Zhi. 2011;13(10):814–6.

    CAS  PubMed  Google Scholar 

  78. Simons FE, Frew AJ, Ansotegui IJ, Bochner BS, Golden DB, Finkelman FD, et al. Risk assessment in anaphylaxis: current and future approaches. J Allergy Clin Immunol. 2007;120(1 Suppl):S2–24. doi:10.1016/j.jaci.2007.05.001.

    Article  CAS  PubMed  Google Scholar 

  79. Fanning LB, Boyce JA. Lipid mediators and allergic diseases. Ann Allergy Asthma Immunol. 2013;111(3):155–62. doi:10.1016/j.anai.2013.06.031.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  80. Boyce JA. Eicosanoid mediators of mast cells: receptors, regulation of synthesis, and pathobiologic implications. Chem Immunol Allergy. 2005;87:59–79. doi:10.1159/000087571.

    Article  CAS  PubMed  Google Scholar 

  81. Morrow JD, Guzzo C, Lazarus G, Oates JA, Roberts 2nd LJ. Improved diagnosis of mastocytosis by measurement of the major urinary metabolite of prostaglandin D2. J Invest Dermatol. 1995;104(6):937–40.

    Article  CAS  PubMed  Google Scholar 

  82. Ravi A, Butterfield J, Weiler CR. Mast cell activation syndrome: improved identification by combined determinations of serum tryptase and 24-hour urine 11beta-prostaglandin2alpha. J Allergy Clin Immunol Pract. 2014;2(6):775–8. doi:10.1016/j.jaip.2014.06.011.

    Article  PubMed  Google Scholar 

  83. Murphy RC, Hammarstrom S, Samuelsson B. Leukotriene C: a slow-reacting substance from murine mastocytoma cells. Proc Natl Acad Sci U S A. 1979;76(9):4275–9.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  84. Kanaoka Y, Boyce JA. Cysteinyl leukotrienes and their receptors; emerging concepts. Allergy Asthma Immunol Res. 2014;6(4):288–95. doi:10.4168/aair.2014.6.4.288.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  85. Butterfield JH. Increased leukotriene E4 excretion in systemic mastocytosis. Prostaglandins Other Lipid Mediat. 2010;92(1–4):73–6. doi:10.1016/j.prostaglandins.2010.03.003.

    Article  CAS  PubMed  Google Scholar 

  86. Valent P, Cerny-Reiterer S, Herrmann H, Mirkina I, George TI, Sotlar K, et al. Phenotypic heterogeneity, novel diagnostic markers, and target expression profiles in normal and neoplastic human mast cells. Best Pract Res Clin Haematol. 2010;23(3):369–78. doi:10.1016/j.beha.2010.07.003.

    Article  CAS  PubMed  Google Scholar 

  87. Pols MS, Klumperman J. Trafficking and function of the tetraspanin CD63. Exp Cell Res. 2009;315(9):1584–92. doi:10.1016/j.yexcr.2008.09.020.

    Article  CAS  PubMed  Google Scholar 

  88. He SH, Zhang HY, Zeng XN, Chen D, Yang PC. Mast cells and basophils are essential for allergies: mechanisms of allergic inflammation and a proposed procedure for diagnosis. Acta Pharmacol Sin. 2013;34(10):1270–83. doi:10.1038/aps.2013.88.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  89. Sturm EM, Kranzelbinder B, Heinemann A, Groselj-Strele A, Aberer W, Sturm GJ. CD203c-based basophil activation test in allergy diagnosis: characteristics and differences to CD63 upregulation. Cytometry B Clin Cytom. 2010;78(5):308–18. doi:10.1002/cyto.b.20526.

    Article  PubMed  CAS  Google Scholar 

  90. Kraft S, Fleming T, Billingsley JM, Lin SY, Jouvin MH, Storz P, et al. Anti-CD63 antibodies suppress IgE-dependent allergic reactions in vitro and in vivo. J Exp Med. 2005;201(3):385–96. doi:10.1084/jem.20042085.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  91. Kraft S, Jouvin MH, Kulkarni N, Kissing S, Morgan ES, Dvorak AM, et al. The tetraspanin CD63 is required for efficient IgE-mediated mast cell degranulation and anaphylaxis. J Immunol. 2013;191(6):2871–8. doi:10.4049/jimmunol.1202323.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  92. Buhring HJ, Streble A, Valent P. The basophil-specific ectoenzyme E-NPP3 (CD203c) as a marker for cell activation and allergy diagnosis. Int Arch Allergy Immunol. 2004;133(4):317–29. doi:10.1159/000077351.

    Article  PubMed  CAS  Google Scholar 

  93. Yano Y, Hayashi Y, Sano K, Shinmaru H, Kuroda Y, Yokozaki H, et al. Expression and localization of ecto-nucleotide pyrophosphatase/phosphodiesterase I-3 (E-NPP3/CD203c/PD-I beta/B10/gp130RB13-6) in human colon carcinoma. Int J Mol Med. 2003;12(5):763–6.

    CAS  PubMed  Google Scholar 

  94. Hauswirth AW, Escribano L, Prados A, Nunez R, Mirkina I, Kneidinger M, et al. CD203c is overexpressed on neoplastic mast cells in systemic mastocytosis and is upregulated upon IgE receptor cross-linking. Int J Immunopathol Pharmacol. 2008;21(4):797–806.

    CAS  PubMed  Google Scholar 

  95. Ono E, Taniguchi M, Higashi N, Mita H, Kajiwara K, Yamaguchi H, et al. CD203c expression on human basophils is associated with asthma exacerbation. J Allergy Clin Immunol. 2010;125(2):483–9. doi:10.1016/j.jaci.2009.10.074. e3.

    Article  CAS  PubMed  Google Scholar 

  96. Perini GF, Pro B. Brentuximab Vedotin in CD30+ Lymphomas. Biol Ther. 2013;3:15–23. doi:10.1007/s13554-013-0008-7.

    Article  PubMed Central  PubMed  Google Scholar 

  97. Morgado JM, Perbellini O, Johnson RC, Teodosio C, Matito A, Alvarez-Twose I, et al. CD30 expression by bone marrow mast cells from different diagnostic variants of systemic mastocytosis. Histopathology. 2013;63(6):780–7. doi:10.1111/his.12221.

    Article  PubMed  Google Scholar 

  98. Malek TR. The biology of interleukin-2. Annu Rev Immunol. 2008;26:453–79. doi:10.1146/annurev.immunol.26.021607.090357.

    Article  CAS  PubMed  Google Scholar 

  99. Schernthaner GH, Hauswirth AW, Baghestanian M, Agis H, Ghannadan M, Worda C, et al. Detection of differentiation- and activation-linked cell surface antigens on cultured mast cell progenitors. Allergy. 2005;60(10):1248–55. doi:10.1111/j.1398-9995.2005.00865.x.

    Article  CAS  PubMed  Google Scholar 

  100. Pawankar R, Okuda M, Yssel H, Okumura K, Ra C. Nasal mast cells in perennial allergic rhinitics exhibit increased expression of the Fc epsilonRI, CD40L, IL-4, and IL-13, and can induce IgE synthesis in B cells. J Clin Invest. 1997;99(7):1492–9. doi:10.1172/JCI119311.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  101. Kobayashi H, Okayama Y, Ishizuka T, Pawankar R, Ra C, Mori M. Production of IL-13 by human lung mast cells in response to Fcepsilon receptor cross-linkage. Clin Exp Allergy. 1998;28(10):1219–27.

    Article  CAS  PubMed  Google Scholar 

  102. Yamaguchi M, Lantz CS, Oettgen HC, Katona IM, Fleming T, Miyajima I, et al. IgE enhances mouse mast cell Fc(epsilon)RI expression in vitro and in vivo: evidence for a novel amplification mechanism in IgE-dependent reactions. J Exp Med. 1997;185(4):663–72.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  103. Pawankar R, Ra C. IgE-Fc epsilonRI-mast cell axis in the allergic cycle. Clin Exp Allergy. 1998;28 Suppl 3:6–14.

    CAS  PubMed  Google Scholar 

  104. Rajakulasingam K, Till S, Ying S, Humbert M, Barkans J, Sullivan M, et al. Increased expression of high affinity IgE (FcepsilonRI) receptor-alpha chain mRNA and protein-bearing eosinophils in human allergen-induced atopic asthma. Am J Respir Crit Care Med. 1998;158(1):233–40. doi:10.1164/ajrccm.158.1.9708106.

    Article  CAS  PubMed  Google Scholar 

  105. Humbert M, Grant JA, Taborda-Barata L, Durham SR, Pfister R, Menz G, et al. High-affinity IgE receptor (FcepsilonRI)-bearing cells in bronchial biopsies from atopic and nonatopic asthma. Am J Respir Crit Care Med. 1996;153(6 Pt 1):1931–7. doi:10.1164/ajrccm.153.6.8665058.

    Article  CAS  PubMed  Google Scholar 

  106. Andersson CK, Tufvesson E, Aronsson D, Bergqvist A, Mori M, Bjermer L, et al. Alveolar mast cells shift to an FcepsilonRI-expressing phenotype in mild atopic asthma: a novel feature in allergic asthma pathology. Allergy. 2011;66(12):1590–7. doi:10.1111/j.1398-9995.2011.02723.x.

    Article  CAS  PubMed  Google Scholar 

  107. Andersson CK, Mori M, Bjermer L, Lofdahl CG, Erjefalt JS. Novel site-specific mast cell subpopulations in the human lung. Thorax. 2009;64(4):297–305. doi:10.1136/thx.2008.101683.

    Article  CAS  PubMed  Google Scholar 

  108. Bergqvist A, Andersson CK, Mori M, Walls AF, Bjermer L, Erjefalt JS. Alveolar T-helper type-2 immunity in atopic asthma is associated with poor clinical control. Clin Sci (Lond). 2015;128(1):47–56. doi:10.1042/CS20140309.

    Article  CAS  Google Scholar 

  109. Kiyokawa H, Matsumoto H, Nakaji H, Niimi A, Ito I, Ono K, et al. Centrilobular opacities in the asthmatic lung successfully treated with inhaled ciclesonide and tiotropium: with assessment of alveolar nitric oxide levels. Allergol Int. 2011;60(3):381–5. doi:10.2332/allergolint.10-CR-0251.

    Article  CAS  PubMed  Google Scholar 

  110. Arock M, Schneider E, Boissan M, Tricottet V, Dy M. Differentiation of human basophils: an overview of recent advances and pending questions. J Leukoc Biol. 2002;71(4):557–64.

    CAS  PubMed  Google Scholar 

  111. Naclerio RM, Baroody FM, Kagey-Sobotka A, Lichtenstein LM. Basophils and eosinophils in allergic rhinitis. J Allergy Clin Immunol. 1994;94(6 Pt 2):1303–9.

    Article  CAS  PubMed  Google Scholar 

  112. Uyttebroek AP, Sabato V, Faber MA, Cop N, Bridts CH, Lapeere H, et al. Basophil activation tests: time for a reconsideration. Expert Rev Clin Immunol. 2014;10(10):1325–35. doi:10.1586/1744666X.2014.959498.

    Article  CAS  PubMed  Google Scholar 

  113. Ebo DG, Bridts CH, Mertens CH, Hagendorens MM, Stevens WJ, De Clerck LS. Analyzing histamine release by flow cytometry (HistaFlow): a novel instrument to study the degranulation patterns of basophils. J Immunol Methods. 2012;375(1–2):30–8. doi:10.1016/j.jim.2011.09.003.

    Article  CAS  PubMed  Google Scholar 

  114. Shamji MH, Layhadi JA, Scadding GW, Cheung DK, Calderon MA, Turka LA, et al. Basophil expression of diamine oxidase: a novel biomarker of allergen immunotherapy response. J Allergy Clin Immunol. 2015;135(4):913–21. doi:10.1016/j.jaci.2014.09.049. e9.

    Article  CAS  PubMed  Google Scholar 

  115. Schleimer RP, Davidson DA, Peters SP, Lichtenstein LM. Inhibition of human basophil leukotriene release by antiinflammatory steroids. Int Arch Allergy Appl Immunol. 1985;77(1–2):241–3.

    Article  CAS  PubMed  Google Scholar 

  116. Schroeder JT. Basophils: emerging roles in the pathogenesis of allergic disease. Immunol Rev. 2011;242(1):144–60. doi:10.1111/j.1600-065X.2011.01023.x.

    Article  CAS  PubMed  Google Scholar 

  117. Austen KF, Maekawa A, Kanaoka Y, Boyce JA. The leukotriene E4 puzzle: finding the missing pieces and revealing the pathobiologic implications. J Allergy Clin Immunol. 2009;124(3):406–14. doi:10.1016/j.jaci.2009.05.046. quiz 15–6.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  118. Schroeder JT, Kagey-Sobotka A. Assay methods for measurement of mediators and markers of allergic inflammation. In: Rose NR, Hamilton RG, Detrick B, editors. Manual of clinical laboratory immunology. 6th ed. Washington: ASM Press; 2002. p. 899–909.

    Google Scholar 

  119. Karasuyama H, Yamanishi Y. Basophils have emerged as a key player in immunity. Curr Opin Immunol. 2014;31:1–7. doi:10.1016/j.coi.2014.07.004.

    Article  CAS  PubMed  Google Scholar 

  120. Plager DA, Weiss EA, Kephart GM, Mocharla RM, Matsumoto R, Checkel JL, et al. Identification of basophils by a mAb directed against pro-major basic protein 1. J Allergy Clin Immunol. 2006;117(3):626–34. doi:10.1016/j.jaci.2005.10.023.

    Article  CAS  PubMed  Google Scholar 

  121. Kepley CL, McFeeley PJ, Oliver JM, Lipscomb MF. Immunohistochemical detection of human basophils in postmortem cases of fatal asthma. Am J Respir Crit Care Med. 2001;164(6):1053–8. doi:10.1164/ajrccm.164.6.2102025.

    Article  CAS  PubMed  Google Scholar 

  122. Foster B, Schwartz LB, Devouassoux G, Metcalfe DD, Prussin C. Characterization of mast-cell tryptase-expressing peripheral blood cells as basophils. J Allergy Clin Immunol. 2002;109(2):287–93.

    Article  CAS  PubMed  Google Scholar 

  123. Cromheecke JL, Nguyen KT, Huston DP. Emerging role of human basophil biology in health and disease. Curr Allergy Asthma Rep. 2014;14(1):408. doi:10.1007/s11882-013-0408-2.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  124. Sturm GJ, Kranzelbinder B, Sturm EM, Heinemann A, Groselj-Strele A, Aberer W. The basophil activation test in the diagnosis of allergy: technical issues and critical factors. Allergy. 2009;64(9):1319–26. doi:10.1111/j.1398-9995.2009.02004.x.

    Article  CAS  PubMed  Google Scholar 

  125. McGowan EC, Saini S. Update on the performance and application of basophil activation tests. Curr Allergy Asthma Rep. 2013;13(1):101–9. doi:10.1007/s11882-012-0324-x.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  126. Boumiza R, Debard AL, Monneret G. The basophil activation test by flow cytometry: recent developments in clinical studies, standardization and emerging perspectives. Clin Mol Allergy. 2005;3:9. doi:10.1186/1476-7961-3-9.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  127. Hauswirth AW, Sonneck K, Florian S, Krauth MT, Bohm A, Sperr WR, et al. Interleukin-3 promotes the expression of E-NPP3/CD203C on human blood basophils in healthy subjects and in patients with birch pollen allergy. Int J Immunopathol Pharmacol. 2007;20(2):267–78.

    CAS  PubMed  Google Scholar 

  128. Hennersdorf F, Florian S, Jakob A, Baumgartner K, Sonneck K, Nordheim A, et al. Identification of CD13, CD107a, and CD164 as novel basophil-activation markers and dissection of two response patterns in time kinetics of IgE-dependent upregulation. Cell Res. 2005;15(5):325–35. doi:10.1038/sj.cr.7290301.

    Article  CAS  PubMed  Google Scholar 

  129. MacGlashan Jr D. Expression of CD203c and CD63 in human basophils: relationship to differential regulation of piecemeal and anaphylactic degranulation processes. Clin Exp Allergy. 2010;40(9):1365–77. doi:10.1111/j.1365-2222.2010.03572.x.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  130. McEuen AR, Calafat J, Compton SJ, Easom NJ, Buckley MG, Knol EF, et al. Mass, charge, and subcellular localization of a unique secretory product identified by the basophil-specific antibody BB1. J Allergy Clin Immunol. 2001;107(5):842–8. doi:10.1067/mai.2001.114650.

    Article  CAS  PubMed  Google Scholar 

  131. Mochizuki A, McEuen AR, Buckley MG, Walls AF. The release of basogranulin in response to IgE-dependent and IgE-independent stimuli: validity of basogranulin measurement as an indicator of basophil activation. J Allergy Clin Immunol. 2003;112(1):102–8.

    Article  CAS  PubMed  Google Scholar 

  132. Agis H, Krauth MT, Bohm A, Mosberger I, Mullauer L, Simonitsch-Klupp I, et al. Identification of basogranulin (BB1) as a novel immunohistochemical marker of basophils in normal bone marrow and patients with myeloproliferative disorders. Am J Clin Pathol. 2006;125(2):273–81. doi:10.1309/M9FQ-MQGF-6616-7N2X.

    Article  CAS  PubMed  Google Scholar 

  133. Nguyen T, Gernez Y, Fuentebella J, Patel A, Tirouvanziam R, Reshamwala N, et al. Immunophenotyping of peripheral eosinophils demonstrates activation in eosinophilic esophagitis. J Pediatr Gastroenterol Nutr. 2011;53(1):40–7. doi:10.1097/MPG.0b013e318212647a.

    Article  PubMed Central  PubMed  Google Scholar 

  134. Heinisch IV, Bizer C, Volgger W, Simon HU. Functional CD137 receptors are expressed by eosinophils from patients with IgE-mediated allergic responses but not by eosinophils from patients with non-IgE-mediated eosinophilic disorders. J Allergy Clin Immunol. 2001;108(1):21–8. doi:10.1067/mai.2001.116864.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

The authors would like to acknowledge the members of the World Allergy Organization’s Committee on Eosinophils, Mast Cells, and Basophils in Allergic Disease, 2012–2013 and 2013–2014. DDM are ADK are supported by the Division of Intramural Research of NIAID/NIH. SJA is supported in part by research grants from the FDA (R01FD004086), NIH (R21HL118588), American Partnership for Eosinophilic Disorders (APFED), and University of Illinois, Chicago. MWJ is mainly supported by research grants from the NIH (P01 HL088594 and 1U10 HL109168 [PI: N. N. Jarjour]).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Dean D. Metcalfe.

Additional information

Competing interests

MWJ, FHF, AK, GN, DDM, have no relevant potential conflict of interest. SJA is a co-founder and CSO for EnteroTrack, LLC, the company developing the Esophageal String Test (EST) for EoE.

Authors’ contributions

DDM and MT developed the concept for this paper. DDM led the development and coordination of the project, wrote the abstract; RP, FLS and LBS were sub-editors on the mast cell, eosinophil and basophil sections, respectively; DDM, RP, CA, AI, GN, YO, LBS, and MT contributed to the mast cell section; SJA, GLG, MWJ, ADK, FLS, and HUS contributed to the eosinophil section; FC, FHF, GJG, KML, CP, JTS, and AFW contributed to the basophil section. All authors read and approved the final manuscript.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Metcalfe, D.D., Pawankar, R., Ackerman, S.J. et al. Biomarkers of the involvement of mast cells, basophils and eosinophils in asthma and allergic diseases. World Allergy Organ J 9, 7 (2016). https://doi.org/10.1186/s40413-016-0094-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40413-016-0094-3

Keywords