Summary
While the origin of the phrase “birds of a feather flock together” is unclear, it has been in use for centuries and is typically employed to describe the phenomenon that people with similar tastes or interests tend to seek each other out and congregate together. In this review, we have co-opted this phrase to compare innate immune cells of related origin, the eosinophil and mast cell, because they very often accumulate together in tissue sites under both homeostatic and inflammatory conditions. To highlight overlapping yet distinct features, their hematopoietic development, cell surface phenotype, mediator release profiles and roles in diseases have been compared and contrasted. What emerges is a sense that these two cell types often interact with each other and their tissue environment to provide synergistic contributions to a variety of normal and pathologic immune responses.
Keywords: Eosinophils, mast cells, receptors, mediators, cytokines, lineage, inflammation, homeostasis
1 Introduction
Mast cells (MCs) and eosinophils are immune cells well known for their roles in initiating allergic responses. These cells are developmentally and functionally similar and participate in many of the same diseases, often in a synergistic manner. They have been described as key players in atopic dermatitis, food allergies, asthma and rhinitis, all of which have increased in prevalence in recent decades. Since the late 1980’s, MCs and eosinophils have been the subject of many human and animal research studies that consistently demonstrate their important contributions to disease pathogenesis. It is therefore not surprising that the majority of approved biologicals for the treatment of these diseases target MCs and eosinophils, for example through IgE and IL-5.
This review will focus on MC and eosinophil similarities during differentiation and maturation and their response to external stimuli. While they do have a comparable repertoire of surface receptors, their responses and reactions often vary. Replete with granules, both MCs and eosinophils harbor a unique collection of pre-stored mediators to be released upon their activation. They also have overlapping but distinct patterns of newly formed lipid and protein mediators. Thus, key similarities and differences will be highlighted while summarizing what is known about their paired roles in allergies and other relevant disorders.
2 Origin and development
2.1 Hematopoietic lineage
MCs and eosinophils both arise from CD34+ hematopoietic stem cells in the bone marrow. Hematopoietic stem cells can exist as long-term self-renewing cells or as cells capable of differentiating into multiple cell lineages, i.e. multipotent progenitors. Classic models of hematopoiesis highlight a bifurcation downstream of multipotent progenitors where commitment towards a myeloid or lymphoid lineage occurs. In humans, eosinophil-committed progenitors branch directly from common myeloid progenitors 1. However, the origin of human MC-committed progenitors is still controversial. Studies in mice suggest they come from granulocyte/monocyte progenitors downstream of common myeloid progenitors 2. To make matters more complicated distinct developmental pathways and tissue microenvironments govern MC phenotypes, i.e. tryptase-positive (MCT) and tryptase/chymase-positive (MCTC) 3.
2.2 Differentiation and maturation
Differentiation of progenitors, towards a terminal MC or eosinophil fate, is a complex process regulated by transcription factors and extrinsic signals such as cytokines. The most pertinent transcription factors in eosinophil differentiation are GATA-1 and C/EBPα, because their forced expression in human myeloid progenitors results in eosinophil formation 4,5. In addition, PU.1 and FOG-1 indirectly contribute to the emergence of eosinophils because augmentation of either one shifts progenitors towards an alternate fate 6–9. Recent studies in mice have identified additional transcription factors important for eosinophil differentiation; Helios and Aelios, which regulate gene expression during development 10 and Xbp1, critical for eosinophil differentiation and granulogenesis 11. Another protein necessary for eosinophil granularity and survival in mice is cystatin F 12. In MCs, GATA-1 and GATA-2 drive differentiation and regulate transcription of FcεRI and kit genes 3. PU.1 also promotes MC development, but expression becomes weaker as MCs mature 9. Similar to eosinophils, FOG-1 inhibits MC differentiation redirecting progenitors towards other cell lineages 9,13.
MCs and eosinophils both originate in the bone marrow, however eosinophils enter the circulation as mature cells while MCs circulate as progenitors and become mature once they reach the tissues where they ultimately reside (as reviewed in 13,14). Growth factors are important for regulating a variety of cellular processes including differentiation and proliferation. Eosinophil maturation and proliferation is dependent on cytokines IL-3, IL-5 and GM-CSF, for which receptors are expressed on eosinophil precursors and feature a common β chain. While different combinations of these cytokines generally can promote eosinophilic differentiation and proliferation, the three together greatly enhance both 15. It is well established that IL-5 is selective for eosinophil production and also mediates their release from the bone marrow 16. The importance of IL-5 to eosinophil development has been further demonstrated in recent trials of now approved anti-IL-5 monoclonal antibodies that attenuated eosinophil numbers 17. Of note, it was recently reported in mice that IL-33 can expand eosinophil progenitors upstream of IL-5 and promote upregulation of IL-5Rα 18. IL-5R is a critical component of eosinophil lineage commitment and is responsible for the occurrence of increased eosinophil numbers observed in allergic inflammation (e.g. increased blood and sputum eosinophils in asthmatic patients following allergen challenge) 19. Interestingly, IL-4Rα supports IL-5 mediated maturation of eosinophils, but in the absence of IL-5 CCL3 (from eosinophil precursors) can promote cell differentiation through CCR1 20. In homeostatic conditions, eosinophils are most commonly found in the circulation. However, eosinophils also home to tissues where eotaxin-1 (CCL11) is constitutively expressed, such as the GI tract, thymus, mammary glands and uterus 21.
Bone marrow and blood MC precursors are CD13+, CD34+and CD117+ (c-Kit, receptor for SCF discussed below) and express low levels of FcεRIα 13,22,23. Other cytokines, including IL-4, IL-9, IL-10, TGFβ and nerve growth factor, can also contribute to the maturation and proliferation of human MCs. Interestingly, a key growth factor for bone marrow-derived mouse MCs, IL-3 24, promotes survival of human cord blood-derived MCs 25 but not human lung MCs 26, suggesting that differences in developmental pathways are species-specific and governed by MC phenotype. During an inflammatory or allergic event, tissue-resident MC numbers increase due to recruitment of precursors 27, which is chemokine-driven22. MC precursors also respond to other chemotactic factors, including histamine leukotriene B4 (LTB4), 28 TGF-β1 and fractalkine, the latter produced by endothelial cells and by smooth muscle cells in human airways during asthma 27,29.
3 Cell Surface Receptors
3.1 Immunoglobulin and immunoglobulin-like receptors
Immunoglobulin or immunoglobulin-like receptors are a family of receptors that feature extracellular immunoglobulin domains comprised of 2 antiparallel β sheets. The most common types are Fc receptors that exist on a variety of cells, including MCs and eosinophils, and serve a protective role by recognizing pathogen-associated antibodies and virally-infected cells. Different classes of Fc receptors exist, named for the antibody to which they bind. Fc gamma (Fcγ) receptors recognizing IgG make up the largest family of Fc receptors. Human MCs express both FcγRI (CD64) and FcγRII (CD32). FcγRI expression can be upregulated by IFNγ 30 and engagement triggers the release of inflammatory mediators such as histamine, prostaglandins, leukotrienes and type 1 cytokines 31. FcγRII is constitutively expressed on human skin MCs 32 and its aggregation is known to inhibit histamine release 33. Human eosinophils lack FcγRI, but do express FcγRII 34. However, unlike MCs, internalization of FcγRII on eosinophils results in activation 35,36 and promotes survival 37. Of note, there are several different isoforms of FcγRII, including FcγRIIa and FcγRIIb 38. Human MCs and eosinophils express FcγRIIa, which features an activating immunoreceptor tyrosine-based activation motifs (ITAM) domain and has been shown to play a role in allergic responses 32,39. ITIM-bearing FcγRIIb is expressed by some but not all MCs and when present and co-engaged can inhibit IgE-dependent activation and proliferation in mice 40,41. The other IgG receptor, FcγRIII (CD16), is normally internally expressed in eosinophils, but can translocate to the surface during allergic inflammation 42 or in the presence of mediators such as IFNγ, C5a or platelet activating factor 43,44. In humans there are two Fc receptors to which IgE can bind. The high-affinity IgE receptor, FcεRI, features an immunoglobulin-binding α chain, while the low-affinity receptor, FcεRII (CD23), does not and is instead characterized as a C-type lectin. Canonical activation of MCs occurs upon antigen cross-linking of FcεRI-bound IgE. MC-expression of FcεRI is upregulated in the presence of IgE, IL-4 and IL-13 45–47 and downregulated by granulocyte macrophage colony-stimulating factor (GM-CSF) and TGFβ1 48,49. In addition, a monoclonal antibody that targets the high-affinity binding site on IgE, called omalizumab, reduces FcεRI expression 50. Human MCs express FcεRI (which consists of α, β, and γ subunits) either as tetrameric αβγ2 or trimeric αγ2 51. Expression of FcεRI is γ chain-dependent, while the β chain enhances MC signaling through ITAM located within the cytoplasmic tails of both the γ and β chains 51,52. Interestingly, when ITAM-containing FcεRI is cross-linked with the ITIM-bearing FcγRII receptor on MCs, IgE-mediated activation is inhibited 53. Expression of FcεRI on the surface of human eosinophils is still controversial, but the majority of studies suggest that surface expression, if present at all, is low and unaffected by IgE levels 54. There may also be an intracellular pool of α chain albeit with unknown function 55. Like for MCs, IL-4 can stimulate FcεRI expression on human eosinophils, however receptor engagement does not induce degranulation 56. Eosinophils also express the low-affinity IgE receptor FcεRII (CD23) intracellularly at homeostasis. Upon activation, FcεRII is translocated to the cell surface, followed quickly by reinternalization 57. There is one IgA-binding receptor, FcαR, which features 2 immunoglobulin domains that are homologous to FcγR and FcεRI 58. IL-4, IL-5 and corticosteroids have been shown to enhance IgA/ FcαR binding on eosinophils, triggering release of peroxidase 59,60. A study by Bracke et al. found that human eosinophil priming with GM-CSF, IL-4 or IL-5 is necessary for IgA binding to occur 59.
Sialic acid-binding immunoglobulin-like lectins (Siglecs) also feature extracellular domains that resemble immunoglobulins. Siglecs are expressed by a variety of immune cells, including MCs and eosinophils. It has been reported that CD34+ MC progenitors express a variety of Siglecs during differentiation and maturation, including Siglec-2, -3, -5, -6, -8 and -10. We found that as MCs mature, expression of Siglec-5 and Siglec-10 decrease, while Siglec-6 and Siglec-8 appear de novo around the same time as FcεRIα, perhaps indicating a more functional role for these Siglecs in MCs 61. Another study reported that Siglec-7 is expressed by human MCs 62. Siglec-8 exists as two distinct isoforms with differing cytoplasmic regions, dependent on splicing 62, while its extracellular domains preferentially bind 6′-sulfo-sialyl Lewis X 63. In humans, Siglec-8 engagement has profound functional consequences on both MCs and eosinophils 64. For example, IL-5 priming in human eosinophils leaves them much more susceptible to Siglec-8 mediated apoptosis 65,66, whereas in MCs Siglec-8 ligation inhibits FcεRIα-dependent MC activation and calcium flux in an ITIM-dependent manner 67. Other studies have highlighted the contribution of IL-33 priming to Siglec-8 mediated eosinophil apoptosis and demonstrated how Siglec-8 ligation promoted eosinophil adhesion via β2 integrins that was necessary for apoptosis 66,68. In addition to Siglec-8, Siglec-10 expression has also been detected on human eosinophils 69, however further studies are needed to determine its functional relevance.
CD300 receptors are transmembrane proteins which feature IgV-like extracellular domains. These receptors are expressed on many different immune cells, including MCs and eosinophils, and can be activating or inhibitory. CD300a features several ITIM domains and, once activated by endogenous ligands (i.e. phosphatidylserine and phosphatidylethanolamine), can inhibit IgE- and SCF-mediated functions in cord blood-derived MCs 70. A bispecific antibody targeting both CD300a and c-Kit inhibits activation in cord blood-derived MCs and in human MC leukaemia cell line (HMC-1) 70. The inhibitory effects of CD300a was also highlighted in CD300a KO mice, where IgE activation of MCs triggered an increased release of cytokines and chemokines compared to MCs in WT control mice 71. In human peripheral blood eosinophils, activation of CD300a inhibits chemotactic responses to eotaxin-1 and IL-5 and GM-CSF-associated survival and cytokine release 72. CD300c is also expressed on human MCs 73 and is characterized by the presence of a cytoplasmic ITAM-bearing FcRγ chain 74 and its ligation results in MC activation 73. Lastly, MCs and eosinophils both express inhibitory CD300f 75. Increased expression of CD300f has been detected on eosinophils from allergic rhinitis patients 76. The primary ligands for CD300f are sphingolipids, such as sphingomyelin and ceramide 77,78. Activated CD300f can inhibit FcεRI-driven MC activation 77. Of note, CD300f can also display an activating phenotype when cross-linked with mutated ITIM-expressing receptors 75.
Leukocyte immunoglobulin-like receptors (LILRs) are another group of cell surface proteins with both activating and inhibitory properties. Human eosinophils express activating LILRA2 and inhibitory LILRB1, B2 and B3 on their surface 79. MC precursors express LILRB1, B2, B3, B4 and A1 80, while mature MCs express LILRB5 on their granules. In MCs, it is suggested that LILRs play a role in down-regulating inflammatory responses 80. However, studies suggest they promote activation in eosinophils 79. To date, therapeutics targeting LILRs have yet to be developed, but theoretically targeting of these receptors might be useful in treating cancer and autoimmune diseases.
3.2 Cytokine receptors
One important form of cell communication is governed by cytokine receptors that trigger cellular responses to external stimuli. When cytokines bind to their receptor, transduced signals lead to changes in gene expression, release of inflammatory mediators and other reactions. Type 2 inflammatory responses prominently feature MCs and eosinophils and are defined by production of the cytokines IL-4, IL-5 and IL-13. The IL-4 receptor is constitutively expressed on eosinophils. A study by Wedi et al. reported that IL-4 may participate in inflammatory resolution by inhibiting eosinophil survival by promoting apoptosis 81. In MCs, IL-4 priming enhances IL-13 and histamine production following IgE-dependent activation 82,83. A study by Oskeritzian et al. found that MCs cultured with recombinant IL-4 experienced increased apoptosis 84. The receptor for IL-5 is a heterodimer complex which consists in a unique α subunit, required for signaling, and a common β subunit shared with IL-3 and GM-CSF receptors, that regulates binding affinity 85. Human eosinophils express IL-5R throughout development and maturation 86 and both IL-5 and its receptor have been successfully targeted in several clinical trials utilizing anti-IL-5 antibodies i.e. reslizumab 87, mepolizumab 88 and benralizumab, the latter unique since it specifically targets the IL-5 receptor α subunit with an antibody engineered to have enhanced antibody-dependent cellular cytotoxicity (ADCC) activity 89. There have been reports that human MCs also express IL-5R; however, its functional role is not yet understood 90. Of note, Otani et al. demonstrated that MCs numbers were reduced in pediatric EoE patients undergoing mepolizumab therapy 91, an effect not observed in mepolizumab-treated adult patients 92. The most common IL-13 receptor contains α chain 1 and has weak binding capacity that is strengthened upon formation of a heterodimer complex with IL-4Rα 93. Human eosinophils express low levels of IL-13Rα1, which can be upregulated by TGFβ or IFNγ 94. Human lung MCs and the immature MC line HMC 1.1 also express IL-13Rα1. MCs primed with IL-13 display increased FcεRI expression and proliferation 45,95.
Together with FcεRI, c-kit is a particularly important receptor on MCs playing a central role in their differentiation, regulation of cell growth, priming and chemotaxis. It is a type 3 tyrosine kinase receptor, which in humans exists in four different isoforms 96. It is expressed by a variety of cells including mature MCs during homeostasis and inflammation. SCF is the high-affinity ligand of c-kit. Interestingly, SCF can be released by eosinophils via a mechanism involving MC-derived chymase 97, highlighting the importance of MC-eosinophil crosstalk (i.e. the allergy effector unit, AEU 98,99, discussed in detail below). Eosinophils have also been shown to express c-kit, where it is suggested to have a role in promoting cellular adhesion 100. In disease, c-kit is involved in cancer and asthma. Activating mutations in c-kit, especially the D816V mutation, is the main cause of systemic mastocytosis, which involves widespread proliferation and accumulation of MCs in numerous organs 101,102. Of note, mutations of c-kit have also been found in patients with hypereosinophilic syndromes 103. Additionally, imatinib, a tyrosine kinase inhibitor mostly used to treat leukemias and some forms of hypereosinophilic syndromes, has recently been shown to modestly decrease MC numbers and airway hyperresponsiveness in patients with severe asthma 104.TSLP signaling is associated with many MC and eosinophil prominent diseases. Receptors for TSLP are heterodimeric complexes that consist in TSLPRα chain and IL-7 receptor α chain 105. TSLP is released from stimulated epithelium and mediates MC-driven type 2 inflammation in response to physical trauma or allergen exposure 106,107. Of note, activated MCs can also produce TSLP 108. In eosinophils, expression of TSLPR is elevated in the presence of IL-3 or TNFα 109. TSLP promotes eosinophil survival, adhesion and release of eosinophil-derived neurotoxin (EDN), IL-6 and several chemokines 110. Tezepelumab (AMG-157), an anti-TSLP monoclonal antibody, has been successful in improving lung function in mild asthmatic patients 111 and decreasing the number of asthma exacerbations, blood eosinophil levels and total serum IgE in subjects with moderate-to-severe asthma 112. Additionally, the finding that Tezepelumab attenuated both the early and late phase response to inhalational allergen challenge in humans implies potential effects of TSLP antagonism on both eosinophils and MCs 111.
Another important cytokine receptor expressed by both MCs and eosinophils is the IL-33 receptor, interleukin-1 receptor-like 1 (IL1R1, commonly called ST2). This receptor also exists as soluble ST2 (sST2) upon differential splicing of the ST2-encoding gene 113,114. ST2 is expressed by MCs, independent of maturation status, and IgE/allergen activation or IL-33 stimulation induces sST2 production 115,116. In mice, IL-33-dependent MC activation triggers release of many pro-inflammatory mediators including IL-1β, IL-6, IL-13, TNFα, CCL2 and PGD2 117,118. IL-33 can induce secretion of IL-5, IL-6, IL-13 and TNFα from human MCs, an effect that can be enhanced by TSLP 119. MCs do not only respond to IL-33, but can produce it upon IgE/allergen activation in a calcium-dependent manner 120,121. Eosinophils also express ST2 and respond to IL-33, but by producing superoxide anion 122. IL-33 is necessary for eosinophil hematopoiesis, homeostasis and promotes cell survival via the MyD88 pathway 18,123. IL-33 promotes eosinophil-mediated inflammation and our lab has recently demonstrated that Siglec-8 engagement on IL-33-primed eosinophils results in apoptosis 68,124. Lately it has been shown that eosinophil progenitors express a functional IL-33 receptor capable of producing pro-inflammatory cytokines at levels that far surpass those observed in mature eosinophils 125.
3.3 Cell migration
Integrins are a family of heterodimeric transmembrane cell adhesion receptors composed of α and β subunits. In humans, 18 α subunits and 8 β subunits can differentially combine to form at least 24 receptors 126,127. Ligation of integrin receptors modulates a variety of cellular responses including proliferation, degranulation, survival and cell motility. Integrin actions via the cytoskeleton drive vascular adhesion, endothelial transmigration and other aspects of cell trafficking and extravasation. Inflammatory pathways often feature α4 integrins. For example, the α4β1 integrin, or very late antigen-4 (VLA-4), is expressed by MCs and eosinophils 128,129 and binds VCAM-1 and fibronectin 130 to promote eosinophil migration and extravascular accumulation, and contributes to the perivascular localization of MCs 131–133. Another α4 integrin expressed by eosinophils and MCs is α4β7, which binds mucosal addressin cell adhesion molecule 1 (MAdCAM-1) and VCAM-1 and is important for homing of cells to mucosal tissues such as the intestine 134–137. Recent studies have shown that β2 integrins are necessary for release of granule proteins and cell death 66,138 and can mediate eosinophil-MC crosstalk through their activation on MCs by eosinophil-produced major basic protein-1 (MBP-1) 139.
Prostaglandin D2 (as described below) is a pro-inflammatory mediator with a role in allergic responses. Its high affinity G-protein coupled receptor, CRTH2, is expressed by eosinophils. Activated MCs are a major producer of PGD2, which promotes eosinophil recruitment and activation 140–142. Eosinophils can also produce PGD2 but at much lower levels per cell 143. Recent studies have reported that MC CRTH2 expression is internal 144 so its function is still unknown. It is worth noting that small molecule CRTH2 antagonists are showing some promising efficacy in early clinical trials 145–148.
Anaphylatoxins C3a and C5a are produced following activation of the complement system, and both eosinophils and MCs can respond to these proteins via expression of C3aR and C5aR. In eosinophils, C3a and C5a can trigger an increase in intracellular Ca2+ and release of reactive oxygen species (ROS) 149. Furthermore, they promote eosinophil rolling and adhesion and C5a influences eosinophil transmigration, integrin expression and superoxide anion production 150,151. Complement proteins C3a and C5a are also MC chemotaxins. Interestingly, C3a is more effective than C5a at recruiting MCs and can also stimulate production of CCL2 and CCL5, which have been shown to be important for early inflammatory responses 152–155. Recently another receptor for C5a was described in MCs, called C5aR2, which can stimulate release of certain cytokines, namely TNF, GM-CSF, CCL2 and CXCL10, and can mediate chemoattractant functions 156. While C5aR2 is expressed by mouse eosinophils, its expression in humans is unknown 157.
Sphingosine-1-phosphate (S1P) is important for cell trafficking, proliferation and survival responses. Crosslinking of high affinity FcεRI on MCs by IgE activates sphingosine kinases (SphK) that produce S1P, whose pleiotropic properties can be attributed to the fact that S1P binds to 5 different G-protein coupled receptors 158. MCs express S1PR1 and when activated it promotes MC movement. S1PR2 activation halts MC movement and initiates degranulation and release of pro-inflammatory mediators 159,160. The MC/S1P/S1PR2 axis was recently shown to be crucial for propagating early inflammatory responses in murine models of pulmonary and cutaneous allergic responses 154,155. Additionally, S1PR2 ligation leads to production of tissue remodeling matrix metalloproteinase-2 (MMP2) and vascular endothelial growth factor (VEGF)-α from human MCs 161. Eosinophils express S1PR1, 3, 4 and 5 and can therefore be recruited to inflamed tissues by MCs and other S1P-producing cells. Eosinophil chemotaxis towards S1P can be abrogated in the presence of the S1P receptor antagonist FTY-720, which has pharmacologically significant affinity for all S1PR except S1PR2 162.
Histamine is another MC-produced mediator that stimulates eosinophil recruitment through G-protein coupled histamine receptors 163. For eosinophils, histamine can act as a chemoattractant through H1R 164 and inhibit chemotaxis through H2R (when histamine concentration is high) 165. Histamine influences eosinophil adhesion, Ca2+ mobilization and cytoskeleton rearrangement via H4R 166,167. MC-expressed H4R regulates calcium mobilization and release of inflammatory mediators 168. Studies in mice found that antagonism of H4R resulted in inhibition of IgE-induced upregulation of FcεRI, suggesting H4R regulates FcεRI expression and function 163 andH4R antagonists have been shown to reduce histamine-mediated pruritus 169 in AD patients 170. In addition to H4R, human skin MCs express H2R, which negatively regulates histamine production, while H1R is expressed on HMC-1 cells 171.
Cannabinoid receptors exist as part of the endocannabinoid system and respond to endogenous ligands anandamide (AEA) and 2-arachidonoylglycerol (2-AG). There are 2 distinct cannabinoid receptors CB1 and CB2, and while AEA can bind to both, 2-AG is a specific agonist of CB1 expressed by cells of the nervous system. MCs express both CB1 and CB2 (Samson, 2003), while eosinophils and most immune cells express CB2 alone (Oka, 2004). Studies have shown that 2-AG acts as a chemoattractant for peripheral blood eosinophils 172, increasing their migration towards inflamed tissues 173.
3.4 Toll-like Receptors and others
Toll like receptors (TLRs) are important for pathogen recognition for innate and adaptive immunity 174,175. Both human MCs and eosinophils express TLRs 1–10, except TLR 8. Studies show contradicting evidence of TLR expression on both MCs and eosinophils as sometimes mRNA is expressed that does not translate to protein production (reviewed in 176,177). MCs and eosinophils respond to different TLR ligands, such as LPS and flaggelin which bind TLR-4 and TLR-5 respectively (reviewed in 176,177). Levels of TLR2 and TLR3 are elevated in the epithelium in fatal asthma specimens, suggesting that they may contribute to disease severity. Moreover, in asthma eosinophil density has been negatively correlated with TLR2 and TLR4 levels, while TLR3 seems to promote eosinophil numbers 178.
CD48 and 2B4 are receptors belonging to the CD2 family that are expressed on MCs and eosinophils. They are rather unique because they are high-affinity ligands for each other. CD48 is a GPI-anchored activating receptor, present in two forms: membrane associate (mCD48) and soluble (sCD48). 2B4 (CD244) is a SLAM family receptor and a CD2-like cell surface protein. CD244 is not expressed on human MCs, but is expressed on murine MCs where it has inhibitory activity. It was shown to have an activating effect on both murine and human eosinophils 179. Triggering of the CD48 on MCs and eosinophils induces activation (186, 188). S. aureus can activate MCs through CD48, causing bacterial uptake and TNF-α release 180, and on eosinophils, causing increasing of CD48 expression,the release of granular proteins, and of cytokines (188). CD48 expression is increased on both peripheral blood and bronchial eosinophils in asthma patients 181. CD48 on eosinophils from the skin of atopic dermatitis patients is upregulated as well 182. Activation of CD244 on eosinophils contributes to cell adhesion and migration 183 and induces degranulation 72. In vivo activation of human eosinophils by the S. aureus endotoxin B (SEB) leads to the formation of sCD48 and upregulates transcription of CD48 182,184. Interactions between CD48 on MCs and CD244 on eosinophils are an important part of the physical formation of the AEU and its pro-inflammatory properties. The AEU comprises physical and soluble interactions between MCs and eosinophils that promotes their survival and activation and can augment the intensity and duration of allergic inflammation (reviewed in 185). The importance of CD48 and CD244 in the AEU and in the interactions of MCs and eosinophils with S. aureus highlight the potential importance of these receptors in allergic inflammatory diseases in which there is superinfection with S. aureus 186.
4 Mediators released upon activation
4.1 Cytokines and chemokines
MCs and eosinophils can both store and synthesize de novo a wide array of cytokines, chemokines, eicosanoids, lipids and other inflammatory mediators. Cytokines and chemokines are most often released upon cell activation during the process of degranulation. The main mechanism for the release of these mediators from MCs is via antigen/allergen cross-linking of IgE receptors 144. Eosinophils on the other hand can release GM-CSF, IL-2, IL-10, IL-12, IL-13, RANTES (CCL5) and eotaxin (CCL11) among others 187, upon their activation 187 by Th2 cytokines such as IL-5 188. Notably, some mediators are produced by both MCs and eosinophils, such as SCF, IL-4, IL-5, IL-6 and TNFα, although quantities released per cell vary and can often be quite small 187,189,190. The evidence that MCs and eosinophils release some common cytokines and chemokines points out their shared capacity to influence similar pathophysiological processes and even each other via paracrine mechanisms. MCs store and release preformed TNFα 191. Eosinophils, on the other hand, do not store TNFα, but can synthesize it once activated 192. Moreover, since both MCs and eosinophils produce TNFα, they can potentially be important regulators of carcinogenesis since TNFα has cytotoxic and cytostatic effects against tumour cell lines 193. IL-4 is another key cytokine of Th2-driven immune responses that, together with SCF, can regulate MC proliferation and cytokine production 194. Eosinophil-derived IL-4 may therefore be important for MC regulation. Interestingly, recent studies have highlighted the importance of the eosinophil/IL-4 axis in limiting obesity in mice by promoting adipocyte beiging (i.e. a healthy and lean phenotype) through activation of M2 macrophages 195–197. IL-5, the main cytokine regulating eosinophils 198, can be produced by MCs, and it has been reported that PrMCs express IL-5R 90, suggesting that this mediator could be involved in the mutual regulation of these two cells.
4.2 Eicosanoids
Prostaglandins are arachidonic acid derivatives produced by the activity of phospholipase A2 and cyclooxygenase enzymes (COX 1 and 2) from membrane phospholipids. MCs and eosinophils have both COX 1 and 2 and release prostaglandins within seconds following their activation. MCs are the main producer of PGD2 involved in allergic reactions 199. It is produced from PGH2, synthesized by COX 1 or 2, which is converted into PGD2 via PGD2 synthase 200. Moreover, since activated MCs are the primary source of PGD2, this mediator and its metabolites are used as biomarkers for those pathologies characterized by activation of MCs, such as mastocytosis 201 and asthma, in which there is enhancement of the PGD2 pathway 202. Eosinophils can also produce PGD2, but in quantities that are much lower than for MCs 143, thus their contribution of PGD2 in disease seems less likely. PGD2 exerts its biological effect via DP1 and DP2/CRTH2, both G-protein coupled receptors (GPCRs) 144. On MCs, PGD2 triggers the synthesis of Th2 mediators through DP1 203, while the functions of CRTH2 receptor on MCs are controversial 144. On eosinophils, DP1 and CRTH2 have some overlapping and cooperative functions, since both receptors are able to induce the egress of eosinophils from bone marrow, and mediate chemotactic activity and production of leukotriene C4 (LTC4) 204. In asthma models, PGD2 activation of eosinophils increases their degranulation, mobilization and recruitment in the lungs through CRTH2. Currently, a CRTH2 antagonist, fevipiprant, is being tested in advanced clinical asthma trials 205,206.
Leukotrienes (LTs) are also rapidly synthesized and released following MC and eosinophil activation 207. The main leukotrienes produced by MCs and eosinophils are products of lipoxygenase 5 activity on arachidonic acid and they comprise the cysteinyl leukotriene (CysLT) LTC4 and LTB4. LTC4 is formed via the addition of a molecule of glutathione to LTA4. LTC4 is then transported outside of the cell where it can be converted to LTD4 and then to LTE4 by specific enzymes 208. Receptors for CysLTs are GPCRs called CysLT1R and CysLT2R. These receptors are expressed on eosinophils and MCs 209. Recently it was found that an ADP receptor, P2Y12, on LAD-2 MCs can also function as a receptor for LTE4, with powerful pro-inflammatory effects 210. On MCs, CysLTs trigger activation and induce proliferation and cytokine secretion 209. LTC4 and LTD4 have been shown to increase Ca2+ influx in MCs 211, enhance cytokine production 212 and, together with LTE4, increase MC proliferation 213. On eosinophils, CysLTs induce chemotaxis, migration and secretion of mediators; moreover, binding of CysLT1R can increase survival of eosinophils. While MCs and eosinophils are important sources of CysLTs, LTB4 is mainly produced by neutrophils and macrophages, with MCs and eosinophils producing only modest quantities 214. The effects of LTB4 are mediated by two GPCRs, BLT1 and BLT2, expressed on human HMC-1 and perhaps normal MCs, while human eosinophils express only BLT1 209. On eosinophils, binding of LTB4 to BLT1 induces their recruitment and chemotaxis; on MCs, both receptors induce chemotaxis and migration in a dose-dependent fashion 215. Given the relevance of leukotrienes and their receptors in inflammation, antagonists of CysLT1R have been developed for treatment of asthma 210 as well as drugs that target 5-lipoxygenase and thereby inhibit the synthesis of multiple leukotrienes 216.
4.3 Reactive oxygen species
Reactive oxygen species (ROS) include hydrogen peroxide (H2O2), nitric oxide (NO) and others 217,218. The primary role of ROS in inflammation is probably to eradicate invaders through their toxic effects 218,219. Both human MCs and eosinophils can produce ROS upon IgE-dependent activation and by activation of FcγR, Siglec-8 or other receptors, respectively 66,149,220,221. ROS plays a role in the regulation of airway inflammation and remodeling by MCs, through their involvement in the VEGF production cascade 222. Unlike MCs, ROS production in eosinophils is involved in the resolution of inflammation. In a preclinical model of asthma, mice with decreased ability to produce ROS showed no resolution compared to WT mice where resolution was observed regularly after 72h 218. Mice treated with H2O2 showed decreased allergic inflammation 218, and death induced by Siglec-8 activation in cytokine-primed eosinophils is completely dependent on internal ROS generation 66. The opposite effects of ROS production by MCs and eosinophils are interesting and raise questions about the possible effect of treatment with ROS scavengers.
4.4 Histamine
MCs, together with basophils, are the only eukaryotic cell sources of preformed histamine that they release promptly upon activation 223. Histamine is involved not only in allergy and anaphylaxis, but it is also important for immunomodulation, cell proliferation, wound healing, neurotransmission, stimulation of gastric secretion, regulation of blood pressure and perception of pain 224. There are four receptors for histamine, H1-4R, some of which are expressed on MCs and eosinophils (discussed above and in another review of this series).
5 Mast cells and eosinophils in allergic disease
5.1 Atopic dermatitis
Atopic dermatitis (AD) is a chronic eczematous disease characterized by itchy, dry and inflamed skin, which affects 10–20% of the world’s population including both adults and children 225. MC and eosinophil activation is a main characteristic of this disease. AD is often the first manifestation of allergies and is considered the first step in the atopic march because affected individuals are likely to develop other allergies such as asthma or rhinitis 226. Mutations in FLG, the gene encoding human filaggrin, are the most known and accepted risk factor for genetic predisposition of AD. Filaggrin is a protein involved in maintenance of the epidermal architecture and keratinocyte development. Thus, mutations in this gene can disrupt the structure of the skin barrier, resulting in increased sensitivity to allergens 226. Interestingly, skin barrier impairment correlates with infections from S. aureus 227, a main player in AD, by producing toxins such as enterotoxin A, B, C, D, etc., which can act like superantigens 224,228,229. Activated MCs are found in AD lesions and eosinophils can be activated by S. aureus directly via TLRs or CD48 180,182. MCs are in part responsible for the itching and skin dryness experienced by AD patients due to their release of histamine, tryptase and other pro-inflammatory mediators 230. Moreover, MCs produce IL-4 and IL-13, which promote eosinophil recruitment, via stimulation of chemokine production from keratinocytes and fibroblasts. Eosinophils recruited to AD lesional skin by MCs, once activated discharge several mediators that contribute to AD symptoms. For example, ECP, EDN and MBP have the ability to increase permeability of blood vessels and neurotoxic properties, while chemokines such as CCL11 can increase eosinophil recruitment 226.
5.2 Asthma
Atopic asthma is characterized by allergic, eosinophilic inflammation (reviewed in 231). One of the main players in the development of atopic asthma are MCs, which have a central role in eliciting the early asthmatic reaction and also contribute to the late phase, likely due to production of substances such as cytokines, chemokines and arachidonic acid metabolites. Even though the number of airway MCs of atopic and non-atopic patients is similar to what is observed in healthy controls, the density of FcεRI on MCs is increased, probably due to the presence of higher levels of serum IgE. The fact that omalizumab can effectively reduce exacerbations further supports the role of MCs in late-phase responses (reviewed in 232). Moreover, MC-derived substances contribute to the recruitment of other immune cells such as lymphocytes, macrophages and eosinophils 190. The release of mediators such as histamine, PGD2, LTC4, cytokines and others by MCs contribute to bronchoconstriction, mucus secretion and mucosal edema 232. The role of eosinophils in asthma, via release of granule proteins and other substances, is probably multifactorial (reviewed by 198). Based on clinical trials employing eosinophil targeting agents anti-IL-5 and anti-IL-5R, the most consistent finding is that of ≈50% reduction in asthma exacerbations, with little to no effect on lung function or airways hyperreactivity 233. The notable role of MCs and eosinophils in asthma makes them useful as biomarkers for diagnosis and treatment. For example different eosinophil and MC products, including Charcot-Leyden crystal protein and carboxypeptidase A3 respectively, have the potential to predict patient responses to oral corticosteroids treatment 234. Patients who successfully respond to treatment have high levels of these proteins initially, which decrease following treatment 234. Another possible marker for asthma is CD48 expressed by MCs and eosinophils. Moderate asthma patients exhibit increased CD48 compared to healthy controls, while CD48 levels in patients with severe asthma are decreased compared to moderate asthma patients. In contrast, sCD48 levels were higher in patients with mild asthma compared to control and decreased levels were detected in moderate patients compared to mild and even lower levels in patients with severe asthma 181. More studies are needed to provide the ideal combination of biomarkers to direct asthma care.
5.3 Rhinitis
Allergic rhinitis, or hay fever, manifests as inflammatory conditions that affect the nasal mucosa, usually accompanied by sneezing, itching, watery discharge and/or blockage 235. Approximately 10–40% of the population within industrialized countries, including 8–15% of children, experiences these symptoms. Episodes usually occur seasonally or perennially in geographical regions with defined seasons, but inflammation and symptoms can exist year-round in situations where continual exposure to allergens such as dust mites and pet dander are relevant. The nasal cavity is lined with epithelium that forms a barrier between the environment and underlying tissues. This epithelial layer comprises basal cells, ciliated cells, and mucus-producing goblet cells and is protective against external allergens (e.g. pollens, molds and others) 236. Epithelial cells play a key role in the pathogenesis of rhinitis, initiating type-2 inflammatory responses through secretion of TSLP, IL-25 and IL-33 237,238. Release of these inflammatory mediators can trigger production of IL-5 and IL-13 from ILC2s, directly and indirectly affecting eosinophils and MCs. Human studies have identified MCs as major effector cells driving early inflammatory responses following activation through their high-affinity IgE receptor, FcεRI 239. Eosinophils, however, are among the initial cells recruited to the nasal mucosa during the late or chronic phase of inflammation 240,241. Eosinophil activation following nasal allergen challenge was reported by Bascom et al. through the detection of eosinophil-derived proteins (MBP and ECP) in nasal fluids 242. Kountakis et al. found that the presence of nasal polyps, often a feature of chronic rhinitis, correlates with increased eosinophil activation and more severe clinical features 243. In some patients, eosinophils can make up 20% or more of total cell counts in nasal smears 244.
5.4 Food allergy and eosinophilic gastrointestinal disorders
Food allergies have recently gained a great deal of attention as the leading cause of anaphylaxis in the United States. Approximately 8% of children and 2–3% of adults are affected by food allergies 245,246. Common allergens include wheat, egg, shellfish, peanuts, tree nuts, soy and cow’s milk. Adverse reactions to food can be classified as food intolerance, a reaction that may not be reproducible and is usually dose-dependent, or food allergy that is reproducible. Patients with food allergies can be further classified into 3 sub-categories: 1) IgE-mediated, 2) cell-mediated, or 3) IgE and cell-mediated. There has been a steady increase in the number of individuals affected by food allergies, particularly in the last 20 years. Generally, symptoms of food allergy and anaphylaxis appear very soon after food consumption and can include urticaria, wheezing or coughing, nausea, vomiting and/or diarrhea, and in extreme cases, hypotension and even death. Whether such immediate reactions are due to MCs activation in tissues, as opposed to basophil activation in the circulation, cannot be easily determined, but in mouse models these are primarily MC-dependent reactions.
Histamine and tryptase levels in the blood can both increase during food-induced anaphylaxis, directly implicating MCs in food allergy 247. Furthermore, platelet activating factor, which has been linked to disease severity, can also be produced and released by MCs 247. MC activation through IgE had been linked to IL-9 and IL-33 production, both of which have been shown to play a role in disease pathogenesis. Eosinophils are more predominantly featured within the inflamed GI tract of patients with non-IgE mediated food allergies (e.g. eosinophilic esophagitis, EoE) 245. EoE features include painful swallowing (dysphagia) and sometimes food impaction. More common in males, EoE is predominantly associated with non-IgE-mediated hypersensitivity to foods 245. Inflammation is primarily driven by eosinophils but also MCs and is defined by presence of at least 15 eosinophils/HPF in biopsy sections 248. The EoE transcriptome was described, with CCL26 (eotaxin-3), an eosinophil chemoattractant, being the most highly induced gene 249. Of note, CD11c, CRTH2, ICAM-1 and the low affinity IgE receptor FcεRII (CD23) are also upregulated on eosinophils in EoE 250,251. As mentioned, MCT are elevated in esophageal tissue of EoE patients compared to healthy controls 252. Surprisingly, MCs may also drive the pathophysiology of EoE as Omani et al. demonstrated that MC numbers were attenuated within a group of pediatric EoE patients undergoing anti-IL-5 treatment 91. They also discovered that both eosinophils and MCs were producing the MC growth factor IL-9 and that MC numbers correlated with disease severity 91.
5.5 Urticaria
Urticaria, or hives, occurs in 15–25% of the population sometime during their lifetime and is characterized by pruritic lesions containing a pale center also known as a wheal with surrounding redness known as a flare 253. Associated inflammation can be acute, but nearly 30% of urticaria patients experience chronic lesions defined by recurrent episodes for at least 6 weeks 253. While urticaria can result from physical stimuli (i.e. temperature or direct stroking of the skin), non-physical cases are often categorized as autoimmune or most commonly idiopathic 254. MCs are considered a primary effector cell responsible for initiation of inflammation, in part due to release of histamine and cell-recruiting chemokines. While the exact contribution and trigger of MCs in urticaria remains controversial 255,256, it has been shown that histamine release correlates with an autoimmune phenotype 254. Recent guidelines agree that anti-histamines are the mainstay of initial treatment for chronic urticaria, even though responses can vary 257. Omalizumab, an anti-IgE monoclonal antibody, is another approved treatment option that is effective in over 60% of antihistamine-refractory patients 258. Eosinophils also participate in chronic urticaria, primarily in patients lacking an autoimmune component, and accumulate in urticarial lesions where MBP and ECP can also be detected 259. Marques et al. recently conducted a study where they found that tissue eosinophilia was linked to disease severity in patients with chronic urticaria 260.
6 Mast cells and eosinophils during bacterial, viral and parasitic infections
Beyond their involvement in allergic inflammation, MCs and eosinophils also serve a protective role during infection and host defense against viruses, bacteria and parasites. MCs and eosinophils are uniquely placed at the interface between the outer environment and tissues, allowing for their response to penetrating pathogens via production of immunoregulatory mediators. Eosinophils can also be recruited by activated MCs in inflamed tissues.
The role of MCs in viral infections is still poorly understood. However, it is known that some viruses, such as HIV, respiratory syncytial virus (RSV) and Dengue virus, can infect MCs 261–263 and remain latent. Troupin et al. recently demonstrated that Dengue virus can directly induce degranulation in human skin MCs. Furthermore, they found that the virus localized to MC secretory granules rendering them infectious, suggesting that MCs are key players in the systemic spread of Dengue 263. Eosinophils can recognize viruses via TLRs and release their mediators after activation. The mechanisms through which eosinophils promote antiviral effects are still unclear, but it is known that eosinophils cannot propagate viruses after being infected and eosinophil-derived nitrous oxide can inhibit viral proteases and alter host cell functions by promoting nitrosylation of thiol and tyrosine groups, which can impair the protein and RNA synthesis 264. Ribonuclease activity in certain eosinophil granules may also be antiviral 187,264. The role of MCs and eosinophils in viral defense still needs clarification, but the finding that reductions of eosinophils via biological therapies do not appear to increase risk of infections is reassuring.
MCs and eosinophils are also participants in fungus-associated inflammation. For example, MCs express PRRs specific for fungi, including Aspergillus and Fusarium species, highlighting their capacity to mediate antifungal responses 265. Some fungi can also infect MCs. Candida albicans can induce MCs degranulation, triggering production of pro-inflammatory cytokines. Interestingly, C. albicans can also be internalized by MCs, where it is able to grow and germinate leading to disruption of MC membranes and ultimately cell death 266. Eosinophils can directly interact with β-glucans, a component of fungal cell walls, via CD11b integrin 267. This interaction leads to eosinophil activation and release of granule proteins resulting in fungal cell death 267. Alternatively, eosinophils can be activated via proteinase-activated receptors (PARs) 268, or by products released by fungal cells 269.
Besides allergies, MCs and eosinophils are most well known for their roles in parasitic infections. When MCs recognize parasites, they form clusters around them and become activated 270, leading to release of parasite-killing mediators and tissue damage 271. For example, histamine released by activated MCs during P. falciparum infection, initiates inflammatory responses and increases vascular permeability and associated damage, driving the pathogenesis of malaria 271. During infections from T. cruzi, the number of MCs, and levels of MC-derived mediators such as chymase, is increased in the myocardium of patients with Chagas disease, resulting in myocardial fibrosis 271. During helminth infections, MCs accumulate at sites of infestation where they are involved in initiating Th2 responses, contributing to helminth expulsion 270. However, the majority of studies focusing on the role of MCs in helminthic infections are conducted in rodents, thus more studies should aim to focus on human infections to better understand the specific contribution of MCs. Helminthic infections are also characterized by high IgE production and marked blood and tissue eosinophilia 272. Comparable to MCs, eosinophils can form aggregates around parasites and kill them by ADCC 273 or through the release of granule proteins and ROS production 272. In contrast, there may be situations where eosinophils may instead benefit parasitosis and be detrimental to host cells and tissues 274, since their granule proteins and other mediators have strong cytotoxic activity 272.
In addition to viral, fungal and parasitic infections, MCs and eosinophils may be involved in bacterial immune responses. Eosinophils granule proteins EPO, ECP and MBP-1 are toxic to microorganisms. For example, ECP can reduce bacterial survival following recognition of LPS and/or peptidoglycans. EPO can generate ROS, leading to the production of hypohalous acids, with bactericidal activity 275. Both MCs and eosinophils are capable of direct and indirect killing of microbes via phagocytosis or enzymes or by recruiting other immune cells. Bacteria can trigger MC degranulation and/or selective cytokine release via pattern recognition receptors (PRRs), such as TLRs, Fc receptors, Nod-like receptors, C-type lectins, and CD48, 276. Furthermore, MC granules contain tryptase and chymase proteases that contribute to microbial killing 277. Eosinophils exhibit phagocytic activity, and both MCs and eosinophils can release their DNA to create traps 187,278,279. Whether the successful targeting of MCs and eosinophils will result in increased risks of developing parasitic or other infections remains to be seen.
7 Conclusions
MCs and eosinophils are complex immune cells that develop and function similarly in many ways. They can be beneficial when protecting us from pathogens, but they can also be detrimental in when activated inappropriately. They have overlapping immunomodulatory roles in both acute and chronic inflammation and can help orchestrate recruitment of other cells through release of pre-stored mediators, with the added capacity to synthesize additional mediators de novo. Within recent decades, the percentage of individuals affected by allergies has steadily increased and several MC/eosinophil-targeting biologics have been approved for use in allergic individuals in the last five years. These drugs are quite effective in select patient cohorts, but they are quite expensive, do not work in everyone and can have undesirable side effects. Thus, it is important to continue studying these cells, especially in humans, because they do vary in important ways from their rodent counterparts. Ultimately, an improved understanding of these closely related cells and their roles in homeostasis, and in both favorable and unfavorable immune responses, should allow us to best determine when targeting of these cells is warranted or best avoided to maintain optimal health.
Table 1.
Mast Cells | Eosinophils | ||
---|---|---|---|
Fc Receptors | FcγRI (CD64) | + 30,31 | — |
FcγRII (CD32) | + 32,33 | + 34–37 | |
FcγRIIa | + 32,39 | + 39 | |
FcγRIIb | + 40,41 | — | |
FcγRIII (CD16) | — | + 42–44 | |
FcεRI | + 45–49,51–53,280 | — | |
FcεRII (CD23) | — | + 57 | |
FcαRI | — | + 59,60 | |
Siglecs | Siglec-2 | + 61 | — |
Siglec-3 | + 61 | + 281 | |
Siglec-5 | + 61 | — | |
Siglec-6 | + 61 | — | |
Siglec-7 | + 62 | + 281 | |
Siglec-8 | + 61,64,67 | + 64–66,68 | |
Siglec-10 | + 61 | + 69 | |
CD300 Receptors | CD300a | + 70,71 | + 72 |
CD300c | + 73 | — | |
CD300f | + 75,77 | + 75,76 | |
Leukocyte Ig-like Receptors (LILR) | LILRA1 | + 80 | — |
LILRA2 | — | + 79 | |
LILRB1 | + 80 | + 79 | |
LILRB2 | + 80 | + 79 | |
LILRB3 | + 80 | + 79 | |
LILRB4 | + 80 | — | |
LILRB5 | + 80 | — | |
Cytokine Receptors | IL-4R | + 82–84 | + 20,81 |
IL-5R | ± 90 | + 19,86 | |
IL-13Rα1 | + 45,95 | + 94 | |
c-kit (CD117) | + 96,101,102 | + 100 | |
TSLPR | + 106,107 | + 109,110 | |
IL1RL1 (ST2) | + 115–119 | + 18,68,122–125 | |
Migratory Receptors and Others | α4β1 integrin (VLA-4) | + 129,131 | + 128,132,133 |
α4β7 integrin | + 131,134,135,139 | + 66,136–138 | |
CRTH2 | ± 144 | + 140–142 | |
C3aR | + 152,153 | + 149,150 | |
C5aR | + 152 | + 150,151 | |
C5aR2 | + 156 | ± 157 | |
S1PR1 | + 159 | + 162 | |
S1PR2 | + 154,155,159–161 | — | |
S1PR3 | — | + 162 | |
S1PR4 | — | + 162 | |
S1PR5 | — | + 162 | |
H1R | + 171 | + 164 | |
H2R | + 171 | + 165 | |
H4R | + 163,168 | + 166,167 | |
CB1 | + 282–284 | — | |
CB2 | + 283 | + 172,173 | |
TLRs | ± 176,177 | ± 176,177 | |
CD48 | + 180 | + 181,182,184 | |
2B4 (CD244) | ± 98 | + 72,179,183 |
Abbreviations: Immunoglobulin (Ig) G receptor (FcγR), IgE receptor (FcεR), IgA receptor (FcαR), (Sialic acid-binding Ig-type lectin (Siglec), Leukocyte Ig-like receptor (LILR), Interleukin 1 receptor-like 1 (IL1RL1), Prostaglandin D2 receptor 2 (CRTH2), Sphingosine-1-phosphate receptor (S1PR), Histamine receptor (HR), Cannabinoid receptor (CB), Toll-like receptor (TLR), Natural killer cell receptor (2B4).
Acknowledgments
Funding:
Israel Science Foundation
U.S. Department of Health and Human Services, National Institutes of Health, National Heart, Lung, and Blood Institute P01 HL107151
U.S. Department of Health and Human Services, National Institutes of Health, National Institute of Allergy and Infectious Diseases R01 AI072265, R01 AI105839 and T32 AI083216
United States-Israel Binational Science Foundation.
Footnotes
Conflict of Interest:
B.S.B. has current or recent consulting or scientific advisory board arrangements with or has received honoraria from Sanofi-Aventis, TEVA, GlaxoSmithKline, AstraZeneca and Allakos, and owns stock in Allakos and Glycomimetics. He receives publication-related royalty payments from Elsevier and UpToDate™ and is a co-inventor on existing Siglec-8-related patents and thus may be entitled to a share of royalties received by Johns Hopkins University on the potential sales of such products. B.S.B. is also a co-founder of Allakos, which makes him subject to certain restrictions under University policy. The terms of this arrangement are being managed by the Johns Hopkins University and Northwestern University in accordance with their conflict of interest policies. The authors have no additional competing financial interests.
References
- 1.Mori Y, Iwasaki H, Kohno K, et al. Identification of the human eosinophil lineage-committed progenitor: revision of phenotypic definition of the human common myeloid progenitor. J Exp Med. 2009;206(1):183–193. doi: 10.1084/jem.20081756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Dahlin JS, Hallgren J. Mast cell progenitors: origin, development and migration to tissues. Mol Immunol. 2015;63(1):9–17. doi: 10.1016/j.molimm.2014.01.018. [DOI] [PubMed] [Google Scholar]
- 3.Schmetzer O, Valentin P, Church MK, Maurer M, Siebenhaar F. Murine and human mast cell progenitors. Eur J Pharmacol. 2016;778:2–10. doi: 10.1016/j.ejphar.2015.07.016. [DOI] [PubMed] [Google Scholar]
- 4.Hirasawa R, Shimizu R, Takahashi S, et al. Essential and instructive roles of GATA factors in eosinophil development. J Exp Med. 2002;195(11):1379–1386. doi: 10.1084/jem.20020170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Iwama A, Osawa M, Hirasawa R, et al. Reciprocal roles for CCAAT/enhancer binding protein (C/EBP) and PU.1 transcription factors in Langerhans cell commitment. J Exp Med. 2002;195(5):547–558. doi: 10.1084/jem.20011465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Lee JJ, Jacobsen EA, Ochkur SI, et al. Human versus mouse eosinophils: “that which we call an eosinophil, by any other name would stain as red”. J Allergy Clin Immunol. 2012;130(3):572–584. doi: 10.1016/j.jaci.2012.07.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.McNagny K, Graf T. Making eosinophils through subtle shifts in transcription factor expression. J Exp Med. 2002;195(11):F43–47. doi: 10.1084/jem.20020636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Babina M, Schulke Y, Kirchhof L, et al. The transcription factor profile of human mast cells in comparison with monocytes and granulocytes. Cell Mol Life Sci. 2005;62(2):214–226. doi: 10.1007/s00018-004-4480-6. [DOI] [PubMed] [Google Scholar]
- 9.Drissen R, Buza-Vidas N, Woll P, et al. Distinct myeloid progenitor-differentiation pathways identified through single-cell RNA sequencing. Nat Immunol. 2016;17(6):666–676. doi: 10.1038/ni.3412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Bouffi C, Kartashov AV, Schollaert KL, et al. Transcription Factor Repertoire of Homeostatic Eosinophilopoiesis. J Immunol. 2015;195(6):2683–2695. doi: 10.4049/jimmunol.1500510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Bettigole SE, Lis R, Adoro S, et al. The transcription factor XBP1 is selectively required for eosinophil differentiation. Nat Immunol. 2015;16(8):829–837. doi: 10.1038/ni.3225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Matthews SP, McMillan SJ, Colbert JD, Lawrence RA, Watts C. Cystatin F Ensures Eosinophil Survival by Regulating Granule Biogenesis. Immunity. 2016;44(4):795–806. doi: 10.1016/j.immuni.2016.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ribatti D. The development of human mast cells. An historical reappraisal. Exp Cell Res. 2016;342(2):210–215. doi: 10.1016/j.yexcr.2016.03.013. [DOI] [PubMed] [Google Scholar]
- 14.Varricchi G, Galdiero MR, Loffredo S, et al. Are Mast Cells MASTers in Cancer? Frontiers in Immunology. 2017:8. doi: 10.3389/fimmu.2017.00424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Paul CC, Tolbert M, Mahrer S, Singh A, Grace MJ, Baumann MA. Cooperative effects of interleukin-3 (IL-3), IL-5, and granulocyte-macrophage colony-stimulating factor: a new myeloid cell line inducible to eosinophils. Blood. 1993;81(5):1193–1199. [PubMed] [Google Scholar]
- 16.Clutterbuck EJ, Hirst EM, Sanderson CJ. Human interleukin-5 (IL-5) regulates the production of eosinophils in human bone marrow cultures: comparison and interaction with IL-1, IL-3, IL-6, and GMCSF. Blood. 1989;73(6):1504–1512. [PubMed] [Google Scholar]
- 17.Yanagibashi T, Satoh M, Nagai Y, Koike M, Takatsu K. Allergic diseases: From bench to clinic - Contribution of the discovery of interleukin-5. Cytokine. 2017;98:59–70. doi: 10.1016/j.cyto.2016.11.011. [DOI] [PubMed] [Google Scholar]
- 18.Johnston LK, Hsu CL, Krier-Burris RA, et al. IL-33 Precedes IL-5 in Regulating Eosinophil Commitment and Is Required for Eosinophil Homeostasis. J Immunol. 2016;197(9):3445–3453. doi: 10.4049/jimmunol.1600611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Sehmi R, Wood LJ, Watson R, et al. Allergen-induced increases in IL-5 receptor alpha-subunit expression on bone marrow-derived CD34+ cells from asthmatic subjects. A novel marker of progenitor cell commitment towards eosinophilic differentiation. J Clin Invest. 1997;100(10):2466–2475. doi: 10.1172/JCI119789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Fulkerson PC, Schollaert KL, Bouffi C, Rothenberg ME. IL-5 triggers a cooperative cytokine network that promotes eosinophil precursor maturation. J Immunol. 2014;193(8):4043–4052. doi: 10.4049/jimmunol.1400732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Rothenberg ME, Hogan SP. The eosinophil. Annu Rev Immunol. 2006;24:147–174. doi: 10.1146/annurev.immunol.24.021605.090720. [DOI] [PubMed] [Google Scholar]
- 22.Ochi H, Hirani WM, Yuan Q, Friend DS, Austen KF, Boyce JA. T helper cell type 2 cytokine-mediated comitogenic responses and CCR3 expression during differentiation of human mast cells in vitro. J Exp Med. 1999;190(2):267–280. doi: 10.1084/jem.190.2.267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Okayama Y, Kawakami T. Development, migration, and survival of mast cells. Immunol Res. 2006;34(2):97–115. doi: 10.1385/IR:34:2:97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Levi-Schaffer F, Austen KF, Gravallese PM, Stevens RL. Coculture of interleukin 3-dependent mouse mast cells with fibroblasts results in a phenotypic change of the mast cells. Proc Natl Acad Sci U S A. 1986;83(17):6485–6488. doi: 10.1073/pnas.83.17.6485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Yanagida M, Fukamachi H, Ohgami K, et al. Effects of T-helper 2-type cytokines, interleukin-3 (IL-3), IL-4, IL-5, and IL-6 on the survival of cultured human mast cells. Blood. 1995;86(10):3705–3714. [PubMed] [Google Scholar]
- 26.Valent P, Besemer J, Sillaber C, et al. Failure to detect IL-3-binding sites on human mast cells. J Immunol. 1990;145(10):3432–3437. [PubMed] [Google Scholar]
- 27.Godot V, Arock M, Garcia G, et al. H4 histamine receptor mediates optimal migration of mast cell precursors to CXCL12. J Allergy Clin Immunol. 2007;120(4):827–834. doi: 10.1016/j.jaci.2007.05.046. [DOI] [PubMed] [Google Scholar]
- 28.Weller CL, Collington SJ, Brown JK, et al. Leukotriene B4, an activation product of mast cells, is a chemoattractant for their progenitors. J Exp Med. 2005;201(12):1961–1971. doi: 10.1084/jem.20042407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.El-Shazly A, Berger P, Girodet PO, et al. Fraktalkine produced by airway smooth muscle cells contributes to mast cell recruitment in asthma. J Immunol. 2006;176(3):1860–1868. doi: 10.4049/jimmunol.176.3.1860. [DOI] [PubMed] [Google Scholar]
- 30.Okayama Y, Kirshenbaum AS, Metcalfe DD. Expression of a functional high-affinity IgG receptor, Fc gamma RI, on human mast cells: Up-regulation by IFN-gamma. J Immunol. 2000;164(8):4332–4339. doi: 10.4049/jimmunol.164.8.4332. [DOI] [PubMed] [Google Scholar]
- 31.Tkaczyk C, Okayama Y, Woolhiser MR, Hagaman DD, Gilfillan AM, Metcalfe DD. Activation of human mast cells through the high affinity IgG receptor. Mol Immunol. 2002;38(16–18):1289–1293. doi: 10.1016/s0161-5890(02)00077-9. [DOI] [PubMed] [Google Scholar]
- 32.Zhao W, Kepley CL, Morel PA, Okumoto LM, Fukuoka Y, Schwartz LB. Fc gamma RIIa, not Fc gamma RIIb, is constitutively and functionally expressed on skin-derived human mast cells. J Immunol. 2006;177(1):694–701. doi: 10.4049/jimmunol.177.1.694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Okayama Y, Hagaman DD, Woolhiser M, Metcalfe DD. Further characterization of FcgammaRII and FcgammaRIII expression by cultured human mast cells. Int Arch Allergy Immunol. 2001;124(1–3):155–157. doi: 10.1159/000053696. [DOI] [PubMed] [Google Scholar]
- 34.Hartnell A, Moqbel R, Walsh GM, Bradley B, Kay AB. Fc gamma and CD11/CD18 receptor expression on normal density and low density human eosinophils. Immunology. 1990;69(2):264–270. [PMC free article] [PubMed] [Google Scholar]
- 35.van der Bruggen T, Kok PT, Raaijmakers JA, Lammers JW, Koenderman L. Cooperation between Fc gamma receptor II and complement receptor type 3 during activation of platelet-activating factor release by cytokine-primed human eosinophils. J Immunol. 1994;153(6):2729–2735. [PubMed] [Google Scholar]
- 36.Kaneko M, Swanson MC, Gleich GJ, Kita H. Allergen-specific IgG1 and IgG3 through Fc gamma RII induce eosinophil degranulation. J Clin Invest. 1995;95(6):2813–2821. doi: 10.1172/JCI117986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Kim JT, Schimming AW, Kita H. Ligation of Fc gamma RII (CD32) pivotally regulates survival of human eosinophils. J Immunol. 1999;162(7):4253–4259. [PubMed] [Google Scholar]
- 38.Van den Herik-Oudijk IE, Capel PJ, van der Bruggen T, Van de Winkel JG. Identification of signaling motifs within human Fc gamma RIIa and Fc gamma RIIb isoforms. Blood. 1995;85(8):2202–2211. [PubMed] [Google Scholar]
- 39.Jonsson F, Mancardi DA, Zhao W, et al. Human FcgammaRIIA induces anaphylactic and allergic reactions. Blood. 2012;119(11):2533–2544. doi: 10.1182/blood-2011-07-367334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Malbec O, Attal JP, Fridman WH, Daeron M. Negative regulation of mast cell proliferation by FcgammaRIIB. Mol Immunol. 2002;38(16–18):1295–1299. doi: 10.1016/s0161-5890(02)00078-0. [DOI] [PubMed] [Google Scholar]
- 41.Mertsching E, Bafetti L, Hess H, et al. A mouse Fcgamma-Fcepsilon protein that inhibits mast cells through activation of FcgammaRIIB, SH2 domain-containing inositol phosphatase 1, and SH2 domain-containing protein tyrosine phosphatases. J Allergy Clin Immunol. 2008;121(2):441–447. e445. doi: 10.1016/j.jaci.2007.08.051. [DOI] [PubMed] [Google Scholar]
- 42.Davoine F, Lavigne S, Chakir J, Ferland C, Boulay ME, Laviolette M. Expression of FcgammaRIII (CD16) on human peripheral blood eosinophils increases in allergic conditions. J Allergy Clin Immunol. 2002;109(3):463–469. doi: 10.1067/mai.2002.121952. [DOI] [PubMed] [Google Scholar]
- 43.Hartnell A, Kay AB, Wardlaw AJ. IFN-gamma induces expression of Fc gamma RIII (CD16) on human eosinophils. J Immunol. 1992;148(5):1471–1478. [PubMed] [Google Scholar]
- 44.Zhu X, Hamann KJ, Munoz NM, et al. Intracellular expression of Fc gamma RIII (CD16) and its mobilization by chemoattractants in human eosinophils. J Immunol. 1998;161(5):2574–2579. [PubMed] [Google Scholar]
- 45.Kaur D, Hollins F, Woodman L, et al. Mast cells express IL-13R alpha 1: IL-13 promotes human lung mast cell proliferation and Fc epsilon RI expression. Allergy. 2006;61(9):1047–1053. doi: 10.1111/j.1398-9995.2006.01139.x. [DOI] [PubMed] [Google Scholar]
- 46.Xia HZ, Du Z, Craig S, et al. Effect of recombinant human IL-4 on tryptase, chymase, and Fc epsilon receptor type I expression in recombinant human stem cell factor-dependent fetal liver-derived human mast cells. J Immunol. 1997;159(6):2911–2921. [PubMed] [Google Scholar]
- 47.Yamaguchi M, Sayama K, Yano K, et al. IgE enhances Fc epsilon receptor I expression and IgE-dependent release of histamine and lipid mediators from human umbilical cord blood-derived mast cells: synergistic effect of IL-4 and IgE on human mast cell Fc epsilon receptor I expression and mediator release. J Immunol. 1999;162(9):5455–5465. [PubMed] [Google Scholar]
- 48.Welker P, Grabbe J, Zuberbier T, Henz BM. GM-CSF downregulates expression of tryptase, Fc epsilon RI and histamine in HMC-1 mast cells. Int Arch Allergy Immunol. 1997;113(1–3):284–286. doi: 10.1159/000237574. [DOI] [PubMed] [Google Scholar]
- 49.Gomez G, Ramirez CD, Rivera J, et al. TGF-beta 1 inhibits mast cell Fc epsilon RI expression. J Immunol. 2005;174(10):5987–5993. doi: 10.4049/jimmunol.174.10.5987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Beck LA, Marcotte GV, MacGlashan D, Togias A, Saini S. Omalizumab-induced reductions in mast cell Fce psilon RI expression and function. J Allergy Clin Immunol. 2004;114(3):527–530. doi: 10.1016/j.jaci.2004.06.032. [DOI] [PubMed] [Google Scholar]
- 51.Ra C, Nunomura S, Okayama Y. Fine-Tuning of Mast Cell Activation by FcepsilonRIbeta Chain. Front Immunol. 2012;3:112. doi: 10.3389/fimmu.2012.00112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ryan JJ, Kinzer CA, Paul WE. Mast cells lacking the high affinity immunoglobulin E receptor are deficient in Fc epsilon RI gamma messenger RNA. J Exp Med. 1995;182(2):567–574. doi: 10.1084/jem.182.2.567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kepley CL, Taghavi S, Mackay G, et al. Co-aggregation of FcgammaRII with FcepsilonRI on human mast cells inhibits antigen-induced secretion and involves SHIP-Grb2-Dok complexes. J Biol Chem. 2004;279(34):35139–35149. doi: 10.1074/jbc.M404318200. [DOI] [PubMed] [Google Scholar]
- 54.Kayaba H, Dombrowicz D, Woerly G, Papin JP, Loiseau S, Capron M. Human eosinophils and human high affinity IgE receptor transgenic mouse eosinophils express low levels of high affinity IgE receptor, but release IL-10 upon receptor activation. J Immunol. 2001;167(2):995–1003. doi: 10.4049/jimmunol.167.2.995. [DOI] [PubMed] [Google Scholar]
- 55.Smith SJ, Ying S, Meng Q, et al. Blood eosinophils from atopic donors express messenger RNA for the alpha, beta, and gamma subunits of the high-affinity IgE receptor (Fc epsilon RI) and intracellular, but not cell surface, alpha subunit protein. J Allergy Clin Immunol. 2000;105(2 Pt 1):309–317. doi: 10.1016/s0091-6749(00)90081-2. [DOI] [PubMed] [Google Scholar]
- 56.Iikura M, Yamaguchi M, Hirai K, et al. Regulation of surface FcepsilonRI expression on human eosinophils by IL-4 and IgE. Int Arch Allergy Immunol. 2001;124(4):470–477. doi: 10.1159/000053782. [DOI] [PubMed] [Google Scholar]
- 57.Sano H, Munoz NM, Sano A, et al. Upregulated surface expression of intracellularly sequestered Igepsilon receptors (FcepsilonRII/CD23) following activation in human peripheral blood eosinophils. Proc Assoc Am Physicians. 1999;111(1):82–91. doi: 10.1046/j.1525-1381.1999.09668.x. [DOI] [PubMed] [Google Scholar]
- 58.Pleass RJ, Dunlop JI, Woof JM. Multiple transcripts of human IgA Fc receptor CD89 in neutrophils, eosinophils and the monocyte-like cell line THP-1. Biochem Soc Trans. 1997;25(2):327S. doi: 10.1042/bst025327s. [DOI] [PubMed] [Google Scholar]
- 59.Bracke M, Dubois GR, Bolt K, et al. Differential effects of the T helper cell type 2-derived cytokines IL-4 and IL-5 on ligand binding to IgG and IgA receptors expressed by human eosinophils. J Immunol. 1997;159(3):1459–1465. [PubMed] [Google Scholar]
- 60.ten Hove W, Houben LA, Raaijmakers JA, Koenderman L, Bracke M. Rapid selective priming of FcalphaR on eosinophils by corticosteroids. J Immunol. 2006;177(9):6108–6114. doi: 10.4049/jimmunol.177.9.6108. [DOI] [PubMed] [Google Scholar]
- 61.Yokoi H, Myers A, Matsumoto K, Crocker PR, Saito H, Bochner BS. Alteration and acquisition of Siglecs during in vitro maturation of CD34+ progenitors into human mast cells. Allergy. 2006;61(6):769–776. doi: 10.1111/j.1398-9995.2006.01133.x. [DOI] [PubMed] [Google Scholar]
- 62.Mizrahi S, Gibbs BF, Karra L, Ben-Zimra M, Levi-Schaffer F. Siglec-7 is an inhibitory receptor on human mast cells and basophils. J Allergy Clin Immunol. 2014;134(1):230–233. doi: 10.1016/j.jaci.2014.03.031. [DOI] [PubMed] [Google Scholar]
- 63.Tateno H, Crocker PR, Paulson JC. Mouse Siglec-F and human Siglec-8 are functionally convergent paralogs that are selectively expressed on eosinophils and recognize 6′-sulfo-sialyl Lewis X as a preferred glycan ligand. Glycobiology. 2005;15(11):1125–1135. doi: 10.1093/glycob/cwi097. [DOI] [PubMed] [Google Scholar]
- 64.Bochner BS. Siglec-8 on human eosinophils and mast cells, and Siglec-F on murine eosinophils, are functionally related inhibitory receptors. Clin Exp Allergy. 2009;39(3):317–324. doi: 10.1111/j.1365-2222.2008.03173.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Nutku-Bilir E, Hudson SA, Bochner BS. Interleukin-5 priming of human eosinophils alters siglec-8 mediated apoptosis pathways. Am J Respir Cell Mol Biol. 2008;38(1):121–124. doi: 10.1165/rcmb.2007-0154OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Carroll DJ, O’Sullivan JA, Nix DB, Cao Y, Tiemeyer M, Bochner BS. Siglec-8 is an activating receptor mediating beta2 integrin-dependent function in human eosinophils. J Allergy Clin Immunol. 2017 doi: 10.1016/j.jaci.2017.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Yokoi H, Choi OH, Hubbard W, et al. Inhibition of FcepsilonRI-dependent mediator release and calcium flux from human mast cells by sialic acid-binding immunoglobulin-like lectin 8 engagement. J Allergy Clin Immunol. 2008;121(2):499–505. e491. doi: 10.1016/j.jaci.2007.10.004. [DOI] [PubMed] [Google Scholar]
- 68.Na HJ, Hudson SA, Bochner BS. IL-33 enhances Siglec-8 mediated apoptosis of human eosinophils. Cytokine. 2012;57(1):169–174. doi: 10.1016/j.cyto.2011.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Munday J, Kerr S, Ni J, et al. Identification, characterization and leucocyte expression of Siglec-10, a novel human sialic acid-binding receptor. Biochem J. 2001;355(Pt 2):489–497. doi: 10.1042/0264-6021:3550489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Bachelet I, Munitz A, Berent-Maoz B, Mankuta D, Levi-Schaffer F. Suppression of normal and malignant kit signaling by a bispecific antibody linking kit with CD300a. J Immunol. 2008;180(9):6064–6069. doi: 10.4049/jimmunol.180.9.6064. [DOI] [PubMed] [Google Scholar]
- 71.Nakahashi-Oda C, Tahara-Hanaoka S, Shoji M, et al. Apoptotic cells suppress mast cell inflammatory responses via the CD300a immunoreceptor. J Exp Med. 2012;209(8):1493–1503. doi: 10.1084/jem.20120096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Munitz A, Bachelet I, Levi-Schaffer F. Reversal of airway inflammation and remodeling in asthma by a bispecific antibody fragment linking CCR3 to CD300a. J Allergy Clin Immunol. 2006;118(5):1082–1089. doi: 10.1016/j.jaci.2006.07.041. [DOI] [PubMed] [Google Scholar]
- 73.Takahashi M, Izawa K, Kashiwakura J, et al. Human CD300C delivers an Fc receptor-gamma-dependent activating signal in mast cells and monocytes and differs from CD300A in ligand recognition. J Biol Chem. 2013;288(11):7662–7675. doi: 10.1074/jbc.M112.434746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Dimitrova M, Zenarruzabeitia O, Borrego F, Simhadri VR. CD300c is uniquely expressed on CD56 bright Natural Killer Cells and differs from CD300a upon ligand recognition. Sci Rep. 2016;6:23942. doi: 10.1038/srep23942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Borrego F. The CD300 molecules: an emerging family of regulators of the immune system. Blood. 2013;121(11):1951–1960. doi: 10.1182/blood-2012-09-435057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Moshkovits I, Karo-Atar D, Itan M, et al. CD300f associates with IL-4 receptor alpha and amplifies IL-4-induced immune cell responses. Proc Natl Acad Sci U S A. 2015;112(28):8708–8713. doi: 10.1073/pnas.1507625112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Izawa K, Isobe M, Matsukawa T, et al. Sphingomyelin and ceramide are physiological ligands for human LMIR3/CD300f, inhibiting FcepsilonRI-mediated mast cell activation. J Allergy Clin Immunol. 2014;133(1):270–273. e271–277. doi: 10.1016/j.jaci.2013.08.008. [DOI] [PubMed] [Google Scholar]
- 78.Bulfone-Paus S, Nilsson G, Draber P, Blank U, Levi-Schaffer F. Positive and Negative Signals in Mast Cell Activation. Trends Immunol. 2017;38(9):657–667. doi: 10.1016/j.it.2017.01.008. [DOI] [PubMed] [Google Scholar]
- 79.Tedla N, Bandeira-Melo C, Tassinari P, et al. Activation of human eosinophils through leukocyte immunoglobulin-like receptor 7. Proc Natl Acad Sci U S A. 2003;100(3):1174–1179. doi: 10.1073/pnas.0337567100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Tedla N, Lee CW, Borges L, Geczy CL, Arm JP. Differential expression of leukocyte immunoglobulin-like receptors on cord-blood-derived human mast cell progenitors and mature mast cells. J Leukoc Biol. 2008;83(2):334–343. doi: 10.1189/jlb.0507314. [DOI] [PubMed] [Google Scholar]
- 81.Wedi B, Raap U, Lewrick H, Kapp A. IL-4-induced apoptosis in peripheral blood eosinophils. J Allergy Clin Immunol. 1998;102(6 Pt 1):1013–1020. doi: 10.1016/s0091-6749(98)70340-9. [DOI] [PubMed] [Google Scholar]
- 82.Toru H, Pawankar R, Ra C, Yata J, Nakahata T. Human mast cells produce IL-13 by high-affinity IgE receptor cross-linking: enhanced IL-13 production by IL-4-primed human mast cells. J Allergy Clin Immunol. 1998;102(3):491–502. doi: 10.1016/s0091-6749(98)70140-x. [DOI] [PubMed] [Google Scholar]
- 83.Niggemann B, Zuberbier T, Herz U, Enssle K, Wahn U, Renz H. Interleukin-4 (IL-4) enhances and soluble interleukin-4 receptor (sIL-4R) inhibits histamine release from peripheral blood basophils and mast cells in vitro and in vivo. Mediators Inflamm. 1997;6(2):111–118. doi: 10.1080/09629359791802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Oskeritzian CA, Wang Z, Kochan JP, et al. Recombinant human (rh)IL-4-mediated apoptosis and recombinant human IL-6-mediated protection of recombinant human stem cell factor-dependent human mast cells derived from cord blood mononuclear cell progenitors. J Immunol. 1999;163(9):5105–5115. [PubMed] [Google Scholar]
- 85.Kotsimbos AT, Hamid Q. IL-5 and IL-5 receptor in asthma. Mem Inst Oswaldo Cruz. 1997;92(Suppl 2):75–91. doi: 10.1590/s0074-02761997000800012. [DOI] [PubMed] [Google Scholar]
- 86.Migita M, Yamaguchi N, Mita S, et al. Characterization of the human IL-5 receptors on eosinophils. Cell Immunol. 1991;133(2):484–497. doi: 10.1016/0008-8749(91)90120-z. [DOI] [PubMed] [Google Scholar]
- 87.Varricchi G, Senna G, Loffredo S, Bagnasco D, Ferrando M, Canonica GW. Reslizumab and Eosinophilic Asthma: One Step Closer to Precision Medicine? Front Immunol. 2017;8:242. doi: 10.3389/fimmu.2017.00242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Albers FC, Price RG, Smith SG, Yancey SW. Mepolizumab efficacy in patients with severe eosinophilic asthma receiving different controller therapies. J Allergy Clin Immunol. 2017 doi: 10.1016/j.jaci.2017.06.010. [DOI] [PubMed] [Google Scholar]
- 89.Ferguson GT, FitzGerald JM, Bleecker ER, et al. Benralizumab for patients with mild to moderate, persistent asthma (BISE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir Med. 2017;5(7):568–576. doi: 10.1016/S2213-2600(17)30190-X. [DOI] [PubMed] [Google Scholar]
- 90.Dahl C, Hoffmann HJ, Saito H, Schiotz PO. Human mast cells express receptors for IL-3, IL-5 and GM-CSF; a partial map of receptors on human mast cells cultured in vitro. Allergy. 2004;59(10):1087–1096. doi: 10.1111/j.1398-9995.2004.00606.x. [DOI] [PubMed] [Google Scholar]
- 91.Otani IM, Anilkumar AA, Newbury RO, et al. Anti-IL-5 therapy reduces mast cell and IL-9 cell numbers in pediatric patients with eosinophilic esophagitis. J Allergy Clin Immunol. 2013;131(6):1576–1582. doi: 10.1016/j.jaci.2013.02.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Conus S, Straumann A, Bettler E, Simon HU. Mepolizumab does not alter levels of eosinophils, T cells, and mast cells in the duodenal mucosa in eosinophilic esophagitis. J Allergy Clin Immunol. 2010;126(1):175–177. doi: 10.1016/j.jaci.2010.04.029. [DOI] [PubMed] [Google Scholar]
- 93.Schnyder B, Lugli S, Feng N, et al. Interleukin-4 (IL-4) and IL-13 bind to a shared heterodimeric complex on endothelial cells mediating vascular cell adhesion molecule-1 induction in the absence of the common gamma chain. Blood. 1996;87(10):4286–4295. [PubMed] [Google Scholar]
- 94.Myrtek D, Knoll M, Matthiesen T, et al. Expression of interleukin-13 receptor alpha 1-subunit on peripheral blood eosinophils is regulated by cytokines. Immunology. 2004;112(4):597–604. doi: 10.1046/j.1365-2567.2004.01897.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Gauchat JF, Schlagenhauf E, Feng NP, et al. A novel 4-kb interleukin-13 receptor alpha mRNA expressed in human B, T, and endothelial cells encoding an alternate type-II interleukin-4/interleukin-13 receptor. Eur J Immunol. 1997;27(4):971–978. doi: 10.1002/eji.1830270425. [DOI] [PubMed] [Google Scholar]
- 96.Reber L, Da Silva CA, Frossard N. Stem cell factor and its receptor c-Kit as targets for inflammatory diseases. Eur J Pharmacol. 2006;533(1–3):327–340. doi: 10.1016/j.ejphar.2005.12.067. [DOI] [PubMed] [Google Scholar]
- 97.Hartman M, Piliponsky AM, Temkin V, Levi-Schaffer F. Human peripheral blood eosinophils express stem cell factor. Blood. 2001;97(4):1086–1091. doi: 10.1182/blood.v97.4.1086. [DOI] [PubMed] [Google Scholar]
- 98.Elishmereni M, Alenius HT, Bradding P, et al. Physical interactions between mast cells and eosinophils: a novel mechanism enhancing eosinophil survival in vitro. Allergy. 2011;66(3):376–385. doi: 10.1111/j.1398-9995.2010.02494.x. [DOI] [PubMed] [Google Scholar]
- 99.Minai-Fleminger Y, Elishmereni M, Vita F, et al. Ultrastructural evidence for human mast cell-eosinophil interactions in vitro. Cell Tissue Res. 2010;341(3):405–415. doi: 10.1007/s00441-010-1010-8. [DOI] [PubMed] [Google Scholar]
- 100.Yuan Q, Austen KF, Friend DS, Heidtman M, Boyce JA. Human peripheral blood eosinophils express a functional c-kit receptor for stem cell factor that stimulates very late antigen 4 (VLA-4)-mediated cell adhesion to fibronectin and vascular cell adhesion molecule 1 (VCAM-1) J Exp Med. 1997;186(2):313–323. doi: 10.1084/jem.186.2.313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Lennartsson J, Ronnstrand L. Stem cell factor receptor/c-Kit: from basic science to clinical implications. Physiol Rev. 2012;92(4):1619–1649. doi: 10.1152/physrev.00046.2011. [DOI] [PubMed] [Google Scholar]
- 102.Metcalfe DD, Mekori YA. Pathogenesis and Pathology of Mastocytosis. Annu Rev Pathol. 2017;12:487–514. doi: 10.1146/annurev-pathol-052016-100312. [DOI] [PubMed] [Google Scholar]
- 103.Schwaab J, Umbach R, Metzgeroth G, et al. KIT D816V and JAK2 V617F mutations are seen recurrently in hypereosinophilia of unknown significance. Am J Hematol. 2015;90(9):774–777. doi: 10.1002/ajh.24075. [DOI] [PubMed] [Google Scholar]
- 104.Cahill KN, Katz HR, Cui J, et al. KIT Inhibition by Imatinib in Patients with Severe Refractory Asthma. N Engl J Med. 2017;376(20):1911–1920. doi: 10.1056/NEJMoa1613125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Reche PA, Soumelis V, Gorman DM, et al. Human thymic stromal lymphopoietin preferentially stimulates myeloid cells. J Immunol. 2001;167(1):336–343. doi: 10.4049/jimmunol.167.1.336. [DOI] [PubMed] [Google Scholar]
- 106.Nagarkar DR, Poposki JA, Tan BK, et al. Thymic stromal lymphopoietin activity is increased in nasal polyps of patients with chronic rhinosinusitis. J Allergy Clin Immunol. 2013;132(3):593–600. e512. doi: 10.1016/j.jaci.2013.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Allakhverdi Z, Comeau MR, Jessup HK, et al. Thymic stromal lymphopoietin is released by human epithelial cells in response to microbes, trauma, or inflammation and potently activates mast cells. J Exp Med. 2007;204(2):253–258. doi: 10.1084/jem.20062211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Okayama Y, Okumura S, Sagara H, et al. FcepsilonRI-mediated thymic stromal lymphopoietin production by interleukin-4-primed human mast cells. Eur Respir J. 2009;34(2):425–435. doi: 10.1183/09031936.00121008. [DOI] [PubMed] [Google Scholar]
- 109.Cook EB, Stahl JL, Schwantes EA, Fox KE, Mathur SK. IL-3 and TNFalpha increase Thymic Stromal Lymphopoietin Receptor (TSLPR) expression on eosinophils and enhance TSLP-stimulated degranulation. Clin Mol Allergy. 2012;10(1):8. doi: 10.1186/1476-7961-10-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Wong CK, Hu S, Cheung PF, Lam CW. Thymic stromal lymphopoietin induces chemotactic and prosurvival effects in eosinophils: implications in allergic inflammation. Am J Respir Cell Mol Biol. 2010;43(3):305–315. doi: 10.1165/rcmb.2009-0168OC. [DOI] [PubMed] [Google Scholar]
- 111.Gauvreau GM, O’Byrne PM, Boulet LP, et al. Effects of an anti-TSLP antibody on allergen-induced asthmatic responses. N Engl J Med. 2014;370(22):2102–2110. doi: 10.1056/NEJMoa1402895. [DOI] [PubMed] [Google Scholar]
- 112.Corren J, Parnes JR, Wang L, et al. Tezepelumab in Adults with Uncontrolled Asthma. N Engl J Med. 2017;377(10):936–946. doi: 10.1056/NEJMoa1704064. [DOI] [PubMed] [Google Scholar]
- 113.Baba Y, Maeda K, Yashiro T, et al. Involvement of PU.1 in mast cell/basophil-specific function of the human IL1RL1/ST2 promoter. Allergol Int. 2012;61(3):461–467. doi: 10.2332/allergolint.12-OA-0424. [DOI] [PubMed] [Google Scholar]
- 114.Tominaga S, Kuroiwa K, Tago K, Iwahana H, Yanagisawa K, Komatsu N. Presence and expression of a novel variant form of ST2 gene product in human leukemic cell line UT-7/GM. Biochem Biophys Res Commun. 1999;264(1):14–18. doi: 10.1006/bbrc.1999.1469. [DOI] [PubMed] [Google Scholar]
- 115.Bandara G, Beaven MA, Olivera A, Gilfillan AM, Metcalfe DD. Activated mast cells synthesize and release soluble ST2-a decoy receptor for IL-33. Eur J Immunol. 2015;45(11):3034–3044. doi: 10.1002/eji.201545501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Moritz DR, Rodewald HR, Gheyselinck J, Klemenz R. The IL-1 receptor-related T1 antigen is expressed on immature and mature mast cells and on fetal blood mast cell progenitors. J Immunol. 1998;161(9):4866–4874. [PubMed] [Google Scholar]
- 117.Ho LH, Ohno T, Oboki K, et al. IL-33 induces IL-13 production by mouse mast cells independently of IgE-FcepsilonRI signals. J Leukoc Biol. 2007;82(6):1481–1490. doi: 10.1189/jlb.0407200. [DOI] [PubMed] [Google Scholar]
- 118.Moulin D, Donze O, Talabot-Ayer D, Mezin F, Palmer G, Gabay C. Interleukin (IL)-33 induces the release of pro-inflammatory mediators by mast cells. Cytokine. 2007;40(3):216–225. doi: 10.1016/j.cyto.2007.09.013. [DOI] [PubMed] [Google Scholar]
- 119.Allakhverdi Z, Smith DE, Comeau MR, Delespesse G. Cutting edge: The ST2 ligand IL-33 potently activates and drives maturation of human mast cells. J Immunol. 2007;179(4):2051–2054. doi: 10.4049/jimmunol.179.4.2051. [DOI] [PubMed] [Google Scholar]
- 120.Hsu CL, Neilsen CV, Bryce PJ. IL-33 is produced by mast cells and regulates IgE-dependent inflammation. PLoS One. 2010;5(8):e11944. doi: 10.1371/journal.pone.0011944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Hsu CL, Bryce PJ. Inducible IL-33 expression by mast cells is regulated by a calcium-dependent pathway. J Immunol. 2012;189(7):3421–3429. doi: 10.4049/jimmunol.1201224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Cherry WB, Yoon J, Bartemes KR, Iijima K, Kita H. A novel IL-1 family cytokine, IL-33, potently activates human eosinophils. J Allergy Clin Immunol. 2008;121(6):1484–1490. doi: 10.1016/j.jaci.2008.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Willebrand R, Voehringer D. IL-33-Induced Cytokine Secretion and Survival of Mouse Eosinophils Is Promoted by Autocrine GM-CSF. PLoS One. 2016;11(9):e0163751. doi: 10.1371/journal.pone.0163751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Stolarski B, Kurowska-Stolarska M, Kewin P, Xu D, Liew FY. IL-33 exacerbates eosinophil-mediated airway inflammation. J Immunol. 2010;185(6):3472–3480. doi: 10.4049/jimmunol.1000730. [DOI] [PubMed] [Google Scholar]
- 125.Tsuzuki H, Arinobu Y, Miyawaki K, et al. Functional interleukin-33 receptors are expressed in early progenitor stages of allergy-related granulocytes. Immunology. 2017;150(1):64–73. doi: 10.1111/imm.12667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Sun Z, Guo SS, Fassler R. Integrin-mediated mechanotransduction. J Cell Biol. 2016;215(4):445–456. doi: 10.1083/jcb.201609037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Takada Y, Ye X, Simon S. The integrins. Genome Biol. 2007;8(5):215. doi: 10.1186/gb-2007-8-5-215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Bochner BS, Luscinskas FW, Gimbrone MA, Jr, et al. Adhesion of human basophils, eosinophils, and neutrophils to interleukin 1-activated human vascular endothelial cells: contributions of endothelial cell adhesion molecules. J Exp Med. 1991;173(6):1553–1557. doi: 10.1084/jem.173.6.1553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Sperr WR, Agis H, Czerwenka K, et al. Differential expression of cell surface integrins on human mast cells and human basophils. Ann Hematol. 1992;65(1):10–16. doi: 10.1007/BF01715119. [DOI] [PubMed] [Google Scholar]
- 130.Elices MJ, Osborn L, Takada Y, et al. VCAM-1 on activated endothelium interacts with the leukocyte integrin VLA-4 at a site distinct from the VLA-4/fibronectin binding site. Cell. 1990;60(4):577–584. doi: 10.1016/0092-8674(90)90661-w. [DOI] [PubMed] [Google Scholar]
- 131.Columbo M, Bochner BS, Marone G. Human skin mast cells express functional beta 1 integrins that mediate adhesion to extracellular matrix proteins. J Immunol. 1995;154(11):6058–6064. [PubMed] [Google Scholar]
- 132.Sriramarao P, von Andrian UH, Butcher EC, Bourdon MA, Broide DH. L-selectin and very late antigen-4 integrin promote eosinophil rolling at physiological shear rates in vivo. J Immunol. 1994;153(9):4238–4246. [PubMed] [Google Scholar]
- 133.Yamamoto H, Nagata M. Regulatory mechanisms of eosinophil adhesion to and transmigration across endothelial cells by alpha4 and beta2 integrins. Int Arch Allergy Immunol. 1999;120(Suppl 1):24–26. doi: 10.1159/000053588. [DOI] [PubMed] [Google Scholar]
- 134.Gurish MF, Tao H, Abonia JP, et al. Intestinal mast cell progenitors require CD49dbeta7 (alpha4beta7 integrin) for tissue-specific homing. J Exp Med. 2001;194(9):1243–1252. doi: 10.1084/jem.194.9.1243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Abonia JP, Hallgren J, Jones T, et al. Alpha-4 integrins and VCAM-1, but not MAdCAM-1, are essential for recruitment of mast cell progenitors to the inflamed lung. Blood. 2006;108(5):1588–1594. doi: 10.1182/blood-2005-12-012781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Brandt EB, Zimmermann N, Muntel EE, et al. The alpha4bbeta7-integrin is dynamically expressed on murine eosinophils and involved in eosinophil trafficking to the intestine. Clin Exp Allergy. 2006;36(4):543–553. doi: 10.1111/j.1365-2222.2006.02456.x. [DOI] [PubMed] [Google Scholar]
- 137.Walsh GM, Symon FA, Lazarovils AL, Wardlaw AJ. Integrin alpha 4 beta 7 mediates human eosinophil interaction with MAdCAM-1, VCAM-1 and fibronectin. Immunology. 1996;89(1):112–119. doi: 10.1046/j.1365-2567.1996.d01-713.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Hennigan K, Conroy PJ, Walsh MT, et al. Eosinophil peroxidase activates cells by HER2 receptor engagement and beta1-integrin clustering with downstream MAPK cell signaling. Clin Immunol. 2016;171:1–11. doi: 10.1016/j.clim.2016.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Ben-Zimra M, Bachelet I, Seaf M, Gleich GJ, Levi-Schaffer F. Eosinophil major basic protein activates human cord blood mast cells primed with fibroblast membranes by integrin-beta1. Allergy. 2013;68(10):1259–1268. doi: 10.1111/all.12232. [DOI] [PubMed] [Google Scholar]
- 140.Gyles SL, Xue L, Townsend ER, Wettey F, Pettipher R. A dominant role for chemoattractant receptor-homologous molecule expressed on T helper type 2 (Th2) cells (CRTH2) in mediating chemotaxis of CRTH2+ CD4+ Th2 lymphocytes in response to mast cell supernatants. Immunology. 2006;119(3):362–368. doi: 10.1111/j.1365-2567.2006.02440.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Hirai H, Tanaka K, Yoshie O, et al. Prostaglandin D2 selectively induces chemotaxis in T helper type 2 cells, eosinophils, and basophils via seven-transmembrane receptor CRTH2. J Exp Med. 2001;193(2):255–261. doi: 10.1084/jem.193.2.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Gervais FG, Cruz RP, Chateauneuf A, et al. Selective modulation of chemokinesis, degranulation, and apoptosis in eosinophils through the PGD2 receptors CRTH2 and DP. J Allergy Clin Immunol. 2001;108(6):982–988. doi: 10.1067/mai.2001.119919. [DOI] [PubMed] [Google Scholar]
- 143.Luna-Gomes T, Magalhaes KG, Mesquita-Santos FP, et al. Eosinophils as a novel cell source of prostaglandin D2: autocrine role in allergic inflammation. J Immunol. 2011;187(12):6518–6526. doi: 10.4049/jimmunol.1101806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Moon TC, Campos-Alberto E, Yoshimura T, et al. Expression of DP2 (CRTh2), a prostaglandin D(2) receptor, in human mast cells. PLoS One. 2014;9(9):e108595. doi: 10.1371/journal.pone.0108595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Diamant Z, Sidharta PN, Singh D, et al. Setipiprant, a selective CRTH2 antagonist, reduces allergen-induced airway responses in allergic asthmatics. Clin Exp Allergy. 2014;44(8):1044–1052. doi: 10.1111/cea.12357. [DOI] [PubMed] [Google Scholar]
- 146.Hall IP, Fowler AV, Gupta A, et al. Efficacy of BI 671800, an oral CRTH2 antagonist, in poorly controlled asthma as sole controller and in the presence of inhaled corticosteroid treatment. Pulm Pharmacol Ther. 2015;32:37–44. doi: 10.1016/j.pupt.2015.03.003. [DOI] [PubMed] [Google Scholar]
- 147.Erpenbeck VJ, Popov TA, Miller D, et al. The oral CRTh2 antagonist QAW039 (fevipiprant): A phase II study in uncontrolled allergic asthma. Pulm Pharmacol Ther. 2016;39:54–63. doi: 10.1016/j.pupt.2016.06.005. [DOI] [PubMed] [Google Scholar]
- 148.Bateman ED, Guerreros AG, Brockhaus F, et al. Fevipiprant, an oral prostaglandin DP2 receptor (CRTh2) antagonist, in allergic asthma uncontrolled on low-dose inhaled corticosteroids. Eur Respir J. 2017;50(2) doi: 10.1183/13993003.00670-2017. [DOI] [PubMed] [Google Scholar]
- 149.Elsner J, Oppermann M, Czech W, et al. C3a activates reactive oxygen radical species production and intracellular calcium transients in human eosinophils. Eur J Immunol. 1994;24(3):518–522. doi: 10.1002/eji.1830240304. [DOI] [PubMed] [Google Scholar]
- 150.DiScipio RG, Daffern PJ, Jagels MA, Broide DH, Sriramarao P. A comparison of C3a and C5a-mediated stable adhesion of rolling eosinophils in postcapillary venules and transendothelial migration in vitro and in vivo. J Immunol. 1999;162(2):1127–1136. [PubMed] [Google Scholar]
- 151.Fujiu T, Kato M, Kimura H, et al. Cellular adhesion is required for effector functions of human eosinophils via G-protein coupled receptors. Ann Allergy Asthma Immunol. 2002;89(1):90–98. doi: 10.1016/S1081-1206(10)61917-5. [DOI] [PubMed] [Google Scholar]
- 152.Nilsson G, Johnell M, Hammer CH, et al. C3a and C5a are chemotaxins for human mast cells and act through distinct receptors via a pertussis toxin-sensitive signal transduction pathway. J Immunol. 1996;157(4):1693–1698. [PubMed] [Google Scholar]
- 153.Venkatesha RT, Berla Thangam E, Zaidi AK, Ali H. Distinct regulation of C3a-induced MCP-1/CCL2 and RANTES/CCL5 production in human mast cells by extracellular signal regulated kinase and PI3 kinase. Mol Immunol. 2005;42(5):581–587. doi: 10.1016/j.molimm.2004.09.009. [DOI] [PubMed] [Google Scholar]
- 154.Oskeritzian CA, Hait NC, Wedman P, et al. The sphingosine-1-phosphate/sphingosine-1-phosphate receptor 2 axis regulates early airway T-cell infiltration in murine mast cell-dependent acute allergic responses. J Allergy Clin Immunol. 2015;135(4):1008–1018. e1001. doi: 10.1016/j.jaci.2014.10.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Wedman PA, Aladhami A, Chumanevich AP, Fuseler JW, Oskeritzian CA. Mast cells and sphingosine-1-phosphate underlie prelesional remodeling in a mouse model of eczema. Allergy. 2017 doi: 10.1111/all.13310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Pundir P, MacDonald CA, Kulka M. The Novel Receptor C5aR2 Is Required for C5a-Mediated Human Mast Cell Adhesion, Migration, and Proinflammatory Mediator Production. J Immunol. 2015;195(6):2774–2787. doi: 10.4049/jimmunol.1401348. [DOI] [PubMed] [Google Scholar]
- 157.Karsten CM, Wiese AV, Mey F, et al. Monitoring C5aR2 Expression Using a Floxed tdTomato-C5aR2 Knock-In Mouse. J Immunol. 2017;199(9):3234–3248. doi: 10.4049/jimmunol.1700710. [DOI] [PubMed] [Google Scholar]
- 158.Spiegel S, Milstien S. The outs and the ins of sphingosine-1-phosphate in immunity. Nat Rev Immunol. 2011;11(6):403–415. doi: 10.1038/nri2974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Jolly PS, Bektas M, Olivera A, et al. Transactivation of sphingosine-1-phosphate receptors by FcepsilonRI triggering is required for normal mast cell degranulation and chemotaxis. J Exp Med. 2004;199(7):959–970. doi: 10.1084/jem.20030680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Oskeritzian CA, Price MM, Hait NC, et al. Essential roles of sphingosine-1-phosphate receptor 2 in human mast cell activation, anaphylaxis, and pulmonary edema. J Exp Med. 2010;207(3):465–474. doi: 10.1084/jem.20091513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Chumanevich A, Wedman P, Oskeritzian CA. Sphingosine-1-Phosphate/Sphingosine-1-Phosphate Receptor 2 Axis Can Promote Mouse and Human Primary Mast Cell Angiogenic Potential through Upregulation of Vascular Endothelial Growth Factor-A and Matrix Metalloproteinase-2. Mediators of Inflammation. 2016;2016:8. doi: 10.1155/2016/1503206. Article ID 1503206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Sugita K, Kabashima K, Sakabe J, Yoshiki R, Tanizaki H, Tokura Y. FTY720 regulates bone marrow egress of eosinophils and modulates late-phase skin reaction in mice. Am J Pathol. 2010;177(4):1881–1887. doi: 10.2353/ajpath.2010.100119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Merves J, Chandramouleeswaran PM, Benitez AJ, et al. Altered esophageal histamine receptor expression in Eosinophilic Esophagitis (EoE): implications on disease pathogenesis. PLoS One. 2015;10(2):e0114831. doi: 10.1371/journal.pone.0114831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Togias A. H1-receptors: localization and role in airway physiology and in immune functions. J Allergy Clin Immunol. 2003;112(4 Suppl):S60–68. doi: 10.1016/s0091-6749(03)01878-5. [DOI] [PubMed] [Google Scholar]
- 165.O’Mahony L, Akdis M, Akdis CA. Regulation of the immune response and inflammation by histamine and histamine receptors. J Allergy Clin Immunol. 2011;128(6):1153–1162. doi: 10.1016/j.jaci.2011.06.051. [DOI] [PubMed] [Google Scholar]
- 166.Ling P, Ngo K, Nguyen S, et al. Histamine H4 receptor mediates eosinophil chemotaxis with cell shape change and adhesion molecule upregulation. Br J Pharmacol. 2004;142(1):161–171. doi: 10.1038/sj.bjp.0705729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.O’Reilly M, Alpert R, Jenkinson S, et al. Identification of a histamine H4 receptor on human eosinophils--role in eosinophil chemotaxis. J Recept Signal Transduct Res. 2002;22(1–4):431–448. doi: 10.1081/rrs-120014612. [DOI] [PubMed] [Google Scholar]
- 168.Jemima EA, Prema A, Thangam EB. Functional characterization of histamine H4 receptor on human mast cells. Mol Immunol. 2014;62(1):19–28. doi: 10.1016/j.molimm.2014.05.007. [DOI] [PubMed] [Google Scholar]
- 169.Kollmeier A, Francke K, Chen B, et al. The histamine H(4) receptor antagonist, JNJ 39758979, is effective in reducing histamine-induced pruritus in a randomized clinical study in healthy subjects. J Pharmacol Exp Ther. 2014;350(1):181–187. doi: 10.1124/jpet.114.215749. [DOI] [PubMed] [Google Scholar]
- 170.Murata Y, Song M, Kikuchi H, et al. Phase 2a, randomized, double-blind, placebo-controlled, multicenter, parallel-group study of a H4 R-antagonist (JNJ-39758979) in Japanese adults with moderate atopic dermatitis. J Dermatol. 2015;42(2):129–139. doi: 10.1111/1346-8138.12726. [DOI] [PubMed] [Google Scholar]
- 171.Lippert U, Artuc M, Grutzkau A, et al. Human skin mast cells express H2 and H4, but not H3 receptors. J Invest Dermatol. 2004;123(1):116–123. doi: 10.1111/j.0022-202X.2004.22721.x. [DOI] [PubMed] [Google Scholar]
- 172.Kishimoto S, Oka S, Gokoh M, Sugiura T. Chemotaxis of human peripheral blood eosinophils to 2-arachidonoylglycerol: comparison with other eosinophil chemoattractants. Int Arch Allergy Immunol. 2006;140(Suppl 1):3–7. doi: 10.1159/000092704. [DOI] [PubMed] [Google Scholar]
- 173.Frei RB, Luschnig P, Parzmair GP, et al. Cannabinoid receptor 2 augments eosinophil responsiveness and aggravates allergen-induced pulmonary inflammation in mice. Allergy. 2016;71(7):944–956. doi: 10.1111/all.12858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Bezemer GF, Sagar S, van Bergenhenegouwen J, et al. Dual role of Toll-like receptors in asthma and chronic obstructive pulmonary disease. Pharmacol Rev. 2012;64(2):337–358. doi: 10.1124/pr.111.004622. [DOI] [PubMed] [Google Scholar]
- 175.Tworek D, Heroux D, O’Byrne SN, Mitchell P, O’Byrne PM, Denburg JA. Toll-like receptor-induced expression of epithelial cytokine receptors on haemopoietic progenitors is altered in allergic asthma. Clin Exp Allergy. 2017;47(7):900–908. doi: 10.1111/cea.12913. [DOI] [PubMed] [Google Scholar]
- 176.Kvarnhammar AM, Cardell LO. Pattern-recognition receptors in human eosinophils. Immunology. 2012;136(1):11–20. doi: 10.1111/j.1365-2567.2012.03556.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Sandig H, Bulfone-Paus S. TLR signaling in mast cells: common and unique features. Front Immunol. 2012;3:185. doi: 10.3389/fimmu.2012.00185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Ferreira DS, Annoni R, Silva LF, et al. Toll-like receptors 2, 3 and 4 and thymic stromal lymphopoietin expression in fatal asthma. Clin Exp Allergy. 2012;42(10):1459–1471. doi: 10.1111/j.1365-2222.2012.04047.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Elishmereni M, Fyhrquist N, Singh Gangwar R, Lehtimaki S, Alenius H, Levi-Schaffer F. Complex 2B4 regulation of mast cells and eosinophils in murine allergic inflammation. J Invest Dermatol. 2014;134(12):2928–2937. doi: 10.1038/jid.2014.280. [DOI] [PubMed] [Google Scholar]
- 180.Rocha-de-Souza CM, Berent-Maoz B, Mankuta D, Moses AE, Levi-Schaffer F. Human mast cell activation by Staphylococcus aureus: interleukin-8 and tumor necrosis factor alpha release and the role of Toll-like receptor 2 and CD48 molecules. Infect Immun. 2008;76(10):4489–4497. doi: 10.1128/IAI.00270-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Gangwar RS, Minai-Fleminger Y, Seaf M, et al. CD48 on blood leukocytes and in serum of asthma patients varies with severity. Allergy. 2017;72(6):888–895. doi: 10.1111/all.13082. [DOI] [PubMed] [Google Scholar]
- 182.Minai-Fleminger Y, Gangwar RS, Migalovich-Sheikhet H, et al. The CD48 receptor mediates Staphylococcus aureus human and murine eosinophil activation. Clin Exp Allergy. 2014;44(11):1335–1346. doi: 10.1111/cea.12422. [DOI] [PubMed] [Google Scholar]
- 183.El-Shazly AE, Henket M, Lefebvre PP, Louis R. 2B4 (CD244) is involved in eosinophil adhesion and chemotaxis, and its surface expression is increased in allergic rhinitis after challenge. Int J Immunopathol Pharmacol. 2011;24(4):949–960. doi: 10.1177/039463201102400413. [DOI] [PubMed] [Google Scholar]
- 184.Gangwar RS, Levi-Schaffer F. sCD48 is anti-inflammatory in Staphylococcus aureus Enterotoxin B-induced eosinophilic inflammation. Allergy. 2016;71(6):829–839. doi: 10.1111/all.12851. [DOI] [PubMed] [Google Scholar]
- 185.Gangwar RS, Landolina N, Arpinati L, Levi-Schaffer F. Mast cell and eosinophil surface receptors as targets for anti-allergic therapy. Pharmacol Ther. 2017;170:37–63. doi: 10.1016/j.pharmthera.2016.10.010. [DOI] [PubMed] [Google Scholar]
- 186.Derycke L, Perez-Novo C, Van Crombruggen K, Corriveau MN, Bachert C. Staphylococcus aureus and Chronic Airway Disease. World Allergy Organ J. 2010;3(8):223–228. doi: 10.1097/WOX.0b013e3181ecd8ae. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Ravin KA, Loy M. The Eosinophil in Infection. Clin Rev Allergy Immunol. 2016;50(2):214–227. doi: 10.1007/s12016-015-8525-4. [DOI] [PubMed] [Google Scholar]
- 188.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Moon TC, Befus AD, Kulka M. Mast cell mediators: their differential release and the secretory pathways involved. Front Immunol. 2014;5:569. doi: 10.3389/fimmu.2014.00569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Galdiero MR, Varricchi G, Seaf M, Marone G, Levi-Schaffer F, Marone G. Bidirectional Mast Cell-Eosinophil Interactions in Inflammatory Disorders and Cancer. Front Med (Lausanne) 2017;4:103. doi: 10.3389/fmed.2017.00103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Olszewski MB, Groot AJ, Dastych J, Knol EF. TNF trafficking to human mast cell granules: mature chain-dependent endocytosis. J Immunol. 2007;178(9):5701–5709. doi: 10.4049/jimmunol.178.9.5701. [DOI] [PubMed] [Google Scholar]
- 192.Finotto S, Ohno I, Marshall JS, et al. TNF-alpha production by eosinophils in upper airways inflammation (nasal polyposis) J Immunol. 1994;153(5):2278–2289. [PubMed] [Google Scholar]
- 193.Sugarman BJ, Aggarwal BB, Hass PE, Figari IS, Palladino MA, Jr, Shepard HM. Recombinant human tumor necrosis factor-alpha: effects on proliferation of normal and transformed cells in vitro. Science. 1985;230(4728):943–945. doi: 10.1126/science.3933111. [DOI] [PubMed] [Google Scholar]
- 194.Lorentz A, Wilke M, Sellge G, et al. IL-4-induced priming of human intestinal mast cells for enhanced survival and Th2 cytokine generation is reversible and associated with increased activity of ERK1/2 and c-Fos. J Immunol. 2005;174(11):6751–6756. doi: 10.4049/jimmunol.174.11.6751. [DOI] [PubMed] [Google Scholar]
- 195.Suarez-Zamorano N, Fabbiano S, Chevalier C, et al. Microbiota depletion promotes browning of white adipose tissue and reduces obesity. Nat Med. 2015;21(12):1497–1501. doi: 10.1038/nm.3994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Withers SB, Forman R, Meza-Perez S, et al. Eosinophils are key regulators of perivascular adipose tissue and vascular functionality. Sci Rep. 2017;7:44571. doi: 10.1038/srep44571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Brestoff JR, Kim BS, Saenz SA, et al. Group 2 innate lymphoid cells promote beiging of white adipose tissue and limit obesity. Nature. 2015;519(7542):242–246. doi: 10.1038/nature14115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.McBrien CN, Menzies-Gow A. The Biology of Eosinophils and Their Role in Asthma. Front Med (Lausanne) 2017;4:93. doi: 10.3389/fmed.2017.00093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Lewis RA, Soter NA, Diamond PT, Austen KF, Oates JA, Roberts LJ., 2nd Prostaglandin D2 generation after activation of rat and human mast cells with anti-IgE. J Immunol. 1982;129(4):1627–1631. [PubMed] [Google Scholar]
- 200.Pettipher R. The roles of the prostaglandin D(2) receptors DP(1) and CRTH2 in promoting allergic responses. Br J Pharmacol. 2008;153(Suppl 1):S191–199. doi: 10.1038/sj.bjp.0707488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Cho C, Nguyen A, Bryant KJ, O’Neill SG, McNeil HP. Prostaglandin D2 metabolites as a biomarker of in vivo mast cell activation in systemic mastocytosis and rheumatoid arthritis. Immun Inflamm Dis. 2016;4(1):64–69. doi: 10.1002/iid3.94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Fajt ML, Gelhaus SL, Freeman B, et al. Prostaglandin D(2) pathway upregulation: relation to asthma severity, control, and TH2 inflammation. J Allergy Clin Immunol. 2013;131(6):1504–1512. doi: 10.1016/j.jaci.2013.01.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Ricciotti E, FitzGerald GA. Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol. 2011;31(5):986–1000. doi: 10.1161/ATVBAHA.110.207449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Peinhaupt M, Sturm EM, Heinemann A. Prostaglandins and Their Receptors in Eosinophil Function and As Therapeutic Targets. Front Med (Lausanne) 2017;4:104. doi: 10.3389/fmed.2017.00104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Fulkerson PC, Rothenberg ME. Targeting eosinophils in allergy, inflammation and beyond. Nat Rev Drug Discov. 2013;12(2):117–129. doi: 10.1038/nrd3838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Gonem S, Berair R, Singapuri A, et al. Fevipiprant, a prostaglandin D2 receptor 2 antagonist, in patients with persistent eosinophilic asthma: a single-centre, randomised, double-blind, parallel-group, placebo-controlled trial. Lancet Respir Med. 2016;4(9):699–707. doi: 10.1016/S2213-2600(16)30179-5. [DOI] [PubMed] [Google Scholar]
- 207.Dennis EA, Norris PC. Eicosanoid storm in infection and inflammation. Nat Rev Immunol. 2015;15(8):511–523. doi: 10.1038/nri3859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Kanaoka Y, Boyce JA. Cysteinyl leukotrienes and their receptors: cellular distribution and function in immune and inflammatory responses. J Immunol. 2004;173(3):1503–1510. doi: 10.4049/jimmunol.173.3.1503. [DOI] [PubMed] [Google Scholar]
- 209.Back M, Dahlen SE, Drazen JM, et al. International Union of Basic and Clinical Pharmacology. LXXXIV: leukotriene receptor nomenclature, distribution, and pathophysiological functions. Pharmacol Rev. 2011;63(3):539–584. doi: 10.1124/pr.110.004184. [DOI] [PubMed] [Google Scholar]
- 210.Paruchuri S, Tashimo H, Feng C, et al. Leukotriene E4-induced pulmonary inflammation is mediated by the P2Y12 receptor. J Exp Med. 2009;206(11):2543–2555. doi: 10.1084/jem.20091240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Mellor EA, Maekawa A, Austen KF, Boyce JA. Cysteinyl leukotriene receptor 1 is also a pyrimidinergic receptor and is expressed by human mast cells. Proc Natl Acad Sci U S A. 2001;98(14):7964–7969. doi: 10.1073/pnas.141221498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Mellor EA, Austen KF, Boyce JA. Cysteinyl leukotrienes and uridine diphosphate induce cytokine generation by human mast cells through an interleukin 4-regulated pathway that is inhibited by leukotriene receptor antagonists. J Exp Med. 2002;195(5):583–592. doi: 10.1084/jem.20020044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Jiang Y, Kanaoka Y, Feng C, Nocka K, Rao S, Boyce JA. Cutting edge: Interleukin 4-dependent mast cell proliferation requires autocrine/intracrine cysteinyl leukotriene-induced signaling. J Immunol. 2006;177(5):2755–2759. doi: 10.4049/jimmunol.177.5.2755. [DOI] [PubMed] [Google Scholar]
- 214.Mitsunobu F, Mifune T, Hosaki Y, et al. Enhanced peripheral leukocyte leukotriene production and bronchial hyperresponsiveness in asthmatics. Eur Respir J. 2000;16(3):504–508. doi: 10.1034/j.1399-3003.2000.016003504.x. [DOI] [PubMed] [Google Scholar]
- 215.Lundeen KA, Sun B, Karlsson L, Fourie AM. Leukotriene B4 receptors BLT1 and BLT2: expression and function in human and murine mast cells. J Immunol. 2006;177(5):3439–3447. doi: 10.4049/jimmunol.177.5.3439. [DOI] [PubMed] [Google Scholar]
- 216.Rao NL, Riley JP, Banie H, et al. Leukotriene A(4) hydrolase inhibition attenuates allergic airway inflammation and hyperresponsiveness. Am J Respir Crit Care Med. 2010;181(9):899–907. doi: 10.1164/rccm.200807-1158OC. [DOI] [PubMed] [Google Scholar]
- 217.Fialkow L, Wang Y, Downey GP. Reactive oxygen and nitrogen species as signaling molecules regulating neutrophil function. Free Radic Biol Med. 2007;42(2):153–164. doi: 10.1016/j.freeradbiomed.2006.09.030. [DOI] [PubMed] [Google Scholar]
- 218.Reis AC, Alessandri AL, Athayde RM, et al. Induction of eosinophil apoptosis by hydrogen peroxide promotes the resolution of allergic inflammation. Cell Death Dis. 2015;6:e1632. doi: 10.1038/cddis.2014.580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Wartha F, Henriques-Normark B. ETosis: a novel cell death pathway. Sci Signal. 2008;1(21):pe25. doi: 10.1126/stke.121pe25. [DOI] [PubMed] [Google Scholar]
- 220.Swindle EJ, Brown JM, Radinger M, DeLeo FR, Metcalfe DD. Interferon-gamma enhances both the anti-bacterial and the pro-inflammatory response of human mast cells to Staphylococcus aureus. Immunology. 2015;146(3):470–485. doi: 10.1111/imm.12524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Swindle EJ, Coleman JW, DeLeo FR, Metcalfe DD. FcepsilonRI- and Fcgamma receptor-mediated production of reactive oxygen species by mast cells is lipoxygenase- and cyclooxygenase-dependent and NADPH oxidase-independent. J Immunol. 2007;179(10):7059–7071. doi: 10.4049/jimmunol.179.10.7059. [DOI] [PubMed] [Google Scholar]
- 222.Lee AJ, Ro M, Kim JH. Leukotriene B4 Receptor 2 Is Critical for the Synthesis of Vascular Endothelial Growth Factor in Allergen-Stimulated Mast Cells. J Immunol. 2016;197(6):2069–2078. doi: 10.4049/jimmunol.1502565. [DOI] [PubMed] [Google Scholar]
- 223.Tiligada E, Ishii M, Riccardi C, et al. The expanding role of immunopharmacology: IUPHAR Review 16. Br J Pharmacol. 2015;172(17):4217–4227. doi: 10.1111/bph.13219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Maintz L, Novak N. Modifications of the innate immune system in atopic dermatitis. J Innate Immun. 2011;3(2):131–141. doi: 10.1159/000323963. [DOI] [PubMed] [Google Scholar]
- 225.Weidinger S, Novak N. Atopic dermatitis. Lancet. 2016;387(10023):1109–1122. doi: 10.1016/S0140-6736(15)00149-X. [DOI] [PubMed] [Google Scholar]
- 226.Mu Z, Zhao Y, Liu X, Chang C, Zhang J. Molecular biology of atopic dermatitis. Clin Rev Allergy Immunol. 2014;47(2):193–218. doi: 10.1007/s12016-014-8415-1. [DOI] [PubMed] [Google Scholar]
- 227.Jinnestal CL, Belfrage E, Back O, Schmidtchen A, Sonesson A. Skin barrier impairment correlates with cutaneous Staphylococcus aureus colonization and sensitization to skin-associated microbial antigens in adult patients with atopic dermatitis. Int J Dermatol. 2014;53(1):27–33. doi: 10.1111/ijd.12198. [DOI] [PubMed] [Google Scholar]
- 228.Sonesson A, Przybyszewska K, Eriksson S, et al. Identification of bacterial biofilm and the Staphylococcus aureus derived protease, staphopain, on the skin surface of patients with atopic dermatitis. Sci Rep. 2017;7(1):8689. doi: 10.1038/s41598-017-08046-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Nakamura Y, Oscherwitz J, Cease KB, et al. Staphylococcus delta-toxin induces allergic skin disease by activating mast cells. Nature. 2013;503(7476):397–401. doi: 10.1038/nature12655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Kasutani K, Fujii E, Ohyama S, et al. Anti-IL-31 receptor antibody is shown to be a potential therapeutic option for treating itch and dermatitis in mice. Br J Pharmacol. 2014;171(22):5049–5058. doi: 10.1111/bph.12823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Casale TB. Biologics and biomarkers for asthma, urticaria, and nasal polyposis. J Allergy Clin Immunol. 2017;139(5):1411–1421. doi: 10.1016/j.jaci.2017.03.006. [DOI] [PubMed] [Google Scholar]
- 232.Bradding P, Arthur G. Mast cells in asthma--state of the art. Clin Exp Allergy. 2016;46(2):194–263. doi: 10.1111/cea.12675. [DOI] [PubMed] [Google Scholar]
- 233.Israel E, Reddel HK. Severe and Difficult-to-Treat Asthma in Adults. N Engl J Med. 2017;377(10):965–976. doi: 10.1056/NEJMra1608969. [DOI] [PubMed] [Google Scholar]
- 234.Berthon BS, Gibson PG, Wood LG, MacDonald-Wicks LK, Baines KJ. A sputum gene expression signature predicts oral corticosteroid response in asthma. Eur Respir J. 2017;49(6) doi: 10.1183/13993003.00180-2017. [DOI] [PubMed] [Google Scholar]
- 235.Eifan AO, Durham SR. Pathogenesis of rhinitis. Clin Exp Allergy. 2016;46(9):1139–1151. doi: 10.1111/cea.12780. [DOI] [PubMed] [Google Scholar]
- 236.Knight DA, Holgate ST. The airway epithelium: structural and functional properties in health and disease. Respirology. 2003;8(4):432–446. doi: 10.1046/j.1440-1843.2003.00493.x. [DOI] [PubMed] [Google Scholar]
- 237.Boita M, Bucca C, Riva G, Heffler E, Rolla G. Release of Type 2 Cytokines by Epithelial Cells of Nasal Polyps. J Immunol Res. 2016;2016:2643297. doi: 10.1155/2016/2643297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Schleimer RP. Immunopathogenesis of Chronic Rhinosinusitis and Nasal Polyposis. Annu Rev Pathol. 2017;12:331–357. doi: 10.1146/annurev-pathol-052016-100401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Miyahara S, Miyahara N, Takeda K, Joetham A, Gelfand EW. Physiologic assessment of allergic rhinitis in mice: role of the high-affinity IgE receptor (FcepsilonRI) J Allergy Clin Immunol. 2005;116(5):1020–1027. doi: 10.1016/j.jaci.2005.08.020. [DOI] [PubMed] [Google Scholar]
- 240.Bachert C, Gevaert E. Advances in rhinitis and rhinosinusitis in 2015. J Allergy Clin Immunol. 2016;138(5):1277–1283. doi: 10.1016/j.jaci.2016.09.008. [DOI] [PubMed] [Google Scholar]
- 241.Bascom R, Pipkorn U, Lichtenstein LM, Naclerio RM. The influx of inflammatory cells into nasal washings during the late response to antigen challenge. Effect of systemic steroid pretreatment. Am Rev Respir Dis. 1988;138(2):406–412. doi: 10.1164/ajrccm/138.2.406. [DOI] [PubMed] [Google Scholar]
- 242.Bascom R, Pipkorn U, Proud D, et al. Major basic protein and eosinophil-derived neurotoxin concentrations in nasal-lavage fluid after antigen challenge: effect of systemic corticosteroids and relationship to eosinophil influx. J Allergy Clin Immunol. 1989;84(3):338–346. doi: 10.1016/0091-6749(89)90418-1. [DOI] [PubMed] [Google Scholar]
- 243.Kountakis SE, Arango P, Bradley D, Wade ZK, Borish L. Molecular and cellular staging for the severity of chronic rhinosinusitis. Laryngoscope. 2004;114(11):1895–1905. doi: 10.1097/01.mlg.0000147917.43615.c0. [DOI] [PubMed] [Google Scholar]
- 244.Ellis AK, Keith PK. Nonallergic rhinitis with eosinophilia syndrome and related disorders. Clin Allergy Immunol. 2007;19:87–100. [PubMed] [Google Scholar]
- 245.Cianferoni A, Spergel JM. Food allergy: review, classification and diagnosis. Allergol Int. 2009;58(4):457–466. doi: 10.2332/allergolint.09-RAI-0138. [DOI] [PubMed] [Google Scholar]
- 246.Tordesillas L, Berin MC, Sampson HA. Immunology of Food Allergy. Immunity. 2017;47(1):32–50. doi: 10.1016/j.immuni.2017.07.004. [DOI] [PubMed] [Google Scholar]
- 247.Vadas P, Perelman B, Liss G. Platelet-activating factor, histamine, and tryptase levels in human anaphylaxis. J Allergy Clin Immunol. 2013;131(1):144–149. doi: 10.1016/j.jaci.2012.08.016. [DOI] [PubMed] [Google Scholar]
- 248.Hill DA, Spergel JM. The Immunologic Mechanisms of Eosinophilic Esophagitis. Curr Allergy Asthma Rep. 2016;16(2):9. doi: 10.1007/s11882-015-0592-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Blanchard C, Wang N, Stringer KF, et al. Eotaxin-3 and a uniquely conserved gene-expression profile in eosinophilic esophagitis. J Clin Invest. 2006;116(2):536–547. doi: 10.1172/JCI26679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Metcalfe DD, Pawankar R, Ackerman SJ, et al. Biomarkers of the involvement of mast cells, basophils and eosinophils in asthma and allergic diseases. World Allergy Organ J. 2016;9:7. doi: 10.1186/s40413-016-0094-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Johnsson M, Bove M, Bergquist H, 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. [DOI] [PubMed] [Google Scholar]
- 252.Abonia JP, Blanchard C, Butz BB, et al. Involvement of mast cells in eosinophilic esophagitis. J Allergy Clin Immunol. 2010;126(1):140–149. doi: 10.1016/j.jaci.2010.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Kanani A, Schellenberg R, Warrington R. Urticaria and angioedema. Allergy Asthma Clin Immunol. 2011;7(Suppl 1):S9. doi: 10.1186/1710-1492-7-S1-S9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Kaplan AP, Greaves M. Pathogenesis of chronic urticaria. Clin Exp Allergy. 2009;39(6):777–787. doi: 10.1111/j.1365-2222.2009.03256.x. [DOI] [PubMed] [Google Scholar]
- 255.Nettis E, Dambra P, Loria MP, et al. Mast-cell phenotype in urticaria. Allergy. 2001;56(9):915. doi: 10.1034/j.1398-9995.2001.00296.x. [DOI] [PubMed] [Google Scholar]
- 256.Smith CH, Kepley C, Schwartz LB, Lee TH. Mast cell number and phenotype in chronic idiopathic urticaria. J Allergy Clin Immunol. 1995;96(3):360–364. doi: 10.1016/s0091-6749(95)70055-2. [DOI] [PubMed] [Google Scholar]
- 257.Bernstein JA, Lang DM, Khan DA, et al. The diagnosis and management of acute and chronic urticaria: 2014 update. J Allergy Clin Immunol. 2014;133(5):1270–1277. doi: 10.1016/j.jaci.2014.02.036. [DOI] [PubMed] [Google Scholar]
- 258.Saini S, Rosen KE, Hsieh HJ, et al. A randomized, placebo-controlled, dose-ranging study of single-dose omalizumab in patients with H1-antihistamine-refractory chronic idiopathic urticaria. J Allergy Clin Immunol. 2011;128(3):567–573. e561. doi: 10.1016/j.jaci.2011.06.010. [DOI] [PubMed] [Google Scholar]
- 259.Sabroe RA, Poon E, Orchard GE, et al. Cutaneous inflammatory cell infiltrate in chronic idiopathic urticaria: comparison of patients with and without anti-FcepsilonRI or anti-IgE autoantibodies. J Allergy Clin Immunol. 1999;103(3 Pt 1):484–493. doi: 10.1016/s0091-6749(99)70475-6. [DOI] [PubMed] [Google Scholar]
- 260.Marques RZ, Criado RF, Machado CDF, Tamanini JM, Mello CV, Speyer C. Correlation between the histopathology of chronic urticaria and its clinical picture. An Bras Dermatol. 2016;91(6):760–763. doi: 10.1590/abd1806-4841.20165066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Sundstrom JB, Ellis JE, Hair GA, et al. Human tissue mast cells are an inducible reservoir of persistent HIV infection. Blood. 2007;109(12):5293–5300. doi: 10.1182/blood-2006-11-058438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Al-Afif A, Alyazidi R, Oldford SA, et al. Respiratory syncytial virus infection of primary human mast cells induces the selective production of type I interferons, CXCL10, and CCL4. J Allergy Clin Immunol. 2015;136(5):1346–1354. e1341. doi: 10.1016/j.jaci.2015.01.042. [DOI] [PubMed] [Google Scholar]
- 263.Troupin A, Shirley D, Londono-Renteria B, et al. A Role for Human Skin Mast Cells in Dengue Virus Infection and Systemic Spread. J Immunol. 2016;197(11):4382–4391. doi: 10.4049/jimmunol.1600846. [DOI] [PubMed] [Google Scholar]
- 264.Drake MG, Bivins-Smith ER, Proskocil BJ, et al. Human and Mouse Eosinophils Have Antiviral Activity against Parainfluenza Virus. Am J Respir Cell Mol Biol. 2016;55(3):387–394. doi: 10.1165/rcmb.2015-0405OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Saluja R, Metz M, Maurer M. Role and relevance of mast cells in fungal infections. Front Immunol. 2012;3:146. doi: 10.3389/fimmu.2012.00146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Lopes JP, Stylianou M, Nilsson G, Urban CF. Opportunistic pathogen Candida albicans elicits a temporal response in primary human mast cells. Sci Rep. 2015;5:12287. doi: 10.1038/srep12287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Yoon J, Ponikau JU, Lawrence CB, Kita H. Innate antifungal immunity of human eosinophils mediated by a beta 2 integrin, CD11b. J Immunol. 2008;181(4):2907–2915. doi: 10.4049/jimmunol.181.4.2907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Kita H. Eosinophils: multifunctional and distinctive properties. Int Arch Allergy Immunol. 2013;161(Suppl 2):3–9. doi: 10.1159/000350662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Inoue Y, Matsuwaki Y, Shin SH, Ponikau JU, Kita H. Nonpathogenic, environmental fungi induce activation and degranulation of human eosinophils. J Immunol. 2005;175(8):5439–5447. doi: 10.4049/jimmunol.175.8.5439. [DOI] [PubMed] [Google Scholar]
- 270.Galli SJ, Tsai M. Mast cells in allergy and infection: versatile effector and regulatory cells in innate and adaptive immunity. Eur J Immunol. 2010;40(7):1843–1851. doi: 10.1002/eji.201040559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Lu F, Huang S. The Roles of Mast Cells in Parasitic Protozoan Infections. Front Immunol. 2017;8:363. doi: 10.3389/fimmu.2017.00363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Klion AD, Nutman TB. The role of eosinophils in host defense against helminth parasites. J Allergy Clin Immunol. 2004;113(1):30–37. doi: 10.1016/j.jaci.2003.10.050. [DOI] [PubMed] [Google Scholar]
- 273.Huang L, Appleton JA. Eosinophils in Helminth Infection: Defenders and Dupes. Trends Parasitol. 2016;32(10):798–807. doi: 10.1016/j.pt.2016.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Cadman ET, Thysse KA, Bearder S, et al. Eosinophils are important for protection, immunoregulation and pathology during infection with nematode microfilariae. PLoS Pathog. 2014;10(3):e1003988. doi: 10.1371/journal.ppat.1003988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Hogan SP, Rosenberg HF, Moqbel R, et al. Eosinophils: biological properties and role in health and disease. Clin Exp Allergy. 2008;38(5):709–750. doi: 10.1111/j.1365-2222.2008.02958.x. [DOI] [PubMed] [Google Scholar]
- 276.Abraham SN, St John AL. Mast cell-orchestrated immunity to pathogens. Nat Rev Immunol. 2010;10(6):440–452. doi: 10.1038/nri2782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Johnzon CF, Ronnberg E, Pejler G. The Role of Mast Cells in Bacterial Infection. Am J Pathol. 2016;186(1):4–14. doi: 10.1016/j.ajpath.2015.06.024. [DOI] [PubMed] [Google Scholar]
- 278.Mollerherm H, von Kockritz-Blickwede M, Branitzki-Heinemann K. Antimicrobial Activity of Mast Cells: Role and Relevance of Extracellular DNA Traps. Front Immunol. 2016;7:265. doi: 10.3389/fimmu.2016.00265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Ueki S, Tokunaga T, Fujieda S, et al. Eosinophil ETosis and DNA Traps: a New Look at Eosinophilic Inflammation. Curr Allergy Asthma Rep. 2016;16(8):54. doi: 10.1007/s11882-016-0634-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Dahlin JS, Malinovschi A, Ohrvik H, et al. Lin- CD34hi CD117int/hi FcepsilonRI+ cells in human blood constitute a rare population of mast cell progenitors. Blood. 2016;127(4):383–391. doi: 10.1182/blood-2015-06-650648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.O’Sullivan JA, Carroll DJ, Bochner BS. Glycobiology of Eosinophilic Inflammation: Contributions of Siglecs, Glycans, and Other Glycan-Binding Proteins. Front Med (Lausanne) 2017;4:116. doi: 10.3389/fmed.2017.00116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Mitchell MD, Sato TA, Wang A, Keelan JA, Ponnampalam AP, Glass M. Cannabinoids stimulate prostaglandin production by human gestational tissues through a tissue- and CB1-receptor-specific mechanism. Am J Physiol Endocrinol Metab. 2008;294(2):E352–356. doi: 10.1152/ajpendo.00495.2007. [DOI] [PubMed] [Google Scholar]
- 283.Samson MT, Small-Howard A, Shimoda LM, Koblan-Huberson M, Stokes AJ, Turner H. Differential roles of CB1 and CB2 cannabinoid receptors in mast cells. J Immunol. 2003;170(10):4953–4962. doi: 10.4049/jimmunol.170.10.4953. [DOI] [PubMed] [Google Scholar]
- 284.Sugawara K, Zákány N, Hundt T, et al. Cannabinoid receptor 1 controls human mucosal-type mast cell degranulation and maturation in situ. J Allergy Clin Immunol. 2013;132(1):182–193. doi: 10.1016/j.jaci.2013.01.002. [DOI] [PubMed] [Google Scholar]