Mast Cell and Basophils: Interaction with IgE and Responses to Toll like Receptor Activators

Jean S. Marshall, Michael G. Brown, and Ruby Pawankar

Mast Cells in Host Defense and Innate Immunity

Mast cells have multiple roles, not only as effector cells in allergic disease, but also as sentinel cells in the regulation of early immune responses to infection and initia- tors of early acquired immune responses [1, 2]. These are fully established as cellular initiators of the symptoms of acute allergic disease and may have more complex immunoregulatory roles through IgE-mediated activation. There is also longstanding evidence, from nematode parasite infection models, that activation can con- tribute to host defense [3–6]. Over the past 11 years evidence has accumulated for a critical role for mast cells in host defense to a variety of bacterial pathogens – primarily as a result of enhanced neutrophil recruitment [7–10]. Mast cell production of TNF has been shown to be of particular importance to several of these processes [8, 9]. However, other multiple mast cell mediators, such as cytokines and proteases, are also of importance in regulating or initiating innate immune responses [11–13]. Mast cells have been shown to respond vigorously to fungal products in vitro [14, 15] and there is an increasing evidence of their important role in models of viral infection [16–18]. Bacterial, viral, and fungal infections have been demonstrated to be associated with the initiation or exacerbation of allergic symptoms. For example, Staphylococcus aureus infection in atopic dermatitis (AD) [19] as well as respiratory syncytial virus or other viral infection in asthma [20–22]. With this in mind, dual roles of mast cells and their mediators in allergic disease and host defense need to be carefully considered. Mast cell activation and production of mediators can occur through multiple mecha- nisms other than classical FcεRI cross-linking. In allergic disease, pathogen-associated mast cell activation may modify sensitization or alter the nature of disease expression. Understanding these processes could provide important opportunities for therapy.

J.S. Marshall and M.G. Brown Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada R. Pawankar () Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, Japan

R. Pawankar et al. (eds.), Allergy Frontiers: Classification and Pathomechanisms, 113 DOI: 10.1007/978-4-431-88315-9_8, © Springer 2009 114 J.S. Marshall et al.

Mast Cell Expression of Innate Immune Receptors

Mast cells express a wide variety of receptors, which enable them to fulfill their role as sentinel cells in early host defense and participate in responses to secondary infec- tions or antigen challenge (see Table 1). When considering these, it is important to distinguish between mast cells derived from different species or distinct anatomical sites, since there are wide variations between mast cell subsets in mediator content and responses to specific activators. It is not surprising that another aspect of such hetero- geneity is the range of innate receptors expressed on cells from specific tissue sources or derived under particular culture conditions. Toll-like receptors (TLRs) are a major family of innate immune receptors that has received a great deal of attention in view of their pivotal role in host defense. These germ-line encoded pattern recognition recep- tors can be activated by a wide variety of pathogen products such as lipopolysaccha- ride (LPS), peptidoglycan (PGN), CpG motifs, and double-stranded RNA, and also by endogenous products produced during tissue injury. Their properties and signaling pathways have been studied in depth and are well reviewed elsewhere [23–26] (see Table 2). However, a number of other innate immune receptors, especially comple- ment component receptors, lectin-like receptors for pathogen products, and adhesion molecules with known roles in innate immune activation are expressed by mast cells and can play an important role in responses to some types of infection [27–30]. TLR1, 2, and 6 have been shown to be expressed by mast cells from a variety of sources although in some cases expression is only observed at the mRNA level. TLR4 is widely expressed on murine mast cells [31–34] and has been described in human- cultured mast cells and ex vivo mast cells from some sites [35, 36], while TLR4 is absent from mast cells from other sites or in alternate conditions [37]. TLR5 has been described as absent from some human mast cells and mast cell lines [14] although reported in cultured peripheral blood stem cell-derived mast cells [38]. TLR3 expres- sion has also been observed in both human and murine mast cells [38–40], while the presence of TLR7 and 9 appears to be highly dependent upon the location of mast cells. A good example of such mast cell source-specific variation in TLR expres- sion within one species comes from studies of fetal mouse -derived mast cells (FSMCs) compared with murine bone marrow-derived murine mast cell (BMMC). FSMCs displayed a connective tissue mast cell phenotype based on granule contents and were found to express high levels of mRNA for TLR1, 2, and 7, a moderate level for TLR3, TLR4, and TLR6, and a low level for TLR9, but lacked TLR5 [39]. In con- trast, BMMCs expressed high levels of TLR2 and TLR6, moderate levels of TLR4, and low levels of TLR1, TLR3, and TLR7, and lacked both TLR5 and TLR9. In human, comparisons of neonatal foreskin mast cells with those derived from adult tissue have proved equally enlightening. Lung mast cells expressed high levels of TLR2, TLR3, moderate levels of TLR4, TLR7, and TLR10, and low levels of TLR5 and TLR9, and lacked TLR1, TLR6, and TLR8. In contrast, foreskin-derived mast cells expressed high levels of TLR3, moderate levels of TLR2, TLR4, and TLR9, and low levels of TLR5, TLR7, TLR10, and were deficient in TLR1, TLR6, and TLR8 [38]. In comparison to cultured human peripheral blood-derived mast cells [36], both of these tissue-derived mast cell preparations exhibited less TLR5, TLR7, Mast Cell and Basophils 115

Table 1 Mast cell Basophil Innate receptor type mouse human mouse human TLR1 mRNA + + + Protein TLR2 mRNA + + + + Protein + + TLR3 mRNA + + Protein TLR4 mRNA + + + + Protein + + + TLR5 mRNA + Protein TLR6 mRNA + + + Protein TLR7 mRNA + + Protein TLR8 mRNA + + Protein TLR9 mRNA + + + Protein TLR10 mRNA + + Protein C1qR mRNA Protein + C3aR mRNA + Protein + C5aR mRNA Protein + + FimH R (CD48) mRNA Protein + RNA helicase (RIG-I, Mda-5) mRNA Protein NOD1 mRNA Protein NOD2 mRNA + Protein + CD11b/CD18 (MAC-1) mRNA Protein + + Dectin-1 mRNA Protein + References: Malaviya et al. (1994), Fureder et al. (1995), Ghebrehiwet et al. (1995), Rosenkranz et al. (1998), Malaviya et al. (1999), McCurdy et al. (2001, 2003), Applequist et al. (2002), Sabroe et al. (2002), Varadaradjalou et al. (2003), Kulka et al. (2004), Matsushima et al. (2004), Bonini et al. (2005), Inomata et al. (2005), Thangam et al. (2005), Florian et al. (2006), Komiya et al. (2006), Kulka and Metcalfe (2006), Kubo et al. (2007), Wu et al. (2007) 116 J.S. Marshall et al.

TLR9, and TLR10, with no TLR1, TLR6, and TLR8 mRNA. An additional study investigating the protein expression of TLR on a number of inflammation-associated immune effector cells, including mast cells, in both normal and allergic conjunctiva, indicated that while mast cells expressed both TLR2 and TLR4, they lacked any detectable TLR9 [41]. It has been suggested that mast cells from certain sites, such as the human intestine, may not express a range of functional TLRs at the protein level; this could represent adaptation of mast cells to these sites rich in microflora [42]. However, extended periods of ex vivo culture are required to obtain pure mast cells from this site, which could alter TLR expression. Both pathogen products and the cytokine microenvironment have been demon- strated to profoundly alter TLR expression in other cell types and this is likely also true for mast cells. In the context of ongoing disease, current evidence suggests that mast cells are capable of expressing a wide range of functional TLRs and thereby responding to multiple pathogen-associated signals. Analysis of protein level expression of sev- eral TLRs has been hampered by the lack of good monoclonal antibodies and by the primarily intracellular location of some TLRs making functional antibody-blocking studies difficult to interpret. However, from what we have learned, in other cell types even quite low levels of TLR expression can mediate substantial responses to certain pathogen products. Inflammatory cytokine and pathogen product exposure can also lead to alterations in the levels or type of TLR expression.

Mast Cell Responses to TLR 1, 2, 4, 5, and 6 Activators

TLR activators are found commonly in our environment, within our natural microbial flora and are a feature of most pathogens. In addition, substances produced during tis- sue damage can serve as TLR activators. Some examples of common TLR activators are provided in Table 2. Multiple studies using TLR-deficient animals have demon- strated the importance of this family of receptors in host defense against bacterial and

Table 2 Pathogen products Receptor References PAM3CSK4 (triacylated LP) TLR1 Marshall (2004), Miyake (2007) PGN, zymosan, LTA TLR2 Marshall (2004) dsRNA TLR3 Kulka et al. (2004), Matsushima et al. (2004), Kawai and Akira (2007) LPS TLR4 Malaviya and Georges (2002), Marshall (2004), Kawai and Akira (2007) Flagellin TLR5 McCurdy et al. (2001), Supajatura et al. (2001), Kulka et al. (2004), Kawai and Akira (2007) MALP-2 (diacylated LP), PGN TLR6 Kawai and Akira (2007), Miyake (2007) ssRNA, R-848 TLR7 Dieboldo et al. (2004), Matsushima et al. (2004) ssRNA, R-848 TLR8 Heil et al. (2004) CpG TLR9 Matsushima et al. (2004) Unknown TLR10 Hasan et al. (2005) Mast Cell and Basophils 117 fungal infection. As part of their “sentinel cell” role, mast cells can respond very rap- idly to activation with TLR ligands. A critical feature of mast cell responses to TLR activators is their selectivity. In both humans and mice, TLR activation usually leads to the production and release of only a subset of potential mast cell mediators and fre- quently occurs in the absence of degranulation [14, 36, 43]. Specific TLR interactions give rise to different mediator profiles, in keeping with a role for mast cell activation by pathogen products in defining the nature of the early immune and inflammatory response to infection. A number of co-receptors such as CD14 and dectin-1 can play an important role in the TLR-dependent mast cell responses and their activities may modify the mediator profile induced by TLR ligands. Since the early 1990s we have been aware of the ability of rodent mast cells to respond to pathogen products such as LPS and give rise to selective cytokine produc- tion in the absence of degranulation [43]. More recent studies of human and rodent mast cells have confirmed a wide range of selective mast cell mediator responses to TLR activators and in several cases the role of specific TLRs have been confirmed using rodent gene deletion models or neutralizing antibodies [31, 32, 36, 44]. TLR2 and TLR4 on murine mast cells have been demonstrated to be critical for mast cell cytokine responses to LPS and bacterial PGN, respectively. There is some controversy surrounding the ability of bacterial PGN or TLR2-activating lipopeptides to induce mast cell degranulation, since there is little evidence to support a calcium signal being generated via TLR-mediated processes [45]. However, PGN is a very effective activa- tor of complement via classical, alternate, and mannose-binding pathways, comple- ment component receptors could lead to an effective degranulation response in vivo even in the absence of TLR2-mediated signals. Recent studies of murine responses to PGN support this concept [30]. Human-cultured mast cells have also been described to selectively respond to some TLR activators without degranulation under some con- ditions. McCurdy et al. [14] described the selective production of the cytokines IL-1β and granulocyte- colony-stimulating factor (GM-CSF) by cord blood- derived human-cultured mast cells in response to PGN, the bacterial lipopeptide ana- logue Pam3CysSer Lys4, and the yeast cell wall component zymosan. Both Zymosan and PGN were also able to induce the selective production of LTC4 from such mast cells under conditions, which did not lead to any significant degranulation. The ability of mast cells to produce leukotrienes in response to certain TLR acti- vators has been established [14, 38]. However, while some of these responses are partially TLR2-dependent there is increasing evidence that they are not all mediated via TLR2 receptors, but can involve other members of a TLR2-containing signaling complex or alternate innate receptors. For example, the mast cell LTC4 response to the yeast cell wall component Zymosan has been shown to be dependent on activa- tion of the dectin-1 receptor [46]. This receptor activates Syk kinase [47, 48] and has the ability to induce a calcium signal, which together with TLR signaling, such as MAPK phosphorylation, provides an appropriate set of combined signals for the generation of LTC4 by mast cells. Dectin-1 signaling has also been demonstrated to be important for lipid mediator production by in response to fungal products [49]. The dependence on co-receptors such as dectin-1 provides additional therapeutic opportunities to inhibit leukotriene generation selectively in response to fungal infection or environmental exposure. 118 J.S. Marshall et al.

Array studies have been performed using several TLR activators and both human and murine mast cells. These have revealed important differences in the range of mediators, particularly cytokines and chemokines, induced by TLR-mediated mast cell activation and those induced by FcεRI cross-linking [44, 50]. The data from such studies is extremely useful as a starting point for elucidating the biological role of mast cell TLR expression and function. In addition, to consider the range of mediators produced, it is also important to recognize that the mast cell can be a particularly rapid source of a number of critical cytokines and chemokines, either because they are found preformed or because the mast cell has specific mechanisms whereby protein level production can be rapidly initiated.

Mast Cell Responses to TLR 3, 7, 8, and 9

TLRs on innate effector cells such as mast cells help to differentiate between vari- ous types of pathogens, directing appropriate responses to a particular pathogen. Evidence in support of this concept is derived from a number of infection models and also from studies of the responses of mast cells to activation of receptors for DNA and RNA sequences associated with infection as compared with responses to bacterial and fungal cell wall components. The nucleic acid sequence receptors TLR3, TLR7, TLR8, and TLR9 have been shown to function primarily intracel- lularly associated with the endoplasmic reticulum and endosomes, and to signal primarily through similar pathways observed for other TLRs [26, 51]. Mast cell activation with polyI:C has been used as a model for examining TLR3 responses. Kulka et al. [36] revealed a potent and selective type 1 interferon response of human mast cells treated in this way that could be blocked by pretreat- ment of mast cells with an antibody to TLR3. This group also observed ablated type 1 interferon responses in BMMC derived from TLR3-deficient animals. This clearly demonstrates a role for TLR3 in the type 1 interferon response. However, it has been suggested that other aspects of the cellular response to polyI:C might be mediated by alternate cellular receptors for double-stranded RNA such as PKR and RIG-I. Treatment of mast cells with a number of viral preparations also gave rise to a type 1 interferon response. A number of other mediators have been demonstrated to be produced by human mast cells following viral infection. For example, dengue virus infection of mast cells is associated with the increased production of IL-1, IL-6, CCL3, CCL4, and most strikingly CCL5, but not CXCL8 [52, 53]. It is not yet known if these effects are TLR3-mediated. Mast cell interactions with the extracellular matrix may also be altered via TLR activation. Following exposure to polyI:C human mast cell adhesion to vitronectin and fibronectin were reduced by an active TLR3-dependent process. In addition, mast cell responsiveness to IgE-mediated activation was inhibited. Such responses may enhance the localization of mast cells to sites of infection and help to focus the mast cell activities towards aiding antiviral responses [38]. In vivo studies of mast cell responses to TLR3 activation are limited, however Orinska et al. [40] observed Mast Cell and Basophils 119 a mast cell-dependent recovery in CD8 T cells following polyI:C treatment or Newcastle disease virus infection, in experiments comparing mast cell-deficient mice with controls. In contrast, granulocyte recruitment to the peritoneal cavity occurred similarly in mast cell-containing and mast cell-deficient animals. As previously indicated, murine skin-derived and bone marrow-derived mast cells have distinct profiles of TLR expression. Both inflammatory cytokine and chemokine secretions by these two mast cell types following stimulation with TLR ligands have been analyzed. TLR3, TLR7, and TLR9 stimulations of fetal skin-derived mast cells with polyI:C, R-848, and CpG respectively resulted in a dose-dependent production of TNF and IL-6. In contrast, BMMC response to these ligands was negligible or absent. Chemokine responses followed a similar pattern, with MIP-1α, MIP-2, and CCL5 produced by poly I:C, R848, and CpG- containing oligonucleotides, while similarly treated mBMMCs produced little of these chemokines [37]. In other studies, using the TLR7 activator imiquimod applied as a cream to the skin, it was shown that mast cells are essential for ensuing inflammatory response, including the mobilization of local populations by mechanisms that involve mast cell-derived IL-1β and TNF [54]. The adjuvant effect of imiqui- mod in the generation of peptide-specific cytotoxic T-cell (CTL) responses was also shown to be mast cell-dependent. There is some controversy surrounding the ability of mast cells to respond effec- tively to CpG-containing oligonucleotides. Early studies of BMMC demonstrated small but significant TNF and IL-6 responses [55], however, TLR9 dependence was not examined. More recent studies of human mast cells also noted modest pro- duction of IL-1β, IFN-α, and TNF in response to CpG-oligonucleotide treatment. Interestingly, CpG treatment in animal models of allergic disease has also revealed decreased mast cell accumulation, but it is not known if this is a direct effect on mast cells or is the result of a decreased severity of disease. Evidence is emerging to suggest that certain pathogens have also acquired the ability to utilize TLRs to their benefit. A major example of this process has been described in mast cells treated with CpG-containing oligonucleotides. It has been demonstrated that the HIV virus is able to reactivate from latent infection in mast cells via stimulation through TLR9 [56]. It is not yet known if the mast cell also serves as a reservoir of other types of latent infection.

Interactions Between IgE and TLR Signaling

Since mast cells reside at classical mucosal surfaces and in the skin there are many situations in which mast cells might concurrently be in contact with allergens and TLR activators. Dual activation or modulation of Fc receptor expression by TLR activators could contribute to the regulation of allergic inflammation especially in the context of food allergies where large amounts of intestinal flora are present or atopic dermatitis when S. aureus colonization is common. Recently, Yoshioka 120 J.S. Marshall et al. et al. [57] examined the ability of the TLR2 activator lipotechoic acid (LTA) to modulate FcεRI expression by pulmonary mast cells and a human mast cell line. They observed substantial down-regulation of FcεRI expression in response to LTA treatment and suggest that this could be employed as a method to control allergic disorders. However, the pro-inflammatory effects of TLR activators may limit the usefulness of this approach. Others have suggested a less beneficial role for TLR activators. Qiao et al. [45] observed that antigen cross-linking of FcεRI interacted synergistically with either TLR2 or TLR4 ligands to enhance production of pro- inflammatory cytokines in murine mast cell lines. Enhanced MAP kinase activation may have contributed to increased cytokine responses that occurred in the absence of enhanced degranulation or leukotriene generation. More recently, Fehrenbach et al. [58] examined the ability of both dipalmitoylated and tripalmitoylated TLR2 activators in enhancing IgE-mediated mast cell activation. They also observed sub- stantial enhancement of cytokine production via a TLR2-dependent pathway. Other opportunities for cross talk between IgE-mediated mast cell activation and TLR-mediated pathways are found when one considered the effects of released mediators in complex tissues. For example, Talreja et al. [59] observed that hista- mine induces TLR2 and 4 expression in endothelial cells suggesting that the pro- inflammatory effects of TLR activators at mucosal sites may be enhanced in the context of ongoing allergic disease. In animal studies, while some TLR activators (such as LPS [60]) have been demonstrated to inhibit the development or expres- sion of allergic disease in a mast cell-dependent manner, other bacterial compo- nents such as PGN have been shown to enhance responses [61].

TLR Activation on Mast Cells Is Only Part of a Complex Integrated Innate Response

While a number of studies have identified key TLR-mediated processes with pro- found effects on the outcome of infection in rodent models, in most disease proc- esses multiple TLRs and co-receptors are activated, in addition to a series of other innate immune responses that work in concert with TLR-mediated processes. For example, during viral infection, mast cells and other innate effector cells might respond to viral-derived double-stranded RNA through both TLR-dependent and TLR-independent mechanisms and also respond to specific viral proteins (e.g., F-protein of RSV) by activation of TLR4. In addition, local tissue damage could liberate substances such as fibronectin breakdown products that can further activate innate immunity in part, through mast cell activation [62]. For many TLR activators, TLRs are only one of a number of receptor systems that can be utilized for initiation of host defense. In vivo, there are often antibody responses to LPS and PGN that can contribute to mast cell activation either directly through Fc receptor cross-linking or indirectly through the activation of the classical complement pathway. Mast cells express multiple complement receptors including α β C3, C4 C3a, and C5a receptors, as well as the 1 2 integrin receptor that serves as a receptor for activated C1q [28, 29]. These receptors can contribute substantially to Mast Cell and Basophils 121 responses to active infection or pathogen products, but are often overlooked during in vitro studies of mast cell activation. Recent studies have demonstrated a critical role for complement in the mast cell-dependent early cell recruitment response to PGN [30]. In addition, mast cells express a number of lectin-like receptors that may contribute to host defense such as peptidoglycan recognition proteins and integrins such as MAC-1, which may contribute. NOD1 and NOD2 expressed by some mast cells might also contribute to responses to Gram-positive bacteria.

Mast Cell Activation by Pathogen Products: Effects on Acquired Immunity

Pathogen product activation by mast cells is thought to modulate acquired immune responses. Studies from Duke University [63] have demonstrated that mast cell activation by whole bacteria or other secretagogues is an important inducer of lymph node hypertrophy following infection. Such lymph node activation, which includes blockade of emigration of T cells from the node, increases the opportunity for antigen laden dendritic cells to come into contact with appropriate T cells and provides an appropriate microenvironment to support optimal antigen presentation and the development of an ensuing acquired immune response. This type of mast cell function may not be dependent primarily on TLR function. Jawdat et al. [64] noted that while S. aureus derived peptidoglycan injection into the ear-induced lymph node swelling by a mast cell-dependent mechanism this response was not inhibited in TLR2-, TLR4-, or MyD88-deficient mice. Mast cell activation by either IgE/antigen or pathogen products also has the ability to mobilize populations and cause them to migrate to draining lymph nodes. This has been most effectively demonstrated in the skin where mast cell activation leads to Langerhans cell migration into the draining lymph node. While the role of mast cells in contact hypersensitivity responses remains highly controversial [65–68]. There appear to be some circumstances in which the mast cell can be pivotal for the development of an effective response. A number of T-cell responses have been shown to be mast cell-dependent by mechanisms which often require the mast cell as an early source of TNF. The role that mast cell activation by either TLRs or IgE have on allergic sensitization or effective responses to vac- cination in humans remains to be determined.

Mast Cells in Allergic Diseases

Mast Cells in Allergic Rhinitis and Asthma

Allergic rhinitis (AR) is the most common allergic disease affecting up to 25–40% of the population. The pathophysiology of AR shares many similarities to allergic asthma and the two diseases are often considered manifestations of “one syndrome, 122 J.S. Marshall et al. one airway, one disease” [69]. Mast cells constitutively reside in the nasal mucosa of normal subjects, but are not normally present in the nasal epithelium. Mast cells comprise of two phenotypes: tryptase (MCT)-positive and tryptase/chymase (MCTC)-positive [70]. While both MCT-positive and MCTC-positive mast cells reside within the subepithelium, the phenotypes of mast cells in the allergic nasal epithelium are predominantly of the MCTC [70–72]. Upon allergen exposure, mast cells migrate to, and proliferate within, the epithelium [73, 74]. Although Stem cell factor (SCF) is a strong mast cell chemoattractant and elevated in the nasal lavage fluid of patients with seasonal AR, the levels of SCF in the allergic nasal epithelium are relatively compared with the lamina propria. By contrast, the levels of RANTES (CCL5) are increased in the epithelium and may instead have a more prominent role in the intraepithelial migration of nasal mast cells (NMCs) as compared to SCF or Eotaxin (CCL11) [74]. A low SCF environment is known to decrease mast cell chymase expression, which may explain the selective accumulation of MCT in the nasal epithelium of AR [74, 75]. Mast cell degranulation is characterized by elevated levels of tryptase, histamine, LTB4, LTC4, and PGD2 in the nasal secretion of individuals with AR following nasal allergen challenge [76–79]. These mediators contribute to the characteristic early-phase symptoms of AR namely sneezing, pruritus, rhinorrhea, and nasal con- gestion. Histamine is a major mediator inducing vasodilation, increased vascular permeability, and increased glandular secretion. In addition, histamine acts on the sensory nerve endings of the trigeminal nerve to cause sneezing. However, more recently mast cells are found to be an important source of a variety of cytokines (TNF-alpha, IL-4, IL-5, IL-6, and IL-13) and mast cell activation results in orches- trating of the late-phase reaction, inducing the infiltration of eosinophils through the release of platelet-activating factor (PAF) and LTB4; and the upregulation of VCAM-1 expression on endothelial cells. The survival of eosinophils is promoted through the mast cell release of granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-5. Additionally, histamine upregulates RANTES (CCL5) and GM-CSF, while IL-4, IL-13, and TNF-α upregulate Eotaxin (CCL11) and TARC (CCL17), further contributing to the late-phase eosinophilic/T-cell infiltration. Clinically, this is displayed as an increase in nasal mucosal swelling with increased nasal airway resistance [74, 80]. As regard to nasal hyperresponsiveness it is the result of exaggerated neural reactivity, and the levels of nerve growth factor (NGF) in the nasal lavage fluids from patients with AR are markedly elevated and ampli- fied through nasal allergen challenge [81, 82]. In addition, IL-4, IgE, and NGF increases the expression of FcεRI and c-Kit on mast cells [83, 84]. Furthermore, the FcεRI expression in NMC correlated well with the levels of serum IgE [83]. These findings taken in concert with the observations of Pastorello et al., who dem- onstrated a strong positive correlation between the levels of serum IgE and clinical symptoms, in symptomatic patients with allergic rhinoconjunctivitis [85], suggest a very important role for NMC in regulating chronic allergic inflammation. Moreover, mast cells can induce IgE synthesis in B cells and IL-4 and IgE can upregulate the FcεRI expression in mast cells. Several studies have demonstrated that local IgE synthesis occurs at sites of allergic inflammation [86–88]. Putting together all these Mast Cell and Basophils 123 observations one can perceive a very important role for the mast cell in perpetuating allergic inflammation via the mast cell-IgE-FcεRI cascade [88]. As in allergic rhinitis, mast cells are primary effector cells in asthma and play crucial roles in the early-phase response, late-phase response, and chronic allergic inflammation [89–92]. Mast cell numbers are increased in the bronchial epithe- lium of atopic asthmatics [93]. Besides the epithelial compartment, mast cells are widely distributed in specific compartments of the asthmatic airways (i.e., the bronchoalveolar space [94]), beneath the basement membrane, in close proximity to blood vessels, near submucosal glands and in the ASM bundles [95]. In fact a close correlation between mast cell numbers in the bronchial smooth muscle of asthmatics and the degree of airway hyperresponsiveness to methacholine is well established [95]. Mast cells in asthmatics release a variety of mediators (histamine, cysteinyl leukotreine C4 (LTC4), PGD2, tryptase, and chymase), as well as cytokines and chemokines, which induce and sustain chronic inflammation in asthma by regulat- ing the recruitment and function of T cells, macrophages, ASM, and epithelial cells. As mentioned earlier in this chapter increasing evidence suggests that his- tamine can regulate the activity and cytokine/chemokine release from other cells. Histamine induces lysosomal enzyme release and IL-6 and TNF-α production from human lung macrophages [96] and CysLTs exert a variety of responses on bron- chial smooth muscle, human lung macrophages, mast cells, and peripheral blood leukocytes via the CysLTR1 and CysLTR2 [69–72]. Besides the above-mentioned mediators, mast cells in asthmatic airways express a wide spectrum of cytokines (e.g., IL-4, SCF, IL-5, IL-6, IL-8, IL-13, IL-16, trans- forming growth factor-beta [TGF-β], IL-25, TNF-α, and granulocyte-macrophage colony-stimulating factor [GM-CSF] [Pawankar R et al.]) [97]. IL-16 is chemo- tactic for CD4 + T cells and IL-25 can induce IL-4 and IL-13 gene expression, and this may contribute to mast cell–ASM interactions, as well as in amplifying allergic inflammation via the mast cell-IgE-FcεRI cascade [98]. IL-3, IL-5, and GM-CSF from mast cells can enhance histamine release and IL-4 synthesis in basophils, whereas IL-4 enhances the production of proinflammatory mediators (PGD2 and LTC4) and various cytokines by mast cells [99, 100]. Thus, Th2 (IL-4, IL-5, and IL-3) cytokines can induce changes in the biosynthetic pathways of mast cells at sites of allergic inflammation. SCF, a principal growth differentiating and chemotactic factor for human mast cells is expressed and released by lung mast cells [101]. Several mast cell mediators like histamine, tryptase, LTC4, TNF-α, and TGF-β1 also exhibit fibrogenic activity [102]. Mast cells that are treated with IL-4 express Toll-like receptor 4 (TLR4), which when activated by lipopolysaccharide (LPS) induces the release of Th2 cytokines [103]. Activation of mast cells via TLR2 and TLR3 selectively induce the release of leukotrienes, GM-CSF, and IL-1β [14, 36, 103]. Human-cultured mast cells also express TLR1, TLR2, TLR4, TLR5, TLR6, TLR7, and TLR9 [36]. These findings highlight several novel mechanisms, whereby endogenous, bacterial, and viral pro- teins can activate human mast cells, thus providing the means by which viruses and bacteria can induce asthma exacerbations. 124 J.S. Marshall et al.

Structural alterations of the airways, defined as “tissue remodeling,” account for a progressive and irreversible loss of lung function in chronic asthma. The positive correlation between the thickness of the tenascin and laminin layers and mast cell density in asthmatics suggests that mast cells may have a role in tissue remodeling [104]. Mast cells, which are closely associated with blood vessels and are increased at angiogenic sites, can contribute to various aspects of angiogenesis [105]. Mast cells synthesize and release various pro-angiogenic factors (histamine, tryptase, TGF-β, IL-8, and vascular endothelial growth factor [VEGF] and the production of VEGF by mast cells is increased by PGE2 and other cAMP-elevating agents) [106–108].

Implications of Mast Cells in Rhinitis and Asthma

The unique microlocalization of mast cells in specific nasal and lung tissue com- partments, the ability of these cells to migrate to the epithelial compartment where they are easily activated by allergens or pathogens, the powerful effector repertoire, the recognition of their different microbial-related activating ligands, and their plasticity in response to various signals suggest that mast cells have a central role in allergic airway diseases like rhinitis and asthma. However, mast cells at different stages of maturation, might have different, or even, in some cases, protective roles in the appearance of the asthmatic phenotype. The extent to which each effector cell contributes toward each phase of allergic inflammation and tissue repair remains to be fully elucidated.

Mast Cells in Allergic Conjunctivitis

Ocular allergy occurs in 45% of the allergic population [109, 110]. The location of mast cells in close proximity to the external environment in the mucosa of the eye allows for exposure of these cells to allergen, thereby facilitating cross-linking of membrane-bound IgE, which leads to degranulation and release of inflammatory mediators. Although there are several types of ocular allergy, seasonal and peren- nial allergic conjunctivitis represent the majority of allergy cases [111]. In normal individuals, mast cells are abundant in the conjunctival stroma with an estimated 50 million cells residing at this environmental interface [112]. In symptomatic allergic patients, an increase in mast cells with evidence of degranulation is seen in conjunctival biopsies [113]. In addition to the increase in mast cells within the conjunctiva, the number of mast cells expressing IL-4 message is increased three- fold in seasonal allergic conjunctivitis [114]. Further, use of a mast cell stabilizer (nedocromil sodium) reduces the amount of histamine and PGD2 by more than 70% after challenge, thereby supporting a major role for mast cells in allergic conjunctivitis [115]. In addition to common mast cell mediators such as histamine and cytokines, chemokines released from activated mast cells mediate late-phase Mast Cell and Basophils 125

reactions by recruitment of additional inflammatory cells. Mast cells residing within the conjunctiva express CCR3 and the use of a CCR3 antagonist in a mouse model of allergic conjuctivitis ablated both the early- and late-phase reactions [116]. In this model, the CCR3 antagonist leads to mast cell stabilization and inhi- bition of immediate hypersensitivity, but also impaired neutrophil and eosinophil influx during the late-phase response [116].

Mast Cells in Atopic Dermatitis

Mast cells are increased in lesions of patients with atopic dermatitis (AD) [117, 118]. They reside in the papillary dermis and undergo migration through where they may influence keratinocyte activation and stimulation of endothelial growth with neoangiogenesis [119]. Although histamine has an established role in other atopic diseases, the effect of histamine in AD is questionable, given that levels are not increased compared with control subjects. Tryptase and activation of proteinase- activated receptor-2 (PAR-2) may contribute to the pruritus seen in AD, as tryptase is reported to be increased up to fourfold in AD patients and PAR-2 expression is markedly enhanced on primary afferent nerve fibers in skin biopsies from patients with AD [120]. On the other hand, chymase may play a role by weakening the skin barrier, in turn allowing an enhanced permeability to allergens and microbes [121]. An association between a promoter polymorphism (rs1800875) of the mast cell chymase gene (CMA-1) and AD has been reported [122]. Significantly elevated levels of total IgE are found in about 80% of patients with AD. Beyond traditional signaling through the FcεRI receptor on mast cells, a novel IgE-independent mast cell activation pathway has been proposed for AD involving CD30 and mast cells were shown to be the predominant CD30 ligand-positive cell in AD lesions and activation through CD30 induced a de novo. As in AR, mast cell–nerve interactions may also play a role in promoting inflammation in AD [123]. Contacts between mast cells and nerves are increased in both lesional and non-lesional samples of AD when compared with normal controls [124]. Inflammation appears to be mediated by neuropeptides such as substance P, calcitonin gene-related peptide, vasoactive intestinal peptide, and NGF [84, 125–127].

Mast Cells in Anaphylaxis

Anaphylaxis is an acute, severe, systemic reaction to a foreign stimulus that is often thought to be associated with mast cell activation. The strongest evidence of a role of mast cells in anaphylaxis is in that serum levels of tryptase, resulting mainly from mast cell degranulation, peaks 1–2 h following the onset of IgE-mediated anaphy- laxis [128–130]. Besides, the classical IgE-dependent anaphylaxis can occur due to IgE-independent mechanisms namely through IgG and complement receptors 126 J.S. Marshall et al.

[131–134]. Although anaphylaxis is considered a systemic event, the presence and activation of mast cells in specific organs may play a critical role in the severity. Within the cardiac mast cells are located between myocardial fibers, around blood vessels and in the arterial intima, activation of these critically positioned mast cells may directly contribute to cardiopulmonary failure. PAF is thought to be a critical factor in the development of anaphylactic shock through its ability to induce hypo- tension and cardiac dysfunction [135–137]. The overall number of mast cells may also be relevant in anaphylaxis in that in those with recurrent anaphylaxis tend to have more dermal mast cells than those without anaphylaxis.

Closing Comments

The mast cell has therefore a central and versatile role in the pathogenesis of allergic diseases, and more recently has been shown to be involved in innate immunity in response to bacterial and parasitic infections [168]. Toll-like receptors and other innate immune receptors on mast cells may participate in effective host immunity and in the exacerbation of allergic inflammation at sites of infection. By better understanding these receptor systems and their function on mast cells we will obtain better tools with which the function of mast cells can be locally modified to enhance appropriate immune responses while limiting adverse inflammatory effects. In aller- gic disease, certain TLRs may hold promise as effective regulators of the develop- ment of allergic disease or as modifiers of ensuing mast cell-mediated responses. More in vivo animal studies and studies of human material are required to come to grips with the complexity of the multiple signals provided to local mast cells at sites of infection. As sentinel cells, the mast cell’s range of mediator production and rapid selective responses are extremely important in dictating the nature of inflam- matory and immune response. Harnessing these responses holds enormous promise as a strategy to modify local inflammatory responses and immune responses. As our understanding of the various roles of mast cells in disease pathogenesis evolves, novel therapeutic targets will continue to be identified.

References

1. Marshall JS and Jawdat DM (2004) Mast cells in innate immunity. J Allergy Clin Immunol 114: 21–7 2. Dawicki W, Marshall JS (2007) New and emerging roles for mast cells in host defence. Curr Opin Immunol 19:31–8 3. Woodbury RG, Miller HR, Huntley JF, et al (1984) Mucosal mast cells are functionally active during spontaneous expulsion of intestinal nematode infections in rat. Nature 312:450–2 4. Knight PA, Wright SH, Lawrence CE, et al (2000) Delayed expulsion of the nematode Trichinella spiralis in mice lacking the mucosal mast cell-specific granule chymase, mouse mast cell protease-1. J Exp Med 192:1849–56 Mast Cell and Basophils 127

5. McDermott, JR., Bartram RE, Knight PA, et al (2003) Mast cells disrupt epithelial barrier function during enteric nematode infection. Proc Natl Acad Sci USA 100:7761–6 6. Pennock JL, Grencis RK (2006) The mast cell and gut nematodes: damage and defence. Chem Immunol Allergy 90:128–40 7. Echtenacher B, Männel DN, Hültner L (1996) Critical protective role of mast cells in a model of acute septic peritonitis. Nature 381:75–7 8. Malaviya R, Ikeda T, Ross E, et al (1996) Mast cell modulation of neutrophil influx and bacte- rial clearance at sites of infection through TNF-alpha. Nature 381:77–80 9. Prodeus AP, Zhou X, Maurer M, et al (1997) Impaired mast cell-dependent natural immunity in complement C3-deficient mice. Nature 39:172–5 10. Xu X, Zhang D, Lyubynska N, et al (2006) Mast cells protect mice from Mycoplasma pneu- monia. Am J Respir Crit Care Med 173:219–25 11. Huang C, De Sanctis GT, O’Brien PJ, et al (2001) Evaluation of the substrate specificity of human mast cell tryptase beta I and demonstration of its importance in bacterial infections of the lung. J Biol Chem 276:26276–84 12. Caughey GH (2007) Mast cell tryptases and chymases in inflammation and host defense. Immunol Rev 217:141–54 13. Lin TJ, Garduno R, Boudreau RT, et al (2002) Pseudomonas aeruginosa activates human mast cells to induce neutrophil transendothelial migration via mast cell-derived IL-1 alpha and beta. J Immunol 169:4522–30 14. McCurdy JD, Olynych TJ, Maher LH, et al (2003) Cutting edge: distinct Toll-like receptor 2 activators selectively induce different classes of mediator production from human mast cells. J Immunol 170:1625–9 15. Olynych TJ, Jakeman DL, Marshall JS (2006) Fungal zymosan induces leukotriene produc- tion by human mast cells through a dectin-1-dependent mechanism. J Allergy Clin Immunol 118:837–43 16. King CA, Anderson R, Marshall JS (2002) Dengue virus selectively induces human mast cell chemokine production. J Virol 76:8408–19 17. Jolly S, Detilleux J, Desmecht D (2004) Extensive mast cell degranulation in bovine res- piratory syncytial virus-associated paroxystic respiratory distress syndrome. Vet Immunol Immunopathol 97:125–36 18. Orinska Z, Bulanova E, Budagian V, et al (2005) TLR3-induced activation of mast cells modulates CD8 + T-cell recruitment. Blood 106:978–87 19. Zollner TM, Wichelhaus TA, Hartung A, et al (2000) Colonization with superantigen-producing Staphylococcus aureus is associated with increased severity of atopic dermatitis. Clin Exp Allergy 30:994–1000 20. Johnston SL, Ferrero F, Garcia ML, et al (2005) Oral oseltamivir improves pulmonary func- tion and reduces exacerbation frequency for influenza-infected children with asthma. Pediatr Infect Dis J 24: 225–32 21. Matsuse H, Kondo Y, Saeki S, et al (2005) Naturally occurring parainfluenza virus 3 infection in adults induces mild exacerbation of asthma associated with increased sputum concentra- tions of cysteinyl leukotrienes. Int Arch Allergy Immunol 138:267–72 22. Lukacs NW, Smit J, Lindell D, et al (2007) Respiratory syncytial virus-induced pulmonary disease and exacerbation of allergic asthma. Contrib Microbiol 14:68–82 23. Qureshi ST, Medzhitov R (2003) Toll-like receptors and their role in experimental models of microbial infection. Genes Immun 4:87–94 24. Pasare C, Medzhitov (2005) Toll-like receptors: linking innate and adaptive immunity. Adv Exp Med Biol 560:11–8 25. Uematsu S, Akira S (2006) Toll-like receptors and innate immunity. J Mol Med 84:712–25 26. Uematsu S, Akira S (2007) Toll-like receptors and type I interferons. J Biol Chem 282:15319–23 27. Malaviya R, Gao Z, Thankavel K, et al (1999) The mast cell tumor necrosis factor alpha response to FimH-expressing Escherichia coli is mediated by the glycosylphosphatidylinositol- anchored molecule CD48. Proc Natl Acad Sci USA 96:8110–5 128 J.S. Marshall et al.

28. Edelson BT, Li Z, Pappan LK, et al (2004). Mast cell-mediated inflammatory responses require the alpha 2 beta 1 integrin. Blood 103:2214–20 29. Edelson BT, Stricker TP, Li Z, et al (2006) Novel collectin/C1q receptor mediates mast cell activation and innate immunity. Blood 107:143–50 30. Mullaly SC, Kubes P (2007) Mast cell-expressed complement receptor, not TLR2, is the main detector of zymosan in peritonitis. Eur J Immunol 37:224–34 31. McCurdy JD, Lin TJ, Marshall JS (2001) Toll-like receptor 4-mediated activation of murine mast cells. J Leukoc Biol 70:977–84 32. Supajatura V, Ushio H, Nakao A, et al (2001) Protective roles of mast cells against enterobac- terial infection are mediated by Toll-like receptor 4. J Immunol 167:2250–6 33. Supajatura V, Ushio H, Nakao A, et al (2002) Differential responses of mast cell Toll-like receptors 2 and 4 in allergy and innate immunity. J Clin Invest 109:1351–9 34. Ikeda T, Funaba M (2003) Altered function of murine mast cells in response to lipopolysac- charide and peptidoglycan. Immunol Lett 88:21–6 35. Varadaradjalou S, Féger F, Thieblemont N, et al (2003) Toll-like receptor 2 (TLR2) and TLR4 differentially activate human mast cells. Eur J Immunol 33:899–906 36. Kulka M, Alexopoulou L, Flavell RA, et al (2004) Activation of mast cells by double- stranded RNA: evidence for activation through Toll-like receptor 3. J Allergy Clin Immunol 114:174–82 37. Bischoff SC, Krämer S (2007) Human mast cells, bacteria, and intestinal immunity. Immunol Rev 217:329–37 38. Kulka M, Metcalfe DD (2006) TLR3 activation inhibits human mast cell attachment to fibronectin and vitronectin. Mol Immunol 43:1579–86 39. Matsushima H., Yamada N, Matsue H, et al (2004) TLR3-, TLR7-, and TLR9-mediated production of proinflammatory cytokines and chemokines from murine connective tis- sue type skin-derived mast cells but not from bone marrow-derived mast cells. J Immunol 173:531–41 40. Orinska Z, Bulanova E, Budagian V, et al (2005) TLR3-induced activation of mast cells modulates CD8 + T-cell recruitment. Blood 106:978–87 41. Bonini S, Micera A, Iovieno A, et al (2005) Expression of Toll-like receptors in healthy and allergic conjunctiva. Ophthalmology 112:1528; discussion 1548–9 42. Bischoff SC (2007) Role of mast cells in allergic and non-allergic immune responses: com- parison of human and murine data. Nat Rev Immunol 7:93–104 43. Leal-Berumen I, Conlon P, Marshall JS (1994) IL-6 production by rat peritoneal mast cells is not necessarily preceded by histamine release and can be induced by bacterial lipopolysac- charide. J Immunol 152:5468–76 44. Okayama Y (2005) Mast cell-derived cytokine expression induced via Fc receptors and Toll- like receptors. Chem Immunol Allergy 87:101–10 45. Qiao H, Andrade MV, Lisboa FA, et al (2006) FcepsilonR1 and toll-like receptors mediate synergistic signals to markedly augment production of inflammatory cytokines in murine mast cells. Blood 107:610–8 46. Olynych TJ, Jakeman DL, Marshall JS (2006) Fungal zymosan induces leukotriene produc- tion by human mast cells through a dectin-1-dependent mechanism. J Allergy Clin Immunol 118:837–43 47. Rogers NC, Slack EC, Edwards AD, et al (2005) Syk-dependent cytokine induction by Dectin-1 reveals a novel pattern recognition pathway for C type lectins. Immunity 22:507–17 48. Brown GD (2006) Dectin-1: a signalling non-TLR pattern-recognition receptor. Nat Rev Immunol 6:33–43 49. Suram S, Brown GD, Ghosh M, et al (2006) Regulation of cytosolic phospholipase A2 acti- vation and cyclooxygenase 2 expression in macrophages by the beta-glucan receptor. J Biol Chem 281:5506–14 50. Okumura S, Kashiwakura J, Tomita H, et al (2003) Identification of specific gene expres- sion profiles in human mast cells mediated by Toll-like receptor 4 and FcepsilonRI. Blood 102:2547–54 Mast Cell and Basophils 129

51. Tkaczyk C, Jensen BM, Iwaki S, et al (2006) Adaptive and innate immune reactions regulating mast cell activation: from receptor-mediated signaling to responses. Immunol Allergy Clin North Am 26:427–50 52. King CA, Marshall JS, Alshurafa H, et al (2000) Release of vasoactive cytokines by antibody- enhanced dengue virus infection of a human mast cell/basophil line. J Virol 74:7146–50 53. King CA, Anderson R, Marshall JS (2002) Dengue virus selectively induces human mast cell chemokine production. J Virol 76:8408–19 54. Heib V, Becker M, Warger T, et al (2007) Mast cells are crucial for early inflammation, migra- tion of Langerhans cells, and CTL responses following topical application of TLR7 ligand in mice. Blood 110:946–53 55. Zhu FG, Marshall JS (2001) CpG-containing oligodeoxynucleotides induce TNF-alpha and IL-6 production but not degranulation from murine bone marrow-derived mast cells. J Leukoc Biol 69:253–62 56. Sundstrom JB, Little DM, Villinger F, et al (2004) Signaling through Toll-like receptors trig- gers HIV-1 replication in latently infected mast cells. J Immunol 172:4391–401 57. Yoshioka M, Fukuishi N, Iriguchi S, et al (2007) Lipoteichoic acid downregulates FcepsilonRI expression on human mast cells through Toll-like receptor 2. J Allergy Clin Immunol 120:452–61 58. Fehrenbach K, Port F, Grochowy G, et al (2007) Stimulation of mast cells via FcvarepsilonR1 and TLR2: the type of ligand determines the outcome. Mol Immunol 44:2087–94 59. Talreja J, Kabir MH, B Filla M, et al (2004) Histamine induces Toll-like receptor 2 and 4 expression in endothelial cells and enhances sensitivity to Gram-positive and Gram-negative bacterial cell wall components. Immunology 113:224–33 60. Nigo YI, Yamashita M, Hirahara K, et al (2006) Regulation of allergic airway inflammation through Toll-like receptor 4-mediated modification of mast cell function. Proc Natl Acad Sci USA 103:2286–91 61. Yamamoto K, Kawamura I, Ito J, et al (2006) Modification of allergic inflammation in murine model of rhinitis by different bacterial ligands: involvement of mast cells and dendritic cells. Clin Exp Allergy 36:760–9 62. Gondokaryono SP, Ushio H, Niyonsaba F, et al (2007) The extra domain A of fibronectin stimulates murine mast cells via Toll-like receptor 4. J Leukoc Biol. 82:657–65 63. McLachlan JB, Hart JP, Pizzo SV, et al (2003) Mast cell-derived tumor necrosis factor induces hypertrophy of draining lymph nodes during infection. Nat Immunol 4:1199–205 64. Jawdat DM, Rowden G, Marshall JS (2006) Mast cells have a pivotal role in TNF-independent lymph node hypertrophy and the mobilization of Langerhans cells in response to bacterial peptidoglycan. J Immunol 177:1755–62 65. Askenase PW, Van Loveren H, Kraeuter-Kops S, et al (1983) Defective elicitation of delayed-type hypersensitivity in W/Wv and SI/SId mast cell-deficient mice. J Immunol 131: 2687–94 66. Galli SJ, Hammel I (1984) Unequivocal delayed hypersensitivity in mast cell-deficient and beige mice. Science 226:710–3 67. Kerdel FA, Belsito DV, Scotto-Chinnici R, et al (1987) Mast cell participation during the elicitation of murine allergic contact hypersensitivity. J Invest Dermatol 88:686–90 68. Bryce PJ, Miller ML, Miyajima I, et al (2005) Immune sensitization in the skin is enhanced by antigen-independent effects of IgE on mast cells. Novartis Found Symp 271:15–24; discus- sion 24–38, 95–9 69. Gelfand EW (2004) Inflammatory mediators in allergic rhinitis. J Allergy Clin Immunol 114:S135–8 70. Irani AMA, Schecter NM, Craig SS, et al (1986) Two types of human mast cell that have distinct neutral protease compositions. Proc Natl Acad Sci USA. 83:4464–9 71. Enerback L, Pipkorn U, Granerus G (1986) Intraepithelial migration of nasal mucosal mast cells in hay fever. Int Arch Allergy Appl Immunol 80:44–51 72. Pawankar R, Ra C (1996) Heterogeneity of mast cells and T cells in the nasal mucosa. J Allergy Clin Immunol 98:S248–62 130 J.S. Marshall et al.

73. Kawabori S, Kanai N, Tosho T (1995) Proliferative activity of mast cells in allergic nasal mucosa. Clin Exp Allergy 25:173–8 74. Pawankar R (2005) Mast cells in allergic airway disease. In: Mast cells in allergic diseases. Saito H, Okayama Y (eds). New York: Karger, Basel 75. Ahn K, Takai S, Pawankar R et al (2000) Regulation of chymase production in human mast cell progenitors. J Allergy Clin Immunol 106:321–8 76. Creticos PS, Peters SP, Adkinson NF, Jr., et al (1984) Peptide leukotriene release after antigen challenge in patients sensitive to ragweed. N Engl J Med 310:1626–30 77. Freeland HS, Pipkorn U, Schleimer RP, et al (1989) Leukotriene B4 as a mediator of early and late reactions to antigen in humans: the effect of systemic glucocorticoid treatment in vivo. J Allergy Clin Immunol 83:634–42 78. Proud D, Bailey GS, Naclerio RM, et al (1992) Tryptase and histamine as markers to evaluate mast cell activation during the responses to nasal challenge with allergen, cold, dry air, and hyperosmolar solutions. J Allergy Clin Immunol 89:1098–110 79. Shaw RJ, Fitzharris P, Cromwell O, et al (1985) Allergen-induced release of sulphidopeptide leukotrienes (SRS-A) and LTB4 in allergic rhinitis. Allergy 40:1–6 80. Baraniuk JN (1997) Pathogenesis of allergic rhinitis. J Allergy Clin Immunol 99:S763–72 81. Sanico AM, Koliatsos VE, Stanisz AM, et al (1999) Neural hyperresponsiveness and nerve growth factor in allergic rhinitis. Int Arch Allergy Immunol 118:154–8 82. Sanico AM, Stanisz AM, Gleeson TD, et al (2000) Nerve growth factor expression and release in allergic inflammatory disease of the upper airways. Am J Respir Crit Care Med 161:1631–5 83. Pawankar R, Okuda M, Yssel H, et al (1997) Nasal mast cells exhibit increased expression of the FcεRI, CD40L, IL-4 and IL-13 and can induce IgE synthesis in B cells. J Clin Invest 99:1492–1499 84. Welker P, Grabbe J, Gibbs B, et al (2000) Nerve growth factor-beta induces mast-cell marker expression during in vitro culture of human umbilical cord blood cells. Immunology 99:418–26 85. Pastorello EA., Incorvaia C, Ortolani C, et al (1995) Studies on the relationship between the level of specific IgE antibodies and the clinical expression of allergy: 1. Definition of levels distinguishing patients with symptomatic from patients with asymptomatic allergy to common allergens. J Allergy Clin Immunol 96:580 86. Pawankar R (2001) Revisting the roles of mast cells and its relation to local IgE synthesis. Am J Rhinol 14:309 87. Durham SR., Gould HJ, Thienes CP, et al (1997) Expression of epsilon germ-line gene tran- scripts and mRNA for the epsilon heavy chain of IgE in nasal B cells and the effects of topical corticosteroid. Eur J Immunol 27:2899 88. Pawankar R, Yamagishi S, Takizawa R, et al (2000) Local IgE synthesis: Its functional sig- nificance and strategy for new therapy. J Rhinol 39:69 89. Kalesnikoff J, Huber M, Lam V, et al (2001) Monomeric IgE stimulates signaling pathways in mast cells that lead to cytokine production and cell survival. Immunity 14:801–1 90. Cruse G, Kaur D, Yang W, et al (2005) Activation of human lung mast cells by monomeric immunoglobulin E. Eur Respir J 25:858–63 91. Matsuda K, Piliponsky AM, Iikura M, et al (2005) Monomeric IgE enhances human mast cell chemokine production: IL-4 augments and dexamethasone suppresses the response. J Allergy Clin Immunol 116:1357–63 92. Burrows B, Martinez FD, Halonen M, et al (1989) Association of asthma with serum IgE levels and skin-test reactivity to allergens. N Engl J Med 320:271–7 93. Sears MR, Burrows B, Flannery EM, et al (1991) Relation between airway responsiveness and serum IgE in children with asthma and in apparently normal children. N Engl J Med 325:1067–71 94. Takeda K, Hamelmann E, Joetham A, et al (2007) Development of eosinophilic airway inflam- mation and airway hyperresponsiveness in mast cell-deficient mice. J Exp Med 186:449–54. The Authors Journal compilation _c Blackwell Publishing Ltd, Clinical and Experimental Allergy, 38:4–18 Mast cells in allergic disease 15 Mast Cell and Basophils 131

95. Williams CM, Galli SJ (2000) Mast cells can amplify airway reactivity and features of chronic inflammation in an asthma model in mice. J Exp Med 192:455–62 96. Taube C, Wei X, Swasey CH, et al (2004) Mast cells, Fc epsilon RI, and IL-13 are required for development of airway hyperresponsiveness after aerosolized allergen exposure in the absence of adjuvant. J Immunol 172:6398–406 97. Saito H, Okayama Y (2005) Mast cells in allergic diseases. New York: Karger, Basel 98. Brightling CE, Symon FA, Holgate ST, et al (2003) Interleukin-4 and -13 expression is colocalized to mast cells within the airway smooth muscle in asthma. Clin Exp Allergy 33:1711–6 99. Hsieh FH, Lam BK, Penrose JF, et al (2001) T helper cell type 2 cytokines coordinately regulate IgE-dependent cysteinyl leukotriene production by human cord blood-derived mast cells: profound induction of leukotriene C4 synthase expression by IL-4. J Exp Med 193:123–33 100. Ochi H, De Jesus NH, Hsieh FH, et al (2000) IL-4 and -5 prime human mast cells for differ- ent profiles of IgE-dependent cytokine production. Proc Natl Acad Sci USA 97:10509–13 101. Galli SJ (2000) The c-kit receptor, stem cell factor, and mast cells. What each is teaching us about the others. Am J Pathol 142:965–74 102. Metcalfe DD (1997) Mast cells. Physiol Rev 77:1033–79 103. Matsushima H, Yamada N, Matsue H, et al (2004) TLR3-, TLR7-, and TLR9-mediated production of proinflammatory cytokines and chemokines from murine connective tissue type skin-derived mast cells but not from bone marrow-derived mast cells. J Immunol 173:531–41 104. Amin K, Ludviksdottir D, Janson C, et al (2000) Inflammation and structural changes in the airways of patients with atopic and nonatopic asthma. BHR Group. Am J Respir Crit Care Med 162:2295–301 105. Ribatti D, Polimeno G, Vacca A, et al (2002) Correlation of bone marrow angiogenesis and mast cells with tryptase activity in myelodysplastic syndromes. Leukemia 16:1680–84 106. Boesiger J, Tsai M, Maurer M, et al (1998) Mast cells can secrete vascular permeability factor/vascular endothelial cell growth factor and exhibit enhanced release after immu- noglobulin E-dependent upregulation of Fc epsilon receptor I expression. J Exp Med 188: 1135–45 107. Grutzkau A, Kruger-Krasagakes S, Baumeister H, et al (1998) Synthesis, storage, and release of vascular endothelial growth factor/vascular permeability factor (VEGF/VPF) by human mast cells: implications for the biological significance of VEGF206. Mol Biol Cell 9:875–84 108. Abdel-Majid RM, Marshall JS (2004) Prostaglandin E2 induces degranulation-independent production of vascular endothelial growth factor by human mast cells. J Immunol 172:1227–36 109. Abelson MB (2001) Allergic diseases of the eye. Philadelphia: W.B. Saunders 110. Friedlaender MH (1991) Current concepts in ocular allergy. Ann Allergy 67:5–10, 13 111. Ono SJ, Abelson MB (2005) Allergic conjunctivitis: update on pathophysiology and pros- pects for future treatment. J Allergy Clin Immunol 115:118–22 112. Allansmith MR (1990) Immunology of the external ocular tissues. J Am Optom Assoc 61:S16–22 113. Macleod JD, Anderson DF, Baddeley SM, et al (1997) Immunolocalization of cytokines to mast cells in normal and allergic conjunctiva. Clin Exp Allergy 27:1328–34 114. Church MK, McGill JI (2002) Human ocular mast cells. Curr Opin Allergy Clin Immunol 2:419–22 115. Ahluwalia P, Anderson DF, Wilson SJ, et al (2001) Nedocromil sodium and levocabastine reduce the symptoms of conjunctival allergen challenge by different mechanisms. J Allergy Clin Immunol 108:449–54 116. Nakamura T, Ohbayashi M, Toda M, et al (2005) A specific CCR3 chemokine recep- tor antagonist inhibits both early and late phase allergic inflammation in the conjunctiva. Immunol Res 33:213–21 132 J.S. Marshall et al.

117. Damsgaard TE, Olesen AB, Sorensen FB, et al (1997) Mast cells and atopic dermatitis. Stereological quantification of mast cells in atopic dermatitis and normal human skin. Arch Dermatol Res 289:256–60 118. Jarvikallio A, Naukkarinen A, Harvima IT, et al (1997) Quantitative analysis of tryptase- and chy- mase containing mast cells in atopic dermatitis and nummular eczema. Br J Dermatol 136:871–7 119. Groneberg DA, Bester C, Grutzkau A, et al (2005) Mast cells and vasculature in atopic dermatitis – potential stimulus of neoangiogenesis. Allergy 60:90–7 120. Steinhoff M, Neisius U, Ikoma A, et al (2003) Proteinase-activated receptor-2 mediates itch: a novel pathway for pruritus in human skin. J Neurosci 23:6176–80 121. Badertscher K, Bronnimann M, Karlen S, et al (2005) Mast cell chymase is increased in chronic atopic dermatitis but not in psoriasis. Arch Dermatol Res 296:503–6 122. Weidinger S, Rummler L, Klopp N, et al (2005) Association study of mast cell chymase polymorphisms with atopy. Allergy 60:1256–61 123. Fischer M, Harvima IT, Carvalho RF, et al (2006) Mast cell CD30 ligand is upregulated in cutaneous inflammation and mediates degranulation-independent chemokine secretion. J Clin Invest 116:2748–56 124. Jarvikallio A, Harvima IT, Naukkarinen A (2003) Mast cells, nerves and neuropeptides in atopic dermatitis and nummular eczema. Arch Dermatol Res 295:2–7 125. Ansel JC, Kaynard AH, Armstrong CA, et al (1996) Skin-nervous system interactions. J Invest Dermatol 106:198–204 126. Groneberg DA, Serowka F, Peckenschneider N, et al (2005) Gene expression and regula- tion of nerve growth factor in atopic dermatitis mast cells and the human mast cell line-1. J Neuroimmunol 161:87–92 127. Groneberg DA, Welker P, Fischer TC, et al (2003) Down-regulation of vasoactive intestinal polypeptide receptor expression in atopic dermatitis. J Allergy Clin Immunol 111:1099–105 128. Caughey GH (2006) Tryptase genetics and anaphylaxis. J Allergy Clin Immunol 117:1411–4 129. Schwartz LB (2004) Effector cells of anaphylaxis: mast cells and basophils. Novartis Found Symp 257:65–74; discussion 74–69, 98–100, 276–85x 130. Schwartz LB, Yunginger JW, Miller J, et al (1989) Time course of appearance and disap- pearance of human mast cell tryptase in the circulation after anaphylaxis. J Clin Invest 83:1551–5 131. Gilfillan AM, Tkaczyk C (2006) Integrated signalling pathways for mast-cell activation. Nat Rev Immunol 6:218–30 132. Tkaczyk C, Okayama Y, Woolhiser MR, et al (2002) Activation of human mast cells through the high affinity IgG receptor. Mol Immunol 38:1289–93 133. Woolhiser MR, Brockow K, Metcalfe DD (2004) Activation of human mast cells by aggre- gated IgG through FcgammaRI: additive effects of C3a. Clin Immunol 110:172–80 134. Okayama Y, Kirshenbaum AS, Metcalfe DD (2000) Expression of a functional high-affinity IgG receptor, Fc gamma RI, on human mast cells: up-regulation by IFN-gamma. J Immunol 164:4332–9 135. Genovese A, Stellato C, Patella V, et al (1996) Contrast media are incomplete secretagogues acting on human basophils and mast cells isolated from heart and lung, but not skin tissue. Int J Clin Lab Res 26:192–8 136. Marone G, Bova M, Detoraki A, et al (2004) The human heart as a shock organ in anaphy- laxis. Novartis Found Symp 257:133–49; discussion 149–60, 276–185 137. Montrucchio G, Alloatti G, Camussi G (2000) pathophysiology. Physiol Rev 80:1669–9 138. McCurdy JD, Olynych TJ, Maher LH, et al (2003) Cutting edge: distinct Toll-like receptor 2 activators selectively induce different classes of mediator production from human mast cells. J Immunol 170:1625–9 139. Kulka M, Alexopoulou L, Flavell RA, et al (2004) Activation of mast cells by double- stranded RNA: evidence for activation through Toll-like receptor 3. J Allergy Clin Immunol 114:174–82 Mast Cell and Basophils 133

140. Applequist SE, Wallin RP, Ljunggren HG (2002) Variable expression of Toll-like receptor in murine innate and adaptive immune cell lines. Int Immunol 14:1065–74 141. Diebold SS, Kaisho T, Hemmi H, et al (2004) Innate antiviral responses by means of TLR7- mediated recognition of single-stranded RNA. Science 303(5663):1529–31 142. Florian S, Sonneck K, Czerny M, et al (2006) Detection of novel leukocyte differentiation antigens on basophils and mast cells by HLDA8 antibodies. Allergy 61:1054–62 143. Füreder W, Agis H, Semper H, et al (1995) Differential response of human basophils and mast cells to recombinant chemokines. Ann Hematol 70:251–8 144. Füreder W, Agis H, Willheim M, et al (1995) Differential expression of complement recep- tors on human basophils and mast cells. Evidence for mast cell heterogeneity and CD88/ C5aR expression on skin mast cells. J Immunol 155:3152–60 145. Ghebrehiwet B, Kew RR, Gruber BL, et al (1995) Murine mast cells express two types of C1q receptors that are involved in the induction of chemotaxis and chemokinesis. J Immunol 155:2614–9 146. Hasan U, Chaffois C, Gaillard C, et al (2005) Human TLR10 is a functional receptor, expressed by B cells and plasmacytoid dendritic cells, which activates gene transcription through MyD88. J Immunol 174:2942–50 147. Heil F, Hemmi H, Hochrein H, et al (2004) Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science 303:1526–9 148. Inomata N, Tomita H, Ikezawa Z, et al (2005) Differential gene expression profile between cord blood progenitor-derived and adult progenitor-derived human mast cells. Immunol Lett 98:265–71 149. Kawai T, Akira S (2007) TLR signaling. Semin Immunol 19:24–32 150. Komiya A, Nagase H, Okugawa S, et al (2006) Expression and function of toll-like receptors in human basophils. Int Arch Allergy Immunol 140:S23–S27 151. Kubo Y, Fukuishi N, Yoshioka M, et al (2007) Bacterial components regulate the expression of Toll-like receptor 4 on human mast cells. Inflamm Res 56:70–5 152. Malaviya R, Georges A (2002) Regulation of mast cell-mediated innate immunity during early response to bacterial infection. Clin Rev Allergy Immunol 22:189–204 153. Malaviya R, Ross EA, MacGregor JI, et al (1994) Mast cell phagocytosis of FimH- expressing enterobacteria. J Immunol 152:1907–14 154. Marshall JS (2004) Mast-cell responses to pathogens. Nat Rev Immunol 4:787–99 155. Miyake K (2007) Innate immune sensing of pathogens and danger signals by cell surface Toll-like receptors. Semin Immunol 19:3–10 156. Rosenkranz AR, Coxon A, Maurer M, et al (1998) Impaired mast cell development and innate immunity in Mac-1 (CD11b/CD18, CR3)-deficient mice. J Immunol 161:6463–7 157. Sabroe I, Jones EC, Usher LR, et al (2002) Toll-like receptor (TLR)2 and TLR4 in human peripheral blood granulocytes: a critical role for monocytes in leukocyte lipopolysaccharide responses. J Immunol 168:4701–10 158. Thangam EB, Venkatesha RT, Zaidi AK, et al (2005) Airway smooth muscle cells enhance C3a-induced mast cell degranulation following cell-cell contact. FASEB J 19:798–800 159. Wu L, Feng BS, He SH, et al (2007) Bacterial peptidoglycan breaks down intestinal toler- ance via mast cell activation: The role of TLR2 and NOD2. Immunol Cell Biol 85:538–45